Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Cancers (Basel) ; 14(3)2022 Jan 20.
Article in English | MEDLINE | ID: mdl-35158774

ABSTRACT

Peptide drugs that target protein-protein interactions have attracted mounting research efforts towards clinical developments over the past decades. Increasing reports have indicated that expression of Musashi 1 (MSI1) is tightly correlated to high grade of cancers as well as enrichment of cancer stem cells. Treatment failure in malignant tumors glioblastoma multiform (GBM) had also been correlated to CSC-regulating properties of MSI1. It is thus imperative to develop new therapeutics that could effectively improve current regimens used in clinics. MSI1 and AGO2 are two emerging oncogenic molecules that both contribute to GBM tumorigenesis through mRNA regulation of targets involved in apoptosis and cell cycle. In this study, we designed peptide arrays covering the C-terminus of MSI1 and identified two peptides (Pep#11 and Pep#26) that could specifically interfere with the binding with AGO2. Our Biacore analyses ascertained binding between the identified peptides and AGO2. Recombinant reporter system Gaussian luciferase and fluorescent bioconjugate techniques were employed to determine biological functions and pharmacokinetic characteristics of these two peptides. Our data suggested that Pep#11 and Pep#26 could function as decoy peptides by mimicking the interaction function of MSI1 with its binding partner AGO2 in vitro and in vivo. Further experiments using GMB animal models corroborated the ability of Pep#11 and Pep#26 in disrupting MSI1/AGO2 interaction and consequently anti-tumorigenicity and prolonged survival rates. These striking therapeutic efficacies orchestrated by the synthetic peptides were attributed to the decoy function to C-terminal MSI1, especially in malignant brain tumors and glioblastoma.

2.
Cancers (Basel) ; 13(8)2021 Apr 09.
Article in English | MEDLINE | ID: mdl-33918794

ABSTRACT

Glioblastoma (GBM) is the most malignant brain tumor which is characterized by high proliferation and migration capacity. The poor survival rate has been attributed to limitations of the current standard therapies. The search for novel biological targets that can effectively hamper tumor progression remains extremely challenging. Previous studies indicated that tumor-associated macrophages (TAMs) are the abundant elements in the tumor microenvironment that are closely implicated in glioma progression and tumor pathogenesis. M2 type TAMs are immunosuppressive and promote GBM proliferation. RNA-binding protein Musashi-1 (MSI1) has recently been identified as a marker of neural stem/progenitor cells, and its high expression has been shown to correlate with the growth of GBM. Nevertheless, the relationship between MSI1 and TAMs in GBM is still unknown. Thus, in our present study, we aimed to investigate the molecular interplay between MSI1 and TAMs in contributing to GBM tumorigenesis. Our data revealed that the secretion of macrophage inhibitory factor 1 (MIF1) is significantly upregulated by MSI1 overexpression in vitro. Importantly, M2 surface markers of THP-1-derived macrophages were induced by recombinant MIF1 and reduced by using MIF1 inhibitor (S,R)-3-(4-hHydroxyphenyl)-4,5-dihydro-5-isoxazole acetic acid (ISO-1). Furthermore, GBM tumor model data suggested that the tumor growth, MIF1 expression and M2 macrophage population were significantly downregulated when MSI1 expression was silenced in vivo. Collectively, our findings identified a novel role of MSI1 in the secretion of MIF1 and the consequent polarization of macrophages into the M2 phenotype in promoting GBM tumor progression.

3.
Int J Mol Sci ; 20(10)2019 May 14.
Article in English | MEDLINE | ID: mdl-31091705

ABSTRACT

Protein-protein interactions (PPIs) execute many fundamental cellular functions and have served as prime drug targets over the last two decades. Interfering intracellular PPIs with small molecules has been extremely difficult for larger or flat binding sites, as antibodies cannot cross the cell membrane to reach such target sites. In recent years, peptides smaller size and balance of conformational rigidity and flexibility have made them promising candidates for targeting challenging binding interfaces with satisfactory binding affinity and specificity. Deciphering and characterizing peptide-protein recognition mechanisms is thus central for the invention of peptide-based strategies to interfere with endogenous protein interactions, or improvement of the binding affinity and specificity of existing approaches. Importantly, a variety of computation-aided rational designs for peptide therapeutics have been developed, which aim to deliver comprehensive docking for peptide-protein interaction interfaces. Over 60 peptides have been approved and administrated globally in clinics. Despite this, advances in various docking models are only on the merge of making their contribution to peptide drug development. In this review, we provide (i) a holistic overview of peptide drug development and the fundamental technologies utilized to date, and (ii) an updated review on key developments of computational modeling of peptide-protein interactions (PepPIs) with an aim to assist experimental biologists exploit suitable docking methods to advance peptide interfering strategies against PPIs.


Subject(s)
Drug Development/methods , Molecular Docking Simulation/methods , Peptides/chemistry , Animals , Humans , Machine Learning , Peptides/pharmacology , Protein Binding
4.
Cell Death Discov ; 5: 74, 2019.
Article in English | MEDLINE | ID: mdl-30854233

ABSTRACT

FUT1 and FUT2 encode alpha 1, 2-fucosyltransferases which catalyze the addition of alpha 1, 2-linked fucose to glycans. Glycan products of FUT1 and FUT2, such as Globo H and Lewis Y, are highly expressed on malignant tissues, including breast cancer. Herein, we investigated the roles of FUT1 and FUT2 in breast cancer. Silencing of FUT1 or FUT2 by shRNAs inhibited cell proliferation in vitro and tumorigenicity in mice. This was associated with diminished properties of cancer stem cell (CSC), including mammosphere formation and CSC marker both in vitro and in xenografts. Silencing of FUT2, but not FUT1, significantly changed the cuboidal morphology to dense clusters of small and round cells with reduced adhesion to polystyrene and extracellular matrix, including laminin, fibronectin and collagen. Silencing of FUT1 or FUT2 suppressed cell migration in wound healing assay, whereas FUT1 and FUT2 overexpression increased cell migration and invasion in vitro and metastasis of breast cancer in vivo. A decrease in mesenchymal like markers such as fibronectin, vimentin, and twist, along with increased epithelial like marker, E-cadherin, was observed upon FUT1/2 knockdown, while the opposite was noted by overexpression of FUT1 or FUT2. As expected, FUT1 or FUT2 knockdown reduced Globo H, whereas FUT1 or FUT2 overexpression showed contrary effects. Exogenous addition of Globo H-ceramide reversed the suppression of cell migration by FUT1 knockdown but not the inhibition of cell adhesion by FUT2 silencing, suggesting that at least part of the effects of FUT1/2 knockdown were mediated by Globo H. Our results imply that FUT1 and FUT2 play important roles in regulating growth, adhesion, migration and CSC properties of breast cancer, and may serve as therapeutic targets for breast cancer.

5.
J Mol Med (Berl) ; 97(3): 325-339, 2019 03.
Article in English | MEDLINE | ID: mdl-30612140

ABSTRACT

Oncogenic N-MYC (MYCN) is widely used as a biomarker in clinics for neuroblastoma (NB) patients; nevertheless, mechanism that underlines MYCN regulation remains elusive. In the present study, we identified calreticulin (CRT) as a novel MYCN suppressor that downregulated MYCN promoter activity and protein expression to modulate neuronal differentiation and stemness. Our data showed that CRT-mediated MYCN suppression led to increased neurite length and commensurate elevation in differentiation marker GAP-43. We examined effect of radiotherapy and discovered that ionizing radiation (IR) was able to augment CRT expression dose-dependently in NB. Interestingly, neuronal differentiation and neurosphere formation (NSF) of NB were not only co-modulated by IR and CRT but were also dependent on Ca2+-buffering domain (C-domain) of CRT. Mutagenesis analysis showed that C-domain was indispensable for CRT-mediated MYCN regulation in NB differentiation and NSF. Of note, IR-induced formation of neural stem-like neurospheres (NS) was significantly impaired in CRT-overexpressed NB cells. The occupancy of CRT on MYCN 5' proximal promoter was confirmed by chromatin immunoprecipitation assays, revealing potential CRT binding sites that coincided with transcription factor E2F1 binding elements. In addition, we identified a physical interaction between CRT and E2F1, and demonstrated that CRT occupancy on MYCN promoter prevented E2F1-mediated MYCN upregulation. In line with in vitro findings, hampered tumor latency and retarded tumor growth in xenograft model corroborated IR and CRT co-mediated neuronal differentiation of NB. Together, our data delineated a novel mechanism of CRT-mediated MYCN regulation and warranted further preclinical investigation towards new therapeutic strategy for NB. CRT suppresses MYCN expression and promotes neuronal differentiation in NB. CRT regulates MYCN via interaction with E2F1 and direct binding to MYCN promoter. Ca2+-buffering domain of CRT is critical in MYCN regulation and NB differentiation. CRT-MYCN axis impacts on NB stemness by modulating neurosphere formation. Xenograft model corroborates in vitro NB differentiation mediated by CRT and IR.


Subject(s)
Calreticulin/metabolism , N-Myc Proto-Oncogene Protein/metabolism , Neuroblastoma/metabolism , Animals , Cell Line, Tumor , Female , Humans , Mice, SCID , N-Myc Proto-Oncogene Protein/genetics , Neuroblastoma/pathology , Neuroblastoma/radiotherapy , Neurogenesis , Neurons/physiology , Promoter Regions, Genetic , Radiation, Ionizing
6.
Comput Biol Chem ; 69: 138-146, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28539271

ABSTRACT

Radiotherapy (RT) is a common cancer treatment approach that accounts for nearly 50% of patient treatment; however, tumor relapse after radiotherapy is still a major issue. To study the crucial role of tumor-associated macrophages (TAMs) in the regulation of tumor progression post-RT, microarray experiments comparing TAM gene expression profiles between unirradiated and irradiated tumors were conducted to discover possible roles of TAMs in initiation or contribution to tumor recurrence following RT, taking into account the relationships among gene expression, tumor microenvironment, and immunology. A single dose of 25Gy was given to TRAMP C-1 prostate tumors established in C57/B6 mice. CD11b-positive macrophages were extracted from the tumors at one, two and three weeks post-RT. Gene ontology (GO) term analysis using the DAVID database revealed that genes that were differentially expressed at one and two weeks after irradiation were associated with biological processes such as morphogenesis of a branching structure, tube development, and cell proliferation. Analysis using Short Time-Series Expression Miner (STEM) revealed the temporal gene expression profiles and identified 13 significant patterns in four main groups of profiles. The genes in the upregulated temporal profile have diverse functions involved in the intracellular signaling cascade, cell proliferation, and cytokine-mediated signaling pathway. We show that tumor irradiation with a single 25-Gy dose can initiate a time-series of differentially expressed genes in TAMs, which are associated with the immune response, DNA repair, cell cycle arrest, and apoptosis. Our study helps to improve our understanding of the function of the group of genes whose expression changes temporally in an irradiated tumor microenvironment.


Subject(s)
Gene Expression Profiling , Macrophages/metabolism , Macrophages/radiation effects , Neoplasms/genetics , Neoplasms/radiotherapy , Animals , Cell Proliferation , Cytokines/metabolism , Mice , Mice, Inbred C57BL , Neoplasms/metabolism , Neoplasms/pathology , X-Rays
7.
Biomaterials ; 94: 31-44, 2016 07.
Article in English | MEDLINE | ID: mdl-27088408

ABSTRACT

It is more challenging to design peptide drugs than small molecules through molecular docking and in silico analysis. Here, we developed a structure-based approach with various computational and analytical techniques to optimize cancer-targeting peptides for molecular imaging and therapy. We first utilized a peptide-binding protein database to identify GRP78, a specific cancer cell-surface marker, as a target protein for the lead, L-peptide. Subsequently, we used homologous modeling and molecular docking to identify a peptide-binding domain within GRP78 and optimized a series of peptides with a new protein-ligand scoring program, HotLig. Binding of these peptides to GRP78 was confirmed using an oriented immobilization technique for the Biacore system. We further examined the ability of the peptides to target cancer cells through in vitro binding studies with cell lines and clinical cancer specimens, and in vivo tumor imaging and targeted chemotherapeutic studies. MicroSPECT/CT imaging revealed significantly greater uptake of (188)Re-liposomes linked to these peptides as compared with non-targeting (188)Re-liposomes. Conjugation with these peptides also significantly increased the therapeutic efficacy of Lipo-Dox. Notably, peptide-conjugated Lipo-Dox significantly reduced stem-cell subpopulation in xenografts of breast cancer. The structure-based optimization strategy for peptides described here may be useful for developing peptide drugs for cancer imaging and therapy.


Subject(s)
Diagnostic Imaging , Heat-Shock Proteins/metabolism , Neoplasms/diagnosis , Neoplasms/drug therapy , Peptides/therapeutic use , Amino Acid Sequence , Animals , Cell Line, Tumor , Doxorubicin/analogs & derivatives , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Drug Design , Endoplasmic Reticulum Chaperone BiP , Humans , Ligands , Mice, SCID , Models, Molecular , Peptides/chemistry , Polyethylene Glycols/pharmacology , Polyethylene Glycols/therapeutic use , Protein Binding , Structure-Activity Relationship , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...