Skip to main content

Home/ WSU Virology/ Group items tagged gene therapy

Rss Feed Group items tagged

sdahlseng10

In Vivo Gene Delivery and Stable Transduction of Nondividing Cells by a Lentiviral Vector - 0 views

  •  
    Supplemental Article to Lesch et al. (2011); Journal club 1: This article provides interesting support to the effectiveness of lentivirus vectors for gene therapy treatment. Not only is HIV or lentivirus efficient at transduction to rapidly dividing cells, it is also capable of transduction to nondividing cells; making it an exciting vector for gene therapy treatment. The range of diseases that could be treatable with a gene therapy vector that can target other cell types than hematopoietic cells could transform the science of gene therapy.
slgoogin8981

HSV carrying WT REST establishes latency but reactivates only if the synthesis of REST ... - 7 views

  • R111 recombinant is not reactivation-defective because it is able to reactivate in the presence of inhibitors of protein synthesis in the same manner as the WT parent virus, and (b) because the only significant difference in the WT and R111 viruses is the presence of the REST gene in the latter virus, the data suggest that expression of this gene blocks reactivation and that suppression of protein synthesis, including that of REST, enables reactivation.
    • becky214
       
      I am confused about when REST blocks reactivation and when it enables it. It says the gene blocks reactivation but continues to say it enables it. Does REST only enable reactivation if it plays a role in suppressing protein synthesis?
  • with dexamethasone or dexamethasone and cycloheximide
  • Specifically, a stress response generated by virus entry recruits or activates REST to enable the assembly of the HCLR complex. Stress responses have been postulated to activate REST
    • becky214
       
      I think this is interesting because REST is not usually found in neurons. I'm curious as to how the neuron recruits this gene. Does the virus actually recruit it? Or is it just a cellular response to the stress of viral entry? 
  • ...13 more annotations...
  • HSV takes advantage of the neuronal stress response to enter into a silent, latent state. To assist in the execution of this plan, HSV evolved a DNA sequence that allows itself to be suppressed in neurons and a mechanism to maintain an equilibrium between total suppression and potential to exit from the latent state.
  • translocated from satellite cells
  • The fundamental question is the identity of the mechanism by which a vigorously replicating virus, on entry into the body, is silenced in neurons harboring latent virus.
    • laceemarie
       
      Just to make sure that I understand this, once the virus enters the body, it quickly replicates and all the new virus particles find a nerve cell to infect and once there, the virus is able to sit in silence, so to speak?
  • Thus, VP16, a virion protein brought into the cells during infection, recruits several cellular proteins, including LSD1, to derepress α gene promoters
    • laceemarie
       
      VP16, a virion protein and a recruiter - this protein sounds pretty important to me. Is there a way or has there been work done with this protein to not allow the derepression at this checkpoint? Is it possible to keep a virus in latency because of alpha gene promoters not getting derepressed so as to not allow the virus to infect the host? I'm not sure if that's a reasonable antiviral therapy or not.
  • In some neurons, the virus establishes a latent, silent state. In other neurons, the virus replicates, and it is most likely that the virus in these neurons is transmitted and replicates in other ganglionic cells.
    • laceemarie
       
      Is this a random event or does this have something to do with the environment of the neuron? I would assume that specific, different environments would be ideal for each.
  • Finally, in contrast to the events following entry of virus after retrograde transport from the periphery, the data suggest that reactivation does not trigger a stress response that leads to activation of REST
    • alexridesducati
       
      Has anyone been able to pinpoint the exact step in infection that activates REST so that it can be studied? If so, perhaps it is possible to manipulate the effects of that step in order to induce an artificial response to the reactivation of HSV1 from its latency period in order to retrigger the stress response that leads to REST activation.
  • Thus, VP16, a virion protein brought into the cells during infection, recruits several cellular proteins, including LSD1, to derepress α gene promoters. One α gene product, infected cell protein 0 (ICP0), derepresses β and γ1 genes. Ultimately, the onset of viral DNA synthesis enables the expression of very late, or γ2, genes (4).
    • Sean Hogan
       
      Is this why the latent infection occurs in the ganglia of the PNS? The necessary proteins for gene derepression can't be recruited in the CNS or other cells?
  • In contrast, simultaneous expression of all viral genes during reactivation from latency is likely to minimize yield, but the mission of the virus is to assemble enough viruses to reach the portal of egress from the body (e.g., mouth, genitals) rather than to overwhelm the host with infectious virus.
    • Sean Hogan
       
      This might be the coolest statement in the whole paper. The discussion above kind of painted a picture of HSV infecting the PNS only but the reason for it's inability to reach the CNS didn't receive as much attention. I think this statement summed it all up though.HSV doesn't care about high virion yields or whether a productive or latent infection is necessary, it just wants to reach the body portals. The virus is "smart," enough to avoid the CNS and keep it's host alive.
  • Between 5 and 24 h after excision, mRNAs representative of all viral gene kinetic groups increase 100-fold in amount. Viral DNA also increases in amount, indicating that viral proteins are made. At the same time, viral LAT and miRNA concentrations decrease at least 10-fold (34). It is convenient to define the initial phase lasting no more than 5 h as the preactivation phase and the remaining time interval as the activation phase.
    • rmeloche10
       
      I'm having trouble grasping why the massive disparity between viral DNA and viral LAT. Obviously there would be some disparity when reactivation occurs, but wouldn't the production of more DNA contribute to even a small amount of LAT production and not a minimal 10 fold decrease?
  • The same concentration of HDAC inhibitor was ineffective in inducing the reactivation of R111 recombinant virus in ganglionic organ cultures maintained in medium containing anti-NGF antibody.
    • ameliaobert
       
      Interesting: That there is an inhibitor for chromatin remodelling (HDAC). Confusing: Is if HDAC can inhibit properly for WT virus, but not for R111 recombinant, that obtains REST. How does REST make the HDAC inhibitor ineffective ia stressed neuron? I understand that REST is what the DNA can be wrapped about and help with latency, so it that why the HDAC cannot inhibit, since REST is already aiding in latency.
    • nleonard11
       
      I thought this process of finding out that the HCLR complex activates from stress was very interesting. Using WT viral genomes appears to be a very effective way to test many virus functions.
    • nleonard11
       
      I was just wondering why the virus goes into a latent after 30 days? What exactly is it waiting to do and what conditions need to be present for it to become active again.
    • slgoogin8981
       
      It would be interesting to know the amount that REST is seen in non-neural cells and nerve cells in the absents of HSV-1. I was state earlier that REST is not normally found in never cells.
sdahlseng10

Improved lentiviral vectors for Wiskott-Aldrich syndrome gene therapy mimic... - 0 views

  •  
    Supplemental Article to Lesch et al. (2011): This article talks about one of the disorders that is treatable by gene therapy: a relatively rare x-linked chromosomal abnormality that causes immunodeficiency as a result of hematopoietic cell damage causing inefficient T-cell receptor binding. Lentiviruses appear to be good vectors for the gene therapy treatment of this disorder as a highly regulated therapy is required for a beneficial outcome.
Sean Hogan

PLOS Pathogens: Different Modes of Retrovirus Restriction by Human APOBEC3A and APOBEC3... - 22 views

  • One such family of restriction factors is the apolipoprotein B editing complex 3 (A3) cellular cytidine deaminases (CDA). While A3 genes are found in all mammals, their number differs from species to species. For example, humans have 7 A3 genes (A3A to A3H) while mice have only one gene. All proteins in this family contain at least one CDA domain that deaminates carbon 4 of cytidine in single-stranded DNA, resulting in a uracil that causes G to A transitions in the opposing strand [3].
    • alexridesducati
       
      Can these genes be exploited for antiviral therapy and if so, can it be done without harm to the host due to mutations?
  • viral cDNA accumulation
  • Packaging of A3G into virions is counteracted by HIV Vif (viral infectivity factor) protein. In virus-producer cells, Vif binds to A3G as well other A3 family members, and recruits cellular E3 ubiquitin ligase complexes, leading to ubiquitination and subsequent proteasomal degradation, thereby preventing packaging of A3G into budding virions [12]–[14]. Lentiviral Vif proteins show strong species-specificity. For example, HIV-1 Vif counteracts human A3G but only certain simian A3G homologues [15], [16]; it also does not interact with mouse A3 [17].
  • ...12 more annotations...
  • Other members of the A3 family are believed to affect other exogenous viruses as well as endogenous retrovirus/retroelement movement within the genome. In particular, human A3A is a potent inhibitor of IAP and MusD and other retrotransposons such as LINE-1 and this inhibition is CDA-independent, at least in cultured cells [18]–[20]. A3A also inhibits adeno-associated virus replication, a nuclear-replicating parvovirus, via CDA-independent means [20]. In monocytes, A3A restricts HIV-1 infection and the decrease in A3A levels that occurs during monocyte-to-macrophage development is concomitant with increased susceptibility to HIV-1 infection [21]. A3A is not packaged into HIV virions and is thought to restrict infection by targeting incoming virus [22]–[24]. In contrast, A3A is packaged in human T-lymphotropic virus type-I virions and restricts infection, at least in transfected cells [25]. A3A preferentially deaminates cytidines that are in a TC motif [26].
  • Different A3 family members block infection by diverse retroviruses from different species, including HIV-2 [27], porcine endogenous retrovirus [28], [29], xenotropic, Friend (F-MLV) and Moloney murine leukemia virus (M-MLV) [30]–[32] and mouse mammary tumor virus (MMTV) [33]. Additionally, A3 proteins may restrict other virus families, including parvoviruses [20], [34], hepatitis B virus [35]–[37], papillomaviruses [38] and herpes simplex virus I [39]. Thus, it has been suggested that A3 proteins exist, at least in part, to prevent zoonotic transmission of viruses [40].
  • Here, we show that transgenic mice expressing the human A3A or A3G proteins restrict murine retrovirus infection in vivo in disparate ways. A3G was packaged into virions in vivo, leading to the deamination of both MLV and MMTV viral genomes. In contrast, A3A was not packaged, and appeared to restrict infection in a largely CDA-independent manner. Finally, we show that Vif/A3G interactions can be studied in this in vivo model, thus providing a potentially useful system for the analysis of small molecule inhibitors of A3 proteins and Vif.
  • To determine the level of transgene expression, we first isolated RNA from different tissues, including peripheral blood mononuclear cells (PBMCs), and performed reverse-transcribed real-time quantitative PCR (RT-qPCR). RNA from human H9 cultured cells and human and C57BL/6 mouse PBMCs served as controls. For each transgene, there was one high- (A3Ghigh, A3Ahigh) and one low- (A3Glow, A3Alow) expressing strain, defined by their relative expression in lymphoid tissues. The A3Ghigh strain expressed higher levels of the transgene than the endogenous mouse gene in spleen and thymus, but similar A3G levels in mouse and human PBMCs, while the A3Glow strain expressed approximately 10-fold lower levels in these tissues (Figure 1A). In contrast, the A3Ahigh strain expressed similar or lower levels than mouse A3; there was also about 2-fold lower expression of A3A in mouse PBMCs than in human PBMCs (Figure 1B). The A3Alow strain had very low but detectable levels of expression in several tissues. Since the β-actin regulatory region was used, transgene expression was seen in many tissues and in several at levels higher than endogenous mouse A3 (e.g. heart, brain and liver) (Figure 1A and 1B). We also performed western blots on different tissues from the 4 different mouse strains, using antiserum that detects both A3A and A3G. The relative protein expression levels were similar to that seen at the RNA level (Figure S1A and S1B).
  • We next determined if the in vivo-produced A3A and A3G proteins were functionally active. Extracts were prepared from primary splenocyte cultures and equal amounts (total protein concentration/volume) were incubated with FAM-labeled substrates containing the A3A- or A3G-preferred target sequence (S50-TTC and S50-CCC, respectively). As controls, we also performed these assays with extracts prepared from 293T cell lines transfected with A3A or A3G. Activity could be readily detected in transgenic mice expressing high levels of A3A or A3G. Further, in accord with the known specificity of the cytidine deaminases, extracts from the A3Ahigh mice deaminated the TTC- more efficiently than CCC-containing substrates, while those from A3Ghigh mice more efficiently deaminated the CCC substrate (Figure 2). For both A3Alow and A3G low, trace amounts of activity were detectable with the preferred substrates, while no activity was detectable with either endogenous mA3 or from mA3 knockout splenocytes. No deaminase activity was detected with WT mouse extracts, perhaps because the mouse protein has lower overall activity or expression. These data show that the transgenic mice expressed catalytically active human deaminases in these heterologous cells.
  • Humans have 7 APOBEC3 genes and determining how each specifically functions to inhibit retroviruses like HIV is complicated, because all 7 can be produced in a given cell type or tissue.
    • laceemarie
       
      What cell/tissue type(s) are these APOBEC3 genes naturally turned on in? 
  • To overcome this limitation, we made transgenic mice that express two of the human proteins, APOBEC3A and APOBEC3G in mice that do not express their own APOBEC3. These mice were able to effectively block infection by several mouse retroviruses
    • laceemarie
       
      What cell type(s) did they use? Does it matter which?
  • We were unable to perform similar assays with in vivo produced MMTV, because the only cell-free virus in mice is found in milk and mammary tumors and we have not yet established breeding colonies of virus-infected human A3 transgenic mice.
    • laceemarie
       
      Was this a screw up, or is it not that important to look at assays with in vivo produced MMTV. And by "not yet," does that mean they are going to? I feel like if your going to use this virus in your experimental studies, you should figure out ways to perform the assays, regardless of how you get the virus. It would appear that they knew this information before hand, so maybe an assay on MMTV is less relevant. 
    • laceemarie
       
      *you're
  • A3G and A3A inhibit retrovirus infection by different means.
    • becky214
       
      Since they use different means of inhibition, do they work together to prevent infection? Or is an and either/or type scenario?
  • Two A3A and A3G mouse strains each were generated, expressing levels of these proteins within the range or at levels lower than that seen in human cells. This likely has relevance to what occurs in individual humans, where non-coding region polymorphisms in A3 genes alter expression levels and may influence progression to HIV-induced disease
    • laceemarie
       
      Could this have something to do with how HIV works in the HIV controllers? Where they still exhibit virus particles, but at a lower amount, don't necessarily spread the virus as much, and don't exhibit as intense of HIV symptoms?
    • ameliaobert
       
      If A3A is less clear, but does not get packaged, where must the A3A involvement with the incoming virus be located (for myeloid cells)? As well as if it is known to use retroelement retotransposition and replication inhibiton in paraoviruses by nh2 to oh independent means, how does the A3A know to be signlle to change the structure of cystine?
  • An additional limitation of previous studies done on human A3 proteins is the reliance on transfecting constructs expressing A3 proteins, which may not reflect the endogenous levels of a protein expression found in vivo
    • Sean Hogan
       
      Couldn't they mimic the in vivo environment by overexpressing necessary proteins in the host?
  •  
    Focus paper for retrovirus presentation.
  •  
    This will be the focus paper for 11/14.
sdahlseng10

Replication-competent Lentivirus Analysis of Clinical Grade Vector Products - 0 views

  •  
    Supplementary Article to Lesch et al. (2011); journal club 1: This article helps us understand the dangers of attenuated lentiviruses used for gene therapy and other clinical applications. It also details a method for screening newly synthesized lentiviruses for replication competency.
sdahlseng10

Production and purification of lentiviral vectors generated in 293T suspens... - 6 views

  •  
    Focus Article; Journal club 1:This is an article that details a novel technique for production of HIV (lentiviral) vectors for use as tools of gene therapy. The fascinating approach that was developed by the authors uses baculovirus as a vector to infect human embryonic kidney cells in culture with the genes necessary to produce a lentiviral vector with therapeutic capabilities.
Casey Finnerty

Research - A Neuro-Oncology Laboratory at Northwestern University in Chicago - 1 views

  • Most adenoviruses that have been historically used for gene therapy have been based on serotype 5 (AdWT). Unfortunately, expression of the primary receptor for Ad5 (the coxsackie-adenovirus receptor, CAR) is highly variable on cancer cells. In fact, several studies have demonstrated a resistance of malignant glioma to adenoviral vectors, a finding that was subsequently attributed to the quantitative deficiency of CAR on brain tumor cells.
  • First, we tested a variety of tumor specific promoters and identified survivin (S) as an excellent tumor specific promoter for transcriptional control of E1a, a gene essential for CRAd replication (J Neurosurg 104:583, 2006; Cancer Biol Ther 6:679, 2007).
  • Based on the above date data, we then created a novel oncolytic adenoviral vector which utilizes the survivin promoter and binds to heparan sulfate proteoglycans expressed on malignant brain tumors and named this new vector CRAd-S-pk7 (Hum Gene Ther 18:589, 2007).
    • Casey Finnerty
       
      How specific is the binding/tropism? HS is fairly widespread throughout the body.
  • ...6 more annotations...
  • Second, we have identified several receptors that are over-expressed on brain tumor cells and created a series of pseudotyped Ad5 vectors that recognize these receptors
  • Our studies with CRAd-S-pk7 indicate that the virus provides the highest level of viral replication and tumor oncolysis in glioma cell lines. At the same time, we observed minimal viral replication and toxicity in normal human brain. Injection of CRAd-S-pk7 inhibited xenograft brain tumor growth by more than 300%.
  • We were the first group in the country to show that human mesenchymal stem cells can be effectively loaded with a replication competent virus and effectively deliver it to an experimental glioma model
  • When oncolytic vectors are loaded onto stem cells, the virus effectively "hides" from the immune system for an extended period of time. The ability of stem cells to suppress the anti-viral immune response in a permissive and tumor-bearing animal model is the subject of one of our latest manuscript
  • Finally, to further enhance the therapeutic efficacy of stem cells, we have optimized them to specifically traffic to intracranial tumors via genetic modification with single-chain antibodies against antigens expressed on gliomas
  • Our latest work in this area supports the development of neural stem cell based cell carriers for oncolytic virotherapy
Haram LEE

BMC Cancer | Full text | Oncolytic Targeting of Androgen-sensitive Prostate Tumor by th... - 4 views

  •  
    Oncolytic virotherapy for cancer treatment utilizes viruses for selective infection and death of cancer cells without any adverse effect on normal cells. We previously reported that the human respiratory syncytial virus (RSV) is a novel oncolytic virus against androgen-independent PC-3 human prostate cancer cells.
  • ...2 more comments...
  •  
    Is there any other virus can using for Oncolytic virotherapy? - Oncolytic viruses identified to date are: adenovirus, reovirus, herpes simplex virus (HSV), Newcastle disease virus (NDV), vaccinia virus, myxoma virus, influenza virus, measles virus, coxsackievirus and vesicular stomatitis virus (VSV) (Anticancer oncolytic activity of respiratory syncytial virus., http://www.ncbi.nlm.nih.gov/pubmed?term=Anti-cancer%20oncolytic%20activity%20of%20respiratory%20syncytial%20virus)
  •  
    Why also using xenograft, not only for cell-culture method? - A human prostate tumor xenograft model (30) was used to examine the oncolytic function of RSV in vivo (Figure 2). -We also investigated the efficacy of intraperitoneally (I.P) delivered RSV for causing tumor regression and determined that intraperitoneally injected RSV also rendered significant reduction in the tumor growth compared to the growth of control, medium-treated tumors (Figure 2c). The significant tumor regression by intraperitoneally delivered RSV is shown in Figure 2d. Similar results were obtained with tumors grown in the dorsal flank (Supplementary Figure S2). Therefore, the RSV-responsive restriction of tumor growth at two sites (ear and flank) demonstrates the versatility of RSV in conferring oncolysis in vivo at different anatomical regions. (Anticancer oncolytic activity of respiratory syncytial virus., http://www.ncbi.nlm.nih.gov/pubmed?term=Anti-cancer%20oncolytic%20activity%20of%20respiratory%20syncytial%20virus)
  •  
    How Oncolytic virus control the inflammation? - Oncolytic virus treatment induced at least a twofold increase or decrease in the expression of 50 genes relative to expression in the PBS-treated tumors (Supplementary Table 1, available online). Of these 50 genes, 48 displayed an increase in expression in the oncolytic virus - treated tumors compared with the controltreated tumors, suggesting that oncolytic virus treatment induced an inflammatory response - To confirm the role of the immune response in oncolytic virus - induced vascular hyperpermeability, we evaluated changes in oncolytic virus - induced vascular leakage in tumor-bearing rats that had been treated with cyclophosphamide before oncolytic virus injection. In addition to its immunosuppressive effects, cyclophosphamide blocks infl ammation and reduces viral clearance, both of which increase the propagation of oncolytic viruses, thereby enhancing therapeutic effi cacy of oncolytic viruses. (Effect of Tumor Microenvironment Modulation on the Efficacy of Oncolytic Virus Therapy, http://www.ncbi.nlm.nih.gov/pubmed?term=Effect%20of%20Tumor%20Microenvironment%20Modulation%20on%20the%20Efficacy%20of%20Oncolytic%20Virus%20Therapy)
Trevor F

Unique molecular signatures of disease brain endothelia provide a novel site for CNS-di... - 5 views

  •  
    Great paper for introductory look at adapting adeno-associated viruses (AAV) to different cell tropisms.  Although the scope of this article is the CNS, and more specifically the brain.  Also goes over how brain vasculature expresses different characteristics in disease states that allows for specification of AAVs to have tropism for the diseased epithelial beds.
sdahlseng10

Possible applications for replicating HIV 1 vectors - 0 views

  •  
    Review Article; Journal club 1: This is a basic review article about HIV virus vectors and their potential uses in clinical science.
1 - 11 of 11
Showing 20 items per page