Open Access

Quantum dot/pMHC multimers vs. phycoerythrin/pMHC tetramers for identification of HLA-A*0201-restricted pHBV core antigen18-27-specific T cells

  • Authors:
    • Jianmeng Zhu
    • Yong Huang
    • Jing Su
    • Jian He
    • Yating Yu
    • Yongxiang Zhao
    • Xiaoling Lu
  • View Affiliations

  • Published online on: August 1, 2017     https://doi.org/10.3892/mmr.2017.7126
  • Pages: 8605-8612
  • Copyright: © Zhu et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Detection of human leukocyte antigens-A2-restricted p-hepatitis B virus (HBV) core antigen‑specific cytotoxic T lymphocytes (CTLs) is important in the study of HBV immunopathogenesis and vaccine design. Currently, major histocompatibility complex (MHC) class I/peptide‑(p) MHCI tetramers are considered the optimal tools to detect antigen‑specific CTLs. However, the MHC‑tetramer technique also has certain drawbacks and is under continuous development. The quantum dot (QD) bioconjugates nanotechnology with its unique inorganic‑biological properties has been developing fast. However, QD/pMHC multimers have seldom been used for the identification of the C18‑27 epitope, which is important in HBV infection. QD/pMHC multimers were synthesized by metal‑affinity coordination and an avidin‑biotin system. In the present study they were characterized by transmission electron microscopy, dynamic light scattering and fluorescence spectrophotometry. C18‑27‑specific CTLs were obtained by ex vivo expansion of CD8+ T cells. Cultured CTLs were tested for the secretion level of interferon (IFN)‑γ by ELISA and for cytotoxicity by lactate dehydrogenase release assay. Then, the performance of phycoerythrin (PE)/pMHC tetramers and QD/pMHC multimers were compared by flow cytometry. The synthesized QD/pMHC multimers dispersed well and their emission spectrum exhibited only slight differences compared with original QDs. C18‑27‑specific CTLs not only secreted IFN‑γ but also effectively targeted T2 cells pulsed with peptide C18‑27. The frequencies of C18‑27‑specific CTLs determined by QD/pMHC multimers were higher compared with PE/pMHC tetramers. The present results suggested that QD/pMHC multimers may be able to characterize greater numbers of C18‑27‑specific CTLs with increased sensitivity compared to conventional strategies.

Introduction

Chronic hepatitis B virus (HBV) infection is an extremely serious health problem worldwide with a number of ensuing complications including chronic hepatitis, liver cirrhosis and hepatocellular carcinoma (1,2). Statistical data indicate that ~400 million people are chronically infected with HBV (3). It has been reported that cellular immune responses serve an important role in preventing the deterioration of HBV infection (4). HBV-specific cytotoxic T lymphocytes (CTLs) are known to serve a critical role in cellular immune responses; efficient estimation of their numbers significantly improves the study of the viral clearance and disease pathogenesis (57). For the identification of HBV-specific CTLs, the human leukocyte antigen (HLA)-A*020-restricted p-HBV core antigen (HBcAg18–27) epitope C18–27 is a perfect candidate. First, HLA-A2 is the most prevalent HLA allele globally so this epitope may be used for targeting virus-specific CTLs in almost half the population (8). Second, the therapeutic C18–27 peptide has been detected in the majority of HLA-A2.1 patients with HBV (9,10). Third, it has been reported that the presence of valine at the C-terminal of C18–27 peptide is associated with severe liver inflammation in Chinese patients with chronic HBV infection (11). Together, these reasons demonstrate that the characterization of C18–27-specific T cells with high sensitivity is vital for the development of immunotherapeutic approaches to diseases caused by HBV.

A wide range of assays have been developed to measure T-cell immune responses (12) and among them peptide-major histocompatibility complex (pMHC) tetramers are the most common and standard methods (13). The fluorescence-labeled pMHC tetramer binds to a specific T-cell receptor (TCR) on the surface of a T cell using the nominal peptide imitating the corresponding antigenic epitope. This enables the detection of T cells by flow cytometry ex vivo (14). The pMHC tetramer technique is a powerful method to study T-cells, allowing direct and rapid visualization and quantification (15). It should be noted that the pMHC tetramers technique has its own drawbacks and remains under improvement. Phycoerythrin (PE) is the most commonly used organic fluorochrome for labeling tetramers (16). Its size is ~0.5–1 nm (17) meaning that normally just one avidin can be attached to a single PE organic fluorophore. The PE/pMHC tetramers usually contain four biotinylated pMHC monomers resulting in an avidin molecule has just four biotin-binding sites (18; as illustrated in Fig. 1A). Thus, they often fail to characterize CTLs where the interaction between pMHC and TCR is relatively weak.

The new emerging inorganic quantum dot-based multimers exhibit promising applications in cellular labeling and possess numerous advantages over conventional tetramers. Semiconductor nanocrystal QDs are spherical particles with diameters typically ranging between 2–20 nm, and consist of a semiconductor core (including CdSe and CdTe) alone or coated with a shell. The shell of the QD (e.g., ZnS) enhances the quantum yield and protects the core from oxidation (19). QD bioconjugates have been found to be 2,600-fold more resistant to photobleaching than PE (20). The narrow emission and broad excitation spectra from different QDs can be excited by a single light source (21). In addition, a single QD can be labeled with several proteins simultaneously. It has been reported that ~15–20 maltose binding proteins can be stably coated on each 6 nm diameter QD (22). Thus, a number of streptavidin molecules may be attached to one QD nanoparticle, leading to the QD/pMHC multimers carrying more pMHC monomers compared with standard pMHC tetramers (as illustrated in Fig. 1B). These QD/pMHC multimers offer a unique architecture for increasing the affinity of the TCR-pMHC interaction and may perform higher detection frequencies than PE/pMHC tetramers.

To date, a number of authors have made the use of QD/pMHC multimers for immunophenotyping (2325). However, to the best of our knowledge, QD/pMHC multimers have not been used for the identification of the C18–27 epitope which serves an important role in the HBV infection. Thus, the present study used the QD/pMHC multimers technique to detect the ex vivo expansion of C18–27-specific T cells by flow cytometry with a single 488-nm excitation light source. The aim of the current study was to examine whether QD/pMHC multimers can be used to stain C18–27-specific T cells and if the technique were preferable to standard pMHC tetramers.

Materials and methods

Materials

The CdTe/CdS/ZnS core/shell/shell QDs were purchased from Beida Jubang Science & Technology Co. Ltd. (Beijing, China) and the streptavidin (SA) from Sigma-Aldrich; Merck KGaA (Darmstadt, Germany). Corning Lymphocyte Serum-free medium was used for T cell culture and purchased from Zhao Sheng Co., Ltd. (Beijing, China). The transporters associated with antigen processing-deficient HLA-A2-positive T2 cell line was obtained from the Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research (Guangxi, China). The K562 cell line (cat no. CCL-243) was purchased from Xiangf Biotechnology Co., Ltd. (Shanghai, Beijing). Peptides SLYNTVATL (SL9) and FLPSDFFPSV (C18–27) were synthesized by the Chinese Peptide Company (Hangzhou, China). Biotinylated pMHC monomers were from Beijing QuantoBio Biotechnology Co., Ltd. (Beijing, China). All chemicals used were of the highest purity available.

Synthesis of water soluble QD/pMHC multimers and PE/pMHC tetramers

QDs with emission maximum of 635 nm were used in the present study. QD-SA conjugates were prepared via metal-affinity interactions between a polyhistidine (His)-tag and Zn on the surface of the QDs. The His-tag SA (HSA) was prepared as previously described (24). Then, 10 µl of 25 µM HSA was incubated with 10 µl of 5 µM QDs in DMSO for 2 h at room temperature to yield QD-SA. The bioconjugates were purified by membrane filtration and washed with DMSO three times. Then, 10 µl QD-SA was added to 50 µl biotinylated pMHC monomers solution (0.1 mM) and stirred in the dark for 10 min; this step was repeated several times and the multimers obtained were purified using a 100 kDa membrane filter. PE/pMHC tetramers were obtained by a similar procedure but by adding PE-SA to biotinylated pMHC monomers solution.

Characterization of QDs and QD bioconjugates

The variety of QDs used in the present study was designated CdTe/CdS/ZnS core/shell/shell QDs. The surface properties of the QDs were characterized by transmission electron microscopy (TEM; H7650; JEOL, Ltd., Tokyo, Japan) and high-resolution TEM (JEM-2100F; JEOL, Ltd.). QD-bioconjugates were characterized by TEM and dynamic light scattering (DLS; Malvern Zetasizer; Malvern Instruments, Ltd., Malvern, UK). A fluorescence spectrophotometer (F-7000; Hitachi, Ltd., Tokyo, Japan) was used to characterize the fluorescence properties of the nanocrystals.

Ex vivo expansion of HBcAg18-27-specific T-cells

Peripheral blood mononuclear cells were isolated from 100 ml whole heparinized blood of healthy HLA-A*0201 volunteers by Ficoll/Hypaque density gradient centrifugation with 120 × g at 25°C for 20 min (Sigma-Aldrich; Merck KGaA). HLA class I typing and A2 subtyping was performed by sequence-specific primer polymerase chain reaction (PCR). T2 cells in serum-free HEPES-buffered RPMI 1640 were pulsed with 40 µg/ml pHBV C18–27 (T2/C18–27) at 37°C for 4 h and irradiated by 200 Gy as stimulating cells. Effector cells were designated into two groups. One group was peripheral blood lymphocyte cells (PBLs), obtained by adherence to plastic for 1 h in complete RPMI-1640 medium (containing 2 mM L-glutamine, 100 U/ml each of penicillin and streptomycin, 1 mM sodium pyruvate, 1X MEM nonessential amino acids and 10% autologous serum). The other group was CD8+ T cells immunobead-purified from PBLs. The effector cells were cocultured with stimulating cells at a ratio of 10:1 at 37°C under 5% CO2 and 20 IU/ml human interleukin-2 and 10 ng/ml human interleukin-7 were added into the coculture medium every 3 days. Lymphocytes were re-stimulated with C18–27-pulsed irradiated T2 cells each week.

The study was approved by the Medical Ethics Committee of Guangxi Medical University (Nanning, China). Written informed consent was obtained from all subjects prior to enrolment in the present study.

Analysis of T2-MHC-I binding affinity

T2/C18–27 cells were incubated at 37°C for 4 h in a 5% CO2 atmosphere and irradiated at 200 Gy. Expression of MHC-I on T2/C18–27 and T2 cells subjected to the same procedures was determined by staining with fluorescein isothiocyanate (FITC)-conjugated anti-HLA-A2 monoclonal antibody (mAb) BB7.2. The fluorescence index (FI) was calculated as follows: FI=(mean FITC fluorescence with peptide C18–27-mean FITC fluorescence without peptide)/(mean FITC fluorescence without peptide). Peptides with FI>1 were considered as high-affinity epitopes.

Interferon (IFN)-γ detection assay

To ensure that the T cells were activated, secretion of IFN-γ by the C18–27-specific T cell cultures was evaluated by ELISA. OptEIA sets (BD Pharmingen, San Diego, CA, USA) were used to measure the concentrations of IFN-γ in supernatants of CTLs restimulated for 40 h with peptide-pulsed T2 cells. T2 were pulsed with peptides at concentrations of 40 µg/ml and used at the T cell: T2 cell ratio of 1:10. The IFN-γ in supernatants of unstimulated T and T2 cells served as negative control.

Peptide-specific CTL cytotoxicity assay

Cytotoxicity of C18–27-specific CTLs was tested by a lactate dehydrogenase (LDH) release assay. The effector cells were C18–27-specific T-cells from ex vivo culture. Target cells were T2 pulsed with FLPSDFFPSV (T2/C18–27) and were cocultured at effector/target ratios of 10:1, 20:1 or 50:1 at 37°C under 5% CO2 for 4 h. K562 and T2 cells pulsed with SLYNTVATL (T2/SL9) were negative control groups. Each assay was performed in triplicate. Percent specific lysis=[(experimental release-spontaneous release)/(maximum release-spontaneous release)] × 100.

Characterization of peptide-specific T-cells by flow cytometry

In order to avoid the interference of non-specific cells, the effector T cells were divided into two groups: Peripheral PBLs and purified CD8+ T cells. Following two weeks in vitro culture, PBL and CD8+ T cells were collected and simultaneously labeled with FITC-conjugated anti CD8 mAb and PE/tetramers or QD/multimers. Four-color flow cytometry analysis was performed on a Coulter Epics XL cytometer with a single 488-nm excitation light source (Beckman Coulter, Inc., Brea, CA, USA). The flow cytometry data were analyzed in real time using Expo32 ADC software version 1.1C (Beckman Coulter).

Quantum dot cytotoxicity assay

For the cytotoxicity experiment, the harvested PBLs were loaded into 96-well microtiter plates and incubated with quantum dot/pMHC multimers (concentrations ranging between 0 and 5 µM) for 24 h at 37°C. Each well contained 5,000 cells and samples were performed in triplicate. Then, 10 µl of the Cell Counting Kit-8 (CCK-8; Sigma-Aldrich; Merck KGaA) solution was added to each well and the cells were incubated for another 2 h at 37°C with 5% CO2. The absorbance of samples was measured at 450 nm by UV-vis with a Multiscan GO (Thermo Fisher Scientific, Inc., Waltham, MA, USA). The number of cells was proportional to the optical density (OD) value and non-treated group was expressed as 100%.

Statistical analysis

Data from the experimental and control groups were analysed using one-way analysis of variance (ANOVA) followed by post hoc least significant differences, Student-Neuman-Keuls or Bonferroni tests for multiple comparisons. Data from the CCK-8 experiment were analysed using two-way ANOVA. P<0.05 was considered to indicate a statistically significant difference. Data are reported as the mean ± standard deviation. Statistical comparisons were performed using GraphPad Prism software version 6.0 (GraphPad Software,. Inc., La Jolla, CA, USA).

Results

Characterization of QDs and QD bioconjugates

TEM and DLS were used for the characterization of QDs, QD-SA and QD/pMHC multimers (Fig. 2). Results demonstrated that the diameter of QDs, QD-SA and QD/pMHC multimers were ~6.0, 18.31±2.01 and 50.04±10.12 nm, respectively (Fig. 2). The polydispersity index of all was <0.5. The fluorescence emission curve of original QD, PE and multimers (Fig. 3) showed that the fluorescence properties of both QD and PE had not been influenced by the surface modification process. These results indicated that the QDs and QD bioconjugate nanoparticles were well-dispersed and that the polymers or protein coated on QDs did not change the surface properties of the CdTe/CdS/ZnS nanocrystals.

Identification of stable HLA-1 expression in T2 pulsed with antigen peptide

To ensure that MHC-I molecules expressed stably on T2 cells, expression of HLA-A0201 on the surface of T2 cell membranes was examined by staining with mAb FITC-BB7.2. As presented in Fig. 4A, a flow cytometry assay demonstrated that T2/C18–27 cells expressed HLA-A0201 with markedly higher frequencies (98.5%) compared with T2 cells treated in similar conditions but without pulsing with antigen peptide, FI>1 (7.9%).

IFN-γ responses of peptide-specific T-cells from in vitro expansion

In order to confirm that the T cells had been activated by the selected functional epitope peptide C18–27, the secretion of IFN-γ in supernatants of CTLs was tested by ELISA. As demonstrated in Fig. 4B, the IFN-γ secretion from C18–27-specific CTLs increased markedly following ex vivo coculture with T2/C18–27 for three weeks. In contrast, the T cells or T2 cells alone released significantly less IFN-γ compared with the T2/T cell experimental group (P<0.01; Fig. 4B).

Cytotoxicity of peptide-specific CTLs

The cytotoxicity of the cultured CTLs was investigated by LDH release assay. The results demonstrated that cultured specific CTLs lysed the T2/C18–27 cells while the negative control groups (K562 and T2/SL9 cells) did not induce a CTL response and spontaneous release was below 10% of the maximum release (P<0.01; Fig. 4C). These results indicated that C18–27 CTLs had been induced successfully ex vivo.

Comparison of QD/pMHC multimers and PE/pMHC tetramers for the detection of peptide-specific T cells

To compare the staining of C18–27-specific T cells with PE/pMHC tetramers and QD/pMHC multimers, SL9 multimers or tetramers were designed as negative control materials. It is notable in Fig. 5 that QD/pMHC multimers detected higher numbers of C18–27-specific CTLs than PE/pMHC tetramers. The frequencies in negative control groups were significantly lower.

Cytotoxicity assessment of pMHCI-QD multimers

To evaluate whether QD toxicity was harmful to T cells, a CCK-8 kit was used. Cytotoxicity data demonstrated that the percentage of cell death upon incubation of T cells with QD/multimers was low (Fig. 6). However, no significant difference in cytotoxicity between QD/multimers and PE/tetramers was detected. It also demonstrated that QDs with shells or coatings minimized Cd2+ release from the particle surface and that the CdTe/CdS/ZnS core/shell/shell QDs can be applied in T cell experiments.

Discussion

For the biological synthesis of QD/pMHC multimers, it is important to choose an appropriate assay for solubilization and stabilization of QD-bioconjugates in solutions (26). There are two main approaches to the surface modification of QDs: Covalent linkage and noncovalent binding (2729). The covalent linkage assay usually results in QD-protein dispersions with a large aggregation and low quantum yield. The present study bound streptavidin to QDs via noncovalent metal-affinity interactions. Studies (28,30,31) have shown that in His-tag motifs to Zn atoms of QDs, the interaction (Zn2+-His) is stronger than the majority of antibody bindings, so streptavidin was labeled with a His-tag to form high-affinity complexes with QDs. Then QD/pMHC multimers were obtained by conjugating monomeric pMHCI biotinylated-proteins to QD-SA. From the TEM images it can be observed that the QD/pMHC multimers nanoparticles are larger than QDs, so it is supposed that one single QD/pMHC multimer may contain several QDs and numerous pMHC monomers (Fig. 2C), although unforeseen aggregation did not appear. The resulting pellets demonstrated suitable stabilization (Fig. 2E) and the photoluminescence spectra did not demonstrate any noticeable difference in the peaks (Fig. 3), indicating that the surface modification did not induce any significant change in the structures of the QDs. One possible reason may be that the exposed charged groups and hydrophobic chains per polymer molecule in the QD-coating bioconjugates had changed little during the QD surface modification process (17).

QD toxicity has always been a great concern in biomedical studies (32). The cytotoxicity of QDs has been demonstrated to occur from released Cd2+ ions and can be greatly reduced by using QDs with shells or coatings (33). Cell viability is a commonly used assay to evaluate the toxicity of nanomaterials. In this study, CCK-8, which was considered to possess a improved sensitivity compared with the well-known MTT assay, was used to evaluate QD toxicity (34). The quantity of the formazan dye is directly proportional to the occurrence of living cells in this method. The relative viability of cells was obtained from the OD value at 450 nm wavelength. The CdTe/CdS/ZnS core/shell/shell QDs used in the present study did not show significant cytotoxicity (Fig. 6), suggesting that the CdS/ZnS coating of QDs greatly reduced the toxicity within the biological system.

For the coculture of T2/C18–27 and T cells, purified CD8+ T cells were developed as one group of effector T cells in order to eliminate non-specificity influence of other cells in PBLs. To determine whether HLA-A*0201 expressed steadily on the surface of T2/C18–27 cells, the T2/C18–27 cells were stained with FITC-BB7.2 mAb. As demonstrated in Fig. 4A the peptide C18–27 bound to T2 with strong affinity. The secretion level of IFN-γ analyzed by ELISA assay demonstrated that C18–27 was markedly efficient at activating C18–27-specific CTL responses (Fig. 4B). The cytotoxicity of peptide-specific CTLs detected by LDH release assay demonstrated that C18–27-specific CTLs were induced successfully and the resulting CTLs had the ability to kill target cells (Fig. 4C). The experiments were designed to ensure that there was a sufficient number of functional C18–27-specific T cells for parallel comparative analyses of T cell staining with PE/pMHC tetramers and QD/pMHC multimers.

During the expansion of C18–27-specific CTL, PE/pMHC tetramers and QD/pMHC multimers were used to detect the frequencies of C18–27 specific CTLs. Fig. 5A and B demonstrate that QD/C18–27 multimers detected a higher number of T cells compared with PE/SL9 tetramers each week during ex vivo expansion. However, PE/SL9 tetramers exhibited a more stable detection result compared with QD/C18–27 multimers. When the effector T cells became purified CD8+ T cells, the performance of the QD/C18–27 multimers was similar to PE/SL9 tetramers following ex vivo expansion for 10 days (data not shown). However, over 15 days the average CTL frequencies determined by QD/C18–27 multimers were markedly higher than PE/SL9 tetramers (Fig. 5C and D). The result may indicate that QD/C18–27 multimers exhibit higher detection efficiency compared with PE/SL9 tetramers when the frequency of C18–27-specific CTL is relatively high. In brief, QD/pMHC multimers detected a higher number of T cells than PE/pMHC tetramers in the majority of cases and the QD/pMHC multimers technique can improve detection sensitivity.

Since Altman et al (18) first constructed MHC-tetramers for tagging T cells in 1996, MHC-tetramers technique have rapidly become the gold standard for T-cell analysis of known antigen specificity (35). The fast-moving development of QD-bioconjugates is stimulating interfaces with nanotechnology as well. The present study compared QD/pMHC multimers and PE/pMHC tetramers for staining C18–27-specific CTLs expanded ex vivo with a single 488 nm argon laser for the first time and the results indicated that QD/pMHC multimer performed better in the analysis of C18–27 specific CTLs, which may improve the understanding of T cell immune response induced by HBV. It may also serve an important role in the development of more effective vaccines.

Acknowledgements

The present study was supported by the National Natural Scientific Foundation of China (grant nos. 81430055, 81172139, 81172138 and 81372452), the International Cooperation Project of the Ministry of Science and Technology of China (grant no. 2015DFA31320), the Project for Innovative Research Team in Guangxi Natural Science Foundation (grant no. 2015GXNSFFA139001) and the Project of Science and Technology of Guangxi (grant nos. 14125008-2-12 and 1599005-2-10).

References

1 

Chisari FV and Ferrari C: Hepatitis B virus immunopathogenesis. Annu Rev Immunol. 13:29–60. 1995. View Article : Google Scholar : PubMed/NCBI

2 

Beasley RP, Hwang LY, Lin CC and Chien CS: Hepatocellular carcinoma and hepatitis B virus. A prospective study of 22 707 men in Taiwan. Lancet. 2:1129–1133. 1981. View Article : Google Scholar : PubMed/NCBI

3 

Noordeen F: Hepatitis B virus infection: An insight into infection outcomes and recent treatment options. Virusdisease. 26:1–8. 2015. View Article : Google Scholar : PubMed/NCBI

4 

Guidotti LG and Chisari FV: Immunobiology and pathogenesis of viral hepatitis. Annu Rev Pathol. 1:23–61. 2006. View Article : Google Scholar : PubMed/NCBI

5 

Wherry EJ and Ahmed R: Memory CD8 T-cell differentiation during viral infection. J Virol. 78:5535–5545. 2004. View Article : Google Scholar : PubMed/NCBI

6 

Virgin HW, Wherry EJ and Ahmed R: Redefining chronic viral infection. Cell. 138:30–50. 2009. View Article : Google Scholar : PubMed/NCBI

7 

Belyakov IM and Ahlers JD: What role does the route of immunization play in the generation of protective immunity against mucosal pathogens? J Immunol. 183:6883–6892. 2009. View Article : Google Scholar : PubMed/NCBI

8 

Varela-Rohena A, Molloy PE, Dunn SM, Li Y, Suhoski MM, Carroll RG, Milicic A, Mahon T, Sutton DH, Laugel B, et al: Control of HIV-1 immune escape by CD8 T cells expressing enhanced T-cell receptor. Nat Med. 14:1390–1395. 2008. View Article : Google Scholar : PubMed/NCBI

9 

Bertoletti A and Gehring AJ: The immune response during hepatitis B virus infection. J Gen Virol. 87:1439–1449. 2006. View Article : Google Scholar : PubMed/NCBI

10 

Engler OB, Dai WJ, Sette A, Hunziker IP, Reichen J, Pichler WJ and Cerny A: Peptide vaccines against hepatitis B virus: From animal model to human studies. Mol Immunol. 38:457–465. 2001. View Article : Google Scholar : PubMed/NCBI

11 

Liu J, Chen KY and Ren EC: Structural insights into the binding of hepatitis B virus core peptide to HLA-A2 alleles: Towards designing better vaccines. Eur J Immunol. 41:2097–2106. 2011. View Article : Google Scholar : PubMed/NCBI

12 

Hobeika AC, Morse MA, Osada T, Ghanayem M, Niedzwiecki D, Barrier R, Lyerly HK and Clay TM: Enumerating antigen-specific T-cell responses in peripheral blood: A comparison of peptide MHC Tetramer, ELISpot, and intracellular cytokine analysis. J Immunother. 28:63–72. 2005. View Article : Google Scholar : PubMed/NCBI

13 

Dolton G, Lissina A, Skowera A, Ladell K, Tungatt K, Jones E, Kronenberg-Versteeg D, Akpovwa H, Pentier JM, Holland CJ, et al: Comparison of peptide-major histocompatibility complex tetramers and dextramers for the identification of antigen-specific T cells. Clin Exp Immunol. 177:47–63. 2014. View Article : Google Scholar : PubMed/NCBI

14 

Meidenbauer N, Hoffmann TK and Donnenberg AD: Direct visualization of antigen-specific T cells using peptide-MHC-class I tetrameric complexes. Methods. 31:160–171. 2003. View Article : Google Scholar : PubMed/NCBI

15 

Sims S, Willberg C and Klenerman P: MHC-peptide tetramers for the analysis of antigen-specific T cells. Expert Rev Vaccines. 9:765–774. 2010. View Article : Google Scholar : PubMed/NCBI

16 

Reguzova AY, Karpenko LI, Mechetina LV and Belyakov IM: Peptide-MHC multimer-based monitoring of CD8 T-cells in HIV-1 infection and AIDS vaccine development. Expert Rev Vaccines. 14:69–84. 2015. View Article : Google Scholar : PubMed/NCBI

17 

Bilan R, Fleury F, Nabiev I and Sukhanova A: Quantum dot surface chemistry and functionalization for cell targeting and imaging. Bioconjug Chem. 26:609–624. 2015. View Article : Google Scholar : PubMed/NCBI

18 

Altman JD, Moss PA, Goulder PJ, Barouch DH, McHeyzer-Williams MG, Bell JI, McMichael AJ and Davis MM: Phenotypic analysis of antigen-specific T lymphocytes. Science. 274:94–96. 1996. View Article : Google Scholar : PubMed/NCBI

19 

Hines MA and Guyot-Sionnest P: Synthesis and characterization of strongly luminescing ZnS-capped CdSe nanocrystals. J Phys Chem. 100:468–471. 1996. View Article : Google Scholar

20 

Sukhanova A, Devy J, Venteo L, Kaplan H, Artemyev M, Oleinikov V, Klinov D, Pluot M, Cohen JH and Nabiev I: Biocompatible fluorescent nanocrystals for immunolabeling of membrane proteins and cells. Anal Biochem. 324:60–67. 2004. View Article : Google Scholar : PubMed/NCBI

21 

Samir TM, Mansour MM, Kazmierczak SC and Azzazy HM: Quantum dots: Heralding a brighter future for clinical diagnostics. Nanomedicine (Lond). 7:1755–1769. 2012. View Article : Google Scholar : PubMed/NCBI

22 

Medintz IL, Uyeda HT, Goldman ER and Mattoussi H: Quantum dot bioconjugates for imaging, labelling and sensing. Nat Mater. 4:435–446. 2005. View Article : Google Scholar : PubMed/NCBI

23 

Chattopadhyay PK, Price DA, Harper TF, Betts MR, Yu J, Gostick E, Perfetto SP, Goepfert P, Koup RA, De Rosa SC, et al: Quantum dot semiconductor nanocrystals for immunophenotyping by polychromatic flow cytometry. Nat Med. 12:972–977. 2006. View Article : Google Scholar : PubMed/NCBI

24 

Andersen RS, Kvistborg P, Frøsig TM, Pedersen NW, Lyngaa R, Bakker AH, Shu CJ, Straten Pt, Schumacher TN and Hadrup SR: Parallel detection of antigen-specific T cell responses by combinatorial encoding of MHC multimers. Nat Protoc. 7:891–902. 2012. View Article : Google Scholar : PubMed/NCBI

25 

Howarth M, Chinnapen DJ, Gerrow K, Dorrestein PC, Grandy MR, Kelleher NL, El-Husseini A and Ting AY: A monovalent streptavidin with a single femtomolar biotin binding site. Nat Methods. 3:267–273. 2006. View Article : Google Scholar : PubMed/NCBI

26 

Wu C, Liu J, Zhang P, Li J, Ji H, Yang X and Wang K: A recognition-before-labeling strategy for sensitive detection of lung cancer cells with a quantum dot-aptamer complex. Analyst. 140:6100–6107. 2015. View Article : Google Scholar : PubMed/NCBI

27 

Pinaud F, King D, Moore HP and Weiss S: Bioactivation and cell targeting of semiconductor CdSe/ZnS nanocrystals with phytochelatin-related peptides. J Am Chem Soc. 126:6115–6123. 2004. View Article : Google Scholar : PubMed/NCBI

28 

Hainfeld JF, Liu W, Halsey CM, Freimuth P and Powell RD: Ni-NTA-gold clusters target His-tagged proteins. J Struct Biol. 127:185–198. 1999. View Article : Google Scholar : PubMed/NCBI

29 

Akerman ME, Chan WC, Laakkonen P, Bhatia SN and Ruoslahti E: Nanocrystal targeting in vivo. Proc Natl Acad Sci USA. 99:pp. 12617–12621. 2002; View Article : Google Scholar : PubMed/NCBI

30 

Goldman ER, Medintz IL, Whitley JL, Hayhurst A, Clapp AR, Uyeda HT, Deschamps JR, Lassman ME and Mattoussi H: A hybrid quantum dot-antibody fragment fluorescence resonance energy transfer-based TNT sensor. J Am Chem Soc. 127:6744–6751. 2005. View Article : Google Scholar : PubMed/NCBI

31 

Medintz IL, Berti L, Pons T, Grimes AF, English DS, Alessandrini A, Facci P and Mattoussi H: A reactive peptidic linker for self-assembling hybrid quantum dot-DNA bioconjugates. Nano Lett. 7:1741–1748. 2007. View Article : Google Scholar : PubMed/NCBI

32 

Hardman R: A toxicologic review of quantum dots: Toxicity depends on physicochemical and environmental factors. Environ Health Perspect. 114:165–172. 2006. View Article : Google Scholar : PubMed/NCBI

33 

Ryman-Rasmussen JP, Riviere JE and Monteiro-Riviere NA: Surface coatings determine cytotoxicity and irritation potential of quantum dot nanoparticles in epidermal keratinocytes. J Invest Dermatol. 127:143–153. 2007. View Article : Google Scholar : PubMed/NCBI

34 

Miyamoto T, Min W and Lillehoj HS: Lymphocyte proliferation response during Eimeria tenella infection assessed by a new, reliable, nonradioactive colorimetric assay. Avian Dis. 46:10–16. 2002. View Article : Google Scholar : PubMed/NCBI

35 

Hadrup SR and Schumacher TN: MHC-based detection of antigen-specific CD8+ T cell responses. Cancer Immunol Immunother. 59:1425–1433. 2010. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2017
Volume 16 Issue 6

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhu J, Huang Y, Su J, He J, Yu Y, Zhao Y and Lu X: Quantum dot/pMHC multimers vs. phycoerythrin/pMHC tetramers for identification of HLA-A*0201-restricted pHBV core antigen18-27-specific T cells. Mol Med Rep 16: 8605-8612, 2017
APA
Zhu, J., Huang, Y., Su, J., He, J., Yu, Y., Zhao, Y., & Lu, X. (2017). Quantum dot/pMHC multimers vs. phycoerythrin/pMHC tetramers for identification of HLA-A*0201-restricted pHBV core antigen18-27-specific T cells. Molecular Medicine Reports, 16, 8605-8612. https://doi.org/10.3892/mmr.2017.7126
MLA
Zhu, J., Huang, Y., Su, J., He, J., Yu, Y., Zhao, Y., Lu, X."Quantum dot/pMHC multimers vs. phycoerythrin/pMHC tetramers for identification of HLA-A*0201-restricted pHBV core antigen18-27-specific T cells". Molecular Medicine Reports 16.6 (2017): 8605-8612.
Chicago
Zhu, J., Huang, Y., Su, J., He, J., Yu, Y., Zhao, Y., Lu, X."Quantum dot/pMHC multimers vs. phycoerythrin/pMHC tetramers for identification of HLA-A*0201-restricted pHBV core antigen18-27-specific T cells". Molecular Medicine Reports 16, no. 6 (2017): 8605-8612. https://doi.org/10.3892/mmr.2017.7126