Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
NPJ Vaccines ; 8(1): 175, 2023 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-37945621

RESUMEN

HIV affects more than 38 million people worldwide. Although HIV can be effectively treated by lifelong combination antiretroviral therapy, only a handful of patients have been cured. Therapeutic vaccines that induce robust de novo immune responses targeting HIV proteins and latent reservoirs will likely be integral for functional HIV cure. Our study shows that immunization of naïve rhesus macaques with arenavirus-derived vaccine vectors encoding simian immunodeficiency virus (SIVSME543 Gag, Env, and Pol) immunogens is safe, immunogenic, and efficacious. Immunization induced robust SIV-specific CD8+ and CD4+ T-cell responses with expanded cellular breadth, polyfunctionality, and Env-binding antibodies with antibody-dependent cellular cytotoxicity. Vaccinated animals had significant reductions in median SIV viral load (1.45-log10 copies/mL) after SIVMAC251 challenge compared with placebo. Peak viral control correlated with the breadth of Gag-specific T cells and tier 1 neutralizing antibodies. These results support clinical investigation of arenavirus-based vectors as a central component of therapeutic vaccination for HIV cure.

2.
Vaccine ; 39(38): 5410-5421, 2021 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-34391593

RESUMEN

Traditional bolus vaccine administration leads to rapid clearance of vaccine from lymphoid tissue. However, there is increasing evidence suggesting that the kinetics of antigen delivery can impact immune responses to vaccines, particularly when tailored to mimic natural infections. Here, we present the specific enhancements sustained release immunization confers to seasonal influenza vaccine, including the magnitude, durability, and breadth of humoral responses. To achieve sustained vaccine delivery kinetics, we have developed a microneedle array patch (MIMIX), with silk fibroin-formulated vaccine tips designed to embed in the dermis after a short application to the skin and release antigen over 1-2 weeks, mimicking the time course of a natural influenza infection. In a preclinical murine model, a single influenza vaccine administration via MIMIX led to faster seroconversion, response-equivalence to prime-boost bolus immunization, higher HAI titers against drifted influenza strains, and improved protective efficacy upon lethal influenza challenge when compared with intramuscular injection. These results highlight infection mimicry, achieved through sustained release silk microneedles, as a powerful approach to improve existing seasonal influenza vaccines, while also suggesting the broader potential of this platform technology to enable more efficacious next-generation vaccines and vaccine combinations.


Asunto(s)
Vacunas contra la Influenza , Gripe Humana , Animales , Humanos , Inmunogenicidad Vacunal , Gripe Humana/prevención & control , Ratones , Agujas , Seda
3.
Cell Rep ; 29(7): 1756-1766.e8, 2019 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-31722194

RESUMEN

The first immunization in a protein prime-boost vaccination is likely to be critical for how the immune response unfolds. Using fine needle aspirates (FNAs) of draining lymph nodes (LNs), we tracked the kinetics of the primary immune response in rhesus monkeys immunized intramuscularly (IM) or subcutaneously (s.c.) with an eOD-GT8 60-mer nanoparticle immunogen to facilitate clinical trial design. Significant numbers of germinal center B (BGC) cells and antigen-specific CD4 T cells were detectable in the draining LN as early as 7 days post-immunization and peaked near day 21. Strikingly, s.c. immunization results in 10-fold larger antigen-specific BGC cell responses compared to IM immunization. Lymphatic drainage studies revealed that s.c. immunization resulted in faster and more consistent axillary LN drainage than IM immunization. These data indicate robust antigen-specific germinal center responses can occur rapidly to a single immunization with a nanoparticle immunogen and vaccine drainage substantially impacts immune responses in local LNs.


Asunto(s)
Formación de Anticuerpos , Linfocitos B/inmunología , Linfocitos T CD4-Positivos/inmunología , Centro Germinal/inmunología , Inmunización , Nanopartículas , Vacunas/farmacología , Animales , Linfocitos B/patología , Biopsia con Aguja Fina , Linfocitos T CD4-Positivos/patología , Centro Germinal/patología , Humanos , Macaca mulatta , Masculino , Vacunas/inmunología
4.
Proc Natl Acad Sci U S A ; 116(33): 16473-16478, 2019 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-31358641

RESUMEN

Sustained exposure of lymphoid tissues to vaccine antigens promotes humoral immunity, but traditional bolus immunizations lead to rapid antigen clearance. We describe a technology to tailor vaccine kinetics in a needle-free platform translatable to human immunization. Solid pyramidal microneedle (MN) arrays were fabricated with silk fibroin protein tips encapsulating a stabilized HIV envelope trimer immunogen and adjuvant, supported on a dissolving polymer base. Upon brief skin application, vaccine-loaded silk tips are implanted in the epidermis/upper dermis where they release vaccine over a time period determined by the crystallinity of the silk matrix. Following MN immunization in mice, Env trimer was released over 2 wk in the skin, correlating with increased germinal center (GC) B cell responses, a ∼1,300-fold increase in serum IgG titers and a 16-fold increase in bone marrow (BM) plasma cells compared with bolus immunization. Thus, implantable MNs provide a practical means to substantially enhance humoral immunity to subunit vaccines.


Asunto(s)
Preparaciones de Acción Retardada/farmacología , Inmunidad Humoral , Agujas , Prótesis e Implantes , Vacunación , Animales , Formación de Anticuerpos/inmunología , Antígenos/inmunología , Bombyx , Centro Germinal/inmunología , Ganglios Linfáticos/inmunología , Ratones Endogámicos BALB C , Seda , Piel
5.
Sci Transl Med ; 10(467)2018 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-30429353

RESUMEN

Important cell populations reside within tissues and are not accessed by traditional blood draws used to monitor the immune system. To address this issue at an essential barrier tissue, the skin, we created a microneedle-based technology for longitudinal sampling of cells and interstitial fluid, enabling minimally invasive parallel monitoring of immune responses. Solid microneedle projections were coated by a cross-linked biocompatible polymer, which swells upon skin insertion, forming a porous matrix for local leukocyte infiltration. By embedding molecular adjuvants and specific antigens encapsulated in nanocapsules within the hydrogel coating, antigen-specific lymphocytes can be enriched in the recovered cell population, allowing for subsequent detailed phenotypic and functional analysis. We demonstrate this approach in mice immunized with a model protein antigen or infected in the skin with vaccinia virus. After vaccination or infection, sampling microneedles allowed tissue-resident memory T cells (TRMs) to be longitudinally monitored in the skin for many months, during which time the antigen-specific T cell population in systemic circulation contracted to low or undetectable counts. Sampling microneedles did not change the immune status of naïve or antigen-exposed animals. We also validated the ability of cell sampling using human skin samples. This approach may be useful in vaccines and immunotherapies to temporally query TRM populations or as a diagnostic platform to sample for biomarkers in chronic inflammatory and autoimmune disorders, allowing information previously accessible only via invasive biopsies to be obtained in a minimally invasive manner from the skin or other mucosal tissues.


Asunto(s)
Líquido Extracelular/metabolismo , Monitorización Inmunológica/métodos , Agujas , Piel/inmunología , Adyuvantes Inmunológicos/farmacología , Alginatos/química , Animales , Antígenos/metabolismo , Humanos , Inmunidad Humoral/efectos de los fármacos , Linfocitos/efectos de los fármacos , Linfocitos/inmunología , Ratones Endogámicos C57BL , Nanocápsulas
6.
Immunity ; 46(6): 1073-1088.e6, 2017 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-28636956

RESUMEN

The development of stabilized recombinant HIV envelope trimers that mimic the virion surface molecule has increased enthusiasm for a neutralizing antibody (nAb)-based HIV vaccine. However, there is limited experience with recombinant trimers as immunogens in nonhuman primates, which are typically used as a model for humans. Here, we tested multiple immunogens and immunization strategies head-to-head to determine their impact on the quantity, quality, and kinetics of autologous tier 2 nAb development. A bilateral, adjuvanted, subcutaneous immunization protocol induced reproducible tier 2 nAb responses after only two immunizations 8 weeks apart, and these were further enhanced by a third immunization with BG505 SOSIP trimer. We identified immunogens that minimized non-neutralizing V3 responses and demonstrated that continuous immunogen delivery could enhance nAb responses. nAb responses were strongly associated with germinal center reactions, as assessed by lymph node fine needle aspiration. This study provides a framework for preclinical and clinical vaccine studies targeting nAb elicitation.


Asunto(s)
Vacunas contra el SIDA/inmunología , Anticuerpos Neutralizantes/uso terapéutico , Centro Germinal/inmunología , Anticuerpos Anti-VIH/uso terapéutico , Infecciones por VIH/terapia , VIH-1/inmunología , Animales , Células Cultivadas , Epítopos de Linfocito B/química , Epítopos de Linfocito B/inmunología , Centro Germinal/virología , Infecciones por VIH/inmunología , Humanos , Inmunización , Inyecciones Subcutáneas , Primates , Multimerización de Proteína , Productos del Gen env del Virus de la Inmunodeficiencia Humana/química , Productos del Gen env del Virus de la Inmunodeficiencia Humana/inmunología
7.
Stem Cells Transl Med ; 5(7): 883-92, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27151913

RESUMEN

UNLABELLED: Children with congenital heart diseases have increased morbidity and mortality, despite various surgical treatments, therefore warranting better treatment strategies. Here we investigate the role of age of human pediatric cardiac progenitor cells (hCPCs) on ventricular remodeling in a model of juvenile heart failure. hCPCs isolated from children undergoing reconstructive surgeries were divided into 3 groups based on age: neonate (1 day to 1 month), infant (1 month to 1 year), and child (1 to 5 years). Adolescent athymic rats were subjected to sham or pulmonary artery banding surgery to generate a model of right ventricular (RV) heart failure. Two weeks after surgery, hCPCs were injected in RV musculature noninvasively. Analysis of cardiac function 4 weeks post-transplantation demonstrated significantly increased tricuspid annular plane systolic excursion and RV ejection fraction and significantly decreased wall thickness and fibrosis in rats transplanted with neonatal hCPCs compared with saline-injected rats. Computational modeling and systems biology analysis were performed on arrays and gave insights into potential mechanisms at the microRNA and gene level. Mechanisms including migration and proliferation assays, as suggested by computational modeling, showed improved chemotactic and proliferative capacity of neonatal hCPCs compared with infant/child hCPCs. In vivo immunostaining further suggested increased recruitment of stem cell antigen 1-positive cells in the right ventricle. This is the first study to assess the role of hCPC age in juvenile RV heart failure. Interestingly, the reparative potential of hCPCs is age-dependent, with neonatal hCPCs exerting the maximum beneficial effect compared with infant and child hCPCs. SIGNIFICANCE: Stem cell therapy for children with congenital heart defects is moving forward, with several completed and ongoing clinical trials. Although there are studies showing how children differ from adults, few focus on the differences among children. This study using human cardiac progenitor cells shows age-related changes in the reparative ability of cells in a model of pediatric heart failure and uses computational and systems biology to elucidate potential mechanisms.


Asunto(s)
Envejecimiento/fisiología , Cardiopatías Congénitas/terapia , Insuficiencia Cardíaca/terapia , Miocardio/citología , Trasplante de Células Madre , Células Madre/citología , Adulto , Animales , Proliferación Celular , Células Cultivadas , Preescolar , Cardiopatías Congénitas/patología , Insuficiencia Cardíaca/patología , Humanos , Lactante , Recién Nacido , Ratas , Ratas Desnudas , Ratas Transgénicas , Remodelación Ventricular
8.
Tissue Eng Part A ; 21(17-18): 2315-22, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25982380

RESUMEN

Myocardial infarction (MI) is the leading cause of death worldwide. Notch1 signaling plays a critical role in cardiac development, in survival, cardiogenic lineage commitment, differentiation of cardiac stem/progenitor cells, and in regenerative responses following myocardial injury. The objective of this study was the evaluation of the therapeutic effect of delivering the Notch ligand-containing hydrogels in a rat model of MI. Self-assembling peptide (SAP) hydrogels were functionalized with a peptide mimic of the Notch1 ligand Jagged1 (RJ). In rats subjected to experimental MI, delivery of RJ-containing hydrogel to the infarcted heart resulted in improvement in cardiac function back to sham-operated levels. A significant decrease in fibrosis and an increase in the endothelial vessel area and Ki67 expression were also observed in rats treated with the RJ hydrogels compared to untreated rats or rats treated with unmodified or scrambled peptide hydrogels. This study demonstrates the functional benefit of Notch1-activating peptide delivered in SAP hydrogels for cardiac repair.


Asunto(s)
Hidrogeles/farmacología , Infarto del Miocardio/fisiopatología , Infarto del Miocardio/terapia , Miocardio/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , Receptores Notch/metabolismo , Animales , Cardiomegalia/complicaciones , Cardiomegalia/patología , Cardiomegalia/fisiopatología , Proliferación Celular/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/patología , Endotelio Vascular/fisiopatología , Femenino , Fibrosis , Pruebas de Función Cardíaca/efectos de los fármacos , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/patología , Ventrículos Cardíacos/fisiopatología , Antígeno Ki-67/metabolismo , Ligandos , Infarto del Miocardio/complicaciones , Infarto del Miocardio/patología , Ratas Sprague-Dawley , Células Madre/citología , Células Madre/efectos de los fármacos
9.
J Cardiovasc Pharmacol Ther ; 20(1): 93-103, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24831254

RESUMEN

In the adult heart, catalase (CAT) activity increases appropriately with increasing levels of hydrogen peroxide, conferring cardioprotection. This mechanism is absent in the newborn for unknown reasons. In the present study, we examined how the posttranslational modification of CAT contributes to its activation during hypoxia/ischemia and the role of c-Abl tyrosine kinase in this process. Hypoxia studies were carried out using primary cardiomyocytes from adult (>8 weeks) and newborn rats. Following hypoxia, the ratio of phosphorylated to total CAT and c-Abl in isolated newborn rat myocytes did not increase and were significantly lower (1.3- and 4.2-fold, respectively; P < .05) than their adult counterparts. Similarly, there was a significant association (P < .0005) between c-Abl and CAT in adult cells following hypoxia (30.9 ± 8.2 to 70.7 ± 13.1 au) that was absent in newborn myocytes. Although ubiquitination of CAT was higher in newborns compared to adults following hypoxia, inhibition of this did not improve CAT activity. When a c-Abl activator (5-(1,3-diaryl-1H-pyrazol-4-yl)hydantoin [DPH], 200 µmol/L) was administered prior to hypoxia, not only CAT activity was significantly increased (P < .05) but also phosphorylation levels were also significantly improved (P < .01) in these newborn myocytes. Additionally, ischemia-reperfusion (IR) studies were performed using newborn (4-5 days) rabbit hearts perfused in a Langendorff method. The DPH given as an intracardiac injection into the right ventricle of newborn rabbit resulted in a significant improvement (P < .002) in the recovery of developed pressure after IR, a key indicator of cardiac function (from 74.6% ± 6.6% to 118.7% ± 10.9%). In addition, CAT activity was increased 3.92-fold (P < .02) in the same DPH-treated hearts. Addition of DPH to adult rabbits in contrast had no significant effect (from 71.3% ± 10.7% to 59.4% ± 12.1%). Therefore, in the newborn, decreased phosphorylation of CAT by c-Abl potentially mediates IR-induced dysfunction, and activation of c-Abl may be a strategy to prevent ischemic injury associated with surgical procedures.


Asunto(s)
Catalasa/metabolismo , Genes abl/fisiología , Miocitos Cardíacos/enzimología , Proteínas Tirosina Quinasas/fisiología , Animales , Animales Recién Nacidos , Hipoxia de la Célula/fisiología , Activación Enzimática/fisiología , Conejos , Ratas , Ratas Sprague-Dawley
10.
Biomaterials ; 35(28): 8103-12, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24974008

RESUMEN

Myocardial infarction is the leading cause of death worldwide and phase I clinical trials utilizing cardiac progenitor cells (CPCs) have shown promising outcomes. Notch1 signaling plays a critical role in cardiac development and in the survival, cardiogenic lineage commitment, and differentiation of cardiac stem/progenitor cells. In this study, we functionalized self-assembling peptide (SAP) hydrogels with a peptide mimic of the Notch1 ligand Jagged1 (RJ) to evaluate the therapeutic benefit of CPC delivery in the hydrogels in a rat model of myocardial infarction. The behavior of CPCs cultured in the 3D hydrogels in vitro including gene expression, proliferation, and growth factor production was evaluated. Interestingly, we observed Notch1 activation to be dependent on hydrogel polymer density/stiffness with synergistic increase in presence of RJ. Our results show that RJ mediated Notch1 activation depending on hydrogel concentration differentially regulated cardiogenic gene expression, proliferation, and growth factor production in CPCs in vitro. In rats subjected to experimental myocardial infarction, improvement in acute retention and cardiac function was observed following cell therapy in RJ hydrogels compared to unmodified or scrambled peptide containing hydrogels. This study demonstrates the potential therapeutic benefit of functionalizing SAP hydrogels with RJ for CPC based cardiac repair.


Asunto(s)
Materiales Biocompatibles/química , Hidrogeles/química , Infarto del Miocardio/metabolismo , Receptor Notch1/metabolismo , Células Madre/citología , Animales , Células CHO , Diferenciación Celular , Movimiento Celular , Colorantes/química , Cricetinae , Cricetulus , Femenino , Citometría de Flujo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Ratones , Miocardio/patología , Miocitos Cardíacos/citología , Péptidos/química , Polímeros/química , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley
11.
Methods Mol Biol ; 1141: 159-64, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24567138

RESUMEN

Drug and cell delivery systems could be modulated to serve as instructive microenvironments in regenerative medicine. Towards this end, several synthetic biomaterials have been developed to mimic the natural extracellular matrix (ECM) for therapeutic use. These include synthetic polymers, decellularized ECM, self-assembling polymers, and cell-responsive hydrogels with varied applications. Here, we describe the development of a self-assembling peptide hydrogel and its potential use as a cell and growth factor delivery vehicle to the infarcted heart in a rodent model of myocardial infarction.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Sistemas de Liberación de Medicamentos/métodos , Hidrogeles/química , Infarto del Miocardio/terapia , Péptidos/química , Secuencia de Aminoácidos , Animales , Materiales Biomiméticos/química , Línea Celular , Matriz Extracelular/química , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Péptidos y Proteínas de Señalización Intercelular/farmacología , Datos de Secuencia Molecular , Infarto del Miocardio/patología , Ratas , Ratas Sprague-Dawley
12.
Biomaterials ; 34(31): 7790-8, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23856052

RESUMEN

Myocardial infarction (MI) is the most common cause of heart failure (HF), the leading cause of death in the developed world. Oxidative stress due to excessive production of reactive oxygen species (ROS) plays a key role in the pathogenesis of cardiac remodeling leading to HF. NADPH oxidase with Nox2 as the catalytic subunit is a major source for cardiac ROS production. Nox2-NADPH expression is significantly increased in the infarcted myocardium, primarily in neutrophils, macrophages and myocytes. Moreover, mice lacking the Nox2 gene are protected from ischemic injury, implicating Nox2 as a potential therapeutic target. RNAi-mediated gene silencing holds great promise as a therapeutic owing to its high specificity and potency. However, in vivo delivery hurdles have limited its effective clinical use. Here, we demonstrate acid-degradable polyketal particles as delivery vehicles for Nox2-siRNA to the post-MI heart. In vitro, Nox2-siRNA particles are effectively taken up by macrophages and significantly knockdown Nox2 expression and activity. Following in vivo intramyocardial injection in experimental mice models of MI, Nox2-siRNA particles prevent upregulation of Nox2 and significantly recovered cardiac function. This study highlights the potential of polyketals as siRNA delivery vehicles to the MI heart and represents a viable therapeutic approach for targeting oxidative stress.


Asunto(s)
Glicoproteínas de Membrana/genética , Infarto del Miocardio/terapia , NADPH Oxidasas/genética , Nanopartículas/administración & dosificación , Nanopartículas/química , ARN Interferente Pequeño/genética , Animales , Línea Celular , Macrófagos/metabolismo , Masculino , Glicoproteínas de Membrana/antagonistas & inhibidores , Ratones , Ratones Endogámicos C57BL , NADPH Oxidasa 2 , NADPH Oxidasas/antagonistas & inhibidores , Nanopartículas/metabolismo , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/química
13.
Stem Cell Res Ther ; 4(2): 43, 2013 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-23597145

RESUMEN

INTRODUCTION: Administration of bone marrow-derived mesenchymal stem cells (MSCs) after myocardial infarction (MI) results in modest functional improvements. However; the effect of microenvironment changes after MI, such as elevated levels of oxidative stress on cardiogenic gene expression of MSCs, remains unclear. METHODS: MSCs were isolated from the bone marrow of adult rats and treated for 1 week with H2O2 (0.1 to 100 µM) or 48 hours with glucose oxidase (GOX; 0 to 5 mU/ml) to mimic long-term pulsed or short-term continuous levels of H2O2, respectively. RESULTS: In 100 µM H2O2 or 5 mU/ml GOX-treated MSCs, mRNA expression of selected endothelial genes (Flt1, vWF, PECAM1), and early cardiac marker (nkx2-5, αMHC) increased significantly, whereas early smooth muscle markers (smooth muscle α-actin and sm22α) and fibroblast marker vimentin decreased, as measured with real-time PCR. Interestingly, mRNA expression and activity of the cell-surface receptor Notch1 were significantly increased, as were its downstream targets, Hes5 and Hey1. Co-treatment of MSCs with 100 µM H2O2 and a γ-secretase inhibitor that prevents Notch signaling abrogated the increase in cardiac and endothelial genes, while augmenting the decrease in smooth muscle markers. Further, on GOX treatment, a significant increase in Wnt11, a downstream target of Notch1, was observed. Similar results were obtained with adult rat cardiac-derived progenitor cells. CONCLUSIONS: These data suggest that H2O2- or GOX-mediated oxidative stress upregulates Notch1 signaling, which promotes cardiogenic gene expression in adult stem/progenitor cells, possibly involving Wnt11. Modulating the balance between Notch activation and H2O2-mediated oxidative stress may lead to improved adult stem cell-based therapies for cardiac repair and regeneration.


Asunto(s)
Células Madre Mesenquimatosas/citología , Estrés Oxidativo , Receptor Notch1/metabolismo , Actinas/metabolismo , Animales , Células de la Médula Ósea/citología , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Dipéptidos/farmacología , Expresión Génica/efectos de los fármacos , Glucosa Oxidasa/farmacología , Peróxido de Hidrógeno/toxicidad , Masculino , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Miocardio/citología , Miocardio/metabolismo , Estrés Oxidativo/efectos de los fármacos , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Sprague-Dawley , Receptor Notch1/antagonistas & inhibidores , Receptor Notch1/genética , Transducción de Señal/efectos de los fármacos , Células Madre/citología , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Proteínas Wnt/metabolismo
14.
Stem Cells Dev ; 22(17): 2414-24, 2013 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-23544670

RESUMEN

There are a limited number of therapies available to prevent heart failure following myocardial infarction. One novel therapy that is currently being pursued is the implantation of cardiac progenitor cells (CPCs); however, their responses to oxidative stress during differentiation have yet to be elucidated. The objective of this study was to determine the effect of hydrogen peroxide (H2O2) treatment on CPC differentiation in vitro, as well as the effect of H2O2 preconditioning before implantation following ischemia-reperfusion (I/R) injury. CPCs were isolated and cloned from adult rat hearts, and then cultured in the absence or presence of H2O2 for 2 or 5 days. CPC survival was assessed with Annexin V, and cellular differentiation was evaluated through mRNA expression for cardiogenic genes. We found that 100 µM H2O2 decreased serum withdrawal-induced apoptosis by at least 45% following both 2 and 5 days of treatment. Moreover, 100 µM H2O2 treatment for 2 days significantly increased endothelial and smooth muscle markers compared to time-matched untreated CPCs. However, continued H2O2 treatment significantly decreased these markers. Left ventricular cardiac function was assessed 28 days after I/R and I/R with the implantation of Luciferase/GFP(+) CPCs, which were preconditioned with 100 µM H2O2 for 2 days. Hearts implanted with Luciferase/GFP(+) CPCs had significant improvement in both positive and negative dP/dT over I/R. Furthermore, cardiac fibrosis was significantly decreased in the preconditioned cells versus both I/R alone and I/R with control cells. We also observed a significant increase in endothelial cell density in the preconditioned CPC hearts compared to untreated CPC hearts, which also coincided with a higher density of Luciferase(+) vessels. These findings suggest that preconditioning of CPCs with H2O2 for 2 days stimulates neoangiogenesis in the peri-infarct area following I/R injury and could be a viable therapeutic option to prevent heart failure.


Asunto(s)
Insuficiencia Cardíaca/prevención & control , Peróxido de Hidrógeno/farmacología , Infarto del Miocardio/tratamiento farmacológico , Daño por Reperfusión/tratamiento farmacológico , Células Madre/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Fibrosis/tratamiento farmacológico , Expresión Génica , Insuficiencia Cardíaca/tratamiento farmacológico , Peróxido de Hidrógeno/metabolismo , Precondicionamiento Isquémico Miocárdico/métodos , Masculino , Contracción Miocárdica/efectos de los fármacos , Neovascularización Fisiológica/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Células Madre/metabolismo , Función Ventricular Izquierda/efectos de los fármacos
15.
PLoS One ; 7(11): e50980, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23226440

RESUMEN

Acute myocardial infarction (MI) caused by ischemia and reperfusion (IR) is the most common cause of cardiac dysfunction due to local cell death and a temporally regulated inflammatory response. Current therapeutics are limited by delivery vehicles that do not address spatial and temporal aspects of healing. The aim of this study was to engineer biotherapeutic delivery materials to harness endogenous cell repair to enhance myocardial repair and function. We have previously engineered poly(ethylene glycol) (PEG)-based hydrogels to present cell adhesive motifs and deliver VEGF to promote vascularization in vivo. In the current study, bioactive hydrogels with a protease-degradable crosslinker were loaded with hepatocyte and vascular endothelial growth factors (HGF and VEGF, respectively) and delivered to the infarcted myocardium of rats. Release of both growth factors was accelerated in the presence of collagenase due to hydrogel degradation. When delivered to the border zones following ischemia-reperfusion injury, there was no acute effect on cardiac function as measured by echocardiography. Over time there was a significant increase in angiogenesis, stem cell recruitment, and a decrease in fibrosis in the dual growth factor delivery group that was significant compared with single growth factor therapy. This led to an improvement in chronic function as measured by both invasive hemodynamics and echocardiography. These data demonstrate that dual growth factor release of HGF and VEGF from a bioactive hydrogel has the capacity to significantly improve cardiac remodeling and function following IR injury.


Asunto(s)
Materiales Biocompatibles/química , Sistemas de Liberación de Medicamentos/métodos , Corazón/fisiopatología , Factor de Crecimiento de Hepatocito/administración & dosificación , Hidrogel de Polietilenoglicol-Dimetacrilato/química , Péptido Hidrolasas/metabolismo , Factor A de Crecimiento Endotelial Vascular/administración & dosificación , Animales , Movimiento Celular/efectos de los fármacos , Separación Celular , Fibrosis , Corazón/efectos de los fármacos , Hemodinámica/efectos de los fármacos , Factor de Crecimiento de Hepatocito/farmacología , Neovascularización Fisiológica/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Daño por Reperfusión/diagnóstico por imagen , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/fisiopatología , Células Madre/citología , Células Madre/efectos de los fármacos , Ultrasonografía , Factor A de Crecimiento Endotelial Vascular/farmacología
16.
Acta Biomater ; 8(12): 4357-64, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22842035

RESUMEN

Myocardial infarction (MI) produces a collagen scar, altering the local microenvironment and impeding cardiac function. Cell therapy is a promising therapeutic option to replace the billions of myocytes lost following MI. Despite early successes, chronic function remains impaired and is likely a result of poor cellular retention, proliferation, and differentiation/maturation. While some efforts to deliver cells with scaffolds have attempted to address these shortcomings, they lack the natural cues required for optimal cell function. The goal of this study was to determine whether a naturally derived cardiac extracellular matrix (cECM) could enhance cardiac progenitor cell (CPC) function in vitro. CPCs were isolated via magnetic sorting of c-kit(+) cells and were grown on plates coated with either cECM or collagen I (Col). Our results show an increase in early cardiomyocyte markers on cECM compared with Col, as well as corresponding protein expression at a later time. CPCs show stronger serum-induced proliferation on cECM compared with Col, as well as increased resistance to apoptosis following serum starvation. Finally, a microfluidic adhesion assay demonstrated stronger adhesion of CPCs to cECM compared with Col. These data suggest that cECM may be optimal for CPC therapeutic delivery, as well as providing potential mechanisms to overcome the shortcomings of naked cell therapy.


Asunto(s)
Diferenciación Celular , Matriz Extracelular/química , Mioblastos Cardíacos/metabolismo , Miocardio/química , Animales , Antígenos de Diferenciación/biosíntesis , Apoptosis , Adhesión Celular , Células Cultivadas , Colágeno Tipo I/química , Masculino , Mioblastos Cardíacos/citología , Ratas , Ratas Sprague-Dawley
17.
Stem Cells Dev ; 21(17): 3136-46, 2012 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-22758933

RESUMEN

Transplantation of cardiac progenitor cells (CPCs) is currently in early clinical testing as a potential therapeutic strategy. Superoxide is increased in the ischemic myocardium and poor survival of cells is one of the major limitations of cell transplantation therapy. Superoxide dismutase (SOD) levels were analyzed in c-kit-positive CPCs isolated from rat myocardium to identify their roles in protection against oxidative stress-induced apoptosis in vitro. CPCs were subjected to oxidative stress using xanthine/xanthine oxidase (XXO) and little apoptosis was detected. CPCs contained significantly higher levels of SOD1 and SOD2 as compared with adult cardiac cell types, both at the protein and activity levels. Both SOD1 and SOD2 were increased by XXO at the mRNA and protein level, suggesting compensatory adaptation. Only knockdown of SOD2 and not SOD1 with siRNA sensitized the cells to XXO-apoptosis, despite only accounting for 10% of total SOD levels. Finally, we found XXO activated Akt within 10 min, and this regulated both SOD2 gene expression and protection against apoptosis. Rat CPCs are resistant to superoxide-induced cell death, primarily through higher levels of SOD2 compared to adult cardiac-derived cells. Exposure to superoxide increases expression of SOD2 in an Akt-dependent manner and regulates CPC survival during oxidative stress.


Asunto(s)
Regulación Enzimológica de la Expresión Génica , Miocardio/citología , Miocitos Cardíacos/enzimología , Células Madre/enzimología , Superóxido Dismutasa/metabolismo , Animales , Apoptosis , Supervivencia Celular , Activación Enzimática , Pruebas de Enzimas , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/enzimología , Técnicas de Silenciamiento del Gen , Miocardio/enzimología , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Estrés Oxidativo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Sprague-Dawley , Células Madre/citología , Células Madre/efectos de los fármacos , Superóxido Dismutasa/genética , Superóxido Dismutasa-1 , Factores de Tiempo , Transfección , Xantina/efectos adversos , Xantina Oxidasa/efectos adversos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA