Network


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

Hotspot


Dive into the research topics where Brij K. Gupta is active.

Publication


Featured researches published by Brij K. Gupta.


Journal of Colloid and Interface Science | 2010

Fabrication of curcumin encapsulated PLGA nanoparticles for improved therapeutic effects in metastatic cancer cells

Murali M. Yallapu; Brij K. Gupta; Meena Jaggi; Subhash C. Chauhan

Curcumin, a natural polyphenolic compound, has shown promising chemopreventive and chemotherapeutic activities in cancer. Although phase I clinical trials have shown curcumin as a safe drug even at high doses, poor bioavailability and suboptimal pharmacokinetics largely moderated its anti-cancer activity in pre-clinical and clinical models. To improve its applicability in cancer therapy, we encapsulated curcumin in poly(lactic-co-glycolide) (PLGA) (biodegradable polymer) nanoparticles, in the presence of poly(vinyl alcohol) and poly(L-lysine) stabilizers, using a nano-precipitation technique. These curcumin nano-formulations were characterized for particle size, zeta potential, drug encapsulation, drug compatibility and drug release. Encapsulated curcumin existed in a highly dispersed state in the PLGA core of the nanoparticles and exhibited good solid-solid compatibility. An optimized curcumin nano-formulation (nano-CUR6) has demonstrated two and sixfold increases in the cellular uptake performed in cisplatin resistant A2780CP ovarian and metastatic MDA-MB-231 breast cancer cells, respectively, compared to free curcumin. In these cells, nano-CUR6 has shown an improved anti-cancer potential in cell proliferation and clonogenic assays compared to free curcumin. This effect was correlated with enhanced apoptosis induced by the nano-CUR6 formulation. Herein, we have also shown antibody conjugation compatibility of our PLGA-NP formulation. Results of this study suggest that therapeutic efficacy of curcumin may be enhanced by such PLGA nanoparticle formulations, and furthermore tumor specific targeted delivery of curcumin is made feasible by coupling of anti-cancer antibody to the NPs.


Biomaterials | 2011

Multi-functional Magnetic Nanoparticles for Magnetic Resonance Imaging and Cancer Therapy

Murali M. Yallapu; Shadi F. Othman; Evan T. Curtis; Brij K. Gupta; Meena Jaggi; Subhash C. Chauhan

We have developed a multi-layer approach for the synthesis of water-dispersible superparamagnetic iron oxide nanoparticles for hyperthermia, magnetic resonance imaging (MRI) and drug delivery applications. In this approach, iron oxide core nanoparticles were obtained by precipitation of iron salts in the presence of ammonia and provided β-cyclodextrin and pluronic polymer (F127) coatings. This formulation (F127250) was highly water dispersible which allowed encapsulation of the anti-cancer drug(s) in β-cyclodextrin and pluronic polymer for sustained drug release. The F127250 formulation has exhibited superior hyperthermia effects over time under alternating magnetic field compared to pure magnetic nanoparticles (MNP) and β-cyclodextrin coated nanoparticles (CD200). Additionally, the improved MRI characteristics were also observed for the F127250 formulation in agar gel and in cisplatin resistant ovarian cancer cells (A12780CP) compared to MNP and CD200 formulations. Furthermore, the drug-loaded formulation of F127250 exhibited many folds of imaging contrast properties. Due to the internalization capacity of the F127250 formulation, its curcumin-loaded formulation (F127250-CUR) exhibited almost equivalent inhibition effects on A2780CP (ovarian), MDA-MB-231 (breast), and PC-3 (prostate) cancer cells even though curcumin release was only 40%. The improved therapeutic effects were verified by examining molecular effects using Western blotting and transmission electron microscopic (TEM) studies. F127250-CUR also exhibited haemocompatibility, suggesting a nanochemo-therapeutic agent for cancer therapy.


Biomaterials | 2014

Anti-cancer activity of curcumin loaded nanoparticles in prostate cancer.

Murali M. Yallapu; Sheema Khan; Diane M. Maher; Mara C. Ebeling; Vasudha Sundram; Neeraj Chauhan; Aditya Ganju; Swathi Balakrishna; Brij K. Gupta; Nadeem Zafar; Meena Jaggi; Subhash C. Chauhan

Prostate cancer is the most commonly diagnosed cancer disease in men in the Unites States and its management remains a challenge in everyday oncology practice. Thus, advanced therapeutic strategies are required to treat prostate cancer patients. Curcumin (CUR) is a promising anticancer agent for various cancer types. The objective of this study was to evaluate therapeutic potential of novel poly(lactic-co-glycolic acid)- CUR nanoparticles (PLGA-CUR NPs) for prostate cancer treatment. Our results indicate that PLGA-CUR NPs efficiently internalize in prostate cancer cells and release biologically active CUR in cytosolic compartment of cells for effective therapeutic activity. Cell proliferation (MTS), clonogenic, and Western blot analyses reveal that PLGA-CUR NPs can effectively inhibit proliferation and colony formation ability of prostate cancer cells than free CUR. PLGA-CUR NPs showed superior tumor regression compared to CUR in xenograft mice. Further investigations reveal that PLGA-CUR NPs inhibit nuclear β-catenin and AR expression in cells and in tumor xenograft tissues. It also suppresses STAT3 and AKT phosphorylation and leads to apoptosis via inhibition of key anti-apoptotic proteins, Mcl-1, Bcl-xL and caused induction of PARP cleavage. Additionally, significant downregulation of oncogenic miR21 and up-regulation of miR-205 was observed with PLGA-CUR NPs treatment as determined by RT-PCR and in situ hybridization analyses. A superior anti-cancer potential was attained with PSMA antibody conjugated PLGA-CUR NPs in prostate cancer cells and a significant tumor targeting of (131)I labeled PSMA antibody was achieved with PLGA-CUR NPs in prostate cancer xenograft mice model. In conclusion, PLGA-CUR NPs can significantly accumulate and exhibit superior anticancer activity in prostate cancer.


Molecular Cancer Therapeutics | 2013

Novel Curcumin-Loaded Magnetic Nanoparticles for Pancreatic Cancer Treatment

Murali M. Yallapu; Mara C. Ebeling; Sheema Khan; Vasudha Sundram; Neeraj Chauhan; Brij K. Gupta; Susan E. Puumala; Meena Jaggi; Subhash C. Chauhan

Curcumin (CUR), a naturally occurring polyphenol derived from the root of Curcuma longa, has showed potent anticancer and cancer prevention activity in a variety of cancers. However, the clinical translation of CUR has been significantly hampered due to its extensive degradation, suboptimal pharmacokinetics, and poor bioavailability. To address these clinically relevant issues, we have developed a novel CUR-loaded magnetic nanoparticle (MNP-CUR) formulation. Herein, we have evaluated the in vitro and in vivo therapeutic efficacy of this novel MNP-CUR formulation in pancreatic cancer. Human pancreatic cancer cells (HPAF-II and Panc-1) exhibited efficient internalization of the MNP-CUR formulation in a dose-dependent manner. As a result, the MNP-CUR formulation effectively inhibited growth of HPAF-II and Panc-1 cells in cell proliferation and colony formation assays. The MNP-CUR formulation suppressed pancreatic tumor growth in an HPAF-II xenograft mouse model and improved the survival of mice by delaying tumor growth. The growth-inhibitory effect of MNP-CUR formulation correlated with the suppression of proliferating cell nuclear antigen (PCNA), B-cell lymphoma-extra large (Bcl-xL), induced myeloid leukemia cell differentiation protein (Mcl-1), cell surface–associated Mucin 1 (MUC1), collagen I, and enhanced membrane β-catenin expression. MNP-CUR formulation did not show any sign of hemotoxicity and was stable after incubation with human serum proteins. In addition, the MNP-CUR formulation improved serum bioavailability of CUR in mice up to 2.5-fold as compared with free CUR. Biodistribution studies show that a significant amount of MNP-CUR formulation was able to reach the pancreatic xenograft tumor(s), which suggests its clinical translational potential. In conclusion, this study suggests that our novel MNP-CUR formulation can be valuable for the treatment of pancreatic cancer. Mol Cancer Ther; 12(8); 1471–80. ©2013 AACR.


Molecular Carcinogenesis | 2011

Curcumin suppresses human papillomavirus oncoproteins, restores p53, Rb, and PTPN13 proteins and inhibits benzo[a]pyrene-induced upregulation of HPV E7.

Diane M. Maher; Maria C. Bell; Emmylu A. O'Donnell; Brij K. Gupta; Meena Jaggi; Subhash C. Chauhan

Curcumin has great potential as a chemopreventive and chemotherapeutic agent; however, its effects on human papillomavirus (HPV)‐associated molecular events are inadequately explored. This study examined the effects of curcumin on HPV‐associated pathways involved in developing cervical cancer. We demonstrate for the first time that curcumin treatment suppresses cervical cancer cell growth in a three‐dimensional raft culture system. Curcumin also inhibits tumorigenic characteristics as shown by decreases in both clonogenic potential and cell motility. Additionally, our findings show that curcumin treatment inhibits the transcription of HPV16 E6/E7 as early as 6 h posttreatment and restores the expression of tumor suppressor proteins p53, retinoblastoma protein, and PTPN13. While smoking is a recognized risk factor for cervical cancer, the molecular effects of smoke carcinogens on the expression of HPV E6/E7 oncogenes are not well known. We show for the first time that exposure to benzo[a]pyrene (BaP), a tobacco carcinogen, increases the expression of HPV E7 oncoprotein suggesting a molecular link between smoking and cervical cancer. Importantly, curcumin decreases the BaP induced increase in the expression of HPV E7 oncoprotein. The results of this study clearly demonstrate that curcumin alters HPV‐associated molecular pathways in cervical cancer cells. These novel findings imply that curcumin may be an effective chemopreventive and therapeutic agent for cervical cancer prevention and treatment. Mol. Carcinog.


Molecular Cancer Research | 2011

Mucin 13: Structure, Function, and Potential Roles in Cancer Pathogenesis

Diane M. Maher; Brij K. Gupta; Satoshi Nagata; Meena Jaggi; Subhash C. Chauhan

Mucin 13 (MUC13) is a high-molecular-weight transmembrane glycoprotein that is frequently and aberrantly expressed in a variety of epithelial carcinomas, including gastric, colorectal, and ovarian cancers. On the basis of the high expression of MUC13 in cancer cells as well as recent laboratory findings suggesting a malignant phenotype of MUC13-transfected cell lines, the oncogenic potential of MUC13 has emerged. The various functional domains of MUC13 may confer oncogenic potential to MUC13. For example, the bulky extracellular domain with extensive modification with glycan chains may prevent cell–cell and cell–extracellular matrix binding whereas the cytoplasmic tail containing serine and tyrosine residues for potential phosphorylation may participate in cell signaling. MUC13 exhibits the characteristics suitable as an early marker for cancer screening and presents a promising target for antibody-guided targeted therapy. Mol Cancer Res; 9(5); 531–7. ©2011 AACR.


Journal of Histochemistry and Cytochemistry | 2012

Increased Expression and Aberrant Localization of Mucin 13 in Metastatic Colon Cancer

Brij K. Gupta; Diane M. Maher; Mara C. Ebeling; Vasudha Sundram; Michael D. Koch; Douglas W. Lynch; Teresa Bohlmeyer; Akira Watanabe; Hiroyuki Aburatani; Susan E. Puumala; Meena Jaggi; Subhash C. Chauhan

MUC13 is a newly identified transmembrane mucin. Although MUC13 is known to be overexpressed in ovarian and gastric cancers, limited information is available regarding the expression of MUC13 in metastatic colon cancer. Herein, we investigated the expression profile of MUC13 in colon cancer using a novel anti-MUC13 monoclonal antibody (MAb, clone ppz0020) by immunohistochemical (IHC) analysis. A cohort of colon cancer samples and tissue microarrays containing adjacent normal, non-metastatic colon cancer, metastatic colon cancer, and liver metastasis tissues was used in this study to investigate the expression pattern of MUC13. IHC analysis revealed significantly higher (p<0.001) MUC13 expression in non-metastatic colon cancer samples compared with faint or very low expression in adjacent normal tissues. Interestingly, metastatic colon cancer and liver metastasis tissue samples demonstrated significantly (p<0.05) higher cytoplasmic and nuclear MUC13 expression compared with non-metastatic colon cancer and adjacent normal colon samples. Moreover, cytoplasmic and nuclear MUC13 expression correlated with larger and poorly differentiated tumors. Four of six tested colon cancer cell lines also expressed MUC13 at RNA and protein levels. These studies demonstrate a significant increase in MUC13 expression in metastatic colon cancer and suggest a correlation between aberrant MUC13 localization (cytoplasmic and nuclear expression) and metastatic colon cancer.


Journal of Gastroenterology | 2014

Functions and regulation of MUC13 mucin in colon cancer cells.

Brij K. Gupta; Diane M. Maher; Mara C. Ebeling; Phillip Stephenson; Susan E. Puumala; Michael R. Koch; Hiroyuki Aburatani; Meena Jaggi; Subhash C. Chauhan

BackgroundMUC13 is overexpressed and aberrantly localized in colon cancer tissue; however, the specific functions and regulation of MUC13 expression are unknown.MethodsStable cell lines with either overexpressed or suppressed MUC13 levels were analyzed to determine cell growth, colony formation, cell migration, and cell invasion assays. The molecular mechanisms involved in MUC13 regulation were elucidated via chromatin immunoprecipitation (ChIP) and analysis of interleukin 6 (IL6) treatments. Colon cancer tissues were analyzed by immunohistochemistry (IHC) for the protein levels of MUC13 and P-STAT5 in colon cancer cells.ResultsOverexpression of MUC13 increased cell growth, colony formation, cell migration, and invasion. In concordance, MUC13 silencing decreased these tumorigenic features. Overexpression of MUC13 also modulated various cancer-associated proteins, including telomerase reverse transcriptase, sonic hedgehog, B cell lymphoma murine like site 1, and GATA like transcription factor 1. Additionally, MUC13-overexpressing cells showed increased HER2 and P-ERK expression. ChIP analysis revealed binding of STAT5 to the predicted MUC13 promoter. IL6 treatment of colon cancer cells increased the expression of MUC13 via activation of the JAK2/STAT5 signaling pathway. Suppression of JAK2 and STAT5 signaling by chemical inhibitors abolished IL6-induced MUC13 expression. IHC analysis showed increased expression of both P-STAT5 and MUC13 in colon cancer as compared to adjacent normal tissue.ConclusionsThe results of this study, for the first time, suggest functional roles of MUC13 in colon cancer progression and provide information regarding the regulation of MUC13 expression via JAK2/STAT5 which may reveal promising therapeutic approaches for colon cancer treatment.


Cancer Research | 2013

Abstract 4503: Novel curcumin loaded magnetic nanoparticles for pancreatic cancer treatment.

Murali M. Yallapu; Mara C. Ebleling; Sheema Khan; Neeraj Chauhan; Brij K. Gupta; Vasudha Sundram; Meena Jaggi; Subhash C. Chauhan

Proceedings: AACR 104th Annual Meeting 2013; Apr 6-10, 2013; Washington, DC Curcumin (CUR), a naturally occurring polyphenol derived from the root of Curcuma longa, has demonstrated potent anti-cancer and cancer prevention activity in a variety of cancers. However, the clinical translation of curcumin has been significantly hampered due its extensive degradation, suboptimal pharmacokinetics and poor bioavailability. To address these clinically relevant issues, we have developed a novel curcumin loaded magnetic nanoparticle (MNP-CUR) formulation. Herein, we have evaluated the in vitro and in vivo therapeutic efficacy of this novel MNP-CUR formulation in pancreatic cancer. Human pancreatic cancer cells (HPAFII and Panc-1) exhibited efficient internalization of the MNP-CUR formulation in a dose dependent manner. As a result, MNP-CUR formulation effectively inhibited growth of HPAFII and Panc-1 cells in cell proliferation and colony formation assays. Similarly, the MNP-CUR formulation suppressed pancreatic tumor growth in an HPAFII xenograft mice model and improved mice survival by delaying tumor growth. The growth inhibitory effect of MNP-CUR formulation was correlated with the suppression of PCNA, Bcl-xL, Mcl-1, MUC1, collagen I and enhanced membrane β-catenin expression. Interestingly, our MNP-CUR formulation did not show any sign of hemotoxicity and stable after incubation with human serum proteins. Additionally, our MNP-CUR formulation improved serum bioavailability of curcumin in mice up to 2.5 fold as compared to curcumin in Tween-20. Furthermore, biodistribution studies of our MNP-CUR formulation demonstrate a significant amount of formulation was able to reach the pancreatic xenograft tumor(s) which suggests its clinical translational potential. In conclusion, our study suggests that our novel MNP-CUR formulation can be valuable for the treatment of pancreatic cancer. Citation Format: Murali M. Yallapu, Mara C. Ebleling, Sheema Khan, Neeraj Chauhan, Brij K. Gupta, Vasudha Sundram, Meena Jaggi, Subhash C. Chauhan. Novel curcumin loaded magnetic nanoparticles for pancreatic cancer treatment. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 4503. doi:10.1158/1538-7445.AM2013-4503


Cancer Research | 2012

Abstract 3589: Comparative expression profile of transmembrane mucin MUC1 in breast cancer from American Indian and Caucasian women

Diane M. Maher; Phillip Stephenson; Brij K. Gupta; Nichole A. Bauer; Michael D. Koch; Susan Eliason; Meena Jaggi; Subhash C. Chauhan

Introduction: Considerable disparity exists in breast cancer incidence and mortality among different racial/ethnic populations and geographic locations. For unknown reasons, American Indian women of the Northern Plains have a greater incidence and higher death rate due to breast cancer than Caucasian women living in the same geographical region. Additionally, American Indian women living in the Northern Plains also have a much higher incidence and death rate due to breast cancer than American Indian women living elsewhere in the United States. To investigate the potential molecular basis for this cancer health disparity, we have compared the expression profile of transmembrane mucin, MUC1 on breast cancer cases from American Indian and Caucasian women living on the Northern Plains. MUC1 is known to be overexpressed in breast cancer and nuclear localization of MUC1 can have downstream effects on cell signaling and increase oncogenic phenotypes. We are also exploring the link between smoking, exposure to HPV infection, and the development of breast cancer in American Indian women. Methods: We have examined the expression profile of MUC1 in breast cancer tissue from American Indian (n=40) and Caucasian (n=60) women. The staining was analyzed by experienced pathologists and scored according to intensity (0-4) and percent of cancer cells stained positive (0-4). A composite score was calculated by multiplying the scores of intensity and percent of cells stained. Tissues were categorized by modified Bloom-Richardson9s histological grade and the mean composite scores were compared amongst American Indian and Caucasian groups. Results and Conclusions: We have observed that grade 3 breast cancer samples from American Indian women had a higher percent of nuclear MUC1 staining and a lower amount of p53 expression. This result correlates with previous data showing that MUC1 inhibits the expression of p53. In conclusion, breast cancer tissue from American Indian women has a different expression profile of molecular markers and this finding may help to explain the cancer health disparity. Further investigation of these differences may promote understanding of the etiological differences and may identify effective treatment strategies for breast cancer in American Indian women. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 3589. doi:1538-7445.AM2012-3589

Collaboration


Dive into the Brij K. Gupta's collaboration.

Top Co-Authors

Avatar

Meena Jaggi

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Subhash C. Chauhan

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Diane M. Maher

University of South Dakota

View shared research outputs
Top Co-Authors

Avatar

Mara C. Ebeling

University of South Dakota

View shared research outputs
Top Co-Authors

Avatar

Murali M. Yallapu

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Susan E. Puumala

University of South Dakota

View shared research outputs
Top Co-Authors

Avatar

Vasudha Sundram

University of South Dakota

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Douglas W. Lynch

University of South Dakota

View shared research outputs
Top Co-Authors

Avatar

Michael D. Koch

University of South Dakota

View shared research outputs
Researchain Logo
Decentralizing Knowledge