Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell ; 78(4): 785-793.e8, 2020 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-32229306

RESUMEN

RNA polymerase II (RNAPII) transcription is governed by the pre-initiation complex (PIC), which contains TFIIA, TFIIB, TFIID, TFIIE, TFIIF, TFIIH, RNAPII, and Mediator. After initiation, RNAPII enzymes pause after transcribing less than 100 bases; precisely how RNAPII pausing is enforced and regulated remains unclear. To address specific mechanistic questions, we reconstituted human RNAPII promoter-proximal pausing in vitro, entirely with purified factors (no extracts). As expected, NELF and DSIF increased pausing, and P-TEFb promoted pause release. Unexpectedly, the PIC alone was sufficient to reconstitute pausing, suggesting RNAPII pausing is an inherent PIC function. In agreement, pausing was lost upon replacement of the TFIID complex with TATA-binding protein (TBP), and PRO-seq experiments revealed widespread disruption of RNAPII pausing upon acute depletion (t = 60 min) of TFIID subunits in human or Drosophila cells. These results establish a TFIID requirement for RNAPII pausing and suggest pause regulatory factors may function directly or indirectly through TFIID.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Regiones Promotoras Genéticas , ARN Polimerasa II/genética , Factor de Transcripción TFIID/metabolismo , Transcripción Genética , Animales , Drosophila/genética , Proteínas de Drosophila/genética , Células HCT116 , Humanos , Unión Proteica , ARN Polimerasa II/metabolismo , Factor de Transcripción TFIID/genética
2.
Nephrology (Carlton) ; 21(2): 81-5, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26461175

RESUMEN

Regulatory T cells (Tregs) have been recognized as having a major role in maintaining peripheral tolerance and preventing and limiting autoimmune and chronic inflammatory diseases. Tregs derive from the thymus and also develop peripherally. In this review, we discuss recent progress in our understanding of the basic mechanisms involved in Treg development and function in protecting against autoimmunity in the periphery, including thymic selection, peripheral induction and the many mechanisms of Treg suppression. Specifically in kidney disease, Tregs have been shown to play a role in limiting injury and may potentially have a therapeutic role.


Asunto(s)
Autoinmunidad , Factores de Transcripción Forkhead/inmunología , Tolerancia Inmunológica , Riñón/inmunología , Linfocitos T Reguladores/inmunología , Animales , Citocinas/inmunología , Citocinas/metabolismo , Metilación de ADN , Factores de Transcripción Forkhead/metabolismo , Glomerulonefritis/inmunología , Glomerulonefritis/metabolismo , Humanos , Riñón/metabolismo , Activación de Linfocitos , Fenotipo , Insuficiencia Renal Crónica/inmunología , Insuficiencia Renal Crónica/metabolismo , Transducción de Señal , Linfocitos T Reguladores/metabolismo
3.
Nucleic Acids Res ; 41(5): 3327-38, 2013 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-23358821

RESUMEN

The chemical strategies used by ribozymes to enhance reaction rates are revealed in part from their metal ion and pH requirements. We find that kinase ribozyme K28(1-77)C, in contrast with previously characterized kinase ribozymes, requires Cu(2+) for optimal catalysis of thiophosphoryl transfer from GTPγS. Phosphoryl transfer from GTP is greatly reduced in the absence of Cu(2+), indicating a specific catalytic role independent of any potential interactions with the GTPγS thiophosphoryl group. In-line probing and ATPγS competition both argue against direct Cu(2+) binding by RNA; rather, these data establish that Cu(2+) enters the active site within a Cu(2+)•GTPγS or Cu(2+)•GTP chelation complex, and that Cu(2+)•nucleobase interactions further enforce Cu(2+) selectivity and position the metal ion for Lewis acid catalysis. Replacing Mg(2+) with [Co(NH3)6](3+) significantly reduced product yield, but not kobs, indicating that the role of inner-sphere Mg(2+) coordination is structural rather than catalytic. Replacing Mg(2+) with alkaline earths of increasing ionic radii (Ca(2+), Sr(2+) and Ba(2+)) gave lower yields and approximately linear rates of product accumulation. Finally, we observe that reaction rates increased with pH in log-linear fashion with an apparent pKa = 8.0 ± 0.1, indicating deprotonation in the rate-limiting step.


Asunto(s)
Complejos de Coordinación/química , Cobre/química , Fosfotransferasas/química , ARN Catalítico/química , Adenosina Trifosfato/análogos & derivados , Adenosina Trifosfato/química , Secuencia de Bases , Tampones (Química) , Catálisis , Dominio Catalítico , Concentración de Iones de Hidrógeno , Ácidos de Lewis , Magnesio/química , Conformación de Ácido Nucleico , Fosforilación
4.
Nucleic Acids Res ; 41(3): 1873-84, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23241386

RESUMEN

Systematic evolution of ligands through exponential enrichment (SELEX) is a well-established method for generating nucleic acid populations that are enriched for specified functions. High-throughput sequencing (HTS) enhances the power of comparative sequence analysis to reveal details of how RNAs within these populations recognize their targets. We used HTS analysis to evaluate RNA populations selected to bind type I human immunodeficiency virus reverse transcriptase (RT). The populations are enriched in RNAs of independent lineages that converge on shared motifs and in clusters of RNAs with nearly identical sequences that share common ancestry. Both of these features informed inferences of the secondary structures of enriched RNAs, their minimal structural requirements and their stabilities in RT-aptamer complexes. Monitoring population dynamics in response to increasing selection pressure revealed RNA inhibitors of RT that are more potent than the previously identified pseudoknots. Improved potency was observed for inhibition of both purified RT in enzymatic assays and viral replication in cell-based assays. Structural and functional details of converged motifs that are obscured by simple consensus descriptions are also revealed by the HTS analysis. The approach presented here can readily be generalized for the efficient and systematic post-SELEX development of aptamers for down-stream applications.


Asunto(s)
Fármacos Anti-VIH/química , Aptámeros de Nucleótidos/química , Transcriptasa Inversa del VIH/antagonistas & inhibidores , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Inhibidores de la Transcriptasa Inversa/química , Análisis de Secuencia de ARN/métodos , Fármacos Anti-VIH/farmacología , Aptámeros de Nucleótidos/farmacología , Secuencia de Bases , Secuencia de Consenso , VIH-1/efectos de los fármacos , VIH-1/fisiología , Motivos de Nucleótidos , Inhibidores de la Transcriptasa Inversa/farmacología , Técnica SELEX de Producción de Aptámeros , Replicación Viral/efectos de los fármacos
5.
J Am Soc Nephrol ; 23(8): 1303-8, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22677553

RESUMEN

Regulatory T cells (Tregs) help protect against autoimmune renal injury. The use of agonist antibodies and antibody/cytokine combinations to expand Tregs in vivo may have therapeutic potential for renal disease. Here, we investigated the effects of administering IL-2/IL-2Ab complexes in mice with adriamycin nephropathy, a model of proteinuric kidney disease that resembles human focal segmental glomerulosclerosis. Injecting IL-2/IL-2Ab complexes before or, to a lesser extent, after induction of disease promoted expansion of Tregs. Furthermore, administration of this complex was renoprotective, evidenced by improved renal function, maintenance of body weight, less histologic injury, and reduced inflammation. IL-2/IL-2Ab reduced serum IL-6 and renal expression of IL-6 and IL-17 but enhanced expression of IL-10 and Foxp3 in the spleen. In vitro, the addition of IL-2/IL-2Ab complexes induced rapid STAT-5 phosphorylation in CD4 T cells. In summary, these data suggest that inducing the expansion of Tregs by administering IL-2/IL-2Ab complexes is a possible strategy to treat renal disease.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Glomeruloesclerosis Focal y Segmentaria/prevención & control , Interleucina-2/uso terapéutico , Proteinuria/prevención & control , Linfocitos T Reguladores/efectos de los fármacos , Animales , Antibióticos Antineoplásicos , Anticuerpos Monoclonales/farmacología , Doxorrubicina , Glomeruloesclerosis Focal y Segmentaria/inducido químicamente , Interleucina-2/farmacología , Masculino , Ratones , Ratones Endogámicos BALB C , Proteinuria/inducido químicamente
6.
J Am Soc Nephrol ; 23(6): 1058-67, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22491420

RESUMEN

Autoreactive T cells play a pivotal role in the pathogenesis of autoimmune kidney disease. T cell vaccination (TCV) may limit autoimmune disease and induce CD8+ regulatory T cells (Tregs). We used Heymann nephritis (HN), a rat model of human membranous nephritis, to study the effects of TCV on autoimmune kidney disease. We harvested CD4+ T cells from renal tubular antigen (Fx1A) -immunized rats and activated these cells in vitro to express the MHC Class Ib molecule Qa-1. Vaccination of Lewis rats with these autoreactive Fx1A-induced T cells protected against HN, whereas control-primed T cells did not. Rats that underwent TCV had lower levels of proteinuria and serum creatinine and significantly less glomerulosclerosis, tubular damage, and interstitial infiltrates. Furthermore, these rats expressed less IFN-γ and IL-6 in splenocytes, whereas the numbers of Tregs and the expression of Foxp3 were unchanged. In vitro cytotoxicity assays showed CD8+ T cell-mediated elimination of Qa-1-expressing CD4+ T cells. In vivo, TCV abrogated the increase in Qa-1-expressing CXCR5+ TFH cells observed in HN compared with controls. Taken together, these results suggest that TCV protects against autoimmune kidney disease by targeting Qa-1-expressing autoreactive CD4+ cells.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Linfocitos T CD8-positivos/metabolismo , Citocinas/metabolismo , Glomerulonefritis Membranosa/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Vacunación/métodos , Análisis de Varianza , Animales , Autoanticuerpos/inmunología , Autoanticuerpos/metabolismo , Enfermedades Autoinmunes/metabolismo , Enfermedades Autoinmunes/patología , Linfocitos T CD4-Positivos/metabolismo , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Glomerulonefritis Membranosa/metabolismo , Glomerulonefritis Membranosa/patología , Humanos , Inmunohistoquímica , Masculino , Ratas , Ratas Endogámicas Lew , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Receptores de Antígenos de Linfocitos T alfa-beta/biosíntesis , Muestreo , Linfocitos T/inmunología
7.
J Exp Med ; 220(6)2023 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-36920308

RESUMEN

The hallmark of tuberculosis (TB) is the formation of immune cell-enriched aggregates called granulomas. While granulomas are pathologically diverse, their tissue-wide heterogeneity has not been spatially resolved at the single-cell level in human tissues. By spatially mapping individual immune cells in every lesion across entire tissue sections, we report that in addition to necrotizing granulomas, the human TB lung contains abundant non-necrotizing leukocyte aggregates surrounding areas of necrotizing tissue. These cellular lesions were more diverse in composition than necrotizing lesions and could be stratified into four general classes based on cellular composition and spatial distribution of B cells and macrophages. The cellular composition of non-necrotizing structures also correlates with their proximity to necrotizing lesions, indicating these are foci of distinct immune reactions adjacent to necrotizing granulomas. Together, we show that during TB, diseased lung tissue develops a histopathological superstructure comprising at least four different types of non-necrotizing cellular aggregates organized as satellites of necrotizing granulomas.


Asunto(s)
Mycobacterium tuberculosis , Tuberculosis , Humanos , Granuloma/patología , Pulmón/patología , Macrófagos
8.
Am J Pathol ; 179(2): 860-8, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21704005

RESUMEN

Thrombospondin (TSP)-2-null mice have an altered brain foreign body response (FBR) characterized by increases in inflammation, extracellular matrix deposition, and leakage of the blood-brain barrier (BBB). In the present study, we investigated the role of TSP-2 in BBB repair during the brain FBR to mixed cellulose ester filters implanted in the cortex of wild-type (WT) and TSP-2-null mice for 2 days to 8 weeks. Histological and immunohistochemical analysis revealed enhanced and prolonged neuroinflammation in TSP-2-null mice up to 8 weeks after implantation. In addition, recovery of the BBB was compromised and was associated with increased gelatinolytic activity and low levels of collagen type IV in the basement membranes of TSP-2-null blood vessels. An analysis of protein extracts from implantation sites revealed elevated levels of matrix metalloproteinase (MMP)-2 and MMP-9 in TSP-2-null brains. TSP-2-null astrocytes secreted higher levels of both MMPs in vitro compared with their WT counterparts. Furthermore, TSP-2-null astrocytes were deficient in supporting the recovery of barrier function in WT endothelial cells. Finally, Western blot analysis of astrocytes and brain endothelial cells revealed TSP-2 expression only in the former. Taken together, our observations suggest that astrocyte-derived TSP-2 is critical for the maintenance of physiological MMP-2 and MMP-9 levels during the FBR and contributes to the repair of the BBB.


Asunto(s)
Astrocitos/citología , Barrera Hematoencefálica/metabolismo , Trombospondinas/metabolismo , Animales , Materiales Biocompatibles , Encéfalo/metabolismo , Circulación Cerebrovascular , Macrófagos/metabolismo , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Microcirculación , Microglía/metabolismo , Albúmina Sérica/metabolismo , Factores de Tiempo
10.
Front Immunol ; 12: 634119, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33868254

RESUMEN

Cystatin F encoded by CST7 is a cysteine peptidase inhibitor known to be expressed in natural killer (NK) and CD8+ T cells during steady-state conditions. However, little is known about its expression during inflammatory disease states in humans. We have developed an analytic approach capable of not only identifying previously poorly characterized disease-associated genes but also defining regulatory mechanisms controlling their expression. By exploring multiple cohorts of public transcriptome data comprising 43 individual datasets, we showed that CST7 is upregulated in the blood during a diverse set of infectious and non-infectious inflammatory conditions. Interestingly, this upregulation of CST7 was neutrophil-specific, as its expression was unchanged in NK and CD8+ T cells during sepsis. Further analysis demonstrated that known microbial products or cytokines commonly associated with inflammation failed to increase CST7 expression, suggesting that its expression in neutrophils is induced by an endogenous serum factor commonly present in human inflammatory conditions. Overall, through the identification of CST7 upregulation as a marker of acute inflammation in humans, our study demonstrates the value of publicly available transcriptome data in knowledge generation and potential biomarker discovery.


Asunto(s)
Biomarcadores de Tumor/genética , Cistatinas/genética , Perfilación de la Expresión Génica , Inflamación/genética , Neutrófilos/metabolismo , Sepsis/genética , Transcriptoma , Enfermedad Aguda , Biomarcadores de Tumor/sangre , Estudios de Casos y Controles , Cistatinas/sangre , Bases de Datos Genéticas , Humanos , Inflamación/sangre , Inflamación/inmunología , Neutrófilos/inmunología , Sepsis/sangre , Sepsis/inmunología , Regulación hacia Arriba
11.
Mol Cancer Ther ; 19(1): 270-281, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31597714

RESUMEN

Combinations of chemotherapy with immunotherapy have seen recent clinical success, including two approvals of anti-PD-1/L1 agents in combination with taxane-based chemotherapy in non-small cell lung cancer and triple-negative breast cancer. Here, we present a study on the combination activity and mechanistic rationale of a novel EphA2-targeted liposomal taxane (EphA2-ILs-DTXp) and anti-PD-1. This combination was highly active in mouse syngeneic tumor models, with complete responses observed in 3 of 5 models. In the EMT-6 tumor model, combination of EphA2-ILs-DTXp with anti-PD-1 resulted in a 60% complete response rate, with durable responses that were resistant to rechallenge. These responses were not observed in the absence of CD8+ T cells. Characterization of the immune infiltrates in EMT-6 tumors reveals increased CD8+ T cells, increased CD8+ IFNγ+ CTLs, and an increased CD8/regulatory T-cell (Treg) ratio. These immunomodulatory effects were not observed in mice treated with a combination of docetaxel and anti-PD-1. Pharmacokinetic analysis revealed that the AUC of docetaxel was increased 15 times, from 52.1 to 785 ng/mL/hour, when delivered by EphA2-ILs-DTXp. A dose reduction study of EphA2-ILs-DTXp showed a dose-response relationship for both tumor growth inhibition and the CD8/Treg ratio. Our data indicate that synergism between docetaxel and anti-PD-1 is achievable with nanoliposomal delivery.


Asunto(s)
Hidrocarburos Aromáticos con Puentes/uso terapéutico , Neoplasias/tratamiento farmacológico , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor EphA2/metabolismo , Taxoides/uso terapéutico , Animales , Hidrocarburos Aromáticos con Puentes/farmacología , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Neoplasias/patología , Taxoides/farmacología
12.
Cell Rep ; 29(11): 3539-3550.e4, 2019 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-31825834

RESUMEN

Interferons (IFN) are pleiotropic cytokines essential for defense against infection, but the identity and tissue distribution of IFN-responsive cells in vivo are poorly defined. In this study, we generate a mouse strain capable of reporting IFN-signaling activated by all three types of IFNs and investigate the spatio-temporal dynamics and identity of IFN-responding cells following IFN injection and influenza virus infection. Despite ubiquitous expression of IFN receptors, cellular responses to IFNs are highly heterogenous in vivo and are determined by anatomical site, cell type, cellular preference to individual IFNs, and activation status. Unexpectedly, type I and II pneumocytes, the primary target of influenza infection, exhibit striking differences in the strength and temporal dynamics of IFN signaling associated with differential susceptibility to the viral infection. Our findings suggest that time- and cell-type-dependent integration of distinct IFN signals govern the specificity and magnitude of IFN responses in vivo.


Asunto(s)
Interferones/metabolismo , Infecciones por Orthomyxoviridae/metabolismo , Transducción de Señal , Células Epiteliales Alveolares/metabolismo , Animales , Células Cultivadas , Femenino , Proteínas de Unión al GTP/genética , Proteínas de Unión al GTP/metabolismo , Células Madre Hematopoyéticas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL
13.
Nat Biomed Eng ; 3(4): 264-280, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30952988

RESUMEN

Antibody-mediated tumour targeting and nanoparticle-mediated encapsulation can reduce the toxicity of antitumour drugs and improve their efficacy. Here, we describe the performance of a nanotherapeutic encapsulating a hydrolytically sensitive docetaxel prodrug and conjugated to an antibody specific for EphA2-a receptor overexpressed in many tumours. Administration of the nanotherapeutic in mice led to slow and sustained release of the prodrug, reduced exposure of active docetaxel in the circulation (compared with administration of the free drug) and maintenance of optimal exposure of the drug in tumour tissue. We also show that administration of the nanotherapeutic in rats and dogs resulted in minimal haematological toxicity, as well as the absence of neutropenia and improved overall tolerability in multiple rodent models. Targeting of the nanotherapeutic to EphA2 improved tumour penetration and resulted in markedly enhanced antitumour activity (compared with administration of free docetaxel and non-targeted nanotherapeutic controls) in multiple tumour-xenografted mice. This nanomedicine could become a potent and safe therapeutic alternative for cancer patients undergoing chemotherapy.


Asunto(s)
Antineoplásicos/uso terapéutico , Nanopartículas/uso terapéutico , Receptor EphA2/metabolismo , Animales , Antineoplásicos/farmacología , Hidrocarburos Aromáticos con Puentes/farmacología , Hidrocarburos Aromáticos con Puentes/uso terapéutico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Docetaxel/sangre , Docetaxel/química , Docetaxel/farmacocinética , Docetaxel/uso terapéutico , Humanos , Liposomas , Ratones Endogámicos NOD , Ratones SCID , Taxoides/farmacología , Taxoides/uso terapéutico , Distribución Tisular/efectos de los fármacos , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Cell Rep ; 25(6): 1458-1468.e4, 2018 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-30404002

RESUMEN

Tumor ecosystems are composed of multiple cell types that communicate by ligand-receptor interactions. Targeting ligand-receptor interactions (for instance, with immune checkpoint inhibitors) can provide significant benefits for patients. However, our knowledge of which interactions occur in a tumor and how these interactions affect outcome is still limited. We present an approach to characterize communication by ligand-receptor interactions across all cell types in a microenvironment using single-cell RNA sequencing. We apply this approach to identify and compare the ligand-receptor interactions present in six syngeneic mouse tumor models. To identify interactions potentially associated with outcome, we regress interactions against phenotypic measurements of tumor growth rate. In addition, we quantify ligand-receptor interactions between T cell subsets and their relation to immune infiltration using a publicly available human melanoma dataset. Overall, this approach provides a tool for studying cell-cell interactions, their variability across tumors, and their relationship to outcome.


Asunto(s)
Comunicación Celular , Neoplasias/patología , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Ligandos , Melanoma/patología , Ratones , Metástasis de la Neoplasia , Fenotipo , Receptores de Superficie Celular/metabolismo , Microambiente Tumoral
15.
Mol Ther Nucleic Acids ; 13: 605-619, 2018 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-30472639

RESUMEN

Aptamer selections often yield distinct subpopulations, each with unique phenotypes that can be leveraged for specialized applications. Although most selections aim to attain ever higher specificity, we sought to identify aptamers that recognize increasingly divergent primate lentiviral reverse transcriptases (RTs). We hypothesized that aptamer subpopulations in libraries pre-enriched against a single RT may exhibit broad-spectrum binding and inhibition, and we devised a multiplexed poly-target selection to elicit those phenotypes against a panel of primate lentiviral RTs. High-throughput sequencing and coenrichment/codepletion analysis of parallel and duplicate selection trajectories rapidly narrowed the list of candidate aptamers by orders of magnitude and identified dozens of priority candidates for further screening. Biochemical characterization validated a novel aptamer motif and several rare and unobserved variants of previously known motifs that inhibited recombinant RTs to varying degrees. These broad-spectrum aptamers also suppressed replication of viral constructs carrying phylogenetically diverse RTs. The poly-target selection and coenrichment/codepletion approach described herein is a generalizable strategy for identifying cross-reactivity among related targets from combinatorial libraries.

16.
Brain Res ; 1180: 121-32, 2007 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-17920047

RESUMEN

This study investigates methods of manipulating the brain extracellular matrix (ECM) to enhance the penetration of nanoparticle drug carriers in convection-enhanced delivery (CED). A probe was fabricated with two independent microfluidic channels to infuse, either simultaneously or sequentially, nanoparticles and ECM-modifying agents. Infusions were performed in the striatum of the normal rat brain. Monodisperse polystyrene particles with a diameter of 54 nm were used as a model nanoparticle system. Because the size of these particles is comparable to the effective pore size of the ECM, their transport may be significantly hindered compared with the transport of low molecular weight molecules. To enhance the transport of the infused nanoparticles, we attempted to increase the effective pore size of the ECM by two methods: dilating the extracellular space and degrading selected constituents of the ECM. Two methods of dilating the extracellular space were investigated: co-infusion of nanoparticles and a hyperosmolar solution of mannitol, and pre-infusion of an isotonic buffer solution followed by infusion of nanoparticles. These treatments resulted in an increase in the nanoparticle distribution volume of 51% and 123%, respectively. To degrade hyaluronan, a primary structural component of the brain ECM, a pre-infusion of hyaluronidase (20,000 U/mL) was followed after 30 min by infusion of nanoparticles. This treatment resulted in an increase in the nanoparticle distribution of 64%. Our results suggest that both dilation and enzymatic digestion can be incorporated into CED protocols to enhance nanoparticle penetration.


Asunto(s)
Dilatación/métodos , Sistemas de Liberación de Medicamentos/métodos , Líquido Extracelular/efectos de los fármacos , Matriz Extracelular/efectos de los fármacos , Nanopartículas/administración & dosificación , Neostriado/efectos de los fármacos , Animales , Convección , Sistemas de Liberación de Medicamentos/instrumentación , Líquido Extracelular/metabolismo , Matriz Extracelular/metabolismo , Hialuronoglucosaminidasa/administración & dosificación , Hialuronoglucosaminidasa/metabolismo , Masculino , Microelectrodos , Peso Molecular , Nanopartículas/química , Neostriado/metabolismo , Concentración Osmolar , Polímeros/administración & dosificación , Polímeros/farmacocinética , Poliestirenos/administración & dosificación , Poliestirenos/farmacocinética , Ratas , Ratas Sprague-Dawley , Distribución Tisular
17.
Vet Parasitol ; 237: 104-109, 2017 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-28259556

RESUMEN

A randomized clinical study was conducted in a total of 45 commercial dairy farms in France (14 farms), Germany (28 farms) and the UK (3 farms) to evaluate the effect of an anthelmintic treatment on milk yield in the subsequent lactation. A total of 1287 animals with suspected exposure to Ostertagia ostertagi were included in the study. Animals were treated during the dry period (7-77days before parturition) with moxidectin pour-on (Cydectin® 0.5% Pour-On, Zoetis; 638 animals) or left untreated (649 animals) according to a randomized block design. Animals were either heifers (n=296) or multiparous cows (n=991). The milk production was monitored at regular intervals after treatment up to 335days after lactation, and analysed using a general linear mixed model with the milk production as outcome variable and several random effects. The effect on milk yield after anthelmintic treatment over the whole subsequent lactation varied from no effect (-0.43kg/day; P=0.35) to an increase of milk yield with 2.35kg/day (P=0.01), depending on the study region and parity of the cows. Lactation curve analysis suggested that the treatment effect was mainly caused by a slower decay of the milk production in the treated animals compared to untreated animals. The present study highlights the beneficial effect of a topical treatment with moxidectin before parturition on milk yield in the subsequent lactation, as well as the importance of a careful evaluation of nematode exposure risk based on local grazing management practices to guide and target production-based anthelmintic treatment decisions.


Asunto(s)
Antihelmínticos/uso terapéutico , Enfermedades de los Bovinos/tratamiento farmacológico , Macrólidos/uso terapéutico , Leche/efectos de los fármacos , Ostertagia/efectos de los fármacos , Ostertagiasis/veterinaria , Animales , Bovinos , Enfermedades de los Bovinos/epidemiología , Enfermedades de los Bovinos/parasitología , Heces/parasitología , Femenino , Francia/epidemiología , Alemania/epidemiología , Ivermectina/uso terapéutico , Lactancia/efectos de los fármacos , Leche/metabolismo , Ostertagia/aislamiento & purificación , Ostertagiasis/tratamiento farmacológico , Ostertagiasis/epidemiología , Ostertagiasis/parasitología , Recuento de Huevos de Parásitos/veterinaria , Embarazo , Reino Unido/epidemiología
18.
Regen Med ; 12(3): 249-261, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28524773

RESUMEN

AIM: Inflammatory myeloid lineage cells mediate neotissue formation in tissue-engineered vascular grafts, but the molecular mechanism is not completely understood. We examined the role of vasculogenic PDGF-B in tissue-engineered vascular graft neotissue development. MATERIALS & METHODS: Myeloid cell-specific PDGF-B knockout mice (PDGF-KO) were generated using bone marrow transplantation, and scaffolds were implanted as inferior vena cava interposition grafts in either PDGF-KO or wild-type mice. RESULTS: After 2 weeks, grafts from PDGF-KO mice had more remaining scaffold polymer and less intimal neotissue development. Increased macrophage apoptosis, decreased smooth muscle cell proliferation and decreased collagen content was also observed. CONCLUSION: Myeloid cell-derived PDGF contributes to vascular neotissue formation by regulating macrophage apoptosis, smooth muscle cell proliferation and extracellular matrix deposition.


Asunto(s)
Bioprótesis , Prótesis Vascular , Linfocinas/metabolismo , Células Mieloides/metabolismo , Neointima/metabolismo , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Ingeniería de Tejidos , Vena Cava Inferior/cirugía , Animales , Diferenciación Celular , Linfocinas/genética , Ratones , Ratones Noqueados , Células Mieloides/patología , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Neointima/genética , Neointima/patología , Factor de Crecimiento Derivado de Plaquetas/genética , Vena Cava Inferior/metabolismo , Vena Cava Inferior/patología
19.
Yale J Biol Med ; 79(3-4): 141-52, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17940624

RESUMEN

Despite advances in diagnostic imaging and drug discovery, primary malignant brain tumors remain fatal. Median survival for patients with the most severe forms is rarely past eight months. The severity of the disease and the lack of substantial improvement in patient survival demand that new approaches be explored in drug delivery to brain tumors. Recently, local delivery of chemotherapy to brain tumors has provided a way to circumvent the blood-brain barrier, allowing delivery of chemotherapy drugs directly to malignant cells in the brain. Two methods of local delivery have been developed: polymeric-controlled release and convection-enhanced delivery. Controlled release utilizes degradable or non-degradable polymers as carriers of chemotherapy; polymer implants or microparticles are implanted locally to introduce a sustained source of drug for periods of days or months. Convection-enhanced delivery employs the bulk flow of drugs dissolved in fluid, which is introduced intracranially using a catheter and pump. The convective fluid flow is capable of delivering drugs great distances within the brain, potentially treating invasive cells at a distance from the catheter infusion site. These two new delivery strategies are capable of delivering both standard chemotherapeutic drugs and new methods of anti-cancer therapy. Taken individually, or used in tandem, they represent a potential revolution in brain cancer treatment.


Asunto(s)
Antineoplásicos/administración & dosificación , Neoplasias Encefálicas/tratamiento farmacológico , Sistemas de Liberación de Medicamentos , Glioma/tratamiento farmacológico , Materiales Biocompatibles/química , Ensayos Clínicos como Asunto , Portadores de Fármacos , Implantes de Medicamentos , Humanos , Ácido Láctico/química , Microscopía Electrónica de Rastreo , Metástasis de la Neoplasia , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Polímeros/química , Poliestirenos/química
20.
Adv Drug Deliv Rev ; 97: 56-68, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26763408

RESUMEN

Extracellular matrix is composed of a complex array of molecules that together provide structural and functional support to cells. These properties are mainly mediated by the activity of collagenous and elastic fibers, proteoglycans, and proteins such as fibronectin and laminin. ECM composition is tissue-specific and could include matricellular proteins whose primary role is to modulate cell-matrix interactions. In adults, matricellular proteins are primarily expressed during injury, inflammation and disease. Particularly, they are closely associated with the progression and prognosis of cardiovascular and fibrotic diseases, and cancer. This review aims to provide an overview of the potential use of matricellular proteins in drug delivery including the generation of therapeutic agents based on the properties and structures of these proteins as well as their utility as biomarkers for specific diseases.


Asunto(s)
Sistemas de Liberación de Medicamentos , Proteínas de la Matriz Extracelular/uso terapéutico , Animales , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA