Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 83
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Ther ; 30(2): 816-830, 2022 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-34371179

RESUMEN

Inflammation plays a key role in the development of age-related diseases. In Alzheimer's disease, neuronal cell death is attributed to amyloidbeta oligomers that trigger microglial activation. Stem cells have shown promise as therapies for inflammatory diseases- because of their paracrine activity combined with their ability to respond to the inflammatory environment. However, the mechanisms underlying stem cell-promoted neurological recovery are poorly understood. To elucidate these mechanisms, we first primed stem cells with the secretome of lipopolysaccharide- or amyloidbeta-activated microglia. Then, we compared the immunomodulatory effects of extracellular vesicles (EVs) secreted from primed and non-primed stem cells. Our results demonstrate that EVs from primed cells are more effective in inhibiting microglia and astrocyte activation, amyloid deposition, demyelination, memory loss and motor and anxiety-like behavioral dysfunction, compared to EVs from non-primed cells. MicroRNA (miRNA) profiling revealed the upregulation of at least 19 miRNAs on primed-stem cell EVs. The miRNA targets were identified, and KEGG pathway analysis showed that the overexpressed miRNAs target key genes on the toll-like receptor-4 (TLR4) signaling pathway. Overall, our results demonstrate that priming mesenchymal stem cells (MSCs) with the secretome of activated microglia results in the release of miRNAs from EVs with enhanced immune regulatory potential able to fight neuroinflammation.


Asunto(s)
Enfermedad de Alzheimer , Encefalitis , Vesículas Extracelulares , Células Madre Mesenquimatosas , MicroARNs , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/terapia , Encefalitis/metabolismo , Vesículas Extracelulares/metabolismo , Humanos , Células Madre Mesenquimatosas/metabolismo , MicroARNs/metabolismo
2.
Int J Mol Sci ; 23(21)2022 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-36361743

RESUMEN

There is currently a growing interest in the use of cannabidiol (CBD) to alleviate the symptoms caused by cancer, including pain, sleep disruption, and anxiety. CBD is often self-administered as an over-the-counter supplement, and patients have reported benefits from its use. However, despite the progress made, the mechanisms underlying CBD's anti-cancer activity remain divergent and unclear. Herein, we provide a comprehensive review of molecular mechanisms to determine convergent anti-cancer actions of CBD from pre-clinical and clinical studies. In vitro studies have begun to elucidate the molecular targets of CBD and provide evidence of CBD's anti-tumor properties in cell and mouse models of cancer. Furthermore, several clinical trials have been completed testing CBD's efficacy in treating cancer-related pain. However, most use a mixture of CBD and the psychoactive, tetrahydrocannabinol (THC), and/or use variable dosing that is not consistent between individual patients. Despite these limitations, significant reductions in pain and opioid use have been reported in cancer patients using CBD or CBD+THC. Additionally, significant improvements in quality-of-life measures and patients' overall satisfaction with their treatment have been reported. Thus, there is growing evidence suggesting that CBD might be useful to improve the overall quality of life of cancer patients by both alleviating cancer symptoms and by synergizing with cancer therapies to improve their efficacy. However, many questions remain unanswered regarding the use of CBD in cancer treatment, including the optimal dose, effective combinations with other drugs, and which biomarkers/clinical presentation of symptoms may guide its use.


Asunto(s)
Cannabidiol , Neoplasias , Animales , Ratones , Cannabidiol/uso terapéutico , Dronabinol/efectos adversos , Calidad de Vida , Neoplasias/tratamiento farmacológico , Neoplasias/inducido químicamente , Dolor/tratamiento farmacológico
3.
Nanomedicine ; 32: 102325, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33186695

RESUMEN

Respiratory Syncytial Virus (RSV) has been a major health concern globally for decades, yet no effective prophylactic or treatment regimen is available. The key viral proteins responsible for RSV pathology include the fusion protein (F), the immunomodulatory non-structural-protein 1 (NS1) and the phosphoprotein (P) involved in viral replication. Herein, we developed a novel shell-core multifunctional nanosystem with dual payload: a plasmid construct encoding for shRNAs against NS1 and P, and an anti-fusion peptide (HR2D). Anti-ICAM1 antibody conjugated on the nanoparticle (NP) surface is used to target RSV infected cells. Our data show the potential of this nanosystem as a prophylactic and/or a therapeutic regimen against RSV infection. Furthermore, therapy of RSV infected mice with this nanosystem, in addition to reducing viral load, modulated expression of Th2 and allergy-associated cytokines such as IL4, IL-13 and IL-17 indicating a direct role of this nanosystem in the mechanisms involved in the immunoregulation of disease pathogenesis.


Asunto(s)
Nanopartículas Multifuncionales/uso terapéutico , Infecciones por Virus Sincitial Respiratorio/prevención & control , Virus Sincitial Respiratorio Humano/fisiología , Animales , Citocinas/metabolismo , Liberación de Fármacos , Femenino , Mediadores de Inflamación/metabolismo , Molécula 1 de Adhesión Intercelular/metabolismo , Ratones Endogámicos BALB C , Nanopartículas Multifuncionales/ultraestructura , Péptidos/farmacología , Plásmidos/genética , ARN Interferente Pequeño/metabolismo , Infecciones por Virus Sincitial Respiratorio/virología , Virus Sincitial Respiratorio Humano/efectos de los fármacos , Transfección , Proteínas Virales de Fusión/metabolismo
4.
Nanomedicine ; 30: 102294, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32861031

RESUMEN

The tumor microenvironment plays a critical role in tumor initiation, progression, metastasis, and drug resistance. However, models recapitulating the complex 3D structure, heterogeneous cell environment, and cell-cell interactions found in vivo are lacking. Herein, we report on a gravitational microfluidic platform (GMP) retrofitted with MEMS sensors, which is integrated with 3D nanofiber scaffold-aided tumoroid culture. The results showed that this GMP for tumoroid growth mimics the tumor microenvironment more precisely than static culture models of colon cancer, including higher drug resistance, enhanced cancer stem cell properties, and increased secretion of pro-tumor cytokines. In addition, the GMP includes an integrated surface acoustic wave-based biosensing to monitor cell growth and pH changes to assess drug efficacy. Thus, this simple-to-use perfused GMP tumoroid culture system for in vitro and ex vivo studies may accelerate the drug development process and be a tool in personalized cancer treatment.


Asunto(s)
Neoplasias Colorrectales/patología , Gravitación , Microfluídica/instrumentación , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Concentración de Iones de Hidrógeno , Medicina de Precisión , Microambiente Tumoral
5.
Nanomedicine ; 29: 102247, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32599163

RESUMEN

Traumatic brain injury (TBI) is a devastating neurological disorder, although the underlying pathophysiology is poorly understood. TBI causes blood-brain barrier (BBB) disruption, immune cell trafficking, neuroinflammation and neurodegeneration. CCL20 is an important chemokine mediating neuroinflammation. Human mesenchymal stem cell (hMSC) therapy is a promising regenerative approach but the inflammatory microenvironment in the brain tends to decrease the efficacy of the hMSC transplantation. Reducing the inflammation prior to hMSC therapy improves the outcome. We developed a combined nano-cell therapy by using dendrimers complexed with plasmids (dendriplexes) targeting CCL20 and its sole receptor CCR6 to reduce inflammation followed by hMSC transplantation. Treatment of TBI mice with shRNA conjugated dendriplexes followed by hMSC administration downregulated the inflammatory markers and significantly increased brain-derived neurotrophic factor (BDNF) expression in the cerebral cortex indicating future possible neurogenesis and improved behavioral deficits. Taken together, this nano-cell therapy ameliorates neuroinflammation and promotes brain tissue repair after TBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo/terapia , Factor Neurotrófico Derivado del Encéfalo/genética , Quimiocina CCL20/genética , Inflamación/terapia , Receptores CCR6/genética , Animales , Barrera Hematoencefálica/efectos de los fármacos , Lesiones Traumáticas del Encéfalo/genética , Lesiones Traumáticas del Encéfalo/patología , Quimiocina CCL20/antagonistas & inhibidores , Dendrímeros/química , Dendrímeros/farmacología , Humanos , Inflamación/genética , Inflamación/patología , Trasplante de Células Madre Mesenquimatosas , Ratones , Plásmidos/química , Plásmidos/genética , Plásmidos/farmacología , ARN Interferente Pequeño/farmacología , Receptores CCR6/antagonistas & inhibidores
6.
J Neuroinflammation ; 16(1): 115, 2019 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-31151410

RESUMEN

BACKGROUND: Traumatic brain injury (TBI) is a major cause of death and disability in the USA and the world; it constitutes 30% of injury-related deaths (Taylor et al., MMWR Surveill Summ 66:1-16, 2017). Contact sports athletes often experience repetitive TBI (rTBI), which exerts a cumulative effect later in life. Visual impairment is a common after-effect of TBI. Previously, we have shown that C-C chemokine 20 (CCL20) plays a critical role in neurodegeneration and inflammation following TBI (Das et al., J Neuroinflammation 8:148, 2011). C-C chemokine receptor 6 (CCR6) is the only receptor that CCL20 interacts with. The objective of the present study was to investigate the role of CCL20-CCR6 axis in mediating rTBI-induced visual dysfunction (TVD). METHODS: Wild type (WT) or CCR6 knock out (CCR6-/-) mice were subjected to closed head rTBI. Pioglitazone (PG) is a peroxisome proliferator-activated receptor γ (PPARγ) agonist which downregulates CCL20 production. Subsets of WT mice were treated with PG following final rTBI. A subset of mice was also treated with anti-CCL20 antibody to neutralize the CCL20 produced after rTBI. Histopathological assessments were performed to show cerebral pathologies, retinal pathologies, and inflammatory changes induced by rTBI. RESULTS: rTBI induced cerebral neurodegeneration, retinal degeneration, microgliosis, astrogliosis, and CCL20 expression. CCR6-/- mice showed reduced retinal degeneration, microgliosis, and inflammation. Treatment with CCL20 neutralization antibody or PG showed reduced CCL20 expression along with reduced retinal degeneration and inflammation. rTBI-induced GFAP-positive glial activation in the optic nerve was not affected by knocking out CCR6. CONCLUSION: The present data indicate that rTBI-induced retinal pathology is mediated at least in part by CCL20 in a CCR6-dependent manner.


Asunto(s)
Lesiones Traumáticas del Encéfalo/metabolismo , Quimiocina CCL20/metabolismo , Receptores CCR6/metabolismo , Retina/metabolismo , Animales , Lesiones Traumáticas del Encéfalo/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores CCR6/deficiencia , Retina/patología
7.
Sensors (Basel) ; 19(8)2019 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-31013700

RESUMEN

The most vital step in the development of novel and existing surface acoustic wave (SAW)-based sensors and transducers is their design and optimization. Demand for SAW devices has been steadily increasing due to their low cost, portability, and versatility in electronics, telecommunications, and biosensor applications. However, a full characterization of surface acoustic wave biosensors in a three-dimensional (3D) finite element model has not yet been developed. In this study, a novel approach is developed for analyzing shear horizontal Love wave resonator devices. The developed modeling methodology was verified using fabricated devices. A thorough analysis of the 3D model and the experimental device was performed in this study including scattering parameters (S-parameters), reflection coefficient parameters, transmission parameters, and phase velocity. The simulated results will be used as a design guideline for future device design and optimization, which has thus far resulted in close matching between prediction and experimental results. This manuscript is the first to demonstrate a 3D finite element model to correlate the sensitivity of the SAW device with the magnitude of the phase shift, the real and imaginary part of the response, insertion loss, and the frequency shift. The results show that the imaginary part of the response shift has a higher sensitivity compared to other parameters.

8.
Sensors (Basel) ; 19(20)2019 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-31635318

RESUMEN

Surface acoustic wave sensors have the advantage of fast response, low-cost, and wireless interfacing capability and they have been used in the medical analysis, material characterization, and other application fields that immerse the device under a liquid environment. The theoretical analysis of the single guided layer shear horizontal acoustic wave based on the perturbation theory has seen developments that span the past 20 years. However, multiple guided layer systems under a liquid environment have not been thoroughly analyzed by existing theoretical models. A dispersion equation previously derived from a system of three rigidly coupled elastic mass layers is extended and developed in this study with multiple guided layers to analyze how the liquid layer's properties affect the device's sensitivity. The combination of the multiple layers to optimize the sensitivity of an acoustic wave sensor is investigated in this study. The Maxwell model of viscoelasticity is applied to represent the liquid layer. A thorough analysis of the complex velocity due to the variations of the liquid layer's properties and thickness is derived and discussed to optimize multilayer Surface acoustic wave (SAW) sensor design. Numerical simulation of the sensitivity with a liquid layer on top of two guided layers is investigated in this study as well. The parametric investigation was conducted by varying the thicknesses for the liquid layer and the guided layers. The effect of the liquid layer viscosity on the sensitivity of the design is also presented in this study. The two guided layer device can achieve higher sensitivity than the single guided layer counterpart in a liquid environment by optimizing the second guided layer thickness. This perturbation analysis is valuable for Love wave sensor optimization to detect the liquid biological samples and analytes.

9.
Int J Mol Sci ; 20(21)2019 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-31731424

RESUMEN

Ashwagandha (Withania Somnifera, WS), belonging to the family Solanaceae, is an Ayurvedic herb known worldwide for its numerous beneficial health activities since ancient times. This medicinal plant provides benefits against many human illnesses such as epilepsy, depression, arthritis, diabetes, and palliative effects such as analgesic, rejuvenating, regenerating, and growth-promoting effects. Several clinical trials of the different parts of the herb have demonstrated safety in patients suffering from these diseases. In the last two decades, an active component of Withaferin A (WFA) has shown tremendous cytotoxic activity suggesting its potential as an anti-carcinogenic agent in treatment of several cancers. In spite of enormous progress, a thorough elaboration of the proposed mechanism and mode of action is absent. Herein, we provide a comprehensive review of the properties of WS extracts (WSE) containing complex mixtures of diverse components including WFA, which have shown inhibitory properties against many cancers, (breast, colon, prostate, colon, ovarian, lung, brain), along with their mechanism of actions and pathways involved.


Asunto(s)
Antineoplásicos Fitogénicos/química , Antineoplásicos Fitogénicos/uso terapéutico , Neoplasias/tratamiento farmacológico , Withania/química , Witanólidos/química , Witanólidos/uso terapéutico , Animales , Humanos , Neoplasias/metabolismo , Neoplasias/patología
10.
J Cell Physiol ; 233(5): 4317-4326, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29139549

RESUMEN

Supplementation of 100% oxygen is a very common intervention in intensive care units (ICU) and critical care centers for patients with dysfunctional lung and lung disorders. Although there is advantage in delivering sufficient levels of oxygen, hyperoxia is reported to be directly associated with increasing in-hospital deaths. Our previous studies reported ventricular and electrical remodeling in hyperoxia treated mouse hearts, and in this article, for the first time, we are investigating the effects of hyperoxia on atrial electrophysiology using whole-cell patch-clamp electrophysiology experiments along with assessment of Kv1.5, Kv4.2, and KChIP2 transcripts and protein profiles using real-time quantitative RT-PCR and Western blotting. Our data showed that induction of hyperoxia for 3 days in mice showed larger outward potassium currents with shorter action potential durations (APD). This increase in current densities is due to significant increase in ultrarapid delayed rectifier outward K+ currents (IKur ) and rapidly activating, rapidly inactivating transient outward K+ current (Ito ) densities. We also observed a significant increase in both transcripts and protein levels of Kv1.5 and KChIP2 in hyperoxia treated atrial cardiomyocytes, whereas no significant change was observed in Kv4.2 transcripts or protein. The data presented here further support our previous findings that hyperoxia induces not only ventricular remodeling, but also atrial electrical remodeling.


Asunto(s)
Proteínas de Interacción con los Canales Kv/genética , Canal de Potasio Kv1.6/genética , Enfermedades Pulmonares/terapia , Oxígeno/efectos adversos , Canales de Potasio Shal/genética , Potenciales de Acción/efectos de los fármacos , Animales , Regulación de la Expresión Génica , Atrios Cardíacos/fisiopatología , Mortalidad Hospitalaria , Humanos , Hiperoxia/etiología , Hiperoxia/fisiopatología , Unidades de Cuidados Intensivos , Pulmón/metabolismo , Pulmón/fisiopatología , Enfermedades Pulmonares/complicaciones , Enfermedades Pulmonares/mortalidad , Enfermedades Pulmonares/fisiopatología , Ratones , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Técnicas de Placa-Clamp , Potasio/metabolismo
11.
Dig Dis Sci ; 63(5): 1123-1138, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29572615

RESUMEN

Colorectal cancer (CRC) accounts for ~9% of all cancers in the Veteran population, a fact which has focused a great deal of the attention of the VA's research and development efforts. A field-based meeting of CRC experts was convened to discuss both challenges and opportunities in precision medicine for CRC. This group, designated as the VA Colorectal Cancer Cell-genomics Consortium (VA4C), discussed advances in CRC biology, biomarkers, and imaging for early detection and prevention. There was also a discussion of precision treatment involving fluorescence-guided surgery, targeted chemotherapies and immunotherapies, and personalized cancer treatment approaches. The overarching goal was to identify modalities that might ultimately lead to personalized cancer diagnosis and treatment. This review summarizes the findings of this VA field-based meeting, in which much of the current knowledge on CRC prescreening and treatment was discussed. It was concluded that there is a need and an opportunity to identify new targets for both the prevention of CRC and the development of effective therapies for advanced disease. Also, developing methods integrating genomic testing with tumoroid-based clinical drug response might lead to more accurate diagnosis and prognostication and more effective personalized treatment of CRC.


Asunto(s)
Neoplasias Colorrectales , Medicina de Precisión/métodos , Salud de los Veteranos , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Neoplasias Colorrectales/diagnóstico , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/terapia , Terapia Combinada , Detección Precoz del Cáncer/métodos , Humanos , Pronóstico
12.
Heart Vessels ; 33(5): 561-572, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29209776

RESUMEN

Hyperoxia, or supplemental oxygen, is regularly used in the clinical setting for critically ill patients in ICU. However, several recent studies have demonstrated the negative impact of this treatment in patients in critical care, including increased rates of lung and cardiac injury, as well as increased mortality. The purpose of this study was to determine the predisposition for arrhythmias and electrical remodeling in a type 2 diabetic mouse model (db/db), as a result of hyperoxia treatment. For this, db/db and their heterozygous controls were treated with hyperoxia (> 90% oxygen) or normoxia (normal air) for 72-h. Immediately following hyperoxia or normoxia treatments, mice underwent surface ECG. Excised left ventricles were used to assess ion channel expression, including for Kv1.4, Kv1.5, Kv4.2, and KChIP2. Serum cardiac markers were also measured, including cardiac troponin I and lactate dehydrogenase. Our results showed that db/db mice have increased sensitivity to arrhythmia. Normoxia-treated db/db mice displayed features of arrhythmia, including QTc and JT prolongation, as well as QRS prolongation. A significant increase in QRS prolongation was also observed in hyperoxia-treated db/db mice, when compared to hyperoxia-treated heterozygous control mice. Db/db mice were also shown to exhibit ion channel dysregulation, as demonstrated by down-regulation in Kv1.5, Kv4.2, and KChIP2 under hyperoxia conditions. From these results, we conclude that: (1) diabetic mice showed distinct pathophysiology, when compared to heterozygous controls, both in normoxia and hyperoxia conditions. (2) Diabetic mice were more susceptible to arrhythmia at normal air conditions; this effect was exacerbated at hyperoxia conditions. (3) Unlike in heterozygous controls, diabetic mice did not demonstrate cardiac hypertrophy as a result of hyperoxia. (4) Ion channel remodeling was also observed in db/db mice under hyperoxia condition similar to its heterozygous controls.


Asunto(s)
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 2/complicaciones , Ventrículos Cardíacos/fisiopatología , Hiperoxia/complicaciones , Síndrome de QT Prolongado/fisiopatología , Remodelación Ventricular/fisiología , Animales , Cardiotoxicidad , Diabetes Mellitus Tipo 2/fisiopatología , Modelos Animales de Enfermedad , Electrocardiografía , Ventrículos Cardíacos/diagnóstico por imagen , Hiperoxia/fisiopatología , Síndrome de QT Prolongado/diagnóstico , Síndrome de QT Prolongado/etiología , Masculino , Ratones
13.
J Drug Deliv Sci Technol ; 43: 453-460, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29805475

RESUMEN

The overall objective of the present research was to develop a nanocarrier system for non-invasive delivery to brain of molecules useful for gene therapy. Manganese-containing nanoparticles (mNPs) carrying anti-eGFP siRNA were tested in cell cultures of eGFP-expressing cell line of mouse fibroblasts (NIH3T3). The optimal mNPs were then tested in vivo in mice. Following intranasal instillation, mNPs were visualized by 7T MRI throughout brain at 24 and 48 hrs. mNPs were effective in significantly reducing GFP mRNA expression in Tg GFP+ mice in olfactory bulb, striatum, hippocampus and cortex. Intranasal instillation of mNPS loaded with dsDNA encoding RFP also resulted in expression of the RFP in multiple brain regions. In conclusion, mNPs carrying siRNA, or dsDNA were capable of delivering the payload from nose to brain. This approach for delivery of gene therapies to humans, if successful, will have a significant impact on disease-modifying therapeutics of neurodegenerative diseases.

15.
Sensors (Basel) ; 15(12): 32045-55, 2015 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-26703604

RESUMEN

Detection and quantification of cell viability and growth in two-dimensional (2D) and three-dimensional (3D) cell cultures commonly involve harvesting of cells and therefore requires a parallel set-up of several replicates for time-lapse or dose-response studies. Thus, developing a non-invasive and touch-free detection of cell growth in longitudinal studies of 3D tumor spheroid cultures or of stem cell regeneration remains a major unmet need. Since surface acoustic waves (SAWs) permit mass loading-based biosensing and have been touted due to their many advantages including low cost, small size and ease of assembly, we examined the potential of SAW-biosensing to detect and quantify cell growth. Herein, we demonstrate that a shear horizontal-surface acoustic waves (SH-SAW) device comprising two pairs of resonators consisting of interdigital transducers and reflecting fingers can be used to quantify mass loading by the cells in suspension as well as within a 3D cell culture platform. A 3D COMSOL model was built to simulate the mass loading response of increasing concentrations of cells in suspension in the polydimethylsiloxane (PDMS) well in order to predict the characteristics and optimize the design of the SH-SAW biosensor. The simulated relative frequency shift from the two oscillatory circuit systems (one of which functions as control) were found to be concordant to experimental data generated with RAW264.7 macrophage and A549 cancer cells. In addition, results showed that SAW measurements per se did not affect viability of cells. Further, SH-SAW biosensing was applied to A549 cells cultured on a 3D electrospun nanofiber scaffold that generate tumor spheroids (tumoroids) and the results showed the device's ability to detect changes in tumor spheroid growth over the course of eight days. Taken together, these results demonstrate the use of SH-SAW device for detection and quantification of cell growth changes over time in 2D suspension cultures and in 3D cell culture models, which may have potential applications in both longitudinal 3D cell cultures in cancer biology and in regenerative medicine.


Asunto(s)
Técnicas Biosensibles/métodos , Recuento de Células/métodos , Técnicas de Cultivo de Célula/métodos , Sonido , Animales , Proliferación Celular , Supervivencia Celular , Ratones , Células RAW 264.7 , Óxido de Zinc
16.
Stem Cells ; 31(7): 1321-9, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23533187

RESUMEN

Natriuretic peptide receptor A (NPRA), the signaling receptor for the cardiac hormone, atrial natriuretic peptide (ANP), is expressed abundantly in inflamed/injured tissues and tumors. NPRA deficiency substantially decreases tissue inflammation and inhibits tumor growth. However, the precise mechanism of NPRA function and whether it links inflammation and tumorigenesis remains unknown. Since both injury repair and tumor growth require stem cell recruitment and angiogenesis, we examined the role of NPRA signaling in tumor angiogenesis as a model of tissue injury repair in this study. In in vitro cultures, aortas from NPRA-KO mice show significantly lower angiogenic response compared to wild-type counterparts. The NPRA antagonist that decreases NPRA expression, inhibits lipopolysaccharide-induced angiogenesis. The reduction in angiogenesis correlates with decreased expression of vascular endothelial growth factor and chemokine (C-X-C motif) receptor 4 (CXCR4) implicating a cell recruitment defect. To test whether NPRA regulates migration of cells to tumors, mesenchymal stem cells (MSCs) were administered i.v., and the results showed that MSCs fail to migrate to the tumor microenvironment in NPRA-KO mice. However, coimplanting tumor cells with MSCs increases angiogenesis and tumorigenesis in NPRA-KO mice, in part by promoting expression of CXCR4 and its ligand, stromal cell-derived factor 1α. Taken together, these results demonstrate that NPRA signaling regulates stem cell recruitment and angiogenesis leading to tumor growth. Thus, NPRA signaling provides a key linkage between inflammation and tumorigenesis, and NPRA may be a target for drug development against cancers and tissue injury repair.


Asunto(s)
Carcinogénesis/metabolismo , Carcinoma Pulmonar de Lewis/irrigación sanguínea , Carcinoma Pulmonar de Lewis/metabolismo , Receptores del Factor Natriurético Atrial/metabolismo , Células Madre/citología , Células Madre/metabolismo , Animales , Carcinogénesis/genética , Carcinogénesis/patología , Carcinoma Pulmonar de Lewis/genética , Carcinoma Pulmonar de Lewis/patología , Femenino , Inmunohistoquímica , Inflamación/metabolismo , Inflamación/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Receptores del Factor Natriurético Atrial/genética , Transducción de Señal , Microambiente Tumoral
17.
Nanomedicine ; 10(7): 1539-48, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24486465

RESUMEN

Traumatic brain injury (TBI) causes significant mortality, long term disability and psychological symptoms. Gene therapy is a promising approach for treatment of different pathological conditions. Here we tested chitosan and polyethyleneimine (PEI)-coated magnetic micelles (CP-mag micelles or CPMMs), a potential MRI contrast agent, to deliver a reporter DNA to the brain after mild TBI (mTBI). CPMM-tomato plasmid (ptd) conjugate expressing a red-fluorescent protein (RFP) was administered intranasally immediately after mTBI or sham surgery in male SD rats. Evans blue extravasation following mTBI suggested CPMM-ptd entry into the brain via the compromised blood-brain barrier. Magnetofection increased the concentration of CPMMs in the brain. RFP expression was observed in the brain (cortex and hippocampus), lung and liver 48 h after mTBI. CPMM did not evoke any inflammatory response by themselves and were excreted from the body. These results indicate the possibility of using intranasally administered CPMM as a theranostic vehicle for mTBI. From the clinical editor: In this study, chitosan and PEI-coated magnetic micelles (CPMM) were demonstrated as potentially useful vehicles in traumatic brain injury in a rodent model. Magnetofection increased the concentration of CPMMs in the brain and, after intranasal delivery, CPMM did not evoke any inflammatory response and were excreted from the body.


Asunto(s)
Lesiones Encefálicas/terapia , ADN/administración & dosificación , Magnetismo , Micelas , Animales , Línea Celular , Medios de Contraste , Imagen por Resonancia Magnética , Masculino , Ratas , Ratas Sprague-Dawley
18.
Viruses ; 16(5)2024 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-38793666

RESUMEN

SARS-CoV-2 primarily infects the lungs via the ACE2 receptor but also other organs including the kidneys, the gastrointestinal tract, the heart, and the skin. SARS-CoV-2 also infects the brain, but the hematogenous route of viral entry to the brain is still not fully characterized. Understanding how SARS-CoV-2 traverses the blood-brain barrier (BBB) as well as how it affects the molecular functions of the BBB are unclear. In this study, we investigated the roles of the receptors ACE2 and DPP4 in the SARS-CoV-2 infection of the discrete cellular components of a transwell BBB model comprising HUVECs, astrocytes, and pericytes. Our results demonstrate that direct infection on the BBB model does not modulate paracellular permeability. Also, our results show that SARS-CoV-2 utilizes clathrin and caveolin-mediated endocytosis to traverse the BBB, resulting in the direct infection of the brain side of the BBB model with a minimal endothelial infection. In conclusion, the BBB is susceptible to SARS-CoV-2 infection in multiple ways, including the direct infection of endothelium, astrocytes, and pericytes involving ACE2 and/or DPP4 and the blood-to-brain transcytosis, which is an event that does not require the presence of host receptors.


Asunto(s)
Enzima Convertidora de Angiotensina 2 , Astrocitos , Barrera Hematoencefálica , COVID-19 , Dipeptidil Peptidasa 4 , Pericitos , SARS-CoV-2 , Transcitosis , Internalización del Virus , Barrera Hematoencefálica/virología , Barrera Hematoencefálica/metabolismo , Humanos , SARS-CoV-2/fisiología , Enzima Convertidora de Angiotensina 2/metabolismo , Pericitos/virología , Pericitos/metabolismo , COVID-19/virología , COVID-19/metabolismo , Astrocitos/virología , Astrocitos/metabolismo , Dipeptidil Peptidasa 4/metabolismo , Encéfalo/virología , Encéfalo/metabolismo , Endocitosis , Células Endoteliales de la Vena Umbilical Humana/virología , Permeabilidad
19.
J Biomed Inform ; 46(2): 341-53, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23376273

RESUMEN

We propose a new method to mitigate (identify and address) adverse interactions (drug-drug or drug-disease) that occur when a patient with comorbid diseases is managed according to two concurrently applied clinical practice guidelines (CPGs). A lack of methods to facilitate the concurrent application of CPGs severely limits their use in clinical practice and the development of such methods is one of the grand challenges for clinical decision support. The proposed method responds to this challenge. We introduce and formally define logical models of CPGs and other related concepts, and develop the mitigation algorithm that operates on these concepts. In the algorithm we combine domain knowledge encoded as interaction and revision operators using the constraint logic programming (CLP) paradigm. The operators characterize adverse interactions and describe revisions to logical models required to address these interactions, while CLP allows us to efficiently solve the logical models - a solution represents a feasible therapy that may be safely applied to a patient. The mitigation algorithm accepts two CPGs and available (likely incomplete) patient information. It reports whether mitigation has been successful or not, and on success it gives a feasible therapy and points at identified interactions (if any) together with the revisions that address them. Thus, we consider the mitigation algorithm as an alerting tool to support a physician in the concurrent application of CPGs that can be implemented as a component of a clinical decision support system. We illustrate our method in the context of two clinical scenarios involving a patient with duodenal ulcer who experiences an episode of transient ischemic attack.


Asunto(s)
Algoritmos , Sistemas de Apoyo a Decisiones Clínicas , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/prevención & control , Guías de Práctica Clínica como Asunto , Enfermedad Aguda , Enfermedad Crónica , Comorbilidad , Interacciones Farmacológicas , Humanos , Modelos Biológicos
20.
Nanomedicine ; 9(7): 903-11, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23352802

RESUMEN

A near infrared (NIR) triggered drug delivery platform based on the chitosan-modified chemically reduced graphene oxide (CRGO) incorporated into a thermosensitive nanogel (CGN) was developed. CGN exhibited an NIR-induced thermal effect similar to that of CRGO, reversible thermo-responsive characteristics at 37-42 °C and high doxorubicin hydrochloride (DOX) loading capacity (48 wt%). The DOX loaded CGN (DOX-CGN) released DOX faster at 42 °C than at 37 °C. The fluorescence images revealed DOX expression in the cytoplasm of cancer cells when incubated with DOX-CGN at 37 °C but in the nucleus at 42 °C. Upon irradiation with NIR light (808 nm), a rapid, repetitive DOX release from the DOX-CGN was observed. Furthermore, the cancer cells incubated with DOX-CGN and irradiated with NIR light displayed significantly greater cytotoxicity than without irradiation owing to NIR-triggered increase in temperature leading to nuclear DOX release. These results demonstrate CGN's promising application for on-demand drug release by NIR light. FROM THE CLINICAL EDITOR: These investigators report the successful development of a novel near infrared triggered drug delivery platform based on chitosan-modified chemically reduced graphene oxide (CRGO) incorporated into a thermosensitive nanogel (CGN).


Asunto(s)
Quitosano/química , Doxorrubicina/farmacología , Grafito/química , Polietilenglicoles/química , Polietileneimina/química , Animales , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Quitosano/toxicidad , Preparaciones de Acción Retardada , Doxorrubicina/química , Endocitosis/efectos de los fármacos , Grafito/toxicidad , Hidrodinámica , Rayos Infrarrojos , Ratones , Nanogeles , Polietilenglicoles/toxicidad , Polietileneimina/toxicidad , Fracciones Subcelulares/efectos de los fármacos , Fracciones Subcelulares/metabolismo , Temperatura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA