Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
RSC Med Chem ; 15(4): 1216-1224, 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38665839

ABSTRACT

ß-Amyloid (Aß) aggregation is increasingly recognized as both a biomarker and an inducer of the progression of Alzheimer's disease (AD). Here, we describe a novel fluorescent probe P14, developed based on the BODIPY structure, capable of simultaneous visualization and inhibition of Aß aggregation in vivo. P14 shows high binding affinity to Aß aggregates and selectively labels Aß plaques in the brain slices of APP/PS1 mice. Moreover, P14 is able to visualize overloaded Aß in both APP/PS1 and 5 × FAD transgenic mice in vivo. From the aspect of potential therapeutic effects, P14 administration inhibits Aß aggregation and alleviates Aß-induced neuronal damage in vitro, as well as reduces central Aß deposition and ameliorates cognitive impairment in APP/PS1 transgenic mice in vivo. Finally, P14 is applied to monitor the progression of Aß aggregation in the brain of 5 × FAD transgenic mice and the intervention effect itself by fluorescence imaging. In summary, the discovery of this fluorescent agent might provide important clues for the future development of theranostic drug candidates targeting Aß aggregation in AD.

2.
Acta Pharm Sin B ; 14(1): 319-334, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38261830

ABSTRACT

Receptor-interacting serine/threonine-protein kinase 1 (RIPK1) functions as a key regulator in inflammation and cell death and is involved in mediating a variety of inflammatory or degenerative diseases. A number of allosteric RIPK1 inhibitors (RIPK1i) have been developed, and some of them have already advanced into clinical evaluation. Recently, selective RIPK1i that interact with both the allosteric pocket and the ATP-binding site of RIPK1 have started to emerge. Here, we report the rational development of a new series of type-II RIPK1i based on the rediscovery of a reported but mechanistically atypical RIPK3i. We also describe the structure-guided lead optimization of a potent, selective, and orally bioavailable RIPK1i, 62, which exhibits extraordinary efficacies in mouse models of acute or chronic inflammatory diseases. Collectively, 62 provides a useful tool for evaluating RIPK1 in animal disease models and a promising lead for further drug development.

3.
Sci Adv ; 9(47): eadj0123, 2023 11 24.
Article in English | MEDLINE | ID: mdl-37992178

ABSTRACT

Transcriptional dysregulation is a recurring pathogenic hallmark and an emerging therapeutic vulnerability in ovarian cancer. Here, we demonstrated that ovarian cancer exhibited a unique dependency on the regulatory machinery of transcriptional termination, particularly, cleavage and polyadenylation specificity factor (CPSF) complex. Genetic abrogation of multiple CPSF subunits substantially hampered neoplastic cell viability, and we presented evidence that their indispensable roles converged on the endonuclease CPSF3. Mechanistically, CPSF perturbation resulted in lengthened 3'-untranslated regions, diminished intronic polyadenylation and widespread transcriptional readthrough, and consequently suppressed oncogenic pathways. Furthermore, we reported the development of specific CPSF3 inhibitors building upon the benzoxaborole scaffold, which exerted potent antitumor activity. Notably, CPSF3 blockade effectively exacerbated genomic instability by down-regulating DNA damage repair genes and thus acted in synergy with poly(adenosine 5'-diphosphate-ribose) polymerase inhibition. These findings establish CPSF3-dependent transcriptional termination as an exploitable driving mechanism of ovarian cancer and provide a promising class of boron-containing compounds for targeting transcription-addicted human malignancies.


Subject(s)
Neoplasm Recurrence, Local , Ovarian Neoplasms , Female , Humans , Cleavage And Polyadenylation Specificity Factor/genetics , Cleavage And Polyadenylation Specificity Factor/metabolism , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics
4.
Nat Chem Biol ; 19(10): 1235-1245, 2023 10.
Article in English | MEDLINE | ID: mdl-37400537

ABSTRACT

Amyloid fibril is an important pharmaceutical target for diagnostic and therapeutic treatment of neurodegenerative diseases. However, rational design of chemical compounds that interact with amyloid fibrils is unachievable due to the lack of mechanistic understanding of the ligand-fibril interaction. Here we used cryoelectron microscopy to survey the amyloid fibril-binding mechanism of a series of compounds including classic dyes, (pre)clinical imaging tracers and newly identified binders from high-throughput screening. We obtained clear densities of several compounds in complex with an α-synuclein fibril. These structures unveil the basic mechanism of the ligand-fibril interaction, which exhibits remarkable difference from the canonical ligand-protein interaction. In addition, we discovered a druggable pocket that is also conserved in the ex vivo α-synuclein fibrils from multiple system atrophy. Collectively, these findings expand our knowledge of protein-ligand interaction in the amyloid fibril state, which will enable rational design of amyloid binders in a medicinally beneficial way.


Subject(s)
Amyloid , alpha-Synuclein , alpha-Synuclein/chemistry , Cryoelectron Microscopy , Amyloid/chemistry , Ligands
5.
J Med Chem ; 66(4): 2608-2621, 2023 02 23.
Article in English | MEDLINE | ID: mdl-36723914

ABSTRACT

There is growing interest in covalent targeted inhibitors in drug discovery against previously "undruggable" sites and targets. These molecules typically feature an electrophilic warhead that reacts with nucleophilic groups of protein residues, most notably the thiol group of cysteines. One main challenge in the field is to develop versatile utilizable warheads. Here, we characterize the unique features of novel arsenous warheads for reaction with thiol species in a reversible manner and further demonstrate that organoarsenic probes can be chemically tuned toward specific molecular targets by developing selective and potent inhibitors of pyruvate kinase M2 (PKM2). We show that compound 24 is a covalent and allosteric inhibitor of PKM2 and its orally bioavailable prodrug 25 exerts efficacious inhibition of PKM2-dependent tumor growth in vitro and in vivo. Our results introduce 25 and its derivatives as useful pharmacological tools and provide a general road map for targeting the protein cysteinome using arsenous warheads.


Subject(s)
Drug Discovery , Pyruvate Kinase , Cysteine/chemistry
6.
Proc Natl Acad Sci U S A ; 119(44): e2214227119, 2022 11.
Article in English | MEDLINE | ID: mdl-36279464

ABSTRACT

LUBAC-mediated linear ubiquitination plays a pivotal role in regulation of cell death and inflammatory pathways. Genetic deficiency in LUBAC components leads to severe immune dysfunction or embryonic lethality. LUBAC has been extensively studied for its role in mediating TNF signaling. However, Tnfr1 knockout is not able to fully rescue the embryonic lethality of LUBAC deficiency, suggesting that LUBAC may modify additional key cellular substrates in promoting cell survival. GPx4 is an important selenoprotein involved in regulating cellular redox homeostasis in defense against lipid peroxidation-mediated cell death known as ferroptosis. Here we demonstrate that LUBAC deficiency sensitizes to ferroptosis by promoting GPx4 degradation and downstream lipid peroxidation. LUBAC binds and stabilizes GPx4 by modulating its linear ubiquitination both in normal condition and under oxidative stress. Our findings identify GPx4 as a key substrate of LUBAC and a previously unrecognized role of LUBAC-mediated linear ubiquitination in regulating cellular redox status and cell death.


Subject(s)
Receptors, Tumor Necrosis Factor, Type I , Ubiquitin , Receptors, Tumor Necrosis Factor, Type I/genetics , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/metabolism , NF-kappa B/metabolism , Ubiquitination
7.
J Med Chem ; 65(20): 13645-13659, 2022 10 27.
Article in English | MEDLINE | ID: mdl-36221183

ABSTRACT

Deubiquitinating enzymes (DUBs) are key regulatory components of the ubiquitination system. Many DUBs have been revealed to play key roles in normal physiology and diseases. However, only very limited DUB members have well-characterized inhibitors. OTUB1 and USP8 are two DUBs reported to promote both immune evasion and tumorigenesis in tumor models, yet their targeted inhibitors are in the early stages of development. Here, we describe the lead identification and optimization of an OTUB1/USP8 dual inhibitor, 61, which exhibits highly potent and selective inhibition of both targets with subnanomolar IC50s in vitro. By inhibiting both DUBs, 61 phenocopies the double knockdown of OTUB1/USP8 and exerts pronounced antiproliferative effects in H1975 and other non-small-cell lung cancer (NSCLC) cell lines. Moreover, 61 efficaciously mitigates tumor growth in vivo. Collectively, our results provide a useful tool for pharmacological perturbation of OTUB1/USP8 and introduce a promising therapeutic strategy of dual DUB inhibition for treating NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Carcinoma, Non-Small-Cell Lung/drug therapy , Proteostasis , Lung Neoplasms/drug therapy , Ubiquitination , Deubiquitinating Enzymes/metabolism , Endopeptidases/metabolism , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/metabolism , Endosomal Sorting Complexes Required for Transport/genetics , Endosomal Sorting Complexes Required for Transport/metabolism
8.
Cancer Res ; 82(19): 3588-3602, 2022 Oct 04.
Article in English | MEDLINE | ID: mdl-35857807

ABSTRACT

Therapeutic perturbation of cyclin-dependent kinase 12 (CDK12) is proposed to have pleiotropic effects in ovarian cancer, including direct cytotoxicity against tumor cells and indirect induction of immunogenicity that confer synthetic sensitivity to immune-based treatment. However, formal testing of this hypothesis has been hindered by an insufficient mechanistic understanding of CDK12 and its close homolog CDK13, as well as generally unfavorable pharmacokinetics of available CDK12/CDK13 covalent inhibitors. In this study, we used an innovative arsenous warhead modality to develop an orally bioavailable CDK12/CDK13 covalent compound. The dual CDK12/CDK13 inhibitors ZSQ836 exerted potent anticancer activity in cell culture and mouse models and induced transcriptional reprogramming, including downregulation of DNA damage response genes. CDK12 and CDK13 were both ubiquitously expressed in primary and metastatic ovarian cancer, and the two kinases performed independent and synergistic functions to promote tumorigenicity. Unexpectedly, although ZSQ836 triggered genomic instability in malignant cells, it counterintuitively impaired lymphocytic infiltration in neoplastic lesions by interfering with T-cell proliferation and activation. These findings highlight the Janus-faced effects of dual CDK12/CDK13 inhibitors by simultaneously suppressing tumor and immune cells, offering valuable insights into the future direction of drug discovery to pharmacologically target CDK12. SIGNIFICANCE: This study dissects the specific roles of CDK12 and CDK13 in ovarian cancer and develops a CDK12/CDK13 inhibitor that impairs both tumor and immune cells, which could guide future CDK12 inhibitor development.


Subject(s)
CDC2 Protein Kinase , Ovarian Neoplasms , Animals , Carcinoma, Ovarian Epithelial/genetics , Cyclin-Dependent Kinases/genetics , Female , Genes, cdc , Humans , Mice , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics
9.
Cell Chem Biol ; 28(6): 855-865.e9, 2021 06 17.
Article in English | MEDLINE | ID: mdl-33979649

ABSTRACT

The COVID-19 pandemic has been disastrous to society and effective drugs are urgently needed. The papain-like protease domain (PLpro) of SARS-CoV-2 (SCoV2) is indispensable for viral replication and represents a putative target for pharmacological intervention. In this work, we describe the development of a potent and selective SCoV2 PLpro inhibitor, 19. The inhibitor not only effectively blocks substrate cleavage and immunosuppressive function imparted by PLpro, but also markedly mitigates SCoV2 replication in human cells, with a submicromolar IC50. We further present a convenient and sensitive activity probe, 7, and complementary assays to readily evaluate SCoV2 PLpro inhibitors in vitro or in cells. In addition, we disclose the co-crystal structure of SCoV2 PLpro in complex with a prototype inhibitor, which illuminates their detailed binding mode. Overall, these findings provide promising leads and important tools for drug discovery aiming to target SCoV2 PLpro.


Subject(s)
Coronavirus Papain-Like Proteases/antagonists & inhibitors , Drug Delivery Systems/methods , Drug Development/methods , Protease Inhibitors/administration & dosage , SARS-CoV-2/drug effects , A549 Cells , Animals , Antiviral Agents/administration & dosage , Antiviral Agents/chemistry , Antiviral Agents/metabolism , COVID-19/enzymology , Coronavirus Papain-Like Proteases/chemistry , Coronavirus Papain-Like Proteases/metabolism , Dose-Response Relationship, Drug , HEK293 Cells , HeLa Cells , Humans , Mice , Molecular Docking Simulation/methods , Protease Inhibitors/chemistry , Protease Inhibitors/metabolism , Protein Structure, Secondary , Protein Structure, Tertiary , SARS-CoV-2/chemistry , SARS-CoV-2/enzymology , COVID-19 Drug Treatment
10.
Biosens Bioelectron ; 172: 112756, 2021 Jan 15.
Article in English | MEDLINE | ID: mdl-33197750

ABSTRACT

Acute kidney injury (AKI) is common in hospital patients. Delayed diagnosis and treatment of AKI due to the lack of efficient early diagnosis is an important cause of its high mortality. While fluorescence imaging seems promising to non-intrusively interrogate AKI-related biomarkers, the low kidney contrast of many fluorophores conferred by their relatively low abundance of distribution in the kidney limits their application for AKI detection. Herein, we discovered a near-infrared fluorophore with inherent kidney-targeting ability. Based on this fluorophore, a fluorogenic probe (KNP-1) was developed by targeting peroxynitrite (ONOO-), which is upregulated at the early onset of AKI. KNP-1 exhibits desirable kidney distribution after intravenous administration and is fluorescent only after activation by ONOO-. These properties lead to excellent kidney contrast imaging results. KNP-1 is capable of detecting both nephrotoxin-induced and ischemia-reperfusion injury-induced AKI in live mice. Temporally resolved imaging of AKI-disease model mice with KNP-1 suggests a gradual increase in renal ONOO- levels with disease progression. Notably, the upregulation of ONOO- can be observed at least 24 h earlier than the clinically popular sCr and BUN methods. Blocking ONOO- generation also proves beneficial. These results highlight the applicability of this inherently tissue targeting-based strategy for designing probes with desirable imaging contrast; potentiate ONOO- as a biomarker and target for AKI early diagnosis and medical intervention; and imply the clinical relevance of KNP-1 for AKI early detection.


Subject(s)
Acute Kidney Injury , Biosensing Techniques , Acute Kidney Injury/diagnosis , Animals , Biomarkers , Early Diagnosis , Fluorescent Dyes , Humans , Kidney/diagnostic imaging , Mice
11.
Proc Natl Acad Sci U S A ; 117(49): 31123-31133, 2020 12 08.
Article in English | MEDLINE | ID: mdl-33229560

ABSTRACT

Membraneless organelles contain a wide spectrum of molecular chaperones, indicating their important roles in modulating the metastable conformation and biological function of membraneless organelles. Here we report that class I and II Hsp40 (DNAJ) proteins possess a high ability of phase separation rendered by the flexible G/F-rich region. Different Hsp40 proteins localize in different membraneless organelles. Specifically, human Hdj1 (DNAJB1), a class II Hsp40 protein, condenses in ubiquitin (Ub)-rich nuclear bodies, while Hdj2 (DNAJA1), a class I Hsp40 protein, condenses in nucleoli. Upon stress, both Hsp40 proteins incorporate into stress granules (SGs). Mutations of the G/F-rich region not only markedly impaired Hdj1 phase separation and SG involvement and disrupted the synergistic phase separation and colocalization of Hdj1 and fused in sarcoma (FUS) in cells. Being cophase separated with FUS, Hdj1 stabilized the liquid phase of FUS against proceeding into amyloid aggregation in vitro and alleviated abnormal FUS aggregation in cells. Moreover, Hdj1 uses different domains to chaperone FUS phase separation and amyloid aggregation. This paper suggests that phase separation is an intrinsic property of Hsp40 proteins, which enables efficient incorporation and function of Hsp40 in membraneless organelles and may further mediate the buildup of chaperone network in membraneless organelles.


Subject(s)
HSP40 Heat-Shock Proteins/genetics , Molecular Chaperones/genetics , Cell Membrane/genetics , Humans , Membranes , Organelles/genetics , Protein Binding/genetics , RNA-Binding Protein FUS/genetics , Ubiquitin/genetics
12.
Theranostics ; 9(19): 5672-5680, 2019.
Article in English | MEDLINE | ID: mdl-31534510

ABSTRACT

Autophagy is a dynamic and complicated catabolic process. Imaging autophagic flux can clearly advance knowledge of its pathophysiology significance. While the most common way autophagy is imaged relies on fluorescent protein-based probes, this method requires substantial genetic manipulation that severely restricts the application. Small fluorescent probes capable of tracking autophagic flux with good spatiotemporal resolution are highly demanable. Methods: In this study, we developed a small-molecule fluorogenic probe (AFG-1) that facilitates real-time imaging of autophagic flux in both intact cells and live mice. AFG-1 is inspired by the cascading nitrosative and acidic microenvironments evolving during autophagy. It operates over two sequential steps. In the first step, AFG-1 responds to the up-regulated peroxynitrite at the initiation of autophagy by its diphenylamino group being oxidatively dearylated to yield a daughter probe. In the second step, the daughter probe responds to the acidic autolysosomes at the late stage of autophagy by being protonated. Results: This pathway-dependent mechanism has been confirmed first by sequentially sensing ONOO- and acid in aqueous solution, and then by imaging autophagic flux in live cells. Furthermore, AFG-1 has been successfully applied to visualize autophagic flux in real-time in live mice following brain ischemic injury, justifying its robustness. Conclusion: Due to the specificity, easy operation, and the dynamic information yielded, AFG-1 should serve as a potential tool to explore the roles of autophagy under various pathological settings.


Subject(s)
Autophagy/drug effects , Fluorescent Dyes/metabolism , Animals , Cell Line , Cellular Microenvironment , Endothelium/metabolism , Endothelium/pathology , Fluorescent Dyes/chemistry , Lysosomes/metabolism , Male , Mice , Mice, Inbred C57BL , Molecular Probes/chemistry , Molecular Probes/metabolism , Nitrosative Stress , Peroxynitrous Acid/metabolism
13.
Bioconjug Chem ; 29(10): 3459-3466, 2018 10 17.
Article in English | MEDLINE | ID: mdl-30189724

ABSTRACT

Fluorescent imaging of ß-amyloid (Aß) is one of the most promising methods for Alzheimer's disease diagnosis. Several fluorescent probes have been reported to detect Aß both in vitro and in vivo. However, highly sensitive and highly selective probes with low background signals are still greatly needed. Herein, we rationally designed and synthesized a PIET quenched near-infrared probe QAD-1 to detect Aß. This probe contains BODIPY as fluorophore and tetrahydroquinoxaline as the quenching group. QAD-1 exhibited significant fluorescent switch-on after binding to soluble and insoluble Aß species, and the probe had the benefit of low background signal to stain Aß plaques without the need of wash-out procedures in vitro, which was specially found by the fluorescence off-on probe. QAD-1 could identify the overproduced Aß in transgenic (APPSWE/PSEN 1dE9) AD mice as early as 6 months old in vivo, which indicated that QAD-1 may be a potential probe for monitoring Aß species at an early stage of AD.


Subject(s)
Amyloid beta-Peptides/metabolism , Boron Compounds/chemistry , Fluorescent Dyes/chemistry , Spectrometry, Fluorescence/methods , Alzheimer Disease/diagnosis , Alzheimer Disease/metabolism , Animals , Brain/metabolism , Disease Models, Animal , Fluorescent Dyes/chemical synthesis , Fluorescent Dyes/metabolism , Male , Mice , Mice, Transgenic , Plaque, Amyloid/metabolism
14.
RSC Adv ; 8(10): 5542-5549, 2018 Jan 29.
Article in English | MEDLINE | ID: mdl-35542398

ABSTRACT

A copper-catalyzed direct C-H arylation or vinylation of BODIPYs at the ß-position by iodonium salts has been developed, which provides facile access to a variety of mono-substituted BODIPY dyes. Interestingly, ß-styryl BODIPY compound 9b exhibits apparent cytotoxicity after laser irradiation, which has great potential for photodynamic therapy.

15.
Biosens Bioelectron ; 75: 136-41, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26313423

ABSTRACT

Here we reported the development of the first photoinduced electron transfer (PeT) probe (1) to directly locate ß-amyloid aggregates (Aß plaques) in the brain without the need of post-washing procedures. The probe showed a high affinity for Aß aggregates with a Kd value of 3.5nM. It is weakly emissive by itself with its fluorescence quenched by electron transfer from PeT donor to the excited fluorophore. But selective binding to Aß plaques would attenuate the PeT process and restore the fluorescence, therefore facilitating the tracking of Aß plaques. The probe is advantageous in that its fluorescence is environment-less-sensitive and no washing procedure is required to provide high contrast fluorescent signal when applied to stain brain tissues. As a proof of concept, its application has been exemplified by staining Aß plaques in slices of brain tissue from double transgenic (APP/PS1) mice of Alzheimer's disease.


Subject(s)
Alzheimer Disease/diagnosis , Amyloid beta-Peptides/isolation & purification , Biosensing Techniques , Plaque, Amyloid/chemistry , Alzheimer Disease/pathology , Amyloid beta-Peptides/chemistry , Animals , Disease Models, Animal , Fluorescent Dyes/chemistry , Humans , Mice , Plaque, Amyloid/diagnosis , Plaque, Amyloid/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...