Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.283
Filter
1.
Eur J Med Chem ; 271: 116444, 2024 May 05.
Article in English | MEDLINE | ID: mdl-38691889

ABSTRACT

The NAPRT-induced increase in NAD+ levels was proposed as a mechanism contributing to hepatocellular carcinoma (HCC) resistance to NAMPT inhibitors. Thus, concurrently targeting NAMPT and NAPRT could be considered to overcome drug resistance. A BRD4 inhibitor downregulates the expression of NAPRT in HCC, and the combination of NAMPT inhibitors with BRD4 inhibitors simultaneously blocks NAD+ generation via salvage and the PH synthesis pathway. Moreover, the combination of the two agents significantly downregulated the expression of tumor-promoting genes and strongly promoted apoptosis. The present work identified various NAMPT/BRD4 dual inhibitors based on the multitargeted drug rationale. Among them, compound A2, which demonstrated the strongest effect, exhibited potent inhibition of NAMPT and BRD4 (IC50 = 35 and 58 nM, respectively). It significantly suppressed the growth and migration of HCC cells and facilitated their apoptosis. Furthermore, compound A2 also manifested a robust anticancer effect in HCCLM3 xenograft mouse models, with no apparent toxic effects. Our findings in this study provide an effective approach to target NAD+ metabolism for HCC treatment.


Subject(s)
Antineoplastic Agents , Apoptosis , Carcinoma, Hepatocellular , Cell Cycle Proteins , Cell Proliferation , Cytokines , Liver Neoplasms , Nicotinamide Phosphoribosyltransferase , Transcription Factors , Nicotinamide Phosphoribosyltransferase/antagonists & inhibitors , Nicotinamide Phosphoribosyltransferase/metabolism , Humans , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Liver Neoplasms/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Cell Proliferation/drug effects , Mice , Apoptosis/drug effects , Structure-Activity Relationship , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/metabolism , Cytokines/metabolism , Cytokines/antagonists & inhibitors , Drug Discovery , Drug Screening Assays, Antitumor , Molecular Structure , Dose-Response Relationship, Drug , Mice, Nude , Cell Line, Tumor , Mice, Inbred BALB C , Bromodomain Containing Proteins
2.
Eur J Med Chem ; 271: 116416, 2024 May 05.
Article in English | MEDLINE | ID: mdl-38657480

ABSTRACT

Targeting polo-box domain (PBD) small molecule for polo-like kinase 1 (PLK1) inhibition is a viable alternative to target kinase domain (KD), which could avoid pan-selectivity and dose-limiting toxicity of ATP-competitive inhibitors. However, their efficacy in these settings is still low and inaccessible to clinical requirement. Herein, we utilized a structure-based high-throughput virtual screen to find novel chemical scaffold capable of inhibiting PLK1 via targeting PBD and identified an initial hit molecule compound 1a. Based on the lead compound 1a, a structural optimization approach was carried out and several series of derivatives with naphthalimide structural motif were synthesized. Compound 4Bb was identified as a new potent PLK1 inhibitor with a KD value of 0.29 µM. 4Bb could target PLK1 PBD to inhibit PLK1 activity and subsequently suppress the interaction of PLK1 with protein regulator of cytokinesis 1 (PRC1), finally leading to mitotic catastrophe in drug-resistant lung cancer cells. Furthermore, 4Bb could undergo nucleophilic substitution with the thiol group of glutathione (GSH) to disturb the redox homeostasis through exhausting GSH. By regulating cell cycle machinery and increasing cellular oxidative stress, 4Bb exhibited potent cytotoxicity to multiple cancer cells and drug-resistant cancer cells. Subcutaneous and oral administration of 4Bb could effectively inhibit the growth of drug-resistant tumors in vivo, doubling the survival time of tumor bearing mice without side effects in normal tissues. Thus, our study offers an orally-available, structurally-novel PLK1 inhibitor for drug-resistant lung cancer therapy.


Subject(s)
Antineoplastic Agents , Cell Cycle Proteins , Cell Proliferation , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Lung Neoplasms , Naphthalimides , Polo-Like Kinase 1 , Protein Kinase Inhibitors , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins , Naphthalimides/chemistry , Naphthalimides/pharmacology , Naphthalimides/chemical synthesis , Humans , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/chemical synthesis , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Animals , Structure-Activity Relationship , Mice , Molecular Structure , Drug Resistance, Neoplasm/drug effects , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Cell Line, Tumor , Mice, Nude , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Neoplasms, Experimental/metabolism
3.
Molecules ; 29(8)2024 Apr 19.
Article in English | MEDLINE | ID: mdl-38675678

ABSTRACT

Bromodomain 4 and 9 (BRD4 and BRD9) have been regarded as important targets of drug designs in regard to the treatment of multiple diseases. In our current study, molecular dynamics (MD) simulations, deep learning (DL) and binding free energy calculations are integrated to probe the binding modes of three inhibitors (H1B, JQ1 and TVU) to BRD4 and BRD9. The MD trajectory-based DL successfully identify significant functional function domains, such as BC-loop and ZA-loop. The information from the post-processing analysis of MD simulations indicates that inhibitor binding highly influences the structural flexibility and dynamic behavior of BRD4 and BRD9. The results of the MM-GBSA calculations not only suggest that the binding ability of H1B, JQ1 and TVU to BRD9 are stronger than to BRD4, but they also verify that van der Walls interactions are the primary forces responsible for inhibitor binding. The hot spots of BRD4 and BRD9 revealed by residue-based free energy estimation provide target sites of drug design in regard to BRD4 and BRD9. This work is anticipated to provide useful theoretical aids for the development of selective inhibitors over BRD family members.


Subject(s)
Bromodomain Containing Proteins , Cell Cycle Proteins , Deep Learning , Molecular Dynamics Simulation , Protein Binding , Transcription Factors , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism , Transcription Factors/chemistry , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , Humans , Binding Sites , Thermodynamics , Triazoles/chemistry , Triazoles/pharmacology , Azepines/chemistry , Azepines/pharmacology , Nuclear Proteins/metabolism , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/chemistry , Molecular Docking Simulation
4.
Eur J Pharmacol ; 972: 176558, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38614382

ABSTRACT

Inhibitors of polo-like kinase (PLK) are currently being evaluated as anticancer drugs. However, the molecular mechanism of PLK inhibitor-induced cell death is not fully understood. In this study, we found that GW843682X and BI2536, two inhibitors of PLK1, significantly induced cell death in multiple type cells. The induction of cell death was related to the preferring expression of PLK1. However, in human umbilical vascular endothelial cells (HUVEC) and human colorectal carcinoma cells, which expressed higher levels of both PLK1 and PLK2, PLK1 inhibitors induced very low levels of cell death. Clinical analysis reveals PLK1 presence in 26 of 30 NPC tumor tissues. In in vivo NPC lung metastasis nude mouse models, PLK1 inhibitors decreased NPC progress. Mechanistically, the PLK1 inhibitor did not activate p53, and the cell death was not reversed by p53 inhibition. Moreover, PLK1 inhibitor-induced cell death was PARP- and caspase-independent. Although PLK1 inhibitors induced down-regulation of calpain inhibitor calpastatin and calpain was activated by PLK1 inhibition, calpain blocking did not reverse cell death induced by PLK1 inhibitors, suggesting the non-involvement of calpain. Surprisingly, we found that PLK1 inhibitors induced the activation of proteasome, and the treatment of cells with PLK1 inhibitors reduced the levels of ubiquitinated proteins. And proteasome inhibitors reversed cell death induced by PLK1 inhibitors in various cell types in which PLK1 was preferentially expressed. Moreover, PLK1 inhibition reversed the degradation of proteins including p53, caspase 8, PARP and calpastatin. These results suggest that the activation of proteasome is critical for cell death induced by PLK1 inhibition.


Subject(s)
Cell Cycle Proteins , Cell Death , Polo-Like Kinase 1 , Proteasome Endopeptidase Complex , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins , Humans , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/metabolism , Animals , Proteasome Endopeptidase Complex/metabolism , Cell Death/drug effects , Mice , Cell Line, Tumor , Protein Kinase Inhibitors/pharmacology , Mice, Nude , Pteridines/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Calpain/antagonists & inhibitors , Calpain/metabolism , Enzyme Activation/drug effects , Xenograft Model Antitumor Assays , Tumor Suppressor Protein p53/metabolism , Antineoplastic Agents/pharmacology
5.
J Med Chem ; 67(8): 6658-6672, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38569135

ABSTRACT

BRD4 is associated with a variety of human diseases, including breast cancer. The crucial roles of amino-terminal bromodomains (BDs) of BRD4 in binding with acetylated histones to regulate oncogene expression make them promising drug targets. However, adverse events impede the development of the BD inhibitors. BRD4 adopts an extraterminal (ET) domain, which recruits proteins to drive oncogene expression. We discovered a peptide inhibitor PiET targeting the ET domain to disrupt BRD4/JMJD6 interaction, a protein complex critical in oncogene expression and breast cancer. The cell-permeable form of PiET, TAT-PiET, and PROTAC-modified TAT-PiET, TAT-PiET-PROTAC, potently inhibits the expression of BRD4/JMJD6 target genes and breast cancer cell growth. Combination therapy with TAT-PiET/TAT-PiET-PROTAC and JQ1, iJMJD6, or Fulvestrant exhibits synergistic effects. TAT-PiET or TAT-PiET-PROTAC treatment overcomes endocrine therapy resistance in ERα-positive breast cancer cells. Taken together, we demonstrated that targeting the ET domain is effective in suppressing breast cancer, providing a therapeutic avenue in the clinic.


Subject(s)
Antineoplastic Agents , Breast Neoplasms , Bromodomain Containing Proteins , Cell Cycle Proteins , Cell Proliferation , Transcription Factors , Humans , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Female , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Animals , Cell Proliferation/drug effects , Peptides/pharmacology , Peptides/chemistry , Cell Line, Tumor , Mice , Protein Domains , Mice, Nude , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/metabolism
7.
Sci Bull (Beijing) ; 69(9): 1286-1301, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38519399

ABSTRACT

Adavosertib (ADA) is a WEE1 inhibitor that exhibits a synthetic lethal effect on p53-mutated gallbladder cancer (GBC). However, drug resistance due to DNA damage response compensation pathways and high toxicity limits further applications. Herein, estrone-targeted ADA-encapsulated metal-organic frameworks (ADA@MOF-EPL) for GBC synthetic lethal treatment by inducing conditional factors are developed. The high expression of estrogen receptors in GBC enables ADA@MOF-EPL to quickly enter and accumulate near the cell nucleus through estrone-mediated endocytosis and release ADA to inhibit WEE1 upon entering the acidic tumor microenvironment. Ultrasound irradiation induces ADA@MOF-EPL to generate reactive oxygen species (ROS), which leads to a further increase in DNA damage, resulting in a higher sensitivity of p53-mutated cancer cells to WEE1 inhibitor and promoting cell death via conditional synthetic lethality. The conditional factor induced by ADA@MOF-EPL further enhances the antitumor efficacy while significantly reducing systemic toxicity. Moreover, ADA@MOF-EPL demonstrates similar antitumor abilities in other p53-mutated solid tumors, revealing its potential as a broad-spectrum antitumor drug.


Subject(s)
Antineoplastic Agents , Gallbladder Neoplasms , Metal-Organic Frameworks , Protein-Tyrosine Kinases , Pyrimidinones , Tumor Suppressor Protein p53 , Metal-Organic Frameworks/chemistry , Metal-Organic Frameworks/pharmacology , Gallbladder Neoplasms/drug therapy , Gallbladder Neoplasms/genetics , Gallbladder Neoplasms/pathology , Humans , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Animals , Cell Line, Tumor , Protein-Tyrosine Kinases/antagonists & inhibitors , Mice , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/chemistry , Pyrazoles/pharmacology , Pyrazoles/therapeutic use , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Synthetic Lethal Mutations , Reactive Oxygen Species/metabolism , Xenograft Model Antitumor Assays , Mutation , Mice, Nude , DNA Damage/drug effects , Female
8.
Biomed Pharmacother ; 174: 116492, 2024 May.
Article in English | MEDLINE | ID: mdl-38537579

ABSTRACT

Targeting epigenetic mechanisms has emerged as a potential therapeutic approach for the treatment of kidney diseases. Specifically, inhibiting the bromodomain and extra-terminal (BET) domain proteins using the small molecule inhibitor JQ1 has shown promise in preclinical models of acute kidney injury (AKI) and chronic kidney disease (CKD). However, its clinical translation faces challenges due to issues with poor pharmacokinetics and side effects. Here, we developed engineered liposomes loaded with JQ1 with the aim of enhancing kidney drug delivery and reducing the required minimum effective dose by leveraging cargo protection. These liposomes efficiently encapsulated JQ1 in both the membrane and core, demonstrating superior therapeutic efficacy compared to freely delivered JQ1 in a mouse model of kidney ischemia-reperfusion injury. JQ1-loaded liposomes (JQ1-NPs) effectively targeted the kidneys and only one administration, one-hour after injury, was enough to decrease the immune cell (neutrophils and monocytes) infiltration to the kidney-an early and pivotal step to prevent damage progression. By inhibiting BRD4, JQ1-NPs suppress the transcription of pro-inflammatory genes, such as cytokines (il-6) and chemokines (ccl2, ccl5). This success not only improved early the kidney function, as evidenced by decreased serum levels of BUN and creatinine in JQ1-NPs-treated mice, along with reduced tissue expression of the damage marker, NGAL, but also halted the production of extracellular matrix proteins (Fsp-1, Fn-1, α-SMA and Col1a1) and the fibrosis development. In summary, this work presents a promising nanotherapeutic strategy for AKI treatment and its progression and provides new insights into renal drug delivery.


Subject(s)
Azepines , Bromodomain Containing Proteins , Disease Progression , Kidney , Liposomes , Mice, Inbred C57BL , Nuclear Proteins , Renal Insufficiency, Chronic , Reperfusion Injury , Triazoles , Animals , Azepines/pharmacology , Azepines/administration & dosage , Reperfusion Injury/drug therapy , Reperfusion Injury/pathology , Triazoles/pharmacology , Triazoles/administration & dosage , Renal Insufficiency, Chronic/drug therapy , Renal Insufficiency, Chronic/pathology , Mice , Kidney/drug effects , Kidney/pathology , Kidney/metabolism , Male , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism , Acute Kidney Injury/drug therapy , Acute Kidney Injury/prevention & control , Disease Models, Animal , Nanoparticles , Cell Cycle Proteins/antagonists & inhibitors
10.
Biochem Biophys Res Commun ; 694: 149388, 2024 Jan 29.
Article in English | MEDLINE | ID: mdl-38150917

ABSTRACT

Despite progress in the application of checkpoint immunotherapy against various tumors, attempts to utilize immune checkpoint blockade (ICB) agents in triple negative breast cancer (TNBC) have yielded limited clinical benefits. The low overall response rate of checkpoint immunotherapy in TNBC may be attributed to the immunosuppressive tumor microenvironment (TME). In this study, we investigated the role of mitogen-associated kinase TTK in reprogramming immune microenvironment in TNBC. Notably, TTK inhibition by BAY-1217389 induced DNA damage and the formation of micronuclei containing dsDNA in the cytosol, resulting in elicition of STING signal pathway and promoted antitumor immunity via the infiltration and activation of CD8+ T cells. Moreover, TTK inhibition also upregulated the expression of PD-L1, demonstrating a synergistic effect with anti-PD1 therapy in 4T1 tumor-bearing mice. Taken together, TTK inhibition facilitated anti-tumor immunity mediated by T cells and enhanced sensitivity to PD-1 blockade, providing a rationale for the combining TTK inhibitors with immune checkpoint blockade in clinical trials.


Subject(s)
CD8-Positive T-Lymphocytes , Triple Negative Breast Neoplasms , Animals , Humans , Mice , B7-H1 Antigen , Cell Cycle Proteins/antagonists & inhibitors , Cell Line, Tumor , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy , Protein Serine-Threonine Kinases , Protein-Tyrosine Kinases , Signal Transduction , Triple Negative Breast Neoplasms/drug therapy , Tumor Microenvironment
11.
Int J Mol Sci ; 24(23)2023 Dec 02.
Article in English | MEDLINE | ID: mdl-38069378

ABSTRACT

Patients receiving cranial radiotherapy for primary and metastatic brain tumors may experience radiation-induced brain injury (RIBI). Thus far, there has been a lack of effective preventive and therapeutic strategies for RIBI. Due to its complicated underlying pathogenic mechanisms, it is rather difficult to develop a single approach to target them simultaneously. We have recently reported that Reprimo (RPRM), a tumor suppressor gene, is a critical player in DNA damage repair, and RPRM deletion significantly confers radioresistance to mice. Herein, by using an RPRM knockout (KO) mouse model established in our laboratory, we found that RPRM deletion alleviated RIBI in mice via targeting its multiple underlying mechanisms. Specifically, RPRM knockout significantly reduced hippocampal DNA damage and apoptosis shortly after mice were exposed to whole-brain irradiation (WBI). For the late-delayed effect of WBI, RPRM knockout obviously ameliorated a radiation-induced decline in neurocognitive function and dramatically diminished WBI-induced neurogenesis inhibition. Moreover, RPRM KO mice exhibited a significantly lower level of acute and chronic inflammation response and microglial activation than wild-type (WT) mice post-WBI. Finally, we uncovered that RPRM knockout not only protected microglia against radiation-induced damage, thus preventing microglial activation, but also protected neurons and decreased the induction of CCL2 in neurons after irradiation, in turn attenuating the activation of microglial cells nearby through paracrine CCL2. Taken together, our results indicate that RPRM plays a crucial role in the occurrence of RIBI, suggesting that RPRM may serve as a novel potential target for the prevention and treatment of RIBI.


Subject(s)
Brain Injuries , Radiation Injuries , Animals , Humans , Mice , Apoptosis , Brain/pathology , Brain Injuries/genetics , Brain Injuries/prevention & control , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/metabolism , Glycoproteins/antagonists & inhibitors , Glycoproteins/metabolism , Inflammation/pathology , Microglia , Radiation Injuries/genetics , Radiation Injuries/prevention & control , Radiation Injuries/pathology
12.
J Med Chem ; 66(4): 2646-2662, 2023 02 23.
Article in English | MEDLINE | ID: mdl-36774555

ABSTRACT

Enhancer of zeste homolog 2 (EZH2) mediates the trimethylation of histone 3 lysine 27 (H3K27) to promote gene silencing. Inhibition of EZH2 is a viable strategy for cancer treatment; however, only a small subset of hematological malignancies are sensitive to small-molecule EZH2 inhibitors. EZH2 inhibitors cause H3K27 acetylation in most solid tumors, leading to drug resistance. Bromodomain-containing protein 4 (BRD4) inhibitors were reported to enhance the sensitivity of solid tumors to EZH2 inhibitors. Thus, we designed and evaluated a series of dual EZH2-BRD4 inhibitors. ZLD-2, the most promising compound, exhibited potent inhibitory activity against EZH2 and BRD4. Compared to the EZH2 inhibitor GSK126, ZLD-2 displayed potent antiproliferation activity against breast, lung, bladder, and pancreatic cancer cells. In vivo, ZLD-2 exhibited antitumor activity in a BxPC-3 mouse xenograft model, whereas GSK126 promoted tumor growth. Thus, ZLD-2 may be a lead compound for treating solid tumors.


Subject(s)
Antineoplastic Agents , Cell Cycle Proteins , Drug Design , Enhancer of Zeste Homolog 2 Protein , Enzyme Inhibitors , Neoplasms , Transcription Factors , Animals , Humans , Mice , Cell Cycle Proteins/antagonists & inhibitors , Cell Line, Tumor , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Histones/metabolism , Neoplasms/drug therapy , Nuclear Proteins/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology
13.
Eur J Med Chem ; 239: 114519, 2022 Sep 05.
Article in English | MEDLINE | ID: mdl-35714446

ABSTRACT

Bromodomain and extraterminal domain (BET) subfamily members are intriguing targets for cancer treatment. Most of the reported BET inhibitors were monovalent inhibitors. Recently, some bivalent inhibitors were disclosed, which bound to two bromodomains simultaneously. They had good activities, however, most of them also showed unsatisfactory pharmacokinetic properties, which were caused by long chain linkers. Based on our previous work on monovalent BRD4 inhibitors, we designed and synthesized a series of novel bivalent inhibitors with short and hydrophilic linkers. These compounds exhibited better activities than the corresponding monovalent inhibitors and good pharmacokinetic properties. Compound 21 showed excellent in vitro activities. And it also demonstrated potent in vivo antitumor efficacy under oral administration and was well tolerated in in vivo tests.


Subject(s)
Cell Cycle Proteins , Nuclear Proteins , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/metabolism , Cell Proliferation , Imidazoles , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/metabolism , Structure-Activity Relationship , Sulfonamides , Thiophenes , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism
14.
Eur J Med Chem ; 238: 114423, 2022 Aug 05.
Article in English | MEDLINE | ID: mdl-35544982

ABSTRACT

Sepsis has long been a major health problem worldwide. It threatens the lives of hospitalized patients and has been one of the leading causes of death in hospitalized patients over the past decades. BRD4 has been regarded as a potential target for sepsis therapy, for its critical role in the transcriptional expression of NF-κB pathway-dependent inflammatory factors. In this study, compound 1 was obtained through virtual screening, and candidate compound 27 was obtained through several rounds of iterative SAR analysis. 27 decreased LPS-induced NO production and expression of the pro-inflammatory factors IL-6, IL-1ß and TNF-α. In vivo, 27 effectively protected mice from LPS-induced sepsis, increased survival rate and decreased the level of pro-inflammatory factors in serum. Collectively, we reported here 27, a BRD4 inhibitor with a new scaffold, as a potential candidate for the treatment of sepsis.


Subject(s)
Cell Cycle Proteins , Nuclear Proteins , Sepsis , Transcription Factors , Animals , Cell Cycle Proteins/antagonists & inhibitors , Humans , Lipopolysaccharides , Mice , NF-kappa B/metabolism , Sepsis/drug therapy , Transcription Factors/antagonists & inhibitors
15.
Cancer Lett ; 539: 215677, 2022 07 28.
Article in English | MEDLINE | ID: mdl-35490917

ABSTRACT

MicroRNAs are noncoding RNAs with a typical length of 22 nucleotides that post-transcriptionally suppress gene expression by inducing target mRNA degradation and/or impairing translation in eukaryotes. Thousands of miRNA genes in the human genome are involved in various physiological and pathological processes. Each miRNA targets many different mRNAs, while each mRNA may be targeted by various miRNAs. Mini-chromosome maintenance (MCM2-7) protein complex functions as essential components of the pre-replicative complex (pre-RC) and forms a helicase together with other proteins to unwind the DNA duplex in S phase. MCM proteins are overexpressed in all cancer cells, while they are strictly regulated in normal cells, with no expression in non-proliferating normal cells. Here we report that miRNA-214-3p (miR-214) targets both MCM5 and MCM7. The level of miR-214 is lower in HepG2 and Hep3B hepatocellular carcinoma cells than the L-02 normal liver cells. Introduction of miRNA-214 mimic into HepG2 and Hep3B cells reduced the mRNA and protein levels of MCM5/7 and inhibited DNA replication, cell cycle progression, cell proliferation and colony formation. Comparatively, miRNA-214 mimic had little effect in L-02 cells. Importantly, miR-214 mimic can also inhibit the growth of HepG2 xenografts in nude mice. Our data suggest that miRNA-214 regulates DNA replication by targeting MCM5/7 and has the potential to be developed into a liver cancer drug. IMPLICATIONS: This study supports the notion that DNA replication-initiation proteins (DRIPs), including MCM2-7 proteins, are attractive anticancer targets. Furthermore, the potential of miR-214 as an anticancer agent, with activity against liver cancer cells but not normal livre cells, may be of high significance.


Subject(s)
Carcinoma, Hepatocellular , Cell Cycle Proteins , DNA Replication , Liver Neoplasms , MicroRNAs , Minichromosome Maintenance Complex Component 7 , Animals , Carcinogenesis/genetics , Carcinoma, Hepatocellular/genetics , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/biosynthesis , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Hep G2 Cells , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Mice , Mice, Nude , MicroRNAs/genetics , MicroRNAs/metabolism , Minichromosome Maintenance Complex Component 7/genetics , RNA, Messenger , Xenograft Model Antitumor Assays
16.
Cell Rep ; 39(7): 110814, 2022 05 17.
Article in English | MEDLINE | ID: mdl-35584676

ABSTRACT

Small cell lung cancers (SCLCs) have high mutational burden but are relatively unresponsive to immune checkpoint blockade (ICB). Using SCLC models, we demonstrate that inhibition of WEE1, a G2/M checkpoint regulator induced by DNA damage, activates the STING-TBK1-IRF3 pathway, which increases type I interferons (IFN-α and IFN-ß) and pro-inflammatory chemokines (CXCL10 and CCL5), facilitating an immune response via CD8+ cytotoxic T cell infiltration. We further show that WEE1 inhibition concomitantly activates the STAT1 pathway, increasing IFN-γ and PD-L1 expression. Consistent with these findings, combined WEE1 inhibition (AZD1775) and PD-L1 blockade causes remarkable tumor regression, activation of type I and II interferon pathways, and infiltration of cytotoxic T cells in multiple immunocompetent SCLC genetically engineered mouse models, including an aggressive model with stabilized MYC. Our study demonstrates cell-autonomous and immune-stimulating activity of WEE1 inhibition in SCLC models. Combined inhibition of WEE1 plus PD-L1 blockade represents a promising immunotherapeutic approach in SCLC.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , B7-H1 Antigen , Cell Cycle Proteins , Lung Neoplasms , Membrane Proteins , Protein-Tyrosine Kinases , STAT1 Transcription Factor , Small Cell Lung Carcinoma , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/immunology , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Drug Synergism , Immune Checkpoint Inhibitors/pharmacology , Lung Neoplasms/drug therapy , Lung Neoplasms/immunology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Membrane Proteins/metabolism , Mice , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , STAT1 Transcription Factor/metabolism , Small Cell Lung Carcinoma/drug therapy , Small Cell Lung Carcinoma/immunology , Small Cell Lung Carcinoma/metabolism , Small Cell Lung Carcinoma/pathology
17.
Biomolecules ; 12(4)2022 03 31.
Article in English | MEDLINE | ID: mdl-35454120

ABSTRACT

While Polo-like kinase 1 (PLK1) inhibitors have shown promise in clinical settings for treating triple-negative breast cancer tumors and other solid tumors, they are limited by their ability to bind non-selectively to the ATP kinase domain. Therefore, we sought to develop a PLK1 allosteric inhibitor targeting the PLK1 T-loop (a switch responsible for activation) and evaluate its effects in triple-negative breast cancer cells. A novel compound, RK-10, was developed based on an in silico model, and its effects on specificity, viability, migration, and cell cycle regulation in MCF-10A and MDA-MB 231 cells were evaluated. When MDA-MB 231 cells were treated with 0−50 µg/mL RK-10, phospho-PLK1 (Thr-210) was decreased in cells cultured adherently and cells cultured as mammospheres. RK-10 significantly inhibited viability after 24 h; however, by 48 h, 25−50 µM RK-10 caused >50% reduction. RK-10 attenuated wound healing by up to 99.7% and caused S and G2/M cell cycle arrest, which was associated with increased p21 expression. We developed a novel allosteric inhibitor which mediates anti-proliferative and anti-migratory properties through targeting phospho-PLK1 (Thr-210) in mammospheres and causing S phase and G2/M cell cycle arrest. Further development of PLK1 allosteric inhibitors may be a promising approach for TNBC treatment.


Subject(s)
Protein Serine-Threonine Kinases , Triple Negative Breast Neoplasms , Apoptosis , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation , Humans , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/metabolism , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/metabolism , Polo-Like Kinase 1
19.
Clin Cancer Res ; 28(10): 2147-2159, 2022 05 13.
Article in English | MEDLINE | ID: mdl-35302600

ABSTRACT

PURPOSE: Uterine leiomyosarcoma is among the most aggressive gynecological malignancies. No effective treatment strategies have been established. This study aimed to identify novel therapeutic targets for uterine leiomyosarcoma based on transcriptome analysis and assess the preclinical efficacy of novel drug candidates. EXPERIMENTAL DESIGN: Transcriptome analysis was performed using fresh-frozen samples of six uterine leiomyosarcomas and three myomas. The Ingenuity Pathway Analysis (IPA) was used to identify potential therapeutic target genes for uterine leiomyosarcoma. Afterward, our results were validated using three independent datasets, including 40 uterine leiomyosarcomas. Then, the inhibitory effects of several selective inhibitors for the candidate genes were examined using SK-UT-1, SK-LMS-1, and SKN cell lines. RESULTS: We identified 512 considerably dysregulated genes in uterine leiomyosarcoma compared with myoma. The IPA revealed that the function of several genes, including CHEK1 and PLK1, were predicted to be activated in uterine leiomyosarcoma. Through an in vitro drug screening, PLK1 or CHEK1 inhibitors (BI-2536 or prexasertib) were found to exert a superior anticancer effect against cell lines at low nanomolar concentrations and induce cell-cycle arrest. In SK-UT-1 tumor-bearing mice, BI-2536 monotherapy remarkably suppressed tumorigenicity. Moreover, the prexasertib and cisplatin combination therapy inhibited tumor proliferation and prolonged the time to tumor progression. CONCLUSIONS: We identified upregulated expressions of PLK1 and CHEK1; their kinase activity was activated in uterine leiomyosarcoma. BI-2536 and prexasertib demonstrated a significant anticancer effect. Therefore, cell-cycle-related kinases may present a promising therapeutic strategy for the treatment of uterine leiomyosarcoma.


Subject(s)
Cell Cycle Proteins , Checkpoint Kinase 1 , Leiomyosarcoma , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins , Uterine Neoplasms , Animals , Cell Cycle Checkpoints , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Cell Line, Tumor , Checkpoint Kinase 1/antagonists & inhibitors , Checkpoint Kinase 1/genetics , Cisplatin/therapeutic use , Female , Humans , Leiomyosarcoma/drug therapy , Leiomyosarcoma/genetics , Leiomyosarcoma/metabolism , Mice , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Uterine Neoplasms/drug therapy , Uterine Neoplasms/genetics , Uterine Neoplasms/metabolism , Polo-Like Kinase 1
20.
Biochem Biophys Res Commun ; 604: 63-69, 2022 05 14.
Article in English | MEDLINE | ID: mdl-35299072

ABSTRACT

Neuroblastoma(NB) is a common childhood solid tumor, and most patients in the high-risk group with MYCN gene amplification have a poor prognosis. Inhibition of bromodomain and extra terminal (BET) proteins has shown considerable promise in the investigation of MYCN-driven malignancies in recent years. MZ1 is a novel BET inhibitor that employs proteolytic-targeting chimera (PROTAC) technology for proteasomal degradation of target proteins and has shown excellent effects in some tumors, but its role in neuroblastoma remains poorly understood. Herein, we observed that MZ1 suppressed MYC-amplified NB cell proliferation and normal cell cycle, while simultaneously boosting cell apoptosis. MZ1 also provides a significant therapeutic impact in vivo. Mechanistically, MZ1 exhibits anti-tumor effect in NB cells by suppressing the expression of N-Myc or C-Myc as well as the MAPK signaling pathway. Overall, our data imply that MZ1 might be exploited as a possible therapeutic method for NB therapy.


Subject(s)
Cell Cycle Proteins , Dipeptides , Heterocyclic Compounds, 3-Ring , N-Myc Proto-Oncogene Protein , Neuroblastoma , Transcription Factors , Cell Cycle Proteins/antagonists & inhibitors , Cell Line, Tumor , Child , Dipeptides/pharmacology , Gene Expression Regulation, Neoplastic , Heterocyclic Compounds, 3-Ring/pharmacology , Humans , N-Myc Proto-Oncogene Protein/genetics , N-Myc Proto-Oncogene Protein/metabolism , Neuroblastoma/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...