Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Reka A. Haraszti is active.

Publication


Featured researches published by Reka A. Haraszti.


Journal of extracellular vesicles | 2016

High-resolution proteomic and lipidomic analysis of exosomes and microvesicles from different cell sources

Reka A. Haraszti; Marie-Cecile Didiot; Ellen Sapp; John D. Leszyk; Scott A. Shaffer; Hannah Rockwell; Fei Gao; Niven Rajin Narain; Marian DiFiglia; Michael Andrew Kiebish; Neil Aronin; Anastasia Khvorova

Extracellular vesicles (EVs), including exosomes and microvesicles (MVs), are explored for use in diagnostics, therapeutics and drug delivery. However, little is known about the relationship of protein and lipid composition of EVs and their source cells. Here, we report high-resolution lipidomic and proteomic analyses of exosomes and MVs derived by differential ultracentrifugation from 3 different cell types: U87 glioblastoma cells, Huh7 hepatocellular carcinoma cells and human bone marrow-derived mesenchymal stem cells (MSCs). We identified 3,532 proteins and 1,961 lipid species in the screen. Exosomes differed from MVs in several different areas: (a) The protein patterns of exosomes were more likely different from their cells of origin than were the protein patterns of MVs; (b) The proteomes of U87 and Huh7 exosomes were similar to each other but different from the proteomes of MSC exosomes, whereas the lipidomes of Huh7 and MSC exosomes were similar to each other but different from the lipidomes of U87 exosomes; (c) exosomes exhibited proteins of extracellular matrix, heparin-binding, receptors, immune response and cell adhesion functions, whereas MVs were enriched in endoplasmic reticulum, proteasome and mitochondrial proteins. Exosomes and MVs also differed in their types of lipid contents. Enrichment in glycolipids and free fatty acids characterized exosomes, whereas enrichment in ceramides and sphingomyelins characterized MVs. Furthermore, Huh7 and MSC exosomes were specifically enriched in cardiolipins; U87 exosomes were enriched in sphingomyelins. This study comprehensively analyses the protein and lipid composition of exosomes, MVs and source cells in 3 different cell types.


Molecular Therapy | 2016

Exosome-mediated Delivery of Hydrophobically Modified siRNA for Huntingtin mRNA Silencing.

Marie-Cecile Didiot; Lauren M Hall; Andrew H. Coles; Reka A. Haraszti; Bruno M.D.C. Godinho; Kathryn Chase; Ellen Sapp; Socheata Ly; Julia F. Alterman; Matthew R. Hassler; Dimas Echeverria; Lakshmi Raj; David V. Morrissey; Marian DiFiglia; Neil Aronin; Anastasia Khvorova

Delivery represents a significant barrier to the clinical advancement of oligonucleotide therapeutics for the treatment of neurological disorders, such as Huntingtons disease. Small, endogenous vesicles known as exosomes have the potential to act as oligonucleotide delivery vehicles, but robust and scalable methods for loading RNA therapeutic cargo into exosomes are lacking. Here, we show that hydrophobically modified small interfering RNAs (hsiRNAs) efficiently load into exosomes upon co-incubation, without altering vesicle size distribution or integrity. Exosomes loaded with hsiRNAs targeting Huntingtin mRNA were efficiently internalized by mouse primary cortical neurons and promoted dose-dependent silencing of Huntingtin mRNA and protein. Unilateral infusion of hsiRNA-loaded exosomes, but not hsiRNAs alone, into mouse striatum resulted in bilateral oligonucleotide distribution and statistically significant bilateral silencing of up to 35% of Huntingtin mRNA. The broad distribution and efficacy of hsiRNA-loaded exosomes delivered to brain is expected to advance the development of therapies for the treatment of Huntingtons disease and other neurodegenerative disorders.


Molecular therapy. Nucleic acids | 2016

Docosahexaenoic Acid Conjugation Enhances Distribution and Safety of siRNA upon Local Administration in Mouse Brain

Mehran Nikan; Maire F. Osborn; Andrew H. Coles; Bruno M.D.C. Godinho; Lauren M Hall; Reka A. Haraszti; Matthew R. Hassler; Dimas Echeverria; Neil Aronin; Anastasia Khvorova

The use of siRNA-based therapies for the treatment of neurodegenerative disease requires efficient, nontoxic distribution to the affected brain parenchyma, notably the striatum and cortex. Here, we describe the synthesis and activity of a fully chemically modified siRNA that is directly conjugated to docosahexaenoic acid (DHA), the most abundant polyunsaturated fatty acid in the mammalian brain. DHA conjugation enables enhanced siRNA retention throughout both the ipsilateral striatum and cortex following a single, intrastriatal injection (ranging from 6-60 μg). Within these tissues, DHA conjugation promotes internalization by both neurons and astrocytes. We demonstrate efficient and specific silencing of Huntingtin mRNA expression in both the ipsilateral striatum (up to 73%) and cortex (up to 51%) after 1 week. Moreover, following a bilateral intrastriatal injection (60 μg), we achieve up to 80% silencing of a secondary target, Cyclophilin B, at both the mRNA and protein level. Importantly, DHA-hsiRNAs do not induce neural cell death or measurable innate immune activation following administration of concentrations over 20 times above the efficacious dose. Thus, DHA conjugation is a novel strategy for improving siRNA activity in mouse brain, with potential to act as a new therapeutic platform for the treatment of neurodegenerative disorders.The use of siRNA-based therapies for the treatment of neurodegenerative disease requires efficient, nontoxic distribution to the affected brain parenchyma, notably the striatum and cortex. Here, we describe the synthesis and activity of a fully chemically modified siRNA that is directly conjugated to docosahexaenoic acid (DHA), the most abundant polyunsaturated fatty acid in the mammalian brain. DHA conjugation enables enhanced siRNA retention throughout both the ipsilateral striatum and cortex following a single, intrastriatal injection (ranging from 6–60 μg). Within these tissues, DHA conjugation promotes internalization by both neurons and astrocytes. We demonstrate efficient and specific silencing of Huntingtin mRNA expression in both the ipsilateral striatum (up to 73%) and cortex (up to 51%) after 1 week. Moreover, following a bilateral intrastriatal injection (60 μg), we achieve up to 80% silencing of a secondary target, Cyclophilin B, at both the mRNA and protein level. Importantly, DHA-hsiRNAs do not induce neural cell death or measurable innate immune activation following administration of concentrations over 20 times above the efficacious dose. Thus, DHA conjugation is a novel strategy for improving siRNA activity in mouse brain, with potential to act as a new therapeutic platform for the treatment of neurodegenerative disorders.


Nucleic Acids Research | 2017

5΄-Vinylphosphonate improves tissue accumulation and efficacy of conjugated siRNAs in vivo

Reka A. Haraszti; Loic Roux; Andrew H. Coles; Anton A. Turanov; Julia F. Alterman; Dimas Echeverria; Bruno M.D.C. Godinho; Neil Aronin; Anastasia Khvorova

Abstract 5΄-Vinylphosphonate modification of siRNAs protects them from phosphatases, and improves silencing activity. Here, we show that 5΄-vinylphosphonate confers novel properties to siRNAs. Specifically, 5΄-vinylphosphonate (i) increases siRNA accumulation in tissues, (ii) extends duration of silencing in multiple organs and (iii) protects siRNAs from 5΄-to-3΄ exonucleases. Delivery of conjugated siRNAs requires extensive chemical modifications to achieve stability in vivo. Because chemically modified siRNAs are poor substrates for phosphorylation by kinases, and 5΄-phosphate is required for loading into RNA-induced silencing complex, the synthetic addition of a 5΄-phosphate on a fully modified siRNA guide strand is expected to be beneficial. Here, we show that synthetic phosphorylation of fully modified cholesterol-conjugated siRNAs increases their potency and efficacy in vitro, but when delivered systemically to mice, the 5΄-phosphate is removed within 2 hours. The 5΄-phosphate mimic 5΄-(E)-vinylphosphonate stabilizes the 5΄ end of the guide strand by protecting it from phosphatases and 5΄-to-3΄ exonucleases. The improved stability increases guide strand accumulation and retention in tissues, which significantly enhances the efficacy of cholesterol-conjugated siRNAs and the duration of silencing in vivo. Moreover, we show that 5΄-(E)-vinylphosphonate stabilizes 5΄ phosphate, thereby enabling systemic delivery to and silencing in kidney and heart.


Bioconjugate Chemistry | 2017

Synthesis and Evaluation of Parenchymal Retention and Efficacy of a Metabolically Stable O-Phosphocholine-N-docosahexaenoyl-l-serine siRNA Conjugate in Mouse Brain.

Mehran Nikan; Maire F. Osborn; Andrew H. Coles; Annabelle Biscans; Bruno M.D.C. Godinho; Reka A. Haraszti; Ellen Sapp; Dimas Echeverria; Marian DiFiglia; Neil Aronin; Anastasia Khvorova

Ligand-conjugated siRNAs have the potential to achieve targeted delivery and efficient silencing in neurons following local administration in the central nervous system (CNS). We recently described the activity and safety profile of a docosahexaenoic acid (DHA)-conjugated, hydrophobic siRNA (DHA-hsiRNA) targeting Huntingtin (Htt) mRNA in mouse brain. Here, we report the synthesis of an amide-modified, phosphocholine-containing DHA-hsiRNA conjugate (PC-DHA-hsiRNA), which closely resembles the endogenously esterified biological structure of DHA. We hypothesized that this modification may enhance neuronal delivery in vivo. We demonstrate that PC-DHA-hsiRNA silences Htt in mouse primary cortical neurons and astrocytes. After intrastriatal delivery, Htt-targeting PC-DHA-hsiRNA induces ∼80% mRNA silencing and 71% protein silencing after 1 week. However, PC-DHA-hsiRNA did not substantially outperform DHA-hsiRNA under the conditions tested. Moreover, at the highest locally administered dose (4 nmol, 50 μg), we observe evidence of PC-DHA-hsiRNA-mediated reactive astrogliosis. Lipophilic ligand conjugation enables siRNA delivery to neural tissues, but rational design of functional, nontoxic siRNA conjugates for CNS delivery remains challenging.


The Lancet | 2014

A new frontier in atherosclerotic coronary imaging

Gregory S. Thomas; Reka A. Haraszti

674 www.thelancet.com Vol 383 February 22, 2014 Ischaemic heart disease resulting from rupture of atherosclerotic plaques is a major cause of death worldwide. Precisely why a plaque ruptures remains a mystery. However, in The Lancet, Nikhil Joshi and colleagues’ fi ndings suggest that we are close to being able to detect when rupture is about to occur. The simple and inexpensive 18F-sodium fl uoride (18F-NaF) PET radioisotope, used for 30 years to image bone formation, was found to signify metabolically active calcifi cation in the aorta by Derlin and colleagues and in the coronary arteries by Beheshti, Dweck, and Li, and their colleagues. In their landmark article, Joshi and coworkers move this nascent fi eld much farther forward. They prospectively studied 40 patients with recent myocardial infarction (mean 8 days earlier) with invasive coronary angiography, CT coronary angiography, coronary calcium scoring, and cardiac gated PET-CT with 18F-NaF and 18F-fl uorodeoxyglucose (18F-FDG). Using invasive coronary angiography as the gold standard for determining the culprit plaque, the area of greatest 18F-NaF uptake in the coronary arteries localised the plaque in 37 of 40 patients (maximum tissue-to-background ratio in the culprit plaque 1·66 [1·40–2·25] vs highest non-culprit plaque 1·24 [1·06–1·38]). By contrast, interpretation of 18F-FDG PET-CT images in the same cohort was technically diffi cult because of the frequent overlap of myocardial 18F-FDG uptake with the adjacent coronary arteries. Of the 55% of vascular territories that were interpretable by 18F-FDG, only a weak correlation was seen with culprit plaque identifi cation. A second cohort of 40 patients with stable angina underwent the same imaging tests and an intracoronary ultrasound. 18 patients had one or more plaques with high 18F-NaF uptake, defi ned as at least 25% greater than a proximal reference lesion. Intracoronary ultrasound identifi ed that microcalcifi cation, necrotic core size, and positive remodelling correlated strongly with plaques of high 18F-NaF activity. Histological correlation was assessed in a third cohort of nine patients who underwent carotid endarterectomy at a mean of 17 days after clinical symptoms. Ex-vivo PET-CT was done on the removed carotid atherosclerotic tissue. Macroscopic plaque rupture was present in each patient, all localised to areas of high 18F-NaF uptake. Plaques with increased 18F-NaF uptake had substantially larger necrotic cores, more cell death and macrophage infi ltration, and, as measured by alkaline phosphatase and osteocalcin staining, more active calcifi cation than those that did not. With the strong in-vivo correlates of coronary plaque rupture seen on intracoronary ultrasound in patients with stable angina, and histological confi rmation of A new frontier in atherosclerotic coronary imaging which might or might not be associated with use of acetylcysteine, it could be prudent in this specifi c clinical setting to avoid use of ondansetron, because other antiemetics are available.


Hepatology | 2018

Extracellular vesicles from mice with alcoholic liver disease carry a distinct protein cargo and induce macrophage activation through heat shock protein 90

Banishree Saha; Fatemeh Momen‐Heravi; Istvan Furi; Karen Kodys; Donna Catalano; Anwesha Gangopadhyay; Reka A. Haraszti; Abhishek Satishchandran; Arvin Iracheta‐Vellve; Adeyinka C Adejumo; Scott A. Shaffer; Gyongyi Szabo

A salient feature of alcoholic liver disease (ALD) is Kupffer cell (KC) activation and recruitment of inflammatory monocytes and macrophages (MØs). These key cellular events of ALD pathogenesis may be mediated by extracellular vesicles (EVs). EVs transfer biomaterials, including proteins and microRNAs, and have recently emerged as important effectors of intercellular communication. We hypothesized that circulating EVs from mice with ALD have a protein cargo characteristic of the disease and mediate biological effects by activating immune cells. The total number of circulating EVs was increased in mice with ALD compared to pair‐fed controls. Mass spectrometric analysis of circulating EVs revealed a distinct signature for proteins involved in inflammatory responses, cellular development, and cellular movement between ALD EVs and control EVs. We also identified uniquely important proteins in ALD EVs that were not present in control EVs. When ALD EVs were injected intravenously into alcohol‐naive mice, we found evidence of uptake of ALD EVs in recipient livers in hepatocytes and MØs. Hepatocytes isolated from mice after transfer of ALD EVs, but not control EVs, showed increased monocyte chemoattractant protein 1 mRNA and protein expression, suggesting a biological effect of ALD EVs. Compared to control EV recipient mice, ALD EV recipient mice had increased numbers of F4/80hi cluster of differentiation 11b (CD11b)lo KCs and increased percentages of tumor necrosis factor alpha–positive/interleukin 12/23–positive (inflammatory/M1) KCs and infiltrating monocytes (F4/80intCD11bhi), while the percentage of CD206+CD163+ (anti‐inflammatory/M2) KCs was decreased. In vitro, ALD EVs increased tumor necrosis factor alpha and interleukin‐1β production in MØs and reduced CD163 and CD206 expression. We identified heat shock protein 90 in ALD EVs as the mediator of ALD‐EV‐induced MØ activation. Conclusion: Our study indicates a specific protein signature of ALD EVs and demonstrates a functional role of circulating EVs containing heat shock protein 90 in mediating KC/MØ activation in the liver. (Hepatology 2018;67:1986‐2000).


Nucleic Acids Research | 2018

Comparison of partially and fully chemically-modified siRNA in conjugate-mediated delivery in vivo

Matthew R. Hassler; Anton A. Turanov; Julia F. Alterman; Reka A. Haraszti; Andrew H. Coles; Maire F. Osborn; Dimas Echeverria; Mehran Nikan; William Salomon; Loic Roux; Bruno M.D.C. Godinho; Sarah M. Davis; David V. Morrissey; Phillip D. Zamore; S. Ananth Karumanchi; Melissa J. Moore; Neil Aronin; Anastasia Khvorova

Abstract Small interfering RNA (siRNA)-based drugs require chemical modifications or formulation to promote stability, minimize innate immunity, and enable delivery to target tissues. Partially modified siRNAs (up to 70% of the nucleotides) provide significant stabilization in vitro and are commercially available; thus are commonly used to evaluate efficacy of bio-conjugates for in vivo delivery. In contrast, most clinically-advanced non-formulated compounds, using conjugation as a delivery strategy, are fully chemically modified (100% of nucleotides). Here, we compare partially and fully chemically modified siRNAs in conjugate mediated delivery. We show that fully modified siRNAs are retained at 100x greater levels in various tissues, independently of the nature of the conjugate or siRNA sequence, and support productive mRNA silencing. Thus, fully chemically stabilized siRNAs may provide a better platform to identify novel moieties (peptides, aptamers, small molecules) for targeted RNAi delivery.


bioRxiv | 2018

Hydrophobicity drives the systemic distribution of lipid-conjugated siRNAs via lipid transport pathways

Maire F. Osborn; Andrew H. Coles; Annabelle Biscans; Reka A. Haraszti; Loic Roux; Sarah M. Davis; Socheata Ly; Dimas Echeverria; Matthew R. Hassler; Bruno M.D.C. Godinho; Mehran Nikan; Anastasia Khvorova

Efficient delivery of therapeutic RNA is the fundamental obstacle preventing its clinical utility. Lipid conjugation improves plasma half-life, tissue accumulation, and cellular uptake of small interfering RNAs (siRNAs). However, the impact of conjugate structure and hydrophobicity on siRNA pharmacokinetics is unclear, impeding the design of clinically relevant lipid-siRNAs. Using a panel of biologically-occurring lipids, we show that lipid conjugation modulates siRNA hydrophobicity and governs spontaneous partitioning into distinct plasma lipoprotein classes in vivo. Lipoprotein binding influences siRNA distribution by delaying renal excretion and promoting uptake into lipoprotein receptor-enriched tissues. Lipid-siRNAs elicit mRNA silencing without causing toxicity in a tissue-specific manner. Lipid-siRNA internalization occurs independently of lipoprotein endocytosis, and is mediated by siRNA phosphorothioate modifications. Although biomimetic lipoprotein nanoparticles have been considered for the enhancement of siRNA delivery, our findings suggest that hydrophobic modifications can be leveraged to incorporate therapeutic siRNA into endogenous lipid transport pathways without the requirement for synthetic formulation.


Journal of Drug Delivery Science and Technology | 2018

Chitosan-Mangafodipir nanoparticles designed for intranasal delivery of siRNA and DNA to brain

Juan Sanchez-Ramos; Shijie Song; Xiaoyuan Kong; Parastou Foroutan; Gary V. Martinez; William Dominguez-Viqueria; Shyam S. Mohapatra; Subhra Mohapatra; Reka A. Haraszti; Anastasia Khvorova; Neil Aronin; Vasyl Sava

The overall objective of the present research was to develop a nanocarrier system for non-invasive delivery to brain of molecules useful for gene therapy. Manganese-containing nanoparticles (mNPs) carrying anti-eGFP siRNA were tested in cell cultures of eGFP-expressing cell line of mouse fibroblasts (NIH3T3). The optimal mNPs were then tested in vivo in mice. Following intranasal instillation, mNPs were visualized by 7T MRI throughout brain at 24 and 48 hrs. mNPs were effective in significantly reducing GFP mRNA expression in Tg GFP+ mice in olfactory bulb, striatum, hippocampus and cortex. Intranasal instillation of mNPS loaded with dsDNA encoding RFP also resulted in expression of the RFP in multiple brain regions. In conclusion, mNPs carrying siRNA, or dsDNA were capable of delivering the payload from nose to brain. This approach for delivery of gene therapies to humans, if successful, will have a significant impact on disease-modifying therapeutics of neurodegenerative diseases.

Collaboration


Dive into the Reka A. Haraszti's collaboration.

Top Co-Authors

Avatar

Anastasia Khvorova

University of Massachusetts Amherst

View shared research outputs
Top Co-Authors

Avatar

Neil Aronin

University of Massachusetts Boston

View shared research outputs
Top Co-Authors

Avatar

Andrew H. Coles

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar

Dimas Echeverria

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar

Bruno M.D.C. Godinho

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Julia F. Alterman

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Marie-Cecile Didiot

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar

Mehran Nikan

University of Massachusetts Medical School

View shared research outputs
Researchain Logo
Decentralizing Knowledge