Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
Leukemia ; 22(4): 708-22, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18337766

ABSTRACT

The Raf/MEK/ERK, PI3K/PTEN/Akt/mTOR and Jak/STAT pathways are frequently activated in leukemia and other hematopoietic disorders by upstream mutations in cytokine receptors, aberrant chromosomal translocations as well as other genetic mechanisms. The Jak2 kinase is frequently mutated in many myeloproliferative disorders. Effective targeting of these pathways may result in suppression of cell growth and death of leukemic cells. Furthermore it may be possible to combine various chemotherapeutic and antibody-based therapies with low molecular weight, cell membrane-permeable inhibitors which target the Raf/MEK/ERK, PI3K/PTEN/Akt/mTOR and Jak/STAT pathways to ultimately suppress the survival pathways, induce apoptosis and inhibit leukemic growth. In this review, we summarize how suppression of these pathways may inhibit key survival networks important in leukemogenesis and leukemia therapy as well as the treatment of other hematopoietic disorders. Targeting of these and additional cascades may also improve the therapy of chronic myelogenous leukemia, which are resistant to BCR-ABL inhibitors. Furthermore, we discuss how targeting of the leukemia microenvironment and the leukemia stem cell are emerging fields and challenges in targeted therapies.


Subject(s)
Apoptosis/drug effects , Drug Delivery Systems , Leukemia/drug therapy , Signal Transduction/drug effects , Humans , Leukemia/pathology
2.
Leukemia ; 22(4): 686-707, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18337767

ABSTRACT

Mutations and chromosomal translocations occur in leukemic cells that result in elevated expression or constitutive activation of various growth factor receptors and downstream kinases. The Raf/MEK/ERK, PI3K/PTEN/Akt/mTOR and Jak/STAT pathways are often activated by mutations in upstream genes. The Raf/MEK/ERK and PI3K/PTEN/Akt/mTOR pathways are regulated by upstream Ras that is frequently mutated in human cancer. Recently, it has been observed that the FLT-3 and Jak kinases and the phosphatase and tensin homologue deleted on chromosome 10 (PTEN) phosphatase are also frequently mutated or their expression is altered in certain hematopoietic neoplasms. Many of the events elicited by the Raf/MEK/ERK, PI3K/PTEN/Akt/mTOR and Jak/STAT pathways have direct effects on survival pathways. Aberrant regulation of the survival pathways can contribute to uncontrolled cell growth and lead to leukemia. In this review, we describe the Raf/MEK/ERK, PI3K/PTEN/Akt/mTOR and Jak/STAT signaling cascades and summarize recent data regarding the regulation and mutation status of these pathways and their involvement in leukemia.


Subject(s)
Leukemia/etiology , Signal Transduction , Humans , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinases/metabolism , STAT Transcription Factors/metabolism
3.
Curr Pharm Biotechnol ; 7(6): 397-405, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17168655

ABSTRACT

Currently, Arsenic Trioxide (ATO) is considered the treatment of choice for patients with relapsed acute promyelocytic leukemia (APL). Recently, a durable remission with minimal toxicity by single agent ATO or ATO + ATRA in newly diagnosed APL was reported by different groups. These regimens have minimal toxicity and can be administered on an outpatient basis after remission induction, thus they could become a real, less toxic and more economic option to ATRA + anthracyclines in particular in low risk APL, or in patients that cannot undergo chemotherapy because of age or comorbid conditions and in patients that refuse chemotherapy. Significantly, these therapies are a successful attempt to cure a tumoral disease without chemotherapy. The results of clinical trials of ATO administration as single agent in multiple myeloma (MM) and myelodisplastic syndromes (MDS) were encouraging and showed clinical effects but they were not close to APL success. On the contrary, results of clinical trials to treat non-APL acute myeloid leukemia (AML) were disappointing. We suggest that a combination therapy with drugs targeting specific pro-survival molecules or capable to enhance pro-apoptotic pathways may lead to an improvement of ATO efficacy against hematological malignancies, in particular AML. Our pre-clinical studies showed that ATO is capable to induce cell death in acute leukemia cells but the apoptotic function is limited since it can induce also a mechanism of cell defense by activating pro-survival molecules such as MEK-ERK, Bcl-xL, Bcl-2. By combining ATO with specific MEK inhibitors, we demonstrated that the block of MEK-ERK phosphorylation, the induction of Bad de-phosphorylation, and activation of p53AIP1 apoptotic pathway interrupt the pro-survival mechanisms of ATO and kill the leukemic cells by apoptotic synergism. Our results provide an experimental basis for combined or sequential treatment with MEK inhibitors and ATO in AML. The renaissance of ATO as a drug in moderne medicine may be considered, together with ATRA success, a victory of empirical analysis, that had (and has) great impact on Chinese culture.


Subject(s)
Arsenicals/therapeutic use , Clinical Trials as Topic/trends , Hematologic Neoplasms/drug therapy , Multiple Myeloma/drug therapy , Oxides/therapeutic use , Antineoplastic Agents/therapeutic use , Arsenic Trioxide , Humans , Practice Patterns, Physicians'/trends , Treatment Outcome
4.
Leukemia ; 19(2): 234-44, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15538402

ABSTRACT

Recent studies suggest that components of the prosurvival signal transduction pathways involving the Ras-mitogen-activated protein kinase (MAPK) can confer an aggressive, apoptosis-resistant phenotype to leukemia cells. In this study, we report that acute promyelocytic leukemia (APL) cells exploit the Ras-MAPK activation pathway to phosphorylate at Ser112 and to inactivate the proapoptotic protein Bad, delaying arsenic trioxide (ATO)-induced apoptosis. Both in APL cell line NB4 and in APL primary blasts, the inhibition of extracellular signal-regulated kinases 1/2 (ERK1/2) and Bad phosphorylation by MEK1 inhibitors enhanced apoptosis in ATO-treated cells. We isolated an arsenic-resistant NB4 subline (NB4-As(R)), which showed stronger ERK1/2 activity (2.7-fold increase) and Bad phosphorylation (2.4-fold increase) compared to parental NB4 cells in response to ATO treatment. Upon ATO exposure, both NB4 and NB4-As(R) cell lines doubled protein levels of the death antagonist Bcl-xL, but the amount of free Bcl-xL that did not heterodimerize with Bad was 1.8-fold greater in NB4-As(R) than in the parental line. MEK1 inhibitors dephosphorylated Bad and inhibited the ATO-induced increase of Bcl-xL, overcoming ATO resistance in NB4-As(R). These results may provide a rationale to develop combined or sequential MEK1 inhibitors plus ATO therapy in this clinical setting.


Subject(s)
Antineoplastic Agents/toxicity , MAP Kinase Kinase 1/antagonists & inhibitors , Oxides/toxicity , Apoptosis/drug effects , Arsenic Trioxide , Arsenicals , Cell Line, Tumor , Drug Resistance, Neoplasm , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Growth Inhibitors/toxicity , Humans , Kinetics , Leukemia, Promyelocytic, Acute , Membrane Potentials/drug effects , Mitochondria/drug effects , Mitochondria/pathology , Mitochondria/physiology , RNA, Small Interfering/genetics , Transfection
5.
Leukemia ; 18(3): 628-35, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14737074

ABSTRACT

The mitogen-activated protein (MAP) cascade leading to the activation of extracellular signal-regulated kinases 1/2 (ERK1/2) is critical for regulating myeloma cell growth; however, the relationship of ERK1/2 activity with vascular endothelial growth factor (VEGF) production and the effects of its downmodulation in myeloma cells are not elucidated. We found that the treatment with MAP/ERK kinase 1 (MEK1) inhibitors PD98059 or PD184352 produced a reduction of phosphorylated ERK1/2 (p-ERK1/2) levels in myeloma cells of more than 80% and prevented the increase of p-ERK1/2 induced by interleukin-6 (IL-6). MEK1 inhibitors also induced a significant inhibition of myeloma cell proliferation and blunted the stimulatory effect induced by IL-6. A significant inhibition of basal VEGF secretion by myeloma cells as well as a suppression of the stimulatory effect of IL-6 on VEGF was observed by either PD98059 or PD184352. Moreover, we also found that the PI3K kinase inhibitors, but not p38 MAPK inhibitors, reduced VEGF secretion by myeloma cells and increase the inhibitory effect of MEK1 inhibitors. In an 'in vitro' model of angiogenesis, we found that MEK1 inhibitors impair vessel formation induced by myeloma cells and restored by VEGF treatment, suggesting that the downmodulation of ERK1/2 activity reduces myeloma-induced angiogenesis by inhibiting VEGF secretion.


Subject(s)
Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Multiple Myeloma/metabolism , Neovascularization, Pathologic/metabolism , Vascular Endothelial Growth Factor A/metabolism , Cell Division/drug effects , Down-Regulation , Flavonoids/pharmacology , Humans , Interleukin-6/metabolism , MAP Kinase Kinase 1 , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Multiple Myeloma/pathology , Phosphorylation/drug effects , Signal Transduction/drug effects , Tumor Cells, Cultured
6.
Leukemia ; 17(9): 1783-93, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12970778

ABSTRACT

MAP kinase/ERK kinase (MEK)-extracellular signal-regulated kinase (ERK) kinases are frequently activated in acute myelogenous leukemia (AML), and can have prosurvival function. The purpose of this study was to induce downmodulation of MEK-ERK activation in AML primary blasts in order to detect the effect on cell cycle progression and on the apoptosis of leukemic cells. We investigated 14 cases of AML with high ERK 1/2 activity and four cases with undetectable or very low activity. After 24 h incubation of the AML blasts with high ERK activity using PD98059 (New England BioLabs, Beverly, MA, USA), a selective inhibitor of MEK1 phosphorylation, at concentrations of 20 and 40 microM, we observed a strong decrease in the levels of ERK1/2 activity. A significant decrease of blast cell proliferation compared with untreated controls was found. In contrast, the proliferation of blast cells that expressed low or undetectable levels of ERK activity was not inhibited. Time-course analysis demonstrated that the downmodulation of MEK1/2, ERK1 and ERK2 dual-phosphorylation was evident even after 3 h of treatment with 20 and 40 microM. The cleavage of poly(ADP-ribose) polymerase (PARP), an early sign of apoptosis, appeared after 18 h of PD98059 treatment at concentrations of 20 and 40 microM in eight of the 14 cases. After 24 h of treatment, cleaved PARP appeared in all 14 cases. Time-course analysis of cell cycle progression and apoptosis showed that PD98059 induced a G1-phase accumulation with low or undetectable levels of apoptosis after 24 h incubation; after 48 and 72 h incubation, a significant increase of apoptosis was observed. Thus, the primary effect of ERK downmodulation was a cell cycle arrest followed by the apoptosis of a significant percentage of the leukemic blasts. The preclinical model of leukemia treatment reported in this paper makes further comment with regard to MEK1 inhibition as a useful antileukemic target, and encourages the conducting of in vivo studies and clinical investigations.


Subject(s)
Apoptosis , Enzyme Inhibitors/pharmacology , Leukemia, Myeloid, Acute/pathology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Adult , Aged , Caspases/metabolism , Cell Differentiation/drug effects , Cell Division , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Cells, Cultured/pathology , Down-Regulation , Female , Flavonoids/pharmacology , Flow Cytometry , G1 Phase/drug effects , Hematopoietic Stem Cells/metabolism , Humans , Leukemia, Myeloid, Acute/enzymology , Male , Middle Aged , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Phosphorylation , Poly(ADP-ribose) Polymerases/metabolism
7.
Hematol J ; 2(2): 70-80, 2001.
Article in English | MEDLINE | ID: mdl-11423998

ABSTRACT

INTRODUCTION: We report the results of a study investigating signaling proteins in 26 cases of primary acute myelogenous leukemia. We studied the Shc adaptor proteins p52/p46Shc, which can activate the RAS/Mitogen Activated Protein kinase pathway, p66Shc which is uncoupled from RAS/MAP kinases and the MAP kinase family members Extracellular signal Regulated Kinase (ERK) and c-Jun NH2-terminal protein Kinase (JNK) or Stress Activated Protein Kinase (SAPK). MATERIAL AND METHODS: CD34+ and CD34- fractions of four human normal bone marrow and unfractionated bone marrow samples were investigated. Immunoblottings, immunoenzymatic and in vitro assays were performed. RESULTS: Shc protein isoforms were constitutively expressed in all the AML cases examined. Tyrosine-phosphorylation of p53/p46Shc isoforms were found in CD34+ but not in the majority of CD34- cases. p66Shc isoform was not tyrosine-phosphorylated in CD34-, and was tyrosine-phosphorylated only in some CD34+ cases. Expression and activation of ERK was constitutively present in the majority of AML patients analysed. JNK/SAPK was expressed but not activated in the AMLs examined. Activation occurred after treatment of the leukemic cells by anisomycin, etoposide, and cytarabine. ERK and JNK/SAPK activation were not detectable in the hematopoietic precursors of human normal bone-marrow. CONCLUSION: These data bear implications for the role of Shc-MAP kinase pathway in normal hemopoiesis and AML leukemogenesis.


Subject(s)
Antineoplastic Agents/pharmacology , Gene Expression Regulation, Leukemic/drug effects , Hematopoietic Stem Cells/metabolism , Leukemia, Myeloid, Acute/metabolism , MAP Kinase Signaling System/drug effects , Adaptor Proteins, Signal Transducing/metabolism , Adult , Aged , Antigens, CD34/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Hematopoietic Stem Cells/pathology , Humans , Leukemia, Myeloid, Acute/pathology , Male , Middle Aged , Shc Signaling Adaptor Proteins , Src Homology 2 Domain-Containing, Transforming Protein 1 , Tumor Cells, Cultured
8.
J Inorg Biochem ; 83(2-3): 169-79, 2001 Jan 15.
Article in English | MEDLINE | ID: mdl-11237256

ABSTRACT

Three new complexes of transition metals as copper, nickel and cobalt with 5-formyluracil thiosemicarbazone (H3ut) have been synthesised and characterised by single-crystal X-ray diffraction. In all compounds the ligand behaves as SNO terdentate. In the copper complex the coordination geometry is square pyramidal with the ligand lying on the basal plane and two water molecules that complete the metal environment, the nickel compound is surrounded by six donor atoms (three of the ligand, two water oxygen atoms and a chlorine atom) in an octahedral fashion, and cobalt also shows an octahedral geometry but determined only by two terdentate ligand molecules. These three compounds have been tested on human leukemic cell lines K562 and CEM. The nickel and cobalt complexes have demonstrated low activity in cell growth, while the copper complex that is more active has been tested also on a third leukemic human cell line (U937), but it was not able to induce apoptosis on all cell lines.


Subject(s)
Cell Division/drug effects , Thiosemicarbazones/chemistry , Thiosemicarbazones/pharmacology , Uracil/analogs & derivatives , Uracil/chemistry , Uracil/pharmacology , Apoptosis/drug effects , Apoptosis/physiology , Cobalt/chemistry , Cobalt/pharmacology , Copper/chemistry , Copper/pharmacology , Crystallography, X-Ray , Drug Screening Assays, Antitumor , Humans , Ligands , Molecular Structure , Mutagens/chemical synthesis , Mutagens/chemistry , Mutagens/pharmacology , Nickel/chemistry , Nickel/pharmacology , Thiosemicarbazones/chemical synthesis , Tumor Cells, Cultured , Uracil/chemical synthesis
9.
Ann Ital Med Int ; 15(3): 206-13, 2000.
Article in Italian | MEDLINE | ID: mdl-11059061

ABSTRACT

The authors discuss the importance that molecular medicine has assumed in recent years. Molecular methodologies have clearly demonstrated that immunological diversity is based fundamentally on the rearrangement of the genes encoding antigen B and T cell receptors. The importance of oncogenes, and their translocation in tumoral pathologies is emphasized, a case in point being the alterations observed in chronic myeloid leukemia and acute promyelocytic leukemia and their implication for innovative therapy. The importance of prothrombin and factor V genetic-molecular alterations in thromboembolic pathology and of the activation of calcineurin phosphatase or other intracellular signal regulator molecules during cardiac insufficiency genesis is also discussed. Particular attention is paid to progress regarding the socially important Alzheimer's syndrome, and the diagnosis of endocrine tumors. Moreover, the authors believe that the identification of new endocrine nuclear receptors, "orphans" of hormonal ligands, will open up interesting prospects--even therapeutic--in endocrinology. The authors conclude by reviewing the therapeutic prospects for immunodeficiency syndromes and malignant tumors, offered by new gene therapy methodologies. They also discuss recent results of studies on the aging process which, until not many years ago, appeared adventuristic. Today they are opening prospects of great interest.


Subject(s)
Genetics, Medical , Acquired Immunodeficiency Syndrome/genetics , Adult , Aging/genetics , Alzheimer Disease/genetics , Cardiovascular Diseases/genetics , Central Nervous System Diseases/genetics , Colonic Neoplasms/genetics , DNA, Neoplasm/genetics , Endocrine System Diseases/genetics , Genetic Therapy , HIV Infections/genetics , Hepatitis C/genetics , Humans , Immunologic Deficiency Syndromes/therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Promyelocytic, Acute/genetics , Mutation , Neoplasms/therapy , Research , Thromboembolism/genetics , Transcription, Genetic , Translocation, Genetic
10.
Bioorg Med Chem ; 8(1): 157-62, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10968274

ABSTRACT

As part of a research programme aimed at the synthesis of compounds with antiviral, antibacterial and antitumor properties and their spectroscopic characterization, new thiosemicarbazones deriving from natural aldehydes have been investigated. These substances contain in the same molecule both a chain with nucleophilic centres N, S with tubercolostatic activity, and a glycosidic or alkyl moiety (modified glycosides and nucleosides have recently received a great deal of attention in the fields of neoplastic diseases and viral infections). In this paper the synthesis and the characterization of these compounds by means of 1H NMR, IR, and MS techniques is reported. Biological studies have involved both inhibition of cell proliferation and apoptosis tests on human leukemia cell line U937.


Subject(s)
Spectrum Analysis/methods , Thiosemicarbazones/chemical synthesis , Thiosemicarbazones/pharmacology , Apoptosis/drug effects , Cell Division/drug effects , Humans , Molecular Structure , Thiosemicarbazones/chemistry , U937 Cells
11.
Cancer Res ; 60(3): 728-32, 2000 Feb 01.
Article in English | MEDLINE | ID: mdl-10676660

ABSTRACT

The BCR/ABL fusion protein is a constitutively active tyrosine kinase that is responsible for the pathogenesis of chronic myelogenous leukemia (CML). Clinically, CML is characterized by a chronic phase (CP) that eventually terminates into a blast crisis (BC). BC transformation is associated with accumulation of CD34+ blasts. We investigated the expression and phosphorylation of Src-homology-2 and collagen-homology domains (SHC) [corrected] proteins in subpopulations of CML primary cells. Shc polypeptides are tyrosine kinase substrates that are constitutively tyrosine-phosphorylated in continuous cell lines of CML origin. High levels of Shc expression were found in the CD34+ cells from CML-BC, CML-CP and normal bone marrow. In contrast, CD34- fractions from CML-CP and normal bone marrow expressed low levels of p46Shc. Shc proteins were constitutively phosphorylated in the CD34+ fractions from CML cells (both CP and BC), but not in normal CD34+ cells. These data bear implications for the role of Shc in normal hemopoiesis and CML leukemogenesis: (a) dramatic changes of Shc expression during terminal differentiation of hemopoietic cells adds a further level of regulation to the signal transduction function of Shc; and (b) constitutive Shc tyrosine-phosphorylation in the rare CD34+ cells of CML-CP might contribute to the selection of this subpopulation during the blast crisis transformation of CMLs.


Subject(s)
Adaptor Proteins, Signal Transducing , Adaptor Proteins, Vesicular Transport , Antigens, CD34/analysis , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Proteins/metabolism , src Homology Domains , Bone Marrow/chemistry , Granulocyte Colony-Stimulating Factor/pharmacology , Humans , Phosphorylation , Proteins/analysis , Shc Signaling Adaptor Proteins , Src Homology 2 Domain-Containing, Transforming Protein 1
12.
J Inorg Biochem ; 76(3-4): 277-84, 1999 Sep 30.
Article in English | MEDLINE | ID: mdl-10605841

ABSTRACT

Reaction of the title ligands (HPyTSC and HS(S)PPh2, respectively) with R2SnO (R = Me, Et, Bu) in ethanol (EtOH) afforded the complexes [SnMe2(PyTSC) (S2PPh2)].EtOH (1) and [SnR2(PyTSC) (S2PPh2)] (R = Et (2), Bu (3)). The structures of 1 and 2 were determined by single-crystal X-ray diffractometry. In both these complexes the tin atom is coordinated to an N,N,S-dentate thiosemicarbazonate ligand, an anisobidentate dithiophosphinato ligand and the two R groups. The coordination polyhedrons can be described as distorted pentagonal bipyramids. A comparative study of the IR spectra of 1, 2 and 3 indicates that the butyl complex has a similar structure. Multinuclear (1H, 13C, 31P and 119Sn) NMR data suggest that the structures of 1 and 2 probably remain in CDCl3 (or DMSO-d6) solution but compound 3 partially decomposes in these media. Preliminary results on the effects of the complexes on the proliferation and differentiation of FLC, CEM, U937, K562 and TOM-1 leukaemia cells, and on the clonogenic activity of K562 cells are also described.


Subject(s)
Organotin Compounds/chemistry , Organotin Compounds/pharmacology , Cell Differentiation/drug effects , Cell Division/drug effects , Crystallography, X-Ray , Humans , K562 Cells , Ligands , Magnetic Resonance Spectroscopy , Molecular Structure , Organotin Compounds/chemical synthesis , Spectrophotometry, Infrared
13.
Lasers Surg Med ; 23(3): 141-50, 1998.
Article in English | MEDLINE | ID: mdl-9779648

ABSTRACT

BACKGROUND AND OBJECTIVE: The purpose of this study is to summarize our use of the Holmium laser as a tool in performing chondroplasties of the knee and to determine whether any untoward affects developed at the site of laser application. STUDY DESIGN/MATERIALS AND METHODS: A retrospective review of 504 laser chondroplasties of the medial femoral condyle was done. Laser parameters and the average number of joules to perform the chondroplasties were recorded. The average follow-up was 11 months. RESULTS: Preoperative MRI interpretation indicated that 8% of the patients had osteonecrosis prior to surgery. 88% of the patients were satisfied with the procedure. All failures were evaluated by X-ray, MRI, bone scan, or biopsy of the medial femoral condyle. No new cases of osteonecrosis were determined. CONCLUSION: No new cases of osteonecrosis were documented of the medial femoral condyle following laser chondroplasty utilizing the parameters in this study. The Holmium laser remains a safe and efficacious tool in performing chondroplasty.


Subject(s)
Cartilage, Articular/surgery , Femur/surgery , Knee Joint/surgery , Laser Therapy , Osteoarthritis/surgery , Adolescent , Adult , Aged , Aged, 80 and over , Arthroscopy , Biopsy , Cartilage, Articular/diagnostic imaging , Cartilage, Articular/pathology , Endoscopy , Female , Femur/diagnostic imaging , Femur/pathology , Follow-Up Studies , Holmium , Humans , Knee Joint/diagnostic imaging , Knee Joint/pathology , Magnetic Resonance Imaging , Male , Middle Aged , Osteoarthritis/diagnosis , Osteoarthritis/diagnostic imaging , Osteonecrosis/diagnosis , Osteonecrosis/surgery , Patient Satisfaction , Radiography , Radionuclide Imaging , Retrospective Studies , Safety , Treatment Failure
14.
J Inorg Biochem ; 70(2): 145-54, 1998 May.
Article in English | MEDLINE | ID: mdl-9666573

ABSTRACT

Three Cu(II) co-ordination compounds with a novel ligand, 5-formyluracil thiosemicarbazone (H3ut), have been synthesised and characterised by single-crystal X-ray diffraction and subsequently tested in vitro on human leukemic cells. The crystal structures revealed, in all three cases, a square pyramidal co-ordination geometry of the copper atom with the ligand lying on the basal plane and behaving as an SNO terdentate ligand. These three compounds have been tested on human leukemic cell line K562 and CEM. In these experiments the complexes have demonstrated to inhibit cell growth and one of them to induce apoptosis. In the paper we also report the spectrophotometric characterization of the free ligand.


Subject(s)
Copper , Organometallic Compounds/chemical synthesis , Uracil/analogs & derivatives , Apoptosis/drug effects , Cell Division/drug effects , Cell Survival/drug effects , Crystallography, X-Ray , Humans , Leukemia , Models, Molecular , Organometallic Compounds/chemistry , Organometallic Compounds/toxicity , Thiosemicarbazones/chemical synthesis , Thiosemicarbazones/chemistry , Thiosemicarbazones/toxicity , Tumor Cells, Cultured , Uracil/chemical synthesis , Uracil/chemistry , Uracil/toxicity
15.
J Inorg Biochem ; 69(4): 283-92, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9654752

ABSTRACT

The complexes [SnR2(L)] (R = Me, Et, Bu, Ph; H2L = pyridoxal thiosemicarbazone) have been prepared and characterized. In the light of the spectral properties of the complexes in the solid state (IR, mass, Mössbauer) the bideprotonated thiosemicarbazonato anion is O(phenolic)-, N(3)-, S-bonded to the tin atom which probably has trigonal bipyramidal coordination with N(3) atom and R groups occupying equatorial positions. NMR ( 1H, 13C and 119Sn) data in CDCl3 or DMSO-d6 suggest that this coordinative picture remains in these solutions. The ethyl, butyl and phenyl derivatives suppress proliferation of Friend erithroleukaemia cells (FLC). Of the pyridoxal thiosemicarbazone complexes so far evaluated. [SnBu2(L)] and [SnPh2(L)] showed the lowest thresholds for inhibition of FLC proliferation. The effects of these compounds on DMSO-induced differentiation of FLC, DNA synthesis and reverse transcriptase were also assayed.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Organotin Compounds/chemical synthesis , Organotin Compounds/pharmacology , Pyridoxal/analogs & derivatives , Thiosemicarbazones/chemical synthesis , Thiosemicarbazones/pharmacology , Animals , Antineoplastic Agents/chemistry , Cell Differentiation/drug effects , Cell Division/drug effects , DNA, Neoplasm/biosynthesis , Dimethyl Sulfoxide/pharmacology , Friend murine leukemia virus , Leukemia, Erythroblastic, Acute/drug therapy , Leukemia, Erythroblastic, Acute/metabolism , Leukemia, Erythroblastic, Acute/pathology , Magnetic Resonance Spectroscopy , Mass Spectrometry , Mice , Organotin Compounds/chemistry , Pyridoxal/chemical synthesis , Pyridoxal/chemistry , Pyridoxal/pharmacology , Reverse Transcriptase Inhibitors/chemical synthesis , Reverse Transcriptase Inhibitors/chemistry , Reverse Transcriptase Inhibitors/pharmacology , Spectroscopy, Mossbauer , Thiosemicarbazones/chemistry , Tumor Cells, Cultured
16.
J Inorg Biochem ; 66(1): 7-17, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9076969

ABSTRACT

The reaction of iron, nickel, copper, and zinc chlorides or acetates with acenaphthenequinone thiosemicarbazone, Haqtsc leads to the formation of novel complexes that have been characterized by spectroscopic studies (NMR, IR) and biological properties. The crystal structures of the free ligand Haqtsc 1 and of the compound [Ni(aqtsc)2].DMF 2, have also been determined by X-ray methods from diffractometer data. In 1, the conformation of the two nonequivalent molecules is governed by intramolecular hydrogen bonds, while an intermolecular hydrogen bond is responsible for dimer-like groups formation. In 2, the coordination geometry about nickel is distorted octahedral, and the two ligand molecules are terdentate monodeprotonated. Biological studies have shown that, for the first time at least up the used doses, a free ligand is active both in the inhibition of cell proliferation and in the induced differentiation on Friend erythroleukemia cells (FLC).


Subject(s)
Acenaphthenes/chemical synthesis , Organometallic Compounds/chemical synthesis , Thiosemicarbazones/chemical synthesis , Acenaphthenes/chemistry , Acenaphthenes/pharmacology , Animals , Cell Differentiation/drug effects , Cell Division/drug effects , Crystallography, X-Ray , DNA, Neoplasm/biosynthesis , Dimethyl Sulfoxide/pharmacology , Friend murine leukemia virus , Leukemia, Erythroblastic, Acute/drug therapy , Leukemia, Erythroblastic, Acute/pathology , Leukemia, Erythroblastic, Acute/virology , Magnetic Resonance Spectroscopy , Mice , Models, Molecular , Molecular Structure , Organometallic Compounds/chemistry , Organometallic Compounds/pharmacology , Thiosemicarbazones/chemistry , Thiosemicarbazones/pharmacology , Tumor Cells, Cultured
17.
Blood ; 88(8): 3091-100, 1996 Oct 15.
Article in English | MEDLINE | ID: mdl-8874208

ABSTRACT

Chronic myelogenous leukemia (CML) is a clonal disorder of the hematopoietic stem cell characterized by a chimeric BCR/ABL gene giving rise to a 210-kD fusion protein with dysregulated tyrosine kinase activity. We investigated the effect of genistein, a protein tyrosine kinase inhibitor, on the in vitro growth of CML and normal marrow-derived multi-potent (colony-forming unit-mix [CFU-Mix]), erythroid (burst-forming unit-erythroid [BFU-E]), and granulocyte-macrophage (colony-forming unit-granulocyte-macrophage [CFU-GM]) hematopoietic progenitors. Continuous exposure of CML and normal marrow to genistein induced a statistically significant and dose-dependent suppression of colony formation. Genistein doses causing 50% inhibition of CML and normal progenitors were not significantly different for CFU-Mix (27 mumol/L v 23 mumol/L), BFU-E (31 mumol/L v 29 mumol/L), and CFU-GM (40 mumol/L v 32 mumol/L v 32 mumol/L). Preincubation of CML and normal marrow with genistein (200 mumol/ L for 1 to 18 hours) induced a time-dependent suppression of progenitor cell growth, while sparing a substantial proportion of long-term culture-initiating cells (LTC-IC) from CML (range, 91% +/- 9% to 32% +/- 3%) and normal marrow (range, 85% +/- 8% to 38% +/- 9%). Analysis of individual CML colonies for the presence of the hybrid BCR/ABL mRNA by reverse transcription-polymerase chain reaction (RT-PCR) showed that genistein treatment significantly reduced the mean +/- SD percentage of marrow BCR/ABL+ progenitors both by continuous exposure (76% +/- 18% v 24% +/- 12%, P < or = .004) or preincubation (75% +/- 16% v 21% +/- 10%, P < or = .002) experiments. Preincubation with genistein reduced the percentage of leukemic LTC-IC from 87% +/- 12% to 37% +/- 12% (P < or = .003). Analysis of individual colonies by cytogenetics and RT-PCR confirmed that genistein-induced increase in the percentage of nonleukemic progenitors was not due to suppression of BCR/ABL transcription. Analysis of nuclear DNA fragmentation by DNA gel electrophoresis and terminal deoxynucleotidyl transferase assay showed that preincubation of CML mononuclear and CD34+ cells with genistein induced significant evidence of apoptosis. These observations show that genistein is capable of (1) exerting a strong antiproliferative effect on CFU-Mix, BFU-E, and CFU-GM while sparing the more primitive LTC-IC and (2) selecting benign hematopoietic progenitors from CML marrow, probably through an apoptotic mechanism.


Subject(s)
Apoptosis/drug effects , Bone Marrow Purging/methods , Enzyme Inhibitors/pharmacology , Fusion Proteins, bcr-abl/genetics , Hematopoietic Stem Cells/drug effects , Isoflavones/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Neoplasm Proteins/genetics , Neoplastic Stem Cells/drug effects , RNA, Messenger/analysis , RNA, Neoplasm/analysis , Adult , Aged , Aged, 80 and over , Cell Division/drug effects , DNA Fragmentation , DNA, Neoplasm/analysis , Female , Fusion Proteins, bcr-abl/antagonists & inhibitors , Genistein , Hematopoietic Stem Cells/enzymology , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Male , Middle Aged , Neoplasm Proteins/analysis , Neoplastic Stem Cells/enzymology , Neoplastic Stem Cells/pathology , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Tumor Cells, Cultured/drug effects , Tumor Stem Cell Assay
18.
Br J Haematol ; 93(3): 551-7, 1996 Jun.
Article in English | MEDLINE | ID: mdl-8652372

ABSTRACT

Receptor and nonreceptor protein tyrosine kinases (PTKs) play a key role in the control of normal and neoplastic cell growth. The availability of PTK inhibitors prompted us to evaluate the effects of genistein, a natural inhibitor of PTKs, on in vitro colony formation by normal multilineage colony-forming units (CFU-Mix), erythroid bursts (BFU-E), granulocyte-macrophage colony-forming units (CFU-GM), long-term culture-initiating cells (LTC-IC) and acute myelogenous leukaemia colony-forming units (CFU-AML). Continuous exposure of normal marrow and blood mononuclear non-adherent cells, blood CD34+CD45RA- cells, and leukaemic blasts to increasing doses of genistein (1-100 microM) resulted in a statistically significant (P < or = 0.05) dose-dependent suppression of CFU-Mix, BFU-E, CFU-GM and CFU-AML growth. Regression analysis showed that growth inhibition was linearly related to genistein concentration. Genistein dose causing 50% inhibition (ID50) of CFU-AML was significantly lower compared to CFU-GM ID50 for marrow (19 v 32 microM, P < or = 0.017), unseparated blood (19 v 44 microM, P < or = 0.028) or CD34+CD45RA- blood (19 v 36, P < or = 0.04). Preincubation of leukaemic blasts with genistein (200 microM) for 1-2h confirmed that CFU-AML were significantly more sensitive than normal marrow and blood CFU-GM to genistein. Preincubation conditions which maximally suppressed leukaemic and normal colony growth spared a substantial percentage of marrow (29 +/- 4%) and blood (40 +/- 3%) LTC-IC. In conclusion, our data demonstrate that: (a) genistein strongly inhibits the growth of normal and leukaemic haemopoietic progenitors; (b) growth inhibition is dose- and time-dependent; (c) leukaemic progenitors are more sensitive than normal progenitors to genistein-induced growth inhibition; (d) genistein exerts a direct toxic effect on haemopoietic cells while sparing a substantial proportion of LTC-IC. The potent CFU-AML growth inhibition associated with the relative resistance of normal LTC-IC strongly supports the use of genistein for marrow purging.


Subject(s)
Hematopoietic Stem Cells/drug effects , Isoflavones/pharmacology , Leukemia, Myeloid, Acute/pathology , Protein-Tyrosine Kinases/antagonists & inhibitors , Cell Division/drug effects , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Genistein , Hematopoietic Stem Cells/pathology , Humans , Tumor Cells, Cultured
19.
Exp Hematol ; 24(3): 459-65, 1996 Feb.
Article in English | MEDLINE | ID: mdl-8599976

ABSTRACT

BCL2 proto-oncogene encodes a 25-kD protein that is characteristically localized in the inner mitochondrial membrane of the cell. It has been reported that BCL2 protein has the unique functional role of blocking programmed cells death without affecting proliferation. We have analyzed the expression of the BCL2 protein in fetal hematopoietic tissues from the 10th week of gestational age onward. Fetal thymus, liver, and bone marrow and cord blood were investigated. The experiments were performed by the alkaline-antialkaline phosphatase (APAAP) technique by staining air-dried acetone-fixed cytospins and by dual-color immunofluorescent assay by staining mononuclear cell suspensions with monoclonal antibodies detecting BCL2 protein and antigens expressed by different hematopoietic subsets. Flow cytometric analyses were performed on FACSort's Comsort 32 (Becton Dickinson, San Jose, CA). The results have shown that the BCL2 protein is expressed in human fetal ontogenesis at the earliest stages examined. The major conceptual aspects of the results are 1) BCL2 is largely expressed in the hematopoietic cells during ontogenesis. BCL2+ cells include both immature and more differentiated subsets. Moreover, the 25-kD protein is expression in cell subsets well known to be high proliferating. This behavior suggests that BCL2 could have more complex functions than those previously described. 2) The expression in the major part of CD34+ cells suggests that BCL2 could play a role in stem cell survival. 3) BCL2 is expressed in not only medullary but also cortical thymocytes, where it could cooperate in positive selection processes. 4) The involvement of BCL2 in the immunosurveillance is indicated not only by its role in B and T cell lineages but also by its expression in particular subsets like that of the cytoplasmic CD3+ fetal liver NK cells. 5) The discrepancy observed between the results of transgenic mice analysis and in vitro inhibition experiments by antisense oligonucleotides performed for understanding BCL2 functions must stress the importance of the direct immunologic analysis of BCL2 in human hematopoietic cells.


Subject(s)
Fetus/metabolism , Hematopoietic Stem Cells/metabolism , Proto-Oncogene Proteins/metabolism , Bone Marrow/embryology , Bone Marrow/metabolism , Fetal Blood/metabolism , Fluorescent Antibody Technique , Gestational Age , Hematopoietic Stem Cells/immunology , Humans , Immunophenotyping , Liver/embryology , Liver/metabolism , Proto-Oncogene Mas , Proto-Oncogene Proteins c-bcl-2 , Thymus Gland/embryology , Thymus Gland/metabolism
20.
Leukemia ; 8(6): 918-23, 1994 Jun.
Article in English | MEDLINE | ID: mdl-8207984

ABSTRACT

T-cell receptor TCR-beta gene expression is an early event during human ontogenesis since the majority of thymocytes express cytoplasmic beta chain as early as the 15th week of gestation, when a complete VDJ rearrangement and functional 1.3-kb beta gene transcript are detectable. We report here our contribution with those of others on the analysis of TCR-beta gene ontogenesis. By sequencing beta gene transcripts we have demonstrated that beta gene N-regions increase dramatically in the thymus after the 20th week and that the period between 20-30 weeks is of critical importance for the acquisition of N-diversity. A correlation between TdT and N-region expression also exists. An ordered expression of TdT and cytoplasmic beta chain occurs in humans starting around the 20th week, similar to the sequence of coordinated expression of TdT and cytoplasmic mu chains detectable in B-cell precursors. TCR-beta gene behavior in T-cell neoplasms, in 'biphenotypic' leukemias and in B-ALL is also discussed. An interesting study of seven cases of B-ALL with complete V(D)J beta gene rearrangement is analyzed, as is its implication for further analysis in B-cell leukemia.


Subject(s)
Gene Rearrangement, beta-Chain T-Cell Antigen Receptor , Leukemia, T-Cell/genetics , T-Lymphocytes/physiology , Gene Expression , Humans , Leukemia, T-Cell/pathology , T-Lymphocytes/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...