Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
Más filtros

País/Región como asunto
País de afiliación
Intervalo de año de publicación
1.
J Neurooncol ; 140(2): 317-328, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30073642

RESUMEN

PURPOSE: Glioblastoma is the most common primary malignant brain tumor. No standard treatment exists for recurrent disease. Glioblastoma is associated with an immunosuppressive tumor microenvironment. Immune checkpoint inhibitors, including atezolizumab (anti-programmed death-ligand 1), have demonstrated clinical activity in various cancers. Here, we present the safety and efficacy of atezolizumab in patients with glioblastoma from the phase 1a PCD4989g clinical trial (NCT01375842). METHODS: Eligible patients had confirmed recurrent glioblastoma and measurable disease per RANO criteria. Atezolizumab (1200 mg) was administered intravenously every 3 weeks until progression or unacceptable toxicity. Patients were monitored for safety; response was evaluated at least every 6 weeks. Baseline biomarkers were evaluated. RESULTS: All 16 patients enrolled had received prior chemotherapy, and 50% prior bevacizumab. Ten patients (63%) experienced a treatment-related event. No treatment-related grade 4-5 events were reported. All deaths occurred due to progression or during follow-up. One patient experienced a partial response (5.3 months); 3 experienced disease stabilization. The median overall survival was 4.2 months (range 1.2 to 18.8+ months). Association between peripheral CD4+ T cells and efficacy was observed. Two patients with IDH1-mutant tumors and 1 with a POLE-mutant tumor experienced ≥ 16 months survival. CONCLUSIONS: Atezolizumab was safe and well tolerated in this group of patients with recurrent glioblastoma. Our preliminary findings suggest that biomarkers, including peripheral CD4+ T cells and hypermutated tumor status, may help guide selection of patients with recurrent glioblastoma who might receive most benefit from atezolizumab therapy, supporting further atezolizumab combination studies in glioblastoma.


Asunto(s)
Anticuerpos Monoclonales/efectos adversos , Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos/efectos adversos , Antineoplásicos/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Glioblastoma/tratamiento farmacológico , Administración Intravenosa , Adulto , Anciano , Anticuerpos Monoclonales Humanizados , Bevacizumab/uso terapéutico , Biomarcadores de Tumor/metabolismo , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/mortalidad , ADN Polimerasa II/genética , Femenino , Estudios de Seguimiento , Glioblastoma/genética , Glioblastoma/metabolismo , Glioblastoma/mortalidad , Humanos , Isocitrato Deshidrogenasa/genética , Lípidos , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/tratamiento farmacológico , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/metabolismo , Recurrencia Local de Neoplasia/mortalidad , Proteínas de Unión a Poli-ADP-Ribosa/genética , Resultado del Tratamiento
2.
Tob Control ; 24(1): 48-53, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23997071

RESUMEN

OBJECTIVES: Exposure of adults to secondhand smoke (SHS) has immediate adverse effects on the cardiovascular system and causes coronary heart disease. The current study evaluated brief self-report screening measures for accurately identifying adult cardiology patients with clinically significant levels of SHS exposure in need of intervention. DESIGN AND SETTING: A cross-sectional study conducted in a university-affiliated cardiology clinic and cardiology inpatient service. PATIENTS: Participants were 118 non-smoking patients (59% male, mean age=63.6 years, SD=16.8) seeking cardiology services. MAIN OUTCOME MEASURES: Serum cotinine levels and self-reported SHS exposure in the past 24 h and 7 days on 13 adult secondhand exposure to smoke (ASHES) items. RESULTS: A single item assessment of SHS exposure in one's own home in the past 7 days was significantly correlated with serum cotinine levels (r=0.41, p<0.001) with sensitivity ≥75%, specificity >85% and correct classification rates >85% at cotinine cut-off points of >0.215 and >0.80 ng/mL. The item outperformed multi-item scales, an assessment of home smoking rules, and SHS exposure assessed in other residential areas, automobiles and public settings. The sample was less accurate at self-reporting lower levels of SHS exposure (cotinine 0.05-0.215 ng/mL). CONCLUSIONS: The single item ASHES-7d Home screener is brief, assesses recent SHS exposure over a week's time, and yielded the optimal balance of sensitivity and specificity. The current findings support use of the ASHES-7d Home screener to detect SHS exposure and can be easily incorporated into assessment of other major vital signs in cardiology.


Asunto(s)
Cotinina/sangre , Exposición a Riesgos Ambientales/análisis , Autoinforme , Fumar , Contaminación por Humo de Tabaco/análisis , Adulto , Anciano , Anciano de 80 o más Años , Automóviles , Estudios Transversales , Femenino , Cardiopatías/terapia , Vivienda , Humanos , Masculino , Persona de Mediana Edad , Encuestas y Cuestionarios , Adulto Joven
3.
J Immunol ; 187(12): 6301-9, 2011 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-22084442

RESUMEN

CTLs and NK cells use the perforin/granzyme cytotoxic pathway to kill virally infected cells and tumors. Human regulatory T cells also express functional granzymes and perforin and can induce autologous target cell death in vitro. Perforin-deficient mice die of excessive immune responses after viral challenges, implicating a potential role for this pathway in immune regulation. To further investigate the role of granzyme B in immune regulation in response to viral infections, we characterized the immune response in wild-type, granzyme B-deficient, and perforin-deficient mice infected with Sendai virus. Interestingly, granzyme B-deficient mice, and to a lesser extent perforin-deficient mice, exhibited a significant increase in the number of Ag-specific CD8(+) T cells in the lungs and draining lymph nodes of virally infected animals. This increase was not the result of failure in viral clearance because viral titers in granzyme B-deficient mice were similar to wild-type mice and significantly less than perforin-deficient mice. Regulatory T cells from WT mice expressed high levels of granzyme B in response to infection, and depletion of regulatory T cells from these mice resulted in an increase in the number of Ag-specific CD8(+) T cells, similar to that observed in granzyme B-deficient mice. Furthermore, granzyme B-deficient regulatory T cells displayed defective suppression of CD8(+) T cell proliferation in vitro. Taken together, these results suggest a role for granzyme B in the regulatory T cell compartment in immune regulation to viral infections.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/virología , Granzimas/fisiología , Virus Sendai/inmunología , Carga Viral/inmunología , Animales , Linfocitos T CD8-positivos/patología , Epítopos de Linfocito T/genética , Epítopos de Linfocito T/inmunología , Granzimas/deficiencia , Granzimas/genética , Pulmón/inmunología , Pulmón/patología , Pulmón/virología , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/patología , Ganglios Linfáticos/virología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Perforina/deficiencia , Perforina/genética , Infecciones por Respirovirus/inmunología , Infecciones por Respirovirus/patología , Infecciones por Respirovirus/virología , Linfocitos T Reguladores/enzimología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/virología , Carga Viral/genética , Pérdida de Peso/genética , Pérdida de Peso/inmunología
4.
J Clin Immunol ; 32(1): 82-8, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22068910

RESUMEN

Severe combined immunodeficiency is a life-threatening primary immune deficiency characterized by low numbers of naïve T cells. Early diagnosis and treatment of this disease decreases mortality. In 2008, Wisconsin began newborn screening of infants for severe combined immunodeficiency and other forms of T-cell lymphopenia by the T-cell receptor excision circle assay. In total, 207,696 infants were screened. Seventy-two infants had an abnormal assay. T-cell numbers were normal in 38 infants, abnormal in 33 infants, and not performed in one infant, giving a positive predictive value for T-cell lymphopenia of any cause of 45.83% and a specificity of 99.98%. Five infants with severe combined immunodeficiency/severe T-cell lymphopenia requiring hematopoietic stem cell transplantation or other therapy were detected. In summary, the T-cell receptor excision circle assay is a sensitive and specific test to identify infants with severe combined immunodeficiency and severe T-cell lymphopenia that leads to life-saving therapies such as hematopoietic stem cell transplantation prior to the acquisition of severe infections.


Asunto(s)
Tamizaje Neonatal , Inmunodeficiencia Combinada Grave/diagnóstico , Humanos , Inmunofenotipificación , Recién Nacido , Recuento de Linfocitos , Linfopenia/diagnóstico , Linfopenia/inmunología , Tamizaje Neonatal/métodos , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Inmunodeficiencia Combinada Grave/epidemiología , Inmunodeficiencia Combinada Grave/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Wisconsin/epidemiología
5.
J Clin Immunol ; 31(3): 323-31, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21424824

RESUMEN

A multi-center, prospective, open-label study was conducted in primary immunodeficiency disease patients to determine the tolerability and pharmacokinetics of a 10% liquid IgG preparation administered subcutaneously. Forty-nine subjects (3-77 years old) were enrolled. Pharmacokinetic equivalence of subcutaneous treatment was achieved at a median dose of 137% of the intravenous dose, with a mean trough IgG level of 1,202 mg/dL at the end of the assessment period. The overall infection rate during subcutaneous treatment was 4.1 per subject-year. Three acute serious bacterial infections were reported, resulting in a rate of 0.067 per subject-year. A low overall rate of temporally associated adverse events (8%), and a very low rate of infusion site adverse events (2.8%), was seen at volumes up to 30 mL/site and rates ≤ 30 mL/h/site. Thus, subcutaneous replacement therapy with a 10% IgG preparation proved effective, safe and well-tolerated in our study population of subjects with primary immunodeficiency disease.


Asunto(s)
Agammaglobulinemia/tratamiento farmacológico , Infecciones Bacterianas/tratamiento farmacológico , Inmunodeficiencia Variable Común/tratamiento farmacológico , Enfermedades Genéticas Ligadas al Cromosoma X/tratamiento farmacológico , Inmunoglobulina G/administración & dosificación , Adolescente , Adulto , Agammaglobulinemia/complicaciones , Agammaglobulinemia/inmunología , Agammaglobulinemia/microbiología , Agammaglobulinemia/patología , Anciano , Bacterias/crecimiento & desarrollo , Infecciones Bacterianas/complicaciones , Infecciones Bacterianas/inmunología , Infecciones Bacterianas/microbiología , Infecciones Bacterianas/patología , Niño , Preescolar , Inmunodeficiencia Variable Común/complicaciones , Inmunodeficiencia Variable Común/inmunología , Inmunodeficiencia Variable Común/microbiología , Inmunodeficiencia Variable Común/patología , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Femenino , Enfermedades Genéticas Ligadas al Cromosoma X/complicaciones , Enfermedades Genéticas Ligadas al Cromosoma X/inmunología , Enfermedades Genéticas Ligadas al Cromosoma X/microbiología , Enfermedades Genéticas Ligadas al Cromosoma X/patología , Humanos , Inmunoglobulina G/efectos adversos , Inmunoglobulina G/inmunología , Inyecciones Intravenosas , Inyecciones Subcutáneas , Cinética , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Soluciones , Resultado del Tratamiento , Estados Unidos
6.
Arterioscler Thromb Vasc Biol ; 30(11): 2212-8, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20705916

RESUMEN

OBJECTIVE: Circulating angiogenic cells (CACs), also termed endothelial progenitor cells, play an integral role in vascular repair and are functionally impaired in coronary artery disease (CAD). The role of nitric oxide (NO) in CAC function is poorly understood. We hypothesized that CAC migration toward angiogenic signals is modulated by both NO synthase (NOS) expression and functional response to NO. METHODS AND RESULTS: Similar to endothelial cells, CAC chemotaxis to vascular endothelial growth factor (VEGF) was blocked by inhibition of NOS, phosphatidylinositol 3-kinase, or guanylyl cyclase or by treatment with an NO scavenger. Addition of an NO donor (S-nitroso-N-acetylpenicillamine) and the NOS substrate l-arginine increased random cell migration (chemokinesis) and enhanced VEGF-dependent chemotaxis. Healthy CACs expressed endothelial NOS, but endothelial NOS was not detected in CAD patient CACs. Both chemokinesis and chemotaxis to VEGF of patient CACs were decreased compared with healthy CACs but were restored to healthy values by S-nitroso-N-acetylpenicillamine. In parallel, CAD patients exhibited lower flow-mediated vasodilation and plasma NO source nitrite than young, healthy subjects, indicating endothelial dysfunction with reduced NO bioavailability. CONCLUSIONS: NOS activity is required for CAC chemotaxis. In CAD patients, impairment of NOS expression and NO bioavailability, rather than response to NO, may contribute to dysfunction of CACs and limit their regenerative capacity.


Asunto(s)
Quimiotaxis/fisiología , Células Endoteliales/fisiología , Óxido Nítrico Sintasa/biosíntesis , Óxido Nítrico/fisiología , Células Madre/fisiología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Movimiento Celular , Humanos , Neovascularización Fisiológica
7.
Physiol Genomics ; 42A(1): 52-60, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20551144

RESUMEN

22q11.2 Deletion syndrome (22q11.2 DS) [DiGeorge syndrome type 1 (DGS1)] occurs in ∼1:3,000 live births; 75% of children with DGS1 have severe congenital heart disease requiring early intervention. The gold standard for detection of DGS1 is fluorescence in situ hybridization (FISH) with a probe at the TUPLE1 gene. However, FISH is costly and is typically ordered in conjunction with a karyotype analysis that takes several days. Therefore, FISH is underutilized and the diagnosis of 22q11.2 DS is frequently delayed, often resulting in profound clinical consequences. Our goal was to determine whether multiplexed, quantitative real-time PCR (MQPCR) could be used to detect the haploinsufficiency characteristic of 22q11.2 DS. A retrospective blinded study was performed on 382 subjects who had undergone congenital heart surgery. MQPCR was performed with a probe localized to the TBX1 gene on human chromosome 22, a gene typically deleted in 22q11.2 DS. Cycle threshold (C(t)) was used to calculate the relative gene copy number (rGCN). Confirmation analysis was performed with the Affymetrix 6.0 Genome-Wide SNP Array. With MQPCR, 361 subjects were identified as nondeleted with an rGCN near 1.0 and 21 subjects were identified as deleted with an rGCN near 0.5, indicative of a hemizygous deletion. The sensitivity (21/21) and specificity (361/361) of MQPCR to detect 22q11.2 deletions was 100% at an rGCN value drawn at 0.7. One of 21 subjects with a prior clinical (not genetically confirmed) DGS1 diagnosis was found not to carry the deletion, while another subject, not previously identified as DGS1, was detected as deleted and subsequently confirmed via microarray. The MQPCR assay is a rapid, inexpensive, sensitive, and specific assay that can be used to screen for 22q11.2 deletion syndrome. The assay is readily adaptable to high throughput.


Asunto(s)
Cromosomas Humanos Par 22/genética , Cardiopatías Congénitas/genética , Reacción en Cadena de la Polimerasa/métodos , Variaciones en el Número de Copia de ADN/genética , Síndrome de DiGeorge/genética , Humanos , Polimorfismo de Nucleótido Simple/genética , Estudios Retrospectivos
8.
Clin Immunol ; 137(1): 21-30, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20675197

RESUMEN

Primary immunodeficiency disease (PIDD) associated with hypogammaglobulinemia is typically treated with immunoglobulin replacement therapy. When administered as intravenous immunoglobulin (IVIG), an IgG trough occurs prior to the next replacement dose. While frequently measured, IgG trough levels required to minimize infection risk are not established. To address this question, all available studies evaluating trough IgG and pneumonia incidence in PIDD patients with hypogammaglobulinemia receiving IVIG were quantitatively combined by meta-analysis. Seventeen studies with 676 total patients and 2,127 patient-years of follow-up were included. Pneumonia incidence declined by 27% with each 100mg/dL increment in trough IgG (incidence rate ratio, 0.726; 95% confidence interval, 0.658-0.801). Pneumonia incidence with maintenance of 500 mg/dL IgG trough levels (0.113 cases per patient-year) was 5-fold that with 1000 mg/dL (0.023 cases per patient-year). This meta-analysis provides evidence that pneumonia risk can be progressively reduced by higher trough IgG levels up to at least 1000 mg/dL.


Asunto(s)
Inmunoglobulina G/inmunología , Inmunoglobulinas Intravenosas/sangre , Inmunoglobulinas Intravenosas/uso terapéutico , Síndromes de Inmunodeficiencia/complicaciones , Síndromes de Inmunodeficiencia/terapia , Neumonía/complicaciones , Neumonía/inmunología , Adolescente , Adulto , Agammaglobulinemia/complicaciones , Agammaglobulinemia/inmunología , Agammaglobulinemia/prevención & control , Agammaglobulinemia/terapia , Anciano , Niño , Preescolar , Ensayos Clínicos como Asunto , Femenino , Humanos , Inmunoglobulina G/sangre , Inmunoglobulinas Intravenosas/administración & dosificación , Síndromes de Inmunodeficiencia/sangre , Síndromes de Inmunodeficiencia/inmunología , Lactante , Masculino , Persona de Mediana Edad , Neumonía/prevención & control , Resultado del Tratamiento , Adulto Joven
9.
Eur J Immunol ; 39(9): 2571-83, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19701891

RESUMEN

The transcription factor Foxp3 is essential for the development of functional, natural Treg (nTreg), which plays a prominent role in self-tolerance. Suppressive Foxp3(+) Treg cells can be generated from naïve T cells ex vivo, following TCR and TGF-beta1 stimulations. However, the molecular contributions from the different arms of these pathways leading to Foxp3 expression are not fully understood. TGF-beta1-activated Smad3 plays a major role in the expression of Foxp3, since TGF-beta1-induced-Treg generation from Smad3(-/-) mice is markedly reduced and abolished by inactivating Smad2. In the TCR pathway, deletion of Bcl10, which activates NF-kappaB, markedly reduces both IL-2 and Foxp3 production. However, partial rescue of Foxp3 expression occurs on addition of exogenous IL-2. TGF-beta1 significantly attenuates NF-kappaB binding to the Foxp3 promoter, while inducing Foxp3 expression. Furthermore, deletion of p50, a NF-kappaB subunit, results in increased Foxp3 expression despite a decline in the IL-2 production. We posit several TCR-NF-kappaB pathways, some increasing (Bcl10-IL-2-Foxp3) while others decreasing (p50-Foxp3) Foxp3 expression, with the former predominating. A better understanding of Foxp3 regulation could be useful in dissecting the cause of Treg dysfunction in several autoimmune diseases and for generating more potent TGF-beta1-induced-Treg cells for therapeutic purposes.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Factores de Transcripción Forkhead/biosíntesis , Subunidad p50 de NF-kappa B/metabolismo , Proteína smad3/metabolismo , Linfocitos T Reguladores/inmunología , Animales , Antineoplásicos/farmacología , Linfocitos T CD4-Positivos/efectos de los fármacos , Células Cultivadas , Factores de Transcripción Forkhead/genética , Interleucina-2/farmacología , Ratones , Ratones Endogámicos C57BL , Subunidad p50 de NF-kappa B/antagonistas & inhibidores , Péptidos/farmacología , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Proteína smad3/genética , Linfocitos T Reguladores/efectos de los fármacos , Factor de Crecimiento Transformador beta1/farmacología
10.
J Card Fail ; 16(8): 669-78, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20670846

RESUMEN

BACKGROUND: Erythropoietin (EPO) and granulocyte colony stimulating factor (GCSF) are potential novel therapies after myocardial infarction (MI). We first established the optimal and clinically applicable dosages of these drugs in mobilizing hematopoietic stem cells (HSC), and then tested the efficacy of monotherapy and combination therapy post-MI. METHODS AND RESULTS: Optimal doses were established in enhanced green fluorescent protein (eGFP) + chimeric mice (n = 30). Next, mice underwent MI and randomized into 4 groups (n = 18/group): 1) GCSF; 2) EPO; 3) EPO+GCSF; and 4) control. Left ventricular (LV) function was analyzed pre-MI, at 4 hours and at 28 days post-MI. Histological assessment of infarct size, blood vessels, apoptotic cardiomyocytes, and engraftment of eGFP+ mobilized cells were analyzed at day 28. LV function in the control group continued to deteriorate, whereas all treatments showed stabilization. The treatment groups resulted in less scarring, increased numbers of mobilized cells to the infarct border zone (BZ), and a reduction in the number of apoptotic cardiomyocytes. Both EPO groups had significantly more capillaries and arterioles at the BZ. CONCLUSION: We have established the optimal doses for EPO and GCSF in mobilizing HSC from the bone marrow and demonstrated that therapy with these agents, either as monotherapy or combination therapy, led to improvement of cardiac function post-MI. Combination therapy does not seem to have additive benefit over monotherapy in this model.


Asunto(s)
Modelos Animales de Enfermedad , Eritropoyetina/administración & dosificación , Factor Estimulante de Colonias de Granulocitos/administración & dosificación , Infarto del Miocardio/tratamiento farmacológico , Función Ventricular Izquierda/efectos de los fármacos , Función Ventricular Izquierda/fisiología , Animales , Citocinas/administración & dosificación , Preparaciones de Acción Retardada/administración & dosificación , Quimioterapia Combinada , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Infarto del Miocardio/fisiopatología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/fisiología , Quimera por Radiación , Distribución Aleatoria
11.
J Card Fail ; 16(7): 579-89, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20610234

RESUMEN

BACKGROUND: Erythropoietin (EPO) has generated interest as a novel therapy after myocardial infarction (MI), but the safety and efficacy of prolonged therapy have not been studied in a large animal model of reperfused MI. METHODS AND RESULTS: MI was induced in pigs by a 90-minute balloon occlusion of the left anterior descending coronary artery. Sixteen animals were randomized to either EPO or saline (control group). Inflammatory markers, bone marrow cell mobilization, and left ventricular function (by both echocardiography and pressure-volume measurements) were assessed at baseline, 1 and 6 weeks post-MI. EPO therapy was associated with a significant increase in hemoglobin and mononuclear counts. D-dimer and C-reactive protein levels did not differ between groups. At week 6, EPO therapy prevented further deterioration of left ventricular ejection fraction (39 +/- 2% vs. 33 +/- 1%, P < .01) and improved wall motion score index (P < .02). Histopathology revealed increased areas of viable myocardium, vascular density, and capillary-to-myocyte ratio in the EPO therapy compared with the control (all P < .05). CONCLUSION: Prolonged EPO therapy after MI in a large animal model is safe and leads to an increase in viable myocardium, increased vascular density, and prevents further deterioration of left ventricular function. These results support future clinical studies in post-MI patients.


Asunto(s)
Modelos Animales de Enfermedad , Eritropoyetina/administración & dosificación , Infarto del Miocardio/tratamiento farmacológico , Disfunción Ventricular Izquierda/prevención & control , Función Ventricular Izquierda/efectos de los fármacos , Animales , Infarto del Miocardio/fisiopatología , Distribución Aleatoria , Porcinos , Factores de Tiempo , Disfunción Ventricular Izquierda/fisiopatología , Función Ventricular Izquierda/fisiología
12.
Cytokine ; 51(3): 278-85, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20609597

RESUMEN

BACKGROUND: We investigated the safety and efficacy of GCSF therapy in a porcine model of ischemia-reperfusion with left ventricle ejection fraction of <45% using a clinically relevant dosing and timing regimen. METHODS: MI was induced in pigs by a 90 min balloon occlusion of the left anterior descending coronary artery. Sixteen animals were randomized to either GCSF (IV bolus of 10 microg/kg at time of reperfusion, followed by SC injections of 5 microg/kg days 5-9 post-MI) or saline (control group). Inflammatory markers, bone marrow cell mobilization and LV function (echocardiography and pressure-volume measurements) were assessed at baseline, 1 and 6 weeks post-MI. Histopathology was performed 6 weeks post-MI. RESULTS: GCSF therapy was associated with a significant increase in white blood cell counts. At week 6, GCSF therapy resulted in less deterioration of LVEF compared to control (38+/-2% vs. 33+/-2%, p<0.02) and improved wall motion score index (p<0.05). Histopathology revealed increased vascular density (p<0.05) and a trend toward increased areas of viable myocardium compared to control (p=0.058). CONCLUSION: GCSF therapy prevents further deterioration of LV function in a porcine model of MI with lower EF (<45%). These results support future clinical trials with GCSF in selected patients with larger MI.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos/uso terapéutico , Infarto del Miocardio/tratamiento farmacológico , Infarto del Miocardio/fisiopatología , Volumen Sistólico/fisiología , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Cateterismo , Movimiento Celular/efectos de los fármacos , Cicatriz/patología , Fibrosis , Factor Estimulante de Colonias de Granulocitos/farmacología , Infarto del Miocardio/diagnóstico por imagen , Infarto del Miocardio/patología , Neovascularización Fisiológica/efectos de los fármacos , Volumen Sistólico/efectos de los fármacos , Sus scrofa , Factores de Tiempo , Ultrasonografía
13.
Cardiovasc Drugs Ther ; 24(5-6): 409-20, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20809214

RESUMEN

PURPOSE: Erythropoietin (EPO) and granulocyte colony stimulating factor (GCSF) have generated interest as novel therapies after myocardial infarction (MI), but the effect of combination therapy has not been studied in the large animal model. We investigated the impact of prolonged combination therapy with EPO and GCSF on cardiac function, infarct size, and vascular density after MI in a porcine model. METHODS: MI was induced in pigs by a 90 min balloon occlusion of the left anterior descending coronary artery. 16 animals were treated with EPO+GCSF, or saline (control group). Cardiac function was assessed by echocardiography and pressure-volume measurements at baseline, 1 and 6 weeks post-MI. Histopathology was performed 6 weeks post-MI. RESULTS: At week 6, EPO+GCSF therapy stabilized left ventricular ejection fraction, (41 ± 1% vs. 33 ± 1%, p < 0.01) and improved diastolic function compared to the control group. Histopathology revealed increased areas of viable myocardium and vascular density in the EPO+GCSF therapy, compared to the control. Despite these encouraging results, in a historical analysis comparing combination therapy with monotherapy with EPO or GCSF, there were no significant additive benefits in the LVEF and volumes overtime using the combination therapy. CONCLUSION: Our findings indicate that EPO+GCSF combination therapy promotes stabilization of cardiac function after acute MI. However, combination therapy does not seem to be superior to monotherapy with either EPO or GCSF.


Asunto(s)
Eritropoyetina/administración & dosificación , Factor Estimulante de Colonias de Granulocitos/administración & dosificación , Infarto del Miocardio/tratamiento farmacológico , Animales , Células de la Médula Ósea/efectos de los fármacos , Modelos Animales de Enfermedad , Quimioterapia Combinada/métodos , Ecocardiografía/métodos , Corazón/efectos de los fármacos , Corazón/fisiopatología , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/fisiopatología , Infarto del Miocardio/diagnóstico por imagen , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Miocardio/patología , Volumen Sistólico/efectos de los fármacos , Porcinos
14.
Mol Ther ; 17(7): 1250-6, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19384293

RESUMEN

We compared therapeutic benefits of intramyocardial injection of unfractionated bone marrow cells (BMCs) versus BMC extract as treatments for myocardial infarction (MI), using closed-chest ultrasound-guided injection at a clinically relevant time post-MI. MI was induced in mice and the animals treated at day 3 with either: (i) BMCs from green fluorescent protein (GFP)-expressing mice (n = 14), (ii) BMC extract (n = 14), or (iii) saline control (n = 14). Six animals per group were used for histology at day 6 and the rest followed to day 28 for functional analysis. Ejection fraction was similarly improved in the BMC and extract groups versus control (40.6 +/- 3.4 and 39.1 +/- 2.9% versus 33.2 +/- 5.0%, P < 0.05) with smaller scar sizes. At day 6 but not day 28, both therapies led to significantly higher capillary area and number of arterioles versus control. At day 6, BMCs increased the number of cycling cardiomyocytes (CMs) versus control whereas extract therapy resulted in significant reduction in the number of apoptotic CMs at the border zone (BZ) versus control. Intracellular components within BMCs can enhance vascularity, reduce infarct size, improve cardiac function, and influence CM apoptosis and cycling early after therapy following MI. Intact cells are not necessary and death of implanted cells may be a major component of the benefit.


Asunto(s)
Células de la Médula Ósea/fisiología , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Corazón/fisiología , Infarto del Miocardio/terapia , Animales , Apoptosis , Células de la Médula Ósea/metabolismo , Ecocardiografía , Masculino , Ratones , Ratones Endogámicos C57BL , Miocitos Cardíacos/citología , Miocitos Cardíacos/fisiología
15.
Public Health Rep ; 125 Suppl 2: 88-95, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20518449

RESUMEN

Severe combined immunodeficiency (SCID) is the result of genetic defects that impair normal T-cell development. SCID babies typically appear normal at birth, but acquire multiple life-threatening infections within a few months. Early diagnosis and treatment with a bone-marrow transplant markedly improves long-term outcomes. On January 1, 2008, the newborn screening (NBS) program in Wisconsin became the first in the world to routinely test all newborns for SCID. A realtime quantitative polymerase chain reaction assay measures T-cell receptor excision circles (TRECs), which are formed during the maturation of normal T-cells. A lack or very low number of TRECs is consistent with T-cell lymphopenia. The development and validation of the TREC assay and the results of the first year of screening have been published. This article describes the process used to add SCID to the NBS panel, the establishment of follow-up capacity, and the integration of SCID screening into routine NBS workflows. The development of this expanded NBS program is described so that other states might benefit from the processes used in Wisconsin.


Asunto(s)
Genes Codificadores de los Receptores de Linfocitos T , Tamizaje Neonatal/métodos , Receptores de Antígenos de Linfocitos T/genética , Inmunodeficiencia Combinada Grave/diagnóstico , ADN/sangre , ADN/genética , Humanos , Recién Nacido , Reacción en Cadena de la Polimerasa , Receptores de Antígenos de Linfocitos T/inmunología , Inmunodeficiencia Combinada Grave/inmunología , Wisconsin
16.
J Allergy Clin Immunol ; 124(3): 522-7, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19482345

RESUMEN

BACKGROUND: Severe combined immunodeficiency (SCID) is characterized by the absence of functional T cells and B cells. Without early diagnosis and treatment, infants with SCID die from severe infections within the first year of life. OBJECTIVE: To determined the feasibility of detecting SCID in newborns by quantitating T-cell receptor excision circles (TRECs) from dried blood spots (DBSs) on newborn screening (NBS) cards. METHODS: DNA was extracted from DBSs on deidentified NBS cards, and real-time quantitative PCR (RT-qPCR) was used to determine the number of TRECs. Positive controls consisted of DBS from a 1-week-old T(-)B(-)NK(+) patient with SCID and whole blood specimens selectively depleted of naive T cells. RESULTS: The mean and median numbers of TRECs from 5766 deidentified DBSs were 827 and 708, respectively, per 3.2-mm punch ( approximately 3 muL whole blood). Ten samples failed to amplify TRECs on initial analysis; all but 1 demonstrated normal TRECs and beta-actin amplification on retesting. No TRECs were detected in either the SCID or naive T-cell-depleted samples, despite the presence of normal levels of beta-actin. CONCLUSIONS: The use of RT-qPCR to quantitate TRECs from DNA extracted from newborn DBSs is a highly sensitive and specific screening test for SCID. This assay is currently being used in Wisconsin for routine screening infants for SCID.


Asunto(s)
Tamizaje Neonatal/métodos , Receptores de Antígenos de Linfocitos T/genética , Inmunodeficiencia Combinada Grave/diagnóstico , Actinas/análisis , Actinas/inmunología , ADN/sangre , ADN/genética , Humanos , Recién Nacido , Reacción en Cadena de la Polimerasa/métodos , Receptores de Antígenos de Linfocitos T/inmunología , Reproducibilidad de los Resultados , Inmunodeficiencia Combinada Grave/inmunología
17.
J Clin Invest ; 116(4): 1005-15, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16585965

RESUMEN

An alpha1-adrenergic receptor (alpha1-AR) antagonist increased heart failure in the Antihypertensive and Lipid-Lowering Treatment to Prevent Heart Attack Trial (ALLHAT), but it is unknown whether this adverse result was due to alpha1-AR inhibition or a nonspecific drug effect. We studied cardiac pressure overload in mice with double KO of the 2 main alpha1-AR subtypes in the heart, alpha 1A (Adra1a) and alpha 1B (Adra1b). At 2 weeks after transverse aortic constriction (TAC), KO mouse survival was only 60% of WT, and surviving KO mice had lower ejection fractions and larger end-diastolic volumes than WT mice. Mechanistically, final heart weight and myocyte cross-sectional area were the same after TAC in KO and WT mice. However, KO hearts after TAC had increased interstitial fibrosis, increased apoptosis, and failed induction of the fetal hypertrophic genes. Before TAC, isolated KO myocytes were more susceptible to apoptosis after oxidative and beta-AR stimulation, and beta-ARs were desensitized. Thus, alpha1-AR deletion worsens dilated cardiomyopathy after pressure overload, by multiple mechanisms, indicating that alpha1-signaling is required for cardiac adaptation. These results suggest that the adverse cardiac effects of alpha1-antagonists in clinical trials are due to loss of alpha1-signaling in myocytes, emphasizing concern about clinical use of alpha1-antagonists, and point to a revised perspective on sympathetic activation in heart failure.


Asunto(s)
Presión Sanguínea/fisiología , Miocardio/metabolismo , Receptores Adrenérgicos alfa 1/metabolismo , Animales , Apoptosis , Cardiomiopatía Dilatada/metabolismo , Células Cultivadas , Fibrosis/patología , Regulación de la Expresión Génica , Hipertensión/metabolismo , Hipertensión/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Ratones Noqueados , Miocardio/patología , Miocitos Cardíacos , Transducción de Señal
18.
J Gene Med ; 11(9): 743-53, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19554624

RESUMEN

BACKGROUND: Transplantation of stem cells from various sources into infarcted hearts has the potential to promote myocardial regeneration. However, the regenerative capacity is limited partly as a result of the low survival rate of the transplanted cells in the ischemic myocardium. In the present study, we tested the hypothesis that combining cell and angiogenic gene therapies would provide additive therapeutic effects via co-injection of bone marrow-derived mesenchymal stem cells (MSCs) with an adeno-associated viral vector (AAV), MLCVEGF, which expresses vascular endothelial growth factor (VEGF) in a cardiac-specific and hypoxia-inducible manner. METHODS: MSCs isolated from transgenic mice expressing green fluorescent protein and MLCVEGF packaged in AAV serotype 1 capsid were injected into mouse hearts at the border of ischemic area, immediately after occlusion of the left anterior descending coronary, individually or together. Engrafted cells were detected and quantified by real-time polymerase chain reaction and immunostaining. Angiogenesis and infarct size were analyzed on histological and immunohistochemical stained sections. Cardiac function was analyzed by echocardiography. RESULTS: We found that co-injection of AAV1-MLCVEGF with MSCs reduced cell loss. Although injection of MSCs and AAV1-MLCVEGF individually improved cardiac function and reduced infarct size, co-injection of MSC and AAV1-MLCVEGF resulted in the best improvement in cardiac function as well as the smallest infarct among all groups. Moreover, injection of AAV1-MLCVEGF induced neovasculatures. Nonetheless, injection of MSCs attracted endogenous stem cell homing and increased scar thickness. CONCLUSIONS: Co-injection of MLCVEGF and MSCs in ischemic hearts can result in better cardiac function and MSC survival, compared to their individual injections, as a result of the additive effects of each therapy.


Asunto(s)
Terapia Genética , Infarto del Miocardio/terapia , Neovascularización Fisiológica/genética , Trasplante de Células Madre , Factor A de Crecimiento Endotelial Vascular/genética , Adenoviridae/genética , Animales , Secuencia de Bases , Western Blotting , Diferenciación Celular , Separación Celular , Cartilla de ADN , Femenino , Masculino , Células Madre Mesenquimatosas/citología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Infarto del Miocardio/cirugía
19.
Am J Ther ; 16(3): 239-46, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19145208

RESUMEN

Enhanced external counterpulsation (EECP) is a noninvasive technique that provides beneficial effects for patients with chronic, symptomatic angina pectoris. However, the direct left ventricular effects of EECP have not been studied invasively. We examined invasive right atrial pressure and left ventricular hemodynamics during EECP. Ten patients referred for diagnostic evaluation underwent left heart catheterization from the radial artery. At baseline and during EECP, left ventricular pressure and volume were measured using a micromanometer pressure-conductance catheter, along with recording of right atrial and central aortic pressures. Hemodynamics were recorded at different lower extremity cuff configuration and cuff inflation pressures. As cuff inflation pressure increased, EECP resulted in a dose-dependent increase in right atrial and aortic diastolic pressure (P < 0.0001). The increase in ventricular preload resulted in increased left ventricular volume. Maximum positive (P = 0.0003) and negative left ventricular dP/dt (P < 0.0001) increased. Left ventricular diastolic pressure decreased. There was a neutral effect on myocardial mechanical efficiency. In conclusion, EECP acutely increased right atrial and central aortic diastolic pressure. The increase in preload attenuated the reduction in left ventricular diastolic pressure resulting from systolic unloading. The increased preload counterbalanced the afterload reduction, resulting in a neutral effect on myocardial efficiency.


Asunto(s)
Contrapulsación/métodos , Función Ventricular Izquierda , Adulto , Anciano , Angina de Pecho/terapia , Cateterismo Cardíaco , Diástole , Femenino , Frecuencia Cardíaca , Humanos , Masculino , Persona de Mediana Edad , Sístole
20.
Am J Respir Crit Care Med ; 177(11): 1268-75, 2008 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-18310479

RESUMEN

RATIONALE: Circulating microparticles (MPs) are submicron membrane fragments shed from damaged or activated vascular cells. Endothelial MPs are a biological marker of dysfunctional endothelium. Vascular remodeling and endothelial dysfunction are involved in pulmonary hypertension (PH). OBJECTIVES: We tested the hypothesis that circulating MPs are increased in patients with PH and that identifiable subgroups of MPs predict the hemodynamic severity of this condition progression. METHODS: Patients (n = 24; age, 54 +/- 4 yr) undergoing right heart catheterization for precapillary PH without any endothelium-active vasodilator therapy participated in the study. Age- and sex-matched healthy control subjects (n = 20) were included. Endothelial (PECAM(+) [CD31(+)]/ CD41(-), VE-cadherin(+) [CD144(+)], and E-selectin(+) [CD62e(+)]), platelet (CD41(+)), leukocyte-derived (CD45(+)), and annexin V(+) MPs were measured by flow cytometry in platelet-free plasma from venous blood. MEASUREMENTS AND MAIN RESULTS: Levels of circulating endothelial PECAM(+), VE-cadherin(+), E-selectin(+), and leukocyte-derived MPs, but not platelet and annexin V(+) MPs, were increased in subjects with PH compared with control subjects (P < 0.01 each). PECAM(+) and VE-cadherin(+) MP levels significantly correlated with mean pulmonary artery pressure (r = 0.92 and r = 0.87, respectively), pulmonary vascular resistance (r = 0.78 and r = 0.73), and mean right atrial pressure (r = 0.43, and r = 0.46) and correlated inversely with cardiac index (r = -0.59 and r = -0.52). These relationships were not observed for other MP subgroups, and persisted in multivariate analysis after adjustment for confounding factors. CONCLUSIONS: In subjects with precapillary PH, levels of circulating endothelial and leukocyte MPs were increased compared with control subjects. In addition, levels of PECAM(+) and VE-cadherin(+), but not E-selectin(+), endothelial MPs predicted hemodynamic severity of the disease.


Asunto(s)
Antígenos CD/sangre , Hipertensión Pulmonar/sangre , Hipertensión Pulmonar/fisiopatología , Glicoproteínas de Membrana/sangre , Presión Sanguínea/fisiología , Estudios de Casos y Controles , Estudios de Cohortes , Femenino , Humanos , Hipertensión Pulmonar/etiología , Masculino , Persona de Mediana Edad , Valor Predictivo de las Pruebas , Índice de Severidad de la Enfermedad , Resistencia Vascular/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA