Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Org Biomol Chem ; 15(36): 7518-7522, 2017 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-28871293

RESUMEN

A modular total synthesis of mycolactone A/B, the exotoxin produced by Mycobacterium ulcerans, has been achieved through the orchestration of several Pd-catalyzed key steps. While this route leads to a mixture of the natural product and its C12 epimer (4 : 1 ratio), this was inconsequential from the biological activity standpoint. Compared to the previously reported routes, this synthetic blueprint allows the late-stage modification of the toxin, as exemplified by the preparation of [22,22,22-2H3]-mycolactone A/B.


Asunto(s)
Macrólidos/síntesis química , Catálisis , Macrólidos/química , Conformación Molecular , Paladio/química
2.
Chimia (Aarau) ; 71(12): 836-840, 2017 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-29289244

RESUMEN

Buruli ulcer, classified as a neglected tropical disease by the World Health Organization, is caused by a mycobacterium which secretes a macrolidic exotoxin called mycolactone A/B. In this article, several synthetic strategies for the preparation of this toxin are discussed, highlighting the importance of total synthesis for the exploration of biological mechanism underpinning relevant human diseases.


Asunto(s)
Úlcera de Buruli/etiología , Macrólidos/síntesis química , Humanos
3.
Front Pharmacol ; 12: 733496, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34603049

RESUMEN

Mycolactone is a diffusible lipid toxin produced by Mycobacterium ulcerans, the causative agent of Buruli ulcer disease. Altough bacterially derived mycolactone has been shown to traffic from cutaneous foci of infection to the bloodstream, the mechanisms underpinning its access to systemic circulation and import by host cells remain largely unknown. Using biophysical and cell-based approaches, we demonstrate that mycolactone specific association to serum albumin and lipoproteins is necessary for its solubilization and is a major mechanism to regulate its bioavailability. We also demonstrate that Scavenger Receptor (SR)-B1 contributes to the cellular uptake of mycolactone. Overall, we suggest a new mechanism of transport and cell entry, challenging the dogma that the toxin enters host cells via passive diffusion.

4.
PLoS Negl Trop Dis ; 14(12): e0008878, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33264290

RESUMEN

Mycobacterium ulcerans, the causative agent of Buruli ulcer (BU) disease, is unique amongst human pathogens in its capacity to produce a lipid toxin called mycolactone. While previous studies have demonstrated that bacterially-released mycolactone diffuses beyond infection foci, the spatiotemporal distribution of mycolactone remained largely unknown. Here, we used the zebrafish model to provide the first global kinetic analysis of mycolactone's diffusion in vivo, and multicellular co-culture systems to address the critical question of the toxin's access to the brain. Zebrafish larvae were injected with a fluorescent-derivative of mycolactone to visualize the in vivo diffusion of the toxin from the peripheral circulation. A rapid, body-wide distribution of mycolactone was observed, with selective accumulation in tissues near the injection site and brain, together with an important excretion through the gastro-intestinal tract. Our conclusion that mycolactone reached the central nervous system was reinforced by an in cellulo model of human blood brain barrier and a mouse model of M. ulcerans-infection. Here we show that mycolactone has a broad but heterogenous profile of distribution in vivo. Our investigations in vitro and in vivo support the view that a fraction of bacterially-produced mycolactone gains access to the central nervous system. The relative persistence of mycolactone in the bloodstream suggests that assays of circulating mycolactone are relevant for BU disease monitoring and treatment optimization.


Asunto(s)
Toxinas Bacterianas/farmacocinética , Sistema Nervioso Central/metabolismo , Macrólidos/farmacocinética , Animales , Astrocitos/fisiología , Toxinas Bacterianas/administración & dosificación , Barrera Hematoencefálica , Línea Celular , Células Endoteliales/fisiología , Humanos , Larva , Macrólidos/administración & dosificación , Mycobacterium ulcerans , Imagen Óptica , Análisis Espacio-Temporal , Pez Cebra
5.
J Med Chem ; 61(8): 3370-3388, 2018 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-29590751

RESUMEN

Macrocyclic inhibitors of rhodesain (RD), a parasitic cysteine protease and drug target for the treatment of human African trypanosomiasis, have shown low metabolic stability at the macrocyclic ether bridge. A series of acyclic dipeptidyl nitriles was developed using structure-based design (PDB ID: 6EX8 ). The selectivity against the closely related cysteine protease human cathepsin L (hCatL) was substantially improved, up to 507-fold. In the S2 pocket, 3,4-dichlorophenylalanine residues provided high trypanocidal activities. In the S3 pocket, aromatic residues provided enhanced selectivity against hCatL. RD inhibition ( Ki values) and in vitro cell-growth of Trypanosoma brucei rhodesiense (IC50 values) were measured in the nanomolar range. Triazole-based ligands, obtained by a safe, gram-scale flow production of ethyl 1 H-1,2,3-triazole-4-carboxylate, showed excellent metabolic stability in human liver microsomes and in vivo half-lives of up to 1.53 h in mice. When orally administered to infected mice, parasitaemia was reduced but without complete removal of the parasites.


Asunto(s)
Cisteína Endopeptidasas/metabolismo , Inhibidores de Cisteína Proteinasa/uso terapéutico , Dipéptidos/uso terapéutico , Nitrilos/uso terapéutico , Triazoles/uso terapéutico , Tripanocidas/uso terapéutico , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Animales , Sitios de Unión , Línea Celular , Cisteína Endopeptidasas/química , Inhibidores de Cisteína Proteinasa/síntesis química , Inhibidores de Cisteína Proteinasa/farmacocinética , Inhibidores de Cisteína Proteinasa/toxicidad , Dipéptidos/síntesis química , Dipéptidos/farmacocinética , Dipéptidos/toxicidad , Diseño de Fármacos , Femenino , Humanos , Leishmania donovani/efectos de los fármacos , Ligandos , Ratones , Microsomas Hepáticos/metabolismo , Estructura Molecular , Nitrilos/síntesis química , Nitrilos/farmacocinética , Nitrilos/toxicidad , Plasmodium falciparum/efectos de los fármacos , Proteínas Protozoarias/antagonistas & inhibidores , Proteínas Protozoarias/química , Ratas , Relación Estructura-Actividad , Porcinos , Triazoles/síntesis química , Triazoles/farmacocinética , Triazoles/toxicidad , Tripanocidas/síntesis química , Tripanocidas/farmacocinética , Tripanocidas/toxicidad , Trypanosoma brucei rhodesiense/efectos de los fármacos , Trypanosoma cruzi/efectos de los fármacos
6.
J Exp Med ; 213(13): 2885-2896, 2016 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-27821549

RESUMEN

Mycolactone, an immunosuppressive macrolide released by the human pathogen Mycobacterium ulcerans, was previously shown to impair Sec61-dependent protein translocation, but the underlying molecular mechanism was not identified. In this study, we show that mycolactone directly targets the α subunit of the Sec61 translocon to block the production of secreted and integral membrane proteins with high potency. We identify a single-amino acid mutation conferring resistance to mycolactone, which localizes its interaction site near the lumenal plug of Sec61α. Quantitative proteomics reveals that during T cell activation, mycolactone-mediated Sec61 blockade affects a selective subset of secretory proteins including key signal-transmitting receptors and adhesion molecules. Expression of mutant Sec61α in mycolactone-treated T cells rescued their homing potential and effector functions. Furthermore, when expressed in macrophages, the mycolactone-resistant mutant restored IFN-γ receptor-mediated antimicrobial responses. Thus, our data provide definitive genetic evidence that Sec61 is the host receptor mediating the diverse immunomodulatory effects of mycolactone and identify Sec61 as a novel regulator of immune cell functions.


Asunto(s)
Macrólidos/farmacología , Receptores de Interferón/inmunología , Canales de Translocación SEC/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Linfocitos T/inmunología , Adhesión Celular/efectos de los fármacos , Adhesión Celular/genética , Adhesión Celular/inmunología , Humanos , Células Jurkat , Receptores de Interferón/genética , Canales de Translocación SEC/genética , Canales de Translocación SEC/inmunología , Transducción de Señal/genética , Transducción de Señal/inmunología , Receptor de Interferón gamma
7.
Sci Transl Med ; 7(289): 289ra85, 2015 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-26019221

RESUMEN

Inflammation adversely affects the health of millions of people worldwide, and there is an unmet medical need for better anti-inflammatory drugs. We evaluated the therapeutic interest of mycolactone, a polyketide-derived macrolide produced by Mycobacterium ulcerans. Bacterial production of mycolactone in human skin causes a combination of ulcerative, analgesic, and anti-inflammatory effects. Whereas ulcer formation is mediated by the proapoptotic activity of mycolactone on skin cells via hyperactivation of Wiskott-Aldrich syndrome proteins, analgesia results from neuronal hyperpolarization via signaling through angiotensin II type 2 receptors. Mycolactone also blunts the capacity of immune cells to produce inflammatory mediators by an independent mechanism of protein synthesis blockade. In an attempt to isolate the structural determinants of mycolactone's immunosuppressive activity, we screened a library of synthetic subunits of mycolactone for inhibition of cytokine production by activated T cells. The minimal structure retaining immunosuppressive activity was a truncated version of mycolactone, missing one of the two core-branched polyketide chains. This compound inhibited the inflammatory cytokine responses of human primary cells at noncytotoxic doses and bound to angiotensin II type 2 receptors comparably to mycolactone in vitro. Notably, it was considerably less toxic than mycolactone in human primary dermal fibroblasts modeling ulcerative activity. In mouse models of human diseases, it conferred systemic protection against chronic skin inflammation and inflammatory pain, with no apparent side effects. In addition to establishing the anti-inflammatory potency of mycolactone in vivo, our study therefore highlights the translational potential of mycolactone core-derived structures as prospective immunosuppressants.


Asunto(s)
Inflamación/tratamiento farmacológico , Macrólidos/uso terapéutico , Animales , Enfermedad Crónica , Células HeLa , Humanos , Inmunomodulación , Inflamación/patología , Células Jurkat , Macrólidos/química , Ratones , Mycobacterium ulcerans/fisiología , Dolor/complicaciones , Dolor/tratamiento farmacológico , Sustancias Protectoras/uso terapéutico , Acetato de Tetradecanoilforbol/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA