Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 10 de 10
1.
Mol Biomed ; 4(1): 32, 2023 Oct 09.
Article En | MEDLINE | ID: mdl-37806992

The proper functioning of diverse biological systems depends on the spatial organization of their cells, a critical factor for biological processes like shaping intricate tissue functions and precisely determining cell fate. Nonetheless, conventional bulk or single-cell RNA sequencing methods were incapable of simultaneously capturing both gene expression profiles and the spatial locations of cells. Hence, a multitude of spatially resolved technologies have emerged, offering a novel dimension for investigating regional gene expression, spatial domains, and interactions between cells. Spatial transcriptomics (ST) is a method that maps gene expression in tissue while preserving spatial information. It can reveal cellular heterogeneity, spatial organization and functional interactions in complex biological systems. ST can also complement and integrate with other omics methods to provide a more comprehensive and holistic view of biological systems at multiple levels of resolution. Since the advent of ST, new methods offering higher throughput and resolution have become available, holding significant potential to expedite fresh insights into comprehending biological complexity. Consequently, a rapid increase in associated research has occurred, using these technologies to unravel the spatial complexity during developmental processes or disease conditions. In this review, we summarize the recent advancement of ST in historical, technical, and application contexts. We compare different types of ST methods based on their principles and workflows, and present the bioinformatics tools for analyzing and integrating ST data with other modalities. We also highlight the applications of ST in various domains of biomedical research, especially development and diseases. Finally, we discuss the current limitations and challenges in the field, and propose the future directions of ST.

2.
Nat Cancer ; 4(8): 1176-1192, 2023 08.
Article En | MEDLINE | ID: mdl-37537298

Detecting and targeting precancerous cells in noncancerous tissues is a major challenge for cancer prevention. Massive stabilization of mutant p53 (mutp53) proteins is a cancer-specific event that could potentially mark precancerous cells, yet in vivo protein-level mutp53 reporters are lacking. Here we developed two transgenic protein-level mutp53 reporters, p53R172H-Akaluc and p53-mCherry, that faithfully mimic the dynamics and function of mutp53 proteins in vivo. Using these reporters, we identified and traced rare precancerous clones in deep noncancerous tissues in various cancer models. In classic mutp53-driven thymic lymphoma models, we found that precancerous clones exhibit broad chromosome number variations, upregulate precancerous stage-specific genes such as Ybx3 and enhance amino acid transport and metabolism. Inhibiting amino acid transporters downstream of Ybx3 at the early but not late stage effectively suppresses tumorigenesis and prolongs survival. Together, these protein-level mutp53 reporters reveal undercharacterized features and vulnerabilities of precancerous cells during early tumorigenesis, paving the way for precision cancer prevention.


Precancerous Conditions , Tumor Suppressor Protein p53 , Humans , Tumor Suppressor Protein p53/genetics , Carcinogenesis/genetics , Clone Cells/metabolism , Precancerous Conditions/genetics
3.
J Med Chem ; 66(17): 12069-12100, 2023 09 14.
Article En | MEDLINE | ID: mdl-37616488

Poly(ADP-ribose) polymerase (PARP) inhibitors have been approved for the treatment of breast cancer (BC) with breast cancer susceptibility (BRCA) gene mutation. Leveraging new synthetic lethal interactions may be an effective way to broaden the indication of PARP inhibitors for BC patients with wild-type BRCA. Vascular endothelial growth factor receptor (VEGFR)-mediated suppression of angiogenesis has been reported to improve the sensitivity of wild-type BRCA cells to PARP inhibitors through synthetic lethality. Herein, we reported the conjugation of a PARP inhibitor with a VEGFR inhibitor pharmacophore to construct dual VEGFR and PARP inhibitors. The most potent compound 14b is identified to exert promising activities against VEGFR and PARP in the nanomolar range and possesses significant in vitro and in vivo antitumor and antimetastasis features. It also presented a favorable pharmacokinetic characteristics in rats with an oral bioavailability of 60.1%. Collectively, 14b may be a promising therapeutic agent of BRCA wild-type BC.


Neoplasms , Poly(ADP-ribose) Polymerases , Animals , Rats , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Vascular Endothelial Growth Factor A , Receptors, Vascular Endothelial Growth Factor
4.
J Med Chem ; 65(9): 6803-6825, 2022 05 12.
Article En | MEDLINE | ID: mdl-35442700

The effective potency and resistance of poly(ADP-ribose) polymerase (PARP) inhibitors limit their application. Here, we exploit a new paradigm that mimics the effects of breast cancer susceptibility genes (BRCA) mutations to trigger the possibility of synthetic lethality, based on the previous discovery of a potential synthetic lethality effect between bromodomain-containing protein 4 (BRD4) and PARP1. Consequently, the present study describes compound BP44 with high selectivity for BRD4 and PARP1. Fortunately, BP44 inhibits the homologous recombination in triple-negative breast cancer (TNBC) and triggers synthetic lethality, thus leading to cell cycle arrest and DNA damage. In conclusion, we optimized the BRD4-PARP1 inhibitor based on previous studies, and we expect it to become a candidate drug for the treatment of TNBC in the future. This strategy aims to expand the use of PARPi in BRCA-competent TNBC, making an innovative approach to address unmet oncology needs.


Antineoplastic Agents , Triple Negative Breast Neoplasms , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , BRCA1 Protein/genetics , Cell Cycle Proteins/genetics , Cell Line, Tumor , Homologous Recombination , Humans , Nuclear Proteins/genetics , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Poly(ADP-ribose) Polymerases/genetics , Quinazolinones , Synthetic Lethal Mutations , Transcription Factors/genetics , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/metabolism
5.
J Med Chem ; 65(7): 5184-5211, 2022 04 14.
Article En | MEDLINE | ID: mdl-35324195

Blocking the interactions between bromodomain and extraterminal (BET) proteins and acetylated lysines of histones by small molecules has important implications for the treatment of cancers and other diseases. Many pan-BET inhibitors have shown satisfactory results in clinical trials, but their potential for poor tolerability and toxicity persist. However, recently reported studies illustrate that some BET bromodomain (BET-BD1 or BET-BD2)-selective inhibitors have advantage over pan-inhibitors, including reduced toxicity concerns. Furthermore, some selective BET inhibitors have similar or even better therapeutic efficacy in inflammatory diseases or cancers. Therefore, the development of selective BET inhibitors has become a hot spot for medicinal chemists. Here, we summarize the known selective BET-BD1 and BET-BD2 inhibitors and review the methods for enhancing the selectivity and potency of these inhibitors based on their different modes of interactions with BET-BD1 or BET-BD2. Finally, we discuss prospective strategies that selectively target the bromodomains of BET proteins.


Neoplasms , Transcription Factors , Drug Discovery , Histones/metabolism , Humans , Prospective Studies , Protein Domains , Transcription Factors/metabolism
6.
J Med Chem ; 64(16): 12022-12048, 2021 08 26.
Article En | MEDLINE | ID: mdl-34351741

We herein report the identification, structural optimization, and structure-activity relationship of thieno[2,3-d]pyrimidine derivatives as a novel kind of selective vascular endothelial growth factor receptor 3 (VEGFR3) inhibitors. N-(4-Chloro-3-(trifluoromethyl)phenyl)-4-(6-(4-(4-methylpiperazin-1-yl)phenyl)thieno[2,3-d]pyrimidin-4-yl)piperazine-1-carboxamide (38k) was the most potent VEGFR3 inhibitor (IC50 = 110.4 nM) among developed compounds. Compared with VEGFR1 and VEGFR2, VEGFR3 was approximately 100 times more selective. Here, compound 38k significantly inhibited proliferation and migration of VEGF-C-induced human dermal lymphatic endothelial cells (HDLEC), MDA-MB-231, and MDA-MB-436 cells by inactivating the VEGFR3 signaling pathway. Additionally, 38k induced cell apoptosis and a prolonged G1/S-phase in MDA-MB-231 and MDA-MB-436 cells. It also presented acceptable pharmacokinetic characteristics in Sprague-Dawley (SD) rats with an oral bioavailability of 30.9%. In the xenograft model in vivo, 38k effectively inhibited breast cancer growth by suppressing the VEGFR3 signaling pathway. 38k pronouncedly resisted the formation of pulmonary metastatic nodules in mice. Collectively, 38k may be a promising therapeutic agent of metastatic breast cancer.


Antineoplastic Agents/therapeutic use , Pyrimidines/therapeutic use , Thiophenes/therapeutic use , Triple Negative Breast Neoplasms/drug therapy , Vascular Endothelial Growth Factor Receptor-3/antagonists & inhibitors , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacokinetics , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Female , G1 Phase Cell Cycle Checkpoints/drug effects , Humans , Male , Mice, Nude , Molecular Docking Simulation , Molecular Structure , Pyrimidines/chemical synthesis , Pyrimidines/metabolism , Pyrimidines/pharmacokinetics , Rats, Sprague-Dawley , Signal Transduction/drug effects , Structure-Activity Relationship , Thiophenes/chemical synthesis , Thiophenes/metabolism , Thiophenes/pharmacokinetics , Triple Negative Breast Neoplasms/pathology , Vascular Endothelial Growth Factor Receptor-3/metabolism , Xenograft Model Antitumor Assays
7.
J Med Chem ; 64(16): 11798-11815, 2021 08 26.
Article En | MEDLINE | ID: mdl-34378389

Existing evidence has demonstrated that epigenetic modifications (including DNA methylation, histone modifications, and microRNAs), which are associated with the occurrence and development of tumors, can directly or indirectly regulate autophagy. In particular, nuclear events induced by several epigenetic regulators can regulate the autophagic process and expression levels of tumor-associated genes, thereby promoting tumor progression. Tumor-associated microRNAs, including oncogenic and tumor-suppressive microRNAs, are of great significance to autophagy during tumor progression. Targeting autophagy with emerging epigenetic drugs is expected to be a promising therapeutic strategy for human tumors. From this perspective, we aim to summarize the role of epigenetic modification in the autophagic process and the underlying molecular mechanisms of tumorigenesis. Furthermore, the regulatory efficacy of epigenetic drugs on the autophagic process in tumors is also summarized. This perspective may provide a theoretical basis for the combined treatment of epigenetic drugs/autophagy mediators in tumors.


Antineoplastic Agents/therapeutic use , Autophagy/drug effects , Enzyme Inhibitors/therapeutic use , Epigenesis, Genetic/drug effects , Neoplasms/drug therapy , Acetylation/drug effects , Antineoplastic Agents/pharmacology , Cell Line, Tumor , DNA/metabolism , DNA Methylation/drug effects , Drug Discovery , Enzyme Inhibitors/pharmacology , Histones/metabolism , Humans , Methyltransferases/antagonists & inhibitors , Neoplasms/metabolism , Protein Processing, Post-Translational/drug effects
8.
Eur J Med Chem ; 209: 112917, 2021 Jan 01.
Article En | MEDLINE | ID: mdl-33077263

Atg4, a pivotal macroautophagy/autophagy-related cysteine protein family, which regulate autophagy through either cleaving Atg8 homologs for its further lipidation or delipidating Atg8 homologs from the autophagosome. There are four homologs, Atg4A, Atg4B, Atg4C, and Atg4D. Among them, an increasing amount of evidence indicates that Atg4B possessed superior catalytic efficiency toward the Atg8 substrate, as well as regulates autophagy process and plays a key role in the development of several human cancers. Recently, efforts have been contributed to the exploration of Atg4B inhibitors or activators. In this review, we comprehensively clarify the function of Atg4B in autophagy and cancer biology, as well as the relationship between pharmacological function and structure-activity of small molecule drugs targeting Atg4B. The development of novel drugs targeting Atg4B could be well applied in the clinical practice.


Antineoplastic Agents/chemistry , Autophagy-Related Proteins/antagonists & inhibitors , Cysteine Proteinase Inhibitors/chemistry , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Autophagy/drug effects , Chloroquine/pharmacology , Cysteine Endopeptidases , Cysteine Proteinase Inhibitors/pharmacology , Drug Screening Assays, Antitumor , Drug Therapy, Combination , Humans , Imidazoles/pharmacology , Models, Molecular , Molecular Targeted Therapy , Protein Binding , Structure-Activity Relationship , Substrate Specificity
9.
Eur J Med Chem ; 204: 112505, 2020 Oct 15.
Article En | MEDLINE | ID: mdl-32717479

Eukaryotic elongation factor 2 kinase (eEF2K) is a key α-kinase that negatively regulates the extension step of protein synthesis, which consumes most of the energy and amino acids required for protein synthesis. Studies have found that eEF2K protein is related to the breast cancer. However, existing inhibitor effect has not achieved the desired effect in cancer therapy. Proteolysis target chimeric (PROTAC) technology is uses proteasome to degrade target protein to achieve the purpose of inhibiting tumour cell growth. Here, we reported that the use of PROTAC strategy in combining with star eEF2K inhibitor A484954 and its potential derivatives. Consequently, candidate compound 11l was found to degrade eEF2K and induce apoptosis in human breast carcinoma MDA-MB-231 cells. Together, these findings demonstrate that our eEF2K-targeting PROTAC small molecule would be a potential new strategy for future breast cancer therapy.


Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Drug Design , Elongation Factor 2 Kinase/metabolism , Proteolysis/drug effects , Small Molecule Libraries/pharmacology , Cell Line, Tumor , Humans
10.
Mikrochim Acta ; 187(1): 16, 2019 12 05.
Article En | MEDLINE | ID: mdl-31802232

A method is described for modulating the anodic photoelectrochemistry of netlike CdS quantum dots through the deposition and dissolution of the electron acceptor manganese dioxide (MnO2) on the surface of the photoelectrode. Specifically, the photocurrent of a CdS-modified indium tin oxide (ITO/CdS) electrode is inhibited by chemical deposition of MnO2. However, the photocurrent becomes recovered by oxidative removal of MnO2 with H2O2. This deposition-dissolution reaction modulates the photoelectrochemistry of CdS effectively. A bioassay for Escherichia coli (E. coli) O157:H7 is designed that uses the antimicrobial peptide magainin I as the recognition element. Glucose oxidase (GOx) acts as a catalytic label tracer to produce the signaling molecule H2O2 in the microwell plates. The enzymatically generated H2O2 etches the deposited MnO2 on the photoelectrode and thus enhances the photocurrent. This detection scheme does not cause any damage to biomolecules. It also avoids the adverse effects of immobilized biomolecules for retarding signal production and leads to improved detection when compared to conventional PEC configurations. E. coli can be detected in the 10 to 5.0 × 106 CFU·mL-1 concentration range, and the limit of detection is 3 CFU·mL-1. Graphical abstractSchematic representation of the photoelectrochemical assay of E. coli through the deposition and dissolution of electron accepting manganese dioxide (MnO2) on the surface of the photoelectrode.

...