Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Commun Biol ; 7(1): 353, 2024 Mar 22.
Article in English | MEDLINE | ID: mdl-38519773

ABSTRACT

Cisplatin-based chemotherapy has associated clinical disadvantages, such as high toxicity and resistance. Thus, the development of new antitumor metallodrugs able to overcome different clinical barriers is a public healthcare priority. Here, we studied the mechanism of action of the isomers trans and cis-[PtI2(isopropylamine)2] (I5 and I6, respectively) against gastrointestinal cancer cells. We demonstrate that I5 and I6 modulate mitochondrial metabolism, decreasing OXPHOS activity and negatively affecting ATP-linked oxygen consumption rate. Consequently, I5 and I6 generated Reactive Oxygen Species (ROS), provoking oxidative damage and eventually the induction of senescence. Thus, herein we propose a loop with three interconnected processes modulated by these iodido agents: (i) mitochondrial dysfunction and metabolic disruptions; (ii) ROS generation and oxidative damage; and (iii) cellular senescence. Functionally, I5 reduces cancer cell clonogenicity and tumor growth in a pancreatic xenograft model without systemic toxicity, highlighting a potential anticancer complex that warrants additional pre-clinical studies.


Subject(s)
Gastrointestinal Neoplasms , Platinum , Humans , Reactive Oxygen Species/metabolism , Cisplatin/pharmacology , Mitochondria/metabolism , Gastrointestinal Neoplasms/metabolism
2.
Arterioscler Thromb Vasc Biol ; 37(2): 237-246, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27856455

ABSTRACT

OBJECTIVE: Map3k8 (Cot/Tpl2) activates the MKK1/2-ERK1/2, MAPK pathway downstream from interleukin-1R, tumor necrosis factor-αR, NOD-2R (nucleotide-binding oligomerization domain-like 2R), adiponectinR, and Toll-like receptors. Map3k8 plays a key role in innate and adaptive immunity and influences inflammatory processes by modulating the functions of different cell types. However, its role in atherogenesis remains unknown. In this study, we analyzed the role of this kinase in this pathology. APPROACH AND RESULTS: We show here that Map3k8 deficiency results in smaller numbers of Ly6ChighCD11clow and Ly6ClowCD11chigh monocytes in ApoE-/- mice fed a high-fat diet (HFD). Map3k8-/-ApoE-/- monocytes displayed high rates of apoptosis and reduced amounts of Nr4a1, a transcription factor known to modulate apoptosis in Ly6ClowCD11chigh monocytes. Map3k8-/-ApoE-/- splenocytes and macrophages showed irregular patterns of cytokine and chemokine expression. Map3k8 deficiency altered cell adhesion and migration in vivo and decreased CCR2 expression, a determinant chemokine receptor for monocyte mobilization, on circulating Ly6ChighCD11clow monocytes. Map3k8-/-ApoE-/- mice fed an HFD showed decreased cellular infiltration in the atherosclerotic plaque, with low lipid content. Lesions had similar size after Map3k8+/+ApoE-/- bone marrow transplant into Map3k8-/-ApoE-/- and Map3k8+/+ApoE-/- mice fed an HFD, whereas smaller plaques were observed after the transplantation of bone marrow lacking both ApoE and Map3k8. CONCLUSIONS: Map3k8 decreases apoptosis of monocytes and enhances CCR2 expression on Ly6ChighCD11clow monocytes of ApoE-/- mice fed an HFD. These findings explain the smaller aortic lesions in ApoE-/- mice with Map3k8-/-ApoE-/- bone marrow cells fed an HFD, supporting further studies of Map3k8 as an antiatherosclerotic target.


Subject(s)
Aorta/metabolism , Aortic Diseases/metabolism , Apolipoproteins E/deficiency , Atherosclerosis/metabolism , MAP Kinase Kinase Kinases/metabolism , Monocytes/metabolism , Plaque, Atherosclerotic , Proto-Oncogene Proteins/metabolism , Animals , Antigens, Ly/metabolism , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/pathology , Aortic Diseases/prevention & control , Apolipoproteins E/genetics , Apoptosis , Atherosclerosis/genetics , Atherosclerosis/pathology , Atherosclerosis/prevention & control , CD11c Antigen/metabolism , Cell Adhesion , Chemotaxis, Leukocyte , Cytokines/metabolism , Diet, High-Fat , Disease Models, Animal , Genetic Predisposition to Disease , MAP Kinase Kinase Kinases/deficiency , MAP Kinase Kinase Kinases/genetics , Macrophages, Peritoneal/metabolism , Male , Mice, Knockout , Monocytes/pathology , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Phenotype , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , Receptors, CCR2/metabolism , Signal Transduction , Spleen/metabolism
3.
Mol Cell Biol ; 36(6): 900-12, 2015 Dec 28.
Article in English | MEDLINE | ID: mdl-26711264

ABSTRACT

Polycomb chromatin modifiers regulate hematopoietic pluripotent stem and progenitor cell self-renewal and expansion. Polycomb complex redundancy and biochemical heterogeneity complicate the unraveling of the functional contributions of distinct components. We have studied the hematopoietic activity of RYBP, a direct interactor and proposed modulator of RING1A/RING1B-dependent histone H2A monoubiquitylation (H2AUb). Using a mouse model to conditionally inactivate Rybp in adult hematopoiesis, we have found that RYBP deletion results in a reversion of B-1-to-B-2 B-cell progenitor ratios, i.e., of the innate (predominantly fetal) to acquired (mostly adult) immunity precursors. Increased numbers of B-1 progenitors correlated with a loss of pre-proB cells, the B-2 progenitors. RYBP-deficient stem and progenitor cell populations (LKS) and isolated common lymphoid progenitors (CLP) gave rise to increased numbers of B-1 progenitors in vitro. Rybp inactivation, however, did not result in changes of global H2AUb and did not interact genetically with Ring1A or Ring1B deletions. These results show that a sustained regulation of the B-1-to-B-2 switch is needed throughout adult life and that RYBP plays an important role in keeping B-2 dominance, most likely independently of its Polycomb affiliation.


Subject(s)
B-Lymphocytes/cytology , Hematopoiesis , Hematopoietic Stem Cells/cytology , Repressor Proteins/metabolism , Animals , B-Lymphocytes/metabolism , Cell Line , Cell Lineage , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Gene Deletion , Hematopoietic Stem Cells/metabolism , Mice , Polycomb Repressive Complex 1/genetics , Polycomb Repressive Complex 1/metabolism , Repressor Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
4.
J Cell Sci ; 128(19): 3660-71, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26272920

ABSTRACT

The functions of polycomb products extend beyond their well-known activity as transcriptional regulators to include genome duplication processes. Polycomb activities during DNA replication and DNA damage repair are unclear, particularly without induced replicative stress. We have used a cellular model of conditionally inactive polycomb E3 ligases (RING1A and RING1B), which monoubiquitylate lysine 119 of histone H2A (H2AK119Ub), to examine DNA replication in unperturbed cells. We identify slow elongation and fork stalling during DNA replication that is associated with the accumulation of mid and late S-phase cells. Signs of replicative stress and colocalisation of double-strand breaks with chromocenters, the sites of coalesced pericentromeric heterocromatic (PCH) domains, were enriched in cells at mid S-phase, the stage at which PCH is replicated. Altered replication was rescued by targeted monoubiquitylation of PCH through methyl-CpG binding domain protein 1. The acute senescence associated with the depletion of RING1 proteins, which is mediated by p21 (also known as CDKN1A) upregulation, could be uncoupled from a response to DNA damage. These findings link cell proliferation and the polycomb proteins RING1A and RING1B to S-phase progression through a specific function in PCH replication.


Subject(s)
Histones/metabolism , Polycomb Repressive Complex 1/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Apoptosis/genetics , Apoptosis/physiology , Cell Differentiation/genetics , Cell Differentiation/physiology , Cells, Cultured , Centromere/metabolism , Mice , Polycomb Repressive Complex 1/genetics , S Phase/physiology , Ubiquitin-Protein Ligases/genetics
5.
Carcinogenesis ; 36(5): 585-97, 2015 May.
Article in English | MEDLINE | ID: mdl-25784375

ABSTRACT

Snail2 is a zinc finger transcription factor involved in driving epithelial to mesenchymal transitions. Snail2 null mice are viable, but display defects in melanogenesis, gametogenesis and hematopoiesis, and are markedly radiosensitive. Here, using mouse genetics, we have studied the contributions of Snail2 to epidermal homeostasis and skin carcinogenesis. Snail2 (-/-) mice presented a defective epidermal terminal differentiation and, unexpectedly, an increase in number, size and malignancy of tumor lesions when subjected to the two-stage mouse skin chemical carcinogenesis protocol, compared with controls. Additionally, tumor lesions from Snail2 (-/-) mice presented a high inflammatory component with an elevated percentage of myeloid precursors in tumor lesions that was further increased in the presence of the anti-inflammatory agent dexamethasone. In vitro studies in Snail2 null keratinocytes showed that loss of Snail2 leads to a decrease in proliferation indicating a non-cell autonomous role for Snail2 in the skin carcinogenic response observed in vivo. Bone marrow (BM) cross-reconstitution assays between Snail2 wild-type and null mice showed that Snail2 absence in the hematopoietic system fully reproduces the tumor behavior of the Snail2 null mice and triggers the accumulation of myeloid precursors in the BM, blood and tumor lesions. These results indicate a new role for Snail2 in preventing myeloid precursors recruitment impairing skin chemical carcinogenesis progression.


Subject(s)
Inflammation/pathology , Keratinocytes/pathology , Myeloid Progenitor Cells/pathology , Neoplasms, Experimental/pathology , Skin Neoplasms/pathology , Transcription Factors/physiology , 9,10-Dimethyl-1,2-benzanthracene/toxicity , Animals , Apoptosis , Blotting, Western , Carcinogens/toxicity , Cell Differentiation , Cell Proliferation , Cells, Cultured , Fluorescent Antibody Technique , Hematopoiesis , Immunoenzyme Techniques , Inflammation/chemically induced , Inflammation/metabolism , Keratinocytes/drug effects , Keratinocytes/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Progenitor Cells/drug effects , Myeloid Progenitor Cells/metabolism , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Skin Neoplasms/chemically induced , Skin Neoplasms/metabolism , Snail Family Transcription Factors
6.
Cell Cycle ; 11(16): 3055-66, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22871742

ABSTRACT

GATA1 is a hematopoietic transcription factor essential for expression of most genes encoding erythro-megakaryocytic proteins, i.e., globins and platelet glycoproteins. A role for GATA1 as a cell proliferation regulator has been proposed, as some of its bona fide targets comprise global regulators, such as c-KIT or c-MYC, or cell cycle factors, i.e., CYCLIN D or p21CIP1. In this study, we describe that GATA1 directly regulates the expression of replication licensing factor CDC6. Using reporter transactivation, electrophoretic mobility shift and chromatin immunoprecipitation assays, we show that GATA1 stimulates CDC6 transcription by binding to a canonical binding site located within a 166bp enhancer region upstream CDC6 promoter. This evolutionary conserved GATA binding site conforms to recently described chromatin occupancy rules, i.e., preferred bases within core WGATAR (TGATAA), 5' and 3' flanking bases (GGTGATAAGG) and distance to the transcription initiation site. We also found adjacent conserved binding sites for ubiquitously expressed transcription factor CP2, needed for GATA activity on CDC6 enhancer. Our results add to the growing evidence for GATA1 acting as a direct transcriptional regulator of the cell cycle machinery, thus linking cell proliferation control and specific gene expression programs during lineage differentiation.


Subject(s)
Cell Cycle Proteins/metabolism , GATA1 Transcription Factor/metabolism , Gene Expression Regulation , Nuclear Proteins/metabolism , Animals , Base Sequence , Binding Sites , Cell Cycle Proteins/genetics , Cell Differentiation , Cell Lineage , Cell Proliferation , Chromatin Immunoprecipitation , Conserved Sequence , Electrophoretic Mobility Shift Assay , Enhancer Elements, Genetic , GATA1 Transcription Factor/genetics , GATA1 Transcription Factor/pharmacology , Genes, Reporter , HEK293 Cells , Humans , Megakaryocytes/cytology , Megakaryocytes/metabolism , Mice , Molecular Sequence Data , NIH 3T3 Cells , Nuclear Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Retroviridae/genetics , Retroviridae/metabolism , Transcription Initiation Site , Transcription, Genetic , Transcriptional Activation , Transfection
7.
Mol Cell Biol ; 28(3): 1018-28, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18039844

ABSTRACT

Polycomb group (PcG) proteins act as positive regulators of cell proliferation. Ring1B is a PcG gene essential for embryonic development, but its contribution to cell turnover in regenerating tissues in not known. Here, we have generated a conditional mouse mutant line to study the Ring1B role in adult hematopoiesis. Mutant mice developed a hypocellular bone marrow that paradoxically contained an enlarged, hyperproliferating compartment of immature cells, with an intact differentiation potential. These alterations were associated with differential upregulation of cyclin D2, which occurred in all mutant bone marrow cells, and of p16(Ink4a), observed only in the differentiated compartment. Concurrent inactivation of Ink4a rescued the defective proliferation of maturing cells but did not affect the hyperproliferative activity of progenitors and resulted in a shortening of the onset of lymphomas induced by Ink4a inactivation. These data show that Ring1B restricts the progenitors' proliferation and promotes the proliferation of their maturing progeny by selectively altering the expression pattern of cell cycle regulators along hematopoietic differentiation. The novel antiproliferative role of Ring1B's downregulation of a cell cycle activator may play an important role in the tight control of hematopoietic cell turnover.


Subject(s)
Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p16/deficiency , DNA-Binding Proteins/physiology , Hematopoietic Stem Cells/cytology , Lymphoma/etiology , Repressor Proteins/physiology , Animals , Cell Cycle Proteins/genetics , Cell Differentiation , Gene Expression Regulation , Mice , Mice, Knockout , Polycomb Repressive Complex 1 , Polycomb-Group Proteins , Ubiquitin-Protein Ligases
8.
Br J Haematol ; 130(6): 902-11, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16156860

ABSTRACT

The 3q21q26 syndrome leukaemias are characterised by dystrophic megakaryocytes, elevated platelet counts, ectopic EVI1 protein production and poor prognosis. To investigate the molecular basis of this disease, we developed a model system to examine the biological activity of EVI1 in a megakaryocyte progenitor cell line. For this purpose, Evi1 was conditionally expressed in human erythroleukaemia cells (HEL) that progress along the megakaryocyte lineage in the presence of 12-O-tetradecanoylphorbol 13-acetate (TPA). TPA-stimulated HEL cells normally undergo: (1) growth arrest; (2) altered morphology; (3) endomitosis and (4) characteristic changes in gene expression, including reduction of the erythroid-specific glycophoryn A and elevation of the specific glycoproteins GPIIIa and GPVI. Enforced Evi1 expression alone had no effect upon HEL cell proliferation or differentiation but a phenotype was manifest upon stimulation to differentiate. Evi1-expressing, TPA-treated HEL cells still showed growth arrest, had reduced and enhanced glycophoryn A and GPIIIa mRNA's, respectively, but failed to significantly elevate GPVI mRNA. This was accompanied by inhibition of endomitosis and altered cell morphology. Sustained CDK2 catalytic activity, typically associated with megakaryocyte endomitosis, was dramatically decreased in TPA-stimulated Evi1-expressing HEL cells because of significantly reduced levels of cyclin A. Therefore, enforced Evi1 expression could inhibit megakaryocyte differentiation although retention of some characteristic molecular changes, in combination with a block in endomitosis and altered morphology, suggest a defect in lineage progression. These results suggest that ectopic Evi1 expression contributes to a defective megakaryocyte differentiation programme and is likely to contribute to the phenotype observed in 3q21q26 syndrome leukaemias.


Subject(s)
CDC2-CDC28 Kinases/antagonists & inhibitors , DNA-Binding Proteins/physiology , Leukemia, Erythroblastic, Acute/pathology , Megakaryocytes/cytology , Proto-Oncogenes/physiology , Transcription Factors/physiology , CDC2-CDC28 Kinases/physiology , Cell Cycle , Cell Differentiation , Cell Proliferation , Cyclin-Dependent Kinase 2 , DNA-Binding Proteins/metabolism , Hematopoiesis , Humans , Leukemia, Erythroblastic, Acute/enzymology , Leukemia, Erythroblastic, Acute/metabolism , MDS1 and EVI1 Complex Locus Protein , Megakaryocytes/enzymology , Mitosis , Neoplasm Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods , Transcription Factors/metabolism , Tumor Cells, Cultured
9.
Nucleic Acids Res ; 32(21): 6454-67, 2004.
Article in English | MEDLINE | ID: mdl-15590906

ABSTRACT

Cdc6 is a key regulator of the strict alternation of S and M phases during the mitotic cell cycle. In mammalian and plant cells that physiologically become polyploid, cdc6 is transcriptionally and post-translationally regulated. We have recently reported that Cdc6 levels are maintained in megakaryoblastic HEL cells, but severely downregulated by ectopic expression of transcriptional repressor Drosophila melanogaster escargot. Here, we show that cdc6 promoter activity is upregulated during megakaryocytic differentiation of HEL endoreplicating cells, and that Escargot interferes with such activation. Transactivation experiments showed that a 1.7 kb region located at 2800 upstream cdc6 transcription initiation site behaved as a potent enhancer in endoreplicating cells only. This activity was mainly dependent on a novel cis-regulatory element composed by an E2 box overlapping a GATA motif. Ectopic Escargot could bind this regulatory element in vitro and endogenous GATA-1 and E2A formed specific complexes in megakaryoblastic cells as well as in primary megakaryocytes. Chromatin Immunoprecipitation analysis revealed that both transcription factors were occupying the E2 box/GATA site in vivo. Altogether, these data suggest that cdc6 expression could be actively maintained during megakaryocytic differentiation through transcriptional mechanisms involving specific cis- and trans-regulatory elements.


Subject(s)
Cell Cycle Proteins/genetics , E-Box Elements , Gene Expression Regulation , Megakaryocytes/metabolism , Nuclear Proteins/genetics , Response Elements , 5' Flanking Region , Animals , Basic Helix-Loop-Helix Transcription Factors , Binding Sites , Cell Cycle Proteins/biosynthesis , Cell Line , DNA Replication , DNA-Binding Proteins/metabolism , Down-Regulation , Drosophila Proteins/metabolism , Erythroid-Specific DNA-Binding Factors , GATA1 Transcription Factor , Humans , Mice , Nuclear Proteins/biosynthesis , Polyploidy , RNA, Messenger/metabolism , Rats , Transcription Factors/metabolism , Transcription, Genetic , Transcriptional Activation
10.
Mol Biol Cell ; 13(11): 3989-4000, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12429841

ABSTRACT

Endomitosis is the process by which mammalian megakaryocytes become polyploid during terminal differentiation. As in other endoreplicating cells, cyclin-cdk complexes are distinctly regulated, probably to overcome the strict mechanisms that prevent rereplication in most somatic cells. We have asked whether key factors involved in the assembly and licensing of replication origins are equally regulated during endomitosis. Cdc6, cdt1, and geminin expression was analyzed during differentiation of two human megakaryoblastic cell lines, HEL and K562, which respectively do and do not establish endoreplication cycles. Geminin was downregulated, whereas cdt1 levels were maintained upon differentiation of both cell lines, independently of whether cells entered extra S-phases. In contrast, cdc6 was present and remained nuclear only in differentiated endoreplicating cells. Interestingly, cdc6 protein expression was reestablished in K562 cells that underwent endomitosis after transient or stable cyclin E overexpression. The high levels of cyclin E reached in these cells appeared to influence the stabilization of cdc6 protein rather than its RNA transcription rate. Finally, cdc6 overexpression drove HEL cells into endoreplication cycles in the absence of differentiation stimuli. Our results show that both cdt1 and cdc6 are differentially regulated during megakaryocytic differentiation and suggest an active role of cdc6 in endomitosis.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Cycle/physiology , Nuclear Proteins/metabolism , Polyploidy , Cell Cycle Proteins/genetics , Cell Differentiation/physiology , Cell Nucleus/metabolism , Cyclin A/metabolism , Cyclin E/metabolism , DNA Replication , Down-Regulation/physiology , Geminin , Humans , Megakaryocytes/cytology , Megakaryocytes/metabolism , Megakaryocytes/physiology , Nuclear Proteins/genetics , Receptor, Nerve Growth Factor/metabolism , Tetradecanoylphorbol Acetate/metabolism , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...