Network


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

Hotspot


Dive into the research topics where Thoudam Debraj Singh is active.

Publication


Featured researches published by Thoudam Debraj Singh.


PLOS ONE | 2013

Construction and application of elastin like polypeptide containing IL-4 receptor targeting peptide.

Vijaya Sarangthem; Eun A. Cho; Sang Mun Bae; Thoudam Debraj Singh; Sun-Ji Kim; Soyoun Kim; Won Bae Jeon; Byung-Heon Lee; Rang-Woon Park

Various human solid tumors highly express IL-4 receptors which amplify the expression of some of anti-apoptotic proteins, preventing drug-induced cancer cell death. Thus, IL-4 receptor targeted drug delivery can possibly increase the therapeutic efficacy in cancer treatment. Macromolecular carriers with multivalent targeting moieties offered great advantages in cancer therapy as they not only increase the plasma half-life of the drug but also allow delivery of therapeutic drugs to the cancer cells with higher specificity, minimizing the deleterious effects of the drug on normal cells. In this study we designed a library of elastin like polypeptide (ELP) polymers containing tumor targeting AP1 peptide using recursive directional ligation method. AP1 was previously discovered as an atherosclerotic plaque and breast tumor tissue homing peptide using phage display screening method, and it can selectively bind to the interleukin 4 receptor (IL-4R). The fluorescently labeled [AP1-V12]6, an ELP polymer containing six AP1 enhanced tumor-specific targeting ability and uptake efficiency in H226 and MDA-MB-231 cancer cell lines in vitro. Surface plasmon resonance analysis showed that multivalent presentation of the targeting ligand in the ELP polymer increased the binding affinity towards IL-4 receptor compared to free peptide. The binding of [AP1-V12]6 to cancer cells was remarkably reduced when IL-4 receptors were blocked by antibody against IL-4 receptor further confirmed its binding. Importantly, the Cy5.5-labeled [AP1-V12]6 demonstrated excellent homing and longer retention in tumor tissues in MDA-MB-231 xenograft mouse model. Immunohistological studies of tumor tissues further validated the targeting efficiency of [AP1-V12]6 to tumor tissue. These results indicate that designed [AP1-V12]6 can serve as a novel carrier for selective delivery of therapeutic drugs to tumors.


FEBS Letters | 2010

MEGF10 functions as a receptor for the uptake of amyloid-β

Thoudam Debraj Singh; Seung-Yoon Park; Jae-sung Bae; Youngeun Yun; Yong-Chul Bae; Rang-Woon Park; In-San Kim

MINT‐7993537: ctxB (uniprotkb:P01556) and Abeta (uniprotkb:P05067) colocalize (MI:0403) by fluorescence microscopy (MI:0416)


Scientific Reports | 2015

Corrigendum: Tracking of dendritic cell migration into lymph nodes using molecular imaging with sodium iodide symporter and enhanced firefly luciferase genes

Ho-Won Lee; Seung Yun Yoon; Thoudam Debraj Singh; Yoon Ju Choi; Hong Je Lee; Ji Young Park; Shin Young Jeong; Sang-Woo Lee; Jeoung-Hee Ha; Byeong-Cheol Ahn; Yong Hyun Jeon; Jaetae Lee

We sought to evaluate the feasibility of molecular imaging using the human sodium iodide symporter (hNIS) gene as a reporter, in addition to the enhanced firefly luciferase (effluc) gene, for tracking dendritic cell (DCs) migration in living mice. A murine dendritic cell line (DC2.4) co-expressing hNIS and effluc genes (DC/NF) was established. For the DC-tracking study, mice received either parental DCs or DC/NF cells in the left or right footpad, respectively, and combined I-124 PET/CT and bioluminescence imaging (BLI) were performed. In vivo PET/CT imaging with I-124 revealed higher activity of the radiotracer in the draining popliteal lymph nodes (DPLN) of the DC/NF injection site at day 1 than DC injection site (p < 0.05). The uptake value further increased at day 4 (p < 0.005). BLI also demonstrated migration of DC/NF cells to the DPLNs at day 1 post-injection, and signals at the DPLNs were much higher at day 4. These data support the feasibility of hNIS reporter gene imaging in the tracking of DC migration to lymphoid organs in living mice. DCs expressing the NIS reporter gene could be a useful tool to optimize various strategies of cell-based immunotherapy.


The Journal of Nuclear Medicine | 2015

Inverse Agonist of Estrogen-Related Receptor γ Enhances Sodium Iodide Symporter Function Through Mitogen-Activated Protein Kinase Signaling in Anaplastic Thyroid Cancer Cells

Thoudam Debraj Singh; Shin Young Jeong; Sang-Woo Lee; Jeoung-Hee Ha; In-Kyu Lee; Seong Heon Kim; Jina Kim; Sung Jin Cho; Byeong-Cheol Ahn; Jaetae Lee; Young Hyun Jeon

Anaplastic thyroid cancer (ATC), a rare thyroid cancer with poor prognosis, is associated with insufficient function of the sodium iodide symporter (NIS). Estrogen-related receptor γ (ERRγ) is a member of the orphan nuclear receptors with important functions in cell development and homeostasis. However, there are no reports that demonstrate whether ERRγ is related to NIS function. Here, we evaluated the role of ERRγ in the regulation of NIS function in ATC cells using GSK5182, an inverse agonist of ERRγ. Methods: Two ATC cell lines, BHT-101 and CAL62, were incubated with GSK5182 at various time points and doses. The NIS function in the ATC cells was serially assessed by their uptake of radioiodine. The effects of GSK5182 on ERRγ and the mitogen-activated protein (MAP) kinase pathway, as well as on NIS protein, were evaluated by immunoblot assay. To examine whether the GSK5182-induced NIS functional activity can be affected by inhibition of the MAP kinase pathway, the MAP kinase activity and levels of radioiodine uptake were determined after application of a mitogen-activated protein kinase kinase (MEK) inhibitor to GSK5182-treated cells. Finally, the cytotoxic effect of 131I was determined by clonogenic assay. Results: Treatment with GSK5182 resulted in dose- and time-dependent increases in iodide uptake in ATC cells, which were accompanied by both the downregulation of ERRγ protein and the activation of extracellular signal-regulated kinase (ERK) 1/2. Both the increased radioiodine uptake and ERK1/2 activation of ATC cells were completely inhibited by the specific MEK inhibitor. GSK5182 treatment enhanced the membrane localization of NIS in both ATC cell lines. Accordingly, preexposure to GSK5182 enhanced the cytotoxic effects of 131I treatment in ATC cells. Conclusion: These findings suggest that the inverse agonist of ERRγ enhances the responsiveness of radioiodine therapy by modulating NIS function in ATC cells via the regulation of ERRγ and the MAP kinase signaling pathway.


The FASEB Journal | 2014

Evaluation of therapeutic effects of natural killer (NK) cell-based immunotherapy in mice using in vivo apoptosis bioimaging with a caspase-3 sensor

Ho-Won Lee; Thoudam Debraj Singh; Sang Woo Lee; Jeoung Hee Ha; Alnawaz Rehemtulla; Byeong-Cheol Ahn; Young Hyun Jeon; Jaetae Lee

Natural killer (NK) cell‐based immunotherapy is a promising strategy for cancer treatment, and caspase‐3 is an important effector molecule in NK cell‐mediated apoptosis in cancers. Here, we evaluated the antitumor effects of NK cell‐based immunotherapy by serial noninvasive imaging of apoptosis using a caspase‐3 sensor in mice with human glioma xenografts. Human glioma cells expressing both a caspase‐3 sensor as a surrogate marker for caspase‐3 activation and Renilla luciferase (Rluc) as a surrogate marker for cell viability were established and referred to as D54‐CR cells. Human NK92 cells were used as effector cells. Treatment with NK92 cells resulted in a time‐ and effector number‐dependent increase in bioluminescence imaging (BLI) activity of the caspase‐3 sensor in D54‐CR cells in vitro. Caspase‐3 activation by NK92 treatment was blocked by Z‐VAD treatment in D54‐CR cells. Transfusion of NK92 cells induced an increase of the BLI signal by caspase‐3 activation in a dose‐ and time‐dependent manner in D54‐CR tumor‐bearing mice but not in PBS‐treated mice. Accordingly, sequential BLI with the Rluc reporter gene revealed marked retardation of tumor growth in the NK92‐treatment group but not in the PBS‐treatment group. These data suggest that noninvasive imaging of apoptosis with a caspase‐3 sensor can be used as an effective tool for evaluation of therapeutic efficacy as well as for optimization of NK cell‐based immunotherapy.—Lee, H. W., Singh, T. D., Lee, S.‐W., Ha, J.‐H., Rehemtulla, A., Ahn, B.‐C., Jeon, Y.‐H., Lee, J. Evaluation of therapeutic effects of natural killer (NK) cell‐based immunotherapy in mice using in vivo apoptosis bioimaging with a caspase‐3 sensor. FASEB J. 28, 2932–2941 (2014). www.fasebj.org


Theranostics | 2017

Visualization of Macrophage Recruitment to Inflammation Lesions using Highly Sensitive and Stable Radionuclide-Embedded Gold Nanoparticles as a Nuclear Bio-Imaging Platform

Sang Bong Lee; Ho-Won Lee; Thoudam Debraj Singh; Yinghua Li; Sang Kyoon Kim; Sung Jin Cho; Sang-Woo Lee; Shin Young Jeong; Byeong-Cheol Ahn; Sangil Choi; In-Kyu Lee; Dong-Kwon Lim; Jaetae Lee; Yong Hyun Jeon

Reliable and sensitive imaging tools are required to track macrophage migration and provide a better understating of their biological roles in various diseases. Here, we demonstrate the possibility of radioactive iodide-embedded gold nanoparticles (RIe-AuNPs) as a cell tracker for nuclear medicine imaging. To demonstrate this utility, we monitored macrophage migration to carrageenan-induced sites of acute inflammation in living subjects and visualized the effects of anti-inflammatory agents on this process. Macrophage labeling with RIe-AuNPs did not alter their biological functions such as cell proliferation, phenotype marker expression, or phagocytic activity. In vivo imaging with positron-emission tomography revealed the migration of labeled macrophages to carrageenan-induced inflammation lesions 3 h after transfer, with highest recruitment at 6 h and a slight decline of radioactive signal at 24 h; these findings were highly consistent with the data of a bio-distribution study. Treatment with dexamethasone (an anti-inflammation drug) or GSK5182 (an ERRγ inverse agonist) hindered macrophage recruitment to the inflamed sites. Our findings suggest that a cell tracking strategy utilizing RIe-AuNPs will likely be highly useful in research related to macrophage-related disease and cell-based therapies.


Oncotarget | 2017

A new bioluminescent reporter system to study the biodistribution of systematically injected tumor-derived bioluminescent extracellular vesicles in mice

Prakash Gangadaran; Xiu Juan Li; Ho-Won Lee; Ji Min Oh; Senthilkumar Kalimuthu; Ramya Lakshmi Rajendran; Seung Hyun Son; Se Hwan Baek; Thoudam Debraj Singh; Liya Zhu; Shin Young Jeong; Sang-Woo Lee; Jaetae Lee; Byeong-Cheol Ahn

In vivo biodistribution and fate of extracellular vesicles (EVs) are still largely unknown and require reliable in vivo tracking techniques. In this study, in vivo bioluminescence imaging (BLI) using Renilla luciferase (Rluc) was developed and applied to monitoring of EVs derived from thyroid cancer (CAL-62 cells) and breast cancer (MDA-MB-231) in nude mice after intravenous administration and was compared with a dye-based labeling method for EV derived from CAL-62 cells. The EVs were successfully labeled with Rluc and visualized by BLI in mice. In vivo distribution of the EVs, as measured by BLI, was consistent with the results of ex vivo organ analysis. EV-CAL-62/Rluc showed strong signals at lung followed by liver, spleen & kidney (P < 0.05). EV-MDA-MB-231/Rluc showed strong signals at liver followed by lung, spleen & kidney (P < 0.05). EV-CAL-62/Rluc and EV-MDA-MB-231/Rluc stayed in animal till day 9 and 3, respectively; showed a differential distribution. Spontaneous EV-CAL-62/Rluc shown distributed mostly to lung followed by liver, spleen & kidney. The new BLI system used to show spontaneous distribution of EV-CAL-62/Rluc in subcutaneous CAL-62/Rluc bearing mice. Dye (DiR)-labeled EV-CAL-62/Rluc showed a different distribution in vivo & ex vivo compared to EV-CAL-62/Rluc. Fluorescent signals were predominately detected in the liver (P < 0.05) and spleen (P < 0.05) regions. The bioluminescent EVs developed in this study may be used for monitoring of EVs in vivo. This novel reporter-imaging approach to visualization of EVs in real time is expected to pave the way for monitoring of EVs in EV-based treatments.


Neoplasia | 2016

Visualization of the Biological Behavior of Tumor-Associated Macrophages in Living Mice with Colon Cancer Using Multimodal Optical Reporter Gene Imaging

Seul-Gi Oh; Thoudam Debraj Singh; Jeoung-Hee Ha; Dong Wook Kim; Sang Woo Lee; Shin Young Jeong; Byeong-Cheol Ahn; Jaetae Lee; Young Hyun Jeon

We sought to visualize the migration of tumor-associated macrophages (TAMs) to tumor lesions and to evaluate the effects of anti-inflammatory drugs on TAM-modulated tumor progression in mice with colon cancer using a multimodal optical reporter gene system. Murine macrophage Raw264.7 cells expressing an enhanced firefly luciferase (Raw/effluc) and murine colon cancer CT26 cells coexpressing Rluc and mCherry (CT26/Rluc-mCherry, CT26/RM) were established. CT26/RM tumor-bearing mice received Raw/effluc via their tail veins, and combination of bioluminescence imaging (BLI) and fluorescence imaging (FLI) was conducted for in vivo imaging of TAMs migration and tumor progression. Dexamethasone (DEX), a potent anti-inflammatory drug, was administered intraperitoneally to tumor-bearing mice following the intravenous transfer of Raw/effluc cells. The migration of TAMs and tumor growth was monitored by serial FLI and BLI. The migration of Raw/effluc cells to tumor lesions was observed at day 1, and BLI signals were still distinct at tumor lesions on day 4. Localization of BLI signals from migrated Raw/effluc cells corresponded to that of FLI signals from CT26/RM tumors. In vivo FLI of tumors demonstrated enhanced tumor growth associated with macrophage migration to tumor lesions. Treatment with DEX inhibited the influx of Raw/effluc cells to tumor lesions and abolished the enhanced tumor growth associated with macrophage migration. These findings suggest that molecular imaging approach for TAM tracking is a valuable tool for evaluating the role of TAMs in the tumor microenvironment as well as for the development of new drugs to control TAM involvement in the modulation of tumor progression.


Theranostics | 2016

Multivalent Targeting Based Delivery of Therapeutic Peptide using AP1-ELP Carrier for Effective Cancer Therapy.

Vijaya Sarangthem; Yunjae Kim; Thoudam Debraj Singh; Boyeon Seo; Sun-Ha Cheon; Young-Jin Lee; Byung-Heon Lee; Rang-Woon Park

Elastin-like polypeptide (ELP)-based drug delivery has been utilized for various applications including cancer therapies for many years. Genetic incorporation of internalization ligands and cell-targeting peptides along with ELP polymer enhanced tumor accumulation and retention time as well as stability and activities of the drug conjugates. Herein, we described a unique delivery system comprised of genetically engineered ELP incorporated with multiple copies of IL-4 receptor targeting peptide (AP1) periodically and proapoptotic peptide (KLAKLAK)2 referred to as AP1-ELP-KLAK. It triggered thermal-responsive self-assembly into a nanoparticle-like structure at physiological body temperature and stabilized its helical conformation, which is critical for its membrane-disrupting activities. Increased IL-4 receptor specific cellular internalization was associated with the enhanced cytotoxic effect of (KLAKLAK)2 peptide. Additionally, multivalent presentation of targeting ligands by AP1-ELP-KLAK significantly enhanced intratumoral localization and prolonged the retention time compared to ELP-KLAK, non-targeted control. Systemic administration of AP1-ELP-KLAK significantly inhibited tumor growth by provoking cell apoptosis in various tumor xenograft models without any specific organ toxicity. Thus, our newly designed AP1-ELP-KLAK polymer nanoparticle is a promising candidate for effective cancer therapy and due to the simple preparative procedures of ELPs, this platform can be used as a good carrier for tumor-specific delivery of other therapeutics.


Translational Oncology | 2017

Multimodality Imaging of Bone Marrow–Derived Dendritic Cell Migration and Antitumor Immunity

Su-Bi Ahn; Sang Bong Lee; Thoudam Debraj Singh; Sung Jin Cho; Sang Kyoon Kim; In-Kyu Lee; Shin Young Jeong; Byeong-Cheol Ahn; Jaetae Lee; Sang-Woo Lee; Yong Hyun Jeon

Here, we sought to monitor bone marrow–derived dendritic cell (BMDC) migration and antitumor effects using a multimodal reporter imaging strategy in living mice. BMDCs were transduced with retroviral vector harboring human sodium iodide symporter (hNIS, nuclear imaging reporter), firefly luc2 (optical imaging reporter), and thy1.1 (surrogate marker of NIS and luc2) genes (BMDC/NF cells). No significant differences in biological functions, including cell proliferation, antigen uptake, phenotype expression, and migration ability, were observed between BMDC and BMDC/NF cells. Combined bioluminescence imaging and I-124 positron emission tomography/computed tomography clearly revealed the migration of BMDC/NF cells to draining popliteal lymph nodes at day 7 postinjection. Interestingly, marked tumor protection was observed in mice immunized with TC-1 lysate-pulsed BMDC/NF cells. Our findings suggested that multimodal reporter gene imaging of NIS and luciferase could provide insights into the biological behaviors of dendritic cells in living organisms and could be a useful tool for the optimization of DC-based immunotherapy protocols.

Collaboration


Dive into the Thoudam Debraj Singh's collaboration.

Top Co-Authors

Avatar

Jaetae Lee

Kyungpook National University

View shared research outputs
Top Co-Authors

Avatar

Byeong-Cheol Ahn

Kyungpook National University

View shared research outputs
Top Co-Authors

Avatar

Sang-Woo Lee

Kyungpook National University

View shared research outputs
Top Co-Authors

Avatar

Shin Young Jeong

Kyungpook National University

View shared research outputs
Top Co-Authors

Avatar

Yong Hyun Jeon

Kyungpook National University

View shared research outputs
Top Co-Authors

Avatar

Ho-Won Lee

Kyungpook National University

View shared research outputs
Top Co-Authors

Avatar

Jeoung-Hee Ha

Kyungpook National University

View shared research outputs
Top Co-Authors

Avatar

In-Kyu Lee

Kyungpook National University

View shared research outputs
Top Co-Authors

Avatar

Sung Jin Cho

Kyungpook National University Hospital

View shared research outputs
Top Co-Authors

Avatar

Sang Bong Lee

Kyungpook National University Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge