Please use this identifier to cite or link to this item: http://hdl.handle.net/123456789/1162
Full metadata record
DC FieldValueLanguage
dc.contributor.authorSengupta, Sagar-
dc.contributor.authorDasgupta, Ujjaini-
dc.contributor.authorBajaj, Avinash-
dc.contributor.authorPal, Sanjay-
dc.contributor.authorMedatwal, Nihal-
dc.contributor.authorKumar, Sandeep-
dc.contributor.authorKar, Animesh-
dc.contributor.authorKomalla, Varsha-
dc.contributor.authorYavvari, Prabhu Srinivas-
dc.contributor.authorMishra, Deepakkumar-
dc.contributor.authorRizvi, Zaigham Abbas-
dc.contributor.authorNandan, Shiv-
dc.contributor.authorMalakar, Dipankar-
dc.contributor.authorPillai, Manoj-
dc.contributor.authorAwasthi, Amit-
dc.contributor.authorDas, Prasenjit-
dc.contributor.authorSharma, Ravi Datta-
dc.contributor.authorSrivastava, Aasheesh-
dc.date.accessioned2021-03-01T10:06:37Z-
dc.date.available2021-03-01T10:06:37Z-
dc.date.issued2019-10-
dc.identifier.urihttp://hdl.handle.net/123456789/1162-
dc.description.abstractRapid proliferation of cancer cells assisted by endothelial cell-mediated angiogenesis and acquired inflammation at the tumor microenvironment (TME) lowers the success rate of chemotherapeutic regimens. Therefore, targeting these processes using localized delivery of a minimally toxic drug combination may be a promising strategy. Here, we present engineering of a biocompatible self-assembled lithocholic acid-dipeptide derived hydrogel (TRI-Gel) that can maintain sustained delivery of antiproliferating doxorubicin, antiangiogenic combretastatin-A4 and anti-inflammatory dexamethasone. Application of TRI-Gel therapy to a murine tumor model promotes enhanced apoptosis with a concurrent reduction in angiogenesis and inflammation, leading to effective abrogation of tumor proliferation and increased median survival with reduced drug resistance. In-depth RNA-sequencing analysis showed that TRI-Gel therapy induced transcriptome-wide alternative splicing of many genes responsible for oncogenic transformation including sphingolipid genes. We demonstrate that TRI-Gel therapy targets the reversal of a unique intron retention event in β-glucocerebrosidase 1 (Gba1), thereby increasing the availability of functional Gba1 protein. An enhanced Gba1 activity elevates ceramide levels responsible for apoptosis and decreases glucosylceramides to overcome drug resistance. Therefore, TRI-Gel therapy provides a unique system that affects the TME via post-transcriptional modulations of sphingolipid metabolic genes, thereby opening a new and rational approach to cancer therapy.en_US
dc.language.isoenen_US
dc.publisherAmerican Chemical Societyen_US
dc.titleA Localized Chimeric Hydrogel Therapy Combats Tumor Progression through Alteration of Sphingolipid Metabolismen_US
dc.typeArticleen_US
dc.journalACS Cent Scien_US
dc.volumeno5en_US
dc.issueno10en_US
dc.pages1648-1662en_US
Appears in Collections:Signal Transduction-II, Publications

Files in This Item:
File Description SizeFormat 
acscentsci.9b00551.pdf8 MBAdobe PDFView/Open    Request a copy


Items in DSpace are protected by copyright, with all rights reserved, unless otherwise indicated.