Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Chem Biol Drug Des ; 103(6): e14566, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38858134

ABSTRACT

The severe acute respiratory syndrome coronavirus (SARS-CoV-2) pandemic has triggered a significant impact on global public health security, it is urgent to develop effective antiviral drugs. Previous studies have found that binding to ACE2 is a key step in the invasion of SARS-CoV-2 into host cells, so virus invasion can be inhibited by blocking ACE2, but there are few reports on this kind of specific inhibitor. Our previous study found that Harringtonine (HT) can inhibit the entry of SARS-CoV-2 spike pseudovirus into ACE2h cells, but its relatively high cytotoxicity limits its further development. Amino acid modification of the active components can increase their solubility and reduce their cytotoxicity. Therefore, in this study, seven new derivatives were synthesized by amino acid modification of its core structure Cephalotaxine. The target compounds were evaluated by cell viability assay and the SARS-CoV-2 spike pseudovirus entry assay. Compound CET-1 significantly inhibited the entry of pseudovirus into ACE2h cells and showed less cytotoxicity than HT. Molecular docking results showed that CET-1 could bind TYR83, an important residue of ACE2, just like HT. In conclusion, our study provided a novel compound with more potential activity and lower toxicity than HT on inhibiting the SARS-CoV-2 spike pseudovirus infection, which makes it possible to be a lead compound as an antiviral drug in the future.


Subject(s)
Angiotensin-Converting Enzyme 2 , Antiviral Agents , COVID-19 Drug Treatment , Homoharringtonine , Molecular Docking Simulation , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Humans , Amino Acids/chemistry , Amino Acids/pharmacology , Angiotensin-Converting Enzyme 2/metabolism , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Antiviral Agents/chemical synthesis , Cell Survival/drug effects , COVID-19/virology , Homoharringtonine/pharmacology , Homoharringtonine/chemistry , SARS-CoV-2/drug effects , Spike Glycoprotein, Coronavirus/metabolism , Spike Glycoprotein, Coronavirus/antagonists & inhibitors , Spike Glycoprotein, Coronavirus/chemistry , Virus Internalization/drug effects , Harringtonines/chemistry , Harringtonines/pharmacology
2.
J Pharm Biomed Anal ; 240: 115927, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38141415

ABSTRACT

Harringtonine (HT) is an anticancer alkaloid early extracted and isolated from cephalotaxus fortunei Hook. f., also has various pharmacological activities such as antiviral, antibacterial, antimalarial, anti-inflammatory, antioxidant, herbicidal and insecticidal. However, the factors affecting the stability of HT, the main degradation sites and mechanisms involved in its disposal process in vivo have not yet been elucidated. This study utilized HPLC-fluorescence detection method to establish a simple quantitative detection method for HT with good accuracy, precision, and high sensitivity. Temperature and pH were the main factors affecting the stability of HT, which underwent significant degradation in high temperature and alkaline environments because of the occurrence of hydrolysis reactions. In isolated biological homogenates of SD rats, except gastrointestinal tract, HT was degraded in other sites, especially respiratory, mainly in airway and lungs, and systemic metabolism, mainly in livers, spleens, and kidneys. Through UPLC-Q-TOF-MS, three forced degradation products were identified as 4'-demethyl HT, cephalotaxine, and dehydrated HT, respectively. However, the degradation product in isolated biological homogenates of SD rats was only 4'-demethyl HT due to the relatively mild environment. Our findings contributed to a necessary study basis for HT in terms of structural optimization, dosage form selection, storage and transportation.


Subject(s)
Antineoplastic Agents , Harringtonines , Rats , Animals , Chromatography, High Pressure Liquid/methods , Rats, Sprague-Dawley , Harringtonines/chemistry , Homoharringtonine/chemistry
3.
Bioengineered ; 12(2): 12940-12953, 2021 12.
Article in English | MEDLINE | ID: mdl-34847838

ABSTRACT

Homoharringtonine (HHT), an Food and Drug Administration (FDA)-approved anti-leukemia drug, exerts anti-tumor activity in several solid tumors, including colorectal cancer (CRC). However, its mechanism of action in CRC progression has not been comprehensively elucidated. The drug-disease targets were obtained using publicly available databases. Protein-protein interaction (PPI) network, Gene ontology (GO) function and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment were performed to reveal the core targets, biological processes and signaling pathways of HHT against CRC. Cell and animal experiments were performed to validate the inhibitory effects of HHT on CRC. A total of 98 overlapping target genes of HHT and CRC were predicted. Through PPI network and topology analysis, we screened out 23 hub genes. Enrichment assays showed 163 biological processes (BP), 18 cell components (CC), 35 molecular functions (MF), and 85 related pathways. Functionally, HHT inhibited CRC cell proliferation, cell cycle progression, colony formation, migration and invasion, and promoted apoptosis. HHT treatment resulted in the inactivation of PI3K/AKT/mTOR signaling in CRC cells. Moreover, activation of PI3K/AKT/mTOR signaling by 740Y-P abated the suppressive effects of HHT on cell malignant phenotypes. Furthermore, HHT repressed CRC tumor growth in nude mice. Our current study demonstrated that HHT repressed CRC progression at least partly by inactivating PI3K/AKT/mTOR signaling pathways, highlighting HHT as a potential therapeutic agent for CRC patients.


Subject(s)
Colorectal Neoplasms/pathology , Homoharringtonine/pharmacology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Disease Progression , Gene Ontology , Homoharringtonine/chemistry , Humans , Male , Mice, Inbred BALB C , Mice, Nude , Models, Biological , Molecular Sequence Annotation , Neoplasm Invasiveness , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism
4.
Int J Mol Sci ; 22(20)2021 Oct 10.
Article in English | MEDLINE | ID: mdl-34681590

ABSTRACT

Lung cancer has the highest rate of incidence and mortality among all cancers. Most chemotherapeutic drugs used to treat lung cancer cause serious side effects and are susceptible to drug resistance. Therefore, exploring novel therapeutic targets for lung cancer is important. In this study, we evaluated the potential of TMEM16A as a drug target for lung cancer. Homoharringtonine (HHT) was identified as a novel natural product inhibitor of TMEM16A. Patch-clamp experiments showed that HHT inhibited TMEM16A activity in a concentration-dependent manner. HHT significantly inhibited the proliferation and migration of lung cancer cells with high TMEM16A expression but did not affect the growth of normal lung cells in the absence of TMEM16A expression. In vivo experiments showed that HHT inhibited the growth of lung tumors in mice and did not reduce their body weight. Finally, the molecular mechanism through which HHT inhibits lung cancer was explored by western blotting. The findings showed that HHT has the potential to regulate TMEM16A activity both in vitro and in vivo and could be a new lead compound for the development of anti-lung-cancer drugs.


Subject(s)
Anoctamin-1/antagonists & inhibitors , Antineoplastic Agents, Phytogenic/pharmacology , Cell Proliferation/drug effects , Homoharringtonine/pharmacology , Animals , Anoctamin-1/metabolism , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/metabolism , Antineoplastic Agents, Phytogenic/therapeutic use , Apoptosis/drug effects , Binding Sites , Cell Line, Tumor , Cell Movement/drug effects , Homoharringtonine/chemistry , Homoharringtonine/metabolism , Homoharringtonine/therapeutic use , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Mice, Inbred BALB C , Molecular Docking Simulation , Transplantation, Heterologous
5.
Molecules ; 26(10)2021 May 18.
Article in English | MEDLINE | ID: mdl-34070111

ABSTRACT

Cephalotaxine (CET) is a natural alkaloid with potent antileukemia effects. However, its underlying molecular mechanism has not been well understood. In this study, we verified that CET significantly inhibited the viability of various leukemia cells, including HL-60, NB4, Jurkat, K562, Raji and MOLT-4. RNA-sequencing and bioinformatics analysis revealed that CET causes mitochondrial function change. Mechanism research indicated that CET activated the mitochondrial apoptosis pathway by reducing the mitochondrial membrane potential, downregulating anti-apoptotic Bcl-2 protein and upregulating pro-apoptotic Bak protein. In addition, the autophagy signaling pathway was highly enriched by RNA-seq analysis. Then, we found that CET blocked the fluorescence colocation of MitoTracker Green and LysoTracker Red and upregulated the level of LC3-II and p62, which indicated that autophagy flow was impaired. Further results demonstrated that CET could impair lysosomal acidification and block autophagy flow. Finally, inhibiting autophagy flow could aggravate apoptosis of HL-60 cells induced by CET. In summary, this study demonstrated that CET exerted antileukemia effects through activation of the mitochondria-dependent pathway and by impairing autophagy flow. Our research provides new insights into the molecular mechanisms of CET in the treatment of leukemia.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Homoharringtonine/pharmacology , Leukemia/pathology , Mitochondria/metabolism , Apoptosis/genetics , Autophagy/genetics , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Gene Expression Regulation, Leukemic/drug effects , Homeostasis/drug effects , Homoharringtonine/chemistry , Humans , Leukemia/genetics , Mitochondria/drug effects , Mitochondria/ultrastructure , Signal Transduction/drug effects , Signal Transduction/genetics , Transcriptome/drug effects , Transcriptome/genetics
6.
Drug Des Devel Ther ; 14: 4749-4763, 2020.
Article in English | MEDLINE | ID: mdl-33177810

ABSTRACT

PURPOSE: Homoharringtonine (HHT) has been used as an antileukemia agent in the clinic which processes a high-potential therapeutic efficacy against multiple myeloma (MM). In this study, we investigated the antimyeloma mechanism of HHT. METHODS: Three MM cell lines and a xenograft model were applied. Mitochondrial function was evaluated by detecting MitoTracker Green, the mtDNA copy number, mitochondrial protein and enzyme activity, the mitochondrial membrane potential and mitochondrial morphology. Mitophagy levels were assessed by monitoring autophagosomes, performing a colocalization analysis and determining the levels of related proteins. An shRNA was applied to knockdown Parkin. RESULTS: Based on the results of the in vitro experiments, HHT exerted a promising antiproliferative effect on the MM.1S, RPMI 8226 and H929 cell lines by increasing mitophagy. In addition, HHT markedly inhibited myeloma tumor growth and prolonged survival by promoting mitophagy in vivo. Furthermore, HHT treatment contributed to notable mitochondrial dysfunction and Parkin-dependent mitophagy, as evidenced by the destruction of mitochondria, the decrease in the mtDNA copy number, decrease in the Bcl-2/Bax ratio, and decrease in the levels of mitochondrial proteins and the optimal expression of Parkin and NDP52. However, the addition of rapamycin did not produce significant synergistic effect with HHT, indicating that HHT reached the threshold level to induce mitophagy. The colocalization analysis and assessment of mitochondrial function examination further confirmed that HHT triggered mitophagy and mitochondrial dysfunction. Moreover, the antiproliferative effect of HHT was reversed by an shRNA targeting Parkin, highlighting the indispensable role of Parkin-dependent mitophagy in the antimyeloma effect of HHT. CONCLUSION: HHT exerts an antimyeloma effect by inducing excess mitophagy, providing new mechanistic insights into a therapeutic strategy for MM.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Homoharringtonine/pharmacology , Ubiquitin-Protein Ligases/antagonists & inhibitors , Animals , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/isolation & purification , Cell Proliferation/drug effects , Cell Survival/drug effects , Cephalotaxus/chemistry , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Homoharringtonine/chemistry , Homoharringtonine/isolation & purification , Humans , Mice , Mice, SCID , Molecular Structure , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Structure-Activity Relationship , Tumor Cells, Cultured , Ubiquitin-Protein Ligases/metabolism
7.
Bioorg Chem ; 105: 104445, 2020 12.
Article in English | MEDLINE | ID: mdl-33197848

ABSTRACT

Rare and endangered plants (REPs) and their associated endophytes survived in unique habitats are promising sources for natural product-derived drug discovery. In this study, six new (cephaloverines A-F, 1-6, resp.) and 16 known (11-26) cephalotaxine-type alkaloids, together with three new (oliverbiflavones A-C, 7-9, resp.) and 11 known (27-37) biflavonoids were isolated and characterized from the twigs and leaves of Cephalotaxus oliveri, an endangered plant endemic to China. Meanwhile, a preliminary investigation on the secondary metabolites from a selected fungal endophyte (i.e., Alternaria alternate Y-4-2) associated with the title plant led to the isolation of 21 structurally distinct polyketides including one new dimeric xanthone (10). The new structures (1-10) with the absolute configurations were determined by detailed spectroscopic analyses, electronic circular dichroism (ECD) or Na2MoO4-induced ECD, the modified Mosher's method, and some chemical transformations. Compounds 1-4 are the first representatives of naturally occurring N-oxides of cephalotaxine esters, while compounds 7-9 have a special structural feature of having a C-methylated biflavonoid skeleton. The Cephalotaxus alkaloids with ester side-chains at C-3 (1-6, 13-22, and 26) and four biflavonoids (27-29 and 34) were found to show pronounced cytotoxicities against a small panel of human cancer cell lines (A549, NCI-H460, HL60, NCI-H929, and RPMI-8226), with IC50 values mainly ranging from 0.003 to 9.34 µM. The most potent compound, deoxyharringtonine (16), generally exhibited IC50 values less than 10 nM. The structure-activity relationship (SAR) of the aforementioned Cephalotaxus alkaloids was briefly discussed.


Subject(s)
Alternaria/drug effects , Antineoplastic Agents/isolation & purification , Biflavonoids/isolation & purification , Cephalotaxus/chemistry , Plant Leaves/chemistry , Antineoplastic Agents/pharmacology , Biflavonoids/pharmacology , Cell Line, Tumor , Drug Screening Assays, Antitumor , Endophytes , Homoharringtonine/chemistry , Humans , Molecular Structure , Polyketides/chemistry , Secondary Metabolism , Structure-Activity Relationship , Xanthones/chemistry
8.
Cell Death Dis ; 11(8): 632, 2020 08 14.
Article in English | MEDLINE | ID: mdl-32801343

ABSTRACT

Overexpressed EphB4 conduce to tumor development and is regarded as a potential anticancer target. Homoharringtonine (HHT) has been approved for hematologic malignancies treatment, but its effect on hepatocellular carcinoma (HCC) has not been studied. This study elucidated HHT could restrain the proliferation and migration of HCC via an EphB4/ß-catenin-dependent manner. We found that the antiproliferative activity of HHT in HCC cells and tumor xenograft was closely related to EphB4 expression. In HepG2, Hep3B and SMMC-7721 cells, EphB4 overexpression or EphrinB2 Fc stimulation augmented HHT-induced inhibitory effect on cell growth and migration ability, and such effect was abrogated when EphB4 was knocked down. The similar growth inhibitory effect of HHT was observed in SMMC-7721 and EphB4+/SMMC-7721 cells xenograft in vivo. Preliminary mechanistic investigation indicated that HHT directly bound to EphB4 and suppressed its expression. Data obtained from HCC patients revealed increased ß-catenin expression and a positive correlation between EphB4 expression and ß-catenin levels. HHT-induced EphB4 suppression promoted the phosphorylation and loss of ß-catenin, which triggered regulation of ß-catenin downstream signaling related to migration, resulting in the reversion of EMT in TGF-ß-induced HepG2 cells. Collectively, this study provided a groundwork for HHT as an effective antitumor agent for HCC in an EphB4/ß-catenin-dependent manner.


Subject(s)
Carcinoma, Hepatocellular/pathology , Cell Movement/drug effects , Homoharringtonine/pharmacology , Liver Neoplasms/pathology , Receptor, EphB4/metabolism , beta Catenin/metabolism , Animals , Cadherins/metabolism , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Epithelial-Mesenchymal Transition/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Hep G2 Cells , Homoharringtonine/chemistry , Humans , Male , Mice, Nude , Phosphorylation/drug effects , Protein Transport/drug effects , Transforming Growth Factor beta/pharmacology
9.
J Mater Chem B ; 8(4): 655-665, 2020 01 28.
Article in English | MEDLINE | ID: mdl-31904073

ABSTRACT

Stimuli response or controlled release is a new research hotspot in nanomedicine; however, there is scarce research on organic nanomedicines with stimuli responses, which limits their practical biological applications. In addition, homoharringtonine (HHT) has been used as an effective anticancer agent, but reducing its toxicity and side effects is an urgent problem to be solved. Herein, an EGFR (epidermal growth factor receptor) aptamer-modified HHT-loaded PLGA-SS-PEG nanomedicine was developed. The nanomaterial possesses spherical morphology and admirable biocompatibility. After targeted endocytosis in tumour cells via the selective recognition between EGFR and its aptamer, the PLGA nanomedicine is triggered by a high GSH level and releases its cargo in lung cancer cells. The in vitro and in vivo results reveal that the PLGA nanomedicine not only inhibited the proliferation and promoted the apoptosis of lung cancer cells, but also possessed better therapeutic efficacy and less toxic side effects compared with the free anticancer agent. Consequently, this study provides a novel approach to construct a biodegradable nanomedicine with targeted recognition and stimuli response. Moreover, it inhibited the proliferation of lung cancer cells with high efficiency and low toxicity. Importantly, the PLGA nanomedicine demonstrates encouraging potential as a multifunctional nano-system applicable for cancer therapy.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Drug Delivery Systems , Glutathione/antagonists & inhibitors , Homoharringtonine/pharmacology , Lung Neoplasms/drug therapy , Nanomedicine , Polyglactin 910/chemistry , Antineoplastic Agents, Phytogenic/chemistry , Aptamers, Nucleotide/chemistry , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Drug Screening Assays, Antitumor , Glutathione/metabolism , Homoharringtonine/chemistry , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Materials Testing , Molecular Structure , Particle Size , Polyethylene Glycols/chemistry , Surface Properties
10.
J Pharm Biomed Anal ; 176: 112801, 2019 Nov 30.
Article in English | MEDLINE | ID: mdl-31430625

ABSTRACT

A rapid ultrahigh-performance liquid chromatography coupled with quadrupole time-of-flight mass spectrometry (UHPLC-Q-TOF-MS) method was developed for separation and characterization of the degradation products in HHT injection. Chromatographic separation was achieved on a ZORBAX Eclipse XDB-C18 HD column (2.1 × 100 mm, 1.8µm) using methanol- ammonium formate (pH 3.0; 30 mM) (30:70, v/v) as mobile phase in an isocratic mode of elution. Forced degradation studies were conducted under hydrolytic (acidic and alkaline), oxidative, photolytic and thermal stress conditions as described in ICH. A total of eleven forced degradation products were detected and the drug was found to be susceptible to all the tested stress conditions. The degradation products were characterized through Q-TOF fragmentation studies and their fragmentation pathways were proposed. Seven of them have not been reported or described as degradation product before, and one of them was further confirmed by reference substance. In addition, plausible mechanisms for the formation of the degradation products were proposed.


Subject(s)
Antineoplastic Agents, Phytogenic/chemistry , Chemistry, Pharmaceutical/methods , Homoharringtonine/chemistry , Chromatography, High Pressure Liquid/methods , Drug Stability , Hydrolysis , Oxidation-Reduction , Spectrometry, Mass, Electrospray Ionization/methods , Tandem Mass Spectrometry/methods
11.
Molecules ; 24(10)2019 May 26.
Article in English | MEDLINE | ID: mdl-31130671

ABSTRACT

The increasing prevalence of drug resistant and/or high-risk cancers indicate further drug discovery research is required to improve patient outcome. This study outlines a simplified approach to identify lead compounds from natural products against several cancer cell lines, and provides the basis to better understand structure activity relationship of the natural product cephalotaxine. Using high-throughput screening, a natural product library containing fractions and pure compounds was interrogated for proliferation inhibition in acute lymphoblastic leukemia cellular models (SUP-B15 and KOPN-8). Initial hits were verified in control and counter screens, and those with EC50 values ranging from nanomolar to low micromolar were further characterized via mass spectrometry, NMR, and cytotoxicity measurements. Most of the active compounds were alkaloid natural products including cephalotaxine and homoharringtonine, which were validated as protein synthesis inhibitors with significant potency against several cancer cell lines. A generated BODIPY-cephalotaxine probe provides insight into the mode of action of cephalotaxine and further rationale for its weaker potency when compared to homoharringtonine. The steroidal natural products (ecdysone and muristerone A) also showed modest biological activity and protein synthesis inhibition. Altogether, these findings demonstrate that natural products continue to provide insight into structure and function of molecules with therapeutic potential against drug resistant cancer cell models.


Subject(s)
Biological Products/pharmacology , Cytostatic Agents/chemistry , Cytostatic Agents/pharmacology , Animals , Apoptosis/drug effects , Biological Products/chemistry , Cell Cycle Checkpoints/drug effects , Cell Survival/drug effects , Drug Discovery , Homoharringtonine/chemistry , Homoharringtonine/pharmacology , Humans , Magnetic Resonance Spectroscopy , Molecular Structure , Structure-Activity Relationship
12.
J Med Virol ; 91(9): 1595-1601, 2019 09.
Article in English | MEDLINE | ID: mdl-31032977

ABSTRACT

Foot-and-mouth disease (FMD) is a highly contagious disease that affects cloven-hoof animals including cattle, swine, sheep, goats, and lots of wild species. Effectively control measures are urged needed. Here, we showed that homoharringtonine treatment exhibited a strong inhibitory effect against two different strains of FMDVs (O/MYA98/BY/2010 and A/GD/MM/2013) in swine kidney (IBRS-2) cells. Further experiments demonstrated that homoharringtonine did not affect virus attachment or entry. Using time-of-addition assays, we found that the antiviral activity of homoharringtonine occurred primarily during the early stage of infection. These results demonstrated that homoharringtonine might be an effective anti-FMDV drug. Further studies are required to explore the antiviral activity of homoharringtonine against FMDV replication in vivo.


Subject(s)
Antiviral Agents/pharmacology , Foot-and-Mouth Disease Virus/drug effects , Foot-and-Mouth Disease/virology , Homoharringtonine/pharmacology , Animals , Antiviral Agents/chemistry , Cell Line , Foot-and-Mouth Disease Virus/physiology , Homoharringtonine/chemistry , Humans , Molecular Structure , Virus Internalization , Virus Replication/drug effects
13.
Proc Natl Acad Sci U S A ; 116(6): 2220-2225, 2019 02 05.
Article in English | MEDLINE | ID: mdl-30659143

ABSTRACT

Homoharringtonine (HHT), a known protein synthesis inhibitor, has an anti-myeloid leukemia effect and potentiates the therapeutic efficacy of anthracycline/cytarabine induction regimens for acute myelogenous leukemia (AML) with favorable and intermediate prognoses, especially in the t(8;21) subtype. Here we provide evidence showing that HHT inhibits the activity of leukemia-initiating cells (Lin-/Sca-1-/c-kit+; LICs) in a t(8;21) murine leukemia model and exerts a down-regulating effect on MYC pathway genes in human t(8;21) leukemia cells (Kasumi-1). We discovered that NF-κB repressing factor (NKRF) is bound directly by HHT via the second double-strand RNA-binding motif (DSRM2) domain, which is the nuclear localization signal of NKRF. A series of deletion and mutagenesis experiments mapped HHT direct binding sites to K479 and C480 amino acids in the DSRM2 domain. HHT treatment shifts NKRF from the nucleus (including nucleoli) to the cytoplasm by occupying the DSRM2 domain, strengthens the p65-NKRF interaction, and interferes with p65-p50 complex formation, thereby attenuating the transactivation activity of p65 on the MYC gene. Moreover, HHT significantly decreases the expression of KIT, a frequently mutated and/or highly expressed gene in t(8;21) AML, in concert with MYC down-regulation. Our work thus identifies a mechanism of action of HHT that is different from, but acts in concert with, the known mode of action of this compound. These results justify further clinical testing of HHT in AML.


Subject(s)
Gene Expression Regulation/drug effects , Genes, myc , Homoharringtonine/pharmacology , Repressor Proteins/metabolism , Animals , Binding Sites , Biomarkers, Tumor , Cell Line, Tumor , Chromosomes, Human, Pair 21 , Chromosomes, Human, Pair 8 , Disease Models, Animal , Dose-Response Relationship, Drug , Homoharringtonine/chemistry , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Mice , Protein Binding , Protein Interaction Domains and Motifs , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/metabolism , Repressor Proteins/chemistry , Transcription Factor RelA/metabolism , Transcription, Genetic , Translocation, Genetic , Xenograft Model Antitumor Assays
14.
J Microbiol ; 57(1): 74-79, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30456755

ABSTRACT

Harringtonine (HT) and homoharringtonine (HHT), alkaloid esters isolated from the genus Cephalotaxus, exhibit antitumor activity. A semisynthetic HHT has been approved for treatment of chronic myelogenous leukemia. In addition to antileukemic activity, HT and HHT are reported to possess potent antiviral activity. In this study, we investigated the effects of HT and HHT on replication of varicella-zoster virus (VZV) in vitro. HT and HHT, but not their biologically inactive parental alkaloid cephalotaxine (CET), significantly inhibited replication of recombinant VZV-pOka luciferase. Furthermore, HT and HHT, but not CET, strongly induced down-regulation of VZV lytic genes and exerted potent antiviral effects against a VZV clinical isolate. The collective data support the utility of HT and HHT as effective antiviral candidates for treatment of VZV-associated diseases.


Subject(s)
Antiviral Agents/pharmacology , Cephalotaxus/chemistry , Esters/pharmacology , Herpesvirus 3, Human/drug effects , Homoharringtonine/pharmacology , Plant Extracts/pharmacology , Antiviral Agents/chemistry , Esters/chemistry , Harringtonines/chemistry , Harringtonines/pharmacology , Herpesvirus 3, Human/genetics , Herpesvirus 3, Human/physiology , Homoharringtonine/chemistry , Humans , Plant Extracts/chemistry , Virus Replication/drug effects
15.
Viruses ; 10(11)2018 11 01.
Article in English | MEDLINE | ID: mdl-30388805

ABSTRACT

To complement traditional antivirals, natural compounds that act via host targets and present high barriers to resistance are of increasing interest. In the work reported here, we detected that homoharringtonine (HHT) presents effective antiviral activity. HHT completely inhibited infections of vesicular stomatitis virus (VSV), Newcastle disease virus (NDV), and porcine epidemic diarrhea virus (PEDV) at concentrations of 50, 100, and 500 nM in cell cultures, respectively. Treatment with HHT at doses of 0.05 or 0.2 mg/kg significantly reduced viral load and relieved severe symptoms in PEDV- or NDV-infected animals. HHT treatment, however, moderately inhibited avian influenza virus (AIV) infection, suggesting its potent antiviral action is restricted to a number of classes of RNA viruses. In this study, we also observed that HHT actively inhibited herpes simplex virus type 1 (HSV-1) replication with a 50% inhibitory concentration (IC50) of 139 nM; the treatment with HHT at 1000 nM led to reductions of three orders of magnitude. Moreover, HHT antagonized the phosphorylation level of endogenous and exogenous eukaryotic initiation factor 4E (p-eIF4E), which might regulate the selective translation of specific messenger RNA (mRNA). HHT provides a starting point for further progress toward the clinical development of broad-spectrum antivirals.


Subject(s)
Antiviral Agents/pharmacology , Biological Products/pharmacology , Homoharringtonine/pharmacology , Virus Replication/drug effects , Animals , Antiviral Agents/chemistry , Biological Products/chemistry , Cells, Cultured , Chick Embryo , Dose-Response Relationship, Drug , Homoharringtonine/chemistry , Humans , Microbial Sensitivity Tests , Molecular Structure , Swine , Transcription Factors/metabolism , Viral Load , Viral Plaque Assay , Virus Physiological Phenomena/drug effects , Viruses/drug effects
16.
J Org Chem ; 83(18): 11318-11322, 2018 09 21.
Article in English | MEDLINE | ID: mdl-30015484

ABSTRACT

A short formal synthesis of ent-Cephalotaxine is achieved. The approach features a new Lewis acid-mediated [2,3]-Stevens rearrangement of N-allylated prolineamide to generate a key quaternary stereogenic center. Additionally, a one-pot Parham-aldol sequence was developed to rapidly assemble two of the four rings in the cephalotaxine core.


Subject(s)
Homoharringtonine/chemistry , Homoharringtonine/chemical synthesis , Amides/chemistry , Chemistry Techniques, Synthetic , Cyclization , Lewis Acids/chemistry , Stereoisomerism
SELECTION OF CITATIONS
SEARCH DETAIL