Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
J Immunol ; 208(9): 2246-2255, 2022 05 01.
Article in English | MEDLINE | ID: mdl-35418467

ABSTRACT

The NKG2A/HLA-E axis is an immune checkpoint that suppresses immune effector activity in the tumor microenvironment. In mice, the ligand for the NKG2A/CD94 inhibitory receptor is the nonclassical MHC molecule Qa-1b, the HLA-E ortholog, which presents the peptide AMAPRTLLL, referred to as Qdm (for Qa-1 determinant modifier). This dominant peptide is derived from the leader sequences of murine classical MHC class I encoded by the H-2D and -L loci. To broaden our understanding of Qa-1b/Qdm peptide complex biology and its tumor protective role, we identified a TCR-like Ab from a single domain VHH library using yeast surface display. The TCR-like Ab (EXX-1) binds only to the Qa-1b/Qdm peptide complex and not to Qa-1b alone or Qa-1b loaded with control peptides. Conversely, currently available Abs to Qa-1b bind independent of peptide loaded. Flow cytometric results revealed that EXX-1 selectively bound to Qa-1b/Qdm-positive B16F10, RMA, and TC-1 mouse tumor cells but only after pretreatment with IFN-γ; no binding was observed following genetic knockdown of Qa-1b or Qdm peptide. Furthermore, EXX-1 Ab blockade promoted NK cell-mediated tumor cell lysis in vitro. Our findings show that EXX-1 has exquisite binding specificity for the Qa-1b/Qdm peptide complex, making it a valuable research tool for further investigation of the Qa-1b/Qdm peptide complex expression and regulation in healthy and diseased cells and for evaluation as an immune checkpoint blocking Ab in syngeneic mouse tumor models.


Subject(s)
Histocompatibility Antigens Class I , Killer Cells, Natural , Animals , Antibodies/metabolism , Mice , NK Cell Lectin-Like Receptor Subfamily C/metabolism , Peptides , Receptors, Antigen, T-Cell/metabolism
2.
Nanotechnology ; 32(4): 045501, 2021 Jan 22.
Article in English | MEDLINE | ID: mdl-33027774

ABSTRACT

A plasmonic nanopore sensor enabling detection of bimodal optical and electrical molecular signatures was fabricated and tested for its ability to characterize low affinity ligand-receptor interactions. This plasmonic nanosensor uses self-induced back-action (SIBA) for optical trapping to enable SIBA-actuated nanopore electrophoresis (SANE) through a nanopore located immediately below the optical trap volume. A natural killer (NK) cell inhibitory receptor heterodimer molecule CD94/NKG2A was synthesized to target a specific peptide-presenting Qa-1b Qdm ligand as a simplified model of low-affinity interactions between immune cells and peptide-presenting cancer cells that occurs during cancer immunotherapy. A cancer-irrelevant Qa-1b GroEL ligand was also targeted by the same receptor as a control experiment to test for non-specific binding. The analysis of different pairs of bimodal SANE sensor signatures enabled discrimination of ligand, receptor and their complexes and enabled differentiating between specific and non-specific ligand interactions. We were able to detect ligand-receptor complex binding at concentrations over 500 times lower than the free solution equilibrium binding constant (K D ). Additionally, SANE sensor measurements enabled estimation of the fast dissociation rate (k off) for this low-affinity specific ligand-receptor system, previously shown to be challenging to quantify with commercial technologies. The k off value of targeted peptide-presenting ligands is known to correlate with the subsequent activation of immune cells in vivo, suggesting the potential utility of the SANE senor as a screening tool in cancer immunotherapy.


Subject(s)
Electrophoresis , Nanopores , Receptors, Natural Killer Cell , Animals , Electrophoresis/instrumentation , Electrophoresis/methods , Kinetics , Ligands , Mice , Mice, Inbred C57BL , Peptides/chemistry , Peptides/metabolism , Protein Binding , Receptors, Natural Killer Cell/chemistry , Receptors, Natural Killer Cell/metabolism
3.
Nanotechnology ; 31(8): 085502, 2019 Nov 01.
Article in English | MEDLINE | ID: mdl-31675752

ABSTRACT

Recent advances in plasmonic nanopore technologies have enabled the use of concurrently acquired bimodal optical-electrical data for improved quantification of molecular interactions. This work presents the use of a new plasmonic nanosensor employing self-induced back-action (SIBA) for optical trapping to enable SIBA-actuated nanopore electrophoresis (SANE) for quantifying antibody-ligand interactions. T-cell receptor-like antibodies (TCRmAbs) engineered to target peptide-presenting major histocompatibility complex (pMHC) ligands, representing a model of target ligands presented on the surface of cancer cells, were used to test the SANE sensor's ability to identify specific antibody-ligand binding. Cancer-irrelevant TCRmAbs targeting the same pMHCs were also tested as a control. It was found that the sensor could provide bimodal molecular signatures that could differentiate between antibody, ligand and the complexes that they formed, as well as distinguish between specific and non-specific interactions. Furthermore, the results suggested an interesting phenomenon of increased antibody-ligand complex bound fraction detected by the SANE sensor compared to that expected for corresponding bulk solution concentrations. A possible physical mechanism and potential advantages for the sensor's ability to augment complex formation near its active sensing volume at concentrations lower than the free solution equilibrium binding constant (K D ) are discussed.

4.
BMC Bioinformatics ; 19(Suppl 20): 504, 2018 Dec 21.
Article in English | MEDLINE | ID: mdl-30577741

ABSTRACT

BACKGROUND: The majority of cancer-related deaths are due to lung cancer, and there is a need for reliable diagnostic biomarkers to predict stages in non-small cell lung cancer cases. Recently, microRNAs were found to have potential as both biomarkers and therapeutic targets for lung cancer. However, some of the microRNA's functions are unknown, and their roles in cancer stage progression have been mostly undiscovered in this clinically and genetically heterogeneous disease. As evidence suggests that microRNA dysregulations are implicated in many diseases, it is essential to consider the changes in microRNA-target regulation across different lung cancer subtypes. RESULTS: We proposed a pipeline to identify microRNA synergistic modules with similar dysregulation patterns across multiple subtypes by constructing the MicroRNA Dysregulational Synergistic Network. From the network, we extracted microRNA modules and incorporated them as prior knowledge to the Sparse Group Lasso classifier. This leads to a more relevant selection of microRNA biomarkers, thereby improving the cancer stage classification accuracy. We applied our method to the TCGA Lung Adenocarcinoma and the Lung Squamous Cell Carcinoma datasets. In cross-validation tests, the area under ROC curve rate for the cancer stages prediction has increased considerably when incorporating the learned microRNA dysregulation modules. The extracted modules from multiple independent subtypes differential analyses were found to have high agreement with microRNA family annotations, and they can also be used to identify mutual biomarkers between different subtypes. Among the top-ranked candidate microRNAs selected by the model, 87% were reported to be related to Lung Adenocarcinoma. The overall result demonstrates that clustering microRNAs from the dysregulation pattern between microRNAs and their targets leads to biomarkers with high precision and recall rate to known differentially expressed disease-associated microRNAs. CONCLUSIONS: The results indicated that our method improves microRNA biomarker selection by detecting similar microRNA dysregulational synergistic patterns across the multiple subtypes. Since microRNA-target dysregulations are implicated in many cancers, we believe this tool can have broad applications for discovery of novel microRNA biomarkers in heterogeneous cancer diseases.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Lung Neoplasms/genetics , MicroRNAs/genetics , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/pathology , Biomarkers, Tumor/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cohort Studies , Gene Expression Profiling , Humans , Lung Neoplasms/pathology , MicroRNAs/metabolism , Neoplasm Staging , ROC Curve , Sample Size
5.
J Immunol ; 196(10): 4263-73, 2016 05 15.
Article in English | MEDLINE | ID: mdl-27183642

ABSTRACT

Class I HLA molecules mark infected cells for immune targeting by presenting pathogen-encoded peptides on the cell surface. Characterization of viral peptides unique to infected cells is important for understanding CD8(+) T cell responses and for the development of T cell-based immunotherapies. Having previously reported a series of West Nile virus (WNV) epitopes that are naturally presented by HLA-A*02:01, in this study we generated TCR mimic (TCRm) mAbs to three of these peptide/HLA complexes-the immunodominant SVG9 (E protein), the subdominant SLF9 (NS4B protein), and the immunorecessive YTM9 (NS3 protein)-and used these TCRm mAbs to stain WNV-infected cell lines and primary APCs. TCRm staining of WNV-infected cells demonstrated that the immunorecessive YTM9 appeared several hours earlier and at 5- to 10-fold greater density than the more immunogenic SLF9 and SVG9 ligands, respectively. Moreover, staining following inhibition of the TAP demonstrated that all three viral ligands were presented in a TAP-dependent manner despite originating from different cellular compartments. To our knowledge, this study represents the first use of TCRm mAbs to define the kinetics and magnitude of HLA presentation for a series of epitopes encoded by one virus, and the results depict a pattern whereby individual epitopes differ considerably in abundance and availability. The observations that immunodominant ligands can be found at lower levels and at later time points after infection suggest that a reevaluation of the factors that combine to shape T cell reactivity may be warranted.


Subject(s)
Antigen Presentation , CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Immunodominant Epitopes/immunology , West Nile virus/immunology , Animals , Antibodies, Monoclonal/immunology , Antibody Specificity , Cell Line, Tumor , Dendritic Cells/virology , Female , HLA-A Antigens/immunology , Humans , Mice , Mice, Inbred BALB C , Receptors, Antigen, T-Cell/immunology
6.
Eur J Immunol ; 44(7): 1936-46, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24723377

ABSTRACT

We used a newly generated T-cell receptor mimic monoclonal antibody (TCRm MAb) that recognizes a known nonself immunodominant peptide epitope from West Nile virus (WNV) NS4B protein to investigate epitope presentation after virus infection in C57BL/6 mice. Previous studies suggested that peptides of different length, either SSVWNATTAI (10-mer) or SSVWNATTA (9-mer) in complex with class I MHC antigen H-2D(b) , were immunodominant after WNV infection. Our data establish that both peptides are presented on the cell surface after WNV infection and that CD8(+) T cells can detect 10- and 9-mer length variants similarly. This result varies from the idea that a given T-cell receptor (TCR) prefers a single peptide length bound to its cognate class I MHC. In separate WNV infection studies with the TCRm MAb, we show that in vivo the 10-mer was presented on the surface of uninfected and infected CD8α(+) CD11c(+) dendritic cells, which suggests the use of direct and cross-presentation pathways. In contrast, CD11b(+) CD11c(-) cells bound the TCRm MAb only when they were infected. Our study demonstrates that TCR recognition of peptides is not limited to certain peptide lengths and that TCRm MAbs can be used to dissect the cell-type specific mechanisms of antigen presentation in vivo.


Subject(s)
Dendritic Cells/immunology , Immunodominant Epitopes , Receptors, Antigen, T-Cell/physiology , West Nile virus/immunology , Animals , CD11b Antigen/analysis , CD11c Antigen/analysis , CD8-Positive T-Lymphocytes/immunology , Mice , Mice, Inbred C57BL , Viral Nonstructural Proteins/immunology
7.
Bioact Mater ; 34: 422-435, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38282968

ABSTRACT

Cell membrane-derived nanoparticles (NPs) have recently gained popularity due to their desirable features in drug delivery such as mimicking properties of native cells, impeding systemic clearance, and altering foreign body responses. Besides NP technology, adoptive immunotherapy has emerged due to its promise in cancer specificity and therapeutic efficacy. In this research, we developed a biomimetic drug carrier based on chimeric antigen receptor (CAR) transduced T-cell membranes. For that purpose, anti-HER2 CAR-T cells were engineered via lentiviral transduction of anti-HER2 CAR coding lentiviral plasmids. Anti-HER2 CAR-T cells were characterized by their specific activities against the HER2 antigen and used for cell membrane extraction. Anti-cancer drug Cisplatin-loaded poly (D, l-lactide-co-glycolic acid) (PLGA) NPs were coated with anti-human epidermal growth factor receptor 2 (HER2)-specific CAR engineered T-cell membranes. Anti-HER2 CAR-T-cell membrane-coated PLGA NPs (CAR-T-MNPs) were characterized and confirmed via fluorescent microscopy and flow cytometry. Membrane-coated NPs showed a sustained drug release over the course of 21 days in physiological conditions. Cisplatin-loaded CAR-T-MNPs also inhibited the growth of multiple HER2+ cancer cells in vitro. In addition, in vitro uptake studies revealed that CAR-T-MNPs showed an increased uptake by A549 cells. These results were also confirmed via in vivo biodistribution and therapeutic studies using a subcutaneous lung cancer model in nude mice. CAR-T-MNPs localized preferentially at tumor areas compared to those of other studied groups and consisted of a significant reduction in tumor growth in tumor-bearing mice. In Conclusion, the new CAR modified cell membrane-coated NP drug-delivery platform has demonstrated its efficacy both in vitro and in vivo. Therefore, CAR engineered membrane-coated NP system could be a promising cell-mimicking drug carrier that could improve therapeutic outcomes of lung cancer treatments.

8.
J Immunol ; 186(5): 3265-76, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21282517

ABSTRACT

mAbs that recognize peptides presented on the cell surface by MHC class I molecules are potential therapeutic agents for cancer therapy. We have previously demonstrated that these Abs, which we termed TCR mimic mAbs (TCRm), reduce tumor growth in models of breast carcinoma. However, mechanisms of TCRm-mediated tumor growth reduction remain largely unknown. In this study, we report that these Abs, in contrast to several mAbs used currently in the clinic, destroy tumor cells independently of immune effector mechanisms such as Ab-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC). We found that TCRm-mediated apoptosis of tumor cells was associated with selective and specific binding of these Abs to peptide/HLA class I complexes, which triggered the activation of JNK and intrinsic caspase pathways. This signaling was accompanied by the release of mitochondrial cytochrome c and apoptosis-inducing factor. TCRm-induced apoptosis in tumor cells was completely inhibited by soluble MHC tetramers loaded with relevant peptide as well as with inhibitors for JNK and caspases. Furthermore, mAbs targeting MHC class I, independent of the peptide bound by HLA, did not stimulate apoptosis, suggesting that the Ab-binding site on the MHC/peptide complex determines cytotoxicity. This study suggests the existence of mechanisms, in addition to ADCC and CDC, through which these therapeutic Abs destroy tumor cells. These mechanisms would appear to be of particular importance in severely immunocompromised patients with advanced neoplastic disease, since immune cell-mediated killing of tumor cells through ADCC and CDC is substantially limited in these individuals.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antibodies, Neoplasm/therapeutic use , Apoptosis/immunology , Molecular Mimicry/immunology , Receptors, Antigen, T-Cell/therapeutic use , Xenograft Model Antitumor Assays/methods , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/metabolism , Antibodies, Neoplasm/administration & dosage , Antibodies, Neoplasm/metabolism , Cell Line, Tumor , Female , HLA-A2 Antigen/metabolism , Humans , Leukemia, Monocytic, Acute/immunology , Leukemia, Monocytic, Acute/pathology , Leukemia, Monocytic, Acute/therapy , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/pathology , Mammary Neoplasms, Experimental/therapy , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Melanoma, Experimental/therapy , Mice , Mice, Nude , Protein Binding/immunology , Receptors, Antigen, T-Cell/metabolism
9.
J Immunol ; 186(11): 6607-16, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21515791

ABSTRACT

This report describes a novel HLA/peptide complex with potential prognostic and therapeutic roles for invasive breast cancer. Macrophage migration inhibitory factor (MIF) mediates inflammation and immunity, and MIF overexpression is observed in breast cancer. We hypothesized that the HLA class I of cancerous breast epithelial cells would present MIF-derived peptides. Consistent with this hypothesis, the peptide FLSELTQQL (MIF(19-27)) was eluted from the HLA-A*0201 (HLA-A2) of breast cancer cell lines. We posited that if this MIF(19-27)/HLA-A2 complex was exclusively found in invasive breast cancer, it could be a useful prognostic indicator. To assess the presentation of MIF peptides by the HLA of various cells and tissues, mice were immunized with the MIF(19-27)/HLA-A2 complex. The resulting mAb (RL21A) stained invasive ductal carcinoma (IDC) but not ductal carcinoma in situ, fibroadenoma, or normal breast tissues. RL21A did not stain WBCs (total WBCs) or normal tissues from deceased HLA-A2 donors, substantiating the tumor-specific nature of this MIF/HLA complex. As this MIF/HLA complex appeared specific to the surface of IDC, RL21A was tested as an immunotherapeutic for breast cancer in vitro and in vivo. In vitro, RL21A killed the MDA-MB-231 cell line via complement and induction of apoptosis. In an in vivo orthotopic mouse model, administration of RL21A reduced MDA-MB-231 and BT-20 tumor burden by 5-fold and by >2-fold, respectively. In summary, HLA-presented MIF peptides show promise as prognostic cell surface indicators for IDC and as targets for immunotherapeutic intervention.


Subject(s)
Antibodies, Monoclonal/immunology , Breast Neoplasms/immunology , HLA-A Antigens/immunology , Macrophage Migration-Inhibitory Factors/immunology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/therapeutic use , Antibody Affinity/immunology , Antibody Specificity/immunology , Apoptosis/drug effects , Apoptosis/immunology , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/drug therapy , Carcinoma, Ductal, Breast/immunology , Carcinoma, Ductal, Breast/pathology , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/immunology , Dose-Response Relationship, Drug , Female , HLA-A Antigens/metabolism , HLA-A2 Antigen , Humans , Kinetics , Macrophage Migration-Inhibitory Factors/chemistry , Macrophage Migration-Inhibitory Factors/metabolism , Mice , Mice, Nude , Peptides/immunology , Peptides/metabolism , Prognosis , Protein Binding/immunology , Xenograft Model Antitumor Assays
10.
J Comput Biol ; 30(7): 766-782, 2023 07.
Article in English | MEDLINE | ID: mdl-37437088

ABSTRACT

The development of tools for the annotation of genes from newly sequenced species has not evolved much from homologous alignment to prior annotated species. While the quality of gene annotations continues to decline as we sequence and assemble more evolutionary distant gut microbiome species, machine learning presents a high quality alternative to traditional techniques. In this study, we investigate the relative performance of common classical and nonclassical machine learning algorithms in the problem of gene annotation using human microbiome-associated species genes from the KEGG database. The majority of the ensemble, clustering, and deep learning algorithms that we investigated showed higher prediction accuracy than CD-Hit in predicting partial KEGG function. Motif-based, machine-learning methods of annotation in new species were faster and had higher precision-recall than methods of homologous alignment or orthologous gene clustering. Gradient boosted ensemble methods and neural networks also predicted higher connectivity in reconstructed KEGG pathways, finding twice as many new pathway interactions than blast alignment. The use of motif-based, machine-learning algorithms in annotation software will allow researchers to develop powerful tools to interact with bacterial microbiomes in ways previously unachievable through homologous sequence alignment alone.


Subject(s)
Algorithms , Genes, Microbial , Humans , Molecular Sequence Annotation , Neural Networks, Computer , Machine Learning
11.
Cell Rep ; 42(12): 113516, 2023 12 26.
Article in English | MEDLINE | ID: mdl-38048225

ABSTRACT

The immune checkpoint NKG2A/CD94 is a promising target for cancer immunotherapy, and its ligand major histocompatibility complex E (MHC-E) is frequently upregulated in cancer. NKG2A/CD94-mediated inhibition of lymphocytes depends on the presence of specific leader peptides in MHC-E, but when and where they are presented in situ is unknown. We apply a nanobody specific for the Qdm/Qa-1b complex, the NKG2A/CD94 ligand in mouse, and find that presentation of Qdm peptide depends on every member of the endoplasmic reticulum-resident peptide loading complex. With a turnover rate of 30 min, the Qdm peptide reflects antigen processing capacity in real time. Remarkably, Qdm/Qa-1b complexes require inflammatory signals for surface expression in situ, despite the broad presence of Qa-1b molecules in homeostasis. Furthermore, we identify LILRB1 as a functional inhibition receptor for MHC-E in steady state. These data provide a molecular understanding of NKG2A blockade in immunotherapy and assign MHC-E as a convergent ligand for multiple immune checkpoints.


Subject(s)
Histocompatibility Antigens Class I , Neoplasms , Mice , Animals , Histocompatibility Antigens Class I/metabolism , Leukocyte Immunoglobulin-like Receptor B1/metabolism , Killer Cells, Natural , Ligands , Peptides/metabolism , Neoplasms/metabolism , NK Cell Lectin-Like Receptor Subfamily C/metabolism
12.
J Immunol ; 184(4): 2156-65, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-20065111

ABSTRACT

Our laboratory has developed a process for generating mAbs with selectivity to unique peptides in the context of MHC molecules. Recently, we reported that RL4B, an mAb that we have called a TCR mimic (TCRm) because it recognizes peptide in the context of MHC, has cytotoxic activity in vitro and prevented growth of tumor cells in a prophylactic setting. When presented in the context of HLA-A2, RL4B TCRm recognizes the peptide GVLPALPQV derived from human chorionic gonadotropin (hCG)-beta. In this study, we show that RL4B TCRm has strong binding affinity for the GVLPALPQV peptide/HLA-A2 epitope and fine binding specificity for cells that express endogenous hCGbeta Ag and HLA-A2. In addition, suppression of tumor growth with RL4B TCRm was observed in orthotopic models for breast cancer. Using two aggressive human tumor cell lines, MDA-MB-231 and MCF-7, we provide evidence that RL4B TCRm significantly retards tumor growth, supporting a possible role for TCRm agents in therapeutic settings. Moreover, tumors in mice responded to RL4B TCRm therapy in a dose-dependent manner, eliminating tumors at the highest dose. RL4B TCRm strongly detects the hCGbeta peptide/HLA-A2 epitope in human primary breast tumor tissue, but does not react or reacts weakly with normal breast tissue from the same patient. These results further illustrate the selective nature of TCRm Abs and the clinical relevance of the GVLPALPQV peptide/HLA-A2 epitope expression in tumor cells, because they provide the first evidence that Abs that mimic the TCR can be used to markedly reduce and suppress tumor growth.


Subject(s)
Antibodies, Monoclonal/metabolism , Antibody Affinity , Antibody Specificity , Breast Neoplasms/therapy , Chorionic Gonadotropin, beta Subunit, Human/immunology , Growth Inhibitors/physiology , Molecular Mimicry/immunology , Peptide Fragments/metabolism , Animals , Antibodies, Monoclonal/physiology , Antibodies, Monoclonal/therapeutic use , Antigen Presentation/genetics , Antigen Presentation/immunology , Antineoplastic Agents/metabolism , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/immunology , Biomarkers, Tumor/metabolism , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Cell Line, Transformed , Cell Line, Tumor , Chorionic Gonadotropin, beta Subunit, Human/metabolism , Cytotoxicity Tests, Immunologic , Epitopes, T-Lymphocyte/biosynthesis , Epitopes, T-Lymphocyte/genetics , Gene Targeting , Growth Inhibitors/therapeutic use , HLA-A Antigens/immunology , HLA-A Antigens/metabolism , HLA-A2 Antigen , Humans , Mice , Mice, Nude , Peptide Fragments/immunology
13.
J Immunol ; 184(8): 4423-30, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20212098

ABSTRACT

The generation of a robust CD8(+) T cell response is an ongoing challenge for the development of DNA vaccines. One problem encountered with classical DNA plasmid immunization is that peptides produced are noncovalently and transiently associated with MHC class I molecules and thus may not durably stimulate CD8(+) T cell responses. To address this and enhance the expression and presentation of the antigenic peptide/MHC complexes, we generated single-chain trimers (SCTs) composed of a single polypeptide chain with a linear composition of antigenic peptide, beta(2)-microglobulin, and H chain connected by flexible linkers. In this study, we test whether the preassembled nature of the SCT makes them effective for eliciting protective CD8(+) T cell responses against pathogens. A DNA plasmid was constructed encoding an SCT incorporating the human MHC class I molecule HLA-A2 and the immunodominant peptide SVG9 derived from the envelope protein of West Nile virus (WNV). HLA-A2 transgenic mice vaccinated with the DNA encoding the SVG9/HLA-A2 SCT generated a robust epitope-specific CD8(+) T cell response and showed enhanced survival rate and lower viral burden in the brain after lethal WNV challenge. Inclusion of a CD4(+) Th cell epitope within the SCT did not increase the frequency of SVG9-specific CD8(+) T cells, but did enhance protection against WNV challenge. Overall, these findings demonstrate that the SCT platform can induce protective CD8(+) T cell responses against lethal virus infection and may be paired with immunogens that elicit robust neutralizing Ab responses to generate vaccines that optimally activate all facets of adaptive immunity.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , HLA-A2 Antigen/immunology , Immunodominant Epitopes/immunology , Vaccines, Subunit/immunology , West Nile Fever/prevention & control , West Nile Virus Vaccines/immunology , West Nile virus/immunology , Animals , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/virology , Cells, Cultured , Cytotoxicity, Immunologic/genetics , Disease Models, Animal , Epitopes, T-Lymphocyte/administration & dosage , Epitopes, T-Lymphocyte/genetics , HLA-A2 Antigen/administration & dosage , HLA-A2 Antigen/genetics , Humans , Immunodominant Epitopes/administration & dosage , Immunodominant Epitopes/genetics , Mice , Mice, Transgenic , Protein Binding/genetics , Protein Binding/immunology , Survival Analysis , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , T-Lymphocytes, Helper-Inducer/virology , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics , Vaccines, DNA/immunology , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/genetics , Viral Load/genetics , Viral Load/immunology , West Nile Fever/immunology , West Nile Fever/mortality , West Nile Virus Vaccines/administration & dosage , West Nile Virus Vaccines/genetics , West Nile virus/pathogenicity
14.
Methods Mol Biol ; 2394: 343-376, 2022.
Article in English | MEDLINE | ID: mdl-35094337

ABSTRACT

We fabricated a novel single molecule nanosensor by integrating a solid-state nanopore and a double nanohole nanoaperture. The nanosensor employs Self-Induced Back-Action (SIBA) for optical trapping and enables SIBA-Actuated Nanopore Electrophoresis (SANE) for concurrent acquisition of bimodal optical and electrical signatures of molecular interactions. This work describes how to fabricate and use the SANE sensor to quantify antibody-ligand interactions. We describe how to analyze the bimodal optical-electrical data to improve upon the discrimination of antibody and ligand versus bound complex compared to electrical measurements alone. Example results for specific interaction detection are described for T-cell receptor-like antibodies (TCRmAbs) engineered to target peptide-presenting Major Histocompatibility Complex (pMHC) ligands, representing a model of target ligands presented on the surface of cancer cells. We also describe how to analyze the bimodal optical-electrical data to discriminate between specific and non-specific interactions between antibodies and ligands. Example results for non-specific interactions are shown for cancer-irrelevant TCRmAbs targeting the same pMHCs, as a control. These example results demonstrate the utility of the SANE sensor as a potential screening tool for ligand targets in cancer immunotherapy, though we believe that its potential uses are much broader.


Subject(s)
Nanopores , Neoplasms , Electrophoresis , Immunotherapy , Ligands , Nanotechnology/methods
15.
J Cell Physiol ; 225(3): 664-72, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20506235

ABSTRACT

Organ-specific vascular targeting, for example, to the blood-brain barrier, requires the identification of unique molecular addresses on a subset of endothelial cells. The present study describes a crucial step towards tapping the exquisite specificity of the peptide/HLA class I system for this goal. We utilized a novel T-cell receptor (TCR) mimic antibody of high affinity and specificity, which is restricted by HLA-A2 and has been generated to recognize a peptide epitope derived from p68 RNA helicase (YLLPAIVHI). The parent protein is highly expressed by brain endothelial cells. Flow cytometry and confocal imaging showed that the antibody binds to HLA-A2-positive human brain-derived endothelial cells, both immortalized hCMEC/D3 cells and primary cells. The TCR mimic antibody undergoes internalization into vesicles, where significant colocalization occurs with the early endosomal marker EEA-1, but barely with caveolin-1. To our knowledge internalization of neither MHC class I protein nor TCR mimics by brain endothelial cells has been previously observed. Knock down of p68 protein expression by siRNA reduced the presentation of YLLPAIVHI-peptide/HLA-A2 complexes on the cell membrane by half as measured by flow cytometry 48 h later. We also found that brain endothelial cells isolated from HLA-A2 transgenic mouse strains express the A2 transgene, and brain endothelial cells of one of these strains also present YLLPAIVHI-peptide/HLA-A2, making these mouse strains suitable models for studying TCR mimic antibodies in vivo. In conclusion, these data strongly support the notion that TCR mimic antibodies could be a new class of therapeutic targeting agents in a wide variety of diseases.


Subject(s)
Antibodies, Monoclonal , Blood-Brain Barrier/immunology , Brain/blood supply , DEAD-box RNA Helicases/immunology , Endothelial Cells/immunology , HLA-A2 Antigen/immunology , Immunomagnetic Separation , Receptors, Antigen, T-Cell/immunology , Animals , Antibodies, Monoclonal/metabolism , Antibody Affinity , Antibody Specificity , Blood-Brain Barrier/cytology , Blood-Brain Barrier/metabolism , Caveolin 1/metabolism , Cells, Cultured , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Endocytosis , Endosomes/immunology , Endosomes/metabolism , Endothelial Cells/metabolism , Flow Cytometry , HLA-A2 Antigen/genetics , Humans , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Transgenic , Microscopy, Confocal , Molecular Mimicry , Platelet Endothelial Cell Adhesion Molecule-1/immunology , RNA Interference , Receptors, Antigen, T-Cell/metabolism , Vesicular Transport Proteins/metabolism
16.
Cancer Immunol Immunother ; 59(4): 563-73, 2010 Apr.
Article in English | MEDLINE | ID: mdl-19779714

ABSTRACT

The identification and validation of new cancer-specific T cell epitopes continues to be a major area of research interest. Nevertheless, challenges remain to develop strategies that can easily discover and validate epitopes expressed in primary cancer cells. Regarded as targets for T cells, peptides presented in the context of the major histocompatibility complex (MHC) are recognized by monoclonal antibodies (mAbs). These mAbs are of special importance as they lend themselves to the detection of epitopes expressed in primary tumor cells. Here, we use an approach that has been successfully utilized in two different infectious disease applications (WNV and influenza). A direct peptide-epitope discovery strategy involving mass spectrometric analysis led to the identification of peptide YLLPAIVHI in the context of MHC A*02 allele (YLL/A2) from human breast carcinoma cell lines. We then generated and characterized an anti-YLL/A2 mAb designated as RL6A TCRm. Subsequently, the TCRm mAb was used to directly validate YLL/A2 epitope expression in human breast cancer tissue, but not in normal control breast tissue. Moreover, mice implanted with human breast cancer cells grew tumors, yet when treated with RL6A TCRm showed a marked reduction in tumor size. These data demonstrate for the first time a coordinated direct discovery and validation strategy that identified a peptide/MHC complex on primary tumor cells for antibody targeting and provide a novel approach to cancer immunotherapy.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Breast Neoplasms/diagnosis , Breast Neoplasms/therapy , Epitopes, T-Lymphocyte/immunology , Peptide Fragments/therapeutic use , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Antibodies, Monoclonal/immunology , Antigens, Neoplasm/immunology , Blotting, Western , Breast/metabolism , Breast/pathology , Breast Neoplasms/immunology , Cancer Vaccines/therapeutic use , DEAD-box RNA Helicases/immunology , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Histocompatibility Antigens Class I/immunology , Humans , Immunoenzyme Techniques , Mice , Mice, Nude , Molecular Mimicry , Peptide Fragments/immunology , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Surface Plasmon Resonance
17.
Article in English | MEDLINE | ID: mdl-32850765

ABSTRACT

Melanoma is one of the most aggressive skin cancers, and the American Cancer Society reports that every hour, one person dies from melanoma. While there are a number of treatments currently available for melanoma (e.g., surgery, chemotherapy, immunotherapy, and radiation therapy), they face several problems including inadequate response rates, high toxicity, severe side effects due to non-specific targeting of anti-cancer drugs, and the development of multidrug resistance during prolonged treatment. To improve chemo-drug therapeutic efficiency and overcome these mentioned limitations, a multifunctional nanoparticle has been developed to effectively target and treat melanoma. Specifically, poly (lactic-co-glycolic acid) (PLGA) nanoparticles (NPs) were coated with a cellular membrane derived from the T cell hybridoma, 19LF6 endowed with a melanoma-specific anti-gp100/HLA-A2 T-cell receptor (TCR) and loaded with an FDA-approved melanoma chemotherapeutic drug Trametinib. T-cell membrane camouflaged Trametinib loaded PLGA NPs displayed high stability, hemo- and cyto-compatibility. They also demonstrated membrane coating dependent drug release profiles with the most sustained release from the NPs proportional with the highest amount of membrane used. 19LF6 membrane-coated NPs produced a threefold increase in cellular uptake toward the melanoma cell line in vitro compared to that of the bare nanoparticle. Moreover, the binding kinetics and cellular uptake of these particles were shown to be membrane/TCR concentration-dependent. The in vitro cancer killing efficiencies of these NPs were significantly higher compared to other NP groups and aligned with binding and uptake characteristics. Particles with the higher membrane content (greater anti-gp100 TCR content) were shown to be more effective when compared to the free drug and negative controls. In vivo biodistribution studies displayed the theragnostic capabilities of these NPs with more than a twofold increase in the tumor retention compared to the uncoated and non-specific membrane coated groups. Based on these studies, these T-cell membrane coated NPs emerge as a potential theragnostic carrier for imaging and therapy applications associated with melanoma.

18.
Int J Oncol ; 34(3): 597-608, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19212664

ABSTRACT

The ability of aspirin to trigger apoptosis in cancer cells is well known and is consistent with the clinical and epidemiological evidence on its chemopreventive effects in curtailing epithelial cancers, including breast cancer. We hypothesized that the anticancer effects of aspirin may involve acetylation of the tumor suppressor protein p53, a known regulator of apoptosis. In the present study, we determined if aspirin at the physiologically achievable concentration of 100 microM acetylates p53 and modulates the expression of p21CIP1, a protein involved in cell cycle arrest, and Bax, a pro-apoptotic protein. Using MDA-MB-231 human breast cancer cells, we demonstrate that aspirin at 100 microM concentration markedly acetylated the p53 protein, which was primarily localized to the nucleus. Aspirin induced p21CIP1 protein levels in a transient fashion in contrast to the sustained induction of Bax. The induction of p21CIP1 protein levels began at 3 h and was maximal at 6-8 h; however, it decreased to control levels by 30 h. In contrast, the anticancer drug, camptothecin (CPT) induced a steady accumulation of p21CIP1 protein. Remarkably, when cells were co-treated with aspirin and CPT, p21CIP1 levels were drastically downregulated, and this phenomenon was observed in many cancer cell lines. Incubation of recombinant p21 with cytoplasmic extracts from aspirin-treated cells caused its degradation suggesting the involvement of proteases in the disappearance of p21CIP1. Consistent with this data, aspirin decreased the survival of CPT-treated cells and greatly increased the extent of apoptosis. Our observation that aspirin has the ability to inhibit p21CIP1 after its initial induction has important implications in chemotherapy, and suggests its potential use to increase the efficacy of anticancer agents.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Aspirin/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Camptothecin/pharmacology , Cyclin-Dependent Kinase Inhibitor p21/biosynthesis , Acetylation/drug effects , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Camptothecin/antagonists & inhibitors , Cell Nucleus/metabolism , Cyclin-Dependent Kinase Inhibitor p21/antagonists & inhibitors , Cyclin-Dependent Kinase Inhibitor p21/genetics , DNA, Neoplasm/metabolism , Drug Synergism , Humans , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Tumor Cells, Cultured , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/metabolism , bcl-2-Associated X Protein/biosynthesis
19.
J Immunol Methods ; 318(1-2): 47-58, 2007 Jan 10.
Article in English | MEDLINE | ID: mdl-17134715

ABSTRACT

Gene and protein expression studies demonstrate that viral-infected and malignant cells undergo a complex series of transcriptional and translational changes. As class I MHC molecules reflect the proteome (and changes therein) by presenting intracellular peptide epitopes, the development of a direct discovery and validation technology for the identification of these epitopes is needed. We developed our technology using HIV-1-infected cells as a model. A combination of hollow fiber class I HLA protein production and mass spectrometric epitope analysis indicated a 3-fold increase in the host-peptide VLMTEDIKL(720-728), [eIF4G((720))] presented by the HLA-A*0201 of HIV-1-infected cells. This peptide is derived from the host-protein translation of eukaryotic initiation factor 4-gamma (eIF4G) that plays a pivotal role in cellular protein synthesis. Direct confirmation of expression of this self-encoded antigen was performed through development of a T cell receptor mimic (TCRm) monoclonal antibody (mAb). The resulting 4F7 TCRm demonstrated specific recognition of the eIF4G((720))-A*0201 complex. Staining of normal PBMCs with 4F7 showed only low levels of endogenous eIF4G((720)) presentation by HLA-A*0201, while 4F7 staining of HIV-1-infected PBMCs revealed an approximately 3-fold increase in eIF4G((720))-A*0201. The MHC-peptide complex was initially detectable by 4F7 at 3 days post-infection, with a steady increase through day 8. We therefore demonstrate the successful development and implementation of an integrated discovery and validation technology system for direct identification and confirmation of class I MHC-peptide epitopes on cells.


Subject(s)
Autoantigens/analysis , Epitopes, T-Lymphocyte/analysis , Histocompatibility Antigens Class I/immunology , Peptides/analysis , Antibodies, Monoclonal/immunology , Antibody Specificity/immunology , Antigen-Antibody Reactions/immunology , Autoantigens/immunology , Binding, Competitive/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , Cell Line, Tumor , Epithelial Cells/immunology , Epitopes, T-Lymphocyte/immunology , Eukaryotic Initiation Factor-4G/immunology , Flow Cytometry , HIV-1/immunology , HLA-A Antigens/immunology , HLA-A2 Antigen , Humans , Kinetics , Mass Spectrometry , Peptide Fragments/analysis , Peptide Fragments/immunology , Peptides/immunology , T-Lymphocytes/immunology , T-Lymphocytes/virology
20.
J Immunol Methods ; 321(1-2): 86-93, 2007 Apr 10.
Article in English | MEDLINE | ID: mdl-17336323

ABSTRACT

No experimental animal model employing a primary human ovarian carcinoma (OC) cell line is presently available that tracks the progression of this cell line with an identifiable marker. This hinders investigations related to developing new approaches for treating OC. Here, we describe the development of a tumor model in NOD/SCID mice for human OC that makes use of the endogenously expressed tumor specific sperm protein 17 (Sp17) cancer testis antigen. In this model, human SKOV-3 OC cell lines were intra-peritoneally seeded. Subsequently viable SKOV-3 cells were recovered from primary organ cell cultures from the liver ovaries, abdomen, and ascitic fluid, and their presence was confirmed by the detection of Sp17 mRNA by RT-PCR and Sp17 protein by immunocytochemistry and FACS analysis. When SKOV-3 tumor cells were administered intravenously the mice developed primarily lung tumor foci. This model makes it possible to evaluate new immunotherapeutic strategies for the treatment of human OC based on the biomarker Sp17.


Subject(s)
Antigens, Surface/metabolism , Biomarkers, Tumor/metabolism , Carrier Proteins/metabolism , Gene Expression Regulation, Neoplastic , Lung Neoplasms/metabolism , Neoplasms, Experimental/metabolism , Ovarian Neoplasms/metabolism , Animals , Antigens, Surface/genetics , Biomarkers, Tumor/genetics , Calmodulin-Binding Proteins , Carrier Proteins/genetics , Cell Line, Tumor , Cell Separation , Disease Progression , Female , Flow Cytometry , Humans , Immunohistochemistry , Injections, Intraperitoneal , Injections, Intravenous , Lung Neoplasms/diagnosis , Lung Neoplasms/genetics , Lung Neoplasms/secondary , Membrane Proteins , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation/methods , Neoplasms, Experimental/diagnosis , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Ovarian Neoplasms/diagnosis , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL