Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Cardiovasc Diabetol ; 22(1): 214, 2023 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-37592236

RESUMEN

BACKGROUND: Cardiac steatosis is an early yet overlooked feature of diabetic cardiomyopathy. There is no available therapy to treat this condition. Tyrosine kinase inhibitors (TKIs) are used as first or second-line therapy in different types of cancer. In cancer patients with diabetes mellitus, TKIs reportedly improved glycemic control, allowing insulin discontinuation. They also reduced liver steatosis in a murine model of non-alcoholic fatty liver disease. The present study aimed to determine the therapeutic effect of the second-generation TKI Dasatinib on lipid accumulation and cardiac function in obese, type 2 diabetic mice. We also assessed if the drug impacts extra-cardiac fat tissue depots. METHODS: Two studies on 21-week-old male obese leptin receptor mutant BKS.Cg-+Leprdb/+Leprdb/OlaHsd (db/db) mice compared the effect of Dasatinib (5 mg/kg) and vehicle (10% DMSO + 90% PEG-300) given via gavage once every three days for a week or once every week for four weeks. Functional and volumetric indices were studied using echocardiography. Post-mortem analyses included the assessment of fat deposits and fibrosis using histology, and senescence using immunohistochemistry and flow cytometry. The anti-adipogenic action of Dasatinib was investigated on human bone marrow (BM)-derived mesenchymal stem cells (MSCs). Unpaired parametric or non-parametric tests were used to compare two and multiple groups as appropriate. RESULTS: Dasatinib reduced steatosis and fibrosis in the heart of diabetic mice. The drug also reduced BM adiposity but did not affect other fat depots. These structural changes were associated with improved diastolic indexes, specifically the E/A ratio and non-flow time. Moreover, Dasatinib-treated mice had lower levels of p16 in the heart compared with vehicle-treated controls, suggesting an inhibitory impact of the drug on the senescence signalling pathway. In vitro, Dasatinib inhibited human BM-MSC viability and adipogenesis commitment. CONCLUSIONS: Our findings suggest that Dasatinib opposes heart and BM adiposity and cardiac fibrosis. In the heart, this was associated with favourable functional consequences, namely improvement in an index of diastolic function. Repurposing TKI for cardiac benefit could address the unmet need of diabetic cardiac steatosis.


Asunto(s)
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 2 , Enfermedad del Hígado Graso no Alcohólico , Humanos , Masculino , Animales , Ratones , Dasatinib/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Fibrosis , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/tratamiento farmacológico
2.
ACS Synth Biol ; 12(6): 1845-1858, 2023 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-37224449

RESUMEN

Synthetic biology applications would benefit from protein modules of reduced complexity that function orthogonally to cellular components. As many subcellular processes depend on peptide-protein or protein-protein interactions, de novo designed polypeptides that can bring together other proteins controllably are particularly useful. Thanks to established sequence-to-structure relationships, helical bundles provide good starting points for such designs. Typically, however, such designs are tested in vitro and function in cells is not guaranteed. Here, we describe the design, characterization, and application of de novo helical hairpins that heterodimerize to form 4-helix bundles in cells. Starting from a rationally designed homodimer, we construct a library of helical hairpins and identify complementary pairs using bimolecular fluorescence complementation in E. coli. We characterize some of the pairs using biophysics and X-ray crystallography to confirm heterodimeric 4-helix bundles. Finally, we demonstrate the function of an exemplar pair in regulating transcription in both E. coli and mammalian cells.


Asunto(s)
Escherichia coli , Biología Sintética , Animales , Escherichia coli/genética , Péptidos/química , Proteínas/química , Mamíferos
3.
Am Surg ; 89(2): 267-276, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34010059

RESUMEN

BACKGROUND: In response to the COVID-19 pandemic, children's hospitals across the country postponed elective surgery beginning in March 2020. As projective curves flattened, administrators and surgeons sought to develop strategies to safely resume non-emergent surgery. This article reviews challenges and solutions specific to a children's hospital related to the resumption of elective pediatric surgeries. We present our tiered reentry approach for pediatric surgery as well as report early data for surgical volume and tracking COVID-19 cases during reentry. METHODS: The experience of shutdown, protocol development, and early reentry of elective pediatric surgery are reported from Levine's Children's Hospital (LCH), a free-leaning children's hospital in Charlotte, North Carolina. Data reported were obtained from de-identified hospital databases. RESULTS: Pediatric surgery experienced a dramatic decrease in case volumes at LCH during the shutdown, variable by specialty. A tiered and balanced reentry strategy was implemented with steady resumption of elective surgery following strict pre-procedural screening and testing. Early outcomes showed a steady thorough fluctuating increase in elective case volumes without evidence of a surgery-associated positive spread through periprocedural tracking. CONCLUSION: Reentry of non-emergent pediatric surgical care requires unique considerations including the impact of COVID-19 on children, each children hospital structure and resources, and preventing undue delay in intervention for age- and disease-specific pediatric conditions. A carefully balanced strategy has been critical for safe reentry following the anticipated surge. Ongoing tracking of resource utilization, operative volumes, and testing results will remain vital as community spread continues to fluctuate across the country.


Asunto(s)
COVID-19 , Cirujanos , Humanos , Niño , COVID-19/epidemiología , Pandemias/prevención & control , Procedimientos Quirúrgicos Electivos , Hospitales
4.
Oncogene ; 41(50): 5347-5360, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36344675

RESUMEN

ARID1a (BAF250), a component of human SWI/SNF chromatin remodeling complexes, is frequently mutated across numerous cancers, and its loss of function has been putatively linked to glucocorticoid resistance. Here, we interrogate the impact of siRNA knockdown of ARID1a compared to a functional interference approach in the HeLa human cervical cancer cell line. We report that ARID1a knockdown resulted in a significant global decrease in chromatin accessibility in ATAC-Seq analysis, as well as affecting a subset of genome-wide GR binding sites determined by analyzing GR ChIP-Seq data. Interestingly, the specific effects on gene expression were limited to a relatively small subset of glucocorticoid-regulated genes, notably those involved in cell cycle regulation and DNA repair. The vast majority of glucocorticoid-regulated genes were largely unaffected by ARID1a knockdown or functional interference, consistent with a more specific role for ARID1a in glucocorticoid function than previously speculated. Using liquid chromatography-mass spectrometry, we have identified a chromatin-associated protein complex comprising GR, ARID1a, and several DNA damage repair proteins including P53 binding protein 1 (P53BP1), Poly(ADP-Ribose) Polymerase 1 (PARP1), DNA damage-binding protein 1 (DDB1), DNA mismatch repair protein MSH6 and splicing factor proline and glutamine-rich protein (SFPQ), as well as the histone acetyltransferase KAT7, an epigenetic regulator of steroid-dependent transcription, DNA damage repair and cell cycle regulation. Not only was this protein complex ablated with both ARID1a knockdown and functional interference, but spontaneously arising DNA damage was also found to accumulate in a manner consistent with impaired DNA damage repair mechanisms. Recovery from dexamethasone-dependent cell cycle arrest was also significantly impaired. Taken together, our data demonstrate that although glucocorticoids can still promote cell cycle arrest in the absence of ARID1a, the purpose of this arrest to allow time for DNA damage repair is hindered.


Asunto(s)
Reparación del ADN , Proteínas Nucleares , Receptores de Glucocorticoides , Proteína 1 de Unión al Supresor Tumoral P53 , Humanos , Ciclo Celular , Puntos de Control del Ciclo Celular , Línea Celular Tumoral , Cromatina/genética , Daño del ADN , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Histona Acetiltransferasas/metabolismo , Proteínas Nucleares/metabolismo , Factores de Transcripción/genética , Receptores de Glucocorticoides/metabolismo , Proteína 1 de Unión al Supresor Tumoral P53/metabolismo
5.
Nat Hum Behav ; 6(7): 919-929, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35637294

RESUMEN

Citations and text analysis are both used to study the distribution and flow of ideas between researchers, fields and countries, but the resulting flows are rarely equal. We argue that the differences in these two flows capture a growing global inequality in the production of scientific knowledge. We offer a framework called 'citational lensing' to identify where citations should appear between countries but are absent given that what is embedded in their published abstract texts is highly similar. This framework also identifies where citations are overabundant given lower similarity. Our data come from nearly 20 million papers across nearly 35 years and 150 fields from the Microsoft Academic Graph. We find that scientific communities increasingly centre research from highly active countries while overlooking work from peripheral countries. This inequality is likely to pose substantial challenges to the growth of novel ideas.


Asunto(s)
Conocimiento , Investigadores , Humanos
6.
Methods Mol Biol ; 2475: 325-337, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35451769

RESUMEN

The transparent, genetically tractable zebrafish is increasingly recognized as a useful model to both live image and uncover mechanistic insight into cell interactions governing tissue homeostasis, pathology, and regeneration. Here, we describe a protocol for the isolation of macrophages from zebrafish wounds using fluorescence-activated cell sorting (FACS), and the identification of specific pro-angiogenic macrophage populations that express high levels of vascular endothelial growth factor (vegf) using quantitative real-time PCR (qPCR). The cell dissociation and FACS sorting techniques have been optimized for immune cells and successfully used to isolate other fluorescently marked populations within the wound such as neutrophils and endothelial cells. More broadly, this protocol can be easily adapted to other contexts where identification of pro-angiogenic immune cells is transformative for understanding, from development to pathologies such as infection, cancer, and diabetes.


Asunto(s)
Factor A de Crecimiento Endotelial Vascular , Pez Cebra , Animales , Células Endoteliales , Citometría de Flujo/métodos , Larva/genética , Macrófagos , Reacción en Cadena en Tiempo Real de la Polimerasa , Factor A de Crecimiento Endotelial Vascular/genética , Pez Cebra/genética
7.
Arterioscler Thromb Vasc Biol ; 41(9): 2454-2468, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34261327

RESUMEN

Objective: Extracellular vesicles (EVs) facilitate molecular transport across extracellular space, allowing local and systemic signaling during homeostasis and in disease. Extensive studies have described functional roles for EV populations, including during cardiovascular disease, but the in vivo characterization of endogenously produced EVs is still in its infancy. Because of their genetic tractability and live imaging amenability, zebrafish represent an ideal but under-used model to investigate endogenous EVs. We aimed to establish a transgenic zebrafish model to allow the in vivo identification, tracking, and extraction of endogenous EVs produced by different cell types. Approach and Results: Using a membrane-tethered fluorophore reporter system, we show that EVs can be fluorescently labeled in larval and adult zebrafish and demonstrate that multiple cell types including endothelial cells and cardiomyocytes actively produce EVs in vivo. Cell-type specific EVs can be tracked by high spatiotemporal resolution light-sheet live imaging and modified flow cytometry methods allow these EVs to be further evaluated. Additionally, cryo electron microscopy reveals the full morphological diversity of larval and adult EVs. Importantly, we demonstrate the utility of this model by showing that different cell types exchange EVs in the adult heart and that ischemic injury models dynamically alter EV production. Conclusions: We describe a powerful in vivo zebrafish model for the investigation of endogenous EVs in all aspects of cardiovascular biology and pathology. A cell membrane fluorophore labeling approach allows cell-type specific tracing of EV origin without bias toward the expression of individual protein markers and will allow detailed future examination of their function.


Asunto(s)
Sistema Cardiovascular/metabolismo , Vesículas Extracelulares/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/metabolismo , Animales , Animales Modificados Genéticamente , Sistema Cardiovascular/embriología , Separación Celular , Microscopía por Crioelectrón , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Células Endoteliales/ultraestructura , Vesículas Extracelulares/genética , Vesículas Extracelulares/ultraestructura , Citometría de Flujo , Regulación del Desarrollo de la Expresión Génica , Larva/metabolismo , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Infarto del Miocardio/genética , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/ultraestructura , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Factores de Tiempo , Pez Cebra/embriología , Pez Cebra/genética , Proteínas de Pez Cebra/genética
8.
AIDS ; 33(12): 1819-1830, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31274540

RESUMEN

OBJECTIVES: IL-27 is known as an antiviral cytokine that inhibits HIV, hepatitis C virus, and other viruses. We have previously demonstrated that, IL-27 posttreatment after HIV-infection inhibits viral replication in primary CD4 T cells. DESIGN: Here, we evaluated the anti-HIV effect of IL-27 pretreatment in CD4 T cells from healthy donors prior to HIV infection with HIVNL4.3 or vesicular stomatitis virus G glycoprotein (VSV-G)-pseudotyped HIV-luciferase virus (HIV-LUC-V). METHODS: IL-27-treated CD4 T cells were infected with HIVNL4.3 or HIV-LUC-V and assessed the anti-HIV effect. HIV infection was monitored by p24 antigen ELISA or luciferase assay. HIV fusion/entry and uncoating were determined by BlaM-Vpr assay and HIV fate of capsid and/or HIV Entry/Uncoating assay based on core-packaged RNA availability and Translation assay, respectively. HIV proviral copy number was determined by real-time PCR. Gene expression profile from IL-27-pretreated CD4 T cells was determined using Genechip array. Posttranslational modification of global proteins from IL-27-pretreated CD4 T cells was determined by a combination of 2-dimensional difference-in-gel-electrophoresis (2D-DIG), western-blot and protein mass spectrometry. RESULTS: IL-27 pretreatment inhibited HIVNL4.3 and HIV-LUC-V infection in CD4 T cells. HIV copy assay demonstrated that IL-27-treatment suppressed an early step of reverse transcription during HIV infection. A combination of 2D-DIG-electrophoresis and western blot assays demonstrated that IL-27-treatment induces a change in posttranslational modification of Y box binding protein-1 (YB-1). Overexpression of domain negative YB-1 mutants illustrated that a residue Lysine at 118 plays a key role in supporting HIV infection in CD4 T cells. CONCLUSION: IL-27-pretreatment inhibits HIV-1 infection by suppressing an HIV-reverse transcription product formation/uncoating step by suppressing the acetylation of YB-1 in primary CD4 T cells.


Asunto(s)
Linfocitos T CD4-Positivos/virología , VIH-1/crecimiento & desarrollo , VIH-1/inmunología , Factores Inmunológicos/metabolismo , Interleucinas/metabolismo , Replicación Viral , Proteína 1 de Unión a la Caja Y/metabolismo , Células Cultivadas , Genes Reporteros , Humanos , Cultivo de Virus
9.
Nanoscale Adv ; 1(8): 2840-2846, 2019 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-36133617

RESUMEN

The development of effective theranostic probes in cancer therapy is hampered due to issues with selectivity and off-target toxicity. We report the selective LED-photothermal ablation of cervical (HeLa) cancer cells over human dermal fibroblasts (HDF) using a new class of green-emissive fluorescent carbon dots (FCDs). The FCDs can be easily prepared in one pot using cheap and commercial starting materials. Physico-chemical characterization revealed that a surface coating of 2,5-deoxyfructosazine on a robust amorphous core gives rise to the nanomaterial's unique properties. We show that intracellular uptake mostly involves passive mechanisms in combination with intracellular DNA interactions to target the nucleus and that cancer cell selective killing is likely due to an increase in intracellular temperature in combination with ATP depletion, which is not observed upon exposure to either the "naked" core FCDs or the surface components individually. The selectivity of these nanoprobes and the lack of apparent production of toxic metabolic byproducts make these new nanomaterials promising agents in cancer therapy.

10.
J Pathol ; 247(4): 422-434, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30565681

RESUMEN

The overall survival for patients with primary glioblastoma is very poor. Glioblastoma contains a subpopulation of glioma stem cells (GSC) that are responsible for tumour initiation, treatment resistance and recurrence. PPARα is a transcription factor involved in the control of lipid, carbohydrate and amino acid metabolism. We have recently shown that PPARα gene and protein expression is increased in glioblastoma and has independent clinical prognostic significance in multivariate analyses. In this work, we report that PPARα is overexpressed in GSC compared to foetal neural stem cells. To investigate the role of PPARα in GSC, we knocked down its expression using lentiviral transduction with short hairpin RNA (shRNA). Transduced GSC were tagged with luciferase and stereotactically xenografted into the striatum of NOD-SCID mice. Bioluminescent and magnetic resonance imaging showed that knockdown (KD) of PPARα reduced the tumourigenicity of GSC in vivo. PPARα-expressing control GSC xenografts formed invasive histological phenocopies of human glioblastoma, whereas PPARα KD GSC xenografts failed to establish viable intracranial tumours. PPARα KD GSC showed significantly reduced proliferative capacity and clonogenic potential in vitro with an increase in cellular senescence. In addition, PPARα KD resulted in significant downregulation of the stem cell factors c-Myc, nestin and SOX2. This was accompanied by downregulation of the PPARα-target genes and key regulators of fatty acid oxygenation ACOX1 and CPT1A, with no compensatory increase in glycolytic flux. These data establish the aberrant overexpression of PPARα in GSC and demonstrate that this expression functions as an important regulator of tumourigenesis, linking self-renewal and the malignant phenotype in this aggressive cancer stem cell subpopulation. We conclude that targeting GSC PPARα expression may be a therapeutically beneficial strategy with translational potential as an adjuvant treatment. © 2018 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Asunto(s)
Neoplasias Encefálicas/patología , Glioblastoma/patología , PPAR alfa/metabolismo , ARN Interferente Pequeño/farmacología , Animales , Biomarcadores de Tumor/metabolismo , Transformación Celular Neoplásica , Regulación hacia Abajo , Femenino , Regulación Neoplásica de la Expresión Génica/fisiología , Técnicas de Silenciamiento del Gen/métodos , Humanos , Lentivirus , Ratones Endogámicos NOD , Ratones SCID , Células Madre Neoplásicas/patología , Fenotipo , Transducción de Señal/fisiología , Trasplante Heterólogo , Células Tumorales Cultivadas
12.
Sci Rep ; 8(1): 2534, 2018 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-29416058

RESUMEN

CD8+ T-cell expansions are the primary manifestation of T-cell large granular lymphocytic leukemia (T-LGLL), which is frequently accompanied by neutropenia and rheumatoid arthritis, and also occur as a secondary phenomenon in leukemia patients treated with dasatinib, notably in association with various drug-induced side-effects. However, the mechanisms that underlie the genesis and maintenance of expanded CD8+ T-cell receptor (TCR)-Vß+ populations in these patient groups have yet to be fully defined. In this study, we performed a comprehensive phenotypic and clonotypic assessment of expanded (TCR-Vß+) and residual (TCR-Vß-) CD8+ T-cell populations in T-LGLL and dasatinib-treated chronic myelogenous leukemia (CML) patients. The dominant CD8+ TCR-Vß+ expansions in T-LGLL patients were largely monoclonal and highly differentiated, whereas the dominant CD8+ TCR-Vß+ expansions in dasatinib-treated CML patients were oligoclonal or polyclonal, and displayed a broad range of memory phenotypes. These contrasting features suggest divergent roles for antigenic drive in the immunopathogenesis of primary versus dasatinib-associated CD8+ TCR-Vß+ expansions.


Asunto(s)
Antineoplásicos/efectos adversos , Linfocitos T CD8-positivos/inmunología , Dasatinib/efectos adversos , Leucemia Linfocítica Granular Grande/tratamiento farmacológico , Leucemia Linfocítica Granular Grande/inmunología , Leucemia Mieloide de Fase Crónica/tratamiento farmacológico , Leucemia Mieloide de Fase Crónica/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Adulto , Anciano , Antineoplásicos/uso terapéutico , Linfocitos T CD8-positivos/citología , Células Clonales , Dasatinib/uso terapéutico , Femenino , Humanos , Masculino , Persona de Mediana Edad , Fenotipo
13.
Sci Rep ; 8(1): 900, 2018 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-29343703

RESUMEN

We have previously demonstrated that Interleukin-27 differentially regulates the expression of seven novel microRNAs. Here we elucidate the functional significance of these novel microRNAs. Of the seven microRNAs, over expression of miRNA-6852 (miR-SX4) mimic induces cell cycle arrest at G2/M phase and induces necrosis in HEK293 and panel of cervical cancer cells (Human Papilloma Virus (HPV) infected cell lines; HeLa, CaSki and SiHa cells). To define the mechanism of the miR-SX4-mediated G2/M arrest, a microarray gene chip array and western blot analysis were performed. FoxM1, a transcription factor is identified as a key protein down-regulated by miR-SX4, even though the miR-SX4 does not target 3'UTR of FoxM1. Knock down of FoxM1 using si-RNA demonstrate that FoxM1 silenced cell induces G2/M cell cycle arrest and necrosis. Our data demonstrated for the first time that miR-SX4 could be a potent anti-cancer microRNA.


Asunto(s)
Regulación hacia Abajo/genética , Proteína Forkhead Box M1/genética , Interleucinas/genética , MicroARNs/genética , Necrosis/genética , Neoplasias del Cuello Uterino/genética , Puntos de Control del Ciclo Celular/genética , Línea Celular , Línea Celular Tumoral , Proliferación Celular/genética , Femenino , Puntos de Control de la Fase G2 del Ciclo Celular/genética , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica/genética , Células HEK293 , Células HeLa , Humanos
14.
J Am Heart Assoc ; 7(2)2018 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-29358198

RESUMEN

BACKGROUND: Transplantation of adventitial pericytes (APCs) promotes cardiac repair in murine models of myocardial infarction. The aim of present study was to confirm the benefit of APC therapy in a large animal model. METHODS AND RESULTS: We performed a blind, randomized, placebo-controlled APC therapy trial in a swine model of reperfused myocardial infarction. A first study used human APCs (hAPCs) from patients undergoing coronary artery bypass graft surgery. A second study used allogeneic swine APCs (sAPCs). Primary end points were (1) ejection fraction as assessed by cardiac magnetic resonance imaging and (2) myocardial vascularization and fibrosis as determined by immunohistochemistry. Transplantation of hAPCs reduced fibrosis but failed to improve the other efficacy end points. Incompatibility of the xenogeneic model was suggested by the occurrence of a cytotoxic response following in vitro challenge of hAPCs with swine spleen lymphocytes and the failure to retrieve hAPCs in transplanted hearts. We next considered sAPCs as an alternative. Flow cytometry, immunocytochemistry, and functional/cytotoxic assays indicate that sAPCs are a surrogate of hAPCs. Transplantation of allogeneic sAPCs benefited capillary density and fibrosis but did not improve cardiac magnetic resonance imaging indices of contractility. Transplanted cells were detected in the border zone. CONCLUSIONS: Immunologic barriers limit the applicability of a xenogeneic swine model to assess hAPC efficacy. On the other hand, we newly show that transplantation of allogeneic sAPCs is feasible, safe, and immunologically acceptable. The approach induces proangiogenic and antifibrotic benefits, though these effects were not enough to result in functional improvements.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Infarto del Miocardio/cirugía , Daño por Reperfusión Miocárdica/cirugía , Miocardio/patología , Neovascularización Fisiológica , Pericitos/trasplante , Función Ventricular Izquierda , Remodelación Ventricular , Anciano , Anciano de 80 o más Años , Células Alogénicas , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Fibrosis , Xenoinjertos , Humanos , Masculino , Persona de Mediana Edad , Contracción Miocárdica , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/fisiopatología , Recuperación de la Función , Volumen Sistólico , Sus scrofa , Trasplante Homólogo
15.
PLoS One ; 10(8): e0133231, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26237588

RESUMEN

Dynamic subcellular distributions of signaling system components are critical regulators of cellular signal transduction through their control of molecular interactions. Understanding how signaling activity depends on such distributions and the cellular structures driving them is required for comprehensive insight into signal transduction. In the activation of primary murine T cells by antigen presenting cells (APC) signaling intermediates associate with various subcellular structures, prominently a transient, wide, and actin-associated lamellum extending from an interdigitated T cell:APC interface several micrometers into the T cell. While actin dynamics are well established as general regulators of cellular organization, their role in controlling signaling organization in primary T cell:APC couples and the specific cellular structures driving it is unresolved. Using modest interference with actin dynamics with a low concentration of Jasplakinolide as corroborated by costimulation blockade we show that T cell actin preferentially controls lamellal signaling localization and activity leading downstream to calcium signaling. Lamellal localization repeatedly related to efficient T cell function. This suggests that the transient lamellal actin matrix regulates T cell signaling associations that facilitate T cell activation.


Asunto(s)
Actinas/metabolismo , Células Presentadoras de Antígenos/metabolismo , Activación de Linfocitos/inmunología , Transducción de Señal/inmunología , Linfocitos T/metabolismo , Animales , Células Presentadoras de Antígenos/efectos de los fármacos , Células Presentadoras de Antígenos/inmunología , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/inmunología , Depsipéptidos/farmacología , Activación de Linfocitos/efectos de los fármacos , Ratones , Fosforilación/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología
16.
PLoS One ; 8(4): e60300, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23565219

RESUMEN

Enucleation is the step in erythroid terminal differentiation when the nucleus is expelled from developing erythroblasts creating reticulocytes and free nuclei surrounded by plasma membrane. We have studied protein sorting during human erythroblast enucleation using fluorescence activated cell sorting (FACS) to obtain pure populations of reticulocytes and nuclei produced by in vitro culture. Nano LC mass spectrometry was first used to determine the protein distribution profile obtained from the purified reticulocyte and extruded nuclei populations. In general cytoskeletal proteins and erythroid membrane proteins were preferentially restricted to the reticulocyte alongside key endocytic machinery and cytosolic proteins. The bulk of nuclear and ER proteins were lost with the nucleus. In contrast to the localization reported in mice, several key erythroid membrane proteins were detected in the membrane surrounding extruded nuclei, including band 3 and GPC. This distribution of key erythroid membrane and cytoskeletal proteins was confirmed using western blotting. Protein partitioning during enucleation was investigated by confocal microscopy with partitioning of cytoskeletal and membrane proteins to the reticulocyte observed to occur at a late stage of this process when the nucleus is under greatest constriction and almost completely extruded. Importantly, band 3 and CD44 were shown not to restrict specifically to the reticulocyte plasma membrane. This highlights enucleation as a stage at which excess erythroid membrane proteins are discarded in human erythroblast differentiation. Given the striking restriction of cytoskeleton proteins and the fact that membrane proteins located in macromolecular membrane complexes (e.g. GPA, Rh and RhAG) are segregated to the reticulocyte, we propose that the membrane proteins lost with the nucleus represent an excess mobile population of either individual proteins or protein complexes.


Asunto(s)
Núcleo Celular/metabolismo , Eritroblastos/metabolismo , Diferenciación Celular , Proteínas del Citoesqueleto/metabolismo , Eritroblastos/citología , Humanos , Proteínas de la Membrana/metabolismo , Unión Proteica , Transporte de Proteínas , Proteoma , Proteómica/métodos
17.
Carcinogenesis ; 34(6): 1286-95, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23358854

RESUMEN

Keratinocyte senescence acts as a barrier to tumor progression but appears to be lost in late pre-malignancy to yield genetically unstable oral squamous cell carcinomas (GU-OSCC); a subset of OSCC possessing wild-type p53 and are genetically stable (GS-OSCC). In this study, fibroblasts from GU-OSCC were senescent relative to fibroblasts from GS-OSCC, epithelial dysplastic tissues or normal oral mucosa, as demonstrated by increased senescence-associated ß-galactosidase (SA ß-Gal) activity and overexpression of p16(INK4A). Keratinocytes from GU-OSCC produced high levels of reactive oxygen species (ROS) and this was associated with an increase in the production of transforming growth factor-ß1 (TGF-ß1) and TGF-ß2 in stromal fibroblasts. Treatment of normal fibroblasts with keratinocyte conditioned media (CM) from GU-OSCC, but not GS-OSCC or dysplastic keratinocytes with dysfunctional p53, induced fibroblast senescence. This phenomenon was inhibited by antioxidants and anti-TGF-ß antibodies. Fibroblast activation by TGF-ß1 preceded cellular senescence and was associated with increased ROS levels; antioxidants inhibited this reaction. Senescent fibroblasts derived from GU-OSCC or normal fibroblasts treated with CM from GU-OSCC or hydrogen peroxide, but not non-senescent fibroblasts derived from GS-OSCC, promoted invasion of keratinocytes in vitro. Epithelial invasion was stimulated by fibroblast activation and amplified further by fibroblast senescence. The data demonstrate that malignant keratinocytes from GU-OSCC, but not their pre-malignant counterparts, produce high levels of ROS, which, in turn, increase TGF-ß1 expression and induce fibroblast activation and senescence in a p5-independent manner. Fibroblasts from GU-OSCC were particularly susceptible to oxidative DNA damage because of high levels of ROS production, downregulation of antioxidant genes and upregulation of pro-oxidant genes. The results demonstrate the functional diversity of cancer-associated fibroblasts and show that malignant keratinocytes from GU-OSCC reinforce their malignant behavior by inducing fibroblast activation and senescence through ROS and TGF-ß-dependent mechanisms.


Asunto(s)
Senescencia Celular , Neoplasias de la Boca/patología , Estrés Oxidativo , Factor de Crecimiento Transformador beta1/metabolismo , Factor de Crecimiento Transformador beta2/metabolismo , Antioxidantes/farmacología , Carcinoma de Células Escamosas/genética , Línea Celular Tumoral , Medios de Cultivo Condicionados/farmacología , Inhibidor p16 de la Quinasa Dependiente de Ciclina/biosíntesis , Daño del ADN , Progresión de la Enfermedad , Fibroblastos/fisiología , Genotipo , Humanos , Peróxido de Hidrógeno/farmacología , Queratinocitos/metabolismo , Queratinocitos/fisiología , Mucosa Bucal/fisiología , Neoplasias de la Boca/genética , Oxidantes/farmacología , Especies Reactivas de Oxígeno/metabolismo , Factor de Crecimiento Transformador beta1/inmunología , Factor de Crecimiento Transformador beta2/inmunología , Proteína p53 Supresora de Tumor/genética , beta-Galactosidasa/metabolismo
18.
Hum Immunol ; 71(3): 225-34, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20034527

RESUMEN

Recent studies have expanded our understanding of the role of the anti-inflammatory cytokine interleukin (IL)-10, produced by multiple lineages of both human and murine T cells, in regulating the immune response. Here, we demonstrate that the small percentage of circulating CD4(+) T cells that secrete IL-10 can be isolated from human peripheral blood and, importantly, we have optimized a protocol to expand these cells in both antigen-specific and polyclonal manners. Expanded CD4(+)IL-10(+) T cells abrogate proliferation and T helper (Th) 1-like cytokine production in an antigen-specific manner, and to a lesser extent exhibit bystander suppressive capacity. CD4(+)IL-10(+) T cells are suppressive in a cell contact-dependent way, though they do not require secretion of IL-10 for their suppressive role in vitro. CD4(+)IL-10(+) T cells have an activated phenotype, with high expression of CD25, CD69, and cytotoxic T-lymphocyte antigen-4, and are largely FoxP3 negative. This novel method for the isolation and expansion of suppressive IL-10-secreting T cells has important implications both for further research and clinical therapeutic development.


Asunto(s)
Efecto Espectador/inmunología , Linfocitos T CD4-Positivos/metabolismo , Técnicas de Cultivo de Célula/métodos , Antígenos/inmunología , Antígenos CD/biosíntesis , Circulación Sanguínea/inmunología , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Antígeno CTLA-4 , Proliferación Celular , Separación Celular , Citometría de Flujo , Factores de Transcripción Forkhead/biosíntesis , Humanos , Inmunofenotipificación , Interleucina-10/metabolismo , Activación de Linfocitos
19.
Circ Res ; 103(11): 1335-43, 2008 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-18927465

RESUMEN

Reduced migratory function of circulating angiogenic progenitor cells (CPCs) has been associated with impaired neovascularization in patients with cardiovascular disease (CVD). Previous findings underline the role of the kallikrein-kinin system in angiogenesis. We now demonstrate the involvement of the kinin B2 receptor (B(2)R) in the recruitment of CPCs to sites of ischemia and in their proangiogenic action. In healthy subjects, B(2)R was abundantly present on CD133(+) and CD34(+) CPCs as well as cultured endothelial progenitor cells (EPCs) derived from blood mononuclear cells (MNCs), whereas kinin B1 receptor expression was barely detectable. In transwell migration assays, bradykinin (BK) exerts a potent chemoattractant activity on CD133(+) and CD34(+) CPCs and EPCs via a B(2)R/phosphoinositide 3-kinase/eNOS-mediated mechanism. Migration toward BK was able to attract an MNC subpopulation enriched in CPCs with in vitro proangiogenic activity, as assessed by Matrigel assay. CPCs from cardiovascular disease patients showed low B(2)R levels and decreased migratory capacity toward BK. When injected systemically into wild-type mice with unilateral limb ischemia, bone marrow MNCs from syngenic B(2)R-deficient mice resulted in reduced homing of sca-1(+) and cKit(+)flk1(+) progenitors to ischemic muscles, impaired reparative neovascularization, and delayed perfusion recovery as compared with wild-type MNCs. Similarly, blockade of the B(2)R by systemic administration of icatibant prevented the beneficial effect of bone marrow MNC transplantation. BK-induced migration represents a novel mechanism mediating homing of circulating angiogenic progenitors. Reduction of BK sensitivity in progenitor cells from cardiovascular disease patients might contribute to impaired neovascularization after ischemic complications.


Asunto(s)
Leucocitos Mononucleares/trasplante , Infarto del Miocardio/terapia , Isquemia Miocárdica/terapia , Revascularización Miocárdica/métodos , Neovascularización Fisiológica/fisiología , Receptor de Bradiquinina B2/fisiología , Trasplante de Células Madre/métodos , Agonistas Adrenérgicos beta/uso terapéutico , Angina de Pecho/fisiopatología , Animales , Bradiquinina/análogos & derivados , Bradiquinina/uso terapéutico , Movimiento Celular , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Citometría de Flujo , Humanos , Leucocitos Mononucleares/fisiología , Ratones , Ratones Noqueados , Infarto del Miocardio/fisiopatología , Receptor de Bradiquinina B2/deficiencia , Receptor de Bradiquinina B2/efectos de los fármacos , Receptor de Bradiquinina B2/genética , Células Madre/citología , Células Madre/fisiología
20.
Arterioscler Thromb Vasc Biol ; 28(1): 68-76, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17962628

RESUMEN

OBJECTIVE: We evaluated whether phosphatidylinositol 3-kinase gamma (PI3Kgamma) plays a role in reparative neovascularization and endothelial progenitor cell (EPC) function. METHODS AND RESULTS: Unilateral limb ischemia was induced in mice lacking the PI3Kgamma gene (PI3Kgamma-/-) or expressing a catalytically inactive mutant (PI3Kgamma(KD/KD)) and wild-type controls (WT). Capillarization and arteriogenesis were reduced in PI3Kgamma-/- ischemic muscles resulting in delayed reperfusion compared with WT, whereas reparative neovascularization was preserved in PI3Kgamma(KD/KD). In PI3Kgamma-/- muscles, endothelial cell proliferation was reduced, apoptosis was increased, and interstitial space was infiltrated with leukocytes but lacked cKit+ progenitor cells that in WT muscles typically surrounded arterioles. PI3Kgamma is constitutively expressed by WT EPCs, with expression levels being upregulated by hypoxia. PI3Kgamma-/- EPCs showed a defect in proliferation, survival, integration into endothelial networks, and migration toward SDF-1. The dysfunctional phenotype was associated with nuclear constraining of FOXO1, reduced Akt and eNOS phosphorylation, and decreased nitric oxide (NO) production. Pretreatment with an NO donor corrected the migratory defect of PI3Kgamma-/- EPCs. PI3Kgamma(KD/KD) EPCs showed reduced Akt phosphorylation, but constitutive activation of eNOS and preserved proliferation, survival, and migration. CONCLUSIONS: We newly demonstrated that PI3Kgamma modulates angiogenesis, arteriogenesis, and vasculogenesis by mechanisms independent from its kinase activity.


Asunto(s)
Células Endoteliales/fisiología , Isquemia/fisiopatología , Neovascularización Fisiológica/fisiología , Fosfatidilinositol 3-Quinasas/fisiología , Células Madre/fisiología , Animales , Fosfatidilinositol 3-Quinasa Clase Ib , Modelos Animales de Enfermedad , Extremidades/irrigación sanguínea , Isoenzimas/genética , Isoenzimas/fisiología , Masculino , Ratones , Ratones Noqueados , Músculo Liso/fisiología , Fosfatidilinositol 3-Quinasas/genética , Trasplantes
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...