Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 135(3): 437-48, 2008 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-18984156

RESUMEN

Interactions between tumorigenic cells and their surrounding microenvironment are critical for tumor progression yet remain incompletely understood. Germline mutations in the NF1 tumor suppressor gene cause neurofibromatosis type 1 (NF1), a common genetic disorder characterized by complex tumors called neurofibromas. Genetic studies indicate that biallelic loss of Nf1 is required in the tumorigenic cell of origin in the embryonic Schwann cell lineage. However, in the physiologic state, Schwann cell loss of heterozygosity is not sufficient for neurofibroma formation and Nf1 haploinsufficiency in at least one additional nonneoplastic lineage is required for tumor progression. Here, we establish that Nf1 heterozygosity of bone marrow-derived cells in the tumor microenvironment is sufficient to allow neurofibroma progression in the context of Schwann cell Nf1 deficiency. Further, genetic or pharmacologic attenuation of c-kit signaling in Nf1+/- hematopoietic cells diminishes neurofibroma initiation and progression. Finally, these studies implicate mast cells as critical mediators of tumor initiation.


Asunto(s)
Neurofibroma/metabolismo , Neurofibromina 1/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Animales , Benzamidas , Médula Ósea/fisiopatología , Trasplante de Médula Ósea , Preescolar , Genes de Neurofibromatosis 1 , Humanos , Mesilato de Imatinib , Mastocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Neurofibroma/tratamiento farmacológico , Neurofibroma/genética , Neurofibroma/patología , Neurofibroma Plexiforme/tratamiento farmacológico , Neurofibroma Plexiforme/metabolismo , Piperazinas/uso terapéutico , Pirimidinas/uso terapéutico , Células de Schwann/metabolismo
2.
Pediatr Blood Cancer ; 67(8): e28372, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32459399

RESUMEN

BACKGROUND: Neurofibromatosis type 1 (NF1) is a common genetic disorder characterized by plexiform neurofibromas (pNF), which are thought to be congenital tumors that arise in utero and enlarge throughout life. Genetic studies in murine models delineated an indispensable role for the stem cell factor (SCF)/c-kit pathway in pNF initiation and progression. A subsequent phase 2 clinical trial using imatinib mesylate to inhibit SCF/c-kit demonstrated tumor shrinkage in a subset of preexisting pNF; however, imatinib's role on preventing pNF development has yet to be explored. PROCEDURE: We evaluated the effect of imatinib dosed at 10-100 mg/kg/day for 12 weeks to one-month-old Nf1flox/flox ;PostnCre(+) mice, prior to onset of pNF formation. To determine durability of response, we then monitored for pNF growth at later time points, comparing imatinib- with vehicle-treated mice. We assessed gross and histopathological analysis of tumor burden. RESULTS: Imatinib administered preventatively led to a significant decrease in pNF number, even at doses as low as 10 mg/kg/day. Tumor development continued to be significantly inhibited after cessation of imatinib dosed at 50 and 100 mg/kg/day. In the cohort of treated mice that underwent prolonged follow-up, the size of residual tumors was significantly reduced as compared with age-matched littermates that received vehicle control. CONCLUSIONS: Early administration of imatinib inhibits pNF genesis in vivo, and effects are sustained after discontinuation of therapy. These findings may guide clinical use of imatinib in young NF1 patients prior to the substantial development of pNF.


Asunto(s)
Mesilato de Imatinib/administración & dosificación , Neoplasias Experimentales/prevención & control , Neurofibroma Plexiforme/prevención & control , Neurofibromatosis 1/prevención & control , Animales , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Ratones , Ratones Transgénicos , Neoplasias Experimentales/genética , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Neurofibroma Plexiforme/genética , Neurofibroma Plexiforme/metabolismo , Neurofibroma Plexiforme/patología , Neurofibromatosis 1/genética , Neurofibromatosis 1/metabolismo , Neurofibromatosis 1/patología
3.
Hum Mol Genet ; 25(6): 1129-39, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26740548

RESUMEN

Persons with neurofibromatosis type 1 (NF1) have a predisposition for premature and severe arterial stenosis. Mutations in the NF1 gene result in decreased expression of neurofibromin, a negative regulator of p21(Ras), and increases Ras signaling. Heterozygous Nf1 (Nf1(+/-)) mice develop a marked arterial stenosis characterized by proliferating smooth muscle cells (SMCs) and a predominance of infiltrating macrophages, which closely resembles arterial lesions from NF1 patients. Interestingly, lineage-restricted inactivation of a single Nf1 allele in monocytes/macrophages is sufficient to recapitulate the phenotype observed in Nf1(+/-) mice and to mobilize proinflammatory CCR2+ monocytes into the peripheral blood. Therefore, we hypothesized that CCR2 receptor activation by its primary ligand monocyte chemotactic protein-1 (MCP-1) is critical for monocyte infiltration into the arterial wall and neointima formation in Nf1(+/-) mice. MCP-1 induces a dose-responsive increase in Nf1(+/-) macrophage migration and proliferation that corresponds with activation of multiple Ras kinases. In addition, Nf1(+/-) SMCs, which express CCR2, demonstrate an enhanced proliferative response to MCP-1 when compared with WT SMCs. To interrogate the role of CCR2 activation on Nf1(+/-) neointima formation, we induced neointima formation by carotid artery ligation in Nf1(+/-) and WT mice with genetic deletion of either MCP1 or CCR2. Loss of MCP-1 or CCR2 expression effectively inhibited Nf1(+/-) neointima formation and reduced macrophage content in the arterial wall. Finally, administration of a CCR2 antagonist significantly reduced Nf1(+/-) neointima formation. These studies identify MCP-1 as a potent chemokine for Nf1(+/-) monocytes/macrophages and CCR2 as a viable therapeutic target for NF1 arterial stenosis.


Asunto(s)
Macrófagos/patología , Monocitos/patología , Neointima/patología , Neurofibromatosis 1/patología , Receptores CCR2/metabolismo , Animales , Arterias Carótidas/metabolismo , Arterias Carótidas/patología , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Genes de Neurofibromatosis 1 , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/metabolismo , Miocitos del Músculo Liso/metabolismo , Neointima/genética , Neointima/metabolismo , Neurofibromatosis 1/genética , Neurofibromatosis 1/metabolismo , Neurofibromina 1/genética , Receptores CCR2/antagonistas & inhibidores , Receptores CCR2/genética , Transducción de Señal
4.
Am J Respir Crit Care Med ; 196(3): 364-374, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28249118

RESUMEN

RATIONALE: Mechanisms contributing to chronic lung disease after preterm birth are incompletely understood. OBJECTIVES: To identify antenatal risk factors associated with increased risk for bronchopulmonary dysplasia (BPD) and respiratory disease during early childhood after preterm birth, we performed a prospective, longitudinal study of 587 preterm infants with gestational age less than 34 weeks and birth weights between 500 and 1,250 g. METHODS: Data collected included perinatal information and assessments during the neonatal intensive care unit admission and longitudinal follow-up by questionnaire until 2 years of age. MEASUREMENTS AND MAIN RESULTS: After adjusting for covariates, we found that maternal smoking prior to preterm birth increased the odds of having an infant with BPD by twofold (P = 0.02). Maternal smoking was associated with prolonged mechanical ventilation and respiratory support during the neonatal intensive care unit admission. Preexisting hypertension was associated with a twofold (P = 0.04) increase in odds for BPD. Lower gestational age and birth weight z-scores were associated with BPD. Preterm infants who were exposed to maternal smoking had higher rates of late respiratory disease during childhood. Twenty-two percent of infants diagnosed with BPD and 34% of preterm infants without BPD had no clinical signs of late respiratory disease during early childhood. CONCLUSIONS: We conclude that maternal smoking and hypertension increase the odds for developing BPD after preterm birth, and that maternal smoking is strongly associated with increased odds for late respiratory morbidities during early childhood. These findings suggest that in addition to the BPD diagnosis at 36 weeks, other factors modulate late respiratory outcomes during childhood. We speculate that measures to reduce maternal smoking not only will lower the risk for preterm birth but also will improve late respiratory morbidities after preterm birth.


Asunto(s)
Displasia Broncopulmonar/epidemiología , Recien Nacido Prematuro , Madres/estadística & datos numéricos , Efectos Tardíos de la Exposición Prenatal/epidemiología , Síndrome de Dificultad Respiratoria del Recién Nacido/epidemiología , Fumar/epidemiología , Causalidad , Colorado/epidemiología , Femenino , Estudios de Seguimiento , Humanos , Hipertensión/epidemiología , Recién Nacido , Estudios Longitudinales , Masculino , Embarazo , Estudios Prospectivos , Factores de Riesgo , Encuestas y Cuestionarios
5.
J Vasc Surg ; 66(6): 1854-1863, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28655551

RESUMEN

OBJECTIVE: Postnatal resident endothelium of blood vessels has been proposed to represent terminally differentiated tissue that does not replicate. We previously isolated endothelial colony-forming cells (ECFCs) from human umbilical cord blood (CB) and term placenta by using colony-forming assays and immunocytochemistry. We showed that ECFCs are highly proliferative and form functioning vessels in vivo, the defining characteristics of a true endothelial progenitor cell. This exploratory investigation was conducted to determine whether the endothelium of healthy adult blood vessels contained resident ECFCs. METHODS: The endothelium of great saphenous vein (GSV) obtained from vein stripping procedures was collected with mechanical scraping, and ECFCs were isolated according to established protocols. RESULTS: GSV ECFCs incorporated acetylated low-density lipoprotein, formed tubules in Matrigel (BD Biosciences, San Jose, Calif) at 24 hours, and expressed endothelial antigens cluster of differentiation (CD) 144, CD31, CD105, and kinase insert domain receptor but not hematopoietic antigen CD45. Using cumulative population doublings and single-cell assays, we demonstrated that GSV ECFCs exhibited comparable proliferative capacities compared with CB ECFCs, including similar numbers of highly proliferative cells. When injected in collagen/fibronectin gels implanted in nonobese diabetic/severe combined immune deficiency mice, GSV ECFCs formed blood vessels with circulating murine red blood cells, demonstrating their vasculogenic potential. CONCLUSIONS: The ECFCs of the GSV contain a hierarchy of progenitor cells with a comparable number of highly proliferative clones as ECFCs of CB. The results of this investigation demonstrate that the adult endothelium contains resident progenitor cells that may have a critical role in vascular homeostasis and repair and could potentially be used as a source of autologous cells for cell therapies focusing on vasculogenesis.


Asunto(s)
Células Madre Adultas/fisiología , Proliferación Celular , Células Progenitoras Endoteliales/fisiología , Neovascularización Fisiológica , Vena Safena/citología , Nicho de Células Madre , Células Madre Adultas/metabolismo , Células Madre Adultas/trasplante , Animales , Biomarcadores/metabolismo , Separación Celular/métodos , Células Cultivadas , Células Progenitoras Endoteliales/metabolismo , Células Progenitoras Endoteliales/trasplante , Humanos , Cinética , Ratones Endogámicos NOD , Ratones SCID , Fenotipo
6.
Pediatr Blood Cancer ; 63(2): 206-13, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26375012

RESUMEN

PURPOSE: Plexiform neurofibromas (pNF) are pathognomonic nerve and soft tissue tumors of neurofibromatosis type I (NF1), which are highly resistant to conventional chemotherapy and associated with significant morbidity/mortality. Disruption of aberrant SCF/c-Kit signaling emanating from the pNF microenvironment induced the first ever objective therapeutic responses in a recent phase 2 trial. Sunitinib malate is a potent, highly selective RTK inhibitor with activity against c-Kit, PDGFR, and VEGFR, which have also been implicated in the pathogenesis of these lesions. Here, we evaluate the efficacy of sunitinib malate in a preclinical Krox20;Nf1(flox/-) pNF murine model. EXPERIMENTAL DESIGN: Proliferation, ß-hexosaminidase release (degranulation), and Erk1/2 phosphorylation were assessed in sunitinib treated Nf1(+/-) mast cells and fibroblasts, respectively. Krox20;Nf1(flox/-) mice with established pNF were treated sunitinib or PBS-vehicle control for a duration of 12 weeks. pNF metabolic activity was monitored by serial [(18)F]DG-PET/CT imaging. RESULTS: Sunitinib suppressed multiple in vitro gain-in-functions of Nf1(+/-) mast cells and fibroblasts and attenuated Erk1/2 phosphorylation. Sunitinib treated Krox20;Nf1(flox/-) mice exhibited significant reductions in pNF size, tumor number, and FDG uptake compared to control mice. Histopathology revealed reduced tumor cellularity and infiltrating mast cells, markedly diminished collagen deposition, and increased cellular apoptosis in sunitinib treated pNF. CONCLUSIONS: Collectively, these results demonstrate the efficacy of sunitinib in reducing tumor burden in Krox20;Nf1(flox/-) mice. These preclinical findings demonstrate the utility of inhibiting multiple RTKs in pNF and provide insights into the design of future clinical trials.


Asunto(s)
Antineoplásicos/farmacología , Indoles/farmacología , Neurofibroma Plexiforme/patología , Pirroles/farmacología , Microambiente Tumoral/efectos de los fármacos , Animales , Western Blotting , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/patología , Masculino , Mastocitos/efectos de los fármacos , Mastocitos/patología , Ratones , Ratones Transgénicos , Tomografía de Emisión de Positrones , Sunitinib
7.
Am J Respir Crit Care Med ; 191(1): 87-95, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25389562

RESUMEN

RATIONALE: Pulmonary hypertension (PH) is associated with poor outcomes among preterm infants with bronchopulmonary dysplasia (BPD), but whether early signs of pulmonary vascular disease are associated with the subsequent development of BPD or PH at 36 weeks post-menstrual age (PMA) is unknown. OBJECTIVES: To prospectively evaluate the relationship of early echocardiogram signs of pulmonary vascular disease in preterm infants to the subsequent development of BPD and late PH (at 36 wk PMA). METHODS: Prospectively enrolled preterm infants with birthweights 500-1,250 g underwent echocardiogram evaluations at 7 days of age (early) and 36 weeks PMA (late). Clinical and echocardiographic data were analyzed to identify early risk factors for BPD and late PH. MEASUREMENTS AND MAIN RESULTS: A total of 277 preterm infants completed echocardiogram and BPD assessments at 36 weeks PMA. The median gestational age at birth and birthweight of the infants were 27 weeks and 909 g, respectively. Early PH was identified in 42% of infants, and 14% were diagnosed with late PH. Early PH was a risk factor for increased BPD severity (relative risk, 1.12; 95% confidence interval, 1.03-1.23) and late PH (relative risk, 2.85; 95% confidence interval, 1.28-6.33). Infants with late PH had greater duration of oxygen therapy and increased mortality in the first year of life (P < 0.05). CONCLUSIONS: Early pulmonary vascular disease is associated with the development of BPD and with late PH in preterm infants. Echocardiograms at 7 days of age may be a useful tool to identify infants at high risk for BPD and PH.


Asunto(s)
Displasia Broncopulmonar/prevención & control , Hipertensión Pulmonar/complicaciones , Recien Nacido Prematuro , Pulmón/fisiopatología , Terapia por Inhalación de Oxígeno/efectos adversos , Respiración Artificial/efectos adversos , Enfermedades Vasculares/complicaciones , Displasia Broncopulmonar/epidemiología , Displasia Broncopulmonar/etiología , Colorado , Femenino , Humanos , Hipertensión Pulmonar/diagnóstico por imagen , Hipertensión Pulmonar/epidemiología , Hipertensión Pulmonar/etiología , Incidencia , Indiana , Recién Nacido de Bajo Peso , Recién Nacido , Modelos Logísticos , Pulmón/diagnóstico por imagen , Pulmón/patología , Masculino , Terapia por Inhalación de Oxígeno/métodos , Estudios Prospectivos , Respiración Artificial/métodos , Factores de Riesgo , Facultades de Medicina , Factores de Tiempo , Ultrasonografía , Enfermedades Vasculares/diagnóstico por imagen , Enfermedades Vasculares/epidemiología
8.
Circulation ; 129(11): 1213-24, 2014 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-24370551

RESUMEN

BACKGROUND: Neurofibromatosis type 1 (NF1) is a genetic disorder resulting from mutations in the NF1 tumor suppressor gene. Neurofibromin, the protein product of NF1, functions as a negative regulator of Ras activity in circulating hematopoietic and vascular wall cells, which are critical for maintaining vessel wall homeostasis. NF1 patients have evidence of chronic inflammation resulting in the development of premature cardiovascular disease, including arterial aneurysms, which may manifest as sudden death. However, the molecular pathogenesis of NF1 aneurysm formation is unknown. METHOD AND RESULTS: With the use of an angiotensin II-induced aneurysm model, we demonstrate that heterozygous inactivation of Nf1 (Nf1(+/-)) enhanced aneurysm formation with myeloid cell infiltration and increased oxidative stress in the vessel wall. Using lineage-restricted transgenic mice, we show that loss of a single Nf1 allele in myeloid cells is sufficient to recapitulate the Nf1(+/-) aneurysm phenotype in vivo. Finally, oral administration of simvastatin or the antioxidant apocynin reduced aneurysm formation in Nf1(+/-) mice. CONCLUSION: These data provide genetic and pharmacological evidence that Nf1(+/-) myeloid cells are the cellular triggers for aneurysm formation in a novel model of NF1 vasculopathy and provide a potential therapeutic target.


Asunto(s)
Aneurisma/metabolismo , Células Mieloides/metabolismo , Neurofibromina 1/deficiencia , Aneurisma/tratamiento farmacológico , Aneurisma/genética , Animales , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Transgénicos , Neurofibromina 1/genética , Simvastatina/uso terapéutico
9.
Hum Mol Genet ; 22(5): 977-88, 2013 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-23197650

RESUMEN

Mutations in the NF1 tumor suppressor gene cause Neurofibromatosis type 1 (NF1). Neurofibromin, the protein product of NF1, functions as a negative regulator of Ras activity. Some NF1 patients develop cardiovascular disease, which represents an underrecognized disease complication and contributes to excess morbidity and mortality. Specifically, NF1 patients develop arterial occlusion resulting in tissue ischemia and sudden death. Murine studies demonstrate that heterozygous inactivation of Nf1 (Nf1(+/-)) in bone marrow cells enhances neointima formation following arterial injury. Macrophages infiltrate Nf1(+/-) neointimas, and NF1 patients have increased circulating inflammatory monocytes in their peripheral blood. Therefore, we tested the hypothesis that heterozygous inactivation of Nf1 in myeloid cells is sufficient for neointima formation. Specific ablation of a single copy of the Nf1 gene in myeloid cells alone mobilizes a discrete pro-inflammatory murine monocyte population via a cell autonomous and gene-dosage dependent mechanism. Furthermore, lineage-restricted heterozygous inactivation of Nf1 in myeloid cells is sufficient to reproduce the enhanced neointima formation observed in Nf1(+/-) mice when compared with wild-type controls, and homozygous inactivation of Nf1 in myeloid cells amplified the degree of arterial stenosis after arterial injury. Treatment of Nf1(+/-) mice with rosuvastatin, a stain with anti-inflammatory properties, significantly reduced neointima formation when compared with control. These studies identify neurofibromin-deficient myeloid cells as critical cellular effectors of Nf1(+/-) neointima formation and propose a potential therapeutic for NF1 cardiovascular disease.


Asunto(s)
Arteriopatías Oclusivas/genética , Fluorobencenos/administración & dosificación , Neointima/tratamiento farmacológico , Neointima/genética , Neurofibromatosis 1/genética , Neurofibromina 1/genética , Pirimidinas/administración & dosificación , Sulfonamidas/administración & dosificación , Animales , Antiinflamatorios/administración & dosificación , Arteriopatías Oclusivas/complicaciones , Arteriopatías Oclusivas/fisiopatología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Heterocigoto , Humanos , Macrófagos/citología , Macrófagos/patología , Redes y Vías Metabólicas/efectos de los fármacos , Ratones , Células Mieloides/citología , Células Mieloides/efectos de los fármacos , Células Mieloides/metabolismo , Neointima/fisiopatología , Neurofibromatosis 1/complicaciones , Neurofibromatosis 1/fisiopatología , Neurofibromina 1/metabolismo , Rosuvastatina Cálcica
10.
Am J Physiol Heart Circ Physiol ; 309(7): H1207-17, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26297224

RESUMEN

The present study was undertaken to establish the role of NADPH oxidase (Nox) in impaired vascular compensation to arterial occlusion that occurs in the presence of risk factors associated with oxidative stress. Diet-induced obese (DIO) mice characterized by multiple comorbidities including diabetes and hyperlipidemia were used as a preclinical model. Arterial occlusion was induced by distal femoral artery ligation in lean and DIO mice. Proximal collateral arteries were identified as the site of major (∼70%) vascular resistance to calf perfusion by distal arterial pressures, which decreased from ∼80 to ∼30 mmHg with ligation in both lean and DIO mice. Two weeks after ligation, significant vascular compensation occurred in lean but not DIO mice as evidenced by increased perfusion (147 ± 48% vs. 49 ± 29%) and collateral diameter (151 ± 30% vs. 44 ± 17%). Vascular mRNA expression of p22(phox), Nox2, Nox4, and p47(phox) were all increased in DIO mice. Treatment of DIO mice with either apocynin or Nox2ds-tat or with whole body ablation of either Nox2 or p47(phox) ameliorated the impairment in both collateral growth and hindlimb perfusion. Multiparametric flow cytometry analysis demonstrated elevated levels of circulating monocytes in DIO mice without impaired mobilization and demargination after femoral artery ligation. These results establish collateral resistance as the major limitation to calf perfusion in this preclinical model, demonstrate than monocyte mobilization and demarginatin is not suppressed, implicate Nox2-p47(phox) interactions in the impairment of vascular compensation to arterial occlusion in DIO mice, and suggest that selective Nox component suppression/inhibition may be effective as either primary or adjuvant therapy for claudicants.


Asunto(s)
Adaptación Fisiológica , Circulación Colateral , Arteria Femoral/cirugía , Glicoproteínas de Membrana/genética , NADPH Oxidasas/genética , Neovascularización Fisiológica , Obesidad/metabolismo , ARN Mensajero/metabolismo , Acetofenonas/farmacología , Animales , Antioxidantes/farmacología , Grupo Citocromo b/genética , Grupo Citocromo b/metabolismo , Miembro Posterior/irrigación sanguínea , Ligadura , Glicoproteínas de Membrana/metabolismo , Ratones , NADPH Oxidasa 2 , NADPH Oxidasa 4 , NADPH Oxidasas/metabolismo , Estrés Oxidativo
11.
Am J Pathol ; 184(1): 79-85, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24211110

RESUMEN

Neurofibromatosis type 1 (NF1) results from mutations in the NF1 tumor-suppressor gene, which encodes neurofibromin, a negative regulator of diverse Ras signaling cascades. Arterial stenosis is a nonneoplastic manifestation of NF1 that predisposes some patients to debilitating morbidity and sudden death. Recent murine studies demonstrate that Nf1 heterozygosity (Nf1(+/-)) in monocytes/macrophages significantly enhances intimal proliferation after arterial injury. However, the downstream Ras effector pathway responsible for this phenotype is unknown. Based on in vitro assays demonstrating enhanced extracellular signal-related kinase (Erk) signaling in Nf1(+/-) macrophages and vascular smooth muscle cells and in vivo evidence of Erk amplification without alteration of phosphatidylinositol 3-kinase signaling in Nf1(+/-) neointimas, we tested the hypothesis that Ras-Erk signaling regulates intimal proliferation in a murine model of NF1 arterial stenosis. By using a well-established in vivo model of inflammatory cell migration and standard cell culture, neurofibromin-deficient macrophages demonstrate enhanced sensitivity to growth factor stimulation in vivo and in vitro, which is significantly diminished in the presence of PD0325901, a specific inhibitor of Ras-Erk signaling in phase 2 clinical trials for cancer. After carotid artery injury, Nf1(+/-) mice demonstrated increased intimal proliferation compared with wild-type mice. Daily administration of PD0325901 significantly reduced Nf1(+/-) neointima formation to levels of wild-type mice. These studies identify the Ras-Erk pathway in neurofibromin-deficient macrophages as the aberrant pathway responsible for enhanced neointima formation.


Asunto(s)
Estenosis Carotídea/patología , Macrófagos/metabolismo , Neointima/patología , Neurofibromatosis 1/metabolismo , Neurofibromina 1/metabolismo , Transducción de Señal/fisiología , Animales , Western Blotting , Estenosis Carotídea/metabolismo , Modelos Animales de Enfermedad , Sistema de Señalización de MAP Quinasas/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Neointima/metabolismo , Neurofibromatosis 1/genética , Neurofibromatosis 1/patología , Neurofibromina 1/genética , Proteínas ras/fisiología
12.
Eur Respir J ; 43(2): 497-504, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23682112

RESUMEN

Angiogenesis is a critical determinant of alveolarisation, which increases alveolar surface area and pulmonary capillary blood volume in infants; however, our understanding of this process is very limited. The purpose of our study was to measure the pulmonary membrane diffusion capacity (DM) and pulmonary capillary blood volume (VC) components of the diffusing capacity of the lung for carbon monoxide (DLCO) in healthy infants and toddlers, and evaluate whether these components were associated with pro-angiogenic circulating haematopoietic stem/progenitor cells (pCHSPCs) early in life. 21 healthy subjects (11 males), 3-25 months of age, were evaluated. DLCO was measured under normoxic and hyperoxic conditions, and DM and VC were calculated. From 1 mL venous blood, pCHSPCs were quantified by multiparametric flow cytometry. DM and VC increased with increasing body length; however, membrane resistance as a fraction of total resistance to pulmonary diffusion remained constant with somatic size. In addition, DLCO and VC, but not DM, increased with an increasing percentage of pCHSPCs. The parallel increase in the membrane and vascular components of pulmonary diffusion is consistent with alveolarisation during this period of rapid lung growth. In addition, the relationship between pCHSPCs and VC suggest that pro-angiogenic cells may contribute to this vascular process.


Asunto(s)
Capilares/fisiología , Pulmón/fisiología , Capacidad de Difusión Pulmonar , Preescolar , Difusión , Femenino , Humanos , Hiperoxia , Lactante , Modelos Lineales , Masculino , Neovascularización Fisiológica , Oxígeno/química , Intercambio Gaseoso Pulmonar
13.
Acad Med ; 99(4): 388-394, 2024 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-38166333

RESUMEN

ABSTRACT: Indiana was the first state to pass legislation severely restricting access to abortion care following the Dobbs v. Jackson Women's Health Organization decision. Indiana Senate Enrolled Act 1 (SEA 1) outlaws all abortions with few exceptions. Indiana University Health (IU Health), the largest and only academic health system in the state, has a unique relationship with the Indiana University School of Medicine and a vision to improve the health of Indiana residents. IU Health employed the Hospital Incident Command System model to create a plan to ensure its patients continue to have access to safe, high-quality family planning, maternal, and neonatal care services and that clinicians are protected against criminal penalties and threats to personal safety. This article provides an overview of the Incident Command structure used to rapidly work across many disciplines, tackle complex issues, respond to concerns, and design and implement changes. The article also outlines the key considerations and decisions made by Incident Command leaders, such as where abortions that met the new law's criteria should be performed, changes to clinical workflows and protocols, and the creation of a rapid response team. The article then examines the operational, legal, and clinical challenges encountered by clinicians and health care team members, including a lack of peer support or idea sharing with other health systems in the state; accurate estimation of abortion, live birth, and neonatal intensive care unit volumes; and ambiguity in the law and lack of guidance from the state government. Recommendations regarding communication with clinicians and other health care team members and engaging information technology early are offered for health systems and medical schools that may face legislative barriers to health care delivery in the future. Finally, IU Health's commitment to tracking the impact of SEA 1 on patients, clinicians, employees, and the state is outlined.


Asunto(s)
Aborto Inducido , Salud de la Mujer , Embarazo , Recién Nacido , Femenino , Humanos , Atención a la Salud , Hospitales , Comunicación , Gobierno Estatal
14.
Arterioscler Thromb Vasc Biol ; 32(4): 1045-53, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22282356

RESUMEN

OBJECTIVE: We sought to identify and characterize 2 distinct populations of bona fide circulating endothelial cells, including the endothelial colony-forming cell (ECFC), by polychromatic flow cytometry (PFC), colony assays, immunomagnetic selection, and electron microscopy. METHODS AND RESULTS: Mononuclear cells from human umbilical cord blood and peripheral blood were analyzed using our recently published PFC protocol. A population of cells containing both ECFCs and mature circulating endothelial cells was determined by varying expressions of CD34, CD31, and CD146 but not AC133 and CD45. After immunomagnetic separation, these cells failed to form hematopoietic colonies, yet clonogenic endothelial colonies with proliferative potential were obtained, thus verifying their identity as ECFCs. The frequency of ECFCs were increased in cord blood and were extremely rare in the peripheral blood of healthy adults. We also detected another mature endothelial cell population in the circulation that was apoptotic. Finally, when comparing this new protocol with a prior method, we determined that the present protocol identifies circulating endothelial cells, whereas the earlier protocol identified extracellular vesicles. CONCLUSIONS: Two populations of circulating endothelial cells, including the functionally characterized ECFC, are now identifiable in human cord blood and peripheral blood by PFC.


Asunto(s)
Separación Celular/métodos , Células Endoteliales/clasificación , Citometría de Flujo , Células Madre/clasificación , Antígeno AC133 , Adulto , Animales , Antígenos CD/análisis , Antígenos CD34/análisis , Apoptosis , Biomarcadores/análisis , Antígeno CD146/análisis , Proliferación Celular , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Células Endoteliales/inmunología , Células Endoteliales/trasplante , Células Endoteliales/ultraestructura , Femenino , Sangre Fetal/citología , Glicoproteínas/análisis , Humanos , Separación Inmunomagnética , Recién Nacido , Antígenos Comunes de Leucocito/análisis , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Microscopía Electrónica de Transmisión , Péptidos/análisis , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/análisis , Trasplante de Células Madre , Células Madre/inmunología , Células Madre/ultraestructura , Adulto Joven
15.
Lancet Oncol ; 13(12): 1218-24, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23099009

RESUMEN

BACKGROUND: Plexiform neurofibromas are slow-growing chemoradiotherapy-resistant tumours arising in patients with neurofibromatosis type 1 (NF1). Currently, there are no viable therapeutic options for patients with plexiform neurofibromas that cannot be surgically removed because of their proximity to vital body structures. We undertook an open-label phase 2 trial to test whether treatment with imatinib mesylate can decrease the volume burden of clinically significant plexiform neurofibromas in patients with NF1. METHODS: Eligible patients had to be aged 3-65 years, and to have NF1 and a clinically significant plexiform neurofibroma. Patients were treated with daily oral imatinib mesylate at 220 mg/m(2) twice a day for children and 400 mg twice a day for adults for 6 months. The primary endpoint was a 20% or more reduction in plexiform size by sequential volumetric MRI imaging. Clinical data were analysed on an intention-to-treat basis; a secondary analysis was also done for those patients able to take imatinib mesylate for 6 months. This trial is registered with ClinicalTrials.gov, number NCT01673009. FINDINGS: Six of 36 patients (17%, 95% CI 6-33), enrolled on an intention-to-treat basis, had an objective response to imatinib mesylate, with a 20% or more decrease in tumour volume. Of the 23 patients who received imatinib mesylate for at least 6 months, six (26%, 95% CI 10-48) had a 20% or more decrease in volume of one or more plexiform tumours. The most common adverse events were skin rash (five patients) and oedema with weight gain (six). More serious adverse events included reversible grade 3 neutropenia (two), grade 4 hyperglycaemia (one), and grade 4 increases in aminotransferase concentrations (one). INTERPRETATION: Imatinib mesylate could be used to treat plexiform neurofibromas in patients with NF1. A multi-institutional clinical trial is warranted to confirm these results. FUNDING: Novartis Pharmaceuticals, the Indiana University Simon Cancer Centre, and the Indiana University Herman B Wells Center for Pediatric Research.


Asunto(s)
Antineoplásicos/uso terapéutico , Neurofibroma Plexiforme/tratamiento farmacológico , Neurofibromatosis 1/complicaciones , Piperazinas/uso terapéutico , Inhibidores de Proteínas Quinasas/uso terapéutico , Pirimidinas/uso terapéutico , Adolescente , Adulto , Benzamidas , Niño , Preescolar , Femenino , Humanos , Mesilato de Imatinib , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Neurofibroma Plexiforme/complicaciones , Neurofibroma Plexiforme/patología , Adulto Joven
16.
Pediatr Res ; 71(2): 156-61, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22258126

RESUMEN

INTRODUCTION: We have identified a novel hierarchy of human endothelial colony-forming cells (ECFCs) that are functionally defined by their proliferative and clonogenic potential and in vivo vessel-forming ability. The rhesus monkey provides an excellent model in which to examine the changes in circulating concentrations and functions of ECFCs since this nonhuman primate possesses a long lifespan and has been used extensively to model age-related processes that occur in humans. RESULTS: Endothelial cells (ECs) derived from rhesus monkey ECFCs share a cell-surface phenotype similar to human cord blood ECFCs, rapidly form capillary-like structures in vitro, and form endothelial-lined vessels in vivo upon implantation in immunodeficient mice in an age-dependent manner. Of interest, although ECFCs from the oldest monkeys formed capillary-like structures in vitro, the cells failed to form inosculating vessels when implanted in vivo and displayed a deficiency in cytoplasmic vacuolation in vitro; a critical first step in vasculogenesis. DISCUSSION: Utilizing previously established clonogenic assays for defining different subpopulations of human ECFCs, we have shown that a hierarchy of ECFCs, identical to human cells, can be isolated from the peripheral blood of rhesus monkeys, and that the frequency of the circulating cells varies with age. These studies establish the rhesus monkey as an important preclinical model for evaluating the role and function of circulating ECFCs in vascular homeostasis and aging. METHODS: Peripheral blood samples were collected from 40 healthy rhesus monkeys from birth to 24 years of age for ECFC analysis including immunophenotyping, clonogenic assays, and in vivo vessel formation.


Asunto(s)
Envejecimiento , Proliferación Celular , Células Endoteliales/fisiología , Neovascularización Fisiológica , Células Madre/fisiología , Factores de Edad , Animales , Animales Recién Nacidos , Biomarcadores/metabolismo , Separación Celular , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Células Endoteliales/metabolismo , Células Endoteliales/trasplante , Inmunofenotipificación , Macaca mulatta , Ratones , Ratones Endogámicos NOD , Ratones SCID , Modelos Animales , Fenotipo , Trasplante de Células Madre , Células Madre/metabolismo , Vacuolas/fisiología
17.
Blood ; 113(12): 2695-705, 2009 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-19124833

RESUMEN

Mast cells are key participants in allergic diseases via activation of high-affinity IgE receptors (FcepsilonRI) resulting in release of proinflammatory mediators. The biochemical pathways linking IgE activation to calcium influx and cytoskeletal changes required for intracellular granule release are incompletely understood. We demonstrate, genetically, that Pak1 is required for this process. In a passive cutaneous anaphylaxis experiment, W(sh)/W(sh) mast cell-deficient mice locally reconstituted with Pak1(-/-) bone marrow-derived mast cells (BMMCs) experienced strikingly decreased allergen-induced vascular permeability compared with controls. Consistent with the in vivo phenotype, Pak1(-/-) BMMCs exhibited a reduction in FcepsilonRI-induced degranulation. Further, Pak1(-/-) BMMCs demonstrated diminished calcium mobilization and altered depolymerization of cortical filamentous actin (F-actin) in response to FcepsilonRI stimulation. These data implicate Pak1 as an essential molecular target for modulating acute mast cell responses that contribute to allergic diseases.


Asunto(s)
Señalización del Calcio/fisiología , Citoesqueleto/ultraestructura , Mastocitos/metabolismo , Quinasas p21 Activadas/fisiología , Actinas/metabolismo , Traslado Adoptivo , Animales , Antígenos CD/genética , Antígenos CD/fisiología , Transporte Biológico , Biopolímeros , Células de la Médula Ósea/citología , Calcimicina/farmacología , Señalización del Calcio/efectos de los fármacos , Citoesqueleto/metabolismo , Activación Enzimática , Femenino , Inmunoglobulina E/inmunología , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Anafilaxis Cutánea Pasiva/inmunología , Glicoproteínas de Membrana Plaquetaria , Quimera por Radiación , Receptores de IgE/fisiología , Proteínas Recombinantes de Fusión/fisiología , Vesículas Secretoras/efectos de los fármacos , Vesículas Secretoras/metabolismo , Transducción de Señal , Tetraspanina 30 , beta-N-Acetilhexosaminidasas/metabolismo , Quinasas p21 Activadas/deficiencia , Quinasas p21 Activadas/genética
18.
Am J Obstet Gynecol ; 204(3): 254.e8-254.e15, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21167470

RESUMEN

OBJECTIVE: The purpose of this study was to examine whether women with gestational diabetes mellitus (GDM) and their offspring have reduced endothelial progenitor cell subsets and vascular reactivity. STUDY DESIGN: Women with GDM, healthy control subjects, and their infants participated. Maternal blood and cord blood were assessed for colony-forming unit-endothelial cells and endothelial progenitor cell subsets with the use of polychromatic flow cytometry. Cord blood endothelial colony-forming cells were enumerated. Vascular reactivity was tested by laser Doppler imaging. RESULTS: Women with GDM had fewer CD34, CD133, CD45, and CD31 cells (circulating progenitor cells [CPCs]) at 24-32 weeks' gestation and 1-2 days after delivery, compared with control subjects. No differences were detected in colony-forming unit-endothelial cells or colony-forming unit-endothelial cells. In control subjects, CPCs were higher in the third trimester, compared with the postpartum period. Cord blood from GDM pregnancies had reduced CPCs. Vascular reactivity was not different between GDM and control subjects. CONCLUSION: The normal physiologic increase in CPCs during pregnancy is impaired in women with GDM, which may contribute to endothelial dysfunction and GDM-associated morbidities.


Asunto(s)
Diabetes Gestacional/fisiopatología , Células Endoteliales/fisiología , Endotelio Vascular/fisiopatología , Piel/irrigación sanguínea , Células Madre/fisiología , Adulto , Femenino , Humanos , Recién Nacido , Flujometría por Láser-Doppler , Embarazo , Flujo Sanguíneo Regional
19.
Circ Res ; 104(12): 1410-20, 2009 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-19443841

RESUMEN

Rapid induction and maintenance of blood flow through new vascular networks is essential for successfully treating ischemic tissues and maintaining function of engineered neo-organs. We have previously shown that human endothelial progenitor cells (EPCs) form functioning vessels in mice, but these are limited in number and persistence; and also that human adipose stromal cells (ASCs) are multipotent cells with pericytic properties which can stabilize vascular assembly in vitro. In this study, we tested whether ASCs would cooperate with EPCs to coassemble vessels in in vivo implants. Collagen implants containing EPCs, ASCs, or a 4:1 mixture of both were placed subcutaneously into NOD/SCID mice. After a range of time periods, constructs were explanted and evaluated with regard to vascular network assembly and cell fate; and heterotypic cell interactions were explored by targeted molecular perturbations. The density and complexity of vascular networks formed by the synergistic dual-cell system was many-fold higher than found in implants containing either ASCs or EPCs alone. Coimplantation of ASCs and EPCs with either pancreatic islets or adipocytes produced neoorgans populated by these parenchymal cells, as well as by chimeric human vessels conducting flow. This study is the first to demonstrate prompt and consistent assembly of a vascular network by human ASCs and endothelial cells and vascularization by these cells of parenchymal cells in implants. Mixture of these 2 readily available, nontransformed human cell types provides a practical approach to tissue engineering, therapeutic revascularization, and in vivo studies of human vasculogenesis.


Asunto(s)
Adipocitos/metabolismo , Comunicación Celular/fisiología , Células Endoteliales/metabolismo , Células Madre Multipotentes/metabolismo , Neovascularización Fisiológica/fisiología , Trasplante de Células Madre , Implantes Absorbibles , Adipocitos/citología , Animales , Colágeno , Células Endoteliales/citología , Humanos , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/trasplante , Ratones , Ratones Endogámicos NOD , Ratones SCID , Células Madre Multipotentes/citología , Células Madre Multipotentes/trasplante , Pericitos/citología , Pericitos/metabolismo , Porcinos , Factores de Tiempo , Ingeniería de Tejidos , Trasplante Heterólogo
20.
Biochim Biophys Acta ; 1796(1): 50-4, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19393290

RESUMEN

Interest in the regulation of blood vessel formation as a mechanism to permit unregulated tumor cell growth was a prescient hypothesis of Dr. Judah Folkman nearly 3 decades ago. Understanding the cellular and molecular mechanisms that affect the recruitment, expansion, and turnover of the tumor microvasculature continues to evolve. While the fundamental paradigms for improving blood flow to growing, injured, diseased, or tumor infiltrated tissues are well known, the potential role of bone marrow derived circulating endothelial progenitor cells (EPCs) to function as postnatal vasculogenic precursors for tumor microvasculature has become a controversial premise. We will briefly review some recently published high profile papers that appear to derive polar interpretations for the role of EPCs in the angiogenic switch and discuss possible reasons for the disparate views in work conducted in both mouse and man.


Asunto(s)
Células de la Médula Ósea/fisiología , Células Endoteliales/fisiología , Neoplasias/irrigación sanguínea , Neoplasias/patología , Neovascularización Patológica , Células Madre/fisiología , Animales , Humanos , Ratones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA