Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters

Affiliation country
Publication year range
1.
Int J Mol Sci ; 24(17)2023 Aug 23.
Article in English | MEDLINE | ID: mdl-37685903

ABSTRACT

Phospholipase C (PLC) enzymes represent crucial participants in the plasma membrane of mammalian cells, including the cardiac sarcolemmal (SL) membrane of cardiomyocytes. They are responsible for the hydrolysis of phosphatidylinositol 4,5-bisphosphate (PtdIns(4,5)P2) into 1,2-diacylglycerol (DAG) and inositol (1,4,5) trisphosphate (Ins(1,4,5)P3), both essential lipid mediators. These second messengers regulate the intracellular calcium (Ca2+) concentration, which activates signal transduction cascades involved in the regulation of cardiomyocyte activity. Of note, emerging evidence suggests that changes in cardiomyocytes' phospholipid profiles are associated with an increased occurrence of cardiovascular diseases, but the underlying mechanisms are still poorly understood. This review aims to provide a comprehensive overview of the significant impact of PLC on the cardiovascular system, encompassing both physiological and pathological conditions. Specifically, it focuses on the relevance of PLCß isoforms as potential cardiac biomarkers, due to their implications for pathological disorders, such as cardiac hypertrophy, diabetic cardiomyopathy, and myocardial ischemia/reperfusion injury. Gaining a deeper understanding of the mechanisms underlying PLCß activation and regulation is crucial for unraveling the complex signaling networks involved in healthy and diseased myocardium. Ultimately, this knowledge holds significant promise for advancing the development of potential therapeutic strategies that can effectively target and address cardiac disorders by focusing on the PLCß subfamily.


Subject(s)
Heart Diseases , Isoenzymes , Animals , Humans , Phospholipase C beta , Myocytes, Cardiac , Biomarkers , Mammals
2.
Cell Mol Life Sci ; 78(6): 2781-2795, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33034697

ABSTRACT

Autosomal-dominant leukodystrophy (ADLD) is a rare fatal neurodegenerative disorder with overexpression of the nuclear lamina component, Lamin B1 due to LMNB1 gene duplication or deletions upstream of the gene. The molecular mechanisms responsible for driving the onset and development of this pathology are not clear yet. Vacuolar demyelination seems to be one of the most significant histopathological observations of ADLD. Considering the role of oligodendrocytes, astrocytes, and leukemia inhibitory factor (LIF)-activated signaling pathways in the myelination processes, this work aims to analyze the specific alterations in different cell populations from patients with LMNB1 duplications and engineered cellular models overexpressing Lamin B1 protein. Our results point out, for the first time, that astrocytes may be pivotal in the evolution of the disease. Indeed, cells from ADLD patients and astrocytes overexpressing LMNB1 show severe ultrastructural nuclear alterations, not present in oligodendrocytes overexpressing LMNB1. Moreover, the accumulation of Lamin B1 in astrocytes induces a reduction in LIF and in LIF-Receptor (LIF-R) levels with a consequential decrease in LIF secretion. Therefore, in both our cellular models, Jak/Stat3 and PI3K/Akt axes, downstream of LIF/LIF-R, are downregulated. Significantly, the administration of exogenous LIF can partially reverse the toxic effects induced by Lamin B1 accumulation with differences between astrocytes and oligodendrocytes, highlighting that LMNB1 overexpression drastically affects astrocytic function reducing their fundamental support to oligodendrocytes in the myelination process. In addition, inflammation has also been investigated, showing an increased activation in ADLD patients' cells.


Subject(s)
Astrocytes/metabolism , Demyelinating Diseases/pathology , Lamin Type B/metabolism , Signal Transduction , Astrocytes/cytology , Cell Nucleus/metabolism , Cell Nucleus/ultrastructure , Cells, Cultured , Demyelinating Diseases/metabolism , Down-Regulation/drug effects , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Hydrogen Peroxide/pharmacology , Inflammation Mediators/metabolism , Lamin Type B/genetics , Leukemia Inhibitory Factor/metabolism , Leukemia Inhibitory Factor/pharmacology , Oligodendroglia/cytology , Oligodendroglia/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Reactive Oxygen Species/metabolism , Receptors, OSM-LIF/metabolism , Up-Regulation/drug effects
3.
FASEB J ; 34(11): 15400-15416, 2020 11.
Article in English | MEDLINE | ID: mdl-32959428

ABSTRACT

MDS are characterized by anemia and transfusion requirements. Transfused patients frequently show iron overload that negatively affects hematopoiesis. Iron chelation therapy can be effective in these MDS cases, but the molecular consequences of this treatment need to be further investigated. That is why we studied the molecular features of iron effect and Deferasirox therapy on PI-PLCbeta1 inositide signaling, using hematopoietic cells and MDS samples. At baseline, MDS patients showing a positive response after iron chelation therapy displayed higher levels of PI-PLCbeta1/Cyclin D3/PKCalpha expression. During treatment, these responder patients, as well as hematopoietic cells treated with FeCl3 and Deferasirox, showed a specific reduction of PI-PLCbeta1/Cyclin D3/PKCalpha expression, indicating that this signaling pathway is targeted by Deferasirox. The treatment was also able to specifically decrease the production of ROS. This effect correlated with a reduction of IL-1A and IL-2, as well as Akt/mTOR phosphorylation. In contrast, cells exposed only to FeCl3 and cells from MDS patients refractory to Deferasirox showed a specific increase of ROS and PI-PLCbeta1/Cyclin D3/PKCalpha expression. All in all, our data show that PI-PLCbeta1 signaling is a target for iron-induced oxidative stress and suggest that baseline PI-PLCbeta1 quantification could predict iron chelation therapy response in MDS.


Subject(s)
Cyclin D3/metabolism , Iron Overload/complications , Iron/adverse effects , Myelodysplastic Syndromes/therapy , Oxidative Stress/drug effects , Phospholipase C beta/metabolism , Protein Kinase C-alpha/metabolism , Aged , Blood Transfusion/statistics & numerical data , Cyclin D3/genetics , Deferasirox/pharmacology , Female , Gene Expression Regulation , Humans , Iron Chelating Agents/pharmacology , Male , Middle Aged , Myelodysplastic Syndromes/pathology , Phospholipase C beta/genetics , Phosphorylation , Protein Kinase C-alpha/genetics , Signal Transduction
4.
Int J Mol Sci ; 22(2)2021 Jan 15.
Article in English | MEDLINE | ID: mdl-33467674

ABSTRACT

Erythropoiesis regulation is essential in normal physiology and pathology, particularly in myelodysplastic syndromes (MDS) and ß-thalassemia. Several signaling transduction processes, including those regulated by inositides, are implicated in erythropoiesis, and the latest MDS or ß-thalassemia preclinical and clinical studies are now based on their regulation. Among others, the main pathways involved are those regulated by transforming growth factor (TGF)-ß, which negatively regulates erythrocyte differentiation and maturation, and erythropoietin (EPO), which acts on the early-stage erythropoiesis. Also small mother against decapentaplegic (SMAD) signaling molecules play a role in pathology, and activin receptor ligand traps are being investigated for future clinical applications. Even inositide-dependent signaling, which is important in the regulation of cell proliferation and differentiation, is specifically associated with erythropoiesis, with phospholipase C (PLC) and phosphatidylinositol 3-kinase (PI3K) as key players that are becoming increasingly important as new promising therapeutic targets. Additionally, Roxadustat, a new erythropoiesis stimulating agent targeting hypoxia inducible factor (HIF), is under clinical development. Here, we review the role and function of the above-mentioned signaling pathways, and we describe the state of the art and new perspectives of erythropoiesis regulation in MDS and ß-thalassemia.


Subject(s)
Erythropoiesis , Myelodysplastic Syndromes/metabolism , Signal Transduction , beta-Thalassemia/metabolism , Animals , Cell Differentiation , Cell Proliferation , Clinical Trials as Topic , Erythropoietin/metabolism , Glycine/analogs & derivatives , Glycine/pharmacology , Hematinics/therapeutic use , Humans , Hypoxia-Inducible Factor 1/metabolism , Isoquinolines/pharmacology , Ligands , Mice , Phosphatidylinositol 3-Kinases/metabolism , Smad Proteins/metabolism , Transforming Growth Factor beta/metabolism , Type C Phospholipases/metabolism
5.
J Cell Physiol ; 235(6): 5413-5428, 2020 06.
Article in English | MEDLINE | ID: mdl-31904116

ABSTRACT

T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive hematological disorder that results from the clonal transformation of T-cell precursors. Phosphatidylinositol 3-kinase (PI3K)/Akt/mechanistic target of rapamycin (mTOR) and canonical Wnt/ß-catenin signaling pathways play a crucial role in T-cell development and in self-renewal of healthy and leukemic stem cells. Notably, ß-catenin is a transcriptional regulator of several genes involved in cancer cell proliferation and survival. In this way, aberrations of components belonging to the aforementioned networks contribute to T-ALL pathogenesis. For this reason, inhibition of both pathways could represent an innovative strategy in this hematological malignancy. Here, we show that combined targeting of Wnt/ß-catenin pathway through ICG-001, a CBP/ß-catenin transcription inhibitor, and of the PI3K/Akt/mTOR axis through ZSTK-474, a PI3K inhibitor, downregulated proliferation, survival, and clonogenic activity of T-ALL cells. ICG-001 and ZSTK-474 displayed cytotoxic effects, and, when combined together, induced a significant increase in apoptotic cells. This induction of apoptosis was associated with the downregulation of Wnt/ß-catenin and PI3K/Akt/mTOR pathways. All these findings were confirmed under hypoxic conditions that mimic the bone marrow niche where leukemic stem cells are believed to reside. Taken together, our findings highlight potentially promising treatment consisting of cotargeting Wnt/ß-catenin and PI3K/Akt/mTOR pathways in T-ALL settings.


Subject(s)
Cell Proliferation/drug effects , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , T-Lymphocytes/drug effects , beta Catenin/genetics , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Humans , Phosphatidylinositol 3-Kinases/genetics , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/genetics , Pyrimidinones/pharmacology , T-Lymphocytes/metabolism , TOR Serine-Threonine Kinases/genetics , Triazines/pharmacology , Wnt Signaling Pathway/drug effects , Wnt Signaling Pathway/genetics , beta Catenin/antagonists & inhibitors
6.
FASEB J ; 32(2): 681-692, 2018 02.
Article in English | MEDLINE | ID: mdl-28970249

ABSTRACT

PI-PLCß1 is involved in cell proliferation, differentiation, and myelodysplastic syndrome (MDS) pathogenesis. Moreover, the increased activity of PI-PLCß1 reduces the expression of PKC-α, which, in turn, delays the cell proliferation and is linked to erythropoiesis. Lenalidomide is currently used in low-risk patients with MDS and del(5q), where it can suppress the del(5q) clone and restore normal erythropoiesis. In this study, we analyzed the effect of lenalidomide on 16 patients with low-risk del(5q) MDS, as well as del(5q) and non-del(5q) hematopoietic cell lines, mainly focusing on erythropoiesis, cell cycle, and PI-PLCß1/PKC-α signaling. Overall, 11 patients were evaluated clinically, and 10 (90%) had favorable responses; the remaining case had a stable disease. At a molecular level, both responder patients and del(5q) cells showed a specific induction of erythropoiesis, with a reduced γ/ß-globin ratio, an increase in glycophorin A, and a nuclear translocation of PKC-α. Moreover, lenalidomide could induce a selective G0/G1 arrest of the cell cycle in del(5q) cells, slowing down the rate proliferation in those cells. Altogether, our results could not only better explain the role of PI-PLCß1/PKC-α signaling in erythropoiesis but also lead to a better comprehension of the lenalidomide effect on del(5q) MDS and pave the way to innovative, targeted therapies.-Poli, A., Ratti, S., Finelli, C., Mongiorgi, S., Clissa, C., Lonetti, A., Cappellini, A., Catozzi, A., Barraco, M., Suh, P.-G., Manzoli, L., McCubrey, J. A., Cocco, L., Follo, M. Y. Nuclear translocation of PKC-α is associated with cell cycle arrest and erythroid differentiation in myelodysplastic syndromes (MDSs).


Subject(s)
Cell Differentiation , Cell Nucleus/enzymology , Erythroid Cells/enzymology , Erythropoiesis , G1 Phase Cell Cycle Checkpoints , Myelodysplastic Syndromes/enzymology , Protein Kinase C-alpha/metabolism , Signal Transduction , Active Transport, Cell Nucleus , Aged , Aged, 80 and over , Cell Line , Cell Nucleus/genetics , Cell Nucleus/pathology , Erythroid Cells/pathology , Female , Humans , Male , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/pathology , Protein Kinase C-alpha/genetics , Resting Phase, Cell Cycle
7.
J Cell Physiol ; 233(3): 1796-1811, 2018 Mar.
Article in English | MEDLINE | ID: mdl-28777460

ABSTRACT

Despite remarkable progress in polychemotherapy protocols, pediatric B-cell acute lymphoblastic leukemia (B-ALL) remains fatal in around 20% of cases. Hence, novel targeted therapies are needed for patients with poor prognosis. Glucocorticoids (GCs) are drugs commonly administrated for B-ALL treatment. Activation of the phosphatidylinositol 3-kinase (PI3K)/Akt/mammalian target of rapamycin signaling pathway is frequently observed in B-ALL and contributes to GC-resistance. Here, we analyzed for the first time to our knowledge, the therapeutic potential of pan and isoform-selective PI3K p110 inhibitors, alone or combined with dexamethasone (DEX), in B-ALL leukemia cell lines and patient samples. We found that a pan PI3K p110 inhibitor displayed the most powerful cytotoxic effects in B-ALL cells, by inducing cell cycle arrest and apoptosis. Both a pan PI3K p110 inhibitor and a dual γ/δ PI3K p110 inhibitor sensitized B-ALL cells to DEX by restoring nuclear translocation of the GC receptor and counteracted stroma-induced DEX-resistance. Finally, gene expression analysis documented that, on one hand the combination consisting of a pan PI3K p110 inhibitor and DEX strengthened the DEX-induced up- or down-regulation of several genes involved in apoptosis, while on the other, it rescued the effects of genes that might be involved in GC-resistance. Overall, our findings strongly suggest that PI3K p110 inhibition could be a promising strategy for treating B-ALL patients by improving GC therapeutic effects and/or overcoming GC-resistance.


Subject(s)
Antineoplastic Agents/pharmacology , Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Dexamethasone/pharmacology , Glucocorticoids/pharmacology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Apoptosis/drug effects , B-Lymphocytes/metabolism , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Child , Child, Preschool , Heterocyclic Compounds, 3-Ring/pharmacology , Humans , Isoquinolines/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Purines/pharmacology , Quinazolinones/pharmacology , Quinoxalines/pharmacology , Thiazolidinediones/pharmacology , Triazines/pharmacology
8.
Biochim Biophys Acta ; 1863(3): 449-463, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26334291

ABSTRACT

The bone marrow (BM) microenvironment regulates the properties of healthy hematopoietic stem cells (HSCs) localized in specific niches. Two distinct microenvironmental niches have been identified in the BM, the "osteoblastic (endosteal)" and "vascular" niches. Nevertheless, these niches provide sanctuaries where subsets of leukemic cells escape chemotherapy-induced death and acquire a drug-resistant phenotype. Moreover, it is emerging that leukemia cells are able to remodel the BM niches into malignant niches which better support neoplastic cell survival and proliferation. This review focuses on the cellular and molecular biology of microenvironment/leukemia interactions in acute lymphoblastic leukemia (ALL) of both B- and T-cell lineage. We shall also highlight the emerging role of exosomes/microvesicles as efficient messengers for cell-to-cell communication in leukemia settings. Studies on the interactions between the BM microenvironment and ALL cells have led to the discovery of potential therapeutic targets which include cytokines/chemokines and their receptors, adhesion molecules, signal transduction pathways, and hypoxia-related proteins. The complex interplays between leukemic cells and BM microenvironment components provide a rationale for innovative, molecularly targeted therapies, designed to improve ALL patient outcome. A better understanding of the contribution of the BM microenvironment to the process of leukemogenesis and leukemia persistence after initial remission, may provide new targets that will allow destruction of leukemia cells without adversely affecting healthy HSCs. This article is part of a Special Issue entitled: Tumor Microenvironment Regulation of Cancer Cell Survival, Metastasis,Inflammation, and Immune Surveillance edited by Peter Ruvolo and Gregg L. Semenza.


Subject(s)
Bone Marrow/pathology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Stem Cell Niche , Tumor Microenvironment , Antineoplastic Agents/therapeutic use , Bone Marrow/drug effects , Bone Marrow/metabolism , Cell Adhesion Molecules/metabolism , Chemokines/metabolism , Humans , Models, Biological , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Signal Transduction/drug effects
9.
Methods Mol Biol ; 2777: 163-176, 2024.
Article in English | MEDLINE | ID: mdl-38478343

ABSTRACT

The hierarchical organization of the leukemic stem cells (LSCs) is identical to that of healthy counterpart cells. It may be split into roughly three stages: a small number of pluripotent stem cells at the top, few lineage-restricted cells in the middle, and several terminally differentiated blood cells at the bottom. Although LSCs can differentiate into the hematopoietic lineage, they can also accumulate as immature progenitor cells, also known as blast cells. Since blast cells are uncommon in healthy bloodstreams, their presence might be a sign of cancer. For instance, a 20% blast cutoff in peripheral blood or bone marrow is formally used to distinguish acute myeloid leukemia from myelodysplastic neoplasms, which is essential to plan the patients' management. Many techniques may be useful for blast enumeration: one of them is flow cytometry, which can perform analyses on many cells by detecting the expression of cell surface markers. Leukemic and non-leukemic blast cells might indeed be characterized by the same surface markers, but these markers are usually differently expressed. Here we propose to use CD45, in combination with CD34 and other cell surface markers, to identify and immunophenotype blast cells in patient-derived samples.


Subject(s)
Leukemia, Myeloid, Acute , Humans , Leukemia, Myeloid, Acute/genetics , Bone Marrow/metabolism , Antigens, CD34/metabolism , Flow Cytometry/methods , Neoplastic Stem Cells/metabolism , Immunophenotyping
10.
Adv Biol Regul ; 91: 101014, 2024 01.
Article in English | MEDLINE | ID: mdl-38242820

ABSTRACT

Myelodysplastic Syndromes, a heterogeneous group of hematological disorders, are characterized by abnormalities in phosphoinositide-dependent signaling, epigenetic regulators, apoptosis, and cytokine interactions within the bone marrow microenvironment, contributing to disease pathogenesis and neoplastic growth. Comprehensive knowledge of these pathways is crucial for the development of innovative therapies that aim to restore normal apoptosis and improve patient outcomes.


Subject(s)
Hematopoietic Stem Cells , Myelodysplastic Syndromes , Humans , Hematopoietic Stem Cells/metabolism , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/metabolism , Bone Marrow/pathology , Cytokines/metabolism , Signal Transduction
11.
Biochim Biophys Acta ; 1803(9): 991-1002, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20399811

ABSTRACT

The phosphatidylinositol 3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) signaling pathway mediates diverse and important physiological cell functions which include proliferation, differentiation, survival, motility, autophagy, and metabolism. However, dysregulated PI3K/Akt/mTOR signaling has been documented in a wide range of neoplasias, including malignant hematological disorders. It is now emerging that this signaling network plays a key role during normal hematopoiesis, a tightly regulated process resulting in the formation of all blood lineages. Blood cell development encompasses a complex series of events which are mainly regulated by actions of cytokines, a family of extracellular ligands which stimulate many biological responses in a wide array of cell types. Hematopoiesis is strictly dependent on the correct function of the bone marrow microenvironment (BMM), as BMM cells secrete most of the cytokines. Several of these cytokines activate the PI3K/Akt/mTOR signaling network and regulate proliferation, survival, and differentiation events during hematopoiesis. Here, we review the evidence that links the signals emanating from the PI3K/Akt/mTOR cascade with the functions of hematopoietic stem cells and the process of myelopoiesis, including lineage commitment. We then highlight the emerging role played by aberrant PI3K/Akt/mTOR signaling during leukemogenesis.


Subject(s)
Leukemia/genetics , Myelopoiesis/genetics , Animals , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/physiology , Leukemia/pathology , Mammals/genetics , Mammals/metabolism , Models, Biological , Myelopoiesis/physiology , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 3-Kinases/physiology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/physiology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-akt/physiology , Signal Transduction/genetics , Signal Transduction/physiology , TOR Serine-Threonine Kinases
12.
Cells ; 10(10)2021 09 28.
Article in English | MEDLINE | ID: mdl-34685544

ABSTRACT

Autosomal dominant leukodystrophy (ADLD) is an extremely rare and fatal neurodegenerative disease due to the overexpression of the nuclear lamina component Lamin B1. Many aspects of the pathology still remain unrevealed. This work highlights the effect of Lamin B1 accumulation on different cellular functions in an ADLD astrocytic in vitro model. Lamin B1 overexpression induces alterations in cell survival signaling pathways with GSK3ß inactivation, but not the upregulation of ß-catenin targets, therefore resulting in a reduction in astrocyte survival. Moreover, Lamin B1 build up affects proliferation and cell cycle progression with an increase of PPARγ and p27 and a decrease of Cyclin D1. These events are also associated to a reduction in cell viability and an induction of apoptosis. Interestingly, ADLD astrocytes trigger a tentative activation of survival pathways that are ineffective. Finally, astrocytes overexpressing Lamin B1 show increased immunoreactivity for both GFAP and vimentin together with NF-kB phosphorylation and c-Fos increase, suggesting astrocytes reactivity and substantial cellular activation. These data demonstrate that Lamin B1 accumulation is correlated to biochemical, metabolic, and morphologic remodeling, probably related to the induction of a reactive astrocytes phenotype that could be strictly associated to ADLD pathological mechanisms.


Subject(s)
Astrocytes/metabolism , Lamin Type B/adverse effects , Neurodegenerative Diseases/physiopathology , Pelizaeus-Merzbacher Disease/physiopathology , Humans
13.
Pharmaceuticals (Basel) ; 14(12)2021 Dec 18.
Article in English | MEDLINE | ID: mdl-34959725

ABSTRACT

The AHCC standardized extract of cultured Lentinula edodes mycelia, and the standardized extract of Asparagus officinalis stem, trademarked as ETAS, are well known supplements with immunomodulatory and anticancer potential. Several reports have described their therapeutic effects, including antioxidant and anticancer activity and improvement of immune response. In this study we aimed at investigating the effects of a combination of AHCC and ETAS on colorectal cancer cells and biopsies from healthy donors to assess the possible use in patients with colorectal cancer. Our results showed that the combination of AHCC and ETAS was synergistic in inducing a significant decrease in cancer cell growth, compared with single agents. Moreover, the combined treatment induced a significant increase in apoptosis, sparing colonocytes from healthy donors, and was able to induce a strong reduction in migration potential, accompanied by a significant modulation of proteins involved in invasiveness. Finally, combined treatment was able to significantly downregulate LGR5 and Notch1 in SW620 cancer stem cell (CSC) colonospheres. Overall, these findings support the potential therapeutic benefits of the AHCC and ETAS combinatorial treatment for patients with colorectal cancer.

14.
Cells ; 9(3)2020 03 12.
Article in English | MEDLINE | ID: mdl-32178280

ABSTRACT

Myelodysplastic syndromes (MDS) are a heterogeneous group of hematological malignancies characterized by peripheral blood cytopenia and abnormal myeloproliferation, as well as a variable risk of evolution into acute myeloid leukemia (AML). The nucleus is a highly organized organelle with several distinct domains where nuclear inositides localize to mediate essential cellular events. Nuclear inositides play a critical role in the modulation of erythropoiesis or myelopoiesis. Here, we briefly review the nuclear structure, the localization of inositides and their metabolic enzymes in subnuclear compartments, and the molecular aspects of nuclear inositides in MDS.


Subject(s)
Cell Nucleus/metabolism , Myelodysplastic Syndromes/immunology , Phosphatidylinositols/metabolism , Humans , Signal Transduction
15.
Cells ; 9(3)2020 03 22.
Article in English | MEDLINE | ID: mdl-32235738

ABSTRACT

A type lamins are fundamental components of the nuclear lamina. Changes in lamin A expression correlate with malignant transformation in several cancers. However, the role of lamin A has not been explored in osteosarcoma (OS). Here, we wanted to investigate the role of lamin A in normal osteoblasts (OBs) and OS cells. Thus, we studied the expression of lamin A/C in OS cells compared to OBs and evaluated the effects of lamin A overexpression in OS cell lines. We show that, while lamin A expression increases during osteoblast differentiation, all examined OS cell lines express lower lamin A levels relative to differentiated OBs. The condition of low LMNA expression confers to OS cells a significant increase in migration potential, while overexpression of lamin A reduces migration ability of OS cells. Moreover, overexpression of unprocessable prelamin A also reduces cell migration. In agreement with the latter finding, OS cells which accumulate the highest prelamin A levels upon inhibition of lamin A maturation by statins, had significantly reduced migration ability. Importantly, OS cells subjected to statin treatment underwent apoptotic cell death in a RAS-independent, lamin A-dependent manner. Our results show that pro-apoptotic effects of statins and statin inhibitory effect on OS cell migration are comparable to those obtained by prelamin A accumulation and further suggest that modulation of lamin A expression and post-translational processing can be a tool to decrease migration potential in OS cells.


Subject(s)
Cell Movement , Lamin Type A/metabolism , Osteosarcoma/metabolism , Osteosarcoma/pathology , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Humans , Lovastatin/pharmacology , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteoblasts/pathology
16.
Cancer Res ; 67(9): 4287-94, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17483341

ABSTRACT

The Akt/mammalian target of rapamycin (mTOR) signaling pathway is important for both cell growth and survival. In particular, an impaired regulation of the Akt/mTOR axis has been strongly implicated in mechanisms related to neoplastic transformation, through enhancement of cell proliferation and survival. Myelodysplastic syndromes (MDS) are a group of heterogeneous hematopoietic stem cell disorders characterized by ineffective hematopoiesis and by a high risk of evolution into acute myelogenous leukemia (AML). The pathogenesis of the MDS evolution into AML is still unclear, although some recent studies indicate that aberrant activation of survival signaling pathways could be involved. In this investigation, done by means of immunofluorescent staining, we report an activation of the Akt/mTOR pathway in high-risk MDS patients. Interestingly, not only mTOR was activated but also its downstream targets, 4E-binding protein 1 and p70 ribosomal S6 kinase. Treatment with the selective mTOR inhibitor, rapamycin, significantly increased apoptotic cell death of CD33(+) (but not CD33(-)) cells from high-risk MDS patients. Rapamycin was ineffective in cells from healthy donors or low-risk MDS. Moreover, incubation of high-risk MDS patient CD34(+) cells with rapamycin decreased the in vitro clonogenic capability of these cells. In contrast, the phosphoinositide 3-kinase inhibitor, LY294002, did not significantly affect the clonogenic activity of high-risk MDS cells. Taken together, our results indicate that the Akt/mTOR pathway is critical for cell survival and proliferation in high-risk MDS patients. Therefore, this signaling network could become an interesting therapeutic target for treating more advanced MDS cases.


Subject(s)
Myelodysplastic Syndromes/metabolism , Myelodysplastic Syndromes/pathology , Protein Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Aged , Antibodies/chemistry , Antibodies/immunology , Antibody Specificity , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Apoptosis/drug effects , Apoptosis/physiology , Cell Cycle Proteins , Cell Growth Processes/physiology , Cell Lineage , Cell Survival/physiology , Female , HL-60 Cells , Humans , Immunohistochemistry , Leukocytes, Mononuclear/enzymology , Leukocytes, Mononuclear/pathology , Male , Middle Aged , Myelodysplastic Syndromes/enzymology , Phosphoproteins/metabolism , Phosphorylation , Protein Kinases/immunology , Proto-Oncogene Proteins c-akt/immunology , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Risk Factors , Sialic Acid Binding Ig-like Lectin 3 , Signal Transduction , Sirolimus/pharmacology , TOR Serine-Threonine Kinases
17.
Mol Pharmacol ; 74(3): 884-95, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18577685

ABSTRACT

Constitutively activated AKT kinase is a common feature of T-cell acute lymphoblastic leukemia (T-ALL). Here, we report that the novel AKT inhibitor (2S)-1-(1H-indol-3-yl)-3-[5-(3-methyl-2H-indazol-5-yl)pyridin-3-yl]oxypropan2-amine (A443654) leads to rapid cell death of T-ALL lines and patient samples. Treatment of CEM, Jurkat, and MOLT-4 cells with nanomolar doses of the inhibitor led to AKT phosphorylation accompanied by dephosphorylation and activation of the downstream target, glycogen synthase kinase-3beta. Effects were time- and dose-dependent, resulting in apoptotic cell death. Treatment of Jurkat cells with A443654 resulted in activation of caspase-2, -3, -6, -8, and -9. Apoptotic cell death was mostly dependent on caspase-2 activation, as demonstrated by preincubation with a selective pharmacological inhibitor. It is remarkable that A443654 was highly effective against the drug-resistant cell line CEM-VBL100, which expresses 170-kDa P-glycoprotein. Moreover, A443654 synergized with the DNA-damaging agent etoposide in both drug-sensitive and drug-resistant cell lines when coadministered [combination index (CI) = 0.39] or when pretreated with etoposide followed by A443654 (CI = 0.689). The efficacy of A443654 was confirmed using blasts from six patients with T-ALL, all of whom displayed low levels of phosphatase and tensin homolog deleted on chromosome 10 (PTEN) and constitutive phosphorylation of Akt on Ser473. At 1 microM, the inhibitor was able to induce apoptotic cell death of T-ALL blast cells, as indicated by flow cytometric analysis of samples immunostained for active (cleaved) caspase-3. Because activated AKT is seen in a large percentage of patients with T-ALL, A443654, either alone or in combination with existing drugs, may be a useful therapy for primary and drug-resistant T-ALL.


Subject(s)
Apoptosis/drug effects , Indazoles/pharmacology , Indoles/pharmacology , Leukemia-Lymphoma, Adult T-Cell/enzymology , Leukemia-Lymphoma, Adult T-Cell/pathology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Caspases/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/drug effects , Drug Screening Assays, Antitumor , Drug Synergism , Enzyme Activation/drug effects , Etoposide/pharmacology , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Phosphorylation/drug effects , Phosphoserine/metabolism , Phosphothreonine/metabolism
18.
PLoS One ; 13(6): e0199312, 2018.
Article in English | MEDLINE | ID: mdl-29958291

ABSTRACT

Radiation therapy is a useful and standard tumor treatment strategy. Despite recent advances in delivery of ionizing radiation, survival rates for some cancer patients are still low because of recurrence and radioresistance. This is why many novel approaches have been explored to improve radiotherapy outcome. Some strategies are focused on enhancement of accuracy in ionizing radiation delivery and on the generation of greater radiation beams, for example with a higher dose rate. In the present study we proposed an in vitro research of the biological effects of very high dose rate beam on SK-Mel28 and A375, two radioresistant human melanoma cell lines. The beam was delivered by a pulsed plasma device, a "Mather type" Plasma Focus for medical applications. We hypothesized that this pulsed X-rays generator is significantly more effective to impair melanoma cells survival compared to conventional X-ray tube. Very high dose rate treatments were able to reduce clonogenic efficiency of SK-Mel28 and A375 more than the X-ray tube and to induce a greater, less easy-to-repair DNA double-strand breaks. Very little is known about biological consequences of such dose rate. Our characterization is preliminary but is the first step toward future clinical considerations.


Subject(s)
Melanoma/radiotherapy , Radiation Tolerance/radiation effects , Radiotherapy/methods , Cell Line, Tumor , Humans , Radiation Dosage , Radiation, Ionizing
19.
Mol Cancer Ther ; 5(6): 1559-70, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16818515

ABSTRACT

Perifosine is an Akt inhibitor displaying strong antineoplastic effects in human tumor cell lines and is currently being tested in phase II clinical trials for treatment of major human cancers. Several recent studies showed the apoptotic effect of perifosine alone or in combination with other anticancer agents. However, this is the first study describing the effects of combining perifosine with the commonly used chemotherapy drug etoposide in cultured human Jurkat T-leukemia cells. Low concentrations of perifosine (5 micromol/L) induced cell death in a synergistic fashion with etoposide if used simultaneously or immediately following exposure to etoposide (posttreatment). The increase in cell death seems to be due to an inactivation of the Akt survival pathway, where treated cells showed a complete dephosphorylation of Akt. Moreover, combined drug-induced Akt deactivation was associated with a parallel decrease in phosphorylation of FoxO1 transcription factor and in expression of antiapoptotic Bcl-xL. Furthermore, the increase in cell death was associated with a specific activation of the caspase-dependent Fas death receptor pathway. These findings might be useful when designing clinical trials where chemotherapy is combined with perifosine for a potential broad use against hematologic malignancies in which the Akt survival pathway is frequently activated.


Subject(s)
Apoptosis/drug effects , Etoposide/pharmacology , Phosphorylcholine/analogs & derivatives , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Signal Transduction , fas Receptor/metabolism , Drug Combinations , Drug Synergism , Enzyme Activation/drug effects , Humans , Jurkat Cells/drug effects , Jurkat Cells/metabolism , Jurkat Cells/pathology , Phosphorylcholine/pharmacology , Proto-Oncogene Proteins c-akt/metabolism
20.
Oncotarget ; 7(2): 1323-40, 2016 Jan 12.
Article in English | MEDLINE | ID: mdl-26593250

ABSTRACT

The proteasome inhibitor bortezomib is a new targeted treatment option for refractory or relapsed acute lymphoblastic leukemia (ALL) patients. However, a limited efficacy of bortezomib alone has been reported. A terminal pro-apoptotic endoplasmic reticulum (ER) stress/unfolded protein response (UPR) is one of the several mechanisms of bortezomib-induced apoptosis. Recently, it has been documented that UPR disruption could be considered a selective anti-leukemia therapy. CX-4945, a potent casein kinase (CK) 2 inhibitor, has been found to induce apoptotic cell death in T-ALL preclinical models, via perturbation of ER/UPR pathway. In this study, we analyzed in T- and B-ALL preclinical settings, the molecular mechanisms of synergistic apoptotic effects observed after bortezomib/CX-4945 combined treatment. We demonstrated that, adding CX-4945 after bortezomib treatment, prevented leukemic cells from engaging a functional UPR in order to buffer the bortezomib-mediated proteotoxic stress in ER lumen. We documented that the combined treatment decreased pro-survival ER chaperon BIP/Grp78 expression, via reduction of chaperoning activity of Hsp90. Bortezomib/CX-4945 treatment inhibited NF-κB signaling in T-ALL cell lines and primary cells from T-ALL patients, but, intriguingly, in B-ALL cells the drug combination activated NF-κB p65 pro-apoptotic functions. In fact in B-cells, the combined treatment induced p65-HDAC1 association with consequent repression of the anti-apoptotic target genes, Bcl-xL and XIAP. Exposure to NEMO (IKKγ)-binding domain inhibitor peptide reduced the cytotoxic effects of bortezomib/CX-4945 treatment. Overall, our findings demonstrated that CK2 inhibition could be useful in combination with bortezomib as a novel therapeutic strategy in both T- and B-ALL.


Subject(s)
Apoptosis/drug effects , Bortezomib/pharmacology , Heat-Shock Proteins/metabolism , Naphthyridines/pharmacology , Transcription Factor RelA/metabolism , Antineoplastic Agents/pharmacology , Blotting, Western , Casein Kinase II/antagonists & inhibitors , Cell Line, Tumor , Cell Survival/drug effects , Drug Synergism , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/drug effects , Humans , Jurkat Cells , Microscopy, Fluorescence , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Phenazines , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/blood , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/blood , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/blood , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Unfolded Protein Response/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL