Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
ACS Med Chem Lett ; 14(7): 955-961, 2023 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-37465311

RESUMEN

Myeloid cell leukemia-1 (MCL-1) is a member of the antiapoptotic BCL-2 proteins family and a key regulator of mitochondrial homeostasis. Overexpression of MCL-1 is found in many cancer cells and contributes to tumor progression, which makes it an attractive therapeutic target. Pursuing our previous study of macrocyclic indoles for the inhibition of MCL-1, we report herein the impact of both pyrazole and indole isomerism on the potency and overall properties of this family of compounds. We demonstrated that the incorporation of a fluorine atom on the naphthalene moiety was a necessary step to improve cellular potency and that, combined with the introduction of various side chains on the pyrazole, it enhanced solubility significantly. This exploration culminated in the discovery of compounds (Ra)-10 and (Ra)-15, possessing remarkable cellular potency and properties.

2.
J Med Chem ; 66(9): 6122-6148, 2023 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-37114951

RESUMEN

Avoidance of apoptosis is critical for the development and sustained growth of tumors. The pro-survival protein myeloid cell leukemia 1 (Mcl-1) is an anti-apoptotic member of the Bcl-2 family of proteins which is overexpressed in many cancers. Upregulation of Mcl-1 in human cancers is associated with high tumor grade, poor survival, and resistance to chemotherapy. Therefore, pharmacological inhibition of Mcl-1 is regarded as an attractive approach to treating relapsed or refractory malignancies. Herein, we disclose the design, synthesis, optimization, and early preclinical evaluation of a potent and selective small-molecule inhibitor of Mcl-1. Our exploratory design tactics focused on structural modifications which improve the potency and physicochemical properties of the inhibitor while minimizing the risk of functional cardiotoxicity. Despite being in the "non-Lipinski" beyond-Rule-of-Five property space, the developed compound benefits from exquisite oral bioavailability in vivo and induces potent pharmacodynamic inhibition of Mcl-1 in a mouse xenograft model.


Asunto(s)
Antineoplásicos , Neoplasias Hematológicas , Humanos , Ratones , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Antineoplásicos/química , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Línea Celular Tumoral , Apoptosis , Neoplasias Hematológicas/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo
3.
Front Immunol ; 12: 754083, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34712241

RESUMEN

Myeloid-derived suppressor cells (MDSCs) are a hetero geneous group of cells, which can suppress the immune response, promote tumor progression and impair the efficacy of immunotherapies. Consequently, the pharmacological targeting of MDSC is emerging as a new immunotherapeutic strategy to stimulate the natural anti-tumor immune response and potentiate the efficacy of immunotherapies. Herein, we leveraged genetically modified models and a small molecule inhibitor to validate Calcium-Calmodulin Kinase Kinase 2 (CaMKK2) as a druggable target to control MDSC accumulation in tumor-bearing mice. The results indicated that deletion of CaMKK2 in the host attenuated the growth of engrafted tumor cells, and this phenomenon was associated with increased antitumor T cell response and decreased accumulation of MDSC. The adoptive transfer of MDSC was sufficient to restore the ability of the tumor to grow in Camkk2-/- mice, confirming the key role of MDSC in the mechanism of tumor rejection. In vitro studies indicated that blocking of CaMKK2 is sufficient to impair the yield of MDSC. Surprisingly, MDSC generated from Camkk2-/- bone marrow cells also showed a higher ability to terminally differentiate toward more immunogenic cell types (e.g inflammatory macrophages and dendritic cells) compared to wild type (WT). Higher intracellular levels of reactive oxygen species (ROS) accumulated in Camkk2-/- MDSC, increasing their susceptibility to apoptosis and promoting their terminal differentiation toward more mature myeloid cells. Mechanistic studies indicated that AMP-activated protein kinase (AMPK), which is a known CaMKK2 proximal target controlling the oxidative stress response, fine-tunes ROS accumulation in MDSC. Accordingly, failure to activate the CaMKK2-AMPK axis can account for the elevated ROS levels in Camkk2-/- MDSC. These results highlight CaMKK2 as an important regulator of the MDSC lifecycle, identifying this kinase as a new druggable target to restrain MDSC expansion and enhance the efficacy of anti-tumor immunotherapy.


Asunto(s)
Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/fisiología , Células Supresoras de Origen Mieloide/enzimología , Proteínas de Neoplasias/fisiología , Proteínas Quinasas Activadas por AMP/fisiología , Traslado Adoptivo , Animales , Apoptosis , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/deficiencia , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/genética , Femenino , Depleción Linfocítica , Linfoma/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Mitocondrias/metabolismo , Células Supresoras de Origen Mieloide/fisiología , Células Supresoras de Origen Mieloide/trasplante , Mielopoyesis , Especies Reactivas de Oxígeno , Microambiente Tumoral
4.
Nat Commun ; 10(1): 2450, 2019 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-31164648

RESUMEN

Tumor-associated myeloid cells regulate tumor growth and metastasis, and their accumulation is a negative prognostic factor for breast cancer. Here we find calcium/calmodulin-dependent kinase kinase (CaMKK2) to be highly expressed within intratumoral myeloid cells in mouse models of breast cancer, and demonstrate that its inhibition within myeloid cells suppresses tumor growth by increasing intratumoral accumulation of effector CD8+ T cells and immune-stimulatory myeloid subsets. Tumor-associated macrophages (TAMs) isolated from Camkk2-/- mice expressed higher levels of chemokines involved in the recruitment of effector T cells compared to WT. Similarly, in vitro generated Camkk2-/- macrophages recruit more T cells, and have a reduced capability to suppress T cell proliferation, compared to WT. Treatment with CaMKK2 inhibitors blocks tumor growth in a CD8+ T cell-dependent manner, and facilitates a favorable reprogramming of the immune cell microenvironment. These data, credential CaMKK2 as a myeloid-selective checkpoint, the inhibition of which may have utility in the immunotherapy of breast cancer.


Asunto(s)
Neoplasias de la Mama/inmunología , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/inmunología , Carcinoma/inmunología , Neoplasias Mamarias Animales/inmunología , Células Mieloides/inmunología , Escape del Tumor/inmunología , Microambiente Tumoral/inmunología , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Linfocitos T CD8-positivos/inmunología , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/antagonistas & inhibidores , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/genética , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/metabolismo , Carcinoma/genética , Carcinoma/metabolismo , Proliferación Celular , Quimiocinas/inmunología , Femenino , Humanos , Inmunohistoquímica , Técnicas In Vitro , Macrófagos/inmunología , Macrófagos/metabolismo , Neoplasias Mamarias Animales/genética , Neoplasias Mamarias Animales/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , Células Mieloides/metabolismo , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/inmunología , Neoplasias de la Mama Triple Negativas/metabolismo , Escape del Tumor/genética
5.
Stem Cells Dev ; 27(21): 1494-1506, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30084753

RESUMEN

The interactions of hematopoietic stem and progenitor cells (HSPCs) with extracellular matrix (ECM) components and cells from the bone marrow (BM) microenvironment control their homeostasis. Regenerative BM conditions can induce expression of the ECM protein transforming growth factor beta-induced gene H3 (TGFBI or BIGH3) in murine HSPCs. In this study, we examined how increased or reduced TGFBI expression in human HSPCs and BM mesenchymal stromal cells (MSCs) affects HSPC maintenance, differentiation, and migration. HSPCs that overexpressed TGFBI showed accelerated megakaryopoiesis, whereas granulocyte differentiation and proliferation of granulocyte, erythrocyte, and monocyte cultures were reduced. In addition, both upregulation and downregulation of TGFBI expression impaired HSPC colony-forming capacity of HSPCs. Interestingly, the colony-forming capacity of HSPCs with reduced TGFBI levels was increased after long-term co-culture with MSCs, as measured by long-term culture-colony forming cell (LTC-CFC) formation. Moreover, TGFBI downregulation in HSPCs resulted in increased cobblestone area-forming cell (CAFC) frequency, a measure for hematopoietic stem cell (HSC) capacity. Concordantly, TGFBI upregulation in HSPCs resulted in a decrease of CAFC and LTC-CFC frequency. These results indicate that reduced TGFBI levels in HSPCs enhanced HSC maintenance, but only in the presence of MSCs. In addition, reduced levels of TGFBI in MSCs affected MSC/HSPC interaction, as observed by an increased migration of HSPCs under the stromal layer. In conclusion, tight regulation of TGFBI expression in the BM niche is essential for balanced HSPC proliferation and differentiation.


Asunto(s)
Proliferación Celular/genética , Proteínas de la Matriz Extracelular/genética , Células Madre Hematopoyéticas/citología , Células Madre Mesenquimatosas/citología , Células Madre/citología , Factor de Crecimiento Transformador beta/genética , Células de la Médula Ósea/citología , Diferenciación Celular/genética , Línea Celular , Movimiento Celular/genética , Lisencefalia de Cobblestone/genética , Técnicas de Cocultivo , Citometría de Flujo , Regulación del Desarrollo de la Expresión Génica , Vectores Genéticos , Hematopoyesis/genética , Células Madre Hematopoyéticas/metabolismo , Humanos , Lentivirus/genética , Células Madre Mesenquimatosas/metabolismo , Células Madre/metabolismo
6.
Cell Death Dis ; 8(10): e3076, 2017 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-28981105

RESUMEN

Hematopoietic stem and progenitor cells (HSPCs) are predominantly quiescent in adults, but proliferate in response to bone marrow (BM) injury. Here, we show that deletion of Ca2+/calmodulin (CaM)-dependent protein kinase kinase 2 (CaMKK2) promotes HSPC regeneration and hematopoietic recovery following radiation injury. Using Camkk2-enhanced green fluorescent protein (EGFP) reporter mice, we found that Camkk2 expression is developmentally regulated in HSPC. Deletion of Camkk2 in HSPC results in a significant downregulation of genes affiliated with the quiescent signature. Accordingly, HSPC from Camkk2 null mice have a high proliferative capability when stimulated in vitro in the presence of BM-derived endothelial cells. In addition, Camkk2 null mice are more resistant to radiation injury and show accelerated hematopoietic recovery, enhanced HSPC regeneration and ultimately a prolonged survival following sublethal or lethal total body irradiation. Mechanistically, we propose that CaMKK2 regulates the HSPC response to hematopoietic damage by coupling radiation signaling to activation of the anti-proliferative AMP-activated protein kinase. Finally, we demonstrated that systemic administration of the small molecule CaMKK2 inhibitor, STO-609, to irradiated mice enhanced HSPC recovery and improved survival. These findings identify CaMKK2 as an important regulator of HSPC regeneration and demonstrate CaMKK2 inhibition is a novel approach to promoting hematopoietic recovery after BM injury.


Asunto(s)
Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/genética , Calmodulina/genética , Células Madre Hematopoyéticas/metabolismo , Traumatismos por Radiación/tratamiento farmacológico , Animales , Bencimidazoles/administración & dosificación , Calcio/metabolismo , Proteínas Fluorescentes Verdes/genética , Ratones , Ratones Noqueados , Naftalimidas/administración & dosificación , Traumatismos por Radiación/genética , Traumatismos por Radiación/patología , Regeneración/genética , Transducción de Señal/efectos de los fármacos , Irradiación Corporal Total
7.
Genes Dev ; 28(9): 995-1004, 2014 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-24788518

RESUMEN

Accidental or deliberate ionizing radiation exposure can be fatal due to widespread hematopoietic destruction. However, little is known about either the course of injury or the molecular pathways that regulate the subsequent regenerative response. Here we show that the Wnt signaling pathway is critically important for regeneration after radiation-induced injury. Using Wnt reporter mice, we show that radiation triggers activation of Wnt signaling in hematopoietic stem and progenitor cells. ß-Catenin-deficient mice, which lack the ability to activate canonical Wnt signaling, exhibited impaired hematopoietic stem cell regeneration and bone marrow recovery after radiation. We found that, as part of the mechanism, hematopoietic stem cells lacking ß-catenin fail to suppress the generation of reactive oxygen species and cannot resolve DNA double-strand breaks after radiation. Consistent with the impaired response to radiation, ß-catenin-deficient mice are also unable to recover effectively after chemotherapy. Collectively, these data indicate that regenerative responses to distinct hematopoietic injuries share a genetic dependence on ß-catenin and raise the possibility that modulation of Wnt signaling may be a path to improving bone marrow recovery after damage.


Asunto(s)
Células Madre Hematopoyéticas/fisiología , Estrés Oxidativo/genética , Regeneración/genética , beta Catenina/genética , beta Catenina/metabolismo , Animales , Antineoplásicos/farmacología , Médula Ósea/efectos de los fármacos , Médula Ósea/efectos de la radiación , Roturas del ADN de Doble Cadena/efectos de la radiación , Fluorouracilo/farmacología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/efectos de la radiación , Estimación de Kaplan-Meier , Ratones , Estrés Oxidativo/efectos de la radiación , Traumatismos por Radiación/genética , Especies Reactivas de Oxígeno/metabolismo , Regeneración/efectos de los fármacos , Regeneración/efectos de la radiación , Transducción de Señal , Vía de Señalización Wnt/efectos de la radiación
8.
Radiat Res ; 181(5): 445-51, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24720754

RESUMEN

Exposure to a nuclear accident or radiological attack can cause death from acute radiation syndrome (ARS), which results from radiation injury to vital organs such as the hematopoietic system. However, the U.S. Food and Drug Administration (FDA) has not approved any medical countermeasures for this specific purpose. With growing concern over nuclear terrorism, there is an urgent need to develop small molecule deliverables that mitigate mortality from ARS. One emerging modulator of hematopoietic stem/progenitor cell (HSPC) activity is glycogen synthase kinase-3 (GSK-3). The inhibition of GSK-3 has been shown to augment hematopoietic repopulation in mouse models of bone marrow transplantation. In this study, we performed an in vitro screen using irradiated bone marrow mononuclear cells (BM-MNCs) to test the effects of four GSK-3 inhibitors: CHIR99021; 6-Bromoindirubin-3'-oxime (BIO); SB415286; and SB216763. This screen showed that SB216763 significantly increased the frequency of c-Kit(+) Lin(-) Sca1(+) (KLS) cells and hematopoietic colony-forming cells in irradiated BM-MNCs. Importantly, administration of a single dose of SB216763 to C57BL/6J mice by subcutaneous injection 24 h after total-body irradiation significantly improved hematopoietic recovery and mitigated hematopoietic ARS. Collectively, our results demonstrate that the GSK-3 inhibitor SB216763 is an effective medical countermeasure against acute radiation injury of the hematopoietic system.


Asunto(s)
Síndrome de Radiación Aguda/tratamiento farmacológico , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Células Madre Hematopoyéticas/efectos de los fármacos , Inhibidores de Proteínas Quinasas/uso terapéutico , Traumatismos Experimentales por Radiación/tratamiento farmacológico , Síndrome de Radiación Aguda/enzimología , Síndrome de Radiación Aguda/patología , Aminofenoles/farmacología , Aminofenoles/uso terapéutico , Animales , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Médula Ósea/patología , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Evaluación Preclínica de Medicamentos , Glucógeno Sintasa Quinasa 3/fisiología , Hematopoyesis/efectos de los fármacos , Hematopoyesis/efectos de la radiación , Células Madre Hematopoyéticas/efectos de la radiación , Indoles/farmacología , Indoles/uso terapéutico , Inyecciones Subcutáneas , Maleimidas/farmacología , Maleimidas/uso terapéutico , Ratones , Ratones Endogámicos C57BL , Oximas/farmacología , Oximas/uso terapéutico , Inhibidores de Proteínas Quinasas/farmacología , Piridinas/farmacología , Piridinas/uso terapéutico , Pirimidinas/farmacología , Pirimidinas/uso terapéutico , Traumatismos Experimentales por Radiación/enzimología , Traumatismos Experimentales por Radiación/patología , Irradiación Corporal Total/efectos adversos
9.
J Leukoc Biol ; 94(6): 1243-51, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23935208

RESUMEN

Chemokine receptor interactions coordinate leukocyte migration in inflammation. Chemokine receptors are GPCRs that when activated, are phosphorylated by GRKs to turn off G protein-mediated signaling yet recruit additional signaling machinery. Recently, GRK3 was identified as a negative regulator of CXCL12/CXCR4 signaling that is defective in human WHIM syndrome. Here, we report that GRK3-/- mice exhibit numerous features of human WHIM, such as impaired CXCL12-mediated desensitization, enhanced CXCR4 signaling to ERK activation, altered granulocyte migration, and a mild myelokathexis. Moreover, GRK3-/- protects mice from two acute models of inflammatory arthritis (K/BxN serum transfer and CAIA). In these granulocyte-dependent disease models, protection of GRK3-/- mice is mediated by retention of cells in the marrow, fewer circulating granulocytes in the peripheral blood, and reduced granulocytes in the joints during active inflammation. In contrast to WHIM, GRK3-/- mice have minimal hypogammaglobulinemia and a peripheral leukocytosis with increased lymphocytes and absent neutropenia. Thus, we conclude that the loss of GRK3-mediated regulation of CXCL12/CXCR4 signaling contributes to some, but not all, of the complete WHIM phenotype and that GRK3 inhibition may be beneficial in the treatment of inflammatory arthritis.


Asunto(s)
Quinasa 3 del Receptor Acoplado a Proteína-G/inmunología , Síndromes de Inmunodeficiencia/inmunología , Sistema de Señalización de MAP Quinasas/inmunología , Verrugas/inmunología , Animales , Línea Celular Transformada , Quimiocina CXCL12/genética , Quimiocina CXCL12/inmunología , Quimiocina CXCL12/metabolismo , Modelos Animales de Enfermedad , Quinasa 3 del Receptor Acoplado a Proteína-G/genética , Quinasa 3 del Receptor Acoplado a Proteína-G/metabolismo , Granulocitos/enzimología , Granulocitos/inmunología , Granulocitos/patología , Humanos , Síndromes de Inmunodeficiencia/enzimología , Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/patología , Inflamación/enzimología , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Sistema de Señalización de MAP Quinasas/genética , Ratones , Ratones Noqueados , Enfermedades de Inmunodeficiencia Primaria , Receptores CXCR4/genética , Receptores CXCR4/inmunología , Receptores CXCR4/metabolismo , Verrugas/enzimología , Verrugas/genética , Verrugas/patología
10.
Artículo en Inglés | MEDLINE | ID: mdl-23378582

RESUMEN

One of the most remarkable characteristics of stem cells is their ability to perpetuate themselves through self-renewal while concomitantly generating differentiated cells. In the hematopoietic system, stem cells balance these mechanisms to maintain steady-state hematopoiesis for the lifetime of the organism, and to effectively regenerate the system following injury. Defects in the proper control of self-renewal and differentiation can be potentially devastating and contribute to the development of malignancies. In this review, we trace the emerging role of Wnt signaling as a critical regulator of distinct aspects of self-renewal and differentiation, its contribution to the maintenance of homeostasis and regeneration, and how the pathway can be hijacked to promote leukemia development. A better understanding of these processes could pave the way to enhancing recovery after injury and to developing better therapeutic approaches for hematologic malignancies.


Asunto(s)
Hematopoyesis , Células Madre Hematopoyéticas/metabolismo , Leucemia/metabolismo , Proteínas Wnt/metabolismo , Vía de Señalización Wnt , Animales , Diferenciación Celular , Humanos , Linfocitos/metabolismo , Ratones , Proteínas Wnt/genética , Proteínas Wnt/fisiología , Xenopus
11.
Development ; 139(10): 1724-33, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22461560

RESUMEN

The WNT pathway plays multiple roles in neural development and is crucial for establishment of the embryonic cerebellum. In addition, WNT pathway mutations are associated with medulloblastoma, the most common malignant brain tumor in children. However, the cell types within the cerebellum that are responsive to WNT signaling remain unknown. Here we investigate the effects of canonical WNT signaling on two important classes of progenitors in the developing cerebellum: multipotent neural stem cells (NSCs) and granule neuron precursors (GNPs). We show that WNT pathway activation in vitro promotes proliferation of NSCs but not GNPs. Moreover, mice that express activated ß-catenin in the cerebellar ventricular zone exhibit increased proliferation of NSCs in that region, whereas expression of the same protein in GNPs impairs proliferation. Although ß-catenin-expressing NSCs proliferate they do not undergo prolonged expansion or neoplastic growth; rather, WNT signaling markedly interferes with their capacity for self-renewal and differentiation. At a molecular level, mutant NSCs exhibit increased expression of c-Myc, which might account for their transient proliferation, but also express high levels of bone morphogenetic proteins and the cyclin-dependent kinase inhibitor p21, which might contribute to their altered self-renewal and differentiation. These studies suggest that the WNT pathway is a potent regulator of cerebellar stem cell growth and differentiation.


Asunto(s)
Cerebelo/citología , Cerebelo/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Vía de Señalización Wnt/fisiología , Animales , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Proliferación Celular , Células Cultivadas , Cerebelo/embriología , Citometría de Flujo , Ratones , Reacción en Cadena en Tiempo Real de la Polimerasa , Vía de Señalización Wnt/genética , beta Catenina/genética , beta Catenina/metabolismo
12.
Mol Biol Cell ; 22(8): 1312-20, 2011 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21346186

RESUMEN

Growth factor erv1-like (Gfer) is an evolutionarily conserved sulfhydryl oxidase that is enriched in embryonic and adult stem cells and plays an essential prosurvival role in pluripotent embryonic stem cells. Here we show that knockdown (KD) of Gfer in hematopoietic stem cells (HSCs) compromises their in vivo engraftment potential and triggers a hyper-proliferative response that leads to their exhaustion. KD of Gfer in HSCs does not elicit a significant alteration of mitochondrial morphology or loss of cell viability. However, these cells possess significantly reduced levels of the cyclin-dependent kinase inhibitor p27(kip1). In contrast, overexpression of Gfer in HSCs results in significantly elevated total and nuclear p27(kip1). KD of Gfer results in enhanced binding of p27(kip1) to its inhibitor, the COP9 signalosome subunit jun activation-domain binding protein 1 (Jab1), leading to its down-regulation. Conversely, overexpression of Gfer results in its enhanced binding to Jab1 and inhibition of the Jab1-p27(kip1) interaction. Furthermore, normalization of p27(kip1) in Gfer-KD HSCs rescues their in vitro proliferation deficits. Taken together, our data demonstrate the presence of a novel Gfer-Jab1-p27(kip1) pathway in HSCs that functions to restrict abnormal proliferation.


Asunto(s)
Proliferación Celular , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Células Madre Hematopoyéticas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/metabolismo , Péptido Hidrolasas/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Animales , Complejo del Señalosoma COP9 , Supervivencia Celular/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Regulación hacia Abajo , Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/citología , Péptidos y Proteínas de Señalización Intracelular/genética , Lentivirus , Ratones , Ratones Endogámicos , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/genética , Péptido Hidrolasas/genética , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Unión Proteica/genética , ARN Interferente Pequeño/metabolismo , Proteínas Recombinantes de Fusión/genética , Transfección , Irradiación Corporal Total
13.
Nature ; 466(7307): 765-8, 2010 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-20639863

RESUMEN

Chronic myelogenous leukaemia (CML) can progress from a slow growing chronic phase to an aggressive blast crisis phase, but the molecular basis of this transition remains poorly understood. Here we have used mouse models of CML to show that disease progression is regulated by the Musashi-Numb signalling axis. Specifically, we find that the chronic phase is marked by high levels of Numb expression whereas the blast crisis phase has low levels of Numb expression, and that ectopic expression of Numb promotes differentiation and impairs advanced-phase disease in vivo. As a possible explanation for the decreased levels of Numb in the blast crisis phase, we show that NUP98-HOXA9, an oncogene associated with blast crisis CML, can trigger expression of the RNA-binding protein Musashi2 (Msi2), which in turn represses Numb. Notably, loss of Msi2 restores Numb expression and significantly impairs the development and propagation of blast crisis CML in vitro and in vivo. Finally we show that Msi2 expression is not only highly upregulated during human CML progression but is also an early indicator of poorer prognosis. These data show that the Musashi-Numb pathway can control the differentiation of CML cells, and raise the possibility that targeting this pathway may provide a new strategy for the therapy of aggressive leukaemias.


Asunto(s)
Diferenciación Celular , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Proteínas de Unión al ARN/metabolismo , Animales , Crisis Blástica/genética , Crisis Blástica/metabolismo , Crisis Blástica/patología , Diferenciación Celular/genética , Progresión de la Enfermedad , Proteínas de Fusión bcr-abl/genética , Proteínas de Fusión bcr-abl/metabolismo , Regulación Neoplásica de la Expresión Génica , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas de Complejo Poro Nuclear/genética , Proteínas de Complejo Poro Nuclear/metabolismo , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , Pronóstico , Proteínas de Unión al ARN/biosíntesis , Proteínas de Unión al ARN/genética , Receptor Notch1/metabolismo , Transducción de Señal , Proteína p53 Supresora de Tumor/metabolismo , Regulación hacia Arriba
14.
Nat Med ; 16(4): 475-82, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20305662

RESUMEN

Hematopoietic stem cell (HSC) self-renewal is regulated by both intrinsic and extrinsic signals. Although some of the pathways that regulate HSC self-renewal have been uncovered, it remains largely unknown whether these pathways can be triggered by deliverable growth factors to induce HSC growth or regeneration. Here we show that pleiotrophin, a neurite outgrowth factor with no known function in hematopoiesis, efficiently promotes HSC expansion in vitro and HSC regeneration in vivo. Treatment of mouse bone marrow HSCs with pleiotrophin caused a marked increase in long-term repopulating HSC numbers in culture, as measured in competitive repopulating assays. Treatment of human cord blood CD34(+)CDCD38(-)Lin(-) cells with pleiotrophin also substantially increased severe combined immunodeficient (SCID)-repopulating cell counts in culture, compared to input and cytokine-treated cultures. Systemic administration of pleiotrophin to irradiated mice caused a pronounced expansion of bone marrow stem and progenitor cells in vivo, indicating that pleiotrophin is a regenerative growth factor for HSCs. Mechanistically, pleiotrophin activated phosphoinositide 3-kinase (PI3K) signaling in HSCs; antagonism of PI3K or Notch signaling inhibited pleiotrophin-mediated expansion of HSCs in culture. We identify the secreted growth factor pleiotrophin as a new regulator of both HSC expansion and regeneration.


Asunto(s)
Proteínas Portadoras/fisiología , Citocinas/fisiología , Células Madre Hematopoyéticas/fisiología , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/fisiología , Proteínas Portadoras/farmacología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Citocinas/farmacología , Relación Dosis-Respuesta a Droga , Células Madre Hematopoyéticas/efectos de los fármacos , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones SCID , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfatidilinositol 3-Quinasas/fisiología , Receptores Notch/fisiología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Factor de Células Madre/farmacología , Trombopoyetina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...