Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
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
2.
Hum Mol Genet ; 28(16): 2752-2762, 2019 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-31091306

RESUMEN

Plexiform neurofibroma (PN) tumors are a hallmark manifestation of neurofibromatosis type 1 (NF1) that arise in the Schwann cell (SC) lineage. NF1 is a common heritable cancer predisposition syndrome caused by germline mutations in the NF1 tumor suppressor, which encodes a GTPase-activating protein called neurofibromin that negatively regulates Ras proteins. Whereas most PN are clinically indolent, a subset progress to atypical neurofibromatous neoplasms of uncertain biologic potential (ANNUBP) and/or to malignant peripheral nerve sheath tumors (MPNSTs). In small clinical series, loss of 9p21.3, which includes the CDKN2A locus, has been associated with the genesis of ANNUBP. Here we show that the Cdkn2a alternate reading frame (Arf) serves as a gatekeeper tumor suppressor in mice that prevents PN progression by inducing senescence-mediated growth arrest in aberrantly proliferating Nf1-/- SC. Conditional ablation of Nf1 and Arf in the neural crest-derived SC lineage allows escape from senescence, resulting in tumors that accurately phenocopy human ANNUBP and progress to MPNST with high penetrance. This animal model will serve as a platform to study the clonal development of ANNUBP and MPNST and to identify new therapies to treat existing tumors and to prevent disease progression.


Asunto(s)
Transformación Celular Neoplásica/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/deficiencia , Neurofibroma/genética , Neurofibroma/patología , Neurofibromatosis 1/genética , Animales , Biomarcadores de Tumor , Línea Celular Tumoral , Transformación Celular Neoplásica/metabolismo , Senescencia Celular/genética , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Genotipo , Xenoinjertos , Humanos , Inmunohistoquímica , Ratones , Mutación , Neoplasias de la Vaina del Nervio/genética , Neoplasias de la Vaina del Nervio/metabolismo , Neoplasias de la Vaina del Nervio/patología , Neurofibroma/metabolismo , Neurofibroma/mortalidad , Neurofibromatosis 1/metabolismo , Células de Schwann/metabolismo , Células de Schwann/patología , Proteínas ras/metabolismo
3.
Hum Mol Genet ; 28(4): 572-583, 2019 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-30335132

RESUMEN

Schwannomas are common, highly morbid and medically untreatable tumors that can arise in patients with germ line as well as somatic mutations in neurofibromatosis type 2 (NF2). These mutations most commonly result in the loss of function of the NF2-encoded protein, Merlin. Little is known about how Merlin functions endogenously as a tumor suppressor and how its loss leads to oncogenic transformation in Schwann cells (SCs). Here, we identify nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB)-inducing kinase (NIK) as a potential drug target driving NF-κB signaling and Merlin-deficient schwannoma genesis. Using a genomic approach to profile aberrant tumor signaling pathways, we describe multiple upregulated NF-κB signaling elements in human and murine schwannomas, leading us to identify a caspase-cleaved, proteasome-resistant NIK kinase domain fragment that amplifies pathogenic NF-κB signaling. Lentiviral-mediated transduction of this NIK fragment into normal SCs promotes proliferation, survival, and adhesion while inducing schwannoma formation in a novel in vivo orthotopic transplant model. Furthermore, we describe an NF-κB-potentiated hepatocyte growth factor (HGF) to MET proto-oncogene receptor tyrosine kinase (c-Met) autocrine feed-forward loop promoting SC proliferation. These innovative studies identify a novel signaling axis underlying schwannoma formation, revealing new and potentially druggable schwannoma vulnerabilities with future therapeutic potential.


Asunto(s)
Neurilemoma/genética , Neurofibromatosis 2/genética , Neurofibromina 2/genética , Proteínas Serina-Treonina Quinasas/genética , Animales , Comunicación Autocrina/genética , Carcinogénesis/genética , Caspasa 1/genética , Proliferación Celular/genética , Modelos Animales de Enfermedad , Regulación Neoplásica de la Expresión Génica , Factor de Crecimiento de Hepatocito/genética , Humanos , Ratones , Terapia Molecular Dirigida , FN-kappa B/genética , Neurilemoma/complicaciones , Neurilemoma/tratamiento farmacológico , Neurilemoma/patología , Neurofibromatosis 2/complicaciones , Neurofibromatosis 2/tratamiento farmacológico , Neurofibromatosis 2/patología , Complejo de la Endopetidasa Proteasomal/genética , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas c-met/genética , Células de Schwann , Transducción de Señal/genética , Quinasa de Factor Nuclear kappa B
4.
Haematologica ; 102(6): 1017-1027, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28341737

RESUMEN

Fanconi anemia is a complex heterogeneous genetic disorder with a high incidence of bone marrow failure, clonal evolution to acute myeloid leukemia and mesenchymal-derived congenital anomalies. Increasing evidence in Fanconi anemia and other genetic disorders points towards an interdependence of skeletal and hematopoietic development, yet the impact of the marrow microenvironment in the pathogenesis of the bone marrow failure in Fanconi anemia remains unclear. Here we demonstrated that mice with double knockout of both Fancc and Fancg genes had decreased bone formation at least partially due to impaired osteoblast differentiation from mesenchymal stem/progenitor cells. Mesenchymal stem/progenitor cells from the double knockout mice showed impaired hematopoietic supportive activity. Mesenchymal stem/progenitor cells of patients with Fanconi anemia exhibited similar cellular deficits, including increased senescence, reduced proliferation, impaired osteoblast differentiation and defective hematopoietic stem/progenitor cell supportive activity. Collectively, these studies provide unique insights into the physiological significance of mesenchymal stem/progenitor cells in supporting the marrow microenvironment, which is potentially of broad relevance in hematopoietic stem cell transplantation.


Asunto(s)
Médula Ósea/patología , Microambiente Celular , Anemia de Fanconi/patología , Animales , Huesos/anomalías , Huesos/fisiopatología , Linaje de la Célula , Anemia de Fanconi/fisiopatología , Proteína del Grupo de Complementación C de la Anemia de Fanconi/genética , Hematopoyesis , Trasplante de Células Madre Hematopoyéticas , Células Madre Mesenquimatosas/patología , Ratones , Ratones Noqueados
6.
Blood ; 123(4): 541-53, 2014 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-24255920

RESUMEN

ASXL1 is mutated/deleted with high frequencies in multiple forms of myeloid malignancies, and its alterations are associated with poor prognosis. De novo ASXL1 mutations cause Bohring-Opitz syndrome characterized by multiple congenital malformations. We show that Asxl1 deletion in mice led to developmental abnormalities including dwarfism, anophthalmia, and 80% embryonic lethality. Surviving Asxl1(-/-) mice lived for up to 42 days and developed features of myelodysplastic syndrome (MDS), including dysplastic neutrophils and multiple lineage cytopenia. Asxl1(-/-) mice had a reduced hematopoietic stem cell (HSC) pool, and Asxl1(-/-) HSCs exhibited decreased hematopoietic repopulating capacity, with skewed cell differentiation favoring granulocytic lineage. Asxl1(+/-) mice also developed mild MDS-like disease, which could progress to MDS/myeloproliferative neoplasm, demonstrating a haploinsufficient effect of Asxl1 in the pathogenesis of myeloid malignancies. Asxl1 loss led to an increased apoptosis and mitosis in Lineage(-)c-Kit(+) (Lin(-)c-Kit(+)) cells, consistent with human MDS. Furthermore, Asxl1(-/-) Lin(-)c-Kit(+) cells exhibited decreased global levels of H3K27me3 and H3K4me3 and altered expression of genes regulating apoptosis (Bcl2, Bcl2l12, Bcl2l13). Collectively, we report a novel ASXL1 murine model that recapitulates human myeloid malignancies, implying that Asxl1 functions as a tumor suppressor to maintain hematopoietic cell homeostasis. Future work is necessary to clarify the contribution of microenvironment to the hematopoietic phenotypes observed in the constitutional Asxl1(-/-) mice.


Asunto(s)
Mutación , Síndromes Mielodisplásicos/genética , Proteínas Represoras/genética , Proteínas Represoras/fisiología , Animales , Apoptosis , Células de la Médula Ósea/citología , Diferenciación Celular , Linaje de la Célula , Proliferación Celular , Cruzamientos Genéticos , Modelos Animales de Enfermedad , Eliminación de Gen , Proteínas Fluorescentes Verdes/metabolismo , Hematopoyesis , Células Madre Hematopoyéticas/citología , Homeostasis , Homocigoto , Humanos , Ratones , Ratones Transgénicos , Mitosis , Síndromes Mielodisplásicos/metabolismo , Fenotipo
7.
Hum Mol Genet ; 22(23): 4818-28, 2013 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-23863460

RESUMEN

Neurofibromatosis type 1 (NF1) is a common genetic disorder affecting 1 in 3500 individuals. Patients with NF1 are predisposed to debilitating skeletal manifestations, including osteopenia/osteoporosis and long bone pseudarthrosis (nonunion fracture). Hyperactivation of the Ras/mitogen-activated protein kinase (MAPK) pathway in NF1 is known to underlie aberrant proliferation and differentiation in cell lineages, including osteoclast progenitors and mesenchymal stem cells (MSCs) also known as osteoblast progenitors (pro-OBLs). Our current study demonstrates the hyper Ras/MAPK as a critical pathway underlying the pathogenesis of NF1-associated fracture repair deficits. Nf1-deficient pro-OBLs exhibit Ras/MAPK hyperactivation. Introduction of the NF1 GTPase activating-related domain (NF1 GAP-related domain) in vitro is sufficient to rescue hyper Ras activity and enhance osteoblast (OBL) differentiation in Nf1(-/-) pro-OBLs and NF1 human (h) MSCs cultured from NF1 patients with skeletal abnormalities, including pseudarthrosis or scoliosis. Pharmacologic inhibition of mitogen-activated protein kinase kinase (MEK) signaling with PD98059 partially rescues aberrant Erk activation while enhancing OBL differentiation and expression of OBL markers, osterix and osteocalcin, in Nf1-deficient murine pro-OBLs. Similarly, MEK inhibition enhances OBL differentiation of hMSCs. In addition, PD98059 rescues aberrant osteoclast maturation in Nf1 haploinsufficient bone marrow mononuclear cells (BMMNCs). Importantly, MEK inhibitor significantly improves fracture healing in an NF1 murine model, Col2.3Cre;Nf1(flox/-). Collectively, these data indicate the Ras/MAPK cascade as a critical pathway in the pathogenesis of bone loss and pseudarthrosis related to NF1 mutations. These studies provide evidence for targeting the MAPK pathway to improve bone mass and treat pseudarthrosis in NF1.


Asunto(s)
Proteínas Quinasas Activadas por Mitógenos/metabolismo , Neurofibromatosis 1/metabolismo , Neurofibromina 1/deficiencia , Seudoartrosis/fisiopatología , Transducción de Señal/fisiología , Proteínas ras/metabolismo , Animales , Linaje de la Célula , Células Cultivadas , Modelos Animales de Enfermedad , Flavonoides/farmacología , Humanos , Ratones , Ratones Transgénicos , Neurofibromatosis 1/genética , Neurofibromatosis 1/patología , Neurofibromina 1/genética , Neurofibromina 1/metabolismo , Osteoblastos/fisiología , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Seudoartrosis/tratamiento farmacológico , Seudoartrosis/genética , Seudoartrosis/patología , Transducción de Señal/efectos de los fármacos , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Fracturas de la Tibia/fisiopatología
8.
Int J Mol Sci ; 16(6): 12345-59, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-26039236

RESUMEN

Neurofibromatosis type 1 (NF1) is an autosomal dominant disease caused by mutations in the NF1 tumor suppressor gene, which affect approximately 1 out of 3000 individuals. Patients with NF1 suffer from a range of malignant and nonmalignant manifestations such as plexiform neurofibromas and skeletal abnormalities. We previously demonstrated that Nf1 haploinsufficiency in mesenchymal stem/progenitor cells (MSPCs) results in impaired osteoblastic differentiation, which may be associated with the skeletal manifestations in NF1 patients. Here we sought to further ascertain the role of Nf1 in modulating the migration and adhesion of MSPCs of the Nf1 haploinsufficient (Nf1(+/-)) mice. Nf1(+/-) MSPCs demonstrated increased nuclear-cytoplasmic ratio, increased migration, and increased actin polymerization as compared to wild-type (WT) MSPCs. Additionally, Nf1(+/-) MSPCs were noted to have significantly enhanced cell adhesion to fibronectin with selective affinity for CH271 with an overexpression of its complimentary receptor, CD49e. Nf1(+/-) MSPCs also showed hyperactivation of phosphoinositide 3-kinase (PI3-K) and mitogen activated protein kinase (MAPK) signaling pathways when compared to WT MSPCs, which were both significantly reduced in the presence of their pharmacologic inhibitors, LY294002 and PD0325901, respectively. Collectively, our study suggests that both PI3-K and MAPK signaling pathways play a significant role in enhanced migration and adhesion of Nf1 haploinsufficient MSPCs.


Asunto(s)
Haploinsuficiencia , Sistema de Señalización de MAP Quinasas , Células Madre Mesenquimatosas/fisiología , Neurofibromina 1/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Actinas/metabolismo , Animales , Benzamidas/farmacología , Adhesión Celular , Movimiento Celular , Células Cultivadas , Cromonas/farmacología , Difenilamina/análogos & derivados , Difenilamina/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Morfolinas/farmacología , Neurofibromina 1/genética , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas ras/metabolismo
9.
J Biol Chem ; 287(16): 12858-66, 2012 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-22362783

RESUMEN

Prion diseases are a group of rare, fatal neurodegenerative disorders associated with a conformational transformation of the cellular prion protein (PrP(C)) into a self-replicating and proteinase K-resistant conformer, termed scrapie PrP (PrP(Sc)). Aggregates of PrP(Sc) deposited around neurons lead to neuropathological alterations. Currently, there is no effective treatment for these fatal illnesses; thus, the development of an effective therapy is a priority. PrP peptide-based ELISA assay methods were developed for detection and immunoaffinity chromatography capture was developed for purification of naturally occurring PrP peptide autoantibodies present in human CSF, individual donor serum, and commercial preparations of pooled intravenous immunoglobulin (IVIg). The ratio of anti-PrP autoantibodies (PrP-AA) to total IgG was ∼1:1200. The binding epitope of purified PrP-AA was mapped to an N-terminal region comprising the PrP amino acid sequence KTNMK. Purified PrP-AA potently blocked fibril formation by a toxic 21-amino acid fragment of the PrP peptide containing the amino acid alanine to valine substitution corresponding to position 117 of the full-length peptide (A117V). Furthermore, PrP-AA attenuated the neurotoxicity of PrP(A117V) and wild-type peptides in rat cerebellar granule neuron (CGN) cultures. In contrast, IgG preparations depleted of PrP-AA had little effect on PrP fibril formation or PrP neurotoxicity. The specificity of PrP-AA was demonstrated by immunoprecipitating PrP protein in brain tissues of transgenic mice expressing the human PrP(A117V) epitope and Sc237 hamster. Based on these intriguing findings, it is suggested that human PrP-AA may be useful for interfering with the pathogenic effects of pathogenic prion proteins and, thereby has the potential to be an effective means for preventing or attenuating human prion disease progression.


Asunto(s)
Amiloide/inmunología , Anticuerpos Bloqueadores/farmacología , Autoanticuerpos/farmacología , Proteínas PrPC/inmunología , Proteínas PrPSc/inmunología , Enfermedades por Prión , Animales , Anticuerpos Bloqueadores/inmunología , Especificidad de Anticuerpos , Autoanticuerpos/inmunología , Cricetinae , Mapeo Epitopo , Epítopos , Heterocigoto , Humanos , Inmunoglobulinas Intravenosas/farmacología , Ratones , Ratones Transgénicos , Neuroglía/citología , Neuroglía/inmunología , Neuroglía/patología , Neuronas/citología , Neuronas/inmunología , Neuronas/patología , Proteínas PrPC/genética , Proteínas PrPC/metabolismo , Proteínas PrPSc/genética , Proteínas PrPSc/metabolismo , Cultivo Primario de Células , Enfermedades por Prión/inmunología , Enfermedades por Prión/prevención & control , Enfermedades por Prión/terapia , Ratas , Ratas Sprague-Dawley
10.
Crit Rev Eukaryot Gene Expr ; 22(3): 205-18, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23140162

RESUMEN

Chronic degenerative diseases are increasing with the aging U.S. population. One consequence of this phenomenon is the need for long-term osteoporosis therapies. Parathyroid hormone (PTH), the only FDA-approved treatment that adds bone to the aged skeleton, loses its potency within two years of initial treatment but the mechanism regulating its limited "anabolic window" is unknown. We have discovered that disabling the nucleocytoplasmic shuttling transcription factor nuclear matrix protein 4/cas interacting zinc finger protein (Nmp4/CIZ) in mice extends the PTH bone-forming capacity. Nmp4 was discovered during our search for nuclear matrix transcription factors that couple this hormone's impact on osteoblast cytoskeletal and nuclear organization with its anabolic capacity. CIZ was independently discovered as a protein that associates with the focal adhesion-associated mechanosensor p130Cas. The Nmp4/CIZ-knockout (KO) skeletal phenotype exhibits a modestly enhanced bone mineral density but manifests an exaggerated response to both PTH and to BMP2 and is resistant to disuse-induced bone loss. The cellular basis of the global Nmp4/CIZ-KO skeletal phenotype remains to be elucidated but may involve an expansion of the bone marrow osteoprogenitor population along with modestly enhanced osteoblast and osteoclast activities supporting anabolic bone turnover. As a shuttling Cys(2)His(2) zinc finger protein, Nmp4/CIZ acts as a repressive transcription factor perhaps associated with epigenetic remodeling complexes, but the functional significance of its interaction with p130Cas is not known. Despite numerous remaining questions, Nmp4/CIZ provides insights into how the anabolic window is regulated, and itself may provide an adjuvant therapy target for the treatment of osteoporosis by extending PTH anabolic efficacy.


Asunto(s)
Huesos/fisiología , Proteínas Asociadas a Matriz Nuclear/metabolismo , Hormona Paratiroidea/fisiología , Factores de Transcripción/metabolismo , Animales , Densidad Ósea , Proteína Morfogenética Ósea 2/genética , Proteína Morfogenética Ósea 2/metabolismo , Adhesión Celular , Proteína Sustrato Asociada a CrK/genética , Proteína Sustrato Asociada a CrK/metabolismo , Regulación de la Expresión Génica , Técnicas de Inactivación de Genes , Humanos , Ratones , Proteínas Asociadas a Matriz Nuclear/genética , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Hormona Paratiroidea/farmacología , Fenotipo , Factores de Transcripción/genética , Dedos de Zinc/genética
11.
Am J Med Genet A ; 155A(5): 1050-9, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21465658

RESUMEN

Skeletal abnormalities including scoliosis, tibial dysplasia, sphenoid wing dysplasia, and decreased bone mineral density (BMD) are associated with neurofibromatosis type 1 (NF1). We report the cellular phenotype of NF1 human-derived osteoclasts and compare the in vitro findings with the clinical phenotype. Functional characteristics (e.g., osteoclast formation, migration, adhesion, resorptive capacity) and cellular mechanistic alterations (e.g., F-actin polymerization, MAPK phosphorylation, RhoGTPase activity) from osteoclasts cultured from peripheral blood of individuals with NF1 (N = 75) were assessed. Osteoclast formation was compared to phenotypic, radiologic, and biochemical data. NF1 osteoprogenitor cells demonstrated increased osteoclast forming capacity. Human NF1-derived osteoclasts demonstrated increased migration, adhesion, and in vitro bone resorption. These activities coincided with increased actin belt formation and hyperactivity in MAPK and RhoGTPase pathways. Although osteoclast formation was increased, no direct correlation of osteoclast formation with BMD, markers of bone resorption, or the clinical skeletal phenotype was observed suggesting that osteoclast formation in vitro cannot directly predict NF1 skeletal phenotypes. While NF1 haploinsufficiency produces a generalized osteoclast gain-in-function and may contribute to increased bone resorption, reduced BMD, and focal skeletal defects associated with NF1, additional and perhaps local modifiers are likely required for the development of skeletal abnormalities in NF1.


Asunto(s)
Neurofibromatosis 1/patología , Osteoclastos/citología , Absorciometría de Fotón , Adolescente , Adulto , Western Blotting , Adhesión Celular , Proliferación Celular , Células Cultivadas , Niño , Preescolar , Activación Enzimática , GTP Fosfohidrolasas/metabolismo , Humanos , Lactante , Microscopía Fluorescente , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Osteoclastos/enzimología
12.
Exp Mol Pathol ; 91(3): 745-52, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21840308

RESUMEN

Clinical trials of suicide gene therapy have achieved limited success, which suggests a need for improvement. Angiogenesis plays a crucial role in the progression of cancers, which is greatly regulated by vascular endothelial growth factor (VEGF).The current study was designed to evaluate the anti-tumor effects of VEGF siRNA in combination with fusion suicide gene yCDglyTK. Introduction of a VEGF-targeted small hairpin RNA (shVEGF) to CDTK/5-FC system could induce cell apoptosis more effectively and decrease micro vessel density in xenograft tissue, thus resulted in a significant tumor growth delay in SGC7901 xenografts. These findings for the first time suggest the potential of combination gene therapy using suicide gene therapy and anti-angiogenesis gene therapy.


Asunto(s)
Genes Transgénicos Suicidas , Terapia Genética/métodos , Neovascularización Patológica/genética , Neovascularización Patológica/terapia , ARN Interferente Pequeño , Neoplasias Gástricas/genética , Neoplasias Gástricas/terapia , Factor A de Crecimiento Endotelial Vascular/genética , Animales , Apoptosis/genética , Línea Celular Tumoral , Femenino , Fusión Génica , Humanos , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Interferencia de ARN , Neoplasias Gástricas/patología , Ensayos Antitumor por Modelo de Xenoinjerto
13.
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
14.
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
15.
Stem Cells ; 27(2): 478-88, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19023032

RESUMEN

Adipose tissue stroma contains a population of mesenchymal stem cells, which support repair when administered to damaged tissues, in large part through secreted trophic factors. We directly tested the ability of media collected from cultured adipose-derived stem cells (ASCs) to protect neurons in a rat model of brain hypoxic-ischemic (HI) injury. Concentrated conditioned medium from cultured rat ASCs (ASC-CM) or control medium was infused through the jugular vein of neonatal Sprague-Dawley rats subjected to HI injury. The ASC-CM was administered either 1 hour before or 24 hours after induction of injury. Analysis at 1 week indicated that administration at both time points significantly protected against hippocampal and cortical volume loss. Analysis of parallel groups for behavioral and learning changes at 2 months postischemia demonstrated that both treated groups performed significantly better than the controls in Morris water maze functional tests. Subsequent post-mortem evaluation of brain damage at the 2-month time point confirmed neuronal loss to be similar to that observed at 1 week for all groups. We have identified several neurotrophic factors in ASC-CM, particularly insulin-like growth factor-1 and brain-derived neurotrophic factor, which are important factors that could contribute to the protective effects of ASCs observed in studies with both in vitro and in vivo neuronal injury models. These data suggest that delivery of the milieu of factors secreted by ASCs may be a viable therapeutic option for treatment of HI, as well as other brain injuries.


Asunto(s)
Tejido Adiposo/citología , Encéfalo/efectos de los fármacos , Medios de Cultivo Condicionados/farmacología , Hipoxia-Isquemia Encefálica/prevención & control , Células del Estroma/fisiología , Animales , Animales Recién Nacidos , Células Cultivadas , Femenino , Humanos , Hipoxia-Isquemia Encefálica/patología , Aprendizaje por Laberinto , Embarazo , Ratas , Ratas Sprague-Dawley , Células del Estroma/metabolismo
16.
JCI Insight ; 5(20)2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-32960816

RESUMEN

Schwannomas are tumors of the Schwann cells that cause chronic pain, numbness, and potentially life-threatening impairment of vital organs. Despite the identification of causative genes, including NF2 (Merlin), INI1/SMARCB1, and LZTR1, the exact molecular mechanism of schwannoma development is still poorly understood. Several studies have identified Merlin as a key regulator of the Hippo, MAPK, and PI3K signaling pathways; however, definitive evidence demonstrating the importance of these pathways in schwannoma pathogenesis is absent. Here, we provide direct genetic evidence that dysregulation of the Hippo pathway in the Schwann cell lineage causes development of multiple schwannomas in mice. We found that canonical Hippo signaling through the effectors YAP/TAZ is required for schwannomagenesis and that MAPK signaling modifies schwannoma formation. Furthermore, cotargeting YAP/TAZ transcriptional activity and MAPK signaling demonstrated a synergistic therapeutic effect on schwannomas. Our new model provides a tractable platform to dissect the molecular mechanisms underpinning schwannoma formation and the role of combinatorial targeted therapy in schwannoma treatment.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Pérdida de Heterocigocidad/genética , Neurilemoma/genética , Neurofibromina 2/genética , Animales , Regulación Neoplásica de la Expresión Génica/genética , Factor de Crecimiento de Hepatocito/genética , Humanos , Ratones , Quinasas de Proteína Quinasa Activadas por Mitógenos/genética , Neurilemoma/patología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas/genética , Proteína SMARCB1/genética , Transducción de Señal/genética , Factores de Transcripción/genética , Proteínas Señalizadoras YAP , Proteínas ras/genética
17.
Exp Mol Pathol ; 87(1): 48-53, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19341727

RESUMEN

Smad signaling pathway plays an important role in tumorigenesis and progression in cancer (Halder, S.K., Rachakonda, G., Deane, N.G., Datta, P.K., 2008. Smad7 induces hepatic metastasis in colorectal cancer. Br. J. Cancer 99, 957-965). The protein level of Smad is associated with growth, inhibition, and metastasis in different cancers. It is unclear if the differentiation, metastasis and apoptosis are reduced by Smad expression pattern in gastric cancer. To determine the effect of Smad on gastric cancer cells, we investigated the relationship of Smad4/Smad7 expression, and differentiation, metastasis, and apoptosis in different gastric cancer. The results show that Smad4 expression in the gastric cancer tissue was dramatically lower than that in the peritumoral tissue. A lower expression of Samd4 was significantly lower in the poorly differentiated tissue than that in the well and middle differentiated tissues (P<0.01). In contrast, Smad7 expression in gastric cancer tissues was significantly higher than that in the peritumoral tissue. Smad7 was overexpressed in poorly differentiated tissue, also higher than those in the middle, and well differentiated tissues (P<0.05). The Smad4 or Smad7 expression obviously related with the lymphatic metastasis in gastric cancer. There were 45 cases with lymphatic metastasis in all 78 patients. Smad4 expression in the cases with lymphatic metastasis was lower than the cases without metastasis (P<0.01), whereas Smad7 expression in the cases with lymphatic metastasis was much higher than the case without metastasis (P<0.01). To better understand the mechanisms involved in tumorigenesis of gastric cancer, we established SGC7901 gastric cancer cell lines transduced with Smad4 or Smad7 plasmid DNA. Apoptosis and survival of cancer cells was induced after Smad4 and Smad7 transduction. This effect is concentration and time dependent. Thus, this study provides a mechanism by which a balance between Smad4 and Smad7 in human gastric cancer is critical for differentiation, metastasis, and apoptosis of tumor cells.


Asunto(s)
Transducción de Señal/fisiología , Proteína Smad4/metabolismo , Proteína smad7/metabolismo , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Animales , Apoptosis/fisiología , Diferenciación Celular/fisiología , Línea Celular , Mucosa Gástrica/metabolismo , Humanos , Neoplasias Hepáticas/secundario , Metástasis Linfática , Proteína Smad4/genética , Proteína smad7/genética , Estómago/patología , Neoplasias Gástricas/genética
18.
Oncotarget ; 9(1): 718-725, 2018 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-29416648

RESUMEN

Mutations in the tumor suppressor gene NF2 lead to Neurofibromatosis type 2 (NF2), a tumor predisposition syndrome characterized by the development of schwannomas, including bilateral vestibular schwannomas with complete penetrance. Recent work has implicated the importance of COX-2 in schwannoma growth. Using a genetically engineered murine model of NF2, we demonstrate that selective inhibition of COX-2 with celecoxib fails to prevent the spontaneous development of schwannomas or sensorineural hearing loss in vivo, despite elevated expression levels of COX-2 in Nf2-deficient tumor tissue. These results suggest that COX-2 is nonessential to schwannomagenesis and that the proposed tumor suppressive effects of NSAIDs on schwannomas may occur through COX-2 independent mechanisms.

19.
Endocrinology ; 158(9): 2722-2740, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28637206

RESUMEN

Combining anticatabolic agents with parathyroid hormone (PTH) to enhance bone mass has yielded mixed results in osteoporosis patients. Toward the goal of enhancing the efficacy of these regimens, we tested their utility in combination with loss of the transcription factor Nmp4 because disabling this gene amplifies PTH-induced increases in trabecular bone in mice by boosting osteoblast secretory activity. We addressed whether combining a sustained anabolic response with an anticatabolic results in superior bone acquisition compared with PTH monotherapy. Additionally, we inquired whether Nmp4 interferes with anticatabolic efficacy. Wild-type and Nmp4-/- mice were ovariectomized at 12 weeks of age, followed by therapy regimens, administered from 16 to 24 weeks, and included individually or combined PTH, alendronate (ALN), zoledronate (ZOL), and raloxifene (RAL). Anabolic therapeutic efficacy generally corresponded with PTH + RAL = PTH + ZOL > PTH + ALN = PTH > vehicle control. Loss of Nmp4 enhanced femoral trabecular bone increases under PTH + RAL and PTH + ZOL. RAL and ZOL promoted bone restoration, but unexpectedly, loss of Nmp4 boosted RAL-induced increases in femoral trabecular bone. The combination of PTH, RAL, and loss of Nmp4 significantly increased bone marrow osteoprogenitor number, but did not affect adipogenesis or osteoclastogenesis. RAL, but not ZOL, increased osteoprogenitors in both genotypes. Nmp4 status did not influence bone serum marker responses to treatments, but Nmp4-/- mice as a group showed elevated levels of the bone formation marker osteocalcin. We conclude that the heightened osteoanabolism of the Nmp4-/- skeleton enhances the effectiveness of diverse osteoporosis treatments, in part by increasing hyperanabolic osteoprogenitors. Nmp4 provides a promising target pathway for identifying barriers to pharmacologically induced bone formation.


Asunto(s)
Huesos/efectos de los fármacos , Huesos/metabolismo , Difosfonatos/administración & dosificación , Imidazoles/administración & dosificación , Osteoporosis/tratamiento farmacológico , Hormona Paratiroidea/administración & dosificación , Clorhidrato de Raloxifeno/administración & dosificación , Animales , Resorción Ósea/tratamiento farmacológico , Resorción Ósea/genética , Resorción Ósea/metabolismo , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Quimioterapia Combinada , Femenino , Ratones , Ratones Noqueados , Proteínas Asociadas a Matriz Nuclear/genética , Osteoporosis/genética , Osteoporosis/patología , Factores de Transcripción/genética , Ácido Zoledrónico
20.
Sci Adv ; 3(1): e1601602, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28116354

RESUMEN

ASXL1 is frequently mutated in a spectrum of myeloid malignancies with poor prognosis. Loss of Asxl1 leads to myelodysplastic syndrome-like disease in mice; however, the underlying molecular mechanisms remain unclear. We report that ASXL1 interacts with the cohesin complex, which has been shown to guide sister chromatid segregation and regulate gene expression. Loss of Asxl1 impairs the cohesin function, as reflected by an impaired telophase chromatid disjunction in hematopoietic cells. Chromatin immunoprecipitation followed by DNA sequencing data revealed that ASXL1, RAD21, and SMC1A share 93% of genomic binding sites at promoter regions in Lin-cKit+ (LK) cells. We have shown that loss of Asxl1 reduces the genome binding of RAD21 and SMC1A and alters the expression of ASXL1/cohesin target genes in LK cells. Our study underscores the ASXL1-cohesin interaction as a novel means to maintain normal sister chromatid separation and regulate gene expression in hematopoietic cells.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Cromátides/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Segregación Cromosómica/fisiología , Regulación de la Expresión Génica/fisiología , Hematopoyesis/fisiología , Proteínas Represoras/metabolismo , Animales , Proteínas de Ciclo Celular/genética , Cromátides/genética , Proteínas Cromosómicas no Histona/genética , Proteínas de Unión al ADN , Ratones , Ratones Noqueados , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Proteínas Represoras/genética , Telofase/fisiología , Cohesinas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA