7 results on '"Bangari, Dinesh S."'
Search Results
2. A trispecific antibody targeting HER2 and T cells inhibits breast cancer growth via CD4 cells.
- Author
-
Seung E, Xing Z, Wu L, Rao E, Cortez-Retamozo V, Ospina B, Chen L, Beil C, Song Z, Zhang B, Levit M, Deng G, Hebert A, Kirby P, Li A, Poulton EJ, Vicente R, Garrigou A, Piepenhagen P, Ulinski G, Sanicola-Nadel M, Bangari DS, Qiu H, Pao L, Wiederschain D, Wei R, Yang ZY, and Nabel GJ
- Subjects
- Animals, CD28 Antigens metabolism, CD4-Positive T-Lymphocytes, CD8-Positive T-Lymphocytes, Female, Humans, Mice, Receptor, ErbB-2 genetics, Breast Neoplasms drug therapy, Breast Neoplasms metabolism
- Abstract
Effective antitumour immunity depends on the orchestration of potent T cell responses against malignancies
1 . Regression of human cancers has been induced by immune checkpoint inhibitors, T cell engagers or chimeric antigen receptor T cell therapies2-4 . Although CD8 T cells function as key effectors of these responses, the role of CD4 T cells beyond their helper function has not been defined. Here we demonstrate that a trispecific antibody to HER2, CD3 and CD28 stimulates regression of breast cancers in a humanized mouse model through a mechanism involving CD4-dependent inhibition of tumour cell cycle progression. Although CD8 T cells directly mediated tumour lysis in vitro, CD4 T cells exerted antiproliferative effects by blocking cancer cell cycle progression at G1/S. Furthermore, when T cell subsets were adoptively transferred into a humanized breast cancer tumour mouse model, CD4 T cells alone inhibited HER2+ breast cancer growth in vivo. RNA microarray analysis revealed that CD4 T cells markedly decreased tumour cell cycle progression and proliferation, and also increased pro-inflammatory signalling pathways. Collectively, the trispecific antibody to HER2 induced T cell-dependent tumour regression through direct antitumour and indirect pro-inflammatory/immune effects driven by CD4 T cells., (© 2022. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2022
- Full Text
- View/download PDF
3. Blood phenylalanine reduction reverses gene expression changes observed in a mouse model of phenylketonuria.
- Author
-
Manek R, Zhang YV, Berthelette P, Hossain M, Cornell CS, Gans J, Anarat-Cappillino G, Geller S, Jackson R, Yu D, Singh K, Ryan S, Bangari DS, Xu EY, and Kyostio-Moore SRM
- Subjects
- Animals, Biomarkers blood, Brain metabolism, Brain pathology, Disease Models, Animal, Down-Regulation, Gene Expression Profiling, Male, Mice, Knockout, Phenylalanine Ammonia-Lyase genetics, Phenylalanine Hydroxylase genetics, Phenylketonurias blood, Phenylketonurias genetics, Phenylketonurias pathology, Proteome, Proteomics, Mice, Genetic Therapy, Liver enzymology, Phenylalanine blood, Phenylalanine Ammonia-Lyase metabolism, Phenylalanine Hydroxylase metabolism, Phenylketonurias therapy, Transcriptome
- Abstract
Phenylketonuria (PKU) is a genetic deficiency of phenylalanine hydroxylase (PAH) in liver resulting in blood phenylalanine (Phe) elevation and neurotoxicity. A pegylated phenylalanine ammonia lyase (PEG-PAL) metabolizing Phe into cinnamic acid was recently approved as treatment for PKU patients. A potentially one-time rAAV-based delivery of PAH gene into liver to convert Phe into tyrosine (Tyr), a normal way of Phe metabolism, has now also entered the clinic. To understand differences between these two Phe lowering strategies, we evaluated PAH and PAL expression in livers of PAH
enu2 mice on brain and liver functions. Both lowered brain Phe and increased neurotransmitter levels and corrected animal behavior. However, PAL delivery required dose optimization, did not elevate brain Tyr levels and resulted in an immune response. The effect of hyperphenylalanemia on liver functions in PKU mice was assessed by transcriptome and proteomic analyses. We observed an elevation in Cyp4a10/14 proteins involved in lipid metabolism and upregulation of genes involved in cholesterol biosynthesis. Majority of the gene expression changes were corrected by PAH and PAL delivery though the role of these changes in PKU pathology is currently unclear. Taken together, here we show that blood Phe lowering strategy using PAH or PAL corrects both brain pathology as well as previously unknown lipid metabolism associated pathway changes in liver., (© 2021. The Author(s).)- Published
- 2021
- Full Text
- View/download PDF
4. CRISPR/Cas9 generated knockout mice lacking phenylalanine hydroxylase protein as a novel preclinical model for human phenylketonuria.
- Author
-
Singh K, Cornell CS, Jackson R, Kabiri M, Phipps M, Desai M, Fogle R, Ying X, Anarat-Cappillino G, Geller S, Johnson J, Roberts E, Malley K, Devlin T, DeRiso M, Berthelette P, Zhang YV, Ryan S, Rao S, Thurberg BL, Bangari DS, and Kyostio-Moore S
- Subjects
- Animals, Male, Mice, Mice, Knockout, CRISPR-Cas Systems, Disease Models, Animal, Gene Knockout Techniques, Phenylalanine Hydroxylase genetics, Phenylketonurias genetics
- Abstract
Phenylketonuria (PKU) is an autosomal recessive inborn error of L-phenylalanine (Phe) metabolism. It is caused by a partial or complete deficiency of the enzyme phenylalanine hydroxylase (PAH), which is necessary for conversion of Phe to tyrosine (Tyr). This metabolic error results in buildup of Phe and reduction of Tyr concentration in blood and in the brain, leading to neurological disease and intellectual deficits. Patients exhibit retarded body growth, hypopigmentation, hypocholesterolemia and low levels of neurotransmitters. Here we report first attempt at creating a homozygous Pah knock-out (KO) (Hom) mouse model, which was developed in the C57BL/6 J strain using CRISPR/Cas9 where codon 7 (GAG) in Pah gene was changed to a stop codon TAG. We investigated 2 to 6-month-old, male, Hom mice using comprehensive behavioral and biochemical assays, MRI and histopathology. Age and sex-matched heterozygous Pah-KO (Het) mice were used as control mice, as they exhibit enough PAH enzyme activity to provide Phe and Tyr levels comparable to the wild-type mice. Overall, our findings demonstrate that 6-month-old, male Hom mice completely lack PAH enzyme, exhibit significantly higher blood and brain Phe levels, lower levels of brain Tyr and neurotransmitters along with lower myelin content and have significant behavioral deficit. These mice exhibit phenotypes that closely resemble PKU patients such as retarded body growth, cutaneous hypopigmentation, and hypocholesterolemia when compared to the age- and sex-matched Het mice. Altogether, biochemical, behavioral, and pathologic features of this novel mouse model suggest that it can be used as a reliable translational tool for PKU preclinical research and drug development.
- Published
- 2021
- Full Text
- View/download PDF
5. Skeletal Characterization of the Fgfr3 Mouse Model of Achondroplasia Using Micro-CT and MRI Volumetric Imaging.
- Author
-
Shazeeb MS, Cox MK, Gupta A, Tang W, Singh K, Pryce CT, Fogle R, Mu Y, Weber WD, Bangari DS, Ying X, and Sabbagh Y
- Subjects
- Achondroplasia genetics, Achondroplasia mortality, Animals, Disease Models, Animal, Humans, Kyphosis etiology, Magnetic Resonance Imaging, Mice, Mice, Transgenic, Mutation, Spine diagnostic imaging, Survival Rate, X-Ray Microtomography, Achondroplasia diagnostic imaging, Kyphosis diagnostic imaging, Receptor, Fibroblast Growth Factor, Type 3 genetics, Spine abnormalities
- Abstract
Achondroplasia, the most common form of dwarfism, affects more than a quarter million people worldwide and remains an unmet medical need. Achondroplasia is caused by mutations in the fibroblast growth factor receptor 3 (FGFR3) gene which results in over-activation of the receptor, interfering with normal skeletal development leading to disproportional short stature. Multiple mouse models have been generated to study achondroplasia. The characterization of these preclinical models has been primarily done with 2D measurements. In this study, we explored the transgenic model expressing mouse Fgfr3 containing the achondroplasia mutation G380R under the Col2 promoter (Ach). Survival and growth rate of the Ach mice were reduced compared to wild-type (WT) littermates. Axial skeletal defects and abnormalities of the sternebrae and vertebrae were observed in the Ach mice. Further evaluation of the Ach mouse model was performed by developing 3D parameters from micro-computed tomography (micro-CT) and magnetic resonance imaging (MRI). The 3-week-old mice showed greater differences between the Ach and WT groups compared to the 6-week-old mice for all parameters. Deeper understanding of skeletal abnormalities of this model will help guide future studies for evaluating novel and effective therapeutic approaches for the treatment of achondroplasia.
- Published
- 2018
- Full Text
- View/download PDF
6. Immunocompetent mouse model of breast cancer for preclinical testing of EphA2-targeted therapy.
- Author
-
Noblitt LW, Bangari DS, Shukla S, Mohammed S, and Mittal SK
- Subjects
- Adenoviridae genetics, Animals, Breast Neoplasms veterinary, Disease Progression, Female, Genetic Therapy methods, Genetic Vectors, Mice, Phosphorylation, Receptor, EphA2 biosynthesis, Receptor, EphA2 metabolism, Tumor Cells, Cultured, Breast Neoplasms genetics, Breast Neoplasms therapy, Disease Models, Animal, Immunocompetence, Receptor, EphA2 genetics
- Abstract
EphA2, a receptor tyrosine kinase, is elevated in many invasive human breast cancers, and the majority of EphA2 remains unphosphorylated. The successful attachment of ligand EphrinA1 present on the surface of adjacent cells to EphA2 initiates EphA2 phosphorylation leading to its turnover. In vivo efficacy of various approaches targeting EphA2 for breast cancer therapy is usually evaluated in nude mice bearing human breast cancer xenografts. In order to establish an immunocompetent mouse model of breast cancer for EphA2-targeted therapies, we evaluated a mouse breast cancer cell line (MT1A2) for EphA2 expression and phosphorylation. Overexpression of EphA2 was observed in MT1A2 cells and the majority of it remained unphosphorylated signifying that EphA2 in MT1A2 cells behaved similar to that of human breast cancer cells. Human adenovirus subtype 5 (HAd5) vectors expressing secretory forms of EphrinA1 were used for in vitro and in vivo targeting of MT1A2-derived EphA2. MT1A2 cells infected with HAd-EphrinA1-Fc (HAd expressing extracellular domain of human EphrinA1 attached to Fc portion of human IgG1 heavy chain) induced EphA2 activation and its turnover. This led to inhibition in MT1A2 cell colony formation in soft agar and cell viability in monolayer culture. In addition, MT1A2 cells-infected with HAd-EphrinA1-Fc failed to form tumors in syngeneic FVB/n mice at least 32 days postinoculation. Moreover, intratumoral inoculation of FVB/n mice-bearing MT1A2-induced tumors with HAd-EphrinA1-Fc slowed the tumor growth and also resulted in the development of vector-specific immune response. These results indicate that FVB/n mice-bearing MT1A2-induced tumors could serve as an immunocompetent model of breast cancer for EphA2-targeted therapeutic strategies.
- Published
- 2005
- Full Text
- View/download PDF
7. Decreased tumorigenic potential of EphA2-overexpressing breast cancer cells following treatment with adenoviral vectors that express EphrinA1.
- Author
-
Noblitt LW, Bangari DS, Shukla S, Knapp DW, Mohammed S, Kinch MS, and Mittal SK
- Subjects
- Breast Neoplasms genetics, Cell Line, Tumor, Humans, Injections, Intralesional, Phosphorylation, Adenoviridae genetics, Breast Neoplasms pathology, Ephrin-A1 genetics, Genetic Vectors, Receptor, EphA2 genetics
- Abstract
The EphA2 receptor tyrosine kinase is frequently overexpressed in invasive breast cancer cells. Moreover, these malignant cells have unstable cell-cell contacts, which preclude EphA2 from interacting with its ligand, EphrinA1, which is anchored to the membrane of adjacent cells. This defect is important because ligand binding causes EphA2 to transmit signals that negatively regulate tumor cell growth and survival, whereas the absence of ligand binding favors these same behaviors. In our present study, human adenoviral type 5 (HAd) vectors were engineered to express secreted-forms of EphrinA1. These vectors were used to infect MDA-MB-231 human breast cancer cells, or MCF-10A human breast epithelial cells providing matched controls. Infection with HAd-EphrinA1-Fc (HAd vector expressing extracellular domain of human EphrinA1 attached to Fc portion of human IgG1 heavy chain) caused increased EphA2 activation and turnover and consequently decreased tumor cell viability in soft agar assays. Consistent with this observation, infection of MDA-MB-231 cells with HAd-EphrinA1-Fc prevented tumor formation in xenograft models. Furthermore, therapeutic modeling via intratumoral inoculation revealed that HAd-EphrinA1-Fc significantly inhibited subsequent tumor growth as compared to matched controls. These results suggest that targeting of EphA2 with adenoviral vectors may have therapeutic value.
- Published
- 2004
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.