Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Blood ; 136(4): 441-454, 2020 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-32369572

RESUMEN

Chemotherapy and irradiation cause DNA damage to hematopoietic stem cells (HSCs), leading to HSC depletion and dysfunction and the risk of malignant transformation over time. Extrinsic regulation of HSC DNA repair is not well understood, and therapies to augment HSC DNA repair following myelosuppression remain undeveloped. We report that epidermal growth factor receptor (EGFR) regulates DNA repair in HSCs following irradiation via activation of the DNA-dependent protein kinase-catalytic subunit (DNA-PKcs) and nonhomologous end joining (NHEJ). We show that hematopoietic regeneration in vivo following total body irradiation is dependent upon EGFR-mediated repair of DNA damage via activation of DNA-PKcs. Conditional deletion of EGFR in hematopoietic stem and progenitor cells (HSPCs) significantly decreased DNA-PKcs activity following irradiation, causing increased HSC DNA damage and depressed HSC recovery over time. Systemic administration of epidermal growth factor (EGF) promoted HSC DNA repair and rapid hematologic recovery in chemotherapy-treated mice and had no effect on acute myeloid leukemia growth in vivo. Further, EGF treatment drove the recovery of human HSCs capable of multilineage in vivo repopulation following radiation injury. Whole-genome sequencing analysis revealed no increase in coding region mutations in HSPCs from EGF-treated mice, but increased intergenic copy number variant mutations were detected. These studies demonstrate that EGF promotes HSC DNA repair and hematopoietic regeneration in vivo via augmentation of NHEJ. EGF has therapeutic potential to promote human hematopoietic regeneration, and further studies are warranted to assess long-term hematopoietic effects.


Asunto(s)
Reparación del ADN por Unión de Extremidades , Receptores ErbB/metabolismo , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/metabolismo , Regeneración , Animales , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Daño del ADN , Proteína Quinasa Activada por ADN/genética , Proteína Quinasa Activada por ADN/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Factor de Crecimiento Epidérmico/genética , Factor de Crecimiento Epidérmico/metabolismo , Receptores ErbB/genética , Células Madre Hematopoyéticas/citología , Humanos , Ratones
2.
Int J Cancer ; 136(6): 1390-401, 2015 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-25047817

RESUMEN

Most solid tumors contain cancer-associated fibroblasts (CAFs) that support tumorigenesis and malignant progression. However, the cellular origins of CAFs in epithelial ovarian cancers (EOCs) remain poorly understood, and their utility as a source of clinical biomarkers for cancer diagnosis has not been explored in great depth. Here, we report establishing in vitro and in vivo models of CAFs in ovarian cancer development. Normal ovarian fibroblasts and mesenchymal stem cells cultured in the presence of EOC cells acquired a CAF-like phenotype, and promoted EOC cell migration in vitro. CAFs also promoted ovarian cancer growth in vivo in both subcutaneous and intraperitoneal murine xenograft assays. Molecular profiling of CAFs identified gene expression signatures that were highly enriched for extracellular and secreted proteins. We identified novel candidate CAF-specific biomarkers for ovarian cancer including NPPB, which was expressed in the stroma of 60% primary ovarian cancer tissues (n = 145) but not in the stroma of normal ovaries (n = 4). NPPB is a secreted protein that was also elevated in the blood of 50% of women with ovarian cancer (n = 8). Taken together, these data suggest that the tumor stroma is a novel source of biomarkers, including NPPB, that may be of clinical utility for detection of EOC.


Asunto(s)
Biomarcadores de Tumor/análisis , Fibroblastos/química , Neoplasias Glandulares y Epiteliales/patología , Nitrobenzoatos/análisis , Neoplasias Ováricas/patología , Animales , Carcinoma Epitelial de Ovario , Línea Celular Tumoral , Femenino , Humanos , Células Madre Mesenquimatosas/química , Células Madre Mesenquimatosas/fisiología , Ratones , Neoplasias Glandulares y Epiteliales/química , Neoplasias Ováricas/química
3.
Cancer Res ; 84(6): 919-934, 2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38231476

RESUMEN

Bone marrow vascular endothelial cells (BM EC) regulate multiple myeloma pathogenesis. Identification of the mechanisms underlying this interaction could lead to the development of improved strategies for treating multiple myeloma. Here, we performed a transcriptomic analysis of human ECs with high capacity to promote multiple myeloma growth, revealing overexpression of the receptor tyrosine kinases, EPHB1 and EPHB4, in multiple myeloma-supportive ECs. Expression of ephrin B2 (EFNB2), the binding partner for EPHB1 and EPHB4, was significantly increased in multiple myeloma cells. Silencing EPHB1 or EPHB4 in ECs suppressed multiple myeloma growth in coculture. Similarly, loss of EFNB2 in multiple myeloma cells blocked multiple myeloma proliferation and survival in vitro, abrogated multiple myeloma engraftment in immune-deficient mice, and increased multiple myeloma sensitivity to chemotherapy. Administration of an EFNB2-targeted single-chain variable fragment also suppressed multiple myeloma growth in vivo. In contrast, overexpression of EFNB2 in multiple myeloma cells increased STAT5 activation, increased multiple myeloma cell survival and proliferation, and decreased multiple myeloma sensitivity to chemotherapy. Conversely, expression of mutant EFNB2 lacking reverse signaling capacity in multiple myeloma cells increased multiple myeloma cell death and sensitivity to chemotherapy and abolished multiple myeloma growth in vivo. Complementary analysis of multiple myeloma patient data revealed that increased EFNB2 expression is associated with adverse-risk disease and decreased survival. This study suggests that EFNB2 reverse signaling controls multiple myeloma pathogenesis and can be therapeutically targeted to improve multiple myeloma outcomes. SIGNIFICANCE: Ephrin B2 reverse signaling mediated by endothelial cells directly regulates multiple myeloma progression and treatment resistance, which can be overcome through targeted inhibition of ephrin B2 to abolish myeloma.


Asunto(s)
Efrina-B2 , Mieloma Múltiple , Animales , Humanos , Ratones , Células Endoteliales/metabolismo , Efrina-B2/genética , Efrina-B2/metabolismo , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/genética , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptor EphB4/genética , Receptor EphB4/metabolismo , Transducción de Señal/fisiología
4.
J Autism Dev Disord ; 51(6): 1995-2003, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32889639

RESUMEN

To examine reliability and validity of the new Social Motor Function Classification System for Children with Autism Spectrum Disorders (SMFCS-ASD). The SMFCS-ASD reliability was examined on 25 children (62.4 months SD 7.8) with ASD among six physical therapists. The validity study involved 1001 children (57.0 months, SD 9.9) with ASD using the gross motor scale (GMS) of the Peabody Developmental Motor Scales (PDMS-2). The indices of agreement and reliability across six examiners were moderate to substantial (Cohen's κ ≤ 0.65 and ICC > 0.90, all p < 0.001). The SMFCS-ASD was significantly correlated with the GMS of PDMS-2 (all rho from 0.61 to 0.76, p < 0.001). The SMFCS-ASD was reliable and significantly correlated with the GMS of the PDMS-2.


Asunto(s)
Trastorno del Espectro Autista/fisiopatología , Trastorno del Espectro Autista/psicología , Psicometría/métodos , Niño , Desarrollo Infantil , Preescolar , Humanos , Masculino , Reproducibilidad de los Resultados
5.
J Clin Invest ; 130(1): 315-328, 2020 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-31613796

RESUMEN

Tyrosine kinase inhibitors (TKIs) induce molecular remission in the majority of patients with chronic myelogenous leukemia (CML), but the persistence of CML stem cells hinders cure and necessitates indefinite TKI therapy. We report that CML stem cells upregulate the expression of pleiotrophin (PTN) and require cell-autonomous PTN signaling for CML pathogenesis in BCR/ABL+ mice. Constitutive PTN deletion substantially reduced the numbers of CML stem cells capable of initiating CML in vivo. Hematopoietic cell-specific deletion of PTN suppressed CML development in BCR/ABL+ mice, suggesting that cell-autonomous PTN signaling was necessary for CML disease evolution. Mechanistically, PTN promoted CML stem cell survival and TKI resistance via induction of Jun and the unfolded protein response. Human CML cells were also dependent on cell-autonomous PTN signaling, and anti-PTN antibody suppressed human CML colony formation and CML repopulation in vivo. Our results suggest that targeted inhibition of PTN has therapeutic potential to eradicate CML stem cells.


Asunto(s)
Proteínas Portadoras/metabolismo , Citocinas/metabolismo , Proteínas de Fusión bcr-abl/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Células Madre Neoplásicas/metabolismo , Transducción de Señal , Animales , Proteínas Portadoras/genética , Supervivencia Celular , Citocinas/genética , Proteínas de Fusión bcr-abl/genética , Humanos , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Ratones , Ratones Transgénicos , Células Madre Neoplásicas/patología
6.
Nat Commun ; 10(1): 3667, 2019 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-31413255

RESUMEN

Receptor type protein tyrosine phosphatase-sigma (PTPσ) is primarily expressed by adult neurons and regulates neural regeneration. We recently discovered that PTPσ is also expressed by hematopoietic stem cells (HSCs). Here, we describe small molecule inhibitors of PTPσ that promote HSC regeneration in vivo. Systemic administration of the PTPσ inhibitor, DJ001, or its analog, to irradiated mice promotes HSC regeneration, accelerates hematologic recovery, and improves survival. Similarly, DJ001 administration accelerates hematologic recovery in mice treated with 5-fluorouracil chemotherapy. DJ001 displays high specificity for PTPσ and antagonizes PTPσ via unique non-competitive, allosteric binding. Mechanistically, DJ001 suppresses radiation-induced HSC apoptosis via activation of the RhoGTPase, RAC1, and induction of BCL-XL. Furthermore, treatment of irradiated human HSCs with DJ001 promotes the regeneration of human HSCs capable of multilineage in vivo repopulation. These studies demonstrate the therapeutic potential of selective, small-molecule PTPσ inhibitors for human hematopoietic regeneration.


Asunto(s)
Apoptosis/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Células Madre Hematopoyéticas/efectos de los fármacos , Proteínas Tirosina Fosfatasas Clase 2 Similares a Receptores/antagonistas & inhibidores , Regeneración/efectos de los fármacos , Regulación Alostérica , Animales , Antimetabolitos Antineoplásicos/farmacología , Apoptosis/efectos de la radiación , Fluorouracilo/farmacología , Células Madre Hematopoyéticas/efectos de la radiación , Humanos , Ratones , Radiación , Regeneración/efectos de la radiación , Proteína bcl-X/efectos de los fármacos , Proteína bcl-X/metabolismo , Proteína de Unión al GTP rac1/efectos de los fármacos , Proteína de Unión al GTP rac1/metabolismo , Proteínas de Unión al GTP rho/efectos de los fármacos , Proteínas de Unión al GTP rho/metabolismo
7.
Cell Stem Cell ; 23(3): 370-381.e5, 2018 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-30100167

RESUMEN

Bone marrow (BM) perivascular stromal cells and vascular endothelial cells (ECs) are essential for hematopoietic stem cell (HSC) maintenance, but the roles of distinct niche compartments during HSC regeneration are less understood. Here we show that Leptin receptor-expressing (LepR+) BM stromal cells and ECs dichotomously regulate HSC maintenance and regeneration via secretion of pleiotrophin (PTN). BM stromal cells are the key source of PTN during steady-state hematopoiesis because its deletion from stromal cells, but not hematopoietic cells, osteoblasts, or ECs, depletes the HSC pool. Following myelosuppressive irradiation, PTN expression is increased in bone marrow endothelial cells (BMECs), and PTN+ ECs are more frequent in the niche. Moreover, deleting Ptn from ECs impairs HSC regeneration whereas Ptn deletion from BM stromal cells does not. These findings reveal dichotomous and complementary regulation of HSC maintenance and regeneration by BM stromal cells and ECs.


Asunto(s)
Médula Ósea/metabolismo , Proteínas Portadoras/metabolismo , Autorrenovación de las Células , Citocinas/metabolismo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Animales , Citocinas/deficiencia , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL
8.
JCI Insight ; 3(11)2018 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-29875320

RESUMEN

Oncogenic Kras expression specifically in hematopoietic stem cells (HSCs) induces a rapidly fatal myeloproliferative neoplasm in mice, suggesting that Kras signaling plays a dominant role in normal hematopoiesis. However, such a conclusion is based on expression of an oncogenic version of Kras. Hence, we sought to determine the effect of simply increasing the amount of endogenous wild-type Kras on HSC fate. To this end, we utilized a codon-optimized version of the murine Kras gene (Krasex3op) that we developed, in which silent mutations in exon 3 render the encoded mRNA more efficiently translated, leading to increased protein expression without disruption to the normal gene architecture. We found that Kras protein levels were significantly increased in bone marrow (BM) HSCs in Krasex3op/ex3op mice, demonstrating that the translation of Kras in HSCs is normally constrained by rare codons. Krasex3op/ex3op mice displayed expansion of BM HSCs, progenitor cells, and B lymphocytes, but no evidence of myeloproliferative disease or leukemia in mice followed for 12 months. BM HSCs from Krasex3op/ex3op mice demonstrated increased multilineage repopulating capacity in primary competitive transplantation assays, but secondary competitive transplants revealed exhaustion of long-term HSCs. Following total body irradiation, Krasex3op/ex3op mice displayed accelerated hematologic recovery and increased survival. Mechanistically, HSCs from Krasex3op/ex3op mice demonstrated increased proliferation at baseline, with a corresponding increase in Erk1/2 phosphorylation and cyclin-dependent kinase 4 and 6 (Cdk4/6) activation. Furthermore, both the enhanced colony-forming capacity and in vivo repopulating capacity of HSCs from Krasex3op/ex3op mice were dependent on Cdk4/6 activation. Finally, BM transplantation studies revealed that augmented Kras expression produced expansion of HSCs, progenitor cells, and B cells in a hematopoietic cell-autonomous manner, independent from effects on the BM microenvironment. This study provides fundamental demonstration of codon usage in a mammal having a biological consequence, which may speak to the importance of codon usage in mammalian biology.


Asunto(s)
Hematopoyesis/genética , Células Madre Hematopoyéticas/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/genética , Animales , Trasplante de Médula Ósea , Células Cultivadas , Codón/genética , Exones/genética , Femenino , Masculino , Ratones , Ratones Transgénicos , Modelos Animales , Mutación , Cultivo Primario de Células , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Quimera por Trasplante , Irradiación Corporal Total
9.
Nat Med ; 23(1): 91-99, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27918563

RESUMEN

The role of osteolineage cells in regulating hematopoietic stem cell (HSC) regeneration following myelosuppression is not well understood. Here we show that deletion of the pro-apoptotic genes Bak and Bax in osterix (Osx, also known as Sp7 transcription factor 7)-expressing cells in mice promotes HSC regeneration and hematopoietic radioprotection following total body irradiation. These mice showed increased bone marrow (BM) levels of the protein dickkopf-1 (Dkk1), which was produced in Osx-expressing BM cells. Treatment of irradiated HSCs with Dkk1 in vitro increased the recovery of both long-term repopulating HSCs and progenitor cells, and systemic administration of Dkk1 to irradiated mice increased hematopoietic recovery and improved survival. Conversely, inducible deletion of one allele of Dkk1 in Osx-expressing cells in adult mice inhibited the recovery of BM stem and progenitor cells and of complete blood counts following irradiation. Dkk1 promoted hematopoietic regeneration via both direct effects on HSCs, in which treatment with Dkk1 decreased the levels of mitochondrial reactive oxygen species and suppressed senescence, and indirect effects on BM endothelial cells, in which treatment with Dkk1 induced epidermal growth factor (EGF) secretion. Accordingly, blockade of the EGF receptor partially abrogated Dkk1-mediated hematopoietic recovery. These data identify Dkk1 as a regulator of hematopoietic regeneration and demonstrate paracrine cross-talk between BM osteolineage cells and endothelial cells in regulating hematopoietic reconstitution following injury.


Asunto(s)
Células de la Médula Ósea/metabolismo , Autorrenovación de las Células , Células Madre Hematopoyéticas/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Osteoblastos/metabolismo , Regeneración , Factores de Transcripción/metabolismo , Irradiación Corporal Total , Animales , Médula Ósea/metabolismo , Citocinas/metabolismo , Células Endoteliales/metabolismo , Ensayo de Inmunoadsorción Enzimática , Factor de Crecimiento Epidérmico/metabolismo , Receptores ErbB/antagonistas & inhibidores , Citometría de Flujo , Perfilación de la Expresión Génica , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/efectos de la radiación , Péptidos y Proteínas de Señalización Intercelular/farmacología , Ratones , Mitocondrias/metabolismo , Traumatismos Experimentales por Radiación , Especies Reactivas de Oxígeno , Factor de Transcripción Sp7 , Proteína Destructora del Antagonista Homólogo bcl-2/genética , Proteína X Asociada a bcl-2/genética
10.
Radiat Res ; 186(2): 141-52, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27387861

RESUMEN

Ionizing radiation exposure can cause acute radiation sickness (ARS) by damaging the hematopoietic compartment. Radiation damages quiescent hematopoietic stem cells (HSCs) and proliferating hematopoietic cells, resulting in neutropenia, thrombocytopenia and increased risk for long-term hematopoietic dysfunction and myelodysplasia. While some aspects of the hematopoietic response to radiation injury are intrinsic to hematopoietic cells, the recovery of the HSC pool and overall hematopoiesis is also dependent on signals from bone marrow endothelial cells (BM ECs) within the HSC vascular niche. The precise mechanisms through which BM ECs regulate HSC regeneration remain unclear. Characterization of the altered EC gene expression that occurs in response to radiation could provide a roadmap to the discovery of EC-derived mechanisms that regulate hematopoietic regeneration. Here, we show that 5 Gy total-body irradiation substantially alters the expression of numerous genes in BM ECs within 24 h and this molecular response largely resolves by day 14 postirradiation. Several unique and nonannotated genes, which encode secreted proteins were upregulated and downregulated in ECs in response to radiation. These results highlight the complexity of the molecular response of BM ECs to ionizing radiation and identify several candidate mechanisms that should be prioritized for functional analysis in models of hematopoietic injury and regeneration.


Asunto(s)
Células Endoteliales/citología , Células Endoteliales/efectos de la radiación , Animales , Células de la Médula Ósea/citología , Muerte Celular/efectos de la radiación , Relación Dosis-Respuesta en la Radiación , Células Endoteliales/metabolismo , Femenino , Regulación de la Expresión Génica/efectos de la radiación , Ratones , Radiación Ionizante , Factores de Tiempo , Irradiación Corporal Total/efectos adversos
11.
Cell Rep ; 17(6): 1584-1594, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27806297

RESUMEN

Imprinted genes are differentially expressed by adult stem cells, but their functions in regulating adult stem cell fate are incompletely understood. Here we show that growth factor receptor-bound protein 10 (Grb10), an imprinted gene, regulates hematopoietic stem cell (HSC) self-renewal and regeneration. Deletion of the maternal allele of Grb10 in mice (Grb10m/+ mice) substantially increased HSC long-term repopulating capacity, as compared to that of Grb10+/+ mice. After total body irradiation (TBI), Grb10m/+ mice demonstrated accelerated HSC regeneration and hematopoietic reconstitution, as compared to Grb10+/+ mice. Grb10-deficient HSCs displayed increased proliferation after competitive transplantation or TBI, commensurate with upregulation of CDK4 and Cyclin E. Furthermore, the enhanced HSC regeneration observed in Grb10-deficient mice was dependent on activation of the Akt/mTORC1 pathway. This study reveals a function for the imprinted gene Grb10 in regulating HSC self-renewal and regeneration and suggests that the inhibition of Grb10 can promote hematopoietic regeneration in vivo.


Asunto(s)
Autorrenovación de las Células/genética , Proteína Adaptadora GRB10/deficiencia , Eliminación de Gen , Impresión Genómica , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Regeneración , Animales , Células de la Médula Ósea/citología , Proliferación Celular , Proteína Adaptadora GRB10/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Irradiación Corporal Total
12.
Anemia ; 2012: 481583, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22693661

RESUMEN

The Fanconi Anemia (FA) pathway consists of proteins involved in repairing DNA damage, including interstrand cross-links (ICLs). The pathway contains an upstream multiprotein core complex that mediates the monoubiquitylation of the FANCD2 and FANCI heterodimer, and a downstream pathway that converges with a larger network of proteins with roles in homologous recombination and other DNA repair pathways. Selective killing of cancer cells with an intact FA pathway but deficient in certain other DNA repair pathways is an emerging approach to tailored cancer therapy. Inhibiting the FA pathway becomes selectively lethal when certain repair genes are defective, such as the checkpoint kinase ATM. Inhibiting the FA pathway in ATM deficient cells can be achieved with small molecule inhibitors, suggesting that new cancer therapeutics could be developed by identifying FA pathway inhibitors to treat cancers that contain defects that are synthetic lethal with FA.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA