Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters










Publication year range
1.
EMBO Mol Med ; 16(6): 1404-1426, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38684864

ABSTRACT

As an important immune stimulator and modulator, IFNγ is crucial for gut homeostasis and its dysregulation links to diverse colon pathologies, such as colitis and colorectal cancer (CRC). Here, we demonstrated that the epigenetic regulator, CBX3 (also known as HP1γ) antagonizes IFNγ signaling in the colon epithelium by transcriptionally repressing two critical IFNγ-responsive genes: STAT1 and CD274 (encoding Programmed death-ligand 1, PD-L1). Accordingly, CBX3 deletion resulted in chronic mouse colon inflammation, accompanied by upregulated STAT1 and CD274 expressions. Chromatin immunoprecipitation indicated that CBX3 tethers to STAT1 and CD274 promoters to inhibit their expression. Reversely, IFNγ significantly reduces CBX3 binding to these promoters and primes gene expression. This antagonist effect between CBX3 and IFNγ on STAT1/PD-L1 expression was also observed in CRC. Strikingly, CBX3 deletion heightened CRC cells sensitivity to IFNγ, which ultimately enhanced their chemosensitivity under IFNγ stimulation in vitro with CRC cells and in vivo with a syngeneic mouse tumor model. Overall, this work reveals that by negatively tuning IFNγ-stimulated immune genes' transcription, CBX3 participates in modulating colon inflammatory response and CRC chemo-resistance.


Subject(s)
B7-H1 Antigen , Chromosomal Proteins, Non-Histone , Colorectal Neoplasms , Interferon-gamma , STAT1 Transcription Factor , Animals , B7-H1 Antigen/metabolism , B7-H1 Antigen/genetics , Interferon-gamma/metabolism , STAT1 Transcription Factor/metabolism , STAT1 Transcription Factor/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Colorectal Neoplasms/genetics , Humans , Mice , Chromosomal Proteins, Non-Histone/metabolism , Chromosomal Proteins, Non-Histone/genetics , Colitis/metabolism , Colitis/pathology , Mice, Inbred C57BL , Signal Transduction , Cell Line, Tumor
2.
J Gen Physiol ; 155(1)2023 01 02.
Article in English | MEDLINE | ID: mdl-36326690

ABSTRACT

Using cell-attached recordings from molecular layer interneurons (MLI) of the cerebellar cortex of adult mice expressing channel rhodopsin 2, we show that wide-field optical activation induces an increase in firing rate during illumination and a firing pause when the illumination ends (post-stimulation silencing; PSS). Significant spike rate changes with respect to basal firing rate were observed for optical activations lasting 200 ms and 1 s as well as for 1 s long trains of 10 ms pulses at 50 Hz. For all conditions, the net effect of optical activation on the integrated spike rate is significantly reduced because of PSS. Three lines of evidence indicate that this PSS is due to intrinsic factors. Firstly, PSS is induced when the optical stimulation is restricted to a single MLI using a 405-nm laser delivering a diffraction-limited spot at the focal plane. Secondly, PSS is not affected by block of GABA-A or GABA-B receptors, ruling out synaptic interactions amongst MLIs. Thirdly, PSS is mimicked in whole-cell recording experiments by step depolarizations under current clamp. Activation of Ca-dependent K channels during the spike trains appears as a likely candidate to underlie PSS. Using immunocytochemistry, we find that one such channel type, KCa1.1, is present in the somato-dendritic and axonal compartments of MLIs. In cell-attached recordings, charybdotoxin and iberiotoxin significantly reduce the optically induced PSS, while TRAM-34 does not affect it, suggesting that KCa1.1 channels, but not KCa3.1 channels, contribute to PSS.


Subject(s)
Cerebellum , Interneurons , Mice , Animals , Interneurons/physiology , Cerebellum/physiology , Patch-Clamp Techniques , Action Potentials
3.
Proc Natl Acad Sci U S A ; 117(20): 11097-11108, 2020 05 19.
Article in English | MEDLINE | ID: mdl-32358199

ABSTRACT

It has been known for a long time that inositol-trisphosphate (IP3) receptors are present in the axon of certain types of mammalian neurons, but their functional role has remained unexplored. Here we show that localized photolysis of IP3 induces spatially constrained calcium rises in Purkinje cell axons. Confocal immunohistology reveals that the axon initial segment (AIS), as well as terminals onto deep cerebellar cells, express specific subtypes of Gα/q and phospholipase C (PLC) molecules, together with the upstream purinergic receptor P2Y1. By contrast, intermediate parts of the axon express another set of Gα/q and PLC molecules, indicating two spatially segregated signaling cascades linked to IP3 generation. This prompted a search for distinct actions of IP3 in different parts of Purkinje cell axons. In the AIS, we found that local applications of the specific P2Y1R agonist MRS2365 led to calcium elevation, and that IP3 photolysis led to inhibition of action potential firing. In synaptic terminals on deep cerebellar nuclei neurons, we found that photolysis of both IP3 and ATP led to GABA release. We propose that axonal IP3 receptors can inhibit action potential firing and increase neurotransmitter release, and that these effects are likely controlled by purinergic receptors. Altogether our results suggest a rich and diverse functional role of IP3 receptors in axons of mammalian neurons.


Subject(s)
Action Potentials/physiology , Axons/metabolism , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Inositol 1,4,5-Trisphosphate/metabolism , Purkinje Cells/metabolism , Calcium/metabolism , Cerebellum/metabolism , Neurons/metabolism , Presynaptic Terminals/metabolism , Receptors, Purinergic P2Y1 , Type C Phospholipases/metabolism
4.
Elife ; 92020 05 13.
Article in English | MEDLINE | ID: mdl-32401196

ABSTRACT

Type 1 metabotropic glutamate receptors (mGluR1s) are key elements in neuronal signaling. While their function is well documented in slices, requirements for their activation in vivo are poorly understood. We examine this question in adult mice in vivo using 2-photon imaging of cerebellar molecular layer interneurons (MLIs) expressing GCaMP. In anesthetized mice, parallel fiber activation evokes beam-like Cai rises in postsynaptic MLIs which depend on co-activation of mGluR1s and ionotropic glutamate receptors (iGluRs). In awake mice, blocking mGluR1 decreases Cai rises associated with locomotion. In vitro studies and freeze-fracture electron microscopy show that the iGluR-mGluR1 interaction is synergistic and favored by close association of the two classes of receptors. Altogether our results suggest that mGluR1s, acting in synergy with iGluRs, potently contribute to processing cerebellar neuronal signaling under physiological conditions.


Subject(s)
Cerebellum/metabolism , Interneurons/metabolism , Receptors, Ionotropic Glutamate/metabolism , Receptors, Metabotropic Glutamate/metabolism , Synaptic Transmission , Animals , Biosensing Techniques , Calcium Signaling , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cerebellum/ultrastructure , Female , Interneurons/ultrastructure , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Microscopy, Fluorescence, Multiphoton , Motor Activity , Walking
5.
Cell ; 179(7): 1590-1608.e23, 2019 12 12.
Article in English | MEDLINE | ID: mdl-31835034

ABSTRACT

Optical interrogation of voltage in deep brain locations with cellular resolution would be immensely useful for understanding how neuronal circuits process information. Here, we report ASAP3, a genetically encoded voltage indicator with 51% fluorescence modulation by physiological voltages, submillisecond activation kinetics, and full responsivity under two-photon excitation. We also introduce an ultrafast local volume excitation (ULoVE) method for kilohertz-rate two-photon sampling in vivo with increased stability and sensitivity. Combining a soma-targeted ASAP3 variant and ULoVE, we show single-trial tracking of spikes and subthreshold events for minutes in deep locations, with subcellular resolution and with repeated sampling over days. In the visual cortex, we use soma-targeted ASAP3 to illustrate cell-type-dependent subthreshold modulation by locomotion. Thus, ASAP3 and ULoVE enable high-speed optical recording of electrical activity in genetically defined neurons at deep locations during awake behavior.


Subject(s)
Brain/physiology , GTPase-Activating Proteins/genetics , Microscopy, Fluorescence, Multiphoton/methods , Optogenetics/methods , Theta Rhythm , Wakefulness , Action Potentials , Animals , Brain/metabolism , CHO Cells , Cells, Cultured , Cricetinae , Cricetulus , Female , GTPase-Activating Proteins/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/metabolism , Rats , Rats, Sprague-Dawley , Running
6.
J Gen Physiol ; 146(6): 477-93, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26621773

ABSTRACT

Axonal ionotropic receptors are present in a variety of neuronal types, and their function has largely been associated with the modulation of axonal activity and synaptic release. It is usually assumed that activation of axonal GABA(A)Rs comes from spillover, but in cerebellar molecular layer interneurons (MLIs) the GABA source is different: in these cells, GABA release activates presynaptic GABA(A) autoreceptors (autoRs) together with postsynaptic targets, producing an autoR-mediated synaptic event. The frequency of presynaptic, autoR-mediated miniature currents is twice that of their somatodendritic counterparts, suggesting that autoR-mediated responses have an important effect on interneuron activity. Here, we used local Ca(2+) photolysis in MLI axons of juvenile rats to evoke GABA release from individual varicosities to study the activation of axonal autoRs in single release sites. Our data show that single-site autoR conductances are similar to postsynaptic dendritic conductances. In conditions of high [Cl(-)](i), autoR-mediated conductances range from 1 to 5 nS; this corresponds to ∼30-150 GABA(A) channels per presynaptic varicosity, a value close to the number of channels in postsynaptic densities. Voltage responses produced by the activation of autoRs in single varicosities are amplified by a Na(v)-dependent mechanism and propagate along the axon with a length constant of 91 µm. Immunolabeling determination of synapse location shows that on average, one third of the synapses produce autoR-mediated signals that are large enough to reach the axon initial segment. Finally, we show that single-site activation of presynaptic GABA(A) autoRs leads to an increase in MLI excitability and thus conveys a strong feedback signal that contributes to spiking activity.


Subject(s)
Cerebellum/physiology , GABAergic Neurons/physiology , Interneurons/physiology , Synaptic Potentials , Animals , Axons/physiology , Calcium/metabolism , Cerebellum/cytology , Female , Male , Rats , Rats, Sprague-Dawley , Receptors, GABA-A/metabolism , Synapses/physiology , Voltage-Gated Sodium Channels/metabolism , gamma-Aminobutyric Acid/metabolism
7.
Front Cell Neurosci ; 9: 275, 2015.
Article in English | MEDLINE | ID: mdl-26236197

ABSTRACT

While it has been proposed that the conventional inhibitory neurotransmitter GABA can be excitatory in the mammalian brain, much remains to be learned concerning the circumstances and the cellular mechanisms governing potential excitatory GABA action. Using a combination of optogenetics and two-photon calcium imaging in vivo, we find that activation of chloride-permeable GABAA receptors in parallel fibers (PFs) of the cerebellar molecular layer of adult mice causes parallel fiber excitation. Stimulation of PFs at submaximal stimulus intensities leads to GABA release from molecular layer interneurons (MLIs), thus creating a positive feedback loop that enhances excitation near the center of an activated PF bundle. Our results imply that elevated chloride concentration can occur in specific intracellular compartments of mature mammalian neurons and suggest an excitatory role for GABAA receptors in the cerebellar cortex of adult mice.

8.
J Mol Med (Berl) ; 91(10): 1207-20, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23744108

ABSTRACT

UNLABELLED: Gap junctions (GJs) enable intercellular communication between adjacent cells through channels of connexins. Using a three-dimensional construct, we previously showed that endothelial and tumor cells formed GJs, allowing melanoma-specific T lymphocytes to recognize and kill melanoma-derived endothelial cells. We demonstrate here on histological sections of melanoma biopsies that GJ formation occurs in vivo between tumor and endothelial cells and between T lymphocytes and target cells. We also show an in vitro increase of GJ formation in melanoma and endothelial cells following dacarbazin and interferon gamma (IFN-γ) treatment or hypoxic stress induction. Our data indicate that although connexin 43 (Cx43), the main GJ protein of the immune system, was localized at the immunological synapse between T lymphocyte and autologous melanoma cells, its over-expression or inhibition of GJs does not interfere with cytotoxic T lymphocyte (CTL) clone lytic function. In contrast, we showed that inhibition of GJs by oleamide during stimulation of resting PBMCs with Melan-A natural and analog peptides resulted in a decrease in antigen (Ag) specific CD8(+) T lymphocyte induction. These Ag-specific CD8(+) cells displayed paradoxically stronger reactivity as revealed by CD107a degranulation and IFN-γ secretion. These findings indicate that Cx43 does not affect lytic function of differentiated CTL, but reveal a major role for GJs in the regulation of antigen CD8(+)-naïve T lymphocyte activation. KEY MESSAGE: GJ formation occurs in vivo between T lymphocytes and tumor cells Cx43 localized at the immunological synapse between T and autologous melanoma cells Inhibition of GJs resulted in a decrease in Ag-specific CD8(+) T lymphocyte induction A role for GJs in the regulation of antigen CD8(+)-naïve T lymphocyte activation.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Gap Junctions/immunology , MART-1 Antigen/immunology , Melanoma/immunology , Antineoplastic Agents, Alkylating/pharmacology , Apoptosis/genetics , Apoptosis/immunology , Cell Line, Tumor , Connexin 43/genetics , Connexin 43/metabolism , Cytotoxicity, Immunologic/genetics , Dacarbazine/pharmacology , Gap Junctions/drug effects , Gap Junctions/metabolism , Gene Expression , Humans , Hypoxia , Immunological Synapses/metabolism , Interferon-gamma/pharmacology , Melanoma/genetics , Melanoma/metabolism , Oxidative Stress , Protein Transport , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism
9.
Cell Cycle ; 11(23): 4385-9, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-23159853

ABSTRACT

Megakaryocyte is the naturally polyploid cell that gives rise to platelets. Polyploidization occurs by endomitosis, a process corresponding to a late failure of cytokinesis with a backward movement of the daughter cells. Generally, a pure defect in cytokinesis produces a multinucleated cell, but megakaryocytes are characterized by a single polylobulated nucleus with a 2 (N) ploidy. Here, we show the existence of a defect in karyokinesis during the endomitotic process. From late telophase until the reversal of cytokinesis, some dipolar mitosis/endomitosis and most multipolar endomitosis present a thin DNA link between the segregated chromosomes surrounded by an incomplete nuclear membrane formation, which implies that sister chromatid separation is not complete. This observation may explain why polyploid megakaryocytes display a single polylobulated nucleus along with an increase in ploidy.


Subject(s)
Megakaryocytes/cytology , Mitosis , Antigens, CD34/metabolism , Cell Nucleus Division , Humans , Megakaryocytes/metabolism , Microscopy, Confocal , Telophase
10.
PLoS One ; 7(6): e38808, 2012.
Article in English | MEDLINE | ID: mdl-22719951

ABSTRACT

BACKGROUND: Isolated limb perfusion with TNF-α and melphalan is used with remarkable efficiency to treat unresectable limb sarcomas. Here we tested the ability of TNF-α to directly induce apoptosis of sarcoma cells. In addition, we investigated the impact of p53 in the regulation of such effect. METHODOLOGY/PRINCIPAL FINDINGS: We first analysed the ability of TNF-α to induce apoptosis in freshly isolated tumour cells. For this purpose, sarcoma tumours (n = 8) treated ex vivo with TNF-α were processed for TUNEL staining. It revealed substantial endothelial cell apoptosis and levels of tumour cell apoptosis that varied from low to high. In order to investigate the role of p53 in TNF-α-induced cell death, human sarcoma cell lines (n = 9) with different TP53 and MDM2 status were studied for their sensitivity to TNF-α. TP53(Wt) cell lines were sensitive to TNF-α unless MDM2 was over-expressed. However, TP53(Mut) and TP53(Null) cell lines were resistant. TP53 suppression in TP53(Wt) cell lines abrogated TNF-α sensitivity and TP53 overexpression in TP53(Null) cell lines restored it. The use of small molecules that restore p53 activity, such as CP-31398 or Nutlin-3a, in association with TNF-α, potentiated the cell death of respectively TP53(Mut) and TP53(Wt)/MDM2(Ampl). In particular, CP-31398 was able to induce p53 as well as some of its apoptotic target genes in TP53(Mut) cells. In TP53(Wt)/MDM2(Ampl) cells, Nutlin-3a effects were associated with a decrease of TNF-α-induced NF-κB-DNA binding and correlated with a differential regulation of pro- and anti-apoptotic genes such as TP53BP2, GADD45, TGF-ß1 and FAIM. CONCLUSION/SIGNIFICANCE: More effective therapeutic approaches are critically needed for the treatment of unresectable limb sarcomas. Our results show that restoring p53 activity in sarcoma cells correlated with increased sensitivity to TNF-α, suggesting that this strategy may be an important determinant of TNF-α-based sarcomas treatment.


Subject(s)
Genes, p53 , Sarcoma/physiopathology , Tumor Necrosis Factor-alpha/pharmacology , Apoptosis , Blotting, Western , Cell Line, Tumor , Flow Cytometry , Humans , In Situ Nick-End Labeling , Mutation , Real-Time Polymerase Chain Reaction , Sarcoma/pathology
11.
Nat Commun ; 3: 717, 2012 Mar 06.
Article in English | MEDLINE | ID: mdl-22395608

ABSTRACT

Megakaryocytes are unique mammalian cells that undergo polyploidization (endomitosis) during differentiation, leading to an increase in cell size and protein production that precedes platelet production. Recent evidence demonstrates that endomitosis is a consequence of a late failure in cytokinesis associated with a contractile ring defect. Here we show that the non-muscle myosin IIB heavy chain (MYH10) is expressed in immature megakaryocytes and specifically localizes in the contractile ring. MYH10 downmodulation by short hairpin RNA increases polyploidization by inhibiting the return of 4N cells to 2N, but other regulators, such as of the G1/S transition, might regulate further polyploidization of the 4N cells. Conversely, re-expression of MYH10 in the megakaryocytes prevents polyploidization and the transition of 2N to 4N cells. During polyploidization, MYH10 expression is repressed by the major megakaryocyte transcription factor RUNX1. Thus, RUNX1-mediated silencing of MYH10 is required for the switch from mitosis to endomitosis, linking polyploidization with megakaryocyte differentiation.


Subject(s)
Core Binding Factor Alpha 2 Subunit/metabolism , Megakaryocytes/cytology , Myosin Heavy Chains/genetics , Nonmuscle Myosin Type IIB/genetics , Polyploidy , Animals , Antigens, CD34/biosynthesis , Cell Line , Core Binding Factor Alpha 2 Subunit/genetics , Cytokinesis , Heterocyclic Compounds, 4 or More Rings/pharmacology , Humans , Megakaryocytes/metabolism , Mice , Mice, Knockout , Mitosis , Myosin Heavy Chains/biosynthesis , Myosin Heavy Chains/metabolism , Nonmuscle Myosin Type IIB/biosynthesis , Nonmuscle Myosin Type IIB/metabolism , RNA Interference , RNA, Small Interfering
12.
J Mol Med (Berl) ; 89(4): 411-21, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21301799

ABSTRACT

Granzyme B plays a key role in cell-mediated programmed cell death. We previously demonstrated that p53 is a functional determinant in the granzyme B-induced cytotoxic T-lymphocyte response. However, the pathways leading to activation of p53 by granzyme B remain incompletely understood. We now demonstrate that granzyme B-induced DNA damage signaling as revealed by histone H2AX phosphorylation and subsequent activation of the stress kinase CHK2. Confocal microscopy analysis indicates that granzyme B treatment of tumor cells induced an early translocation of endonuclease caspase-activated DNase. DNA microarray-based global transcriptional profiling and RT-PCR indeed revealed genes related to DNA damage. Among these genes, hSMG-1, a genotoxic stress-activated protein, was constantly upregulated in tumor cells following granzyme B treatment. Knockdown of the hSMG-1 gene in T1 tumor target cell line resulted in a significant inhibition of granzyme B- and CTL-induced killing. Our data suggest that granzyme B may exert cell death through DNA damage signaling and uncover a novel molecular link between the DNA damage pathway and granzyme B-induced cell death.


Subject(s)
Granzymes/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , T-Lymphocytes, Cytotoxic/drug effects , Cell Death/drug effects , Cell Line, Tumor , Cell Nucleus/enzymology , DNA Damage/drug effects , Deoxyribonucleases/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Granzymes/metabolism , Humans , Phosphatidylinositol 3-Kinases/genetics , Protein Serine-Threonine Kinases , Protein Transport/drug effects , RNA Interference , Stress, Physiological/drug effects , T-Lymphocytes, Cytotoxic/immunology
13.
Cancer Res ; 71(2): 328-38, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-21224355

ABSTRACT

Interaction of the integrin αE(CD103)ß7 expressed on tumor-infiltrating lymphocytes (TIL) with E-cadherin on epithelial tumor cells is required to trigger polarized exocytosis of cytotoxic granules in TIL that elicit tumor cell lysis. In this study, we investigated the functional and signaling properties of CD103 and its individual contribution to T-cell-mediated cancer-cell killing. Our results indicated that the binding of CD103 on tumor-specific CTL to immobilized recombinant E-cadherin-Fc is sufficient to induce the polarization of cytolytic granules, whereas the degranulation of cytolytic granules also requires the coengagement of the T-cell receptor. Moreover, minimal CD103 triggering promotes the phosphorylation of the ERK1/2 kinases and phospholipase Cγ1 (PLCγ1). Inhibiting PLCγ blocks granule relocalization, decreasing T-cell receptor-mediated cytotoxicity. Thus, our results emphasize a unique costimulatory role of CD103 in tumor-specific CTL activation by providing signals that promote T-cell effector functions needed to specifically target and lyse cancer cells.


Subject(s)
Antigens, CD/immunology , Cadherins/immunology , Immunoglobulin Fc Fragments/immunology , Integrin alpha Chains/immunology , Lung Neoplasms/immunology , Phospholipase C gamma/immunology , T-Lymphocytes, Cytotoxic/immunology , Antigens, CD/metabolism , Cadherins/chemistry , Cell Adhesion/immunology , Cell Line, Tumor , Cytoplasmic Granules/immunology , Cytoplasmic Granules/metabolism , Cytotoxicity, Immunologic , Humans , Immobilized Proteins/chemistry , Immobilized Proteins/immunology , Immunoglobulin Fc Fragments/chemistry , Integrin alpha Chains/metabolism , Lung Neoplasms/metabolism , Mitogen-Activated Protein Kinase 1/immunology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/immunology , Mitogen-Activated Protein Kinase 3/metabolism , Phosphatidylinositol 3-Kinases/immunology , Phosphatidylinositol 3-Kinases/metabolism , Phospholipase C gamma/antagonists & inhibitors , Phospholipase C gamma/metabolism , Phosphorylation , Protein Kinase C/immunology , Protein Kinase C/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/immunology , T-Lymphocytes, Cytotoxic/metabolism
14.
Blood ; 116(13): 2345-55, 2010 Sep 30.
Article in English | MEDLINE | ID: mdl-20548097

ABSTRACT

Polyploidization of megakaryocytes (MKs), the platelet precursors, occurs by endomitosis, a mitotic process that fails at late stages of cytokinesis. Expression and function of Aurora B kinase during endomitosis remain controversial. Here, we report that Aurora B is normally expressed during the human MK endomitotic process. Aurora B localized normally in the midzone or midbody during anaphase and telophase in low ploidy megakaryocytes and in up to 16N rare endomitotic MKs was observed. Aurora B was also functional during cytokinesis as attested by phosphorylation of both its activation site and MgcRacGAP, its main substrate. However, despite its activation, Aurora B did not prevent furrow regression. Inhibition of Aurora B by AZD1152-HQPA decreased cell cycle entry both in 2N to 4N and polyploid MKs and induced apoptosis mainly in 2N to 4N cells. In both MK classes, AZD1152-HQPA induced p53 activation and retinoblastoma hypophosphorylation. Resistance of polyploid MKs to apoptosis correlated to a high BclxL level. Aurora B inhibition did not impair MK polyploidization but profoundly modified the endomitotic process by inducing a mis-segregation of chromosomes and a mitotic failure in anaphase. This indicates that Aurora B is dispensable for MK polyploidization but is necessary to achieve a normal endomitotic process.


Subject(s)
Megakaryocytes/cytology , Megakaryocytes/enzymology , Mitosis/genetics , Mitosis/physiology , Polyploidy , Protein Serine-Threonine Kinases/physiology , Apoptosis/drug effects , Apoptosis/physiology , Aurora Kinase B , Aurora Kinases , Chromosome Segregation/drug effects , Chromosome Segregation/physiology , G1 Phase/drug effects , G1 Phase/physiology , Humans , In Vitro Techniques , Inhibitor of Apoptosis Proteins , Megakaryocytes/drug effects , Microtubule-Associated Proteins/metabolism , Mitosis/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , S Phase/drug effects , S Phase/physiology , Spindle Apparatus/enzymology , Survivin
15.
Cancer Res ; 69(15): 6249-55, 2009 Aug 01.
Article in English | MEDLINE | ID: mdl-19638592

ABSTRACT

We have reported previously that the interaction of alpha(E)(CD103)beta(7) integrin, expressed on a CD8(+) tumor-infiltrating lymphocyte (TIL) clone but not on a peripheral blood lymphocyte (PBL) counterpart, with the epithelial marker E-cadherin on human lung tumor cells plays a crucial role in T-cell receptor-mediated cytotoxicity. We show here that both TIL and PBL clones are able to migrate toward autologous tumor cells and that chemokine receptor CCR5 is involved in this process. Adoptive transfer of the PBL clone in the cognate tumor engrafted in nonobese diabetic/severe combined immunodeficient mice and subsequent coengagement of T-cell receptor and transforming growth factor-beta1 receptor triggers CD103 expression on T-cell surface resulting in strong potentiation of antitumor lytic function. Moreover, interaction of alpha(E)beta(7) integrin with E-cadherin, but not lymphocyte function-associated antigen-1 with intercellular adhesion molecule-1, promotes CCR5 recruitment at the immunologic synapse formed between TIL and tumor cells, leading to inhibition of T-cell sensitivity to CCL5 chemotactic gradient. These results provide evidence for a role of tumor microenvironment, namely MHC class I-restricted antigen presentation and transforming growth factor-beta1 secretion, in regulating the effector phase of tumor-specific CTL response. They also suggest a unique role of CD103 in T-cell retention at the tumor site by a CCR5-dependent mechanism.


Subject(s)
Antigens, CD/biosynthesis , Carcinoma, Non-Small-Cell Lung/immunology , Integrin alpha Chains/biosynthesis , Lung Neoplasms/immunology , Receptors, CCR5/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Antigens, CD/immunology , Cadherins/immunology , Carcinoma, Non-Small-Cell Lung/blood , Cell Line, Tumor , Cell Movement/immunology , Humans , Integrin alpha Chains/immunology , Intercellular Adhesion Molecule-1/immunology , Lung Neoplasms/blood , Lymphocyte Function-Associated Antigen-1/immunology , Lymphocytes/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Receptors, Antigen, T-Cell/biosynthesis , Receptors, Antigen, T-Cell/immunology , Receptors, CCR5/biosynthesis
16.
J Immunol ; 182(6): 3510-21, 2009 Mar 15.
Article in English | MEDLINE | ID: mdl-19265129

ABSTRACT

Hypoxia is an essential component of tumor microenvironment. In this study, we investigated the influence of hypoxia (1% PO(2)) on CTL-mediated tumor cell lysis. We demonstrate that exposure of target tumor cells to hypoxia has an inhibitory effect on the CTL clone (Heu171)-induced autologous target cell lysis. Such inhibition correlates with hypoxia-inducible factor-1alpha (HIF-1alpha) induction but is not associated with an alteration of CTL reactivity as revealed by granzyme B polarization or morphological change. Western blot analysis indicates that although hypoxia had no effect on p53 accumulation, it induced the phosphorylation of STAT3 in tumor cells by a mechanism at least in part involving vascular endothelial growth factor secretion. We additionally show that a simultaneous nuclear translocation of HIF-1alpha and phospho-STAT3 was observed. Interestingly, gene silencing of STAT3 by small interfering RNA resulted in HIF-1alpha inhibition and a significant restoration of target cell susceptibility to CTL-induced killing under hypoxic conditions by a mechanism involving at least in part down-regulation of AKT phosphorylation. Moreover, knockdown of HIF-1alpha resulted in the restoration of target cell lysis under hypoxic conditions. This was further supported by DNA microarray analysis where STAT3 inhibition resulted in a partly reversal of the hypoxia-induced gene expression profile. The present study demonstrates that the concomitant hypoxic induction of phospho-STAT3 and HIF-1alpha are functionally linked to the alteration of non-small cell lung carcinoma target susceptibility to CTL-mediated killing. Considering the eminent functions of STAT3 and HIF-1alpha in the tumor microenvironment, their targeting may represent novel strategies for immunotherapeutic intervention.


Subject(s)
Cytotoxicity, Immunologic , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Hypoxia/immunology , Lung Neoplasms/immunology , STAT3 Transcription Factor/biosynthesis , T-Lymphocytes, Cytotoxic/immunology , Cell Line, Tumor , Clone Cells , Gene Expression Regulation, Neoplastic/immunology , Humans , Hypoxia/metabolism , Hypoxia/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Immunity, Innate , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/physiology , T-Lymphocytes, Cytotoxic/metabolism , T-Lymphocytes, Cytotoxic/pathology
17.
J Immunol ; 182(5): 2654-64, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-19234159

ABSTRACT

Cellular interactions in the tumor stroma play a major role in cancer progression but can also induce tumor rejection. To explore the role of endothelial cells in these interactions, we used an in vitro three-dimensional collagen matrix model containing a cytotoxic T lymphocyte CTL clone (M4.48), autologous tumor cells (M4T), and an endothelial cell (M4E) line that are all derived from the same tumor. We demonstrate in this study that specific killing of the endothelial cells by the CTL clone required the autologous tumor cells and involved Ag cross-presentation. The formation of gap junctions between endothelial and tumor cells is required for antigenic peptide transfer to endothelial cells that are then recognized and eliminated by CTL. Our results indicate that gap junctions facilitate an effective CTL-mediated destruction of endothelial cells from the tumor microenvironment that may contribute to the control of tumor progression.


Subject(s)
Cell Communication/immunology , Cross-Priming/immunology , Endothelial Cells/immunology , Endothelial Cells/pathology , Gap Junctions/immunology , Melanoma/immunology , Melanoma/pathology , T-Lymphocytes, Cytotoxic/immunology , Antigens, Neoplasm/metabolism , Biomarkers/metabolism , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Clone Cells , Coculture Techniques , Cytosol/immunology , Cytosol/metabolism , Cytotoxicity, Immunologic/immunology , Endothelial Cells/metabolism , Gap Junctions/metabolism , Gap Junctions/pathology , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphocytes, Tumor-Infiltrating/pathology , Melanoma/therapy , T-Lymphocytes, Cytotoxic/metabolism , T-Lymphocytes, Cytotoxic/pathology
18.
Cancer Res ; 68(23): 9854-64, 2008 Dec 01.
Article in English | MEDLINE | ID: mdl-19047166

ABSTRACT

Human primary melanoma cells (T1) were found to be more susceptible to lysis by a Melan-A/MART-1-specific CTL clone (LT12) than their metastatic derivative (G1). We show that this differential susceptibility does not involve antigen presentation by target cells, synapse formation between the metastatic target and CTL clone, or subsequent granzyme B (GrB) polarization. Although PI-9, an inhibitor of GrB, was found to be overexpressed in metastatic G1 cells, knockdown of the PI-9 gene did not result in the attenuation of G1 resistance to CTL-induced killing. Interestingly, we show that whereas T1 cells express high levels of intercellular adhesion molecule-1 (ICAM-1), a dramatically reduced expression was noted on G1 cells. We also showed that sorted ICAM-1+ G1 cells were highly sensitive to CTL-induced lysis compared with ICAM-1- G1 cells. Furthermore, incubation of metastatic G1 cells with IFN-gamma resulted in the induction of ICAM-1 and the potentiation of their susceptibility to lysis by LT12. More importantly, we found that the level of ICAM-1 expression by melanoma cells correlated with decreased PTEN activity. ICAM-1 knockdown in T1 cells resulted in increased phosphorylation of PTEN and the subsequent activation of AKT. We have additionally shown that inhibition of the phosphatidylinositol (3,4,5)-triphosphate kinase (PI3K)/AKT pathway by the specific inhibitor wortmannin induced a significant potentiation of susceptibility of G1 and ICAM-1 small interfering RNA-treated T1 cells to CTL-induced lysis. The present study shows that a shift in ICAM-1 expression, which was associated with an activation of the PI3K/AKT pathway, can be used by metastatic melanoma cells to escape CTL-mediated killing.


Subject(s)
Intercellular Adhesion Molecule-1/immunology , Melanoma/immunology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , T-Lymphocytes, Cytotoxic/immunology , Antigen Presentation , Antigens, Neoplasm/immunology , Cell Line, Tumor , Down-Regulation , Enzyme Activation , G1 Phase/immunology , Humans , Intercellular Adhesion Molecule-1/biosynthesis , Intercellular Adhesion Molecule-1/genetics , Interferon-gamma/pharmacology , MART-1 Antigen , Melanoma/enzymology , Melanoma/secondary , Neoplasm Proteins/immunology , PTEN Phosphohydrolase/metabolism , RNA, Small Interfering/genetics , Serpins/biosynthesis , Serpins/immunology
19.
J Biol Chem ; 283(46): 31665-72, 2008 Nov 14.
Article in English | MEDLINE | ID: mdl-18779334

ABSTRACT

Tumor cells evade adaptive immunity by a variety of mechanisms, including selection of variants that are resistant to specific cytotoxic T lymphocyte (CTL) pressure. Recently, we have reported that the reorganization of the actin cytoskeleton can be used by tumor cells as a strategy to promote their resistance to CTL-mediated lysis. In this study, we further examined the functional features of a CTL-resistant tumor variant and investigated the relationship between cytoskeleton alteration, the acquisition of tumor resistance to CTL-induced cell death, Rho-GTPases, and focal adhesion kinase (FAK) pathways. Our data indicate that although the resistant cells do not display an increased migratory potential, an alteration of adhesion to the extracellular matrix was observed. When Rho-GTPases were activated in cells by the bacterial CNF1 (cytotoxic necrotizing factor 1), striking changes in the cell morphology, including actin cytoskeleton, focal adhesions, and membrane extensions, were observed. More importantly, such activation also resulted in a significant attenuation of resistance to CTL-induced cell death. Furthermore, we demonstrate that FAK signaling pathways were constitutively defective in the resistant cells. Silencing of FAK in the sensitive target cells resulted in the inhibition of immune synapse formation with specific CTLs and their subsequent lysis. Expression of the FAK mutant (Y397F) resulted in an inhibition of IGR-Heu cell adhesion and of their susceptibility to specific lysis. These results suggest that FAK activation plays a role in the control of tumor cell susceptibility to CTL-mediated lysis.


Subject(s)
Focal Adhesion Protein-Tyrosine Kinases/metabolism , Neoplasms/enzymology , Neoplasms/immunology , T-Lymphocytes, Cytotoxic/enzymology , T-Lymphocytes, Cytotoxic/immunology , rho GTP-Binding Proteins/metabolism , Cell Adhesion , Cell Line , Cell Shape , Enzyme Activation , Extracellular Matrix/metabolism , Focal Adhesion Protein-Tyrosine Kinases/genetics , Gene Expression Regulation, Enzymologic , Macrophages/enzymology , Mutation/genetics , Neoplasms/genetics , Protein Binding
20.
Blood ; 112(8): 3164-74, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18684864

ABSTRACT

Megakaryocyte (MK) is the naturally polyploid cell that gives rise to platelets. Polyploidization occurs by endomitosis, which was a process considered to be an incomplete mitosis aborted in anaphase. Here, we used time-lapse confocal video microscopy to visualize the endomitotic process of primary human megakaryocytes. Our results show that the switch from mitosis to endomitosis corresponds to a late failure of cytokinesis accompanied by a backward movement of the 2 daughter cells. No abnormality was observed in the central spindle of endomitotic MKs. A furrow formation was present, but the contractile ring was abnormal because accumulation of nonmuscle myosin IIA was lacking. In addition, a defect in cell elongation was observed in dipolar endomitotic MKs during telophase. RhoA and F-actin were partially concentrated at the site of furrowing. Inhibition of the Rho/Rock pathway caused the disappearance of F-actin at midzone and increased MK ploidy level. This inhibition was associated with a more pronounced defect in furrow formation as well as in spindle elongation. Our results suggest that the late failure of cytokinesis responsible for the endomitotic process is related to a partial defect in the Rho/Rock pathway activation.


Subject(s)
Megakaryocytes/cytology , Mitosis , rho-Associated Kinases/metabolism , Actins/metabolism , Antigens, CD34/biosynthesis , Cell Separation , Cytokinesis , Humans , Lentivirus/metabolism , Microscopy, Confocal , Microscopy, Video , Ploidies , Signal Transduction , Telophase
SELECTION OF CITATIONS
SEARCH DETAIL
...