Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Cell Mol Life Sci ; 80(11): 316, 2023 Oct 06.
Article in English | MEDLINE | ID: mdl-37801090

ABSTRACT

Nuclear deformability plays a critical role in cell migration. During this process, the remodeling of internal components of the nucleus has a direct impact on DNA damage and cell behavior; however, how persistent migration promotes nuclear changes leading to phenotypical and functional consequences remains poorly understood. Here, we described that the persistent migration through physical barriers was sufficient to promote permanent modifications in migratory-altered cells. We found that derived cells from confined migration showed changes in lamin B1 localization, cell morphology and transcription. Further analysis confirmed that migratory-altered cells showed functional differences in DNA repair, cell response to chemotherapy and cell migration in vivo homing experiments. Experimental modulation of actin polymerization affected the redistribution of lamin B1, and the basal levels of DNA damage in migratory-altered cells. Finally, since major nuclear changes were present in migratory-altered cells, we applied a multidisciplinary biochemical and biophysical approach to identify that confined conditions promoted a different biomechanical response of the nucleus in migratory-altered cells. Our observations suggest that mechanical compression during persistent cell migration has a role in stable nuclear and genomic alterations that might handle the genetic instability and cellular heterogeneity in aging diseases and cancer.


Subject(s)
Leukemia , Neoplasms , Humans , Stress, Mechanical , Cell Movement , DNA Repair , Leukemia/genetics , Cell Nucleus/physiology
2.
Eur J Cell Biol ; 102(3): 151343, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37494871

ABSTRACT

Acute lymphoblastic leukemia (ALL) is the most common pediatric cancer, and the infiltration of leukemic cells is critical for disease progression and relapse. Nuclear deformability plays a critical role in cancer cell invasion through confined spaces; however, the direct impact of epigenetic changes on the nuclear deformability of leukemic cells remains unclear. Here, we characterized how 3D collagen matrix conditions induced H3K4 methylation in ALL cell lines and clinical samples. We used specific shRNA and chemical inhibitors to target WDR5 (a core subunit involved in H3K4 methylation) and determined that targeting WDR5 reduced the H3K4 methylation induced by the 3D environment and the invasiveness of ALL cells in vitro and in vivo. Intriguingly, targeting WDR5 did not reduce the adhesion or the chemotactic response of leukemia cells, suggesting a different mechanism by which H3K4 methylation might govern ALL cell invasiveness. Finally, we conducted biochemical, and biophysical experiments to determine that 3D environments promoted the alteration of the chromatin, the morphology, and the mechanical behavior of the nucleus in ALL cells. Collectively, our data suggest that 3D environments control an upregulation of H3K4 methylation in ALL cells, and targeting WDR5 might serve as a promising therapeutic target against ALL invasiveness in vivo.


Subject(s)
Histones , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Child , Humans , Histones/metabolism , Methylation , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Epigenesis, Genetic , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism
3.
Oncogene ; 41(9): 1324-1336, 2022 02.
Article in English | MEDLINE | ID: mdl-34999734

ABSTRACT

T-acute lymphoblastic leukemia (T-ALL) is an aggressive hematological malignancy that comprises the accumulation of malignant T-cells. Despite current therapies, failure to conventional treatments and relapse are frequent in children with T-ALL. It is known that the chemokine CXCL12 modulates leukemia survival and dissemination; however, our understanding of molecular mechanisms used by T-ALL cells to infiltrate and respond to leukemia cells-microenvironment interactions is still vague. In the present study, we showed that CXCL12 promoted H3K9 methylation in cell lines and primary T-ALL cells within minutes. We thus identified that CXCL12-mediated H3K9 methylation affected the global chromatin configuration and the nuclear mechanics of T-ALL cells. Importantly, we characterized changes in the genomic profile of T-ALL cells associated with rapid CXCL12 stimulation. We showed that blocking CXCR4 and protein kinase C (PKC) impaired the H3K9 methylation induced by CXCL12 in T-ALL cells. Finally, blocking H3K9 methyltransferases reduced the efficiency of T-ALL cells to deform their nuclei, migrate across confined spaces, and home to spleen and bone marrow in vivo models. Together, our data show novel functions for CXL12 as a master regulator of nuclear deformability and epigenetic changes in T-ALL cells, and its potential as a promising pharmacological target against T-ALL dissemination.


Subject(s)
Chemokine CXCL12
4.
Front Cell Dev Biol ; 8: 595849, 2020.
Article in English | MEDLINE | ID: mdl-33381504

ABSTRACT

Phosphoinositides, which are membrane-bound phospholipids, are critical signaling molecules located at the interface between the extracellular matrix, cell membrane, and cytoskeleton. Phosphoinositides are essential regulators of many biological and cellular processes, including but not limited to cell migration, proliferation, survival, and differentiation, as well as cytoskeletal rearrangements and actin dynamics. Over the years, a multitude of studies have uniquely implicated phosphoinositide signaling as being crucial in cardiovascular biology and a dominant force in the development of cardiovascular disease and its progression. Independently, the cellular transduction of mechanical forces or mechanotransduction in cardiovascular cells is widely accepted to be critical to their homeostasis and can drive aberrant cellular phenotypes and resultant cardiovascular disease. Given the versatility and diversity of phosphoinositide signaling in the cardiovascular system and the dominant regulation of cardiovascular cell functions by mechanotransduction, the molecular mechanistic overlap and extent to which these two major signaling modalities converge in cardiovascular cells remain unclear. In this review, we discuss and synthesize recent findings that rightfully connect phosphoinositide signaling to cellular mechanotransduction in the context of cardiovascular biology and disease, and we specifically focus on phosphatidylinositol-4,5-phosphate, phosphatidylinositol-4-phosphate 5-kinase, phosphatidylinositol-3,4,5-phosphate, and phosphatidylinositol 3-kinase. Throughout the review, we discuss how specific phosphoinositide subspecies have been shown to mediate biomechanically sensitive cytoskeletal remodeling in cardiovascular cells. Additionally, we discuss the direct interaction of phosphoinositides with mechanically sensitive membrane-bound ion channels in response to mechanical stimuli. Furthermore, we explore the role of phosphoinositide subspecies in association with critical downstream effectors of mechanical signaling in cardiovascular biology and disease.

5.
Sci Rep ; 10(1): 6707, 2020 04 21.
Article in English | MEDLINE | ID: mdl-32317728

ABSTRACT

The nucleus is fundamentally composed by lamina and nuclear membranes that enclose the chromatin, nucleoskeletal components and suspending nucleoplasm. The functional connections of this network integrate external stimuli into cell signals, including physical forces to mechanical responses of the nucleus. Canonically, the morphological characteristics of the nucleus, as shape and size, have served for pathologists to stratify and diagnose cancer patients; however, novel biophysical techniques must exploit physical parameters to improve cancer diagnosis. By using multiple particle tracking (MPT) technique on chromatin granules, we designed a SURF (Speeded Up Robust Features)-based algorithm to study the mechanical properties of isolated nuclei and in living cells. We have determined the apparent shear stiffness, viscosity and optical density of the nucleus, and how the chromatin structure influences on these biophysical values. Moreover, we used our MPT-SURF analysis to study the apparent mechanical properties of isolated nuclei from patients of acute lymphoblastic leukemia. We found that leukemia cells exhibited mechanical differences compared to normal lymphocytes. Interestingly, isolated nuclei from high-risk leukemia cells showed increased viscosity than their counterparts from normal lymphocytes, whilst nuclei from relapsed-patient's cells presented higher density than those from normal lymphocytes or standard- and high-risk leukemia cells. Taken together, here we presented how MPT-SURF analysis of nuclear chromatin granules defines nuclear mechanical phenotypic features, which might be clinically relevant.


Subject(s)
Cell Nucleus/pathology , Leukemia/pathology , Algorithms , Chromatin/metabolism , Elasticity , Humans , Jurkat Cells , Osmotic Pressure , Phenotype , Rheology , Viscosity
6.
Front Immunol ; 10: 156, 2019.
Article in English | MEDLINE | ID: mdl-30787933

ABSTRACT

The trafficking of neoplastic cells represents a key process that contributes to progression of hematologic malignancies. Diapedesis of neoplastic cells across endothelium and perivascular cells is facilitated by adhesion molecules and chemokines, which act in concert to tightly regulate directional motility. Intravital microscopy provides spatio-temporal views of neoplastic cell trafficking, and is crucial for testing and developing therapies against hematologic cancers. Multiple myeloma (MM), chronic lymphocytic leukemia (CLL), and acute lymphoblastic leukemia (ALL) are hematologic malignancies characterized by continuous neoplastic cell trafficking during disease progression. A common feature of these neoplasias is the homing and infiltration of blood cancer cells into the bone marrow (BM), which favors growth and survival of the malignant cells. MM cells traffic between different BM niches and egress from BM at late disease stages. Besides the BM, CLL cells commonly home to lymph nodes (LNs) and spleen. Likewise, ALL cells also infiltrate extramedullary organs, such as the central nervous system, spleen, liver, and testicles. The α4ß1 integrin and the chemokine receptor CXCR4 are key molecules for MM, ALL, and CLL cell trafficking into and out of the BM. In addition, the chemokine receptor CCR7 controls CLL cell homing to LNs, and CXCR4, CCR7, and CXCR3 contribute to ALL cell migration across endothelia and the blood brain barrier. Some of these receptors are used as diagnostic markers for relapse and survival in ALL patients, and their level of expression allows clinicians to choose the appropriate treatments. In CLL, elevated α4ß1 expression is an established adverse prognostic marker, reinforcing its role in the disease expansion. Combining current chemotherapies with inhibitors of malignant cell trafficking could represent a useful therapy against these neoplasias. Moreover, immunotherapy using humanized antibodies, CAR-T cells, or immune check-point inhibitors together with agents targeting the migration of tumor cells could also restrict their survival. In this review, we provide a view of the molecular players that regulate the trafficking of neoplastic cells during development and progression of MM, CLL, and ALL, together with current therapies that target the malignant cells.


Subject(s)
Cell Movement , Hematologic Neoplasms/metabolism , Hematologic Neoplasms/pathology , ADP-ribosyl Cyclase 1/metabolism , Animals , Chemokines/metabolism , Humans , Hyaluronan Receptors/metabolism , Integrins/metabolism , Matrix Metalloproteinases/metabolism , Selectins/metabolism
7.
Nucleus ; 10(1): 42-47, 2019 12.
Article in English | MEDLINE | ID: mdl-30784352

ABSTRACT

Migrating cells have to cross many physical barriers and confined in 3D environments. The surrounding environment promotes mechano- and biological signals that orchestrate cellular changes, such as cytoskeletal and adhesion rearrangements and proteolytic digestion. Recent studies provide new insights into how the nucleus must alter its shape, localization and mechanical properties in order to promote nuclear deformability, chromatin compaction and gene reprogramming. It is known that the chromatin structure contributes directly to genomic and non-genomic functions, such as gene transcription and the physical properties of the nucleus. Here, we appraise paradigms and novel insights regarding the functional role of chromatin during nuclear deformation. In so doing, we review how constraint and mechanical conditions influence the structure, localization and chromatin decompaction. Finally, we highlight the emerging roles of mechanogenomics and the molecular basis of nucleoskeletal components, which open unexplored territory to understand how cells regulate their chromatin and modify the nucleus.


Subject(s)
Cell Nucleus/genetics , Cell Nucleus/metabolism , Epigenesis, Genetic/genetics , Humans
8.
Cancers (Basel) ; 10(9)2018 Sep 12.
Article in English | MEDLINE | ID: mdl-30213075

ABSTRACT

Acute lymphoblastic leukemia (ALL) is the most common pediatric cancer. As ALL progresses, leukemic cells cross the endothelial barrier and infiltrate other tissues. Epigenetic enzymes represent novel therapeutic targets in hematological malignancies, and might contribute to cells' capacity to migrate across physical barriers. Although many molecules drive this process, the role of the nucleus and its components remain unclear. We report here, for the first time, that the expression of G9a (a histone methyltransferase related with gene silencing) correlates with the expression of the integrin subunit α4 in children with ALL. We have demonstrated that G9a depletion or its inhibition with BIX01294 abrogated the ability of ALL cells to migrate through an endothelial monolayer. Moreover, G9a-depleted and BIX01294-treated cells presented bigger nuclei and more adherent phenotype than control cells on endothelial monolayers. Blocking G9a did not affect the cell cytoskeleton or integrin expression of ALL cell lines, and only its depletion reduced slightly F-actin polymerization. Similarly to the transendothelial migration, G9a inhibition impaired the cell migration induced by the integrin VLA-4 (α4ß1) of primary cells and ALL cell lines through narrow spaces in vitro. Our results suggest a cellular connection between G9a and VLA-4, which underlies novel functions of G9a during ALL cell migration.

9.
Proc Natl Acad Sci U S A ; 115(34): 8581-8586, 2018 08 21.
Article in English | MEDLINE | ID: mdl-29987046

ABSTRACT

Cell migration through extracellular matrices requires nuclear deformation, which depends on nuclear stiffness. In turn, chromatin structure contributes to nuclear stiffness, but the mechanosensing pathways regulating chromatin during cell migration remain unclear. Here, we demonstrate that WD repeat domain 5 (WDR5), an essential component of H3K4 methyltransferase complexes, regulates cell polarity, nuclear deformability, and migration of lymphocytes in vitro and in vivo, independent of transcriptional activity, suggesting nongenomic functions for WDR5. Similarly, depletion of RbBP5 (another H3K4 methyltransferase subunit) promotes similar defects. We reveal that a 3D environment increases the H3K4 methylation dependent on WDR5 and results in a globally less compacted chromatin conformation. Further, using atomic force microscopy, nuclear particle tracking, and nuclear swelling experiments, we detect changes in nuclear mechanics that accompany the epigenetic changes induced in 3D conditions. Indeed, nuclei from cells in 3D environments were softer, and thereby more deformable, compared with cells in suspension or cultured in 2D conditions, again dependent on WDR5. Dissecting the underlying mechanism, we determined that actomyosin contractility, through the phosphorylation of myosin by MLCK (myosin light chain kinase), controls the interaction of WDR5 with other components of the methyltransferase complex, which in turn up-regulates H3K4 methylation activation in 3D conditions. Taken together, our findings reveal a nongenomic function for WDR5 in regulating H3K4 methylation induced by 3D environments, physical properties of the nucleus, cell polarity, and cell migratory capacity.


Subject(s)
Cell Movement , Cell Polarity , Chromatin/metabolism , Epigenesis, Genetic , Histone-Lysine N-Methyltransferase/metabolism , Nuclear Proteins/metabolism , Chromatin/genetics , Chromatin/ultrastructure , DNA-Binding Proteins , Histone-Lysine N-Methyltransferase/chemistry , Histone-Lysine N-Methyltransferase/genetics , Humans , Intracellular Signaling Peptides and Proteins , Jurkat Cells , Microscopy, Atomic Force , Myosin-Light-Chain Kinase/genetics , Myosin-Light-Chain Kinase/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/genetics
10.
Cancers (Basel) ; 9(7)2017 Jul 06.
Article in English | MEDLINE | ID: mdl-28684679

ABSTRACT

Cancer cell migration is a complex process that requires coordinated structural changes and signals in multiple cellular compartments. The nucleus is the biggest and stiffest organelle of the cell and might alter its physical properties to allow cancer cell movement. Integrins are transmembrane receptors that mediate cell-cell and cell-extracellular matrix interactions, which regulate numerous intracellular signals and biological functions under physiological conditions. Moreover, integrins orchestrate changes in tumor cells and their microenvironment that lead to cancer growth, survival and invasiveness. Most of the research efforts have focused on targeting integrin-mediated adhesion and signaling. Recent exciting data suggest the crucial role of integrins in controlling internal cellular structures and nuclear alterations during cancer cell migration. Here we review the emerging role of integrins in nuclear biology. We highlight increasing evidence that integrins are critical for changes in multiple nuclear components, the positioning of the nucleus and its mechanical properties during cancer cell migration. Finally, we discuss how integrins are integral proteins linking the plasma membrane and the nucleus, and how they control cell migration to enable cancer invasion and infiltration. The functional connections between these cell receptors and the nucleus will serve to define new attractive therapeutic targets.

11.
Nucleic Acids Res ; 44(7): 3031-44, 2016 Apr 20.
Article in English | MEDLINE | ID: mdl-26657637

ABSTRACT

The mechanical properties of the cell nucleus change to allow cells to migrate, but how chromatin modifications contribute to nuclear deformability has not been defined. Here, we demonstrate that a major factor in this process involves epigenetic changes that underpin nuclear structure. We investigated the link between cell adhesion and epigenetic changes in T-cells, and demonstrate that T-cell adhesion to VCAM1 via α4ß1 integrin drives histone H3 methylation (H3K9me2/3) through the methyltransferase G9a. In this process, active G9a is recruited to the nuclear envelope and interacts with lamin B1 during T-cell adhesion through α4ß1 integrin. G9a activity not only reorganises the chromatin structure in T-cells, but also affects the stiffness and viscoelastic properties of the nucleus. Moreover, we further demonstrated that these epigenetic changes were linked to lymphocyte movement, as depletion or inhibition of G9a blocks T-cell migration in both 2D and 3D environments. Thus, our results identify a novel mechanism in T-cells by which α4ß1 integrin signaling drives specific chromatin modifications, which alter the physical properties of the nucleus and thereby enable T-cell migration.


Subject(s)
Cell Movement , Cell Nucleus/physiology , Epigenesis, Genetic , Histocompatibility Antigens/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Integrin alpha4beta1/metabolism , Lymphocytes/immunology , Animals , Cell Adhesion , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cells, Cultured , Chromatin/chemistry , HEK293 Cells , Histones/metabolism , Humans , Jurkat Cells , Methylation , Mice, Inbred C57BL , Vascular Cell Adhesion Molecule-1/metabolism
12.
Mol Cell Biol ; 35(1): 249-63, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25348717

ABSTRACT

The nuclear envelope (NE) forms a barrier between the nucleus and the cytosol that preserves genomic integrity. The nuclear lamina and nuclear pore complexes (NPCs) are NE components that regulate nuclear events through interaction with other proteins and DNA. Defects in the nuclear lamina are associated with the development of laminopathies. As cells depleted of phosphoinositide 3-kinase beta (PI3Kß) showed an aberrant nuclear morphology, we studied the contribution of PI3Kß to maintenance of NE integrity. pik3cb depletion reduced the nuclear membrane tension, triggered formation of areas of lipid bilayer/lamina discontinuity, and impaired NPC assembly. We show that one mechanism for PI3Kß regulation of NE/NPC integrity is its association with RCC1 (regulator of chromosome condensation 1), the activator of nuclear Ran GTPase. PI3Kß controls RCC1 binding to chromatin and, in turn, Ran activation. These findings suggest that PI3Kß regulates the nuclear envelope through upstream regulation of RCC1 and Ran.


Subject(s)
Cell Cycle Proteins/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Nuclear Envelope/metabolism , Nuclear Proteins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , ran GTP-Binding Protein/metabolism , Animals , Cell Cycle , Chromatin/metabolism , Class I Phosphatidylinositol 3-Kinases , Fibroblasts/metabolism , HEK293 Cells , Humans , Lipid Bilayers , Mice , Microscopy, Confocal , Microscopy, Electron , NIH 3T3 Cells , Protein Binding
13.
Biol Open ; 3(10): 924-36, 2014 Sep 12.
Article in English | MEDLINE | ID: mdl-25217619

ABSTRACT

The acquisition of invasiveness is characteristic of tumor progression. Numerous genetic changes are associated with metastasis, but the mechanism by which a cell becomes invasive remains unclear. Expression of p85ß, a regulatory subunit of phosphoinositide-3-kinase, markedly increases in advanced carcinoma, but its mode of action is unknown. We postulated that p85ß might facilitate cell invasion. We show that p85ß localized at cell adhesions in complex with focal adhesion kinase and enhanced stability and maturation of cell adhesions. In addition, p85ß induced development at cell adhesions of an F-actin core that extended several microns into the cell z-axis resembling the skeleton of invadopodia. p85ß lead to F-actin polymerization at cell adhesions by recruiting active Cdc42/Rac at these structures. In accordance with p85ß function in invadopodium-like formation, p85ß levels increased in metastatic melanoma and p85ß depletion reduced invadopodium formation and invasion. These results show that p85ß enhances invasion by inducing cell adhesion development into invadopodia-like structures explaining the metastatic potential of tumors with increased p85ß levels.

14.
Mol Cell Biol ; 34(18): 3359-73, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-24958106

ABSTRACT

The phosphoinositide 3-kinase (PI3K)/PTEN (phosphatase and tensin homolog) pathway is one of the central routes that enhances cell survival, division, and migration, and it is frequently deregulated in cancer. PI3K catalyzes formation of phosphatidylinositol 3,4,5-triphosphate [PI(3,4,5)P3] after cell activation; PTEN subsequently reduces these lipids to basal levels. Activation of the ubiquitous p110α isoform precedes that of p110ß at several points during the cell cycle. We studied the potential connections between p110α and p110ß activation, and we show that cell stimulation promotes p110α and p110ß association, demonstrating oligomerization of PI3K catalytic subunits within cells. Cell stimulation also promoted PTEN incorporation into this complex, which was necessary for PTEN activation. Our results show that PI3Ks dimerize in vivo and that PI3K and PTEN activities modulate each other in a complex that controls cell PI(3,4,5)P3 levels.


Subject(s)
Class Ia Phosphatidylinositol 3-Kinase/metabolism , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol Phosphates/metabolism , Animals , Cell Cycle , Cell Line , Class Ia Phosphatidylinositol 3-Kinase/genetics , Dimerization , Gene Expression Regulation , HEK293 Cells , Humans , Mice , NIH 3T3 Cells , PTEN Phosphohydrolase/genetics , Signal Transduction/physiology
15.
Cell Signal ; 25(4): 848-59, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23333462

ABSTRACT

Recent studies have suggested a regulatory role for the dioxin receptor (AhR) in cell adhesion and migration. Following our previous work, we report here that the C-terminal Src kinase-binding protein (Cbp) signaling pathway controls ß1 integrin activation and that this mechanism is AhR dependent. T-FGM AhR-/- fibroblasts displayed higher integrin ß1 activation, revealed by the increased binding of the activation reporter 9EG7 anti-ß1 mAb and of a soluble fibronectin fragment, as well as by enhanced talin-ß1 association. AhR-/- fibroblasts also showed increased fibronectin secretion and impaired directional migration. Notably, interfering Cbp expression in AhR-/- fibroblasts reduced ß1 integrin activation, improved cell migration and rescued wild-type cell morphology. Cbp over-expression in T-FGM AhR-/- cells enhanced the formation of inhibitory Csk-Cbp complexes which in turn reduced c-Src p-Tyr(416) activation and focal adhesion kinase (FAK) phosphorylation at the c-Src-responsive residues p-Tyr(576) and p-Tyr(577). The c-Src target and migration-related protein Cav1 was also hypophosphorylated at p-Tyr(14) in AhR-/- cells, and such effect was rescued by down-modulating Cbp levels. Thus, AhR regulates fibroblast migration by modulating ß1 integrin activation via Cbp-dependent, Src-mediated signaling.


Subject(s)
Integrin beta1/metabolism , Membrane Proteins/metabolism , Phosphoproteins/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Sialoglycoproteins/metabolism , src-Family Kinases/metabolism , Actins/metabolism , Animals , CSK Tyrosine-Protein Kinase , Caveolin 1/metabolism , Cell Adhesion , Cell Movement , Fibroblasts/metabolism , Fibronectins/metabolism , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Mice , Phosphorylation , RNA Interference , RNA, Small Interfering/metabolism , Receptors, Aryl Hydrocarbon/antagonists & inhibitors , Receptors, Aryl Hydrocarbon/genetics , Signal Transduction
16.
J Pathol ; 229(1): 36-48, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22711564

ABSTRACT

Myeloma cell adhesion dependent on α4ß1 integrin is crucial for the progression of multiple myeloma (MM). The α4ß1-dependent myeloma cell adhesion is up-regulated by the chemokine CXCL12, and pharmacological blockade of the CXCL12 receptor CXCR4 leads to defective myeloma cell homing to bone marrow (BM). Sphingosine-1-phosphate (S1P) regulates immune cell trafficking upon binding to G-protein-coupled receptors. Here we show that myeloma cells express S1P1, a receptor for S1P. We found that S1P up-regulated the α4ß1-mediated myeloma cell adhesion and transendothelial migration stimulated by CXCL12. S1P promoted generation of high-affinity α4ß1 that efficiently bound the α4ß1 ligand VCAM-1, a finding that was associated with S1P-triggered increase in talin-ß1 integrin association. Furthermore, S1P cooperated with CXCL12 for enhancement of α4ß1-dependent adhesion strengthening and spreading. CXCL12 and S1P activated the DOCK2-Rac1 pathway, which was required for stimulation of myeloma cell adhesion involving α4ß1. Moreover, in vivo analyses indicated that S1P contributes to optimizing the interactions of MM cells with the BM microvasculture and for their lodging inside the bone marrow. The regulation of α4ß1-dependent adhesion and migration of myeloma cells by CXCL12-S1P combined activities might have important consequences for myeloma disease progression.


Subject(s)
Bone Marrow/metabolism , Cell Adhesion , Chemokine CXCL12/metabolism , Integrin alpha4beta1/metabolism , Lysophospholipids/metabolism , Multiple Myeloma/metabolism , Sphingosine/analogs & derivatives , Stromal Cells/metabolism , Transendothelial and Transepithelial Migration , Animals , Bone Marrow/blood supply , Bone Marrow/immunology , Bone Marrow/pathology , Cell Shape , Coculture Techniques , GTPase-Activating Proteins , Guanine Nucleotide Exchange Factors/metabolism , Humans , Integrin alpha5beta1/metabolism , K562 Cells , Mice , Mice, Inbred NOD , Mice, SCID , Multiple Myeloma/genetics , Multiple Myeloma/immunology , Multiple Myeloma/pathology , RNA Interference , Receptors, Lysosphingolipid/genetics , Receptors, Lysosphingolipid/metabolism , Signal Transduction , Sphingosine/metabolism , Stromal Cells/immunology , Stromal Cells/pathology , Talin/metabolism , Time Factors , Transfection , Tumor Cells, Cultured , Vascular Cell Adhesion Molecule-1/metabolism , rac1 GTP-Binding Protein/metabolism
17.
Nucleic Acids Res ; 41(2): 855-68, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23175608

ABSTRACT

Genomic integrity is preserved by the action of protein complexes that control DNA homeostasis. These include the sliding clamps, trimeric protein rings that are arranged around DNA by clamp loaders. Replication factor C (RFC) is the clamp loader for proliferating cell nuclear antigen, which acts on DNA replication. Other processes that require mobile contact of proteins with DNA use alternative RFC complexes that exchange RFC1 for CTF18 or RAD17. Phosphoinositide 3-kinases (PI3K) are lipid kinases that generate 3-poly-phosphorylated-phosphoinositides at the plasma membrane following receptor stimulation. The two ubiquitous isoforms, PI3Kalpha and PI3Kbeta, have been extensively studied due to their involvement in cancer and nuclear PI3Kbeta has been found to regulate DNA replication and repair, processes controlled by molecular clamps. We studied here whether PI3Kbeta directly controls the process of molecular clamps loading. We show that PI3Kbeta associated with RFC1 and RFC1-like subunits. Only when in complex with PI3Kbeta, RFC1 bound to Ran GTPase and localized to the nucleus, suggesting that PI3Kbeta regulates RFC1 nuclear import. PI3Kbeta controlled not only RFC1- and RFC-RAD17 complexes, but also RFC-CTF18, in turn affecting CTF18-mediated chromatid cohesion. PI3Kbeta thus has a general function in genomic stability by controlling the localization and function of RFC complexes.


Subject(s)
Class Ia Phosphatidylinositol 3-Kinase/metabolism , Replication Protein C/metabolism , Amino Acid Motifs , Animals , Cell Line , Cell Nucleus/metabolism , Class Ia Phosphatidylinositol 3-Kinase/chemistry , Class Ia Phosphatidylinositol 3-Kinase/physiology , DNA Repair , DNA Replication , Humans , Protein Subunits/metabolism , Replication Protein C/antagonists & inhibitors , Replication Protein C/chemistry , ran GTP-Binding Protein/metabolism
18.
Mol Biol Cell ; 23(23): 4526-42, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23051731

ABSTRACT

Class I(A) phosphoinositide 3-kinases (PI3K) are enzymes composed of a p85 regulatory and a p110 catalytic subunit that control formation of 3-poly-phosphoinositides (PIP(3)). The PI3K pathway regulates cell survival, migration, and division, and is mutated in approximately half of human tumors. For this reason, it is important to define the function of the ubiquitous PI3K subunits, p110α and p110ß. Whereas p110α is activated at G1-phase entry and promotes protein synthesis and gene expression, p110ß activity peaks in S phase and regulates DNA synthesis. PI3K activity also increases at the onset of mitosis, but the isoform activated is unknown; we have examined p110α and p110ß function in mitosis. p110α was activated at mitosis entry and regulated early mitotic events, such as PIP(3) generation, prometaphase progression, and spindle orientation. In contrast, p110ß was activated near metaphase and controlled dynein/dynactin and Aurora B activities in kinetochores, chromosome segregation, and optimal function of the spindle checkpoint. These results reveal a p110ß function in preserving genomic stability during mitosis.


Subject(s)
Class Ia Phosphatidylinositol 3-Kinase , DNA , Phosphatidylinositol 3-Kinases , Animals , Aurora Kinase B , Aurora Kinases , Cell Cycle , Cell Survival , Chromosome Segregation , Class Ia Phosphatidylinositol 3-Kinase/genetics , Class Ia Phosphatidylinositol 3-Kinase/metabolism , DNA/biosynthesis , DNA/genetics , Dyneins/metabolism , HeLa Cells , Humans , Kinetochores/metabolism , Mice , Mitosis , NIH 3T3 Cells , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Spindle Apparatus/genetics
19.
Mol Cell Biol ; 31(10): 2122-33, 2011 May.
Article in English | MEDLINE | ID: mdl-21383062

ABSTRACT

Class I(A) phosphoinositide 3-kinases (PI3Ks) are heterodimeric enzymes composed of a p85 regulatory and a p110 catalytic subunit that induce the formation of 3-polyphosphoinositides, which mediate cell survival, division, and migration. There are two ubiquitous PI3K isoforms p110α and p110ß that have nonredundant functions in embryonic development and cell division. However, whereas p110α concentrates in the cytoplasm, p110ß localizes to the nucleus and modulates nuclear processes such as DNA replication and repair. At present, the structural features that determine p110ß nuclear localization remain unknown. We describe here that association with the p85ß regulatory subunit controls p110ß nuclear localization. We identified a nuclear localization signal (NLS) in p110ß C2 domain that mediates its nuclear entry, as well as a nuclear export sequence (NES) in p85ß. Deletion of p110ß induced apoptosis, and complementation with the cytoplasmic C2-NLS p110ß mutant was unable to restore cell survival. These studies show that p110ß NLS and p85ß NES regulate p85ß/p110ß nuclear localization, supporting the idea that nuclear, but not cytoplasmic, p110ß controls cell survival.


Subject(s)
Cell Nucleus/enzymology , Cell Survival , Class Ia Phosphatidylinositol 3-Kinase/metabolism , Nuclear Export Signals , Nuclear Localization Signals , Phosphatidylinositol 3-Kinases/metabolism , Amino Acid Sequence , Animals , Apoptosis , Blotting, Western , Cell Line , Class I Phosphatidylinositol 3-Kinases , Class Ia Phosphatidylinositol 3-Kinase/chemistry , Class Ia Phosphatidylinositol 3-Kinase/genetics , Cytosol/enzymology , Fluorescent Antibody Technique , Humans , Immunoprecipitation , Mice , Molecular Sequence Data , NIH 3T3 Cells , Phosphatidylinositol 3-Kinases/chemistry , Phosphatidylinositol 3-Kinases/genetics , Signal Transduction
20.
Clin Cancer Res ; 16(17): 4382-91, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20534739

ABSTRACT

PURPOSE: Arsenic trioxide (ATO) induces B-cell chronic lymphocytic leukemia (B-CLL) cell apoptosis in vitro. We sought to study the mechanism involved in this effect and whether ATO is suitable for combination therapies with protein kinase inhibitors. EXPERIMENTAL DESIGN: B-CLL cells were isolated from the peripheral blood of 28 patients. Cell viability studies with ATO alone or in combination with kinase inhibitors were done by flow cytometry, Western blotting, and immunofluorescence analyses. RESULTS: After 48 hours, 3 mumol/L ATO induced apoptosis (average 75%) in all B-CLL samples studied and with minimal effect on normal peripheral blood lymphocytes. Apoptosis entailed Akt and NF-kappaB inactivation, XIAP downregulation, and PTEN upregulation, thus implying inhibition of the phosphoinositide 3-kinase (PI3K) survival pathway. Indeed, the combination of ATO and PI3K inhibitors increased the apoptotic effect of either agent alone. ATO also induced c-jun-NH(2) terminal kinase (JNK) activation, and this was crucial and required for subsequent apoptotic events, as inhibiting JNK activity by either gene silencing or specific inhibitors prevented Akt and NF-kappaB inactivation, caspase activation, and mitochondrial damage. Moreover, JNK activation was the earliest response to ATO, preceding and determining reactive oxygen species production. CONCLUSIONS: We identified the mechanism involved in ATO action on B-CLL cells and show that the combination of low doses of ATO and PI3K inhibitors efficiently induces B-CLL cell death. ATO may therefore constitute an efficient treatment for B-CLL, particularly in combined therapies.


Subject(s)
Apoptosis/drug effects , Arsenicals/pharmacology , JNK Mitogen-Activated Protein Kinases/metabolism , Oxides/pharmacology , PTEN Phosphohydrolase/metabolism , Signal Transduction/drug effects , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/pharmacology , Arsenic Trioxide , Blotting, Western , Cell Survival/drug effects , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Female , Humans , JNK Mitogen-Activated Protein Kinases/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Male , Membrane Potential, Mitochondrial/drug effects , Microscopy, Confocal , Middle Aged , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , Tumor Cells, Cultured , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...