1
|
Cao F, You Q, Zhu F, Zhang Y. Serum exosomal small nucleolar RNA (snoRNA) signatures as a predictive biomarker for benign and malignant pulmonary nodules. Cancer Cell Int 2024; 24:341. [PMID: 39402628 PMCID: PMC11475846 DOI: 10.1186/s12935-024-03522-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 10/05/2024] [Indexed: 10/19/2024] Open
Abstract
Low-dose CT (LDCT) is increasingly recognized as the preferred method for detecting pulmonary nodules. However, distinguishing whether a nodule is benign or malignant often necessitates repeated scans or invasive tissue sampling procedures. Therefore, there is a pressing need for non-invasive techniques to minimize unnecessary interventions. This study aim to investigate the expression profile of exosomal snoRNA in the serum of patients with benign and malignant pulmonary nodules. We identified a total of 278 snoRNAs in serum exosomes, revealing significant differences in snoRNA levels between patients with malignant and benign nodules. Specifically, the upregulated snoRNAs U78 and U37 were validated through qRT-PCR and were found significantly elevated in the serum of patients with malignant pulmonary nodules, positioning them as promising biomarkers for the early detection of lung cancer. This study underscores the potential of serum exosomal U78 and U37 as critical tools for assessing the risk of pulmonary nodules identified through CT screening.
Collapse
Affiliation(s)
- Fei Cao
- Department of Oncology, The Fifth People's Hospital of Wuxi Affiliated to Jiangnan University, Wuxi, China
| | - Qian You
- Department of Respiratory, The Fifth People's Hospital of Wuxi Affiliated to Jiangnan University, Wuxi, China
| | - Feng Zhu
- Department of Respiratory, The Fifth People's Hospital of Wuxi Affiliated to Jiangnan University, Wuxi, China.
| | - Yu Zhang
- Department of Respiratory, Wuxi 9th Affiliated Hospital of Soochow University, Wuxi, China.
| |
Collapse
|
2
|
Shen LP, Zhang WC, Deng JR, Qi ZH, Lin ZW, Wang ZD. Advances in the mechanism of small nucleolar RNA and its role in DNA damage response. Mil Med Res 2024; 11:53. [PMID: 39118131 PMCID: PMC11308251 DOI: 10.1186/s40779-024-00553-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 07/08/2024] [Indexed: 08/10/2024] Open
Abstract
Small nucleolar RNAs (snoRNAs) were previously regarded as a class of functionally conserved housekeeping genes, primarily involved in the regulation of ribosome biogenesis by ribosomal RNA (rRNA) modification. However, some of them are involved in several biological processes via complex molecular mechanisms. DNA damage response (DDR) is a conserved mechanism for maintaining genomic stability to prevent the occurrence of various human diseases. It has recently been revealed that snoRNAs are involved in DDR at multiple levels, indicating their relevant theoretical and clinical significance in this field. The present review systematically addresses four main points, including the biosynthesis and classification of snoRNAs, the mechanisms through which snoRNAs regulate target molecules, snoRNAs in the process of DDR, and the significance of snoRNA in disease diagnosis and treatment. It focuses on the potential functions of snoRNAs in DDR to help in the discovery of the roles of snoRNAs in maintaining genome stability and pathological processes.
Collapse
Affiliation(s)
- Li-Ping Shen
- Department of Radiobiology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Wen-Cheng Zhang
- Department of Radiobiology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Jia-Rong Deng
- Graduate Collaborative Training Base of Academy of Military Sciences, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Zhen-Hua Qi
- Department of Radiobiology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Zhong-Wu Lin
- Department of Radiobiology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Zhi-Dong Wang
- Department of Radiobiology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, China.
- Graduate Collaborative Training Base of Academy of Military Sciences, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
| |
Collapse
|
3
|
Wang K, Li Y, Lin J. Identification of diagnostic biomarkers for osteoarthritis through bioinformatics and machine learning. Heliyon 2024; 10:e27506. [PMID: 38496843 PMCID: PMC10944228 DOI: 10.1016/j.heliyon.2024.e27506] [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: 09/09/2023] [Revised: 01/20/2024] [Accepted: 02/29/2024] [Indexed: 03/19/2024] Open
Abstract
Osteoarthritis (OA) is a prevalent degenerative joint disease characterized by cartilage degradation, inflammatory arthritis, and joint dysfunction. Currently, there is a lack of effective early diagnostic methods and treatment strategies for OA. Bioinformatics and biomarker research provide new possibilities for early detection and personalized therapy of OA. In this study, we investigated the molecular mechanisms of OA and important signaling pathways involved in disease progression through bioinformatics analysis. Firstly, using the limma package, we analyzed the differentially expressed genes (DEGs) between normal healthy samples and OA cartilage tissue samples. These DEGs were found to be primarily involved in biological processes such as extracellular matrix (ECM) binding, immune receptor activity, and cytokine activity, as well as signaling pathways including cytokine receptors, ECM-receptor interaction, and PI3K-Akt. Gene set enrichment analysis revealed that in the OA group, signaling pathways such as AMPK, B cell receptor, IL-17, and PPAR were downregulated, while calcium signaling pathway, cell adhesion molecules, ECM-receptor interaction, TGF-β signaling pathway, and Wnt signaling pathway were upregulated. Additionally, we constructed a co-expression module network using WGCNA and identified key modules associated with OA, from which we selected 7 most predictive OA characteristic genes. Among them, ANTXR1, KCNS3, SGCD, and LIN7A were correlated with the level of immune cell infiltration. This study elucidates the mechanisms underlying the development of OA and identifies potential diagnostic markers and therapeutic targets, providing important insights for early diagnosis and treatment of OA.
Collapse
Affiliation(s)
| | | | - JinXiu Lin
- Department of Orthopedics, Zibo First Hospital, Zibo, 255200, China
| |
Collapse
|
4
|
Kharaz YA, Zamboulis DE, Fang Y, Welting TJM, Peffers MJ, Comerford EJ. Small RNA signatures of the anterior cruciate ligament from patients with knee joint osteoarthritis. Front Mol Biosci 2023; 10:1266088. [PMID: 38187089 PMCID: PMC10768046 DOI: 10.3389/fmolb.2023.1266088] [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: 07/24/2023] [Accepted: 12/05/2023] [Indexed: 01/09/2024] Open
Abstract
Introduction: The anterior cruciate ligament (ACL) is susceptible to degeneration, resulting in joint pain, reduced mobility, and osteoarthritis development. There is currently a paucity of knowledge on how anterior cruciate ligament degeneration and disease leads to osteoarthritis. Small non-coding RNAs (sncRNAs), such as microRNAs and small nucleolar RNA (snoRNA), have diverse roles, including regulation of gene expression. Methods: We profiled the sncRNAs of diseased osteoarthritic ACLs to provide novel insights into osteoarthritis development. Small RNA sequencing from the ACLs of non- or end-stage human osteoarthritic knee joints was performed. Significantly differentially expressed sncRNAs were defined, and bioinformatics analysis was undertaken. Results and Discussion: A total of 184 sncRNAs were differentially expressed: 68 small nucleolar RNAs, 26 small nuclear RNAs (snRNAs), and 90 microRNAs. We identified both novel and recognized (miR-206, -365, and -29b and -29c) osteoarthritis-related microRNAs and other sncRNAs (including SNORD72, SNORD113, and SNORD114). Significant pathway enrichment of differentially expressed miRNAs includes differentiation of the muscle, inflammation, proliferation of chondrocytes, and fibrosis. Putative mRNAs of the microRNA target genes were associated with the canonical pathways "hepatic fibrosis signaling" and "osteoarthritis." The establishing sncRNA signatures of ACL disease during osteoarthritis could serve as novel biomarkers and potential therapeutic targets in ACL degeneration and osteoarthritis development.
Collapse
Affiliation(s)
- Yalda A. Kharaz
- Department of Musculoskeletal Ageing Sciences, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Danae E. Zamboulis
- Comparative Biomedical Sciences, Royal Veterinary College, London, United Kingdom
| | - Yongxiang Fang
- Centre for Genomic Research, Institute of Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Tim J. M. Welting
- Department of Orthopaedic Surgery, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Mandy J. Peffers
- Department of Musculoskeletal Ageing Sciences, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Eithne J. Comerford
- Department of Musculoskeletal Ageing Sciences, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
- Institute of Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
5
|
Zhang Y, Zhao Z, Huang LA, Liu Y, Yao J, Sun C, Li Y, Zhang Z, Ye Y, Yuan F, Nguyen TK, Garlapati NR, Wu A, Egranov SD, Caudle AS, Sahin AA, Lim B, Beretta L, Calin GA, Yu D, Hung MC, Curran MA, Rezvani K, Gan B, Tan Z, Han L, Lin C, Yang L. Molecular mechanisms of snoRNA-IL-15 crosstalk in adipocyte lipolysis and NK cell rejuvenation. Cell Metab 2023; 35:1457-1473.e13. [PMID: 37329887 PMCID: PMC10712687 DOI: 10.1016/j.cmet.2023.05.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 03/03/2023] [Accepted: 05/18/2023] [Indexed: 06/19/2023]
Abstract
Obesity, in which the functional importance of small nucleolar RNAs (snoRNAs) remains elusive, correlates with risk for many cancer types. Here, we identify that the serum copies of adipocyte-expressed SNORD46 correlate with body mass index (BMI), and serum SNORD46 antagonizes interleukin-15 (IL-15) signaling. Mechanically, SNORD46 binds IL-15 via G11, and G11A (a mutation that significantly enhances binding affinity) knockin drives obesity in mice. Functionally, SNORD46 blocks IL-15-induced, FER kinase-dependent phosphorylation of platelet glycoprotein 4 (CD36) and monoglyceride lipase (MGLL) in adipocytes, leading to inhibited lipolysis and browning. In natural killer (NK) cells, SNORD46 suppresses the IL-15-dependent autophagy, leading to reduced viability of obese NK. SNORD46 power inhibitors exhibit anti-obesity effects, concurring with improved viability of obese NK and anti-tumor immunity of CAR-NK cell therapy. Hence, our findings demonstrate the functional importance of snoRNAs in obesity and the utility of snoRNA power inhibitors for antagonizing obesity-associated immune resistance.
Collapse
Affiliation(s)
- Yaohua Zhang
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Zilong Zhao
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Lisa A Huang
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yuan Liu
- Center for Epigenetics and Disease Prevention, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, USA
| | - Jun Yao
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Chengcao Sun
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yajuan Li
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Zhao Zhang
- Department of Biochemistry and Molecular Biology, University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX 77030, USA
| | - Youqiong Ye
- Department of Biochemistry and Molecular Biology, University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX 77030, USA
| | - Fei Yuan
- Center for Drug Discovery, Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Tina K Nguyen
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Nikhil Reddy Garlapati
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Andrew Wu
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sergey D Egranov
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Abigail S Caudle
- Department of Breast Surgical Oncology, Division of Surgery, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Aysegul A Sahin
- Department of Pathology, Division of Pathology and Laboratory Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Bora Lim
- Oncology/Medicine, Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Laura Beretta
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - George A Calin
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Center for RNA Interference and Non-Coding RNAs, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Dihua Yu
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Graduate School of Biomedical Sciences, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Mien-Chie Hung
- Graduate Institute of Biomedical Sciences, Institute of Biochemistry and Molecular Biology, Research Center for Cancer Biology, Cancer Biology and Precision Therapeutics Center, and Center for Molecular Medicine, China Medical University, Taichung 406, Taiwan
| | - Michael A Curran
- Graduate School of Biomedical Sciences, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Katayoun Rezvani
- Graduate School of Biomedical Sciences, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Boyi Gan
- Graduate School of Biomedical Sciences, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Experimental Radiation Oncology, Division of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Zhi Tan
- Center for Drug Discovery, Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030, USA; Department of Pharmacology and Chemical Biology, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Leng Han
- Center for Epigenetics and Disease Prevention, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, USA; Brown Center for Immunotherapy, School of Medicine, Indiana University, Indianapolis, IN 46202, USA; Department of Biostatistics and Health Data Science, School of Medicine, Indiana University, Indianapolis, IN 46202, USA.
| | - Chunru Lin
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Graduate School of Biomedical Sciences, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Liuqing Yang
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Center for RNA Interference and Non-Coding RNAs, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Graduate School of Biomedical Sciences, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
6
|
Li X, Shen L, Deng Z, Huang Z. New treatment for osteoarthr: pbad014itis: Gene therapy. PRECISION CLINICAL MEDICINE 2023; 6:pbad014. [PMID: 37333626 PMCID: PMC10273835 DOI: 10.1093/pcmedi/pbad014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 05/24/2023] [Indexed: 06/20/2023] Open
Abstract
Osteoarthritis is a complex degenerative disease that affects the entire joint tissue. Currently, non-surgical treatments for osteoarthritis focus on relieving pain. While end-stage osteoarthritis can be treated with arthroplasty, the health and financial costs associated with surgery have forced the search for alternative non-surgical treatments to delay the progression of osteoarthritis and promote cartilage repair. Unlike traditional treatment, the gene therapy approach allows for long-lasting expression of therapeutic proteins at specific sites. In this review, we summarize the history of gene therapy in osteoarthritis, outlining the common expression vectors (non-viral, viral), the genes delivered (transcription factors, growth factors, inflammation-associated cytokines, non-coding RNAs) and the mode of gene delivery (direct delivery, indirect delivery). We highlight the application and development prospects of the gene editing technology CRISPR/Cas9 in osteoarthritis. Finally, we identify the current problems and possible solutions in the clinical translation of gene therapy for osteoarthritis.
Collapse
Affiliation(s)
- Xinyu Li
- Department of Orthopaedic Surgery, West China Hospital, West China Medical School, Sichuan University, Chengdu 610041, China
| | - Leyao Shen
- School of Dentistry, University of Michigan, Ann Arbor, MI 48109-1078, USA
| | | | | |
Collapse
|
7
|
Behnia M, Bradfute SB. The Host Non-Coding RNA Response to Alphavirus Infection. Viruses 2023; 15:v15020562. [PMID: 36851776 PMCID: PMC9967650 DOI: 10.3390/v15020562] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 02/10/2023] [Accepted: 02/15/2023] [Indexed: 02/22/2023] Open
Abstract
Alphaviruses are important human and animal pathogens that can cause a range of debilitating symptoms and are found worldwide. These include arthralgic diseases caused by Old-World viruses and encephalitis induced by infection with New-World alphaviruses. Non-coding RNAs do not encode for proteins, but can modulate cellular response pathways in a myriad of ways. There are several classes of non-coding RNAs, some more well-studied than others. Much research has focused on the mRNA response to infection against alphaviruses, but analysis of non-coding RNA responses has been more limited until recently. This review covers what is known regarding host cell non-coding RNA responses in alphavirus infections and highlights gaps in the knowledge that future research should address.
Collapse
|
8
|
Xiao F, Wang C, Peng J, Zhou X, Ma D, Wang Y, Li Y, Chen X, Wang C. Changes in Small Noncoding RNA Expression during Chondrocyte Senescence. Cartilage 2022; 13:19476035221118165. [PMID: 35993268 PMCID: PMC9403477 DOI: 10.1177/19476035221118165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVE Osteoarthritis (OA) is characterized by the chronic and progressive deterioration of articular cartilage. Chondrocyte senescence could lead to a shift in the balance between extracellular matrix (ECM) component synthesis and degradation. Small noncoding RNAs (sncRNAs), including microRNAs (miRNAs), P-element-induced wimpy testis-(PIWI-) interacting RNAs (piRNAs), small nucleolar RNAs (snoRNAs), small nuclear RNAs (snRNAs), and repeat-associated siRNAs (rasiRNAs), are a class of important epigenetic molecules. We aimed to gain insights into the changes and roles of sncRNA in chondrocyte senescence. DESIGN Healthy mouse postnatal chondrocytes were isolated, and a replicative aging model was constructed. We used small RNA sequencing (small RNA-seq) to generate extensive small RNA data. We identified differentially expressed sncRNAs and performed tissue-specific analysis using real-time quantitative polymerase chain reaction (qRT-PCR). β-galactosidase staining was used to detect chondrocyte senescence. The results showed that the expression profiles of sncRNA in passage 5 chondrocytes were significantly different from those in passage 0 chondrocytes. The expression of sncRNA was tissue specific. We found that 40 miRNAs were upregulated and 70 miRNAs were downregulated during chondrocyte senescence, and that miR-132-5p expression inhibition prevented chondrocyte senescence. We found that 8 piRNAs were upregulated and 17 piRNAs were downregulated during chondrocyte senescence, and that piRNA piR_025576 overexpression delayed chondrocyte senescence. We found that 24 snoRNAs were upregulated and 28 snoRNAs were downregulated during chondrocyte senescence, and that snoRNA ENSMUSG00000087935 overexpression delayed chondrocyte senescence. We found that 5 snRNAs were upregulated and 6 snRNAs were downregulated during chondrocyte senescence, and that snRNA ENSMUSG00000064682 overexpression delayed chondrocyte senescence. We found that 1 rasiRNA was upregulated and 4 rasiRNAs were downregulated during chondrocyte senescence. CONCLUSIONS These findings might provide novel insights into OA pathogenesis and contribute to the development of candidates for targeted therapeutics in OA.
Collapse
Affiliation(s)
- Fei Xiao
- Department of Orthopedic Surgery,
Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine,
Shanghai, China
| | - Chenglong Wang
- Department of Orthopedic Surgery,
Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine,
Shanghai, China
| | - Jianping Peng
- Department of Orthopedic Surgery,
Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine,
Shanghai, China
| | - Xing Zhou
- Guangxi Collaborative Innovation Center
for Biomedicine, Guangxi Medical University, Nanning, China
| | - Ding Ma
- Department of Orthopedic Surgery,
Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine,
Shanghai, China
| | - Yu Wang
- Department of Cardiology, Shidong
Hospital, Shanghai, China
| | - Yanpeng Li
- Department of Spine Surgery, Affiliated
Hospital of Jining Medical University, Jining, China
| | - Xiaodong Chen
- Department of Orthopedic Surgery,
Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine,
Shanghai, China
| | - Chuandong Wang
- Department of Orthopedic Surgery,
Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine,
Shanghai, China,Xiaodong Chen, Department of Orthopedic
Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of
Medicine, 1665 Kongjiang Road, Shanghai 200092, China.
| |
Collapse
|
9
|
Anderson JR, Jacobsen S, Walters M, Bundgaard L, Diendorfer A, Hackl M, Clarke EJ, James V, Peffers MJ. Small non-coding RNA landscape of extracellular vesicles from a post-traumatic model of equine osteoarthritis. Front Vet Sci 2022; 9:901269. [PMID: 36003409 PMCID: PMC9393553 DOI: 10.3389/fvets.2022.901269] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 07/18/2022] [Indexed: 12/12/2022] Open
Abstract
Extracellular vesicles comprise an as yet inadequately investigated intercellular communication pathway in the field of early osteoarthritis. We hypothesised that the small non-coding RNA expression pattern in synovial fluid and plasma would change during progression of experimental osteoarthritis. In this study, we conducted small RNA sequencing to provide a comprehensive overview of the temporal expression profiles of small non-coding transcripts carried by extracellular vesicles derived from plasma and synovial fluid for the first time in a posttraumatic model of equine osteoarthritis. Additionally, we characterised synovial fluid and plasma-derived extracellular vesicles with respect to quantity, size, and surface markers. The different temporal expressions of seven microRNAs in plasma and synovial fluid-derived extracellular vesicles, eca-miR-451, eca-miR-25, eca-miR-215, eca-miR-92a, eca-miR-let-7c, eca-miR-486-5p, and eca-miR-23a, and four snoRNAs, U3, snord15, snord46, and snord58, represent potential biomarkers for early osteoarthritis. Bioinformatics analysis of the differentially expressed microRNAs in synovial fluid highlighted that in early osteoarthritis these related to the inhibition of cell cycle, cell cycle progression, DNA damage and cell proliferation as well as increased cell viability and differentiation of stem cells. Plasma and synovial fluid-derived extracellular vesicle small non-coding signatures have been established for the first time in a temporal model of osteoarthritis. These could serve as novel biomarkers for evaluation of osteoarthritis progression or act as potential therapeutic targets.
Collapse
Affiliation(s)
- James R. Anderson
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Stine Jacobsen
- Department of Veterinary Clinical Sciences, University of Copenhagen, Taastrup, Denmark
| | - Marie Walters
- Department of Veterinary Clinical Sciences, University of Copenhagen, Taastrup, Denmark
| | - Louise Bundgaard
- Department of Veterinary Clinical Sciences, University of Copenhagen, Taastrup, Denmark
| | | | | | - Emily J. Clarke
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Victoria James
- School of Veterinary Medicine and Science, University of Nottingham, Loughborough, United Kingdom
| | - Mandy J. Peffers
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
10
|
Koch CM, Prigge AD, Anekalla KR, Shukla A, Do Umehara HC, Setar L, Chavez J, Abdala-Valencia H, Politanska Y, Markov NS, Hahn GR, Heald-Sargent T, Sanchez- Pinto LN, Muller WJ, Singer BD, Misharin AV, Ridge KM, Coates BM. Age-related Differences in the Nasal Mucosal Immune Response to SARS-CoV-2. Am J Respir Cell Mol Biol 2022; 66:206-222. [PMID: 34731594 PMCID: PMC8845137 DOI: 10.1165/rcmb.2021-0292oc] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 11/01/2021] [Indexed: 11/24/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has infected more than 180 million people since the onset of the pandemic. Despite similar viral load and infectivity rates between children and adults, children rarely develop severe illness. Differences in the host response to the virus at the primary infection site are among the mechanisms proposed to account for this disparity. Our objective was to investigate the host response to SARS-CoV-2 in the nasal mucosa in children and adults and compare it with the host response to respiratory syncytial virus (RSV) and influenza virus. We analyzed clinical outcomes and gene expression in the nasal mucosa of 36 children with SARS-CoV-2, 24 children with RSV, 9 children with influenza virus, 16 adults with SARS-CoV-2, and 7 healthy pediatric and 13 healthy adult controls. In both children and adults, infection with SARS-CoV-2 led to an IFN response in the nasal mucosa. The magnitude of the IFN response correlated with the abundance of viral reads, not the severity of illness, and was comparable between children and adults infected with SARS-CoV-2 and children with severe RSV infection. Expression of ACE2 and TMPRSS2 did not correlate with age or presence of viral infection. SARS-CoV-2-infected adults had increased expression of genes involved in neutrophil activation and T-cell receptor signaling pathways compared with SARS-CoV-2-infected children, despite similar severity of illness and viral reads. Age-related differences in the immune response to SARS-CoV-2 may place adults at increased risk of developing severe illness.
Collapse
Affiliation(s)
| | - Andrew D. Prigge
- Department of Pediatrics
- Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, Illinois
| | | | - Avani Shukla
- Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, Illinois
| | | | - Leah Setar
- Department of Pediatrics
- Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, Illinois
| | - Jairo Chavez
- Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, Illinois
| | | | | | | | - Grant R. Hahn
- Department of Pediatrics
- Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, Illinois
| | - Taylor Heald-Sargent
- Department of Pediatrics
- Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, Illinois
| | - L. Nelson Sanchez- Pinto
- Department of Pediatrics
- Department of Preventive Medicine
- Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, Illinois
| | - William J. Muller
- Department of Pediatrics
- Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, Illinois
| | - Benjamin D. Singer
- Department of Medicine
- Department of Biochemistry and Molecular Genetics, and
| | | | - Karen M. Ridge
- Department of Medicine
- Department of Cell and Developmental Biology, Northwestern University, Evanston, Illinois; and
| | - Bria M. Coates
- Department of Pediatrics
- Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, Illinois
| |
Collapse
|
11
|
Abstract
PURPOSE OF REVIEW Translation of genetic information encoded within mRNA molecules by ribosomes into proteins is a key part of the central dogma of molecular biology. Despite the central position of the ribosome in the translation of proteins, and considering the major proteomic changes that occur in the joint during osteoarthritis development and progression, the ribosome has received very limited attention as driver of osteoarthritis pathogenesis. RECENT FINDINGS We provide an overview of the limited literature regarding this developing topic for the osteoarthritis field. Recent key findings that connect ribosome biogenesis and activity with osteoarthritis include: ribosomal RNA transcription, processing and maturation, ribosomal protein expression, protein translation capacity and preferential translation. SUMMARY The ribosome as the central cellular protein synthesis hub is largely neglected in osteoarthritis research. Findings included in this review reveal that in osteoarthritis, ribosome aberrations have been found from early-stage ribosome biogenesis, through ribosome build-up and maturation, up to preferential translation. Classically, osteoarthritis has been explained as an imbalance between joint tissue anabolism and catabolism. We postulate that osteoarthritis can be interpreted as an acquired ribosomopathy. This hypothesis fine-tunes the dogmatic anabolism/katabolism point-of-view, and may provide novel molecular opportunities for the development of osteoarthritis disease-modifying treatments.
Collapse
Affiliation(s)
- Guus G.H. van den Akker
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University
| | - Marjolein M.J. Caron
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University
| | - Mandy J. Peffers
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Tim J.M. Welting
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University Medical Center, Maastricht, the Netherlands
| |
Collapse
|
12
|
Castanheira CIGD, Anderson JR, Fang Y, Milner PI, Goljanek-Whysall K, House L, Clegg PD, Peffers MJ. Mouse microRNA signatures in joint ageing and post-traumatic osteoarthritis. OSTEOARTHRITIS AND CARTILAGE OPEN 2021; 3:100186. [PMID: 34977596 PMCID: PMC8683752 DOI: 10.1016/j.ocarto.2021.100186] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 05/19/2021] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVE This study investigated mice serum and joint microRNA expression profiles in ageing and osteoarthritis to elucidate the role of microRNAs in the development and progression of disease, and provide biomarkers for ageing and osteoarthritis. DESIGN Whole joints and serum samples were collected from C57BL6/J male mice and subjected to small RNA sequencing. Groups used included; surgically-induced post-traumatic osteoarthritis, (DMM; 24 months-old); sham surgery (24 months-old); old mice (18 months-old); and young mice (8 months-old). Differentially expressed microRNAs between the four groups were identified and validated using real-time quantitative PCR. MicroRNA differential expression data was used for target prediction and pathway analysis. RESULTS In joint tissues, miR-140-5p, miR-205-5p, miR-682, miR-208b-3p, miR-499-5p, miR-455-3p and miR-6238 were differentially expressed between young and old groups; miR-146a-5p, miR-3474, miR-615-3p and miR-151-5p were differentially expressed between DMM and Sham groups; and miR-652-3p, miR-23b-3p, miR-708-5p, miR-5099, miR-23a-3p, miR-214-3p, miR-6238 and miR-148-3p between the old and DMM groups. The number of differentially expressed microRNAs in serum was higher, some in common with joint tissues including miR-140-5p and miR-455-3p between young and old groups; and miR-23b-3p, miR-5099 and miR-6238 between old and DMM groups.We confirmed miR-140-5p, miR-499-5p and miR-455-3p expression to be decreased in old mouse joints compared to young, suggesting their potential use as biomarkers of joint ageing in mice. CONCLUSIONS MiR-140-5p, miR-499-5p and miR-455-3p could be used as joint ageing biomarkers in mice. Further research into these specific molecules in human tissues is now warranted to check their potential suitability as human biomarkers of ageing.
Collapse
Affiliation(s)
- Catarina I G D Castanheira
- Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, L7 8TX, UK
| | - James R Anderson
- Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, L7 8TX, UK
| | - Yongxiang Fang
- Centre for Genomic Research, Institute of Systems, Molecular and Integrative Biology, Biosciences Building, Crown Street, University of Liverpool, Liverpool, L69 7ZB, UK
| | - Peter I Milner
- Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, L7 8TX, UK
| | - Katarzyna Goljanek-Whysall
- Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, L7 8TX, UK
| | - Louise House
- Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, L7 8TX, UK
| | - Peter D Clegg
- Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, L7 8TX, UK
| | - Mandy J Peffers
- Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, L7 8TX, UK
| |
Collapse
|
13
|
Fitz NF, Wang J, Kamboh MI, Koldamova R, Lefterov I. Small nucleolar RNAs in plasma extracellular vesicles and their discriminatory power as diagnostic biomarkers of Alzheimer's disease. Neurobiol Dis 2021; 159:105481. [PMID: 34411703 PMCID: PMC9382696 DOI: 10.1016/j.nbd.2021.105481] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 07/20/2021] [Accepted: 08/09/2021] [Indexed: 12/13/2022] Open
Abstract
The clinical diagnosis of Alzheimer's disease, at its early stage, remains a difficult task. Advanced imaging technologies and laboratory assays to detect Aβ peptides Aβ42 and Aβ40, total and phosphorylated tau in CSF provide a set of biomarkers of developing AD brain pathology and facilitate the diagnostic process. The search for biofluid biomarkers, other than in CSF, and the development of biomarker assays have accelerated significantly and now represent the fastest-growing field in AD research. The goal of this study was to determine the differential enrichment of noncoding RNAs (ncRNAs) in plasma-derived extracellular vesicles (EV) of AD patients and Cognitively Normal controls (NC). Using RNA-seq, we profiled four significant classes of ncRNAs: miRNAs, snoRNAs, tRNAs, and piRNAs. We report a significant enrichment of SNORDs - a group of snoRNAs, in AD samples compared to NC. To verify the differential enrichment of two clusters of SNORDs - SNORD115 and SNORD116, localized on human chromosome 15q11-q13, we used plasma samples of an independent group of AD patients and NC. We applied ddPCR technique and identified SNORD115 and SNORD116 with a high discriminatory power to differentiate AD samples from NC. The results of our study present evidence that AD is associated with changes in the enrichment of SNORDs, transcribed from imprinted genomic loci, in plasma EV and provide a rationale to further explore the validity of those SNORDs as plasma biomarkers of AD.
Collapse
Affiliation(s)
- Nicholas F Fitz
- Department of Environmental & Occupational Health, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, United States of America
| | - Jiebiao Wang
- Department of Biostatistics, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, United States of America
| | - M Ilyas Kamboh
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, United States of America
| | - Radosveta Koldamova
- Department of Environmental & Occupational Health, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, United States of America.
| | - Iliya Lefterov
- Department of Environmental & Occupational Health, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, United States of America.
| |
Collapse
|
14
|
Abdel Salam L, Aldarwesh AQ, Eleishi HH. Whole exome sequencing (WES) of methotrexate response/adverse event profile in rheumatoid arthritis patients. THE EGYPTIAN RHEUMATOLOGIST 2021. [DOI: 10.1016/j.ejr.2020.08.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/10/2023]
|
15
|
Kharaz YA, Birch H, Chester A, Alchorne E, Simpson D, Clegg P, Comerford E. The effect of exercise on the protein profile of rat knee joint intra- and extra-articular ligaments. Scand J Med Sci Sports 2021; 31:2033-2043. [PMID: 34271594 DOI: 10.1111/sms.14023] [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: 06/14/2021] [Accepted: 07/15/2021] [Indexed: 01/13/2023]
Abstract
Injuries to the intra-articular anterior cruciate ligament (ACL) and the extra-articular medial collateral ligament (MCL) result in significant knee joint instability, pain, and immobility. Moderate endurance-type exercise can increase ligament strength but little is known on the effect of short-term regular bouts of high-intensity exercise on the extracellular matrix (ECM) structure of knee ligaments. Therefore, this study aimed to identify the effect of short-term regular bouts high exercise on the proteome of the rat ACL and MCL using mass spectrometry. Sprague-Dawley male rats (n = 6) were split into control and exercise groups, and subjected to high-intensity training for four 4 weeks followed by proteomic analyses of the ACL and MCL. Knee joint health status was assessed using OARSI and a validated histological scoring system. Histopathological analyses demonstrated no significant changes in either in cruciate, collateral ligaments, or cartilage between the control and exercised knee joints. However, significant proteins were found to be more abundant in the exercised ACL compared to ACL control group but not between the exercised MCL and control MCL groups. The significant abundant proteins in ACL exercise groups were mostly cytoskeletal, ribosomal and enzymes with several abundant matrisomal proteins such as collagen proteins and proteoglycans being found in this group. In conclusion, our results indicate that short-term regular bouts of high-intensity exercise have an impact on the intra-articular ACL but not extra-articular MCL ECM protein expression.
Collapse
Affiliation(s)
- Yalda A Kharaz
- Department of Musculoskeletal and Ageing Sciences, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Helen Birch
- Department of Orthopaedics and Musculoskeletal Science, University College London, London, UK
| | | | | | - Deborah Simpson
- Centre for Proteome Research, Institute of Integrative Biology, University of Liverpool, Liverpool, UK
| | - Peter Clegg
- Department of Musculoskeletal and Ageing Sciences, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK.,School of Veterinary Science, Leahurst Campus, Neston, UK
| | - Eithne Comerford
- Department of Musculoskeletal and Ageing Sciences, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK.,School of Veterinary Science, Leahurst Campus, Neston, UK
| |
Collapse
|
16
|
Koch CM, Prigge AD, Anekalla KR, Shukla A, Do-Umehara HC, Setar L, Chavez J, Abdala-Valencia H, Politanska Y, Markov NS, Hahn GR, Heald-Sargent T, Sanchez-Pinto LN, Muller WJ, Misharin AV, Ridge KM, Coates BM. Immune response to SARS-CoV-2 in the nasal mucosa in children and adults. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021:2021.01.26.21250269. [PMID: 33532801 PMCID: PMC7852252 DOI: 10.1101/2021.01.26.21250269] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
RATIONALE Despite similar viral load and infectivity rates between children and adults infected with SARS-CoV-2, children rarely develop severe illness. Differences in the host response to the virus at the primary infection site are among the proposed mechanisms. OBJECTIVES To investigate the host response to SARS-CoV-2, respiratory syncytial virus (RSV), and influenza virus (IV) in the nasal mucosa in children and adults. METHODS Clinical outcomes and gene expression in the nasal mucosa were analyzed in 36 children hospitalized with SARS-CoV-2 infection, 24 children with RSV infection, 9 children with IV infection, 16 adults with mild to moderate SARS-CoV-2 infection, and 7 healthy pediatric and 13 healthy adult controls. RESULTS In both children and adults, infection with SARS-CoV-2 leads to an interferon response in the nasal mucosa. The magnitude of the interferon response correlated with the abundance of viral reads and was comparable between symptomatic children and adults infected with SARS-CoV-2 and symptomatic children infected with RSV and IV. Cell type deconvolution identified an increased abundance of immune cells in the samples from children and adults with a viral infection. Expression of ACE2 and TMPRSS2 - key entry factors for SARS-CoV-2 - did not correlate with age or presence or absence of viral infection. CONCLUSIONS Our findings support the hypothesis that differences in the immune response to SARS-CoV-2 determine disease severity, independent of viral load and interferon response at the primary infection primary site.
Collapse
Affiliation(s)
| | - Andrew D Prigge
- Department of Pediatrics, Northwestern University
- Ann & Robert H. Lurie Children's Hospital of Chicago
| | | | - Avani Shukla
- Ann & Robert H. Lurie Children's Hospital of Chicago
| | | | - Leah Setar
- Department of Pediatrics, Northwestern University
- Ann & Robert H. Lurie Children's Hospital of Chicago
| | - Jairo Chavez
- Ann & Robert H. Lurie Children's Hospital of Chicago
| | | | | | | | - Grant R Hahn
- Department of Pediatrics, Northwestern University
- Ann & Robert H. Lurie Children's Hospital of Chicago
| | - Taylor Heald-Sargent
- Department of Pediatrics, Northwestern University
- Ann & Robert H. Lurie Children's Hospital of Chicago
| | - L Nelson Sanchez-Pinto
- Department of Pediatrics, Northwestern University
- Ann & Robert H. Lurie Children's Hospital of Chicago
- Department of Preventive Medicine, Northwestern University
| | - William J Muller
- Department of Pediatrics, Northwestern University
- Ann & Robert H. Lurie Children's Hospital of Chicago
| | | | - Karen M Ridge
- Department of Medicine, Northwestern University
- Department of Cell and Developmental Biology, Northwestern University
| | - Bria M Coates
- Department of Pediatrics, Northwestern University
- Ann & Robert H. Lurie Children's Hospital of Chicago
| |
Collapse
|
17
|
Castanheira C, Balaskas P, Falls C, Ashraf-Kharaz Y, Clegg P, Burke K, Fang Y, Dyer P, Welting TJM, Peffers MJ. Equine synovial fluid small non-coding RNA signatures in early osteoarthritis. BMC Vet Res 2021; 17:26. [PMID: 33422071 PMCID: PMC7796526 DOI: 10.1186/s12917-020-02707-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 12/02/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Osteoarthritis remains one of the greatest causes of morbidity and mortality in the equine population. The inability to detect pre-clinical changes in osteoarthritis has been a significant impediment to the development of effective therapies against this disease. Synovial fluid represents a potential source of disease-specific small non-coding RNAs (sncRNAs) that could aid in the understanding of the pathogenesis of osteoarthritis. We hypothesised that early stages of osteoarthritis would alter the expression of sncRNAs, facilitating the understanding of the underlying pathogenesis and potentially provide early biomarkers. METHODS Small RNA sequencing was performed using synovial fluid from the metacarpophalangeal joints of both control and early osteoarthritic horses. A group of differentially expressed sncRNAs was selected for further validation through qRT-PCR using an independent cohort of synovial fluid samples from control and early osteoarthritic horses. Bioinformatic analysis was performed in order to identify putative targets of the differentially expressed microRNAs and to explore potential associations with specific biological processes. RESULTS Results revealed 22 differentially expressed sncRNAs including 13 microRNAs; miR-10a, miR-223, let7a, miR-99a, miR-23b, miR-378, miR-143 (and six novel microRNAs), four small nuclear RNAs; U2, U5, U11, U12, three small nucleolar RNAs; U13, snoR38, snord96, and one small cajal body-specific RNA; scarna3. Five sncRNAs were validated; miR-223 was significantly reduced in early osteoarthritis and miR-23b, let-7a-2, snord96A and snord13 were significantly upregulated. Significant cellular actions deduced by the differentially expressed microRNAs included apoptosis (P < 0.0003), necrosis (P < 0.0009), autophagy (P < 0.0007) and inflammation (P < 0.00001). A conservatively filtered list of 57 messenger RNA targets was obtained; the top biological processes associated were regulation of cell population proliferation (P < 0.000001), cellular response to chemical stimulus (P < 0.000001) and cell surface receptor signalling pathway (P < 0.000001). CONCLUSIONS Synovial fluid sncRNAs may be used as molecular biomarkers for early disease in equine osteoarthritic joints. The biological processes they regulate may play an important role in understanding early osteoarthritis pathogenesis. Characterising these dynamic molecular changes could provide novel insights on the process and mechanism of early osteoarthritis development and is critical for the development of new therapeutic approaches.
Collapse
Affiliation(s)
- Catarina Castanheira
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, William Henry Duncan Building, 6 West Derby Street, Liverpool, L7 8TX UK
| | - Panagiotis Balaskas
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, William Henry Duncan Building, 6 West Derby Street, Liverpool, L7 8TX UK
| | - Charlotte Falls
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, William Henry Duncan Building, 6 West Derby Street, Liverpool, L7 8TX UK
| | - Yalda Ashraf-Kharaz
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, William Henry Duncan Building, 6 West Derby Street, Liverpool, L7 8TX UK
| | - Peter Clegg
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, William Henry Duncan Building, 6 West Derby Street, Liverpool, L7 8TX UK
| | - Kim Burke
- Institute of Veterinary Science, University of Liverpool, Chester High Road, Neston, CH64 7TE UK
| | - Yongxiang Fang
- Centre for Genomic Research, Institute of Integrative Biology, University of Liverpool, Biosciences Building, Crown Street, Liverpool, L69 7ZB UK
| | - Philip Dyer
- Institute of Infection and Global Health, University of Liverpool, 8 West Derby Street, Liverpool, L7 3EA UK
| | - Tim J. M. Welting
- Department of Orthopaedic Surgery, Maastricht University Medical Centre, Maastricht, AZ 6202 The Netherlands
| | - Mandy J. Peffers
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, William Henry Duncan Building, 6 West Derby Street, Liverpool, L7 8TX UK
| |
Collapse
|
18
|
Impaired chondrocyte U3 snoRNA expression in osteoarthritis impacts the chondrocyte protein translation apparatus. Sci Rep 2020; 10:13426. [PMID: 32778764 PMCID: PMC7417995 DOI: 10.1038/s41598-020-70453-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Accepted: 07/23/2020] [Indexed: 12/18/2022] Open
Abstract
Although pathways controlling ribosome activity have been described to regulate chondrocyte homeostasis in osteoarthritis, ribosome biogenesis in osteoarthritis is unexplored. We hypothesized that U3 snoRNA, a non-coding RNA involved in ribosomal RNA maturation, is critical for chondrocyte protein translation capacity in osteoarthritis. U3 snoRNA was one of a number of snoRNAs with decreased expression in osteoarthritic cartilage and osteoarthritic chondrocytes. OA synovial fluid impacted U3 snoRNA expression by affecting U3 snoRNA gene promoter activity, while BMP7 was able to increase its expression. Altering U3 snoRNA expression resulted in changes in chondrocyte phenotype. Interference with U3 snoRNA expression led to reduction of rRNA levels and translational capacity, whilst induced expression of U3 snoRNA was accompanied by increased 18S and 28S rRNA levels and elevated protein translation. Whole proteome analysis revealed a global impact of reduced U3 snoRNA expression on protein translational processes and inflammatory pathways. For the first time we demonstrate implications of a snoRNA in osteoarthritis chondrocyte biology and investigated its role in the chondrocyte differentiation status, rRNA levels and protein translational capacity.
Collapse
|
19
|
Balaskas P, Green JA, Haqqi TM, Dyer P, Kharaz YA, Fang Y, Liu X, Welting TJ, Peffers MJ. Small Non-Coding RNAome of Ageing Chondrocytes. Int J Mol Sci 2020; 21:E5675. [PMID: 32784773 PMCID: PMC7461137 DOI: 10.3390/ijms21165675] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/03/2020] [Accepted: 08/04/2020] [Indexed: 12/27/2022] Open
Abstract
Ageing is a leading risk factor predisposing cartilage to osteoarthritis. However, little research has been conducted on the effect of ageing on the expression of small non-coding RNAs (sncRNAs). RNA from young and old chondrocytes from macroscopically normal equine metacarpophalangeal joints was extracted and subjected to small RNA sequencing (RNA-seq). Differential expression analysis was performed in R using package DESeq2. For transfer RNA (tRNA) fragment analysis, tRNA reads were aligned to horse tRNA sequences using Bowtie2 version 2.2.5. Selected microRNA (miRNAs or miRs) and small nucleolar RNA (snoRNA) findings were validated using real-time quantitative Polymerase Chain Reaction (qRT-PCR) in an extended cohort of equine chondrocytes. tRNA fragments were further investigated in low- and high-grade OA human cartilage tissue. In total, 83 sncRNAs were differentially expressed between young and old equine chondrocytes, including miRNAs, snoRNAs, small nuclear RNAs (snRNAs), and tRNAs. qRT-PCR analysis confirmed findings. tRNA fragment analysis revealed that tRNA halves (tiRNAs), tiRNA-5035-GluCTC and tiRNA-5031-GluCTC-1 were reduced in both high grade OA human cartilage and old equine chondrocytes. For the first time, we have measured the effect of ageing on the expression of sncRNAs in equine chondrocytes. Changes were detected in a number of different sncRNA species. This study supports a role for sncRNAs in ageing cartilage and their potential involvement in age-related cartilage diseases.
Collapse
Affiliation(s)
- Panagiotis Balaskas
- Institute of Life Course and Medical Sciences, William Henry Duncan Building, 6 West Derby Street, Liverpool L7 8TX, UK; (P.D.); (Y.A.K.)
| | - Jonathan A. Green
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University, Rootstown, OH 44272, USA; (J.A.G.); (T.M.H.)
| | - Tariq M. Haqqi
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University, Rootstown, OH 44272, USA; (J.A.G.); (T.M.H.)
| | - Philip Dyer
- Institute of Life Course and Medical Sciences, William Henry Duncan Building, 6 West Derby Street, Liverpool L7 8TX, UK; (P.D.); (Y.A.K.)
| | - Yalda A. Kharaz
- Institute of Life Course and Medical Sciences, William Henry Duncan Building, 6 West Derby Street, Liverpool L7 8TX, UK; (P.D.); (Y.A.K.)
| | - Yongxiang Fang
- Centre for Genomic Research, Institute of Integrative Biology, Biosciences Building, Crown Street, University of Liverpool, Liverpool L69 7ZB, UK; (Y.F.); (X.L.)
| | - Xuan Liu
- Centre for Genomic Research, Institute of Integrative Biology, Biosciences Building, Crown Street, University of Liverpool, Liverpool L69 7ZB, UK; (Y.F.); (X.L.)
| | - Tim J.M. Welting
- Department of Orthopaedic Surgery, Maastricht University Medical Centre, 6202 AZ Maastricht, The Netherlands;
| | - Mandy J. Peffers
- Institute of Life Course and Medical Sciences, William Henry Duncan Building, 6 West Derby Street, Liverpool L7 8TX, UK; (P.D.); (Y.A.K.)
| |
Collapse
|
20
|
Peffers MJ, Chabronova A, Balaskas P, Fang Y, Dyer P, Cremers A, Emans PJ, Feczko PZ, Caron MM, Welting TJM. SnoRNA signatures in cartilage ageing and osteoarthritis. Sci Rep 2020; 10:10641. [PMID: 32606371 PMCID: PMC7326970 DOI: 10.1038/s41598-020-67446-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 06/08/2020] [Indexed: 01/23/2023] Open
Abstract
Osteoarthritis presents as a change in the chondrocyte phenotype and an imbalance between anabolic and catabolic processes. Age affects its onset and progression. Small nucleolar RNAs (SnoRNAs) direct chemical modification of RNA substrates to fine-tune spliceosomal and rRNA function, accommodating changing requirements for splicing and protein synthesis during health and disease. Articular cartilage from young, old and OA knees was used in a microarray study to identify alterations in snoRNA expression. Changes in snoRNAs in osteoarthritis-like conditions were studied in chondrocytes using interleukin-1 and osteoarthritic synovial fluid. SNORD26 and SNORD96A knockdown and overexpression were undertaken using antisense oligonucleotides and overexpression plasmids. We identified panels of snoRNAs differentially expressed due to ageing (including SNORD96A, SNORD44) and osteoarthritis (including SNORD26 and SNORD116). In vitro experiments using osteoarthritis-like conditions affected snoRNA expression. Knockdown or overexpression of SNORD26 or SNORD96A resulted in changes in chondrogenic, hypertrophic, rRNA and osteoarthritis related gene expression. We demonstrate that snoRNA expression changes in cartilage ageing, and osteoarthritis and in osteoarthritis-like conditions, and when the expression of these snoRNAs is altered this affects chondrogenic and hypertrophic gene expression. Thus, we propose an additional dimension in the molecular mechanisms underlying cartilage ageing and osteoarthritis through the dysregulation of snoRNAs.
Collapse
Affiliation(s)
- Mandy J Peffers
- Institute of Ageing and Chronic Disease, William Henry Duncan Building, 6 West Derby Street, Liverpool, L7 8TX, UK.
| | - Alzbeta Chabronova
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University Medical Centre, 6202 AZ, Maastricht, The Netherlands
| | - Panagiotis Balaskas
- Institute of Ageing and Chronic Disease, William Henry Duncan Building, 6 West Derby Street, Liverpool, L7 8TX, UK
| | - Yongxiang Fang
- Centre for Genomic Research, Institute of Integrative Biology, University of Liverpool, Biosciences Building, Crown Street, Liverpool, L69 7ZB, UK
| | - Philip Dyer
- Institute of Ageing and Chronic Disease, William Henry Duncan Building, 6 West Derby Street, Liverpool, L7 8TX, UK
| | - Andy Cremers
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University Medical Centre, 6202 AZ, Maastricht, The Netherlands
| | - Pieter J Emans
- Department of Orthopaedic Surgery, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Peter Z Feczko
- Department of Orthopaedic Surgery, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Marjolein M Caron
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University Medical Centre, 6202 AZ, Maastricht, The Netherlands
| | - Tim J M Welting
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University Medical Centre, 6202 AZ, Maastricht, The Netherlands.,Department of Orthopaedic Surgery, Maastricht University Medical Center, Maastricht, The Netherlands
| |
Collapse
|
21
|
Kuksa PP, Amlie-Wolf A, Katanić Ž, Valladares O, Wang LS, Leung YY. DASHR 2.0: integrated database of human small non-coding RNA genes and mature products. Bioinformatics 2019; 35:1033-1039. [PMID: 30668832 PMCID: PMC6419920 DOI: 10.1093/bioinformatics/bty709] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 05/31/2018] [Accepted: 08/20/2018] [Indexed: 12/23/2022] Open
Abstract
Motivation Small non-coding RNAs (sncRNAs, <100 nts) are highly abundant RNAs that regulate diverse and often tissue-specific cellular processes by associating with transcription factor complexes or binding to mRNAs. While thousands of sncRNA genes exist in the human genome, no single resource provides searchable, unified annotation, expression and processing information for full sncRNA transcripts and mature RNA products derived from these larger RNAs. Results Our goal is to establish a complete catalog of annotation, expression, processing, conservation, tissue-specificity and other biological features for all human sncRNA genes and mature products derived from all major RNA classes. DASHR (Database of small human non-coding RNAs) v2.0 database is the first that integrates human sncRNA gene and mature products profiles obtained from multiple RNA-seq protocols. Altogether, 185 tissues/cell types and sncRNA annotations and >800 curated experiments from ENCODE and GEO/SRA across multiple RNA-seq protocols for both GRCh38/hg38 and GRCh37/hg19 assemblies are integrated in DASHR. Moreover, DASHR is the first to contain both known and novel, previously un-annotated sncRNA loci identified by unsupervised segmentation (13 times more loci with 1 678 800 total). Additionally, DASHR v2.0 adds >3 200 000 annotations for non-small RNA genes and other genomic features (long-noncoding RNAs, mRNAs, promoters, repeats). Furthermore, DASHR v2.0 introduces an enhanced user interface, interactive experiment-by-locus table view, sncRNA locus sorting and filtering by biological features. All annotation and expression information directly downloadable and accessible as UCSC genome browser tracks. Availability and implementation DASHR v2.0 is freely available at https://lisanwanglab.org/DASHRv2. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Pavel P Kuksa
- Penn Neurodegeneration Genomics Center, Department of Pathology and Laboratory Medicine
| | - Alexandre Amlie-Wolf
- Penn Neurodegeneration Genomics Center, Department of Pathology and Laboratory Medicine.,Genomics and Computational Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Živadin Katanić
- Penn Neurodegeneration Genomics Center, Department of Pathology and Laboratory Medicine
| | - Otto Valladares
- Penn Neurodegeneration Genomics Center, Department of Pathology and Laboratory Medicine
| | - Li-San Wang
- Penn Neurodegeneration Genomics Center, Department of Pathology and Laboratory Medicine.,Genomics and Computational Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yuk Yee Leung
- Penn Neurodegeneration Genomics Center, Department of Pathology and Laboratory Medicine
| |
Collapse
|
22
|
Razmara E, Bitaraf A, Yousefi H, Nguyen TH, Garshasbi M, Cho WCS, Babashah S. Non-Coding RNAs in Cartilage Development: An Updated Review. Int J Mol Sci 2019; 20:E4475. [PMID: 31514268 PMCID: PMC6769748 DOI: 10.3390/ijms20184475] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 09/02/2019] [Accepted: 09/02/2019] [Indexed: 02/06/2023] Open
Abstract
In the development of the skeleton, the long bones are arising from the process of endochondral ossification (EO) in which cartilage is replaced by bone. This complex process is regulated by various factors including genetic, epigenetic, and environmental elements. It is recognized that DNA methylation, higher-order chromatin structure, and post-translational modifications of histones regulate the EO. With emerging understanding, non-coding RNAs (ncRNAs) have been identified as another mode of EO regulation, which is consist of microRNAs (miRNAs or miRs) and long non-coding RNAs (lncRNAs). There is expanding experimental evidence to unlock the role of ncRNAs in the differentiation of cartilage cells, as well as the pathogenesis of several skeletal disorders including osteoarthritis. Cutting-edge technologies such as epigenome-wide association studies have been employed to reveal disease-specific patterns regarding ncRNAs. This opens a new avenue of our understanding of skeletal cell biology, and may also identify potential epigenetic-based biomarkers. In this review, we provide an updated overview of recent advances in the role of ncRNAs especially focus on miRNA and lncRNA in the development of bone from cartilage, as well as their roles in skeletal pathophysiology.
Collapse
Affiliation(s)
- Ehsan Razmara
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran P.O. Box 14115-111, Iran
| | - Amirreza Bitaraf
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran P.O. Box 14115-111, Iran
| | - Hassan Yousefi
- Department of Biochemistry and Molecular Biology, LSUHSC School of Medicine, New Orleans, LA 70112, USA
| | - Tina H Nguyen
- Department of Biochemistry and Molecular Biology, LSUHSC School of Medicine, New Orleans, LA 70112, USA
| | - Masoud Garshasbi
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran P.O. Box 14115-111, Iran
| | | | - Sadegh Babashah
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran P.O. Box 14115-111, Iran.
| |
Collapse
|
23
|
Kuksa PP, Amlie-Wolf A, Katanic Ž, Valladares O, Wang LS, Leung YY. SPAR: small RNA-seq portal for analysis of sequencing experiments. Nucleic Acids Res 2019; 46:W36-W42. [PMID: 29733404 PMCID: PMC6030839 DOI: 10.1093/nar/gky330] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 04/17/2018] [Indexed: 02/01/2023] Open
Abstract
The introduction of new high-throughput small RNA sequencing protocols that generate large-scale genomics datasets along with increasing evidence of the significant regulatory roles of small non-coding RNAs (sncRNAs) have highlighted the urgent need for tools to analyze and interpret large amounts of small RNA sequencing data. However, it remains challenging to systematically and comprehensively discover and characterize sncRNA genes and specifically-processed sncRNA products from these datasets. To fill this gap, we present Small RNA-seq Portal for Analysis of sequencing expeRiments (SPAR), a user-friendly web server for interactive processing, analysis, annotation and visualization of small RNA sequencing data. SPAR supports sequencing data generated from various experimental protocols, including smRNA-seq, short total RNA sequencing, microRNA-seq, and single-cell small RNA-seq. Additionally, SPAR includes publicly available reference sncRNA datasets from our DASHR database and from ENCODE across 185 human tissues and cell types to produce highly informative small RNA annotations across all major small RNA types and other features such as co-localization with various genomic features, precursor transcript cleavage patterns, and conservation. SPAR allows the user to compare the input experiment against reference ENCODE/DASHR datasets. SPAR currently supports analyses of human (hg19, hg38) and mouse (mm10) sequencing data. SPAR is freely available at https://www.lisanwanglab.org/SPAR.
Collapse
Affiliation(s)
- Pavel P Kuksa
- Penn Neurodegeneration Genomics Center, University of Pennsylvania, Philadelphia, PA 19104, USA.,Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Alexandre Amlie-Wolf
- Penn Neurodegeneration Genomics Center, University of Pennsylvania, Philadelphia, PA 19104, USA.,Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.,Genomics and Computational Biology Graduate Group, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Živadin Katanic
- Penn Neurodegeneration Genomics Center, University of Pennsylvania, Philadelphia, PA 19104, USA.,Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Otto Valladares
- Penn Neurodegeneration Genomics Center, University of Pennsylvania, Philadelphia, PA 19104, USA.,Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Li-San Wang
- Penn Neurodegeneration Genomics Center, University of Pennsylvania, Philadelphia, PA 19104, USA.,Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.,Genomics and Computational Biology Graduate Group, University of Pennsylvania, Philadelphia, PA 19104, USA.,Institute on Aging, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yuk Yee Leung
- Penn Neurodegeneration Genomics Center, University of Pennsylvania, Philadelphia, PA 19104, USA.,Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
24
|
McDermott BT, Peffers MJ, McDonagh B, Tew SR. Translational regulation contributes to the secretory response of chondrocytic cells following exposure to interleukin-1β. J Biol Chem 2019; 294:13027-13039. [PMID: 31300557 PMCID: PMC6721953 DOI: 10.1074/jbc.ra118.006865] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 06/12/2019] [Indexed: 01/18/2023] Open
Abstract
Osteoarthritis is a chronic disease characterized by the loss of articular cartilage in synovial joints through a process of extracellular matrix destruction that is strongly associated with inflammatory stimuli. Chondrocytes undergo changes to their protein translational capacity during osteoarthritis, but a study of how disease-relevant signals affect chondrocyte protein translation at the transcriptomic level has not previously been performed. In this study, we describe how the inflammatory cytokine interleukin 1-β (IL-1β) rapidly affects protein translation in the chondrocytic cell line SW1353. Using ribosome profiling we demonstrate that IL-1β induced altered translation of inflammatory-associated transcripts such as NFKB1, TNFAIP2, MMP13, CCL2, and CCL7, as well as a number of ribosome-associated transcripts, through differential translation and the use of multiple open reading frames. Proteomic analysis of the cellular layer and the conditioned media of these cells identified changes in a number of the proteins that were differentially translated. Translationally regulated secreted proteins included a number of chemokines and cytokines, underlining the rapid, translationally mediated inflammatory cascade that is initiated by IL-1β. Although fewer cellular proteins were found to be regulated in both ribosome profiling and proteomic data sets, we did find increased levels of SOD2, indicative of redox changes within SW1353 cells being modulated at the translational level. In conclusion, we have produced combined ribosome profiling and proteomic data sets that provide a valuable resource in understanding the processes that occur during cytokine stimulation of chondrocytic cells.
Collapse
Affiliation(s)
- Benjamin T McDermott
- Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, University of Liverpool, William Henry Duncan Building, 6 West Derby Street, Liverpool L7 8TX, United Kingdom.
| | - Mandy J Peffers
- Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, University of Liverpool, William Henry Duncan Building, 6 West Derby Street, Liverpool L7 8TX, United Kingdom
| | - Brian McDonagh
- Department of Physiology, School of Medicine, National University of Ireland (NUI), Galway H91 TK33, Ireland
| | - Simon R Tew
- Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, University of Liverpool, William Henry Duncan Building, 6 West Derby Street, Liverpool L7 8TX, United Kingdom
| |
Collapse
|
25
|
Joilin G, Leigh PN, Newbury SF, Hafezparast M. An Overview of MicroRNAs as Biomarkers of ALS. Front Neurol 2019; 10:186. [PMID: 30899244 PMCID: PMC6416171 DOI: 10.3389/fneur.2019.00186] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 02/13/2019] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS; MND, motor neuron disease) is a debilitating neurodegenerative disease affecting 4.5 per 100,000 people per year around the world. There is currently no cure for this disease, and its causes are relatively unknown. Diagnosis is based on a battery of clinical tests up to a year after symptom onset, with no robust markers of diagnosis or disease progression currently identified. A major thrust of current research is to identify potential non-invasive markers (“biomarkers”) in body fluids such as blood and/or cerebrospinal fluid (CSF) to use for diagnostic or prognostic purposes. Non-coding RNAs (ncRNAs), including microRNAs (miRNAs), are found at detectable and stable levels in blood and other bodily fluids. Specific ncRNAs can vary in levels between ALS patients and non-ALS controls without the disease. In this review, we will provide an overview of early findings, demonstrate the potential of this new class as biomarkers, and discuss future challenges and opportunities taking this forward to help patients with ALS.
Collapse
Affiliation(s)
- Greig Joilin
- School of Life Sciences, University of Sussex, Brighton, United Kingdom
| | - P Nigel Leigh
- Brighton and Sussex Medical School, University of Sussex, Brighton, United Kingdom
| | - Sarah F Newbury
- Brighton and Sussex Medical School, University of Sussex, Brighton, United Kingdom
| | - Majid Hafezparast
- School of Life Sciences, University of Sussex, Brighton, United Kingdom
| |
Collapse
|
26
|
Guide snoRNAs: Drivers or Passengers in Human Disease? BIOLOGY 2018; 8:biology8010001. [PMID: 30577491 PMCID: PMC6466398 DOI: 10.3390/biology8010001] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 12/16/2018] [Accepted: 12/18/2018] [Indexed: 01/17/2023]
Abstract
In every domain of life, RNA-protein interactions play a significant role in co- and post-transcriptional modifications and mRNA translation. RNA performs diverse roles inside the cell, and therefore any aberrancy in their function can cause various diseases. During maturation from its primary transcript, RNA undergoes several functionally important post-transcriptional modifications including pseudouridylation and ribose 2′-O-methylation. These modifications play a critical role in the stability of the RNA. In the last few decades, small nucleolar RNAs (snoRNAs) were revealed to be one of the main components to guide these modifications. Due to their active links to the nucleoside modification, deregulation in the snoRNA expressions can cause multiple disorders in humans. Additionally, host genes carrying snoRNA-encoding sequences in their introns also show differential expression in disease. Although few reports support a causal link between snoRNA expression and disease manifestation, this emerging field will have an impact on the way we think about biomarkers or identify novel targets for therapy. This review focuses on the intriguing aspect of snoRNAs that function as a guide in post-transcriptional RNA modification, and regulation of their host genes in human disease.
Collapse
|
27
|
Trzybulska D, Vergadi E, Tsatsanis C. miRNA and Other Non-Coding RNAs as Promising Diagnostic Markers. EJIFCC 2018; 29:221-226. [PMID: 30479608 PMCID: PMC6247131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Since the discovery of non-coding RNAs (ncRNAs) a new area has emerged in the field of biomarkers. NcRNAs are RNA molecules of different sizes that are transcribed as independent genes or as part of protein coding genes and are not translated, therefore they do not produce proteins. They have been classified according to their size and function and include microRNAs (miRNAs), piwiRNAs (piRNAs), snoRNAs and long non-coding RNAs (lncRNAs). These non-coding RNAs are present in different cell compartments participating in multiple cell functions, but they have also been identified in biological fluids, also known as cell-free or circulating ncRNAs, where they can be detected in exosomes, bound on lipoproteins as well as free circulating molecules. The role of circulating ncRNAs is still under investigation but are believed to be paracrine or endocrine messengers to systematically deliver signals between cells and tissues. Detecting ncRNAs in biological fluids has opened a new field in Clinical Chemistry utilizing them as biomarkers of diseases or prognostic markers for different pathological conditions. Herein, the different types of ncRNAs and their potential in the field of diagnostics are outlined.
Collapse
Affiliation(s)
- Dorota Trzybulska
- Molecular Reproductive Medicine, Department of Translational Medicine, Lund University, Malmö, Sweden
| | - Eleni Vergadi
- Department of Clinical Chemistry, School of Medicine, University of Crete, Heraklion, Greece
| | - Christos Tsatsanis
- Molecular Reproductive Medicine, Department of Translational Medicine, Lund University, Malmö, Sweden, Department of Clinical Chemistry, School of Medicine, University of Crete, Heraklion, Greece,Corresponding author: Christos Tsatsanis Medical School University of Crete Voutes, 70013 Heraklion, Crete Greece Phone: +30 2810394833 E-mail:
| |
Collapse
|
28
|
Nucleotide Modifications Decrease Innate Immune Response Induced by Synthetic Analogs of snRNAs and snoRNAs. Genes (Basel) 2018; 9:genes9110531. [PMID: 30400232 PMCID: PMC6266926 DOI: 10.3390/genes9110531] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 10/26/2018] [Indexed: 12/25/2022] Open
Abstract
Short nuclear regulatory RNAs play a key role in the main stages of maturation of the precursors of the major RNA species. Small nuclear RNAs (snRNAs) form the core of the spliceosome and are responsible for the splicing of pre-mRNA molecules. Small nucleolar RNAs (snoRNAs) direct post-transcriptional modification of pre-rRNAs. A promising strategy for the development of non-coding RNA (ncRNAs) mimicking molecules is the introduction of modified nucleotides, which are normally present in natural ncRNAs, into the structure of synthetic RNAs. We have created a set of snoRNAs and snRNA analogs and studied the effect of base modifications, specifically, pseudouridine (Ψ) and 5-methylcytidine (m⁵C), on the immune-stimulating and cytotoxic properties of these RNAs. Here, we performed a whole-transcriptome study of the influence of synthetic snoRNA analogs with various modifications on gene expression in human cells. Moreover, we confirmed the role of PKR in the recognition of snoRNA and snRNA analogs using the short hairpin RNA (shRNA) technique. We believe that the data obtained will contribute to the understanding of the role of nucleotide modification in ncRNA functions, and can be useful for creating the agents for gene regulation based on the structure of natural snoRNAs and snRNAs.
Collapse
|
29
|
Kemp GJ, Jackson MJ, McCloskey EV, Mathers JC, Birrell F, Clegg PD, Cuthbertson DJ, De Vito G, van Dieën JH, Del Din S, Eastell R, Garnero P, Goljanek-Whysall K, Hackl M, Hodgson R, Lord S, Mazzà C, McArdle A, Narici M, Peffers MJ, Schiaffino S. Towards a toolkit for the assessment and monitoring of musculoskeletal ageing. Age Ageing 2018; 47:774-777. [PMID: 30202858 PMCID: PMC6201831 DOI: 10.1093/ageing/afy078] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Indexed: 11/29/2022] Open
Abstract
The complexities and heterogeneity of the ageing process have slowed the development of consensus on appropriate biomarkers of healthy ageing. The MRC-Arthritis Research UK Centre for Integrated research into Musculoskeletal Ageing (CIMA) is a collaboration between researchers and clinicians at the Universities of Liverpool, Sheffield and Newcastle. One of CIMA’s objectives is to ‘Identify and share optimal techniques and approaches to monitor age-related changes in all musculoskeletal tissues, and to provide an integrated assessment of musculoskeletal function’, i.e. to develop a toolkit for assessing musculoskeletal ageing. This toolkit is envisaged as an instrument that can be used to characterise and quantify musculoskeletal function during ‘normal’ ageing, lend itself to use in large-scale, internationally important cohorts, and provide a set of biomarker outcome measures for epidemiological and intervention studies designed to enhance healthy musculoskeletal ageing. Such potential biomarkers include: biochemical measurements in biofluids or tissue samples, in vivo measurements of body composition, imaging of structural and physical properties, and functional tests. The CIMA Toolkit Working Group assessed candidate biomarkers of musculoskeletal ageing under these four headings, detailed their biological bases, strengths and limitations, and made practical recommendations for their use. In addition, the CIMA Toolkit Working Group identified gaps in the evidence base and suggested priorities for further research on biomarkers of musculoskeletal ageing.
Collapse
Affiliation(s)
- Graham J Kemp
- Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease (IACD), Faculty of Health and Life Sciences, University of Liverpool, Liverpool, UK
- The MRC-Arthritis Research UK Centre for Integrated Research into Musculoskeletal Ageing (CIMA), Newcastle upon Tyne, UK
| | - Malcolm J Jackson
- Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease (IACD), Faculty of Health and Life Sciences, University of Liverpool, Liverpool, UK
- The MRC-Arthritis Research UK Centre for Integrated Research into Musculoskeletal Ageing (CIMA), Newcastle upon Tyne, UK
| | - Eugene V McCloskey
- The MRC-Arthritis Research UK Centre for Integrated Research into Musculoskeletal Ageing (CIMA), Newcastle upon Tyne, UK
- Mellanby Centre for Bone Research, University of Sheffield, Sheffield, UK
| | - John C Mathers
- The MRC-Arthritis Research UK Centre for Integrated Research into Musculoskeletal Ageing (CIMA), Newcastle upon Tyne, UK
- Human Nutrition Research Centre, Institute of Cellular Medicine and Newcastle University Institute for Ageing, Newcastle University, Newcastle upon Tyne, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Kemp GJ, Birrell F, Clegg PD, Cuthbertson DJ, De Vito G, van Dieën JH, Del Din S, Eastell R, Garnero P, Goljanek–Whysall K, Hackl M, Hodgson R, Jackson MJ, Lord S, Mazzà C, McArdle A, McCloskey EV, Narici M, Peffers MJ, Schiaffino S, Mathers JC. Developing a toolkit for the assessment and monitoring of musculoskeletal ageing. Age Ageing 2018; 47:iv1-iv19. [PMID: 30203052 PMCID: PMC6127513 DOI: 10.1093/ageing/afy143] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 08/03/2018] [Indexed: 12/12/2022] Open
Abstract
The complexities and heterogeneity of the ageing process have slowed the development of consensus on appropriate biomarkers of healthy ageing. The Medical Research Council–Arthritis Research UK Centre for Integrated research into Musculoskeletal Ageing (CIMA) is a collaboration between researchers and clinicians at the Universities of Liverpool, Sheffield and Newcastle. One of CIMA’s objectives is to ‘Identify and share optimal techniques and approaches to monitor age-related changes in all musculoskeletal tissues, and to provide an integrated assessment of musculoskeletal function’—in other words to develop a toolkit for assessing musculoskeletal ageing. This toolkit is envisaged as an instrument that can be used to characterise and quantify musculoskeletal function during ‘normal’ ageing, lend itself to use in large-scale, internationally important cohorts, and provide a set of biomarker outcome measures for epidemiological and intervention studies designed to enhance healthy musculoskeletal ageing. Such potential biomarkers include: biochemical measurements in biofluids or tissue samples, in vivo measurements of body composition, imaging of structural and physical properties, and functional tests. This review assesses candidate biomarkers of musculoskeletal ageing under these four headings, details their biological bases, strengths and limitations, and makes practical recommendations for their use. In addition, we identify gaps in the evidence base and priorities for further research on biomarkers of musculoskeletal ageing.
Collapse
Affiliation(s)
- Graham J Kemp
- Department of Musculoskeletal Biology, Faculty of Health and Life Sciences, Institute of Ageing and Chronic Disease (IACD), University of Liverpool, William Duncan Building, 6 West Derby Street, Liverpool, UK
- The MRC-Arthritis Research UK Centre for Integrated Research into Musculoskeletal Ageing (CIMA)
| | - Fraser Birrell
- Institute of Cellular Medicine, Musculoskeletal Research Group, Newcastle University, Newcastle upon Tyne, UK
- The MRC-Arthritis Research UK Centre for Integrated Research into Musculoskeletal Ageing (CIMA)
| | - Peter D Clegg
- Department of Musculoskeletal Biology, Faculty of Health and Life Sciences, Institute of Ageing and Chronic Disease (IACD), University of Liverpool, William Duncan Building, 6 West Derby Street, Liverpool, UK
- The MRC-Arthritis Research UK Centre for Integrated Research into Musculoskeletal Ageing (CIMA)
| | - Daniel J Cuthbertson
- Department of Musculoskeletal Biology, Faculty of Health and Life Sciences, Institute of Ageing and Chronic Disease (IACD), University of Liverpool, William Duncan Building, 6 West Derby Street, Liverpool, UK
- The MRC-Arthritis Research UK Centre for Integrated Research into Musculoskeletal Ageing (CIMA)
| | - Giuseppe De Vito
- School of Public Health, Physiotherapy and Sports Science, Institute for Sport and Health, University College Dublin, Belfield, Dublin, Ireland
| | - Jaap H van Dieën
- Department of Human Movement Sciences, VU University Amsterdam, Amsterdam Movement Sciences, Van der Boechorststraat 9, Amsterdam, The Netherlands
| | - Silvia Del Din
- Clinical Ageing Research Unit, Institute of Neuroscience/Newcastle University Institute for Ageing, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, UK
| | - Richard Eastell
- Mellanby Centre for Bone Research, University of Sheffield, Sheffield, UK
- The MRC-Arthritis Research UK Centre for Integrated Research into Musculoskeletal Ageing (CIMA)
| | - Patrick Garnero
- Division of Bone Diseases, Geneva University Hospital and Faculty of Medicine, 1205 Geneva, Switzerland
| | - Katarzyna Goljanek–Whysall
- Department of Musculoskeletal Biology, Faculty of Health and Life Sciences, Institute of Ageing and Chronic Disease (IACD), University of Liverpool, William Duncan Building, 6 West Derby Street, Liverpool, UK
- The MRC-Arthritis Research UK Centre for Integrated Research into Musculoskeletal Ageing (CIMA)
| | | | - Richard Hodgson
- Centre for Imaging Sciences, University of Manchester, Stopford Building, Oxford Road, Manchester, UK
| | - Malcolm J Jackson
- Department of Musculoskeletal Biology, Faculty of Health and Life Sciences, Institute of Ageing and Chronic Disease (IACD), University of Liverpool, William Duncan Building, 6 West Derby Street, Liverpool, UK
- The MRC-Arthritis Research UK Centre for Integrated Research into Musculoskeletal Ageing (CIMA)
| | - Sue Lord
- Clinical Ageing Research Unit, Institute of Neuroscience/Newcastle University Institute for Ageing, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, UK
| | - Claudia Mazzà
- Department of Mechanical Engineering & INSIGNEO Institute for in silico Medicine, University of Sheffield, Sheffield, UK
- The MRC-Arthritis Research UK Centre for Integrated Research into Musculoskeletal Ageing (CIMA)
| | - Anne McArdle
- Department of Musculoskeletal Biology, Faculty of Health and Life Sciences, Institute of Ageing and Chronic Disease (IACD), University of Liverpool, William Duncan Building, 6 West Derby Street, Liverpool, UK
- The MRC-Arthritis Research UK Centre for Integrated Research into Musculoskeletal Ageing (CIMA)
| | - Eugene V McCloskey
- Mellanby Centre for Bone Research, University of Sheffield, Sheffield, UK
- The MRC-Arthritis Research UK Centre for Integrated Research into Musculoskeletal Ageing (CIMA)
| | - Marco Narici
- MRC-ARUK Centre of Excellence for Musculoskeletal Ageing Research, Derby Royal Hospital, Uttoxeter Road, Derby, UK
| | - Mandy J Peffers
- Department of Musculoskeletal Biology, Faculty of Health and Life Sciences, Institute of Ageing and Chronic Disease (IACD), University of Liverpool, William Duncan Building, 6 West Derby Street, Liverpool, UK
- The MRC-Arthritis Research UK Centre for Integrated Research into Musculoskeletal Ageing (CIMA)
| | - Stefano Schiaffino
- Venetian Institute of Molecular Medicine (VIMM), Via Orus 2, Padova, Italy
| | - John C Mathers
- Human Nutrition Research Centre, Institute of Cellular Medicine and Newcastle University Institute for Ageing, Newcastle University, Newcastle upon Tyne, UK
- The MRC-Arthritis Research UK Centre for Integrated Research into Musculoskeletal Ageing (CIMA)
| |
Collapse
|
31
|
Noncoding RNA Transcripts during Differentiation of Induced Pluripotent Stem Cells into Hepatocytes. Stem Cells Int 2018; 2018:5692840. [PMID: 30210551 PMCID: PMC6120260 DOI: 10.1155/2018/5692840] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 05/20/2018] [Accepted: 06/12/2018] [Indexed: 02/01/2023] Open
Abstract
Recent advances in the stem cell field allow to obtain many human tissues in vitro. However, hepatic differentiation of induced pluripotent stem cells (iPSCs) still remains challenging. Hepatocyte-like cells (HLCs) obtained after differentiation resemble more fetal liver hepatocytes. MicroRNAs (miRNA) play an important role in the differentiation process. Here, we analysed noncoding RNA profiles from the last stages of differentiation and compare them to hepatocytes. Our results show that HLCs maintain an epithelial character and express miRNA which can block hepatocyte maturation by inhibiting the epithelial-mesenchymal transition (EMT). Additionally, we identified differentially expressed small nucleolar RNAs (snoRNAs) and discovered novel noncoding RNA (ncRNA) genes.
Collapse
|
32
|
Peffers MJ, Balaskas P, Smagul A. Osteoarthritis year in review 2017: genetics and epigenetics. Osteoarthritis Cartilage 2018; 26:304-311. [PMID: 28989115 PMCID: PMC6292677 DOI: 10.1016/j.joca.2017.09.009] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 09/06/2017] [Accepted: 09/08/2017] [Indexed: 02/02/2023]
Abstract
OBJECTIVE The purpose of this review is to describe highlights from original research publications related to osteoarthritis (OA), epigenetics and genomics with the intention of recognising significant advances. DESIGN To identify relevant papers a Pubmed literature search was conducted for articles published between April 2016 and April 2017 using the search terms 'osteoarthritis' together with 'genetics', 'genomics', 'epigenetics', 'microRNA', 'lncRNA', 'DNA methylation' and 'histone modification'. RESULTS The search term OA generated almost 4000 references. Publications using the combination of descriptors OA and genetics provided the most references (82 references). However this was reduced compared to the same period in the previous year; 8.1-2.1% (expressed as a percentage of the total publications combining the terms OA and genetics). Publications combining the terms OA with genomics (29 references), epigenetics (16 references), long non-coding RNA (lncRNA) (11 references; including the identification of novel lncRNAs in OA), DNA methylation (21 references), histone modification (3 references) and microRNA (miR) (79 references) were reviewed. Potential OA therapeutics such as histone deacetylase (HDAC) inhibitors have been identified. A number of non-coding RNAs may also provide targets for future treatments. CONCLUSION There continues to be a year on year increase in publications researching miRs in OA (expressed as a percentage of the total publications), with a doubling over the last 4 years. An overview on the last year's progress within the fields of epigenetics and genomics with respect to OA will be given.
Collapse
Affiliation(s)
- M J Peffers
- Institute of Ageing and Chronic Disease, William Henry Duncan Building, 6 West Derby Street, Liverpool, L7 8TX, UK.
| | - P Balaskas
- Institute of Ageing and Chronic Disease, William Henry Duncan Building, 6 West Derby Street, Liverpool, L7 8TX, UK.
| | - A Smagul
- Institute of Ageing and Chronic Disease, William Henry Duncan Building, 6 West Derby Street, Liverpool, L7 8TX, UK.
| |
Collapse
|
33
|
Abstract
INTRODUCTION Osteoarthritis (OA), a chronic, debilitating and degenerative disease of the joints, is the most common form of arthritis. The seriousness of this prevalent and chronic disease is often overlooked. Disease modifying OA drug development is hindered by the lack of soluble biomarkers to detect OA early. The objective of OA biomarker research is to identify early OA prior to the appearance of radiographic signs and the development of pain. Areas covered: This review has focused on extracellular genomic material that could serve as biomarkers of OA. Recent studies have examined the expression of extracellular genomic material such as miRNA, lncRNA, snoRNA, mRNA and cell-free DNA, which are aberrantly expressed in the body fluids of OA patients. Changes in genomic content of peripheral blood mononuclear cells in OA could also function as biomarkers of OA. Expert commentary: There is an unmet need for soluble biomarkers for detecting and then monitoring OA disease progression. Extracellular genomic material research may also reveal more about the underlying pathophysiology of OA. Minimally-invasive liquid biopsies such as synovial fluid and blood sampling of genomic material may be more sensitive over radiography in the detection, diagnosis and monitoring of OA in the future.
Collapse
Affiliation(s)
- Emma Budd
- a The D-BOARD European Consortium for Biomarker Discovery, School of Veterinary Medicine , University of Surrey , Guildford , UK.,b Department of Veterinary Pre-Clinical Sciences, School of Veterinary Medicine, Faculty of Health and Medical Sciences , University of Surrey , Guildford , UK
| | - Giovanna Nalesso
- b Department of Veterinary Pre-Clinical Sciences, School of Veterinary Medicine, Faculty of Health and Medical Sciences , University of Surrey , Guildford , UK
| | - Ali Mobasheri
- a The D-BOARD European Consortium for Biomarker Discovery, School of Veterinary Medicine , University of Surrey , Guildford , UK.,b Department of Veterinary Pre-Clinical Sciences, School of Veterinary Medicine, Faculty of Health and Medical Sciences , University of Surrey , Guildford , UK.,c Arthritis Research UK Centre for Sport, Exercise and Osteoarthritis , Queen's Medical Centre , Nottingham , UK
| |
Collapse
|
34
|
Falaleeva M, Welden JR, Duncan MJ, Stamm S. C/D-box snoRNAs form methylating and non-methylating ribonucleoprotein complexes: Old dogs show new tricks. Bioessays 2017; 39:10.1002/bies.201600264. [PMID: 28505386 PMCID: PMC5586538 DOI: 10.1002/bies.201600264] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
C/D box snoRNAs (SNORDs) are an abundantly expressed class of short, non-coding RNAs that have been long known to perform 2'-O-methylation of rRNAs. However, approximately half of human SNORDs have no predictable rRNA targets, and numerous SNORDs have been associated with diseases that show no defects in rRNAs, among them Prader-Willi syndrome, Duplication 15q syndrome and cancer. This apparent discrepancy has been addressed by recent studies showing that SNORDs can act to regulate pre-mRNA alternative splicing, mRNA abundance, activate enzymes, and be processed into shorter ncRNAs resembling miRNAs and piRNAs. Furthermore, recent biochemical studies have shown that a given SNORD can form both methylating and non-methylating ribonucleoprotein complexes, providing an indication of the likely physical basis for such diverse new functions. Thus, SNORDs are more structurally and functionally diverse than previously thought, and their role in gene expression is under-appreciated. The action of SNORDs in non-methylating complexes can be substituted with oligonucleotides, allowing devising therapies for diseases like Prader-Willi syndrome.
Collapse
Affiliation(s)
- Marina Falaleeva
- University Kentucky, Institute for Biochemistry, Lexington, KY, USA
| | - Justin R. Welden
- University Kentucky, Institute for Biochemistry, Lexington, KY, USA
| | | | - Stefan Stamm
- University Kentucky, Institute for Biochemistry, Lexington, KY, USA
| |
Collapse
|