Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 713
Filtrar
1.
Proc Natl Acad Sci U S A ; 121(16): e2309211121, 2024 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-38593081

RESUMEN

Vesicular release of neurotransmitters and hormones relies on the dynamic assembly of the exocytosis/trans-SNARE complex through sequential interactions of synaptobrevins, syntaxins, and SNAP-25. Despite SNARE-mediated release being fundamental for intercellular communication in all excitable tissues, the role of auxiliary proteins modulating the import of reserve vesicles to the active zone, and thus, scaling repetitive exocytosis remains less explored. Secretagogin is a Ca2+-sensor protein with SNAP-25 being its only known interacting partner. SNAP-25 anchors readily releasable vesicles within the active zone, thus being instrumental for 1st phase release. However, genetic deletion of secretagogin impedes 2nd phase release instead, calling for the existence of alternative protein-protein interactions. Here, we screened the secretagogin interactome in the brain and pancreas, and found syntaxin-4 grossly overrepresented. Ca2+-loaded secretagogin interacted with syntaxin-4 at nanomolar affinity and 1:1 stoichiometry. Crystal structures of the protein complexes revealed a hydrophobic groove in secretagogin for the binding of syntaxin-4. This groove was also used to bind SNAP-25. In mixtures of equimolar recombinant proteins, SNAP-25 was sequestered by secretagogin in competition with syntaxin-4. Kd differences suggested that secretagogin could shape unidirectional vesicle movement by sequential interactions, a hypothesis supported by in vitro biological data. This mechanism could facilitate the movement of transport vesicles toward release sites, particularly in the endocrine pancreas where secretagogin, SNAP-25, and syntaxin-4 coexist in both α- and ß-cells. Thus, secretagogin could modulate the pace and fidelity of vesicular hormone release by differential protein interactions.


Asunto(s)
Fusión de Membrana , Secretagoginas , Proteínas Qa-SNARE/genética , Proteínas Qa-SNARE/metabolismo , Secretagoginas/metabolismo , Membrana Celular/metabolismo , Proteína 25 Asociada a Sinaptosomas/metabolismo , Exocitosis , Comunicación Celular , Sintaxina 1/metabolismo , Unión Proteica
2.
Proc Natl Acad Sci U S A ; 121(2): e2309161121, 2024 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-38170748

RESUMEN

In neuronal cell types, vesicular exocytosis is governed by the SNARE (soluble NSF attachment receptor) complex consisting of synaptobrevin2, SNAP25, and syntaxin1. These proteins are required for vesicle priming and fusion. We generated an improved SNAP25-based SNARE COmplex Reporter (SCORE2) incorporating mCeruelan3 and Venus and overexpressed it in SNAP25 knockout embryonic mouse chromaffin cells. This construct rescues vesicle fusion with properties indistinguishable from fusion in wild-type cells. Combining electrochemical imaging of individual release events using electrochemical detector arrays with total internal reflection fluorescence resonance energy transfer (TIR-FRET) imaging reveals a rapid FRET increase preceding individual fusion events by 65 ms. The experiments are performed under conditions of a steady-state cycle of docking, priming, and fusion, and the delay suggests that the FRET change reflects tight docking and priming of the vesicle, followed by fusion after ~65 ms. Given the absence of wt SNAP25, SCORE2 allows determination of the number of molecules at fusion sites and the number that changes conformation. The number of SNAP25 molecules changing conformation in the priming step increases with vesicle size and SNAP25 density in the plasma membrane and equals the number of copies present in the vesicle-plasma membrane contact zone. We estimate that in wt cells, 6 to 7 copies of SNAP25 change conformation during the priming step.


Asunto(s)
Células Cromafines , Proteínas SNARE , Animales , Ratones , Membrana Celular/metabolismo , Células Cromafines/metabolismo , Exocitosis/fisiología , Fusión de Membrana/fisiología , Proteínas SNARE/metabolismo , Proteína 25 Asociada a Sinaptosomas/genética , Proteína 25 Asociada a Sinaptosomas/metabolismo
3.
Commun Biol ; 7(1): 34, 2024 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-38182732

RESUMEN

SNARE-mediated vesicular transport is thought to play roles in photoreceptor glutamate exocytosis and photopigment delivery. However, the functions of Synaptosomal-associated protein (SNAP) isoforms in photoreceptors are unknown. Here, we revisit the expression of SNAP-23 and SNAP-25 and generate photoreceptor-specific knockout mice to investigate their roles. Although we find that SNAP-23 shows weak mRNA expression in photoreceptors, SNAP-23 removal does not affect retinal morphology or vision. SNAP-25 mRNA is developmentally regulated and undergoes mRNA trafficking to photoreceptor inner segments at postnatal day 9 (P9). SNAP-25 knockout photoreceptors develop normally until P9 but degenerate by P14 resulting in severe retinal thinning. Photoreceptor loss in SNAP-25 knockout mice is associated with abolished electroretinograms and vision loss. We find mistrafficked photopigments, enlarged synaptic vesicles, and abnormal synaptic ribbons which potentially underlie photoreceptor degeneration. Our results conclude that SNAP-25, but not SNAP-23, mediates photopigment delivery and synaptic functioning required for photoreceptor development, survival, and function.


Asunto(s)
Células Fotorreceptoras de Vertebrados , Proteínas Qb-SNARE , Proteínas Qc-SNARE , Proteína 25 Asociada a Sinaptosomas , Animales , Ratones , Transporte Biológico , Citoesqueleto , Ácido Glutámico , Ratones Noqueados , ARN Mensajero , Proteínas Qb-SNARE/metabolismo , Proteínas Qc-SNARE/metabolismo , Proteína 25 Asociada a Sinaptosomas/metabolismo , Células Fotorreceptoras de Vertebrados/citología , Células Fotorreceptoras de Vertebrados/metabolismo
4.
Cell Commun Signal ; 21(1): 356, 2023 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-38102610

RESUMEN

BACKGROUND: Synaptosomal-associated protein 25 (SNAP25) exerts protective effects against postoperative cognitive dysfunction (POCD) by promoting PTEN-induced kinase 1 (PINK1)/Parkin-mediated mitophagy and repressing caspase-3/gasdermin E (GSDME)-mediated pyroptosis. However, the regulatory mechanisms of SNAP25 protein remain unclear. METHODS: We employed recombinant adeno-associated virus 9 (AAV9)-hSyn to knockdown tumor necrosis factor α-induced protein 1 (TNFAIP1) or SNAP25 and investigate the role of TNFAIP1 in POCD. Cognitive performance, hippocampal injury, mitophagy, and pyroptosis were assessed. Co-immunoprecipitation (co-IP) and ubiquitination assays were conducted to elucidate the mechanisms by which TNFAIP1 stabilizes SNAP25. RESULTS: Our results demonstrated that the ubiquitin ligase TNFAIP1 was upregulated in the hippocampus of mice following isoflurane (Iso) anesthesia and laparotomy. The N-terminal region (residues 1-96) of TNFAIP1 formed a conjugate with SNAP25, leading to lysine (K) 48-linked polyubiquitination of SNAP25 at K69. Silencing TNFAIP1 enhanced SH-SY5Y cell viability and conferred antioxidant, pro-mitophagy, and anti-pyroptosis properties in response to Iso and lipopolysaccharide (LPS) challenges. Conversely, TNFAIP1 overexpression reduced HT22 cell viability, increased reactive oxygen species (ROS) accumulation, impaired PINK1/Parkin-dependent mitophagy, and induced caspase-3/GSDME-dependent pyroptosis by suppressing SNAP25 expression. Neuron-specific knockdown of TNFAIP1 ameliorated POCD, restored mitophagy, and reduced pyroptosis, which was reversed by SNAP25 depletion. CONCLUSIONS: In summary, our findings demonstrated that inhibiting TNFAIP1-mediated degradation of SNAP25 might be a promising therapeutic approach for mitigating postoperative cognitive decline. Video Abstract.


Asunto(s)
Neuroblastoma , Complicaciones Cognitivas Postoperatorias , Humanos , Ratones , Animales , Complicaciones Cognitivas Postoperatorias/metabolismo , Complicaciones Cognitivas Postoperatorias/patología , Proteína 25 Asociada a Sinaptosomas/metabolismo , Caspasa 3/metabolismo , Proteínas Quinasas/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Mitocondrias/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Neuronas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo
5.
Anal Chem ; 94(44): 15307-15314, 2022 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-36301050

RESUMEN

Impaired insulin release is the key feature of type 2 diabetes. Insulin secretion, mainly mediated by SNARE proteins, is closely related to the blood glucose level. However, the mechanism underlying how glucose controls SNARE proteins to regulate insulin release is largely unexplained. Herein, we investigated the effects of glucose on the subcellular localization and spatial distribution on the plasma membrane (PM) of t-SNAREs (SNAP-25 and STX-1A) using a live-cell confocal microscope and the single-molecule localization imaging technique. Live-cell confocal and dSTORM imaging first revealed that SNAP-25 was mostly localized to the PM as clusters under the basal glucose concentration condition and demonstrated significant colocalization with STX-1A clusters. Furthermore, our data showed that the elevated glucose concentration increased the expression of SNAP-25 and induced more and larger SNAP-25 clusters on the PM, whereas glucotoxicity severely inhibited SNAP-25 transport to the PM and caused fewer and smaller SNAP-25 clusters on the PM. Additionally, we found that glucotoxicity also had an inhibitory effect on the colocalization between SNAP-25 and STX-1A, indicating a decrease of their interactions. Our study sheds light on the regulatory effects of glucose on the functional organization of t-SNAREs at a subcellular and molecular level, thus providing new insights into the mechanisms by which SNAREs regulate insulin release.


Asunto(s)
Diabetes Mellitus Tipo 2 , Insulina , Humanos , Insulina/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Proteína 25 Asociada a Sinaptosomas/metabolismo , Secreción de Insulina , Membrana Celular/metabolismo , Glucosa/metabolismo
6.
Int J Mol Sci ; 23(19)2022 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-36232926

RESUMEN

Recent animal experiments suggested that centrally transported botulinum toxin type A (BoNT-A) might reduce an abnormal muscle tone, though with an unknown contribution to the dominant peripheral muscular effect observed clinically. Herein, we examined if late BoNT-A antispastic actions persist due to possible central toxin actions in rats. The early effect of intramuscular (i.m.) BoNT-A (5, 2 and 1 U/kg) on a reversible tetanus toxin (TeNT)-induced calf muscle spasm was examined 7 d post-TeNT and later during recovery from flaccid paralysis (TeNT reinjected on day 49 post-BoNT-A). Lumbar intrathecal (i.t.) BoNT-A-neutralizing antiserum was used to discriminate the transcytosis-dependent central toxin action of 5 U/kg BoNT-A. BoNT-A-truncated synaptosomal-associated protein 25 immunoreactivity was examined in the muscles and spinal cord at day 71 post-BoNT-A. All doses (5, 2 and 1 U/kg) induced similar antispastic actions in the early period (days 1-14) post-BoNT-A. After repeated TeNT, only the higher two doses prevented the muscle spasm and associated locomotor deficit. Central trans-synaptic activity contributed to the late antispastic effect of 5 U/kg BoNT-A. Ongoing BoNT-A enzymatic activity was present in both injected muscle and the spinal cord. These observations suggest that the treatment duration in sustained or intermittent muscular hyperactivity might be maintained by higher doses and combined peripheral and central BoNT-A action.


Asunto(s)
Toxinas Botulínicas Tipo A , Animales , Toxinas Botulínicas Tipo A/farmacología , Hipertonía Muscular/tratamiento farmacológico , Ratas , Espasmo/tratamiento farmacológico , Proteína 25 Asociada a Sinaptosomas/metabolismo , Toxina Tetánica/metabolismo , Toxina Tetánica/farmacología
7.
Life Sci ; 309: 120995, 2022 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-36167148

RESUMEN

Ca2+-triggered neurotransmitter release involves complex regulatory mechanisms, including a series of protein-protein interactions. Three proteins, synaptobrevin (VAMP), synaptosomal-associated protein of 25kDa (SNAP-25) and syntaxin, constitute the soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) core complex that plays key roles in controlling vesicle fusion and exocytosis. Many other proteins participate in the regulation of the processes via direct and/or indirect interaction with the SNARE complex. Although much effort has been made, the regulatory mechanism for exocytosis is still not completely clear. Accumulated evidence indicates that the small GTPase Rab3 and synaptotagmin proteins play important regulatory roles during vesicle fusion and neurotransmitter release. This review outlines our present understanding of the two regulatory proteins, with the focus on the interaction of Rab3 with synaptotagmin in the regulatory process.


Asunto(s)
Proteínas de Unión al GTP Monoméricas , Proteínas del Tejido Nervioso , Sinaptotagminas , Proteínas Solubles de Unión al Factor Sensible a la N-Etilmaleimida/metabolismo , Proteína 25 Asociada a Sinaptosomas/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Exocitosis/fisiología , Proteínas R-SNARE , Proteínas Qa-SNARE/metabolismo , Neurotransmisores , Proteínas de Unión al GTP Monoméricas/metabolismo , Calcio/metabolismo
8.
Elife ; 112022 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-36173100

RESUMEN

Exocytosis of secretory vesicles requires the soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) proteins and small GTPase Rabs. As a Rab3/Rab27 effector protein on secretory vesicles, Rabphilin 3A was implicated to interact with SNAP-25 to regulate vesicle exocytosis in neurons and neuroendocrine cells, yet the underlying mechanism remains unclear. In this study, we have characterized the physiologically relevant binding sites between Rabphilin 3A and SNAP-25. We found that an intramolecular interplay between the N-terminal Rab-binding domain and C-terminal C2AB domain enables Rabphilin 3A to strongly bind the SNAP-25 N-peptide region via its C2B bottom α-helix. Disruption of this interaction significantly impaired docking and fusion of vesicles with the plasma membrane in rat PC12 cells. In addition, we found that this interaction allows Rabphilin 3A to accelerate SNARE complex assembly. Furthermore, we revealed that this interaction accelerates SNARE complex assembly via inducing a conformational switch from random coils to α-helical structure in the SNAP-25 SNARE motif. Altogether, our data suggest that the promotion of SNARE complex assembly by binding the C2B bottom α-helix of Rabphilin 3A to the N-peptide of SNAP-25 underlies a pre-fusion function of Rabphilin 3A in vesicle exocytosis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Exocitosis , Fusión de Membrana , Proteínas del Tejido Nervioso , Proteína 25 Asociada a Sinaptosomas , Proteínas de Transporte Vesicular , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Exocitosis/fisiología , Proteínas del Tejido Nervioso/metabolismo , Ratas , Proteínas SNARE/metabolismo , Vesículas Secretoras/metabolismo , Proteínas Solubles de Unión al Factor Sensible a la N-Etilmaleimida/metabolismo , Proteína 25 Asociada a Sinaptosomas/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Proteínas de Unión al GTP rab3/metabolismo
9.
Toxins (Basel) ; 14(2)2022 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-35202143

RESUMEN

Chimeras of botulinum neurotoxin (BoNT) serotype A (/A) combined with /E protease might possess improved analgesic properties relative to either parent, due to inheriting the sensory neurotropism of the former with more extensive disabling of SNAP-25 from the latter. Hence, fusions of /E protease light chain (LC) to whole BoNT/A (LC/E-BoNT/A), and of the LC plus translocation domain (HN) of /E with the neuronal acceptor binding moiety (HC) of /A (BoNT/EA), created previously by gene recombination and expression in E. coli., were used. LC/E-BoNT/A (75 units/kg) injected into the whisker pad of rats seemed devoid of systemic toxicity, as reflected by an absence of weight loss, but inhibited the nocifensive behavior (grooming, freezing, and reduced mobility) induced by activating TRPV1 with capsaicin, injected at various days thereafter. No sex-related differences were observed. c-Fos expression was increased five-fold in the trigeminal nucleus caudalis ipsi-lateral to capsaicin injection, relative to the contra-lateral side and vehicle-treated controls, and this increase was virtually prevented by LC/E-BoNT/A. In vitro, LC/E-BoNT/A or /EA diminished CGRP exocytosis from rat neonate trigeminal ganglionic neurons stimulated with up to 1 µM capsaicin, whereas BoNT/A only substantially reduced the release in response to 0.1 µM or less of the stimulant, in accordance with the /E protease being known to prevent fusion of exocytotic vesicles.


Asunto(s)
Analgésicos/farmacología , Toxinas Botulínicas Tipo A/farmacología , Capsaicina/farmacología , Neurotoxinas/farmacología , Células Receptoras Sensoriales/efectos de los fármacos , Animales , Animales Recién Nacidos , Conducta Animal/efectos de los fármacos , Péptido Relacionado con Gen de Calcitonina/metabolismo , Femenino , Masculino , Dolor/inducido químicamente , Dolor/tratamiento farmacológico , Dolor/metabolismo , Ratas Sprague-Dawley , Células Receptoras Sensoriales/metabolismo , Proteína 25 Asociada a Sinaptosomas/metabolismo , Ganglio del Trigémino/citología
10.
Int J Mol Sci ; 23(2)2022 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-35055082

RESUMEN

Nerve growth factor (NGF) is known to intensify pain in various ways, so perturbing pertinent effects without negating its essential influences on neuronal functions could help the search for much-needed analgesics. Towards this goal, cultured neurons from neonatal rat trigeminal ganglia-a locus for craniofacial sensory nerves-were used to examine how NGF affects the Ca2+-dependent release of a pain mediator, calcitonin gene-related peptide (CGRP), that is triggered by activating a key signal transducer, transient receptor potential vanilloid 1 (TRPV1) with capsaicin (CAP). Measurements utilised neurons fed with or deprived of NGF for 2 days. Acute re-introduction of NGF induced Ca2+-dependent CGRP exocytosis that was inhibited by botulinum neurotoxin type A (BoNT/A) or a chimera of/E and/A (/EA), which truncated SNAP-25 (synaptosomal-associated protein with Mr = 25 k) at distinct sites. NGF additionally caused a Ca2+-independent enhancement of the neuropeptide release evoked by low concentrations (<100 nM) of CAP, but only marginally increased the peak response to ≥100 nM. Notably, BoNT/A inhibited CGRP exocytosis evoked by low but not high CAP concentrations, whereas/EA effectively reduced responses up to 1 µM CAP and inhibited to a greater extent its enhancement by NGF. In addition to establishing that sensitisation of sensory neurons to CAP by NGF is dependent on SNARE-mediated membrane fusion, insights were gleaned into the differential ability of two regions in the C-terminus of SNAP-25 (181-197 and 198-206) to support CAP-evoked Ca2+-dependent exocytosis at different intensities of stimulation.


Asunto(s)
Péptido Relacionado con Gen de Calcitonina/biosíntesis , Capsaicina/farmacología , Factor de Crecimiento Nervioso/metabolismo , Células Receptoras Sensoriales/efectos de los fármacos , Células Receptoras Sensoriales/metabolismo , Ganglio del Trigémino/efectos de los fármacos , Ganglio del Trigémino/metabolismo , Animales , Toxinas Botulínicas Tipo A/farmacología , Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Exocitosis/efectos de los fármacos , Factor de Crecimiento Nervioso/farmacología , Proteolisis , Ratas , Proteína 25 Asociada a Sinaptosomas/metabolismo
11.
Proc Natl Acad Sci U S A ; 118(49)2021 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-34857632

RESUMEN

Syntaxin helps in catalyzing membrane fusion during exocytosis. It also forms clusters in the plasma membrane, where both its transmembrane and SNARE domains are thought to homo-oligomerize. To study syntaxin clustering in live PC12 cells, we labeled granules with neuropeptide-Y-mCherry and syntaxin clusters with syntaxin-1a green fluorescent protein (GFP). Abundant clusters appeared under total internal reflection (TIRF) illumination, and some of them associated with granules ("on-granule clusters"). Syntaxin-1a-GFP or its mutants were expressed at low levels and competed with an excess of endogenous syntaxin for inclusion into clusters. On-granule inclusion was diminished by mutations known to inhibit binding to Munc18-1 in vitro. Knock-down of Munc18-1 revealed Munc18-dependent and -independent on-granule clustering. Clustering was inhibited by mutations expected to break salt bridges between syntaxin's Hb and SNARE domains and was rescued by additional mutations expected to restore them. Most likely, syntaxin is in a closed conformation when it clusters on granules, and its SNARE and Hb domains approach to within atomic distances. Pairwise replacements of Munc18-contacting residues with alanines had only modest effects, except that the pair R114A/I115A essentially abolished on-granule clustering. In summary, an on-granule cluster arises from the specific interaction between a granule and a dense cluster of syntaxin-Munc18-1 complexes. Off-granule clusters, by contrast, were resistant to even the strongest mutations we tried and required neither Munc18-1 nor the presence of a SNARE domain. They may well form through the nonstoichiometric interactions with membrane lipids that others have observed in cell-free systems.


Asunto(s)
Membrana Celular/metabolismo , Proteínas Munc18/metabolismo , Proteínas Qa-SNARE/metabolismo , Proteína 25 Asociada a Sinaptosomas/metabolismo , Glándulas Suprarrenales/citología , Animales , Membrana Celular/química , Regulación de la Expresión Génica/fisiología , Modelos Moleculares , Proteínas Munc18/genética , Mutación , Células PC12 , Unión Proteica , Conformación Proteica , Proteínas Qa-SNARE/genética , Ratas , Proteína 25 Asociada a Sinaptosomas/genética
12.
Elife ; 102021 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-34779769

RESUMEN

SNARE proteins have been described as the effectors of fusion events in the secretory pathway more than two decades ago. The strong interactions between SNARE domains are clearly important in membrane fusion, but it is unclear whether they are involved in any other cellular processes. Here, we analyzed two classical SNARE proteins, syntaxin 1A and SNAP25. Although they are supposed to be engaged in tight complexes, we surprisingly find them largely segregated in the plasma membrane. Syntaxin 1A only occupies a small fraction of the plasma membrane area. Yet, we find it is able to redistribute the far more abundant SNAP25 on the mesoscale by gathering crowds of SNAP25 molecules onto syntaxin clusters in a SNARE-domain-dependent manner. Our data suggest that SNARE domain interactions are not only involved in driving membrane fusion on the nanoscale, but also play an important role in controlling the general organization of proteins on the mesoscale. Further, we propose these mechanisms preserve active syntaxin 1A-SNAP25 complexes at the plasma membrane.


Asunto(s)
Proteínas SNARE/genética , Proteína 25 Asociada a Sinaptosomas/genética , Sintaxina 1/genética , Animales , Células Hep G2 , Humanos , Células PC12 , Mapas de Interacción de Proteínas , Ratas , Proteínas SNARE/metabolismo , Proteína 25 Asociada a Sinaptosomas/metabolismo , Sintaxina 1/metabolismo
13.
Pharmacol Res Perspect ; 9(5): e00857, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34632725

RESUMEN

Clinically used botulinum neurotoxins (BoNTs) are natural products of Clostridium botulinum. A novel, recombinant BoNT type A1 (rBoNT/A1; IPN10260) has been synthesized using the native amino acid sequence expressed in Escherichia coli and has previously been characterized in vitro and ex vivo. Here, we aimed to characterize rBoNT/A1 in vivo and evaluate its effects on skeletal muscle. The properties of rBoNT/A1 following single, intramuscular administration were evaluated in the mouse and rat digit abduction score (DAS) assays and compared with those of natural BoNT/A1 (nBoNT/A1). rBoNT/A1-injected tibialis anterior was assessed in the in situ muscle force test in rats. rBoNT/A1-injected gastrocnemius lateralis (GL) muscle was assessed in the compound muscle action potential (CMAP) test in rats. The rBoNT/A1-injected GL muscle was evaluated for muscle weight, volume, myofiber composition and immunohistochemical detection of cleaved SNAP25 (c-SNAP25). Results showed that rBoNT/A1 and nBoNT/A1 were equipotent and had similar onset and duration of action in both mouse and rat DAS assays. rBoNT/A1 caused a dose-dependent inhibition of muscle force and a rapid long-lasting reduction in CMAP amplitude that lasted for at least 30 days. Dose-dependent reductions in GL weight and volume and increases in myofiber atrophy were accompanied by immunohistochemical detection of c-SNAP25. Overall, rBoNT/A1 and nBoNT/A1 exhibited similar properties following intramuscular administration. rBoNT/A1 inhibited motoneurons neurotransmitter release, which was robust, long-lasting, and accompanied by cleavage of SNAP25. rBoNT/A1 is a useful tool molecule for comparison with current natural and future modified recombinant neurotoxins products.


Asunto(s)
Toxinas Botulínicas Tipo A/farmacología , Fibras Musculares Esqueléticas/efectos de los fármacos , Músculo Esquelético/efectos de los fármacos , Proteínas Recombinantes/farmacología , Potenciales de Acción/efectos de los fármacos , Animales , Inyecciones Intramusculares , Ratones , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patología , Fuerza Muscular/efectos de los fármacos , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Tamaño de los Órganos , Ratas , Proteína 25 Asociada a Sinaptosomas/efectos de los fármacos , Proteína 25 Asociada a Sinaptosomas/metabolismo
14.
Int J Mol Sci ; 22(20)2021 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-34681775

RESUMEN

Botulinum neurotoxin serotype A (BoNT/A) is the most potent protein toxin to humans. BoNT/A light chain (LC/A) cleavage of the membrane-bound SNAP-25 has been well-characterized, but how LC/A traffics to the plasma membrane to target SNAP-25 is unknown. Of the eight BoNT/A subtypes (A1-A8), LC/A3 has a unique short duration of action and low potency that correlate to the intracellular steady state of LC/A, where LC/A1 is associated with the plasma membrane and LC/A3 is present in the cytosol. Steady-state and live imaging of LC/A3-A1 chimeras identified a two-step process where the LC/A N terminus bound intracellular vesicles, which facilitated an internal α-helical-rich domain to mediate LC/A plasma membrane association. The propensity of LC/A variants for membrane association correlated with enhanced BoNT/A potency. Understanding the basis for light chain intracellular localization provides insight to mechanisms underlying BoNT/A potency, which can be extended to applications as a human therapy.


Asunto(s)
Toxinas Botulínicas Tipo A/metabolismo , Membrana Celular/metabolismo , Membranas Intracelulares/metabolismo , Animales , Toxinas Botulínicas Tipo A/farmacocinética , Membrana Celular/efectos de los fármacos , Femenino , Humanos , Membranas Intracelulares/efectos de los fármacos , Ratones , Ratones Endogámicos ICR , Unión Proteica , Proteína 25 Asociada a Sinaptosomas/metabolismo , Células Tumorales Cultivadas
15.
Int J Mol Sci ; 22(16)2021 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-34445536

RESUMEN

Chronic pain is a leading health and socioeconomic problem and an unmet need exists for long-lasting analgesics. SNAREs (soluble N-ethylmaleimide-sensitive factor attachment protein receptors) are required for neuropeptide release and noxious signal transducer surface trafficking, thus, selective expression of the SNARE-cleaving light-chain protease of botulinum neurotoxin A (LCA) in peripheral sensory neurons could alleviate chronic pain. However, a safety concern to this approach is the lack of a sensory neuronal promoter to prevent the expression of LCA in the central nervous system. Towards this, we exploit the unique characteristics of Pirt (phosphoinositide-interacting regulator of TRP), which is expressed in peripheral nociceptive neurons. For the first time, we identified a Pirt promoter element and cloned it into a lentiviral vector driving transgene expression selectively in peripheral sensory neurons. Pirt promoter driven-LCA expression yielded rapid and concentration-dependent cleavage of SNAP-25 in cultured sensory neurons. Moreover, the transcripts of pain-related genes (TAC1, tachykinin precursor 1; CALCB, calcitonin gene-related peptide 2; HTR3A, 5-hydroxytryptamine receptor 3A; NPY2R, neuropeptide Y receptor Y2; GPR52, G protein-coupled receptor 52; SCN9A, sodium voltage-gated channel alpha subunit 9; TRPV1 and TRPA1, transient receptor potential cation channel subfamily V member 1 and subfamily A member 1) in pro-inflammatory cytokines stimulated sensory neurons were downregulated by viral mediated expression of LCA. Furthermore, viral expression of LCA yielded long-lasting inhibition of pain mediator release. Thus, we show that the engineered Pirt-LCA virus may provide a novel means for long lasting pain relief.


Asunto(s)
Toxinas Botulínicas Tipo A/farmacología , Neuropéptidos/metabolismo , Dolor/prevención & control , Sistema Nervioso Periférico/metabolismo , Células Receptoras Sensoriales/metabolismo , Proteína 25 Asociada a Sinaptosomas/metabolismo , Animales , Animales Recién Nacidos , Femenino , Humanos , Masculino , Fusión de Membrana , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Nociceptores/efectos de los fármacos , Nociceptores/metabolismo , Dolor/genética , Dolor/metabolismo , Dolor/patología , Sistema Nervioso Periférico/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Células Receptoras Sensoriales/efectos de los fármacos , Proteína 25 Asociada a Sinaptosomas/genética
16.
Biochem Biophys Res Commun ; 570: 162-168, 2021 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-34284142

RESUMEN

Mammalian taste buds comprise types I, II, and III taste cells, with each type having specific characteristics: glia-like supporting cells (type I), taste receptor cells (type II), and presynaptic cells (type III). In this study, to characterize the peripheral taste-sensing systems in chickens, we analyzed the distributions of the mammalian types I, II, and III taste cell markers in chicken taste buds: glutamate-aspartate transporter (GLAST) for type I; taste receptor type 1 members 1 and 3 (T1R1 and T1R3), taste receptor type 2 member 7 (T2R7), and α-gustducin for type II; and synaptosomal protein 25 (SNAP25) and neural cell adhesion molecule (NCAM) for type III. We found that most GLAST+ taste cells expressed α-gustducin and SNAP25 and that high percentages of T1R3+ or α-gustducin+ taste cells expressed SNAP25 and NCAM. These results demonstrated a unique subset of chicken taste cells expressing multiple taste cell type marker proteins. Taken together, these results provide new insights into the taste-sensing mechanisms in vertebrate taste buds.


Asunto(s)
Biomarcadores/metabolismo , Pollos/metabolismo , Mamíferos/metabolismo , Papilas Gustativas/metabolismo , Gusto , Animales , Especificidad de Anticuerpos/inmunología , Moléculas de Adhesión de Célula Nerviosa/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Proteína 25 Asociada a Sinaptosomas/metabolismo , Transducina/metabolismo , Vimentina/metabolismo
17.
J Mol Neurosci ; 71(12): 2593-2607, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34151409

RESUMEN

Alzheimer's disease (AD) is characterized by progressive synaptic dysfunction, deterioration of neuronal transmission, and consequently neuronal death. Although there is no treatment for AD, exposure to enriched environment (EE) in mice, as well as physical and mental activity in human subjects have been shown to have a protective effect by slowing the disease's progression and reducing AD-like cognitive impairment. However, the molecular mechanism of this mitigating effect is still not understood. One of the mechanisms that has recently been shown to be involved in neuronal degeneration is microRNAs (miRNAs) regulation, which act as a post-transcriptional regulators of gene expression. miR-128 has been shown to be significantly altered in individuals with AD and in mice following exposure to EE. Here, we focused on elucidating the possible role of miR-128 in AD pathology and found that miR-128 regulates the expression of two proteins essential for synaptic transmission, SNAP-25, and synaptotagmin1 (Syt1). Clinically relevant, in 5xFAD mouse model for AD, this miRNA's expression was found as downregulated, resembling the alteration found in the hippocampi of individuals with AD. Interestingly, exposing WT mice to EE also resulted in downregulation of miR-128 expression levels, although EE and AD conditions demonstrate opposing effects on neuronal functioning and synaptic plasticity. We also found that miR-128 expression downregulation in primary hippocampal cultures from 5xFAD mice results in increased neuronal network activity and neuronal excitability. Altogether, our findings place miR-128 as a synaptic player that may contribute to synaptic functioning and plasticity through regulation of synaptic protein expression and function.


Asunto(s)
Enfermedad de Alzheimer/genética , Hipocampo/metabolismo , MicroARNs/metabolismo , Sinapsis/metabolismo , Proteína 25 Asociada a Sinaptosomas/genética , Sinaptotagmina I/genética , Enfermedad de Alzheimer/metabolismo , Animales , Células Cultivadas , Hipocampo/citología , Ratones , MicroARNs/genética , Neuronas/metabolismo , Proteína 25 Asociada a Sinaptosomas/metabolismo , Sinaptotagmina I/metabolismo
18.
Behav Brain Res ; 412: 113417, 2021 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-34157371

RESUMEN

ß-adrenoceptor (ß-AR), especially the ß1- and ß2-AR subtypes, is known to participate in stress-related behavioral changes. Recently, SR58611A, a brain-penetrant ß3-AR agonist, exhibits anxiolytic- and antidepressant-like effects. In this study, we sought to study the role of SR58611A in behavioral changes and its potential cellular and molecular mechanism in the prefrontal cortex (PFC). We found that rats with SR58611A (1 mg/kg) enhanced PFC-mediated recognition memory, whereas administration of higher dosage of SR58611A (20 mg/kg) caused hyperlocomotion, and exhibited an impairment effect on recognition memory. Electrophysiological data also indicated that SR58611A (1 mg/kg) selectively enhanced NMDA receptor-mediated excitatory postsynaptic currents (EPSC) through interacting with norepinephrine (NE) system and activating ß3-AR, whereas higher dosage of SR58611A (20 mg/kg) reduced both AMPA receptor- and NMDA receptor-mediated EPSC. SR58611A-induced different effects on EPSC linked with the change of the surface expression quantity of NMDA receptor and/or AMPA receptor subunits. Synaptosomal-associated protein 25 (SNAP-25), which is a key soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) protein involved in incorporation of NMDA receptor to postsynaptic membrane, contributed to SR58611A (1 mg/kg)-induced enhancement of recognition memory and NMDA receptor function. Moreover, SR58611A (1 mg/kg) could rescue repeated stress-induced defect of both recognition memory and NMDA receptor function through a SNAP-25-dependent mechanism. These results provide a potential mechanism underlying the cognitive-enhancing effects of SR58611A (1 mg/kg).


Asunto(s)
Corteza Prefrontal , Receptores Adrenérgicos beta 3 , Receptores de Glutamato , Animales , Masculino , Ratas , Agonistas de Receptores Adrenérgicos beta 3/metabolismo , Agonistas de Receptores Adrenérgicos beta 3/farmacología , Ansiolíticos/farmacología , Conducta Animal/fisiología , Encéfalo/metabolismo , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Ácido Glutámico/metabolismo , Norepinefrina/metabolismo , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/fisiología , Ratas Sprague-Dawley , Receptores Adrenérgicos beta 3/metabolismo , Receptores Adrenérgicos beta 3/fisiología , Receptores AMPA/metabolismo , Receptores de Glutamato/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Membranas Sinápticas/metabolismo , Proteína 25 Asociada a Sinaptosomas/metabolismo
19.
FASEB J ; 35(7): e21724, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34133802

RESUMEN

Neuromuscular junctions (NMJ) regulate cholinergic exocytosis through the M1 and M2 muscarinic acetylcholine autoreceptors (mAChR), involving the crosstalk between receptors and downstream pathways. Protein kinase C (PKC) regulates neurotransmission but how it associates with the mAChRs remains unknown. Here, we investigate whether mAChRs recruit the classical PKCßI and the novel PKCε isoforms and modulate their priming by PDK1, translocation and activity on neurosecretion targets. We show that each M1 and M2 mAChR activates the master kinase PDK1 and promotes a particular priming of the presynaptic PKCßI and ε isoforms. M1 recruits both primed-PKCs to the membrane and promotes Munc18-1, SNAP-25, and MARCKS phosphorylation. In contrast, M2 downregulates PKCε through a PKA-dependent pathway, which inhibits Munc18-1 synthesis and PKC phosphorylation. In summary, our results discover a co-dependent balance between muscarinic autoreceptors which orchestrates the presynaptic PKC and their action on ACh release SNARE-SM mechanism. Altogether, this molecular signaling explains previous functional studies at the NMJ and guide toward potential therapeutic targets.


Asunto(s)
Proteínas Quinasas Dependientes de 3-Fosfoinosítido/metabolismo , Unión Neuromuscular/metabolismo , Proteína Quinasa C/metabolismo , Receptores Muscarínicos/metabolismo , Acetilcolina/metabolismo , Animales , Regulación hacia Abajo/fisiología , Exocitosis/fisiología , Fosforilación/fisiología , Terminales Presinápticos/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal/fisiología , Transmisión Sináptica/fisiología , Proteína 25 Asociada a Sinaptosomas/metabolismo
20.
Sci Rep ; 11(1): 8252, 2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33859251

RESUMEN

Herein proteomic profiling of the rat hippocampus from the kindling and pilocarpine models of epilepsy was performed to achieve new potential targets for treating epileptic seizures. A total of 144 differently expressed proteins in both left and right hippocampi by two-dimensional electrophoresis coupled to matrix-assisted laser desorption-mass spectrometry were identified across the rat models of epilepsy. Based on network analysis, the majority of differentially expressed proteins were associated with Ca2+ homeostasis. Changes in ADP-ribosyl cyclase (ADPRC), lysophosphatidic acid receptor 3 (LPAR3), calreticulin, ubiquitin carboxyl-terminal hydrolase L1 (UCH-L1), synaptosomal nerve-associated protein 25 (SNAP 25) and transgelin 3 proteins were probed by Western blot analysis and validated using immunohistochemistry. Inhibition of calcium influx by 8-Bromo-cADP-Ribose (8-Br-cADPR) and 2-Aminoethyl diphenylborinate (2-APB) which act via the ADPRC and LPAR3, respectively, attenuated epileptic seizures. Considering a wide range of molecular events and effective role of calcium homeostasis in epilepsy, polypharmacy with multiple realistic targets should be further explored to reach the most effective treatments.


Asunto(s)
Calcio/metabolismo , Epilepsia/genética , Epilepsia/metabolismo , Hipocampo/metabolismo , Excitación Neurológica , Pilocarpina , Proteómica , ADP-Ribosil Ciclasa/metabolismo , Animales , ADP-Ribosa Cíclica/análogos & derivados , ADP-Ribosa Cíclica/fisiología , Modelos Animales de Enfermedad , Electroforesis/métodos , Epilepsia/terapia , Homeostasis , Masculino , Terapia Molecular Dirigida , Ratas Wistar , Receptores del Ácido Lisofosfatídico/metabolismo , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción/métodos , Proteína 25 Asociada a Sinaptosomas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA