Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
3.
Nanomedicine (Lond) ; 15(3): 303-318, 2020 02.
Article in English | MEDLINE | ID: mdl-31802702

ABSTRACT

In the development of cancer chemotherapy, besides the discovery of new anticancer drugs, a variety of nanocarrier systems for the delivery of previously developed and new chemotherapeutic drugs have currently been explored. Liposome is one of the most studied nanocarrier systems because of its biodegradability, simple preparation method, high efficacy and low toxicity. To make the best use of this vehicle, a number of multifunctionalized liposomal formulations have been investigated. The objective of this review is to summarize the current development of novel active targeting liposomal formulations, and to give insight into the challenges and future direction of the field. The recent studies in active targeting liposomes suggest the great potential of precise targeted anticancer drug delivery in cancer therapeutics.


Subject(s)
Antineoplastic Agents/therapeutic use , Drug Delivery Systems , Liposomes/therapeutic use , Neoplasms/drug therapy , Antineoplastic Agents/chemistry , Drug Carriers/chemistry , Drug Carriers/therapeutic use , Humans , Liposomes/chemistry
4.
World J Gastroenterol ; 24(27): 2949-2973, 2018 Jul 21.
Article in English | MEDLINE | ID: mdl-30038463

ABSTRACT

MicroRNAs (miRNAs) are small, single-stranded, noncoding RNAs that can post-transcriptionally regulate the expression of various oncogenes and tumor suppressor genes. Dysregulated expression of many miRNAs have been shown to mediate the signaling pathways critical in the multistep carcinogenesis of colorectal cancer (CRC). MiRNAs are stable and protected from RNase-mediated degradation, thereby enabling its detection in biological fluids and archival tissues for biomarker studies. This review focuses on the role and application of miRNAs in the prognosis and therapy of CRC. While stage II CRC is potentially curable by surgical resection, a significant percentage of stage II CRC patients do develop recurrence. MiRNA biomarkers may be used to stratify such high-risk population for adjuvant chemotherapy to provide better prognoses. Growing evidence also suggests that miRNAs are involved in the metastatic process of CRC. Certain of these miRNAs may thus be used as prognostic biomarkers to identify patients more likely to have micro-metastasis, who could be monitored more closely after surgery and/or given more aggressive adjuvant chemotherapy. Intrinsic and acquired resistance to chemotherapy severely hinders successful chemotherapy in CRC treatment. Predictive miRNA biomarkers for response to chemotherapy may identify patients who will benefit the most from a particular regimen and also spare the patients from unnecessary side effects. Selection of patients to receive the new targeted therapy is becoming possible with the use of predictive miRNA biomarkers. Lastly, forced expression of tumor suppressor miRNA or silencing of oncogenic miRNA in tumors by gene therapy can also be adopted to treat CRC alone or in combination with other chemotherapeutic drugs.


Subject(s)
Biomarkers, Tumor/analysis , Colorectal Neoplasms/genetics , MicroRNAs/analysis , Neoplasm Recurrence, Local/genetics , Translational Research, Biomedical/trends , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/antagonists & inhibitors , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinogenesis/genetics , Chemotherapy, Adjuvant/methods , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Colorectal Neoplasms/therapy , Gene Expression Regulation, Neoplastic , Gene Silencing , Genetic Therapy/methods , Humans , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , MicroRNAs/metabolism , Neoplasm Recurrence, Local/prevention & control , Neoplasm Staging , Patient Selection , Prognosis , Signal Transduction/genetics
5.
Methods Mol Biol ; 1012: 77-84, 2013.
Article in English | MEDLINE | ID: mdl-24006059

ABSTRACT

Protein-protein interaction is an essential biochemical event that mediates various cellular processes including gene expression, intracellular signaling, and intercellular interaction. Understanding such interaction is key to the elucidation of mechanisms of cellular processes in biology and diseases. The hypoxia-inducible transcription factor HIF-1α possesses a non-transcriptional activity that competes with c-Myc for Sp1 binding, whereas its isoform HIF-2α lacks Sp1-binding activity due to phosphorylation. Here, we describe the use of in vitro translation to effectively investigate the dynamics of protein-protein interactions among HIF-1α, c-Myc, and Sp1 and to demonstrate protein phosphorylation as a molecular determinant that functionally distinguishes HIF-2α from HIF-1α.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Protein Interaction Mapping/methods , Proto-Oncogene Proteins c-myc/metabolism , Sp1 Transcription Factor/metabolism , Binding, Competitive , Protein Binding , Transcriptional Activation
6.
Chin Med ; 5: 26, 2010 Jul 25.
Article in English | MEDLINE | ID: mdl-20653978

ABSTRACT

Multi-drug resistance (MDR) of cancer cells severely limits therapeutic outcomes. A proposed mechanism for MDR involves the efflux of anti-cancer drugs from cancer cells, primarily mediated by ATP-binding cassette (ABC) membrane transporters including P-glycoprotein. This article reviews the recent progress of using active ingredients, extracts and formulae from Chinese medicine (CM) in circumventing ABC transporters-mediated MDR. Among the ABC transporters, Pgp is the most extensively studied for its role in MDR reversal effects. While other MDR reversal mechanisms remain unclear, Pgp inhibition is a criterion for further mechanistic study. More mechanistic studies are needed to fully establish the pharmacological effects of potential MDR reversing agents.

7.
Cell Cycle ; 6(15): 1919-26, 2007 Aug 01.
Article in English | MEDLINE | ID: mdl-17671423

ABSTRACT

Hypoxia, a key microenvironmental factor for tumor development, not only stimulates angiogenesis and glycolysis for tumor expansion, but also induces cell cycle arrest and genetic instability for tumor progression. Several independent studies have shown hypoxic blockade of cell cycle progression at the G1/S transition, arising from the inactivation of S-phase-promoting cyclin E-CDK2 kinase complex. Despite these findings, the biochemical pathways leading to the cell cycle arrest remain poorly defined. We recently showed that hypoxic activates the expression of CDNK1A encoding the CDK2 inhibitor p21Cip1, through a novel HIF-1alpha-Myc pathway that involves Myc displacement from the CDNK1A promoter by the hypoxia-inducible transcription factor HIF-1alpha. In pursuit of further understanding of the hypoxic effects on cell cycle in tumor cells, here we report that hypoxia inhibits the expression of CDC25A, another cell cycle gene encoding a tyrosine phosphatase that maintains CDK2 activity. In accordance with the HIF-1alpha-Myc pathway, hypoxia requires HIF-1alpha for CDC25A repression, resulting in a selective displacement of an activating Myc from the CDC25A promoter without affecting a canonical Myc binding in the intron. Intriguingly, HIF-1alpha alone fails to recapitulate the hypoxic effect, indicating that HIF-1alpha is necessary but insufficient for the hypoxic repression. Taken together, our studies indicate that hypoxia inhibits cell cycle progression by controlling the expression of various cell cycle genes.


Subject(s)
Down-Regulation/genetics , Gene Expression Regulation, Neoplastic/genetics , Neoplasms/enzymology , cdc25 Phosphatases/genetics , cdc25 Phosphatases/metabolism , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/metabolism , Cell Differentiation , Cell Hypoxia/genetics , Cell Line, Tumor , Cell Proliferation , Checkpoint Kinase 1 , Gene Expression Profiling , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Neoplasms/genetics , Neoplasms/pathology , Promoter Regions, Genetic/genetics , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Signal Transduction , Transcription, Genetic/genetics , Tumor Suppressor Protein p53/metabolism
8.
EMBO J ; 25(20): 4784-94, 2006 Oct 18.
Article in English | MEDLINE | ID: mdl-17024177

ABSTRACT

Hypoxia promotes genetic instability for tumor progression. Recent evidence indicates that the transcription factor HIF-1alpha impairs DNA mismatch repair, yet the role of HIF-1alpha isoform, HIF-2alpha, in tumor progression remains obscure. In pursuit of the involvement of HIF-alpha in chromosomal instability, we report here that HIF-1alpha, specifically its PAS-B, induces DNA double-strand breaks at least in part by repressing the expression of NBS1, a crucial DNA repair gene constituting the MRE11A-RAD50-NBS1 complex. Despite strong similarities between the two isoforms, HIF-2alpha fails to do so. We demonstrate that this functional distinction stems from phosphorylation of HIF-2alpha Thr-324 by protein kinase D1, which discriminates between subtle differences of the two PAS-B in amino-acid sequence, thereby precluding NBS1 repression. Hence, our findings delineate a molecular pathway that functionally distinguishes HIF-1alpha from HIF-2alpha, and arguing a unique role for HIF-1alpha in tumor progression by promoting genomic instability.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Cycle Proteins/metabolism , Chromosomal Instability , Gene Expression Regulation, Neoplastic , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Nuclear Proteins/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Cycle Proteins/genetics , Cell Line , Chromosomal Instability/genetics , DNA Repair/genetics , Down-Regulation/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Neoplasm Proteins/genetics , Neoplasms/genetics , Nuclear Proteins/genetics , Phosphorylation , Protein Processing, Post-Translational , Protein Structure, Tertiary/genetics , Rabbits
9.
Mol Cell ; 17(6): 793-803, 2005 Mar 18.
Article in English | MEDLINE | ID: mdl-15780936

ABSTRACT

Hypoxia promotes genetic instability by undefined mechanisms. The transcription factor HIF-1alpha is crucial for the cellular response to hypoxia and is frequently overexpressed in human cancers, resulting in the activation of genes essential for cell survival. Here, we demonstrate that HIF-1alpha is responsible for genetic instability at the nucleotide level by inhibiting MSH2 and MSH6, thereby decreasing levels of the MSH2-MSH6 complex, MutSalpha, which recognizes base mismatches. HIF-1alpha displaces the transcriptional activator Myc from Sp1 binding to repress MutSalpha expression in a p53-dependent manner; Sp1 serves as a molecular switch by recruiting HIF-1alpha to the gene promoter under hypoxia. Furthermore, in human sporadic colon cancers, HIF-1alpha overexpression is statistically associated with the loss of MSH2 expression, especially when p53 is immunochemically undetectable. These findings indicate that the regulation of DNA repair is an integral part of the hypoxic response, providing molecular insights into the mechanisms underlying hypoxia-induced genetic instability.


Subject(s)
Cell Hypoxia , Chromosomal Instability , DNA-Binding Proteins/genetics , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins/genetics , Transcription Factors/metabolism , Transcription, Genetic , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , DNA Repair , DNA-Binding Proteins/metabolism , Down-Regulation , Humans , Hypoxia-Inducible Factor 1, alpha Subunit , MutS Homolog 2 Protein , Protein Binding , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism , Transcription Factors/genetics , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...