scholarly journals Adeno-Associated Virus Type 2 (AAV2) Capsid-Specific Cytotoxic T Lymphocytes Eliminate Only Vector-Transduced Cells Coexpressing the AAV2 Capsid In Vivo

2007 ◽  
Vol 81 (14) ◽  
pp. 7540-7547 ◽  
Author(s):  
Chengwen Li ◽  
Matthew Hirsch ◽  
Aravind Asokan ◽  
Brian Zeithaml ◽  
Hong Ma ◽  
...  

ABSTRACT A recent clinical trial has suggested that recombinant adeno-associated virus (rAAV) vector transduction in humans induces a cytotoxic T-lymphocyte (CTL) response against the AAV2 capsid. To directly address the ability of AAV capsid-specific CTLs to eliminate rAAV-transduced cells in vitro and in vivo in mice, we first demonstrated that AAV2 capsid-specific CTLs could be induced by dendritic cells with endogenous AAV2 capsid expression or pulsed with AAV2 vectors. These CTLs were able to kill a cell line stable for capsid expression in vitro and also in a mouse tumor xenograft model in vivo. Parent colon carcinoma (CT26) cells transduced with a large amount of AAV2 vectors in vitro were also destroyed by these CTLs. To determine the effect of CTLs on the elimination of target cells transduced by AAV2 vectors in vivo, we carried out adoptive transfer experiments. CTLs eliminated liver cells with endogenous AAV2 capsid expression but not liver cells transduced by AAV2 vectors, regardless of the reporter genes. Similar results were obtained for rAAV2 transduction in muscle. Our data strongly suggest that AAV vector-transduced cells are rarely eliminated by AAV2 capsid-specific CTLs in vivo, even though the AAV capsid can induce a CTL response. In conclusion, AAV capsid-specific CTLs do not appear to play a role in elimination of rAAV-transduced cells in a mouse model. In addition, our data suggest that the mouse model may not mimic the immune response noted in humans and additional modification to AAV vectors may be required for further study in order to elicit a similar cellular immune response.

2009 ◽  
Vol 83 (13) ◽  
pp. 6817-6824 ◽  
Author(s):  
Chengwen Li ◽  
Matt Hirsch ◽  
Nina DiPrimio ◽  
Aravind Asokan ◽  
Kevin Goudy ◽  
...  

ABSTRACT A recent clinical trial in patients with hemophilia B has suggested that adeno-associated virus (AAV) capsid-specific cytotoxic T lymphocytes (CTLs) eliminated AAV-transduced hepatocytes and resulted in therapeutic failure. AAV capsids elicit a CTL response in animal models; however, these capsid-specific CTLs fail to kill AAV-transduced target cells in mice. To better model the human clinical trial data in mice, we introduced an immunodominant epitope derived from ovalbumin (OVA; SIINFEKL) into the AAV capsid and tested CTL-mediated killing of AAV2-transduced target tissues in vivo. Initially, in vitro experiments demonstrated both classical class I and cross-presentation of the OVA antigen, following endogenous expression or AAV2-OVA vector transduction, respectively. Furthermore, an OVA-specific CTL response was elicited after muscular or systemic injection of the AAV2-OVA vector. Finally, CTL reactivity was enhanced in mice with established SIINFEKL-specific immunity after AAV2-OVA/α1 anti-trypsin (AAT) administration. Most importantly, these OVA-specific CTLs decreased AAT expression in mice treated with AAV2-OVA/AAT vector that followed a time course mimicking uncoating kinetics of AAV2 transduction in OVA-immunized mice. These results demonstrate that AAV capsid-derived antigens elicit CD8+ CTL reactivity, and these CTLs eliminated AAV-transduced target cells in mice. Notably, this model system can be exploited to study the kinetics of capsid presentation from different serotypes of AAV and permit the design of novel strategies to block CTL-mediated killing of AAV-transduced cells.


Author(s):  
Shuhong Ma ◽  
Wenjian Jiang ◽  
Xujie Liu ◽  
Wen-Jing Lu ◽  
Tao Qi ◽  
...  

Rationale: Genetic editing has shown great potential for the treatment of human hereditary disorders via the elimination of mutations in embryos. However, the efficiency and safety of germline gene editing are not well understood. Objective: We aimed to examine the preclinical efficacy/safety of embryonic base editing in a mouse model of hypertrophic cardiomyopathy (HCM) using a novel adenine base editor (ABE) platform. Methods and Results: Here, we described the use of an ABEmax-NG to directly correct the pathogenic R404Q/+ mutation (Myh6 c.1211C>T) in embryos for a mouse model of HCM, increasing the number of wild-type embryos for in vitro fertilization. Delivery of the ABEmax-NG mRNA to embryos from R404Q/+ HCM mice resulted in 62.5-70.8% correction of the Myh6 c.1211C>T, reducing the level of mutant RNA and eliminating HCM in the post-natal mice as well as their offspring. In addition, the same sgRNA was also used to target an intronic locus (TGG PAM) with an overall editing rate of 86.7%, thus confirming that ABEmax-NG can efficiently edit target loci with different PAMs (NG) and genomic distribution in vivo. Compared with CRISPR/ssODN-mediated correction, ABEmax-NG displayed a much higher correction rate without introducing indels. DNA and RNA off-target analysis did not detect off-target editing in treated embryos and founder mice. In utero injection of adeno-associated virus 9 (AAV9) encoding the ABEmax-NG also resulted in around 25.3% correction of the pathogenic mutation and reduced of mutant RNA, thereby indicating ABEmax-NG has the potential to correct the HCM mutation in vivo. Conclusions: We developed an ABEmax-NG system, which efficiently corrected a pathogenic Myh6 HCM mutation in mouse embryos without off target lesions, thus safely eliminating HCM in derived mice and their progeny.


2020 ◽  
Vol 7 (8) ◽  
pp. 2410-2422
Author(s):  
Irene Brandts ◽  
Marlid Garcia-Ordoñez ◽  
Lluis Tort ◽  
Mariana Teles ◽  
Nerea Roher

Polystyrene nanoplastics are internalized in zebrafish liver cells, accumulating in lysosomes, and in zebrafish larvae but do not affect the larval suvival to a lethal infection.


1977 ◽  
Vol 146 (2) ◽  
pp. 606-610 ◽  
Author(s):  
R D Gordon ◽  
L E Samelson ◽  
E Simpson

T-cell mediated cytotoxic responses to H-Y antigen require co-recognition of H-Y and H-2 gene products. F1 mael stimulating cells and target cells express H-Y antigen in association with both parental H-2 haplotypes. However, F1 females primed in vivo and challenged in vitro with F1 male cells lyse male target cells of F1 and only one parental H-2 haplotype. Thus, (CBA X B10)F1 females sensitized to (CBA X B10)F1 male cells lyse (CBA X B10)F1 and CBA but not B10 male target cells, and (BALB/c X B10)F1 females sensitized to (BALB/c X B10)F1 male cells will lyse (BALB/c X B10)F1 and B10 but not BALB/c male target cells. It is suggested that this may represent an effect of immune response or suppressor genes mapping in the major histocompatibility gene complex which regulate responsiveness to H-Y antigen.


2000 ◽  
Vol 74 (17) ◽  
pp. 8003-8010 ◽  
Author(s):  
Yi Zhang ◽  
Narendra Chirmule ◽  
Guang-ping Gao ◽  
James Wilson

ABSTRACT Recombinant adeno-associated virus type 2 (rAAV) is being explored as a vector for gene therapy because of its broad host range, good safety profile, and persistent transgene expression in vivo. However, accumulating evidence indicates that administration of AAV vector may initiate a detectable cellular and humoral immune response to its transduced neo-antigen in vivo. To elucidate the cellular basis of the AAV-mediated immune response, C57BL/6 mouse bone marrow-derived immature and mature dendritic cells (DCs) were infected with AAV encoding β-galactosidase (AAV-lacZ) and adoptively transferred into mice that had received an intramuscular injection of AAV-lacZ 10 days earlier. Unexpectedly, C57BL/6 mice but not CD40 ligand-deficient (CD40L−/−) mice adoptively transferred with AAV-lacZ-infected immature DCs developed a β-galactosidase-specific cytotoxic T-lymphocyte (CTL) response that markedly diminished AAV-lacZ-transduced gene expression in muscle fibers. In contrast, adoptive transfer of AAV-lacZ-infected mature DCs failed to elicit a similar CTL response in vivo. Our findings indicate, for the first time, that immature DCs may be able to elicit a CD40L-dependent T-cell immunity to markedly diminish AAV-lacZ transduced gene expression in vivo when a sufficient number of DCs capturing rAAV vector and/or its transduced gene products is recruited.


2007 ◽  
Vol 81 (21) ◽  
pp. 11982-11991 ◽  
Author(s):  
Judith N. Mandl ◽  
Roland R. Regoes ◽  
David A. Garber ◽  
Mark B. Feinberg

ABSTRACT Antiviral CD8+ T cells are thought to play a significant role in limiting the viremia of human and simian immunodeficiency virus (HIV and SIV, respectively) infections. However, it has not been possible to measure the in vivo effectiveness of cytotoxic T cells (CTLs), and hence their contribution to the death rate of CD4+ T cells is unknown. Here, we estimated the ability of a prototypic antigen-specific CTL response against a well-characterized epitope to recognize and kill infected target cells by monitoring the immunodominant Mamu-A*01-restricted Tat SL8 epitope for escape from Tat-specific CTLs in SIVmac239-infected macaques. Fitting a mathematical model that incorporates the temporal kinetics of specific CTLs to the frequency of Tat SL8 escape mutants during acute SIV infection allowed us to estimate the in vivo killing rate constant per Tat SL8-specific CTL. Using this unique data set, we show that at least during acute SIV infection, certain antiviral CD8+ T cells can have a significant impact on shortening the longevity of infected CD4+ T cells and hence on suppressing virus replication. Unfortunately, due to viral escape from immune pressure and a dependency of the effectiveness of antiviral CD8+ T-cell responses on the availability of sufficient CD4+ T cells, the impressive early potency of the CTL response may wane in the transition to the chronic stage of the infection.


2008 ◽  
Vol 77 (3) ◽  
pp. 1182-1188 ◽  
Author(s):  
Charlotte Andreasen ◽  
Nicholas H. Carbonetti

ABSTRACT Pertussis is an acute respiratory disease caused by the bacterium Bordetella pertussis, for which humans are the only known reservoir. During infection, B. pertussis releases several toxins, including pertussis toxin (PT) and adenylate cyclase toxin (ACT), which have both been shown to play roles in promoting bacterial growth during early infection in a mouse model. Furthermore, in vitro and in vivo studies suggest that PT and ACT affect neutrophil chemotaxis and/or function, thereby altering the innate immune response. In this study we depleted animals of neutrophils to investigate whether neutrophils play a protective role during B. pertussis infection in mice. In addition, by infection with toxin-deficient strains, we investigated whether neutrophils are the main targets for PT and/or ACT activity in promoting bacterial growth. Surprisingly, we found no role for neutrophils during B. pertussis infection in naïve mice. However, in previously infected (immune) mice or in mice receiving immune serum, we observed a significant role for neutrophils during infection. Furthermore, in this immune mouse model our evidence indicates that neutrophils appear to be the main target cells for ACT, but not for PT.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 4061-4061
Author(s):  
Anna Sergeeva ◽  
Hong He ◽  
Kathryn Cox ◽  
Lisa St John ◽  
Mei Sang ◽  
...  

Abstract Previously we developed a T cell receptor (TCR) -like murine IgG2a monoclonal antibody, m8F4, which binds to the PR1 peptide/HLA-A2 complex, and targets both HLA-A2+ leukemia cell lines and primary HLA-A2+ patient blasts of various AML subtypes. We reported that m8F4 mediated complement dependent cytotoxicity (CDC) of AML in vitro and actively depleted AML in vivoin AML patient-derived xenograft (PDX) models. Due to the therapeutic potential of 8F4, we humanized 8F4 for clinical development. Here we report the specificity of this humanized IgG1κ 8F4 (h8F4) to PR1/HLA-A2 and document the activity of h8F4 against AML. Both ELISA and surface plasmon resonance (SPR) were used to study h8F4 specificity to recombinant monomer, consisting of PR1 peptide, HLA-A2 and β2 microglobulin. Like mouse 8F4, human 8F4 bound PR1/HLA-A2 monomer with high affinity (KD= 6.5 nM), while no binding was detected to control monomers pp65/HLA-A2 and WT1/HLA-A2. Moreover, FACS of peptide-pulsed T2 cells showed that h8F4 bound PR1- pulsed T2, but did not bind T2 cells loaded with other HLA-A2-restricted peptides, including MART1, pp65, WT1 and gp100, thus confirming h8F4 specificity to PR1/HLA-A2. To test whether h8F4 retains the in vivoanti-leukemia activity seen with m8F4, we used an NSG mouse model. Similar to m8F4, h8F4 treatment reduced leukemia growth in NSG mice with established disease in 3 out of 4 AML PDX models tested. To determine the mechanism of action (MOA) of humanized 8F4, we first tested h8F4 activity in the presence of complement because we had previously demonstrated the primary MOA of m8F4 to be CDC. However, unlike m8F4, even when tested at high concentrations up to 20 µg/ml, h8F4 did not mediate CDC of HLA-A2 transduced U937, HL60, or PR1-loaded T2 cells. To further examine h8F4 MOA, we tested antibody dependent cellular cytotoxicity (ADCC) of h8F4 using an lactase dehydrogenase (LDH) assay with pooled human PBMC as effectors. H8F4 induced ADCC of U937-A2 at E:T ratio 50 (EC50=0.95 µg/ml), thus indicating improved ADCC anti-AML activity of h8F4. Introduction of alanine substitutions in positions 234-235 (AAh8F4), which disrupted the binding of h8F4 to Fc receptor (FcR), abolished ADCC activity against U937-A2 in vitro. In contrast, AA8F4 still retained partial in vivo activity against the same target cells in the NSG mouse model, suggesting that ADCC is important, but not the exclusive mechanism of h8F4 in vivo. To investigate whether h8F4 had direct activity against AML, as possible alternative MOA, we measured apoptosis of target cells by h8F4 with and without crosslinking anti-Fc antibody fragments (CLA). Indeed, we found, by using annexin V and propidium iodide staining, that a 24 hours incubation with h8F4 (1µg/ml) in the presence of CLA induced apoptosis of 55.5 ± 3.5% of U937-A2 cells, as compared to 23.5 ± 2.1% in CLA-only control (p < 0.05). Moreover, caspase 3 activation was observed as early as 10 minutes after adding h8F4+CLA, and was time- and h8F4 concentration- dependent. Our data demonstrate that specificity of humanized anti-PR1/HLA-A2 TCR-like antibody, h8F4, is preserved and that h8F4 has a similar affinity compared with m8F4. In addition to inducing apoptosis, h8F4 mediates greater ADCC compared with m8F4, but lacks CDC activity in vitro. Together, our data is consistent with multiple mechanisms of action in vivo, including ADCC. Our results support further testing of h8F4 for patients with AML. Disclosures Sergeeva: Astellas Pharma: Patents & Royalties. Molldrem:Astellas Pharma: Patents & Royalties.


2021 ◽  
Vol 8 ◽  
Author(s):  
Ramazan Rezaei ◽  
Kaveh Baghaei ◽  
Seyed Mahmoud Hashemi ◽  
Mohammad Reza Zali ◽  
Hossein Ghanbarian ◽  
...  

Exosomes have been introduced as a new alternative delivery system for the transmission of small molecules. Tumor-derived exosomes (TEXs) not only contain tumor-associated antigens to stimulate antitumor immune responses but also act as natural carriers of microRNAs. The aim of the current study was to evaluate the efficacy of miR-124-3p-enriched TEX (TEXomiR) as cell-free vaccine in the induction of antitumor immune responses in a mouse model of colorectal cancer. Briefly, the exosomes were isolated from cultured CT-26 cell line, and modified calcium chloride method was used to deliver miR-124-3p mimic into the exosomes. We used a CT-26-induced BALB/c mouse model of colorectal cancer and analyzed the effect of TEXomiR on survival, tumor size, spleen and tumor-infiltrated lymphocytes, and splenocyte proliferation. Furthermore, intra-tumor regulatory T cells, cytotoxic activity of the splenocytes, and cytokine secretion was also evaluated to describe the anti-tumor immune response. When the tumor size reached 100 mm3, the mice were injected with TEXomiR, TEX, and/or phosphate-buffered saline (PBS) subcutaneously three times with 3-day interval, and then tumor size was monitored every 2 days. The in vitro results indicated that TEXs could efficiently deliver functional miR-124-3p mimic. The in vivo evaluation in tumor-bearing mice showed that treatment with TEXomiR can elicit a stronger anti-tumor immune response than unloaded TEX and PBS. Significant tumor growth inhibition and increased median survival time was achieved in tumor-bearing mice treated with TEXomiR. A significant decrease in CD4/CD8 and Treg/CD8 ratio in tumor tissue was demonstrated. Moreover, increased cytotoxicity and proliferation of splenocytes in the TEXomiR group compared to the TEX and PBS groups were identified. Taken together, our data demonstrated that tumor-derived exosomes efficiently deliver miR-124-3p mimic, and TEXomiR promotes anti-tumor immune responses.


2020 ◽  
Author(s):  
Xiaobo Zhang ◽  
Jun Xie ◽  
Hongmei Sun ◽  
Qing Wei ◽  
Ying Tao ◽  
...  

Abstract Background Neutrophilic asthma (NA) was a subtype of asthma. Soluble advanced glycosylation receptor (sRAGE) was considered to be associated with the neutrophilic airway. However, the role of sRAGE in NA still limited. Methods A NA mouse model was established and the levels of sRAGE in the bronchoalveolar lavage fluid (BALF) were measured by ELISA. Hematoxylin-eosin (HE) and Masson trichrome staining were used to identifying airway remodeling. Adeno-associated virus 9 (AAV9) overexpressed sRAGE and inhibitors for HMGB1, RAGE, and PI3K were used to intervene NA mouse model via tail-vein injection and intraperitoneally injection. Expressions of airway remodeling, EMT, and signaling markers were detected using qRT-PCR or western blotting. The levels of IL-17 and IL-6 in BALF were measured by ELISA. HMGB1 was applied to induce EMT of human bronchial epithelial cells (16HBE), then E-cadherin and vimentin expressions were examined after sRAGE, RAGE inhibitor, and PI3K inhibitor administration. Results sRAGE levels were significantly reduced in BALF and the airway remodeling was observed in the NA mouse model. AAV9-sRAGE significantly inhibited the neutrophilic airway inflammation, airway remodeling, and the expression of IL-17, IL-6, TGF-β1, RAGE, PI3K, and EMT markers -E-cadherin and vimentin in vivo. HMGB1 inhibitor, RAGE inhibitor, and PI3K inhibitor upregulated E-cadherin level. Moreover, HMGB1 promoted the EMT process via RAGE/PI3K in 16HBE cells and sRAGE reversed HMGB1- induced EMT in vitro. Conclusion sRAGE levels decrease in the mouse model with NA. sRAGE treatment attenuates neutrophilic airway inflammation, airway remodeling, and EMT. This suggests sRAGE may yield benefits in the treatment of NA.


Sign in / Sign up

Export Citation Format

Share Document