Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Biochem Pharmacol ; 182: 114230, 2020 12.
Article in English | MEDLINE | ID: mdl-32979352

ABSTRACT

L-asparaginase (ASNase) from Escherichia coli (EcAII) is used in the treatment of acute lymphoblastic leukaemia (ALL). EcAII activity in vivo has been described to be influenced by the human lysosomal proteases asparaginyl endopeptidase (AEP) and cathepsin B (CTSB); these hydrolases cleave and could expose epitopes associated with the immune response against EcAII. In this work, we show that ASNase resistance to CTSB and/or AEP influences the formation of anti-ASNase antibodies, one of the main causes of hypersensitivity reactions in patients. Error-prone polymerase chain reaction was used to produce variants of EcAII more resistant to proteolytic cleavage by AEP and CTSB. The variants with enzymatic activity and cytotoxicity levels equivalent to or better than EcAII WT were submitted to in vivo assays. Only one of the mutants presented increased serum half-life, so resistance to these proteases is not the only feature involved in EcAII stability in vivo. Our results showed alteration of the phenotypic profile of B cells isolated after animal treatment with different protease-resistant proteoforms. Furthermore, mice that were exposed to the protease-resistant proteoforms presented lower anti-asparaginase antibodies production in vivo. Our data suggest that modulating resistance to lysosomal proteases can result in less immunogenic protein drugs.


Subject(s)
Antineoplastic Agents/pharmacology , Asparaginase/pharmacology , Biological Products/pharmacology , Immunogenetic Phenomena/drug effects , Lysosomes/immunology , Peptide Hydrolases/pharmacology , Amino Acid Sequence , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Asparaginase/chemistry , Asparaginase/therapeutic use , Biological Products/chemistry , Biological Products/therapeutic use , Cattle , Cell Survival/drug effects , Cell Survival/physiology , Chickens , Dose-Response Relationship, Drug , Escherichia coli , Female , Horses , Humans , Immunogenetic Phenomena/physiology , Jurkat Cells , Lysosomes/chemistry , Mice , Mice, Inbred BALB C , Peptide Hydrolases/chemistry , Peptide Hydrolases/therapeutic use , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Protein Structure, Secondary
2.
Cell Death Discov ; 5: 135, 2019.
Article in English | MEDLINE | ID: mdl-31552142

ABSTRACT

Annexin A1 (AnxA1) modulates neutrophil life span and bone marrow/blood cell trafficking thorough activation of formyl-peptide receptors (FPRs). Here, we investigated the effect of exogenous AnxA1 on haematopoiesis in the mouse. Treatment of C57BL/6 mice with recombinant AnxA1 (rAnxA1) reduced the granulocyte-macrophage progenitor (GMP) population in the bone marrow, enhanced the number of mature granulocytes Gr-1+Mac-1+ in the bone marrow as well as peripheral granulocytic neutrophils and increased expression of mitotic cyclin B1 on hematopoietic stem cells (HSCs)/progenitor cells (Lin-Sca-1+c-Kit+: LSK). These effects were abolished by simultaneous treatment with Boc-2, an FPR pan-antagonist. In in vitro studies, rAnxA1 reduced both HSC (LSKCD90lowFLK-2-) and GMP populations while enhancing mature cells (Gr1+Mac1+). Moreover, rAnxA1 induced LSK cell proliferation (Ki67+), increasing the percentage of cells in the S/G2/M cell cycle phases and reducing Notch-1 expression. Simultaneous treatment with WRW4, a selective FPR2 antagonist, reversed the in vitro effects elicited by rAnxA1. Treatment of LSK cells with rAnxA1 led to phosphorylation of PCLγ2, PKC, RAS, MEK, and ERK1/2 with increased expression of NFAT2. In long-term bone marrow cultures, rAnxA1 did not alter the percentage of LSK cells but enhanced the Gr-1+Mac-1+ population; treatment with a PLC (U73122), but not with a PKC (GF109203), inhibitor reduced rAnxA1-induced phosphorylation of ERK1/2 and Elk1. Therefore, we identify here rAnxA1 as an inducer of HSC/progenitor cell differentiation, favouring differentiation of the myeloid/granulocytic lineage, via Ca2+/MAPK signalling transduction pathways.

3.
Peptides ; 85: 41-45, 2016 11.
Article in English | MEDLINE | ID: mdl-27614284

ABSTRACT

Due to the cytotoxic effect of antimicrobial peptides (AMP) against several microorganism and tumor cells has been proposed their association with the immune system. However, just a few reports have shown this relationship. In this study, mice were treated with gomesin, a ß-hairpin AMP that exhibit high cytotoxicity against bacterial and tumor cells. Different effects in the immune system were observed, such as, decrease of CD3+ in T lymphocytes (Control: 17.7±1.4%; Gomesin: 7.67±1.2%) and in hematopoietic progenitors and increase of hematopoietic stem cell (Control: 0.046±0.004%; Gomesin: 0.067±0.003%), B220+ B lymphocytes (Control: 38.63±1.5%; Gomesin: 47.83±0.48%), and Mac-1+F4/80+ macrophages (Control: 11.76±3.4%; Gomesin: 27.13±4.0%). Additionally, macrophage increase was accompanied by an increase of macrophage phagocytosis (Control 20.85±1.53; Gomesin 31.32±1 Geometric mean), interleukin 6 (Control: 47.24±1.9ng/mL; Gomesin: 138.68±33.68ng/mL) and monocyte chemoattractant protein-1 (Control: 0.872±0.093ng/mL; Gomesin: 1.83±0.067ng/mL). Thus, this report showed immunomodulatory activity of gomesin in the immune system of mice.


Subject(s)
Antimicrobial Cationic Peptides/immunology , Cell Differentiation/genetics , Macrophage Activation/genetics , Myeloid Cells/metabolism , Animals , Antimicrobial Cationic Peptides/administration & dosage , Immune System/metabolism , Immunomodulation/genetics , Macrophage Activation/immunology , Mice , Monocytes/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
4.
Eur J Med Chem ; 79: 24-33, 2014 May 22.
Article in English | MEDLINE | ID: mdl-24709226

ABSTRACT

The search for new compounds that induce p53-independent apoptosis is the focus of many studies in cancer biology because these compounds could be more specific and would overcome chemotherapy resistance. In this study, we evaluated the in vitro antitumour activity of a Biphosphinic Palladacycle Complex (BPC) and extended preclinical studies to an in vivo model. Saos-2 cells, a p53-null human osteosarcoma drug-resistant cell line, were treated with BPC in the presence or absence of a cathepsin B inhibitor and a calcium chelator (CA074 and BAPTA-AM, respectively), and several parameters related to apoptosis were evaluated. Preclinical studies were performed with mice that were intravenously inoculated with murine melanoma B16F10-Nex2 cells and treated intraperitoneally (i.p.) with BPC (8 mg/kg/day) for ten consecutive days, when lung metastatic nodules were counted. In vitro data show that BPC induces cell death in Saos-2 cells mainly by apoptosis, which was accompanied by the effector caspase-3 activation. These events are most likely related to Bax translocation and increased cytosolic calcium mobilisation, mainly from intracellular compartments. Lysosomal Membrane Permeabilisation (LMP) was also observed after 12 h of BPC exposure. Interestingly, BAPTA-AM and CA074 significantly decreased BPC cytotoxicity, suggesting that both calcium and cathepsin B are required for BPC antitumour activity. In vivo studies demonstrated that BPC protects mice against murine metastatic melanoma. In conclusion, BPC complex is an effective anticancer compound against metastatic murine melanoma. This complex is cytotoxic to the drug-resistant osteosarcoma Saos-2 human tumour cells by inducing apoptosis triggered by calcium signalling and a lysosomal-dependent pathway.


Subject(s)
Antineoplastic Agents/pharmacology , Calcium/metabolism , Cathepsin B/metabolism , Cytosol/metabolism , Neoplasms, Experimental/drug therapy , Organometallic Compounds/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Caspase 3/metabolism , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Enzyme Activation/drug effects , Humans , Injections, Intraperitoneal , Lysosomes/metabolism , Mice , Molecular Structure , Neoplasms, Experimental/pathology , Organometallic Compounds/administration & dosage , Organometallic Compounds/chemistry , Signal Transduction/drug effects , Structure-Activity Relationship
5.
J Cancer Res Clin Oncol ; 140(6): 909-20, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24659340

ABSTRACT

BACKGROUND: As the molecular mechanisms of Cytarabine,one of the most important drugs used in the leukaemia's treatment, are only partially understood and the role of autophagy on leukaemia development and treatment is only recently being investigated, in this study, by using Chloroquine (CQ) and 3-methyladenine (3MA) as autophagy inhibitors, we aim to evaluate the contribution of an autophagic mechanism to Cytarabine (AraC)-induced death of HL60 leukaemia cells. METHODS: Trypan blue exclusion and AnnexinV/PI assays were used to evaluate HL60 cell death under AraC treatment in the presence or absence of 3MA and CQ. Western blotting and immunofluorescence experiments were performed to show the involvement of apoptosis and autophagy protein expressions. Phenotypic characterization of HL60-treated cells was performed by using immunophenotyping. Clonogenic assays were applied to analyse clonal function of HL60-treated cells. RESULTS: We observed that although autophagy inhibition by 3MA, but not CQ, increased the death of HL60 AraC cells after 24 h of treatment, no significant differences between AraC and AraC + 3MA-treated groups were observed by using clonogenic assay. In addition, increased number of immature (CD34(+)/CD38(−)Lin(−/low)) HL60 cells was found in AraC and AraC-3MA groups when compared with control untreated cells. CONCLUSIONS: Although AraC anti-leukaemia effects could be potentiated by 3MA autophagy inhibition after 24 h of exposure, leukaemia cell resistance, the main causes of treatment failure, is also promoted by autophagy initial stage impairment by 3MA, denoting the complex role of autophagy in leukaemia cells' response to chemotherapy.


Subject(s)
Adenine/analogs & derivatives , Apoptosis/drug effects , Autophagy/drug effects , Chloroquine/pharmacology , Cytarabine/pharmacology , Leukemia/drug therapy , Adenine/pharmacology , Antimalarials/pharmacology , Antimetabolites, Antineoplastic/pharmacology , Antineoplastic Combined Chemotherapy Protocols , Blotting, Western , Caspase 3/metabolism , Cell Proliferation/drug effects , Flow Cytometry , Fluorescent Antibody Technique , Humans , Immunophenotyping , Leukemia/enzymology , Leukemia/pathology , Receptors, Retinoic Acid/antagonists & inhibitors , Receptors, Retinoic Acid/metabolism , Retinoic Acid Receptor alpha , Tumor Cells, Cultured , Tumor Stem Cell Assay
6.
Peptides ; 50: 24-7, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24090593

ABSTRACT

Leptin is a cytokine that regulates food intake, energy expenditure and hematopoiesis. Based on the tridimensional structure of the human leptin molecule, six fragments have been synthesized, (Ac-Lep23-47-NH2, [LEP1]; Ac-Lep48-71-NH2, [LEP2]; Ac-Lep72-88-NH2, [LEP3]; Ac-Lep92-115-NH2, [LEP4], Ac-[Ser(117)]-Lep116-140-NH2, [LEP5] and Ac-Lep141-164-NH2, [LEP6]), and their effects on hematopoiesis were evaluated. The mice were treated with 1mg/kg LEP5 for 3 days. The mature and primitive hematopoietic populations were quantified. We observed that the mature populations from the bone marrow and spleen were not affected by LEP5. However, the peptide caused at least a two-fold increase in the number of hematopoietic stem cells, the most primitive population of the bone marrow. Additionally, the number of granulocyte/macrophage colony-forming units produced by bone marrow cells in methylcellulose also increased by 40% after treatment with LEP5, and the leptin receptor was activated. These results show that the leptin fragment LEP5 is a positive modulator of the in vivo expansion of hematopoietic stem cells.


Subject(s)
Bone Marrow Cells/drug effects , Hematopoiesis/drug effects , Hematopoietic Stem Cells/drug effects , Leptin/pharmacology , Peptide Fragments/pharmacology , Spleen/drug effects , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Proliferation/drug effects , Gene Expression/drug effects , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Injections, Intraperitoneal , Male , Mice , Mice, Inbred C57BL , Peptide Fragments/chemical synthesis , Receptors, Leptin/agonists , Receptors, Leptin/genetics , Receptors, Leptin/metabolism , Spleen/cytology , Spleen/metabolism
7.
Brain Behav Immun ; 29: 39-50, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23246529

ABSTRACT

The reports regarding the mutual influence between the central nervous system and the immune system constitute a vast and somewhat controversial body of literature. Stress is known to disturb homeostasis, impairing immunological functions. In this study, we investigated the hematopoietic response of Chlorella vulgaris (CV)-treated mice exposed to single (SST) and repeated stress (RST). We observed a reduction in the numbers of hematopoietic progenitors (HP) in the bone marrow and long-term bone marrow cultures (LTBMC) using flow cytometry and a coinciding decrease in the number of granulocyte-macrophage colonies (CFU-GM) after treatment with both stressors, but SST caused a more profound suppression. We observed a proportional increase in the colony-stimulating activity (CSA) of the serum of animals subjected to SST or RST. In the bone marrow, SST and RST induced a decrease in both mature myeloid and lymphoid populations but did not affect pluripotent hematopoietic progenitors (Lin(-)Sca-1(+)c-kit(+), LSK), and again, a more profound suppression was observed after SST. We further quantified the levels of interleukin-1α (IL-1α) and interleukin-6 (IL-6) and the number of myeloid cells in LTBMC. Both SST and RST reduced the levels of these cytokines to similar degrees. The myeloid population was also reduced in LTBMC, and SST induced a more intense suppression. Importantly, CV treatment prevented the changes produced by SST and RST in all of the parameters evaluated. Together, our results suggest that CV treatment is an effective tool for the prophylaxis of myelosuppression caused by single or repeated stressors.


Subject(s)
Chlorella vulgaris/chemistry , Hematopoiesis/physiology , Stress, Psychological/drug therapy , Stress, Psychological/physiopathology , Animals , Bone Marrow/metabolism , Bone Marrow Cells/drug effects , Flow Cytometry , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Interleukin-1alpha/biosynthesis , Interleukin-6/biosynthesis , Male , Mice , Mice, Inbred BALB C , Myeloid Cells/drug effects , Stem Cells/drug effects , Stress, Psychological/cerebrospinal fluid
8.
Eur J Pharmacol ; 678(1-3): 6-14, 2012 Mar 05.
Article in English | MEDLINE | ID: mdl-22227332

ABSTRACT

Apoptosis induction is often associated with increased autophagy, indicating interplay between these two important cellular events in cell death and survival. In this study, the programmed cell death and autophagy induced by two nitrostyrene derivative compounds (NTS1 and NTS2) was studied using the tumorigenic Ehrlich ascitic tumor (EAT) cells. EAT cells were highly sensitive to NTS1 and NTS2 cytotoxicity in a dose-dependent manner. NTS1 and NTS2 IC(50) was less than 15.0µM post 12h incubation. Apoptosis was primarily induced by both compounds, as demonstrated by an increase in Annexin-V positive cells, concurrently with cytochrome c release from mitochondria to cytosol and caspase-3 activation. Although cytosolic Ca(2+) mobilization is involved in autophagy as well as apoptosis in response to cellular stress in many cancer cell types, from the two nitrostyrene derivative compounds studied, mainly NTS1 mobilized this ion and disparate autophagy in EAT cells. These results suggest that EAT induced cell death by NTS1 and NTS2 involved a Ca(2+)-dependent and a Ca(2+)-independent pathways, respectively. In accordance with these results, the treatment of EAT cells with 3 methyladenine (3-MA), an autophagy inhibitor; significantly increased the number of apoptotic cells after NTS1 treatment, suggesting that pharmacological modulation of autophagy augments the NTS1 efficacy. Thus, we denote the importance of studies involving autophagy and apoptosis during pre-clinical studies of new drugs with anticancer properties.


Subject(s)
Apoptosis/physiology , Autophagy/physiology , Calcium/metabolism , Enzyme Inhibitors/pharmacology , Nitro Compounds/pharmacology , Styrenes/pharmacology , Adenine/analogs & derivatives , Adenine/pharmacology , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Autophagy/drug effects , Carcinoma, Ehrlich Tumor , Caspase 3/metabolism , Cell Line, Tumor , Cytochromes c/metabolism , Cytosol/metabolism , Dose-Response Relationship, Drug
9.
J Leukoc Biol ; 90(6): 1111-7, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21954285

ABSTRACT

Tocopherols promote or inhibit growth in different cell types. In the hematopoietic system, the radioprotective property of tocopherols is thought to act through the expansion of primitive hematopoietic cells. However, the mechanisms activated by tocopherols and which HPs are affected remain poorly understood. To better address these questions, mice were treated with α-tocopherol, and its effects were investigated in the BM microenvironment. α-Tocopherol induced increased proliferation in HSC/HP cells, leading to BM hyperplasia. In addition, differentiation to the granulocytic/monocytic lineage was enhanced by α-tocopherol treatment. α-Tocopherol treatment resulted in decreased basal phosphorylation of ERK1/2, PKC, and STAT-5 in HSC/HP cells. In contrast, α-tocopherol enhanced ERK1/2 activation in response to IL-3 stimulation in HSC/HP cells without altering the expression of IL-3Rs. Moreover, α-tocopherol-induced differentiation and ERK1/2 activation were abolished in mice pretreated with a MEK inhibitor (PD98059); however, pretreatment with PD98059 did not reduce the α-tocopherol-mediated increase in HSC/HP cells but instead, further enhanced their proliferation. Therefore, α-tocopherol induces expansion of HSC/HP cells by a nonidentified intracellular pathway and granulocytic/monocytic differentiation through ERK1/2 activation.


Subject(s)
Cell Differentiation/drug effects , Hematopoietic Stem Cells/drug effects , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , alpha-Tocopherol/pharmacology , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/enzymology , Bone Marrow Cells/pathology , Cell Differentiation/physiology , Cell Proliferation/drug effects , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/enzymology , Hyperplasia , Male , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/metabolism
10.
Food Chem Toxicol ; 49(11): 2934-41, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21820028

ABSTRACT

Chlorella vulgaris (CV) was examined for its modulating effects on the reduction induced by lead (Pb) on the numbers of marrow hematopoietic stem cells (HSCs) (c-Kit(+)Lin(-)), granulocyte-macrophage progenitors (Gr1(+)Mac1(+)) and total bone marrow cellularity. In mice gavage-treated daily with 50mg/kg dose of CV for 10 days, concomitant to a continuous offering of 1300 ppm lead acetate in drinking water, the treatment with the algae recovered the significantly reduced numbers of these cell populations to control values. As CV may have a myelostimulating effect through the induction of cytokines, we evaluated its modulating effects on the production of IL-1α, TNF-α, IFN-γ, IL-10 and IL-6. Our results demonstrated that lead significantly impairs the production of IFN-γ, IL-1α and TNF-α and increases the production of IL-10 and IL-6 and that these effects are successfully modulated by the CV treatment. The activity of NK cells, reduced in Pb-exposed animals, was raised to levels higher than those of controls in the exposed group treated with CV. Treatment with the algae also stimulated the production of IFN-γ, IL-1α, TNF-α and NK cells activity in normal mice. In addition, zinc bone concentrations, reduced in lead-exposed mice, were partially, but significantly, reversed by the treatment with CV.


Subject(s)
Bone Marrow Cells/physiology , Chlorella vulgaris , Cytokines/metabolism , Lead/toxicity , Animals , Bone Marrow Cells/drug effects , Cell Proliferation , Cytokines/genetics , Gene Expression Regulation/drug effects , Killer Cells, Natural/drug effects , Killer Cells, Natural/physiology , Male , Mice , Mice, Inbred BALB C , Phytotherapy , Zinc
11.
J Cell Physiol ; 226(7): 1780-92, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21506110

ABSTRACT

Even though the involvement of intracellular Ca(2+) Ca(i)(2+) in hematopoiesis has been previously demonstrated, the relationship between Ca(i)(2+) signaling and cytokine-induced intracellular pathways remains poorly understood. Herein, the molecular mechanisms integrating Ca(2+) signaling with the extracellular signal-regulated kinase 1/2 (ERK1/2) pathway in primary murine and human hematopoietic stem/progenitor cells stimulated by IL-3 and GM-CSF were studied. Our results demonstrated that IL-3 and GM-CSF stimulation induced increased inositol 1,4,5-trisphosphate (IP(3) ) levels and Ca(i)(2+) release in murine and human hematopoietic stem/progenitor cells. In addition, Ca(i)(2+) signaling inhibitors, such as inositol 1,4,5-trisphosphate receptor antagonist (2-APB), PKC inhibitor (GF109203), and CaMKII inhibitor (KN-62), blocked phosphorylation of MEK activated by IL-3 and GM-CSF, suggesting the participation of Ca(2+) -dependent kinases in MEK activation. In addition, we identify phospholipase Cγ2 (PLCγ2) as a PLCγ responsible for the induction of Ca(2+) release by IL-3 and GM-CSF in hematopoietic stem/progenitor cells. Furthermore, the PLCγ inhibitor U73122 significantly reduced the numbers of granulocyte-macrophage colony-forming units after cytokine stimulation. Similar results were obtained in both murine and human hematopoietic stem/progenitor cells. Taken together, these data indicate a role for PLCγ2 and Ca(2+) signaling through the modulation of MEK in both murine and human hematopoietic stem/progenitor cells.


Subject(s)
Calcium Signaling , Extracellular Signal-Regulated MAP Kinases/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Hematopoietic Stem Cells/enzymology , Interleukin-3/metabolism , MAP Kinase Kinase Kinases/metabolism , Phospholipase C gamma/metabolism , Adult , Animals , Biomarkers/metabolism , Calcium Signaling/drug effects , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cell Separation , Cells, Cultured , Enzyme Activation , Enzyme Inhibitors/pharmacology , Flow Cytometry , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/immunology , Humans , Inositol 1,4,5-Trisphosphate/metabolism , Mice , Mice, Inbred C57BL , Middle Aged , Phospholipase C gamma/antagonists & inhibitors , Phosphorylation , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Recombinant Proteins , Time Factors , Young Adult
12.
Chem Biol Interact ; 184(3): 388-95, 2010 Mar 30.
Article in English | MEDLINE | ID: mdl-20096676

ABSTRACT

Angiotensin I-converting enzyme (ACE), a common element of renin-angiotensin system (RAS) and kallikrein-kinin system (KKS), is involved in myelopoiesis modulation, mainly by cleaving the tetrapeptide N-acetyl-seryl-aspartyl-lysyl-proline (AcSDKP). Based on this finding and in our results showing B1 and B2 kinin receptors expression in murine bone marrow (BM) cells, we evaluated the ACE influence on myelopoiesis of kinin B1 receptor knockout mice (B1KO) using long-term bone marrow cultures (LTBMCs). Captopril and AcSDKP were used as controls. Enhanced ACE activity, expressed by non-hematopoietic cells (Ter-199(-) and CD45(-)), was observed in B1KO LTBMCs when compared to wild-type (WT) cells. ACE hyperfunction in B1KO cells was maintained when LTBMCs from B1KO mice were treated with captopril (1.0microM) or AcSDKP (1.0nM). Although no alterations were observed in ACE mRNA and protein levels under these culture conditions, 3.0nM of AcSDKP increased ACE mRNA levels in WT LTBMCs. No alteration in the number of GM-CFC was seen in B1KO mice compared to WT animals, even when the former were treated with AcSDKP (10microg/kg) or captopril (100mg/kg) for 4 consecutive days. Hematological data also revealed no differences between WT and B1KO mice under basal conditions. When the animals received 4 doses of lipopolysaccharide (LPS), a decreased number of blood cells was detected in B1KO mice in relation to WT. We also found a decreased percentage of Gr1(+)/Mac-1(+), Ter119(+), B220(+), CD3(+), and Lin(-)Sca1(+)c-Kit(+) (LSK) cells in the BM of B1KO mice compared to WT animals. Low AcSDKP levels were observed in BM cultures from B1KO in comparison to WT cultures. We conclude that ACE hyperfunction in B1KO mice resulted in faster hydrolysis of AcSDKP peptide, which in turn decreased in BM tissues allowing HSC to enter the S stage of the cell cycle.


Subject(s)
Myelopoiesis/drug effects , Oligopeptides/pharmacology , Peptidyl-Dipeptidase A/metabolism , Receptor, Bradykinin B1/metabolism , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Captopril/pharmacology , Granulocytes/cytology , Granulocytes/drug effects , Kallikrein-Kinin System , Lipopolysaccharides/toxicity , Macrophages/cytology , Macrophages/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Peptidyl-Dipeptidase A/genetics , Receptor, Bradykinin B1/deficiency , Receptor, Bradykinin B1/genetics , Renin-Angiotensin System
13.
Chem Biol Interact ; 177(3): 181-9, 2009 Feb 12.
Article in English | MEDLINE | ID: mdl-19026616

ABSTRACT

Previous studies reported by our group have introduced a new antitumoural drug called Biphosphinic Palladacycle Complex (BPC). In this paper we show that BPC causes apoptosis in leukaemia cells (HL60 and Jurkat), but not in normal human lymphocytes. IC(50) values obtained for both cell lines using the MTT and trypan blue exclusion assays 5h after BPC treatment were lower than 8.0 microM. Using metachromatic fluorophore, acridine orange, we observed that BPC elicited lysosomal rupture of leukaemic cells. Furthermore, BPC triggered caspase-3 and caspase-6 activation and apoptosis in cell lines, inducing chromatin condensation, apoptotic bodies, and DNA fragmentation. Interestingly, the lysosomal cathepsin B inhibitor CA074 markedly decreased BPC-induced caspase-3 and caspase-6 activation as well as cell death. Lysosomal BPC-induced membrane destabilisation was not dependent on reactive oxygen species generation, which was consistent with the absence of cellular HL60 and Jurkat membrane lipid peroxidation. We conclude that, following BPC treatment, lysosomal membrane rupture precedes cell death and the apoptotic signalling pathway is initiated by the release of cathepsin B in the cytoplasm of leukaemia cells. As no toxic effects for human lymphocytes were observed, we suggest that BPC is more selective for transformed cells, mainly due to their exacerbated lysosome expression.


Subject(s)
Antineoplastic Agents/pharmacology , Leukemia/drug therapy , Organometallic Compounds/pharmacology , Apoptosis/drug effects , Caspases/metabolism , Drug Screening Assays, Antitumor , HL-60 Cells , Humans , Jurkat Cells , Leukemia/enzymology , Leukemia/pathology , Lipid Peroxidation , Reactive Oxygen Species/metabolism
14.
Eur J Pharmacol ; 542(1-3): 37-47, 2006 Aug 07.
Article in English | MEDLINE | ID: mdl-16831419

ABSTRACT

The cell death mechanism of cytotoxicity induced by the Biphosphinic Palladacycle Complex (BPC) was studied using a K562 leukaemia cell line. The IC50 values obtained for K562 cells post-72 h of BPC were less than 5.0 microM by using 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyl tetrazolium bromide (MTT) and trypan blue assays. Using the Acridine Orange vital staining combining fluorescence microscopy it was observed that the complex triggers apoptosis in K562 cells, inducing DNA fragmentation, as analysed through electrophoresis. Lysosomal-membrane permeabilization was also observed in K562 cells post-5 h of BPC, which suggests intralysosomal accumulation by proton-trapping, since its pKa value ranged from 5.1 to 6.5. Caspase-3, and -6 activity induced by BPC in K562 cells was prevented by the cathepsin-B inhibitor [N-(L-3-trans-propylcarbamoyl-oxirane-2-carbonyl)-L-isoleucyl-L-proline] (CA074). These events occurred in the presence of endogenous bcl-2 and bax expression. Acute toxicological studies demonstrated that BPC produces no lesions for liver and kidney fourteen-days after drug administration (100 mg/kg--i.p.). White and red blood cells of BPC-treated mice presented normal morphological characteristics. Taken together, these data suggest a novel lysosomal pathway for BPC-induced apoptosis, in which lysosomes are the primary target and cathepsin B acts as death mediator.


Subject(s)
Apoptosis/drug effects , Intracellular Membranes/metabolism , Lysosomes/metabolism , Organometallic Compounds/pharmacology , Animals , Caspase 3/metabolism , Caspase 6/metabolism , Cell Survival/drug effects , Diphosphonates , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Gene Expression/genetics , Humans , Hydrogen-Ion Concentration , K562 Cells , Kidney/drug effects , Kidney/pathology , Leukemia/genetics , Leukemia/metabolism , Leukemia/pathology , Liver/drug effects , Liver/pathology , Mice , Microscopy, Confocal , Organometallic Compounds/chemistry , Organometallic Compounds/toxicity , Organophosphonates/chemistry , Palladium/chemistry , Permeability/drug effects , Proto-Oncogene Proteins c-bcl-2/genetics , Spectrophotometry, Infrared , bcl-2-Associated X Protein/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...