Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Subst Use Misuse ; 59(2): 193-207, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-37822106

RESUMEN

BACKGROUND: While growing evidence has identified mental and physical health-related cannabis use motives as significant mechanisms between childhood trauma and problematic cannabis use (PCU) for emerging adults (EA), there is a need to understand the longitudinal stability of these pathways and how they impact PCU as cannabis users age into later adulthood. METHODS: The current study extends an analysis examining the impact of childhood trauma (e.g., emotional abuse, sexual abuse) on multiple indicators of PCU through a range of cannabis use motives. 339 medical cannabis patient and non-patient EA users from the Los Angeles area were sampled at baseline (mean age = 21.23; SD = 2.48). The present analysis used four waves of follow-up data collected from 2016 to 2018 (W3, W4) and 2019-2020 (W5, W6). RESULTS: Use of cannabis to cope with nausea, sleep, pain, and emotional distress mediated the relationships between some types of childhood abuse and PCU at W4, though most associations attenuated by later adulthood (W6). Specifically, greater emotional distress and nausea motives were associated with greater PCU in models of emotional abuse and neglect and sexual abuse, with emotional distress continuing to mediate at W6. Conversely, sleep and pain motives were associated with lower PCU in models for emotional neglect. CONCLUSIONS: Mental and physical health-related motives reflect potential intervenable factors that predict PCU in emerging adulthood among EA cannabis users with histories of childhood trauma. Results highlight the importance of and value for assessing a wide range of motives and PCU outcomes to target and address areas for intervention.


Asunto(s)
Experiencias Adversas de la Infancia , Cannabis , Adulto , Humanos , Niño , Adulto Joven , Motivación , Dolor , Náusea
2.
Exp Eye Res ; 178: 91-98, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30268699

RESUMEN

Optical coherence tomography (OCT) angiography is a dye-free and non-invasive angiography which allows visualization of retinal and choroid vascular flow, enabling observation of highly permeable and three dimensional vasculature. Although OCT angiography is providing new insights in human retinal and choroidal diseases, a few studies have been reported in experimental mice. In this study, to determine the potential of OCT angiography in experimental mice, we sought to examine whether OCT angiography can detect vascular change in type I diabetic mice. To conduct age dependent analysis, 2 and 6 month old male type 1 diabetic Ins2Akita/+ and age matched C57BL/6J mice were used. OCT angiography was performed by Heidelberg Spectralis OCT Angiography Module with 30° lens + mouse adapter lens. We acquired the OCT angiography image from the peripheral nasal position. For analysis of OCT angiography images, OCT angiography positive area were used for vascular density. We analyzed vascular density from the retinal surface (inner limiting membrane) to 120 µm depth with 4 µm steps in order to correlate vascular density vs depth (N = 4 per group). Vascular density of both mouse strains demonstrated three different peaks. By comparing with the OCT image, the first peak (superficial), second peak (intermediate) and third peak (deep) were located in nerve fiber layer/ganglion cell layer, inner plexiform layer/inner nuclear layer and outer plexiform layer/outer nuclear layer, respectively. We calculated vascular density of these peaks separately. In C57BL/6J mice, the vascular density in all three layers do not show significant difference between 2- and 6-month-old. On the other hand, 6-month-old Ins2Akita/+ mice showed a significant decrease of the vascular density in all three layers compared to 2-month-old Ins2Akita/+ mice. Also, the vascular density of 6-month-old Ins2Akita/+ mice in the deep layer showed a significant decrease compared to 2- and 6-month-old C57BL/6J mice. Thus, OCT angiography successfully detects retinal vascular difference between type I diabetic mice and control mice, and age-dependent vasculature change in type I diabetic mice. The diabetic mice demonstrated reduced vascular density due to reduced density of flowing deep vessels. Importantly, we observed this difference without retinal blood leakage, hemorrhage or neovascularization. Our analysis (vascular density vs retinal depth) suggests that OCT angiography is useful for in vivo detection of retinal vasculature alteration in experimental mice.


Asunto(s)
Diabetes Mellitus Tipo 1/diagnóstico , Retinopatía Diabética/diagnóstico , Vasos Retinianos/patología , Envejecimiento/fisiología , Animales , Diabetes Mellitus Experimental/diagnóstico , Angiografía con Fluoresceína , Masculino , Ratones , Ratones Endogámicos C57BL , Microvasos/patología , Vasos Retinianos/diagnóstico por imagen , Tomografía de Coherencia Óptica
3.
Bioorg Med Chem Lett ; 27(24): 5473-5480, 2017 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-29150397

RESUMEN

In this work, we describe the use of the rule of 3 fragment-based strategies from biochemical screening data of 1100 in-house, small, low molecular weight fragments. The sequential combination of in silico fragment hopping and fragment linking based on S160/Y161/A162 hinge residues hydrogen bonding interactions leads to the identification of novel 1H-benzo[d]imidazol-2-yl)-1H-indazol class of Phosphoinositide-Dependent Kinase-1 (PDK1) inhibitors. Consequent SAR and follow-up screening data led to the discovery of two potent PDK1 inhibitors: compound 32 and 35, with an IC50 of 80 nM and 94 nM, respectively. Further biological evaluation showed that, at the low nanomolar concentration, the drug had potent ability to inhibit phosphorylation of AKT and p70S6, and selectively kill the cancer cells with mutations in both PTEN and PI3K. The microarray data showed that DUSP6, DUSP4, and FOSL1 were down-regulated in the sensitive cell lines with the compound treatment. The in vivo test showed that 35 can significantly inhibit tumor growth without influencing body weight growth. Our results suggest that these compounds, especially 35, merit further pre-clinical evaluation.


Asunto(s)
Diseño de Fármacos , Indazoles/química , Inhibidores de Proteínas Quinasas/síntesis química , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Apoptosis/efectos de los fármacos , Sitios de Unión , Línea Celular Tumoral , Regulación hacia Abajo/efectos de los fármacos , Fosfatasa 6 de Especificidad Dual/genética , Fosfatasa 6 de Especificidad Dual/metabolismo , Fosfatasas de Especificidad Dual/genética , Fosfatasas de Especificidad Dual/metabolismo , Humanos , Imidazoles/química , Indazoles/síntesis química , Indazoles/farmacología , Concentración 50 Inhibidora , Fosfatasas de la Proteína Quinasa Activada por Mitógenos/genética , Fosfatasas de la Proteína Quinasa Activada por Mitógenos/metabolismo , Simulación del Acoplamiento Molecular , Fosfohidrolasa PTEN/antagonistas & inhibidores , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Relación Estructura-Actividad
4.
Exp Eye Res ; 145: 248-257, 2016 04.
Artículo en Inglés | MEDLINE | ID: mdl-26775053

RESUMEN

To assess whether Tie2-mediated vascular stabilization ameliorates neovascular age-related macular degeneration (AMD), we investigated the impact of adeno-associated virus-mediated gene therapy with cartilage oligomeric matrix protein angiopoietin-1 (AAV2.COMP-Ang1) on choroidal neovascularization (CNV), vascular endothelial growth factor (VEGF), and hypoxia-inducible factor (HIF) in a mouse model of the disease. We treated mice with subretinal injections of AAV2.COMP-Ang1 or control (AAV2.AcGFP, AAV2.LacZ, and phosphate-buffered saline). Subretinal AAV2 localization and plasmid protein expression was verified in the retinal pigment epithelium (RPE)/choroid of mice treated with all AAV2 constructs. Laser-assisted simulation of neovascular AMD was performed and followed by quantification of HIF, VEGF, and CNV in each experimental group. We found that AAV2.COMP-Ang1 was associated with a significant reduction in VEGF levels (29-33%, p < 0.01) and CNV volume (60-70%, p < 0.01), without a concomitant decrease in HIF1-α, compared to all controls. We concluded that a) AAV2 is a viable vector for delivering COMP-Ang1 to subretinal tissues, b) subretinal COMP-Ang1 holds promise as a prospective treatment for neovascular AMD, and c) although VEGF suppression in the RPE/choroid may be one mechanism by which AAV2.COMP-Ang1 reduces CNV, this therapeutic effect may be hypoxia-independent. Taken together, these findings suggest that AAV2.COMP-Ang1 has potential to serve as an alternative or complementary option to anti-VEGF agents for the long-term amelioration of neovascular AMD.


Asunto(s)
Proteína de la Matriz Oligomérica del Cartílago/uso terapéutico , Neovascularización Coroidal/terapia , Terapia Genética/métodos , Degeneración Macular/terapia , Factor A de Crecimiento Endotelial Vascular/metabolismo , Angiopoyetina 1/metabolismo , Animales , Western Blotting , Proteína de la Matriz Oligomérica del Cartílago/metabolismo , Neovascularización Coroidal/metabolismo , Dependovirus/genética , Modelos Animales de Enfermedad , Vectores Genéticos/administración & dosificación , Factor 1 Inducible por Hipoxia/metabolismo , Degeneración Macular/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Epitelio Pigmentado de la Retina/metabolismo
5.
Bioorg Med Chem Lett ; 21(17): 5071-4, 2011 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-21531555

RESUMEN

Riboswitches are noncoding RNA elements embedded in 5'-untranslated region of many bacterial mRNAs regulating gene expression in response to essential metabolites. They are unique from other RNA targets because they have evolved to form specific structural receptors for the purpose of binding small molecular metabolites suggesting that structure-based rational drug design approach may be used in designing metabolite mimics targeting riboswitches. We have developed a fluorescence binding assay for SAM-II riboswitch aptamer and identified an S-adenosylmethionine (SAM) analogue that selectively binds to SAM-II riboswitch aptamer with comparable binding affinity to its native metabolite using structure-based design approach.


Asunto(s)
Diseño de Fármacos , S-Adenosilmetionina/análogos & derivados , Cristalografía por Rayos X , Estructura Molecular , S-Adenosilmetionina/química
6.
Elife ; 102021 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-34100716

RESUMEN

A missense mutation of collagen type VIII alpha 2 chain (COL8A2) gene leads to early-onset Fuchs' endothelial corneal dystrophy (FECD), which progressively impairs vision through the loss of corneal endothelial cells. We demonstrate that CRISPR/Cas9-based postnatal gene editing achieves structural and functional rescue in a mouse model of FECD. A single intraocular injection of an adenovirus encoding both the Cas9 gene and guide RNA (Ad-Cas9-Col8a2gRNA) efficiently knocked down mutant COL8A2 expression in corneal endothelial cells, prevented endothelial cell loss, and rescued corneal endothelium pumping function in adult Col8a2 mutant mice. There were no adverse sequelae on histology or electroretinography. Col8a2 start codon disruption represents a non-surgical strategy to prevent vision loss in early-onset FECD. As this demonstrates the ability of Ad-Cas9-gRNA to restore the phenotype in adult post-mitotic cells, this method may be widely applicable to adult-onset diseases, even in tissues affected with disorders of non-reproducing cells.


Asunto(s)
Sistemas CRISPR-Cas/genética , Codón Iniciador/genética , Distrofia Endotelial de Fuchs , Edición Génica/métodos , Animales , Colágeno Tipo VIII/genética , Modelos Animales de Enfermedad , Distrofia Endotelial de Fuchs/genética , Distrofia Endotelial de Fuchs/prevención & control , Masculino , Ratones , Ratones Endogámicos C57BL , ARN Guía de Kinetoplastida/genética
7.
Plast Reconstr Surg Glob Open ; 7(9): e2442, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31942400

RESUMEN

The radial artery forearm free flap (RFFF) is the workhorse technique for phallus reconstruction. The RFFF provides good cosmesis and potential sensory recovery. However, the donor site is large in comparison to other applications of the RFFF which may increase the potential for donor site morbidity, such as nerve injury, delayed wound healing, and decreased hand strength. This study systematically reviewed the current literature to assess the donor site morbidity associated with RFFF phalloplasty (RFFFP). METHODS: A systematic review utilizing Preferred Reporting Items for Systematic Review and Meta-Analyses guidelines was completed of the current literature pertaining to donor site morbidity after RFFFP. Two investigators independently reviewed the literature to determine eligibility for inclusion. Two hundred sixty-seven studies were reviewed and 10 were included in the final analysis after application of exclusion criteria. RESULTS: Nine hundred forty flap reconstructions were identified. Gender affirming surgery was the indication in 77.7% (n = 730) of patients. The overall donor site complication rate was 7.9% (n = 74). Skin graft failure occurred in 41 patients (4.5%) and was the most frequent complication. Donor site infection (n = 3, 15.8%), hematoma (n = 1, 0.8%), neuroma (n = 1, 10%), compartment syndrome (n = 1, 0.8%), decreased strength or sensation (n = 15, 4.9%), lymphedema or limb swelling (n = 10, 3.9%), and contracture (n = 2, 6.5%) were also found. CONCLUSIONS: The most common donor site complication after RFFFP is skin graft failure. Decreased forearm sensation and strength affected a significant proportion of patients within each reported cohort. Prospective studies should continue to evaluate donor site morbidity with objective measures, such as grip strength evaluation, and long-term follow-up for vascular changes following radial artery harvest.

8.
Invest Ophthalmol Vis Sci ; 60(7): 2494-2502, 2019 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-31185088

RESUMEN

Purpose: We determine whether intravitreal angiopoietin-1 combined with the short coiled-coil domain of cartilage oligomeric matrix protein by adeno-associated viral serotype 2 (AAV2.COMP-Ang1) delivery following the onset of vascular damage could rescue or repair damaged vascular beds and attenuate neuronal atrophy and dysfunction in the retinas of aged diabetic mice. Methods: AAV2.COMP-Ang1 was bilaterally injected into the vitreous of 6-month-old male Ins2Akita mice. Age-matched controls consisted of uninjected C57BL/6J and Ins2Akita males, and of Ins2Akita males injected with PBS or AAV2.REPORTER (AcGFP or LacZ). Retinal thickness and visual acuity were measured in vivo at baseline and at the 10.5-month endpoint. Ex vivo vascular parameters were measured from retinal flat mounts, and Western blot was used to detect protein expression. Results: All three Ins2Akita control groups showed significantly increased deep vascular density at 10.5 months compared to uninjected C57BL/6J retinas (as measured by vessel area, length, lacunarity, and number of junctions). In contrast, deep microvascular density of Ins2Akita retinas treated with AAV2.COMP-Ang1 was more similar to uninjected C57BL/6J retinas for all parameters. However, no significant improvement in retinal thinning or diabetic retinopathy-associated visual loss was found in treated diabetic retinas. Conclusions: Deep retinal microvasculature of diabetic Ins2Akita eyes shows late stage changes consistent with disorganized vascular proliferation. We show that intravitreally injected AAV2.COMP-Ang1 blocks this increase in deep microvascularity, even when administered subsequent to development of the first detectable vascular defects. However, improving vascular normalization did not attenuate neuroretinal degeneration or loss of visual acuity. Therefore, additional interventions are required to address neurodegenerative changes that are already underway.


Asunto(s)
Angiopoyetina 1/administración & dosificación , Proteína de la Matriz Oligomérica del Cartílago/administración & dosificación , Retinopatía Diabética/prevención & control , Vectores Genéticos , Parvovirinae/genética , Neovascularización Retiniana/prevención & control , Vasos Retinianos/efectos de los fármacos , Animales , Glucemia/metabolismo , Western Blotting , Capilares/efectos de los fármacos , Dependovirus , Diabetes Mellitus Tipo 1/complicaciones , Retinopatía Diabética/fisiopatología , Portadores de Fármacos , Combinación de Medicamentos , Femenino , Terapia Genética , Insulina/genética , Inyecciones Intravítreas , Masculino , Ratones , Ratones Endogámicos C57BL , Retina/patología , Neovascularización Retiniana/fisiopatología , Vasos Retinianos/patología , Agudeza Visual/fisiología
9.
AMIA Jt Summits Transl Sci Proc ; 2019: 202-211, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31258972

RESUMEN

As the appeal and use of mobile health (mHealth) technologies continues to grow, where does mHealth fit into clinical practice? This article explores the approach and obstacles encountered when integrating mHealth data into existing clinical frameworks and explores data visualization design tradeoffs. Specifically, this paper discusses the successes and challenges that arose when using commercial mHealth technologies, synthesizing multiple mHealth device data, and tailoring visualizations based on iterative feedback from type II diabetes mellitus patients. This research aims to influence the development of patient portals within electronic health records by understanding and addressing the challenges involved in acquiring, interpreting, and displaying this data set. In particular, we need to ensure that the presentation of these data is accessible and understandable by diverse populations.

10.
PLoS One ; 13(3): e0193590, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29509796

RESUMEN

Short-activating RNA (saRNA), which targets gene promoters, has been shown to increase the target gene expression. In this study, we describe the use of an saRNA (Flt a-1) to target the flt-1 promoter, leading to upregulation of the soluble isoform of Flt-1 and inhibition of angiogenesis. We demonstrate that Flt a-1 increased sFlt-1 mRNA and protein levels, while reducing VEGF expression. This was associated with suppression of human umbilical vascular endothelial cell (HUVEC) proliferation and cell cycle arrest at the G0/G1 phase. HUVEC migration and tube formation were also suppressed by Flt a-1. An siRNA targeting Flt-1 blocked the effects of Flt a-1. Flt a-1 effects were not mediated via argonaute proteins. However, trichostatin A and 5'-deoxy-5'-(methylthio) adenosine inhibited Flt a-1 effects, indicating that histone acetylation and methylation are mechanistically involved in RNA activation of Flt-1. In conclusion, RNA activation of sFlt-1 can be used to inhibit angiogenesis.


Asunto(s)
Proliferación Celular/fisiología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , ARN Bicatenario/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Ciclo Celular/fisiología , Línea Celular , Movimiento Celular/fisiología , Metilación de ADN , Histonas/metabolismo , Humanos , Regiones Promotoras Genéticas , ARN Mensajero/metabolismo , ARN Interferente Pequeño , Regulación hacia Arriba , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética
11.
Transl Vis Sci Technol ; 6(3): 9, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28553563

RESUMEN

PURPOSE: We previously showed that intravitreal injection of the sFLT morpholino-oligomer (FLT-MO) suppresses laser-induced choroidal neovascularization (CNV) in mice by decreasing the membrane bound form of Flt-1 while increasing the soluble form of Flt-1 via alternative splicing shift. In this study, we examined whether cyclic RGD peptide (cRGD) can promote morpholino-oligomer accumulation in CNV following tail vein injection, and whether systemic cRGD conjugated FLT-MO (cRGD-FLT-MO) suppresses CNV growth. METHODS: cRGD conjugated fluorescent morpholino-oligomer (cRGD-F-MO) was injected via tail vein into mice with previous retinal laser photocoagulation and examined for cRGD-F-MO accumulation in CNV. To examine whether cRGD-FLT-MO suppresses CNV growth, mice were tail-vein injected with cRGD-FLT-MO, cRGD conjugated standard morpholino-oligomer (cRGD-STD-MO), or Dulbecco's Phosphate-Buffered Saline (DPBS) 1 and 4 days postlaser photocoagulation. Seven days postlaser photocoagulation, eyes were harvested and laser CNV was stained with isolectin GS-IB4, allowing quantification of CNV size by confocal microscopy. RESULTS: cRGD-F-MO accumulation in CNV commenced immediately after tail vein injection and could be observed even 1 day after injection. cRGD-FLT-MO tail vein injection significantly suppressed CNV size (2.7 × 105 ± 0.3 × 105 µm3, P < 0.05 by Student's t-test) compared with controls (DPBS: 5.1 × 105 ± 0.6 × 105 µm3 and cRGD-STD-MO: 5.5 × 105 ± 0.8 × 105 µm3). CONCLUSIONS: cRGD peptide facilitates morpholino-oligomer accumulation in CNV following systemic delivery. cRGD-FLT-MO suppressed CNV growth after tail-vein injection, demonstrating the potential utility of cRGD peptide for morpholino-oligomer delivery to CNV. TRANSLATIONAL RELEVANCE: Current therapy for neovascular age-related macular degeneration involves intravitreal injection of anti-vascular endothelial growth factor drugs. Our results indicate that CNV can be treated systemically, thus eliminating risks and hazards associated with intravitreal injection.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA