1
|
Yang Q, Hong J, Fu J, Liu X, Zheng X, Jiang J, Zhu A, Chen L, Lin H, Sun P. Integrated multi-omics analysis and experimental verification reveal the involvement of the PI3K/Akt signaling pathway in myometrial fibrosis of adenomyosis. Sci Rep 2025; 15:13637. [PMID: 40254638 PMCID: PMC12010003 DOI: 10.1038/s41598-025-98369-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 04/10/2025] [Indexed: 04/22/2025] Open
Abstract
Adenomyosis (AM) is characterized as a chronic and progressive disorder with limited therapeutic strategies available. Myometrial fibrosis is a prominent pathological feature of AM, yet the underlying molecular mechanisms remain elusive. The present study conducted a comparative analysis using proteomics and metabolomics to investigate myometrial fibrosis and its underlying mechanisms. Liquid chromatography-tandem mass spectrometry (LC-MS/MS) was utilized to analyze adenomyotic and normal myometrial tissues from ten AM patients who underwent hysterectomy with myometrial fibrosis confirmed by Masson staining. This analysis established comprehensive proteomic and metabolomic profiles of AM patients and revealed widespread alterations in the proteome and metabolome within normal and fibrotic myometrium. Key proteins and signaling pathways linked to myometrial fibrogenesis were identified based on proteomic data. The integrated analysis showed significant associations between proteomic and metabolomic data and highlighted the critical role of the PI3K/AKT signaling pathway. Immunohistochemistry and Spearman's correlation analysis suggested a relationship between myometrial fibrosis and the metaplasia of myometrial stromal cells into myofibroblasts. Subsequent experiments identified crucial proteins and signaling pathways involved in myometrial fibrosis, indicating an association with the activation of the PI3K/AKT signaling pathway in myofibroblasts. Notably, PI3K/AKT inhibitors may contribute to the effective alleviation of myometrial fibrosis. This study is the first to demonstrate that myometrial fibrosis represents a critical pathological mechanism in AM through multi-omics methods and to elucidate the crucial role of the PI3K/AKT signaling pathway in this process. These findings provide valuable insights into the pathophysiology of AM and suggest antifibrotic treatment as a promising therapeutic strategy.
Collapse
Affiliation(s)
- Qiaomei Yang
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, Fujian, China
- Department of Gynecology, Fujian Maternity and Child Health Hospital, Fuzhou, Fujian, China
- National Key Clinical Specialty Construction Program of China (Gynecology), Fujian Maternity and Child Health Hospital (Fujian Obstetrics and Gynecology Hospital), Fuzhou, Fujian, China
| | - Jingxuan Hong
- Department of Cardiology, Fujian Provincial Hospital Facilitated to Fuzhou University, Fujian Provincial Center for Cardiovascular Epidemiology Research and Prevention, Fuzhou, China
- Department of Cardiology, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, China
| | - Jianhui Fu
- Department of Pathology, Fujian Maternity and Child Health Hospital College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, Fujian, China
| | - Xianhua Liu
- Department of Pathology, Fujian Maternity and Child Health Hospital College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, Fujian, China
| | - Xinye Zheng
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, Fujian, China
- Department of Gynecology, Fujian Maternity and Child Health Hospital, Fuzhou, Fujian, China
- National Key Clinical Specialty Construction Program of China (Gynecology), Fujian Maternity and Child Health Hospital (Fujian Obstetrics and Gynecology Hospital), Fuzhou, Fujian, China
| | - Junying Jiang
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, Fujian, China
- Department of Gynecology, Fujian Maternity and Child Health Hospital, Fuzhou, Fujian, China
- National Key Clinical Specialty Construction Program of China (Gynecology), Fujian Maternity and Child Health Hospital (Fujian Obstetrics and Gynecology Hospital), Fuzhou, Fujian, China
| | - An Zhu
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Li Chen
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, Fujian, China
- Department of Gynecology, Fujian Maternity and Child Health Hospital, Fuzhou, Fujian, China
- National Key Clinical Specialty Construction Program of China (Gynecology), Fujian Maternity and Child Health Hospital (Fujian Obstetrics and Gynecology Hospital), Fuzhou, Fujian, China
| | - Hao Lin
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, Fujian, China
- Department of Gynecology, Fujian Maternity and Child Health Hospital, Fuzhou, Fujian, China
- National Key Clinical Specialty Construction Program of China (Gynecology), Fujian Maternity and Child Health Hospital (Fujian Obstetrics and Gynecology Hospital), Fuzhou, Fujian, China
| | - PengMing Sun
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, Fujian, China.
- Department of Gynecology, Fujian Maternity and Child Health Hospital, Fuzhou, Fujian, China.
- National Key Clinical Specialty Construction Program of China (Gynecology), Fujian Maternity and Child Health Hospital (Fujian Obstetrics and Gynecology Hospital), Fuzhou, Fujian, China.
| |
Collapse
|
2
|
Tang X, Ran X, Liang Z, Zhuang H, Yan X, Feng C, Qureshi A, Gao Y, Shen L. Screening biomarkers for autism spectrum disorder using plasma proteomics combined with machine learning methods. Clin Chim Acta 2025; 565:120018. [PMID: 39481511 DOI: 10.1016/j.cca.2024.120018] [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: 05/20/2024] [Revised: 10/22/2024] [Accepted: 10/23/2024] [Indexed: 11/02/2024]
Abstract
BACKGROUND AND AIMS Autism spectrum disorder (ASD) is a common neurodevelopmental disorder in children. Early intervention is effective. Investigation of novel blood biomarkers of ASD facilitates early detection and intervention. MATERIALS AND METHODS Sequential window acquisition of all theoretical spectra-mass spectrometry (SWATH-MS)-based proteomics technology and 30 DSM-V defined ASD cases versus age- and sex-matched controls were initially evaluated, and candidate biomarkers were screened using machine learning methods. Candidate biomarkers were validated by targeted proteomics multiple reaction monitoring (MRM) analysis using an independent group of 30 ASD cases vs. controls. RESULTS Fifty-one differentially expressed proteins (DEPs) were identified by SWATH analysis. They were associated with the immune response, complements and coagulation cascade pathways, and apolipoprotein-related metabolic pathways. Machine learning analysis screened 10 proteins as biomarker combinations (TFRC, PPBP, APCS, ALDH1A1, CD5L, SPARC, FGG, SHBG, S100A9, and PF4V1). In the MRM analysis, four proteins (PPBP, APCS, FGG, and PF4V1) were significantly different between the groups, and their combination as a screening indicator showed high potential (AUC = 0.8087, 95 % confidence interval 0.6904-0.9252, p < 0.0001). CONCLUSIONS Our study provides data that suggests that a few plasma proteins have potential use in screening for ASD.
Collapse
Affiliation(s)
- Xiaoxiao Tang
- College of Life Science and Oceanography, Shenzhen University, Shenzhen 518071, PR China
| | - Xiaoqian Ran
- College of Life Science and Oceanography, Shenzhen University, Shenzhen 518071, PR China
| | - Zhiyuan Liang
- College of Life Science and Oceanography, Shenzhen University, Shenzhen 518071, PR China
| | - Hongbin Zhuang
- College of Life Science and Oceanography, Shenzhen University, Shenzhen 518071, PR China
| | - Xi Yan
- College of Life Science and Oceanography, Shenzhen University, Shenzhen 518071, PR China
| | - Chengyun Feng
- Maternal and Child Health Hospital of Baoan, Shenzhen 518100, PR China
| | - Ayesha Qureshi
- College of Life Science and Oceanography, Shenzhen University, Shenzhen 518071, PR China
| | - Yan Gao
- Maternal and Child Health Hospital of Baoan, Shenzhen 518100, PR China
| | - Liming Shen
- College of Life Science and Oceanography, Shenzhen University, Shenzhen 518071, PR China; Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research, Institutions, Shenzhen 518055, PR China.
| |
Collapse
|
3
|
Hewitson L, Mathews JA, Devlin M, Schutte C, Lee J, German DC. Blood biomarker discovery for autism spectrum disorder: A proteomic analysis. PLoS One 2024; 19:e0302951. [PMID: 39700097 DOI: 10.1371/journal.pone.0302951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 09/25/2024] [Indexed: 12/21/2024] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by deficits in social communication and social interaction and restricted, repetitive patterns of behavior, interests, or activities. Given the lack of specific pharmacological therapy for ASD and the clinical heterogeneity of the disorder, current biomarker research efforts are geared mainly toward identifying markers for determining ASD risk or for assisting with a diagnosis. A wide range of putative biological markers for ASD are currently being investigated. Proteomic analyses indicate that the levels of many proteins in plasma/serum are altered in ASD, suggesting that a panel of proteins may provide a blood biomarker for ASD. Serum samples from 76 boys with ASD and 78 typically developing (TD) boys, 2-10 years of age, were analyzed to identify possible early biological markers for ASD. Proteomic analysis of serum was performed using SomaLogic's SOMAScanTM assay 1.3K platform. A total of 1,125 proteins were analyzed. There were 86 downregulated proteins and 52 upregulated proteins in ASD (FDR < 0.05). Combining three different algorithms, we found a panel of 12 proteins that identified ASD with an area under the curve (AUC) = 0.8790±0.0572, with specificity and sensitivity of 0.8530±0.1076 and 0.8324±0.1137, respectively. All 12 proteins were significantly different in ASD compared with TD boys, and 4 were significantly correlated with ASD severity as measured by ADOS total scores. Using machine learning methods, a panel of serum proteins was identified that may be useful as a blood biomarker for ASD in boys. Further verification of the protein biomarker panel with independent test sets is warranted.
Collapse
Affiliation(s)
- Laura Hewitson
- The Johnson Center for Child Health and Development, Austin, TX, United States of America
| | - Jeremy A Mathews
- Bioinformatics & Computational Biology Program, Departments of Mathematical Sciences and Biological Sciences, University of Texas at Dallas, Dallas, TX, United States of America
| | - Morgan Devlin
- The Johnson Center for Child Health and Development, Austin, TX, United States of America
| | - Claire Schutte
- The Johnson Center for Child Health and Development, Austin, TX, United States of America
| | - Jeon Lee
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX, United States of America
| | - Dwight C German
- Department of Psychiatry, UT Southwestern Medical Center, Dallas, TX, United States of America
| |
Collapse
|
4
|
Ghunaim L, Ali Agha ASA, Aburjai T. Integrating Artificial Intelligence and Advanced Genomic Technologies in Unraveling Autism Spectrum Disorder and Gastrointestinal Comorbidities: A Multidisciplinary Approach to Precision Medicine. JORDAN JOURNAL OF PHARMACEUTICAL SCIENCES 2024; 17:567-581. [DOI: 10.35516/jjps.v17i3.2410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
This article explores the potential impact of Artificial Intelligence (AI), Machine Learning (ML), CRISPR-Cas9 gene editing, and single-cell RNA sequencing on improving our understanding and management of Autism Spectrum Disorder (ASD) and its gastrointestinal (GI) comorbidities. It examines how these technologies illuminate the complex interplay between the gut and the brain, identifying specific enzyme deficiencies and microbial imbalances linked to GI symptoms in ASD. By leveraging AI and ML, personalized intervention strategies are developed through the analysis of genomic, proteomic, and environmental data, enhancing our ability to predict and address GI issues in ASD. Additionally, CRISPR-Cas9 gene editing holds promise for correcting genetic abnormalities related to enzyme production, potentially offering precise treatments. Single-cell RNA sequencing provides critical insights into the cellular diversity of the ASD gut, uncovering new therapeutic targets. The article highlights the transformative potential of these technologies while addressing the associated challenges and ethical considerations. It underscores the necessity of a multidisciplinary approach to fully harness their benefits and discusses the significant progress and emerging trends in the field, emphasizing the role of technological advancements in advancing precision medicine for ASD and its GI comorbidities.
Collapse
|
5
|
Gulati S, Narayan CL, Mahesan A, Kamila G, Kapoor S, Chaturvedi PK, Scaria V, Velpandian T, Jauhari P, Chakrabarty B, Datta SKR, Pandey RM. Transmethylation and Oxidative Biomarkers in Children with Autism Spectrum Disorder: A Cross Sectional Study. J Autism Dev Disord 2024:10.1007/s10803-024-06542-9. [PMID: 39230783 DOI: 10.1007/s10803-024-06542-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/23/2024] [Indexed: 09/05/2024]
Abstract
We aimed to investigate the potential role of biomarkers of transmethylation, oxidative stress, and mitochondrial dysfunction in children with Autism Spectrum Disorder (ASD) by comparing them with that of typically developing children (TDC) controls. We also tried to correlate them with severity of autism, sensory issues, behavioural comorbidities and developmental quotients 119 with ASD and 52 age and sex matched typically developing children (TDC) controls were enrolled excluding those with chronic-illness or on any antioxidant therapy/multivitamins/anti-epileptic drugs. Median levels of biomarkers - serum homocysteine, cysteine, methionine, urine uric acid-to-creatinine ratio, arterial lactate, serum vitamin E, vitamin B12, folate, Nε-carboxymethyllysine, Nω- carboxymethylarginine (CMA), dityrosine and MTHFR C677T polymorphism were calculated. Children with ASD were further characterised using Childhood Autism Rating Scale-2, Childhood behavioural checklist, child sensory profile 2 caregiver questionnaire, Developmental Profile 3 for any correlation with the various biomarker levels. The median level of serum homocysteine in ASD group was 9 μmol/L(Range, 7- 16μmol/L), which was significantly higher than controls 7 μmol/L(Range, 4- 11μmol/L)(p=0.01). The prevalence of hyper-homocystinemia(>15μmol/L) was 13.4% in ASD as compared to 3.8% in controls with a significant difference(p=0.04). Dityrosine level was higher among ASD children when compared to TDC (9.8 vs 2.2 counts per second(cps), p<0.001). No significant correlation was found between prevalence of hyperhomocysteinemia and severity of autism/DQ/behavioural issues. No significant difference was found between the median levels of other biomarkers. Results support possible role of transmethylation defects and oxidative stress in ASD pathogenesis. Further studies are warranted for a better understanding of ASD pathogenesis.
Collapse
Affiliation(s)
- Sheffali Gulati
- Centre of Excellence & Advanced Research for Childhood Neurodevelopmental Disorders, Child Neurology Division, Department of Pediatrics, All India Institute of Medical Sciences, New Delhi, India.
| | - Chinthana L Narayan
- Centre of Excellence & Advanced Research for Childhood Neurodevelopmental Disorders, Child Neurology Division, Department of Pediatrics, All India Institute of Medical Sciences, New Delhi, India
| | - Aakash Mahesan
- Centre of Excellence & Advanced Research for Childhood Neurodevelopmental Disorders, Child Neurology Division, Department of Pediatrics, All India Institute of Medical Sciences, New Delhi, India
| | - Gautam Kamila
- Centre of Excellence & Advanced Research for Childhood Neurodevelopmental Disorders, Child Neurology Division, Department of Pediatrics, All India Institute of Medical Sciences, New Delhi, India
| | - Seema Kapoor
- Division of Genetics, Department of Pediatrics, Maulana Azad Medical College, New Delhi, India
| | | | - Vinod Scaria
- Institue of Genomics and Integrative Biology, New Delhi, India
| | | | - Prashant Jauhari
- Centre of Excellence & Advanced Research for Childhood Neurodevelopmental Disorders, Child Neurology Division, Department of Pediatrics, All India Institute of Medical Sciences, New Delhi, India
| | - Biswaroop Chakrabarty
- Centre of Excellence & Advanced Research for Childhood Neurodevelopmental Disorders, Child Neurology Division, Department of Pediatrics, All India Institute of Medical Sciences, New Delhi, India
| | | | - R M Pandey
- Department of Biostatistics, AIIMS, New Delhi, India
| |
Collapse
|
6
|
Deng M, Li X, Shi D, Fan Q, Zhang H, Wang Z, Wang Y, Xiao Z. iTRAQ-Based Serum Proteomic Analysis Reveals Multifactorial Cellular Function Impairment and Aggravated Systematic Inflammation in Drug-free Obsessive-Compulsive Disorders. ACS Chem Neurosci 2024; 15:3053-3063. [PMID: 39120470 DOI: 10.1021/acschemneuro.4c00317] [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: 08/10/2024] Open
Abstract
Obsessive-compulsive disorder (OCD) is a debilitating mental disorder with obvious difficulties in treatment. Its pathogenesis has not been fully elucidated. Further understanding of etiology and mechanism needs to be explored further. We employed the isobaric tag for relative and absolute quantitation (iTRAQ)-based proteomic analysis to compare serum proteome profile between OCD patients and healthy controls, in order to find out the possible mechanism of OCD in the downstream biological process. Eighty-one drug-free OCD patients and 78 healthy controls were enrolled. A total of 475 proteins were identified. Totally, 80 proteins with p < 0.05 were selected for gene set enrichment analysis (GSEA), and only those with a fold change ≥1.2 and q value <0.2 between groups were accepted as differentially expressed proteins (DEPs). We observed a significant enrichment of immuno-inflammation-related pathways, along with intriguing expression trends that immuno-inflammation-related proteins were upregulated in GSEA. After that, 2 up-regulated proteins and 13 down-regulated ones were accepted as DEP. According to the available literature, most of the DEPs have not been reported in OCD. These DEPs were enriched in 121 gene ontology (GO) terms, including hepatocyte growth factor receptor activity, angiogenin-PRI complex, and so on. DEPs were enriched in pathways including adherens junction in the Kyoto Encyclopedia of Genes and Genomes (KEGG) database. Alterations in DEPs including STXBP5L, GRN, and ANG were validated in OCD animal models. Our study suggested that OCD patients manifested multifactorial impairment in neuronal or non-neuronal cellular function under the inflammatory background. Further research employing larger sample sizes, longitudinal design, stratified analysis, and multiomics methodology will be needed. Experiments in laboratories were essential in illuminating the mechanism.
Collapse
Affiliation(s)
- Miaohan Deng
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Xia Li
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Dongdong Shi
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Qing Fan
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Haiyin Zhang
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Zhen Wang
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Yuan Wang
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Zeping Xiao
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| |
Collapse
|
7
|
Nussinov R, Yavuz BR, Demirel HC, Arici MK, Jang H, Tuncbag N. Review: Cancer and neurodevelopmental disorders: multi-scale reasoning and computational guide. Front Cell Dev Biol 2024; 12:1376639. [PMID: 39015651 PMCID: PMC11249571 DOI: 10.3389/fcell.2024.1376639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 06/10/2024] [Indexed: 07/18/2024] Open
Abstract
The connection and causality between cancer and neurodevelopmental disorders have been puzzling. How can the same cellular pathways, proteins, and mutations lead to pathologies with vastly different clinical presentations? And why do individuals with neurodevelopmental disorders, such as autism and schizophrenia, face higher chances of cancer emerging throughout their lifetime? Our broad review emphasizes the multi-scale aspect of this type of reasoning. As these examples demonstrate, rather than focusing on a specific organ system or disease, we aim at the new understanding that can be gained. Within this framework, our review calls attention to computational strategies which can be powerful in discovering connections, causalities, predicting clinical outcomes, and are vital for drug discovery. Thus, rather than centering on the clinical features, we draw on the rapidly increasing data on the molecular level, including mutations, isoforms, three-dimensional structures, and expression levels of the respective disease-associated genes. Their integrated analysis, together with chromatin states, can delineate how, despite being connected, neurodevelopmental disorders and cancer differ, and how the same mutations can lead to different clinical symptoms. Here, we seek to uncover the emerging connection between cancer, including pediatric tumors, and neurodevelopmental disorders, and the tantalizing questions that this connection raises.
Collapse
Affiliation(s)
- Ruth Nussinov
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Cancer Innovation Laboratory, National Cancer Institute, Frederick, MD, United States
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv-Yafo, Israel
| | - Bengi Ruken Yavuz
- Cancer Innovation Laboratory, National Cancer Institute, Frederick, MD, United States
| | | | - M. Kaan Arici
- Graduate School of Informatics, Middle East Technical University, Ankara, Türkiye
| | - Hyunbum Jang
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Cancer Innovation Laboratory, National Cancer Institute, Frederick, MD, United States
| | - Nurcan Tuncbag
- Department of Chemical and Biological Engineering, Koc University, Istanbul, Türkiye
- School of Medicine, Koc University, Istanbul, Türkiye
- Koc University Research Center for Translational Medicine (KUTTAM), Istanbul, Türkiye
| |
Collapse
|
8
|
Xie X, Zhou R, Fang Z, Zhang Y, Wang Q, Liu X. Seeing beyond words: Visualizing autism spectrum disorder biomarker insights. Heliyon 2024; 10:e30420. [PMID: 38694128 PMCID: PMC11061761 DOI: 10.1016/j.heliyon.2024.e30420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 05/04/2024] Open
Abstract
Objective This study employs bibliometric and visual analysis to elucidate global research trends in Autism Spectrum Disorder (ASD) biomarkers, identify critical research focal points, and discuss the potential integration of diverse biomarker modalities for precise ASD assessment. Methods A comprehensive bibliometric analysis was conducted using data from the Web of Science Core Collection database until December 31, 2022. Visualization tools, including R, VOSviewer, CiteSpace, and gCLUTO, were utilized to examine collaborative networks, co-citation patterns, and keyword associations among countries, institutions, authors, journals, documents, and keywords. Results ASD biomarker research emerged in 2004, accumulating a corpus of 4348 documents by December 31, 2022. The United States, with 1574 publications and an H-index of 213, emerged as the most prolific and influential country. The University of California, Davis, contributed significantly with 346 publications and an H-index of 69, making it the leading institution. Concerning journals, the Journal of Autism and Developmental Disorders, Autism Research, and PLOS ONE were the top three publishers of ASD biomarker-related articles among a total of 1140 academic journals. Co-citation and keyword analyses revealed research hotspots in genetics, imaging, oxidative stress, neuroinflammation, gut microbiota, and eye tracking. Emerging topics included "DNA methylation," "eye tracking," "metabolomics," and "resting-state fMRI." Conclusion The field of ASD biomarker research is dynamically evolving. Future endeavors should prioritize individual stratification, methodological standardization, the harmonious integration of biomarker modalities, and longitudinal studies to advance the precision of ASD diagnosis and treatment.
Collapse
Affiliation(s)
- Xinyue Xie
- The First Affiliated Hospital of Henan University of Chinese Medicine, Pediatrics Hospital, Zhengzhou, Henan, 450000, China
- Henan University of Chinese Medicine, School of Pediatrics, Zhengzhou, Henan, 450046, China
| | - Rongyi Zhou
- The First Affiliated Hospital of Henan University of Chinese Medicine, Pediatrics Hospital, Zhengzhou, Henan, 450000, China
- Henan University of Chinese Medicine, School of Pediatrics, Zhengzhou, Henan, 450046, China
| | - Zihan Fang
- Henan University of Chinese Medicine, School of Pediatrics, Zhengzhou, Henan, 450046, China
| | - Yongting Zhang
- The First Affiliated Hospital of Henan University of Chinese Medicine, Pediatrics Hospital, Zhengzhou, Henan, 450000, China
- Henan University of Chinese Medicine, School of Pediatrics, Zhengzhou, Henan, 450046, China
| | - Qirong Wang
- Henan University of Chinese Medicine, School of Pediatrics, Zhengzhou, Henan, 450046, China
| | - Xiaomian Liu
- Henan University of Chinese Medicine, School of Medicine, Zhengzhou, Henan, 450046, China
| |
Collapse
|
9
|
Liu J, Tan Y, Zhang F, Wang Y, Chen S, Zhang N, Dai W, Zhou L, Li J. Metabolomic analysis of plasma biomarkers in children with autism spectrum disorders. MedComm (Beijing) 2024; 5:e488. [PMID: 38420161 PMCID: PMC10901282 DOI: 10.1002/mco2.488] [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: 06/28/2023] [Revised: 01/10/2024] [Accepted: 01/22/2024] [Indexed: 03/02/2024] Open
Abstract
Autism spectrum disorder (ASD) presents a significant risk to human well-being and has emerged as a worldwide public health concern. Twenty-eight children with ASD and 33 healthy children (HC) were selected for the quantitative determination of their plasma metabolites using an ultraperformance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS) platform. A total of 1997 metabolites were detected in the study cohort, from which 116 metabolites were found to be differentially expressed between the ASD and HC groups. Through analytical algorithms such as least absolute shrinkage selection operator (LASSO), support vector machine (SVM), and random forest (RF), three potential metabolic markers were identified as FAHFA (18:1(9Z)/9-O-18:0), DL-2-hydroxystearic acid, and 7(S),17(S)-dihydroxy-8(E),10(Z),13(Z),15(E),19(Z)-docosapentaenoic acid. These metabolites demonstrated superior performance in distinguishing the ASD group from the HC group, as indicated by the area under curves (AUCs) of 0.935, 0.897, and 0.963 for the three candidate biomarkers, respectively. The samples were divided into training and validation sets according to 7:3. Diagnostic models were constructed using logistic regression (LR), SVM, and RF. The constructed three-biomarker diagnostic model also exhibited strong discriminatory efficacy. These findings contribute to advancing our understanding of the underlying mechanisms involved in the occurrence of ASD and provide a valuable reference for clinical diagnosis.
Collapse
Affiliation(s)
- Jun Liu
- Medical Research CenterYue Bei People's Hospital, Shantou University Medical CollegeShaoguanChina
| | - Yuhua Tan
- Shaoguan Maternal and Child Health HospitalShaoguanChina
| | - Fan Zhang
- Medical Research CenterYue Bei People's Hospital, Shantou University Medical CollegeShaoguanChina
| | - Yan Wang
- Shaoguan Maternal and Child Health HospitalShaoguanChina
| | - Shu Chen
- Shaoguan Maternal and Child Health HospitalShaoguanChina
| | - Na Zhang
- Shaoguan Maternal and Child Health HospitalShaoguanChina
| | - Wenjie Dai
- Medical Research CenterYue Bei People's Hospital, Shantou University Medical CollegeShaoguanChina
| | - Liqing Zhou
- Medical Research CenterYue Bei People's Hospital, Shantou University Medical CollegeShaoguanChina
| | - Ji‐Cheng Li
- Medical Research CenterYue Bei People's Hospital, Shantou University Medical CollegeShaoguanChina
- Institute of Cell BiologyZhejiang UniversityHangzhouChina
- Major Disease Biomarkers Research LaboratorySchool of Basic Medical Science, Henan UniversityKaifengChina
| |
Collapse
|
10
|
Bhuvaneshwar K, Gusev Y. Translational bioinformatics and data science for biomarker discovery in mental health: an analytical review. Brief Bioinform 2024; 25:bbae098. [PMID: 38493340 PMCID: PMC10944574 DOI: 10.1093/bib/bbae098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 01/23/2024] [Accepted: 02/18/2024] [Indexed: 03/18/2024] Open
Abstract
Translational bioinformatics and data science play a crucial role in biomarker discovery as it enables translational research and helps to bridge the gap between the bench research and the bedside clinical applications. Thanks to newer and faster molecular profiling technologies and reducing costs, there are many opportunities for researchers to explore the molecular and physiological mechanisms of diseases. Biomarker discovery enables researchers to better characterize patients, enables early detection and intervention/prevention and predicts treatment responses. Due to increasing prevalence and rising treatment costs, mental health (MH) disorders have become an important venue for biomarker discovery with the goal of improved patient diagnostics, treatment and care. Exploration of underlying biological mechanisms is the key to the understanding of pathogenesis and pathophysiology of MH disorders. In an effort to better understand the underlying mechanisms of MH disorders, we reviewed the major accomplishments in the MH space from a bioinformatics and data science perspective, summarized existing knowledge derived from molecular and cellular data and described challenges and areas of opportunities in this space.
Collapse
Affiliation(s)
- Krithika Bhuvaneshwar
- Innovation Center for Biomedical Informatics (ICBI), Georgetown University, Washington DC, 20007, USA
| | - Yuriy Gusev
- Innovation Center for Biomedical Informatics (ICBI), Georgetown University, Washington DC, 20007, USA
| |
Collapse
|
11
|
Lin P, Zhang Q, Sun J, Li Q, Li D, Zhu M, Fu X, Zhao L, Wang M, Lou X, Chen Q, Liang K, Zhu Y, Qu C, Li Z, Ma P, Wang R, Liu H, Dong K, Guo X, Cheng X, Sun Y, Sun J. A comparison between children and adolescents with autism spectrum disorders and healthy controls in biomedical factors, trace elements, and microbiota biomarkers: a meta-analysis. Front Psychiatry 2024; 14:1318637. [PMID: 38283894 PMCID: PMC10813399 DOI: 10.3389/fpsyt.2023.1318637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 12/13/2023] [Indexed: 01/30/2024] Open
Abstract
Introduction Autism spectrum disorder (ASD) is a multifaceted developmental condition that commonly appears during early childhood. The etiology of ASD remains multifactorial and not yet fully understood. The identification of biomarkers may provide insights into the underlying mechanisms and pathophysiology of the disorder. The present study aimed to explore the causes of ASD by investigating the key biomedical markers, trace elements, and microbiota factors between children with autism spectrum disorder (ASD) and control subjects. Methods Medline, PubMed, ProQuest, EMBASE, Cochrane Library, PsycINFO, Web of Science, and EMBSCO databases have been searched for publications from 2012 to 2023 with no language restrictions using the population, intervention, control, and outcome (PICO) approach. Keywords including "autism spectrum disorder," "oxytocin," "GABA," "Serotonin," "CRP," "IL-6," "Fe," "Zn," "Cu," and "gut microbiota" were used for the search. The Joanna Briggs Institute (JBI) critical appraisal checklist was used to assess the article quality, and a random model was used to assess the mean difference and standardized difference between ASD and the control group in all biomedical markers, trace elements, and microbiota factors. Results From 76,217 records, 43 studies met the inclusion and exclusion criteria and were included in this meta-analysis. The pooled analyses showed that children with ASD had significantly lower levels of oxytocin (mean differences, MD = -45.691, 95% confidence interval, CI: -61.667, -29.717), iron (MD = -3.203, 95% CI: -4.891, -1.514), and zinc (MD = -6.707, 95% CI: -12.691, -0.722), lower relative abundance of Bifidobacterium (MD = -1.321, 95% CI: -2.403, -0.238) and Parabacteroides (MD = -0.081, 95% CI: -0.148, -0.013), higher levels of c-reactive protein, CRP (MD = 0.401, 95% CI: 0.036, 0.772), and GABA (MD = 0.115, 95% CI: 0.045, 0.186), and higher relative abundance of Bacteroides (MD = 1.386, 95% CI: 0.717, 2.055) and Clostridium (MD = 0.281, 95% CI: 0.035, 0.526) when compared with controls. The results of the overall analyses were stable after performing the sensitivity analyses. Additionally, no substantial publication bias was observed among the studies. Interpretation Children with ASD have significantly higher levels of CRP and GABA, lower levels of oxytocin, iron, and zinc, lower relative abundance of Bifidobacterium and Parabacteroides, and higher relative abundance of Faecalibacterium, Bacteroides, and Clostridium when compared with controls. These results suggest that these indicators may be a potential biomarker panel for the diagnosis or determining therapeutic targets of ASD. Furthermore, large, sample-based, and randomized controlled trials are needed to confirm these results.
Collapse
Affiliation(s)
- Ping Lin
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai, China
- Department of Clinical Laboratory, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qianwen Zhang
- Key Laboratory of Digital Technology in Medical Diagnostics of Zhejiang Province, Hangzhou, China
- Hangzhou Calibra Diagnostics, Hangzhou, China
| | - Junyu Sun
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Qingtian Li
- College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dan Li
- Department of Clinical Laboratory, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mengyuan Zhu
- Department of Clinical Laboratory, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaomei Fu
- Department of Clinical Laboratory, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ling Zhao
- Department of Clinical Laboratory, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mengxia Wang
- Department of Clinical Laboratory, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoyan Lou
- Department of Clinical Laboratory, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qing Chen
- Department of Clinical Laboratory, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kangyi Liang
- Department of Clinical Laboratory, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuxin Zhu
- Department of Clinical Laboratory, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Caiwei Qu
- Department of Clinical Laboratory, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhenhua Li
- Department of Clinical Laboratory, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Peijun Ma
- Department of Clinical Laboratory, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Renyu Wang
- Department of Clinical Laboratory, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huafen Liu
- Key Laboratory of Digital Technology in Medical Diagnostics of Zhejiang Province, Hangzhou, China
- Hangzhou Calibra Diagnostics, Hangzhou, China
| | - Ke Dong
- Institute for Global Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaokui Guo
- Institute for Global Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xunjia Cheng
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yang Sun
- Institute of Arthritis Research, Shanghai Academy of Chinese Medical Sciences, Shanghai, China
| | - Jing Sun
- School of Medicine and Dentistry, Institute for Integrated Intelligence and Systems, Griffith University, Gold Coast Campus, Gold Coast, QLD, Australia
- Charles Sturt University, Orange, NSW, Australia
| |
Collapse
|
12
|
Tiao MM, Chang YC, Ou LS, Hung CF, Khwepeya M. An Exploration of Pediatricians' Professional Identities: A Q-Methodology Study. Healthcare (Basel) 2024; 12:144. [PMID: 38255033 PMCID: PMC10815713 DOI: 10.3390/healthcare12020144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 12/22/2023] [Accepted: 01/04/2024] [Indexed: 01/24/2024] Open
Abstract
Professional identities may influence a wide range of attitudes, ethical standards, professional commitments and patient safety. This study aimed to explore the important elements that comprise pediatricians' professional identities. A Q-methodology was used to identify the similarities and differences in professional identity. Forty pediatricians were recruited from two tertiary referral hospitals in Taiwan. A list of statements was developed by five attending physicians and three residents. R software was used to analyze the Q-sorts to load the viewpoints and formulate the viewpoint arrays. Additional qualitative data-one-to-one personal interviews-were analyzed. Twenty-eight of forty pediatricians, 11 males and 17 females, with an average age of 39.9 (27-62) years, were associated with four viewpoints. We labeled the four viewpoints identified for professional identity as (1) professional recognition, (2) patient communication, (3) empathy and (4) insight. The professional recognition viewpoint comprised of youngest participants-28-36 years-with the majority as residents (77.8%), while the empathy viewpoint comprised the oldest participants-38-62 years-with all as attending physicians. All participants in the empathy and insight viewpoints were married. This study found professional identity to be a multifaceted concept for pediatricians, especially in the areas of professional recognition, patient communication, empathy and insight into patient care.
Collapse
Affiliation(s)
- Mao-Meng Tiao
- Department of Pediatrics, Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan;
| | - Yu-Che Chang
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan;
- Chang Gung Medical Education Research Centre, Chang Gung Memorial Hospital, Linkou, Taoyuan 333, Taiwan;
- Department of Emergency Medicine, Chang Gung Memorial Hospital, Linkou, Taoyuan 333, Taiwan
| | - Liang-Shiou Ou
- Chang Gung Medical Education Research Centre, Chang Gung Memorial Hospital, Linkou, Taoyuan 333, Taiwan;
| | - Chi-Fa Hung
- Department of Psychology, Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan;
| | - Madalitso Khwepeya
- Chang Gung Medical Education Research Centre, Chang Gung Memorial Hospital, Linkou, Taoyuan 333, Taiwan;
| |
Collapse
|
13
|
Vernocchi P, Marangelo C, Guerrera S, Del Chierico F, Guarrasi V, Gardini S, Conte F, Paci P, Ianiro G, Gasbarrini A, Vicari S, Putignani L. Gut microbiota functional profiling in autism spectrum disorders: bacterial VOCs and related metabolic pathways acting as disease biomarkers and predictors. Front Microbiol 2023; 14:1287350. [PMID: 38192296 PMCID: PMC10773764 DOI: 10.3389/fmicb.2023.1287350] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 11/14/2023] [Indexed: 01/10/2024] Open
Abstract
Background Autism spectrum disorder (ASD) is a multifactorial neurodevelopmental disorder. Major interplays between the gastrointestinal (GI) tract and the central nervous system (CNS) seem to be driven by gut microbiota (GM). Herein, we provide a GM functional characterization, based on GM metabolomics, mapping of bacterial biochemical pathways, and anamnestic, clinical, and nutritional patient metadata. Methods Fecal samples collected from children with ASD and neurotypical children were analyzed by gas-chromatography mass spectrometry coupled with solid phase microextraction (GC-MS/SPME) to determine volatile organic compounds (VOCs) associated with the metataxonomic approach by 16S rRNA gene sequencing. Multivariate and univariate statistical analyses assessed differential VOC profiles and relationships with ASD anamnestic and clinical features for biomarker discovery. Multiple web-based and machine learning (ML) models identified metabolic predictors of disease and network analyses correlated GM ecological and metabolic patterns. Results The GM core volatilome for all ASD patients was characterized by a high concentration of 1-pentanol, 1-butanol, phenyl ethyl alcohol; benzeneacetaldehyde, octadecanal, tetradecanal; methyl isobutyl ketone, 2-hexanone, acetone; acetic, propanoic, 3-methyl-butanoic and 2-methyl-propanoic acids; indole and skatole; and o-cymene. Patients were stratified based on age, GI symptoms, and ASD severity symptoms. Disease risk prediction allowed us to associate butanoic acid with subjects older than 5 years, indole with the absence of GI symptoms and low disease severity, propanoic acid with the ASD risk group, and p-cymene with ASD symptoms, all based on the predictive CBCL-EXT scale. The HistGradientBoostingClassifier model classified ASD patients vs. CTRLs by an accuracy of 89%, based on methyl isobutyl ketone, benzeneacetaldehyde, phenyl ethyl alcohol, ethanol, butanoic acid, octadecane, acetic acid, skatole, and tetradecanal features. LogisticRegression models corroborated methyl isobutyl ketone, benzeneacetaldehyde, phenyl ethyl alcohol, skatole, and acetic acid as ASD predictors. Conclusion Our results will aid the development of advanced clinical decision support systems (CDSSs), assisted by ML models, for advanced ASD-personalized medicine, based on omics data integrated into electronic health/medical records. Furthermore, new ASD screening strategies based on GM-related predictors could be used to improve ASD risk assessment by uncovering novel ASD onset and risk predictors.
Collapse
Affiliation(s)
- Pamela Vernocchi
- Research Unit of Human Microbiome, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Chiara Marangelo
- Research Unit of Human Microbiome, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Silvia Guerrera
- Child and Adolescent Neuropsychiatry Unit, Department of Neuroscience, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Federica Del Chierico
- Research Unit of Human Microbiome, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | | | | | - Federica Conte
- Institute for Systems Analysis and Computer Science “Antonio Ruberti”, National Research Council, Rome, Italy
| | - Paola Paci
- Department of Computer, Control and Management Engineering, Sapienza University of Rome, Rome, Italy
| | - Gianluca Ianiro
- CEMAD Digestive Disease Center, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Antonio Gasbarrini
- CEMAD Digestive Disease Center, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Stefano Vicari
- Child and Adolescent Neuropsychiatry Unit, Department of Neuroscience, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
- Life Sciences and Public Health Department, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Lorenza Putignani
- Unit of Microbiomics and Research Unit of Human Microbiome, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| |
Collapse
|
14
|
Kaupper CS, Blaauwendraad SM, Cecil CAM, Mulder RH, Gaillard R, Goncalves R, Borggraefe I, Koletzko B, Jaddoe VWV. Cord Blood Metabolite Profiles and Their Association with Autistic Traits in Childhood. Metabolites 2023; 13:1140. [PMID: 37999236 PMCID: PMC10672851 DOI: 10.3390/metabo13111140] [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: 09/28/2023] [Revised: 11/02/2023] [Accepted: 11/03/2023] [Indexed: 11/25/2023] Open
Abstract
Autism Spectrum Disorder (ASD) is a diverse neurodevelopmental condition. Gene-environmental interactions in early stages of life might alter metabolic pathways, possibly contributing to ASD pathophysiology. Metabolomics may serve as a tool to identify underlying metabolic mechanisms contributing to ASD phenotype and could help to unravel its complex etiology. In a population-based, prospective cohort study among 783 mother-child pairs, cord blood serum concentrations of amino acids, non-esterified fatty acids, phospholipids, and carnitines were obtained using liquid chromatography coupled with tandem mass spectrometry. Autistic traits were measured at the children's ages of 6 (n = 716) and 13 (n = 648) years using the parent-reported Social Responsiveness Scale. Lower cord blood concentrations of SM.C.39.2 and NEFA16:1/16:0 were associated with higher autistic traits among 6-year-old children, adjusted for sex and age at outcome. After more stringent adjustment for confounders, no significant associations of cord blood metabolites and autistic traits at ages 6 and 13 were detected. Differences in lipid metabolism (SM and NEFA) might be involved in ASD-related pathways and are worth further investigation.
Collapse
Affiliation(s)
- Christin S. Kaupper
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, 3000 CA Rotterdam, The Netherlands (R.G.)
- Department of Pediatrics, Sophia’s Children’s Hospital, Erasmus MC, University Medical Center Rotterdam, 3000 CA Rotterdam, The Netherlands
| | - Sophia M. Blaauwendraad
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, 3000 CA Rotterdam, The Netherlands (R.G.)
- Department of Pediatrics, Sophia’s Children’s Hospital, Erasmus MC, University Medical Center Rotterdam, 3000 CA Rotterdam, The Netherlands
| | - Charlotte A. M. Cecil
- Department of Child and Adolescent Psychiatry, Erasmus MC, University Medical Center Rotterdam, 3000 CA Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus MC, 3000 CA Rotterdam, The Netherlands
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, 2333 ZC Leiden, The Netherlands
| | - Rosa H. Mulder
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, 3000 CA Rotterdam, The Netherlands (R.G.)
- Department of Child and Adolescent Psychiatry, Erasmus MC, University Medical Center Rotterdam, 3000 CA Rotterdam, The Netherlands
| | - Romy Gaillard
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, 3000 CA Rotterdam, The Netherlands (R.G.)
- Department of Pediatrics, Sophia’s Children’s Hospital, Erasmus MC, University Medical Center Rotterdam, 3000 CA Rotterdam, The Netherlands
| | - Romy Goncalves
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, 3000 CA Rotterdam, The Netherlands (R.G.)
- Department of Pediatrics, Sophia’s Children’s Hospital, Erasmus MC, University Medical Center Rotterdam, 3000 CA Rotterdam, The Netherlands
| | - Ingo Borggraefe
- Division of Pediatric Neurology, Developmental Medicine and Social Pediatrics, Comprehensive Epilepsy Center for Children and Adolescents, Dr. von Hauner Children’s Hospital, LMU University Hospitals, LMU—Ludwig-Maximilians Universität, 80337 Munich, Germany
| | - Berthold Koletzko
- Division of Metabolic and Nutritional Medicine, Department of Pediatrics, Dr. von Hauner Children’s Hospital, LMU University Hospitals, LMU—Ludwig-Maximilians Universität, 80337 Munich, Germany
| | - Vincent W. V. Jaddoe
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, 3000 CA Rotterdam, The Netherlands (R.G.)
- Department of Pediatrics, Sophia’s Children’s Hospital, Erasmus MC, University Medical Center Rotterdam, 3000 CA Rotterdam, The Netherlands
| |
Collapse
|
15
|
Cao X, Tang X, Feng C, Lin J, Zhang H, Liu Q, Zheng Q, Zhuang H, Liu X, Li H, Khan NU, Shen L. A Systematic Investigation of Complement and Coagulation-Related Protein in Autism Spectrum Disorder Using Multiple Reaction Monitoring Technology. Neurosci Bull 2023; 39:1623-1637. [PMID: 37031449 PMCID: PMC10603015 DOI: 10.1007/s12264-023-01055-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Accepted: 02/02/2023] [Indexed: 04/10/2023] Open
Abstract
Autism spectrum disorder (ASD) is one of the common neurodevelopmental disorders in children. Its etiology and pathogenesis are poorly understood. Previous studies have suggested potential changes in the complement and coagulation pathways in individuals with ASD. In this study, using multiple reactions monitoring proteomic technology, 16 of the 33 proteins involved in this pathway were identified as differentially-expressed proteins in plasma between children with ASD and controls. Among them, CFHR3, C4BPB, C4BPA, CFH, C9, SERPIND1, C8A, F9, and F11 were found to be altered in the plasma of children with ASD for the first time. SERPIND1 expression was positively correlated with the CARS score. Using the machine learning method, we obtained a panel composed of 12 differentially-expressed proteins with diagnostic potential for ASD. We also reviewed the proteins changed in this pathway in the brain and blood of patients with ASD. The complement and coagulation pathways may be activated in the peripheral blood of children with ASD and play a key role in the pathogenesis of ASD.
Collapse
Affiliation(s)
- Xueshan Cao
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, 518060, China
- College of Physics and Optoelectronics Engineering, Shenzhen University, Shenzhen, 518060, China
| | - Xiaoxiao Tang
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, 518060, China
| | - Chengyun Feng
- Maternal and Child Health Hospital of Baoan, Shenzhen, 518100, China
| | - Jing Lin
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, 518060, China
| | - Huajie Zhang
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, 518060, China
| | - Qiong Liu
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, 518060, China
- College of Physics and Optoelectronics Engineering, Shenzhen University, Shenzhen, 518060, China
| | - Qihong Zheng
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, 518060, China
| | - Hongbin Zhuang
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, 518060, China
| | - Xukun Liu
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, 518060, China
| | - Haiying Li
- Department of Endocrinology, Guiyang First People's Hospital, Guiyang, 550002, China
| | - Naseer Ullah Khan
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, 518060, China
| | - Liming Shen
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, 518060, China.
- Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 518055, China.
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, Shenzhen, 518060, China.
| |
Collapse
|
16
|
Malesci R, Lombardi M, Abenante V, Fratestefano F, Del Vecchio V, Fetoni AR, Troisi J. A Systematic Review on Metabolomics Analysis in Hearing Impairment: Is It a Possible Tool in Understanding Auditory Pathologies? Int J Mol Sci 2023; 24:15188. [PMID: 37894867 PMCID: PMC10607298 DOI: 10.3390/ijms242015188] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 09/27/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023] Open
Abstract
With more than 466 million people affected, hearing loss represents the most common sensory pathology worldwide. Despite its widespread occurrence, much remains to be explored, particularly concerning the intricate pathogenic mechanisms underlying its diverse phenotypes. In this context, metabolomics emerges as a promising approach. Indeed, lying downstream from molecular biology's central dogma, the metabolome reflects both genetic traits and environmental influences. Furthermore, its dynamic nature facilitates well-defined changes during disease states, making metabolomic analysis a unique lens into the mechanisms underpinning various hearing impairment forms. Hence, these investigations may pave the way for improved diagnostic strategies, personalized interventions and targeted treatments, ultimately enhancing the clinical management of affected individuals. In this comprehensive review, we discuss findings from 20 original articles, including human and animal studies. Existing literature highlights specific metabolic changes associated with hearing loss and ototoxicity of certain compounds. Nevertheless, numerous critical issues have emerged from the study of the current state of the art, with the lack of standardization of methods, significant heterogeneity in the studies and often small sample sizes being the main limiting factors for the reliability of these findings. Therefore, these results should serve as a stepping stone for future research aimed at addressing the aforementioned challenges.
Collapse
Affiliation(s)
- Rita Malesci
- Department of Neuroscience, Reproductive Sciences and Dentistry (Audiology and Vestibology Service), University of Naples Federico II, 80138 Napoli, Italy; (V.D.V.); (A.R.F.)
| | - Martina Lombardi
- Theoreo srl, Spin off Company of the University of Salerno, Via Degli Ulivi 3, 84090 Montecorvino Pugliano, Italy; (V.A.); (F.F.); (J.T.)
- Department of Chemistry and Biology “A. Zambelli”, University of Salerno, 84084 Fisciano, Italy
- European Institute of Metabolomics (EIM) Foundation ETS, G. Puccini, 2, 84081 Baronissi, Italy
| | - Vera Abenante
- Theoreo srl, Spin off Company of the University of Salerno, Via Degli Ulivi 3, 84090 Montecorvino Pugliano, Italy; (V.A.); (F.F.); (J.T.)
| | - Federica Fratestefano
- Theoreo srl, Spin off Company of the University of Salerno, Via Degli Ulivi 3, 84090 Montecorvino Pugliano, Italy; (V.A.); (F.F.); (J.T.)
| | - Valeria Del Vecchio
- Department of Neuroscience, Reproductive Sciences and Dentistry (Audiology and Vestibology Service), University of Naples Federico II, 80138 Napoli, Italy; (V.D.V.); (A.R.F.)
| | - Anna Rita Fetoni
- Department of Neuroscience, Reproductive Sciences and Dentistry (Audiology and Vestibology Service), University of Naples Federico II, 80138 Napoli, Italy; (V.D.V.); (A.R.F.)
| | - Jacopo Troisi
- Theoreo srl, Spin off Company of the University of Salerno, Via Degli Ulivi 3, 84090 Montecorvino Pugliano, Italy; (V.A.); (F.F.); (J.T.)
- Department of Chemistry and Biology “A. Zambelli”, University of Salerno, 84084 Fisciano, Italy
- European Institute of Metabolomics (EIM) Foundation ETS, G. Puccini, 2, 84081 Baronissi, Italy
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy
| |
Collapse
|
17
|
Hollstein R, Peron A, Wendt KS, Parenti I. Editorial: Pathogenic mechanisms in neurodevelopmental disorders: advances in cellular models and multi-omics approaches. Front Cell Dev Biol 2023; 11:1296885. [PMID: 37868909 PMCID: PMC10588624 DOI: 10.3389/fcell.2023.1296885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 09/27/2023] [Indexed: 10/24/2023] Open
Affiliation(s)
- R. Hollstein
- Institute of Human Genetics, University of Bonn and University Hospital Bonn, Bonn, Germany
| | - A. Peron
- Medical Genetics, Meyer Children’s Hospital IRCCS, Florence, Italy
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, Università Degli Studi Di Firenze, Florence, Italy
| | - K. S. Wendt
- Department of Cell Biology, Erasmus MC, Rotterdam, Netherlands
| | - I. Parenti
- Institute of Human Genetics, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| |
Collapse
|
18
|
Smith AM, Donley ELR, Ney DM, Amaral DG, Burrier RE, Natowicz MR. Metabolomic biomarkers in autism: identification of complex dysregulations of cellular bioenergetics. Front Psychiatry 2023; 14:1249578. [PMID: 37928922 PMCID: PMC10622772 DOI: 10.3389/fpsyt.2023.1249578] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 08/30/2023] [Indexed: 11/07/2023] Open
Abstract
Autism Spectrum Disorder (ASD or autism) is a phenotypically and etiologically heterogeneous condition. Identifying biomarkers of clinically significant metabolic subtypes of autism could improve understanding of its underlying pathophysiology and potentially lead to more targeted interventions. We hypothesized that the application of metabolite-based biomarker techniques using decision thresholds derived from quantitative measurements could identify autism-associated subpopulations. Metabolomic profiling was carried out in a case-control study of 499 autistic and 209 typically developing (TYP) children, ages 18-48 months, enrolled in the Children's Autism Metabolome Project (CAMP; ClinicalTrials.gov Identifier: NCT02548442). Fifty-four metabolites, associated with amino acid, organic acid, acylcarnitine and purine metabolism as well as microbiome-associated metabolites, were quantified using liquid chromatography-tandem mass spectrometry. Using quantitative thresholds, the concentrations of 4 metabolites and 149 ratios of metabolites were identified as biomarkers, each identifying subpopulations of 4.5-11% of the CAMP autistic population. A subset of 42 biomarkers could identify CAMP autistic individuals with 72% sensitivity and 90% specificity. Many participants were identified by several metabolic biomarkers. Using hierarchical clustering, 30 clusters of biomarkers were created based on participants' biomarker profiles. Metabolic changes associated with the clusters suggest that altered regulation of cellular metabolism, especially of mitochondrial bioenergetics, were common metabolic phenotypes in this cohort of autistic participants. Autism severity and cognitive and developmental impairment were associated with increased lactate, many lactate containing ratios, and the number of biomarker clusters a participant displayed. These studies provide evidence that metabolic phenotyping is feasible and that defined autistic subgroups can lead to enhanced understanding of the underlying pathophysiology and potentially suggest pathways for targeted metabolic treatments.
Collapse
Affiliation(s)
- Alan M. Smith
- Stemina Biomarker Discovery, Inc, Madison, WI, United States
| | | | - Denise M. Ney
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, United States
| | - David G. Amaral
- Department of Psychiatry and Behavioral Sciences, The MIND Institute, University of California, Davis, Davis, CA, United States
| | | | - Marvin R. Natowicz
- Pathology and Laboratory Medicine, Genomic Medicine, Neurological and Pediatrics Institutes, Cleveland Clinic, Cleveland, OH, United States
| |
Collapse
|
19
|
Tang X, Feng C, Zhao Y, Zhang H, Gao Y, Cao X, Hong Q, Lin J, Zhuang H, Feng Y, Wang H, Shen L. A study of genetic heterogeneity in autism spectrum disorders based on plasma proteomic and metabolomic analysis: multiomics study of autism heterogeneity. MedComm (Beijing) 2023; 4:e380. [PMID: 37752942 PMCID: PMC10518435 DOI: 10.1002/mco2.380] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 08/04/2023] [Accepted: 08/24/2023] [Indexed: 09/28/2023] Open
Abstract
Genetic heterogeneity poses a challenge to research and clinical translation of autism spectrum disorder (ASD). In this study, we conducted a plasma proteomic and metabolomic study of children with ASD with and without risk genes (de novo mutation) and controls to explore the impact of genetic heterogeneity on the search for biomarkers for ASD. In terms of the proteomic and metabolomic profiles, the groups of children with ASD carrying and those not carrying de novo mutation tended to cluster and overlap, and integrating them yielded differentially expressed proteins and differential metabolites that effectively distinguished ASD from controls. The mechanisms associated with them focus on several common and previously reported mechanisms. Proteomics results highlight the role of complement, inflammation and immunity, and cell adhesion. The main pathways of metabolic perturbations include amino acid, vitamin, glycerophospholipid, tryptophan, and glutamates metabolic pathways and solute carriers-related pathways. Integrating the two omics analyses revealed that L-glutamic acid and malate dehydrogenase may play key roles in the pathogenesis of ASD. These results suggest that children with ASD may have important underlying common mechanisms. They are not only potential therapeutic targets for ASD but also important contributors to the study of biomarkers for the disease.
Collapse
Affiliation(s)
- Xiaoxiao Tang
- College of Life Science and OceanographyShenzhen UniversityShenzhenP. R. China
| | - Chengyun Feng
- Maternal and Child Health Hospital of BaoanShenzhenP. R. China
| | - Yuxi Zhao
- College of Life Science and OceanographyShenzhen UniversityShenzhenP. R. China
| | - Huajie Zhang
- College of Life Science and OceanographyShenzhen UniversityShenzhenP. R. China
| | - Yan Gao
- Maternal and Child Health Hospital of BaoanShenzhenP. R. China
| | - Xueshan Cao
- College of Life Science and OceanographyShenzhen UniversityShenzhenP. R. China
| | - Qi Hong
- Maternal and Child Health Hospital of BaoanShenzhenP. R. China
| | - Jing Lin
- College of Life Science and OceanographyShenzhen UniversityShenzhenP. R. China
| | - Hongbin Zhuang
- College of Life Science and OceanographyShenzhen UniversityShenzhenP. R. China
| | - Yuying Feng
- College of Life Science and OceanographyShenzhen UniversityShenzhenP. R. China
| | - Hanghang Wang
- College of Life Science and OceanographyShenzhen UniversityShenzhenP. R. China
| | - Liming Shen
- College of Life Science and OceanographyShenzhen UniversityShenzhenP. R. China
- Shenzhen‐Hong Kong Institute of Brain Science‐Shenzhen Fundamental Research InstitutionsShenzhenP. R. China
- Shenzhen Key Laboratory of Marine Biotechnology and EcologyShenzhenP. R. China
| |
Collapse
|
20
|
Siracusano M, Arturi L, Riccioni A, Noto A, Mussap M, Mazzone L. Metabolomics: Perspectives on Clinical Employment in Autism Spectrum Disorder. Int J Mol Sci 2023; 24:13404. [PMID: 37686207 PMCID: PMC10487559 DOI: 10.3390/ijms241713404] [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: 07/06/2023] [Revised: 08/09/2023] [Accepted: 08/25/2023] [Indexed: 09/10/2023] Open
Abstract
Precision medicine is imminent, and metabolomics is one of the main actors on stage. We summarize and discuss the current literature on the clinical application of metabolomic techniques as a possible tool to improve early diagnosis of autism spectrum disorder (ASD), to define clinical phenotypes and to identify co-occurring medical conditions. A review of the current literature was carried out after PubMed, Medline and Google Scholar were consulted. A total of 37 articles published in the period 2010-2022 was included. Selected studies involve as a whole 2079 individuals diagnosed with ASD (1625 males, 394 females; mean age of 10, 9 years), 51 with other psychiatric comorbidities (developmental delays), 182 at-risk individuals (siblings, those with genetic conditions) and 1530 healthy controls (TD). Metabolomics, reflecting the interplay between genetics and environment, represents an innovative and promising technique to approach ASD. The metabotype may mirror the clinical heterogeneity of an autistic condition; several metabolites can be expressions of dysregulated metabolic pathways thus liable of leading to clinical profiles. However, the employment of metabolomic analyses in clinical practice is far from being introduced, which means there is a need for further studies for the full transition of metabolomics from clinical research to clinical diagnostic routine.
Collapse
Affiliation(s)
- Martina Siracusano
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
- Child Neurology and Psychiatry Unit, Department of Neurosciences, Policlinico Tor Vergata Hospital, Viale Oxford 81, 00133 Rome, Italy; (L.A.); (A.R.); (L.M.)
| | - Lucrezia Arturi
- Child Neurology and Psychiatry Unit, Department of Neurosciences, Policlinico Tor Vergata Hospital, Viale Oxford 81, 00133 Rome, Italy; (L.A.); (A.R.); (L.M.)
| | - Assia Riccioni
- Child Neurology and Psychiatry Unit, Department of Neurosciences, Policlinico Tor Vergata Hospital, Viale Oxford 81, 00133 Rome, Italy; (L.A.); (A.R.); (L.M.)
| | - Antonio Noto
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria, SS 554, Km 4.5, 09042 Monserrato, Italy
| | - Michele Mussap
- Department of Surgical Sciences, School of Medicine, University of Cagliari, Cittadella Universitaria, SS 554, Km 4.5, 09042 Monserrato, Italy
| | - Luigi Mazzone
- Child Neurology and Psychiatry Unit, Department of Neurosciences, Policlinico Tor Vergata Hospital, Viale Oxford 81, 00133 Rome, Italy; (L.A.); (A.R.); (L.M.)
- Systems Medicine Department, University of Rome Tor Vergata, Montpellier Street 1, 00133 Rome, Italy
| |
Collapse
|
21
|
Quillet JC, Siani-Rose M, McKee R, Goldstein B, Taylor M, Kurek I. A machine learning approach for understanding the metabolomics response of children with autism spectrum disorder to medical cannabis treatment. Sci Rep 2023; 13:13022. [PMID: 37608004 PMCID: PMC10444802 DOI: 10.1038/s41598-023-40073-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Accepted: 08/03/2023] [Indexed: 08/24/2023] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental condition impacting behavior, communication, social interaction and learning abilities. Medical cannabis (MC) treatment can reduce clinical symptoms in individuals with ASD. Cannabis-responsive biomarkers are metabolites found in saliva that change in response to MC treatment. Previously we showed levels of these biomarkers in children with ASD successfully treated with MC shift towards the physiological levels detected in typically developing (TD) children, and potentially can quantify the impact. Here, we tested for the first time the capabilities of machine learning techniques applied to our dynamic, high-resolution and rich feature dataset of cannabis-responsive biomarkers from a limited number of children with ASD before and after MC treatment and a TD group to identify: (1) biomarkers distinguishing ASD and TD groups; (2) non-cannabinoid plant molecules with synergistic effects; and (3) biomarkers associated with specific cannabinoids. We found: (1) lysophosphatidylethanolamine can distinguish between ASD and TD groups; (2) novel phytochemicals contribute to the therapeutic effects of MC treatment by inhibition of acetylcholinesterase; and (3) THC- and CBD-associated cannabis-responsive biomarkers are two distinct groups, while CBG is associated with some biomarkers from both groups.
Collapse
Affiliation(s)
| | - Michael Siani-Rose
- Cannformatics, Inc., 3859 Cesar Chavez St, San Francisco, CA, 94131, USA
| | - Robert McKee
- Cannformatics, Inc., 3859 Cesar Chavez St, San Francisco, CA, 94131, USA
| | - Bonni Goldstein
- Cannformatics, Inc., 3859 Cesar Chavez St, San Francisco, CA, 94131, USA
| | - Myiesha Taylor
- Cannformatics, Inc., 3859 Cesar Chavez St, San Francisco, CA, 94131, USA
| | - Itzhak Kurek
- Cannformatics, Inc., 3859 Cesar Chavez St, San Francisco, CA, 94131, USA.
| |
Collapse
|
22
|
Washington P, Wall DP. A Review of and Roadmap for Data Science and Machine Learning for the Neuropsychiatric Phenotype of Autism. Annu Rev Biomed Data Sci 2023; 6:211-228. [PMID: 37137169 PMCID: PMC11093217 DOI: 10.1146/annurev-biodatasci-020722-125454] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Autism spectrum disorder (autism) is a neurodevelopmental delay that affects at least 1 in 44 children. Like many neurological disorder phenotypes, the diagnostic features are observable, can be tracked over time, and can be managed or even eliminated through proper therapy and treatments. However, there are major bottlenecks in the diagnostic, therapeutic, and longitudinal tracking pipelines for autism and related neurodevelopmental delays, creating an opportunity for novel data science solutions to augment and transform existing workflows and provide increased access to services for affected families. Several efforts previously conducted by a multitude of research labs have spawned great progress toward improved digital diagnostics and digital therapies for children with autism. We review the literature on digital health methods for autism behavior quantification and beneficial therapies using data science. We describe both case-control studies and classification systems for digital phenotyping. We then discuss digital diagnostics and therapeutics that integrate machine learning models of autism-related behaviors, including the factors that must be addressed for translational use. Finally, we describe ongoing challenges and potential opportunities for the field of autism data science. Given the heterogeneous nature of autism and the complexities of the relevant behaviors, this review contains insights that are relevant to neurological behavior analysis and digital psychiatry more broadly.
Collapse
Affiliation(s)
- Peter Washington
- Department of Information and Computer Sciences, University of Hawai'i at Mānoa, Honolulu, Hawai'i, USA
| | - Dennis P Wall
- Departments of Pediatrics (Systems Medicine), Biomedical Data Science, and Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, California, USA;
| |
Collapse
|
23
|
Zhang H, Tang X, Feng C, Gao Y, Hong Q, Zhang J, Zhang X, Zheng Q, Lin J, Liu X, Shen L. The use of data independent acquisition based proteomic analysis and machine learning to reveal potential biomarkers for autism spectrum disorder. J Proteomics 2023; 278:104872. [PMID: 36898611 DOI: 10.1016/j.jprot.2023.104872] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 02/08/2023] [Accepted: 03/03/2023] [Indexed: 03/12/2023]
Abstract
Autism spectrum disorder (ASD) is a complex neurological developmental disorder in children, and is associated with social isolation and restricted interests. The etiology of this disorder is still unknown. There is neither any confirmed laboratory test nor any effective therapeutic strategy to diagnose or cure it. We performed data independent acquisition (DIA) and multiple reaction monitoring (MRM) analysis of plasma from children with ASD and controls. The result showed that 45 differentially expressed proteins (DEPs) were identified between autistic subjects and controls. Among these, only one DEP was down-regulated in ASD; other DEPs were up-regulated in ASD children's plasma. These proteins are found associated with complement and coagulation cascades, vitamin digestion and absorption, cholesterol metabolism, platelet degranulation, selenium micronutrient network, extracellular matrix organization and inflammatory pathway, which have been reported to be related to ASD. After MRM verification, five key proteins in complement pathway (PLG, SERPINC1, and A2M) and inflammatory pathway (CD5L, ATRN, SERPINC1, and A2M) were confirmed to be significantly up-regulated in ASD group. Through the screening of machine learning model and MRM verification, we found that two proteins (biotinidase and carbonic anhydrase 1) can be used as early diagnostic markers of ASD (AUC = 0.8, p = 0.0001). SIGNIFICANCE: ASD is the fastest growing neurodevelopmental disorder in the world and has become a major public health problem worldwide. Its prevalence has been steadily increasing, with a global prevalence rate of 1%. Early diagnosis and intervention can achieve better prognosis. In this study, data independent acquisition (DIA) and multiple reaction monitoring (MRM) analysis was applied to analyze the plasma proteome of ASD patients (31 (±5) months old), and 378 proteins were quantified. 45 differentially expressed proteins (DEPs) were identified between the ASD group and the control group. They mainly were associated with platelet degranulation, ECM proteoglycar, complement and coagulation cascades, selenium micronutrient network, regulation of insulin-like growth factor (IGF) transport and uptake by insulin-like growth factor binding proteins (IGFBPs), cholesterol metabolism, vitamin metabolism, and inflammatory pathway. Through the integrated machine learning methods and the MRM verification of independent samples, it is considered that biotinidase and carbon anhydrase 1 have the potential to become biomarkers for the early diagnosis of ASD. These results complement proteomics database of the ASD patients, broaden our understanding of ASD, and provide a panel of biomarkers for the early diagnosis of ASD.
Collapse
Affiliation(s)
- Huajie Zhang
- College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518071, PR China
| | - Xiaoxiao Tang
- College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518071, PR China
| | - Chengyun Feng
- Maternal and Child Health Hospital of Baoan, Shenzhen 518100, PR China
| | - Yan Gao
- Maternal and Child Health Hospital of Baoan, Shenzhen 518100, PR China
| | - Qi Hong
- Maternal and Child Health Hospital of Baoan, Shenzhen 518100, PR China
| | - Jun Zhang
- College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518071, PR China
| | - Xinglai Zhang
- College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518071, PR China
| | - Qihong Zheng
- College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518071, PR China
| | - Jing Lin
- College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518071, PR China
| | - Xukun Liu
- College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518071, PR China
| | - Liming Shen
- College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518071, PR China; Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research, Institutions, Shenzhen 518055, PR China; Shenzhen Key Laboratory of Marine Biotechnology and Ecology, Shenzhen 518071, PR China.
| |
Collapse
|
24
|
Boksha IS, Prokhorova TA, Tereshkina EB, Savushkina OK, Burbaeva GS. Differentiated Approach to Pharmacotherapy of Autism Spectrum Disorders: Biochemical Aspects. BIOCHEMISTRY (MOSCOW) 2023; 88:303-318. [PMID: 37076279 DOI: 10.1134/s0006297923030021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
Abstract
Autism Spectrum Disorders (ASD) are highly heterogeneous neurodevelopmental disorders caused by a complex interaction of numerous genetic and environmental factors and leading to deviations in the nervous system formation at the very early developmental stages. Currently, there are no accepted pharmacological treatments for the so-called core symptoms of ASD, such as social communication disorders and restricted and repetitive behavior patterns. Lack of knowledge about biological basis of ASD, absence of the clinically significant biochemical parameters reflecting abnormalities in the signaling cascades controlling the nervous system development and functioning, and lack of methods for selection of clinically and biologically homogeneous subgroups are considered as causes for the failure of clinical trials of ASD pharmacotherapy. This review considers the possibilities of applying differentiated clinical and biological approaches to the targeted search for ASD pharmacotherapy with emphasis on biochemical markers associated with ASD and attempts to stratify patients by biochemical parameters. The use of such approach as "the target-oriented therapy and assessment of the target status before and during the treatment to identify patients with a positive response to treatment" is discussed using the published results of clinical trials as examples. It is concluded that identification of biochemical parameters for selection of the distinct subgroups among the ASD patients requires research on large samples reflecting clinical and biological diversity of the patients with ASD, and use of unified approaches for such studies. An integrated approach, including clinical observation, clinical-psychological assessment of the patient behavior, study of medical history and description of individual molecular profiles should become a new strategy for stratifying patients with ASD for clinical pharmacotherapeutic trials, as well as for evaluating their efficiency.
Collapse
|
25
|
New insight of metabolomics in ocular diseases in the context of 3P medicine. EPMA J 2023; 14:53-71. [PMID: 36866159 PMCID: PMC9971428 DOI: 10.1007/s13167-023-00313-9] [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: 11/01/2022] [Accepted: 01/09/2023] [Indexed: 02/19/2023]
Abstract
Metabolomics refers to the high-through untargeted or targeted screening of metabolites in biofluids, cells, and tissues. Metabolome reflects the functional states of cells and organs of an individual, influenced by genes, RNA, proteins, and environment. Metabolomic analyses help to understand the interaction between metabolism and phenotype and reveal biomarkers for diseases. Advanced ocular diseases can lead to vision loss and blindness, reducing patients' quality of life and aggravating socio-economic burden. Contextually, the transition from reactive medicine to the predictive, preventive, and personalized (PPPM / 3P) medicine is needed. Clinicians and researchers dedicate a lot of efforts to explore effective ways for disease prevention, biomarkers for disease prediction, and personalized treatments, by taking advantages of metabolomics. In this way, metabolomics has great clinical utility in the primary and secondary care. In this review, we summarized much progress achieved by applying metabolomics to ocular diseases and pointed out potential biomarkers and metabolic pathways involved to promote 3P medicine approach in healthcare.
Collapse
|
26
|
Chen Z, Shi J, Zhang Y, Zhang J, Li S, Guan L, Jia G. Lipidomics Profiles and Lipid Metabolite Biomarkers in Serum of Coal Workers' Pneumoconiosis. TOXICS 2022; 10:496. [PMID: 36136461 PMCID: PMC9500698 DOI: 10.3390/toxics10090496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 08/20/2022] [Accepted: 08/25/2022] [Indexed: 06/16/2023]
Abstract
As a serious occupational pulmonary fibrosis disease, pneumoconiosis still lacks effective biomarkers. Previous studies suggest that pneumoconiosis may affect the body's lipid metabolism. The purpose of this study was to explore lipidomics profiles and lipid metabolite biomarkers in the serum of coal workers' pneumoconiosis (CWP) by a population case-control study. A total of 150 CWP cases and 120 healthy controls from Beijing, China were included. Blood lipids were detected in serum biochemistry. Lipidomics was performed in serum samples for high-throughput detection of lipophilic metabolites. Serum high density lipoprotein cholesterol (HDL-C) decreased significantly in CWP cases. Lipidomics data found 131 differential lipid metabolites between the CWP case and control groups. Further, the top eight most important differential lipid metabolites were screened. They all belonged to differential metabolites of CWP at different stages. However, adjusting for potential confounding factors, only three of them were significantly related to CWP, including acylhexosylceramide (AHEXCER 43:5), diacylglycerol (DG 34:8) and dimethyl-phosphatidylethanolamine (DMPE 36:0|DMPE 18:0_18:0), of which good sensitivity and specificity were proven. The present study demonstrated that lipidomics profiles could change significantly in the serum of CWP patients and that the lipid metabolites represented by AHEXCER, DG and DMPE may be good biomarkers of CWP.
Collapse
Affiliation(s)
- Zhangjian Chen
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing 100191, China
| | - Jiaqi Shi
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing 100191, China
| | - Yi Zhang
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing 100191, China
| | - Jiahe Zhang
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing 100191, China
| | - Shuqiang Li
- Department of Occupational Disease, Peking University Third Hospital, Beijing 100191, China
| | - Li Guan
- Department of Occupational Disease, Peking University Third Hospital, Beijing 100191, China
| | - Guang Jia
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing 100191, China
| |
Collapse
|
27
|
Piazzesi A, Putignani L. Extremely small and incredibly close: Gut microbes as modulators of inflammation and targets for therapeutic intervention. Front Microbiol 2022; 13:958346. [PMID: 36071979 PMCID: PMC9441770 DOI: 10.3389/fmicb.2022.958346] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 07/25/2022] [Indexed: 11/15/2022] Open
Abstract
Chronic inflammation is a hallmark for a variety of disorders and is at least partially responsible for disease progression and poor patient health. In recent years, the microbiota inhabiting the human gut has been associated with not only intestinal inflammatory diseases but also those that affect the brain, liver, lungs, and joints. Despite a strong correlation between specific microbial signatures and inflammation, whether or not these microbes are disease markers or disease drivers is still a matter of debate. In this review, we discuss what is known about the molecular mechanisms by which the gut microbiota can modulate inflammation, both in the intestine and beyond. We identify the current gaps in our knowledge of biological mechanisms, discuss how these gaps have likely contributed to the uncertain outcome of fecal microbiota transplantation and probiotic clinical trials, and suggest how both mechanistic insight and -omics-based approaches can better inform study design and therapeutic intervention.
Collapse
Affiliation(s)
- Antonia Piazzesi
- Multimodal Laboratory Medicine Research Area, Unit of Human Microbiome, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Lorenza Putignani
- Department of Diagnostic and Laboratory Medicine, Unit of Microbiology and Diagnostic Immunology, Unit of Microbiomics and Multimodal Laboratory Medicine Research Area, Unit of Human Microbiome, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
- *Correspondence: Lorenza Putignani,
| |
Collapse
|
28
|
Chen Z, Shi J, Zhang Y, Zhang J, Li S, Guan L, Jia G. Screening of Serum Biomarkers of Coal Workers' Pneumoconiosis by Metabolomics Combined with Machine Learning Strategy. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19127051. [PMID: 35742299 PMCID: PMC9222502 DOI: 10.3390/ijerph19127051] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/04/2022] [Accepted: 06/07/2022] [Indexed: 12/03/2022]
Abstract
Pneumoconiosis remains one of the most serious global occupational diseases. However, effective treatments are lacking, and early detection is crucial for disease prevention. This study aimed to explore serum biomarkers of occupational coal workers’ pneumoconiosis (CWP) by high-throughput metabolomics, combining with machine learning strategy for precision screening. A case–control study was conducted in Beijing, China, involving 150 pneumoconiosis patients with different stages and 120 healthy controls. Metabolomics found a total of 68 differential metabolites between the CWP group and the control group. Then, potential biomarkers of CWP were screened from these differential metabolites by three machine learning methods. The four most important differential metabolites were identified as benzamide, terazosin, propylparaben and N-methyl-2-pyrrolidone. However, after adjusting for the influence of confounding factors, including age, smoking, drinking and chronic diseases, only one metabolite, propylparaben, was significantly correlated with CWP. The more severe CWP was, the higher the content of propylparaben in serum. Moreover, the receiver operating characteristic curve (ROC) of propylparaben showed good sensitivity and specificity as a biomarker of CWP. Therefore, it was demonstrated that the serum metabolite profiles in CWP patients changed significantly and that the serum metabolites represented by propylparaben were good biomarkers of CWP.
Collapse
Affiliation(s)
- Zhangjian Chen
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing 100191, China; (Z.C.); (J.S.); (Y.Z.); (J.Z.)
| | - Jiaqi Shi
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing 100191, China; (Z.C.); (J.S.); (Y.Z.); (J.Z.)
| | - Yi Zhang
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing 100191, China; (Z.C.); (J.S.); (Y.Z.); (J.Z.)
| | - Jiahe Zhang
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing 100191, China; (Z.C.); (J.S.); (Y.Z.); (J.Z.)
| | - Shuqiang Li
- Department of Occupational Disease, Peking University Third Hospital, Beijing 100191, China;
| | - Li Guan
- Department of Occupational Disease, Peking University Third Hospital, Beijing 100191, China;
- Correspondence: (L.G.); (G.J.)
| | - Guang Jia
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing 100191, China; (Z.C.); (J.S.); (Y.Z.); (J.Z.)
- Correspondence: (L.G.); (G.J.)
| |
Collapse
|
29
|
Shen L, Zhang H, Lin J, Gao Y, Chen M, Khan NU, Tang X, Hong Q, Feng C, Zhao Y, Cao X. A Combined Proteomics and Metabolomics Profiling to Investigate the Genetic Heterogeneity of Autistic Children. Mol Neurobiol 2022; 59:3529-3545. [PMID: 35348996 DOI: 10.1007/s12035-022-02801-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 03/16/2022] [Indexed: 11/30/2022]
Abstract
Autism spectrum disorder (ASD) has become one of the most common neurological developmental disorders in children. However, the study of ASD diagnostic markers faces significant challenges due to the existence of heterogeneity. In this study, genetic testing was performed on children who were clinically diagnosed with ASD. Children with ASD susceptibility genes and healthy controls were studied. The proteomics of plasma and peripheral blood mononuclear cells (PBMCs) as well as plasma metabolomics were carried out. The results showed that although there was genetic heterogeneity in children with ASD, the differentially expressed proteins (DEPs) in plasma, peripheral blood mononuclear cells, and differential metabolites in plasma could still effectively distinguish autistic children from controls. The mechanism associated with them focuses on several common and previously reported mechanisms of ASD. The biomarkers for ASD diagnosis could be found by taking differentially expressed proteins and differential metabolites into consideration. Integrating omics data, glycerophospholipid metabolism and N-glycan biosynthesis might play a critical role in the pathogenesis of ASD.
Collapse
Affiliation(s)
- Liming Shen
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, 518071, People's Republic of China
| | - Huajie Zhang
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, 518071, People's Republic of China.,Brain Disease and Big Data Research Institute, Shenzhen University, Shenzhen, 518071, People's Republic of China
| | - Jing Lin
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, 518071, People's Republic of China.,Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 518055, People's Republic of China
| | - Yan Gao
- Maternal and Child Health Hospital of Baoan, Shenzhen, 518100, People's Republic of China
| | - Margy Chen
- Department of Psychology, Emory University, Atlanta, GA, 30322, USA
| | - Naseer Ullah Khan
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, 518071, People's Republic of China
| | - Xiaoxiao Tang
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, 518071, People's Republic of China
| | - Qi Hong
- Maternal and Child Health Hospital of Baoan, Shenzhen, 518100, People's Republic of China
| | - Chengyun Feng
- Maternal and Child Health Hospital of Baoan, Shenzhen, 518100, People's Republic of China
| | - Yuxi Zhao
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, 518071, People's Republic of China.
| | - Xueshan Cao
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, 518071, People's Republic of China.
| |
Collapse
|
30
|
Potential protein markers in children with Autistic Spectrum Disorder (ASD) revealed by salivary proteomics. Int J Biol Macromol 2022; 199:243-251. [PMID: 35016969 DOI: 10.1016/j.ijbiomac.2022.01.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 12/22/2021] [Accepted: 01/03/2022] [Indexed: 12/26/2022]
Abstract
The lack of specific pharmacological therapy for Autistic Spectrum Disorder (ASD) and its clinical heterogeneity demand efforts directed toward the identification of biomarkers to aid in diagnosis. Proteomics offers a new perspective for studying the altered proteins associated with autism spectrum disorders (ASD) and we have saliva as an easy-to-collect biological fluid with important biomolecules for investigating biomarkers in various diseases. In this sense, saliva could be used to identify potential biomarkers of ASD. In the current work, saliva samples were collected from children with different degrees of ASD and healthy children and proteomics approaches were applied to generate data on differentially expressed proteins between groups which will serve as a basis for future validation studies as protein markers. Data are available via ProteomeXchange with identifier PXD030065. As results, 132 proteins were present in 80% of the saliva pools of all analyzed groups. Twenty-five proteins were identified as overexpressed in the group of severe and mild/moderate ASD carriers, among which, eight were identified as potential biomarkers for ASD.
Collapse
|
31
|
Panisi C, Marini M. Dynamic and Systemic Perspective in Autism Spectrum Disorders: A Change of Gaze in Research Opens to A New Landscape of Needs and Solutions. Brain Sci 2022; 12:250. [PMID: 35204013 PMCID: PMC8870276 DOI: 10.3390/brainsci12020250] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 02/08/2022] [Accepted: 02/09/2022] [Indexed: 12/21/2022] Open
Abstract
The first step for a harmonious bio-psycho-social framework in approaching autism spectrum disorders (ASD) is overcoming the conflict between the biological and the psychosocial perspective. Biological research can provide clues for a correct approach to clinical practice, assuming that it would lead to the conceptualization of a pathogenetic paradigm able to account for epidemiologic and clinical findings. The upward trajectory in ASD prevalence and the systemic involvement of other organs besides the brain suggest that the epigenetic paradigm is the most plausible one. The embryo-fetal period is the crucial window of opportunity for keeping neurodevelopment on the right tracks, suggesting that women's health in pregnancy should be a priority. Maladaptive molecular pathways beginning in utero, in particular, a vicious circle between the immune response, oxidative stress/mitochondrial dysfunction, and dysbiosis-impact neurodevelopment and brain functioning across the lifespan and are the basis for progressive multisystemic disorders that account for the substantial health loss and the increased mortality in ASD. Therefore, the biological complexity of ASD and its implications for health requires the enhancement of clinical skills on these topics, to achieve an effective multi-disciplinary healthcare model. Well-balanced training courses could be a promising starting point to make a change.
Collapse
Affiliation(s)
- Cristina Panisi
- Fondazione Istituto Sacra Famiglia ONLUS, Cesano Boscone, 20090 Milan, Italy
| | - Marina Marini
- Department of Experimental, Diagnostic and Specialty Medicine, School of Medicine, University of Bologna, 40126 Bologna, Italy;
| |
Collapse
|
32
|
“Silicon-On-Insulator”-Based Nanosensor for the Revelation of MicroRNA Markers of Autism. Genes (Basel) 2022; 13:genes13020199. [PMID: 35205244 PMCID: PMC8872218 DOI: 10.3390/genes13020199] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 01/10/2022] [Accepted: 01/19/2022] [Indexed: 02/04/2023] Open
Abstract
MicroRNAs (miRNAs), which represent short (20 to 22 nt) non-coding RNAs, were found to play a direct role in the development of autism in children. Herein, a highly sensitive “silicon-on-insulator”-based nanosensor (SOI-NS) has been developed for the revelation of autism-associated miRNAs. This SOI-NS comprises an array of nanowire sensor structures fabricated by complementary metal–oxide–semiconductor (CMOS)-compatible technology, gas-phase etching, and nanolithography. In our experiments described herein, we demonstrate the revelation of ASD-associated miRNAs in human plasma with the SOI-NS, whose sensor elements were sensitized with oligonucleotide probes. In order to determine the concentration sensitivity of the SOI-NS, experiments on the detection of synthetic DNA analogues of autism-associated miRNAs in purified buffer were performed. The lower limit of miRNA detection attained in our experiments amounted to 10−17 M.
Collapse
|
33
|
Levi Mortera S, Vernocchi P, Basadonne I, Zandonà A, Chierici M, Durighello M, Marzano V, Gardini S, Gasbarrini A, Urbani A, Vicari S, Roncada P, Furlanello C, Venuti P, Putignani L. A metaproteomic-based gut microbiota profiling in children affected by autism spectrum disorders. J Proteomics 2022; 251:104407. [PMID: 34763095 DOI: 10.1016/j.jprot.2021.104407] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 09/02/2021] [Accepted: 10/13/2021] [Indexed: 12/15/2022]
Abstract
During the last decade, the evidences on the relationship between neurodevelopmental disorders and the microbial communities of the intestinal tract have considerably grown. Particularly, the role of gut microbiota (GM) ecology and predicted functions in Autism Spectrum Disorders (ASD) has been especially investigated by 16S rRNA targeted and shotgun metagenomics, trying to assess disease signature and their correlation with cognitive impairment or gastrointestinal (GI) manifestations of the disease. Herein we present a metaproteomic approach to point out the microbial gene expression profiles, their functional annotations, and the taxonomic distribution of gut microbial communities in ASD children. We pursued a LC-MS/MS based investigation, to compare the GM profiles of patients with those of their respective relatives and aged-matched controls, providing a quantitative evaluation of bacterial metaproteins by SWATH analysis. All data were managed by a multiple step bioinformatic pipeline, including network analysis. In particular, comparing ASD subjects with CTRLs, up-regulation was found for some metaproteins associated with Clostridia and with carbohydrate metabolism (glyceraldehyde-3-phosphate and glutamate dehydrogenases), while down-regulation was observed for others associated with Bacteroidia (SusC and SusD family together with the TonB dependent receptor). Moreover, network analysis highlighted specific microbial correlations among ASD subgroups characterized by different functioning levels and GI symptoms. SIGNIFICANCE: To the best of our knowledge, this study represents the first metaproteomic investigation on the gut microbiota of ASD children compared with relatives and age-matched CTRLs. Remarkably, the applied SWATH methodology allowed the attribution of differentially regulated functions to specific microbial taxa, offering a novel and complementary point of view with respect to previous studies.
Collapse
Affiliation(s)
- Stefano Levi Mortera
- Multimodal Laboratory Medicine Research Area, Unit of Human Microbiome, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Pamela Vernocchi
- Multimodal Laboratory Medicine Research Area, Unit of Human Microbiome, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Ilaria Basadonne
- Department of Psychology and Cognitive Sciences, University of Trento, Rovereto, Italy
| | | | | | - Martina Durighello
- Multimodal Laboratory Medicine Research Area, Unit of Human Microbiome, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Valeria Marzano
- Multimodal Laboratory Medicine Research Area, Unit of Human Microbiome, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | | | - Antonio Gasbarrini
- Istituto di Patologia Speciale Medica, Università Cattolica del Sacro Cuore, Rome, Italy; UOC Medicina Interna e Gastroenterologia, Area Gastroenterologia ed Oncologia Medica, Dipartimento di Scienze Gastroenterologiche, Endocrino-Metaboliche e Nefro-Urologiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Andrea Urbani
- Clinical Chemistry, Biochemistry and Molecular Biology Operations (UOC), Agostino Gemelli Foundation University Hospital IRCCS, Rome, Italy; Department of Basic Biotechnological Sciences, Intensivological and Perioperative Clinics, Catholic University of Sacred Heart, Rome, Italy
| | - Stefano Vicari
- Department of Neuroscience, Unit of Head Child & Adolescent Psychiatry, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy; Department of Life Sciences and Public Health, Catholic University of Sacred Heart, Rome, Italy
| | - Paola Roncada
- Department of Health Sciences, University 'Magna Græcia' of Catanzaro, Catanzaro, Italy
| | | | - Paola Venuti
- Department of Psychology and Cognitive Sciences, University of Trento, Rovereto, Italy
| | - Lorenza Putignani
- Department of Diagnostics and Laboratory Medicine, Unit of Microbiology and Diagnostic Immunology, Unit of Microbiomics and Multimodal Laboratory Medicine Research Area, Unit of Human Microbiome, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy.
| |
Collapse
|
34
|
Tayanloo-Beik A, Hamidpour SK, Abedi M, Shojaei H, Tavirani MR, Namazi N, Larijani B, Arjmand B. Zebrafish Modeling of Autism Spectrum Disorders, Current Status and Future Prospective. Front Psychiatry 2022; 13:911770. [PMID: 35911241 PMCID: PMC9329562 DOI: 10.3389/fpsyt.2022.911770] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 06/22/2022] [Indexed: 11/13/2022] Open
Abstract
Autism spectrum disorder (ASD) refers to a complicated range of childhood neurodevelopmental disorders which can occur via genetic or non-genetic factors. Clinically, ASD is associated with problems in relationships, social interactions, and behaviors that pose many challenges for children with ASD and their families. Due to the complexity, heterogeneity, and association of symptoms with some neuropsychiatric disorders such as ADHD, anxiety, and sleep disorders, clinical trials have not yielded reliable results and there still remain challenges in drug discovery and development pipeline for ASD patients. One of the main steps in promoting lead compounds to the suitable drug for commercialization is preclinical animal testing, in which the efficacy and toxicity of candidate drugs are examined in vivo. In recent years, zebrafish have been able to attract the attention of many researchers in the field of neurological disorders such as ASD due to their outstanding features. The presence of orthologous genes for ASD modeling, the anatomical similarities of parts of the brain, and similar neurotransmitter systems between zebrafish and humans are some of the main reasons why scientists draw attention to zebrafish as a prominent animal model in preclinical studies to discover highly effective treatment approaches for the ASD through genetic and non-genetic modeling methods.
Collapse
Affiliation(s)
- Akram Tayanloo-Beik
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Shayesteh Kokabi Hamidpour
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mina Abedi
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamide Shojaei
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Nazli Namazi
- Diabetes Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Babak Arjmand
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
35
|
Requena-Jimenez A, Nabiuni M, Miyan JA. Profound changes in cerebrospinal fluid proteome and metabolic profile are associated with congenital hydrocephalus. J Cereb Blood Flow Metab 2021; 41:3400-3414. [PMID: 34415213 PMCID: PMC8669293 DOI: 10.1177/0271678x211039612] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 07/07/2021] [Accepted: 07/12/2021] [Indexed: 11/15/2022]
Abstract
The aetiology of congenital hydrocephalus (cHC) has yet to be resolved. cHC manifests late in rodent gestation, and by 18-22 weeks in human fetuses, coinciding with the start of the major phase of cerebral cortex development. Previously we found that cerebrospinal fluid (CSF) accumulation is associated with compositional changes, folate metabolic impairment and consequential arrest in cortical development. Here, we report a proteomics study on hydrocephalic and normal rat CSF using LC-MSMS and a metabolic pathway analysis to determine the major changes in metabolic and signalling pathways. Non-targeted analysis revealed a proteome transformation across embryonic days 17-20, with the largest changes between day 19 and 20. This provides evidence for a physiological shift in CSF composition and identifies some of the molecular mechanisms unleashed during the onset of cHC. Top molecular regulators that may control the shift in the CSF metabolic signature are also predicted, with potential key biomarkers proposed for early detection of these changes that might be used to develop targeted early therapies for this condition. This study confirms previous findings of a folate metabolic imbalance as well as providing more in depth metabolic analysis and understanding of cHC CSF.
Collapse
Affiliation(s)
- Alicia Requena-Jimenez
- Faculty of Biology, Medicine and Health, The University of Manchester, Division of Neuroscience & Experimental Psychology, The University of Manchester, Manchester, UK
| | - Mohammad Nabiuni
- Faculty of Biology, Medicine and Health, The University of Manchester, Division of Neuroscience & Experimental Psychology, The University of Manchester, Manchester, UK
| | - Jaleel A Miyan
- Faculty of Biology, Medicine and Health, The University of Manchester, Division of Neuroscience & Experimental Psychology, The University of Manchester, Manchester, UK
| |
Collapse
|
36
|
Dean DD, Agarwal S, Muthuswamy S, Asim A. Brain exosomes as minuscule information hub for Autism Spectrum Disorder. Expert Rev Mol Diagn 2021; 21:1323-1331. [PMID: 34720032 DOI: 10.1080/14737159.2021.2000395] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Autism spectrum disorder (ASD) is a neurodevelopmental disorder initiating in the first three years of life. Early initiation of management therapies can significantly improve the health and quality of life of ASD subjects. Thus, indicating the need for suitable biomarkers for the early identification of ASD. Various biological domains were investigated in the quest for reliable biomarkers. However, most biomarkers are in the preliminary stage, and clinical validation is yet to be defined. Exosome based research gained momentum in various Central Nervous System disorders for biomarker identification. However, the utility and prospect of exosomes in ASD is still underexplored. AREAS COVERED In the present review, we summarized the biomarker discovery current status and the future of brain-specific exosomes in understanding pathophysiology and its potential as a biomarker. The studies reviewed herein were identified via systematic search (dated: June 2021) of PubMed using variations related to autism (ASD OR autism OR Autism spectrum disorder) AND exosomes AND/OR biomarkers. EXPERT OPINION As exosomess are highly relevant in brain disorders like ASD, direct access to brain tissue for molecular assessment is ethically impossible. Thus investigating the brain-derived exosomes would undoubtedly answer many unsolved aspects of the pathogenesis and provide reliable biomarkers.
Collapse
Affiliation(s)
- Deepika Delsa Dean
- Deptartment of Medical Genetics, Sanjay Gandhi Post Graduate Institute of Medical Sciences (Sgpgims), Lucknow, India
| | - Sarita Agarwal
- Deptartment of Medical Genetics, Sanjay Gandhi Post Graduate Institute of Medical Sciences (Sgpgims), Lucknow, India
| | | | - Ambreen Asim
- Deptartment of Medical Genetics, Sanjay Gandhi Post Graduate Institute of Medical Sciences (Sgpgims), Lucknow, India
| |
Collapse
|
37
|
Mahony C, O’Ryan C. Convergent Canonical Pathways in Autism Spectrum Disorder from Proteomic, Transcriptomic and DNA Methylation Data. Int J Mol Sci 2021; 22:ijms221910757. [PMID: 34639097 PMCID: PMC8509728 DOI: 10.3390/ijms221910757] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 09/27/2021] [Accepted: 10/01/2021] [Indexed: 12/20/2022] Open
Abstract
Autism Spectrum Disorder (ASD) is a complex neurodevelopmental disorder with extensive genetic and aetiological heterogeneity. While the underlying molecular mechanisms involved remain unclear, significant progress has been facilitated by recent advances in high-throughput transcriptomic, epigenomic and proteomic technologies. Here, we review recently published ASD proteomic data and compare proteomic functional enrichment signatures with those of transcriptomic and epigenomic data. We identify canonical pathways that are consistently implicated in ASD molecular data and find an enrichment of pathways involved in mitochondrial metabolism and neurogenesis. We identify a subset of differentially expressed proteins that are supported by ASD transcriptomic and DNA methylation data. Furthermore, these differentially expressed proteins are enriched for disease phenotype pathways associated with ASD aetiology. These proteins converge on protein–protein interaction networks that regulate cell proliferation and differentiation, metabolism, and inflammation, which demonstrates a link between canonical pathways, biological processes and the ASD phenotype. This review highlights how proteomics can uncover potential molecular mechanisms to explain a link between mitochondrial dysfunction and neurodevelopmental pathology.
Collapse
|
38
|
Ning L, Huixin H. Topic Evolution Analysis for Omics Data Integration in Cancers. Front Cell Dev Biol 2021; 9:631011. [PMID: 33898421 PMCID: PMC8058380 DOI: 10.3389/fcell.2021.631011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 02/04/2021] [Indexed: 12/02/2022] Open
Abstract
One of the vital challenges for cancer diseases is efficient biomarkers monitoring formation and development are limited. Omics data integration plays a crucial role in the mining of biomarkers in the human condition. As the link between omics study on biomarkers discovery and cancer diseases is deepened, defining the principal technologies applied in the field is a must not only for the current period but also for the future. We utilize topic modeling to extract topics (or themes) as a probabilistic distribution of latent topics from the dataset. To predict the future trend of related cases, we utilize the Prophet neural network to perform a prediction correction model for existing topics. A total of 2,318 pieces of literature (from 2006 to 2020) were retrieved from MEDLINE with the query on “omics” and “cancer.” Our study found 20 topics covering current research types. The topic extraction results indicate that, with the rapid development of omics data integration research, multi-omics analysis (Topic 11) and genomics of colorectal cancer (Topic 10) have more studies reported last 15 years. From the topic prediction view, research findings in multi-omics data processing and novel biomarker discovery for cancer prediction (Topic 2, 3, 10, 11) will be heavily focused in the future. From the topic visuallization and evolution trends, metabolomics of breast cancer (Topic 9), pharmacogenomics (Topic 15), genome-guided therapy regimens (Topic 16), and microRNAs target genes (Topic 17) could have more rapidly developed in the study of cancer treatment effect and recurrence prediction.
Collapse
Affiliation(s)
- Li Ning
- Business School of Huaqiao University, Quan Zhou, China.,Business School of Huaqiao University, Quan Zhou, China
| | - He Huixin
- Management Science and Engineering Department, Management School, Xiamen University, Xiamen, China
| |
Collapse
|
39
|
Hewitson L, Mathews JA, Devlin M, Schutte C, Lee J, German DC. Blood biomarker discovery for autism spectrum disorder: A proteomic analysis. PLoS One 2021; 16:e0246581. [PMID: 33626076 PMCID: PMC7904196 DOI: 10.1371/journal.pone.0246581] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 01/22/2021] [Indexed: 12/12/2022] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by deficits in social communication and social interaction and restricted, repetitive patterns of behavior, interests, or activities. Given the lack of specific pharmacological therapy for ASD and the clinical heterogeneity of the disorder, current biomarker research efforts are geared mainly toward identifying markers for determining ASD risk or for assisting with a diagnosis. A wide range of putative biological markers for ASD is currently being investigated. Proteomic analyses indicate that the levels of many proteins in plasma/serum are altered in ASD, suggesting that a panel of proteins may provide a blood biomarker for ASD. Serum samples from 76 boys with ASD and 78 typically developing (TD) boys, 18 months-8 years of age, were analyzed to identify possible early biological markers for ASD. Proteomic analysis of serum was performed using SomaLogic’s SOMAScanTM assay 1.3K platform. A total of 1,125 proteins were analyzed. There were 86 downregulated proteins and 52 upregulated proteins in ASD (FDR < 0.05). Combining three different algorithms, we found a panel of 9 proteins that identified ASD with an area under the curve (AUC) = 0.8599±0.0640, with specificity and sensitivity of 0.8217±0.1178 and 0.835±0.1176, respectively. All 9 proteins were significantly different in ASD compared with TD boys, and were significantly correlated with ASD severity as measured by ADOS total scores. Using machine learning methods, a panel of serum proteins was identified that may be useful as a blood biomarker for ASD in boys. Further verification of the protein biomarker panel with independent test sets is warranted.
Collapse
Affiliation(s)
- Laura Hewitson
- The Johnson Center for Child Health and Development, Austin, TX, United States of America
| | - Jeremy A Mathews
- Departments of Mathematical Sciences and Biological Sciences, Bioinformatics & Computational Biology Program, University of Texas at Dallas, Dallas, TX, United States of America
| | - Morgan Devlin
- The Johnson Center for Child Health and Development, Austin, TX, United States of America
| | - Claire Schutte
- The Johnson Center for Child Health and Development, Austin, TX, United States of America
| | - Jeon Lee
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX, United States of America
| | - Dwight C German
- Department of Psychiatry, UT Southwestern Medical Center, Dallas, TX, United States of America
| |
Collapse
|
40
|
Malsagova KA, Pleshakova TO, Popov VP, Kupriyanov IN, Galiullin RA, Kozlov AF, Shumov ID, Kaysheva AL, Tikhonenko FV, Archakov AI, Ivanov YD. Optical Monitoring of the Production Quality of Si-Nanoribbon Chips Intended for the Detection of ASD-Associated Oligonucleotides. MICROMACHINES 2021; 12:mi12020147. [PMID: 33546438 PMCID: PMC7913754 DOI: 10.3390/mi12020147] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 01/25/2021] [Accepted: 01/29/2021] [Indexed: 02/07/2023]
Abstract
Gas-phase etching and optical lithography were employed for the fabrication of a silicon nanoribbon chip (Si-NR chip). The quality of the so-fabricated silicon nanoribbons (Si-NRs) was monitored by optical Raman scattering spectroscopy. It was demonstrated that the structures of the Si-NRs were virtually defect-free, meaning they could be used for highly sensitive detection of biological macromolecules. The Si-NR chips were then used for the highly sensitive nanoelectronics detection of DNA oligonucleotides (oDNAs), which represent synthetic analogs of 106a-5p microRNA (miR-106a-5p), associated with the development of autism spectrum disorders in children. The specificity of the analysis was attained by the sensitization of the Si-NR chip sur-face by covalent immobilization of oDNA probes, whose nucleotide sequence was complementary to the known sequence of miR-106a-5p. The use of the Si-NR chip was demonstrated to al-low for the rapid label-free real-time detection of oDNA at ultra-low (~10−17 M) concentrations.
Collapse
Affiliation(s)
- Kristina A. Malsagova
- Laboratory of Nanobiotechnology, Institute of Biomedical Chemistry, 119121 Moscow, Russia; (T.O.P.); (R.A.G.); (A.F.K.); (I.D.S.); (A.L.K.); (A.I.A.); (Y.D.I.)
- Correspondence: ; Tel.: +7-499-246-3761
| | - Tatyana O. Pleshakova
- Laboratory of Nanobiotechnology, Institute of Biomedical Chemistry, 119121 Moscow, Russia; (T.O.P.); (R.A.G.); (A.F.K.); (I.D.S.); (A.L.K.); (A.I.A.); (Y.D.I.)
| | - Vladimir P. Popov
- Rzhanov Institute of Semiconductor Physics, Siberian Branch of Russian Academy of Sciences, Laboratory of Silicon Material Science, 630090 Novosibirsk, Russia; (V.P.P.); (F.V.T.)
| | - Igor N. Kupriyanov
- Sobolev Institute of Geology and Mineralogy, Siberian Branch of Russian Academy of Sciences, Laboratory of Experimental Mineralogy and Crystallogenesis, 630090 Novosibirsk, Russia;
| | - Rafael A. Galiullin
- Laboratory of Nanobiotechnology, Institute of Biomedical Chemistry, 119121 Moscow, Russia; (T.O.P.); (R.A.G.); (A.F.K.); (I.D.S.); (A.L.K.); (A.I.A.); (Y.D.I.)
| | - Andrey F. Kozlov
- Laboratory of Nanobiotechnology, Institute of Biomedical Chemistry, 119121 Moscow, Russia; (T.O.P.); (R.A.G.); (A.F.K.); (I.D.S.); (A.L.K.); (A.I.A.); (Y.D.I.)
| | - Ivan D. Shumov
- Laboratory of Nanobiotechnology, Institute of Biomedical Chemistry, 119121 Moscow, Russia; (T.O.P.); (R.A.G.); (A.F.K.); (I.D.S.); (A.L.K.); (A.I.A.); (Y.D.I.)
| | - Anna L. Kaysheva
- Laboratory of Nanobiotechnology, Institute of Biomedical Chemistry, 119121 Moscow, Russia; (T.O.P.); (R.A.G.); (A.F.K.); (I.D.S.); (A.L.K.); (A.I.A.); (Y.D.I.)
| | - Fedor V. Tikhonenko
- Rzhanov Institute of Semiconductor Physics, Siberian Branch of Russian Academy of Sciences, Laboratory of Silicon Material Science, 630090 Novosibirsk, Russia; (V.P.P.); (F.V.T.)
| | - Alexander I. Archakov
- Laboratory of Nanobiotechnology, Institute of Biomedical Chemistry, 119121 Moscow, Russia; (T.O.P.); (R.A.G.); (A.F.K.); (I.D.S.); (A.L.K.); (A.I.A.); (Y.D.I.)
| | - Yuri D. Ivanov
- Laboratory of Nanobiotechnology, Institute of Biomedical Chemistry, 119121 Moscow, Russia; (T.O.P.); (R.A.G.); (A.F.K.); (I.D.S.); (A.L.K.); (A.I.A.); (Y.D.I.)
| |
Collapse
|
41
|
Micera A, Balzamino BO, Di Zazzo A, Dinice L, Bonini S, Coassin M. Biomarkers of Neurodegeneration and Precision Therapy in Retinal Disease. Front Pharmacol 2021; 11:601647. [PMID: 33584278 PMCID: PMC7873955 DOI: 10.3389/fphar.2020.601647] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 11/09/2020] [Indexed: 12/11/2022] Open
Abstract
Vision-threatening retinal diseases affect millions of people worldwide, representing an important public health issue (high social cost) for both technologically advanced and new-industrialized countries. Overall RD group comprises the retinitis pigmentosa, the age-related macular degeneration (AMD), the diabetic retinopathy (DR), and idiopathic epiretinal membrane formation. Endocrine, metabolic, and even lifestyles risk factors have been reported for these age-linked conditions that represent a "public priority" also in this COVID-19 emergency. Chronic inflammation and neurodegeneration characterize the disease evolution, with a consistent vitreoretinal interface impairment. As the vitreous chamber is significantly involved, the latest diagnostic technologies of imaging (retina) and biomarker detection (vitreous) have provided a huge input at both medical and surgical levels. Complement activation and immune cell recruitment/infiltration as well as detrimental intra/extracellular deposits occur in association with a reactive gliosis. The cell/tissue aging route shows a specific signal path and biomolecular profile characterized by the increased expression of several glial-derived mediators, including angiogenic/angiostatic, neurogenic, and stress-related factors (oxidative stress metabolites, inflammation, and even amyloid formation). The possibility to access vitreous chamber by collecting vitreous reflux during intravitreal injection or obtaining vitreous biopsy during a vitrectomy represents a step forward for an individualized therapy. As drug response and protein signature appear unique in each single patient, therapies should be individualized. This review addresses the current knowledge about biomarkers and pharmacological targets in these vitreoretinal diseases. As vitreous fluids might reflect the early stages of retinal sufferance and/or late stages of neurodegeneration, the possibility to modulate intravitreal levels of growth factors, in combination to anti-VEGF therapy, would open to a personalized therapy of retinal diseases.
Collapse
Affiliation(s)
- Alessandra Micera
- Research and Development Laboratory for Biochemical, Molecular and Cellular Applications in Ophthalmological Sciences, IRCCS - Fondazione Bietti, Rome, Italy
| | - Bijorn Omar Balzamino
- Research and Development Laboratory for Biochemical, Molecular and Cellular Applications in Ophthalmological Sciences, IRCCS - Fondazione Bietti, Rome, Italy
| | - Antonio Di Zazzo
- Ophthalmology Operative Complex Unit, University Campus Bio-Medico, Rome, Italy
| | - Lucia Dinice
- Research and Development Laboratory for Biochemical, Molecular and Cellular Applications in Ophthalmological Sciences, IRCCS - Fondazione Bietti, Rome, Italy
| | - Stefano Bonini
- Ophthalmology Operative Complex Unit, University Campus Bio-Medico, Rome, Italy
| | - Marco Coassin
- Ophthalmology Operative Complex Unit, University Campus Bio-Medico, Rome, Italy
| |
Collapse
|