Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
1.
Int J STD AIDS ; 23(2): 77-82, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22422679

RESUMEN

Tobacco smoking and HIV infection increase the risk of epidermoid anal cancer (EAC). No published studies have examined smoking and EAC outcomes, and the literature is discrepant regarding outcomes of HIV-positive patients with EAC. The goal of this study was to examine smoking history, HIV status and outcomes in EAC patients. We conducted a retrospective analysis of adults with invasive EAC treated in the University of Washington hospital system from 1 January 1994 to 31 December 2008. Sixty-three patients were included. Forty-seven patients (75%) had primary chemoradiation, of whom 42 (89%) completed therapy. Two patients (3%) received radiotherapy alone. Fourteen patients (22%) underwent primary surgery, of whom 11 (79%) underwent tumour excision and three (21%) abdominoperineal resection (APR). We analysed smoking history, HIV status and CD4 count (≥ 200 cells/µL/<200 cells/µL for HIV-positive patients) versus outcomes. Forty-five patients (71%) were in remission, and 44 (70%) were alive at last follow-up. Overall survival was significantly better for never-smokers than for ever-smokers. There were no differences in outcomes according to HIV status or CD4 counts. Patients with anal cancer who smoke have worse overall survival than non-smoking patients. HIV infection does not appear to affect anal cancer outcomes.


Asunto(s)
Neoplasias del Ano/epidemiología , Carcinoma de Células Escamosas/epidemiología , Seropositividad para VIH/epidemiología , Fumar/epidemiología , Adulto , Neoplasias del Ano/terapia , Neoplasias del Ano/virología , Recuento de Linfocito CD4 , Carcinoma de Células Escamosas/terapia , Carcinoma de Células Escamosas/virología , Femenino , Seropositividad para VIH/inmunología , Humanos , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Fumar/efectos adversos , Tasa de Supervivencia , Resultado del Tratamiento , Washingtón/epidemiología
2.
Cytokine ; 15(4): 188-95, 2001 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-11563879

RESUMEN

Stem cell factor (SCF) initiates its biological effects by binding to its receptor Kit. Cell surface Kit is proteolytically cleaved to generate soluble Kit. Structure-function analysis of the extracellular region of Kit has implicated the first three immunoglobulin-like domains in SCF binding, and the fourth immunoglobulin-like domain in receptor dimerization. However, the role of the fifth immunoglobulin-like domain is unknown. To test the hypothesis that the fifth immunoglobulin-like domain is important for proteolytic cleavage of Kit from the cell surface, we constructed a mutant form of Kit in which the first four immunoglobulin-like domains are linked to the transmembrane and cytoplasmic domains (designated Kit-Del5). Kit-wild type (Kit-WT) and Kit-Del5 were expressed in the murine mast cell line IC2. Flow cytometry demonstrated that both Kit-WT and Kit-Del5 are displayed on the IC2 cell surface, and immunoblotting confirmed the presence of Kit proteins of the expected molecular weights, 154 kDa and 134 kDa, respectively. Although IC2-Kit-WT cells proteolytically cleave cell surface Kit, generating a 98 kDa soluble form of Kit, IC2-Kit-Del5 cells do not. These findings demonstrate that the fifth immunoglobulin-like domain of Kit is required for proteolytic cleavage of Kit from the cell surface.


Asunto(s)
Inmunoglobulinas/química , Proteínas Proto-Oncogénicas c-kit/química , Animales , División Celular , Línea Celular , Membrana Celular/metabolismo , Citoplasma/metabolismo , Citometría de Flujo , Eliminación de Gen , Humanos , Ratones , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-kit/genética , Proteínas Proto-Oncogénicas c-kit/metabolismo , Factor de Células Madre/metabolismo , Acetato de Tetradecanoilforbol/farmacología
3.
Blood ; 98(7): 2077-83, 2001 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-11567993

RESUMEN

Cellular trafficking of growth factor receptors, including cross-talk among receptors at the cell surface, may be important for signal transduction in normal hematopoietic cells. To test this idea, the signaling domain of Mpl (the thrombopoietin receptor) was targeted to the plasma membrane, or to the cytoplasm of murine marrow cells, and the ability of the cells to proliferate and differentiate in response to Mpl dimerized at the plasma membrane or free in the cytoplasm was assessed. Constructs encoding the signaling domain of Mpl linked to an FK506 binding protein domain (to permit dimerization by the membrane-permeable ligand AP20187) with or without a myristylation sequence (to target the receptor to the plasma membrane) and a hemagglutinin epitope tag were generated and introduced into murine marrow cells using a murine stem cell virus (MSCV)-based retroviral vector. Both populations of transduced marrow cells proliferated in Iscoves modified Dulbecco medium-10% FCS-100 nM AP20187 without exogenous growth factors for more than 100 days and achieved greater than a 10(7)-fold expansion of cells by day 50 (n = 4 transductions). Growth was dimerizer dependent, and myeloid, erythroid, and megakaryocytic progenitors were generated. Activation of Mpl either at the plasma membrane or in the cytoplasm allowed for the terminal maturation of transduced progenitor cells. Introduction of membrane-targeted or cytoplasmic Mpl into fetal liver cells from homozygous JAK2 knock-out mice or wild-type littermates demonstrated that both forms of Mpl require JAK2 for signaling. These data show that the activation of Mpl independent of its normal plasma membrane location can support production of the full range of normal hematopoietic progenitor cells in vitro.


Asunto(s)
Membrana Celular/metabolismo , Células Madre Hematopoyéticas/efectos de los fármacos , Proteínas de la Leche , Proteínas de Neoplasias , Proteínas Proto-Oncogénicas/fisiología , Receptores de Citocinas , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Línea Celular , Membrana Celular/química , Citoplasma/química , Proteínas de Unión al ADN/farmacología , Proteínas de Unión al ADN/fisiología , Dimerización , Células Madre Hematopoyéticas/citología , Janus Quinasa 2 , Ratones , Microscopía Fluorescente , Transporte de Proteínas , Proteínas Tirosina Quinasas/farmacología , Proteínas Tirosina Quinasas/fisiología , Proteínas Proto-Oncogénicas/farmacología , Receptores de Trombopoyetina , Factor de Transcripción STAT5 , Transducción de Señal , Transactivadores/farmacología , Transactivadores/fisiología
4.
Leuk Res ; 25(5): 413-21, 2001 May.
Artículo en Inglés | MEDLINE | ID: mdl-11301110

RESUMEN

Stem cell factor (SCF) is a growth factor that promotes the survival, proliferation, and differentiation of hematopoietic cells. SCF and its receptor, Kit, are normally present in both cell surface and soluble forms. Both forms of Kit can bind SCF. However, the function of soluble Kit is unknown. In order to determine if soluble Kit can modulate SCF activity, we produced a fusion protein, Kit-Fc, comprised of the extracellular domain of murine Kit and the Fc portion of human IgG(1) and investigated its ability to bind 125I-SCF and to inhibit SCF-stimulated hematopoietic colony growth in vitro. Stable cell lines expressing Kit-Fc were generated and Kit-Fc was purified to greater than 95% purity. Scatchard analysis demonstrated that Kit-Fc binds iodinated SCF with high affinity (Kd 570 pM). Kit-Fc also bound to transmembrane SCF displayed on the surface of fibroblasts. The murine mast cell line IC2 was engineered to express murine Kit on the cell surface and was demonstrated to proliferate in the presence of SCF. Kit-Fc completely blocked SCF-stimulated proliferation of IC2-Kit cells, but not IL-3-stimulated growth of IC2-Kit cells, demonstrating the specificity of Kit-Fc. We investigated the ability of Kit-Fc to block SCF-stimulated murine hematopoietic colony growth. Kit-Fc blocked SCF-stimulated erythroid colony growth as effectively as a neutralizing anti-Kit monoclonal antibody, ACK2, but did not block erythropoietin-stimulated erythroid colony growth. Likewise, Kit-Fc blocked SCF-stimulated myeloid colony growth as effectively as ACK2 antibody, but did not block IL-3- or GM-CSF-stimulated myeloid colony growth. These results indicate that a form of soluble Kit binds SCF with high affinity, and can specifically block the ability of SCF to stimulate hematopoietic colony growth, suggesting that one function of soluble Kit may be to modulate SCF bioactivity.


Asunto(s)
Proteínas Proto-Oncogénicas c-kit/farmacología , Factor de Células Madre/antagonistas & inhibidores , Células 3T3/metabolismo , Animales , Línea Celular , Cricetinae , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/efectos de los fármacos , Humanos , Inmunoglobulina G/genética , Inmunoglobulina G/metabolismo , Inmunoglobulina G/farmacología , Ratones , Proteínas Proto-Oncogénicas c-kit/genética , Proteínas Proto-Oncogénicas c-kit/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes de Fusión/farmacología , Solubilidad , Factor de Células Madre/metabolismo , Factor de Células Madre/farmacología
5.
Blood ; 97(7): 1990-8, 2001 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-11264163

RESUMEN

Platelet-derived growth factor (PDGF)-B and PDGF beta-receptor (PDGFR beta) deficiency in mice is embryonic lethal and results in cardiovascular, renal, placental, and hematologic disorders. The hematologic disorders are described, and a correlation with hepatic hypocellularity is demonstrated. To explore possible causes, the colony-forming activity of fetal liver cells in vitro was assessed, and hematopoietic chimeras were demonstrated by the transplantation of mutant fetal liver cells into lethally irradiated recipients. It was found that mutant colony formation is equivalent to that of wild-type controls. Hematopoietic chimeras reconstituted with PDGF-B(-/-), PDGFR beta(-/-), or wild-type fetal liver cells show complete engraftment (greater than 98%) with donor granulocytes, monocytes, B cells, and T cells and display none of the cardiovascular or hematologic abnormalities seen in mutants. In mouse embryos, PDGF-B is expressed by vascular endothelial cells and megakaryocytes. After birth, expression is seen in macrophages and neurons. This study demonstrates that hematopoietic PDGF-B or PDGFR beta expression is not required for hematopoiesis or integrity of the cardiovascular system. It is argued that metabolic stress arising from mutant defects in the placenta, heart, or blood vessels may lead to impaired liver growth and decreased production of blood cells. The chimera models in this study will serve as valuable tools to test the role of PDGF in inflammatory and immune responses. (Blood. 2001;97:1990-1998)


Asunto(s)
Vasos Sanguíneos/anomalías , Enfermedades Fetales/genética , Hematopoyesis/fisiología , Proteínas Proto-Oncogénicas c-sis/fisiología , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/fisiología , Anemia/embriología , Anemia/genética , Anemia/metabolismo , Animales , Vasos Sanguíneos/embriología , Trasplante de Médula Ósea , Desarrollo Embrionario y Fetal/genética , Eritroblastosis Fetal/genética , Eritroblastosis Fetal/metabolismo , Femenino , Enfermedades Fetales/sangre , Enfermedades Fetales/patología , Corazón Fetal/anomalías , Trasplante de Tejido Fetal , Genes Letales , Prueba de Complementación Genética , Genotipo , Trasplante de Células Madre Hematopoyéticas , Inflamación , Riñón/anomalías , Riñón/embriología , Hígado/citología , Hígado/embriología , Masculino , Megacariocitos/citología , Ratones , Ratones Noqueados , Neovascularización Fisiológica/genética , Placenta/fisiopatología , Embarazo , Proteínas Proto-Oncogénicas c-sis/deficiencia , Proteínas Proto-Oncogénicas c-sis/genética , Quimera por Radiación , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/deficiencia , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Organismos Libres de Patógenos Específicos , Estrés Fisiológico/embriología , Estrés Fisiológico/genética , Estrés Fisiológico/metabolismo
6.
Cytokine ; 12(2): 127-33, 2000 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-10671297

RESUMEN

The ligand binding site of Mpl, the thrombopoietin (Tpo) receptor, has not been determined. Tyr(462)of murine Mpl corresponds to Tyr(421)of the common beta chain of the human IL-3, IL-5 and GM-CSF receptors. Tyr(421)has been identified as essential for high-affinity ligand binding. To determine whether Tyr(462)is similarly required for Tpo binding, wild-type murine Mpl (Mpl-WT) or mutant receptors containing an alanine (Y462A) or lysine (Y462K) in place of Tyr(462)were expressed in BaF3 cells. In proliferation studies, the Y462A mutation had no effect on Tpo-induced growth. In contrast, the Y462K mutation led to an attenuated proliferative response to Tpo. In single-point binding studies, both Mpl-WT and Y462A cells were able to bind [(125)I]Tpo in a specific manner. In contrast, there was a marked reduction in binding of [(125)I]Tpo by Y462K cells. Mpl-WT cells bound Tpo with a K(d)of approximately 330 pM, while Y462A cells bound Tpo with a K(d)of approximately 268 pM. The binding affinity of Y462K cells was below that quantifiable by Scatchard analysis. This study suggests that unlike the corresponding Tyr(421)of the common human beta chain, Tyr(462)of murine Mpl is not required for high-affinity ligand binding, although it may be located in proximity to the ligand binding site.


Asunto(s)
Proteínas de Neoplasias , Proteínas Proto-Oncogénicas/química , Proteínas Proto-Oncogénicas/metabolismo , Receptores de Citocinas/química , Receptores de Citocinas/metabolismo , Trombopoyetina/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Sitios de Unión/genética , División Celular/genética , Línea Celular , Cartilla de ADN/genética , Humanos , Cinética , Ligandos , Ratones , Datos de Secuencia Molecular , Mutación Puntual , Proteínas Proto-Oncogénicas/genética , Receptores de Citocinas/genética , Receptores de Trombopoyetina , Homología de Secuencia de Aminoácido , Transfección , Tirosina/química
8.
Blood ; 94(6): 1979-86, 1999 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-10477727

RESUMEN

Stem cell factor (SCF) exerts its biological effects by binding to a specific receptor, the tyrosine kinase c-Kit, which is expressed on the cell surface. Although normal cellular trafficking of growth factor receptors may play a critical role in the modulation of receptor function, the mechanisms that regulate the distribution of c-Kit on the cell surface and the internalization of c-Kit have not been fully defined. We investigated whether signal transduction via Src family kinases is required for normal c-Kit trafficking. Treatment of the SCF-responsive human hematopoietic cell line MO7e with the inhibitor of Src family kinases PP1 blocked SCF-induced capping of c-Kit and internalization of c-Kit. c-Kit was able to associate with clathrin in the presence of PP1, suggesting that entry of c-Kit into clathrin-coated pits occurs independently of Src family kinases. SCF-induced internalization of c-Kit was also diminished in the D33-3 lymphoid cell line in which expression of Lyn kinase was disrupted by homologous recombination. These results indicate that Src family kinases play a role in ligand-induced trafficking of c-Kit.


Asunto(s)
Invaginaciones Cubiertas de la Membrana Celular/fisiología , Proteínas Proto-Oncogénicas c-kit/metabolismo , Transducción de Señal/fisiología , Factor de Células Madre/fisiología , Familia-src Quinasas/metabolismo , Membrana Celular/fisiología , Quimiotaxis/efectos de los fármacos , Quimiotaxis/fisiología , Clatrina/metabolismo , Regulación Enzimológica de la Expresión Génica , Humanos , Cinética , Leucemia , Proteínas Proto-Oncogénicas c-kit/efectos de los fármacos , Pirazoles/farmacología , Pirimidinas/farmacología , Recombinación Genética , Factor de Células Madre/farmacología , Células Tumorales Cultivadas , Familia-src Quinasas/antagonistas & inhibidores , Familia-src Quinasas/genética
9.
Blood ; 94(1): 365-7, 1999 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-10381535

RESUMEN

The thrombopoietin receptor, Mpl, is a member of the cytokine receptor superfamily. The extracellular domain of Mpl contains two copies of the cytokine receptor homology module (CRM). Mpl is encoded by c-mpl, the cellular homologue of the oncogene v-mpl. The oncogenic potential of v-mpl may arise from deletion of all but the 43 most membrane-proximal amino acids of the extracellular domain of the wild-type receptor. To test the hypothesis that the extracellular domain of Mpl plays a role in controlling receptor activity, we created mutants of murine Mpl in which the membrane-distal CRM was either deleted or replaced by the membrane-proximal CRM. Introduction of these mutant receptors into factor-dependent BaF3 cells led to constitutive cell growth in the absence of growth factor. Both mutant receptors failed to bind 125I-Tpo. These results suggest that the membrane-distal CRM of Mpl acts as a brake on cell proliferation and that this region is required for ligand binding.


Asunto(s)
Transformación Celular Neoplásica/genética , Proteínas de Neoplasias , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Trombopoyetina/metabolismo , Animales , División Celular/genética , Línea Celular , Ratones , Receptores de Citocinas/genética , Receptores de Citocinas/metabolismo , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo , Receptores de Trombopoyetina , Eliminación de Secuencia
10.
Curr Opin Pediatr ; 10(1): 60-4, 1998 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-9529640

RESUMEN

Thrombopoietin is a hematopoietic growth factor that stimulates megakaryopoiesis. Recent results indicate that thrombopoietin has multilineage hematopoietic effects both in vitro and in vivo. Clinical studies of thrombopoietin have begun, and are reviewed.


Asunto(s)
Trombopoyetina/fisiología , Animales , Plaquetas/fisiología , Hematopoyesis/fisiología , Humanos , Hígado/fisiología , Ensayos Clínicos Controlados Aleatorios como Asunto , Proteínas Recombinantes , Trombopoyetina/uso terapéutico
11.
Blood ; 91(3): 898-906, 1998 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-9446650

RESUMEN

Stem cell factor (SCF) binding to the c-kit receptor triggers homodimerization and intermolecular tyrosine phosphorylation of the c-kit receptor, thus initiating signal transduction. Receptor dimerization is a critical early step in this process. Prior biochemical studies of c-kit receptor dimerization have mainly used affinity cross-linking techniques, which are beset with problems including low efficiency of cross-linking and the usual requirement for radiolabeled SCF to detect the cross-linked complex. We used the fluorescence resonance energy transfer (FRET) technique to examine the effects of SCF and other hematopoietic cytokines on c-kit receptor dimerization. The nonneutralizing anti-c-kit receptor monoclonal antibody 104D2 was directly conjugated to fluorescein isothiocyanate (FITC) or to the carbocyanine dye Cy3 and used to label cytokine-responsive human hematopoietic cell lines. The ability of SCF to induce c-kit receptor dimerization was assessed by flow cytometric analysis of FRET between the donor fluorochrome FITC and the acceptor fluorochrome Cy3. SCF induced a dose-dependent increase in c-kit receptor dimerization that correlated well with the concentrations of SCF required to stimulate cell proliferation. Receptor dimerization was detectable within 3 minutes after the addition of SCF and was maximal 30 minutes after the addition of SCF. Confocal microscopy showed redistribution of the c-kit receptor (from a diffuse distribution on the cell surface to "caps" at one end of the cell) within 3 minutes after SCF addition, followed by receptor internalization. Reappearance of the c-kit receptor on the cell surface required new protein synthesis, suggesting that the c-kit receptor is not recycled to the cell surface after internalization. Finally, erythropoietin (Epo), but not the structurally and functionally related cytokine thrombopoietin (Tpo), stimulated c-kit receptor dimerization detectable by FRET, and tyrosine phosphorylation of the c-kit receptor. These results suggest that exposure to Epo can activate the c-kit receptor and provide further evidence for cross-talk between the Epo and c-kit receptors in human hematopoietic cell lines. Studies with progeny of burst-forming unit-erythroid (BFU-E) suggest that the FRET technique is sufficiently sensitive to detect c-kit receptor dimerization on normal human hematopoietic cells.


Asunto(s)
Proteínas Proto-Oncogénicas c-kit/química , Carbocianinas , División Celular , Línea Celular , Citocinas/farmacología , Dimerización , Citometría de Flujo , Fluoresceína-5-Isotiocianato , Colorantes Fluorescentes , Células Madre Hematopoyéticas/química , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Humanos , Cinética , Microscopía Confocal , Espectrometría de Fluorescencia , Factor de Células Madre/metabolismo , Factor de Células Madre/farmacología
12.
Transplantation ; 66(12): 1770-9, 1998 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-9884275

RESUMEN

Posttransplant lymphoproliferative disorder (PTLD) has been treated with decreased immunosuppression, antiviral medications, anti-B lymphocyte agents, radiation therapy, and/or chemotherapy. However, a standardized stepwise approach to treatment has not been previously evaluated. In the present study, 19 consecutive patients presenting to a single institution with newly diagnosed PTLD were treated according to a sequential protocol that consisted of (1) a reduction in immunosuppressive medications plus, if feasible, resection or definitive radiation therapy of localized disease, (2) interferon-alpha, and (3) systemic chemotherapy. Of the 3 patients presenting exclusively with localized disease, two were treated with resection of pulmonary parenchymal nodules and one was treated with radiation therapy to a paraspinous mass, without evidence of recurrence at a mean follow-up of 31 months (range, 8 to 46 months). Sixteen patients presented with PTLD not amenable to local therapy, and they were treated daily with 3x10(6) units/m2 subcutaneous interferon-alpha. Total regression of PTLD (defined as disappearance of the tumor mass by physical examination or computed tomography scanning) was found in 8 of 14 patients who received at least 3 weeks of interferon therapy. Interferon-alpha therapy was continued for 6 to 9 months in the eight patients judged to be responders. None of these patients have relapsed to date with the same neoplastic clone. Two patients, however, developed new neoplastic clones. Seven patients received systemic chemotherapy with CHOP (cyclophosphamide, doxorubicin, vincristine, and prednisone) (n=1), EPOCH (etoposide, vincristine, and doxorubicin administered as a continuous infusion, with an intravenous bolus of cyclophosphamide and oral prednisone) (n=4), or EPOCH followed by DHAP (dexamethasone, cytarabine, and cisplatin) (n=2) after failure of interferon-alpha; five patients had a complete response. Only 1 of the 19 patients died of uncontrolled PTLD. These results suggest that the majority of solid organ transplant recipients who develop PTLD can be safely and successfully treated using a sequential approach to therapy.


Asunto(s)
Interferón-alfa/uso terapéutico , Trastornos Linfoproliferativos/terapia , Trasplante de Órganos/efectos adversos , Adulto , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Femenino , Humanos , Interferón-alfa/efectos adversos , Trastornos Linfoproliferativos/patología , Masculino , Persona de Mediana Edad , Recurrencia
14.
Blood ; 89(6): 1896-904, 1997 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-9058709

RESUMEN

Thrombopoietin (Tpo) is a major regulator of megakaryopoiesis both in vivo and in vitro. Tpo initiates its biologic effects by binding to the Mpl receptor, which is a member of the hematopoietin receptor family. To define the Tpo binding characteristics of the Mpl receptor, we iodinated purified 70-kD recombinant human Tpo using the Bolton-Hunter reagent. Autoradiographic analysis of (125)I-Tpo binding to normal human marrow mononuclear cells showed many grains specifically associated with megakaryocytes; there were no grains specifically associated with myeloblasts or erythroblasts. Equilibrium binding experiments with (125)I-Tpo and normal human platelets showed a single class of high-affinity receptors (kd, 190 pmol/L) with approximately 30 Mpl receptors per platelet. Affinity cross-linking with (125)I-Tpo showed that the Mpl receptor on platelets is of molecular weight approximately 98 kD. Despite their sequence similarity, erythropoietin and Tpo did not cross-compete for binding to BaF3 cells engineered to coexpress Mpl receptor and erythropoietin receptor. Progeny of normal human burst-forming units-erythroid (BFU-E) contained Mpl receptor mRNA, and flow cytometric analysis showed the presence of Mpl receptor protein on the surface of these cells. These data indicate that display of the Mpl receptor is not limited to the megakaryocytic lineage, but also includes progeny of BFU-E. Like receptors for other hematopoietic cytokines, the binding affinity of the Mpl receptor for Tpo is high, with relatively few receptors displayed per cell. These results suggest that the effects of Tpo to speed red blood cell recovery after myelosuppressive therapy in vivo and to enhance colony-forming unit-erythroid generation in vitro may be mediated by direct interaction of Tpo and erythroid progenitor cells.


Asunto(s)
Plaquetas/metabolismo , Proteínas de Neoplasias , Proteínas Proto-Oncogénicas/biosíntesis , Receptores de Citocinas , Trombopoyetina/sangre , Adulto , Autorradiografía , Unión Competitiva , Células de la Médula Ósea , Diferenciación Celular , Línea Celular , Ensayo de Unidades Formadoras de Colonias , Células Precursoras Eritroides/metabolismo , Humanos , Unión Proteica , Proteínas Proto-Oncogénicas/metabolismo , Receptores de Trombopoyetina
15.
Cancer ; 78(10): 2223-8, 1996 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-8918418

RESUMEN

BACKGROUND: An increased incidence of posttransplant lymphoproliferative disorders (PTLD) as a result of immunosuppression has been suggested in combined heart-lung transplantation compared with other solid organ transplants, and represents a significant source of morbidity and mortality among lung transplant patients. Although infection with the Epstein-Barr virus has been implicated in the pathogenesis of PTLD, it is unclear if the PTLD is of donor or recipient origin in lung transplant patients. METHODS: The case histories and histologic, immunophenotypic, and molecular genetic findings for the three patients who developed PTLD in the authors' transplantation program are presented. RESULTS: All three patients developed PTLD of B-cell immunophenotype with two patients having evidence of monoclonality by Southern blotting DNA gene rearrangement analysis. The site of involvement by PTLD was the lung in two patients and the small bowel in the third. In one patient, two separate pulmonary lesions were found to be comprised of distinct clonal populations, one exhibiting kappa and the other lambda light chain restriction and each having different immunoglobulin heavy chain gene rearrangements. The Epstein-Barr virus genome was present in the PTLD from all three patients. Polymerase chain reaction (PCR) amplification of a polymorphic huntington gene or annexin III locus was used to identify the PTLD in all three patients as being of recipient origin. CONCLUSIONS: PCR amplification of polymorphic genetic loci demonstrated the recipient origin of PTLD in these three pulmonary transplant patients.


Asunto(s)
Trasplante de Pulmón/efectos adversos , Trastornos Linfoproliferativos/etiología , Adulto , Southern Blotting , ADN Viral/análisis , Herpesvirus Humano 4/aislamiento & purificación , Humanos , Inmunofenotipificación , Terapia de Inmunosupresión/efectos adversos , Enfermedades Intestinales/etiología , Enfermedades Intestinales/inmunología , Enfermedades Intestinales/virología , Enfermedades Pulmonares/etiología , Enfermedades Pulmonares/inmunología , Enfermedades Pulmonares/virología , Trastornos Linfoproliferativos/inmunología , Trastornos Linfoproliferativos/virología , Masculino , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa
16.
Blood ; 88(9): 3383-90, 1996 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-8896403

RESUMEN

FLT3 ligand is a hematopoietic growth factor that plays a key role in growth of primitive hematopoietic cells. FLT3 receptor mRNA is found in early hematopoietic progenitors and in human myeloid leukemia blasts. Much less is known about the surface expression of FLT3 receptor on human hematopoietic cells. Using human 125I-FLT3 ligand, we have identified and characterized surface FLT3 receptors on normal and malignant human hematopoietic cells and cell lines. Our results showed that surface display of FLT3 receptor was greatest in fresh myeloid leukemia blast cells and myeloid leukemia cell lines. Erythroleukemic and megakaryocytic leukemia cell lines (n = 5) bound little to no 125I-FLT3 ligand. Scatchard analysis of 125I-FLT3 ligand binding data shows that three myeloid leukemia cell lines, ML-1, AML-193, and HL-60, as well as normal human marrow mononuclear cells, exhibit high affinity FLT3 receptors. Crosslinking of 125I-FLT3 ligand to FLT3 receptors on the surface of ML-1 myeloid leukemia cells indicates that the FLT3 ligand. The rates of FLT3 ligand internalization and degradation were determined by binding 125I-FLT3 ligand to ML-1 cells and acid stripping to distinguish surface bound from internalized ligand. Internalized 125I-FLT3 ligand was detected within 5 minutes after binding to ML-1 cells. In addition, we evaluated the effect of FLT3 ligand on megakaryocytic colony growth and nuclear endoreduplication, alone or in the presence of thrombopoietin. FLT3 ligand did not promote colony forming unit megakaryocyte (CFU-Meg) colony growth or megakaryocyte nuclear maturation, nor did FLT3 ligand augment the effects of thrombopoietin on these measures of megakaryopoiesis. These data indicate that the FLT3 receptor shares several characteristics with the c-kit receptor including dimerization and rapid internalization. However, the more restricted cellular distribution of the FLT3 receptor may target the effects of FLT3 ligand to primitive hematopoietic cells and to myeloid and lymphoid progenitor cells, in contrast to the pleiotropic effects of the c-kit receptor ligand, stem cell factor.


Asunto(s)
Células Madre Hematopoyéticas/metabolismo , Leucemia Mieloide/metabolismo , Proteínas Tirosina Quinasas Receptoras/biosíntesis , Autorradiografía , Línea Celular , Humanos , Ligandos , Ensayo de Unión Radioligante , Proteínas Tirosina Quinasas Receptoras/análisis
17.
Blood ; 88(6): 2026-32, 1996 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-8822921

RESUMEN

Thrombopoietin (Tpo), the ligand for the c-Mpl receptor, is a major regulator of megakaryopoiesis. Treatment of mice with Tpo raises the platelet count fourfold within a few days. Conversely, c-mpl knock-out mice have platelet counts that are 15% that of normal. The subunit structure of the c-Mpl receptor is not fully understood. Some cytokines that stimulate megakaryopoiesis (IL-6, IL-11, leukemia inhibitory factor, and oncostatin M) bind to receptors that use gp130 as a signal transduction subunit. For these reasons, we determined whether gp130 function was required for Tpo-induced signal transduction. Murine marrow cells were cultured in semi-solid media in the presence of Tpo or IL-3, with or without a neutralizing anti-gp130 monoclonal antibody (RX187) or a soluble form of c-Mpl receptor (soluble Mpl) that blocks Tpo bioactivity, and the numbers of colony-forming unit-megakaryocyte (CFU-Meg) colonies were counted on day 5. Murine marrow cells were also cultured in suspension under serum-free conditions for 5 days, and megakaryocyte DNA content was measured by flow cytometry, as an index of nuclear maturation. The addition of RX187 did not block Tpo-induced CFU-Meg colony growth nor CFU-Meg nuclear maturation in suspension culture. However, IL-3-induced CFU-Meg colony growth and megakaryocyte nuclear maturation decreased in the presence of RX187. Soluble Mpl completely ablated Tpo-induced CFU-Meg growth, and partially blocked IL-3-stimulated CFU-Meg growth. Thus the effects of Tpo on megakaryopoiesis in vitro do not depend on cytokines that signal through gp130. Furthermore, it is unlikely that gp 130 serves as a beta chain for the c-Mpl receptor, as Tpo signalling is unimpaired in the presence of RX187. In contrast, the effects of IL-3 on CFU-Meg growth are mediated in part through Tpo and through gp130-signalling cytokines.


Asunto(s)
Antígenos CD/fisiología , Hematopoyesis/efectos de los fármacos , Megacariocitos/citología , Glicoproteínas de Membrana/fisiología , Proteínas de Neoplasias , Proteínas Proto-Oncogénicas/fisiología , Receptores de Citocinas , Trombopoyetina/farmacología , Animales , Diferenciación Celular/efectos de los fármacos , Ensayo de Unidades Formadoras de Colonias , Receptor gp130 de Citocinas , ADN/metabolismo , Heterocigoto , Técnicas Inmunológicas , Interleucina-3/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Receptores de Trombopoyetina , Transducción de Señal
18.
Blood ; 88(1): 75-81, 1996 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-8704204

RESUMEN

The phenotypes of mice that harbor a defect in the genes encoding either stem cell factor (SCF) or its receptor, c-kit, indicate that this ligand/receptor pair is necessary for maintenance of normal hematopoiesis in the adult. Our objective was to determine whether SCF, like erythropoietin, is necessary for acute erythroid expansion during recovery from hemolytic anemia. Monoclonal antibody ACK2, which recognizes the murine c-kit receptor, was used to selectively block the hematopoietic growth-promoting effects of SCF. Mice were treated with phenylhydrazine on day 0 and day 1 to induce hemolytic anemia and also received no antibody, control IgG, or ACK2 on day 0. The mice were killed on day 3 and the hematocrit (Hct), reticulocyte count, and numbers of erythroid and myeloid hematopoietic progenitor cells (colony-forming unit-erythroid [CFU-E], burst-forming unit [BFU]-E, and CFU-granulocyte-macrophage [GM]) were quantitated in the femoral marrow and spleen using hematopoietic colony-forming assays. Induction of hemolytic anemia with phenylhydrazine resulted in a drop in the Hct from approximately 50% to 30%, and an approximate 8- to 10-fold increase in the reticulocyte count. The numbers of CFU-E increased modestly in the femur, and approximately 25- to 50-fold in the spleen, in comparison with normal mice. BFU-E and CFU-GM values did not increase in the femur but expanded 6- to 10-fold in the spleen, in comparison with normal mice. This confirms that much of the erythroid expansion in response to hemolytic anemia occurs in the murine spleen. Neutralizing quantities of the ACK2 antibody reduced femoral CFU-E, BFU-E, and CFU-GM content to less than half that found in phenylhydrazine-treated control mice and nearly totally ablated splenic hematopoiesis. These results suggest that c-kit receptor function may be required for optimal response to acute erythropoietic demand and that erythropoiesis in the splenic microenvironment is more dependent on SCF/c-kit receptor interaction than is erythropoiesis in the marrow microenvironment. Because expansion of late erythropoiesis in the spleen was preferentially blocked, we tested the hypothesis that homing of more primitive hematopoietic cells to the spleen was dependent on c-kit receptor function. Lethally irradiated mice were injected with marrow cells obtained from mice that had received phenylhydrazine plus control IgG or with marrow cells obtained from mice that had received phenylhydrazine plus ACK2. In parallel experiments, normal murine marrow cells were treated in vitro with control IgG or with ACK2 and were injected into lethally irradiated mice. The fraction of BFU-E and CFU-GM retrieved from the marrow and spleen of the recipient mice 4 hours later was reduced by approximately 75% when progenitor cells had been exposed to ACK2, in comparison with control IgG. These data suggest that interaction of SCF with the c-kit receptor affects the homing behavior of hematopoietic progenitor cells in the adult animal.


Asunto(s)
Células Madre Hematopoyéticas/patología , Proteínas Proto-Oncogénicas c-kit/fisiología , Bazo/patología , Factor de Células Madre/fisiología , Anemia Hemolítica/inducido químicamente , Anemia Hemolítica/patología , Animales , Anticuerpos Monoclonales/farmacología , Médula Ósea/patología , Movimiento Celular/fisiología , Ensayo de Unidades Formadoras de Colonias , Eritropoyesis/fisiología , Femenino , Células Madre Hematopoyéticas/metabolismo , Inmunoglobulina G/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Fenotipo , Fenilhidrazinas/toxicidad , Proteínas Proto-Oncogénicas c-kit/efectos de los fármacos , Quimera por Radiación , Ratas , Factor de Células Madre/antagonistas & inhibidores
19.
Blood ; 87(12): 4998-5005, 1996 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-8652812

RESUMEN

In this study, we explored whether thrombopoietin (Tpo) has a direct in vitro effect on the proliferation and differentiation of long-term repopulating hematopoietic stem cells (LTR-HSC). We previously reported a cell separation method that uses the fluorescence-activated cell sorter selection of low Hoescht 33342/low Rhodamine 123 (low Ho/low Rh) fluorescence cell fractions that are highly enriched for LTR-HSC and can reconstitute lethally irradiated recipients with fewer than 20 cells. Low Ho/low Rh cells clone with high proliferative potential in vitro in the presence of stem cell factor (SCF) + interleukin-3 (IL-3) + IL-6 (90% to 100% HPP-CFC). Tpo alone did not induce proliferation of these low Ho/low Rh cells. However, in combination with SCF or IL-3, Tpo had several synergistic effects on cell proliferation. When Tpo was added to single growth factors (either SCF or IL-3 or the combination of both), the time required for the first cell division of low Ho/low Rh cells was significantly shortened and their cloning efficiency increased substantially. Moreover, the subsequent clonal expansion at the early time points of culture was significantly augmented by Tpo. Low Ho/low Rh cells, when assayed in agar directly after sorting, did not form megakaryocyte colonies in any growth condition tested. Several days of culture in the presence of multiple cytokines were required to obtain colony-forming units-megakaryocyte (CFU-Mk). In contrast, more differentiated, low Ho/high Rh cells, previously shown to contain short-term repopulating hematopoietic stem cells (STR-HSC), were able to form megakaryocyte colonies in agar when cultured in Tpo alone directly after sorting. These data establish that Tpo acts directly on primitive hematopoietic stem cells selected using the Ho/Rh method, but this effect is dependent on the presence of pluripotent cytokines. These cells subsequently differentiate into CFU-Mk, which are capable of responding to Tpo alone. Together with the results of previous reports of its effects on erythroid progenitors, these results suggest that the effects of Tpo on hematopoiesis are greater than initially anticipated.


Asunto(s)
Células Madre Hematopoyéticas/efectos de los fármacos , Trombopoyetina/farmacología , Animales , Diferenciación Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Sinergismo Farmacológico , Femenino , Factores de Crecimiento de Célula Hematopoyética/farmacología , Células Madre Hematopoyéticas/citología , Masculino , Megacariocitos/citología , Megacariocitos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Organismos Libres de Patógenos Específicos
20.
Ann Hematol ; 72(4): 253-9, 1996 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-8624380

RESUMEN

Stem cell factor (SCF), also termed mast cell growth factor or c-kit ligand, plays a central role in the regulation of hematopoiesis and maintenance of viability of hematopoietic cells. We used a new murine monoclonal antibody (MAb) specific for canine SCF to further dissect the role of SCF in vitro and in vivo. This neutralizing MAb, RG7.6 (IgG1), recognizes the soluble form as well as the membrane-bound form of SCF on marrow-derived stromal cells. Treatment of long-term bone marrow cultures (LTMC) with RG7.6 suppressed or stimulated the production of CFU-GM, depending on the MAb concentration and the time of addition to cultures. At concentrations of 0.1-10 micrograms/ml given on the day of recharge of the LTMC, RG7.6 resulted in sustained suppression of CFU-GM grown from nonadherent cells. In contrast, higher doses of RG7.6 (20-100 micrograms/ml) led to a two- to threefold increase in CFU-GM formation from nonadherent cells after 3 days of RG7.6 exposure; after longer RG7.6 exposure there was a rapid decline in the number of CFU-GM. The early increase of CFU-GM was even more distinct when RG7.6 addition to LTMCs was delayed until 1 day before cells were plated for the CFU-GM assay. The early increase of CFU-GMs in the presence of high-dose RG7.6 was mimicked by the addition of granulocyte colony-stimulating factor (G-CSF) to cultures containing suboptimal concentrations of RG7.6, suggesting the possibility that the "positive" response to high-dose RG7.6 was due to an overriding effect of other growth factors, e.g., G-CSF. In stromal cells expressing the membrane-bound form of SCF, the presence of MAb RG7.6, even at low concentrations, interfered with thymidine uptake and proliferation. RG7.6 was also tested in vivo. RG7.6 was given intravenously immediately (days 0-4) after total body irradiation and autologous bone marrow transplantation, and granulocyte counts were followed. The post-irradiation nadir of peripheral blood granulocytes was indistinguishable from controls at low doses of RG7.6 but became more shallow as higher doses of RG7.6 were infused, again suggesting a positive effect on granulocyte differentiation. Thus, the SCF-specific MAb appears to interfere with both stromal and hematopoietic cell function. While only inhibition was observed at lower concentrations, a transient increase in granulocyte production was seen at higher MAb concentrations.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Células Madre Hematopoyéticas/fisiología , Factor de Células Madre/antagonistas & inhibidores , Células del Estroma/fisiología , Animales , Diferenciación Celular/efectos de los fármacos , Perros , Células Madre Hematopoyéticas/efectos de los fármacos , Ratones , Monocitos/citología , Monocitos/efectos de los fármacos , Factor de Células Madre/fisiología , Células del Estroma/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA