Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
In Vivo ; 31(2): 159-168, 2017.
Article in English | MEDLINE | ID: mdl-28358695

ABSTRACT

BACKGROUND/AIM: Total-body irradiation and/or administration of chemotherapy drugs in bone marrow transplantation induce cytokines that can suppress engraftment. Fanconi Anemia (FA) patients have a hyperactive responsiveness to the inhibitory cytokine, transforming growth factor-beta (TGF-ß). Small molecule radiation mitigator drugs, JP4-039 and MMS350, were evaluated for suppression of irradiation or drug-induced TGF-ß. MATERIALS AND METHODS: In vivo induction of TGF-ß by total-body ionizing irradiation (TBI), L-phenylalanine mustard (L-PAM), busulfan or fludarabine, was quantified. In parallel, mitigator drug amelioration of TGF-ß induction in FA D2-/- (FANCD2-/-) mouse bone marrow, was studied in vitro. Tissue culture medium, cell lysates, and mouse plasma were analyzed for TGF-ß levels. RESULTS: Induction of TGF-ß levels in FANCD2-/- and FANCD2+/+ mice and in mouse bone marrow were modulated by both JP4-039 and MMS350. CONCLUSION: Bone marrow transplantation in FA recipients may benefit from administration of small molecule agents that suppress TGF-ß induction.


Subject(s)
Bone Marrow/drug effects , Ethers, Cyclic/pharmacology , Fanconi Anemia/drug therapy , Fanconi Anemia/radiotherapy , Nitrogen Oxides/pharmacology , Sulfoxides/pharmacology , Transforming Growth Factor beta/metabolism , Animals , Blotting, Western , Bone Marrow/metabolism , Busulfan/pharmacology , Cell Line , Cells, Cultured , Drug Therapy/methods , Fanconi Anemia/metabolism , Melphalan/pharmacology , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Myeloablative Agonists/pharmacology , Radiation-Protective Agents/pharmacology , Tissue Culture Techniques , Transforming Growth Factor beta/blood , Transforming Growth Factor beta/genetics , Vidarabine/analogs & derivatives , Vidarabine/pharmacology , Whole-Body Irradiation/methods
2.
In Vivo ; 28(5): 699-708, 2014.
Article in English | MEDLINE | ID: mdl-25189880

ABSTRACT

AIM: To determine if the small-molecule radioprotector GS-nitroxide, JP4-039, improved hematopoiesis in long-term bone marrow cultures (LTBMCs), explanted marrow from in vivo drug-treated C57BL/6NTac mice was maintained in JP4-039 for 25 weeks. Hematopoietic cell production and radiobiology of derived stromal cell lines was measured. MATERIALS AND METHODS: Groups of LTBMCs were established from mouse groups. Stromal cell lines were established from the adherent layer of JP4-039-treated and untreated control groups. RESULTS: LTBMCs maintained in JP4-039 exhibited increased production of total non-adherent and 7-day and 14-day hematopoietic colony-forming cells. Stromal cell lines derived from JP4-039-treated cultures were radioresistant in vitro, demonstrated a distinct squamous/epithelial morphology and overexpressed Nrf2, Ctgf, Lox, Tlr1, collagen 1a, Brd3, and Brd4. CONCLUSION: Chronic treatment of bone marrow cultures and derived stromal cell lines with JP4-039 was non-toxic, and conferred resistance to oxidative stress.


Subject(s)
Hematopoiesis/drug effects , Hematopoiesis/radiation effects , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/radiation effects , Nitrogen Oxides/pharmacology , Radiation Tolerance/drug effects , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Bone Marrow Cells/radiation effects , Cell Line , Cells, Cultured , Colony-Forming Units Assay , Gene Expression Profiling , Hematopoiesis/genetics , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/radiation effects , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Radiation Tolerance/genetics , Transcription, Genetic
3.
In Vivo ; 28(4): 441-8, 2014.
Article in English | MEDLINE | ID: mdl-24982208

ABSTRACT

AIM: We measured long-term hematopoiesis in continuous bone marrow cultures derived from Toll-like receptor-4 (Tlr4(-/-))(C57BL/6J) mice. MATERIALS AND METHODS: We measured hematopoiesis in vitro over 27 weeks in long-term bone marrow cultures from Tlr4(-/-) and control mice, and irradiation-induced pulmonary fibrosis in mice irradiated to 20 Gy to the thorax. RESULTS: There was a significant increase in the duration of hematopoiesis in long-term bone marrow cultures from Tlr4(-/-) mice in production of total non-adherent cells and day 7 and day 14 multi-lineage colony-forming cells. The histology of bone marrow hematopoietic and stromal cell lines was indistinguishable between different mouse strains. There was no detectable late irradiation pulmonary fibrosis in Tlr4(-/-) mice. CONCLUSION: Homozygous deletion of both alleles of Tlr4, encoding for an inflammatory mediator receptor, improves the duration of hematopoiesis in vitro and reduces irradiation-induced lung fibrosis.


Subject(s)
Gene Deletion , Hematopoiesis/genetics , Hematopoiesis/radiation effects , Pulmonary Fibrosis/etiology , Radiation , Toll-Like Receptor 4/genetics , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Colony-Forming Units Assay , Disease Models, Animal , Homozygote , Mice , Mice, Knockout , Pulmonary Fibrosis/genetics
4.
In Vivo ; 28(4): 449-56, 2014.
Article in English | MEDLINE | ID: mdl-24982209

ABSTRACT

AIM: We investigated whether homologous recombinant deletion of the endothelial cell-specific protein Von Willebrand factor (vWF) affected hematopoiesis in long-term bone marrow cultures, and irradiation induction of pulmonary fibrosis/organizing alveolitis. MATERIALS AND METHODS: We established long-term bone marrow cultures from vWF(-/-) (C57BL/6 background) and vWF(+/+) littermate mice. Non-adherent cells removed weekly were tested for formation of multi-lineage hematopoietic stem cells forming colonies at 7 and 14 days in secondary semi-solid medium cultures. Irradiation fibrosis in the lungs of 20-Gy thoracic irradiated mice was quantitated and scored. RESULTS: Hematopoiesis was increased over 20 weeks in vWF(-/-) compared to vWF(+/+) cultures in production of non-adherent cells, and cells forming colonies at 7 or 14 days in secondary semi-solid medium culture. The irradiated lungs showed no increased fibrosis. CONCLUSION: Absence of vWF increases hematopoiesis in long-term bone marrow cultures and has a protective effect in irradiated lungs.


Subject(s)
Gene Deletion , Hematopoiesis/genetics , Hematopoiesis/radiation effects , Pulmonary Fibrosis/etiology , Radiation , von Willebrand Factor/genetics , Animals , Bone Marrow Cells/metabolism , Colony-Forming Units Assay , Disease Models, Animal , Homozygote , Mice, Knockout , Pulmonary Fibrosis/genetics
5.
In Vivo ; 28(4): 457-65, 2014.
Article in English | MEDLINE | ID: mdl-24982210

ABSTRACT

The ionizing irradiation mitigator MMS350 prolongs survival of mice treated with total-body irradiation and prevents radiation-induced pulmonary fibrosis when added to drinking water at day 100 after thoracic irradiation. The effects of MMS350 on hematopoiesis in long-term bone marrow culture and on the radiobiology of derived bone marrow stromal cell lines were tested. Long-term bone marrow cultures were established from C57BL/6NTac mice and maintained in a high-humidity incubator, with 7% CO2 and the addition of 100 µM MMS350 at the weekly media change. Over 10 weeks in culture, MMS350 had no significant effect on maintenance of hematopoietic stem cell production, or on nonadherent cells or colony-forming units of hematopoietic progenitor cells. Stromal cell lines derived from non MMS350-treated long-term cultures or control stromal cells treated with MMS350 were radioresistant in the clonogenic survival curve assay. MMS350 is a non-toxic, highly water-soluble radiation mitigator that exhibits radioprotective effects on bone marrow stromal cells.


Subject(s)
Bone Marrow Cells/drug effects , Bone Marrow Cells/radiation effects , Ethers, Cyclic/pharmacology , Hematopoiesis/drug effects , Radiation Tolerance/drug effects , Radiation-Protective Agents/pharmacology , Sulfoxides/pharmacology , Animals , Cell Line , Cell Survival , Colony-Forming Units Assay , Gene Expression Profiling , Hematopoiesis/genetics , Hematopoiesis/radiation effects , Mice , Radiation Tolerance/genetics , Radiation, Ionizing , Stromal Cells , Transcription, Genetic
6.
Radiat Res ; 181(1): 76-89, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24397476

ABSTRACT

FancD2 plays a central role in the human Fanconi anemia DNA damage response (DDR) pathway. Fancd2(-/-) mice exhibit many features of human Fanconi anemia including cellular DNA repair defects. Whether the DNA repair defect in Fancd2(-/-) mice results in radiologic changes in all cell lineages is unknown. We measured stress of hematopoiesis in long-term marrow cultures and radiosensitivity in clonogenic survival curves, as well as comet tail intensity, total antioxidant stores and radiation-induced gene expression in hematopoietic progenitor compared to bone marrow stromal cell lines. We further evaluated radioprotection by a mitochondrial-targeted antioxidant GS-nitroxide, JP4-039. Hematopoiesis longevity in Fancd2(-/-) mouse long-term marrow cultures was diminished and bone marrow stromal cell lines were radiosensitive compared to Fancd2(+/+) stromal cells (Fancd2(-/-) D0 = 1.4 ± 0.1 Gy, ñ = 5.0 ± 0.6 vs. Fancd2(+/+) D0 = 1.6 ± 0.1 Gy, ñ = 6.7 ± 1.6), P = 0.0124 for D0 and P = 0.0023 for ñ, respectively). In contrast, Fancd2(-/-) IL-3-dependent hematopoietic progenitor cells were radioresistant (D0 = 1.71 ± 0.04 Gy and ñ = 5.07 ± 0.52) compared to Fancd2(+/+) (D0 = 1.39 ± 0.09 Gy and ñ = 2.31 ± 0.85, P = 0.001 for D0). CFU-GM from freshly explanted Fancd2(-/-) marrow was also radioresistant. Consistent with radiosensitivity, irradiated Fancd2(-/-) stromal cells had higher DNA damage by comet tail intensity assay compared to Fancd2(+/+) cells (P < 0.0001), slower DNA damage recovery, lower baseline total antioxidant capacity, enhanced radiation-induced depletion of antioxidants, and increased CDKN1A-p21 gene transcripts and protein. Consistent with radioresistance, Fancd2(-/-) IL-3-dependent hematopoietic cells had higher baseline and post irradiation total antioxidant capacity. While, there was no detectable alteration of radiation-induced cell cycle arrest with Fancd2(-/-) stromal cells, hematopoietic progenitor cells showed reduced G2/M cell cycle arrest. The absence of the mouse Fancd2 gene product confers radiosensitivity to bone marrow stromal but not hematopoietic progenitor cells.


Subject(s)
Fanconi Anemia Complementation Group D2 Protein/deficiency , Fanconi Anemia/pathology , Hematopoietic Stem Cells/pathology , Hematopoietic Stem Cells/radiation effects , Mesenchymal Stem Cells/pathology , Mesenchymal Stem Cells/radiation effects , Animals , Antioxidants/metabolism , Cell Cycle/drug effects , Cell Cycle/radiation effects , Cell Line , Fanconi Anemia/metabolism , Free Radical Scavengers/pharmacology , Granulocyte-Macrophage Progenitor Cells/metabolism , Hematopoiesis/drug effects , Hematopoiesis/radiation effects , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Humans , Interleukin-3/pharmacology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/radiation effects , Nitrogen Oxides/pharmacology , Oxidative Stress/drug effects , Oxidative Stress/radiation effects , Radiation Tolerance , Reactive Oxygen Species/metabolism , Time Factors
7.
Radiat Res ; 180(5): 474-90, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24125487

ABSTRACT

A water-soluble ionizing radiation mitigator would have considerable advantages for the management of acute and chronic effects of ionizing radiation. We report that a novel oxetanyl sulfoxide (MMS350) is effective both as a protector and a mitigator of clonal mouse bone marrow stromal cell lines in vitro, and is an effective in vivo mitigator when administered 24 h after 9.5 Gy (LD100/30) total-body irradiation of C57BL/6NHsd mice, significantly improving survival (P = 0.0097). Furthermore, MMS350 (400 µM) added weekly to drinking water after 20 Gy thoracic irradiation significantly decreased: expression of pulmonary inflammatory and profibrotic gene transcripts and proteins; migration into the lungs of bone marrow origin luciferase+/GFP+ (luc+/GFP+) fibroblast progenitors (in both luc+ marrow chimeric and luc+ stromal cell line injected mouse models) and decreased radiation-induced pulmonary fibrosis (P < 0.0001). This nontoxic and orally administered small molecule may be an effective therapeutic in clinical radiotherapy and as a counter measure against the acute and chronic effects of ionizing radiation.


Subject(s)
Ethers, Cyclic/pharmacology , Lung/drug effects , Radiation Pneumonitis/drug therapy , Radiation-Protective Agents/administration & dosage , Safrole/analogs & derivatives , Sulfoxides/pharmacology , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/radiation effects , Cell Line , Humans , Lung/radiation effects , Mice , Radiation Pneumonitis/pathology , Radiation, Ionizing , Safrole/administration & dosage , Water/chemistry , Whole-Body Irradiation
8.
In Vivo ; 27(5): 571-82, 2013.
Article in English | MEDLINE | ID: mdl-23988890

ABSTRACT

AIM: We determined whether bone marrow from Nrf2(-/-) compared with Nrf2(+/+) mice differed in response to the oxidative stress of continuous marrow culture, and in radiosensitivity of derived stromal and interleukin-3 (IL-3)-dependent hematopoietic progenitor cells. MATERIALS AND METHODS: Hematopoiesis longevity in Nrf2(-/-) was compared with Nrf2(+/+) mice in long-term bone marrow cultures. Clonogenic irradiation survival curves were performed on derived cell lines. Total antioxidant capacity at baseline in nonirradiated cells and at 24 hours after 5 Gy and 10 Gy irradiation was quantitated using an antioxidant reductive capacity assay. RESULTS: Long-term cultures of bone marrow from Nrf2(-/-) compared to Nrf2(+/+) mice demonstrated equivalent longevity of production of total cells and hematopoietic progenitor cells forming multi-lineage hematopoietic colonies over 26 weeks in culture. Both bone marrow stromal cell lines and Il-3-dependent hematopoietic progenitor cell lines derived from Nrf2(-/-) mouse marrow cultures were radioresistant compared to Nrf2(+/+)-derived cell lines. Both DNA repair assay and total antioxidant capacity assay showed no defect in Nrf2(-/-) compared to Nrf2(+/+) stromal cells and IL-3-dependent cells. CONCLUSION: The absence of a functional Nrf2 gene product does not alter cellular interactions in continuous marrow culture, nor response to dsDNA damage repair and antioxidant response. However, lack of the Nrf2 gene does confer radioresistance on marrow stromal and hematopoietic cells.


Subject(s)
Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/radiation effects , Homozygote , NF-E2-Related Factor 2/deficiency , Radiation Tolerance/genetics , Stromal Cells/metabolism , Stromal Cells/radiation effects , Animals , Bone Marrow Cells/metabolism , Bone Marrow Cells/radiation effects , Cells, Cultured , Colony-Forming Units Assay , Hematopoiesis/genetics , Hematopoiesis/radiation effects , Mice , Mice, Knockout , NF-E2-Related Factor 2/genetics
9.
In Vivo ; 27(4): 419-30, 2013.
Article in English | MEDLINE | ID: mdl-23812211

ABSTRACT

AIM: We determined whether absence of caspase-1 altered the stress response of hematopoietic and bone marrow stromal cells in vitro. MATERIALS AND METHODS: Long-term bone marrow cultures from caspase-1 -/- and control caspase-1 +/+ mice were established and the derived bone marrow stromal and interleukin-3 (Il-3)-dependent hematopoietic progenitor cell lines were evaluated for radiosensitivity. RESULTS: Long-term bone marrow cultures from caspase-1 -/- mice generated hematopoietic cells for over 30 weeks in vitro, significantly longer than controls did (p=0.0018). Bone marrow stromal (mesenchymal stem cell) and Il-3-dependent hematopoietic progenitor cell lines from caspase-1-/- marrow cultures compared to caspase-1 +/+ were radioresistant (p=0.0486 and p=0.0235 respectively). Total-body irradiated caspase-1 -/- mice were not significantly radioresistant compared to controls (p=0.6542). CONCLUSION: Caspase-1 deletion increases hematopoiesis and radioresistance of bone marrow cells in vitro.


Subject(s)
Caspase 1/genetics , Hematopoiesis/genetics , Hematopoiesis/radiation effects , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/radiation effects , Radiation Tolerance/genetics , Stromal Cells/metabolism , Stromal Cells/radiation effects , Animals , Antioxidants/pharmacology , Bone Marrow Cells/metabolism , Bone Marrow Cells/radiation effects , Cell Culture Techniques , Cell Survival/drug effects , Cell Survival/genetics , Cell Survival/radiation effects , DNA Repair , Genotyping Techniques , Hematopoiesis/drug effects , Hematopoietic Stem Cells/drug effects , Homozygote , Mice , Mice, Knockout , Stromal Cells/drug effects , Survival Analysis , Time Factors
10.
Exp Hematol ; 40(6): 499-509, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22326715

ABSTRACT

The senescence accelerated-prone mouse variant 6 (SAMP6) shows normal growth followed by rapid aging, development of osteopenia, and shortened lifespan, compared with control R1 mice. Because oxidative stress is a fundamental mechanism of tissue aging, we tested whether cellular parameters that are associated with oxidative stress are impaired with marrow from SAMP6 mice. We compared in vitro hematopoiesis, irradiation sensitivity, proliferative potential, and osteoblastogenesis with marrow cells from SAMP6 and R1 mice. Marrow cells from SAMP6 mice showed shortened in vitro hematopoiesis; their stromal cells showed greater radiation sensitivity and decreased proliferation. Consistent with those properties, there was constitutive upregulation of transforming growth factor-ß(1), an inhibitor of hematopoiesis, and of cell cycle inhibitory genes, p16(INK4A) and p19(ARF). Paradoxically, there was constitutive expression of osteoblast genes in stromal cells from SAMP6 mice, but in vitro matrix mineralization was impaired. These studies and data included in other reports indicate that impaired proliferation of osteoblast progenitors in SAMP6 marrow may be a major factor contributing to accelerated loss of bone mass. In sum, marrow from SAMP6 mice had diminished capacity for long-term hematopoiesis, increased radiosensitivity, and reduced proliferative capacity.


Subject(s)
Bone Marrow Cells/pathology , Hematopoiesis , Osteoblasts/cytology , Radiation Tolerance , Animals , Cells, Cultured , In Vitro Techniques , Mice , Oxidative Stress , Reverse Transcriptase Polymerase Chain Reaction
11.
In Vivo ; 26(1): 9-18, 2012.
Article in English | MEDLINE | ID: mdl-22210711

ABSTRACT

AIM: The effect of lung irradiation on reduction of lung stem cells and repopulation with bone marrow-derived cells was measured. MATERIALS AND METHODS: Expression of green fluorescent protein positive cells (GFP(+)) in the lungs of thoracic irradiated FVB/NHsd mice (Harlan Sprague Dawley, Indianapolis, IN, USA) was determined. This was compared to the repopulation of bone marrow-derived cells found in the lungs from naphthalene treated male FVB/NHsd mice and gangciclovir (GCV) treated FeVBN GFP(+) male marrow chimeric HSV-TK-CCSP. The level of mRNA for lung stem cell markers clara cell (CCSP), epithelium 1 (FOXJ1) and surfactant protein C (SP-C), and sorted single cells positive for marrow origin epithelial cells (GFP(+)CD45(-)) was measured. RESULTS: The expression of pulmonary stem cells as determined by PCR was reduced most by GCV, then naphthalene, and least by thoracic irradiation. Irradiation, like GCV, reduced mRNA expression of CCSP, CYP2F2, and FOXJ1, while naphthalene reduced that of CCSP and CYP2F2. Ultrastructural analysis showed GFP(+) pulmonary cells of bone marrow origin, with the highest frequency being found in GCV-treated groups. CONCLUSION: Bone marrow progenitor cells may not participate in the repopulation of the lung following irradiation.


Subject(s)
Bone Marrow Cells/metabolism , Bone Marrow Transplantation/methods , Epithelial Cells/metabolism , Lung/metabolism , Animals , Antiviral Agents/pharmacology , Bone Marrow Cells/ultrastructure , Cytochrome P-450 Enzyme System/genetics , Epithelial Cells/ultrastructure , Female , Forkhead Transcription Factors/genetics , Ganciclovir/pharmacology , Gene Expression/drug effects , Gene Expression/radiation effects , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Lung/drug effects , Lung/radiation effects , Male , Mice , Mice, Transgenic , Microscopy, Confocal , Microscopy, Electron, Transmission , Naphthalenes/pharmacology , Pulmonary Surfactant-Associated Protein C/genetics , Radiation Injuries, Experimental/genetics , Radiation Injuries, Experimental/metabolism , Radiation Injuries, Experimental/pathology , Reverse Transcriptase Polymerase Chain Reaction , Uteroglobin/genetics
12.
Radiat Res ; 174(3): 297-312, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20726721

ABSTRACT

The effect of deletion of the nitric oxide synthase 1 gene (NOS1(-/-)) on radiosensitivity was determined. In vitro, long-term cultures of bone marrow stromal cells derived from NOS1(-/-) were more radioresistant than cells from C57BL/6NHsd (wild-type), NOS2(-/-) or NOS3(-/-) mice. Mice from each strain received 20 Gy thoracic irradiation or 9.5 Gy total-body irradiation (TBI), and NOS1(-/-) mice were more sensitive to both. To determine the etiology of radiosensitivity, studies of histopathology, lower esophageal contractility, gastrointestinal transit, blood counts, electrolytes and inflammatory markers were performed; no significant differences between irradiated NOS1(-/-) and control mice were found. Video camera surveillance revealed the cause of death in NOS1(-/-) mice to be grand mal seizures; control mice died with fatigue and listlessness associated with low blood counts after TBI. NOS1(-/-) mice were not sensitive to brain-only irradiation. MnSOD-PL therapy delivered to the esophagus of wild-type and NOS1(-/-) mice resulted in equivalent biochemical levels in both; however, in NOS1(-/-) mice, MnSOD-PL significantly increased survival after both thoracic and total-body irradiation. The mechanism of radiosensitivity of NOS1(-/-) mice and its reversal by MnSOD-PL may be related to the developmental esophageal enteric neuronal innervation abnormalities described in these mice.


Subject(s)
Esophagus/enzymology , Liposomes , Nitric Oxide Synthase Type I/metabolism , Plasmids , Superoxide Dismutase/genetics , Animals , Bone Marrow Cells/radiation effects , Electric Stimulation , Gastrointestinal Transit , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase Type I/genetics , Radiography, Thoracic , Reverse Transcriptase Polymerase Chain Reaction
13.
In Vivo ; 23(5): 669-77, 2009.
Article in English | MEDLINE | ID: mdl-19779099

ABSTRACT

Fluorescent yellow direct repeat (FYDR) mice carry a transgenic reporter for homologous recombination (HR) and have been used to reveal an age-dependent increase in HR in the pancreas. An established in vitro model system for accelerated aging of the marrow is the mouse long-term bone marrow culture (LTBMC) system. To determine whether the FYDR system, in which an HR event can lead to a fluorescent cell, can be used to study the effects of aging in LTBMCs, clonally expanded hematopoietic and marrow stromal cells in FYDR, positive control FYDR-Recombined (FYDR-Rec), and negative control wild-type C57BL/6NHsd (WT) LTBMCs were analysed. All groups of cultures demonstrated equivalent parameters of continuous hematopoiesis including generation of multilineage colony forming CFU-GM progenitor cells for over 22 weeks and age associated senescence of hematopoiesis. Results indicate that low expression of the FYDR transgene in bone marrow cells in vivo and in vitro prevents the use of the FYDR mice to study rare combination events in bone marrow. Using an alternative approach for detecting HR, namely the sister chromatid exchange (SCE) assay, a statistically significant increase in the number of SCEs per chromosome was observed in adherent cells subcultured from 20-week-compared to 4-week-old LTBMCs. These data suggest that adherent marrow stromal cells from LTBMCs become increasingly susceptible to HR events during aging.


Subject(s)
Bone Marrow Cells/cytology , Cellular Senescence/genetics , Hematopoiesis/genetics , Recombination, Genetic , Animals , Bone Marrow Cells/physiology , Cells, Cultured , Clone Cells , Female , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Mice , Sister Chromatid Exchange , Stromal Cells/cytology , Stromal Cells/physiology
14.
Radiat Res ; 168(5): 552-9, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17973549

ABSTRACT

Ethyl pyruvate (EP), a simple aliphatic ester of pyruvic acid, has been shown to improve survival and ameliorate organ damage in animal models of sepsis, ischemia/reperfusion injury and hemorrhagic shock. Incubating IL3-dependent mouse hematopoietic progenitor cell 32Dcl3 cells before or after irradiation with 10 mM EP increased resistance to radiation as assessed by clonogenic radiation survival curves, decreased release of mitochondrial cytochrome C into the cytoplasm, and decreased apoptosis. EP inhibited radiation-induced caspase 3 activation and poly(ADP-ribose) polymerase (PARP) cleavage in 32Dcl3 cells in a concentration-dependent fashion. EP was given i.p. to C57BL/6NHsd mice irradiated with 9.75 Gy total-body irradiation (TBI). This treatment significantly improved survival. The survival benefit was apparent irrespective of whether treatment with EP was started 1 h before TBI and continued for 5 consecutive days after TBI or the compound was injected only 1 h before or only for 5 days after TBI. In all of the in vitro and in vivo experiments, ethyl lactate, an inactive analogue of EP, had no detectable radioprotective or mitigating effects. EP may be an effective radioprotector and mitigator of the hematopoietic syndrome induced by TBI.


Subject(s)
Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/radiation effects , Pyruvates/administration & dosage , Radiation-Protective Agents/administration & dosage , Survival Rate , Whole-Body Irradiation , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Cell Line , Cell Survival/drug effects , Cell Survival/radiation effects , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , Female , Hematopoietic Stem Cells/cytology , Mice , Mice, Inbred C57BL , Survival Analysis
15.
Exp Hematol ; 35(1): 137-45, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17198882

ABSTRACT

OBJECTIVE: Neuronal nitric oxide synthase (NOS1, mitochondrial NOS, neuronal NOS) homozygous deletion recombinant negative mice demonstrate ionizing irradiation resistance in vivo, attributable to the decrease in mitochondrial-localized production of peroxynitrite, a potent lipid toxic free radical species resulting from the combination of nitric oxide and superoxide. The present studies were designed to determine whether reduced mitochondrial generation of toxic radical oxygen species in NOS1-/- mice also increased the longevity of hematopoiesis in continuous bone marrow cultures and conferred radioresistance to cells in vitro. MATERIALS AND METHODS: Long-term bone marrow cultures (LTBMCs) were established from NOS1-/- and NOS1+/+ littermate mice. Radiation resistance of hematopoietic and marrow stromal cells was measured. Cell cycle analysis and measurement of glutathione and glutathione peroxidase were carried out on irradiated clonal bone marrow stromal cell lines. RESULTS: A significant increase in longevity of hematopoiesis was detected in NOS1-/- mouse LTBMCs for over 64 weeks in culture compared to 20 weeks for NOS1+/+ mouse LTBMCs (p < 0.001). Permanent bone marrow stromal cell lines derived from NOS1-/- mouse LTBMCs demonstrated increased radioresistance in vitro reflected by an increased shoulder on the survival curve with n = 32.15 +/- 1.21 compared to NOS1+/+ cells n = 10.47 +/- 3.2 (p = 0.0026), interleukin-3-dependent NOS1-/- hematopoietic progenitor cell lines also demonstrated decreased apoptosis after 10 Gy irradiation. Both pre- and postirradiation stabilization of the cellular antioxidant pool was detected in NOS1-/- cells. NOS1-/- cells showed a prolonged G1 cell cycle arrest after 10 Gy. CONCLUSIONS: Prolonged hematopoiesis in LTBMCs correlates with intrinsic radioresistance of hematopoietic and marrow stromal cells from NOS1-/- mice. The data confirm the importance to hematopoiesis of mitochondrial localized nitric oxide in both radioresistance and longevity of hematopoiesis in continuous bone marrow cultures.


Subject(s)
Bone Marrow Cells/cytology , Hematopoiesis , Hematopoietic Stem Cells/radiation effects , Nitric Oxide Synthase Type I/genetics , Stromal Cells/radiation effects , Animals , Bone Marrow Cells/enzymology , Cell Culture Techniques , Cells, Cultured , Homozygote , Mice , Mice, Mutant Strains , Mitochondria/metabolism , Mutation , Nitric Oxide/metabolism , Nitric Oxide/physiology , Radiation Effects , Time Factors
16.
Cancer Res ; 66(1): 343-51, 2006 Jan 01.
Article in English | MEDLINE | ID: mdl-16397248

ABSTRACT

Cell cycle inhibitors are important regulators in normal tissue regeneration and disruption of the regulators are involved in cancer development. Our recent study showed that the absence of the CDK inhibitor p18(INK4C) (p18) enhances self-renewal of normal hematopoietic stem cell (HSC) in vivo, whereas previous studies by others showed an increased incidence of leukemogenesis in older p18-null mice. Here, we have examined potential leukemogenesis during experimentally induced regeneration of HSC in the absence of p18 in order to gauge the relation between these two processes. Reconstituted mice with p18-deficient HSCs under the condition of repetitive proliferative stress (serial transplantation) were followed for >3 years. T cell leukemia from the p18-/- origin was recapitulated 24 months after secondary transplantation. However, no myeloid leukemia was found in the recipients. The T cell leukemia-initiating cells (mainly in a CD3(lo) cell subset) did not share the same immunophenotype with normal HSCs and, in fact, the function of HSCs was significantly compromised with decreased abundance in the leukemic mice. Furthermore, we found that the p15 or p16 gene promoters were frequently methylated in the leukemic cells but not in HSCs. Our present study argues against the possibility of overgrowth of p18-null HSCs leading to a leukemic phenotype. The data also support the notion that p18 has an independent role in T cell maintenance such that CD3+ CD8+ cells, unlike HSCs, are more accessible to leukemogenic transformation after the loss of p18.


Subject(s)
Cell Transformation, Neoplastic/pathology , Cyclin-Dependent Kinase Inhibitor p18/deficiency , Hematopoietic Stem Cells/pathology , Leukemia, T-Cell/pathology , Animals , Cell Cycle/physiology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cyclin-Dependent Kinase Inhibitor p18/genetics , Hematopoietic Stem Cells/metabolism , Immunophenotyping , Leukemia, T-Cell/genetics , Leukemia, T-Cell/metabolism , Mice , Mice, Inbred C57BL , T-Lymphocytes/metabolism , T-Lymphocytes/pathology
17.
In Vivo ; 19(6): 965-74, 2005.
Article in English | MEDLINE | ID: mdl-16277008

ABSTRACT

BACKGROUND: Intraesophageal manganese superoxide dismutase plasmid liposome (MnSOD-PL) gene therapy protects against irradiation damage. MATERIALS AND METHODS: To determine whether esophageal side population (SP) stem cells were protected, epitope-tagged (hemagglutinin) (HA) MnSOD-PL was administered to C57BL/6J mice 24 hours prior to 30 Gy esophageal irradiation. SP cells were isolated, and apoptosis and multi-lineage vimentin/endothelin/F4/80 (macrophage) colonies in vitro were quantitated. RESULTS: The number and percent of SP cells, apoptotic cells, or numbers of multi-lineage vimentin/endothelin/F4/80-positive in vitro colonies isolated from non-irradiated HA-MnSOD-PL-treated or 30 Gy-irradiated esophagus did not differ between groups. Irradiation in vitro significantly increased apoptosis in explanted non-SP cells from control (p = 0.021) compared to MnSOD-PL-treated mice. Irradiation-induced cell division was significantly increased in SP cells from control-irradiated mice (p = 0.001), but not MnSOD-PL-treated mice. Irradiation-induced apoptosis detected in vivo at 5 days was decreased by MnSOD-PL. CONCLUSION: MnSOD-PL gene therapy protects esophageal SP cells from irradiation in vitro and in vivo.


Subject(s)
Esophagus/radiation effects , Genetic Therapy , Radiation Injuries, Experimental/pathology , Radiation Protection , Stem Cells/radiation effects , Superoxide Dismutase/administration & dosage , Animals , Annexin A5/metabolism , Antigens, Differentiation/metabolism , Apoptosis/radiation effects , Biomarkers/metabolism , Cell Culture Techniques , Cell Division/radiation effects , Cell Lineage , Cells, Cultured , Endothelins/metabolism , Esophagus/enzymology , Esophagus/pathology , Fluorescein-5-isothiocyanate , Fluorescent Dyes , Liposomes , Male , Mice , Mice, Inbred C57BL , Plasmids , Proliferating Cell Nuclear Antigen/metabolism , Radiation Injuries, Experimental/prevention & control , Radiation, Ionizing , Stem Cells/cytology , Superoxide Dismutase/genetics , Vimentin/metabolism
18.
Exp Hematol ; 33(3): 353-62, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15730859

ABSTRACT

OBJECTIVE: To determine the role of Smad3 in modulating hematopoiesis, continuous bone marrow cultures were established from Smad-/- mice, and the longevity of hematopoiesis and extent of adipogenesis in the supportive hematopoietic microenvironment were compared to those from cultures of control, Smad3+/+ or heterozygous Smad3+/- mice. MATERIALS AND METHODS: Long-term bone marrow cultures (LTBMCs) were established from Smad3+/+, Smad3+/-, or Smad3-/- mice. On a weekly basis, the number of cobblestone islands, number of nonadherent cells, confluence of the adherent cells, or CFU-GEMM colonies was determined. Bone marrow stromal cell lines were established and cobblestone island production on these cell lines determined in the presence of nonadherent cells from week-42 Smad3-/- or week-4 C57BL/6J LTBMCs. RESULTS: Initial proliferative capacity of the LTBMCs was similar in all groups through week 20, at which time there was an increase in cobblestone islands and production of nonadherent cells and CFU-GEMM colonies in the Smad3-/- group. By week 28, only the Smad3-/- LTBMCs had significantly maintained increased production of these parameters. Maintenance of cobblestone islands indicative of the most primitive hematopoietic progenitor cells persisted past 45 weeks in Smad3-/- cultures. The Smad3-/- stromal cell line also demonstrated increased support of cobblestone island production when incubated with nonadherent cells from week-42 Smad3-/- or week-4 C57BL/6J LTBMCs. Evaluation of adipocytogenesis in stromal cells showed significantly greater accumulation of adipocytes in lines from Smad3-/- than from Smad3+/+ mice. CONCLUSIONS: These data provide evidence for a significant effect of deletion of the Smad3 signaling pathway in increased hematopoiesis in LTBMCs and support the negative regulatory influence of TGFbeta signaling on adipocytogenesis and long-term hematopoiesis in vitro.


Subject(s)
Adipocytes/physiology , Bone Marrow/physiology , DNA-Binding Proteins/metabolism , Hematopoiesis/physiology , Hematopoietic Stem Cells/physiology , Signal Transduction/physiology , Trans-Activators/metabolism , Animals , Cells, Cultured , DNA-Binding Proteins/genetics , Hematopoiesis/genetics , Male , Mice , Mice, Knockout , Signal Transduction/genetics , Smad3 Protein , Stromal Cells/physiology , Time Factors , Trans-Activators/genetics , Transforming Growth Factor beta/physiology
19.
Hum Gene Ther ; 15(4): 415-9, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15053866

ABSTRACT

Protein transduction domains (PTDs) have been used increasingly to deliver biologically active agents to a variety of cell types in vitro and in vivo. To define the most effective PTDs for transducing hematopoietic cells, we have screened a panel of PTD peptides in human CD34(+) cells for delivery of a 60-kd marker protein and assessed its impact on phenotypic maintenence in vitro. Compared to the HIV-TAT peptide, most peptide complexes displayed high efficiency in transducing the CD34(+) cells, except for those based on shorter peptides (4R, 4K, and 5RQ). In particular, the arginine homopolymers including 8R, 10R, and 12R, were internalized by the cells to a greater extent than the other PTDs. Transduction was significantly potentiated by preincubation of cells with dextran sulfate. Importantly, colony forming ability and CD34(+) CD38(-) primitive phenotype were not significantly altered in the presence of these peptides during a short-term liquid culture. Together, these data suggest the potential usefulness of arginine homopolymers in hematopoietic stem and progenitor cell manipulations.


Subject(s)
Antigens, CD34/metabolism , Dextran Sulfate/pharmacology , Peptide Fragments/metabolism , Transduction, Genetic , Colony-Forming Units Assay , Fetal Blood , Humans , Peptide Fragments/chemistry , Protein Structure, Tertiary , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...