Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Angew Chem Int Ed Engl ; 63(8): e202314791, 2024 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-38109686

RESUMEN

Photolipids have emerged as attractive tools for the optical control of lipid functions. They often contain an azobenzene photoswitch that imparts a cis double-bond upon irradiation. Herein, we present the application of photoswitching to a lipidated natural product, the potent proteasome inhibitor cepafungin I. Several azobenzene-containing lipids were attached to the cyclopeptide core, yielding photoswitchable derivatives. Most notably, PhotoCep4 exhibited a 10-fold higher cellular potency in its light-induced cis-form, matching the potency of natural cepafungin I. The length of the photolipid tail and distal positioning of the azobenzene photoswitch with respect to the macrocycle is critical for this activity. In a proteome-wide experiment, light-triggered PhotoCep4 modulation showed high overlap with constitutively active cepafungin I. The mode of action was studied using crystallography and revealed an identical binding of the cyclopeptide in comparison to cepafungin I, suggesting that differences in their cellular activity originate from switching the tail structure. The photopharmacological approach described herein could be applicable to many other natural products as lipid conjugation is common and often necessary for potent activity. Such lipids are often introduced late in synthetic routes, enabling facile chemical modifications.


Asunto(s)
Compuestos Azo , Lipopéptidos , Lipopéptidos/farmacología , Proteolisis , Compuestos Azo/química , Péptidos Cíclicos/farmacología
2.
ACS Cent Sci ; 9(2): 239-251, 2023 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-36844499

RESUMEN

The cepafungins are a class of highly potent and selective eukaryotic proteasome inhibitor natural products with potential to treat refractory multiple myeloma and other cancers. The structure-activity relationship of the cepafungins is not fully understood. This Article chronicles the development of a chemoenzymatic approach to cepafungin I. A failed initial route involving derivatization of pipecolic acid prompted us to examine the biosynthetic pathway for the production of 4-hydroxylysine, which culminated in the development of a 9-step synthesis of cepafungin I. An alkyne-tagged analogue enabled chemoproteomic studies of cepafungin and comparison of its effects on global protein expression in human multiple myeloma cells to the clinical drug bortezomib. A preliminary series of analogues elucidated critical determinants of potency in proteasome inhibition. Herein we report the chemoenzymatic syntheses of 13 additional analogues of cepafungin I guided by a proteasome-bound crystal structure, 5 of which are more potent than the natural product. The lead analogue was found to have 7-fold greater proteasome ß5 subunit inhibitory activity and has been evaluated against several multiple myeloma and mantle cell lymphoma cell lines in comparison to the clinical drug bortezomib.

3.
Angew Chem Int Ed Engl ; 61(18): e202115193, 2022 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-35170181

RESUMEN

For the discovery of novel chemical matter generally endowed with bioactivity, strategies may be particularly efficient that combine previous insight about biological relevance, e.g., natural product (NP) structure, with methods that enable efficient coverage of chemical space, such as fragment-based design. We describe the de novo combination of different 5-membered NP-derived N-heteroatom fragments to structurally unprecedented "pseudo-natural products" in an efficient complexity-generating and enantioselective one-pot synthesis sequence. The pseudo-NPs inherit characteristic elements of NP structure but occupy areas of chemical space not covered by NP-derived chemotypes, and may have novel biological targets. Investigation of the pseudo-NPs in unbiased phenotypic assays and target identification led to the discovery of the first small-molecule ligand of the RHO GDP-dissociation inhibitor 1 (RHOGDI1), termed Rhonin. Rhonin inhibits the binding of the RHOGDI1 chaperone to GDP-bound RHO GTPases and alters the subcellular localization of RHO GTPases.


Asunto(s)
Productos Biológicos , Productos Biológicos/química , Ligandos , Proteínas de Unión al GTP rho , Inhibidor alfa de Disociación del Nucleótido Guanina rho , Inhibidores de la Disociación del Nucleótido Guanina rho-Específico
4.
J Am Chem Soc ; 143(48): 20332-20342, 2021 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-34817176

RESUMEN

Chemoproteomic profiling of cysteines has emerged as a powerful method for screening the proteome-wide targets of cysteine-reactive fragments, drugs, and natural products. Herein, we report the development and an in-depth evaluation of a tetrafluoroalkyl benziodoxole (TFBX) as a cysteine-selective chemoproteomic probe. We show that this probe features numerous key improvements compared to the traditionally used cysteine-reactive probes, including a superior target occupancy, faster labeling kinetics, and broader proteomic coverage, thus enabling profiling of cysteines directly in live cells. In addition, the fluorine "signature" of probe 7 constitutes an additional advantage resulting in a more confident adduct-amino acid site assignment in mass-spectrometry-based identification workflows. We demonstrate the utility of our new probe for proteome-wide target profiling by identifying the cellular targets of (-)-myrocin G, an antiproliferative fungal natural product with a to-date unknown mechanism of action. We show that this natural product and a simplified analogue target the X-ray repair cross-complementing protein 5 (XRCC5), an ATP-dependent DNA helicase that primes DNA repair machinery for nonhomologous end joining (NHEJ) upon DNA double-strand breaks, making them the first reported inhibitors of this biomedically highly important protein. We further demonstrate that myrocins disrupt the interaction of XRCC5 with DNA leading to sensitization of cancer cells to the chemotherapeutic agent etoposide as well as UV-light-induced DNA damage. Altogether, our next-generation cysteine-reactive probe enables broader and deeper profiling of the cysteinome, rendering it a highly attractive tool for elucidation of targets of electrophilic small molecules.


Asunto(s)
Cisteína/química , Compuestos Heterocíclicos con 2 Anillos/química , Hidrocarburos Fluorados/química , Sondas Moleculares/química , Proteómica/métodos , Alquilación , Reparación del ADN por Unión de Extremidades/efectos de los fármacos , Diterpenos/farmacología , Inhibidores Enzimáticos/farmacología , Células HEK293 , Células HeLa , Humanos , Autoantígeno Ku/antagonistas & inhibidores , Autoantígeno Ku/química
5.
ACS Chem Biol ; 16(12): 2845-2851, 2021 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-34792325

RESUMEN

Arbidol (ARB) is a broad-spectrum antiviral drug approved in Russia and China for the treatment of influenza. ARB was tested in patients as a drug candidate for the treatment at the early onset of COVID-19 caused by the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Despite promising clinical results and multiple ongoing trials, preclinical data are lacking and the molecular mechanism of action of ARB against SARS-CoV-2 remains unknown. Here, we demonstrate that ARB binds to the spike viral fusion glycoprotein of the SARS-CoV-2 Wuhan strain as well as its more virulent variants from the United Kingdom (strain B.1.1.7) and South Africa (strain B.1.351). We pinpoint the ARB binding site on the S protein to the S2 membrane fusion domain and use an infection assay with Moloney murine leukemia virus (MLV) pseudoviruses (PVs) pseudotyped with the S proteins of the Wuhan strain and the new variants to show that this interaction is sufficient for the viral cell entry inhibition by ARB. Finally, our experiments reveal that the ARB interaction leads to a significant destabilization and eventual lysosomal degradation of the S protein in cells. Collectively, our results identify ARB as the first clinically approved small molecule drug binder of the SARS-CoV-2 S protein and place ARB among the more promising drug candidates for COVID-19.


Asunto(s)
Antivirales/farmacología , Indoles/farmacología , SARS-CoV-2/efectos de los fármacos , Glicoproteína de la Espiga del Coronavirus/metabolismo , Células A549 , Animales , Antivirales/metabolismo , Sitios de Unión , Chlorocebus aethiops , Células HEK293 , Humanos , Indoles/metabolismo , Lisosomas/metabolismo , Mutación , Dominios Proteicos , Proteolisis/efectos de los fármacos , Glicoproteína de la Espiga del Coronavirus/química , Glicoproteína de la Espiga del Coronavirus/genética , Células Vero , Internalización del Virus/efectos de los fármacos
6.
Angew Chem Int Ed Engl ; 60(6): 3071-3079, 2021 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-33035395

RESUMEN

Herein, we report arylazopyrazole ureas and sulfones as a novel class of photoswitchable serine hydrolase inhibitors and present a chemoproteomic platform for rapid discovery of optically controlled serine hydrolase targets in complex proteomes. Specifically, we identify highly potent and selective photoswitchable inhibitors of the drug-metabolizing enzymes carboxylesterases 1 and 2 and demonstrate their pharmacological application by optically controlling the metabolism of the immunosuppressant drug mycophenolate mofetil. Collectively, this proof-of-concept study provides a first example of photopharmacological tools to optically control drug metabolism by modulating the activity of a metabolizing enzyme. Our arylazopyrazole ureas and sulfones offer synthetically accessible scaffolds that can be expanded to identify specific photoswitchable inhibitors for other serine hydrolases, including lipases, peptidases, and proteases. Our chemoproteomic platform can be applied to other photoswitches and scaffolds to achieve optical control over diverse protein classes.


Asunto(s)
Carboxilesterasa/antagonistas & inhibidores , Inhibidores Enzimáticos/química , Preparaciones Farmacéuticas/metabolismo , Rayos Ultravioleta , Células CACO-2 , Carboxilesterasa/metabolismo , Hidrolasas de Éster Carboxílico/antagonistas & inhibidores , Hidrolasas de Éster Carboxílico/genética , Hidrolasas de Éster Carboxílico/metabolismo , Evaluación Preclínica de Medicamentos , Inhibidores Enzimáticos/metabolismo , Humanos , Hidrólisis , Microscopía Fluorescente , Preparaciones Farmacéuticas/química , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Estereoisomerismo , Sulfonas/química , Sulfonas/metabolismo , Ureasa/química , Ureasa/metabolismo
7.
Cell Chem Biol ; 27(10): 1318-1326.e18, 2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-32763140

RESUMEN

The natural product cepafungin I was recently reported to be one of the most potent covalent inhibitors of the 20S proteasome core particle through a series of in vitro activity assays. Here, we report a short chemoenzymatic total synthesis of cepafungin I featuring the use of a regioselective enzymatic oxidation to prepare a key hydroxylated amino acid building block in a scalable fashion. The strategy developed herein enabled access to a chemoproteomic probe, which in turn revealed the exceptional selectivity and potency of cepafungin I toward the ß2 and ß5 subunits of the proteasome. Further structure-activity relationship studies suggest the key role of the hydroxyl group in the macrocycle and the identity of the lipid tail in modulating the potency of this natural product family. This study lays the groundwork for further medicinal chemistry exploration to fully realize the anticancer potential of cepafungin I.


Asunto(s)
Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma/farmacología , Células Cultivadas , Humanos , Estructura Molecular , Péptidos Cíclicos/síntesis química , Péptidos Cíclicos/química , Péptidos Cíclicos/farmacología , Inhibidores de Proteasoma/síntesis química , Inhibidores de Proteasoma/química , Estereoisomerismo , Relación Estructura-Actividad
8.
Cell Chem Biol ; 27(5): 586-597.e12, 2020 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-32330443

RESUMEN

In this study, we identify the natural product gambogic acid as well as structurally related synthetic xanthones as first-in-class covalent inhibitors of the de novo sphingolipid biosynthesis. We apply chemoproteomics to determine that gambogic acid binds to the regulatory small subunit B of the serine palmitoyltransferase complex (SPTSSB). We then test structurally related synthetic xanthones to identify 18 as an equally potent but more selective binder of SPTSSB and show that 18 reduces sphingolipid levels in situ and in vivo. Finally, using various biological methods, we demonstrate that 18 induces cellular responses characteristic for diminished sphingosine-1-phosphate (S1P) signaling. This study demonstrates that SPTSSB may become a viable therapeutic target in various diseases with pathological S1P signaling. Furthermore, we believe that our compound will become a valuable tool for studying the sphingolipid metabolism and serve as a blueprint for the development of a new generation of sphingolipid biosynthesis inhibitors.


Asunto(s)
Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Serina C-Palmitoiltransferasa/antagonistas & inhibidores , Xantonas/química , Xantonas/farmacología , Animales , Descubrimiento de Drogas , Células HEK293 , Humanos , Lisofosfolípidos/metabolismo , Células MCF-7 , Ratones , Ratones Endogámicos ICR , Proteómica , Serina C-Palmitoiltransferasa/metabolismo , Esfingolípidos/metabolismo , Esfingosina/análogos & derivados , Esfingosina/metabolismo
9.
J Am Chem Soc ; 142(15): 6970-6982, 2020 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-32233464

RESUMEN

Many RNAs are processed into biologically active transcripts, the aberrant expression of which can contribute to disease phenotypes. For example, the primary microRNA-17-92 (pri-miR-17-92) cluster contains six microRNAs (miRNAs) that collectively act in several disease settings. Herein, we used sequence-based design of structure-specific ligands to target a common structure in the Dicer processing sites of three miRNAs in the cluster, miR-17, miR-18a, and miR-20a, thereby inhibiting their biogenesis. The compound was optimized to afford a dimeric molecule that binds the Dicer processing site and an adjacent bulge, affording a 100-fold increase in potency. The dimer's mode of action was then extended from simple binding to direct cleavage by conjugation to bleomycin A5 in a manner that imparts RNA-selective cleavage or to indirect cleavage by recruiting an endogenous nuclease, or a ribonuclease targeting chimera (RIBOTAC). Interestingly, the dimer-bleomycin conjugate cleaves the entire pri-miR-17-92 cluster and hence functionally inhibits all six miRNAs emanating from it. The compound selectively reduced levels of the cluster in three disease models: polycystic kidney disease, prostate cancer, and breast cancer, rescuing disease-associated phenotypes in the latter two. Further, the bleomycin conjugate exerted selective effects on the miRNome and proteome in prostate cancer cells. In contrast, the RIBOTAC only depleted levels of pre- and mature miR-17, -18a, and 20a, with no effect on the primary transcript, in accordance with the cocellular localization of RNase L, the pre-miRNA targets, and the compound. These studies demonstrate a strategy to tune RNA structure-targeting compounds to the cellular localization of the target.


Asunto(s)
Carcinogénesis/metabolismo , Ligandos , MicroARNs/metabolismo , Humanos , Estructura Molecular
10.
J Am Chem Soc ; 139(1): 231-238, 2017 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-28001050

RESUMEN

In this study, we demonstrate that appendage of a single asparagusic acid residue (AspA tag) is sufficient to ensure efficient cellular uptake and intracellular distribution of fully unprotected peptides. We apply this new delivery method to induce apoptotic response in cancer cells using long (up to 20mer) BH3 domain peptides. Moreover, to understand the molecular mechanism of the cellular uptake, we perform chemical proteomics experiments and identify the direct molecular targets of the asparagusic acid tag. Our findings document covalent bond formation between the asparagusic acid moiety and the cysteines 556 and 558 on the surface of the transferrin receptor resulting in subsequent endocytic uptake of the payload. We believe that the small size, low cellular toxicity and the efficient transferrin receptor-mediated uptake render the AspA tag highly attractive for various life science applications.


Asunto(s)
Disulfuros/metabolismo , Receptores de Transferrina/química , Receptores de Transferrina/metabolismo , Tiofenos/metabolismo , Apoptosis , Sitios de Unión , Disulfuros/química , Relación Dosis-Respuesta a Droga , Sistemas de Liberación de Medicamentos , Células HeLa , Humanos , Estructura Molecular , Relación Estructura-Actividad , Tiofenos/química
11.
Chimia (Aarau) ; 70(11): 764-767, 2016 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-28661335

RESUMEN

Our laboratory focuses on chemical proteomics-enabled discovery of new cysteine-reactive small molecules with intriguing biomedical activities as well as identification and detailed characterization of their proteomic targets. In this overview article, we summarize our progress since 2013 in this research field. We have developed a novel mass spectrometry-based chemoproteomic method that allows detection and monitoring of up to ~3000 reactive cysteines in any cellular proteome. This is achieved via strategic use of two clickable, cysteine-reactive chemical probes with complementary substrate selectivity profiles, iodoacetamide and ethynyl benziodoxolone. Using this method, we have been able to identify the direct biological targets of curcumin, a diarylheptanoid natural product with anticancer activity, and deoxyelephantopin, a highly cytotoxic natural sesquiterpene lactone. Furthermore, we have developed chloromethyl triazoles (CMTs) as a novel chemical scaffold for cysteine-reactive inhibitors that can be accessed from commercially available substrates in only two chemical steps. From a small collection of chloromethyl triazoles, we have identified compound AA-CW236 as the first non-pseudosubstrate inhibitor of MGMT, a DNA repair protein that renders several devastating cancer forms resistant to chemotherapy.


Asunto(s)
Cisteína/análisis , Cisteína/química , Descubrimiento de Drogas , Proteómica , Cisteína/antagonistas & inhibidores , Humanos , Espectrometría de Masas , Modelos Moleculares , Estructura Molecular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...