Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Front Cell Neurosci ; 16: 1108593, 2022.
Article in English | MEDLINE | ID: covidwho-2199041

ABSTRACT

[This corrects the article DOI: 10.3389/fncel.2022.954912.].

2.
Front Genet ; 13: 1085155, 2022.
Article in English | MEDLINE | ID: covidwho-2198792
3.
Cells ; 11(15)2022 07 25.
Article in English | MEDLINE | ID: covidwho-1993936

ABSTRACT

Skeletal muscle is a pivotal organ in humans that maintains locomotion and homeostasis. Muscle atrophy caused by sarcopenia and cachexia, which results in reduced muscle mass and impaired skeletal muscle function, is a serious health condition that decreases life longevity in humans. Recent studies have revealed the molecular mechanisms by which long non-coding RNAs (lncRNAs) regulate skeletal muscle mass and function through transcriptional regulation, fiber-type switching, and skeletal muscle cell proliferation. In addition, lncRNAs function as natural inhibitors of microRNAs and induce muscle hypertrophy or atrophy. Intriguingly, muscle atrophy modifies the expression of thousands of lncRNAs. Therefore, although their exact functions have not yet been fully elucidated, various novel lncRNAs associated with muscle atrophy have been identified. Here, we comprehensively review recent knowledge on the regulatory roles of lncRNAs in skeletal muscle atrophy. In addition, we discuss the issues and possibilities of targeting lncRNAs as a treatment for skeletal muscle atrophy and muscle wasting disorders in humans.


Subject(s)
Muscular Diseases , RNA, Long Noncoding , Humans , Muscle Development/genetics , Muscle, Skeletal/metabolism , Muscular Atrophy/genetics , Muscular Atrophy/metabolism , Muscular Diseases/genetics , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism
4.
Theranostics ; 12(8): 3946-3962, 2022.
Article in English | MEDLINE | ID: covidwho-1934549

ABSTRACT

Rationale: Viral infections are complex processes based on an intricate network of molecular interactions. The infectious agent hijacks components of the cellular machinery for its profit, circumventing the natural defense mechanisms triggered by the infected cell. The successful completion of the replicative viral cycle within a cell depends on the function of viral components versus the cellular defenses. Non-coding RNAs (ncRNAs) are important cellular modulators, either promoting or preventing the progression of viral infections. Among these ncRNAs, the long non-coding RNA (lncRNA) family is especially relevant due to their intrinsic functional properties and ubiquitous biological roles. Specific lncRNAs have been recently characterized as modulators of the cellular response during infection of human host cells by single stranded RNA viruses. However, the role of host lncRNAs in the infection by human RNA coronaviruses such as SARS-CoV-2 remains uncharacterized. Methods: In the present work, we have performed a transcriptomic study of a cohort of patients with different SARS-CoV-2 viral load and analyzed the involvement of lncRNAs in supporting regulatory networks based on their interaction with RNA-binding proteins (RBPs). Results: Our results revealed the existence of a SARS-CoV-2 infection-dependent pattern of transcriptional up-regulation in which specific lncRNAs are an integral component. To determine the role of these lncRNAs, we performed a functional correlation analysis complemented with the study of the validated interactions between lncRNAs and RBPs. This combination of in silico functional association studies and experimental evidence allowed us to identify a lncRNA signature composed of six elements - NRIR, BISPR, MIR155HG, FMR1-IT1, USP30-AS1, and U62317.2 - associated with the regulation of SARS-CoV-2 infection. Conclusions: We propose a competition mechanism between the viral RNA genome and the regulatory lncRNAs in the sequestering of specific RBPs that modulates the interferon response and the regulation of RNA surveillance by nonsense-mediated decay (NMD).


Subject(s)
COVID-19 , RNA, Long Noncoding , COVID-19/genetics , Fragile X Mental Retardation Protein , Genome, Viral , Humans , Immunity , Mitochondrial Proteins/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , RNA, Untranslated/genetics , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , SARS-CoV-2/genetics , Thiolester Hydrolases/metabolism
5.
Biochim Biophys Acta Mol Basis Dis ; 1868(2): 166291, 2022 02 01.
Article in English | MEDLINE | ID: covidwho-1525693

ABSTRACT

OBJECTIVES: To investigate in silico the presence of nucleotide sequence complementarity between the RNA genome of Severe Acute Respiratory Syndrome CoronaVirus-2 (SARS-CoV-2) and human non-coding (nc)RNA genes. METHODS: The FASTA sequence (NC_045512.2) of each of the 11 SARS-CoV-2 isolate Wuhan-Hu-1 genes was retrieved from NCBI.nlm.nih.gov/gene and the Ensembl.org library interrogated for any base-pair match with human ncRNA genes. SARS-CoV-2 gene-matched human ncRNAs were screened for functional activity using bioinformatic analysis. Finally, associations between identified ncRNAs and human diseases were searched in GWAS databases. RESULTS: A total of 252 matches were found between the nucleotide sequence of SARS-CoV-2 genes and human ncRNAs. With the exception of two small nuclear RNAs, all of them were long non-coding (lnc)RNAs expressed mainly in testis and central nervous system under physiological conditions. The percentage of alignment ranged from 91.30% to 100% with a mean nucleotide alignment length of 17.5 ± 2.4. Thirty-three (13.09%) of them contained predicted R-loop forming sequences, but none of these intersected the complementary sequences of SARS-CoV-2. However, in 31 cases matches fell on ncRNA regulatory sites, whose adjacent coding genes are mostly involved in cancer, immunological and neurological pathways. Similarly, several polymorphic variants of detected non-coding genes have been associated with neuropsychiatric and proliferative disorders. CONCLUSION: This pivotal in silico study shows that SARS-CoV-2 genes have Watson-Crick nucleotide complementarity to human ncRNA sequences, potentially disrupting ncRNA epigenetic control of target genes. It remains to be elucidated whether this could result in the development of human disease in the long term.


Subject(s)
COVID-19/genetics , COVID-19/virology , RNA, Untranslated/genetics , SARS-CoV-2/genetics , Base Sequence , Epigenesis, Genetic , Genes, Viral , Humans , Neoplasms/genetics , RNA, Long Noncoding/genetics , Sequence Alignment , Sequence Homology, Nucleic Acid
6.
Biomed J ; 44(5): 515-520, 2021 10.
Article in English | MEDLINE | ID: covidwho-1482466

ABSTRACT

In the current issue of the Biomedical Journal we gain an insight into the regeneration of liver tissue and how an over-the-counter supplement, stem cells and two plant extracts counteract liver damage. Furthermore the advances against hepatitis C virus are presented, the role of long non-coding RNA elucidated as well as the potential of an adhesion G protein-coupled receptor. In another contribution, the definition and evolutionary impact of copy number variants is clarified. Also, the polymorphism of a scaffolding caspase is investigated. We furthermore learn about the relation between SARS-CoV2 mutants in dependence of geography and explore the challenges of telemedicine in a complex healthcare field. A novel approach to engineering artificial grafts is presented, the challenges of total knee arthroplasty discussed as well as a possible mean of sinus floor elevation for dental implants. At last the concept of flipped classroom is scrutinized in terms of usefulness for a hospital in Taiwan.


Subject(s)
COVID-19 , Dental Implants , Liver Regeneration , Sinus Floor Augmentation , Humans , RNA, Viral , SARS-CoV-2
7.
PeerJ ; 9: e11169, 2021.
Article in English | MEDLINE | ID: covidwho-1227167

ABSTRACT

BACKGROUND: Kawasaki disease (KD) is an acute and febrile systemic vasculitis of unknown etiology. This study aimed to identify the competing endogenous RNA (ceRNA) networks of lncRNAs, miRNAs, and genes in KD and explore the molecular mechanisms underlying KD. METHODS: GSE68004 and GSE73464 datasets were downloaded from the Gene Expression Omnibus. Differentially expressed lncRNAs (DElncRNAs) and genes (DEGs) in KD were identified using the criteria of p < 0.05 and | log2 (fold change) | ≥ 1. MicroRNAs (miRNAs) related to KD were searched from databases. The lncRNA-miRNA-mRNA networks involving the DElncRNAs and DEGs were constructed. RESULTS: A total of 769 common upregulated, 406 common downregulated DEGs, and six DElncRNAs were identified in the KD samples. The lncRNA-miRNA-mRNA network consisted of four miRNAs, three lncRNAs (including the upregulated PSORS1C3, LINC00999, and the downregulated SNHG5) and four DEGs (including the downregulated GATA3 and the upregulated SOD2, MAPK14, and PPARG). Validation in the GSE18606 dataset showed that intravenous immunoglobulin treatment significantly alleviated the deregulated profiles of the above RNAs in KD patients. Three ceRNA networks of LINC00999-hsa-miR-6780-SOD2, PSORS1C3-hsa-miR-216a-PPARG/MAPK14, and SNHG5-hsa-miR-132/hsa-miR-92-GATA3 were identified. Four genes were associated with functional categories, such as inflammatory response and vascular endothelial cell. CONCLUSIONS: The ceRNA networks involve genes, such as SOD2, MAPK14, and PPARG, and lncRNAs, including PSORS1C3, LINC00999, and SNHG5, which might play a key role in the pathogenesis and development of KD by regulating inflammation.

8.
J Cell Mol Med ; 25(12): 5823-5827, 2021 06.
Article in English | MEDLINE | ID: covidwho-1221608

ABSTRACT

The long non-coding RNAs (lncRNAs) play a critical regulatory role in the host response to the viral infection. However, little is understood about the transcriptome architecture, especially lncRNAs pattern during the SARS-CoV-2 infection. In the present study, using publicly available RNA sequencing data of bronchoalveolar lavage fluid (BALF) and peripheral blood mononuclear cells (PBMC) samples from COVID-19 patients and healthy individuals, three interesting findings highlighted: (a) More than half of the interactions between lncRNAs-PCGs of BALF samples established by three trans-acting lncRNAs (HOTAIRM1, PVT1 and AL392172.1), which also exhibited the high affinity for binding to the SARS-CoV-2 genome, suggesting the major regulatory role of these lncRNAs during the SARS-CoV-2 infection. (b) lncRNAs of MALAT1 and NEAT1 are possibly contributed to the inflammation development in the SARS-CoV-2 infected cells. (c) In contrast to the 3' part of the SARS-CoV-2 genome, the 5' part can interact with many human lncRNAs. Therefore, the mRNA-based vaccines will not show any side effects because of the off-label interactions with the human lncRNAs. Overall, the putative functionalities of lncRNAs can be promising to design the non-coding RNA-based drugs and to inspect the efficiency of vaccines to overcome the current pandemic.


Subject(s)
COVID-19 , RNA, Long Noncoding/metabolism , RNA, Viral/metabolism , SARS-CoV-2/genetics , Bronchoalveolar Lavage Fluid/immunology , Bronchoalveolar Lavage Fluid/virology , COVID-19/immunology , COVID-19/virology , Databases, Nucleic Acid , Humans , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/virology
9.
Cell ; 184(7): 1790-1803.e17, 2021 04 01.
Article in English | MEDLINE | ID: covidwho-1139467

ABSTRACT

The long non-coding RNA (lncRNA) XIST establishes X chromosome inactivation (XCI) in female cells in early development and thereafter is thought to be largely dispensable. Here, we show XIST is continually required in adult human B cells to silence a subset of X-linked immune genes such as TLR7. XIST-dependent genes lack promoter DNA methylation and require continual XIST-dependent histone deacetylation. XIST RNA-directed proteomics and CRISPRi screen reveal distinctive somatic cell-type-specific XIST complexes and identify TRIM28 that mediates Pol II pausing at promoters of X-linked genes in B cells. Single-cell transcriptome data of female patients with either systemic lupus erythematosus or COVID-19 infection revealed XIST dysregulation, reflected by escape of XIST-dependent genes, in CD11c+ atypical memory B cells (ABCs). XIST inactivation with TLR7 agonism suffices to promote isotype-switched ABCs. These results indicate cell-type-specific diversification and function for lncRNA-protein complexes and suggest expanded roles for XIST in sex-differences in biology and medicine.


Subject(s)
B-Lymphocytes/immunology , COVID-19 , Lupus Erythematosus, Systemic , RNA, Long Noncoding/physiology , Toll-Like Receptor 7/immunology , X Chromosome Inactivation , COVID-19/genetics , COVID-19/immunology , Cell Line , DNA Methylation , Female , Gene Silencing , Humans , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/immunology
10.
Viruses ; 12(12)2020 12 01.
Article in English | MEDLINE | ID: covidwho-954243

ABSTRACT

The emergence of SARS-CoV-2 in 2019 has caused a major health and economic crisis around the globe. Gaining knowledge about its attributes and interactions with human host cells is crucial. Non-coding RNAs (ncRNAs) are involved in the host cells' innate antiviral immune response. In RNA interference, microRNAs (miRNAs) may bind to complementary sequences of the viral RNA strand, forming an miRNA-induced silencing complex, which destroys the viral RNA, thereby inhibiting viral protein expression. There are several targets for human miRNAs on SARS-CoV-2's RNA, most of which are in the 5' and 3' untranslated regions. Mutations of the viral genome causing the creation or loss of miRNA binding sites may have crucial effects on SARS-CoV-2 pathogenicity. In addition to mediating immunity, the ncRNA landscape of host cells further influences their susceptibility to virus infection, as certain miRNAs are essential in the regulation of cellular receptors that are necessary for virus invasion. Conversely, virus infection also changes the host ncRNA expression patterns, possibly augmenting conditions for viral replication and dissemination. Hence, ncRNAs typically upregulated in SARS-CoV-2 infection could be useful biomarkers for disease progression and severity. Understanding these mechanisms could provide further insight into the pathogenesis and possible treatment options against COVID-19.


Subject(s)
Coronavirus Infections/immunology , Coronavirus Infections/virology , Coronavirus/pathogenicity , RNA, Untranslated/metabolism , Animals , Antiviral Agents/metabolism , Antiviral Agents/therapeutic use , Coronavirus/genetics , Coronavirus Infections/drug therapy , Coronavirus Infections/genetics , Humans , Immune Evasion/genetics , Mutation , RNA Interference , RNA, Untranslated/genetics , RNA, Untranslated/therapeutic use , RNA, Viral/genetics
11.
Front Immunol ; 11: 590870, 2020.
Article in English | MEDLINE | ID: covidwho-902405

ABSTRACT

The COVID-19 pandemic exerts inflammation-related parasympathetic complications and post-infection manifestations with major inter-individual variability. To seek the corresponding transcriptomic origins for the impact of COVID-19 infection and its aftermath consequences, we sought the relevance of long and short non-coding RNAs (ncRNAs) for susceptibility to COVID-19 infection. We selected inflammation-prone men and women of diverse ages among the cohort of Genome Tissue expression (GTEx) by mining RNA-seq datasets from their lung, and blood tissues, followed by quantitative qRT-PCR, bioinformatics-based network analyses and thorough statistics compared to brain cell culture and infection tests with COVID-19 and H1N1 viruses. In lung tissues from 57 inflammation-prone, but not other GTEx donors, we discovered sharp declines of the lung pathology-associated ncRNA DANCR and the nuclear paraspeckles forming neuroprotective ncRNA NEAT1. Accompanying increases in the acetylcholine-regulating transcripts capable of controlling inflammation co-appeared in SARS-CoV-2 infected but not H1N1 influenza infected lung cells. The lung cells-characteristic DANCR and NEAT1 association with inflammation-controlling transcripts could not be observed in blood cells, weakened with age and presented sex-dependent links in GTEx lung RNA-seq dataset. Supporting active involvement in the inflammatory risks accompanying COVID-19, DANCR's decline associated with decrease of the COVID-19-related cellular transcript ACE2 and with sex-related increases in coding transcripts potentiating acetylcholine signaling. Furthermore, transcription factors (TFs) in lung, brain and cultured infected cells created networks with the candidate transcripts, indicating tissue-specific expression patterns. Supporting links of post-infection inflammatory and cognitive damages with cholinergic mal-functioning, man and woman-originated cultured cholinergic neurons presented differentiation-related increases of DANCR and NEAT1 targeting microRNAs. Briefly, changes in ncRNAs and TFs from inflammation-prone human lung tissues, SARS-CoV-2-infected lung cells and man and woman-derived differentiated cholinergic neurons reflected the inflammatory pathobiology related to COVID-19. By shifting ncRNA differences into comparative diagnostic and therapeutic profiles, our RNA-sequencing based Resource can identify ncRNA regulating candidates for COVID-19 and its associated immediate and predicted long-term inflammation and neurological complications, and sex-related therapeutics thereof. Our findings encourage diagnostics of involved tissue, and further investigation of NEAT1-inducing statins and anti-cholinergic medications in the COVID-19 context.


Subject(s)
COVID-19/genetics , Cholinergic Neurons/metabolism , MicroRNAs/genetics , RNA, Long Noncoding/genetics , SARS-CoV-2 , Transcriptome , A549 Cells , Acetylcholine/metabolism , Brain/metabolism , Brain/pathology , COVID-19/virology , Databases, Genetic , Female , Humans , Inflammation/genetics , Influenza A Virus, H1N1 Subtype , Influenza, Human/genetics , Influenza, Human/virology , Lung/metabolism , Lung/pathology , Male , RNA-Seq , Transcription Factors/genetics
12.
Brief Bioinform ; 22(3)2021 05 20.
Article in English | MEDLINE | ID: covidwho-787100

ABSTRACT

Recent advances in transcriptomics have uncovered lots of novel transcripts in plants. To annotate such transcripts, dissecting their coding potential is a critical step. Computational approaches have been proven fruitful in this task; however, most current tools are designed/optimized for mammals and only a few of them have been tested on a limited number of plant species. In this work, we present NAMS webserver, which contains a novel coding potential classifier, NAMS, specifically optimized for plants. We have evaluated the performance of NAMS using a comprehensive dataset containing more than 3 million transcripts from various plant species, where NAMS demonstrates high accuracy and remarkable performance improvements over state-of-the-art software. Moreover, our webserver also furnishes functional annotations, aiming to provide users informative clues to the functions of their transcripts. Considering that most plant species are poorly characterized, our NAMS webserver could serve as a valuable resource to facilitate the transcriptomic studies. The webserver with testing dataset is freely available at http://sunlab.cpy.cuhk.edu.hk/NAMS/.


Subject(s)
Computational Biology/methods , Gene Expression Profiling/methods , Gene Expression Regulation, Plant , Internet , Molecular Sequence Annotation/methods , Plants/genetics , Genetic Code/genetics , Plants/classification , RNA, Messenger/genetics , RNA, Plant/genetics , Reproducibility of Results , Species Specificity , Support Vector Machine
SELECTION OF CITATIONS
SEARCH DETAIL