Daniel E. Strongin’s research while affiliated with University of Washington and other places

What is this page?


This page lists works of an author who doesn't have a ResearchGate profile or hasn't added the works to their profile yet. It is automatically generated from public (personal) data to further our legitimate goal of comprehensive and accurate scientific recordkeeping. If you are this author and want this page removed, please let us know.

Publications (6)


Design of a viral gene drive targeting HSV-1 UL37-38 region
a Gene drive viruses carry Cas9 and a gRNA targeting the same location in a wild-type genome. After coinfection of cells by wild-type (WT) and gene drive (GD) viruses, Cas9 cleaves the wild-type sequence and homology-directed repair –using the gene drive sequence as a repair template– causes the conversion of the wild-type locus into a new gene drive sequence and the formation of new recombinant gene drive viruses (rGD). Artwork was modified from ref. ¹¹, ¹². b Modified and unmodified UL37-38 region. The gene drive cassette was inserted between the UL37 and UL38 viral genes and was composed of spCas9 under the control of a CBH promoter followed by the SV40 polyA signal, a CMV promoter driving an mCherry reporter, followed by the beta-globin polyA signal, and a U6-driven gRNA. c Localizations of the gene drive sequence and YFP/CFP reporters on HSV-1 genomes. GD represents a functional gene drive virus, GD-ns carries a non-specific gRNA, and Cas9 is deleted in GD-ΔCas9. UL/US: unique long/short genome segments. d, e Recombination products and examples of viral plaques after cellular co-infection with HSV1-WT expressing YFP and gene drive viruses expressing mCherry and CFP. Representative images from more than n > 10 experiments. Scale bars: 100 μm.
Gene drive spread in cell culture
a Viral titers in the supernatant after infection of N2a cells with WT, GD, GD-ns or GD-ΔCas9. Cells were infected with a single virus at MOI = 1. n = 4. b, c Viral titers in the supernatant after co-infection of N2a cells with WT + GD, WT + GD-ns or WT + GD-ΔCas9, with a starting proportion of gene drive virus of 20% (b) or 40% (c). MOI = 1, n = 4. d–g Evolution of the viral population after co-infection with WT + GD, WT + GD-ns or WT + GD-ΔCas9, with a starting proportion of gene drive virus of 20% (d, e) or 40% (f, g). Panels (d and f) show the proportion of viruses expressing mCherry, representing gene drive virus. Panels (e and g) show the proportion of viruses expressing the different fluorophore combinations. Viral titers are expressed in log-transformed PFU (plaque-forming unit) per mL of supernatant. Error bars represent the standard error of the mean (SEM) between biological replicates. n = 4. Source data are provided as a Source Data file.
Gene drive spread during herpes simplex encephalitis
a Infection routes along the optic, oculomotor and trigeminal nerves (cranial nerves II, III and V, respectively) following ocular inoculation of HSV-1. Male and female Balb/c mice were infected with 10⁶ PFU in the left eye. b, c Viral titers after four days in the eye, TG and whole brain after (b) infection with a single virus, n = 5 mice, or (c) with a starting proportion of gene drive virus of 15%, n = 6 mice. d, e Viral population in the eye, TG and whole brain after co-infection with WT + GD or WT + GD-ns, after four days. n = 6. f Proportion of viral genomes with a mutated target site in the brain after four days. n = 3. g Viral titers in the spinal cord and brain after inoculation of WT, WT + GD, or WT + GD-ns in the right hind leg footpad, after 5–7 days. n = 8 for WT and WT + GD, n = 4 for WT + GD-ns. h, i Viral population in the spinal cord and whole brain after co-infection with WT + GD or WT + GD-ns, after 5–7 days. n = 5 for WT + GD, n = 1 for WT + GD-ns. Viral titers are expressed in log-transformed PFU. In panels (b, c and g) black lines indicate the median. n.d.: non-detected. Panels (d, e, f, h, and i) show the average and SEM between biological replicates. Source data are provided as a Source Data file.
High heterogeneity between brain regions during gene drive spread
a Infection routes following ocular inoculation of HSV-1. Male and female Balb/c mice were co-infected with 10⁶ PFU of WT + GD in the left eye, with a starting proportion of gene drive virus of 15%. b Viral titers over time. Black lines indicate the median. n.d.: non-detected. c, d Proportion of gene drive viruses over time. Data show the average and SEM between biological replicates. e Heatmap summarizing panels (b and c). n = 4 mice for day 2 and 3, n = 6 mice for day 4. Source data are provided as a Source Data file.
High levels of co-infection in the TG during HSV-1 infection
a Balb/c mice were co-infected with equivalent amounts of three viruses expressing YFP, CFP and RFP, respectively, with a total of 10⁶ PFU in the left eye. b. YFP and CFP cellular intensity after machine learning-assisted cell segmentation of TG sections. Datapoints represent individual cells and were colored by converting YFP and CFP signals into the CYMK color space. 4035 cells were detected, originating from 53 images and n = 4 mice. c Percentage of infected cells expressing YFP, CFP, or both. n = 4 mice. d Percentage of infected cells expressing one or two fluorescent markers. n = 4 mice. e, f Representative images of TG sections from four biological replicates, highlighting high levels of co-infection. Arrows indicate cells co-expressing YFP, CFP and RFP together. Scale bars: 100 μm. Panels (c and d) show the average and standard deviation (SD) between biological replicates. Source data are provided in Supplementary data 2 and as a Source Data file.

+4

Viral gene drive spread during herpes simplex virus 1 infection in mice
  • Article
  • Full-text available

September 2024

·

112 Reads

·

3 Citations

·

Anoria K. Haick

·

·

[...]

·

Gene drives are genetic modifications designed to propagate efficiently through a population. Most applications rely on homologous recombination during sexual reproduction in diploid organisms such as insects, but we recently developed a gene drive in herpesviruses that relies on co-infection of cells by wild-type and engineered viruses. Here, we report on a viral gene drive against human herpes simplex virus 1 (HSV-1) and show that it propagates efficiently in cell culture and during HSV-1 infection in mice. We describe high levels of co-infection and gene drive-mediated recombination in neuronal tissues during herpes encephalitis as the infection progresses from the site of inoculation to the peripheral and central nervous systems. In addition, we show evidence that a superinfecting gene drive virus could recombine with wild-type viruses during latent infection. These findings indicate that HSV-1 achieves high rates of co-infection and recombination during viral infection, a phenomenon that is currently underappreciated. Overall, this study shows that a viral gene drive could spread in vivo during HSV-1 infection, paving the way toward therapeutic applications.

Download

Gene editing for latent herpes simplex virus infection reduces viral load and shedding in vivo

May 2024

·

142 Reads

·

9 Citations

Anti-HSV therapies are only suppressive because they do not eliminate latent HSV present in ganglionic neurons, the source of recurrent disease. We have developed a potentially curative approach against HSV infection, based on gene editing using HSV-specific meganucleases delivered by adeno-associated virus (AAV) vectors. Gene editing performed with two anti-HSV-1 meganucleases delivered by a combination of AAV9, AAV-Dj/8, and AAV-Rh10 can eliminate 90% or more of latent HSV DNA in mouse models of orofacial infection, and up to 97% of latent HSV DNA in mouse models of genital infection. Using a pharmacological approach to reactivate latent HSV-1, we demonstrate that ganglionic viral load reduction leads to a significant decrease of viral shedding in treated female mice. While therapy is well tolerated, in some instances, we observe hepatotoxicity at high doses and subtle histological evidence of neuronal injury without observable neurological signs or deficits. Simplification of the regimen through use of a single serotype (AAV9) delivering single meganuclease targeting a duplicated region of the HSV genome, dose reduction, and use of a neuron-specific promoter each results in improved tolerability while retaining efficacy. These results reinforce the curative potential of gene editing for HSV disease.


Viral gene drive spread during herpes simplex virus 1 infection in mice

December 2023

·

51 Reads

·

1 Citation

Gene drives are genetic modifications designed to propagate efficiently through a population. Most applications rely on homologous recombination during sexual reproduction in diploid organisms such as insects, but we recently developed a gene drive in herpesviruses that relies on the co-infection of cells by wild-type and engineered viruses. Here, we developed a viral gene drive against human herpes simplex virus 1 (HSV-1) and showed that it propagated efficiently in vitro and during HSV-1 infection in mice. We observed high levels of gene drive-mediated recombination in neuronal tissues as the infection progressed from the site of inoculation to the peripheral and central nervous systems. These findings indicated that HSV-1 achieves high rates of co-infection and recombination during an infection, a phenomenon that is currently underappreciated. Together, this study shows that a viral gene drive can spread in vivo during HSV-1 infection, paving the way toward therapeutic applications.


A multiplexed barcode approach to simultaneously evaluate gene delivery by adeno-associated virus capsid variants in nonhuman primates

January 2023

·

72 Reads

·

1 Citation

Hepatology Communications

Background and aims: Adeno-associated virus (AAV) vectors are widely used to deliver therapeutic transgenes to distinct tissues, including the liver. Vectors based on naturally occurring AAV serotypes as well as vectors using engineered capsids have shown variations in tissue tropism and level of transduction between different mouse models. Moreover, results obtained in rodents frequently lack translatability into large animal studies. In light of the increasing interest in AAV vectors for human gene therapy, an increasing number of studies are being performed in nonhuman primates. To keep animal numbers to a minimum and thus optimize the process of AAV capsid selection, we developed a multiplex barcoding approach to simultaneously evaluate the in vivo vector performance for a set of serotypes and capsid-engineered AAV vectors across multiple organs. Approach and results: Vector biodistribution and transgene expression were assessed by quantitative PCR, quantitative reverse transcription PCR, vector DNA amplicon Illumina sequencing and vRNAseq in male and female rhesus macaques simultaneously dosed with a mixture of barcoded naturally occurring or engineered AAV vectors encoding the same transgene. As expected, our findings show animal-to-animal variation in both the biodistribution and tissue transduction pattern, which was partly influenced by each animal's distinctive serological status. Conclusions: This method offers a robust approach to AAV vector optimization that can be used to identify and validate AAV vectors for gene delivery to potentially any anatomical site or cell type.


AAV-delivered gene editing for latent genital or orofacial herpes simplex virus infection reduces ganglionic viral load and minimizes subsequent viral shedding in mice

September 2022

·

290 Reads

·

1 Citation

Herpes simplex virus (HSV) establishes latency in ganglionic neurons of the peripheral nervous system, from which it can reactivate, causing recurrent disease and possible transmission to a new host. Current anti-HSV therapy does not eliminate latent HSV, and thus is only suppressive rather than curative. We developed a potentially curative approach to latent HSV infection and pathogenesis, based on gene editing using HSV-specific meganucleases delivered by adeno-associated virus (AAV) vectors. Our results demonstrated that a dual meganuclease therapy, composed of two anti-HSV-1 meganucleases delivered by a triple AAV serotype combination (AAV9, AAV-Dj/8, AAV-Rh10), can eliminate up to 97% of latent HSV DNA from ganglia in both ocular and vaginal mouse models of latent HSV infection. Using a novel pharmacological approach to reactivate latent HSV-1 in mice with the bromodomain inhibitor JQ-1, we demonstrated that this reduction in ganglionic viral load leads to a significant reduction of viral shedding from treated vs. control mice, with many treated mice showing no detectable virus shedding. In general, therapy was well tolerated, although dose-ranging studies showed hepatotoxicity at high AAV doses, consistent with previous observations in animals and humans. Also in agreement with previous literature, we observed subtle histological evidence of neuronal injury in some experimental mice, although none of the mice demonstrated observable neurological signs or deficits. These results reinforce the curative potential of gene editing for latent orofacial and genital HSV disease, and provide a framework for additional safety studies before human trials can begin.


Gene editing and elimination of latent herpes simplex virus in vivo

August 2020

·

2,203 Reads

·

62 Citations

We evaluate gene editing of HSV in a well-established mouse model, using adeno-associated virus (AAV)-delivered meganucleases, as a potentially curative approach to treat latent HSV infection. Here we show that AAV-delivered meganucleases, but not CRISPR/Cas9, mediate highly efficient gene editing of HSV, eliminating over 90% of latent virus from superior cervical ganglia. Single-cell RNA sequencing demonstrates that both HSV and individual AAV serotypes are non-randomly distributed among neuronal subsets in ganglia, implying that improved delivery to all neuronal subsets may lead to even more complete elimination of HSV. As predicted, delivery of meganucleases using a triple AAV serotype combination results in the greatest decrease in ganglionic HSV loads. The levels of HSV elimination observed in these studies, if translated to humans, would likely significantly reduce HSV reactivation, shedding, and lesions. Further optimization of meganuclease delivery and activity is likely possible, and may offer a pathway to a cure for HSV infection.

Citations (5)


... Methods based on genetic interference represent starting points for the design of novel antiviral strategies (Tanner et al., 2016), as the targeting of viral proteins would limit the risk to off-targets effects. Very recently, interference based on the co-infection of an engineered virus with the corresponding wt virus was applied to HSV-1 in mice to spread a foreign gene to the wt viruses in the infected cells via CRISPR-mediated homologous recombination (Walter et al., 2024). While our study was based on a simpler dominant-negative approach, the design and use of engineered herpesviruses to combat against themselves was expanded here to a congenital context: dramatic prevention of the postnatal consequences of CMV infection of the fetal brain was obtained by prenatal co-infection assays with a mutant CMV encoding a dominant-negative isoform of viral chemokine RCK3. ...

Reference:

Virus-against-virus dominant-negative interference strategy targeting a viral CC chemokine prevents cytomegalovirus-related neurodevelopmental pathogenesis
Viral gene drive spread during herpes simplex virus 1 infection in mice

... For instance, nanoparticle-based drug delivery systems have shown potential in delivering antiviral medications or immunomodulatory agents directly to the cornea, improving both drug bioavailability and patient outcomes (26). Similarly, gene therapy targeting the reactivation pathways of herpes simplex virus (HSV) could provide long-term solutions to prevent recurrent episodes of HSK (27). Developing these advanced therapeutic strategies could transform the clinical management of HSK, offering patients more effective and personalized treatment options. ...

Gene editing for latent herpes simplex virus infection reduces viral load and shedding in vivo

... While gene drive development has been explored not only in model species such as fly [12][13][14] , mice 15,16 and Arabidopsis 17,18 , as well as non-model organisms like yeast 19 , herpesviruses 20 , and agriculture pests 21 , the efficiency in insect species beyond a few studies in Drosophila 12,13,22 , Aedes 23 and especially Anopheles mosquitoes [24][25][26] has been relatively lower due to varies factors such as low conversion rate, high fitness cost, parental effects and resistant allele formation. Several potential solutions have been proposed to address these issues. ...

Viral gene drive spread during herpes simplex virus 1 infection in mice

... [8][9][10] The meganuclease-based therapeutic approach inhibited HSV-1 replication, disrupted latent HSV-1 reservoirs in the mouse model, and led to a reduction in viral reactivation. 11,12 Recently, Wei and colleagues employed a combinatorial therapeutic approach, using CRISPR-Cas9-mediated gene editing alongside corneal transplantation, to treat patients with herpes keratitis in a clinical trial. 13,14 The Cas9-coding mRNA and the guide RNAs were delivered in lentiviral particles directly into the graft bed of the eye that received a new corneal transplantation. ...

AAV-delivered gene editing for latent genital or orofacial herpes simplex virus infection reduces ganglionic viral load and minimizes subsequent viral shedding in mice

... Recently, the editing of the herpes simplex virus (HSV) genome by virus-specific meganucleases delivered by adeno-associated virus (AAV) vectors could eliminate over 90% of the HSV genome in mouse models [90,91]. These large and highly specific endonucleases possess the ability to recognize and cleave DNA sequences ranging from 18 to 24 base pairs in length, a characteristic that sets them apart from other gene-editing technologies [92]. ...

Gene editing and elimination of latent herpes simplex virus in vivo