scholarly journals Ago2-seq identifies new microRNA targets for seizure control

2019 ◽  
Author(s):  
Morten T. Venø ◽  
Cristina R. Reschke ◽  
Gareth Morris ◽  
Niamh M. C. Connolly ◽  
Junyi Su ◽  
...  

AbstractMicroRNAs (miRNAs) are short noncoding RNAs that shape the gene expression landscape, including during the pathogenesis of temporal lobe epilepsy (TLE). In order to provide a full catalog of the miRNA changes that happen during experimental TLE, we sequenced Argonaute 2-loaded miRNAs in the hippocampus of three different animal models at regular intervals between the time of the initial precipitating insult to the establishment of spontaneous recurrent seizures. The commonly upregulated miRNAs were selected for a functional in vivo screen using oligonucleotide inhibitors. This revealed anti-seizure phenotypes upon inhibition of miR-10a-5p, miR-21a-5p and miR-142a-5p as well as neuroprotection-only effects for inhibition of miR-27a-3p and miR-431-5p. Proteomic data and pathway analysis on predicted and validated targets of these miRNAs indicated a role for TGFβ signaling in a shared seizure-modifying mechanism. Together, these results identify functional miRNAs in the hippocampus and a pipeline of new targets for seizure control in epilepsy.

2021 ◽  
Author(s):  
Qingsong Guo ◽  
Yuhua Lu ◽  
Yan Huang ◽  
Yibing Guo ◽  
Shajun Zhu ◽  
...  

Abstract Purpose Exosome-based therapeutic approaches have been applied in diabetes. In the present study, we explored the effect of exosomes on iPSCs differentiation into insulin-producing cells and its underlying mechanisms. Methods Exosomes were isolated by ultracentrifugation from MIN6 cells and identified by Transmission electron microscopy (TEM), nanoparticle tracking analysis (NTA) and Western blot. PKH67 tracer and transwell assay were used to confirm exosome delivery into iPSCs. QRT-PCR was applied to detect key pancreatic gene expression and miRNAs expression in differentiated iPSCs. Insulin expression was assessed by flow cytometry (FCM) and immunofluorescence. The mechanism underlying exosome induction capacity for iPSCs was determined via RNA-interference of Argonaute-2 (Ago2). Streptozotozin(STZ) was used to establish diabetic mouse model to verify the function of differentiated β-like cells. Results MIN6-derived exosomes promoted the key pancreatic gene expression and immunofluorescence for Nkx6.1 and insulin remarkably, confirming the capability of exosomes for iPSCs differentiation. Moreover, transplantation of differentiated iPSCs efficiently enhanced IPGTT and partially control hyperglycemia in T1D mice. Knockdown of Ago2 in MIN6 cells affect exosomal miRNAs expression and pancreatic gene expression and insulin secretion in iPSCs.The therapeutic effect in vivo was weakened, further indicating decreased exosomal miRNA affect iPSCs differentiation.7 specific exosomal miRNAs were selected for single-assay validation. MiR-706, miR-709, miR-466c-5p and miR-423-5p were found dynamic changed during differentiation stages. Conclusion Exosomes is an effective and convenient induction approach for iPSCs differentiation into functional insulin secreting cells.The effect was downregulated via Ago2 knockdown illustrates the mechanisms are highly relevant to specific miRNAs enriched in exosomes.


2012 ◽  
Vol 78 (17) ◽  
pp. 6372-6376 ◽  
Author(s):  
Mariko Matsui ◽  
Marie-Estelle Soupé ◽  
Jérôme Becam ◽  
Cyrille Goarant

ABSTRACTTranscripts ofLeptospira16S rRNA, FlaB, LigB, LipL21, LipL32, LipL36, LipL41, and OmpL37 were quantified in the blood of susceptible (hamsters) and resistant (mice) animal models of leptospirosis. We first validated adequate reference genes and then evaluated expression patternsin vivocompared toin vitrocultures. LipL32 expression was downregulatedin vivoand differentially regulated in resistant and susceptible animals. FlaB expression was also repressed in mice but not in hamsters. In contrast, LigB and OmpL37 were upregulatedin vivo. Thus, we demonstrated that a virulent strain ofLeptospiradifferentially adapts its gene expression in the blood of infected animals.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 3246-3246 ◽  
Author(s):  
Monica Galli ◽  
Hila Magen ◽  
Hermann Einsele ◽  
Manik Chatterjee ◽  
Mariella Grasso ◽  
...  

Abstract Background: HPSE is an endo-ß-d-glucuronidase that trims the heparan sulfate (HS) chains of proteoglycans, releasing biologically active fragments of HS. HPSE activity impacts cell signaling, gene expression and promotes extracellular matrix remodeling within the tumor microenvironment; high HPSE expression is associated with enhanced tumor growth, angiogenesis and metastases in several cancer types. As a result of its tumor promoting activities, HPSE is a promising new and unexploited target for anti-cancer therapy. There is a single enzymatically active HPSE in humans and HPSE knockout mice appear to be healthy, thus therapeutic neutralization of HPSE activity would likely have limited negative side effects. In MM preclinical models HPSE was shown to be a master regulator of aggressive tumor behavior. Preclinical evidence also indicates that HPSE promotes chemoresistance suggesting it plays a pivotal role in regulating myeloma response to therapy (Ramani VPC et al., AACR 2014, Abstract nr. 1708). In preclinical studies, bortezomib or melphalan were found to enhance HPSE expression and secretion. High HPSE expressing MM cells were less susceptible to the cytotoxic effects of those drugs. Likewise, a very significant increase in HPSE gene expression following chemotherapy was observed in patient-derived tumor samples, indicating a potential role for HPSE in regulating myeloma response to therapy. Roneparstat (SST0001), a 100% N-acetylated and glycol split heparin, is a potent HPSE inhibitor devoid of any significant anticoagulant activity. In an in vivo model of disseminated myeloma, Roneparstat in combination with either bortezomib or melphalan, significantly decreased both the number of animals with detectable tumor and the tumor burden when compared with animals treated with either of these drugs alone. In addition, studies in animal models of MM indicated that the mechanism of action of Roneparstat was consistent with it having anti-HPSE activity in vivo (reduced angiogenesis and diminished expression of HGF, VEGF and MMP-9 and diminished HPSE induced shedding of syndecan-1, a HS proteoglycan known to be a potent promoter of myeloma growth). Patients and Methods: A First in Man, multicenter, international, phase I clinical study is currently ongoing in advanced heavily pre-treated refractory MM patients (pts) who have exhausted all available anti-MM therapies. Roneparstat is administered subcutaneously, with a starting flat dose defined according to ICH S9 guidelines. A schedule DX5W1,W2 Q28D is being tested. Each cohort plans 3 + 3 pts. A direct fluorescence method (Heparin Red assay) is used in pharmacokinetic studies along with aPTT, used as a surrogate (indirect) measurement of Roneparstat plasma concentration. The pharmacodynamic effect of the drug on the coagulation cascade and any antitumor effect are also evaluated. Results: 15 pts have been enrolled to date. 5 cohorts (doses ranging from 25 to 200 mg/day) have been evaluated, while a 400 mg cohort has just been opened. Five pts have received 1 cycle of therapy, six pts 2 cycles, one 3 cycles, one 5 cycles, one 9 cycles; one patient is currently on treatment, one is not evaluable. Roneparstat administration was found to be safe with only minimal transient side effects. No DLTs and no bleeding complications have been observed. Roneparstat has been well tolerated both systemically and locally. The only side effect observed was minor reactions (redness, bruising) at the injection site (in 6 pts, all grade 1). A decrease > 50% in the serum monoclonal component was observed in one patient, lasting for 6 cycles. Conclusions: Preclinical studies in MM lines and animal models have demonstrated Roneparstat as a potent anti-myeloma compound, particularly when used in combination with other drugs. In the ongoing Phase I escalating dose study (n. pts = 15), Roneparstat administration (at a dose of up to 200 mg/day) was found to be safe with only minimal local side effects. Based on these results, Roneparstat, at a defined dose, in combination with other anti-myeloma agents, will be evaluated in relapsed/resistant MM pts. Disclosures Galli: sigma-tau Research Switzerland SA: Consultancy. Einsele:Novartis: Consultancy, Honoraria, Speakers Bureau; Celgene: Consultancy, Honoraria, Research Funding, Speakers Bureau; Janssen: Consultancy, Honoraria, Research Funding, Speakers Bureau; Amgen/Onyx: Consultancy, Honoraria, Speakers Bureau. Barbieri:sigma-tau Research Switzerland SA, Mendrisio, Switzerland: Employment. Paoletti:sigma-tau Research Switzerland SA, Mendrisio, Switzerland: Employment. Pace:sigma-sau Industrie Farmaceutiche Riunite SpA, Pomezia (RM), Italy: Employment. Sanderson:Sigma-tau Research S.P.A.: Consultancy, Research Funding. Nagler:Novaratis Pharmaceuticals Corporation: Consultancy, Honoraria, Research Funding.


2019 ◽  
Vol 10 (1) ◽  
Author(s):  
Jordan P. Lewandowski ◽  
James C. Lee ◽  
Taeyoung Hwang ◽  
Hongjae Sunwoo ◽  
Jill M. Goldstein ◽  
...  

Abstract RNA has been classically known to play central roles in biology, including maintaining telomeres, protein synthesis, and in sex chromosome compensation. While thousands of long noncoding RNAs (lncRNAs) have been identified, attributing RNA-based roles to lncRNA loci requires assessing whether phenotype(s) could be due to DNA regulatory elements, transcription, or the lncRNA. Here, we use the conserved X chromosome lncRNA locus Firre, as a model to discriminate between DNA- and RNA-mediated effects in vivo. We demonstrate that (i) Firre mutant mice have cell-specific hematopoietic phenotypes, and (ii) upon exposure to lipopolysaccharide, mice overexpressing Firre exhibit increased levels of pro-inflammatory cytokines and impaired survival. (iii) Deletion of Firre does not result in changes in local gene expression, but rather in changes on autosomes that can be rescued by expression of transgenic Firre RNA. Together, our results provide genetic evidence that the Firre locus produces a trans-acting lncRNA that has physiological roles in hematopoiesis.


Author(s):  
Paula Marincola Smith ◽  
Yash A Choksi ◽  
Nicholas O Markham ◽  
David N Hanna ◽  
Jinghuan Zi ◽  
...  

Background: Defective barrier function is a predisposing factor in inflammatory bowel disease (IBD) and colitis-associated cancer (CAC). While TGFβ signaling defects have been associated with IBD and CAC, few studies have examined the relationship between TGFβ and intestinal barrier function. Here, we examine the role of TGFβ signaling via SMAD4 in modulation of colon barrier function. Methods: The Smad4 gene was conditionally deleted in the intestines of adult mice and intestinal permeability assessed using an in vivo 4kD FITC-Dextran (FD4) permeability assay. Mouse colon was isolated for gene expression (RNA-sequencing), western blot, and immunofluorescence analysis. In vitro colon organoid culture was utilized to assess junction-related gene expression by qPCR and trans-epithelial resistance (TER). In silico analyses of human IBD and colon cancer databases were performed. Results: Mice lacking intestinal expression of Smad4 demonstrate increased colonic permeability to FD4 without gross mucosal damage. mRNA/protein expression analyses demonstrate significant increases in Cldn2/Claudin 2 and Cldn8/Claudin 8, and decreases in Cldn3, Cldn4, and Cldn7/Claudin 7 with intestinal SMAD4 loss in vivo without changes in Claudin protein localization. TGFb1/BMP2 treatment of polarized SMAD4+ colonoids increases TER. Cldn2, Cldn4, Cldn7, and Cldn8 are regulated by canonical TGFβ signaling, and TGFβ-dependent regulation of these genes is dependent on nascent RNA transcription (Cldn2, Cldn4, Cldn8) but not nascent protein translation (Cldn4, Cldn8). Human IBD/colon cancer specimens demonstrate decreased SMAD4, CLDN4, CLDN7, and CLDN8 and increased CLDN2 compared to healthy controls. Conclusion: Canonical TGFβ signaling modulates the expression of tight junction proteins and barrier function in mouse colon.


2019 ◽  
Author(s):  
Jordan P. Lewandowski ◽  
James C. Lee ◽  
Taeyoung Hwang ◽  
Hongjae Sunwoo ◽  
Jill M. Goldstein ◽  
...  

ABSTRACTRNA has been classically known to play central roles in biology, including maintaining telomeres1, protein synthesis2, and in sex chromosome compensation in certain species3,4. At the center of these important biological systems are noncoding RNAs. While thousands of long noncoding RNAs (lncRNAs) have been identified in mammalian genomes5–8, attributing RNA-based roles to lncRNA loci requires an assessment of whether the observed effect could be due to DNA regulatory elements, the act of transcription, or the lncRNA transcript. Here, we use the syntenically conserved lncRNA locus, Functional intergenic repeating RNA element (Firre), that is located on the X chromosome as a model to discriminate between DNA- and RNA-mediated effects in vivo. To this end, we generated genetically defined loss-of-function, gain-of-function, and rescue mouse models for Firre and provide genetic evidence that the Firre locus produces a trans-acting RNA. We report that: (i) Firre mutant mice have cell-specific defects during hematopoiesis and changes in gene expression that can be rescued by induction of Firre RNA from a transgene in the Firre knockout background, (ii) mice overexpressing Firre from a transgene exhibit increased levels of pro-inflammatory cytokines and impaired survival upon exposure to lipopolysaccharide, and (iii) deletion of the Firre locus did not result in changes in local gene expression on the X chromosome in 9 different biological contexts, suggesting that Firre does not function by cis-acting RNA or DNA elements. Together, our results provide genetic evidence that the Firre locus produces a trans-acting lncRNA that has physiological roles in hematopoiesis and immune function.


2010 ◽  
Vol 15 (10) ◽  
pp. 987-995 ◽  
Author(s):  
K A Garbett ◽  
S Horváth ◽  
P J Ebert ◽  
M J Schmidt ◽  
K Lwin ◽  
...  

2020 ◽  
Vol 21 (10) ◽  
pp. 3696 ◽  
Author(s):  
Deyanira Contartese ◽  
Matilde Tschon ◽  
Monica De Mattei ◽  
Milena Fini

Osteoarthritis (OA) is a highly prevalent joint disease that primarily affects about 10% of the world’s population over 60 years old. The purpose of this study is to systematically review the preclinical studies regarding sex differences in OA, with particular attention to the molecular aspect and gene expression, but also to the histopathological aspects. Three databases (PubMed, Scopus, and Web of Knowledge) were screened for eligible studies. In vitro and in vivo papers written in English, published in the last 11 years (2009–2020) were eligible. Participants were preclinical studies, including cell cultures and animal models of OA, evaluating sex differences. Independent extraction of articles and quality assessments were performed by two authors using predefined data fields and specific tools (Animals in Research Reporting In Vivo Experiments (ARRIVE) guideline and Systematic Review Centre for Laboratory animal Experimentation (SYRCLE) tool). Twenty-three studies were included in the review: 4 in vitro studies, 18 in vivo studies, and 1 both in vitro and in vivo study. From in vitro works, sex differences were found in the gene expression of inflammatory molecules, hormonal receptors, and in responsiveness to hormonal stimulation. In vivo research showed a great heterogeneity of animal models mainly focused on the histopathological aspects rather than on the analysis of sex-related molecular mechanisms. This review highlights that many gaps in knowledge still exist; improvementsin the selection and reporting of animal models, the use of advanced in vitro models, and multiomics analyses might contribute to developing a personalized gender-based medicine.


Sign in / Sign up

Export Citation Format

Share Document