Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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.

9.
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
10.
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
11.
Sci Rep ; 6: 33277, 2016 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-27615159

RESUMEN

Cell-cell fusion is fundamental to a multitude of biological processes ranging from cell differentiation and embryogenesis to cancer metastasis and biomaterial-tissue interactions. Fusogenic cells are exposed to biochemical and biophysical factors, which could potentially alter cell behavior. While biochemical inducers of fusion such as cytokines and kinases have been identified, little is known about the biophysical regulation of cell-cell fusion. Here, we designed experiments to examine cell-cell fusion using bulk metallic glass (BMG) nanorod arrays with varying biophysical cues, i.e. nanotopography and stiffness. Through independent variation of stiffness and topography, we found that nanotopography constitutes the primary biophysical cue that can override biochemical signals to attenuate fusion. Specifically, nanotopography restricts cytoskeletal remodeling-associated signaling, which leads to reduced fusion. This finding expands our fundamental understanding of the nanoscale biophysical regulation of cell fusion and can be exploited in biomaterials design to induce desirable biomaterial-tissue interactions.


Asunto(s)
Macrófagos/fisiología , Nanoestructuras/ultraestructura , Óxido de Aluminio/química , Animales , Técnicas de Cultivo de Célula , Fusión Celular , Línea Celular , Medios de Cultivo , Citoesqueleto/metabolismo , Activación Enzimática , Sistema de Señalización de MAP Quinasas , Ratones , Nanoestructuras/química , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
13.
J Control Release ; 232: 103-12, 2016 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-27063424

RESUMEN

Glioblastoma multiforme (GBM) is a fatal brain tumor characterized by infiltration beyond the margins of the main tumor mass and local recurrence after surgery. The blood-brain barrier (BBB) poses the most significant hurdle to brain tumor treatment. Convection-enhanced delivery (CED) allows for local administration of agents, overcoming the restrictions of the BBB. Recently, polymer nanoparticles have been demonstrated to penetrate readily through the healthy brain when delivered by CED, and size has been shown to be a critical factor for nanoparticle penetration. Because these brain-penetrating nanoparticles (BPNPs) have high potential for treatment of intracranial tumors since they offer the potential for cell targeting and controlled drug release after administration, here we investigated the intratumoral CED infusions of PLGA BPNPs in animals bearing either U87 or RG2 intracranial tumors. We demonstrate that the overall volume of distribution of these BPNPs was similar to that observed in healthy brains; however, the presence of tumors resulted in asymmetric and heterogeneous distribution patterns, with substantial leakage into the peritumoral tissue. Together, our results suggest that CED of BPNPs should be optimized by accounting for tumor geometry, in terms of location, size and presence of necrotic regions, to determine the ideal infusion site and parameters for individual tumors.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Convección , Sistemas de Liberación de Medicamentos , Ácido Láctico/administración & dosificación , Nanopartículas/administración & dosificación , Ácido Poliglicólico/administración & dosificación , Animales , Encéfalo/metabolismo , Encéfalo/patología , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Glioblastoma/tratamiento farmacológico , Glioblastoma/metabolismo , Proteínas Fluorescentes Verdes/administración & dosificación , Proteínas Fluorescentes Verdes/farmacocinética , Humanos , Masculino , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Ratas Endogámicas F344 , Ratas Desnudas , Ratas Sprague-Dawley , Carga Tumoral
14.
Biomaterials ; 89: 127-35, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26967647

RESUMEN

The foreign body response (FBR) begins with injury acquired during implantation of a biomaterial (BM) and is detrimental due to the eventual encapsulation of the implant. Fusion of macrophages to form foreign body giant cells (FBGC), a hallmark of the FBR, is the consequence of a multistep mechanism induced by interleukin (IL)-4 that includes the acquisition of a fusion competent state and subsequent cytoskeletal rearrangements. However, the precise mechanism, regulation, and interplay among molecular mediators to generate FBGCs are insufficiently understood. Seeking novel mediators of fusion that might be regulated at the post-transcriptional level, we examined the role of microRNAs (miRs) in this process. A miR microarray was screened and identified miR-223 as a negative regulator of macrophage fusion. In addition, transfection of primary macrophages with a mir-223 mimic attenuated IL-4-induced fusion. Furthermore, miR-223 KO mice and mir-223 deficient cells displayed increased fusion in vivo and in vitro, respectively. Finally, we developed a method for in vivo delivery of miR-223 mimic utilizing PLGA nanoparticles, which inhibited FBGC formation in a biomaterial implant model. Our results identify miR-223 as a negative regulator of fusion and demonstrate miR-223 mimic-loaded nanoparticles as a therapeutic inhibitor of macrophage fusion.


Asunto(s)
Células Gigantes de Cuerpo Extraño/metabolismo , Macrófagos/metabolismo , MicroARNs/genética , Animales , Fusión Celular , Células Cultivadas , Regulación de la Expresión Génica , Células Gigantes de Cuerpo Extraño/citología , Ácido Láctico/química , Macrófagos/citología , Ratones , Ratones Noqueados , MicroARNs/administración & dosificación , Nanopartículas/química , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico
15.
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
16.
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
17.
Acta Biomater ; 11: 37-47, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25242651

RESUMEN

Implantation of biomaterials elicits a foreign body response characterized by fusion of macrophages to form foreign body giant cells and fibrotic encapsulation. Studies of the macrophage polarization involved in this response have suggested that alternative (M2) activation is associated with more favorable outcomes. Here we investigated this process in vivo by implanting mixed cellulose ester filters or polydimethylsiloxane disks in the peritoneal cavity of wild-type (WT) and monocyte chemoattractant protein-1 (MCP-1) knockout mice. We analyzed classical (M1) and alternative (M2) gene expression via quantitative polymerase chain reaction, immunohistochemistry and enzyme-linked immunosorbent assay in both non-adherent cells isolated by lavage and implant-adherent cells. Our results show that macrophages undergo unique activation that displays features of both M1 and M2 polarization including induction of tumor necrosis factor α (TNF), which induces the expression and nuclear translocation of p50 and RelA determined by immunofluorescence and Western blot. Both processes were compromised in fusion-deficient MCP-1 KO macrophages in vitro and in vivo. Furthermore, inclusion of BAY 11-7028, an inhibitor of NFκB activation, reduced nuclear translocation of RelA and fusion in WT macrophages. Our studies suggest that peritoneal implants elicit a unique macrophage polarization phenotype leading to induction of TNF and activation of the NFκB pathway.


Asunto(s)
Núcleo Celular/metabolismo , Celulosa/análogos & derivados , Quimiocina CCL2/metabolismo , Dimetilpolisiloxanos/toxicidad , Reacción a Cuerpo Extraño/metabolismo , Macrófagos/metabolismo , Subunidad p50 de NF-kappa B/metabolismo , Factor de Transcripción ReIA/metabolismo , Transporte Activo de Núcleo Celular/efectos de los fármacos , Transporte Activo de Núcleo Celular/genética , Animales , Núcleo Celular/genética , Núcleo Celular/patología , Celulosa/toxicidad , Quimiocina CCL2/genética , Reacción a Cuerpo Extraño/inducido químicamente , Reacción a Cuerpo Extraño/genética , Reacción a Cuerpo Extraño/patología , Regulación de la Expresión Génica/efectos de los fármacos , Macrófagos/patología , Ratones , Ratones Noqueados , Subunidad p50 de NF-kappa B/genética , Nitrilos/farmacología , Sulfonas/farmacología , Factor de Transcripción ReIA/genética
18.
Biomaterials ; 35(25): 6698-706, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24881026

RESUMEN

Intracranial implants elicit neurodegeneration via the foreign body response (FBR) that includes BBB leakage, macrophage/microglia accumulation, and reactive astrogliosis, in addition to neuronal degradation that limit their useful lifespan. Previously, monocyte chemoattractant protein 1 (MCP-1, also CCL2), which plays an important role in monocyte recruitment and propagation of inflammation, was shown to be critical for various aspects of the FBR in a tissue-specific manner. However, participation of MCP-1 in the brain FBR has not been evaluated. Here we examined the FBR to intracortical silicon implants in MCP-1 KO mice at 1, 2, and 8 weeks after implantation. MCP-1 KO mice had a diminished FBR compared to WT mice, characterized by reductions in BBB leakage, macrophage/microglia accumulation, and astrogliosis, and an increased neuronal density. Moreover, pharmacological inhibition of MCP-1 in implant-bearing WT mice maintained the increased neuronal density. To elucidate the relative contribution of microglia and macrophages, bone marrow chimeras were generated between MCP-1 KO and WT mice. Increased neuronal density was observed only in MCP-1 knockout mice transplanted with MCP-1 knockout marrow, which indicates that resident cells in the brain are major contributors. We hypothesized that these improvements are the result of a phenotypic switch of the macrophages/microglia polarization state, which we confirmed using PCR for common activation markers. Our observations suggest that MCP-1 influences neuronal loss, which is integral to the progression of neurological disorders like Alzheimer's and Parkinson disease, via BBB leakage and macrophage polarization.


Asunto(s)
Quimiocina CCL2/metabolismo , Reacción a Cuerpo Extraño/terapia , Enfermedades Neurodegenerativas/terapia , Neuronas/metabolismo , Animales , Benzoxazinas/farmacología , Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Supervivencia Celular/efectos de los fármacos , Enfermedad Crónica , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/metabolismo , Piperidinas/farmacología , Prótesis e Implantes , Receptores CCR2/antagonistas & inhibidores , Receptores CCR2/metabolismo , Ingeniería de Tejidos
19.
ACS Nano ; 8(5): 4366-75, 2014 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-24724817

RESUMEN

Nanopatterning of biomaterials is rapidly emerging as a tool to engineer cell function. Bulk metallic glasses (BMGs), a class of biocompatible materials, are uniquely suited to study nanopattern-cell interactions as they allow for versatile fabrication of nanopatterns through thermoplastic forming. Work presented here employs nanopatterned BMG substrates to explore detection of nanopattern feature sizes by various cell types, including cells that are associated with foreign body response, pathology, and tissue repair. Fibroblasts decreased in cell area as the nanopattern feature size increased, and fibroblasts could detect nanopatterns as small as 55 nm in size. Macrophages failed to detect nanopatterns of 150 nm or smaller in size, but responded to a feature size of 200 nm, resulting in larger and more elongated cell morphology. Endothelial cells responded to nanopatterns of 100 nm or larger in size by a significant decrease in cell size and elongation. On the basis of these observations, nondimensional analysis was employed to correlate cellular morphology and substrate nanotopography. Analysis of the molecular pathways that induce cytoskeletal remodeling, in conjunction with quantifying cell traction forces with nanoscale precision using a unique FIB-SEM technique, enabled the characterization of underlying biomechanical cues. Nanopatterns altered serum protein adsorption and effective substrate stiffness, leading to changes in focal adhesion density and compromised activation of Rho-A GTPase in fibroblasts. As a consequence, cells displayed restricted cell spreading and decreased collagen production. These observations suggest that topography on the nanoscale can be designed to engineer cellular responses to biomaterials.


Asunto(s)
Materiales Biocompatibles/química , Vidrio/química , Metales/química , Nanotecnología/métodos , Animales , Fenómenos Biomecánicos , Adhesión Celular , Supervivencia Celular , Colágeno/química , Citoesqueleto/metabolismo , Fibroblastos/metabolismo , Fibronectinas/química , Reacción a Cuerpo Extraño , GTP Fosfohidrolasas/química , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Microscopía Fluorescente , Células 3T3 NIH , Nanoestructuras/química , Ingeniería de Proteínas , Proteína de Unión al GTP rhoA/química
20.
Int J Pharm ; 453(2): 651-5, 2013 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-23727592

RESUMEN

Management of skin wound infections presents a serious problem in the clinic, in the community, and in both civilian and military clinical treatment centers. Staphylococcus aureus is one of the most common microbial pathogens in cutaneous wounds. Peptide-morpholino oligomer (PMO) conjugates targeted to S. aureus gyrase A mRNA have shown the ability to reduce bacterial viability by direct site-specific mRNA cleavage via RNase P. As a treatment, these conjugates have the added advantages of not being susceptible to resistance due to genetic mutations and are effective against drug resistant strains. While this strategy has proven effective in liquid culture, it has yet to be evaluated in an animal model of infected surface wounds. In the present study, we combined PMO conjugates with a thermoresponsive gel delivery system to treat full-thickness mouse cutaneous wounds infected with S. aureus. Wounds treated with a single dose of PMO conjugate displayed improved healing that was associated with increased epithelialization, reduced bacterial load, and increased matrix deposition. Taken together, our findings demonstrate the efficacy and flexibility of the PMO conjugate drug delivery system and make it an attractive and novel topical antimicrobial agent.


Asunto(s)
Antibacterianos/administración & dosificación , Girasa de ADN/genética , Morfolinos/administración & dosificación , Péptidos/administración & dosificación , Staphylococcus aureus , Infección de Heridas/terapia , Animales , Sistemas de Liberación de Medicamentos , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/genética , Piel/patología , Cicatrización de Heridas/efectos de los fármacos , Infección de Heridas/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA