Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Blood ; 85(12): 3478-86, 1995 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-7780134

RESUMEN

Cocultivation of cells from the gamma-irradiated D2XRII murine bone marrow stromal cell line with an interleukin-3/granulocyte-macrophage colony-stimulating factor (GM-CSF)-dependent hematopoietic progenitor cell line FDC-P1JL26 stimulates the emergence of factor-independent hematopoietic cell sublines. Several lines of evidence suggested that M-CSF or a protein antigenically related to M-CSF, termed leukemogenic stromal factor (LSF), that was expressed by D2XRII cells may have played a role in the emergence of the factor-independent sublines. In an effort to isolate a factor antigenically related to M-CSF, molecular clones were isolated from a D2XRII cDNA library that hybridized to a mouse M-CSF genetic probe. Two of these molecular clones, designated 60.8.2 and 6452, contained an 885-bp deletion in the M-CSF coding region. Such a cDNA clone has not been previously described in the mouse, but a cDNA clone homologous to it has been isolated from a human pancreatic tumor cell line, MIA-PaCa-2. Three transcripts (4.8, 3.4, and 1.8 kb) were detected that hybridized to an oligonucleotide probe that was specific to RNA transcripts containing the 60.8.2 deletion. The level of the 1.8-kb transcript was not detectably induced by ionizing irradiation; however, the levels of the 4.8-kb and 3.4-kb transcripts and two other M-CSF transcripts of sizes to 4.4 kb and 2.3 kb showed a 1.4- to 2.2-fold increase after gamma irradiation. Reverse transcription-polymerase chain reaction showed that the deletion-specific transcript(s) was detected in multiple mouse bone marrow stromal cell lines and in normal mouse tissues. The present studies establish the existence of an increased spectrum of murine M-CSF transcripts in bone marrow stromal cells and other tissues. This complexity of transcripts along with their increased accumulation after irradiation provides additional evidence for a role of proteins encoded by M-CSF transcripts in the response of bone marrow stromal cells to ionizing irradiation.


Asunto(s)
Factor Estimulante de Colonias de Macrófagos/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Médula Ósea/metabolismo , Células Cultivadas , Clonación Molecular , ADN Complementario/aislamiento & purificación , Biblioteca de Genes , Humanos , Factor Estimulante de Colonias de Macrófagos/biosíntesis , Factor Estimulante de Colonias de Macrófagos/aislamiento & purificación , Ratones , Datos de Secuencia Molecular , Alineación de Secuencia
2.
Stem Cells ; 12(1): 87-94, 1994 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-8142925

RESUMEN

The effects of cytokines produced by bone marrow stromal cells on closely associated hematopoietic cells constitute a major component of the physiology of the hematopoietic microenvironment. A major cytokine produced by marrow stromal cells is macrophage colony-stimulating factor (M-CSF). To determine the effect of gamma-irradiation on the M-CSF promoter in bone marrow stromal cells, we selected a clonal cell line from the C3H/HeJ mouse marrow stromal cell line D2XRII and stably transfected a reporter construct containing the murine M-CSF-promoter linked to a chloramphenicol aminoacyl transferase (CAT) gene. CAT activity was measured at serial time points after gamma-irradiation in vitro to doses between 500 and 10,000 cGy at a dose rate of 116 cGy/min. D2XRII marrow stromal cells treated with phorbol myristate acetate (40 micrograms/ml, four h), demonstrated a significant two-fold increase in CAT activity. In contrast, CAT activity measured immediately, 24 h, 72 h or 1 week after gamma-irradiation, showed no significant increase or decrease in CAT activity. An increase in CAT activity was detected 48 h after irradiation with cells that received 5,000 cGy. Thus, single fraction gamma-irradiation of plateau phase bone marrow stromal cells did not decrease M-CSF-promoter activity. These results are consistent with prior experimental data demonstrating stable levels of release of M-CSF protein following gamma-irradiation of bone marrow stromal cells and imply that the stability of transcription of the gene for this important cytokine is protected from irradiation.


Asunto(s)
Cloranfenicol O-Acetiltransferasa/efectos de la radiación , Rayos gamma , Factor Estimulante de Colonias de Granulocitos y Macrófagos/efectos de la radiación , Animales , Médula Ósea/efectos de la radiación , Células de la Médula Ósea , Línea Celular , Cloranfenicol O-Acetiltransferasa/genética , Cloranfenicol O-Acetiltransferasa/metabolismo , Expresión Génica , Genes Reporteros/fisiología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Técnicas In Vitro , Ratones
3.
Int J Radiat Oncol Biol Phys ; 26(5): 831-6, 1993 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-8344852

RESUMEN

PURPOSE: The cytogenetic finding of the Ph1+ chromosome and its molecular biologic marker bcr/abl gene rearrangement in cells from patients with chronic myeloid leukemia are associated with a proliferative advantage of the Ph1+ clone in vivo. Although the transition to the acute terminal phase or blastic crisis is often associated with additional cytogenetic abnormalities, the molecular events which correlate the initial cytogenetic lesion with the terminal phase are poorly understood. Defective cellular DNA repair capacity is often associated with chromosomal instability, increased mutation frequency, and biologic alterations. METHODS AND MATERIALS: We, therefore, tested whether the protein product of the bcr/abl translocation (p210) could alter DNA repair after gamma-irradiation of murine cell lines expressing the bcr/abl cDNA. RESULTS: The 32D cl 3 parent, 32D cl 3 pYN (containing the control vector plasmid) and each of two sources of 32D cl 3 cells expressing p210 bcr/abl cDNA (32D-PC1 cell line and 32D-LG7 subclone) showed a D0 of 1.62, 1.57, 1.16, and 1.27 Gy, respectively. Thus, expression of the p210 bcr/abl product induced a significant (p < 0.05) increase in radiosensitivity at the clinically relevant radiation therapy dose-rate (1.16 Gy/min). The increased radiosensitivity of p210 bcr/abl expressing cells persisted if cells were held before plating in a density-inhibited state for 8 hr after gamma-irradiation, indicating little effect on the repair of potentially lethal gamma-irradiation damage. The IL-3 dependent parent 32D cl 3 cells demonstrated programmed cell death in the absence of growth factor or following gamma-irradiation to 200 cGy. Expression of bcr/abl cDNA in the 32D-PC1 and 32D-LG7 sub clones abrogated IL-3 requirement of these cell lines and inhibited gamma-irradiation induced programmed cell death. CONCLUSION: These data suggest a role for bcr/abl p210 in amplifying gamma-irradiation DNA damage or broadly inhibiting DNA repair, conditions that may stimulate further cytogenetic alterations in hematopoietic cells.


Asunto(s)
Reparación del ADN , Expresión Génica , Células Madre Hematopoyéticas/efectos de la radiación , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Tolerancia a Radiación , Translocación Genética , Biomarcadores , Humanos , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Cromosoma Filadelfia , Proteínas Proto-Oncogénicas , Genética de Radiación
4.
Leukemia ; 6(7): 626-33, 1992 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-1385639

RESUMEN

Gamma irradiation of plateau-phase clonal bone marrow stromal cell lines produces factor-independent growth of cocultivated clonal interleukin-3/granulocyte-macrophage colony-stimulating factor-dependent hematopoietic progenitor cell lines. The process is associated with three biologic changes including: (i) adherence of hematopoietic cells to stromal cells forming 'cobblestone islands'; (ii) an intermediate stage [during which the cells show proliferation in suspension in the presence in leukemogenic stromal factor (LSF), a factor similar to macrophage colony-stimulating factor (M-CSF) released by irradiated stromal cells, and transient hematopoietic cell surface expression of MAC-1, and c-fms (M-CSF receptor)]; and (iii) a third stage of factor-independence. A monoclonal antibody to M-CSF receptor inhibited proliferation of intermediate stage but not all factor-independent cell subclones. In the present studies, a subclonal factor-independent malignant subline of FDC-P1JL26 derived by cocultivation with gamma-irradiated stromal cells as well as the parent clone and intermediate stage cells were shown to express significant levels of M-CSF polyA+ mRNA and M-CSF of at least two sizes (23 and 15 kDa) as detected by 35S-methionine labelling and immunoprecipitation with polyclonal anti-M-CSF antiserum. There was no significant difference in intracellular M-CSF protein size between cells at each of the three stages of biologic change. This M-CSF was not detected on the cell surface by fluorescence-activated cell sorting (FACS). In contrast, c-fms expression at the cell surface was detected by FACS analysis and c-fms polyA+ mRNA was only detected during the intermediate stage of induction of factor-independence. FDC-P1JL26 parent cells, the subclone stimulated by LSF, and the factor-independent subclone, showed little or no detectable autophosphorylation of the c-fms receptor at tyrosine. There was no detectable rearrangement of the M-CSF or c-fms genes by Southern analysis between clonal lines during the three stages. While we cannot rule out an autocrine mechanism or mutated c-fms receptor mechanism, the data also suggest that evolution of hemopoietic cell factor-independence during cocultivation with irradiated stromal cells may involve a mechanism distal to the c-fms receptor/M-CSF interaction.


Asunto(s)
Genes fms , Células Madre Hematopoyéticas/fisiología , Factor Estimulante de Colonias de Macrófagos/metabolismo , Receptor de Factor Estimulante de Colonias de Macrófagos/metabolismo , Animales , Southern Blotting , Células de la Médula Ósea , Línea Celular , Expresión Génica , Reordenamiento Génico , Genes , Sustancias de Crecimiento/metabolismo , Humanos , Factor Estimulante de Colonias de Macrófagos/genética , Ratones , Neoplasias Experimentales/genética , Fosforilación , ARN Mensajero/genética , Transducción de Señal
5.
Radiat Res ; 129(3): 297-303, 1992 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-1311863

RESUMEN

Infection of the IL-3-dependent, myeloid progenitor cell line 32D cl 3 with murine retroviruses that contain either the wild-type or a temperature-sensitive mutant v-src can render these cells growth-factor independent. These cells also became resistant to gamma irradiation administered at the low-dose rate of 0.05 Gy/min, which is used clinically. The v-src-dependent nature of resistance to gamma irradiation was examined by studying four clones of 32D cl 3 cells that had been infected with a retrovirus carrying the tsLA31A mutant of v-src. The tyrosine-specific kinase activity of this mutant is dramatically reduced at the nonpermissive temperature of 39 degrees C. Cells transformed by v-src and grown at either 34 or 39 degrees C, in the presence or absence of IL-3, demonstrated a significantly higher D0 compared to parental cells examined under identical conditions. In addition, expression of v-src abrogated the synergistic killing effect of heat and gamma irradiation. The D0 of parental 32D cl 3 cells kept at 39 degrees C after gamma irradiation was reduced significantly compared to the D0 of these cells kept at 34 degrees C. This contrasts with data from 32D cl 3 cells infected with either the wild-type v-src or the temperature-sensitive mutant, neither exhibited a synergistic effect in the D0 at either 34 or 39 degrees C. Therefore, while continuous expression of a v-src gene product is required for maintenance of the growth-factor-independent state, v-src does not appear to be responsible for the increased gamma-radiation resistance of these cells at low dose rate.


Asunto(s)
Expresión Génica , Células Madre Hematopoyéticas/efectos de la radiación , Oncogenes/genética , Tolerancia a Radiación/genética , Animales , Línea Celular , Supervivencia Celular/efectos de la radiación , Relación Dosis-Respuesta en la Radiación , Calor , Interleucina-3/fisiología , Ratones , Mutación , Infecciones por Retroviridae
6.
Exp Hematol ; 20(1): 92-102, 1992 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-1533594

RESUMEN

Gamma-irradiation of plateau phase cultures of the clonal murine bone marrow stromal cell line D2XRII followed by cocultivation of a clonal interleukin 3 (IL-3) (granulocyte-macrophage colony-stimulating factor (GM-CSF)-dependent hematopoietic progenitor cell line FDC-P1JL26 results in a significant increase in "cobblestone islands" of attachment and emergence of subclonal factor-independent malignant sublines. Biochemical purification of conditioned medium from irradiated D2XRII cells yielded a 75,000-dalton glycoprotein termed leukemogenic stromal factor (LSF) that was neutralized by a polyclonal antiserum to murine macrophage colony-stimulating factor (M-CSF). A monoclonal antibody to the murine M-CSF receptor (c-fms) neutralized the biological activity of this molecule in a manner comparable to its effect on recombinant human or murine M-CSF. FDC-P1JL26 parent cells were positive for Ly5, MEL-14, mGR, VLA-4, PGP-1 (CD44), and Thy1.2. After culture in LSF, Thy1.2, MEL-14, and mGR became undetectable; however, significant cell surface MAC-1 antigen and c-fms (M-CSF receptor) were expressed. Neither line was positive for Ly6, Ly22, I-CAM-1, or B220 antigen. LSF-precultured FDC-P1JL26 cells transferred as single cells to microwell culture with 5000-cGy-irradiated D2XRII cells revealed a 60-fold increase in frequency of cobblestone island formation and evolution of factor-independent subclones compared to the parent line. Both parent and LSF-precultured cells became factor independent at a 100-fold lower frequency if kept in suspension in LSF in the absence of stromal cells. Antiserum to M-CSF or monoclonal antibody to the murine M-CSF receptor (c-fms) did not inhibit or displace cobblestone island formation by either clone of FDC-P1 on irradiated stromal cells indicating a mechanism of binding not involving the M-CSF receptor. However, anti-serum to the M-CSF receptor inhibited growth of one factor-independent subclone. In separate studies, a subclone of IL-3-dependent 32Dc13 cells, expressing the transfected murine c-fms protooncogene but not the parent 32Dc13 cell line or another subclone expressing the transfected gene for the human M-CSF receptor, showed adherence and became factor independent when cocultivated with irradiated D2XRII stromal cells. Thus, irradiated stromal cells bind M-CSF receptor-positive hematopoietic progenitor cells and induce c-fms-dependent factor-independent tumorigenic subclones. The cellular interactions in this model may be relevant to gamma-irradiation leukemogenesis in vivo.


Asunto(s)
Células de la Médula Ósea , Médula Ósea/efectos de la radiación , Leucemia Experimental/etiología , Neoplasias Inducidas por Radiación/etiología , Animales , Anticuerpos Monoclonales , Médula Ósea/fisiología , Comunicación Celular/fisiología , Membrana Celular/fisiología , Membrana Celular/ultraestructura , Separación Celular , Células Cultivadas , Fluorescencia , Rayos gamma , Glicoproteínas/farmacología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Hematopoyesis/efectos de los fármacos , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/fisiología , Células Madre Hematopoyéticas/efectos de la radiación , Interleucina-3/farmacología , Leucemia Experimental/patología , Factor Estimulante de Colonias de Macrófagos/farmacología , Antígeno de Macrófago-1/análisis , Ratones , Neoplasias Inducidas por Radiación/patología , Fenotipo , Receptor de Factor Estimulante de Colonias de Macrófagos/análisis , Receptor de Factor Estimulante de Colonias de Macrófagos/fisiología
7.
Exp Hematol ; 18(5): 408-15, 1990 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-2186923

RESUMEN

The hematopoietic and stromal cell-specific properties of the cells involved in gamma irradiation leukemogenesis in vitro were defined. Cocultivation of clonal factor-dependent (FD), interleukin 3 (IL-3)-dependent cell lines 32D cl 3 or B6SUtA, or dual IL-3-/granulocyte-macrophage colony-stimulating factor (GM-CSF)-dependent cell lines FDC-P1 or bg/bg d64 was carried out with clonal stromal cell lines D2XRII, GB1/6, +/+ 2.4, or Sld3. FD cell lines were added to control or 5000-cGy-irradiated plateau phase monolayer cultures of each stromal cell line, and parameters of stem cell engraftment and malignant transformation in vitro were quantitated. Cobblestone island formation by FD cells, cumulative production of nonadherent hematopoietic cells, and evolution of tumorigenic factor-independent (FI) subclonal lines were quantitated over 5-8 weeks. There was no detectable evolution of FI sublines with 32D cl 3, B6SUtA, or bg/bg d64 cells cocultivated with control or irradiated Sld3 stromal cells. IL-3-dependent cell lines 32D cl 3 or B6SUtA formed small 10- to 49-cell cobblestone "clusters" at low frequency on control or irradiated D2XRII, showed limited proliferation for less than 1 week, and showed no detectable evolution of FI cell lines. Subclones of 32D cl 3 derived by transfection and expression of recombinant oncogenes v-sis, or c-myc, or the epidermal growth factor receptor remained factor dependent and did not transform to factor independence after cocultivation with irradiated stromal cell lines. In contrast, cell line bg/bg d64, and each of seven subclonal lines of FDC-P1, including subclones selected for growth in GM-CSF, formed abundant cobblestone island colonies of greater than or equal to 50 cells on irradiated D2XRII stromal cells, produced non-adherent cells over 5-8 weeks, and showed evolution of tumorigenic FI subclonal lines. The data provide evidence for stable biological differences in both the hematopoietic and stromal cell components of the in vitro model of gamma irradiation leukemogenesis.


Asunto(s)
Médula Ósea/patología , Células Madre Hematopoyéticas/fisiología , Leucemia Inducida por Radiación/patología , Animales , Adhesión Celular , División Celular , Línea Celular , Transformación Celular Neoplásica , Células Clonales , Factores Estimulantes de Colonias/farmacología , ADN/biosíntesis , Rayos gamma , Factor Estimulante de Colonias de Granulocitos y Macrófagos , Sustancias de Crecimiento/farmacología , Interleucina-3/farmacología , Ratones , Proteínas Recombinantes/farmacología
8.
Int J Radiat Oncol Biol Phys ; 18(5): 1083-92, 1990 May.
Artículo en Inglés | MEDLINE | ID: mdl-2347717

RESUMEN

There is evidence for differences in the gamma-irradiation response of different cellular lineages within the bone marrow microenvironment. We previously reported that heterogeneity is demonstrable in the gamma-irradiation response of five clonal stromal cell lines, derived from one human bone marrow specimen, despite morphological, histochemical, cytogenetic, and functional similarity. In the present study we tested whether one stromal cell line could affect the intrinsic radiosensitivity of another. Two clonal stromal cell lines, which display distinct gamma-irradiation responses relative to dose rate were used: KM 101, which shows the same radiosensitivity at a low dose rate of 5 cGy/min (LDR) and a high dose rate of 120 cGy/min (HDR) and KM 104 which shows significant gamma-irradiation resistance at LDR. To facilitate the study of the gamma-irradiation response of each cell line during cocultivation, we derived stable subclones of each, expressing the transfected neomycin resistance (neo-r) gene, which confers resistance to the neomycin analog: G 418. Introduction of the neo-r gene did not alter cell lines radiosensitivity. The results show that cocultivation of stromal cell lines before, during, and after gamma-irradiation induces changes in repair of radiation-induced damage, with a dominant effect of a resistant cell line at LDR. In fact, the radiation survival curves of cocultivated stromal cell lines were always characteristic of KM 104, and a dose rate effect was observed, even when KM 101 was present in large excess. Moreover, our results are consistent with preferential killing of the more radiosensitive stromal cell line: both LDR and HDR Do values of the neo-r KM 101, cocultivated with the parent KM 104 for 24 hr before, and during gamma irradiation were significantly lower compared to the neo-r subclone irradiated alone. The LDR Do value of the neo-r KM 104 cocultivated for 24 hr before, and during gamma irradiation with excess of parent KM 101, was significantly higher, compared to the neo-r cells irradiated alone.


Asunto(s)
Médula Ósea/efectos de la radiación , Tolerancia a Radiación , Línea Celular , Supervivencia Celular/efectos de la radiación , Células Clonales , Ensayo de Unidades Formadoras de Colonias , Relación Dosis-Respuesta en la Radiación , Rayos gamma , Humanos
9.
Radiat Res ; 122(1): 44-52, 1990 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-2320725

RESUMEN

To determine the effect of oncogene expression on gamma radiation sensitivity of hematopoietic compared to fibroblastic cells, we selected clonal sublines of an interleukin-3 (IL-3)-dependent hematopoietic progenitor cell line 32D cl 3 and NIH/3T3 embryo fibroblastic cells following transfection with each oncogene linked to the mycophenolic acid resistance gene. Each mycophenolic acid-resistant subclone demonstrated high levels of specific poly(A)+ mRNA for each oncogene. The parent line 32D cl 3 demonstrated similar radiosensitivity at 116 cGy/min (D0 126, n 1.17) compared to 5 cGy/min (D0 123, n 1.65). This pattern was not altered in subclones of 32D cl 3 cells transfected with the epidermal growth factor (EGF) receptor gene and grown in EGF (at 116 cGy/min D0 104, n 0.998, at 5 cGy/min D0 115, n 1.09), or in 32D cl 3 cells expressing the v-sis oncogene (at 116 cGy/min D0 122.4, n 1.79, at 5 cGy/min D0 135, n 1.43). In contrast, expression of the transfected oncogenes v-erb-B, v-abl, or v-src conferred significant radioresistance at 5 cGy/min dose rate (D0 194, n 1.77; D0 165.5, n 1.56; D0 171, n 1.28, respectively). With the exception of v-sis, oncogene expression resulted in nonautocrine factor independence of 32D cl 3 subclones, and production of donor origin tumors in syngeneic new-born or adult mice. Two rare spontaneous factor-independent subclones of 32D cl 3 were also tested. Nonautocrine clone 32D cl 2 demonstrated significantly increased radioresistance at low dose rate (D0 186, n 1.63), while autocrine (IL-3 producing) subclone 32D cl 4 revealed no significant increase in radioresistance at 5 cGy/min. The parent fibroblast cell line NIH/3T3 showed an intrinsic relative radioresistance at low dose rate (at 5 cGy/min D0 157.3, n 1.81, compared to 116 cGy/min D0 134.3, n 1.57). Expression in NIH/3T3 of transfected oncogenes v-abl, v-fms, v-fos, or H-ras increased radioresistance at low dose rate (D0 208.6, n 1.61; D0 206.6, n 1.51; D0 167.5, n 1.85; and D0 206.8, n 1.08, respectively). Thus expression of each of several oncogenes induces resistance to gamma irradiation at 5 cGy/min in hematopoietic and fibroblast cell lines. These data may help explain the clinical recurrence of oncogene-expressing leukemia and lymphoma cells after marrow stem cell ablative doses of low-dose-rate total-body irradiation.


Asunto(s)
ADN Recombinante , Células Madre Hematopoyéticas/efectos de la radiación , Oncogenes , Tolerancia a Radiación , Animales , Línea Celular , Supervivencia Celular , Relación Dosis-Respuesta en la Radiación , Fibroblastos/efectos de la radiación , Células Madre Hematopoyéticas/efectos de los fármacos , Humanos , Interleucina-3/farmacología , Proteínas Recombinantes/farmacología , Transfección
10.
Radiother Oncol ; 15(3): 285-93, 1989 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-2772256

RESUMEN

The relative radioresponsiveness of human prostate cancer compared to malignant melanoma is well known. The effects of beta-estradiol or testosterone on the X-irradiation survival of several human cell lines were studied, including: human prostate carcinoma cell lines PC3 and DU145 and human malignant melanoma cell lines A375 and A875. Lines PC3 and DU145 demonstrated 55-61 fmol per 10(6) cells of androgen receptor with no detectable estrogen or progesterone receptor. Cells were irradiated at 120 cGy/min dose rate. There was no detectable toxicity of up to 10(-4) M testosterone or beta-estradiol on PC3 or DU145 cells in the absence of X-irradiation. At plating efficiencies from 11-13%, and plating densities of 1 x 10(4) cells per 60 cm2 flask, cell lines PC3 and DU145 demonstrated a Do of 108.5 +/- 6.5, n 2.1 +/- 0.7 cGy, and Do of 143.5 +/- 1.5 cGy, n 2.4 +/- 0.5, respectively. The addition of testosterone or beta-estradiol at 10(-4) to 10(-10) M prior to or after, X-irradiation did not alter radiosensitivity. At the same dose rate of 120 cGy/min, malignant melanoma cell lines A375 and A875 had a Do of 125 +/- 2.5 cGy, n 1.56 +/- 0.8 SF2 0.65 +/- 0.03 and line A875 demonstrated a Do of 129 +/- 4.5 cGy, n 1.58 +/- 0.4 SF2 0.55 +/- 0.04, respectively. The radiosensitivity of melanoma cell lines did not decrease at low dose rate 5 cGy/min. Thus, the in vitro radiosensitivity of androgen receptor positive prostate cancer cell lines is not necessarily altered by the presence of androgen before or after irradiation. The data support the concept that all malignant melanoma cell lines do not show a broad-shouldered cell survival curve in vitro and intrinsic cellular radioresistance.


Asunto(s)
Melanoma/patología , Neoplasias de la Próstata/patología , Tolerancia a Radiación , Supervivencia Celular/efectos de la radiación , Estradiol/farmacología , Humanos , Masculino , Testosterona/farmacología , Células Tumorales Cultivadas/efectos de la radiación
11.
Acta Haematol ; 82(3): 136-43, 1989.
Artículo en Inglés | MEDLINE | ID: mdl-2554632

RESUMEN

Stability and expression of the bacterial neomycin resistance gene (neor) transferred to human continuous marrow cultures by a retroviral vector [pZIP-NeoSV(X)] was evaluated over 4 weeks. Following infection of long-term human marrow cultures with pZIP-NeoSV(X), 10-15% of the stromal cells demonstrated high replating efficiency in a dose of the neomycin analogue G418 that was toxic to stromal cells from uninfected cultures. In contrast, G418 resistance was detected in less than or equal to 1% of GM-CFUc and CFU-GEMM derived from the same virus-infected compared to control cultures. Infection of human CFU-GEMM enriched 100 X by monoclonal antibody selection with pZIP-NeoSV(X) did not increase the percentage of neor progenitors. Marrow cells from cultures infected with pZIP-NeoSV(X) and a replication competent amphotropic virus transferred the vector and G418 resistance to HeLa cells at a frequency of 1/10(5) for nonadherent and 1/10(4) for adherent cells. Two established human hematopoietic (HL60 and K562) and one stromal cell line (KM101) stably expressed the neor gene. Thus, a higher efficiency of infection and expression of a gene transferred by pZIP-NeoSV(X) to permanent human hematopoietic tumor cell lines and fresh marrow stromal cells contrasts with a lower level of expression in fresh CSF-dependent human hematopoietic stem cells.


Asunto(s)
Médula Ósea/metabolismo , Neomicina/farmacología , Fosfotransferasas/genética , Transfección , Células Cultivadas , Expresión Génica , Vectores Genéticos , Células Madre Hematopoyéticas , Humanos , Kanamicina Quinasa , Retroviridae/genética
12.
Int J Radiat Oncol Biol Phys ; 15(5): 1153-9, 1988 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-3182348

RESUMEN

In contrast to the dose-rate independent X ray killing observed with human bone marrow hematopoietic stem cells, bone marrow adherent stromal cells from the same fresh marrow harvests demonstrate increased radiation resistance at low dose rate (LDR) (5 cGy/min), compared to high dose rate (HDR) irradiation (120-200 cGy/min). Physiologic changes observed in plateau phase bone marrow cells after LDR irradiation in vivo and in vitro suggested that marrow stromal cells might be heterogeneous in LDR irradiation repair. Five permanent clonal bone marrow stromal lines were derived from a single human marrow donor. Each cell line was positive for markers of fibroblasts including: immunohistochemically detectable fibronectin, collagen, acid phosphatase, and nonspecific esterase, and was negative for Factor VIII, alkaline phosphatase, lysozyme and several markers of marrow macrophages. The x-irradiation survival curve of each cell line was determined at LDR and HDR in vitro. Cell lines KM102, KM103, KM104, and KM105 each demonstrated a significant (p less than .05) increase in radioresistance at LDR (D0 = 142, n = 2.9; D0 = 131, n = 2.5; D0 = 145, n = 2.1 and D0 = 127, n = 2.1 respectively) compared to HDR: (D0 = 111, n = 2.1; D0 = 94, n = 3.5; D0 = 99, n = 3.5 and D0 = 95, n = 2.1 respectively). In contrast, cell line KM101 demonstrated no significant change in radiosensitivity relative to dose rate at LDR (D0 = 113, n = 3.3) compared to HDR, D0 = 114, n = 3.3. Cell line KM101 was more supportive than the other lines of cocultivated hemopoietic cells in vitro. Subclones of KM101 and KM104 selected by retroviral vector transfer of the neor gene for growth in the antibiotic neomycin-analogue G418, maintained the stably associated radiobiologic properties of each parent clonal line. These data indicate significant heterogeneity in the LDR irradiation response of clonal stromal cell lines derived from human bone marrow.


Asunto(s)
Médula Ósea/efectos de la radiación , Dosis de Radiación , Tolerancia a Radiación , Células de la Médula Ósea , Línea Celular , Supervivencia Celular/efectos de la radiación , Relación Dosis-Respuesta en la Radiación , Humanos , Técnicas In Vitro
13.
Exp Hematol ; 16(10): 820-6, 1988 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-3049129

RESUMEN

The x-irradiation biology of supportive stromal cells of the bone marrow microenvironment was investigated by using cloned permanent cell lines that were established from hematopoietically active murine long-term bone marrow cultures. X-irradiation survival curves were derived for each cell line at either 120 cGy/min or clinical low dose rate (LDR) (5 cGy/min) that is used in total body irradiation protocols prior to bone marrow transplantation. Four cell lines, MBA-1, MBA-13, 14F2.1, and D2XRII (Group I) demonstrated a significant increase in D0 at 5 cGy/min (280 cGy, 270 cGy, 210 cGy, and 240 cGy, respectively) compared to 120 cGy/min (215 cGy, 210 cGy, 157 cGy, and 210 cGy, respectively) (p less than 0.05). In contrast, three other clonal cell lines, +/+ #2 cl 4, S1d #3, and GPI alpha-1 (Group II) showed no significant dose rate dependent change in D0 at 5 cGy/min (164 cGy, 174 cGy, and 159 cGy, respectively) compared to 120 cGy/min (159 cGy, 167 cGy, and 143 cGy, respectively). Group I and II cell lines could not be distinguished by differences in synthesis of extracellular matrix proteins including laminin, collagen types I and IV, or histochemically detectable enzymes. Three Group I lines (MBA-1, MBA-13, and D2XRII) and one Group II line, Sld #3, showed decreased support capacity for cocultivated hematopoietic stem cells in vitro. All seven lines had detectable polyA+ mRNA for monocyte colony-stimulating factor (M-CSF) as detected by molecular hybridization; one had detectable levels of polyA+ mRNA for granulocyte-macrophage colony-stimulating factor (GM-CSF) (D2XRII); one, detectable levels of polyA+ mRNA for interleukin 1 (IL-1); and none of the seven had detectable polyA+ mRNA for granulocyte colony-stimulating factor (G-CSF) or IL-3 (multi-CSF). The data indicate that some cells of the hematopoietic microenvironment may not be selectively protected by clinical low dose rate irradiation.(ABSTRACT TRUNCATED AT 250 WORDS)


Asunto(s)
Médula Ósea/efectos de la radiación , Tejido Conectivo/efectos de la radiación , Tolerancia a Radiación , Animales , Médula Ósea/metabolismo , Trasplante de Médula Ósea , Línea Celular , Supervivencia Celular , Colágeno/metabolismo , Tejido Conectivo/metabolismo , Relación Dosis-Respuesta en la Radiación , Fibronectinas/metabolismo , Supervivencia de Injerto/efectos de la radiación , Laminina/metabolismo , Ratones , Factores de Tiempo
14.
Int J Radiat Oncol Biol Phys ; 14(1): 85-94, 1988 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-2891657

RESUMEN

The mechanism of physiologic alteration by high (HDR) or low dose rate (LDR) (5 or 120 cGy/min) irradiation of plateau-phase bone marrow stromal cell cultures was investigated using a technique of in vitro bone marrow transplantation. Purified stromal cell cultures from C57BL/6J, C3H/HeJ, or (C57BL/6J X DBA2/J)F1 (B6D2F1) mouse marrow were irradiated to doses of 2.5 to 10 Gy at LDR or 10-100 Gy at HDR and were then engrafted in vitro with nonadherent hematopoietic cells from murine continuous bone marrow cultures. Parameters of engraftment quantitated included: (1) numbers of adherent proliferating hematopoietic cell colonies, "cobblestone islands" (2) cumulative production of nonadherent hematopoietic cells over 8 weeks after engraftment, (3) M-CSF, GM-CSF and multi-CSF (IL-3) dependent hematopoietic progenitor cells forming greater than or equal to 50 cell colonies in semisolid medium, (4) cumulative production of CFUs, and (5) number of adherent stromal cells positive for detectable extracellular laminin or collagen type IV (markers of endothelial cells, reticular adventitial cells, or sinus lining cells). There was a decrease in cobblestone island formation between 5 and 10 Gy and this parameter possibly increased at doses of 50 and 100 Gy. There was no difference between HDR and LDR irradiation to 10 Gy. Irradiation to doses above 10 Gy decreased support of engrafted cells forming CFU-GM and CFU-GEMM. Measures of CFUs after 10 Gy were variable but indicated a possible increase with HDR and no effect of LDR at 1 week and a decrease in both HDR and LDR groups at 3 weeks after engraftment. Thus, LDR and HDR irradiation in vitro alter several specific parameters of marrow stromal cell support for engrafted hematopoietic stem cells.


Asunto(s)
Médula Ósea/efectos de la radiación , Células Madre Hematopoyéticas/efectos de la radiación , Animales , Trasplante de Médula Ósea , Adhesión Celular/efectos de la radiación , Células Cultivadas , Cruzamientos Genéticos , Relación Dosis-Respuesta en la Radiación , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/citología , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Endogámicos
15.
Proc Natl Acad Sci U S A ; 84(23): 8662-6, 1987 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-2825203

RESUMEN

To analyze the transcriptional activity of retroviral enhancer sequences in hematopoietic lineages, we determined the effect of enhancer sequences on the expression of the neomycin resistance gene transferred by two retroviral vectors to primary hematopoietic lineages. We constructed the vector pFr-SV(X). The Moloney murine leukemia virus enhancer region of a vector, pZIP-SV(X), was replaced by a 380-nucleotide-long fragment containing the enhancer sequences of the Friend murine leukemia virus. The enhancer sequences of Friend murine leukemia virus were used because these sequences have been shown to target the disease specificity of the virus to the erythroid lineage. Hematopoietic progenitors in murine continuous marrow cultures were infected with identical numbers of pure defective, infectious viral vector particles of either pFr-SV(X) or pZIP-SV(X). Expression of the transferred neomycin resistance gene in multipotential stem cells and their differentiated progeny was assayed as the ability of infected progenitors to form colonies (greater than 50 cells) in G418. Expression of the neomycin resistance gene in multipotential progenitor cells during the entire 11 weeks of the cultures was independent of the vector used to transfer the gene. Conversely, committed hemoglobinized erythroid bursts and myeloid colonies resistant to G418 were consistently produced by pFr-SV(X)-infected cultures but not pZIP-SV(X)-infected cultures. These results demonstrate that both pFr-SV(X) and pZIP-SV(X) were stably integrated and expressed in more primitive, multilineage, hematopoietic progenitor cells and suggest that the enhancer sequences of a vector affects expression of the transferred neomycin resistance gene when these cells differentiate to committed myeloid and erythroid cells.


Asunto(s)
Elementos de Facilitación Genéticos , Eritropoyesis , Células Madre Hematopoyéticas/fisiología , Virus de la Leucemia Murina/genética , Animales , Células Cultivadas , Resistencia a Medicamentos , Virus de la Leucemia Murina de Friend/genética , Regulación de la Expresión Génica , Vectores Genéticos , Ratones , Virus de la Leucemia Murina de Moloney/genética , Neomicina/farmacología , Transfección
16.
Proc Natl Acad Sci U S A ; 84(21): 7681-5, 1987 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-2890167

RESUMEN

Whether bone marrow stromal cells of donors contribute physiologically to hematopoietic stem cell reconstitution after marrow transplantation is unknown. To determine the transplantability of nonhematopoietic marrow stromal cells, stable clonal stromal cell line (GB1/6) expressing the a isoenzyme of glucose-6-phosphate isomerase (Glu6PI-a, D-glucose-6-phosphate ketol-isomerase; EC 5.3.1.9) was derived from murine long-term bone marrow cultures and made resistant to neomycin analogue G418 by retroviral gene transfer. GB1/6 cells were fibronectin+, laminin+, and collagen-type IV+ and collagen type I-; these GB1/6 cells supported in vitro growth of hematopoietic stem cells forming colony-forming units of spleen cells (CFU-S) and of granulocytes, erythrocytes, and macrophage/megakarocytes (CFU-GEMM) in the absence of detectable growth factors interleukin 3 (multi-colony-stimulating factor), granulocyte/macrophage colony-stimulating factor, granulocyte-stimulating factor, or their poly(A)+ mRNAs. The GB1/6 cells produced macrophage colony-stimulating factor constitutively. Recipient C57BL/6J (glucose-6-phosphate isomerase b) mice that received 3-Gy total-body irradiation and 13 Gy to the right hind limb were injected i.v. with GB1/6 cells. Engrafted mice demonstrated donor-originating Glu6PI-a+ stromal cells in marrow sinuses in situ 2 mo after transplantation and a significantly enhanced hematopoietic recovery compared with control irradiated nontransplanted mice. Continuous (over numerous passages) marrow cultures derived from transplanted mice demonstrated G418-resistant, Glu6PI-a+ stromal colony-forming cells and greater cumulative production of multipotential stem cells of recipient origin compared with cultures established from irradiated, nontransplanted control mice. These data are evidence for physiological function in vivo of a transplanted bone marrow stromal cell line.


Asunto(s)
Trasplante de Médula Ósea , Hematopoyesis/efectos de la radiación , Trasplante de Células Madre Hematopoyéticas , Animales , Células de la Médula Ósea , Línea Celular , Células Cultivadas , Células Madre Hematopoyéticas/citología , Cinética , Ratones , Ratones Endogámicos C57BL , Irradiación Corporal Total
17.
Exp Hematol ; 15(6): 636-44, 1987 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-3595763

RESUMEN

Permanent cloned bone marrow stromal cell lines, designated Sld1, Sld2, Sld3, were derived from long-term bone marrow cultures (LTBMC) of Sl/Sld mice and littermate WCB6F1 mice (designated +/+1, +/+2, +/+1.10, and +/+2.4). Production of extracellular matrix proteins by these cell lines was detected using specific antibodies. Fibronectin, laminin, and collagen type IV were detected in all clonal cell lines tested. Each cloned stromal cell line or parent stromal cell culture was also tested in vitro for support of engrafted hemopoietic stem cells. Engrafted hemopoietic stem cells from C57BL/6J LTBMCs forming CFU-GEMM and BFUe colonies were supported at 76% and 44% efficiency by lines +/+2.4 and +/+1.10, respectively, compared to the number recovered from parent uncloned C57BL/6J stromal cell cultures. In contrast, cell lines Sld1 and Sld3 were significantly less supportive of CFU-GEMM progenitor cells (14% and 18% efficiency, respectively) and erythroid progenitors (6% and less than 0.1% efficiency, respectively). Some Sld lines also showed reduced support compared to +/+ cell lines of a clonal multipotential erythroid cell line B6SUtA. These permanent stromal cell lines should be useful in elucidation of the specific microenvironmental factors required for support of multilineage and erythroid progenitor cells.


Asunto(s)
Anemia Macrocítica/genética , Ensayo de Unidades Formadoras de Colonias , Células Madre Hematopoyéticas/citología , Ratones Endogámicos/genética , Animales , Células de la Médula Ósea , Comunicación Celular , Diferenciación Celular , Línea Celular , Células Clonales/citología , Eritropoyesis , Granulocitos/citología , Hematopoyesis , Macrófagos/citología , Ratones , Factores de Tiempo
18.
Scanning Microsc ; 1(1): 247-53, 1987 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-3589603

RESUMEN

Plateau-phase mouse clonal bone marrow stromal cell lines D2XRII and C3H cl 11 produce decreasing levels of M-CSF (CSF-1), a specific macrophage progenitor cell humoral regulator, following X-irradiation in vitro. The decrease did not go below 40% of control levels, even after irradiation doses of 50,000 rad (500 Gy). In contrast, a distinct humoral regulator stimulating growth of GM-CSF/IL-3 factor-dependent (FD) hematopoietic progenitor cell lines was detected following radiation to doses above 2,000 rad. This humoral factor was not detectable in conditioned medium from irradiated cells, weakly detected using factor-dependent target cell populations in agar overlay, and was prominently detected by liquid co-cultivation of factor-dependent cells with irradiated stromal cell cultures. Subclonal lines of FD cells, derived after co-cultivation revealed karyotypic abnormalities and induced myeloblastic tumors in syngeneic mice. Five-eight weeks co-cultivation was required for induction of factor independence and malignancy and was associated with dense cell to cell contact between FD cells and stromal cells demonstrated by light and electron microscopy. Increases in hematopoietic to stromal cell surface area, total number of adherent cells per flask, total non-adherent cell colonies per flask, and cumulative non-adherent cell production were observed after irradiation. The present data may prove very relevant to an understanding of the cell to cell interactions during X-irradiation-induced leukemia.


Asunto(s)
Médula Ósea/efectos de la radiación , Transformación Celular Neoplásica , Células Madre Hematopoyéticas/efectos de la radiación , Animales , Células de la Médula Ósea , Línea Celular , Células Cultivadas , Bandeo Cromosómico , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/ultraestructura , Cariotipificación , Ratones , Ratones Endogámicos , Microscopía Electrónica
19.
Exp Hematol ; 15(2): 195-202, 1987 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-3817049

RESUMEN

Retroviral-mediated gene transfer to multipotent and committed hematopoietic stem cells and marrow stromal cells was evaluated in long-term bone marrow cultures (LTBMCs). The retroviral vector pZIP-Neo(SV)(X) carrying the bacterial neomycin resistance (neor) gene that confers resistance to the neomycin analog G418 in mammalian cells was packaged in a Moloney envelope either as a replication-competent or replication-defective virus. Virus was introduced by infection of long-term marrow cultures at day 7. During a period of 12 weeks in culture, 10%-50% of harvested hematopoietic progenitor cells that formed differentiated CFU-GEMM colonies in response to pokeweed mitogen-containing spleen cell-conditioned medium (SCCM) and erythropoietin expressed the neor gene. In contrast, 1%-10% of hematopoietic progenitor cells that formed colonies in agar in response to WEHI-3B- or L-cell-conditioned medium expressed resistance to G418. The percentage of resistant progenitors was not detectably enhanced when replication-competent Moloney murine leukemia virus (M-MuLV) was present as helper virus, even though M-MuLV infected greater than 90% of cells in the long-term marrow cultures. In a separate CFU-F assay, 12%-17% of the adherent stromal cells in LTBMCs were found to express the neor gene. Thus gene transfer is limited by the fraction of progenitor cells that can integrate and express the transferred genetic sequences, rather than by the fraction of cells that are initially infected by the vector.


Asunto(s)
Células de la Médula Ósea , Regulación de la Expresión Génica , Animales , Transformación Celular Viral , Células Cultivadas , Genes Dominantes , Ratones , Retroviridae/fisiología
20.
Leuk Res ; 11(1): 51-61, 1987.
Artículo en Inglés | MEDLINE | ID: mdl-3492638

RESUMEN

C3H/HeJ mouse long-term bone marrow cultures infected at initiation with a cloned polycythemic strain of Friend spleen focus forming virus in a cloned N-tropic murine leukemia virus helper virus coat, persistently produced: colony-forming unit spleen (CFUs) for 55 weeks that formed macroscopic spleen colonies in syngeneic or allogeneic C57B10.Br/J mice; and L-cell or WEHI-3 cell conditioned medium-dependent granulocyte-macrophage colony forming unit culture (GM-CFUc); and morphologically normal granulocytes for over 245 weeks. Colony stimulating factor (CSF)-independent colony forming progenitor cells were first detectably produced in vitro at 75 weeks, and when subcultured generated karyotypically distinct permanent factor-independent tumorigenic cell lines. Nonadherent cells removed from long-term marrow cultures at 19 but not at 77 weeks reconstituted donor origin hematopoiesis in C57B10.Br/J mice as measured by B-cell lineage surface immunoglobin allotype. Nonadherent cells removed at 77 weeks produced lethal splenomegaly and marrow infiltration with culture origin cells in C57B10.Br/J mice. Despite generation of clonal malignant cell lines, L-cell DSF (CSF-1, M-CSF) responsive GM-CFUc that were simultaneously produced over 4 years in the same long-term marrow cultures, grew to 7 day colonies in semisolid medium and terminally differentiated. Thus, adherent stromal cells in Friend virus-infected long-term bone marrow cultures simultaneously support CSF-responsive and malignant CSF-independent hematopoietic progenitor cells.


Asunto(s)
Células de la Médula Ósea , Transformación Celular Viral , Granulocitos/citología , Hematopoyesis , Células Madre Hematopoyéticas/citología , Macrófagos/citología , Animales , Diferenciación Celular , Células Cultivadas , Factores Estimulantes de Colonias/farmacología , Virus de la Leucemia Murina de Friend , Alotipos de Inmunoglobulinas/análisis , Ratones , Receptores de Antígenos de Linfocitos B/análisis , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA