Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Cancer Res ; 80(6): 1268-1278, 2020 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-31941698

RESUMEN

Targeted delivery of chemotherapeutics aims to increase efficacy and lower toxicity by concentrating drugs at the site-of-action, a method embodied by the seven current FDA-approved antibody-drug conjugates (ADC). However, a variety of pharmacokinetic challenges result in relatively narrow therapeutic windows for these agents, hampering the development of new drugs. Here, we use a series of prostate-specific membrane antigen-binding single-domain (Humabody) ADC constructs to demonstrate that tissue penetration of protein-drug conjugates plays a major role in therapeutic efficacy. Counterintuitively, a construct with lower in vitro potency resulted in higher in vivo efficacy than other protein-drug conjugates. Biodistribution data, tumor histology images, spheroid experiments, in vivo single-cell measurements, and computational results demonstrate that a smaller size and slower internalization rate enabled higher tissue penetration and more cell killing. The results also illustrate the benefits of linking an albumin-binding domain to the single-domain ADCs. A construct lacking an albumin-binding domain was rapidly cleared, leading to lower tumor uptake (%ID/g) and decreased in vivo efficacy. In conclusion, these results provide evidence that reaching the maximum number of cells with a lethal payload dose correlates more strongly with in vivo efficacy than total tumor uptake or in vitro potency alone for these protein-drug conjugates. Computational modeling and protein engineering can be used to custom design an optimal framework for controlling internalization, clearance, and tissue penetration to maximize cell killing. SIGNIFICANCE: A mechanistic study of protein-drug conjugates demonstrates that a lower potency compound is more effective in vivo than other agents with equal tumor uptake due to improved tissue penetration and cellular distribution.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Inmunoconjugados/farmacocinética , Modelos Biológicos , Neoplasias de la Próstata/tratamiento farmacológico , Anticuerpos de Dominio Único/farmacología , Animales , Antineoplásicos Alquilantes/química , Antineoplásicos Alquilantes/uso terapéutico , Línea Celular Tumoral , Simulación por Computador , Femenino , Humanos , Inmunoconjugados/química , Inmunoconjugados/uso terapéutico , Masculino , Ratones , Microscopía Confocal , Neoplasias de la Próstata/diagnóstico por imagen , Neoplasias de la Próstata/patología , Anticuerpos de Dominio Único/química , Anticuerpos de Dominio Único/uso terapéutico , Esferoides Celulares , Relación Estructura-Actividad , Distribución Tisular , Ensayos Antitumor por Modelo de Xenoinjerto
2.
J Mol Cell Cardiol ; 117: 1-18, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29470978

RESUMEN

The shortening of sarcomeres that co-ordinates the pump function of the heart is stimulated by electrically-mediated increases in [Ca2+]. This process of excitation-contraction coupling (ECC) is subject to modulation by neurohormonal mediators that tune the output of the heart to meet the needs of the organism. Endothelin-1 (ET-1) is a potent modulator of cardiac function with effects on contraction amplitude, chronotropy and automaticity. The actions of ET-1 are evident during normal adaptive physiological responses and increased under pathophysiological conditions, such as following myocardial infarction and during heart failure, where ET-1 levels are elevated. In myocytes, ET-1 acts through ETA- or ETB-G protein-coupled receptors (GPCRs). Although well studied in atrial myocytes, the influence and mechanisms of action of ET-1 upon ECC in ventricular myocytes are not fully resolved. We show in rat ventricular myocytes that ET-1 elicits a biphasic effect on fractional shortening (initial transient negative and sustained positive inotropy) and increases the peak amplitude of systolic Ca2+ transients in adult rat ventricular myocytes. The negative inotropic phase was ETB receptor-dependent, whereas the positive inotropic response and increase in peak amplitude of systolic Ca2+ transients required ETA receptor engagement. Both effects of ET-1 required phospholipase C (PLC)-activity, although distinct signalling pathways downstream of PLC elicited the effects of each ET receptor. The negative inotropic response involved inositol 1,4,5-trisphosphate (InsP3) signalling and protein kinase C epsilon (PKCε). The positive inotropic action and the enhancement in Ca2+ transient amplitude induced by ET-1 were independent of InsP3 signalling, but suppressed by PKCε. Serine 302 in cardiac myosin binding protein-C was identified as a PKCε substrate that when phosphorylated contributed to the suppression of contraction and Ca2+ transients by PKCε following ET-1 stimulation. Thus, our data provide a new role and mechanism of action for InsP3 and PKCε in mediating the negative inotropic response and in restraining the positive inotropy and enhancement in Ca2+ transients following ET-1 stimulation.


Asunto(s)
Proteínas Portadoras/metabolismo , Endotelina-1/farmacología , Ventrículos Cardíacos/citología , Contracción Miocárdica , Miocitos Cardíacos/metabolismo , Proteína Quinasa C-epsilon/metabolismo , Animales , Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Cardiotónicos/farmacología , Citosol/metabolismo , Acoplamiento Excitación-Contracción/efectos de los fármacos , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Masculino , Contracción Miocárdica/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Fosforilación/efectos de los fármacos , Proteína Quinasa C-epsilon/antagonistas & inhibidores , Ratas Wistar , Receptores de Endotelina/metabolismo , Retículo Sarcoplasmático/efectos de los fármacos , Retículo Sarcoplasmático/metabolismo , Fosfolipasas de Tipo C/metabolismo
3.
J Physiol ; 595(5): 1533-1546, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-27859356

RESUMEN

KEY POINTS: In human sperm, proton flux across the membrane is controlled by the voltage-gated proton channel Hv1. We show that sperm harbour both Hv1 and an N-terminally cleaved isoform termed Hv1Sper. The pH-control of Hv1Sper and Hv1 is distinctively different. Hv1Sper and Hv1 can form heterodimers that combine features of both constituents. Cleavage and heterodimerization of Hv1 might represent an adaptation to the specific requirements of pH control in sperm. ABSTRACT: In human sperm, the voltage-gated proton channel Hv1 controls the flux of protons across the flagellar membrane. Here, we show that sperm harbour Hv1 and a shorter isoform, termed Hv1Sper. Hv1Sper is generated from Hv1 by removal of 68 amino acids from the N-terminus by post-translational proteolytic cleavage. The pH-dependent gating of the channel isoforms is distinctly different. In both Hv1 and Hv1Sper, the conductance-voltage relationship is determined by the pH difference across the membrane (∆pH). However, simultaneous changes in intracellular and extracellular pH that leave ΔpH constant strongly shift the activation curve of Hv1Sper but not that of Hv1, demonstrating that cleavage of the N-terminus tunes pH sensing in Hv1. Moreover, we show that Hv1 and Hv1Sper assemble as heterodimers that combine features of both constituents. We suggest that cleavage and heterodimerization of Hv1 represents an adaptation to the specific requirements of pH control in sperm.


Asunto(s)
Activación del Canal Iónico/fisiología , Canales Iónicos/fisiología , Espermatozoides/fisiología , Animales , Línea Celular , Células HEK293 , Humanos , Concentración de Iones de Hidrógeno , Canales Iónicos/metabolismo , Masculino , Ratones Endogámicos C57BL , Oocitos/fisiología , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Mucosa Respiratoria , Serina Proteasas/metabolismo , Inhibidores de Serina Proteinasa/farmacología , Espermatozoides/efectos de los fármacos , Espermatozoides/metabolismo , Sulfonas/farmacología , Xenopus laevis
4.
EMBO J ; 34(3): 379-92, 2015 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-25535245

RESUMEN

Sperm guidance is controlled by chemical and physical cues. In many species, Ca(2+) bursts in the flagellum govern navigation to the egg. In Arbacia punctulata, a model system of sperm chemotaxis, a cGMP signaling pathway controls these Ca(2+) bursts. The underlying Ca(2+) channel and its mechanisms of activation are unknown. Here, we identify CatSper Ca(2+) channels in the flagellum of A. punctulata sperm. We show that CatSper mediates the chemoattractant-evoked Ca(2+) influx and controls chemotactic steering; a concomitant alkalization serves as a highly cooperative mechanism that enables CatSper to transduce periodic voltage changes into Ca(2+) bursts. Our results reveal intriguing phylogenetic commonalities but also variations between marine invertebrates and mammals regarding the function and control of CatSper. The variations probably reflect functional and mechanistic adaptations that evolved during the transition from external to internal fertilization.


Asunto(s)
Canales de Calcio/metabolismo , Señalización del Calcio/fisiología , Quimiotaxis/fisiología , Evolución Molecular , Potenciales de la Membrana/fisiología , Erizos de Mar/metabolismo , Animales , Canales de Calcio/genética , Masculino , Erizos de Mar/genética
5.
J Cell Biol ; 206(4): 541-57, 2014 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-25135936

RESUMEN

Guanylyl cyclases (GCs), which synthesize the messenger cyclic guanosine 3',5'-monophosphate, control several sensory functions, such as phototransduction, chemosensation, and thermosensation, in many species from worms to mammals. The GC chemoreceptor in sea urchin sperm can decode chemoattractant concentrations with single-molecule sensitivity. The molecular and cellular underpinnings of such ultrasensitivity are not known for any eukaryotic chemoreceptor. In this paper, we show that an exquisitely high density of 3 × 10(5) GC chemoreceptors and subnanomolar ligand affinity provide a high ligand-capture efficacy and render sperm perfect absorbers. The GC activity is terminated within 150 ms by dephosphorylation steps of the receptor, which provides a means for precise control of the GC lifetime and which reduces "molecule noise." Compared with other ultrasensitive sensory systems, the 10-fold signal amplification by the GC receptor is surprisingly low. The hallmarks of this signaling mechanism provide a blueprint for chemical sensing in small compartments, such as olfactory cilia, insect antennae, or even synaptic boutons.


Asunto(s)
Arbacia/metabolismo , GMP Cíclico/biosíntesis , Guanilato Ciclasa/metabolismo , Receptores Acoplados a la Guanilato-Ciclasa/metabolismo , Espermatozoides/metabolismo , Animales , Células Quimiorreceptoras/metabolismo , Factores Quimiotácticos/fisiología , Células HEK293 , Humanos , Masculino , Fosforilación , Unión Proteica , Transducción de Señal
6.
EMBO J ; 31(7): 1654-65, 2012 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-22354039

RESUMEN

The sperm-specific CatSper channel controls the intracellular Ca(2+) concentration ([Ca(2+)](i)) and, thereby, the swimming behaviour of sperm. In humans, CatSper is directly activated by progesterone and prostaglandins-female factors that stimulate Ca(2+) influx. Other factors including neurotransmitters, chemokines, and odorants also affect sperm function by changing [Ca(2+)](i). Several ligands, notably odorants, have been proposed to control Ca(2+) entry and motility via G protein-coupled receptors (GPCRs) and cAMP-signalling pathways. Here, we show that odorants directly activate CatSper without involving GPCRs and cAMP. Moreover, membrane-permeable analogues of cyclic nucleotides that have been frequently used to study cAMP-mediated Ca(2+) signalling also activate CatSper directly via an extracellular site. Thus, CatSper or associated protein(s) harbour promiscuous binding sites that can host various ligands. These results contest current concepts of Ca(2+) signalling by GPCR and cAMP in mammalian sperm: ligands thought to activate metabotropic pathways, in fact, act via a common ionotropic mechanism. We propose that the CatSper channel complex serves as a polymodal sensor for multiple chemical cues that assist sperm during their voyage across the female genital tract.


Asunto(s)
Canales de Calcio/metabolismo , Señalización del Calcio/fisiología , Feromonas/metabolismo , Espermatozoides/metabolismo , Aldehídos/farmacología , Bencimidazoles/farmacología , Bloqueadores de los Canales de Calcio/farmacología , Señalización del Calcio/efectos de los fármacos , AMP Cíclico/metabolismo , Ciclopropanos/farmacología , Humanos , Masculino , Mibefradil/farmacología , Naftalenos/farmacología , Receptores Acoplados a Proteínas G/metabolismo , Espermatozoides/efectos de los fármacos
7.
Nature ; 471(7338): 382-6, 2011 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-21412338

RESUMEN

In the oviduct, cumulus cells that surround the oocyte release progesterone. In human sperm, progesterone stimulates a Ca(2+) increase by a non-genomic mechanism. The Ca(2+) signal has been proposed to control chemotaxis, hyperactivation and acrosomal exocytosis of sperm. However, the underlying signalling mechanism has remained mysterious. Here we show that progesterone activates the sperm-specific, pH-sensitive CatSper Ca(2+) channel. We found that both progesterone and alkaline pH stimulate a rapid Ca(2+) influx with almost no latency, incompatible with a signalling pathway involving metabotropic receptors and second messengers. The Ca(2+) signals evoked by alkaline pH and progesterone are inhibited by the Ca(v) channel blockers NNC 55-0396 and mibefradil. Patch-clamp recordings from sperm reveal an alkaline-activated current carried by mono- and divalent ions that exhibits all the hallmarks of sperm-specific CatSper Ca(2+) channels. Progesterone substantially enhances the CatSper current. The alkaline- and progesterone-activated CatSper current is inhibited by both drugs. Our results resolve a long-standing controversy over the non-genomic progesterone signalling. In human sperm, either the CatSper channel itself or an associated protein serves as the non-genomic progesterone receptor. The identification of CatSper channel blockers will greatly facilitate the study of Ca(2+) signalling in sperm and help to define further the physiological role of progesterone and CatSper.


Asunto(s)
Canales de Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Calcio/metabolismo , Progesterona/farmacología , Espermatozoides/efectos de los fármacos , Espermatozoides/metabolismo , Alprostadil/farmacología , Bencimidazoles/farmacología , Bloqueadores de los Canales de Calcio/farmacología , AMP Cíclico , Ciclopropanos/farmacología , Conductividad Eléctrica , Humanos , Concentración de Iones de Hidrógeno , Masculino , Mibefradil/farmacología , Naftalenos/farmacología , Técnicas de Placa-Clamp , Progesterona/metabolismo
8.
J Am Chem Soc ; 131(11): 4027-30, 2009 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-19256499

RESUMEN

Ketalization of the biomolecule progesterone with (6-bromo-7-hydroxycoumarin-4-yl)ethane-1,2-diol gives the photolabile progesterone derivatives 3 and 4. These compounds display dramatically reduced bioactivity and release progesterone upon irradiation with UV/vis or IR light. In particular, 4 can be used to perform concentration-jump experiments with high temporal and spatial resolution that allows one to study elegantly the mechanisms of rapid nongenomic cellular events evoked by progesterone. The usefulness of 4 was demonstrated by measurement of changes in swimming behavior of single human sperm caused by progesterone-induced Ca(2+) influx in the sperm flagellum.


Asunto(s)
Procesos Fotoquímicos , Progesterona/análogos & derivados , Progesterona/farmacología , Alcoholes , Calcio/metabolismo , Flagelos/efectos de los fármacos , Humanos , Luz , Masculino , Progesterona/química , Progesterona/efectos de la radiación , Proyectos de Investigación , Espermatozoides/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA