Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
J Pain Symptom Manage ; 61(2): 384-394, 2021 02.
Article in English | MEDLINE | ID: mdl-32822755

ABSTRACT

CONTEXT: Virtual reality (VR) has emerged as a novel form of nonpharmacological analgesia therapy. We wished to review the use of VR to treat pain and anxiety in cancer-related medical procedures and chemotherapy. OBJECTIVES: To determine if immersive VR influences pain and/or anxiety outcomes in patients with cancer undergoing medical interventions. To discuss critical limitations in the current evidence base and provide suggestions for future areas of research. METHODS: A systematic review was performed on Ovid MEDLINE, PubMed, and Google Scholar from 1999 to December 2019. The following search terms were run in each of the databases: Virtual Reality and pain or anxiety. Articles were assessed by two independent authors for inclusion. RESULTS: From 999 retrieved citations, nine studies met inclusion criteria for review. Methodological limitations and small sample sizes preclude strong guidance for clinical applications. Although studies demonstrated a trend toward improvement in pain and anxiety, only two studies reached statistical significance. CONCLUSION: There is inconclusive evidence on the significance of immersive VR in reducing pain (five studies) or anxiety (six studies) for patients with cancer undergoing medical interventions or receiving chemotherapy. Further research on the effect of immersive VR as a tool for medical procedures and/or patients with cancer undergoing treatment is required.


Subject(s)
Neoplasms , Virtual Reality Exposure Therapy , Virtual Reality , Anxiety/etiology , Anxiety/therapy , Humans , Neoplasms/complications , Neoplasms/therapy , Pain/etiology
3.
Blood ; 123(16): 2518-29, 2014 Apr 17.
Article in English | MEDLINE | ID: mdl-24591202

ABSTRACT

FMS-like tyrosine kinase 3 (FLT3) is expressed in human hematopoietic stem and progenitor cells (HSPCs) but its role during embryogenesis is unclear. In acute myeloid leukemia (AML), internal tandem duplication (ITD) of FLT3 at the juxtamembrane (JMD) and tyrosine kinase (TKD) domains (FLT3-ITD(+)) occurs in 30% of patients and is associated with inferior clinical prognosis. TKD mutations (FLT3-TKD(+)) occur in 5% of cases. We made use of zebrafish to examine the role of flt3 in developmental hematopoiesis and model human FLT3-ITD(+) and FLT3-TKD(+) AML. Zebrafish flt3 JMD and TKD were remarkably similar to their mammalian orthologs. Morpholino knockdown significantly reduced the expression of l-plastin (pan-leukocyte), csf1r, and mpeg1 (macrophage) as well as that of c-myb (definitive HSPCs), lck, and rag1 (T-lymphocyte). Expressing human FLT3-ITD in zebrafish embryos resulted in expansion and clustering of myeloid cells (pu.1(+), mpo(+), and cebpα(+)) which were ameliorated by AC220 and associated with stat5, erk1/2, and akt phosphorylation. Human FLT3-TKD (D835Y) induced significant, albeit modest, myeloid expansion resistant to AC220. This study provides novel insight into the role of flt3 during hematopoiesis and establishes a zebrafish model of FLT3-ITD(+) and FLT3-TKD(+) AML that may facilitate high-throughput screening of novel and personalized agents.


Subject(s)
Hematopoiesis/genetics , Leukemia, Myeloid, Acute/genetics , Zebrafish Proteins/physiology , Zebrafish/physiology , fms-Like Tyrosine Kinase 3/physiology , Amino Acid Sequence , Animals , Animals, Genetically Modified , Conserved Sequence , Embryo, Nonmammalian , Humans , Molecular Sequence Data , Protein Structure, Tertiary/genetics , Sequence Homology, Amino Acid , Tandem Repeat Sequences , Transcriptome , Zebrafish/embryology , Zebrafish Proteins/chemistry , fms-Like Tyrosine Kinase 3/chemistry
4.
Blood ; 123(16): 2530-9, 2014 Apr 17.
Article in English | MEDLINE | ID: mdl-24608976

ABSTRACT

Internal tandem duplication (ITD) of fms-like tyrosine kinase 3 (FLT3) in acute myeloid leukemia (AML) is associated with inferior clinical prognosis. Sorafenib is effective in clearing leukemic blasts in chemorefractory FLT3-ITD(+) AML, but leukemia progression invariably occurs. Mechanisms of drug resistance are not completely understood. We hypothesized that a gene encoding tescalcin (TESC), known to be upregulated at leukemia progression during continuous sorafenib treatment and activate an Na(+)/H(+) exchanger type-1 (NHE1), may underlie tyrosine kinase inhibitor resistance. TESC was highly expressed in FLT3-ITD(+) AML lines MOLM-13 and MV4-11, and its knockdown by small-interfering RNA lowered intracellular pH (pHi) and induced apoptosis. The results were recapitulated by treatment with an NHE1 inhibitor, 5-(N,N-hexamethylene) amiloride (HMA). Induction of sorafenib resistance in the MOLM-13 cell line (M13-RE) significantly increased its sensitivity to HMA. The later also enhanced suppression of FLT3 signaling by sorafenib in otherwise resistant cell lines. HMA treatment of MOLM-13 and MV4-11 as well as primary FLT3-ITD(+) AML cells significantly reduced leukemia initiation in anti-CD122-primed NOD/SCID mouse xenotransplantation. These observations provided novel information about the pathogenetic role of a TESC-NHE1-pHi axis in mediating sorafenib resistance in AML.


Subject(s)
Antineoplastic Agents/therapeutic use , Calcium-Binding Proteins/physiology , Cation Transport Proteins/physiology , Drug Resistance, Neoplasm/genetics , Leukemia, Myeloid, Acute/drug therapy , Niacinamide/analogs & derivatives , Phenylurea Compounds/therapeutic use , Sodium-Hydrogen Exchangers/physiology , Animals , Cells, Cultured , Humans , Hydrogen-Ion Concentration , Intracellular Space/metabolism , K562 Cells , Leukemia, Myeloid, Acute/genetics , Mice , Mice, Inbred NOD , Mice, SCID , Niacinamide/therapeutic use , Signal Transduction/genetics , Sodium-Hydrogen Exchanger 1 , Sorafenib , Tandem Repeat Sequences/genetics , fms-Like Tyrosine Kinase 3/chemistry , fms-Like Tyrosine Kinase 3/genetics
5.
Stem Cells Dev ; 23(14): 1704-16, 2014 07 15.
Article in English | MEDLINE | ID: mdl-24564569

ABSTRACT

Self-renewable human pluripotent stem cells (hPSCs) serve as a potential unlimited ex vivo source of human cardiomyocytes (CMs) for cell-based disease modeling and therapies. Although recent advances in directed differentiation protocols have enabled more efficient derivation of hPSC-derived CMs with an efficiency of ∼50%-80% CMs and a final yield of ∼1-20 CMs per starting undifferentiated hPSC, these protocols are often not readily transferrable across lines without first optimizing multiple parameters. Further, the resultant populations are undefined for chamber specificity or heterogeneous containing mixtures of atrial, ventricular (V), and pacemaker derivatives. Here we report a highly cost-effective and reproducibly efficient system for deriving hPSC-ventricular cardiomyocytes (VCMs) from all five human embryonic stem cell (HES2, H7, and H9) and human induced PSC (hiPSC) (reprogrammed from human adult peripheral blood CD34(+) cells using nonintegrating episomal vectors) lines tested. Cardiogenic embryoid bodies could be formed by the sequential addition of BMP4, Rho kinase inhibitor, activin-A, and IWR-1. Spontaneously contracting clusters appeared as early as day 8. At day 16, up to 95% of cells were cTnT(+). Of which, 93%, 94%, 100%, 92%, and 92% of cardiac derivatives from HES2, H7, H9, and two iPSC lines, respectively, were VCMs as gauged by signature ventricular action potential and ionic currents (INa(+)/ICa,L(+)/IKr(+)/IKATP(+)); Ca(2+) transients showed positive chronotropic responses to ß-adrenergic stimulation. Our simple, cost-effective protocol required the least amounts of reagents and time compared with others. While the purity and percentage of PSC-VCMs were comparable to a recently published protocol, the present yield and efficiency with a final output of up to 70 hPSC-VCMs per hPSC was up to 5-fold higher and without the need of performing line-specific optimization. These differences were discussed. The results may lead to mass production of hPSC-VCMs in bioreactors.


Subject(s)
Embryonic Stem Cells/cytology , Heart Ventricles/cytology , Induced Pluripotent Stem Cells/cytology , Myocytes, Cardiac/cytology , Bioreactors , Bone Morphogenetic Protein 4/biosynthesis , Cell Differentiation/genetics , Humans
6.
Zhonghua Zhong Liu Za Zhi ; 34(9): 652-7, 2012 Sep.
Article in Chinese | MEDLINE | ID: mdl-23159076

ABSTRACT

OBJECTIVE: To observe the effect of rosiglitazone (RGZ) and all-trans-retinoic acid (ATRA) on the growth of myeloma xenograft in nude mice and to explore the influence of RGZ and ATRA on VEGF expression and angiogenesis in the tumor. METHODS: VEGF gene expression in myeloma cell line U266 cells was analyzed by semi-quantitative RT-PCR after incubation with RGZ, ATRA, or RGZ + ATRA for 24 h. Myeloma xenograft was established by subcutaneous injection of 10(7) U266 cells in the scapula area of 4-week old nude mice. 7 days later, the nude mice were administered with RGZ, ATRA or RGZ + ATRA, respectively, by intraperitoneal injection once every day for 21 days. The control mice were given equal volume of normal saline instead of the drug. On the 21(st) day of treatment, the mice were sacrificed and the tumors were taken off, and the tumor volume and weight were measured. The tumors were examined by histopathology with HE staining, and microvessel density (MVD), CD34 and VEGF expression in the tumors were analyzed by immunohistochemical staining. RESULTS: VEGF mRNA was highly expressed in U266 cells and was decreased in a dose-dependent manner after incubation with RGZ. The VEGF mRNA level was further more decreased after RGZ + ATRA treatment. Xenografts of U266 cells were developed in all nude mice. The volume and weight of xenografts in the RGZ group were (785 ± 262) mm(3) and (1748 ± 365) mg, respectively, significantly lower than those of the control group (both P < 0.01). More significant inhibition was in the RGZ + ATRA group, (154 ± 89) mm(3) and (626 ± 102) mg, respectively, both were P < 0.05 vs. the RGZ group. RGZ inhibited the angiogenesis in U266 xenografts and immunohistochemical staining showed that the tumor MVD and VEGF expression were significantly decreased by RGZ treatment, and further more inhibited in the RGZ + ATRA group. VEGF protein was expressed in all xenografts in the nude mice. Its immunohistochemical staining intensity was 2.20 ± 0.40 in the control group, significantly higher than that of 1.48 ± 0.37 in the RGZ group (P < 0.01), and that of RGZ + ATRA group was 0.58 ± 0.26, further significantly lower than that of the RGZ group (P < 0.01). CD34 was expressed in all xenografts, most highly in the control group and lowest in the RGZ + ATRA group. The microvessel density (MVD) was highest in the control group (56.4 ± 15.2), significantly lower in the RGZ group (44.6 ± 11.2) (P < 0.05), and lowest in the RGZ + ATRA group (21.5 ± 8.6, P < 0.01). CONCLUSIONS: The growth of myeloma cells can also be inhibited by RGZ and ATRA in nude mice in vivo. In addition to differentiation and apoptosis induction, RGZ can inhibit the formation of myeloma xenograft probably also through the downregulation of VEGF expression and subsequent angiogenesis.


Subject(s)
Multiple Myeloma/pathology , Neovascularization, Pathologic , Thiazolidinediones/pharmacology , Tretinoin/pharmacology , Tumor Burden/drug effects , Animals , Antigens, CD34/metabolism , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Synergism , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Microvessels/pathology , Multiple Myeloma/metabolism , Neoplasm Transplantation , RNA, Messenger/metabolism , Rosiglitazone , Thiazolidinediones/administration & dosage , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Xenograft Model Antitumor Assays
7.
Blood ; 119(22): 5133-43, 2012 May 31.
Article in English | MEDLINE | ID: mdl-22368270

ABSTRACT

Internal tandem duplication (ITD) of the fms-related tyrosine kinase-3 (FLT3) gene occurs in 30% of acute myeloid leukemias (AMLs) and confers a poor prognosis. Thirteen relapsed or chemo-refractory FLT3-ITD(+) AML patients were treated with sorafenib (200-400 mg twice daily). Twelve patients showed clearance or near clearance of bone marrow myeloblasts after 27 (range 21-84) days with evidence of differentiation of leukemia cells. The sorafenib response was lost in most patients after 72 (range 54-287) days but the FLT3 and downstream effectors remained suppressed. Gene expression profiling showed that leukemia cells that have become sorafenib resistant expressed several genes including ALDH1A1, JAK3, and MMP15, whose functions were unknown in AML. Nonobese diabetic/severe combined immunodeficiency mice transplanted with leukemia cells from patients before and during sorafenib resistance recapitulated the clinical results. Both ITD and tyrosine kinase domain mutations at D835 were identified in leukemia initiating cells (LICs) from samples before sorafenib treatment. LICs bearing the D835 mutant have expanded during sorafenib treatment and dominated during the subsequent clinical resistance. These results suggest that sorafenib have selected more aggressive sorafenib-resistant subclones carrying both FLT3-ITD and D835 mutations, and might provide important leads to further improvement of treatment outcome with FLT3 inhibitors.


Subject(s)
Antineoplastic Agents/administration & dosage , Benzenesulfonates/administration & dosage , Drug Resistance, Neoplasm , Gene Expression Regulation, Leukemic , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Mutation , Pyridines/administration & dosage , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , fms-Like Tyrosine Kinase 3/genetics , Adult , Aldehyde Dehydrogenase/biosynthesis , Aldehyde Dehydrogenase/genetics , Aldehyde Dehydrogenase 1 Family , Animals , Antineoplastic Agents/adverse effects , Benzenesulfonates/adverse effects , Bone Marrow/enzymology , Bone Marrow/pathology , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Female , Gene Expression Profiling , Humans , Janus Kinase 3/biosynthesis , Janus Kinase 3/genetics , Leukemia, Myeloid, Acute/enzymology , Leukemia, Myeloid, Acute/pathology , Male , Matrix Metalloproteinase 15/biosynthesis , Matrix Metalloproteinase 15/genetics , Mice , Mice, Inbred NOD , Mice, SCID , Middle Aged , Neoplasm Transplantation , Niacinamide/analogs & derivatives , Phenylurea Compounds , Protein Structure, Tertiary , Pyridines/adverse effects , Retinal Dehydrogenase , Sorafenib , Time Factors , Transplantation, Heterologous , fms-Like Tyrosine Kinase 3/metabolism
8.
Exp Hematol ; 38(1): 3-10, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19837128

ABSTRACT

OBJECTIVE: Xenogeneic transplantation has been the gold standard for enumeration of leukemia initiating cells in acute myeloid and lymphoblastic leukemia (ALL). Most transplantation models have required conditioning in which the recipients were either irradiated or treated with chemotherapy prior to injection of human leukemia cells. In this study, we reported an undescribed model in which adult ALL cells were injected into unconditioned newborn nonobese diabetic severe combined immunodeficient mice via an intrahepatic route. MATERIALS AND METHODS: Bone marrow (BM) and peripheral blood were collected from patients with ALL at diagnosis or relapse. CD34(+) selected lymphoblasts or mononuclear cells were transplanted as mentioned previously. Cells were also transplanted into sublethally irradiated adult mice via intravenous route for comparison. Leukemia engraftment was enumerated from mouse BM 6 to 18 weeks after transplantation. Clonality of the engrafting cells was examined based on IGH rearrangement and fluorescent in situ hybridization. RESULTS: Five of 13 ALL samples engrafted into the recipient BM 6 to 18 weeks after transplantation. Engrafted cells recapitulated the immunophenotype and cytogenetic characteristics of the original samples. Engraftment in BM and peripheral blood was significantly correlated. Importantly, there was significant correlation of engraftment between this and the conventional adult nonobese diabetic severe combined immunodeficient mouse model involving irradiation. CONCLUSION: Our results demonstrated that this unconditioned newborn mouse model could be used for enumeration of leukemia initiating cells in ALL and should be further evaluated.


Subject(s)
Neoplasm Transplantation , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Adult , Animals , Animals, Newborn , Base Sequence , Cell Lineage , DNA Primers , Female , Humans , In Situ Hybridization, Fluorescence , Male , Mice , Mice, Inbred NOD , Mice, SCID , Middle Aged , Spectral Karyotyping , Transplantation, Heterologous
9.
Leuk Res ; 34(1): 119-22, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19683812

ABSTRACT

In this study, we tested if FLT3/internal tandem duplication (ITD) in acute myeloid leukemia (AML) might occur at different hierarchical stages during leukemogenesis. In 56 AML cases, 10 showed FLT3/ITD (single ITD=5; multiple ITD=5). Myeloblasts from seven cases (CD34-selected=4; unselected=3) were transplanted into NOD/SCID mice. Five cases engrafted successfully into 14 mice. Two patients carried single FLT3/ITD subclones, which were maintained during primary and secondary transplantations. In three patients with multiple FLT3/ITD subclones, some subclones persisted or expanded while others diminished upon transplantation. Their different engraftment capabilities in NOD/SCID mice supported the proposition that FLT3/ITD might occur at different stages during leukemogenesis.


Subject(s)
Gene Duplication , Leukemia, Myeloid, Acute/genetics , fms-Like Tyrosine Kinase 3/genetics , Amino Acid Sequence , Animals , Base Sequence , DNA Primers , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Molecular Sequence Data , Polymerase Chain Reaction , fms-Like Tyrosine Kinase 3/chemistry
10.
Eur J Haematol ; 83(3): 191-202, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19467017

ABSTRACT

OBJECTIVE: Activation of PPARgamma by its ligands has shown potential anti-neoplastic effects in solid tumors. In this study, we investigate the effects of rosiglitazone (RGZ) alone as well as in combination with all trans-retinoic acid (ATRA) on human myeloma cell lines and try to address its potential mechanism. METHODS: U266, RPMI-8226 and primary myeloma cells from patients were treated with different concentrations of RGZ in the presence or absence of ATRA and various biological responses were studied by the methods of [3H] thymidine incorporation, MTT, cell cycle analysis, Annexin V-PI staining, Wright-Giemsa staining, CD49e expression assay, light chain protein detection, RT-PCR and caspase-3 activity assay. RESULTS: We report that exposure to RGZ induced proliferation inhibition and viability reduction in a dose-dependent manner in both U266 and RPMI-8226 cells. A similar exposure to RGZ also induced cell cycle arrest and cell apoptosis of myeloma cells. A combination of RGZ with ATRA enhanced the effects of RGZ and induced cell cycle arrest and apoptosis more profoundly in both cell lines. RGZ treated cells displayed morphological characteristics of cell differentiation, and more evident signs of differentiation were observed when combined with ATRA. These changes were confirmed by the detection of CD49e expression and light chain protein secretion. Similar cell apoptosis and differentiation were observed when primary CD138+ myeloma cells were treated with RGZ and ATRA. The mRNA expressions of FLIP, XIAP and survivin were detected in both cell lines and the levels decreased significantly after culture with RGZ. The addition of ATRA in culture medium made these changes more apparently. Caspase-3 activity was increased upon exposure to RGZ in both U266 and RPMI-8226 cells while combination of RGZ and ATRA brought out more effective activation of caspase-3. Similar apoptosis and cell differentiation induced by RGZ and ATRA can also be observed in primary CD138+ cells from myeloma patients. CONCLUSION: Concomitant RXRalpha activation by ATRA enhanced the inhibitory effects of RGZ on myeloma cell proliferation, cell cycle, apoptosis and differentiation. Combination of RGZ and ATRA may be a useful therapy for human multiple myeloma.


Subject(s)
Multiple Myeloma/drug therapy , Multiple Myeloma/pathology , Thiazolidinediones/pharmacology , Tretinoin/pharmacology , Aged , Apoptosis , Caspase 3/metabolism , Cell Differentiation , Cell Line, Tumor , Cell Survival , Female , Humans , Hypoglycemic Agents/pharmacology , Male , Middle Aged , PPAR gamma/metabolism , Rosiglitazone
11.
Br J Haematol ; 144(4): 484-91, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19055666

ABSTRACT

To examine the differences between primitive bone marrow (BM) and peripheral blood (PB) myeloblasts in acute myeloid leukaemia (AML), we compared CD34(+) myeloblasts of paired BM and PB samples from 14 AML patients in terms of surface phenotype, homing and engraftment in a xenogeneic transplantation model, and gene expression, based on microarray studies and quantitative polymerase chain reaction. While there was no significant difference in surface phenotypes between these two populations, in vivo assay showed significantly better homing potential of PB CD34(+) cells than BM CD34(+) cells. Significant correlation between homing and engraftment in AML samples was also noted. In addition, gene expression profiling of CD34(+) cells from five paired BM and PB leukaemic samples showed that genes involved in G-protein and prostaglandin signalling, chemotaxis and stress response, cell proliferation and apoptosis were down-regulated in PB CD34(+) myeloblasts. These data suggested that circulating primitive myeloblasts in AML are functionally different from those residing in the marrow compartment, and such differences may be partly regulated by the BM microenvironment.


Subject(s)
Antigens, CD34/analysis , Bone Marrow Cells/physiology , Granulocyte Precursor Cells/physiology , Leukemia, Myeloid, Acute/pathology , Adult , Aged , Animals , Antigens, Neoplasm/analysis , Female , Gene Expression Profiling/methods , Graft Survival , Humans , Immunophenotyping , Leukemia, Myeloid, Acute/blood , Male , Mice , Mice, SCID , Middle Aged , Neoplasm Transplantation , Prospective Studies , Reverse Transcriptase Polymerase Chain Reaction/methods , Transplantation, Heterologous
12.
Zhonghua Zhong Liu Za Zhi ; 31(12): 885-9, 2009 Dec.
Article in Chinese | MEDLINE | ID: mdl-20193324

ABSTRACT

OBJECTIVE: To investigate the effects of PPARgamma ligand (rosiglitazone, RGZ) as well as combined with all trans-retinoic acid (ATRA) on human myeloma cells and try to explore the possible mechanism. METHODS: Human myeloma cell lines U266 and RPMI-8226 cells were treated with RGZ in the presence or absence of ATRA. Cell proliferation was evaluated by [(3)H] thymidine incorporation, cell cycle distribution and CD49e expression were analyzed by flow cytometry, morphology changes were evaluated by Wright-Giemsa staining, and p27(Kip1) and p21(Waf1) expression was detected by Western blotting. RESULTS: The exposure to RGZ induced proliferation inhibition in both cell lines in a dose-dependent manner. After cultured with 5 micromol/L RGZ, the proportion of U266 and RPMI-8226 cells in phase G(0)/G(1) was (45.2 +/- 6.7)% and (40.3 +/- 7.3)%, respectively (P < 0.05). The proportion of the cells in phase G(2)/M and S was (52.2 +/- 7.4)% and (57.4 +/- 9.5)%, respectively (P < 0.05). These changes were more evident when the RGZ concentration was increased to 10 micromol/L. A combination of RGZ with ATRA enhanced the growth inhibition and cell cycle arrest effects of RGZ. The RGZ-treated myeloma cells displayed morphological characteristics of cell differentiation, and more evident signs of differentiation were observed when RGZ was combined with ATRA. These changes were confirmed by the detection of CD49e expression. The expression of p27(Kip1) and p21(Waf1) in myeloma cells was up-regulated by RGZ and this change was more apparent when RGZ was used in combination with ATRA. CONCLUSION: RGZ can induce cell cycle arrest and cell differentiation in myeloma cells which maybe caused by up-regulation of p27(Kip1) and p21(Waf1) expression. ATRA can enhance these effects of RGZ on multiple myeloma cells and combined use of these two drugs may show a synergistic effect on myeloma cells.


Subject(s)
Cell Proliferation/drug effects , Multiple Myeloma/pathology , Thiazolidinediones/pharmacology , Tretinoin/pharmacology , Cell Cycle/drug effects , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p27 , Dose-Response Relationship, Drug , Drug Synergism , Humans , Integrin alpha5/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Multiple Myeloma/metabolism , PPAR gamma/agonists , Rosiglitazone , Thiazolidinediones/administration & dosage , Up-Regulation
13.
Exp Hematol ; 35(1): 56-63, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17198874

ABSTRACT

OBJECTIVE: We have previously shown that all-trans retinoic acid (ATRA) enhanced the maintenance of early human hematopoietic progenitor cells (HPCs) in the presence of an irradiated stromal cell line AFT024. In this study, we examined the effects of ATRA on the stromal cell component with particular reference to cellular proliferation and gene expression. METHODS: Irradiated AFT024 cells were cultured in Dulbecco's Modified Eagle's Medium supplemented with fetal bovine serum and were incubated with ATRA at 1 mumol/L up to 21 days. The cells were examined in terms of immunostaining for proliferative cell nuclear antigen (PCNA) and BrdU incorporation, apoptosis assay, cell cycle analysis, and gene expression using semiquantitative reverse-transcriptase polymerase chain reaction. RESULTS: In the control experiments, AFT024 cells lost their confluence in culture after 15-Gy irradiation and were arrested in G2/M phase on days 7 and 21. ATRA restored the cellular confluence with an increase in proliferation on day 21 (BrdU incorporation: 20.6-fold; PCNA staining: 51.7-fold) with reversal of cell cycle arrest (S phase: 2.7-fold increase; G2/M phase: 2.0-fold decrease). There was no effect on apoptosis as shown by terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling staining. ATRA significantly upregulated the expression of cell cycle genes for checkpoint transition, including cyclin A2, B2, and aurora kinase B, as well as genes associated with a putative role in HPC maintenance, including osteopontin, HoxA5, enhancer of zeste homolog 2, and peroxisome proliferator-activated receptor gamma. CONCLUSION: We concluded that ATRA induced cellular proliferation of irradiated AFT024 cells and expression of a number of genes whose relevance to HPC homeostasis would have to be further examined.


Subject(s)
Cell Proliferation/drug effects , Gene Expression Regulation/drug effects , Stromal Cells/cytology , Tretinoin/pharmacology , Animals , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Line , G2 Phase/drug effects , G2 Phase/radiation effects , Gene Expression Regulation/genetics , Genes, cdc/drug effects , Mice , Stem Cells/cytology , Up-Regulation/drug effects , Up-Regulation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...