Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
PLoS One ; 16(7): e0252048, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34264955

RESUMEN

Neurofibromatosis Type 2 (NF2) is an autosomal dominant genetic syndrome caused by mutations in the NF2 tumor suppressor gene resulting in multiple schwannomas and meningiomas. There are no FDA approved therapies for these tumors and their relentless progression results in high rates of morbidity and mortality. Through a combination of high throughput screens, preclinical in vivo modeling, and evaluation of the kinome en masse, we identified actionable drug targets and efficacious experimental therapeutics for the treatment of NF2 related schwannomas and meningiomas. These efforts identified brigatinib (ALUNBRIG®), an FDA-approved inhibitor of multiple tyrosine kinases including ALK, to be a potent inhibitor of tumor growth in established NF2 deficient xenograft meningiomas and a genetically engineered murine model of spontaneous NF2 schwannomas. Surprisingly, neither meningioma nor schwannoma cells express ALK. Instead, we demonstrate that brigatinib inhibited multiple tyrosine kinases, including EphA2, Fer and focal adhesion kinase 1 (FAK1). These data demonstrate the power of the de novo unbiased approach for drug discovery and represents a major step forward in the advancement of therapeutics for the treatment of NF2 related malignancies.


Asunto(s)
Neoplasias Meníngeas/genética , Meningioma/genética , Neurilemoma/genética , Neurofibromina 2/deficiencia , Neurofibromina 2/genética , Compuestos Organofosforados/farmacología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Pirimidinas/farmacología , Proliferación Celular , Humanos , Mutación , Neurilemoma/patología
2.
Neurooncol Adv ; 3(1): vdab063, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34131650

RESUMEN

BACKGROUND: Genetically susceptible individuals can develop malignancies after irradiation of normal tissues. In the context of therapeutic irradiation, it is not known whether irradiating benign neoplasms in susceptible individuals promotes neoplastic transformation and worse clinical outcomes. Individuals with Neurofibromatosis 1 (NF1) are susceptible to both radiation-induced second malignancies and spontaneous progression of plexiform neurofibromas (PNs) to malignant peripheral nerve sheath tumors (MPNSTs). The role of radiotherapy in the treatment of benign neoplasms such as PNs is unclear. METHODS: To test whether radiotherapy promotes neoplastic progression of PNs and reduces overall survival, we administered spinal irradiation (SI) to conditional knockout mouse models of NF1-associated PNs in 2 germline contexts: Nf1 fllfl ; PostnCre + and Nf1 fl/- ; PostnCre + . Both genotypes develop extensive Nf1 null spinal PNs, modeling PNs in NF1 patients. A total of 101 mice were randomized to 0 Gy, 15 Gy (3 Gy × 5), or 30 Gy (3 Gy × 10) of spine-focused, fractionated SI and aged until signs of illness. RESULTS: SI decreased survival in both Nf1 fllfl mice and Nf1 fl/- mice, with the worst overall survival occurring in Nf1 fl/- mice receiving 30 Gy. SI was also associated with increasing worrisome histologic features along the PN-MPNST continuum in PNs irradiated to higher radiation doses. CONCLUSIONS: This preclinical study provides experimental evidence that irradiation of pre-existing PNs reduces survival and may shift PNs to higher grade neoplasms.

3.
Nat Med ; 27(1): 165-173, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33442015

RESUMEN

Neurofibromatosis type 1 (NF1) plexiform neurofibromas (PNs) are progressive, multicellular neoplasms that cause morbidity and may transform to sarcoma. Treatment of Nf1fl/fl;Postn-Cre mice with cabozantinib, an inhibitor of multiple tyrosine kinases, caused a reduction in PN size and number and differential modulation of kinases in cell lineages that drive PN growth. Based on these findings, the Neurofibromatosis Clinical Trials Consortium conducted a phase II, open-label, nonrandomized Simon two-stage study to assess the safety, efficacy and biologic activity of cabozantinib in patients ≥16 years of age with NF1 and progressive or symptomatic, inoperable PN ( NCT02101736 ). The trial met its primary outcome, defined as ≥25% of patients achieving a partial response (PR, defined as ≥20% reduction in target lesion volume as assessed by magnetic resonance imaging (MRI)) after 12 cycles of therapy. Secondary outcomes included adverse events (AEs), patient-reported outcomes (PROs) assessing pain and quality of life (QOL), pharmacokinetics (PK) and the levels of circulating endothelial cells and cytokines. Eight of 19 evaluable (42%) trial participants achieved a PR. The median change in tumor volume was 15.2% (range, +2.2% to -36.9%), and no patients had disease progression while on treatment. Nine patients required dose reduction or discontinuation of therapy due to AEs; common AEs included gastrointestinal toxicity, hypothyroidism, fatigue and palmar plantar erythrodysesthesia. A total of 11 grade 3 AEs occurred in eight patients. Patients with PR had a significant reduction in tumor pain intensity and pain interference in daily life but no change in global QOL scores. These data indicate that cabozantinib is active in NF1-associated PN, resulting in tumor volume reduction and pain improvement.


Asunto(s)
Anilidas/uso terapéutico , Neurofibroma Plexiforme/tratamiento farmacológico , Neurofibromatosis 1/tratamiento farmacológico , Piridinas/uso terapéutico , Adolescente , Adulto , Anilidas/efectos adversos , Anilidas/farmacocinética , Animales , Modelos Animales de Enfermedad , Femenino , Genes de Neurofibromatosis 1 , Humanos , Masculino , Ratones , Ratones Mutantes , Neurofibroma Plexiforme/genética , Neurofibroma Plexiforme/patología , Neurofibromatosis 1/genética , Neurofibromatosis 1/patología , Dimensión del Dolor , Estudios Prospectivos , Inhibidores de Proteínas Quinasas/efectos adversos , Inhibidores de Proteínas Quinasas/farmacocinética , Inhibidores de Proteínas Quinasas/uso terapéutico , Piridinas/efectos adversos , Piridinas/farmacocinética , Calidad de Vida , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Investigación Biomédica Traslacional , Adulto Joven
5.
J Biol Chem ; 295(29): 9948-9958, 2020 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-32471868

RESUMEN

Neurofibromatosis type 1 (NF1) is a common cancer predisposition syndrome caused by mutations in the NF1 tumor suppressor gene. NF1 encodes neurofibromin, a GTPase-activating protein for RAS proto-oncogene GTPase (RAS). Plexiform neurofibromas are a hallmark of NF1 and result from loss of heterozygosity of NF1 in Schwann cells, leading to constitutively activated p21RAS. Given the inability to target p21RAS directly, here we performed an shRNA library screen of all human kinases and Rho-GTPases in a patient-derived NF1-/- Schwann cell line to identify novel therapeutic targets to disrupt PN formation and progression. Rho family members, including Rac family small GTPase 1 (RAC1), were identified as candidates. Corroborating these findings, we observed that shRNA-mediated knockdown of RAC1 reduces cell proliferation and phosphorylation of extracellular signal-regulated kinase (ERK) in NF1-/- Schwann cells. Genetically engineered Nf1flox/flox;PostnCre+ mice, which develop multiple PNs, also exhibited increased RAC1-GTP and phospho-ERK levels compared with Nf1flox/flox;PostnCre- littermates. Notably, mice in which both Nf1 and Rac1 loci were disrupted (Nf1flox/floxRac1flox/flox;PostnCre+) were completely free of tumors and had normal phospho-ERK activity compared with Nf1flox/flox ;PostnCre+ mice. We conclude that the RAC1-GTPase is a key downstream node of RAS and that genetic disruption of the Rac1 allele completely prevents PN tumor formation in vivo in mice.


Asunto(s)
Técnicas de Silenciamiento del Gen , Neoplasias Primarias Secundarias , Neurofibroma Plexiforme , Neurofibromatosis 1 , Neuropéptidos/deficiencia , Proteína de Unión al GTP rac1/deficiencia , Animales , Ratones , Ratones Noqueados , Neoplasias Primarias Secundarias/enzimología , Neoplasias Primarias Secundarias/genética , Neoplasias Primarias Secundarias/patología , Neoplasias Primarias Secundarias/prevención & control , Neurofibroma Plexiforme/enzimología , Neurofibroma Plexiforme/genética , Neurofibroma Plexiforme/prevención & control , Neurofibromatosis 1/enzimología , Neurofibromatosis 1/genética , Neurofibromatosis 1/patología , Neurofibromina 1/deficiencia , Neurofibromina 1/metabolismo , Neuropéptidos/metabolismo , Proto-Oncogenes Mas , Proteína de Unión al GTP rac1/metabolismo
6.
Mol Cancer Ther ; 18(12): 2321-2330, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31527226

RESUMEN

Neurofibromatosis Type 1 (NF1) is one of the most common genetic tumor predisposition syndromes in humans. Mutant NF1 results in dysregulated RAS allowing neoplasms throughout the neuroaxis. Plexiform neurofibromas (pNF) afflict up to 50% of patients with NF1. They are complex tumors of the peripheral nerve that cause major morbidity via nerve dysregulation and mortality via conversion to malignant sarcoma. Genetically engineered mouse models (GEMM) of NF1 provide valuable insights for the identification of therapies that have utility in people with pNF. Preclinical studies in GEMMs implicate mast cells and the c-Kit/Kit ligand pathway in pNF tumorigenesis. Kit ligand is a potent chemokine secreted by tumorigenic, Nf1-deficient Schwann cells. Ketotifen is an FDA-approved drug for the treatment of allergic conjunctivitis and asthma that promotes mast cell stabilization and has been used in prior case studies to treat or prevent pNFs. This study investigated the effect of ketotifen on mast cell infiltration and degranulation in the presence and absence of Kit ligand provocation and the effect of ketotifen on shrinking or preventing pNF formation in the Nf1flox/flox ;PostnCre + GEMM. Ketotifen decreased mast cell infiltration in response to exogenous Kit ligand administration, but did not affect mast cell degranulation. Importantly, ketotifen did not reduce mast cells numbers or activity in pNF and did not prevent pNF formation or decrease the volume of established pNF despite administration of pharmacologically active doses. These findings suggest that ketotifen has limited use as monotherapy to prevent or reduce pNF burden in the setting of Nf1 mutations.


Asunto(s)
Quimiotaxis/fisiología , Antagonistas de los Receptores Histamínicos H1/uso terapéutico , Cetotifen/uso terapéutico , Mastocitos/efectos de los fármacos , Neurofibroma/genética , Neurofibromina 1/deficiencia , Animales , Antagonistas de los Receptores Histamínicos H1/farmacología , Cetotifen/farmacología , Ratones , Factor de Células Madre
7.
Free Radic Biol Med ; 97: 212-222, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27266634

RESUMEN

Neurofibromatosis type 1 (NF1) predisposes individuals to early and debilitating cardiovascular disease. Loss of function mutations in the NF1 tumor suppressor gene, which encodes the protein neurofibromin, leads to accelerated p21(Ras) activity and phosphorylation of multiple downstream kinases, including Erk and Akt. Nf1 heterozygous (Nf1(+/-)) mice develop a robust neointima that mimics human disease. Monocytes/macrophages play a central role in NF1 arterial stenosis as Nf1 mutations in myeloid cells alone are sufficient to reproduce the enhanced neointima observed in Nf1(+/-) mice. Though the molecular mechanisms underlying NF1 arterial stenosis remain elusive, macrophages are important producers of reactive oxygen species (ROS) and Ras activity directly regulates ROS production. Here, we use compound mutant and lineage-restricted mice to demonstrate that Nf1(+/-) macrophages produce excessive ROS, which enhance Nf1(+/-) smooth muscle cell proliferation in vitro and in vivo. Further, use of a specific NADPH oxidase-2 inhibitor to limit ROS production prevents neointima formation in Nf1(+/-) mice. Finally, mononuclear cells from asymptomatic NF1 patients have increased oxidative DNA damage, an indicator of chronic exposure to oxidative stress. These data provide genetic and pharmacologic evidence that excessive exposure to oxidant species underlie NF1 arterial stenosis and provide a platform for designing novels therapies and interventions.


Asunto(s)
NADPH Oxidasa 2/genética , Neurofibromatosis 1/genética , Neurofibromina 1/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Animales , Estenosis Carotídea/genética , Estenosis Carotídea/fisiopatología , Proliferación Celular/genética , Daño del ADN/genética , Heterocigoto , Humanos , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , NADPH Oxidasa 2/metabolismo , Neointima/genética , Neointima/metabolismo , Neointima/fisiopatología , Neurofibromatosis 1/metabolismo , Neurofibromatosis 1/fisiopatología , Estrés Oxidativo/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Especies Reactivas de Oxígeno/metabolismo
8.
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
9.
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
10.
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
11.
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
12.
J Clin Invest ; 120(3): 859-70, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20160346

RESUMEN

Neurofibromatosis type 1 (NF1) results from mutations in the NF1 tumor suppressor gene, which encodes the protein neurofibromin. NF1 patients display diverse clinical manifestations, including vascular disease, which results from neointima formation and vessel occlusion. However, the pathogenesis of NF1 vascular disease remains unclear. Vessel wall homeostasis is maintained by complex interactions between vascular and bone marrow-derived cells (BMDCs), and neurofibromin regulates the function of each cell type. Therefore, utilizing cre/lox techniques and hematopoietic stem cell transplantation to delete 1 allele of Nf1 in endothelial cells, vascular smooth muscle cells, and BMDCs alone, we determined which cell lineage is critical for neointima formation in vivo in mice. Here we demonstrate that heterozygous inactivation of Nf1 in BMDCs alone was necessary and sufficient for neointima formation after vascular injury and provide evidence of vascular inflammation in Nf1+/- mice. Further, analysis of peripheral blood from NF1 patients without overt vascular disease revealed increased concentrations of inflammatory cells and cytokines previously linked to vascular inflammation and vasoocclusive disease. These data provide genetic and cellular evidence of vascular inflammation in NF1 patients and Nf1+/- mice and provide a framework for understanding the pathogenesis of NF1 vasculopathy and potential therapeutic and diagnostic interventions.


Asunto(s)
Células Endoteliales/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Neurofibromina 1 , Vasculitis/metabolismo , Adolescente , Adulto , Animales , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Células Endoteliales/patología , Femenino , Trasplante de Células Madre Hematopoyéticas , Humanos , Masculino , Ratones , Ratones Noqueados , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/patología , Neurofibromatosis 1/genética , Neurofibromatosis 1/metabolismo , Neurofibromatosis 1/patología , Vasculitis/genética , Vasculitis/patología
13.
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
14.
Blood ; 112(12): 4646-54, 2008 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-18768391

RESUMEN

Neurofibromatosis type 1 (NF1) is a common genetic disorder caused by mutations in the NF1 locus, which encodes neurofibromin, a negative regulator of Ras. Patients with NF1 develop numerous neurofibromas, which contain many inflammatory mast cells that contribute to tumor formation. Subsequent to c-Kit stimulation, signaling from Ras to Rac1/2 to the MAPK pathway appears to be responsible for multiple hyperactive mast cell phenotypes; however, the specific effectors that mediate these functions remain uncertain. p21-activated kinase 1 (Pak1) is a downstream mediator of Rac1/2 that has been implicated as a positive regulator of MAPK pathway members and is a modulator of cell growth and cytoskeletal dynamics. Using an intercross of Pak 1(-/-) mice with Nf1(+/-) mice, we determined that Pak1 regulates hyperactive Ras-dependent proliferation via a Pak1/Erk pathway, whereas a Pak1/p38 pathway is required for the increased migration in Nf1(+/-) mast cells. Furthermore, we confirmed that loss of Pak1 corrects the dermal accumulation of Nf1(+/-) mast cells in vivo to levels found in wild-type mice. Thus, Pak1 is a novel mast cell mediator that functions as a key node in the MAPK signaling network and potential therapeutic target in NF1 patients.


Asunto(s)
Genes de Neurofibromatosis 1 , Mastocitos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/fisiología , Proteínas Proto-Oncogénicas c-kit/fisiología , Proteínas Proto-Oncogénicas p21(ras)/fisiología , Quinasas p21 Activadas/fisiología , Animales , Biomarcadores/metabolismo , Diferenciación Celular/genética , Proliferación Celular , Células Cultivadas , Genes de Neurofibromatosis 1/fisiología , Heterocigoto , Mastocitos/patología , Mastocitos/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Neurofibromatosis 1/genética , Neurofibromatosis 1/metabolismo , Neurofibromatosis 1/patología , Neurofibromina 1/metabolismo , Fenotipo , Proteínas Proto-Oncogénicas c-kit/genética , Proteínas Proto-Oncogénicas c-kit/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Transducción de Señal/genética , Transducción de Señal/fisiología , Quinasas p21 Activadas/genética , Quinasas p21 Activadas/metabolismo
15.
Hum Mol Genet ; 17(15): 2336-44, 2008 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-18442999

RESUMEN

Neurofibromatosis type I (NF1) is a genetic disorder caused by mutations in the NF1 tumor suppressor gene. Neurofibromin is encoded by NF1 and functions as a negative regulator of Ras activity. Somatic mutations in the residual normal NF1 allele within cancers of NF1 patients is consistent with NF1 functioning as a tumor-suppressor. However, the prevalent non-malignant manifestations of NF1, including learning and bone disorders emphasize the importance of dissecting the cellular and biochemical effects of NF1 haploinsufficiency in multiple cell lineages. One of the least studied complications of NF1 involves cardiovascular disorders, including arterial occlusions that result in cerebral and visceral infarcts. NF1 vasculopathy is characterized by vascular smooth muscle cell (VSMC) accumulation in the intima area of vessels resulting in lumen occlusion. We recently showed that Nf1 haploinsufficiency increases VSMC proliferation and migration via hyperactivation of the Ras-Erk pathway, which is a signaling axis directly linked to neointima formation in diverse animal models of vasculopathy. Given this observation, we tested whether heterozygosity of Nf1 would lead to vaso-occlusive disease in genetically engineered mice in vivo. Strikingly, Nf1+/- mice have increased neointima formation, excessive vessel wall cell proliferation and Erk activation after vascular injury in vivo. Further, this effect is directly dependent on a Gleevec sensitive molecular pathway. Therefore, these studies establish an Nf1 model of vasculopathy, which mirrors features of human NF1 vaso-occlusive disease, identifies a potential therapeutic target and provides a platform to further dissect the effect of Nf1 haploinsufficiency in cardiovascular disease.


Asunto(s)
Arteriopatías Oclusivas/genética , Enfermedades Cardiovasculares/genética , Trastornos Cerebrovasculares/genética , Resistencia a Antineoplásicos/genética , Genes de Neurofibromatosis 1 , Neurofibromatosis 1/complicaciones , Animales , Antineoplásicos/farmacología , Arteriopatías Oclusivas/etiología , Arteriopatías Oclusivas/patología , Benzamidas , Enfermedades Cardiovasculares/etiología , Enfermedades Cardiovasculares/patología , Arterias Carótidas/efectos de los fármacos , Arterias Carótidas/patología , Proliferación Celular , Trastornos Cerebrovasculares/etiología , Trastornos Cerebrovasculares/patología , Modelos Animales de Enfermedad , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Mesilato de Imatinib , Ratones , Ratones Mutantes , Neurofibromatosis 1/genética , Neurofibromatosis 1/patología , Piperazinas/farmacología , Pirimidinas/farmacología , Túnica Íntima/efectos de los fármacos , Túnica Íntima/patología
16.
Pediatr Res ; 64(1): 68-73, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18360305

RESUMEN

Endothelial progenitor cells (EPCs) are used for angiogenic therapies and as biomarkers of cardiovascular disease. Human umbilical cord blood (UCB) is a rich source of endothelial colony forming cells (ECFCs), which are EPCs with robust proliferative potential that may be useful for clinical vascular regeneration. Previous studies show that hematopoietic progenitor cells are increased in premature UCB compared with term controls. Based on this paradigm, we hypothesized that premature UCB would be an enriched source of ECFCs. Thirty-nine UCB samples were obtained from premature infants (24-37 wk gestational age (GA)) and term controls. ECFC colonies were enumerated, clonally isolated, and identified by expression of endothelial cell surface antigens and functional analysis. GA of 33-36 wk UCB yielded predominantly ECFC colonies at equivalent numbers to term infants. UCB from 24 to 28 wk GA infants had significantly fewer ECFCs. Surprisingly, 24-28 wk GA UCB yielded predominantly mesenchymal stem cell (MSC) colonies, capable of differentiating into adipocytes, chondrocytes, and osteocytes. MSCs were rarely identified in 37-40 wk GA UCB. These studies demonstrate that circulating MSCs and ECFCs appear at different GA in the human UCB, and that 24-28 wk GA UCB may be a novel source of MSCs for therapeutic use in human diseases.


Asunto(s)
Células Endoteliales/fisiología , Sangre Fetal/citología , Células Madre Fetales/fisiología , Células Madre Mesenquimatosas/fisiología , Adipocitos/fisiología , Antígenos CD/análisis , Diferenciación Celular , Proliferación Celular , Forma de la Célula , Células Cultivadas , Condrocitos/fisiología , Células Endoteliales/inmunología , Células Madre Fetales/inmunología , Edad Gestacional , Humanos , Recién Nacido , Recien Nacido Prematuro , Células Madre Mesenquimatosas/inmunología , Neovascularización Fisiológica , Osteocitos/fisiología , Fenotipo
17.
Exp Hematol ; 35(7): 1109-18, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17588480

RESUMEN

OBJECTIVE: Endothelial progenitor cells (EPCs) are used for angiogenic therapies or as biomarkers to assess cardiovascular disease risk. However, there is no uniform definition of an EPC, which confounds EPC studies. EPCs are widely described as cells that coexpress the cell-surface antigens CD34, AC133, and vascular endothelial growth factor receptor-2 (VEGFR-2). These antigens are also expressed on primitive hematopoietic progenitor cells (HPCs). Remarkably, despite their original identification, CD34+AC133+VEGFR-2+ cells have never been isolated and simultaneously plated in hematopoietic and endothelial cell (EC) clonogenic assays to assess the identity of their clonal progeny, which are presumably the cellular participants in vascular regeneration. METHODS: CD34+AC133+VEGFR-2+ cells were isolated from human umbilical cord blood (CB) or granulocyte colony-stimulating factor-mobilized peripheral blood and assayed for either EPCs or HPCs. RESULTS: CD34+AC133+VEGFR-2+ cells did not form EPCs and were devoid of vessel forming activity. However, CD34+AC133+VEGFR-2+ cells formed HPCs and expressed the hematopoietic lineage-specific antigen, CD45. We next tested whether EPCs could be separated from HPCs by immunoselection for CD34 and CD45. CD34+CD45+ cells formed HPCs but not EPCs, while CD34+CD45- cells formed EPCs but not HPCs. CONCLUSIONS: Therefore, CD34+AC133+VEGFR-2+ cells are HPCs that do not yield EC progeny, and the biological mechanism for their correlation with cardiovascular disease needs to be reexamined.


Asunto(s)
Antígenos CD34/análisis , Antígenos CD/análisis , Células Endoteliales/citología , Glicoproteínas/análisis , Células Madre Hematopoyéticas/citología , Péptidos/análisis , Células Madre/citología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/análisis , Antígeno AC133 , Adulto , Separación Celular , Células Cultivadas , Femenino , Humanos , Antígenos Comunes de Leucocito/análisis , Masculino , Persona de Mediana Edad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA