Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Bioconjug Chem ; 35(6): 737-743, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38738511

ABSTRACT

Radiation therapy is one of the most common treatments for cancer. However, enhancing tumors' radiation sensitivity and overcoming tolerance remain a challenge. Previous studies have shown that the Ras signaling pathway directly influences tumor radiation sensitivity. Herein, we designed a series of Ras-targeting stabilized peptides, with satisfactory binding affinity (KD = 0.13 µM with HRas) and good cellular uptake. Peptide H5 inhibited downstream phosphorylation of ERK and increased radio-sensitivity in HeLa cells, resulting in significantly reduced clonogenic survival. The stabilized peptides, designed with an N-terminal nucleation strategy, acted as potential radio-sensitizers and broadened the applications of this kind of molecule. This is the first report of using stabilized peptides as radio-sensitizers, broadening the applications of this kind of molecule.


Subject(s)
Peptides , Radiation Tolerance , ras Proteins , Humans , Peptides/chemistry , Peptides/pharmacology , HeLa Cells , Radiation Tolerance/drug effects , ras Proteins/metabolism , Radiation-Sensitizing Agents/pharmacology , Radiation-Sensitizing Agents/chemistry , Cell Survival/drug effects , Phosphorylation/drug effects , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/radiotherapy
2.
ACS Chem Biol ; 19(5): 1161-1168, 2024 05 17.
Article in English | MEDLINE | ID: mdl-38662199

ABSTRACT

Targeted protein degradation is becoming more and more important in the field of drug development. Compared with proteasomal-based degraders, lysosomal-based degraders have a broader target spectrum of targets, which have been demonstrated to have great potential, especially in degrading undruggable proteins. Recently, we developed a programmable and facile screening PROTAC development platform based on peptide self-assembly termed split-and-mix PROTAC (SM-PROTAC). In this study, we applied this technology for the development of lysosome-based degraders, named a split-and-mix chaperone-mediated autophagy-based degrader (SM-CMAD). We successfully demonstrated SM-CMAD as a universal platform by degrading several targets, including ERα, AR, MEK1/2, and BCR-ABL. Different from other lysosomal-based degraders, SM-CMAD was capable of facile screening with programmable ligand ratios. We believe that our work will promote the development of other multifunctional molecules and clinical translation for lysosomal-based degraders.


Subject(s)
Lysosomes , Proteolysis , Lysosomes/metabolism , Proteolysis/drug effects , Humans , Peptides/chemistry , Peptides/metabolism , Peptides/pharmacology , Autophagy/drug effects
3.
J Am Chem Soc ; 146(4): 2624-2633, 2024 01 31.
Article in English | MEDLINE | ID: mdl-38239111

ABSTRACT

Herein, we report a versatile reaction platform for tracelessly cleavable cysteine-selective peptide/protein modification. This platform offers highly tunable and predictable conjugation and cleavage by rationally estimating the electron effect on the nucleophilic halopyridiniums. Cleavable peptide stapling, antibody conjugation, enzyme masking/de-masking, and proteome labeling were achieved based on this facile pyridinium-thiol-exchange protocol.


Subject(s)
Peptides , Proteome , Cysteine/metabolism
4.
Chem Commun (Camb) ; 60(6): 686-689, 2024 Jan 16.
Article in English | MEDLINE | ID: mdl-38054347

ABSTRACT

Covalent proteolysis-targeting chimeras (PROTACs) offer enhanced selectivity, prolonged action, and increased efficacy against challenging target proteins. The conventional approach relies on covalent ligands, but our study presents an innovative method employing an N-sulfonyl pyridone warhead to selectively target tyrosine (Tyr) residues. The von Hippel-Lindau (VHL) moiety is transferred from the warhead to the exposed Tyr, allowing us to design a STING degrader (DC50 0.53 µM, Dmax 56.65%). This approach showcases the potential of nucleophilic amino acid labeling probes, particularly for proteins lacking easily accessible cysteine residues, opening new possibilities for covalent PROTAC design and targeted protein degradation therapies.


Subject(s)
Pyridones , Ubiquitin-Protein Ligases , Ubiquitin-Protein Ligases/metabolism , Proteolysis
5.
J Med Chem ; 66(22): 15409-15423, 2023 11 23.
Article in English | MEDLINE | ID: mdl-37922441

ABSTRACT

Lysine-specific demethylase 1 (LSD1) is a promising therapeutic target, especially in cancer treatment. Despite several LSD1 inhibitors being discovered for the cofactor pocket, none are FDA-approved. We aimed to develop stabilized peptides for irreversible LSD1 binding, focusing on unique cysteine residue Cys360 in LSD1 and SNAIL1. We created LSD1 C360-targeting peptides, like cyclic peptide S9-CMC1, using our Cysteine-Methionine cyclization strategy. S9-CMC1 effectively inhibited LSD1 at the protein level, as confirmed by MS analysis showing covalent bonding to Cys360. In cells, S9-CMC1 inhibited LSD1 activity, increasing H3K4me1 and H3K4me2 levels, leading to G1 cell cycle arrest and apoptosis and inhibiting cell proliferation. Remarkably, S9-CMC1 showed therapeutic potential in A549 xenograft animal models, regulating LSD1 activity and significantly inhibiting tumor growth with minimal organ damage. These findings suggest LSD1 C360 as a promising site for covalent LSD1 inhibitors' development.


Subject(s)
Cysteine , Neoplasms , Animals , Humans , Peptides/pharmacology , Peptides, Cyclic/pharmacology , Peptides, Cyclic/therapeutic use , Cell Proliferation , Histone Demethylases/metabolism , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry , Cell Line, Tumor
6.
Nat Commun ; 14(1): 6132, 2023 10 02.
Article in English | MEDLINE | ID: mdl-37783727

ABSTRACT

Cyclic GMP-AMP synthase (cGAS) is an essential sensor of aberrant cytosolic DNA for initiating innate immunity upon invading pathogens and cellular stress, which is considered as a potential drug target for autoimmune and autoinflammatory diseases. Here, we report the discovery of a class of cyclopeptide inhibitors of cGAS identified by an in vitro screening assay from a focused library of cyclic peptides. These cyclopeptides specifically bind to the DNA binding site of cGAS and block the binding of dsDNA with cGAS, subsequently inhibit dsDNA-induced liquid phase condensation and activation of cGAS. The specificity and potency of one optimal lead XQ2B were characterized in cellular assays. Concordantly, XQ2B inhibited herpes simplex virus-1 (HSV-1)-induced antiviral immune responses and enhanced HSV-1 infection in vitro and in vivo. Furthermore, XQ2B significantly suppressed the elevated levels of type I interferon and proinflammatory cytokines in primary macrophages from Trex1-/- mice and systemic inflammation in Trex1-/- mice. XQ2B represents the specific cGAS inhibitor targeting protein-DNA interaction and phase separation and serves as a scaffold for the development of therapies in the treatment of cGAS-dependent inflammatory diseases.


Subject(s)
DNA , Peptides, Cyclic , Animals , Mice , Peptides, Cyclic/pharmacology , DNA/metabolism , Nucleotidyltransferases/metabolism , Immunity, Innate , Cytokines
7.
J Am Chem Soc ; 145(40): 21860-21870, 2023 Oct 11.
Article in English | MEDLINE | ID: mdl-37708462

ABSTRACT

Proteolysis Targeting Chimera (PROTAC) technology represents a promising new approach for target protein degradation using a cellular ubiquitin-proteasome system. Recently, we developed a split-and-mix nanoplatform based on peptide self-assembly, which could serve as a self-adjustable platform for multifunctional applications. However, the lower drug efficacy limits further biomedical applications of peptide-based SM-PROTAC. In this study, we develop a novel split-and-mix PROTAC system based on liposome self-assembly (LipoSM-PROTAC), concurrent with modification of FA (folate) to enhance its tumor-targeting capabilities. Estrogen receptors (ERα) were chosen as the protein of interest (POI) to validate the efficacy of Lipo degraders. Results demonstrate that this PROTAC can be efficiently and selectively taken up into the cells by FA receptor-positive cells (FR+) and degrade the POI with significantly reduced concentration. Compared to the peptide-based SM-PROTACs, our designed LipoSM-PROTAC system could achieve therapeutic efficacy with a lower concentration and provide opportunities for clinical translational potential. Overall, the LipoSM-based platform shows a higher drug efficacy, which offers promising potential applications for PROTAC and other biomolecule regulations.

8.
J Am Chem Soc ; 145(14): 7879-7887, 2023 04 12.
Article in English | MEDLINE | ID: mdl-37001133

ABSTRACT

The development of bifunction al molecules, which can enable targeted RNA degradation, targeted protein acetylation, or targeted protein degradation, remains a time-consuming process that requires tedious optimization. We propose a split-and-mix nanoplatform that serves as a self-adjustable platform capable of facile screening, programmable ligand ratios, self-optimized biomolecule spatial recognition, and multifunctional applications. Herein, we demonstrate the potential of our proposed nanoplatform by showcasing proteolysis-targeting chimeras (PROTACs), namely, split-and-mix PROTAC (SM-PROTAC). We highlight the scope of our platform through the targeted disruption of intracellular therapeutic targets involving ERα, CDK4/6, AR, MEK1/2, BRD2/4, BCR-ABL, etc. These studies confirm the effectiveness and universality of the SM-PROTAC platform for proximity-induced applications. This platform is programmable, with significant potential applications to biomolecule regulation, including the fields of epigenetics, gene editing, and biomolecule modification regulation.


Subject(s)
Protein Processing, Post-Translational , Proteolysis
9.
Eur J Med Chem ; 243: 114796, 2022 Dec 05.
Article in English | MEDLINE | ID: mdl-36198216

ABSTRACT

Cancer immunotherapy is a powerful weapon in the fight against cancers. Cyclic dinucleotides (CDNs) have demonstrated the great potential by evoking the immune system to fight cancers. There are still a lot of unmet needs for highly active CDNs in clinical applications due to low cell permeation and serum stability. Here we reported S-acylthioalkyl ester (SATE)-based prodrugs of deoxyribose cyclic dinucleotides (dCDNs) with three different types of internucleotide linkages (3',3':11a; 2',3':11b; 2',2':11c). The parent dCDNs could be efficiently released from SATE-dCDNs by cellular esterases. Compared to 2',3'-cGAMP and ADU-S100, 11a exhibited much higher potency of activating STING pathway and higher serum stability. In a CT26-Luc tumor-bearing animal model, 11a showed the efficient antitumor activity in eliminating the established tumor and induced significant increase of mRNA expression of IFN-ß and other related inflammatory cytokines. Hence, SATE-dCDN prodrugs demonstrated their benefits in promoting cell penetration, improving serum stability, and thus enhancing bioactivity, suggesting their potential application as immunotherapy in a variety of malignancies.


Subject(s)
Neoplasms , Prodrugs , Animals , Prodrugs/pharmacology , Deoxyribose , Esters/pharmacology , Immunotherapy , Immunologic Factors , Neoplasms/drug therapy
10.
Front Genet ; 13: 934223, 2022.
Article in English | MEDLINE | ID: mdl-36017491

ABSTRACT

N6-methyladenosine (m6A) is the most abundant internal chemical modification of eukaryotic mRNA and plays diverse roles in gene regulation. The m6A modification plays a significant role in numerous cancer types, including kidney, stomach, lung, bladder tumors, and melanoma, through varied mechanisms. As direct m6A readers, the YT521-B homology domain family proteins (YTHDFs) play a key role in tumor transcription, translation, protein synthesis, tumor stemness, epithelial-mesenchymal transition (EMT), immune escape, and chemotherapy resistance. An in-depth understanding of the molecular mechanism of YTHDFs is expected to provide new strategies for tumor treatment. In this review, we provide a systematic description of YTHDF protein structure and its function in tumor progression.

11.
Org Lett ; 24(19): 3532-3537, 2022 05 20.
Article in English | MEDLINE | ID: mdl-35546524

ABSTRACT

The diversity of cyclic peptides was expanded by elaborating Mitsunobu macrocyclization, tethering various hydroxy acid building blocks with different Nε-amine substituents. This new strategy was then applied in synthesizing peptidomimetic estrogen receptor modulator (PERM) analogs on the solid support. The PERM analogs exhibited increased serum peptidase stability, cell penetration, and estrogen receptor α binding affinity. Studying diversity-oriented methods for preparing azacyclopeptides provides a new tool for macrocycle construction and further structural information for optimizing ERα modulators for ER positive breast cancers.


Subject(s)
Breast Neoplasms , Peptidomimetics , Breast Neoplasms/metabolism , Estrogen Receptor alpha/chemistry , Estrogen Receptor alpha/metabolism , Female , Humans , Peptides, Cyclic , Protein Binding
12.
Cancer Lett ; 450: 110-122, 2019 05 28.
Article in English | MEDLINE | ID: mdl-30790684

ABSTRACT

Cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway is a key regulator in innate immunity and has emerged as a promising drug target in cancer treatment, but the utility of this pathway in therapeutic development is complicated by its dichotomous roles in tumor development and immunity. The activation of the STING pathway and the induced antitumor immunity could be attenuated by the feedback activation of IL-6/STAT3 pathway. Here we reported that STAT3 inhibition significantly enhanced the intensity and duration of STING signaling induced by the STING agonist c-diAM(PS)2. Such sensitization effect of STAT3 inhibition on STING signaling depended on STING rather than cGAS, which was mediated by simultaneously upregulating the positive modulators and downregulating the negative modulators of the STING pathway. Furthermore, the combination treatment with the STAT3 inhibitor and STING agonist markedly regressed tumor growth in syngeneic mice by increasing CD8+ T cells and reducing regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs) in the tumor microenvironment. Our work provides a rationale for the combination of STAT3 inhibitors and STING agonists in cancer immunotherapy.


Subject(s)
Adenosine Monophosphate/analogs & derivatives , Benzamides/pharmacology , Mammary Neoplasms, Experimental/drug therapy , Membrane Proteins/agonists , STAT3 Transcription Factor/antagonists & inhibitors , Adenosine Monophosphate/pharmacology , Animals , Drug Synergism , Female , Humans , Interferon-beta/biosynthesis , Interferon-beta/immunology , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/metabolism , Membrane Proteins/metabolism , Mice , Mice, Inbred BALB C , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects
13.
ChemMedChem ; 13(19): 2057-2064, 2018 10 08.
Article in English | MEDLINE | ID: mdl-30079976

ABSTRACT

The xanthone derivate 5',6'-dimethylxanthenone-4-acetic acid (DMXAA, also known as ASA404 or vadimezan) is a potent agonist of murine STING (stimulator of interferon genes), but cannot activate human STING. Herein we report that α-mangostin, which bears the xanthone skeleton, is an agonist of human STING, but activates murine STING to a lesser extent. Biochemical and cell-based assays indicate that α-mangostin binds to and activates human STING, leading to activation of the downstream interferon regulatory factor (IRF) pathway and production of type I interferons. Furthermore, our studies show that α-mangostin has the potential to repolarize human monocyte-derived M2 macrophages to the M1 phenotype. The agonist effect of α-mangostin in the STING pathway might account for its antitumor and antiviral activities.


Subject(s)
Antineoplastic Agents/pharmacology , Antiviral Agents/pharmacology , Membrane Proteins/agonists , Xanthones/pharmacology , Animals , Cell Line, Tumor , Cellular Reprogramming/drug effects , Humans , Interferon Regulatory Factor-3/metabolism , Interferon Type I/metabolism , Macrophages/drug effects , Membrane Proteins/chemistry , Mice , Protein Domains , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...