Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
J Inflamm Res ; 17: 1467-1480, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38476468

RESUMEN

Background: Bronchopulmonary dysplasia (BPD) has become a major cause of morbidity and mortality in preterm infants worldwide, yet its pathogenesis and underlying mechanisms remain poorly understood. The present study sought to explore microRNA-mRNA regulatory networks and immune cells involvement in BPD through a combination of bioinformatic analysis and experimental validation. Methods: MicroRNA and mRNA microarray datasets were obtained from the Gene Expression Omnibus (GEO) database. Differentially expressed microRNAs (DEMs) were identified in BPD patients compared to control subjects, and their target genes were predicted using miRWalk, miRNet, miRDB, and TargetScan databases. Subsequently, protein-protein interaction (PPI) and functional enrichment analyses were conducted on the target genes. 30 hub genes were screened using the Cytohubba plugin of the Cytoscape software. Additionally, mRNA microarray data was utilized to validate the expression of hub genes and to perform immune infiltration analysis. Finally, real-time PCR (RT-PCR), immunohistochemistry (IHC), and flow cytometry were conducted using a mouse model of BPD to confirm the bioinformatics findings. Results: Two DEMs (miR-15b-5p and miR-20a-5p) targeting genes primarily involved in the regulation of cell cycle phase transition, ubiquitin ligase complex, protein serine/threonine kinase activity, and MAPK signaling pathway were identified. APP and four autophagy-related genes (DLC1, PARP1, NLRC4, and NRG1) were differentially expressed in the mRNA microarray dataset. Analysis of immune infiltration revealed significant differences in levels of neutrophils and naive B cells between BPD patients and control subjects. RT-PCR and IHC confirmed reduced expression of APP in a mouse model of BPD. Although the proportion of total neutrophils did not change appreciably, the activation of neutrophils, marked by loss of CD62L, was significantly increased in BPD mice. Conclusion: Downregulation of APP mediated by miR-15b-5p and miR-20a-5p may be associated with the development of BPD. Additionally, increased CD62L- neutrophil subset might be important for the immune-mediated injury in BPD.

2.
J Exp Clin Cancer Res ; 42(1): 322, 2023 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-38012650

RESUMEN

BACKGROUND: Lines of evidence indicated that, immune checkpoints (ICs) inhibitors enhanced T cell immune response to exert anti-tumor effects. However, T cell exhaustion has been so far a major obstacle to antitumor immunotherapy in colorectal cancer patients. Our previous studies showed that ginseng-derived nanoparticles (GDNPs) inhibited the growth of various tumors by reprograming tumor-associated macrophages (TAMs) and downregulated the ICs expression on T cells in tumor microenvironment (TME), but the underlying effector mechanisms remained unclear. METHODS: The correlation between arginase-1 (ARG1) and T cells was computed based on the colorectal cancer patients in TCGA database. In vitro, we observed that GDNPs reprogrammed TAMs inhibited ARG1 release and ultimately ameliorated T cell exhaustion according to several techniques including WB, PCR, ELISA and flow cytometry. We also used an in vivo MC38 tumor-bearing model and administered GDNPs to assess their anti-tumor effects through multiple indices. The mechanism that GDNPs improved T cell exhaustion was further clarified using the bioinformatics tools and flow cytometry. RESULTS: GDNPs reprogramed TAMs via reducing ARG1 production. Moreover, normalized arginine metabolism ameliorated T cell exhaustion through mTOR-T-bet axis, resulting in reduced ICs expression and enhanced CD8+ T cells expansion. CONCLUSIONS: By regulating the mTOR-T-bet axis, GDNPs reprogramed macrophages to regulate ARG1 release, which further ameliorated T cell exhaustion in TME. These findings provided new insights into comprehending the mechanisms underlying the mitigation of T cell exhaustion, which may facilitate the development of innovative therapeutic strategies in the field of cancer treatment.


Asunto(s)
Arginasa , Neoplasias Colorrectales , Nanopartículas , Panax , Agotamiento de Células T , Humanos , Arginasa/metabolismo , Linfocitos T CD8-positivos/metabolismo , Neoplasias Colorrectales/patología , Macrófagos/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Microambiente Tumoral
3.
Nano Lett ; 23(21): 10058-10065, 2023 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-37877757

RESUMEN

Currently, mainstream lanthanide probes with fluorescence located in the second near-infrared subwindow of 1500-1700 nm (NIR-IIb) are predominantly Er(III)-based nanoparticles (NPs). Here we report a newly developed NIR-IIb fluorescent nanoprobe, α-Tm NP (cubic-phase NaYF4@NaYF4:Tm@NaYF4), with an emission at 1630 nm. We activate the 1630 nm emission of Tm(III) in α-Tm NP through the large spread of the Stark split sublevels induced by the crystal-field effect of the α-NaYF4 host. Further, we systematically investigated the effect of crystalline structure of the host NaYF4 NP (cubic phase (α) or hexagonal phase (ß)), the type and concentrations of dopants (Yb(III), Tm(III), and Ca(II) ions) in the α-phase host, and the thicknesses of the interlayer and inert shell on the NIR-IIb fluorescence of Tm(III). The ultimate nanostructure presents a significant enhancement factor of the NIR-IIb photoluminescence intensity of Tm(III) up to ∼315. With this bright NIR-IIb fluorescent nanoprobe, we demonstrate high-spatial-resolution time-coursing imaging of breast cancer bone metastasis.

4.
Chem Asian J ; 18(23): e202300847, 2023 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-37842968

RESUMEN

Aggregation-induced emission luminogens (AIEgens) in the second near-infrared region (NIR-II,1000-1700 nm) have shown tremendous potential as theragnostic probe for tumor multimodal diagnostic imaging and combined treatment owing to their programmable optical, structural and functional properties. Herein, we presented a radionuclide 177 Lu-labeled AIEgen, 177 Lu-2TT-oC6B dots, for NIR-II fluorescence and SPECT/CT imaging-guided tumor photothermal and radiopharmaceutical therapy. Intriguingly, 177 Lu-2TT-oC6B self-assembled into 10 nm dots, exhibited high NIR-II fluorescence quantum yield (QY, 1.34 %) and unprecedented photothermal conversion efficiency (PCE, 70.3 %) in vitro, furtherly performed extremely long blood circulation (T1/2 =52.4 h), persistent tumor accumulation and retention in tumor (NIR-II SNR=5.56; SPECT SNR=36.59) via intravenous administration in vivo. Furthermore, upon NIR light activation and 177 Lu irradiation, 177 Lu-2TT-oC6B demonstrated great application potential in synergistic photothermal/radiopharmaceutical tumor therapy.


Asunto(s)
Nanopartículas , Neoplasias , Humanos , Radiofármacos/farmacología , Neoplasias/diagnóstico por imagen , Neoplasias/tratamiento farmacológico , Terapia Fototérmica , Imagen Óptica/métodos , Imagen Multimodal , Nanopartículas/química
5.
Chem Asian J ; 18(11): e202300189, 2023 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-37032315

RESUMEN

Breast cancer has become a huge burden with continued rise of incidence and death rate worldwide. Various methods for diagnosis and therapy of breast cancer have met the challenges of lack of complete information about the tumor location and limited therapy efficacy. Although aggregation-induced emission luminogens (AIEgens) have shown great promise for various cancer treatment applications, they may be incompetent for deep-seated tumor diagnosis due to the limited penetration depth. Herein, we designed and prepared a radiolabeled AIEgen-based organic photothermal agent for bimodal PET/fluorescence imaging-guided breast tumor photothermal therapy. The prepared multifunctional nanoparticles (68 Ga-TPA-TTINC NPs) with NIR-II fluorescence, gamma irradiation and photothermal conversion property could be efficiently taken up by tumor cells and induce reactive oxygen species burst in vitro, further boosting the photothermal treatment of tumor in vivo. More importantly, the nanoprobe could target and clearly visualize 4T1 tumor xenografts through PET and NIR-II fluorescence imaging with high tumor/muscle ratio up to 4.8, which provides a promising tool and solution for breast tumor theranostics.


Asunto(s)
Neoplasias de la Mama , Nanopartículas , Neoplasias , Humanos , Femenino , Terapia Fototérmica , Fluorescencia , Nanomedicina Teranóstica/métodos , Neoplasias/patología , Neoplasias de la Mama/diagnóstico por imagen , Neoplasias de la Mama/terapia , Fototerapia/métodos , Imagen Óptica/métodos , Línea Celular Tumoral
6.
J Nanobiotechnology ; 21(1): 41, 2023 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-36740689

RESUMEN

Clinically, activated EGFR mutation associated chemo-drugs resistance has severely threaten NSCLC patients. Nanoparticle based small interfering RNA (siRNA) therapy representing another promising alternative by silencing specific gene while still suffered from charge associated toxicity, strong immunogenicity and poor targetability. Herein, we reported a novel EGFR-mutant NSCLC therapy relying on edible and cation-free kiwi-derived extracellular vesicles (KEVs), which showed sevenfold enhancement of safe dosage compared with widely used cationic liposomes and could be further loaded with Signal Transducer and Activator of Transcription 3 interfering RNA (siSTAT3). siSTAT3 loaded KEVs (STAT3/KEVs) could be easily endowed with EGFR targeting ability (STAT3/EKEVs) and fluorescence by surface modification with tailor-making aptamer through hydrophobic interaction. STAT3/EKEVs with a controlled size of 186 nm displayed excellent stability, high specificity and good cytotoxicity towards EGFR over-expressing and mutant PC9-GR4-AZD1 cells. Intriguingly, the systemic administration of STAT3/EKEVs significantly suppressed subcutaneous PC9-GR4-AZD1 tumor xenografts in nude mice by STAT3 mediated apoptosis. This safe and robust KEVs has emerged as the next generation of gene delivery platform for NSCLC therapy after multiple drug-resistance.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Animales , Ratones , Humanos , ARN Interferente Pequeño/química , Ratones Desnudos , Frutas/metabolismo , Línea Celular Tumoral , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Receptores ErbB/genética , Receptores ErbB/metabolismo , Resistencia a Antineoplásicos/genética , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo
7.
J Hazard Mater ; 430: 128483, 2022 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-35739666

RESUMEN

2,2',4,4'-tetrabromodiphenyl ether (PBDE-47), the widely used brominated flame retardant, has remarkable neurotoxicity which is associated with autophagy disorder. However, the mechanism remains unclear. The results showed that PBDE-47 damaged lysosomal biogenesis and interfered with autophagy-lysosome fusion both in vivo and in vitro. Our investigation further demonstrated that PBDE-47 could downregulate TFEB expression and inhibit the nuclear translocation of TFEB. Knockdown of TFEB in PC12 cells increased the reduction of lysosomal-associated proteins and the expression of STX17-SNAP29-VAMP8 proteins involved in autophagy-lysosomal fusion. Conversely, Overexpression TFEB in vitro significantly improved lysosomal abundance and ameliorated the autophagosome-lysosome fusion inhibition, thus restoring autophagic flux and improving PC12 cells survival. In addition, TFEB biologically interacted with STX17 by not inducing or inducing TFEB overexpression. Collectively, our results indicate that the autophagy flux compromised by PBDE-47 is related to the defective fusion of autophagosome and lysosome. TFEB may serve as a promising molecular target for future study of PBDE-47 developmental neurotoxicity.


Asunto(s)
Autofagia , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice , Lisosomas , Síndromes de Neurotoxicidad , Animales , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Éteres Difenilos Halogenados/metabolismo , Éteres Difenilos Halogenados/toxicidad , Lisosomas/metabolismo , Síndromes de Neurotoxicidad/metabolismo , Proteínas Qb-SNARE/metabolismo , Proteínas Qc-SNARE/metabolismo , Ratas
8.
Mol Ther ; 30(1): 327-340, 2022 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-34450250

RESUMEN

Cold tumor microenvironment (TME) marked with low effector T cell infiltration leads to weak response to immune checkpoint inhibitor (ICI) treatment. Thus, switching cold to hot TME is critical to improve potent ICI therapy. Previously, we reported extracellular vesicle (EV)-like ginseng-derived nanoparticles (GDNPs) that were isolated from Panax ginseng C.A. Mey and can alter M2 polarization to delay the hot tumor B16F10 progression. However, the cold tumor is more common and challenging in the real world. Here, we explored a combinatorial strategy with both GDNPs and PD-1 (programmed cell death protein-1) monoclonal antibody (mAb), which exhibited the ability to alter cold TME and subsequently induce a durable systemic anti-tumor immunity in multiple murine tumor models. GDNPs enhanced PD-1 mAb anti-tumor efficacy in activating tumor-infiltrated T lymphocytes. Our results demonstrated that GDNPs could reprogram tumor-associated macrophages (TAMs) to increase CCL5 and CXCL9 secretion for recruiting CD8+ T cells into the tumor bed, which have the synergism to PD-1 mAb therapy with no detected systemic toxicity. In situ activation of TAMs by GDNPs may broadly serve as a facile platform to modulate the suppressive cold TME and optimize the PD-1 mAb immunotherapy in future clinical application.


Asunto(s)
Nanopartículas , Panax , Animales , Linfocitos T CD8-positivos , Línea Celular Tumoral , Inmunoterapia , Ratones , Microambiente Tumoral
9.
Biomed Mater ; 17(1)2021 12 24.
Artículo en Inglés | MEDLINE | ID: mdl-34879355

RESUMEN

Near-infrared (NIR) fluorescence imaging technology (NIR-I region, 650-950 nm and NIR-II region, 1000-1700 nm), with deeper tissue penetration and less disturbance from auto-fluorescence than that in visible region (400-650 nm), is playing a more and more extensive role in the field of biomedical imaging. With the development of precise medicine, intelligent NIR fluorescent probes have been meticulously designed to provide more sensitive, specific and accurate feedback on detection. Especially, recently developed ratiometric fluorescent probes have been devoted to quantify physiological and pathological parameters with a combination of responsive fluorescence changes and self-calibration. Herein, we systemically introduced the construction strategies of NIR ratiometric fluorescent probes and their applications in biological imagingin vivo, such as molecular detection, pH and temperature measurement, drug delivery monitoring and treatment evaluation. We further summarized possible optimization on the design of ratiometric probes for quantitative analysis with NIR fluorescence, and prospected the broader optical applications of ratiometric probes in life science and clinical translation.


Asunto(s)
Colorantes Fluorescentes , Imagen Óptica , Colorantes Fluorescentes/química , Imagen Óptica/métodos
10.
Adv Mater ; 33(37): e2103953, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34308556

RESUMEN

Fluorescence located in 1500-1700 nm (denoted as the near-infrared IIb window, NIR-IIb) has drawn great interest for bioimaging, owing to its ultrahigh tissue penetration depth and spatiotemporal resolution. Therefore, NIR-IIb fluorescent probes with high photoluminescence quantum yield (PLQY) and stability along with high biocompatibility are urgently pursued. Herein, a novel NIR-IIb fluorescent probe of Au-doped Ag2 Te (Au:Ag2 Te) quantum dots (QDs) is developed via a facile cation exchange method. The Au dopant concentration in the Ag2 Te QDs is tunable from 0% to 10% by controlling the ratio of supplied Au precursor to Ag2 Te QDs, resulting in a wide range of PL emission in the NIR-IIb window and a much-enhanced PL intensity. After surface modification, the Au:Ag2 Te QDs possess bright NIR-IIb emission, high colloidal stability and photostability, and decent biocompatibility. Further, in vivo monitoring of the process of angiogenesis and arteriogenesis in an ischemic hindlimb is successfully performed.


Asunto(s)
Materiales Biocompatibles/química , Isquemia/diagnóstico por imagen , Puntos Cuánticos/química , Espectroscopía Infrarroja Corta , Animales , Materiales Biocompatibles/farmacología , Línea Celular , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Oro/química , Miembro Posterior/irrigación sanguínea , Miembro Posterior/diagnóstico por imagen , Humanos , Ratones , Ratones Endogámicos BALB C , Neovascularización Patológica , Puntos Cuánticos/toxicidad , Relación Señal-Ruido , Plata/química , Telurio/química
11.
Nano Lett ; 21(15): 6576-6583, 2021 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-34304558

RESUMEN

Rational design of efficient lanthanide-doped down-shifting nanoparticles (DSNPs) has attracted tremendous attention. However, energy loss was inevitable in the multiple Ln3+ doping systems owing to complex energy migration processes. Here, an efficient NaErF4@NaYF4@NaYF4:10%Nd@NaYF4 DSNP was tactfully designed, in which a buffer layer of NaYF4 was modulated to restrict the interionic energy migration between Er3+ and Nd3+; meanwhile, the surface defects were passivated by an outermost layer of NaYF4. Therefore, the as-prepared DSNPs exhibited two intensive near-infrared-II fluorescence emissions of 1525 nm from Er3+ and 1060 nm from doped Nd3+ under 808 nm excitation. Further, a novel ratiometric nanoprobe NaErF4@NaYF4@NaYF4:10%Nd@NaYF4@A1094 was fabricated by coupling an organic dye of A1094 onto the DSNP surface to quench the 1060 nm emission by the efficient Förster resonance energy transfer, while emission at 1525 nm retained. Thereafter, these activatable ratiometric nanoprobes were used for rapid and sensitive detection of peroxynitrite (ONOO-) in vivo.


Asunto(s)
Elementos de la Serie de los Lantanoides , Nanopartículas , Transferencia Resonante de Energía de Fluorescencia
12.
Brain Res Bull ; 170: 81-89, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33581311

RESUMEN

Aberrant expression or dysfunction of a number of genes in the brain contributes to epilepsy, a common neurological disorder characterized by recurrent seizures. Local overexpression of arachidonate lipoxygenase 3 (ALOXE3), a key enzyme for arachidonic acid (AA) metabolic pathway, alleviates seizure severities. However, the relationship between the ALOXE3 gene mutation and epilepsy has not been reported until now. Here we firstly characterized the promoter of human ALOXE3 gene and found that the ALOXE3 promoter could drive luciferase gene expression in the human HEK-293 and SH-SY5Y cells. We then screened the ALOXE3 promoter region and all coding exons from those patients with Dravet syndrome and identified 5 variants c.-163T > C, c.-50C > G, c.-37G > A, c. + 228G > A and c. + 290G > T in the promoter region and one missense variant c.1939A > G (p.I647 V) in the exon. Of these variants in the promoter region, only -50C > G was a novel variant located on the transcriptional factor NFII-I binding element. Luciferase reporter gene analyses indicated that the c.-50C > G could decrease gene expression by preventing the TFII-I's binding. In addition, the variant p.I647 V was conserved among all analyzed species and located within the ALOXE3 functional domain for catalyzing its substrate. In cultured cell lines, overexpression of ALOXE3 significantly decreased the cellular AA levels and overexpression of ALOXE3-I647 V could restore the AA levels, suggesting that the p.I647 V mutant led to a decrease in enzyme activity. Taken together, the present study proposes that the identified ALOXE3 variants potentially contribute to the AA-pathway-mediated epileptogenesis, which should provide a novel avenue for clinical diagnosis of epilepsy.


Asunto(s)
Encéfalo/metabolismo , Epilepsias Mioclónicas/genética , Lipooxigenasa/genética , Mutación , Alelos , Epilepsias Mioclónicas/metabolismo , Células HEK293 , Humanos , Lipooxigenasa/metabolismo , Fenotipo , Regiones Promotoras Genéticas
13.
Small ; 17(8): e2006111, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33522125

RESUMEN

Ag2 Se quantum dots (QDs) as an effective biological probe in the second near-infrared window (NIR-II, 1000-1700 nm) have been widely applied in bioimaging with high tissue penetration depth and high spatiotemporal resolution. However, the ions deficiency and crystal defects caused by the high Ag+ mobility in Ag2 Se crystals are mainly responsible for the inefficient photoluminescence (PL) of Ag2 Se QDs. Herein, a tailored route is reported to achieve controllable doping of Ag2 Se QDs in which Ag is exchanged by Pb via cation exchange (CE), which is unattainable by direct synthetic methods. The Pb-doped Ag2 Se QDs (denoted as Pb:Ag2 Se QDs) present fire-new optical features with significantly enhanced PL intensity of 4.2 folds. Photoelectron spectroscopy confirms that Pb acts as an n-type dopant for Ag2 Se QDs and therefore the electronic impurities provide additional carriers to fill the traps. Moreover, the general validity of this method is demonstrated to convert different sized Ag2 Se into Pb:Ag2 Se QDs, so that a wide range of NIR-II PL with high intensity is obtained. The bright NIR-II emission of Pb:Ag2 Se QDs is further successfully performed in lymphatic system mapping.


Asunto(s)
Puntos Cuánticos , Plomo , Plata
14.
Adv Healthc Mater ; 10(5): e2001544, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33002345

RESUMEN

Ischemic stroke is a devastating disease resulting in high morbidity and mortality. To date, its early diagnosis still faces challenges. Herein, an efficient detection strategy is proposed, in which a targeted activatable NIR-IIb nanoprobe (V&C/PbS@Ag2 Se) is constructed for in vivo highly sensitive detection of early ischemic stroke in a photothrombotic stroke model. At first, the fluorescence of V&C/PbS@Ag2 Se displays an "off" state due to the competitive absorption of excitation irradiation between Cy7.5 fluorophores and PbS@Ag2 Se quantum dots (QDs). Upon intravenous injection, the V&C/PbS@Ag2 Se quickly accumulates in the lesion regions based on VCAM1 binding peptide target to the inflamed vascular endothelium of ischemic stroke. Later, the nanoprobes can be rapidly activated via Cy7.5 oxidation by peroxynitrite (ONOO- ), the prodromal biomarker of ischemic stroke, instantly illuminating the lesion regions. Such a targeted activatable strategy offers a favorable approach for in vivo early real-time assessment of ischemic stroke, which can be expanded to other diseases as a general mothed for in vivo precise diagnosis.


Asunto(s)
Isquemia Encefálica , Accidente Cerebrovascular Isquémico , Puntos Cuánticos , Accidente Cerebrovascular , Isquemia Encefálica/diagnóstico , Fluorescencia , Humanos , Accidente Cerebrovascular/diagnóstico
15.
Angew Chem Int Ed Engl ; 60(5): 2637-2642, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33015947

RESUMEN

Accurate intraoperative tissue identification is critical to tumor surgery. However, conventional methods are labor- and time-intensive, which greatly delay the intraoperative decision-making. Herein, a matrix metalloproteinase (MMP)14-activated NIR-II nanoprobe (A&MMP@Ag2 S-AF7P) is presented for rapid unperturbed-tissue analysis for ex vivo and in vivo neuroblastoma diagnosis. A&MMP@Ag2 S-AF7P displays negligible fluorescence in normal tissues but is activated quickly by inhibiting the fluorescence resonance energy transfer (FRET) between Ag2 S QDs and A1094 mediated by MMP14 overexpressed in neuroblastoma; meanwhile, the exposure of the membrane penetrating peptide R9 (TAT-peptide) results in efficient internalization of nanoprobes in the cancer cells, providing superior tumor-to-normal (T/N) tissue ratio. Instant illumination of the lesion and well-defined tumor margins make the nanoprobes a suitable rapid diagnostic reagent for cancer surgical or tissue biopsy procedures.


Asunto(s)
Rayos Infrarrojos/uso terapéutico , Nanopartículas/química , Neoplasias/diagnóstico , Humanos
16.
Front Med ; 13(5): 602-609, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31321611

RESUMEN

Panic disorder (PD) is an acute paroxysmal anxiety disorder with poorly understood pathophysiology. The dorsal periaqueductal gray (dPAG) is involved in the genesis of PD. However, the downstream neurofunctional changes of the dPAG during panic attacks have yet to be evaluated in vivo. In this study, optogenetic stimulation to the dPAG was performed to induce panic-like behaviors, and in vivo positron emission tomography (PET) imaging with 18F-flurodeoxyglucose (18F-FDG) was conducted to evaluate neurofunctional changes before and after the optogenetic stimulation. Compared with the baseline, post-optogenetic stimulation PET imaging demonstrated that the glucose metabolism significantly increased (P < 0.001) in dPAG, the cuneiform nucleus, the cerebellar lobule, the cingulate cortex, the alveus of the hippocampus, the primary visual cortex, the septohypothalamic nucleus, and the retrosplenial granular cortex but significantly decreased (P < 0.001) in the basal ganglia, the frontal cortex, the forceps minor corpus callosum, the primary somatosensory cortex, the primary motor cortex, the secondary visual cortex, and the dorsal lateral geniculate nucleus. Taken together, these data indicated that in vivo PET imaging can successfully detect downstream neurofunctional changes involved in the panic attacks after optogenetic stimulation to the dPAG.


Asunto(s)
Ansiedad de Separación/complicaciones , Trastorno de Pánico/etiología , Sustancia Gris Periacueductal/diagnóstico por imagen , Sustancia Gris Periacueductal/fisiopatología , Animales , Masculino , Optogenética , Tomografía de Emisión de Positrones , Ratas , Ratas Sprague-Dawley
17.
Neuroscience ; 404: 282-296, 2019 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-30742966

RESUMEN

Fragile X mental retardation protein (FMRP), a key determinant of normal brain development and neuronal plasticity, plays critical roles in nucleocytoplasmic shuttling of mRNAs. However, the factors involved in FMRP nuclear localization remain to be determined. Using cross-species sequence comparison, we show that an aspartate in position 132 (D132), located within the conserved nuclear localization signal (NLS) of FMRP, appears in human and other mammals, while glutamate 132 (E132) appears in rodents and birds. Human FMRP-D132E alters the secondary structure of the protein and reduces its nuclear localization, while the reciprocal substitution in mouse FMRP-E132D promotes its nuclear localization. Human FMRP could interact with poly(A)-binding protein 1 (PABP1) which is impeded by the D132E mutation. Reversely, mouse FMRP could not interact with PABP1, but the E132D mutation leads to the FMRP-PABP1 interaction. We further show that overexpression of human FMRP-D132E mutant promotes the formation of cytoplasmic aggregates of PABP1 in human cells, but not of mouse FMRP-E132D in mouse cells. PABP1 knockdown reduces the nuclear localization of human FMRP, but not mouse FMRP. Furthermore, RNase A treatment decreases the PABP1 levels in the anti-V5-immunoprecipitates using the V5-hFMRP-transfected cells, suggesting an interaction between human FMRP and PABP1 in an RNA-dependent fashion. Thus, our data suggest that the FMRP protein with the human-used D132 accommodates a novel protein-RNA-protein interaction which may implicate a connection between FMRP residue transition and neural evolution.


Asunto(s)
Núcleo Celular/metabolismo , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , Proteína I de Unión a Poli(A)/metabolismo , ARN/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular Tumoral , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/química , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Células HEK293 , Humanos , Ratones , Proteína I de Unión a Poli(A)/química , Proteína I de Unión a Poli(A)/genética , Unión Proteica/fisiología , Estructura Secundaria de Proteína , ARN/química , ARN/genética , Especificidad de la Especie
18.
J Proteomics ; 185: 39-50, 2018 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-29953961

RESUMEN

Physical exercise has been reported to increase neurotrophin in brain tissues as hippocampus as well as increased neurotrophic level peripherally in blood plasma and might have an effect on/or affect molecular processes of energy metabolism (and homeostasis). In this study, using quantitative proteomic analysis, we obtained a plasma protein profile from the rat with long-term moderate exercise. A total of 752 proteins were identified in the plasma. Among them, 54 proteins were significant up-regulated and 47 proteins were down-regulated in the plasma of exercise group compared with the control group. Bioinformatic analyses showed that these altered proteins are widely involved in multiple biological processes, molecular functions and cellular components, which connect with 11 signaling pathways. Interestingly, 5 up-regulated proteins Rap1b, PTPN11, ARHGDIA, Cdc42 and YWHAE, confirmed by Western blots, are involved in the neurotrophin signaling pathway which shows the lowest P value among the identified pathways. Further analyses showed that the 5 neurotrophin-signaling-pathway-related proteins participate in two important protein-protein interaction networks associated to cell survival and apoptosis, axonal development, synapse formation and plasticity. This study provides an exercise-induced plasma protein profile, suggesting that long-term exercise enhances the proteins involved in neurotrophin signaling pathway which may contribute to health benefit. SIGNIFICANCE: Physical activity contributes to myriad benefits on body health across the lifespan. The changes in plasma proteins after chronic moderate exercise may be used as biomarkers for health and may also play important roles in increase of cardiovascular fitness, enhancement of immune competence, prevention of obesity, decrease of risk for neurological disorders, cancer, stroke, diabetes and other metabolic disorders. Using a TMT-based proteomic method, this study identified 101 altered proteins in the plasma of rats after long-term moderate treadmill running, which may provide novel biomarkers for further investigation of the underlying mechanism of physical exercise. We confirmed that exercise enhances 5 proteins of the neurotrophin signaling pathway that may contribute to health benefits.


Asunto(s)
Proteínas Sanguíneas/análisis , Factores de Crecimiento Nervioso/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Condicionamiento Físico Animal/métodos , Proteómica/métodos , Animales , Proteínas Sanguíneas/química , Proteínas Sanguíneas/metabolismo , Estudios de Casos y Controles , Cromatografía Liquida , Masculino , Neurogénesis/fisiología , Plasma/química , Plasma/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal/fisiología , Espectrometría de Masas en Tándem , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA