26 results on '"Mahadeo Gorain"'
Search Results
2. Antiproliferative and apoptotic potential of methotrexate lipid nanoparticles in a murine breast cancer model
- Author
-
Chaitali Surve, Ananya Banerjee, Anupriya S, Rajasree Chakraborty, Dhiraj Kumar, Ramesh Butti, Mahadeo Gorain, Sabyasachi Parida, Gopal C Kundu, Supriya Shidhaye, and Srinivas Patnaik
- Subjects
Cell Survival ,Biomedical Engineering ,Medicine (miscellaneous) ,Breast Neoplasms ,Bioengineering ,Development ,Mice ,Methotrexate ,Liposomes ,Animals ,Humans ,Nanoparticles ,Female ,General Materials Science - Abstract
Aim: To evaluate the efficacy of novel methotrexate-loaded nanoparticles (MTX-NPs) in vitro and in vivo in the treatment of breast cancer. Materials & methods: MTX-NPs were tested for cellular uptake, cell viability, cell cycle, cellular wound migration and changes in tumor volume using characterized NPs. Results: The solid lipid NPs (SLNPs) showed strong cellular uptake, increased apoptosis, controlled cytotoxicity at lower IC50 of methotrexate and a sizable reduction in tumor burden. Conclusion: MTX-NP oral formulation can be a promising candidate in breast cancer treatment with improved cellular uptake and in vivo efficacy.
- Published
- 2022
- Full Text
- View/download PDF
3. Identification of a physiologic vasculogenic fibroblast state to achieve tissue repair
- Author
-
Durba Pal, Subhadip Ghatak, Kanhaiya Singh, Ahmed Safwat Abouhashem, Manishekhar Kumar, Mohamed S El Masry, Sujit K. Mohanty, Ravichand Palakurti, Yashika Rustagi, Saba Tabasum, Dolly K. Khona, Savita Khanna, Sedat Kacar, Rajneesh Srivastava, Pramod Bhasme, Sumit S. Verma, Edward Hernandez, Anu Sharma, Diamond Reese, Priyanka Verma, Nandini Ghosh, Mahadeo Gorain, Jun Wan, Sheng Liu, Yunlong Liu, Natalia Higuita Castro, Surya C. Gnyawali, William Lawrence, Jordan Moore, Daniel Gallego Perez, Sashwati Roy, Mervin C. Yoder, and Chandan K. Sen
- Subjects
Multidisciplinary ,General Physics and Astronomy ,General Chemistry ,General Biochemistry, Genetics and Molecular Biology - Abstract
Tissue injury to skin diminishes miR-200b in dermal fibroblasts. Fibroblasts are widely reported to directly reprogram into endothelial-like cells and we hypothesized that miR-200b inhibition may cause such changes. We transfected human dermal fibroblasts with anti-miR-200b oligonucleotide, then using single cell RNA sequencing, identified emergence of a vasculogenic subset with a distinct fibroblast transcriptome and demonstrated blood vessel forming function in vivo. Anti-miR-200b delivery to murine injury sites likewise enhanced tissue perfusion, wound closure, and vasculogenic fibroblast contribution to perfused vessels in a FLI1 dependent manner. Vasculogenic fibroblast subset emergence was blunted in delayed healing wounds of diabetic animals but, topical tissue nanotransfection of a single anti-miR-200b oligonucleotide was sufficient to restore FLI1 expression, vasculogenic fibroblast emergence, tissue perfusion, and wound healing. Augmenting a physiologic tissue injury adaptive response mechanism that produces a vasculogenic fibroblast state change opens new avenues for therapeutic tissue vascularization of ischemic wounds.
- Published
- 2023
- Full Text
- View/download PDF
4. Ultrahigh Penetration and Retention of Graphene Quantum Dot Mesoporous Silica Nanohybrids for Image Guided Tumor Regression
- Author
-
Amit S. Yadav, Mahadeo Gorain, N. N. V. Radharani, João Conde, Nishant Kumar Jain, Gopal C. Kundu, Rajendra Prasad, Rohit Srivastava, and Manali Jadhav
- Subjects
Materials science ,Nanostructure ,Biomedical Engineering ,Nanotechnology ,02 engineering and technology ,010402 general chemistry ,01 natural sciences ,Biomaterials ,Light responsive ,Neoplasms ,Quantum Dots ,Tumor regression ,Humans ,Biochemistry (medical) ,General Chemistry ,Penetration (firestop) ,Photothermal therapy ,Mesoporous silica ,021001 nanoscience & nanotechnology ,Silicon Dioxide ,Graphene quantum dot ,0104 chemical sciences ,Nanomedicine ,Nanoparticles ,Graphite ,0210 nano-technology - Abstract
So far, near-infrared (NIR) light responsive nanostructures have been well-defined in cancer nanomedicine. However, poor penetration and retention in tumors are the limiting factors. Here, we report the ultrahigh penetration and retention of carbanosilica (graphene quantum dots, GQDs embedded mesoporous silica) in solid tumors. After NIR light exposure, quick (0.5 h) emission from the tumor area is observed that is further retained up to a week (tested up to 10 days) with a single dose administration of nanohybrids. Emissive and photothermally active GQDs and porous silica shell (about 31% drug loading) make carbanosilica a promising nanotheranostic agent exhibiting 68.75% tumor shrinking compared to without NIR light exposure (34.48%). Generated heat (∼52 °C) alters the permeability of tumor enhancing the accumulation of nanotheranostics into the tumor environment. Successive tumor imaging ensures the prolonged follow-up of image guided tumor regression due to synergistic therapeutic effect of nanohybrids.
- Published
- 2022
5. RGD functionalized chitosan nanoparticle mediated targeted delivery of raloxifene selectively suppresses angiogenesis and tumor growth in breast cancer
- Author
-
Thejus Baby, Mahadeo Gorain, Anuradha Bulbule, Gaurab Roy, Dattatrya Shetti, N Naga Venkata Radharani, Amit S. Yadav, and Gopal C. Kundu
- Subjects
Angiogenesis ,Breast Neoplasms ,02 engineering and technology ,03 medical and health sciences ,Drug Delivery Systems ,Breast cancer ,In vivo ,Cell Line, Tumor ,Tumor Microenvironment ,medicine ,Humans ,General Materials Science ,Raloxifene ,030304 developmental biology ,Chitosan ,0303 health sciences ,Tumor microenvironment ,Chemistry ,021001 nanoscience & nanotechnology ,medicine.disease ,In vitro ,Targeted drug delivery ,Cell culture ,Raloxifene Hydrochloride ,Cancer research ,Nanoparticles ,0210 nano-technology ,Oligopeptides ,medicine.drug - Abstract
Acidic pH is a crucial intrinsic property of the microenvironment of most solid tumors. Hence, the use of pH sensitive tumor targeting nanoparticles is an attractive approach to enhance the therapeutic efficacy of anti-cancer agents in solid tumors. Chitosan nanoparticles (CHNPs) have been widely explored in the area of cancer drug delivery; nevertheless their true potential as a pH responsive targeted drug delivery vehicle in cancer therapy has not been deciphered yet as most of the research is limited to pH dependent stability and drug release. In the present study, we investigate the direct effect of pH in synergy with RGD peptide based targeting on the therapeutic efficacy of chitosan nanoparticles (RGD-CHNPs) in breast cancer. Furthermore, for the first time we performed a comprehensive study showing the anti-tumor, anti-migratory and anti-angiogenic effect of raloxifene (Rlx) loaded CHNPs in breast cancer. We prepared stable formulations of raloxifene encapsulated CHNPs and RGD-CHNPs by the nontoxic ionic gelation method. pH dependent studies revealed that NPs possess higher stability and zeta potential along with enhanced cellular uptake at acidic pH (as present in solid tumors) compared to physiological pH. Furthermore, RGD conjugation enhanced the in vitro cellular uptake of CHNPs in αvβ3 integrin expressing breast cancer cells and induced higher cellular apoptosis in breast cancer cells which was further augmented by lower pH. Moreover, Rlx-RGD-CHNPs significantly inhibited breast cancer cell migration and angiogenesis. In vivo studies showed that Cy5.5 conjugated RGD-CHNPs can distinctly visualize tumors and Rlx-RGD-CHNPs significantly inhibit breast tumor growth without causing any toxic effect to normal tissue as confirmed by hematology and blood biochemical studies. Therefore, RGD-CHNPs could potentially enhance the therapeutic efficacy of chemotherapeutic drugs due to the synergistic effect of pH responsiveness and tumor specific targeting in breast cancer.
- Published
- 2020
- Full Text
- View/download PDF
6. Functional design of pH-responsive folate-targeted polymer-coated gold nanoparticles for drug delivery and in vivo therapy in breast cancer
- Author
-
Mahadeo Gorain, Suresh W. Gosavi, Ranveig Braathen, Amit S. Yadav, Bjarne Bogen, Sneha Mahalunkar, Gopal C. Kundu, Siegfried Weiss, and Vinay Pawar
- Subjects
Organic Chemistry ,Biophysics ,Pharmaceutical Science ,Bioengineering ,02 engineering and technology ,General Medicine ,010402 general chemistry ,021001 nanoscience & nanotechnology ,01 natural sciences ,In vitro ,0104 chemical sciences ,Bioavailability ,Biomaterials ,chemistry.chemical_compound ,chemistry ,In vivo ,Cell culture ,Colloidal gold ,mental disorders ,Drug Discovery ,Drug delivery ,Curcumin ,Cancer research ,0210 nano-technology ,Cytotoxicity - Abstract
Background Curcumin has been widely used owing to its various medicinal properties including antitumor effects. However, its clinical application is limited by its instability, poor solubility and low bioavailability. Folic acid (FA)-functionalized nanoformulations may enhance the sustained release of an anticancer drug (curcumin) by tumor-specific targeting to improve therapeutic benefit. This study aims to design a nanoconjugate (NC) comprised of folate-curcumin-loaded gold-polyvinylpyrrolidone nanoparticles (FA-CurAu-PVP NPs) for targeted delivery in breast cancer model systems. Methods We developed curcumin-loaded FA-functionalized Au-PVP NCs by layer-by-layer assembly. The folic acid-curcumin Au-PVP NCs (FA-CurAu-PVP NCs) were characterized by ultraviolet-visible spectra, Fourier transform infrared spectroscopy, X-ray powder diffraction and thermogravimetric analysis. In vitro anticancer and antimigratory effects of NCs were examined by performing MTT and wound migration assays. The in vivo antitumor efficacy of NCs was investigated using a preclinical breast cancer orthotopic mouse model. Results Curcumin (40 µg/mL) was loaded along with conjugation of folate onto Au-PVP NPs to form FA-CurAu-PVP NCs. The size and charge of the NCs were increased gradually through layer-by-layer assembly and showed 80% release of curcumin at acidic pH. The NC did not show aggregation when incubated with human serum and mimicked an intrinsic peroxidase-like property in the presence of 3,3',5,5'-tetramethylbenzidine substrate. The MTT data using these NCs showed efficient anticancer activity at lower doses in estrogen/progesterone receptor (ER/PR)-negative cells compared with ER/PR-positive cells. Furthermore, the NCs did not show cytotoxicity at the investigated concentration in human breast epithelial and mouse fibroblast cell lines. They showed inhibitory effects on cell migration and high antitumor efficacy in in vivo analysis. Conclusion These results suggest that folate-based tumor targeting using CurAu-PVP NCs is a promising approach for tumor-specific therapy of breast cancer without harming normal cells.
- Published
- 2019
- Full Text
- View/download PDF
7. In Vivo Wound Healing Performance of Halloysite Clay and Gentamicin-Incorporated Cellulose Ether-PVA Electrospun Nanofiber Mats
- Author
-
Gopal C. Kundu, Manohar V. Badiger, Mahadeo Gorain, S. R. Inamdar, and Ashwini Wali
- Subjects
Biochemistry (medical) ,technology, industry, and agriculture ,Biomedical Engineering ,Ether ,Context (language use) ,General Chemistry ,engineering.material ,Halloysite ,Electrospinning ,Biomaterials ,chemistry.chemical_compound ,Gentamicin Sulfate ,Chemical engineering ,chemistry ,In vivo ,parasitic diseases ,engineering ,Cellulose ,Wound healing - Abstract
Wound healing is a dynamic and complex process that requires a suitable environment to enhance the rapid healing process. In this context, fabrications of nanofibrous materials with antibiotic and antibacterial properties are becoming extremely important. In this present work, we report on the fabrication and characterization of electro-spun cellulose ether-PVA nanofiber mats loaded with halloysite clay (HNT) and gentamicin sulfate (GS) for faster wound healing applications. The morphology of nanofiber mats was examined by SEM and TEM. The average diameter of the nanofiber mats were in the range of 325 ± 30 nm. The physicochemical characterizations were done by FT-IR and XRD, which reveal the presence of HNT and GS into the nanofibers. The incorporation of halloysite gave good mechanical strength to the nanofiber mats. Swelling studies indicated the hydrophilicity of the mats. In vitro studies revealed that HNTs are nontoxic to L929 fibroblast cells and also promote cell growth and proliferation. The antibacterial property of HNT was also studied. The slow release of GS from the nanofiber mats was observed for a period of 18 days. The in vivo wound healing studies on the wistar rats for 21 days revealed the wound healing faster within 2 weeks by the incorporation of HNT and GS into the nanofiber mats and hence these nanofiber mats show great potential in acute and chronic wound healing applications.
- Published
- 2019
- Full Text
- View/download PDF
8. Oncostatin M Improves Cutaneous Wound Re-Epithelialization and Is Deficient under Diabetic Conditions
- Author
-
Amitava Das, Mahadeo Gorain, Atul Rawat, Nirupam Biswas, Amit Kumar Madeshiya, Sanskruti P. Mahajan, Nandini Ghosh, Savita Khanna, Chandan K. Sen, and Sashwati Roy
- Subjects
Dermatology ,Oncostatin M ,Biochemistry ,Article ,Mice ,Re-Epithelialization ,Glycation ,Diabetes Mellitus ,Medicine ,Animals ,Keratinocyte migration ,STAT3 ,Molecular Biology ,Wound Healing ,integumentary system ,biology ,business.industry ,fungi ,Cell Biology ,Mice, Inbred C57BL ,medicine.anatomical_structure ,STAT protein ,biology.protein ,Cancer research ,Signal transduction ,Janus kinase ,business ,Keratinocyte - Abstract
Impaired re-epithelialization characterized by hyperkeratotic nonmigratory wound epithelium is a hallmark of nonhealing diabetic wounds. In chronic wounds, the copious release of oncostatin M (OSM) from wound macrophages is evident. OSM is a potent keratinocyte (KC) activator. This work sought to understand the signal transduction pathway responsible for wound re-epithelialization, the primary mechanism underlying wound closure. Daily topical treatment of full-thickness excisional wounds of C57BL/6 mice with recombinant murine OSM improved wound re-epithelialization and accelerated wound closure by bolstering KC proliferation and migration. OSM activated the Jak-signal transducer and activator of transcription pathway as manifested by signal transducer and activator of transcription 3 phosphorylation. Such signal transduction in the human KC induced TP63, the master regulator of KC function. Elevated TP63 induced ITGB1, a known effector of KC migration. In diabetic wounds, OSM was more abundant than the level in nondiabetic wounds. However, in diabetic wounds, OSM activity was compromised by glycation. Aminoguanidine, a deglycation agent, rescued the compromised KC migration caused by glycated OSM. Finally, topical application of recombinant OSM improved KC migration and accelerated wound closure in db/db mice. This work recognizes that despite its abundance at the wound site, OSM is inactivated by glycation, and topical delivery of exogenous OSM is likely to be productive in accelerating diabetic wound closure.
- Published
- 2020
9. Urolithin A augments angiogenic pathways in skeletal muscle by bolstering NAD+ and SIRT1
- Author
-
Mahadeo Gorain, Nirupam Biswas, Savita Khanna, Carly Polcyn, Nandini Ghosh, Chandan K. Sen, Kanhaiya Singh, Amitava Das, Surya Gnyawali, and Sashwati Roy
- Subjects
medicine.medical_specialty ,Multidisciplinary ,Chemistry ,Angiogenesis ,lcsh:R ,lcsh:Medicine ,Skeletal muscle ,Urolithin ,Vascular endothelial growth factor A ,Endocrinology ,medicine.anatomical_structure ,Downregulation and upregulation ,In vivo ,Internal medicine ,medicine ,lcsh:Q ,NAD+ kinase ,lcsh:Science ,C2C12 - Abstract
Urolithin A (UA) is a natural compound that is known to improve muscle function. In this work we sought to evaluate the effect of UA on muscle angiogenesis and identify the underlying molecular mechanisms. C57BL/6 mice were administered with UA (10 mg/body weight) for 12–16 weeks. ATP levels and NAD+ levels were measured using in vivo 31P NMR and HPLC, respectively. UA significantly increased ATP and NAD+ levels in mice skeletal muscle. Unbiased transcriptomics analysis followed by Ingenuity Pathway Analysis (IPA) revealed upregulation of angiogenic pathways upon UA supplementation in murine muscle. The expression of the differentially regulated genes were validated using quantitative real-time polymerase chain reaction (qRT-PCR) and immunohistochemistry (IHC). Angiogenic markers such as VEGFA and CDH5 which were blunted in skeletal muscles of 28 week old mice were found to be upregulated upon UA supplementation. Such augmentation of skeletal muscle vascularization was found to be bolstered via Silent information regulator 1 (SIRT1) and peroxisome proliferator-activated receptor-gamma coactivator-1-alpha (PGC-1α) pathway. Inhibition of SIRT1 by selisistat EX527 blunted UA-induced angiogenic markers in C2C12 cells. Thus this work provides maiden evidence demonstrating that UA supplementation bolsters skeletal muscle ATP and NAD+ levels causing upregulated angiogenic pathways via a SIRT1-PGC-1α pathway.
- Published
- 2020
- Full Text
- View/download PDF
10. Exosome-Mediated Crosstalk between Keratinocytes and Macrophages in Cutaneous Wound Healing
- Author
-
Lava Timsina, Puneet Khandelwal, Mohamed S. El Masry, Stephen C. Jacobson, Subhadip Ghatak, Amanda P. Siegel, Amitava Das, Mahadeo Gorain, David E. Clemmer, Xuyao Zeng, Sashwati Roy, Robert J. Lee, Brooke A. Brown, Xiaoju Zhou, Andrew G. Clark, Chandan K. Sen, Woran Song, Kanhaiya Singh, Yi Xuan, Mangilal Agarwal, Poornachander R. Guda, and Milos V. Novotny
- Subjects
Keratinocytes ,General Physics and Astronomy ,02 engineering and technology ,010402 general chemistry ,Exosomes ,01 natural sciences ,Exosome ,Article ,Mice ,microRNA ,Animals ,General Materials Science ,Skin ,Wound Healing ,Chemistry ,Vesicle ,Macrophages ,General Engineering ,021001 nanoscience & nanotechnology ,Microvesicles ,0104 chemical sciences ,Cell biology ,Crosstalk (biology) ,Cutaneous wound ,0210 nano-technology ,Wound healing - Abstract
Bidirectional cell-cell communication involving exosome-borne cargo such as miRNA, has emerged as a critical mechanism for wound healing. Unlike other shedding vesicles, exosomes selectively package miRNA by SUMOylation of heterogeneous nuclear ribonucleoproteinA2B1 (hnRNPA2B1). In this work, we elucidate the significance of exosome in keratinocyte-macrophage crosstalk following injury. Keratinocyte-derived exosomes were genetically labeled with GFP reporter (Exo(κ-GFP)) using tissue nanotransfection and were isolated from dorsal murine skin and wound-edge tissue by affinity selection using magnetic beads. Surface N-glycans of Exo(κ-GFP) were also characterized. Unlike skin exosome, wound-edge Exo(κ-GFP) demonstrated characteristic N-glycan ions with abundance of low base pair RNA and were selectively engulfed by wound-macrophages (ωmϕ) in granulation tissue. In vitro addition of wound-edge Exo(κ-GFP) to proinflammatory ωmϕ resulted in conversion to a proresolution phenotype. To selectively inhibit miRNA packaging within Exo(κ-GFP) in vivo, pH-responsive keratinocyte-targeted siRNA-hnRNPA2B1 functionalized lipid nanoparticles (TLNP(κ)) were designed with 94.3% encapsulation efficiency. Application of TLNP(κ/si-hnRNPA2B1) to murine dorsal wound-edge significantly inhibited expression of hnRNPA2B1 by 80% in epidermis compared to TLNP(κ/si-control) group. Although no significant difference in wound closure or re-epithelialization was observed, TLNP(κ/si-hnRNPA2B1) treated group showed significant increase in ωmϕ displaying proinflammatory markers in the granulation tissue at day 10 post-wounding compared to TLNP(κ/si-control) group. Furthermore, TLNP(κ/si-hnRNPA2B1) treated mice showed impaired barrier function with diminished expression of epithelial junctional proteins, lending credence to the notion that unresolved inflammation results in leaky skin. This work provides insight wherein Exo(κ-GFP) are recognized as a major contributor that regulates macrophage trafficking and epithelial barrier properties post-injury.
- Published
- 2020
11. Urolithin A augments angiogenic pathways in skeletal muscle by bolstering NAD
- Author
-
Nandini, Ghosh, Amitava, Das, Nirupam, Biswas, Surya, Gnyawali, Kanhaiya, Singh, Mahadeo, Gorain, Carly, Polcyn, Savita, Khanna, Sashwati, Roy, and Chandan K, Sen
- Subjects
Male ,Gene Expression Profiling ,Administration, Oral ,Neovascularization, Physiologic ,Reproducibility of Results ,NAD ,Senescence ,Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha ,Article ,Mice, Inbred C57BL ,Ageing ,Adenosine Triphosphate ,Sirtuin 1 ,Coumarins ,Animals ,Angiogenesis ,Muscle, Skeletal ,Transcriptomics - Abstract
Urolithin A (UA) is a natural compound that is known to improve muscle function. In this work we sought to evaluate the effect of UA on muscle angiogenesis and identify the underlying molecular mechanisms. C57BL/6 mice were administered with UA (10 mg/body weight) for 12–16 weeks. ATP levels and NAD+ levels were measured using in vivo 31P NMR and HPLC, respectively. UA significantly increased ATP and NAD+ levels in mice skeletal muscle. Unbiased transcriptomics analysis followed by Ingenuity Pathway Analysis (IPA) revealed upregulation of angiogenic pathways upon UA supplementation in murine muscle. The expression of the differentially regulated genes were validated using quantitative real-time polymerase chain reaction (qRT-PCR) and immunohistochemistry (IHC). Angiogenic markers such as VEGFA and CDH5 which were blunted in skeletal muscles of 28 week old mice were found to be upregulated upon UA supplementation. Such augmentation of skeletal muscle vascularization was found to be bolstered via Silent information regulator 1 (SIRT1) and peroxisome proliferator-activated receptor-gamma coactivator-1-alpha (PGC-1α) pathway. Inhibition of SIRT1 by selisistat EX527 blunted UA-induced angiogenic markers in C2C12 cells. Thus this work provides maiden evidence demonstrating that UA supplementation bolsters skeletal muscle ATP and NAD+ levels causing upregulated angiogenic pathways via a SIRT1-PGC-1α pathway.
- Published
- 2020
12. A Plasmonic Supramolecular Nanohybrid as a Contrast Agent for Site‐Selective Computed Tomography Imaging of Tumor
- Author
-
Anivind Kaur Bindra, Yanli Zhao, Sivaramapanicker Sreejith, Mui Hoon Nai, Mahadeo Gorain, Rohit Srivastava, Dongdong Wang, Chwee Teck Lim, Deblin Jana, Rijil Thomas, Sabu Thomas, Gopal C. Kundu, Rajendra Prasad, Abhimanyu Tharayil, School of Physical and Mathematical Sciences, and School of Electrical and Electronic Engineering
- Subjects
Mammary Carcinoma ,Materials science ,medicine.diagnostic_test ,media_common.quotation_subject ,Supramolecular chemistry ,Computed tomography ,Condensed Matter Physics ,Bioimaging ,Electronic, Optical and Magnetic Materials ,Biomaterials ,Mammary carcinoma ,Nuclear magnetic resonance ,Chemistry [Science] ,Electrochemistry ,medicine ,Site selective ,Contrast (vision) ,Plasmon ,media_common - Abstract
Design of organic–inorganic hybrids by anchoring of plasmonic materials such as gold nanoparticles (AuNPs) on self-assembled organic substrates is useful but challenging. Herein, in situ anchoring of plasmonic nanoparticles on the surface of a designed spherical assembly via Au-S bond formation is presented. First, a thiol tailed pyrene derivative (2) undergoes solvent dependent self-assembly, transforming into an organic spherical aggregate (2agg). The thiol (-SH) rich surface of the organic assembly allows cumulative anchoring of AuNPs on the surface to form an organic–inorganic hybrid (Au@2agg). Further coating of biocompatible polyethylene glycol (PEG) leads to the construction of the final multicomponent system (PEG-Au@2agg) exhibiting morphological and spectroscopic features. The potential of PEG-Au@2agg as a bioprobe and a contrast agent is investigated by X-ray computed tomography (CT) experiments in vivo. High X-ray attenuation of directly anchored AuNP clusters on the surface of this supramolecular nanohybrids enhances the X-ray CT contrast and allows tracing of site-selective accumulation in mouse 4T1 breast tumor. Thus, this approach of designing organic–inorganic nanohybrids paves the way for developing future intelligent multifunctional nanosystems capable of cancer detection and imaging. Agency for Science, Technology and Research (A*STAR) National Research Foundation (NRF) Submitted/Accepted version A.K.B. and Y.Z thank the Singapore Agency for Science, Technology and Research (A*STAR) AME IRG grant (No. A20E5c0081), and the Singapore National Research Foundation Investigatorship (No. NRF-NRFI2018-03). S.S. and C.T.L. thank iHealthtech-NUS for financial support. R.P. acknowledges IIT-Bombay for awarding Institute Post-Doctoral Fellowship. R.S. thanks the Department of Biotechnology National Bioscience Award, Govt. of India.
- Published
- 2021
- Full Text
- View/download PDF
13. A biodegradable fluorescent nanohybrid for photo-driven tumor diagnosis and tumor growth inhibition
- Author
-
Deepak S. Chauhan, Mahadeo Gorain, Jayesh R. Bellare, Janhavi Devrukhkar, Gopal C. Kundu, Mayur Temgire, Amit S. Yadav, Rohit Srivastava, Rajendra Prasad, and Barkha Singh
- Subjects
Biocompatibility ,Infrared Rays ,Nanoparticle ,02 engineering and technology ,QUANTUM DOTS ,010402 general chemistry ,CARBON DOTS ,01 natural sciences ,Mice ,GOLD NANORODS ,INORGANIC NANOPARTICLES ,Cell Line, Tumor ,MESOPOROUS SILICA NANOPARTICLES ,Animals ,Humans ,LIVING MICE ,General Materials Science ,DRUG-DELIVERY ,PHOTOTRIGGERED LOCAL-ANESTHESIA ,chemistry.chemical_classification ,Reactive oxygen species ,Liposome ,Neoplasms, Experimental ,Phototherapy ,Photothermal therapy ,021001 nanoscience & nanotechnology ,Fluorescence ,0104 chemical sciences ,chemistry ,Liposomes ,Cancer cell ,Biophysics ,PHOTOTHERMAL CANCER-THERAPY ,Tumor growth inhibition ,Female ,Reactive Oxygen Species ,0210 nano-technology ,ORGANIC NANOPARTICLES - Abstract
Specific targeting and phototriggered therapy in mouse model have recently emerged as the starting point of cancer theragnosis. Herein, we report a bioresponsive and degradable nanohybrid, a liposomal nanohybrid decorated with red emissive carbon dots, for localized tumor imaging and light-mediated tumor growth inhibition. Unsaturated carbon dots (C-dots) anchored to liposomes convert near-infrared (NIR) light into heat and also produce reactive oxygen species (ROS), demonstrating the capability of phototriggered cancer cell death and tumor regression. The photothermal and oxidative damage of breast tumor by the nonmetallic nanohybrid has also been demonstrated. Designed nanoparticles show excellent aqueous dispersibility, biocompatibility, light irradiated enhanced cellular uptake, release of reactive oxygen species, prolonged and specific tumor binding ability and good photothermal response (62 degrees C in 5 minutes). Safe and localized irradiation of 808 nm light demonstrates significant tumor growth inhibition and bioresponsive degradation of the fluorescent nanohybrid without affecting the surrounding healthy tissues.
- Published
- 2018
- Full Text
- View/download PDF
14. Green synthesis of selenium nanoparticles using Acinetobacter sp. SW30: optimization, characterization and its anticancer activity in breast cancer cells
- Author
-
Pinaki Banerjee, Utkarsha U. Shedbalkar, Gopal C. Kundu, Mahadeo Gorain, Richa Singh, Balu A. Chopade, and Sweety A. Wadhwani
- Subjects
0301 basic medicine ,Chemistry ,Organic Chemistry ,HEK 293 cells ,Biophysics ,Pharmaceutical Science ,Nanoparticle ,chemistry.chemical_element ,Bioengineering ,02 engineering and technology ,General Medicine ,021001 nanoscience & nanotechnology ,Suspension culture ,Microbiology ,Biomaterials ,03 medical and health sciences ,030104 developmental biology ,Average size ,Biochemistry ,Drug Discovery ,Nanorod ,Breast cancer cells ,Acinetobacter sp ,0210 nano-technology ,Selenium - Abstract
The aim of this study was to synthesize selenium nanoparticles (SeNPs) using cell suspension and total cell protein of Acinetobacter sp. SW30 and optimize its synthesis by studying the influence of physiological and physicochemical parameters. Also, we aimed to compare its anticancer activity with that of chemically synthesized SeNPs in breast cancer cells. Cell suspension of Acinetobacter sp. SW30 was exposed to various physiological and physicochemical conditions in the presence of sodium selenite to study their effects on the synthesis and morphology of SeNPs. Breast cancer cells (4T1, MCF-7) and noncancer cells (NIH/3T3, HEK293) were exposed to different concentrations of SeNPs. The 18 h grown culture with 2.7×109 cfu/mL could synthesize amorphous nanospheres of size 78 nm at 1.5 mM and crystalline nanorods at above 2.0 mM Na2SeO3 concentration. Polygonal-shaped SeNPs of average size 79 nm were obtained in the supernatant of 4 mg/mL of total cell protein of Acinetobacter sp. SW30. Chemical SeNPs showed more anticancer activity than SeNPs synthesized by Acinetobacter sp. SW30 (BSeNPs), but they were found to be toxic to noncancer cells also. However, BSeNPs were selective against breast cancer cells than chemical ones. Results suggest that BSeNPs are a good choice of selection as anticancer agents.
- Published
- 2017
- Full Text
- View/download PDF
15. Notch1-MAPK Signaling Axis Regulates CD133+ Cancer Stem Cell-Mediated Melanoma Growth and Angiogenesis
- Author
-
Mahadeo Gorain, Hirekodathakallu V. Thulasiram, Deepti Tomar, Harshal S. Patil, Dhiraj Kumar, Tushar Patil, Totakura V. S. Kumar, Gopal C. Kundu, N. N. V. Radharani, and Santosh Kumar
- Subjects
0301 basic medicine ,Angiogenesis ,Cellular differentiation ,Dermatology ,Biology ,Biochemistry ,03 medical and health sciences ,chemistry.chemical_compound ,Cancer stem cell ,medicine ,Epithelial–mesenchymal transition ,Protein kinase A ,neoplasms ,Molecular Biology ,Melanoma ,Cell Biology ,medicine.disease ,Cell biology ,Vascular endothelial growth factor ,030104 developmental biology ,chemistry ,embryonic structures ,cardiovascular system ,Cancer research ,biological phenomena, cell phenomena, and immunity ,Stem cell - Abstract
Functional characterization and understanding of the intricate signaling mechanisms in stem-like cells is crucial for the development of effective therapies in melanoma. We have studied whether melanoma cells are phenotypically distinct and hierarchically organized according to their tumorigenic nature. We report that melanoma-specific CD133+ cancer stem cells exhibit increased tumor-initiating potential, tumor-endothelial cell interaction, and lung metastasis. These cells are able to transdifferentiate into an endothelial-like phenotype when cultured under endothelial differentiation-promoting conditions. Mechanistically, Notch1 upregulates mitogen-activated protein kinase activation through CD133, which ultimately controls vascular endothelial growth factor and matrix metalloproteinase expression in CD133+ stem cells leading to melanoma growth, angiogenesis, and lung metastasis. Blockade or genetic ablation of Notch1 and mitogen-activated protein kinase pathways abolishes melanoma cell migration and angiogenesis. Chromatin immunoprecipitation and reporter assays revealed that Notch1 intracellular domain regulates CD133 expression at the transcriptional level. Andrographolide inhibits Notch1 intracellular domain expression, Notch1 intracellular domain-dependent CD133-mediated mitogen-activated protein kinase and activator protein-1 activation, and epithelial to mesenchymal-specific gene expression, ultimately attenuating melanoma growth and lung metastasis. Human malignant melanoma specimen analyses revealed a strong correlation between Notch1 intracellular domain, CD133, and p-p38 mitogen-activated protein kinase expression and malignant melanoma progression. Thus, targeting Notch1 and its regulated signaling network may have potential therapeutic implications for the management of cancer stem cell-mediated melanoma progression.
- Published
- 2016
- Full Text
- View/download PDF
16. The Biology and Therapeutic Implications of Tumor Dormancy and Reactivation
- Author
-
Amit S. Yadav, Poonam R. Pandey, Ramesh Butti, N. N. V. Radharani, Shamayita Roy, Shaileshkumar R. Bhalara, Mahadeo Gorain, Gopal C. Kundu, and Dhiraj Kumar
- Subjects
0301 basic medicine ,reactivation ,Cancer Research ,dormancy ,medicine.medical_treatment ,Cancer metastasis ,Review ,Biology ,lcsh:RC254-282 ,03 medical and health sciences ,cancer metastasis ,Adjuvant therapy ,medicine ,tumor microenvironment ,Epithelial–mesenchymal transition ,Cause of death ,Tumor microenvironment ,epithelial to mesenchymal transition ,Cancer ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,Radiation therapy ,030104 developmental biology ,Oncology ,Cancer research ,Dormancy - Abstract
Advancements in the early detection of cancer coupled with improved surgery, radiotherapy, and adjuvant therapy led to substantial increase in patient survival. Nevertheless, cancer metastasis is the leading cause of death in several cancer patients. The majority of these deaths are associated with metastatic relapse kinetics after a variable period of clinical remission. Most of the cancer recurrences are thought to be associated with the reactivation of dormant disseminated tumor cells (DTCs). In this review, we have summarized the cellular and molecular mechanisms related to DTCs and the role of microenvironmental niche. These mechanisms regulate the dormant state and help in the reactivation, which leads to metastatic outgrowth. Identification of novel therapeutic targets to eliminate these dormant tumor cells will be highly useful in controlling the metastatic relapse-related death with several cancers.
- Published
- 2018
- Full Text
- View/download PDF
17. Non-migratory tumorigenic intrinsic cancer stem cells ensure breast cancer metastasis by generation of CXCR4+ migrating cancer stem cells
- Author
-
Tanya Das, Shravanti Mukherjee, Gopal C. Kundu, Arghya Adhikary, Pushpak Bhattacharjee, Mahadeo Gorain, Deblina Guha, Sanhita Mukherjee, Argha Manna, Samik Chakraborty, Minakshi Mazumdar, Diptendra K. Sarkar, Sudipto Saha, and Debarshi Jana
- Subjects
0301 basic medicine ,Receptors, CXCR4 ,Cancer Research ,Carcinogenesis ,Breast Neoplasms ,Biology ,medicine.disease_cause ,CXCR4 ,Metastasis ,03 medical and health sciences ,Cell Movement ,Cancer stem cell ,Genetics ,medicine ,Humans ,Neoplasm Metastasis ,Molecular Biology ,medicine.disease ,Primary tumor ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,Tumor progression ,Immunology ,Cancer cell ,MCF-7 Cells ,Neoplastic Stem Cells ,Cancer research ,Female ,Lymph Nodes ,Stem cell ,Signal Transduction - Abstract
Although the role of metastatic cancer stem cells (mCSCs) in tumor progression has been well documented, our study reveals a hitherto unidentified role of tumorigenic intrinsic CSCs (iCSCs) in breast cancer metastasis. We show that unlike highly migratory mCSCs residing in the breast tumor disseminating/peripheral regions, iCSCs populate the inner mass of the tumor and are non-migratory. However iCSCs, via paracrine signaling, induce conversion of non-stem cancer cells to CSCs that (i) are identical to the previously reported mCSCs, and (ii) in contrast to iCSCs, express chemokine receptor, chemokine (C-X-C motif) receptor 4 (CXCR4), which is crucial for their metastatic potential. These mCSCs also demonstrate high in vivo tumorigenicity. Physical non-participation of iCSCs in metastasis is further validated in vivo, where only mCSCs are found to exist in the metastatic sites, lymph nodes and bone marrow, whereas the primary tumor retains both iCSCs and mCSCs. However, iCSCs ensure metastasis since their presence is crucial for deliverance of highly metastatic CXCR4(+) mCSCs to the migrating fraction of cells. Cumulatively, these results unveil a novel role of iCSCs in breast cancer metastasis as parental regulators of CXCR4(+) mCSCs, and highlight the therapeutic requisite of targeting iCSCs, but not CXCR4(+) mCSCs, to restrain breast cancer metastasis from the root by inhibiting the generation of mCSCs from iCSCs. Considering the pivotal role of iCSCs in tumor metastasis, the possibility of metastasis to be a 'stem cell phenomena' is suggested.
- Published
- 2016
- Full Text
- View/download PDF
18. Additional file 1: Figure S1. of Epoxyazadiradione suppresses breast tumor growth through mitochondrial depolarization and caspase-dependent apoptosis by targeting PI3K/Akt pathway
- Author
-
Dhiraj Kumar, Saikat Haldar, Mahadeo Gorain, Kumar, Santosh, Fayaj Mulani, Yadav, Amit, Miele, Lucio, Hirekodathakallu Thulasiram, and Kundu, Gopal
- Subjects
skin and connective tissue diseases - Abstract
Epoxyazadiradione inhibits breast cancer cell viability. Figure S2. Epoxyazadiradione induces cell death through ROS and AIF independent pathway in MCF-7 cells. Figure S3. Epoxyazadiradione attenuates breast cancer cell migration through downregulation of PI3K/Akt pathway. Table S1. List of antibodies used for western blot and immunofluorescence. Table S2. IC50 of epoxyazadiradione in breast cancer (MDA-MB-231 and MCF-7) cells. (PDF 3912 kb)
- Published
- 2018
- Full Text
- View/download PDF
19. Cross-talk between Endoplasmic Reticulum (ER) Stress and the MEK/ERK Pathway Potentiates Apoptosis in Human Triple Negative Breast Carcinoma Cells
- Author
-
Mahadeo Gorain, Swatilekha Ghosh, Salil Putatunda, Arghya Adhikary, Pushpak Bhattacharjee, Supriya Chakraborty, Gopal C. Kundu, Minakshi Mazumder, Arijit Chakraborty, Parimal C. Sen, and Tanya Das
- Subjects
MAPK/ERK pathway ,biology ,Endoplasmic reticulum ,Cell Biology ,Biochemistry ,Cell biology ,Transcription (biology) ,Apoptosis ,biology.protein ,Unfolded protein response ,Triple-Negative Breast Carcinoma ,Signal transduction ,Molecular Biology ,Caspase - Abstract
Triple negative breast cancers (TNBC) are among the most aggressive and therapy-resistant breast tumors and currently possess almost no molecular targets for therapeutic options in this horizon. In the present study we discerned the molecular mechanisms of potential interaction between the endoplasmic reticulum (ER) stress response and the MEK/ERK pathway in inducing apoptosis in TNBC cells. Here we observed that induction of ER stress alone was not sufficient to trigger significant apoptosis but simultaneous inhibition of the MEK/ERK pathway enhanced ER stress-induced apoptosis via a caspase-dependent mechanism. Our study also demonstrated nifetepimine, a dihydropyrimidone derivative as a potent anti-cancer agent in TNBC cells. Nifetepimine down-regulated the MEK/ERK pathway in MDAMB-231 and MDAMB-468 cells and resulted in blockage of ER stress-mediated GRP78 up-regulation. Detailed mechanistic studies also revealed that nifetepimine by down-regulating pERK expression also declined the promoter binding activity of TFII-I to the GRP78 promoter and in turn regulated GRP78 transcription. Studies further extended to in vivo Swiss albino and SCID mice models also revalidated the anti-carcinogenic property of nifetepimine. Thus our findings cumulatively suggest that nifetepimine couples two distinct signaling pathways to induce the apoptotic death cascade in TNBC cells and raises the possibility for the use of nifetepimine as a potent anti-cancer agent with strong immune-restoring properties for therapeutic intervention for this group of cancer bearers.
- Published
- 2015
- Full Text
- View/download PDF
20. Therapeutic implications of cellular and molecular biology of cancer stem cells in melanoma
- Author
-
Mahadeo Gorain, Gopal C. Kundu, Dhiraj Kumar, and Gautam Kundu
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Microenvironment ,Angiogenesis ,medicine.medical_treatment ,Antineoplastic Agents ,Review ,Tumor initiation ,Biology ,Metastasis ,03 medical and health sciences ,0302 clinical medicine ,Cancer stem cell ,Internal medicine ,Tumor Microenvironment ,medicine ,Humans ,Melanoma ,Melanoma growth ,Cancer ,medicine.disease ,Embryonic stem cell ,Signaling ,Radiation therapy ,030104 developmental biology ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Neoplastic Stem Cells ,CSCs ,Molecular Medicine ,Signal Transduction - Abstract
Melanoma is a form of cancer that initiates in melanocytes. Melanoma has multiple phenotypically distinct subpopulation of cells, some of them have embryonic like plasticity which are involved in self-renewal, tumor initiation, metastasis and progression and provide reservoir of therapeutically resistant cells. Cancer stem cells (CSCs) can be identified and characterized based on various unique cell surface and intracellular markers. CSCs exhibit different molecular pattern with respect to non-CSCs. They maintain their stemness and chemoresistant features through specific signaling cascades. CSCs are weak in immunogenicity and act as immunosupressor in the host system. Melanoma treatment becomes difficult and survival is greatly reduced when the patient develop metastasis. Standard conventional oncology treatments such as chemotherapy, radiotherapy and surgical resection are only responsible for shrinking the bulk of the tumor mass and tumor tends to relapse. Thus, targeting CSCs and their microenvironment niche addresses the alternative of traditional cancer therapy. Combined use of CSCs targeted and traditional therapies may kill the bulk tumor and CSCs and offer a promising therapeutic strategy for the management of melanoma.
- Published
- 2017
- Full Text
- View/download PDF
21. Epoxyazadiradione suppresses breast tumor growth through mitochondrial depolarization and caspase-dependent apoptosis by targeting PI3K/Akt pathway
- Author
-
Mahadeo Gorain, Dhiraj Kumar, Hirekodathakallu V. Thulasiram, Lucio Miele, Fayaj A. Mulani, Gopal C. Kundu, Santosh Kumar, Saikat Haldar, and Amit S. Yadav
- Subjects
0301 basic medicine ,Limonins ,Vascular Endothelial Growth Factor A ,Cancer Research ,Angiogenesis ,Apoptosis ,Breast Neoplasms ,lcsh:RC254-282 ,PI3K ,Metastasis ,03 medical and health sciences ,Mice ,Phosphatidylinositol 3-Kinases ,0302 clinical medicine ,Breast cancer ,Cell Movement ,Genetics ,medicine ,Animals ,Humans ,Limonoids ,PI3K/AKT/mTOR pathway ,Triple-negative breast cancer ,Cell Proliferation ,Membrane Potential, Mitochondrial ,Cell growth ,Chemistry ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,Xenograft Model Antitumor Assays ,Mitochondria ,Gene Expression Regulation, Neoplastic ,Vascular endothelial growth factor A ,030104 developmental biology ,Oncology ,Matrix Metalloproteinase 9 ,Cyclooxygenase 2 ,030220 oncology & carcinogenesis ,Cancer research ,MCF-7 Cells ,Female ,Proto-Oncogene Proteins c-akt ,Research Article - Abstract
Background Breast cancer is one of the most commonly diagnosed invasive cancers among women around the world. Among several subtypes, triple negative breast cancer (TNBC) is highly aggressive and chemoresistant. Treatment of TNBC patients has been challenging due to heterogeneity and devoid of well-defined molecular targets. Thus, identification of novel effective and selective agents against TNBC is essential. Methods We used epoxyazadiradione to assess the cell viability, mitochondrial potential, ROS level, cell migration, apoptosis and protein expression in cell culture models of TNBC MDA-MB-231 and ER+ MCF-7 breast cancer cells. The molecular mechanism was examined in two different type of breast cancer cells in response to epoxyazadiradione. We have also analyzed the effect of epoxyazadiradione on breast tumor growth using in vivo mice model. Results In this study, we for the first time investigated that out of 10 major limonoids isolated from Azadirachta indica, epoxyazadiradione exhibits most potent anti-cancer activity in both TNBC and ER+ breast cancer cells. Epoxyazadiradione induces apoptosis and inhibits PI3K/Akt-mediated mitochondrial potential, cell viability, migration and angiogenesis. It also inhibits the expression of pro-angiogenic and pro-metastatic genes such as Cox2, OPN, VEGF and MMP-9 in these cells. Furthermore, epoxyazadiradione attenuates PI3K/Akt-mediated AP-1 activation. Our in vivo data revealed that epoxyazadiradione suppresses breast tumor growth and angiogenesis in orthotopic NOD/SCID mice model. Conclusion Our findings demonstrate that epoxyazadiradione inhibits PI3K/Akt-dependent mitochondrial depolarisation, induces apoptosis and attenuates cell migration, angiogenesis and breast tumor growth suggesting that this compound may act as a potent therapeutic agent for the management of breast cancer. Electronic supplementary material The online version of this article (10.1186/s12885-017-3876-2) contains supplementary material, which is available to authorized users.
- Published
- 2017
22. Commercial Non-Timber Forest Products collected by Arid Region and Marketed by Pawata and Basani in Jodhpur Town of Jodhpur District, Rajasthan
- Author
-
Mahadeo Gorain and Anamika Sharma
- Abstract
The study documents the commercial Non-Timber Forest Products, collected by arid and semi-arid region and marketed by the tribals in Jodhpur town of Jodhpur District, Rajasthan. These tribals collecting a total of 32 products as commercial NTFPs which are sold in the open daily and highly in weekly tribal market by the tribal people. Buyers come from within and around the town. Women constitute over 60% of the traders in these markets.
- Published
- 2012
- Full Text
- View/download PDF
23. Diosgenin Functionalized Iron Oxide Nanoparticles as Novel Nanomaterial Against Breast Cancer
- Author
-
Sangeeta Kale, Jayesh R. Bellare, Sougata Ghosh, Rohini Kitture, Mahadeo Gorain, Dilip D. Dhavale, Balu A. Chopade, Gopal C. Kundu, Abhishek Derle, Piyush More, Pramod R. Markad, Trupti Kale, and Ashish Avasthi
- Subjects
Materials science ,Stereochemistry ,Cell Lines ,Cytotoxicity ,Biomedical Engineering ,Bioengineering ,Apoptosis ,Breast Neoplasms ,Diosgenin ,chemistry.chemical_compound ,Medicinal-Plants ,Breast Cancer ,Gold Nanoparticles ,Humans ,General Materials Science ,MTT assay ,Magnetite Nanoparticles ,Alexa Fluor ,Drug Carriers ,Iron Oxide Nanoparticles ,Mcf-7 ,Drug-Delivery ,General Chemistry ,Condensed Matter Physics ,Antineoplastic Agents, Phytogenic ,chemistry ,MCF-7 ,Colloidal gold ,Fe3o4 Magnetite Nanoparticles ,Antimicrobial Agents ,MCF-7 Cells ,Issue-On Reviews ,Antiproliferative ,Citric acid ,Iron oxide nanoparticles ,Nuclear chemistry - Abstract
Iron oxide nanoparticles (IONPs) have gained immense importance recently as drug nanocarriers due to easy multifunctionalization, simultaneous targeting, imaging and cancer hyperthermia. Herein, we report a novel nanomedicine comprising of IONPs core functionalized with a potent anticancer bioactive principle, diosgenin from medicinal plant Dioscorea bulbifera via citric acid linker molecule. IONPs were synthesized by reverse co-precipitation and characterized using field emission scanning electron microscopy (FESEM), high resolution transmission electron microscopy (HRTEM) and dynamic light scattering (DLS). Diosgenin functionalization was confirmed using fourier transform infrared spectroscopy (FTIR) and biochemical methods. Synthesized IONPs, citrate linked IONPs (IONPs-CA), diosgenin functionalized IONPs (IONPs-D) along with free citric acid and diosgenin were checked for anticancer activity against MCF7 breast cancer cells by MU assay, wound migration assay, confocal microscopy and protein expression by western blotting. Size of IONPs, IONPs-CA and IONPs-D gradually increased ranging from 12 to 21 nm as confirmed by FESEM and HRTEM. Signature peaks of diosgenin at 2914, 1166 and 1444 cm(-1) IONPs-D, revealed in FTIR indicated the presence of functionalized diosgenin. IONPs-D exhibited 51.08 +/- 0.37% antiproliferative activity against MCF7 cells, which was found to be superior to free citric acid (17.71 +/- 0.58%) and diosgenin (33.31 +/- 0.37%). Treatment with IONPs-D exhibited reduced wound migration upto 40.83 +/- 2.91% compared to bare IONPs (89.03 +/- 2.58%) and IONPs-CA (50.35 +/- 0.48%). IONPs-D and diosgenin exhibited apoptosis induction, confirmed by Alexa Fluor 488 annexin V/PI double-stained cells indicating extensive cell membrane damage coupled with PI influx leading to nuclear staining in treated cells. IONPs-D mediated selective PARP cleavage strongly rationalized it as superior apoptotic inducers. Based on these findings, IONPs-D can be considered as first diosgenin functionalized novel magnetic nanomedicine with antiproliferative, migration inhibiting and apoptosis inducing properties against breast cancer.
- Published
- 2015
24. Notch1-MAPK Signaling Axis Regulates CD133
- Author
-
Dhiraj, Kumar, Santosh, Kumar, Mahadeo, Gorain, Deepti, Tomar, Harshal S, Patil, Nalukurthi N V, Radharani, Totakura V S, Kumar, Tushar V, Patil, Hirekodathakallu V, Thulasiram, and Gopal C, Kundu
- Subjects
Neovascularization, Pathologic ,Carcinogenesis ,MAP Kinase Signaling System ,Cell Differentiation ,Neoplasms, Experimental ,Immunohistochemistry ,Tumor Burden ,Gene Expression Regulation, Neoplastic ,Mice, Inbred C57BL ,Disease Models, Animal ,Mice ,Random Allocation ,Cell Line, Tumor ,Neoplastic Stem Cells ,Animals ,Humans ,AC133 Antigen ,Receptor, Notch1 - Abstract
Functional characterization and understanding of the intricate signaling mechanisms in stem-like cells is crucial for the development of effective therapies in melanoma. We have studied whether melanoma cells are phenotypically distinct and hierarchically organized according to their tumorigenic nature. We report that melanoma-specific CD133
- Published
- 2015
25. MiRNA199a-3p suppresses tumor growth, migration, invasion and angiogenesis in hepatocellular carcinoma by targeting VEGFA, VEGFR1, VEGFR2, HGF and MMP2
- Author
-
Debanjali Dasgupta, Soma Banerjee, Gopal Krishna Dhali, Simanti Datta, Dhiraj Kumar, Thomas D. Schmittgen, Alip Ghosh, Ramesh Butti, Mahadeo Gorain, Subash Gupta, Shaleen Agarwal, Amit Ghosh, Shrabasti Roychoudhury, Gopal C. Kundu, and Abhijit Chowdhury
- Subjects
Vascular Endothelial Growth Factor A ,0301 basic medicine ,Cancer Research ,Pathology ,medicine.medical_specialty ,Carcinoma, Hepatocellular ,MMP2 ,Angiogenesis ,Immunology ,Biology ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Cell Movement ,Human Umbilical Vein Endothelial Cells ,medicine ,Humans ,Neoplasm Invasiveness ,RNA, Neoplasm ,Tube formation ,Tumor microenvironment ,Vascular Endothelial Growth Factor Receptor-1 ,Neovascularization, Pathologic ,Hepatocyte Growth Factor ,Liver Neoplasms ,Kinase insert domain receptor ,Hep G2 Cells ,Cell Biology ,Vascular Endothelial Growth Factor Receptor-2 ,Neoplasm Proteins ,Gene Expression Regulation, Neoplastic ,MicroRNAs ,Vascular endothelial growth factor A ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,Matrix Metalloproteinase 2 ,Original Article ,Hepatocyte growth factor ,medicine.drug - Abstract
Increasing significance of tumor–stromal interaction in development and progression of cancer implies that signaling molecules in the tumor microenvironment (TME) might be the effective therapeutic targets for hepatocellular carcinoma (HCC). Here, the role of microRNA miR-199a-3p in the regulation of TME and development of HCC has been investigated by several in vitro and in vivo assays. Expression of miR-199a-3p was observed significantly low in HCC tissues and its overexpression remarkably inhibited in vivo tumor growth and metastasis to lung in NOD-SCID mice. In vitro restoration of miR-199a-3p expression either in endothelial cells (ECs) or in cancer cells (CACs) significantly diminished migration of ECs in co-culture assay. Again incubation of miR-199a-3p transfected ECs with either conditioned media (CM) of CACs or recombinant VEGF has reduced tube formation, in ECs and it was also dropped upon growth in CM of either anti-VEGF antibody-treated or miR-199a-3p-transfected CACs. In addition, bioinformatics and luciferase-reporter assays revealed that miR-199a-3p inhibited VEGF secretion from CACs and VEGFR1 and VEGFR2 expression on ECs and thus restricted cross talk between CACs and ECs. Again, restoration of miR-199a-3p in hepatic stellate cells (HSCs) reduced migration and invasion of CACs in co-culture assay, while it was enhanced by the overexpression of HGF suggesting miR-199a-3p has hindered HSC-CACs cross talk probably by inhibiting HGF and regulating matrix metalloproteinase MMP2, which were found as targets of miR-199a-3p subsequently by luciferase-reporter assay and gelatin zymography, respectively. Thus, these findings collectively highlight that miR-199a-3p restricts metastasis, invasion and angiogenesis in HCC and hence it may be considered as one of the powerful effective therapeutics for management of HCC patients.
- Published
- 2017
- Full Text
- View/download PDF
26. Osteopontin as a therapeutic target for cancer
- Author
-
Rosalin Mishra, Gopal C. Kundu, Kumar V. S. Totakura, Goutam Chakraborty, Gowrishankar Soundararajan, Smita Kale, Ramesh Butti, Anuradha Bulbule, Remya Raja, Monalisa Bandopadhyay, Radha Nalukurthi, Gaurab Roy, Priyanka Ghorpade, Mahadeo Gorain, Pompom Ghosh, Priyanka Sharma, Santosh Kumar, Dhiraj Kumar, Deepti Tomar, Amit S. Yadav, Dhanashri Thorat, and Dattatrya Shetti
- Subjects
Angiogenesis ,medicine.medical_treatment ,Clinical Biochemistry ,Targeted therapy ,Metastasis ,stomatognathic system ,Neoplasms ,Drug Discovery ,medicine ,Tumor Microenvironment ,Animals ,Humans ,Osteopontin ,Molecular Targeted Therapy ,Neoplasm Metastasis ,Cell Proliferation ,Pharmacology ,Tumor microenvironment ,biology ,Neovascularization, Pathologic ,Melanoma ,medicine.disease ,Cell biology ,HIF1A ,Tumor progression ,biology.protein ,Disease Progression ,Molecular Medicine ,Stromal Cells ,Signal Transduction - Abstract
Cancer is a complex pathological disorder, established as a result of accumulation of genetic and epigenetic changes, which lead to adverse alterations in the cellular phenotype. Tumor progression involves intricate signaling mediated through crosstalk between various growth factors, cytokines and chemokines. Osteopontin (OPN), a chemokine-like protein, is involved in promotion of neoplastic cancer into higher grade malignancies by regulating various facets of tumor progression such as cell proliferation, angiogenesis and metastasis.Tumors as well as stroma-derived OPN play key roles in various signaling pathways involved in tumor growth, angiogenesis and metastasis. OPN derived from tumor-activated macrophages modulates the tumor microenvironment and thereby regulate melanoma growth and angiogenesis. OPN also regulates hypoxia-inducible factor-1α-dependent VEGF expression leading to breast tumor growth and angiogenesis in response to hypoxia. Thus, a clear understanding of the molecular mechanism underlying OPN-mediated regulation will shed light on exciting avenues for further investigation of targeted therapies. Silencing of OPN using RNAi technology, blocking OPN activity using specific antibodies and small-molecule inhibitors might provide novel strategies, which would aid in developing effective therapeutics for the treatment of various types of cancer.This review focuses on new possibilities to exploit OPN as a tumor and stroma-derived therapeutic target to combat cancer.
- Published
- 2014
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.