1
|
Luo M, Xie X, Wang D, Li X, Ling X, Yuan W, Cong J, Huang H, Zhang Y, Kong Z, Wan B. Senecavirus A nonstructural protein 3A Inhibits Ago2-mediated RNAi antiviral response. Vet Microbiol 2025; 306:110547. [PMID: 40359781 DOI: 10.1016/j.vetmic.2025.110547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 04/30/2025] [Accepted: 05/04/2025] [Indexed: 05/15/2025]
Abstract
Senecavirus A (SVA) can infect pigs of all ages and poses a significant risk to the swine industry. RNA interference (RNAi) is a conserved antiviral immune defense pathway in eukaryotes, and many viruses have been proven to encode viral proteins as viral RNAi suppressors (VSRs) to evade RNAi-mediated antiviral response. However, it is unclear whether SVA regulates RNAi for its infection and replication. In this study, it is found that SVA nonstructural protein 3 A has a VSR function. Mechanistically, 3 A effectively inhibited double-stranded RNA and small interfering RNA-induced RNAi, degrading Argonaute2 (Ago2) through autophagy. This paper demonstrated that SVA encoded 3 A as VSR to resist RNAi pathway, escaping host immunity and promoting viral replication, which expands the understanding of the interaction between SVA and its host.
Collapse
Affiliation(s)
- Mengru Luo
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Henan Agricultural University, Zhengzhou, China
| | - Xingyi Xie
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Henan Agricultural University, Zhengzhou, China
| | - Dengkun Wang
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Henan Agricultural University, Zhengzhou, China
| | - Xuechen Li
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Henan Agricultural University, Zhengzhou, China
| | - Xianghui Ling
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Henan Agricultural University, Zhengzhou, China
| | - Wenju Yuan
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Henan Agricultural University, Zhengzhou, China
| | - Jiahao Cong
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Henan Agricultural University, Zhengzhou, China
| | - Huihui Huang
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Henan Agricultural University, Zhengzhou, China
| | - Yuhang Zhang
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Henan Agricultural University, Zhengzhou, China.
| | - Zhengjie Kong
- School of Advanced Agricultural Sciences, Peking University, Beijing, China.
| | - Bo Wan
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Henan Agricultural University, Zhengzhou, China.
| |
Collapse
|
2
|
Modafferi S, Esposito F, Tavella S, Gioia U, Francia S. Traffic light at DSB-transit regulation between gene transcription and DNA repair. FEBS Lett 2025; 599:177-189. [PMID: 39333024 PMCID: PMC11771567 DOI: 10.1002/1873-3468.15024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 08/12/2024] [Accepted: 08/15/2024] [Indexed: 09/29/2024]
Abstract
Transcription of actively expressed genes is dampened for kilobases around DNA lesions via chromatin modifications. This is believed to favour repair and prevent genome instability. Nonetheless, mounting evidence suggests that transcription may be induced by DNA breakage, resulting in the local de novo synthesis of non-coding RNAs (ncRNAs). Such transcripts have been proposed to play important functions in both DNA damage signalling and repair. Here, we review the recently identified mechanistic details of transcriptional silencing at damaged chromatin, highlighting how post-translational histone modifications can also be modulated by the local synthesis of DNA damage-induced ncRNAs. Finally, we envision that these entangled transcriptional events at DNA breakages can be targeted to modulate DNA repair, with potential implications for locus-specific therapeutic strategies.
Collapse
Affiliation(s)
- Stefania Modafferi
- Istituto di Genetica Molecolare “Luigi Luca Cavalli Sforza”– Consiglio Nazionale delle RicerchePaviaItaly
- PhD Program in Biomolecular Sciences and Biotechnology (SBB)Istituto Universitario di Studi Superiori (IUSS)PaviaItaly
| | - Francesca Esposito
- Istituto di Genetica Molecolare “Luigi Luca Cavalli Sforza”– Consiglio Nazionale delle RicerchePaviaItaly
- PhD Program in Genetics, Molecular and Cellular Biology (GMCB)University of PaviaPaviaItaly
| | - Sara Tavella
- Istituto di Genetica Molecolare “Luigi Luca Cavalli Sforza”– Consiglio Nazionale delle RicerchePaviaItaly
- IFOM‐ETS – The AIRC Institute of Molecular OncologyMilanItaly
| | - Ubaldo Gioia
- Istituto di Genetica Molecolare “Luigi Luca Cavalli Sforza”– Consiglio Nazionale delle RicerchePaviaItaly
- IFOM‐ETS – The AIRC Institute of Molecular OncologyMilanItaly
| | - Sofia Francia
- Istituto di Genetica Molecolare “Luigi Luca Cavalli Sforza”– Consiglio Nazionale delle RicerchePaviaItaly
- IFOM‐ETS – The AIRC Institute of Molecular OncologyMilanItaly
| |
Collapse
|
3
|
Kong J, Bie Y, Ji W, Xu J, Lyu B, Xiong X, Qiu Y, Zhou X. Alphavirus infection triggers antiviral RNAi immunity in mammals. Cell Rep 2023; 42:112441. [PMID: 37104090 DOI: 10.1016/j.celrep.2023.112441] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 03/29/2023] [Accepted: 04/11/2023] [Indexed: 04/28/2023] Open
Abstract
RNA interference (RNAi) is a well-established antiviral immunity. However, for mammalian somatic cells, antiviral RNAi becomes evident only when viral suppressors of RNAi (VSRs) are disabled by mutations or VSR-targeting drugs, thereby limiting its scope as a mammalian immunity. We find that a wild-type alphavirus, Semliki Forest virus (SFV), triggers the Dicer-dependent production of virus-derived small interfering RNAs (vsiRNAs) in both mammalian somatic cells and adult mice. These SFV-vsiRNAs are located at a particular region within the 5' terminus of the SFV genome, Argonaute loaded, and active in conferring effective anti-SFV activity. Sindbis virus, another alphavirus, also induces vsiRNA production in mammalian somatic cells. Moreover, treatment with enoxacin, an RNAi enhancer, inhibits SFV replication dependent on RNAi response in vitro and in vivo and protects mice from SFV-induced neuropathogenesis and lethality. These findings show that alphaviruses trigger the production of active vsiRNA in mammalian somatic cells, highlighting the functional importance and therapeutic potential of antiviral RNAi in mammals.
Collapse
Affiliation(s)
- Jing Kong
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuanyuan Bie
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wenting Ji
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China; School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Jiuyue Xu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Bao Lyu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaobei Xiong
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Yang Qiu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Xi Zhou
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China; University of Chinese Academy of Sciences, Beijing 100049, China; School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China.
| |
Collapse
|
4
|
Välikangas T, Junttila S, Rytkönen KT, Kukkonen-Macchi A, Suomi T, Elo LL. COVID-19-specific transcriptomic signature detectable in blood across multiple cohorts. Front Genet 2022; 13:929887. [PMID: 35991542 PMCID: PMC9388772 DOI: 10.3389/fgene.2022.929887] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 06/27/2022] [Indexed: 01/08/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is spreading across the world despite vast global vaccination efforts. Consequently, many studies have looked for potential human host factors and immune mechanisms associated with the disease. However, most studies have focused on comparing COVID-19 patients to healthy controls, while fewer have elucidated the specific host factors distinguishing COVID-19 from other infections. To discover genes specifically related to COVID-19, we reanalyzed transcriptome data from nine independent cohort studies, covering multiple infections, including COVID-19, influenza, seasonal coronaviruses, and bacterial pneumonia. The identified COVID-19-specific signature consisted of 149 genes, involving many signals previously associated with the disease, such as induction of a strong immunoglobulin response and hemostasis, as well as dysregulation of cell cycle-related processes. Additionally, potential new gene candidates related to COVID-19 were discovered. To facilitate exploration of the signature with respect to disease severity, disease progression, and different cell types, we also offer an online tool for easy visualization of the selected genes across multiple datasets at both bulk and single-cell levels.
Collapse
Affiliation(s)
- Tommi Välikangas
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Sini Junttila
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Kalle T. Rytkönen
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Anu Kukkonen-Macchi
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Tomi Suomi
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Laura L. Elo
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
- Institute of Biomedicine, University of Turku, Turku, Finland
| |
Collapse
|
5
|
Van de Vyver T, De Smedt SC, Raemdonck K. Modulating intracellular pathways to improve non-viral delivery of RNA therapeutics. Adv Drug Deliv Rev 2022; 181:114041. [PMID: 34763002 DOI: 10.1016/j.addr.2021.114041] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 10/12/2021] [Accepted: 11/02/2021] [Indexed: 12/12/2022]
Abstract
RNA therapeutics (e.g. siRNA, oligonucleotides, mRNA, etc.) show great potential for the treatment of a myriad of diseases. However, to reach their site of action in the cytosol or nucleus of target cells, multiple intra- and extracellular barriers have to be surmounted. Several non-viral delivery systems, such as nanoparticles and conjugates, have been successfully developed to meet this requirement. Unfortunately, despite these clear advances, state-of-the-art delivery agents still suffer from relatively low intracellular delivery efficiencies. Notably, our current understanding of the intracellular delivery process is largely oversimplified. Gaining mechanistic insight into how RNA formulations are processed by cells will fuel rational design of the next generation of delivery carriers. In addition, identifying which intracellular pathways contribute to productive RNA delivery could provide opportunities to boost the delivery performance of existing nanoformulations. In this review, we discuss both established as well as emerging techniques that can be used to assess the impact of different intracellular barriers on RNA transfection performance. Next, we highlight how several modulators, including small molecules but also genetic perturbation technologies, can boost RNA delivery by intervening at differing stages of the intracellular delivery process, such as cellular uptake, intracellular trafficking, endosomal escape, autophagy and exocytosis.
Collapse
Affiliation(s)
- Thijs Van de Vyver
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium.
| | - Stefaan C De Smedt
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium.
| | - Koen Raemdonck
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium.
| |
Collapse
|
6
|
Ahmadi A, Moradi S. In silico analysis suggests the RNAi-enhancing antibiotic enoxacin as a potential inhibitor of SARS-CoV-2 infection. Sci Rep 2021; 11:10271. [PMID: 33986351 PMCID: PMC8119475 DOI: 10.1038/s41598-021-89605-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 04/29/2021] [Indexed: 02/06/2023] Open
Abstract
COVID-19 has currently become the biggest challenge in the world. There is still no specific medicine for COVID-19, which leaves a critical gap for the identification of new drug candidates for the disease. Recent studies have reported that the small-molecule enoxacin exerts an antiviral activity by enhancing the RNAi pathway. The aim of this study is to analyze if enoxacin can exert anti-SARS-CoV-2 effects. We exploit multiple computational tools and databases to examine (i) whether the RNAi mechanism, as the target pathway of enoxacin, could act on the SARS-CoV-2 genome, and (ii) microRNAs induced by enoxacin might directly silence viral components as well as the host cell proteins mediating the viral entry and replication. We find that the RNA genome of SARS-CoV-2 might be a suitable substrate for DICER activity. We also highlight several enoxacin-enhanced microRNAs which could target SARS-CoV-2 components, pro-inflammatory cytokines, host cell components facilitating viral replication, and transcription factors enriched in lung stem cells, thereby promoting their differentiation and lung regeneration. Finally, our analyses identify several enoxacin-targeted regulatory modules that were critically associated with exacerbation of the SARS-CoV-2 infection. Overall, our analysis suggests that enoxacin could be a promising candidate for COVID-19 treatment through enhancing the RNAi pathway.
Collapse
Affiliation(s)
- Amirhossein Ahmadi
- Department of Biological Science and Technology, Faculty of Nano and Bio Science and Technology, Persian Gulf University, Bushehr, 75169,, Iran
| | - Sharif Moradi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
7
|
Zika Virus Pathogenesis: A Battle for Immune Evasion. Vaccines (Basel) 2021; 9:vaccines9030294. [PMID: 33810028 PMCID: PMC8005041 DOI: 10.3390/vaccines9030294] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 03/12/2021] [Accepted: 03/13/2021] [Indexed: 12/13/2022] Open
Abstract
Zika virus (ZIKV) infection and its associated congenital and other neurological disorders, particularly microcephaly and other fetal developmental abnormalities, constitute a World Health Organization (WHO) Zika Virus Research Agenda within the WHO’s R&D Blueprint for Action to Prevent Epidemics, and continue to be a Public Health Emergency of International Concern (PHEIC) today. ZIKV pathogenicity is initiated by viral infection and propagation across multiple placental and fetal tissue barriers, and is critically strengthened by subverting host immunity. ZIKV immune evasion involves viral non-structural proteins, genomic and non-coding RNA and microRNA (miRNA) to modulate interferon (IFN) signaling and production, interfering with intracellular signal pathways and autophagy, and promoting cellular environment changes together with secretion of cellular components to escape innate and adaptive immunity and further infect privileged immune organs/tissues such as the placenta and eyes. This review includes a description of recent advances in the understanding of the mechanisms underlying ZIKV immune modulation and evasion that strongly condition viral pathogenesis, which would certainly contribute to the development of anti-ZIKV strategies, drugs, and vaccines.
Collapse
|
8
|
Raue R, Frank AC, Syed SN, Brüne B. Therapeutic Targeting of MicroRNAs in the Tumor Microenvironment. Int J Mol Sci 2021; 22:ijms22042210. [PMID: 33672261 PMCID: PMC7926641 DOI: 10.3390/ijms22042210] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 02/16/2021] [Accepted: 02/18/2021] [Indexed: 02/06/2023] Open
Abstract
The tumor-microenvironment (TME) is an amalgamation of various factors derived from malignant cells and infiltrating host cells, including cells of the immune system. One of the important factors of the TME is microRNAs (miRs) that regulate target gene expression at a post transcriptional level. MiRs have been found to be dysregulated in tumor as well as in stromal cells and they emerged as important regulators of tumorigenesis. In fact, miRs regulate almost all hallmarks of cancer, thus making them attractive tools and targets for novel anti-tumoral treatment strategies. Tumor to stroma cell cross-propagation of miRs to regulate protumoral functions has been a salient feature of the TME. MiRs can either act as tumor suppressors or oncogenes (oncomiRs) and both miR mimics as well as miR inhibitors (antimiRs) have been used in preclinical trials to alter cancer and stromal cell phenotypes. Owing to their cascading ability to regulate upstream target genes and their chemical nature, which allows specific pharmacological targeting, miRs are attractive targets for anti-tumor therapy. In this review, we cover a recent update on our understanding of dysregulated miRs in the TME and provide an overview of how these miRs are involved in current cancer-therapeutic approaches from bench to bedside.
Collapse
Affiliation(s)
- Rebecca Raue
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany; (R.R.); (A.-C.F.)
| | - Ann-Christin Frank
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany; (R.R.); (A.-C.F.)
| | - Shahzad Nawaz Syed
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany; (R.R.); (A.-C.F.)
- Correspondence: (S.N.S.); (B.B.); Tel.: +49-69-6301-7424 (B.B.)
| | - Bernhard Brüne
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany; (R.R.); (A.-C.F.)
- Project Group Translational Medicine and Pharmacology TMP, Fraunhofer Institute for Molecular Biology and Applied Ecology, 60596 Frankfurt, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt, 60590 Frankfurt, Germany
- Frankfurt Cancer Institute, Goethe-University Frankfurt, 60596 Frankfurt, Germany
- Correspondence: (S.N.S.); (B.B.); Tel.: +49-69-6301-7424 (B.B.)
| |
Collapse
|
9
|
Dalhoff A. Selective toxicity of antibacterial agents-still a valid concept or do we miss chances and ignore risks? Infection 2021; 49:29-56. [PMID: 33367978 PMCID: PMC7851017 DOI: 10.1007/s15010-020-01536-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 10/04/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Selective toxicity antibacteribiotics is considered to be due to interactions with targets either being unique to bacteria or being characterized by a dichotomy between pro- and eukaryotic pathways with high affinities of agents to bacterial- rather than eukaryotic targets. However, the theory of selective toxicity oversimplifies the complex modes of action of antibiotics in pro- and eukaryotes. METHODS AND OBJECTIVE This review summarizes data describing multiple modes of action of antibiotics in eukaryotes. RESULTS Aminoglycosides, macrolides, oxazolidinones, chloramphenicol, clindamycin, tetracyclines, glycylcyclines, fluoroquinolones, rifampicin, bedaquillin, ß-lactams inhibited mitochondrial translation either due to binding to mitosomes, inhibition of mitochondrial RNA-polymerase-, topoisomerase 2ß-, ATP-synthesis, transporter activities. Oxazolidinones, tetracyclines, vancomycin, ß-lactams, bacitracin, isoniazid, nitroxoline inhibited matrix-metalloproteinases (MMP) due to chelation with zinc and calcium, whereas fluoroquinols fluoroquinolones and chloramphenicol chelated with these cations, too, but increased MMP activities. MMP-inhibition supported clinical efficacies of ß-lactams and daptomycin in skin-infections, and of macrolides, tetracyclines in respiratory-diseases. Chelation may have contributed to neuroprotection by ß-lactams and fluoroquinolones. Aminoglycosides, macrolides, chloramphenicol, oxazolidins oxazolidinones, tetracyclines caused read-through of premature stop codons. Several additional targets for antibiotics in human cells have been identified like interaction of fluoroquinolones with DNA damage repair in eukaryotes, or inhibition of mucin overproduction by oxazolidinones. CONCLUSION The effects of antibiotics on eukaryotes are due to identical mechanisms as their antibacterial activities because of structural and functional homologies of pro- and eukaryotic targets, so that the effects of antibiotics on mammals are integral parts of their overall mechanisms of action.
Collapse
Affiliation(s)
- Axel Dalhoff
- Christian-Albrechts-University of Kiel, Institue for Infection Medicine, Brunswiker Str. 4, D-24105, Kiel, Germany.
| |
Collapse
|
10
|
Scroggs SLP, Gass JT, Chinnasamy R, Widen SG, Azar SR, Rossi SL, Arterburn JB, Vasilakis N, Hanley KA. Evolution of resistance to fluoroquinolones by dengue virus serotype 4 provides insight into mechanism of action and consequences for viral fitness. Virology 2021; 552:94-106. [PMID: 33120225 PMCID: PMC7528753 DOI: 10.1016/j.virol.2020.09.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 08/30/2020] [Accepted: 09/08/2020] [Indexed: 02/07/2023]
Abstract
Drugs against flaviviruses such as dengue (DENV) and Zika (ZIKV) virus are urgently needed. We previously demonstrated that three fluoroquinolones, ciprofloxacin, enoxacin, and difloxacin, suppress replication of six flaviviruses. To investigate the barrier to resistance and mechanism(s) of action of these drugs, DENV-4 was passaged in triplicate in HEK-293 cells in the presence or absence of each drug. Resistance to ciprofloxacin was detected by the seventh passage and to difloxacin by the tenth, whereas resistance to enoxacin did not occur within ten passages. Two putative resistance-conferring mutations were detected in the envelope gene of ciprofloxacin and difloxacin-resistant DENV-4. In the absence of ciprofloxacin, ciprofloxacin-resistant viruses sustained a significantly higher viral titer than control viruses in HEK-293 and HuH-7 cells and resistant viruses were more stable than control viruses at 37 °C. These results suggest that the mechanism of action of ciprofloxacin and difloxacin involves interference with virus binding or entry.
Collapse
Affiliation(s)
- Stacey L P Scroggs
- Department of Biology, New Mexico State University, Las Cruces, NM, USA.
| | - Jordan T Gass
- Department of Biology, New Mexico State University, Las Cruces, NM, USA
| | - Ramesh Chinnasamy
- Department of Chemistry and Biochemistry, New Mexico State University, Las Cruces, NM, USA
| | - Steven G Widen
- Department of Biochemistry & Molecular Biology, The University of Texas Medical Branch, Galveston, TX, USA
| | - Sasha R Azar
- Department of Pathology, The University of University of Texas Medical Branch, Galveston, TX, USA
| | - Shannan L Rossi
- Department of Pathology, The University of University of Texas Medical Branch, Galveston, TX, USA; Institute for Human Infection and Immunity, The University of University of Texas Medical Branch, Galveston, TX, USA
| | - Jeffrey B Arterburn
- Department of Chemistry and Biochemistry, New Mexico State University, Las Cruces, NM, USA
| | - Nikos Vasilakis
- Department of Pathology, The University of University of Texas Medical Branch, Galveston, TX, USA; Center for Biodefense and Emerging Infectious Diseases, The University of University of Texas Medical Branch, Galveston, TX, USA; Center for Tropical Diseases, The University of University of Texas Medical Branch, Galveston, TX, USA; Institute for Human Infection and Immunity, The University of University of Texas Medical Branch, Galveston, TX, USA
| | - Kathryn A Hanley
- Department of Biology, New Mexico State University, Las Cruces, NM, USA
| |
Collapse
|
11
|
Rocha AL, de Lima TI, de Souza GP, Corrêa RO, Ferrucci DL, Rodrigues B, Lopes-Ramos C, Nilsson D, Knittel TL, Castro PR, Fernandes MF, Dos Santos Martins F, Parmigiani RB, Silveira LR, Carvalho HF, Auwerx J, Vinolo MAR, Boucher J, Mori MA. Enoxacin induces oxidative metabolism and mitigates obesity by regulating adipose tissue miRNA expression. SCIENCE ADVANCES 2020; 6:eabc6250. [PMID: 33268375 PMCID: PMC7710362 DOI: 10.1126/sciadv.abc6250] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 10/14/2020] [Indexed: 05/03/2023]
Abstract
MicroRNAs (miRNAs) have been implicated in oxidative metabolism and brown/beige adipocyte identity. Here, we tested whether widespread changes in miRNA expression promoted by treatment with the small-molecule enoxacin cause browning and prevent obesity. Enoxacin mitigated diet-induced obesity in mice, and this was associated with increased energy expenditure. Consistently, subcutaneous white and brown adipose tissues and skeletal muscle of enoxacin-treated mice had higher levels of markers associated with thermogenesis and oxidative metabolism. These effects were cell autonomous since they were recapitulated in vitro in murine and human cell models. In preadipocytes, enoxacin led to a reduction of miR-34a-5p expression and up-regulation of its target genes (e.g., Fgfr1, Klb, and Sirt1), thus increasing FGF21 signaling and promoting beige adipogenesis. Our data demonstrate that enoxacin counteracts obesity by promoting thermogenic signaling and inducing oxidative metabolism in adipose tissue and skeletal muscle in a mechanism that involves, at least in part, miRNA-mediated regulation.
Collapse
Affiliation(s)
- Andréa Livia Rocha
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, Brazil
- Program in Genetics and Molecular Biology, Institute of Biology, University of Campinas, Campinas, Brazil
- Department of Biophysics, São Paulo School of Medicine, Federal University of São Paulo, São Paulo, Brazil
- Program in Biotechnology, Federal University of São Paulo, São Paulo, Brazil
| | - Tanes Imamura de Lima
- Obesity and Comorbidities Research Center (OCRC), University of Campinas, Campinas, Brazil
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, Campinas, Brazil
- Laboratory of Integrative Systems Physiology, Interfaculty Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Gerson Profeta de Souza
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, Brazil
- Program in Genetics and Molecular Biology, Institute of Biology, University of Campinas, Campinas, Brazil
- Obesity and Comorbidities Research Center (OCRC), University of Campinas, Campinas, Brazil
| | - Renan Oliveira Corrêa
- Program in Genetics and Molecular Biology, Institute of Biology, University of Campinas, Campinas, Brazil
- Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Danilo Lopes Ferrucci
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, Brazil
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, Campinas, Brazil
- National Institute of Science and Technology in Photonics Applied to Cell Biology (INFABiC), University of Campinas, Campinas, Brazil
| | - Bruno Rodrigues
- Department of Adapted Physical Activity, School of Physical Education, University of Campinas, Campinas, Brazil
| | - Camila Lopes-Ramos
- Center of Molecular Oncology, Sírio-Libanês Hospital, São Paulo, Brazil
- Department of Biostatistics, Harvard TH Chan School of Public Health, Boston, USA
| | - Daniel Nilsson
- Wallenberg Center for Molecular and Translational Medicine, University of Gothenburg, Sweden
- The Lundberg Laboratory for Diabetes Research, University of Gothenburg, Sweden
| | - Thiago Leite Knittel
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, Brazil
- Program in Genetics and Molecular Biology, Institute of Biology, University of Campinas, Campinas, Brazil
- Obesity and Comorbidities Research Center (OCRC), University of Campinas, Campinas, Brazil
| | - Pollyana Ribeiro Castro
- Program in Genetics and Molecular Biology, Institute of Biology, University of Campinas, Campinas, Brazil
- Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Mariane Font Fernandes
- Program in Genetics and Molecular Biology, Institute of Biology, University of Campinas, Campinas, Brazil
- Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Flaviano Dos Santos Martins
- Laboratory of Biotherapeutics Agents, Department of Microbiology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | | | - Leonardo Reis Silveira
- Obesity and Comorbidities Research Center (OCRC), University of Campinas, Campinas, Brazil
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Hernandes F Carvalho
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, Campinas, Brazil
- National Institute of Science and Technology in Photonics Applied to Cell Biology (INFABiC), University of Campinas, Campinas, Brazil
| | - Johan Auwerx
- Laboratory of Integrative Systems Physiology, Interfaculty Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Marco Aurélio R Vinolo
- Program in Genetics and Molecular Biology, Institute of Biology, University of Campinas, Campinas, Brazil
- Obesity and Comorbidities Research Center (OCRC), University of Campinas, Campinas, Brazil
- Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas, Brazil
- Experimental Medicine Research Cluster (EMRC), Campinas, Brazil
| | - Jeremie Boucher
- Wallenberg Center for Molecular and Translational Medicine, University of Gothenburg, Sweden
- The Lundberg Laboratory for Diabetes Research, University of Gothenburg, Sweden
- Bioscience Metabolism, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Marcelo A Mori
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, Brazil.
- Program in Genetics and Molecular Biology, Institute of Biology, University of Campinas, Campinas, Brazil
- Department of Biophysics, São Paulo School of Medicine, Federal University of São Paulo, São Paulo, Brazil
- Program in Biotechnology, Federal University of São Paulo, São Paulo, Brazil
- Obesity and Comorbidities Research Center (OCRC), University of Campinas, Campinas, Brazil
- Experimental Medicine Research Cluster (EMRC), Campinas, Brazil
| |
Collapse
|
12
|
Scroggs SLP, Andrade CC, Chinnasamy R, Azar SR, Schirtzinger EE, Garcia EI, Arterburn JB, Hanley KA, Rossi SL. Old Drugs with New Tricks: Efficacy of Fluoroquinolones to Suppress Replication of Flaviviruses. Viruses 2020; 12:v12091022. [PMID: 32933138 PMCID: PMC7551155 DOI: 10.3390/v12091022] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 09/10/2020] [Accepted: 09/11/2020] [Indexed: 02/06/2023] Open
Abstract
Repurposing FDA-approved compounds could provide the fastest route to alleviate the burden of disease caused by flaviviruses. In this study, three fluoroquinolones, enoxacin, difloxacin and ciprofloxacin, curtailed replication of flaviviruses Zika (ZIKV), dengue (DENV), Langat (LGTV) and Modoc (MODV) in HEK-293 cells at low micromolar concentrations. Time-of-addition assays suggested that enoxacin suppressed ZIKV replication at an intermediate step in the virus life cycle, whereas ciprofloxacin and difloxacin had a wider window of efficacy. A129 mice infected with 1 × 105 plaque-forming units (pfu) ZIKV FSS13025 (n = 20) or phosphate buffered saline (PBS) (n = 11) on day 0 and treated with enoxacin at 10 mg/kg or 15 mg/kg or diluent orally twice daily on days 1–5 did not differ in weight change or virus titer in serum or brain. However, mice treated with enoxacin showed a significant, five-fold decrease in ZIKV titer in testes relative to controls. Mice infected with 1 × 102 pfu ZIKV (n = 13) or PBS (n = 13) on day 0 and treated with 15 mg/kg oral enoxacin or diluent twice daily pre-treatment and days 1–5 post-treatment also did not differ in weight and viral load in the serum, brain, and liver, but mice treated with enoxacin showed a significant, 2.5-fold decrease in ZIKV titer in testes relative to controls. ZIKV can be sexually transmitted, so reduction of titer in the testes by enoxacin should be further investigated.
Collapse
Affiliation(s)
- Stacey L. P. Scroggs
- Department of Biology, New Mexico State University, Las Cruces, NM 88003, USA; (C.C.A.); (E.E.S.); (E.I.G.); (K.A.H.)
- Biology of Vector-Borne Viruses Section, Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA
- Correspondence:
| | - Christy C. Andrade
- Department of Biology, New Mexico State University, Las Cruces, NM 88003, USA; (C.C.A.); (E.E.S.); (E.I.G.); (K.A.H.)
- Department of Biology, Gonzaga University, Spokane, WA 99258, USA
| | - Ramesh Chinnasamy
- Department of Chemistry and Biochemistry, New Mexico State University, Las Cruces, NM 88003, USA; (R.C.); (J.B.A.)
| | - Sasha R. Azar
- Institute for Translational Sciences, The University of University of Texas Medical Branch, Galveston, TX 77555, USA;
| | - Erin E. Schirtzinger
- Department of Biology, New Mexico State University, Las Cruces, NM 88003, USA; (C.C.A.); (E.E.S.); (E.I.G.); (K.A.H.)
- Arthropod-borne Animal Diseases Research Unit, United States Department of Agriculture, Agricultural Research Service, Manhattan, KS 66506, USA
| | - Erin I. Garcia
- Department of Biology, New Mexico State University, Las Cruces, NM 88003, USA; (C.C.A.); (E.E.S.); (E.I.G.); (K.A.H.)
- Science News, Washington, DC 20036, USA
| | - Jeffrey B. Arterburn
- Department of Chemistry and Biochemistry, New Mexico State University, Las Cruces, NM 88003, USA; (R.C.); (J.B.A.)
| | - Kathryn A. Hanley
- Department of Biology, New Mexico State University, Las Cruces, NM 88003, USA; (C.C.A.); (E.E.S.); (E.I.G.); (K.A.H.)
| | - Shannan L. Rossi
- Department of Pathology, The University of Texas Medical Branch, Galveston, TX 77555, USA;
| |
Collapse
|
13
|
Huang X, Chen Z, Liu Y. RNAi-mediated control of CRISPR functions. Theranostics 2020; 10:6661-6673. [PMID: 32550896 PMCID: PMC7295050 DOI: 10.7150/thno.44880] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 04/30/2020] [Indexed: 12/20/2022] Open
Abstract
CRISPR-Cas9 has become a versatile tool for genome editing and regulation, and strategies to effectively control its activity have attracted much attention. RNAi, also a gene-regulating tool, is used as another mechanism by which eukaryotes resist the invasion of foreign genetic material. Methods: In this study, we analyzed the quantitative inhibition of the CRISPR system by using artificial miRNAs (amiRNAs) combined with the RNAi enhancer enoxacin to improve the targeting specificity of the CRISPR system. Furthermore, we examined the feasibility of improving the efficiency of gene editing and regulation by blocking the effects of natural intracellular miRNAs on sgRNAs. Results: amiRNAs targeting the sgRNA were used to control its expression, and the small molecule drug denoxacin was utilized to enhance this effect, especially in the presence of Cas9. amiRNA/enoxacin inhibited CRISPR-mediated gene editing and regulation both in vitro and in vivo and could tune sgRNA-targeting specificity. Furthermore, CRISPR efficiency was increased by blocking the effects of endogenous miRNAs. Conclusion: Our study provides an efficient molecular switch for conditional regulation of CRISPR activities in mammalian cells and also presents potentially useful approaches for solving current key issues of off-target effects and low targeting efficiency.
Collapse
Affiliation(s)
- Xinbo Huang
- National and Local Joint Engineering Laboratory of Medical Synthetic Biology, Shenzhen, Second People's Hospital, The First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen518035, China
- Department of Urology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen518035, China
- Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University Health, Science Center, Shenzhen 518035, China
| | - Zhicong Chen
- National and Local Joint Engineering Laboratory of Medical Synthetic Biology, Shenzhen, Second People's Hospital, The First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen518035, China
- Department of Urology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen518035, China
- Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University Health, Science Center, Shenzhen 518035, China
| | - Yuchen Liu
- National and Local Joint Engineering Laboratory of Medical Synthetic Biology, Shenzhen, Second People's Hospital, The First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen518035, China
- Department of Urology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen518035, China
- Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University Health, Science Center, Shenzhen 518035, China
| |
Collapse
|
14
|
Xu YP, Qiu Y, Zhang B, Chen G, Chen Q, Wang M, Mo F, Xu J, Wu J, Zhang RR, Cheng ML, Zhang NN, Lyu B, Zhu WL, Wu MH, Ye Q, Zhang D, Man JH, Li XF, Cui J, Xu Z, Hu B, Zhou X, Qin CF. Zika virus infection induces RNAi-mediated antiviral immunity in human neural progenitors and brain organoids. Cell Res 2019; 29:265-273. [PMID: 30814679 PMCID: PMC6461993 DOI: 10.1038/s41422-019-0152-9] [Citation(s) in RCA: 123] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 01/29/2019] [Indexed: 12/27/2022] Open
Abstract
The re-emergence of Zika virus (ZIKV) in the Western Hemisphere has resulted in global public health crisis since 2015. ZIKV preferentially infects and targets human neural progenitor cells (hNPCs) and causes fetal microcephaly upon maternal infection. hNPCs not only play critical roles during fetal brain development, but also persist in adult brain throughout life. Yet the mechanism of innate antiviral immunity in hNPCs remains largely unknown. Here, we show that ZIKV infection triggers the abundant production of virus-derived small interfering RNAs in hNPCs, but not in the more differentiated progenies or somatic cells. Ablation of key RNAi machinery components significantly enhances ZIKV replication in hNPCs. Furthermore, enoxacin, a broad-spectrum antibiotic that is known as an RNAi enhancer, exerts potent anti-ZIKV activity in hNPCs and other RNAi-competent cells. Strikingly, enoxacin treatment completely prevents ZIKV infection and circumvents ZIKV-induced microcephalic phenotypes in brain organoid models that recapitulate human fetal brain development. Our findings highlight the physiological importance of RNAi-mediated antiviral immunity during the early stage of human brain development, uncovering a novel strategy to combat human congenital viral infections through enhancing RNAi.
Collapse
Affiliation(s)
- Yan-Peng Xu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, 100071, Beijing, China
| | - Yang Qiu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences (CAS), Wuhan, 430071, Hubei, China
| | - Boya Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, CAS, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Guilai Chen
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, CAS, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Qi Chen
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, 100071, Beijing, China
| | - Miao Wang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences (CAS), Wuhan, 430071, Hubei, China
| | - Fan Mo
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, CAS, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Jiuyue Xu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences (CAS), Wuhan, 430071, Hubei, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Jin Wu
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, 100850, Beijing, China
| | - Rong-Rong Zhang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, 100071, Beijing, China
- School of Basic Sciences, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Meng-Li Cheng
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, 100071, Beijing, China
- School of Basic Sciences, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Na-Na Zhang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, 100071, Beijing, China
| | - Bao Lyu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences (CAS), Wuhan, 430071, Hubei, China
- College of Life Sciences, Wuhan University, Wuhan, 430072, Hubei, China
| | - Wen-Liang Zhu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, CAS, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Meng-Hua Wu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, CAS, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Qing Ye
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, 100071, Beijing, China
| | - Da Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, CAS, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Jiang-Hong Man
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, 100850, Beijing, China
| | - Xiao-Feng Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, 100071, Beijing, China
| | - Jie Cui
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences (CAS), Wuhan, 430071, Hubei, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Zhiheng Xu
- University of Chinese Academy of Sciences, 100049, Beijing, China
- State Key Laboratory of Molecular Developmental Biology, Center for Excellence in Brain Science and Intelligence Technology, Institute of Genetics and Developmental Biology, CAS, 100101, Beijing, China
| | - Baoyang Hu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, CAS, 100101, Beijing, China.
- University of Chinese Academy of Sciences, 100049, Beijing, China.
| | - Xi Zhou
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences (CAS), Wuhan, 430071, Hubei, China.
- University of Chinese Academy of Sciences, 100049, Beijing, China.
- College of Life Sciences, Wuhan University, Wuhan, 430072, Hubei, China.
| | - Cheng-Feng Qin
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, 100071, Beijing, China.
- School of Basic Sciences, Anhui Medical University, Hefei, 230032, Anhui, China.
| |
Collapse
|
15
|
Enoxacin and bis-enoxacin stimulate 4T1 murine breast cancer cells to release extracellular vesicles that inhibit osteoclastogenesis. Sci Rep 2018; 8:16182. [PMID: 30385810 PMCID: PMC6212457 DOI: 10.1038/s41598-018-34698-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 10/23/2018] [Indexed: 12/21/2022] Open
Abstract
Enoxacin and its bone-seeking bisphosphonate derivative, bis-enoxacin, have recently captured attention as potential therapeutic agents for the treatment of cancer and bone disease. No differences in growth or survival of 4T1 murine breast cancer cells were detected at a concentration of 50 µM of enoxacin or bis-enoxacin. Growth was perturbed at higher concentrations. Both 50 µM enoxacin and bis-enoxacin stimulated increases in the number of GW/Processing bodies, but there were minimal changes in microRNA levels. Extracellular vesicles (EVs) released from 4T1 cells treated with 50 µM enoxacin or 50 µM bis-enoxacin stimulated proliferation of RAW 264.7 cells, and both significantly inhibited osteoclastogenesis in calcitriol-stimulated mouse marrow. EVs from 4T1 cells treated with enoxacin and bis-enoxacin displayed small reductions in the amount of microRNA (miR)-146a-5p and let-7b-5p. In marked contrast, miR-214-3p, which has been shown to regulate bone remodeling, was increased 22-fold and 30-fold respectively. We conclude that enoxacin and bis-enoxacin trigger the release of EVs from 4T1 cancer cells that inhibit osteoclastogenesis.
Collapse
|
16
|
Pinto S, Sato VN, De-Souza EA, Ferraz RC, Camara H, Pinca APF, Mazzotti DR, Lovci MT, Tonon G, Lopes-Ramos CM, Parmigiani RB, Wurtele M, Massirer KB, Mori MA. Enoxacin extends lifespan of C. elegans by inhibiting miR-34-5p and promoting mitohormesis. Redox Biol 2018; 18:84-92. [PMID: 29986212 PMCID: PMC6037660 DOI: 10.1016/j.redox.2018.06.006] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 06/17/2018] [Indexed: 11/29/2022] Open
Abstract
Alterations in microRNA (miRNA) processing have been previously linked to aging. Here we used the small molecule enoxacin to pharmacologically interfere with miRNA biogenesis and study how it affects aging in C. elegans. Enoxacin extended worm lifespan and promoted survival under normal and oxidative stress conditions. Enoxacin-induced longevity required the transcription factor SKN-1/Nrf2 and was blunted by the antioxidant N-acetyl-cysteine, suggesting a prooxidant-mediated mitohormetic response. The longevity effects of enoxacin were also dependent on the miRNA pathway, consistent with changes in miRNA expression elicited by the drug. Among these differentially expressed miRNAs, the widely conserved miR-34-5p was found to play an important role in enoxacin-mediated longevity. Enoxacin treatment down-regulated miR-34-5p and did not further extend lifespan of long-lived mir-34 mutants. Moreover, N-acetyl-cysteine abrogated mir-34(gk437)-induced longevity. Evidence also points to double-stranded RNA-specific adenosine deaminases (ADARs) as new targets of enoxacin since ADAR loss-of-function abrogates enoxacin-induced lifespan extension. Thus, enoxacin increases lifespan by reducing miR-34-5p levels, interfering with the redox balance and promoting healthspan.
Collapse
Affiliation(s)
- Silas Pinto
- Department of Biophysics, Program in Molecular Biology, São Paulo School of Medicine, Federal University of São Paulo (UNIFESP), São Paulo, SP, Brazil; Laboratory of Aging Biology (LaBE), Department of Biochemistry and Tissue Biology, Program in Genetics and Molecular Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Vitor N Sato
- Department of Biophysics, Program in Molecular Biology, São Paulo School of Medicine, Federal University of São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Evandro A De-Souza
- Department of Biophysics, Program in Molecular Biology, São Paulo School of Medicine, Federal University of São Paulo (UNIFESP), São Paulo, SP, Brazil; Laboratory of Aging Biology (LaBE), Department of Biochemistry and Tissue Biology, Program in Genetics and Molecular Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Rafael C Ferraz
- Department of Biophysics, Program in Molecular Biology, São Paulo School of Medicine, Federal University of São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Henrique Camara
- Department of Biophysics, Program in Molecular Biology, São Paulo School of Medicine, Federal University of São Paulo (UNIFESP), São Paulo, SP, Brazil; Laboratory of Aging Biology (LaBE), Department of Biochemistry and Tissue Biology, Program in Genetics and Molecular Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Ana Paula F Pinca
- Department of Biophysics, Program in Molecular Biology, São Paulo School of Medicine, Federal University of São Paulo (UNIFESP), São Paulo, SP, Brazil; Laboratory of Aging Biology (LaBE), Department of Biochemistry and Tissue Biology, Program in Genetics and Molecular Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Diego R Mazzotti
- Department of Psychobiology. São Paulo School of Medicine. Federal University of São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Michael T Lovci
- Center for Molecular Biology and Genetic Engineering (CBMEG), University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Guilherme Tonon
- Laboratory of Aging Biology (LaBE), Department of Biochemistry and Tissue Biology, Program in Genetics and Molecular Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | | | | | - Martin Wurtele
- Institute of Science and Technology, Federal University of Sao Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Katlin B Massirer
- Center for Molecular Biology and Genetic Engineering (CBMEG), University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Marcelo A Mori
- Department of Biophysics, Program in Molecular Biology, São Paulo School of Medicine, Federal University of São Paulo (UNIFESP), São Paulo, SP, Brazil; Laboratory of Aging Biology (LaBE), Department of Biochemistry and Tissue Biology, Program in Genetics and Molecular Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil.
| |
Collapse
|
17
|
Holliday LS. Vacuolar H +-ATPases (V-ATPases) as therapeutic targets: a brief review and recent developments. ACTA ACUST UNITED AC 2017; 1. [PMID: 30957075 DOI: 10.21037/biotarget.2017.12.01] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Vacuolar H+-ATPases (V-ATPases) are multi-subunit enzymes that play housekeeping roles in eukaryotic cells by acidifying lysosomes, late endosomes, Golgi, and other membrane-bounded compartments. Beyond that, V-ATPases have specialized functions in certain cell types linked to diseases including osteoporosis and cancer. Efforts to identify strategies to develop inhibitors selective for V-ATPases that are involved in disease progression have been ongoing for more than two decades, but so far have not yielded a therapeutic agent that has been translated to the clinic. Recent basic science studies have identified unexpected roles for V-ATPases in nutrient and energy sensing, and renin/angiotensin signaling, which offer additional incentives for considering V-ATPases as therapeutic targets. This article briefly reviews efforts to utilize inhibitors of V-ATPases as drugs. Primary focus is on recent "rational" efforts to identify small molecule inhibitors of the V-ATPases that are selectively expressed in osteoclasts and cancer cells. Enoxacin and bis-enoxacin are two molecules that emerged from these efforts. These molecules block a binding interaction between V-ATPases and microfilaments that occurs in osteoclasts, but not most other cell types, which relates to the specialized function of V-ATPases in bone resorption. Enoxacin and bis-enoxacin have proven useful in the treatment of bone diseases and cancer in animal models and display therapeutic effects that are different, and perhaps better, than current drugs. These results provide evidence that agents targeting subsets of V-ATPases may prove useful in the clinic.
Collapse
Affiliation(s)
- L Shannon Holliday
- Departments of Orthodontics and Anatomy & Cell Biology, University of Florida College of Dentistry, Gainesville, Florida, USA
| |
Collapse
|
18
|
Abell NS, Mercado M, Cañeque T, Rodriguez R, Xhemalce B. Click Quantitative Mass Spectrometry Identifies PIWIL3 as a Mechanistic Target of RNA Interference Activator Enoxacin in Cancer Cells. J Am Chem Soc 2017; 139:1400-1403. [PMID: 28094937 DOI: 10.1021/jacs.6b11751] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Enoxacin is a small molecule that stimulates RNA interference (RNAi) and acts as a growth inhibitor selectively in cancer but not in untransformed cells. Here, we used alkenox, a clickable enoxacin surrogate, coupled with quantitative mass spectrometry, to identify PIWIL3 as a mechanistic target of enoxacin. PIWIL3 is an Argonaute protein of the PIWI subfamily that is mainly expressed in the germline and that mediates RNAi through piRNAs. Our results suggest that cancer cells re-express PIWIL3 to repress RNAi through miRNAs and thus open a new opportunity for cancer-specific targeting.
Collapse
Affiliation(s)
- Nathan S Abell
- Department of Molecular Biosciences, University of Texas at Austin , 2500 Speedway, Austin, Texas 78712, United States
| | - Marvin Mercado
- Department of Molecular Biosciences, University of Texas at Austin , 2500 Speedway, Austin, Texas 78712, United States
| | - Tatiana Cañeque
- UPR2301, Institut de Chimie des Substances Naturelles , 1 avenue de la Terrasse, 91198 Gif-sur-Yvette Cedex, France.,Organic Synthesis and Cell Biology Group, Institut Curie, PSL Research University , 26 rue d'Ulm, 75248 Paris Cedex 05, France.,CNRS UMR3666 , 75005 Paris, France.,INSERM U1143 , 75005 Paris, France
| | - Raphaël Rodriguez
- UPR2301, Institut de Chimie des Substances Naturelles , 1 avenue de la Terrasse, 91198 Gif-sur-Yvette Cedex, France.,Organic Synthesis and Cell Biology Group, Institut Curie, PSL Research University , 26 rue d'Ulm, 75248 Paris Cedex 05, France.,CNRS UMR3666 , 75005 Paris, France.,INSERM U1143 , 75005 Paris, France
| | - Blerta Xhemalce
- Department of Molecular Biosciences, University of Texas at Austin , 2500 Speedway, Austin, Texas 78712, United States
| |
Collapse
|
19
|
miRNA-based therapies: strategies and delivery platforms for oligonucleotide and non-oligonucleotide agents. Future Med Chem 2015; 6:1967-84. [PMID: 25495987 DOI: 10.4155/fmc.14.116] [Citation(s) in RCA: 218] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The discovery of miRNAs as important regulatory agents for gene expression has expanded the therapeutic opportunities for oligonucleotides. In contrast to siRNA, miRNA-targeted therapy is able to influence not only a single gene, but entire cellular pathways or processes. It is possible to supplement downregulated or non-functional miRNAs by synthetic oligonucleotides, as well as alleviating effects caused by overexpression of malignant miRNAs through artificial antagonists, either oligonucleotides or small molecules. Chemical oligonucleotide modifications together with an efficient delivery system seem to be mandatory for successful therapeutic application. While miRNA-based therapy benefits from the decades of research spent on other therapeutic oligonucleotides, there are some specific challenges associated with miRNA therapy, mainly caused by the short target sequence. The current status and recent progress of miRNA-targeted therapeutics is described and future challenges and potential applications in treatment of cancer and viral infections are discussed.
Collapse
|
20
|
Qiu L, Tan EK, Zeng L. microRNAs and Neurodegenerative Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 888:85-105. [PMID: 26663180 DOI: 10.1007/978-3-319-22671-2_6] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
microRNAs (miRNAs) are small, noncoding RNA molecules that through imperfect base-pairing with complementary sequences of target mRNA molecules, typically cleave target mRNA, causing subsequent degradation or translation inhibition. Although an increasing number of studies have identified misregulated miRNAs in the neurodegenerative diseases (NDDs) Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis, which suggests that alterations in the miRNA regulatory pathway could contribute to disease pathogenesis, the molecular mechanisms underlying the pathological implications of misregulated miRNA expression and the regulation of the key genes involved in NDDs remain largely unknown. In this chapter, we provide evidence of the function and regulation of miRNAs and their association with the neurological events in NDDs. This will help improve our understanding of how miRNAs govern the biological functions of key pathogenic genes in these diseases, which potentially regulate several pathways involved in the progression of neurodegeneration. Additionally, given the growing interest in the therapeutic potential of miRNAs, we discuss current clinical challenges to developing miRNA-based therapeutics for NDDs.
Collapse
Affiliation(s)
- Lifeng Qiu
- Neural Stem Cell Research Lab, Department of Research, National Neuroscience Institute, Singapore, 308433, Singapore
| | - Eng King Tan
- Department of Neurology, National Neuroscience Institute, SGH Campus, Singapore, 169856, Singapore
- Department of Research, National Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore, 308433, Singapore
- Neuroscience and Behavioral Disorders program, Duke-National University of Singapore, Graduate Medical School, Singapore, 169857, Singapore
| | - Li Zeng
- Neural Stem Cell Research Lab, Department of Research, National Neuroscience Institute, Singapore, 308433, Singapore.
- Department of Research, National Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore, 308433, Singapore.
- Neuroscience and Behavioral Disorders program, Duke-National University of Singapore, Graduate Medical School, Singapore, 169857, Singapore.
| |
Collapse
|
21
|
Li J, Tan S, Kooger R, Zhang C, Zhang Y. MicroRNAs as novel biological targets for detection and regulation. Chem Soc Rev 2014; 43:506-17. [PMID: 24161958 DOI: 10.1039/c3cs60312a] [Citation(s) in RCA: 211] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
MicroRNAs are being considered as a novel type of bio-markers and potential therapeutic targets for various diseases. Diverse chemical tools are being developed for the detection or regulation of microRNAs with bio-medical implications. Chemical probes have been developed for use in combination with in situ signal amplification strategies to realize sensitive detection of microRNAs of low abundance. Regulation of microRNAs aberrantly expressed in tumours represents a new approach to cancer chemotherapy. Synthetic oligonucleotides including antisense oligonucleotides and microRNA mimics have been successfully delivered into cells or tissues to inhibit or enhance the function of specific endogenous microRNAs. Small-molecule modifiers of microRNAs that modify the expression or function of endogenous microRNAs are emerging not only as useful probes to explore microRNA-involved regulatory networks, but also as potential therapeutic reagents. In this tutorial review, we discuss the strategies developed by chemists in recent years for microRNA detection and regulation, with a focus on the potential of these chemical tools in microRNA-related biomedical applications.
Collapse
Affiliation(s)
- Jinbo Li
- School of Chemistry and Chemical Engineering, State Key Lab of Analytical Chemistry for Life Science, Nanjing University, Nanjing, 210093, P. R. China.
| | | | | | | | | |
Collapse
|
22
|
Abstract
The therapeutic importance of microRNA (miRNA) regulation has recently been realized as these small, noncoding RNAs have been demonstrated to be involved with a plethora of diseases and disorders. Due to the complex miRNA maturation process, the expression of these important biomolecules can be manipulated at various stages of the pathway. This review examines both in vivo and in vitro mechanisms and assays that have been developed to regulate miRNA levels. Modulation of miRNA maturation can be accomplished via several therapeutic agents, including small molecules and oligonucleotides, in both specific and nonspecific fashions. Due to the relevance of miRNAs, these novel therapeutic approaches represent new tools for the treatment of various cancers and other deleterious disorders.
Collapse
Affiliation(s)
- Valerie T Tripp
- Integrated Science Center, College of William and Mary, Williamsburg, VA, USA
| | | | | |
Collapse
|
23
|
Börner K, Niopek D, Cotugno G, Kaldenbach M, Pankert T, Willemsen J, Zhang X, Schürmann N, Mockenhaupt S, Serva A, Hiet MS, Wiedtke E, Castoldi M, Starkuviene V, Erfle H, Gilbert DF, Bartenschlager R, Boutros M, Binder M, Streetz K, Kräusslich HG, Grimm D. Robust RNAi enhancement via human Argonaute-2 overexpression from plasmids, viral vectors and cell lines. Nucleic Acids Res 2013; 41:e199. [PMID: 24049077 PMCID: PMC3834839 DOI: 10.1093/nar/gkt836] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Revised: 08/21/2013] [Accepted: 08/25/2013] [Indexed: 12/31/2022] Open
Abstract
As the only mammalian Argonaute protein capable of directly cleaving mRNAs in a small RNA-guided manner, Argonaute-2 (Ago2) is a keyplayer in RNA interference (RNAi) silencing via small interfering (si) or short hairpin (sh) RNAs. It is also a rate-limiting factor whose saturation by si/shRNAs limits RNAi efficiency and causes numerous adverse side effects. Here, we report a set of versatile tools and widely applicable strategies for transient or stable Ago2 co-expression, which overcome these concerns. Specifically, we engineered plasmids and viral vectors to co-encode a codon-optimized human Ago2 cDNA along with custom shRNAs. Furthermore, we stably integrated this Ago2 cDNA into a panel of standard human cell lines via plasmid transfection or lentiviral transduction. Using various endo- or exogenous targets, we demonstrate the potential of all three strategies to boost mRNA silencing efficiencies in cell culture by up to 10-fold, and to facilitate combinatorial knockdowns. Importantly, these robust improvements were reflected by augmented RNAi phenotypes and accompanied by reduced off-targeting effects. We moreover show that Ago2/shRNA-co-encoding vectors can enhance and prolong transgene silencing in livers of adult mice, while concurrently alleviating hepatotoxicity. Our customizable reagents and avenues should broadly improve future in vitro and in vivo RNAi experiments in mammalian systems.
Collapse
Affiliation(s)
- Kathleen Börner
- Department of Infectious Diseases, Virology, Heidelberg University Hospital, Im Neuenheimer Feld 324, D-69120 Heidelberg, Germany, Cluster of Excellence CellNetworks, Heidelberg University, Im Neuenheimer Feld 267, D-69120 Heidelberg, Germany, Department of Medicine III, University Hospital Aachen, Pauwelstrasse 30, D-52074 Aachen, Germany, Department of Infectious Diseases, Molecular Virology, Heidelberg University, Im Neuenheimer Feld 345, D-69120 Heidelberg, Germany, Division Signaling and Functional Genomics, German Cancer Research Center, Im Neuenheimer Feld 580, D-69120 Heidelberg, Germany, BioQuant, Heidelberg University, Im Neuenheimer Feld 267, D-69120 Heidelberg, Germany and Department of Pediatric Oncology, Hematology, Immunology and Pulmonology, Heidelberg University Hospital, Im Neuenheimer Feld 350, D-69120 Heidelberg, Germany
| | - Dominik Niopek
- Department of Infectious Diseases, Virology, Heidelberg University Hospital, Im Neuenheimer Feld 324, D-69120 Heidelberg, Germany, Cluster of Excellence CellNetworks, Heidelberg University, Im Neuenheimer Feld 267, D-69120 Heidelberg, Germany, Department of Medicine III, University Hospital Aachen, Pauwelstrasse 30, D-52074 Aachen, Germany, Department of Infectious Diseases, Molecular Virology, Heidelberg University, Im Neuenheimer Feld 345, D-69120 Heidelberg, Germany, Division Signaling and Functional Genomics, German Cancer Research Center, Im Neuenheimer Feld 580, D-69120 Heidelberg, Germany, BioQuant, Heidelberg University, Im Neuenheimer Feld 267, D-69120 Heidelberg, Germany and Department of Pediatric Oncology, Hematology, Immunology and Pulmonology, Heidelberg University Hospital, Im Neuenheimer Feld 350, D-69120 Heidelberg, Germany
| | - Gabriella Cotugno
- Department of Infectious Diseases, Virology, Heidelberg University Hospital, Im Neuenheimer Feld 324, D-69120 Heidelberg, Germany, Cluster of Excellence CellNetworks, Heidelberg University, Im Neuenheimer Feld 267, D-69120 Heidelberg, Germany, Department of Medicine III, University Hospital Aachen, Pauwelstrasse 30, D-52074 Aachen, Germany, Department of Infectious Diseases, Molecular Virology, Heidelberg University, Im Neuenheimer Feld 345, D-69120 Heidelberg, Germany, Division Signaling and Functional Genomics, German Cancer Research Center, Im Neuenheimer Feld 580, D-69120 Heidelberg, Germany, BioQuant, Heidelberg University, Im Neuenheimer Feld 267, D-69120 Heidelberg, Germany and Department of Pediatric Oncology, Hematology, Immunology and Pulmonology, Heidelberg University Hospital, Im Neuenheimer Feld 350, D-69120 Heidelberg, Germany
| | - Michaela Kaldenbach
- Department of Infectious Diseases, Virology, Heidelberg University Hospital, Im Neuenheimer Feld 324, D-69120 Heidelberg, Germany, Cluster of Excellence CellNetworks, Heidelberg University, Im Neuenheimer Feld 267, D-69120 Heidelberg, Germany, Department of Medicine III, University Hospital Aachen, Pauwelstrasse 30, D-52074 Aachen, Germany, Department of Infectious Diseases, Molecular Virology, Heidelberg University, Im Neuenheimer Feld 345, D-69120 Heidelberg, Germany, Division Signaling and Functional Genomics, German Cancer Research Center, Im Neuenheimer Feld 580, D-69120 Heidelberg, Germany, BioQuant, Heidelberg University, Im Neuenheimer Feld 267, D-69120 Heidelberg, Germany and Department of Pediatric Oncology, Hematology, Immunology and Pulmonology, Heidelberg University Hospital, Im Neuenheimer Feld 350, D-69120 Heidelberg, Germany
| | - Teresa Pankert
- Department of Infectious Diseases, Virology, Heidelberg University Hospital, Im Neuenheimer Feld 324, D-69120 Heidelberg, Germany, Cluster of Excellence CellNetworks, Heidelberg University, Im Neuenheimer Feld 267, D-69120 Heidelberg, Germany, Department of Medicine III, University Hospital Aachen, Pauwelstrasse 30, D-52074 Aachen, Germany, Department of Infectious Diseases, Molecular Virology, Heidelberg University, Im Neuenheimer Feld 345, D-69120 Heidelberg, Germany, Division Signaling and Functional Genomics, German Cancer Research Center, Im Neuenheimer Feld 580, D-69120 Heidelberg, Germany, BioQuant, Heidelberg University, Im Neuenheimer Feld 267, D-69120 Heidelberg, Germany and Department of Pediatric Oncology, Hematology, Immunology and Pulmonology, Heidelberg University Hospital, Im Neuenheimer Feld 350, D-69120 Heidelberg, Germany
| | - Joschka Willemsen
- Department of Infectious Diseases, Virology, Heidelberg University Hospital, Im Neuenheimer Feld 324, D-69120 Heidelberg, Germany, Cluster of Excellence CellNetworks, Heidelberg University, Im Neuenheimer Feld 267, D-69120 Heidelberg, Germany, Department of Medicine III, University Hospital Aachen, Pauwelstrasse 30, D-52074 Aachen, Germany, Department of Infectious Diseases, Molecular Virology, Heidelberg University, Im Neuenheimer Feld 345, D-69120 Heidelberg, Germany, Division Signaling and Functional Genomics, German Cancer Research Center, Im Neuenheimer Feld 580, D-69120 Heidelberg, Germany, BioQuant, Heidelberg University, Im Neuenheimer Feld 267, D-69120 Heidelberg, Germany and Department of Pediatric Oncology, Hematology, Immunology and Pulmonology, Heidelberg University Hospital, Im Neuenheimer Feld 350, D-69120 Heidelberg, Germany
| | - Xian Zhang
- Department of Infectious Diseases, Virology, Heidelberg University Hospital, Im Neuenheimer Feld 324, D-69120 Heidelberg, Germany, Cluster of Excellence CellNetworks, Heidelberg University, Im Neuenheimer Feld 267, D-69120 Heidelberg, Germany, Department of Medicine III, University Hospital Aachen, Pauwelstrasse 30, D-52074 Aachen, Germany, Department of Infectious Diseases, Molecular Virology, Heidelberg University, Im Neuenheimer Feld 345, D-69120 Heidelberg, Germany, Division Signaling and Functional Genomics, German Cancer Research Center, Im Neuenheimer Feld 580, D-69120 Heidelberg, Germany, BioQuant, Heidelberg University, Im Neuenheimer Feld 267, D-69120 Heidelberg, Germany and Department of Pediatric Oncology, Hematology, Immunology and Pulmonology, Heidelberg University Hospital, Im Neuenheimer Feld 350, D-69120 Heidelberg, Germany
| | - Nina Schürmann
- Department of Infectious Diseases, Virology, Heidelberg University Hospital, Im Neuenheimer Feld 324, D-69120 Heidelberg, Germany, Cluster of Excellence CellNetworks, Heidelberg University, Im Neuenheimer Feld 267, D-69120 Heidelberg, Germany, Department of Medicine III, University Hospital Aachen, Pauwelstrasse 30, D-52074 Aachen, Germany, Department of Infectious Diseases, Molecular Virology, Heidelberg University, Im Neuenheimer Feld 345, D-69120 Heidelberg, Germany, Division Signaling and Functional Genomics, German Cancer Research Center, Im Neuenheimer Feld 580, D-69120 Heidelberg, Germany, BioQuant, Heidelberg University, Im Neuenheimer Feld 267, D-69120 Heidelberg, Germany and Department of Pediatric Oncology, Hematology, Immunology and Pulmonology, Heidelberg University Hospital, Im Neuenheimer Feld 350, D-69120 Heidelberg, Germany
| | - Stefan Mockenhaupt
- Department of Infectious Diseases, Virology, Heidelberg University Hospital, Im Neuenheimer Feld 324, D-69120 Heidelberg, Germany, Cluster of Excellence CellNetworks, Heidelberg University, Im Neuenheimer Feld 267, D-69120 Heidelberg, Germany, Department of Medicine III, University Hospital Aachen, Pauwelstrasse 30, D-52074 Aachen, Germany, Department of Infectious Diseases, Molecular Virology, Heidelberg University, Im Neuenheimer Feld 345, D-69120 Heidelberg, Germany, Division Signaling and Functional Genomics, German Cancer Research Center, Im Neuenheimer Feld 580, D-69120 Heidelberg, Germany, BioQuant, Heidelberg University, Im Neuenheimer Feld 267, D-69120 Heidelberg, Germany and Department of Pediatric Oncology, Hematology, Immunology and Pulmonology, Heidelberg University Hospital, Im Neuenheimer Feld 350, D-69120 Heidelberg, Germany
| | - Andrius Serva
- Department of Infectious Diseases, Virology, Heidelberg University Hospital, Im Neuenheimer Feld 324, D-69120 Heidelberg, Germany, Cluster of Excellence CellNetworks, Heidelberg University, Im Neuenheimer Feld 267, D-69120 Heidelberg, Germany, Department of Medicine III, University Hospital Aachen, Pauwelstrasse 30, D-52074 Aachen, Germany, Department of Infectious Diseases, Molecular Virology, Heidelberg University, Im Neuenheimer Feld 345, D-69120 Heidelberg, Germany, Division Signaling and Functional Genomics, German Cancer Research Center, Im Neuenheimer Feld 580, D-69120 Heidelberg, Germany, BioQuant, Heidelberg University, Im Neuenheimer Feld 267, D-69120 Heidelberg, Germany and Department of Pediatric Oncology, Hematology, Immunology and Pulmonology, Heidelberg University Hospital, Im Neuenheimer Feld 350, D-69120 Heidelberg, Germany
| | - Marie-Sophie Hiet
- Department of Infectious Diseases, Virology, Heidelberg University Hospital, Im Neuenheimer Feld 324, D-69120 Heidelberg, Germany, Cluster of Excellence CellNetworks, Heidelberg University, Im Neuenheimer Feld 267, D-69120 Heidelberg, Germany, Department of Medicine III, University Hospital Aachen, Pauwelstrasse 30, D-52074 Aachen, Germany, Department of Infectious Diseases, Molecular Virology, Heidelberg University, Im Neuenheimer Feld 345, D-69120 Heidelberg, Germany, Division Signaling and Functional Genomics, German Cancer Research Center, Im Neuenheimer Feld 580, D-69120 Heidelberg, Germany, BioQuant, Heidelberg University, Im Neuenheimer Feld 267, D-69120 Heidelberg, Germany and Department of Pediatric Oncology, Hematology, Immunology and Pulmonology, Heidelberg University Hospital, Im Neuenheimer Feld 350, D-69120 Heidelberg, Germany
| | - Ellen Wiedtke
- Department of Infectious Diseases, Virology, Heidelberg University Hospital, Im Neuenheimer Feld 324, D-69120 Heidelberg, Germany, Cluster of Excellence CellNetworks, Heidelberg University, Im Neuenheimer Feld 267, D-69120 Heidelberg, Germany, Department of Medicine III, University Hospital Aachen, Pauwelstrasse 30, D-52074 Aachen, Germany, Department of Infectious Diseases, Molecular Virology, Heidelberg University, Im Neuenheimer Feld 345, D-69120 Heidelberg, Germany, Division Signaling and Functional Genomics, German Cancer Research Center, Im Neuenheimer Feld 580, D-69120 Heidelberg, Germany, BioQuant, Heidelberg University, Im Neuenheimer Feld 267, D-69120 Heidelberg, Germany and Department of Pediatric Oncology, Hematology, Immunology and Pulmonology, Heidelberg University Hospital, Im Neuenheimer Feld 350, D-69120 Heidelberg, Germany
| | - Mirco Castoldi
- Department of Infectious Diseases, Virology, Heidelberg University Hospital, Im Neuenheimer Feld 324, D-69120 Heidelberg, Germany, Cluster of Excellence CellNetworks, Heidelberg University, Im Neuenheimer Feld 267, D-69120 Heidelberg, Germany, Department of Medicine III, University Hospital Aachen, Pauwelstrasse 30, D-52074 Aachen, Germany, Department of Infectious Diseases, Molecular Virology, Heidelberg University, Im Neuenheimer Feld 345, D-69120 Heidelberg, Germany, Division Signaling and Functional Genomics, German Cancer Research Center, Im Neuenheimer Feld 580, D-69120 Heidelberg, Germany, BioQuant, Heidelberg University, Im Neuenheimer Feld 267, D-69120 Heidelberg, Germany and Department of Pediatric Oncology, Hematology, Immunology and Pulmonology, Heidelberg University Hospital, Im Neuenheimer Feld 350, D-69120 Heidelberg, Germany
| | - Vytaute Starkuviene
- Department of Infectious Diseases, Virology, Heidelberg University Hospital, Im Neuenheimer Feld 324, D-69120 Heidelberg, Germany, Cluster of Excellence CellNetworks, Heidelberg University, Im Neuenheimer Feld 267, D-69120 Heidelberg, Germany, Department of Medicine III, University Hospital Aachen, Pauwelstrasse 30, D-52074 Aachen, Germany, Department of Infectious Diseases, Molecular Virology, Heidelberg University, Im Neuenheimer Feld 345, D-69120 Heidelberg, Germany, Division Signaling and Functional Genomics, German Cancer Research Center, Im Neuenheimer Feld 580, D-69120 Heidelberg, Germany, BioQuant, Heidelberg University, Im Neuenheimer Feld 267, D-69120 Heidelberg, Germany and Department of Pediatric Oncology, Hematology, Immunology and Pulmonology, Heidelberg University Hospital, Im Neuenheimer Feld 350, D-69120 Heidelberg, Germany
| | - Holger Erfle
- Department of Infectious Diseases, Virology, Heidelberg University Hospital, Im Neuenheimer Feld 324, D-69120 Heidelberg, Germany, Cluster of Excellence CellNetworks, Heidelberg University, Im Neuenheimer Feld 267, D-69120 Heidelberg, Germany, Department of Medicine III, University Hospital Aachen, Pauwelstrasse 30, D-52074 Aachen, Germany, Department of Infectious Diseases, Molecular Virology, Heidelberg University, Im Neuenheimer Feld 345, D-69120 Heidelberg, Germany, Division Signaling and Functional Genomics, German Cancer Research Center, Im Neuenheimer Feld 580, D-69120 Heidelberg, Germany, BioQuant, Heidelberg University, Im Neuenheimer Feld 267, D-69120 Heidelberg, Germany and Department of Pediatric Oncology, Hematology, Immunology and Pulmonology, Heidelberg University Hospital, Im Neuenheimer Feld 350, D-69120 Heidelberg, Germany
| | - Daniel F. Gilbert
- Department of Infectious Diseases, Virology, Heidelberg University Hospital, Im Neuenheimer Feld 324, D-69120 Heidelberg, Germany, Cluster of Excellence CellNetworks, Heidelberg University, Im Neuenheimer Feld 267, D-69120 Heidelberg, Germany, Department of Medicine III, University Hospital Aachen, Pauwelstrasse 30, D-52074 Aachen, Germany, Department of Infectious Diseases, Molecular Virology, Heidelberg University, Im Neuenheimer Feld 345, D-69120 Heidelberg, Germany, Division Signaling and Functional Genomics, German Cancer Research Center, Im Neuenheimer Feld 580, D-69120 Heidelberg, Germany, BioQuant, Heidelberg University, Im Neuenheimer Feld 267, D-69120 Heidelberg, Germany and Department of Pediatric Oncology, Hematology, Immunology and Pulmonology, Heidelberg University Hospital, Im Neuenheimer Feld 350, D-69120 Heidelberg, Germany
| | - Ralf Bartenschlager
- Department of Infectious Diseases, Virology, Heidelberg University Hospital, Im Neuenheimer Feld 324, D-69120 Heidelberg, Germany, Cluster of Excellence CellNetworks, Heidelberg University, Im Neuenheimer Feld 267, D-69120 Heidelberg, Germany, Department of Medicine III, University Hospital Aachen, Pauwelstrasse 30, D-52074 Aachen, Germany, Department of Infectious Diseases, Molecular Virology, Heidelberg University, Im Neuenheimer Feld 345, D-69120 Heidelberg, Germany, Division Signaling and Functional Genomics, German Cancer Research Center, Im Neuenheimer Feld 580, D-69120 Heidelberg, Germany, BioQuant, Heidelberg University, Im Neuenheimer Feld 267, D-69120 Heidelberg, Germany and Department of Pediatric Oncology, Hematology, Immunology and Pulmonology, Heidelberg University Hospital, Im Neuenheimer Feld 350, D-69120 Heidelberg, Germany
| | - Michael Boutros
- Department of Infectious Diseases, Virology, Heidelberg University Hospital, Im Neuenheimer Feld 324, D-69120 Heidelberg, Germany, Cluster of Excellence CellNetworks, Heidelberg University, Im Neuenheimer Feld 267, D-69120 Heidelberg, Germany, Department of Medicine III, University Hospital Aachen, Pauwelstrasse 30, D-52074 Aachen, Germany, Department of Infectious Diseases, Molecular Virology, Heidelberg University, Im Neuenheimer Feld 345, D-69120 Heidelberg, Germany, Division Signaling and Functional Genomics, German Cancer Research Center, Im Neuenheimer Feld 580, D-69120 Heidelberg, Germany, BioQuant, Heidelberg University, Im Neuenheimer Feld 267, D-69120 Heidelberg, Germany and Department of Pediatric Oncology, Hematology, Immunology and Pulmonology, Heidelberg University Hospital, Im Neuenheimer Feld 350, D-69120 Heidelberg, Germany
| | - Marco Binder
- Department of Infectious Diseases, Virology, Heidelberg University Hospital, Im Neuenheimer Feld 324, D-69120 Heidelberg, Germany, Cluster of Excellence CellNetworks, Heidelberg University, Im Neuenheimer Feld 267, D-69120 Heidelberg, Germany, Department of Medicine III, University Hospital Aachen, Pauwelstrasse 30, D-52074 Aachen, Germany, Department of Infectious Diseases, Molecular Virology, Heidelberg University, Im Neuenheimer Feld 345, D-69120 Heidelberg, Germany, Division Signaling and Functional Genomics, German Cancer Research Center, Im Neuenheimer Feld 580, D-69120 Heidelberg, Germany, BioQuant, Heidelberg University, Im Neuenheimer Feld 267, D-69120 Heidelberg, Germany and Department of Pediatric Oncology, Hematology, Immunology and Pulmonology, Heidelberg University Hospital, Im Neuenheimer Feld 350, D-69120 Heidelberg, Germany
| | - Konrad Streetz
- Department of Infectious Diseases, Virology, Heidelberg University Hospital, Im Neuenheimer Feld 324, D-69120 Heidelberg, Germany, Cluster of Excellence CellNetworks, Heidelberg University, Im Neuenheimer Feld 267, D-69120 Heidelberg, Germany, Department of Medicine III, University Hospital Aachen, Pauwelstrasse 30, D-52074 Aachen, Germany, Department of Infectious Diseases, Molecular Virology, Heidelberg University, Im Neuenheimer Feld 345, D-69120 Heidelberg, Germany, Division Signaling and Functional Genomics, German Cancer Research Center, Im Neuenheimer Feld 580, D-69120 Heidelberg, Germany, BioQuant, Heidelberg University, Im Neuenheimer Feld 267, D-69120 Heidelberg, Germany and Department of Pediatric Oncology, Hematology, Immunology and Pulmonology, Heidelberg University Hospital, Im Neuenheimer Feld 350, D-69120 Heidelberg, Germany
| | - Hans-Georg Kräusslich
- Department of Infectious Diseases, Virology, Heidelberg University Hospital, Im Neuenheimer Feld 324, D-69120 Heidelberg, Germany, Cluster of Excellence CellNetworks, Heidelberg University, Im Neuenheimer Feld 267, D-69120 Heidelberg, Germany, Department of Medicine III, University Hospital Aachen, Pauwelstrasse 30, D-52074 Aachen, Germany, Department of Infectious Diseases, Molecular Virology, Heidelberg University, Im Neuenheimer Feld 345, D-69120 Heidelberg, Germany, Division Signaling and Functional Genomics, German Cancer Research Center, Im Neuenheimer Feld 580, D-69120 Heidelberg, Germany, BioQuant, Heidelberg University, Im Neuenheimer Feld 267, D-69120 Heidelberg, Germany and Department of Pediatric Oncology, Hematology, Immunology and Pulmonology, Heidelberg University Hospital, Im Neuenheimer Feld 350, D-69120 Heidelberg, Germany
| | - Dirk Grimm
- Department of Infectious Diseases, Virology, Heidelberg University Hospital, Im Neuenheimer Feld 324, D-69120 Heidelberg, Germany, Cluster of Excellence CellNetworks, Heidelberg University, Im Neuenheimer Feld 267, D-69120 Heidelberg, Germany, Department of Medicine III, University Hospital Aachen, Pauwelstrasse 30, D-52074 Aachen, Germany, Department of Infectious Diseases, Molecular Virology, Heidelberg University, Im Neuenheimer Feld 345, D-69120 Heidelberg, Germany, Division Signaling and Functional Genomics, German Cancer Research Center, Im Neuenheimer Feld 580, D-69120 Heidelberg, Germany, BioQuant, Heidelberg University, Im Neuenheimer Feld 267, D-69120 Heidelberg, Germany and Department of Pediatric Oncology, Hematology, Immunology and Pulmonology, Heidelberg University Hospital, Im Neuenheimer Feld 350, D-69120 Heidelberg, Germany
| |
Collapse
|
24
|
Li Y, Ji P, Jin P. Probing the microRNA pathway with small molecules. Bioorg Med Chem 2013; 21:6119-23. [PMID: 23791866 DOI: 10.1016/j.bmc.2013.05.030] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Revised: 05/03/2013] [Accepted: 05/17/2013] [Indexed: 01/21/2023]
Abstract
MicroRNA (miRNA)/RNA interference (RNAi) is recognized as one of the most important mechanisms regulating gene expression at the posttranscriptional level in eukaryotic cells. The main components within the miRNA/RNAi pathway are now known and well characterized, but studies on the molecular mechanisms that regulate the activity of the miRNA/RNAi pathway are just beginning to emerge. High-throughput reporter assays have been developed to monitor the activity of the miRNA/RNAi pathway and applied in a proof-of-concept pilot screening, which has led to the identification of some inhibitors and activators that either generally or specifically regulate the activity of the miRNA/RNAi pathway. In addition, combined with multidisciplinary approaches like proteomics, biochemistry, and genetics, some protein co-factors were found to play important roles in the regulation of the miRNA/RNAi pathway. Herein we highlight the high-throughput reporter assays developed in recent years and the resulting discovery of miRNA/RNAi enhancers and inhibitors.
Collapse
Affiliation(s)
- Yujing Li
- Department of Human Genetics, Emory University School of Medicine, 615 Michael Street, Suite 301, Atlanta, GA 30322, USA
| | | | | |
Collapse
|
25
|
Wang L, Yang Y, Hong B. Advances in the role of microRNAs in lipid metabolism-related anti-atherosclerotic drug discovery. Expert Opin Drug Discov 2013; 8:977-90. [DOI: 10.1517/17460441.2013.798639] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
26
|
Wei L, Cao L, Xi Z. Highly Potent and Stable Capped siRNAs with Picomolar Activity for RNA Interference. Angew Chem Int Ed Engl 2013; 52:6501-3. [DOI: 10.1002/anie.201301122] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Indexed: 01/07/2023]
|
27
|
Wei L, Cao L, Xi Z. Highly Potent and Stable Capped siRNAs with Picomolar Activity for RNA Interference. Angew Chem Int Ed Engl 2013. [DOI: 10.1002/ange.201301122] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
28
|
Cao L, Xi Z. Fast thiol–maleamic methyl ester addition for facile covalent cross-linking of oligonucleotides. Tetrahedron Lett 2013. [DOI: 10.1016/j.tetlet.2013.01.112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
29
|
Toro EJ, Ostrov DA, Wronski TJ, Holliday LS. Rational identification of enoxacin as a novel V-ATPase-directed osteoclast inhibitor. Curr Protein Pept Sci 2012; 13:180-91. [PMID: 22044158 PMCID: PMC3409362 DOI: 10.2174/138920312800493151] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2011] [Revised: 06/15/2011] [Accepted: 06/16/2011] [Indexed: 11/22/2022]
Abstract
Binding between vacuolar H+-ATPases (V-ATPases) and microfilaments is mediated by an actin binding domain in the B-subunit. Both isoforms of mammalian B-subunit bind microfilaments with high affinity. A similar actin-binding activity has been demonstrated in the B-subunit of yeast. A conserved “profilin-like” domain in the B-subunit mediates this actin-binding activity, named due to its sequence and structural similarity to an actin-binding surface of the canonical actin binding protein profilin. Subtle mutations in the “profilin-like” domain eliminate actin binding activity without disrupting the ability of the altered protein to associate with the other subunits of V-ATPase to form a functional proton pump. Analysis of these mutated B-subunits suggests that the actin-binding activity is not required for the “housekeeping” functions of V-ATPases, but is important for certain specialized roles. In osteoclasts, the actin-binding activity is required for transport of V-ATPases to the plasma membrane, a prerequisite for bone resorption. A virtual screen led to the identification of enoxacin as a small molecule that bound to the actin-binding surface of the B2-subunit and competitively inhibited B2-subunit and actin interaction. Enoxacin disrupted osteoclastic bone resorption in vitro, but did not affect osteoblast formation or mineralization. Recently, enoxacin was identified as an inhibitor of the virulence of Candidaalbicans and more importantly of cancer growth and metastasis. Efforts are underway to determine the mechanisms by which enoxacin and other small molecule inhibitors of B2 and microfilament binding interaction selectively block bone resorption, the virulence of Candida, cancer growth, and metastasis.
Collapse
Affiliation(s)
- Edgardo J Toro
- Department of Orthodontics, University of Florida College of Dentistry, Gainesville, FL 32610, USA
| | | | | | | |
Collapse
|
30
|
Zhang J, Zheng J, Lu C, Du Q, Liang Z, Xi Z. Modification of the siRNA passenger strand by 5-nitroindole dramatically reduces its off-target effects. Chembiochem 2012; 13:1940-5. [PMID: 22887813 DOI: 10.1002/cbic.201200349] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Indexed: 12/16/2022]
Abstract
During the formation of RNA-induced silencing complex (RISC), the passenger and guide strand of an siRNA duplex separate from each other to generate an active RISC complex. Accumulating evidence shows that an siRNA passenger strand can also assemble into a RISC complex and mediate RNA interference, thereby causing undesired off-target effects. To reduce this effect, the so-called "universal base" 5-nitroindole nucleotides were incorporated into an siRNA passenger strand. Melting temperature and circular dichroism spectrum measurements showed no significant changes compared to the unmodified duplex, thus indicating the formation of normal A-form conformation. Using a dual luciferase reporter assay, we have further shown that 5-nitroindole modification at position 15 of the siRNA passenger strand drastically decreased the RNAi (RNA interfering) potency of this strand, whereas the potency of the RNA guide strand was not much affected. These results could provide a practical approach for reducing off-target effects mediated by the siRNA passenger strand.
Collapse
Affiliation(s)
- Junbin Zhang
- Department of Chemical Biology and State Key Laboratory of Elemento-organic Chemistry, Nankai University, Tianjin, China
| | | | | | | | | | | |
Collapse
|
31
|
Gao S, Zhang R, Yu Z, Xi Z. Antofine Analogues Can Inhibit Tobacco Mosaic Virus Assembly through Small-Molecule-RNA Interactions. Chembiochem 2012; 13:1622-7. [DOI: 10.1002/cbic.201200313] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Indexed: 11/10/2022]
|
32
|
Im HI, Kenny PJ. MicroRNAs in neuronal function and dysfunction. Trends Neurosci 2012; 35:325-34. [PMID: 22436491 PMCID: PMC3565236 DOI: 10.1016/j.tins.2012.01.004] [Citation(s) in RCA: 328] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2011] [Revised: 01/24/2012] [Accepted: 01/24/2012] [Indexed: 01/07/2023]
Abstract
MicroRNAs (miRNAs) are small noncoding RNA transcripts expressed throughout the brain that can regulate neuronal gene expression at the post-transcriptional level. Here, we provide an overview of the role for miRNAs in brain development and function, and review evidence suggesting that dysfunction in miRNA signaling contributes to neurodevelopment disorders such as Rett and fragile X syndromes, as well as complex behavioral disorders including schizophrenia, depression and drug addiction. A better understanding of how miRNAs influence the development of neuropsychiatric disorders may reveal fundamental insights into the causes of these devastating illnesses and offer novel targets for therapeutic development.
Collapse
Affiliation(s)
- Heh-In Im
- Laboratory of Behavioral and Molecular Neuroscience, Department of Molecular Therapeutics, The Scripps Research Institute - Scripps Florida, Jupiter, FL 33458, USA
| | | |
Collapse
|
33
|
Toro EJ, Zuo J, Ostrov DA, Catalfamo D, Bradaschia-Correa V, Arana-Chavez V, Caridad AR, Neubert JK, Wronski TJ, Wallet SM, Holliday LS. Enoxacin directly inhibits osteoclastogenesis without inducing apoptosis. J Biol Chem 2012; 287:17894-17904. [PMID: 22474295 DOI: 10.1074/jbc.m111.280511] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Enoxacin has been identified as a small molecule inhibitor of binding between the B2-subunit of vacuolar H+-ATPase (V-ATPase) and microfilaments. It inhibits bone resorption by calcitriol-stimulated mouse marrow cultures. We hypothesized that enoxacin acts directly and specifically on osteoclasts by disrupting the interaction between plasma membrane-directed V-ATPases, which contain the osteoclast-selective a3-subunit of V-ATPase, and microfilaments. Consistent with this hypothesis, enoxacin dose-dependently reduced the number of multinuclear cells expressing tartrate-resistant acid phosphatase (TRAP) activity produced by RANK-L-stimulated osteoclast precursors. Enoxacin (50 μM) did not induce apoptosis as measured by TUNEL and caspase-3 assays. V-ATPases containing the a3-subunit, but not the "housekeeping" a1-subunit, were isolated bound to actin. Treatment with enoxacin reduced the association of V-ATPase subunits with the detergent-insoluble cytoskeleton. Quantitative PCR revealed that enoxacin triggered significant reductions in several osteoclast-selective mRNAs, but levels of various osteoclast proteins were not reduced, as determined by quantitative immunoblots, even when their mRNA levels were reduced. Immunoblots demonstrated that proteolytic processing of TRAP5b and the cytoskeletal protein L-plastin was altered in cells treated with 50 μM enoxacin. Flow cytometry revealed that enoxacin treatment favored the expression of high levels of DC-STAMP on the surface of osteoclasts. Our data show that enoxacin directly inhibits osteoclast formation without affecting cell viability by a novel mechanism that involves changes in posttranslational processing and trafficking of several proteins with known roles in osteoclast function. We propose that these effects are downstream to blocking the binding interaction between a3-containing V-ATPases and microfilaments.
Collapse
Affiliation(s)
- Edgardo J Toro
- Department of Orthodontics, University of Florida College of Dentistry, Gainesville, Florida 32610
| | - Jian Zuo
- Department of Orthodontics, University of Florida College of Dentistry, Gainesville, Florida 32610
| | - David A Ostrov
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida College of Medicine, Gainesville, Florida 32610
| | - Dana Catalfamo
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, Florida 32610
| | - Vivian Bradaschia-Correa
- Laboratory of Oral Biology, Department of Dental Materials, School of Dentistry, University of São Paulo, 05508-900 São Paulo SP, Brazil
| | - Victor Arana-Chavez
- Laboratory of Oral Biology, Department of Dental Materials, School of Dentistry, University of São Paulo, 05508-900 São Paulo SP, Brazil
| | - Aliana R Caridad
- Department of Orthodontics, University of Florida College of Dentistry, Gainesville, Florida 32610
| | - John K Neubert
- Department of Orthodontics, University of Florida College of Dentistry, Gainesville, Florida 32610
| | - Thomas J Wronski
- Department of Physiological Sciences, University of Florida, Gainesville, Florida 32610
| | - Shannon M Wallet
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, Florida 32610
| | - L Shannon Holliday
- Department of Orthodontics, University of Florida College of Dentistry, Gainesville, Florida 32610; Department of Anatomy and Cell Biology, University of Florida College of Medicine, Gainesville, Florida 32610.
| |
Collapse
|
34
|
Georgianna WE, Young DD. Development and utilization of non-coding RNA–small molecule interactions. Org Biomol Chem 2011; 9:7969-78. [DOI: 10.1039/c1ob06324c] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
35
|
Li Y, He C, Jin P. Emergence of chemical biology approaches to the RNAi/miRNA pathway. ACTA ACUST UNITED AC 2010; 17:584-9. [PMID: 20609408 DOI: 10.1016/j.chembiol.2010.05.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2010] [Revised: 05/17/2010] [Accepted: 05/24/2010] [Indexed: 11/26/2022]
Abstract
RNA interference (RNAi) is a well-conserved mechanism that uses small noncoding RNAs to silence gene expression posttranscriptionally. Gene regulation by RNAi is now recognized as one of the major regulatory pathways in eukaryotic cells. Although the main components of the RNAi/miRNA pathway have been identified, the molecular mechanisms regulating the activity of the RNAi/miRNA pathway have only begun to emerge within the last couple of years. Recently, high-throughput reporter assays to monitor the activity of the RNAi/miRNA pathway have been developed and used for proof-of-concept pilot screens. Both inhibitors and activators of the RNAi/miRNA pathway have been found. Although still in its infancy, a chemical biology approach using high-throughput chemical screens should open up a new avenue for dissecting the RNAi/miRNA pathway, as well as developing novel RNAi- or miRNA-based therapeutic interventions.
Collapse
Affiliation(s)
- Yujing Li
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | | |
Collapse
|
36
|
Ostrov DA, Magis AT, Wronski TJ, Chan EKL, Toro EJ, Donatelli RE, Sajek K, Haroun IN, Nagib MI, Piedrahita A, Harris A, Holliday LS. Identification of enoxacin as an inhibitor of osteoclast formation and bone resorption by structure-based virtual screening. J Med Chem 2010; 52:5144-51. [PMID: 19630402 DOI: 10.1021/jm900277z] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
An interaction between the B2 subunit of vacuolar H(+)-ATPase (V-ATPase) and microfilaments is required for osteoclast bone resorption. An atomic homology model of the actin binding site on B2 was generated and molecular docking simulations were performed. Enoxacin, a fluoroquinolone antibiotic, was identified and in vitro testing demonstrated that enoxacin blocked binding between purified B2 and microfilaments. Enoxacin dose dependently reduced the number of osteoclasts differentiating in mouse marrow cultures stimulated with 1,25-dihydroxyvitamin D(3), as well as markers of osteoclast activity, and the number of resorption lacunae formed on bone slices. Enoxacin inhibited osteoclast formation at concentrations where osteoblast formation was not altered. In summary, enoxacin is a novel small molecule inhibitor of osteoclast bone resorption that acts by an unique mechanism and is therefore an attractive lead molecule for the development of a new class of antiosteoclastic agents.
Collapse
Affiliation(s)
- David A Ostrov
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Chowdhury SR, Islam MM, Kumar GS. Binding of the anticancer alkaloid sanguinarine to double stranded RNAs: insights into the structural and energetics aspects. MOLECULAR BIOSYSTEMS 2010; 6:1265-76. [PMID: 20442937 DOI: 10.1039/b927001a] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Elucidation of the molecular aspects of small molecule-RNA complexation is of prime importance for rational RNA targeted drug design strategies. Towards this, the interaction of the cytotoxic plant alkaloid sanguinarine to three double stranded ribonucleic acids, poly (A).poly(U), poly(I).poly(C) and poly(C).poly(G) was studied using various biophysical and thermodynamic techniques. Absorbance and fluorescence studies showed that the alkaloid bound cooperatively to these RNAs with binding affinities of the order 10(4) M(-1). Fluorescence quenching and hydrodynamic studies gave evidence for intercalation of sanguinarine to these RNA duplexes. Isothermal titration calorimetric studies revealed that the binding was characterized by negative enthalpy and positive entropy changes and the affinity constants derived were in agreement with the overall binding affinity values obtained from spectroscopic data. The binding of sanguinarine stabilized the melting of poly(A). poly(U) and poly(I).poly(C) and the binding data evaluated from the melting data were in agreement with that obtained from other techniques. The overall binding affinity of sanguinarine to these double stranded RNAs varied in the order, poly(A).poly(U) > poly(I).poly(C) >> poly(C).poly(G). The temperature dependence of the enthalpy changes afforded negative values of heat capacity changes for the binding of sanguinarine to poly(A).poly(U) and poly(I).poly(C), suggesting substantial hydrophobic contribution in the binding process. Further, enthalpy-entropy compensation phenomena was also seen in poly(A).poly(U) and poly(I).poly(C) systems that correlated to the strong binding involving a multiplicity of weak noncovalent interactions compared to the weak binding with poly(C).poly(G). These results further advance our understanding on the binding of small molecules that are specific binders to double stranded RNA sequences.
Collapse
Affiliation(s)
- Sebanti Roy Chowdhury
- Biophysical Chemistry Laboratory, Indian Institute of Chemical Biology (CSIR), 4, Raja S. C. Mullick Road, Kolkata 700032, India
| | | | | |
Collapse
|