Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Deepika Rajesh is active.

Publication


Featured researches published by Deepika Rajesh.


Journal of Virology | 2011

A new model of Epstein-Barr virus infection reveals an important role for early lytic viral protein expression in the development of lymphomas

Shi Dong Ma; Subramanya Hegde; Ken H. Young; Ruth Sullivan; Deepika Rajesh; Ying Zhou; Ewa Jankowska-Gan; William J. Burlingham; Xiaoping Sun; Margaret L. Gulley; Weihua Tang; Jenny E. Gumperz; Shannon C. Kenney

ABSTRACT Epstein-Barr virus (EBV) infects cells in latent or lytic forms, but the role of lytic infection in EBV-induced lymphomas is unclear. Here, we have used a new humanized mouse model, in which both human fetal CD34+ hematopoietic stem cells and thymus/liver tissue are transplanted, to compare EBV pathogenesis and lymphoma formation following infection with a lytic replication-defective BZLF1-deleted (Z-KO) virus or a lytically active BZLF1+ control. Both the control and Z-KO viruses established long-term viral latency in all infected animals. The infection appeared well controlled in some animals, but others eventually developed CD20+ diffuse large B cell lymphomas (DLBCL). Animals infected with the control virus developed tumors more frequently than Z-KO virus-infected animals. Specific immune responses against EBV-infected B cells were generated in mice infected with either the control virus or the Z-KO virus. In both cases, forms of viral latency (type I and type IIB) were observed that are less immunogenic than the highly transforming form (type III) commonly found in tumors of immunocompromised hosts, suggesting that immune pressure contributed to the outcome of the infection. These results point to an important role for lytic EBV infection in the development of B cell lymphomas in the context of an active host immune response.


PLOS ONE | 2010

Derivation of Induced Pluripotent Stem Cells from Human Peripheral Blood T Lymphocytes

Matthew E. Brown; Elizabeth Rondon; Deepika Rajesh; Amanda Mack; Rachel Lewis; Xuezhu Feng; Laura J. Zitur; Randall D. Learish; Emile Nuwaysir

Induced pluripotent stem cells (iPSCs) hold enormous potential for the development of personalized in vitro disease models, genomic health analyses, and autologous cell therapy. Here we describe the generation of T lymphocyte-derived iPSCs from small, clinically advantageous volumes of non-mobilized peripheral blood. These T-cell derived iPSCs (“TiPS”) retain a normal karyotype and genetic identity to the donor. They share common characteristics with human embryonic stem cells (hESCs) with respect to morphology, pluripotency-associated marker expression and capacity to generate neurons, cardiomyocytes, and hematopoietic progenitor cells. Additionally, they retain their characteristic T-cell receptor (TCR) gene rearrangements, a property which could be exploited for iPSC clone tracking and T-cell development studies. Reprogramming T-cells procured in a minimally invasive manner can be used to characterize and expand donor specific iPSCs, and control their differentiation into specific lineages.


Human Immunology | 2010

Th1 and Th17 immunocompetence in humanized NOD/SCID/IL2rγnull mice

Deepika Rajesh; Ying Zhou; Ewa Jankowska-Gan; Drew A. Roenneburg; Melanie L. Dart; Jose Torrealba; William J. Burlingham

We evaluated the immunocompetence of human T cells in humanized NOD-SCID interleukin (IL)-2r-gamma-null (hu-NSG) mice bearing a human thymic organoid, after multilineage reconstitution with isogeneic human leukocytes. Delayed type hypersensitivity (DTH) response was assessed by a direct footpad challenge of the immunized hu-NSG host, or by transfer of splenocytes from immunized hu-NSG, along with antigen, into footpads of C.B-17 scid mice (trans vivo [tv] DTH). Both methods revealed cellular immunity to tetanus toxoid (TT) or collagen type V (ColV). Immunohistochemical analysis of the swollen footpads revealed infiltration of human CD45(+) cells, including CD3(+) T cells, CD68(+) macrophages, and murine Ly6G(+) neutrophils. We observed a significant correlation between the percentage of circulating human CD4(+) cells and the direct DTH swelling response to TT. The tvDTH response to TT was inhibited by anti-interferon-gamma, whereas the tvDTH response to collagen V was inhibited by anti-IL-17 antibody, mimicking the cytokine bias of adult human T cells to these antigens. hu-NSG mice were also capable of mounting a B-cell response (primarily IgM) to TT antigen. The activation of either Th1- or Th17-dependent cellular immune response supports the utility of hu-NSG mice as a surrogate model of allograft rejection and autoimmunity.


Blood | 2011

Human lymphoblastoid B cell lines reprogrammed to EBV-free induced pluripotent stem cells

Deepika Rajesh; Sarah J. Dickerson; Junying Yu; Matthew E. Brown; James A. Thomson; Nicholas Seay

Generation of patient-specific induced pluripotent cells (iPSCs) holds great promise for regenerative medicine. Epstein-Barr virus immortalized lymphoblastoid B-cell lines (LCLs) can be generated from a minimal amount of blood and are banked worldwide as cellular reference material for immunologic or genetic analysis of pedigreed study populations. We report the generation of iPSCs from 2 LCLs (LCL-iPSCs) via a feeder-free episomal method using a cocktail of transcription factors and small molecules. LCL-derived iPSCs exhibited normal karyotype, expressed pluripotency markers, lost oriP/EBNA-1 episomal vectors, generated teratomas, retained donor identity, and differentiated in vitro into hematopoietic, cardiac, neural, and hepatocyte-like lineages. Significantly, although the parental LCLs express viral EBNA-1 and other Epstein-Barr virus latency-related elements for their survival, their presence was not detectable in LCL-iPSCs. Thus, reprogramming LCLs could offer an unlimited source for patient-specific iPSCs.


PLOS ONE | 2011

Generation of Induced Pluripotent Stem Cells from CD34+ Cells across Blood Drawn from Multiple Donors with Non-Integrating Episomal Vectors

Amanda Mack; Stacie Kroboth; Deepika Rajesh; Wen Bo Wang

The methodology to create induced pluripotent stem cells (iPSCs) affords the opportunity to generate cells specific to the individual providing the host tissue. However, existing methods of reprogramming as well as the types of source tissue have significant limitations that preclude the ability to generate iPSCs in a scalable manner from a readily available tissue source. We present the first study whereby iPSCs are derived in parallel from multiple donors using episomal, non-integrating, oriP/EBNA1-based plasmids from freshly drawn blood. Specifically, successful reprogramming was demonstrated from a single vial of blood or less using cells expressing the early lineage marker CD34 as well as from unpurified peripheral blood mononuclear cells. From these experiments, we also show that proliferation and cell identity play a role in the number of iPSCs per input cell number. Resulting iPSCs were further characterized and deemed free of transfected DNA, integrated transgene DNA, and lack detectable gene rearrangements such as those within the immunoglobulin heavy chain and T cell receptor loci of more differentiated cell types. Furthermore, additional improvements were made to incorporate completely defined media and matrices in an effort to facilitate a scalable transition for the production of clinic-grade iPSCs.


PLOS ONE | 2011

A Defined, Feeder-Free, Serum-Free System to Generate In Vitro Hematopoietic Progenitors and Differentiated Blood Cells from hESCs and hiPSCs

Giorgia Salvagiotto; Sarah A. Burton; Christine A. Daigh; Deepika Rajesh; Igor I. Slukvin; Nicholas Seay

Human ESC and iPSC are an attractive source of cells of high quantity and purity to be used to elucidate early human development processes, for drug discovery, and in clinical cell therapy applications. To efficiently differentiate pluripotent cells into a pure population of hematopoietic progenitors we have developed a new 2-dimentional, defined and highly efficient protocol that avoids the use of feeder cells, serum or embryoid body formation. Here we showed that a single matrix protein in combination with growth factors and a hypoxic environment is sufficient to generate from pluripotent cells hematopoietic progenitors capable of differentiating further in mature cell types of different lineages of the blood system. We tested the differentiation method using hESCs and 9 iPSC lines generated from different tissues. These data indicate the robustness of the protocol providing a valuable tool for the generation of clinical-grade hematopoietic cells from pluripotent cells.


Neurological Research | 2005

Are gadolinium contrast agents suitable for gadolinium neutron capture therapy

Gelsomina De Stasio; Deepika Rajesh; Patrizia Casalbore; Matthew J. Daniels; Robert J. Erhardt; Bradley H. Frazer; Lisa M. Wiese; Katherine L. Richter; Brandon R. Sonderegger; Benjamin Gilbert; Sébastien Schaub; Rachel J. Cannara; John F. Crawford; Mary K. Gilles; Tolek Tyliszczak; Jack F. Fowler; Luigi Maria Larocca; Steven P. Howard; Delio Mercanti; Minesh P. Mehta; Roberto Pallini

Abstract Objective: Gadolinium neutron capture therapy (GdNCT) is a potential treatment for malignant tumors based on two steps: (1) injection of a tumor-specific 157Gd compound; (2) tumor irradiation with thermal neutrons. The GdNC reaction can induce cell death provided that Gd is proximate to DNA. Here, we studied the nuclear uptake of Gd by glioblastoma (GBM) tumor cells after treatment with two Gd compounds commonly used for magnetic resonance imaging, to evaluate their potential as GdNCT agents. Methods: Using synchrotron X-ray spectromicroscopy, we analyzed the Gd distribution at the subcellular level in: (1) human cultured GBM cells exposed to Gd-DTPA or Gd-DOTA for 0–72hours; (2) intracerebrally implanted C6 glioma tumors in rats injected with one or two doses of Gd-DOTA, and (3) tumor samples from GBM patients injected with Gd-DTPA. Results: In cell cultures, Gd-DTPA and Gd-DOTA were found in 84% and 56% of the cell nuclei, respectively. In rat tumors, Gd penetrated the nuclei of 47% and 85% of the tumor cells, after single and double injection of Gd-DOTA, respectively. In contrast, in human GBM tumors 6.1% of the cell nuclei contained Gd-DTPA. Discussion: Efficacy of Gd-DTPA and Gd-DOTA as GdNCT agents is predicted to be low, due to the insufficient number of tumor cell nuclei incorporating Gd. Although multiple administration schedules in vivo might induce Gd penetration into more tumor cell nuclei, a search for new Gd compounds with higher nuclear affinity is warranted before planning GdNCT in animal models or clinical trials.


Clinical Cancer Research | 2006

Motexafin-Gadolinium Taken Up In vitro by at Least 90% of Glioblastoma Cell Nuclei

Gelsomina De Stasio; Deepika Rajesh; Judith Ford; Matthew J. Daniels; Robert J. Erhardt; Bradley H. Frazer; Tolek Tyliszczak; Mary K. Gilles; Robert L. Conhaim; Steven P. Howard; Jack F. Fowler; François Estève; Minesh P. Mehta

Purpose: We present preclinical data showing the in vitro intranuclear uptake of motexafin gadolinium by glioblastoma multiforme cells, which could serve as a prelude to the future development of radiosensitizing techniques, such as gadolinium synchrotron stereotactic radiotherapy (GdSSR), a new putative treatment for glioblastoma multiforme. Experimental Design: In this approach, administration of a tumor-seeking Gd-containing compound would be followed by stereotactic external beam radiotherapy with 51-keV photons from a synchrotron source. At least two criteria must be satisfied before this therapy can be established: Gd must accumulate in cancer cells and spare the normal tissue; Gd must be present in almost all the cancer cell nuclei. We address the in vitro intranuclear uptake of motexafin gadolinium in this article. We analyzed the Gd distribution with subcellular resolution in four human glioblastoma cell lines, using three independent methods: two novel synchrotron spectromicroscopic techniques and one confocal microscopy. We present in vitro evidence that the majority of the cell nuclei take up motexafin gadolinium, a drug that is known to selectively reach glioblastoma multiforme. Results: With all three methods, we found Gd in at least 90% of the cell nuclei. The results are highly reproducible across different cell lines. The present data provide evidence for further studies, with the goal of developing GdSSR, a process that will require further in vivo animal and future clinical studies.


PLOS ONE | 2011

Analysis of the CD1 Antigen Presenting System in Humanized SCID Mice

Jennifer L. Lockridge; Xiuxu Chen; Ying Zhou; Deepika Rajesh; Drew A. Roenneburg; Subramanya Hegde; Sarah Gerdts; Tan-Yun Cheng; Regan J. Anderson; Gavin F. Painter; D. Branch Moody; William J. Burlingham; Jenny E. Gumperz

CD1 molecules are glycoproteins that present lipids and glycolipids for recognition by T cells. CD1-dependent immune activation has been implicated in a wide range of immune responses, however, our understanding of the role of this pathway in human disease remains limited because of species differences between humans and other mammals: whereas humans express five different CD1 gene products (CD1a, CD1b, CD1c, CD1d, and CD1e), muroid rodents express only one CD1 isoform (CD1d). Here we report that immune deficient mice engrafted with human fetal thymus, liver, and CD34+ hematopoietic stem cells develop a functional human CD1 compartment. CD1a, b, c, and d isoforms were highly expressed by human thymocytes, and CD1a+ cells with a dendritic morphology were present in the thymic medulla. CD1+ cells were also detected in spleen, liver, and lungs. APCs from spleen and liver were capable of presenting bacterial glycolipids to human CD1-restricted T cells. ELISpot analyses of splenocytes demonstrated the presence of CD1-reactive IFN-γ producing cells. CD1d tetramer staining directly identified human iNKT cells in spleen and liver samples from engrafted mice, and injection of the glycolipid antigen α-GalCer resulted in rapid elevation of human IFN-γ and IL-4 levels in the blood indicating that the human iNKT cells are biologically active in vivo. Together, these results demonstrate that the human CD1 system is present and functionally competent in this humanized mouse model. Thus, this system provides a new opportunity to study the role of CD1-related immune activation in infections to human-specific pathogens.


Stem Cells | 2007

Differential requirements for hematopoietic commitment between human and rhesus embryonic stem cells

Deepika Rajesh; Nachimuthu Chinnasamy; Shoukhrat Mitalipov; Don P. Wolf; Igor I. Slukvin; James A. Thomson; Aimen F. Shaaban

Progress toward clinical application of ESC‐derived hematopoietic cellular transplantation will require rigorous evaluation in a large animal allogeneic model. However, in contrast to human ESCs (hESCs), efforts to induce conclusive hematopoietic differentiation from rhesus macaque ESCs (rESCs) have been unsuccessful. Characterizing these poorly understood functional differences will facilitate progress in this area and likely clarify the critical steps involved in the hematopoietic differentiation of ESCs. To accomplish this goal, we compared the hematopoietic differentiation of hESCs with that of rESCs in both EB culture and stroma coculture. Initially, undifferentiated rESCs and hESCs were adapted to growth on Matrigel without a change in their phenotype or karyotype. Subsequent differentiation of rESCs in OP9 stroma led to the development of CD34+CD45− cells that gave rise to endothelial cell networks in methylcellulose culture. In the same conditions, hESCs exhibited convincing hematopoietic differentiation. In cytokine‐supplemented EB culture, rESCs demonstrated improved hematopoietic differentiation with higher levels of CD34+ and detectable levels of CD45+ cells. However, these levels remained dramatically lower than those for hESCs in identical culture conditions. Subsequent plating of cytokine‐supplemented rhesus EBs in methylcellulose culture led to the formation of mixed colonies of erythroid, myeloid, and endothelial cells, confirming the existence of bipotential hematoendothelial progenitors in the cytokine‐supplemented EB cultures. Evaluation of four different rESC lines confirmed the validity of these disparities. Although rESCs have the potential for hematopoietic differentiation, they exhibit a pause at the hemangioblast stage of hematopoietic development in culture conditions developed for hESCs.

Collaboration


Dive into the Deepika Rajesh's collaboration.

Top Co-Authors

Avatar

Aimen F. Shaaban

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Steven P. Howard

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Emily T. Durkin

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Matthew E. Brown

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Kelly A. Jones

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Lasya Gaur

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Rachel Lewis

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

William J. Burlingham

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Ying Zhou

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Bradley H. Frazer

University of Wisconsin-Madison

View shared research outputs
Researchain Logo
Decentralizing Knowledge