36 results on '"Vivek K. Kashyap"'
Search Results
2. Figure S2 from Ormeloxifene Suppresses Prostate Tumor Growth and Metastatic Phenotypes via Inhibition of Oncogenic β-catenin Signaling and EMT Progression
- Author
-
Meena Jaggi, Subhash C. Chauhan, Murali M. Yallapu, Man M. Singh, Nadeem Zafar, Fathi T. Halaweish, Manish K. Tripathi, Andrew E. Massey, Shabnam Malik, Neeraj Chauhan, Zubair Bin Hafeez, Vivek K. Kashyap, Mohammed Sikander, Aditya Ganju, and Bilal Bin Hafeez
- Abstract
Effect of ORM on colonigenic and invasive potential of C4-2 cells: A. Effect of ORM on proliferation of C4-2 cells as determined by colony formation assay. B. Effect of ORM anchorage-independent growth of C4-2 cells as determined by soft agar colony Formation. Representative images showing colony formation in control and ORM treated groups. Bar graph indicates number of colony formed in control and ORM treated groups. Values represent mean {plus minus}SE of 3 plates. C. Effect of ORM on invasion of C4-2 cells as determined by Boyden chamber assay. i) Representative images showing invaded cells in control and ORM treated groups after 24 hrs. ii) Bar graph indicating average number of invaded cells in control and ORM treated groups.
- Published
- 2023
- Full Text
- View/download PDF
3. Data from Ormeloxifene Suppresses Prostate Tumor Growth and Metastatic Phenotypes via Inhibition of Oncogenic β-catenin Signaling and EMT Progression
- Author
-
Meena Jaggi, Subhash C. Chauhan, Murali M. Yallapu, Man M. Singh, Nadeem Zafar, Fathi T. Halaweish, Manish K. Tripathi, Andrew E. Massey, Shabnam Malik, Neeraj Chauhan, Zubair Bin Hafeez, Vivek K. Kashyap, Mohammed Sikander, Aditya Ganju, and Bilal Bin Hafeez
- Abstract
Ormeloxifene is a clinically approved selective estrogen receptor modulator, which has also shown excellent anticancer activity, thus it can be an ideal repurposing pharmacophore. Herein, we report therapeutic effects of ormeloxifene on prostate cancer and elucidate a novel molecular mechanism of its anticancer activity. Ormeloxifene treatment inhibited epithelial-to-mesenchymal transition (EMT) process as evident by repression of N-cadherin, Slug, Snail, vimentin, MMPs (MMP2 and MMP3), β-catenin/TCF-4 transcriptional activity, and induced the expression of pGSK3β. In molecular docking analysis, ormeloxifene showed proficient docking with β-catenin and GSK3β. In addition, ormeloxifene induced apoptosis, inhibited growth and metastatic potential of prostate cancer cells and arrested cell cycle in G0–G1 phase via modulation of cell-cycle regulatory proteins (inhibition of Mcl-1, cyclin D1, and CDK4 and induction of p21 and p27). In functional assays, ormeloxifene remarkably reduced tumorigenic, migratory, and invasive potential of prostate cancer cells. In addition, ormeloxifene treatment significantly (P < 0.01) regressed the prostate tumor growth in the xenograft mouse model while administered through intraperitoneal route (250 μg/mouse, three times a week). These molecular effects of ormeloxifene were also observed in excised tumor tissues as shown by immunohistochemistry analysis. Our results, for the first time, demonstrate repurposing potential of ormeloxifene as an anticancer drug for the treatment of advanced stage metastatic prostate cancer through a novel molecular mechanism involving β-catenin and EMT pathway. Mol Cancer Ther; 16(10); 2267–80. ©2017 AACR.
- Published
- 2023
- Full Text
- View/download PDF
4. Figure S4 from Ormeloxifene Suppresses Prostate Tumor Growth and Metastatic Phenotypes via Inhibition of Oncogenic β-catenin Signaling and EMT Progression
- Author
-
Meena Jaggi, Subhash C. Chauhan, Murali M. Yallapu, Man M. Singh, Nadeem Zafar, Fathi T. Halaweish, Manish K. Tripathi, Andrew E. Massey, Shabnam Malik, Neeraj Chauhan, Zubair Bin Hafeez, Vivek K. Kashyap, Mohammed Sikander, Aditya Ganju, and Bilal Bin Hafeez
- Abstract
Effect of ORM on cell cycle analysis and β-catenin localization. A. Histogram (i) and table (ii) represent the cell cycle distribution in DU145 cells. B. Confocal images of β-catenin, DAPI and merged images of control (i) and ORM treated (ii) PC3 cells.
- Published
- 2023
- Full Text
- View/download PDF
5. Figure S1 from Ormeloxifene Suppresses Prostate Tumor Growth and Metastatic Phenotypes via Inhibition of Oncogenic β-catenin Signaling and EMT Progression
- Author
-
Meena Jaggi, Subhash C. Chauhan, Murali M. Yallapu, Man M. Singh, Nadeem Zafar, Fathi T. Halaweish, Manish K. Tripathi, Andrew E. Massey, Shabnam Malik, Neeraj Chauhan, Zubair Bin Hafeez, Vivek K. Kashyap, Mohammed Sikander, Aditya Ganju, and Bilal Bin Hafeez
- Abstract
Effect of ORM on proliferation of various human PrCa cell lines as determined by counting the cells.
- Published
- 2023
- Full Text
- View/download PDF
6. Stem cell therapy: a novel approach against emerging and re-emerging viral infections with special reference to SARS-CoV-2
- Author
-
Vishal Khandelwal, Tarubala Sharma, Saurabh Gupta, Shoorvir Singh, Manish Kumar Sharma, Deepak Parashar, and Vivek K. Kashyap
- Subjects
Genetics ,General Medicine ,Molecular Biology - Abstract
The past several decades have witnessed the emergence and re-emergence of many infectious viral agents, flaviviruses, influenza, filoviruses, alphaviruses, and coronaviruses since the advent of human deficiency virus (HIV). Some of them even become serious threats to public health and have raised major global health concerns. Several different medicinal compounds such as anti-viral, anti-malarial, and anti-inflammatory agents, are under investigation for the treatment of these viral diseases. These therapies are effective improving recovery rates and overall survival of patients but are unable to heal lung damage caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Therefore, there is a critical need to identify effective treatments to combat this unmet clinical need. Due to its antioxidant and immunomodulatory properties, stem cell therapy is considered a novel approach to regenerate damaged lungs and reduce inflammation. Stem cell therapy uses a heterogeneous subset of regenerative cells that can be harvested from various adult tissue types and is gaining popularity due to its prodigious regenerative potential as well as immunomodulatory and anti-inflammatory properties. These cells retain expression of cluster of differentiation markers (CD markers), interferon-stimulated gene (ISG), reduce expression of pro-inflammatory cytokines and, show a rapid proliferation rate, which makes them an attractive tool for cellular therapies and to treat various inflammatory and viral-induced injuries. By examining various clinical studies, this review demonstrates positive considerations for the implications of stem cell therapy and presents a necessary approach for treating virally induced infections in patients.
- Published
- 2022
7. A topography of immunotherapies against gastrointestinal malignancies
- Author
-
Rinaldo Pellicano, Anukriti A, Partha Laskar, Sharmila Fagoonee, Poornima D. Shaji, Anupam Dhasmana, Subhash C. Chauhan, Vivek K. Kashyap, Sudhir Kotnala, Shafiul Haque, Swati Dhasmana, Murali M. Yallapu, Sheema Khan, and Meena Jaggi
- Subjects
Mechanism (biology) ,business.industry ,medicine.medical_treatment ,Cancer ,General Medicine ,Disease ,Immunotherapy ,medicine.disease ,Bioinformatics ,Clinical trial ,Immune system ,Cancer immunotherapy ,Cancer cell ,medicine ,Humans ,Immunologic Factors ,business ,Gastrointestinal Neoplasms - Abstract
Gastrointestinal (GI) cancers are one of the leading causes of death worldwide. Although various approaches are implemented to improve the health condition of GI patients, none of the treatment protocols promise for eradicating cancer. However, a treatment mechanism against any kind of disease condition is already existing executing inside the human body. The 'immune system' is highly efficient to detect and destroy the unfavourable events of the body including tumor cells. The immune system can restrict the growth and proliferation of cancer. Cancer cells behave much smarter and adopt new mechanisms for hiding from the immune cells. Thus, cancer immunotherapy might play a decisive role to train the immune system against cancer. In this review, we have discussed the immunotherapy permitted for the treatment of GI cancers. We have discussed various methods and mechanisms, periodic development of cancer immunotherapies, approved biologicals, completed and ongoing clinical trials, role of various biopharmaceuticals, and epigenetic factors involved in GI cancer immunotherapies (graphical abstract Figure 1).
- Published
- 2022
- Full Text
- View/download PDF
8. Cross-Linked Polyphenol-Based Drug Nano-Self-Assemblies Engineered to Blockade Prostate Cancer Senescence
- Author
-
Prashanth K.B. Nagesh, Subhash C. Chauhan, Manish K. Tripathi, Vivek K. Kashyap, Pallabita Chowdhury, Elham Hatami, Murali M. Yallapu, Meena Jaggi, Sonam Kumari, Bernd Meibohm, and Santosh Wagh
- Subjects
Male ,Senescence ,Drug ,Materials science ,media_common.quotation_subject ,medicine.medical_treatment ,Transplantation, Heterologous ,Cancer relapse ,Receptor, Transforming Growth Factor-beta Type I ,Mice, Nude ,Antineoplastic Agents ,Apoptosis ,Docetaxel ,Article ,Mice ,03 medical and health sciences ,Prostate cancer ,0302 clinical medicine ,Cell Line, Tumor ,medicine ,Animals ,Humans ,General Materials Science ,Cellular Senescence ,030304 developmental biology ,media_common ,0303 health sciences ,Chemotherapy ,Forkhead Box Protein O1 ,Polyphenols ,Prostatic Neoplasms ,Cancer ,medicine.disease ,Nanostructures ,Blockade ,Polyphenol ,030220 oncology & carcinogenesis ,Cancer research ,Tannins ,Signal Transduction - Abstract
Cellular senescence is one of the prevailing issues in cancer therapeutics that promotes cancer relapse, chemoresistance, and recurrence. Patients undergoing persistent chemotherapy often develop drug-induced senescence. Docetaxel, an FDA-approved treatment for prostate cancer, is known to induce cellular senescence which often limits the overall survival of patients. Strategic therapies that counter the cellular and drug-induced senescence are an unmet clinical need. Towards this an effort was made to develop a novel therapeutic strategy that targets and removes senescent cells from the tumors, we developed a nanoformulation of tannic acid−docetaxel self-assemblies (DSAs). The construction of DSAs was confirmed through particle size measurements, spectroscopy, thermal, and biocompatibility studies. This formulation exhibited enhanced in vitro therapeutic activity in various biological functional assays with respect to native docetaxel treatments. Microarray and immunoblot analysis results demonstrated that DSAs exposure selectively deregulated senescence associated TGFβR1/FOXO1/p21 signaling. Decrease in β-galactosidase staining further suggested reversion of drug-induced senescence after DSAs exposure. Additionally, DSAs induced profound cell death by activation of apoptotic signaling through bypassing senescence. Furthermore, in vivo and ex vivo imaging analysis demonstrated the tumor targeting behavior of DSAs in mice bearing PC-3 xenograft tumors. The antisenescence and anticancer activity of DSAs was further shown in vivo by inhibiting TGFβR1 proteins and regressing tumor growth through apoptotic induction in the PC-3 xenograft mouse model. Overall, DSAs exhibited such advanced features due to a natural compound in the formulation as a matrix/binder for docetaxel. Overall, DSAs showed superior tumor targeting and improved cellular internalization, promoting docetaxel efficacy. These findings may have great implications in prostate cancer therapy.
- Published
- 2019
- Full Text
- View/download PDF
9. Emerging Roles and Potential Applications of Non-Coding RNAs in Cervical Cancer
- Author
-
Deepak Parashar, Anupam Singh, Saurabh Gupta, Aishwarya Sharma, Manish K. Sharma, Kuldeep K. Roy, Subhash C. Chauhan, and Vivek K. Kashyap
- Subjects
RNA, Untranslated ,Neoplastic Stem Cells ,Tumor Microenvironment ,Genetics ,Humans ,Uterine Cervical Neoplasms ,Female ,Exosomes ,Prognosis ,Genetics (clinical) - Abstract
Cervical cancer (CC) is a preventable disease using proven interventions, specifically prophylactic vaccination, pervasive disease screening, and treatment, but it is still the most frequently diagnosed cancer in women worldwide. Patients with advanced or metastatic CC have a very dismal prognosis and current therapeutic options are very limited. Therefore, understanding the mechanism of metastasis and discovering new therapeutic targets are crucial. New sequencing tools have given a full visualization of the human transcriptome’s composition. Non-coding RNAs (NcRNAs) perform various functions in transcriptional, translational, and post-translational processes through their interactions with proteins, RNA, and even DNA. It has been suggested that ncRNAs act as key regulators of a variety of biological processes, with their expression being tightly controlled under physiological settings. In recent years, and notably in the past decade, significant effort has been made to examine the role of ncRNAs in a variety of human diseases, including cancer. Therefore, shedding light on the functions of ncRNA will aid in our better understanding of CC. In this review, we summarize the emerging roles of ncRNAs in progression, metastasis, therapeutics, chemo-resistance, human papillomavirus (HPV) regulation, metabolic reprogramming, diagnosis, and as a prognostic biomarker of CC. We also discussed the role of ncRNA in the tumor microenvironment and tumor immunology, including cancer stem cells (CSCs) in CC. We also address contemporary technologies such as antisense oligonucleotides, CRISPR–Cas9, and exosomes, as well as their potential applications in targeting ncRNAs to manage CC.
- Published
- 2022
- Full Text
- View/download PDF
10. Abstract 952: Attenuation of β-Catenin signaling by ormeloxifene in hepatocellular carcinoma
- Author
-
Mohammed Sikander, Shabnam Malik, Anyssa Rodriguez, Molly Vela, Daniel Zubieta, Vivek K. Kashyap, Anupam Dhasmana, Bilal B. Hafeez, Sheema Khan, Fathi T. Halaweish, Subhash C. Chauhan, and Meena Jaggi
- Subjects
Cancer Research ,Oncology - Abstract
Background: Hepatocellular carcinoma (HCC) is the most frequent type of primary liver cancer with approximately 41,000 new diagnosis and 29,000 fatalities every year only in the U.S. Severe toxicity and poor response to available chemotherapeutic agents make HCC clinical management extremely difficult. Aberrant β-catenin signaling is found to play a critical role in differentiation grades of HCC patients. Thus, attempts to find more potent and non-toxic β-Catenin inhibitors is urgently needed for cancer therapy. Notably, ormeloxifene (ORM), clinically approved selective estrogen receptor modulator with therapeutic index, has shown excellent anticancer activity, rendering it as an ideal candidate for repurposing. In this study, we investigated the anti-cancer potential of ORM against hepatocellular carcinoma. Methodology: Cell proliferation and colony formation assays were performed to assess the therapeutic activity of ORM in human hepatocellular carcinoma (HepG2, SK-HEP-1, Hep3B, and C3A) cells. Boyden chamber and Matrigel assays were carried out for investigating the effect of ORM on migration and invasion abilities of HCC cells, respectively. The effects of ORM on β-catenin and EMT associated proteins were analyzed through Western blotting and qPCR. Confocal microscopy was used to determine the β-catenin nuclear localization following ORM treatment in HCC cells. Results: As compared to vehicle-treated group, ORM treatment (2.5-20 µM) suppressed proliferation and colony formation in human hepatocellular carcinoma cells in a dose and time-dependent manner. Moreover, ORM treatment suppresses the migration and invasion of human hepatocellular carcinoma cells as shown by wound healing and Matrigel invasion assay, respectively. ORM effectively inhibited the protein levels and mRNA expression of total β-catenin. Additionally, our confocal microscopy results further showed reduced nuclear translocation of β-catenin following the ORM treatment. ORM treatment inhibited epithelial-to-mesenchymal transition (EMT) process as evident by repression of N-cadherin, Slug, Snail, vimentin, MMPs (MMP2 and MMP3), and induced the expression of pGSK3β. Experiments are also being conducted to see how ORM affects epigenetic markers linked to EMT and β-catenin signaling. Conclusion: Taken together, ORM inhibited β-catenin signaling and exhibited potent anticancer effects against HCC and could be investigated further as a novel therapeutic modality for HCC treatment. Citation Format: Mohammed Sikander, Shabnam Malik, Anyssa Rodriguez, Molly Vela, Daniel Zubieta, Vivek K. Kashyap, Anupam Dhasmana, Bilal B. Hafeez, Sheema Khan, Fathi T. Halaweish, Subhash C. Chauhan, Meena Jaggi. Attenuation of β-Catenin signaling by ormeloxifene in hepatocellular carcinoma [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 952.
- Published
- 2022
- Full Text
- View/download PDF
11. Withania somnifera as a potential future drug molecule for COVID-19
- Author
-
Murali M. Yallapu, Subhash C. Chauhan, Vivek K. Kashyap, Anupam Dhasmana, and Meena Jaggi
- Subjects
2019-20 coronavirus outbreak ,biology ,Coronavirus disease 2019 (COVID-19) ,SARS-CoV-2 ,business.industry ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,acute respiratory distress syndrome ,Withania somnifera ,biology.organism_classification ,Drug molecule ,Virology ,angiotensin-converting enzyme 2 ,Angiotensin-converting enzyme 2 ,Commentary ,Medicine ,intranasal delivery ,business - Published
- 2020
- Full Text
- View/download PDF
12. Reprogramming of pancreatic adenocarcinoma immunosurveillance by a microbial probiotic siderophore
- Author
-
Mehdi Chaib, Bilal B. Hafeez, Hassan Mandil, Deidre Daria, Ajeeth K. Pingili, Sonam Kumari, Mohammed Sikander, Vivek K. Kashyap, Guo-Yun Chen, Emmanuel Anning, Manish K. Tripathi, Sheema Khan, Stephen Behrman, Murali M. Yallapu, Meena Jaggi, Liza Makowski, and Subhash C. Chauhan
- Subjects
Pancreatic Neoplasms ,Mice ,Monitoring, Immunologic ,Probiotics ,Tumor Microenvironment ,Medicine (miscellaneous) ,Animals ,Siderophores ,Adenocarcinoma ,General Agricultural and Biological Sciences ,Immune Checkpoint Inhibitors ,General Biochemistry, Genetics and Molecular Biology ,Ferrichrome - Abstract
There is increasing evidence suggesting the role of microbiome alterations in relation to pancreatic adenocarcinoma and tumor immune functionality. However, molecular mechanisms of the interplay between microbiome signatures and/or their metabolites in pancreatic tumor immunosurveillance are not well understood. We have identified that a probiotic strain (Lactobacillus casei) derived siderophore (ferrichrome) efficiently reprograms tumor-associated macrophages (TAMs) and increases CD8 + T cell infiltration into tumors that paralleled a marked reduction in tumor burden in a syngeneic mouse model of pancreatic cancer. Interestingly, this altered immune response improved anti-PD-L1 therapy that suggests promise of a novel combination (ferrichrome and immune checkpoint inhibitors) therapy for pancreatic cancer treatment. Mechanistically, ferrichrome induced TAMs polarization via activation of the TLR4 pathway that represses the expression of iron export protein ferroportin (FPN1) in macrophages. This study describes a novel probiotic based molecular mechanism that can effectively induce anti-tumor immunosurveillance and improve immune checkpoint inhibitors therapy response in pancreatic cancer.
- Published
- 2020
13. Topological and system-level protein interaction network (PIN) analyses to deduce molecular mechanism of curcumin
- Author
-
Uma Bhardwaj, Subhash C. Chauhan, Meena Jaggi, Murali M. Yallapu, Meenu Gupta, Vivek K. Kashyap, Pallavi Somvanshi, Anukriti, Anupam Dhasmana, Shafiul Haque, and Swati Uniyal
- Subjects
0301 basic medicine ,Curcumin ,lcsh:Medicine ,Molecular Dynamics Simulation ,Topology ,Article ,Cancer prevention ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Interaction network ,Protein Interaction Mapping ,System level ,Protein Interaction Maps ,lcsh:Science ,Mode of action ,Cancer ,Multidisciplinary ,Dose-Response Relationship, Drug ,Extramural ,lcsh:R ,Computational Biology ,Molecular Sequence Annotation ,Molecular Docking Simulation ,Kinetics ,030104 developmental biology ,chemistry ,030220 oncology & carcinogenesis ,Molecular mechanism ,lcsh:Q ,Protein network ,Signalling pathways ,Protein Binding ,Signal Transduction - Abstract
Curcumin is an important bioactive component of turmeric and also one of the important natural products, which has been investigated extensively. The precise mode of action of curcumin and its impact on system level protein networks are still not well studied. To identify the curcumin governed regulatory action on protein interaction network (PIN), an interectome was created based on 788 key proteins, extracted from PubMed literatures, and constructed by using STRING and Cytoscape programs. The PIN rewired by curcumin was a scale-free, extremely linked biological system. MCODE plug-in was used for sub-modulization analysis, wherein we identified 25 modules; ClueGo plug-in was used for the pathway’s enrichment analysis, wherein 37 enriched signalling pathways were obtained. Most of them were associated with human diseases groups, particularly carcinogenesis, inflammation, and infectious diseases. Finally, the analysis of topological characteristic like bottleneck, degree, GO term/pathways analysis, bio-kinetics simulation, molecular docking, and dynamics studies were performed for the selection of key regulatory proteins of curcumin-rewired PIN. The current findings deduce a precise molecular mechanism that curcumin might exert in the system. This comprehensive in-silico study will help to understand how curcumin induces its anti-cancerous, anti-inflammatory, and anti-microbial effects in the human body.
- Published
- 2020
- Full Text
- View/download PDF
14. Neutralization of SARS-CoV-2 Spike Protein via Natural Compounds: A Multilayered High Throughput Virtual Screening Approach
- Author
-
Sudhir Kotnala, Ghulam Md Ashraf, Shafiul Haque, Anupam Dhasmana, Swati Dhasmana, Murali M. Yallapu, Vivek K. Kashyap, Subhash C. Chauhan, and Meena Jaggi
- Subjects
Pharmacology ,0303 health sciences ,Virtual screening ,Coronavirus disease 2019 (COVID-19) ,Drug discovery ,Computer science ,SARS-CoV-2 ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Druggability ,Spike Protein ,COVID-19 ,Computational biology ,01 natural sciences ,Antiviral Agents ,Neutralization ,0104 chemical sciences ,Molecular Docking Simulation ,010404 medicinal & biomolecular chemistry ,03 medical and health sciences ,Drug Discovery ,Pandemic ,Spike Glycoprotein, Coronavirus ,Humans ,030304 developmental biology - Abstract
Background: Previously human society has faced various unprecedented pandemics in the history and viruses have majorly held the responsibilities of those outbreaks. Furthermore, due to amplified global connection and speedy modernization, epidemic outbreaks caused by novel and re-emerging viruses signify potential risk to community health. Despite great advancements in immunization and drug discovery processes, various viruses still lack prophylactic vaccines and efficient antiviral therapies. Although, vaccine is a prophylaxes option, but it cannot be applied to infected patients, hence therapeutic interventions are urgently needed to control the ongoing global SARS- CoV-2 pandemic condition. To spot the novel antiviral therapy is of decisive importance and Mother Nature is an excellent source for such discoveries. Methodology: In this article, prompt high through-put virtual screening for vetting the best possible drug candidates from natural compounds’ databases has been implemented. Herein, time tested rigorous multi-layered drug screening process to narrow down 66,969 natural compounds for the identification of potential lead(s) is implemented. Druggability parameters, different docking approaches and neutralization tendency of the natural products were employed in this study to screen the best possible natural compounds from the digital libraries. Conclusion: The results of this study conclude that compounds PALA and HMCA are potential inhibitors of SARS-CoV-2 spike protein and can be further explored for experimental validation. Overall, the methodological approach reported in this article can be suitably used to find the potential drug candidates against SARS-CoV2 in the burning situation of COVID-19 with less expenditure and a concise span of time.
- Published
- 2020
15. Tannic acid inhibits lipid metabolism and induce ROS in prostate cancer cells
- Author
-
Murali M. Yallapu, Vivek K. Kashyap, Pallabita Chowdhury, Shashi Jain, Nirnoy Dan, Prashanth K.B. Nagesh, Subhash C. Chauhan, Elham Hatami, and Meena Jaggi
- Subjects
Male ,0301 basic medicine ,Membrane permeability ,Cell Survival ,Cell ,lcsh:Medicine ,Apoptosis ,Article ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,medicine ,Humans ,lcsh:Science ,Cell Proliferation ,chemistry.chemical_classification ,Reactive oxygen species ,Multidisciplinary ,Pharmaceutics ,Endoplasmic reticulum ,lcsh:R ,Prostate ,Prostatic Neoplasms ,Lipid metabolism ,Lipid signaling ,Endoplasmic Reticulum Stress ,Lipid Metabolism ,Cancer metabolism ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,chemistry ,030220 oncology & carcinogenesis ,Lipogenesis ,lcsh:Q ,Reactive Oxygen Species ,Tannins ,Signal Transduction - Abstract
Prostate cancer (PCa) cells exploit the aberrant lipid signaling and metabolism as their survival advantage. Also, intracellular storage lipids act as fuel for the PCa proliferation. However, few studies were available that addressed the topic of targeting lipid metabolism in PCa. Here, we assessed the tannic acid (TA) lipid-targeting ability and its capability to induce endoplasmic reticulum (ER) stress by reactive oxygen species (ROS) in PCa cells. TA exhibited dual effects by inhibiting lipogenic signaling and suppression of lipid metabolic pathways. The expression of proteins responsible for lipogenesis was down regulated. The membrane permeability and functionality of PCa were severely affected and caused nuclear disorganization during drug exposure. Finally, these consolidated events shifted the cell’s survival balance towards apoptosis. These results suggest that TA distinctly interferes with the lipid signaling and metabolism of PCa cells.
- Published
- 2020
- Full Text
- View/download PDF
16. Withania somnifera: Progress towards a Pharmaceutical Agent for Immunomodulation and Cancer Therapeutics
- Author
-
Vivek K. Kashyap, Godwin Peasah-Darkwah, Anupam Dhasmana, Meena Jaggi, Murali M. Yallapu, and Subhash C. Chauhan
- Subjects
Pharmaceutical Science - Abstract
Chemotherapy is one of the prime treatment options for cancer. However, the key issues with traditional chemotherapy are recurrence of cancer, development of resistance to chemotherapeutic agents, affordability, late-stage detection, serious health consequences, and inaccessibility. Hence, there is an urgent need to find innovative and cost-effective therapies that can target multiple gene products with minimal adverse reactions. Natural phytochemicals originating from plants constitute a significant proportion of the possible therapeutic agents. In this article, we reviewed the advances and the potential of Withania somnifera (WS) as an anticancer and immunomodulatory molecule. Several preclinical studies have shown the potential of WS to prevent or slow the progression of cancer originating from various organs such as the liver, cervix, breast, brain, colon, skin, lung, and prostate. WS extracts act via various pathways and provide optimum effectiveness against drug resistance in cancer. However, stability, bioavailability, and target specificity are major obstacles in combination therapy and have limited their application. The novel nanotechnology approaches enable solubility, stability, absorption, protection from premature degradation in the body, and increased circulation time and invariably results in a high differential uptake efficiency in the phytochemical’s target cells. The present review primarily emphasizes the insights of WS source, chemistry, and the molecular pathways involved in tumor regression, as well as developments achieved in the delivery of WS for cancer therapy using nanotechnology. This review substantiates WS as a potential immunomodulatory, anticancer, and chemopreventive agent and highlights its potential use in cancer treatment.
- Published
- 2022
- Full Text
- View/download PDF
17. Disparity in rates of HPV infection and cervical cancer in underserved US populations
- Author
-
Nadeem Zafar, Satish Kedia, Meena Jaggi, Murali M. Yallapu, Vivek K. Kashyap, Subhash C. Chauhan, and Asok Ranjan Karuri
- Subjects
Adult ,0301 basic medicine ,medicine.medical_specialty ,Population ,Uterine Cervical Neoplasms ,Article ,Health Services Accessibility ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Underserved Population ,0302 clinical medicine ,Humans ,Medicine ,education ,Cervical cancer ,Appalachian Region ,Pap smear screening ,education.field_of_study ,General Immunology and Microbiology ,business.industry ,Obstetrics ,Incidence ,Mortality rate ,Incidence (epidemiology) ,Papillomavirus Infections ,HPV infection ,medicine.disease ,United States ,Black or African American ,030104 developmental biology ,030220 oncology & carcinogenesis ,Female ,business ,Alcohol consumption - Abstract
There is a higher rate of HPV infection and cervical cancer incidence and mortality in underserved US population who reside in Appalachian mountain region compared to Northern Plains. Social and behavioral factors such as smoking and alcohol consumption are for such a high incidence. However, by and large, the reasons for these discrepancies lie in the reluctance of the underserved population to adopt preventive measures such as prophylactic Human papilloma virus (HPV) vaccines and Pap smear screening that have significantly reduced the incidence and mortality rate of cervical cancer in Caucasian women. Thus, it is clear that drastic change in social behavior and implementation of preventive measures is required to effectively reduce the incidence and mortality from cervical cancer in this underserved population.
- Published
- 2017
- Full Text
- View/download PDF
18. Therapeutic efficacy of a novel βIII/βIV-tubulin inhibitor (VERU-111) in pancreatic cancer
- Author
-
Neeraj Chauhan, Bilal Bin Hafeez, Miller Duane D, Vivek K. Kashyap, Wei Li, Subhash C. Chauhan, Saini Setua, Meena Jaggi, Murali M. Yallapu, Prashanth K.B. Nagesh, Qinghui Wang, Pallabita Chowdhury, and Sonam Kumari
- Subjects
0301 basic medicine ,Cancer Research ,Paclitaxel ,Carcinogenesis ,β –tubulins ,Apoptosis ,lcsh:RC254-282 ,miR-200c ,Metastasis ,Mice ,03 medical and health sciences ,βIII/βIV-tubulin inhibitor ,0302 clinical medicine ,Western blot ,Tubulin ,Cell Line, Tumor ,Pancreatic cancer ,medicine ,Animals ,Humans ,Neoplasm Metastasis ,Cyclin B1 ,Cell Proliferation ,Cyclin-dependent kinase 1 ,medicine.diagnostic_test ,biology ,Chemistry ,Panca ,Research ,Cancer ,VERU-111 ,Cell cycle ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,biology.organism_classification ,Xenograft Model Antitumor Assays ,Tubulin Modulators ,3. Good health ,Pancreatic Neoplasms ,030104 developmental biology ,Oncology ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Cancer research - Abstract
Background The management of pancreatic cancer (PanCa) is exceptionally difficult due to poor response to available therapeutic modalities. Tubulins play a major role in cell dynamics, thus are important molecular targets for cancer therapy. Among various tubulins, βIII and βIV-tubulin isoforms have been primarily implicated in PanCa progression, metastasis and chemo-resistance. However, specific inhibitors of these isoforms that have potent anti-cancer activity with low toxicity are not readily available. Methods We determined anti-cancer molecular mechanisms and therapeutic efficacy of a novel small molecule inhibitor (VERU-111) using in vitro (MTS, wound healing, Boyden chamber and real-time xCELLigence assays) and in vivo (xenograft studies) models of PanCa. The effects of VERU-111 treatment on the expression of β-tubulin isoforms, apoptosis, cancer markers and microRNAs were determined by Western blot, immunohistochemistry (IHC), confocal microscopy, qRT-PCR and in situ hybridization (ISH) analyses. Results We have identified a novel small molecule inhibitor (VERU-111), which preferentially represses clinically important, βIII and βIV tubulin isoforms via restoring the expression of miR-200c. As a result, VERU-111 efficiently inhibited tumorigenic and metastatic characteristics of PanCa cells. VERU-111 arrested the cell cycle in the G2/M phase and induced apoptosis in PanCa cell lines via modulation of cell cycle regulatory (Cdc2, Cdc25c, and Cyclin B1) and apoptosis - associated (Bax, Bad, Bcl-2, and Bcl-xl) proteins. VERU-111 treatment also inhibited tumor growth (P
- Published
- 2019
- Full Text
- View/download PDF
19. A bird eye view on cystic fibrosis: An underestimated multifaceted chronic disorder
- Author
-
Vivek K. Kashyap, Anupam Dhasmana, Sudhir Kotnala, Subhash C. Chauhan, Murali M. Yallapu, and Meena Jaggi
- Subjects
0301 basic medicine ,Cystic Fibrosis ,Cystic Fibrosis Transmembrane Conductance Regulator ,Disease ,medicine.disease_cause ,030226 pharmacology & pharmacy ,Cystic fibrosis ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Animals ,Humans ,General Pharmacology, Toxicology and Pharmaceutics ,Regulation of gene expression ,Mutation ,Lung ,biology ,SARS-CoV-2 ,business.industry ,Mucin ,Mucins ,COVID-19 ,General Medicine ,medicine.disease ,Mucus ,Cystic fibrosis transmembrane conductance regulator ,MicroRNAs ,030104 developmental biology ,medicine.anatomical_structure ,Gene Expression Regulation ,Immunology ,biology.protein ,RNA, Long Noncoding ,business - Abstract
Cystic fibrosis (CF) is an autosomal recessive disease which involves the mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene. CF involves in the inflammatory processes and is considered as a multisystem disorder that is not confined to lungs, but it also affects other vital organs that leads to numerous co-morbidities. The respiratory disorder in the CF results in mortality and morbidity which is characterized by series of serious events involving mucus hypersecretion, microbial infections, airways obstruction, inflammation, destruction of epithelium, tissue remodeling and terminal lung diseases. Mucins are the high molecular weight glycoproteins important for the viscoelastic properties of the mucus, play a significant role in the disease mechanisms. Determining the functional association between the CFTR and mucins might help to identify the putative target for specific therapeutic approach. In fact, furin enzyme which helps in the entry of novel COVID-19 virus into the cell, is upregulated in CF and this can also serve as a potential target for CF treatment. Moreover, the use of nano-formulations for CF treatment is an area of research being widely studied as they have also demonstrated promising outcomes. The in-depth knowledge of non-coding RNAs like miRNAs and lncRNAs and their functional association with CFTR gene expression and mutation can provide a different range of opportunity to identify the promising therapeutic approaches for CF.
- Published
- 2021
- Full Text
- View/download PDF
20. Smoking and COVID-19: Adding Fuel to the Flame
- Author
-
Anupam Dhasmana, Vivek K. Kashyap, Andrew E. Massey, Meena Jaggi, Sudhir Kotnala, Subhash C. Chauhan, Nadeem Zafar, and Murali M. Yallapu
- Subjects
0301 basic medicine ,medicine.medical_specialty ,medicine.medical_treatment ,Pneumonia, Viral ,Ethnic group ,α7-nAChR ,Review ,Disease ,smoking ,Catalysis ,lcsh:Chemistry ,Inorganic Chemistry ,03 medical and health sciences ,0302 clinical medicine ,Epidemiology ,Pandemic ,medicine ,Humans ,vaping ,030212 general & internal medicine ,Physical and Theoretical Chemistry ,Risk factor ,Intensive care medicine ,Pandemics ,lcsh:QH301-705.5 ,Molecular Biology ,Spectroscopy ,SARS-CoV-2 ,business.industry ,Organic Chemistry ,COVID-19 ,e-cigarettes ,General Medicine ,medicine.disease ,Health equity ,Computer Science Applications ,030104 developmental biology ,lcsh:Biology (General) ,lcsh:QD1-999 ,Smoking cessation ,hooka and cytokine storm ,Coronavirus Infections ,Cytokine storm ,business ,ACE-2 - Abstract
The coronavirus disease 2019 (COVID-19) pandemic, an infection caused by the severe acute respiratory syndrome coronavirus (SARS-CoV-2), has led to more than 771,000 deaths worldwide. Tobacco smoking is a major known risk factor for severe illness and even death from many respiratory infections. The effects of smoking on COVID-19 are currently controversial. Here, we provide an overview of the current knowledge on the effects of smoking on the clinical manifestations, disease progression, inflammatory responses, immunopathogenesis, racial ethnic disparities, and incidence of COVID-19. This review also documents future directions of smoking related research in COVID-19. The current epidemiological finding suggests that active smoking is associated with an increased severity of disease and death in hospitalized COVID-19 patients. Smoking can upregulate the angiotensin-converting enzyme-2 (ACE-2) receptor utilized by SARS-CoV-2 to enter the host cell and activate a ‘cytokine storm’ which can lead to worsen outcomes in COVID-19 patients. This receptor can also act as a potential therapeutic target for COVID-19 and other infectious diseases. The COVID-19 pandemic sheds light on a legacy of inequalities regarding gender, racial, and ethnic health disparities associated with active smoking, thus, smoking cessation may help in improving outcomes. In addition, to flatten the COVID-19 curve, staying indoors, avoiding unnecessary social contact, and bolstering the immune defense system by maintaining a healthy diet/living are highly desirable.
- Published
- 2020
- Full Text
- View/download PDF
21. Next-generation paclitaxel-nanoparticle formulation for pancreatic cancer treatment
- Author
-
Neeraj Chauhan, Andrew E. Massey, Bilal Bin Hafeez, Sheema Khan, Saini Setua, Subhash C. Chauhan, Hassan Mandil, Sonam Kumari, Vivek K. Kashyap, Meena Jaggi, Mohammed Sikander, Murali M. Yallapu, and Advait B. Shetty
- Subjects
Paclitaxel ,media_common.quotation_subject ,Biomedical Engineering ,Mice, Nude ,Pharmaceutical Science ,Medicine (miscellaneous) ,Apoptosis ,Bioengineering ,Vimentin ,02 engineering and technology ,Article ,Metastasis ,03 medical and health sciences ,chemistry.chemical_compound ,Membrane Microdomains ,Polylactic Acid-Polyglycolic Acid Copolymer ,Cell Movement ,Cell Line, Tumor ,Pancreatic cancer ,medicine ,Animals ,Humans ,Neoplasm Invasiveness ,General Materials Science ,Neoplasm Metastasis ,Internalization ,Cell Proliferation ,030304 developmental biology ,media_common ,0303 health sciences ,biology ,Panca ,Chemistry ,Cell Cycle Checkpoints ,021001 nanoscience & nanotechnology ,medicine.disease ,biology.organism_classification ,Xenograft Model Antitumor Assays ,Endocytosis ,In vitro ,Pancreatic Neoplasms ,Drug Resistance, Neoplasm ,biology.protein ,Cancer research ,Nanoparticles ,Molecular Medicine ,Immunohistochemistry ,0210 nano-technology - Abstract
Pancreatic cancer (PanCa) is a major cause of cancer-related death due to limited therapeutic options. As pancreatic tumors are highly desmoplastic, they prevent appropriate uptake of therapeutic payloads. Thus, our objective is to develop a next-generation nanoparticle system for treating PanCa. We generated a multi-layered Pluronic F127 and polyvinyl alcohol stabilized and poly-L-lysine coated paclitaxel loaded poly(lactic-co-glycolic acid) nanoparticle formulation (PPNPs). This formulation exhibited optimal size (~160 nm) and negative Zeta potential (−6.02 mV), efficient lipid raft mediated internalization, pronounced inhibition in growth and metastasis in vitro, and in chemo-naive and chemo-exposed orthotopic xenograft mouse models. Additionally, PPNPs altered nanomechanical properties of PanCa cells as suggested by the increased elastic modulus in nanoindentation analyses. Immunohistochemistry of orthotopic tumors demonstrated decreased expression of tumorigenic and metastasis associated proteins (ki67, vimentin and slug) in PPNPs treated mice. These results suggest that PPNPs represent a viable and robust platform for (PanCa).
- Published
- 2019
- Full Text
- View/download PDF
22. Abstract 3634: Tannic acid-docetaxel scaffold nanoparticles for improved prostate cancer therapy
- Author
-
Sonam Kumari, Elham Hatami, Pallabita Chowdhury, Sheema Khan, Vivek K. Kashyap, Prashanth Kumar Bhusetty Nagesh, Bilal Bin Hafeez, Subhash C. Chauhan, Murali M. Yallapu, Manish K. Tripathi, and Meena Jaggi
- Subjects
Cancer Research ,Cancer ,medicine.disease ,In vitro ,chemistry.chemical_compound ,Prostate cancer ,Oncology ,chemistry ,Docetaxel ,In vivo ,Apoptosis ,Tannic acid ,Cancer cell ,medicine ,Cancer research ,medicine.drug - Abstract
Purpose: Prostate cancer (PrCa) is one of the leading causes of cancer-related deaths among men in the United States. Chemotherapy with docetaxel holds great promise and important therapeutic option for PrCa treatment. However, chemotherapy is often associated with incomplete cell death and leads to persistent senescent phenotype. Such phenotypic cancer cells survive and promotes tumor development, recurrence, and drug resistance. Altogether, targeting these cells can enhance chemotherapy outcomes. In the current study, we established a tannic acid-docetaxel scaffold nanoparticles (TDS NP) platform that allows for facile targeting and inhibition of drug induced senescence in prostate cancer. Methods: TDS NP were prepared by solvent evaporation and extrusion process using a series of tannic acid and docetaxel weight ratios. The scaffold self-assembly formation was determined using a steady-state fluorescence quenching, FT-IR, XRD, DSC, and TGA. The surface and morphological properties of TDS NPs were determined by dynamic light scattering and transmission electron microscopy. The biocompatibility assessment was characterized by hemolysis assay. C4-2 and PC-3 cell lines were used as PrCa model systems for in vitro and in vivo studies. The cellular internalization (in vitro) and accumulation (in vivo) was examined using dye-tagged TDS NPs. The superior in vitro anti-cancer and metastatic potential of TDS NPs were evaluated using proliferation, colony formation, migration, invasion, and immunoblotting assays. The anti-senescence ability of TDS NPs was examined through β-Galactosidase Staining Kit. A PC-3 xenograft mouse model was used to examine its superior therapeutic activity over free docetaxel treatment. Results: A series of physico-chemical analyses confirmed the integrity of TDS NP. An optimized formulation exhibited a spherical shape particle formulation with 124.13 ± 2.16 nm with a negative zeta potential of -14.0 mV. TDS NPs formulation exhibited superior internalization capacity in PrCa cells in a dose- and time-dependent manner. In vitro functional studies confirm superior therapeutic activity of TDS NPs over free docetaxel treatment. Enhanced pro-apoptotic while downregulating anti-apoptotic effects with treatment of TDS NPs in comparison to native drug. A profound inhibition of β-galactosidase activity was noticed with TDS NPs treatment. In vivobiodistribution studies confirm efficient accumulation of TDS NPs in mice. In addition, TDS NPs showed robust antitumor activity against PC-3 xenografts. Such improved activity was correlated with increased inhibition of senescence and induced apoptosis. Conclusion: In summary, we developed a novel tannic acid-docetaxel scaffold nanoparticle formulation that is capable of inhibiting senescence, minimizing drug resistance, and inducing apoptosis in prostate cancer. Citation Format: Prashanth Kumar Bhusetty Nagesh, Pallabita Chowdhury, Elham Hatami, Sonam Kumari, Vivek Kumar Kashyap, Bilal Hafeez, Sheema Khan, Manish Kumar Tripathi, Meena Jaggi, Subhash C. Chauhan, Murali M. Yallapu. Tannic acid-docetaxel scaffold nanoparticles for improved prostate cancer therapy [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 3634.
- Published
- 2019
- Full Text
- View/download PDF
23. Abstract 3619: Nanoparticle formulation of VERU 111 for pancreatic cancer treatment
- Author
-
Duane D. Miller, Subhash C. Chauhan, Vivek K. Kashyap, Qinghui Wang, Bilal Bin Hafeez, Wei Li, Murali M. Yallapu, Prashanth K.B. Nagesh, Meena Jaggi, and Neeraj Chauhan
- Subjects
Cancer Research ,medicine.diagnostic_test ,biology ,Chemistry ,Panca ,media_common.quotation_subject ,Cancer ,medicine.disease ,biology.organism_classification ,In vitro ,Oncology ,Western blot ,Apoptosis ,In vivo ,Pancreatic cancer ,Cancer research ,medicine ,Internalization ,media_common - Abstract
Background: Pancreatic cancer (PanCa) is one of the leading causes of cancer-related mortality in the United States due to very limited therapeutic options. Thus, developing novel therapeutic strategies will help for the management of this disease. We recently identified VERU-111, a novel synthetic molecule which showed potent anti-cancer effect against PanCa via targeting clinically important βIII and βIV tubulin isoforms. In this study, we synthesized and characterized its novel nanoformulation (MNP-VERU) and evaluated its therapeutic effects in vitro and xenograft mouse model. Methods: MNPs were prepared by chemical precipitation method and loaded with VERU-111 using diffusion method. This formulation was characterized for particle size, chemical composition, and drug loading efficiency, using various physico-chemical methods (TEM, FT-IR, DSC, TGA, and HPLC). The internalization of MNP-VERU was achieved after 6 hours incubation with MNP-VERU in PanCa cells. To determine therapeutic efficacy of MNP-VERU, we performed various in vitro (MTS, wound healing, boyden chamber real-time xCELLigence, and apoptosis assays) and in vivo (mouse tumor xenograft) studies using PanCa. Effect of MNP-VERU on various key oncogenic signaling pathways, and miRNAs was evaluated by Western blot, immunohistochemistry (IHC), confocal microscopy, qRT-PCR and in situ hybridization (ISH) analyses respectively. Result: Our novel MNP-VERU formulation provided average size of 110 nm in dynamic light scattering (DLS) and exhibited -8.23 to -11.65 mV zeta potential with an outstanding loading efficiency (94%). Cellular uptake and internalization studies demonstrate that MNP-VERU escape lysosomal degradation, providing efficient endosomal release to cytosol. MNP-VERU showed remarkable anti-cancer potential in various PanCa cells (Panc-1, AsPC-1, HPAF-II, BxPC-3, MiaPaca) and more effectively repressed βIII and βIV tubulin isoforms via restoring the expression of miR-200c. MNP-VERU more effectively suppressed AsPC-1 cells derived xenograft tumors in athymic nude mice. Conclusions: Taken together, our results suggest that MNP-VERU has more anti-cancer potential than free VERU-111 against PanCa. MNP-VERU may reduce the toxicity and improve the bioavailability of free VERU-111 and could be used for the management of PanCa. Citation Format: Vivek K. Kashyap, Bilal B. Hafeez, Qinghui Wang, Neeraj Chauhan, Prashanth K. Nagesh, Murali M. Yallapu, Duane D. Miller, Wei Li, Meena Jaggi, Subhash C. Chauhan. Nanoparticle formulation of VERU 111 for pancreatic cancer treatment [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 3619.
- Published
- 2019
- Full Text
- View/download PDF
24. Abstract 5777: Ormeloxifene augments the therapeutic response of enzalutamide via targeting androgen receptor splice variant 7
- Author
-
Shabnam Malik, Advit Shetty, Mohammed Sikander, Meena Jaggi, Hassan Mandil, Vivek K. Kashyap, Andrew E. Massey, Mehdi Chaib, Mohan M. Yallapu, Bilal Bin Hafeez, and Subhash C. Chauhan
- Subjects
Cancer Research ,chemistry.chemical_compound ,Oncology ,chemistry ,business.industry ,medicine ,Cancer research ,Enzalutamide ,Androgen Receptor Splice Variant 7 ,Ormeloxifene ,business ,medicine.drug - Abstract
Enzalutamide (ENZ) is a second generation anti-androgen drug in clinical use for the treatment of advanced prostate cancer (PrCa). Use of ENZ has some limitations because of its toxic side effect and developing chemoresistance. It has been shown that expression of androgen receptor splice variant 7 (ARV7) in advanced PrCa is involved in ENZ therapy resistance. It is noteworthy that ENZ does not target AR splice variants because these lack the ligand binding domain of AR. Thus, identification of non-toxic agents which can target ARV7 can improve the enzalutamide therapy response and could be used for the management of advanced stage PrCa. Herein, we identified a clinically approved selective estrogen receptor modulator ormeloxifene (ORM), which augments ENZ therapy response via suppression of ARV7 expression in PrCa cells. We used androgen-independent human prostate cancer cells (22Rv1), which express ARV7 as a model system to study the effect of ORM alone or in combination with ENZ. ORM treatment showed a more potent effect in 22Rv1 cells as compared to ENZ alone. ORM treatment also sensitized the effects of ENZ in 22Rv1 cells. ORM treatment effectively inhibited colony formation ability of 22Rv1 cells; this effect was additive in combination with ENZ. We next performed Western blot analysis of ARV7 in control and ORM treated 22Rv1 cells. ORM effectively inhibited the protein level of AR V7 at 12 hrs post-treatment in 22Rv1 cells; however, no effect was observed with the treatment of ENZ. ORM treatment also inhibited the promoter activity of the AR target gene PSA as determined by luciferase assay. To translate these finding to an in vivo model system, we investigated the chemo-sensitization potential of ORM in a 22Rv1 cell-derived xenograft mouse model. ORM treatment (200 µg/mouse three times a week for 8 consecutive weeks) significantly (P Citation Format: Bilal B. Hafeez, Andrew E. Massey, Vivek K. Kashyap, Mohammed Sikander, Advit Shetty, Mehdi Chaib, Hassan Mandil, Shabnam Malik, Mohan M. Yallapu, Meena Jaggi, Subhash C. Chauhan. Ormeloxifene augments the therapeutic response of enzalutamide via targeting androgen receptor splice variant 7 [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 5777.
- Published
- 2018
- Full Text
- View/download PDF
25. Abstract LB-400: Tannic acid induces prostate cancer cell death via unfolded protein response (UPR) and modulation of CHOP
- Author
-
Vivek K. Kashyap, Bilal Bin Hafeez, Prashanth Kumar Bhusetty Nagesh, Subhash C. Chauhan, Meena Jaggi, Pallabita Chowdhury, Elham Hatami, Murali M. Yallapu, and Sheema Khan
- Subjects
Cancer Research ,Programmed cell death ,XBP1 ,Oncology ,DU145 ,Cell growth ,Chemistry ,Apoptosis ,ATF6 ,Endoplasmic reticulum ,Cancer research ,Unfolded protein response - Abstract
Objectives: Endoplasmic reticulum (ER) is an intricate organelle that is crucial for cellular function and survival. Cellular environments that interfere with ER functioning can lead to the accumulation of unfolded proteins, which are sensed by transmembrane sensors that instigate the unfolded protein response (UPR) to reinstate ER proteostasis. When the UPR is perturbed or not sufficient to deal with the stress conditions, apoptotic cell death is ensued. Often, prostate tumor cells (PrCa) are exposed to intrinsic and external factors that alter protein homeostasis producing endoplasmic reticulum (ER) stress. IRE1, PERK, and ATF6 are key signaling stems of the UPR during ER stress. At molecular level, dimerization of PERK kinase phosphorylates eIF2α, resulting in translation attenuation. This selectively enhances translation of the ATF4 transcription factor. Also, activated IRE1 stimulates the nuclear translocation of XBP1. Both ATF4 and XBP1 induces expression of UPR genes. The UPR stimulates and protects tumor cells against stressful conditions within the tumor microenvironment. Therapeutic intervention on this aspect ER Stress mediated apoptosis is much needed. Here, we examined the mechanistic role of tannic acid (TA) through novel targeting inhibition of UPR via modulation of ER stress in PrCa cell death. Methods: In vitro therapeutic perspective of tannic acid was evaluated using clinically relevant human prostate cancer cell line models (C4-2, PC3 and DU145) through cell proliferation and clonogenic assays. The anti-metastatic potential of TA in PrCa cells was determined using invasive and migration studies. The phase arrest and concomitant apoptosis in PrCa cells were examined using Propidium Iodide based flow cytometric studies. TA induced ER stress-mediated UPR cellular apoptotic activity was confirmed using Western blot, real-time studies, and nuclear distortion studies. Results: Protein profiling results have showed yin approach of inhibition ER stress during TA treatments with the inhibition of CHOP protein. Also, we observed TA treated PrCa cells was able to induce ER stress response proteins, such as PERK and IRE1. Its downstream signaling events include the induction of XBP1, EIF2α expression key mediators by alteration of Bcl2/Bax ratio from shifting survival towards apoptosis. With this line we assessed the expression profile of apoptosis-associated markers such as Bid and Bim (upregulated), and Bcl-2 and Bcl-xL which ratifies the induction of apoptosis during treatments. TA exhibited prominent growth arrest at G1 phase and increase of concentration led to elevated sub G1, annotating the increase of apoptotic cells. The expression of P18 and P21 were increased during TA treatments and cyclin D1 expression was inhibited supporting the G1 phase cell arrest. The expression of MMP2 and MMP9 were reduced, signifying the superior anti-metastatic ability of this compound. Cell proliferation studies showed a dose-dependent inhibitory profile of TA. Distinct results of anti-migratory and anti-metastatic effects of TA was observed during treatments. Conclusion: This study suggests that TA can efficiently induce UPR and promote ER stress signaling in cells that renders apoptosis in PrCa cells by the over expression of CHOP. We believe this novel therapeutic paradigm have clinical significance in advanced PrCa. Citation Format: Elham Hatami, Prashanth Kumar Bhusetty Nagesh, Pallabita Chowdhury, Vivek K. Kashyap, Sheema Khan, Bilal Hafeez, Meena Jaggi, Subhash C. Chauhan, Murali M. Yallapu. Tannic acid induces prostate cancer cell death via unfolded protein response (UPR) and modulation of CHOP [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr LB-400.
- Published
- 2018
- Full Text
- View/download PDF
26. Abstract LB-011: Novel nano-formulation of paclitaxel for pancreatic cancer therapy
- Author
-
Advit Shetty, Bilal Bin Hafeez, Mohammad Sikander, Subhash C. Chauhan, Mehdi Chaib, Mohan M. Yallapu, Meena Jaggi, Andrew E. Massey, Hassan Mandil, and Vivek K. Kashyap
- Subjects
Cancer Research ,Combination therapy ,biology ,Panca ,Cancer ,Pharmacology ,biology.organism_classification ,medicine.disease ,In vitro ,Gemcitabine ,PLGA ,chemistry.chemical_compound ,Oncology ,chemistry ,Paclitaxel ,Pancreatic cancer ,medicine ,medicine.drug - Abstract
Pancreatic cancer (PanCa) is one of the leading causes of cancer-related mortality in the United States due to very limited therapeutic options. Thus, there is an urgent need to develop novel therapeutic strategies for the management of this disease. Paclitaxel (PTX), a natural product derived from the Pacific Yew tree, is an agent typically used in combination therapy for PanCa. Currently, PTX nanoparticles (Abraxane®) along with gemcitabine are in use for the treatment of advanced PanCa. Herein, we have generated and characterized a novel nano-formulation of PTX comprised of PLGA core, stabilized with polyvinyl alcohol (PVA) and coated with poly-L-lysine (PLL). Dynamic light scattering (DLS) characterization indicated that the average particle size of this formulation was approximately 160 nm, and exhibited Zeta potentials of ~ -6.02 mV in 10% serum, along with an outstanding PL loading efficiency. We also evaluated the uptake mechanism of PPNPs in PanCa cells using different endocytosis inhibitors (Genistein, amiloride, nocodazole, chlorpromazine, and methyl beta cyclodextrin). Our results demonstrated that PPNPs were taken up mainly by lipid raft mediated endocytosis pathway in an energy-dependent manner. Next, we evaluated its therapeutic efficacy and underlying molecular mechanisms in human PanCa cells (HPAF-II, Panc1, and MiaPaCa-2) by performing various in vitro experiments. We first performed MTS and colony formation assays to determine the effects of free PTX and PPNPs on growth of PanCa cells. In this experiment, cells were treated with various concentrations of free PTX or abraxane, or PPNPs (2.5-160 nM) for 48 hrs. As a result, the PPNPs formulation showed more therapeutic effect over free abraxane as determined by decrease in cell proliferation and enhanced apoptosis induction in PanCa cells. PPNPs also showed a significant (P Citation Format: Bilal B. Hafeez, Andrew E. Massey, Vivek K. Kashyap, Mohammad Sikander, Advit Shetty, Mehdi Chaib, Hassan Mandil, Mohan Yallapu, Meena Jaggi, Subhash C. Chauhan. Novel nano-formulation of paclitaxel for pancreatic cancer therapy [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr LB-011.
- Published
- 2018
- Full Text
- View/download PDF
27. Abstract 679: ABI-231: A novel small molecule suppresses tumor growth and metastatic phenotypes of cervical cancer cells via targeting HPV E6 and E7
- Author
-
Subhash C. Chauhan, Meena Jaggi, Qinghai Wang, Neeraj Chauhan, Saini Setua, Aditya Ganju, Bilal Bin Hafeez, Murali M. Yallapu, Vivek K. Kashyap, Duane D. Miller, Sonam Kumari, Shabnam Malik, Nirnoy Dan, Prashanth K.B. Nagesh, and Wei Li
- Subjects
Cervical cancer ,Cancer Research ,Vinca ,biology ,business.industry ,Cell growth ,Cancer ,medicine.disease_cause ,biology.organism_classification ,medicine.disease ,In vitro ,chemistry.chemical_compound ,Oncology ,Paclitaxel ,chemistry ,In vivo ,medicine ,Cancer research ,Carcinogenesis ,business - Abstract
Objective: Cervical cancer is one of the most common and deadly cancers among women worldwide and is associated with persistent Human Papillomavirus (HPV) infection. Human papilloma virus (HPV) expressing E6 and E7 oncoproteins involved in carcinogenesis through their interactions with the p53 and pRB pathways, respectively. Therefore, non-toxic agents that have potential to inhibit the expression of E6 and E7 oncoproteins and their regulated oncogenic signaling pathways could be used in the management of cervical cancer. Microtubule targeting agents including paclitaxel, colchicine and vinca alkaloids are widely used in the treatment of various cancers but most of these agents have toxic side effects and develop chemoresistance. Herein, we investigated the potential anti-cancer effects of a novel tubulin targeting agent (ABI-231) in in vitro and in vivo model systems. Method: ABI-231 ((2-(1H-indol-3-yl)-1H-imidazol-4-yl)(3,4,5-trimethoxyphenyl))- methanone was synthesized in our home institution. Human cervical cancer cells (CaSki and SiHa) were used as in vitro model system. To determine the effect of ABI-231 on cell proliferation, migration and invasion, we performed MTS and wound healing assays. Effect of ABI-231 on the expression of HPV E6/E7 was determined by Western blot, qRT-PCR, and confocal microscopy. Xenograft study was performed to determine the effect of ABI-231 on cervical tumor growth. Results: ABI-231 treatment significantly (P Conclusions: Taken together, our results demonstrate the potential anti-cancer efficacy of ABI-231 in in vivo and in vivo. ABI-231 can be explored as a potent therapeutic agent for the treatment of cervical cancer. Citation Format: Vivek K. Kashyap, Bilal B. Hafeez, Qinghai Wang, Neeraj Chauhan, Prashanth K. Nagesh, Nirnoy Dan, sonam kumari, Shabnam Malik, Saini Setua, Aditya Ganju, Murali M. Yallapu, Duane D. Miller, Wei Li, Meena Jaggi, Subhash C. Chauhan C. Chauhan. ABI-231: A novel small molecule suppresses tumor growth and metastatic phenotypes of cervical cancer cells via targeting HPV E6 and E7 [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 679.
- Published
- 2018
- Full Text
- View/download PDF
28. Abstract 4657: Docetaxel nanoformulation reverts drug resistance in prostate cancer
- Author
-
Pallabita Chowdhury, Bilal Bin Hafeez, Elham Hatami, Prashanth Kumar Bhusetty Nagesh, Subhash C. Chauhan, Sheema Khan, Meena Jaggi, Vivek K. Kashyap, and Murali M. Yallapu
- Subjects
Cancer Research ,Chemistry ,Wild type ,Cancer ,Drug resistance ,medicine.disease ,In vitro ,Oncology ,Docetaxel ,Apoptosis ,Cell culture ,Cancer cell ,medicine ,Cancer research ,medicine.drug - Abstract
Objectives: Docetaxel (Dtx) is the primary choice for the treatment of castrate-resistant prostate cancer (PrCa). Besides its therapeutic benefits, many men with castrate-resistant PCa fail to respond to treatment due to Dtx resistance. There are multiple signaling mechanisms that play pivotal role in the development of Dtx resistance. Seminal reports stated that Tau, highly soluble microtubule-associated protein play a crucial role in Dtx resistance development. Disruption Tau signaling affects its chemoresistance. To revert the drug-resistance and target specific pathway, development of novel and strategic therapies are much needed. In this line, we employed magnetic nanoparticle (MNP) loaded Dtx (MNP-Dtx) to revert the chemo resistance of Dtx in PrCa. Methods: Docetaxel loaded magnetic nanoparticle (MNP-Dtx) formulation is composed of an iron oxide core coated with cyclodextrin (for drug loading) and F127 polymer (for particle stability and chemosensitization). Dtx-resistant PrCa cell line models (PC-3R and DU145R) were developed in our laboratory by repeatedly treating cancer cells in a dose escalation manner for over 24 months. Intracellular drug levels of MNP-Dtx in wild type and drug resistant cancer cells was determined by flow cytometry, TEM and HPLC methods. The superior in vitro anti-cancer activity of MNP-Dtx was examined by MTS, proliferation kinetics, colony formation, immunoblotting and qPCR studies in both wild type and drug resistant PrCa cell lines. The interaction between tau and tubulin studies were assessed through immunoprecipitation studies. Results: MNP-Dtx formulation showed optimal particle size and zeta potential which can efficiently be internalized in PrCa/Dtx resistant PrCa cells. MNP-Dtx exhibited enhanced cellular uptake in drug resistant cell lines over wild type cells in a dose and time dependent manner, with 2 to 3 folds increase. Immunoblot and q PCR studies showed the expression of Tau, whole β tubulin, β tubulin III, β tubulin IV and MDR1 were found to be deregulated in both wild type and resistant cells. To delineate the mechanism of chemosensitization by MNP-Dtx on resistant cells, we performed immunoprecipitation studies, found the expression of Tau was significantly reduced during MNP-Dtx treatments w.r.t control and Dtx. HPLC studies have shown higher amounts of Dtx levels were detected in cells after MNP-Dtx treatments, annotating the sustain drug release ability of MNP nanoformulation. Further, these results were confirmed through tubulin polymerization studies. Conclusion: Overall, sustained drug release of Dtx aided in high intracellular levels of drug, thus facilitating high probability of Dtx-tubulin interaction instead of Tau-tubulin interactions resulting in tubulin fibers stabilization. This promotes Dtx induced apoptosis in wild type and Dtx resistant PrCa cells. This novel therapeutic modality might be effective to treat patients with Dtx resistant PrCa. Citation Format: Prashanth K. B. Nagesh, Pallabita Chowdhury, Elham Hatami, Vivek K. Kashyap, Bilal B. Hafeez, Sheema Khan, Subhash C. Chauhan, Meena Jaggi, Murali M. Yallapu. Docetaxel nanoformulation reverts drug resistance in prostate cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 4657.
- Published
- 2018
- Full Text
- View/download PDF
29. Therapeutic efficacy of a novel βIII/βIV-tubulin inhibitor (VERU-111) in pancreatic cancer
- Author
-
Sonam Kumari, Bilal Bin Hafeez, Wei Li, Qinghui Wang, Miller Duane D, Subhash C. Chauhan, Andrew E. Massey, Murali M. Yallapu, Vivek K. Kashyap, Saini Setua, and Meena Jaggi
- Subjects
Cancer Research ,Tubulin ,Oncology ,biology ,Panca ,business.industry ,Pancreatic cancer ,medicine ,Cancer research ,biology.protein ,medicine.disease ,biology.organism_classification ,business - Abstract
e14578Background: The management of pancreatic cancer (PanCa) is exceptionally difficult due to poor response to available therapeutic approaches. Tubulins play a major role in cell dynamics and ar...
- Published
- 2018
- Full Text
- View/download PDF
30. Abstract 4399: MUC13 induced NFκB activation regulates metabolic reprograming by promoting its crosstalk with GLUT-1 receptor
- Author
-
Sonam Kumari, Subash C. Gupta, Meena Jaggi, Vivek K. Kashyap, Subhash C. Chauhan, Murali M. Yallapu, and Sheema Khan
- Subjects
Cancer Research ,Crosstalk (biology) ,Oncology ,Chemistry ,Receptor ,Cell biology - Abstract
Objective: Pancreatic cancer (PanCa) is the fourth most common cause of cancer-related deaths in the US. MUC13, mucin is aberrantly expressed in PanCa and promotes tumor growth and progression. Herein, we investigate the fundamental role of MUC13 in glucose metabolism and delineate the molecular interplay of various molecules governing MUC13 mediated metabolic reprograming that may be involved in pancreatic tumor maintenance. Methods: MUC13 expressing (Panc-1) and knockdown PanCa cells (HPAF-II) were generated for the study. Immunoblotting and qRT-PCR assays were performed to assess the expression of protein and mRNA levels, respectively, of key signaling molecules involved in glucose metabolism of PanCa. MUC13 and Glut-1 interaction was studied using reciprocal co-immunoprecipitation, immunofluorescence, proximity ligation, Western blotting, co-capping assays in cell lines. Lactate and glucose assays were performed using commercially available kits. In vitro functional assays using wound healing scratch assay (migration), and cell Matrigel assay (invasion) were performed in presence or absence of Lactate and 2DG supplementation. Results: Our results demonstrate that MUC13 expression leads to the TNF-induced activation/nuclear translocation of NFκB p-65 and phosphorylation of IkB which in turn upregulates additional key proteins, Glut-1, c-Myc, Bcl-2. This recruits the Glut-1 to MUC13, wherein MUC13 functionally interacts with Glut-1 and stabilizes it, initiating downstream events that result in altered glucose metabolism. MUC13 expression in PanCa cells increases glucose uptake, lactate secretion which is reduced upon MUC13 knockdown. Additionally, MUC13 mediates increased cell migratory and invasion potential which can be potentiated by supplementing the culture media with lactate, an end product of aerobic glycolysis. However, treatment of cells with NFκB inhibitor, Sulfasalazine, inhibits the MUC13 and Glut-1 interaction and abrogates all these events associated with glucose metabolism. Conclusion: These outcomes from our study suggest that MUC13 plays an important role in metabolic reprogramming of PanCa cells metabolism to induce cancer growth and enhanced cellular invasion and motility. NFκB acts downstream of MUC13 to coordinate the events leading to its interation with Glut-1 and metabolic reprogramming. Overall, these findings illustrate mechanisms by which MUC13 coordinates the shift in metabolism to sustain cancer growth and invasion in PanCa. Citation Format: Sonam Kumari, Sheema Khan, Subash C. Gupta, Vivek K. Kashyap, Murali M. Yallapu, Subhash C. Chauhan, Meena Jaggi. MUC13 induced NFκB activation regulates metabolic reprograming by promoting its crosstalk with GLUT-1 receptor [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 4399. doi:10.1158/1538-7445.AM2017-4399
- Published
- 2017
- Full Text
- View/download PDF
31. Abstract 1206: miR-205 replenishment in prostate cancer cells: A novel nanoparticle approach
- Author
-
Bilal Bin Hafeez, Vivek K. Kashyap, Vijayakumar N. Boya, Sheema Khan, Pallabita Chowdhury, Murali M. Yallapu, Prashanth K.B. Nagesh, Nadeem Zafar, Subhash C. Chauhan, Stephen W. Behrman, and Meena Jaggi
- Subjects
0301 basic medicine ,Cancer Research ,business.industry ,Nanoparticle ,medicine.disease ,03 medical and health sciences ,Prostate cancer ,030104 developmental biology ,0302 clinical medicine ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,medicine ,business - Abstract
Introduction Prostate cancer (PrCa) is the most common male malignancy among men in the United States. Recent studies suggest that low expression of miR-205 is seen in PrCa cell lines and tumors in comparison to normal prostatic epithelial cells. A numer of studies have shown that restoration of miR-205 in PrCa cells resulted in supression of cell growth, epithelial-to-mesenchymal transition, and chemosensitization. However, due to the poor pharmacological kinetics and low in vivo stability of miR-205, serious limitations are being experienced at the clinical level. Therefore, we have chosen a novel nanoparticle-based approach to deliver miR-205, for improved therapeutic benefits in PrCa. Methods A novel miR-205 nanoparticle formulation (named miR-MPG) was generated which is composed of an iron oxide core layered with polyethylene imine (PEI), and NHS-PEG-NHS (PEG) polymer. The miR-205 withholding and release characteristic of miR-PEG were examined through fluorescence quenching and agarose gel electrophoresis. Hemocompatibility of this formulation was examined using a hemolysis assay. Cellular uptake of miR-MPG formulation was evaluated using flow cytometry and confocal studies. Further, therapeutic and chemosensitization activity of miR-205 were assessed using cell-culture based assays. Molecular effects associated with the PrCa cells growth inhibiton were evaluated through protein profiling and qRT-PCR analyses. Results and Dissusion miR-MPG formulation exhibited optimal particle size and zeta potential, which are suitable for cancer therapeutics. Agarose gel electrophoresis binding studies suggested 5 µg of nanoparticle formulation is optimum to hold 1 µg of miR-205 mimic. Release of miR-205 from miR-MPG was determined with respect to concentration of anionic molecules and in a time-dependent manner. We observed no hemolysis during miR-MPG interaction with the red blood cells indicating its hemocompatibility. In addition, miR-MPG particles exhibited superior internalization and endosmal escape in PrCa cells. This formulation displayed enhanced sensitization of PrCa cells to docetaxel. Additionally, it induced the expression of apoptotic proteins (Bax, Bim, cleaved PARP, and caspase 3), and downregulated the anti-apoptotic proteins (Bcl-2 and survivin). Moreover, the expression of the chemoresistance-associated proteins (PSMA and MDR1) was profoundly inhibited in cells treated with miR-MPG in the presence of docetaxel. Further dataset of qRT-PCR studies showed induced expression of the miR-205 and affected the expression of its downstream genes. These results suggest that miR-MPG formulation may serve as an ideal delivery vehicle to deliver miR-205. Conclusion Results from this study suggests that successful delivery of miR-205 through miR-MPG nanoparticles can induce sensitization potential for docetaxel treatment. This novel therapeutic modality might be effective for PrCa patients undergoing chemotherapy. Citation Format: Prashanth K. Nagesh, Pallabita Chowdhury, Vijayakumar N. Boya, Vivek K. Kashyap, Sheema Khan, Bilal B. Hafeez, Nadeem Zafar, Stephen W. Behrman, Subhash C. Chauhan, Meena Jaggi, Murali M. Yallapu. miR-205 replenishment in prostate cancer cells: A novel nanoparticle approach [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 1206. doi:10.1158/1538-7445.AM2017-1206
- Published
- 2017
- Full Text
- View/download PDF
32. Abstract 2101: Ormeloxifene suppresses the growth of prostate tumor via inhibition of β-catenin induced AR signaling
- Author
-
Subhash C. Chauhan, Aditya Ganju, Bilal Bin Hafeez, Murali M. Yallapu, Vivek K. Kashyap, Mohammed Sikander, and Meena Jaggi
- Subjects
Cancer Research ,Activator (genetics) ,Chemistry ,Cell ,Cancer ,urologic and male genital diseases ,medicine.disease ,Prostate cancer ,Transactivation ,medicine.anatomical_structure ,Oncology ,Catenin ,medicine ,Cancer research ,Luciferase ,Ormeloxifene ,medicine.drug - Abstract
Background: Prostate cancer (PrCa) first manifests as an androgen-dependent disease and can be treated with androgen-deprivation therapy. Despite the initial success of androgen ablation therapy, resistance to anti-androgen therapy displays by progression to hormone refractory (androgen-independent) advanced stage PrCa which is primary cause of patient’s death. Main underlying cause for the onset of hormone refractory cancer is ligand independent activation of AR signaling in PrCa cells. It has been shown that β-catenin acts as a non-androgen activator of AR which enhances AR transactivation in PrCa cells. Thus, identification of agents with excellent pharmacokinetics and pharmacodynamics parameters that can inhibit ligand independent activation of AR signaling might be highly useful for the treatment of advanced stage PrCa. Herein, we identified a synthetic molecule, ormeloxifene (ORM), which efficiently represses β-catenin mediated ligand independent activation of AR signaling, thus, inhibits growth and metastatic features of PrCa cells. Methods: Androgen-refractory but AR positive PrCa cell (C4-2) was used as an in vitro and in vivo model systems. Effect of ORM on AR and PSA protein levels was determined by Western blot analysis. Effect of ORM treatment was analyzed on AR and PSA luciferase activities by transiently transfecting the C4-2 cells by AR and PSA luciferase plasmids. Renilla construct was used as an internal control. C4-2 cells nuclear and cytoplasmic lysates were prepared using Active Motif kit. Immunoprecipitation analysis was performed to determine if ORM inhibits physical interaction of β-catenin with AR. Therapeutic efficacy of ORM was evaluated in cell lines and PrCa xenograft mouse models. Results: ORM dose-dependently (10, 15 and 20 µM) inhibited the protein levels of AR and its downstream target protein PSA. ORM (10 and 20μM) treatment also inhibited AR transactivation as determined by decreased promoter activities of AR and its target gene PSA. ORM (10 and 20μM) treatment inhibited protein levels of nuclear β-catenin and physical interaction of β-catenin with AR in PrCa cells. ORM administration dose- dependently (intra-peritoneal; 100 and/or 500µg/mouse; thrice/week) significantly (P Conclusion: Our study demonstrates that ORM is a potent inhibitor of β-catenin-mediated activation of AR signaling. Based on its safety profile, ORM might be an ideal candidate for repurposing to treat advanced stage PrCa alone or in combination with other therapies. Citation Format: Aditya Ganju, Bilal Bin Hafeez, Mohammed Sikander, Vivek Kumar Kashyap, Murali Mohan Yallapu, Subhash C. Chauhan, Meena Jaggi. Ormeloxifene suppresses the growth of prostate tumor via inhibition of β-catenin induced AR signaling [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 2101. doi:10.1158/1538-7445.AM2017-2101
- Published
- 2017
- Full Text
- View/download PDF
33. Abstract 3216: Attenuation of pancreatic tumor growth by a small molecule tubulin inhibitor
- Author
-
Wei Li, Saini Setua, Vivek K. Kashyap, Andrew E. Massey, Subhash C. Chauhan, Duane D. Miller, Aditya Ganju, Qinghui Wang, Murali M. Yallapu, Bilal Bin Hafeez, and Meena Jaggi
- Subjects
Cancer Research ,biology ,Cell growth ,business.industry ,Panca ,Cancer ,medicine.disease ,biology.organism_classification ,Vinblastine ,chemistry.chemical_compound ,Oncology ,Docetaxel ,Paclitaxel ,chemistry ,Pancreatic cancer ,Cancer cell ,medicine ,Cancer research ,business ,medicine.drug - Abstract
Introduction: Pancreatic cancer (PanCa) is one of the most fatal cancers and is ranked as the fourth common cause of cancer-related deaths among both men and women in the US. The management of PanCa is exceptionally difficult due to the extremely poor response to available chemotherapeutic drugs. Microtubules are dynamic structures composed of α-β-tubulin heterodimers that are essential in cell division and are important targets for several clinical drugs (paclitaxel, docetaxel and vinblastine). However, clinical use of these tubulin-targeting drugs have toxicity and drug resistance issues in cancer patients. Thus, identification of more potent non-toxic inhibitors of β-tubulin is urgently required for cancer therapy purposes. In this study, we have identified a synthetic compound (ABI-231) which is a potent inhibitor of β-tubulin and evaluated its therapeutic efficacy against PanCa in vitro, and in vivo model systems. Methods: ABI-231 ((2-(1H-indol-3-yl)-1H-imidazol-4-yl) (3, 4, 5-trimethoxyphenyl)) - methanone was synthesized and characterized in our department. Effect of ABI-231 on proliferation, migration and invasion of human PanCa cells (ASPC1, HPAFII, and PANC1) was performed by in vitro functional assays (MTS, wound healing, and Boyden chamber migrations). Effect of ABI-231 on the expression of β-tubulin isoforms was determined and compared with other clinical inhibitors of β-tubulin by Western blot, and qRT-PCR. Moreover, the effect of ABI-231 on the expression of β-tubulin III in PanCa cells was determined by confocal microscopy. Therapeutic efficacy of ABI-231 against PanCa was evaluated in an ectopic xenograft mouse model. Results: ABI-231 treatment inhibited cell proliferation, invasion, migration and colony formation abilities of PanCa cells in a dose-dependent manner (1-100 nM) compared to vehicle treated group. Aberrant expression of β-tubulin III is involved in aggressiveness and drug resistance of various type of cancers including PanCa. ABI-231 effectively inhibited the protein levels and mRNA expression of total β-tubulin (TBB), TBB1, TBB2c, TBB3 and TBB4 in PanCa cells via destabilization. Our confocal microscopy results further showed inhibition of β-tubulin in ABI-231 treated PanCa cells. Upregulation of micro RNA 200c (miR-200c) has been shown to inhibit the expression of β-tubulin III in cancer cells. ABI-231 treatment of PanCa cells showed significant (p Conclusion: Taken together, our results suggest that ABI-231 is a potent β-tubulin inhibitor and chemotherapeutic agent which could be used for the treatment of pancreatic cancer. Citation Format: Vivek K. Kashyap, Bilal B. Hafeez, Qinghui Wang, Saini Setua, Andrew Massey, Aditya Ganju, Murali M. Yallapu, Duane D. Miller, Wei Li, Meena Jaggi, Subhash C. Chauhan. Attenuation of pancreatic tumor growth by a small molecule tubulin inhibitor [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 3216. doi:10.1158/1538-7445.AM2017-3216
- Published
- 2017
- Full Text
- View/download PDF
34. Abstract IA30: Comorbidity factors associated with human papillomavirus infectivity: Implications in cervical cancer health disparity
- Author
-
Namita Sinha, Vivek K. Kashyap, Subhash C. Chauhan, Samtosh K. Kumar, Murali M. Yallapu, Meena Jaggi, Sheema Khan, Diane M. Maher, Mohammad Sikander, and Nadeem Zafar
- Subjects
Infectivity ,Cervical cancer ,Epidemiology ,Cell growth ,business.industry ,Cancer ,Cell cycle ,medicine.disease ,Phenotype ,Oncology ,Apoptosis ,Immunology ,Cancer research ,medicine ,business ,Carcinogen - Abstract
Objective: High-risk strains of human papillomavirus (HPV), HPV E6/E7 cause cervical cancer (CxCa). Certain underserved populations in the United States, such as American Indian and African American women disproportionately suffer from CxCa compared to their Caucasian counter parts. However, precise etiology and comorbidity factors associated with CxCa health disparity are not fully uncovered. Understanding of these factors at molecular level will entail developing novel strategies to reduce this health disparity. In this study, we have investigated the molecular interplay existing between various comorbidity factors, namely, smoking, alcohol and HIV co-infection on the HPV infectivity which are primarily known for the progression of CxCa. Method: In order to define a molecular association of smoking, alcohol and HIV co-infection with CxCa, Caski and SiHa (HPV infected cervical cancer cells) cells were treated with a smoking carcinogens Benzo[a]Pyrene (BaP) or alcohol (EthOH) or both for different time periods. Effects of these treatment was analyzed on cell proliferation, clonogenicity, cell migration, cell cycle and the expression of HPV E6/E7 was determined by qRT-PCR, immunoblotting and confocal microscopy. The effect of HIV co-infection on the expression of HPV E6/E7 was also investigated by incubating CxCa cells with conditioned medium derived from HIV infected U937 monocytic cells (U1). Additionally, we examined effect of these cofactors on the expression enzymes associated with cellular oxidative stress using immunoblotting and confocal microscopy analyses. Results: Our results show that the exposure of BaP or EthOH or their combination enhances the expression of HPV E6/E7 oncogenes. Additionally, cells treated with BaP and EthOH alone or in combination show higher oncogenic phenotypes as evident by increased cell proliferation, clonogenicity and cell migration and invasion. These cofactors in presence of HIV co-infection also augment the expression of HPVE6/E7 oncogenes. Exposure of these cofactors alter cellular oxidative stress via modulation of the expression of PRDX6 enzyme. Interestingly, curcumin and its nanoparticle formulation (Nano-Cur) effectively inhibit BaP/EthOH induced expression of E6/E7 oncogenes, tumorigenic characteristics of CxCa cells and induce apoptosis. Conclusions: The study suggests a molecular link between smoking, alcohol and HIV infection with HPV infectivity and their potential association with CxCa health disparity. These events however, can be effectively attenuated by curcumin/nano-curcumin treatment, implying its role in CxCa prevention/treatment. This provides hope for developing a feasible approach to effectively reduce CxCa health disparity among underserved populations. Citation Format: Vivek K. Kashyap, Sheema Khan, Mohammad Sikander, Diane M. Maher, Samtosh K. Kumar, Namita Sinha, Murali M. Yallapu, Nadeem Zafar, Meena Jaggi, Subhash C. Chauhan. Comorbidity factors associated with human papillomavirus infectivity: Implications in cervical cancer health disparity. [abstract]. In: Proceedings of the Ninth AACR Conference on the Science of Cancer Health Disparities in Racial/Ethnic Minorities and the Medically Underserved; 2016 Sep 25-28; Fort Lauderdale, FL. Philadelphia (PA): AACR; Cancer Epidemiol Biomarkers Prev 2017;26(2 Suppl):Abstract nr IA30.
- Published
- 2017
- Full Text
- View/download PDF
35. Abstract 2208: Novel nanoparticle formulation of Plumbagin for pancreatic cancer treatment
- Author
-
Aditya Ganju, Subhash C. Chauhan, Murali M. Yallapu, Vijayakumar N. Boya, Bilal Bin Hafeez, Vivek K. Kashyap, Mohammad Sikander, and Meena Jaggi
- Subjects
Cancer Research ,Stromal cell ,biology ,Panca ,Chemistry ,Plumbagin ,biology.organism_classification ,medicine.disease ,Bioavailability ,chemistry.chemical_compound ,Oncology ,Pancreatic cancer ,Toxicity ,Drug delivery ,medicine ,Cancer research ,IC50 - Abstract
Pancreatic cancer (PanCa) is one of the most fatal of all cancers and is ranked as the fourth most common cause of cancer related deaths among both men and women in the US. The management of PanCa, is exceptionally difficult due to the extremely poor response to available therapeutic modalities. Highly desmoplastic microenvironment in pancreatic tumor causes suboptimal drug delivery and increases chemo-resistance. Plumbagin (PL), a naturally occurring napthoquinone derived from the root of Plumbago zeylanica L., has showed potent cancer preventive and therapeutic activity against variety of cancers. However, the clinical translation of PL has been significantly hampered due to its toxicity and suboptimal bioavailability. To address these clinically relevant issues, we have developed and characterized a novel PL-loaded magnetic nanoparticle (MNP-PL) formulation. This MNP-PL formulation was prepared using Magnetic nanoparticles (MNPs) composed of an iron oxide core which has distinct advantages in i) bio/hemo-compatibility, ii) biodegradation, iii) higher drug loading capacity and iv) improved bioavailability. Our novel MNP-PL formulation provided average size of 125 nm in dynamic light scattering (DLS) and exhibited -9.42 to -10.79 mV zeta potential with an outstanding PL loading efficiency. We have evaluated anti-cancer potential of MNP-PL formulation in human PanCa cells (HPAF-II, AsPc1 and Panc-1). We first performed MTS and colony formation assays to determine the effects of free PL and MNP-PL formulation on growth of PanCa cells. In this experiment, cells were treated with various concentrations of free PL (1-15 μM) or MNP-PL (1-15 μM) for 24 hrs. Results exhibited efficient internalization of the MNP-PL formulation in a dose-dependent manner. As a result, the MNP-PL formulation showed four fold dose advantage over free PL. IC50 of free PL was recorded 10 μM which was significantly reduced to 2.5 μM in MNP-PL. MNP-PL also showed four fold inhibition in colony formation compared to free PL. MNP-PL treatment more efficiently inhibited oncogenic CXCL12/CXCR4 signaling pathway in both PanCa and patient derived stromal fibroblast cells. MNP-PL treatment also showed decreased expression of CXCR4 protein levels in PanCa cells. Moreover, MNP-PL treatment inhibited stromal derived factor 1 (SDF-1)/CXCL12 expression in stromal fibroblasts. These results suggest that our novel MNP-PL formulation has more anti-cancer potential than free PL against PanCa. In conclusion, MNP-PL formulation may reduce the toxicity and improve the bioavailability of free PL and could be used for the treatment of PanCa. Citation Format: Bilal B. Hafeez, Vivek K. Kashyap, Vijayakumar N. Boya, Aditya Ganju, Mohammad Sikander, Murali M. Yallapu, Meena Jaggi, Subhash C. Chauhan. Novel nanoparticle formulation of Plumbagin for pancreatic cancer treatment. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 2208.
- Published
- 2016
- Full Text
- View/download PDF
36. Abstract 1745: Comorbidity factors associated with human papillomavirus infectivity: Implications in cervical cancer health disparity
- Author
-
Nadeem Zafar, Mohammad Sikander, Diane M. Maher, Sheema Khan, Vivek K. Kashyap, Santosh Kumar, Namita Sinha, Subhash C. Chauhan, Meena Jaggi, and Murali M. Yallapu
- Subjects
Cervical cancer ,Infectivity ,Cancer Research ,business.industry ,Cell growth ,Cancer ,Cell migration ,Cell cycle ,medicine.disease ,Virology ,Oncology ,Apoptosis ,Cancer research ,Medicine ,business ,Carcinogen - Abstract
Objective: High-risk strains of human papillomavirus (HPV), HPV E6/E7 cause cervical cancer (CxCa). Certain underserved populations in the United States, such as American Indian and African American women disproportionately suffer from CxCa compared to their Caucasian counter parts. However, precise etiology and comorbidity factors associated with CxCa health disparity are not fully uncovered. Understanding of these factors at molecular level will entail developing novel strategies to reduce this health disparity. In this study, we have investigated the molecular interplay existing between various comorbidity factors, namely, smoking, alcohol and HIV co-infection on the HPV infectivity which are primarily known for the progression of CxCa. Method: In order to define a molecular association of smoking, alcohol and HIV co-infection with CxCa, Caski and SiHa (HPV infected cervical cancer cells) cells were treated with a smoking carcinogens Benzo[a]Pyrene (BaP) or alcohol (EthOH) or both for different time periods. Effects of these treatment was analyzed on cell proliferation, clonogenicity, cell migration, cell cycle and the expression of HPV E6/E7 was determined by qRT-PCR, immunoblotting and confocal microscopy. The effect of HIV co-infection on the expression of HPV E6/E7 was also investigated by incubating CxCa cells with conditioned medium derived from HIV infected U937 monocytic cells (U1). Additionally, we examined effect of these cofactors on the expression enzymes associated with cellular oxidative stress using immunoblotting and confocal microscopy analyses. Result: Our results show that the exposure of BaP or EthOH or their combination enhances the expression of HPV E6/E7 oncogenes. Additionally, cells treated with BaP and EthOH alone or in combination show higher oncogenic phenotypes as evident by increased cell proliferation, clonogenicity and cell migration andinvasion. These cofactors in presence of HIV co-infection also augment the expression of HPVE6/E7 oncogenes. Exposure of these cofactors alter cellular oxidative stress via modulation of the expression of PRDX6 enzyme. Interestingly, curcumin and its nanoparticle formulation (Nano-Cur) effectively inhibits BaP/EthOH induced expression of E6/E7 oncogenes, growth, migration of CxCa cells and induces apoptosis. Conclusions: The study suggests a molecular link between smoking, alcohol and HIV infection with HPV infectivity and their potential association with CxCa health disparity. These events however, can be effectively attenuated by curcumin/nano-curcumin treatment, implying its role in CxCa prevention/treatment. This provides hope for developing a feasible approach to effectively reduce CxCa health disparity among underserved populations. Citation Format: Vivek K. Kashyap, Sheema Khan, Mohammad Sikander, Diane M. Maher, Santosh Kumar, Namita Sinha, Murali M. Yallapu, Nadeem Zafar, Meena Jaggi, Subhash C. Chauhan. Comorbidity factors associated with human papillomavirus infectivity: Implications in cervical cancer health disparity. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 1745.
- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.