Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Rep ; 9(1): 16838, 2019 11 14.
Article in English | MEDLINE | ID: mdl-31727959

ABSTRACT

Hemophilia A is a monogenic disease with a blood clotting factor VIII (FVIII) deficiency caused by mutation in the factor VIII (F8) gene. Current and emerging treatments such as FVIII protein injection and gene therapies via AAV-delivered F8 transgene in an episome are costly and nonpermanent. Here, we describe a CRISPR/Cas9-based in vivo genome editing method, combined with non-homologous end joining, enabling permanent chromosomal integration of a modified human B domain deleted-F8 (BDD-F8) at the albumin (Alb) locus in liver cells. To test the approach in mice, C57BL/6 mice received tail vein injections of two vectors, AAV8-SaCas9-gRNA, targeting Alb intron 13, and AAV8-BDD-F8. This resulted in BDD-F8 insertion at the Alb locus and FVIII protein expression in the liver of vector-, but not vehicle-, treated mice. Using this approach in hemophilic mice, BDD-F8 was expressed in liver cells as functional human FVIII, leading to increased plasma levels of FVIII and restoration of blood clotting properties in a dose-dependent manor for at least 7 months, with no detectable liver toxicity or meaningful off-target effects. Based on these findings, our BDD-F8 genome editing approach may offer an efficacious, long-term and safe treatment for patients with hemophilia A.


Subject(s)
Dependovirus/genetics , Factor VIII/genetics , Gene Editing/methods , Hemophilia A/therapy , Albumins/genetics , Animals , CRISPR-Cas Systems , Disease Models, Animal , Factor VIII/chemistry , Genetic Therapy , Genetic Vectors/administration & dosage , Hemophilia A/genetics , Humans , Mice , Mice, Inbred C57BL , Protein Domains , Treatment Outcome
2.
Eur J Endocrinol ; 181(5): 565-577, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31539877

ABSTRACT

DESIGN: Gestational diabetes mellitus (GDM) is one of the most common pregnancy complications and its prevalence is constantly rising worldwide. Diagnosis is commonly in the late second or early third trimester of pregnancy, though the development of GDM starts early; hence, first-trimester diagnosis is feasible. OBJECTIVE: Our objective was to identify microRNAs that best distinguish GDM samples from those of healthy pregnant women and to evaluate the predictive value of microRNAs for GDM detection in the first trimester. METHODS: We investigated the abundance of circulating microRNAs in the plasma of pregnant women in their first trimester. Two populations were included in the study to enable population-specific as well as cross-population inspection of expression profiles. Each microRNA was tested for differential expression in GDM vs control samples, and their efficiency for GDM detection was evaluated using machine-learning models. RESULTS: Two upregulated microRNAs (miR-223 and miR-23a) were identified in GDM vs the control set, and validated on a new cohort of women. Using both microRNAs in a logistic-regression model, we achieved an AUC value of 0.91. We further demonstrated the overall predictive value of microRNAs using several types of multivariable machine-learning models that included the entire set of expressed microRNAs. All models achieved high accuracy when applied on the dataset (mean AUC = 0.77). The significance of the classification results was established via permutation tests. CONCLUSIONS: Our findings suggest that circulating microRNAs are potential biomarkers for GDM in the first trimester. This warrants further examination and lays the foundation for producing a novel early non-invasive diagnostic tool for GDM.


Subject(s)
Circulating MicroRNA/blood , Diabetes, Gestational/blood , Diabetes, Gestational/diagnosis , Adipose Tissue/chemistry , Adult , Case-Control Studies , Early Diagnosis , Female , Humans , Machine Learning , MicroRNAs/blood , Placenta/chemistry , Predictive Value of Tests , Pregnancy , Pregnancy Trimester, First , Reproducibility of Results
3.
Sci Rep ; 8(1): 3401, 2018 02 21.
Article in English | MEDLINE | ID: mdl-29467498

ABSTRACT

Preeclampsia is one of the most dangerous pregnancy complications, and the leading cause of maternal and perinatal mortality and morbidity. Although the clinical symptoms appear late, its origin is early, and hence detection is feasible already at the first trimester. In the current study, we investigated the abundance of circulating small non-coding RNAs in the plasma of pregnant women in their first trimester, seeking transcripts that best separate the preeclampsia samples from those of healthy pregnant women. To this end, we performed small non-coding RNAs sequencing of 75 preeclampsia and control samples, and identified 25 transcripts that were differentially expressed between preeclampsia and the control groups. Furthermore, we utilized those transcripts and created a pipeline for a supervised classification of preeclampsia. Our pipeline generates a logistic regression model using a 5-fold cross validation on numerous random partitions into training and blind test sets. Using this classification procedure, we achieved an average AUC value of 0.86. These findings suggest the predictive value of circulating small non-coding RNA in the first trimester, warranting further examination, and lay the foundation for producing a novel early non-invasive diagnostic tool for preeclampsia, which could reduce the life-threatening risk for both the mother and fetus.


Subject(s)
Pre-Eclampsia/blood , Pre-Eclampsia/diagnosis , RNA, Small Untranslated/blood , Adult , Biomarkers/blood , Case-Control Studies , Early Diagnosis , Female , Gestational Age , Humans , Infant, Newborn , Male , Pregnancy , Pregnancy Outcome , Pregnancy Trimester, First/blood , Pregnancy Trimester, Second/blood , Prospective Studies , Risk Factors
4.
Oncotarget ; 8(16): 25963-25976, 2017 Apr 18.
Article in English | MEDLINE | ID: mdl-28404930

ABSTRACT

Hormone receptor status is of significant value when deciding on anti-estrogenic adjuvant therapy for breast cancer tumors. However, while estrogen receptor (ER) regulation was intensively studied, the regulation of progesterone receptor (PR) levels has not been extensively investigated. MicroRNAs (miRNAs, miRs) are post-transcriptional negative regulators of gene expression involved in diverse cellular processes. The aim of this study was to identify miRNAs that regulate PR in breast cancer.We mapped potential miRNA binding sites for miR-181a, miR-23a and miR-26b on PR mRNA and demonstrated a direct regulation of PR by these three miRNAs by in-vitro Luciferase binding assays. Over-expression of each miRNA in MCF-7 cells resulted in a reduction in the expression levels of PR mRNA. Then, expression levels of these miRNAs were measured in Formalin-Fixed, Paraffin-Embedded (FFPE) samples of 29 ER-positive breast cancer tumors and adjacent normal breast tissues. A significant reciprocal correlation between PR mRNA and the miRNA levels were identified suggesting a role for miR-181a, miR-23a and miR-26b in PR regulation in breast cancer. Moreover, the average expression fold-changes of the three miRNAs between cancerous and normal tissues displayed an opposite trend when analyzing according to Immuno-histochemistry(IHC) status. Furthermore, miR-181a and miR-26b were found to be over-expressed in most tumor tissues supporting their role in ER-positive breast cancer development. We conclude that miR-181a, miR-23a and miR-26b act as negative regulators of PR expression in ER-positive breast cancer. The diagnostic and prognostic potential of these miRNAs in breast cancer should be further evaluated.


Subject(s)
Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , RNA Interference , Receptors, Progesterone/genetics , 3' Untranslated Regions , Adult , Aged , Aged, 80 and over , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Gene Expression , Genes, Reporter , Humans , Middle Aged , Neoplasm Staging , RNA, Messenger/genetics
5.
Nat Commun ; 7: 12868, 2016 09 19.
Article in English | MEDLINE | ID: mdl-27641360

ABSTRACT

Metastasis is the primary cause for mortality in breast cancer. MicroRNAs, gene expression master regulators, constitute an attractive candidate to control metastasis. Here we show that breast cancer metastasis can be prevented by miR-96 or miR-182 treatment, and decipher the mechanism of action. We found that miR-96/miR-182 downregulate Palladin protein levels, thereby reducing breast cancer cell migration and invasion. A common SNP, rs1071738, at the miR-96/miR-182-binding site within the Palladin 3'-UTR abolishes miRNA:mRNA binding, thus diminishing Palladin regulation by these miRNAs. Regulation is successfully restored by applying complimentary miRNAs. A hydrogel-embedded, gold-nanoparticle-based delivery vehicle provides efficient local, selective, and sustained release of miR-96/miR-182, markedly suppressing metastasis in a breast cancer mouse model. Combined delivery of the miRNAs with a chemotherapy drug, cisplatin, enables significant primary tumour shrinkage and metastasis prevention. Our data corroborate the role of miRNAs in metastasis, and suggest miR-96/miR-182 delivery as a potential anti-metastatic drug.


Subject(s)
Breast Neoplasms/drug therapy , Carcinoma/drug therapy , Cytoskeletal Proteins/metabolism , MicroRNAs/therapeutic use , Phosphoproteins/metabolism , Animals , Breast Neoplasms/metabolism , Carcinoma/metabolism , Cell Movement , Cell Proliferation , Gene Expression Regulation, Neoplastic , HEK293 Cells , HeLa Cells , Humans , MCF-7 Cells , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/metabolism , Mice, Inbred BALB C , MicroRNAs/metabolism , Neoplasm Metastasis , Polymorphism, Single Nucleotide , Xenograft Model Antitumor Assays
6.
PLoS One ; 11(1): e0146236, 2016.
Article in English | MEDLINE | ID: mdl-26730965

ABSTRACT

Stress research has progressively become more integrative in nature, seeking to unfold crucial relations between the different phenotypic levels of stress manifestations. This study sought to unravel stress-induced variations in expression of human microRNAs sampled in peripheral blood mononuclear cells and further assess their relationship with neuronal and psychological indices. We obtained blood samples from 49 healthy male participants before and three hours after performing a social stress task, while undergoing functional magnetic resonance imaging (fMRI). A seed-based functional connectivity (FC) analysis was conducted for the ventro-medial prefrontal cortex (vmPFC), a key area of stress regulation. Out of hundreds of microRNAs, a specific increase was identified in microRNA-29c (miR-29c) expression, corresponding with both the experience of sustained stress via self-reports, and alterations in vmPFC functional connectivity. Explicitly, miR-29c expression levels corresponded with both increased connectivity of the vmPFC with the anterior insula (aIns), and decreased connectivity of the vmPFC with the left dorso-lateral prefrontal cortex (dlPFC). Our findings further revealed that miR-29c mediates an indirect path linking enhanced vmPFC-aIns connectivity during stress with subsequent experiences of sustained stress. The correlative patterns of miR-29c expression and vmPFC FC, along with the mediating effects on subjective stress sustainment and the presumed localization of miR-29c in astrocytes, together point to an intriguing assumption; miR-29c may serve as a biomarker in the blood for stress-induced functional neural alterations reflecting regulatory processes. Such a multi-level model may hold the key for future personalized intervention in stress psychopathology.


Subject(s)
Brain/physiopathology , MicroRNAs/metabolism , Stress, Psychological/metabolism , Epigenesis, Genetic , Functional Neuroimaging , Humans , Magnetic Resonance Imaging , Male , MicroRNAs/genetics , Neural Pathways/physiopathology , Stress, Psychological/genetics , Stress, Psychological/physiopathology , Young Adult
7.
Genet Res (Camb) ; 95(2-3): 53-6, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23938110

ABSTRACT

A recent E-Rare workshop reviewed the ethical aspects of whole exome and whole genome-sequencing studies (WES and WGS, respectively) in rare diseases. Leveraging new genomic technologies, which output vast amounts of known and novel genetic variants, researchers are learning more about the genetic basis and mechanisms involved in rare diseases. In some cases, these findings are translated into diagnostic tools for the benefit of rare disease patients. Among the disclosed data, which can assist in treatment management, incidental findings await, bringing with them ethical concerns for the clinicians, researchers and patients.


Subject(s)
Ethics, Medical , Exome , Genome, Human , Sequence Analysis, DNA , Humans , Israel
8.
Breast Cancer Res Treat ; 138(3): 753-60, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23549953

ABSTRACT

Several lines of evidence indicate that sequence alterations within microRNA (miRNA)-binding sites can modify the binding to its target gene resulting in altered expression patterns. We hypothesized that a single nucleotide polymorphism (SNP) located in the miR-515-5p binding site of igf-1r gene may alter IGF-1R regulation, with consequent effects on breast cancer risk in BRCA1 mutation carriers. Computational prediction revealed that the rs28674628 SNP in the igf-1r 3' UTR is located within a predicted binding site for miR-515-5p. The effect of this SNP on breast cancer risk was evaluated by genotyping 115 Jewish Ashkenazi carriers of the 185delAG mutation in the BRCA1 gene using the Sequenom platform followed by Kaplan-Meier analysis. Additional data set of 378 Jewish BRCA1 carriers was analyzed to validate our results. MiRNA transfection, Western blot analysis, luciferase reporter assay, real time PCR, and immunohistochemistry were performed to assess direct regulation of igf-1r by miR-515-5p. We show direct regulation of IGF-1R by miR-515-5p. We identified that disrupting miR-515-5p and igf-1r 3' UTR binding by SNP may cause elevated IGF-1R protein levels. Interestingly, miR-515-5p is downregulated in tumor tissue compared to its non-neoplastic surrounding tissue while IGF-1R levels are elevated. This igf-1r SNP was found to be significantly associated with age at diagnosis of breast cancer in Jewish Ashkenazi BRCA1 mutation carriers. These findings support the hypothesis that a SNP located in igf-1r gene may alter miRNA regulation of IGF-1R, with a putative effect on BRCA1 penetrance and breast cancer risk.


Subject(s)
Breast Neoplasms/genetics , Genes, BRCA1 , Heterozygote , MicroRNAs/genetics , Receptor, IGF Type 1/genetics , 3' Untranslated Regions , Adult , Age Factors , Aged , Aged, 80 and over , Base Sequence , Binding Sites , Breast Neoplasms/mortality , Female , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Humans , Jews/genetics , Kaplan-Meier Estimate , Middle Aged , Molecular Sequence Data , Polymorphism, Single Nucleotide , Receptor, IGF Type 1/metabolism , Reproducibility of Results
SELECTION OF CITATIONS
SEARCH DETAIL
...