Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters











Database
Language
Publication year range
1.
Oncoimmunology ; 11(1): 2109861, 2022.
Article in English | MEDLINE | ID: mdl-35979386

ABSTRACT

Tyrosine kinase inhibitors (TKIs) have dramatically improved the survival in chronic myeloid leukemia (CML), but residual disease typically persists even after prolonged treatment. Several lines of evidence suggest that TKIs administered to CML patients upregulate interferon γ (IFNγ) production, which may counteract the anti-tumorigenic effects of the therapy. We now show that activated T cell-conditioned medium (TCM) enhanced proliferation and counteracted imatinib-induced apoptosis of CML cells, and addition of a neutralizing anti-IFNγ antibody at least partially inhibited the anti-apoptotic effect. Likewise, recombinant IFNγ also reduced imatinib-induced apoptosis of CML cells. This anti-apoptotic effect of IFNγ was independent of alternative IFNγ signaling pathways, but could be notably diminished by STAT1-knockdown. Furthermore, IFNγ upregulated the expression of several anti-apoptotic proteins, including MCL1, PARP9, and PARP14, both in untreated and imatinib-treated primary human CD34+ CML stem/progenitor cells. Our results suggest that activated T cells in imatinib-treated CML patients can directly rescue CML cells from imatinib-induced apoptosis at least partially through the secretion of IFNγ, which exerts a rapid, STAT1-dependent anti-apoptotic effect potentially through the simultaneous upregulation of several key hematopoietic survival factors. These mechanisms may have a major clinical impact, when targeting residual leukemic stem/progenitor cells in CML.


Subject(s)
Interferon-gamma , Leukemia, Myelogenous, Chronic, BCR-ABL Positive , Antigens, CD34/metabolism , Antigens, CD34/pharmacology , Apoptosis , Cell Line, Tumor , Humans , Imatinib Mesylate/pharmacology , Imatinib Mesylate/therapeutic use , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Stem Cells/metabolism , Up-Regulation
2.
Cell Death Dis ; 12(10): 875, 2021 09 25.
Article in English | MEDLINE | ID: mdl-34564697

ABSTRACT

Tyrosine kinase inhibitor (TKI) treatment has dramatically improved the survival of chronic myeloid leukemia (CML) patients, but measurable residual disease typically persists. To more effectively eradicate leukemia cells, simultaneous targeting of BCR-ABL1 and additional CML-related survival proteins has been proposed. Notably, several highly specific myeloid cell leukemia 1 (MCL1) inhibitors have recently entered clinical trials for various hematologic malignancies, although not for CML, reflecting the insensitivity of CML cell lines to single MCL1 inhibition. Here, we show that combining TKI (imatinib, nilotinib, dasatinib, or asciminib) treatment with the small-molecule MCL1 inhibitor S63845 exerted strong synergistic antiviability and proapoptotic effects on CML lines and CD34+ stem/progenitor cells isolated from untreated CML patients in chronic phase. Using wild-type BCR-ABL1-harboring CML lines and their T315I-mutated sublines (generated by CRISPR/Cas9-mediated homologous recombination), we prove that the synergistic proapoptotic effect of the drug combination depended on TKI-mediated BCR-ABL1 inhibition, but not on TKI-related off-target mechanisms. Moreover, we demonstrate that colony formation of CML but not normal hematopoietic stem/progenitor cells became markedly reduced upon combination treatment compared to imatinib monotherapy. Our results suggest that dual targeting of MCL1 and BCR-ABL1 activity may efficiently eradicate residual CML cells without affecting normal hematopoietic stem/progenitors.


Subject(s)
Antineoplastic Agents/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Thiophenes/pharmacology , Antigens, CD34/metabolism , Antineoplastic Combined Chemotherapy Protocols , Apoptosis/drug effects , Caspase 3/metabolism , Caspase 7/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Clone Cells , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Humans , Imatinib Mesylate/administration & dosage , Imatinib Mesylate/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Myeloid Cell Leukemia Sequence 1 Protein/antagonists & inhibitors , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Neoplasm Proteins/metabolism , Phosphate-Binding Proteins/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Pore Forming Cytotoxic Proteins/metabolism , Pyroptosis/drug effects , Small Molecule Libraries/pharmacology , bcl-X Protein/metabolism
3.
Oncol Rep ; 40(2): 902-908, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29845257

ABSTRACT

The clinical outcome for patients with chronic myeloid leukemia (CML) has improved significantly with the introduction of tyrosine kinase inhibitors (TKIs). However, their curative potential appears limited, probably as a consequence of TKI-resistant leukemic stem cells (LSCs) that persist as a result of aberrant pathways independent of the well-established oncoprotein Bcr-Abl. One such pathway involves signaling through leukotrienes (LTs), bioactive compounds that have been suggested to play a role in several other malignancies. Cysteinyl LT1 receptor (CysLT1R) has been reported to be overexpressed in a number of solid cancers, and blocking of this receptor with the antagonist montelukast (treatment approved for bronchial asthma) has resulted in the killing of cancer cells. We recently demonstrated that montelukast, alone or in combination with imatinib, can effectively reduce the growth of CML cells, while normal bone marrow cells were left unaffected. Herein, we further investigated the importance of CysLT1R for the survival of CML cells and the mechanisms by which montelukast induces cell death. Knockdown of the CysLT1R of K562 cells with siRNA reduced their growth by 25%. Montelukast had no effect on these cells, while it killed more than 50% of CysLT1R-expressing cells. Growth inhibition exerted by imatinib was unaffected by CysLT1R status. Montelukast-induced killing of K562/JURL-MK1 CML cells was paralleled by Bax overexpression, cytochrome c release, PARP-1 cleavage, and caspase-3 activation, an event further increased in a setting where montelukast was added to imatinib. Wnt/ß-catenin signaling was activated by CysLT1R and we observed that montelukast could induce proteins in this pathway, a finding of relevance for LSC survival. Thus, montelukast, employed at in vivo-like concentrations, induces the killing of CML cells through apoptotic pathways and may provide an additional, novel therapeutic possibility in CML.


Subject(s)
Acetates/pharmacology , Apoptosis/drug effects , Cell Proliferation/drug effects , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukotriene Antagonists/pharmacology , Quinolines/pharmacology , Receptors, Leukotriene/metabolism , Cell Line, Tumor , Cyclopropanes , Drug Resistance, Neoplasm/drug effects , Fusion Proteins, bcr-abl/metabolism , Humans , Imatinib Mesylate/pharmacology , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Sulfides
4.
Biochem Biophys Res Commun ; 490(2): 378-384, 2017 08 19.
Article in English | MEDLINE | ID: mdl-28623130

ABSTRACT

Tyrosine kinase inhibitors targeting the BCR-ABL oncoprotein in chronic myeloid leukemia (CML) are remarkably effective inducing deep molecular remission in most patients. However, they are less effective to eradicate the leukemic stem cells (LSC), resulting in disease persistence. Therefore, there is great need to develop novel therapeutic strategies to specifically target the LSC. In an experimental mouse CML model system, the leukotriene pathway, and specifically, the expression ALOX5, encoding 5-lipoxygenase (5-LO), has been reported as a critical regulator of the LSC. Based on these results, the 5-LO inhibitor zileuton has been introduced in clinical trials as a therapeutic option to target the LSC although its effect on primary human CML LSC has not been studied. We have here by using multiplex single cell PCR analyzed the expression of the mediators of the leukotriene pathway in bone marrow (BM) BCR-ABL+CD34+CD38- cells at diagnosis, and found low or undetectable expression of ALOX5. In line with this, zileuton did not exert significant overall growth inhibition in the long-term culture-initiating cell (LTC-IC) and colony (CFU-C) assays of BM CD34+CD38- cells from 7 CML patients. The majority of the single leukemic BCR-ABL+CD34+CD38- cells expressed cysteinyl leukotriene receptors CYSLT1 and CYSLT2. However, montelukast, an inhibitor of CYSLT1, also failed to significantly suppress CFU-C and LTC-IC growth. These findings indicate that targeting ALOX5 or CYSLT1 signaling with leukotriene antagonists, introduced into the clinical practice primarily as prophylaxis and treatment for asthma, may not be a promising pharmacological strategy to eradicate persisting LSC in CML patients.


Subject(s)
ADP-ribosyl Cyclase 1/analysis , Antigens, CD34/analysis , Arachidonate 5-Lipoxygenase/immunology , Bone Marrow Cells/pathology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Neoplastic Stem Cells/pathology , Receptors, Leukotriene/immunology , ADP-ribosyl Cyclase 1/immunology , Adult , Antigens, CD34/immunology , Bone Marrow Cells/immunology , Cell Proliferation , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology , Neoplastic Stem Cells/immunology , Signal Transduction , Tumor Cells, Cultured
5.
Leuk Lymphoma ; 58(8): 1903-1913, 2017 08.
Article in English | MEDLINE | ID: mdl-27931141

ABSTRACT

Although tyrosine kinase inhibitors (TKIs) have dramatically improved clinical outcome in chronic myeloid leukemia (CML), cure rarely occurs. This may be due to BCR-ABL-independent, aberrant signaling pathways, one of which leads to leukotriene (LT) formation. Well-recognized as inflammatory mediators, LT can also affect oncogenic mechanisms of several tumors. We have previously discovered elevated LT-synthesis and up-regulated cysteinyl-LT-inducing enzyme in CML. Here we report on dose-dependent inhibition of CML cell growth exerted by specific blockers of LT-signaling. Thus, the cysteinyl-LT1-receptor-antagonist montelukast significantly reduced the growth of K562, KCL22, and KU812 cells, as well as primary CD34+ blood cells from two CML patients. Adding montelukast to the TKI imatinib caused combined inhibition. No effect was seen on normal bone marrow cells. Similarly, growth inhibition was also observed with the 5-lipoxygenase (LO)-inhibitor BWA4C, the 5-LO-activating-protein-(FLAP)-inhibitor licofelone and the LTB4(BLT1)-receptor-antagonist LY293111. Thus, blocking of aberrant LT-signaling may provide an additional, novel therapeutic possibility in CML.


Subject(s)
Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukotrienes/metabolism , Signal Transduction , Biosynthetic Pathways/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Leukotriene Antagonists/pharmacology , Protein Kinase Inhibitors/pharmacology , Receptors, Leukotriene/metabolism , Receptors, Leukotriene B4/metabolism , Signal Transduction/drug effects
6.
Pediatr Allergy Immunol ; 18(8): 643-51, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18078418

ABSTRACT

The birth process induces fetal stress. Stress has profound effects on the immune system, also by acting on the trafficking of leukocytes, a process in which adhesion and chemotaxis are primordial and critical events for the development of effective antimicrobial defenses. The newborn is rapidly challenged by a microflora at the epithelia linings and therefore depending on early, innate immunity onset. The objective of the study was to investigate the immune response in cord blood from newborns in relation to different degrees of fetal stress, with focus on neutrophil chemotaxis. We analyzed in vitro transmigration ability of neutrophils and their CD11b expression, measured total white blood count (WBC) and the major leukocyte populations, interleukin (IL)-8, interferon (IFN)-gamma, and soluble E-Selectin, as well as relevant immuno-modulating hormones in infants born at term after Cesarean section prior to the start of labor (n = 55), normal vaginal delivery (n = 87), and assisted delivery (n = 26). Arterial pH and lactate were used as stress markers. We found that spontaneous and IL-8-induced transmigration ability of neutrophils from newborns after normal delivery was significantly higher compared with that of neutrophils from Cesarean section or from adults. With a progressive increase in fetal stress, there were significant elevations in total WBC, in particular neutrophils and monocytes, as well as an enhanced IL-8 and soluble E-Selectin level. Assisted delivery, associated with the highest degree of fetal stress in addition had an enhanced lymphocyte and monocytes count as well as an increased IFN-gamma level. There were significant direct correlations between neutrophils and monocytes, respectively, with cortisol, beta-endorphin, and prolactin. Interferon-gamma was directly related to dopamine, as well as to the lymphocyte and monocyte count. The setting of the HPA-axis at birth is a promoter of an alarm response and a surge of neuroendocrine immuno-modulating factors that enhances antimicrobial defenses of the newborn. We speculate that IL-8 induced by normal labor may be a priming factor for an increased neutrophil chemotaxis through the pre-activated endothelium of the fetus. Assisted delivery may trigger excessive recruitment of additional inflammatory cells and IFN-gamma release.


Subject(s)
Chemotaxis, Leukocyte , Fetal Blood/immunology , Immunity, Innate , Interleukin-8/immunology , Neutrophils/physiology , Parturition , Adult , CD11b Antigen/analysis , Cesarean Section , Delivery, Obstetric , Female , Humans , Infant, Newborn , Leukocyte Count , Male , Pregnancy , Stress, Physiological/immunology , beta-Endorphin/analysis
7.
Pediatr Dermatol ; 24(5): 474-82, 2007.
Article in English | MEDLINE | ID: mdl-17958791

ABSTRACT

At birth, commensal microbes penetrate into the skin of the human newborn, eliciting an acute rash, erythema toxicumn neonatorum. Histologically, the rash is characterized by an upregulation of proinflammatory activity and a local recruitment of immunocytes, including macrophages. High mobility group box chromosomal protein 1, a nuclear and cytosolic protein, is also a pro-inflammatory cytokine released by macrophages in response to microbial stimulation. Here, we reasoned that macrophages but also keratinocytes might upregulate this protein in response to the first colonization and that high mobility group box chromosomal protein 1 might play a role as a proinflammatory mediator in the development and progression of erythema toxicum. Punch biopsy specimens from 1-day-old healthy infants, seven with and four without erythema toxicum were analyzed with indirect immunohistochemistry and two different antihigh mobility group box chromosomal protein 1 antibodies, immunofluorescence, nuclear counterstaining, confocal and immunoelectron imaging. We found relocation of nuclear high mobility group box chromosomal protein 1 into the cytoplasm in keratinocytes and macrophages in erythema toxicum. Cytoplasmatic high mobility group box chromosomal protein 1 was also found in melanocytes and did neither co-locate with lysosomal-associated membrane proteins nor with melanosomes. We speculate that terrestrial adaptation triggers the induction of the endogenous "danger signal" high mobility group box chromosomal protein 1 in the skin of the newborn infant, perhaps in response to the first commensal colonization and that this signal may contribute to alert the immune system and promote a protective immune response.


Subject(s)
Erythema/pathology , HMGB1 Protein/metabolism , Infant, Newborn, Diseases/pathology , Keratinocytes/metabolism , Skin/pathology , Bacteria/immunology , Biopsy , Erythema/immunology , Erythema/microbiology , Fluorescent Antibody Technique , Humans , Infant, Newborn , Infant, Newborn, Diseases/immunology , Infant, Newborn, Diseases/microbiology , Keratinocytes/pathology , Keratinocytes/ultrastructure , Lysosomal-Associated Membrane Protein 2 , Lysosomal Membrane Proteins/metabolism , Macrophages/metabolism , Macrophages/pathology , Macrophages/ultrastructure , Membrane Proteins/metabolism , Microscopy, Confocal , Microscopy, Immunoelectron , Skin/immunology , Skin/microbiology , Vesicular Transport Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL