Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Biochem Biophys Res Commun ; 577: 103-109, 2021 11 05.
Article in English | MEDLINE | ID: mdl-34509721

ABSTRACT

As essential phospholipid signaling regulators, phospholipase C (PLC)s are activated by various extracellular ligands and mediate intracellular signal transduction. PLCγ1 is involved in regulating various cancer cell functions. However, the precise in vivo link between PLCγ1 and cancer behavior remains undefined. To investigate the role of PLCγ1 in colorectal carcinogenesis, we generated an intestinal tissue-specific Plcg1 knock out (KO) in adenomatous polyposis coli (Apc) Min/+ mice. Plcg1 deficiency in ApcMin/+ mice showed earlier death, with a higher colorectal tumor incidence in both number and size than in wild-type mice. Mechanistically, inhibition of PLCγ1 increased the levels of its substrate phosphoinositol 4,5-bisphosphate (PIP2) at the plasma membrane and promoted the activation of Wnt receptor low-density lipoprotein receptor-related protein 6 (LRP6) by glycogen synthase kinase 3ß (GSK3ß) to enhance ß-catenin signaling. Enhanced cell proliferation and Wnt/ß-catenin signaling were observed in colon tumors from Plcg1 KO mice. Furthermore, low PLCγ1 expression was associated with a poor prognosis of colon cancer patients. Collectively, we demonstrated the role of PLCγ1 in vivo as a tumor suppressor relationship between the regulation of the PIP2 level and Wnt/ß-catenin-dependent intestinal tumor formation.


Subject(s)
Cell Proliferation/genetics , Colorectal Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Phospholipase C gamma/genetics , Wnt Signaling Pathway/genetics , beta Catenin/genetics , Animals , Cell Line, Tumor , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/pathology , Disease Progression , Glycogen Synthase Kinase 3 beta/genetics , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Intestines/enzymology , Intestines/pathology , Kaplan-Meier Estimate , Low Density Lipoprotein Receptor-Related Protein-6/genetics , Low Density Lipoprotein Receptor-Related Protein-6/metabolism , Mice, Inbred C57BL , Mice, Knockout , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phospholipase C gamma/deficiency , beta Catenin/metabolism
2.
Artif Cells Nanomed Biotechnol ; 47(1): 3786-3792, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31549850

ABSTRACT

Phospholipase Cγ2 (PLCG2) has been implicated in the regulation of cell proliferation, transformation, and tumor growth. In this study, we investigate the mechanism of PLCG2 action using a short interference RNA (siRNA) method. The effects of PLCG2 on rat liver BRL-3A cells treated siRNA were studied by 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide (MTT assay), bromodeoxyuridine (BrdU) labelling assay, flow cytometry method (FCM), quantitative real-time polymerase chain reaction (qRT-PCR) and western blot. The results showed when PLCG2 was reduced, cell vitality and proliferation rate were significantly decreased (p < .05 vs. control). FCM analysis showed that the number of cell division phase (G2 + M) was declined (p < .05 vs. control). RT-PCR and western blot revealed that the expression of signalling related genes NF-κB, FOS, JUN and ELK, target genes BCL2, CCNB1 and CCND1 were remarkably down-regulated in cells treated with PLCG2 siRNAs. Based on these results, we conclude PLCG2 plays an important role in rat liver cell proliferation via ERK and NF-κB pathway by regulating the expression of BCl2, MYC and CCND1.


Subject(s)
Cyclin D1/metabolism , Hepatocytes/cytology , MAP Kinase Signaling System , NF-kappa B/metabolism , Phospholipase C gamma/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Animals , Cell Proliferation , Phospholipase C gamma/deficiency , Phospholipase C gamma/genetics , RNA, Small Interfering/genetics , Rats
3.
Exp Dermatol ; 28(9): 1051-1057, 2019 09.
Article in English | MEDLINE | ID: mdl-31338881

ABSTRACT

Differentiation and proliferation of keratinocyte are controlled by various signalling pathways. The epidermal growth factor receptor (EGFR) is known to be an important regulator of multiple epidermal functions. Inhibition of EGFR signalling disturbs keratinocyte proliferation, differentiation and migration. Previous studies have revealed that one of the EGFR downstream signalling molecules, phospholipase Cγ1 (PLCγ1), regulates differentiation, proliferation and migration of keratinocytes in in vitro cell culture system. However, the role of PLCγ1 in the regulation of keratinocyte functions in animal epidermis remains unexplored. In this study, we generated keratinocyte-specific PLCγ1 knockout (KO) mice (PLCγ1 cKO mice). Contrary to our expectations, loss of PLCγ1 did not affect differentiation, proliferation and migration of interfollicular keratinocytes. We further examined the role of PLCγ1 in irritant contact dermatitis (ICD), in which epidermal cells play a pivotal role. Upon irritant stimulation, PLCγ1 cKO mice showed exaggerated ICD responses. Further study revealed that epidermal loss of PLCγ1 induced sebaceous gland hyperplasia, indicating that PLCγ1 regulates homeostasis of one of the epidermal appendages. Taken together, our results indicate that, although PLCγ1 is dispensable in interfollicular keratinocyte for normal differentiation, proliferation and migration, it is required for normal ICD responses. Our results also indicate that PLCγ1 regulates homeostasis of sebaceous glands.


Subject(s)
Dermatitis, Irritant/enzymology , Keratinocytes/enzymology , Phospholipase C gamma/physiology , Sebaceous Glands/enzymology , Animals , Cell Differentiation , Cell Movement , Cell Proliferation , Croton Oil/toxicity , Dermatitis, Irritant/etiology , Epidermis/drug effects , Epidermis/enzymology , Epidermis/pathology , Homeostasis , Hyperplasia , Irritants , Keratinocytes/drug effects , Mice , Mice, Knockout , Mice, Transgenic , Phospholipase C gamma/deficiency , Phospholipase C gamma/genetics , Sebaceous Glands/drug effects , Sebaceous Glands/pathology
4.
Nat Commun ; 8(1): 1457, 2017 11 13.
Article in English | MEDLINE | ID: mdl-29133930

ABSTRACT

The precise molecular mechanism underlying the regulation of early B cell lymphopoiesis is unclear. The PLCγ signaling pathway is critical for antigen receptor-mediated lymphocyte activation, but its function in cytokine signaling is unknown. Here we show that PLCγ1/PLCγ2 double deficiency in mice blocks early B cell development at the pre-pro-B cell stage and renders B cell progenitors unresponsive to IL-7. PLCγ pathway inhibition blocks IL-7-induced activation of mTOR, but not Stat5. The PLCγ pathway activates mTOR through the DAG/PKC signaling branch, independent of the conventional Akt/TSC/Rheb signaling axis. Inhibition of PLCγ/PKC-induced mTOR activation impairs IL-7-mediated B cell development. PLCγ1/PLCγ2 double-deficient B cell progenitors have reduced expression of genes related to B cell lineage, IL-7 signaling, and cell cycle. Thus, IL-7 receptor controls early B lymphopoiesis through activation of mTOR via PLCγ/DAG/PKC signaling, not via Akt/Rheb signaling.


Subject(s)
Interleukin-7/immunology , Lymphopoiesis/immunology , Phospholipase C gamma/deficiency , Phospholipase C gamma/immunology , Precursor Cells, B-Lymphoid/cytology , TOR Serine-Threonine Kinases/metabolism , Animals , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phospholipase C gamma/genetics , Precursor Cells, B-Lymphoid/immunology , STAT5 Transcription Factor/metabolism , Signal Transduction/immunology
5.
Sci Rep ; 7(1): 13912, 2017 10 24.
Article in English | MEDLINE | ID: mdl-29066806

ABSTRACT

Phosphoinositide-specific phospholipase C (PLC) γ1 has been reported to be involved in cancer cell proliferation and metastasis. However, whether PLCγ1 modulates autophagy and the underlying mechanism remains unclear. Here, we investigated the relationship between PLCγ1 and autophagy in the human colon cancer cell line HCT116 and hepatocellular carcinoma cell line HepG2. The results indicated that PLCγ1 inhibition via lentivirus-mediated transduction with shRNA/PLCγ1 or transient transfection with pRK5-PLCγ1 (Y783A) vector increased LC3B-II levels and the number of autophagic vacuoles and decreased p62 levels. Addition of an autophagy inhibitor led to LC3B and p62 accumulation. Furthermore, AMPK activation promoted the autophagy induced by PLCγ1 inhibition by blocking the FAK/PLCγ1 axis. In addition, PLCγ1 inhibition either blocked the mTOR/ULK1 axis or enhanced dissociation of the Beclin1-IP3R-Bcl-2 complex to induce autophagy. Taken together, our findings revealed that PLCγ1 inhibition induced autophagy and the FAK/PLCγ1 axis is a potential downstream effector of the AMPK activation-dependent autophagy signalling cascade. Both blockade of the mTOR/ULK1 axis and dissociation of the Beclin1-IP3R-Bcl-2 complex contributed to the induction of autophagy by PLCγ1 inhibition. Consequently, these findings provide novel insight into autophagy regulation by PLCγ1 in colon cancer and hepatocellular carcinoma cells.


Subject(s)
Autophagy/genetics , Carcinoma, Hepatocellular/pathology , Colonic Neoplasms/pathology , Enzyme Inhibitors/pharmacology , Liver Neoplasms/pathology , Phospholipase C gamma/deficiency , Phospholipase C gamma/genetics , AMP-Activated Protein Kinases/metabolism , HCT116 Cells , Hep G2 Cells , Humans , RNA, Small Interfering/genetics
6.
Mol Psychiatry ; 22(10): 1473-1482, 2017 10.
Article in English | MEDLINE | ID: mdl-28138157

ABSTRACT

Manic episodes are one of the major diagnostic symptoms in a spectrum of neuropsychiatric disorders that include schizophrenia, obsessive-compulsive disorder and bipolar disorder (BD). Despite a possible association between BD and the gene encoding phospholipase Cγ1 (PLCG1), its etiological basis remains unclear. Here, we report that mice lacking phospholipase Cγ1 (PLCγ1) in the forebrain (Plcg1f/f; CaMKII) exhibit hyperactivity, decreased anxiety-like behavior, reduced depressive-related behavior, hyperhedonia, hyperphagia, impaired learning and memory and exaggerated startle responses. Inhibitory transmission in hippocampal pyramidal neurons and striatal dopamine receptor D1-expressing neurons of Plcg1-deficient mice was significantly reduced. The decrease in inhibitory transmission is likely due to a reduced number of γ-aminobutyric acid (GABA)-ergic boutons, which may result from impaired localization and/or stabilization of postsynaptic CaMKII (Ca2+/calmodulin-dependent protein kinase II) at inhibitory synapses. Moreover, mutant mice display impaired brain-derived neurotrophic factor-tropomyosin receptor kinase B-dependent synaptic plasticity in the hippocampus, which could account for deficits of spatial memory. Lithium and valproate, the drugs presently used to treat mania associated with BD, rescued the hyperactive phenotypes of Plcg1f/f; CaMKII mice. These findings provide evidence that PLCγ1 is critical for synaptic function and plasticity and that the loss of PLCγ1 from the forebrain results in manic-like behavior.


Subject(s)
Bipolar Disorder/enzymology , Bipolar Disorder/genetics , Phospholipase C gamma/metabolism , Prosencephalon/enzymology , Animals , Bipolar Disorder/parasitology , Brain-Derived Neurotrophic Factor/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Hippocampus/enzymology , Hippocampus/metabolism , Mice , Neuronal Plasticity/physiology , Neurons/enzymology , Neurons/metabolism , Phospholipase C gamma/deficiency , Phospholipase C gamma/genetics , Prosencephalon/pathology , Pyramidal Cells/metabolism , Receptor, trkB/metabolism , Receptors, Dopamine D1 , Synapses/enzymology , Synapses/pathology , Synaptic Transmission/physiology , gamma-Aminobutyric Acid/metabolism
7.
Eur J Immunol ; 47(1): 74-83, 2017 01.
Article in English | MEDLINE | ID: mdl-27759161

ABSTRACT

Pre-T cell receptor (TCR) signaling is required for pre-T cell survival, proliferation, and differentiation from the CD4 and CD8 double negative (DN) to the double positive (DP) stage. However, the pre-TCR signal transduction pathway is not fully understood and the signaling molecules involved have not been completely identified. Phospholipase Cγ (PLCγ) 1 is an important signaling molecule that generates two second messengers, diacylglycerol and inositol 1,4,5-trisphosphate, that are important to mediate PKC activation and intracellular Ca2+ flux in many signaling pathways. Previously, we have shown that PLCγ1 is important for TCR-mediated signaling, development and T-cell activation, but the role of PLCγ1 in pre-TCR signal transduction and pre-T cell development is not known. In this study, we demonstrated that PLCγ1 expression level in pre-T cells was comparable to that in mature T cells. Deletion of PLCγ1 prior to the pre-TCR signaling stage partially blocked the DN3 to DN4 transition and reduced thymic cellularity. We also demonstrated that deletion of PLCγ1 impaired pre-T cell proliferation without affecting cell survival. Further study showed that deficiency of PLCγ1 impaired pre-TCR mediated Ca2+ flux and Erk activation. Thus our studies demonstrate that PLCγ1 is important for pre-TCR mediated signal transduction and pre-T cell development.


Subject(s)
Cell Differentiation , Phospholipase C gamma/metabolism , Precursor Cells, T-Lymphoid/cytology , Precursor Cells, T-Lymphoid/metabolism , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , Animals , Biomarkers , Calcium/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Proliferation , Cell Survival/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression , Genotype , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice , Mice, Transgenic , Phospholipase C gamma/deficiency , Phospholipase C gamma/genetics , Phosphorylation , Precursor Cells, T-Lymphoid/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Thymocytes/cytology , Thymocytes/immunology , Thymocytes/metabolism
8.
PLoS One ; 11(3): e0150521, 2016.
Article in English | MEDLINE | ID: mdl-26950550

ABSTRACT

Phospholipase Cγ2 (PLCγ2)-deficient mice exhibit misconnections of blood and lymphatic vessels, and male infertility. However, the cell type responsible for vascular partitioning and the mechanism for male infertility remain unknown. Accordingly, we generated a mouse line that conditionally expresses endogenous Plcg2 in a Cre/loxP recombination-dependent manner, and found that Tie2-Cre- or Pf4-Cre-driven reactivation of Plcg2 rescues PLCγ2-deficient mice from the vascular phenotype. By contrast, male mice rescued from the vascular phenotype exhibited epididymal sperm granulomas. As judged from immunostaining, PLCγ2 was expressed in clear cells in the epididymis. PLCγ2 deficiency did not compromise differentiation of epididymal epithelial cells, including clear cells, and tube formation at postnatal week 2. However, luminal expansion of the epididymal duct was impaired during the prepubertal period, regardless of epithelial cell polarity and tube architecture. These results suggest that PLCγ2-deficient clear cells cause impaired luminal expansion, stenosis of the epididymal duct, attenuation of luminal flow, and subsequent sperm granulomas. Clear cell-mediated luminal expansion is also supported by the observation that PLCγ2-deficient males were rescued from infertility by epididymal epithelium-specific reactivation of Plcg2, although the edematous and hemorrhagic phenotype associated with PLCγ2 deficiency also caused spontaneous epididymal sperm granulomas in aging males. Collectively, our findings demonstrate that PLCγ2 in clear cells plays an essential role in luminal expansion of the epididymis during the prepubertal period in mice, and reveal an unexpected link between PLCγ2, clear cells, and epididymal development.


Subject(s)
Epididymis/growth & development , Phospholipase C gamma/metabolism , Animals , Epididymis/blood supply , Epididymis/cytology , Gene Expression Regulation, Developmental , Genetic Engineering , Granuloma/enzymology , Hemorrhage/enzymology , Lymphatic Vessels/physiology , Male , Mice , Phospholipase C gamma/deficiency , Phospholipase C gamma/genetics , Recombination, Genetic
9.
PLoS One ; 10(3): e0119739, 2015.
Article in English | MEDLINE | ID: mdl-25793864

ABSTRACT

Receptor-mediated platelet activation requires phospholipase C (PLC) activity to elevate intracellular calcium and induce actin cytoskeleton reorganization. PLCs are classified into structurally distinct ß, γ, δ, ε, ζ, and η isoforms. There are two PLCγ isoforms (PLCγ1, PLCγ2), which are critical for activation by tyrosine kinase-dependent receptors. Platelets express both PLCγ1 and PLCγ2. Although PLCγ2 has been shown to play a dominant role in platelet activation, the extent to which PLCγ1 contributes has not been evaluated. To ascertain the relative contributions of PLCγ1 and PLCγ2 to platelet activation, we generated conditionally PLCγ1-deficient, wild-type (WT), PLCγ2-deficient, and PLCγ1/PLCγ2 double-deficient mice and measured the ability of platelets to respond to different agonists. We found that PLCγ2 deficiency abrogated αIIbß3-dependent platelet spreading, GPVI-dependent platelet aggregation, and thrombus formation on collagen-coated surfaces under shear conditions, which is dependent on both GPVI and αIIbß3. Addition of exogenous ADP overcame defective spreading of PLCγ2-deficient platelets on immobilized fibrinogen, suggesting that PLCγ2 is required for granule secretion in response to αIIbß3 ligation. Consistently, αIIbß3-mediated release of granule contents was impaired in the absence of PLCγ2. In contrast, PLCγ1-deficient platelets spread and released granule contents normally on fibrinogen, exhibited normal levels of GPVI-dependent aggregation, and formed thrombi normally on collagen-coated surfaces. Interestingly, enforced expression of PLCγ1 fully restored GPVI-dependent aggregation and αIIbß3-dependent spreading of PLCγ2-deficient platelets. We conclude that platelet activation through GPVI and αIIbß3 utilizes PLCγ2 because PLCγ1 levels are insufficient to support responsiveness, but that PLCγ1 can restore responsiveness if expressed at levels normally achieved by PLCγ2.


Subject(s)
Blood Platelets/metabolism , Gene Expression , Phospholipase C gamma/deficiency , Animals , Collagen/pharmacology , Enzyme Activation , Isoenzymes , Mice , Mice, Knockout , Mice, Transgenic , Phospholipase C gamma/genetics , Phospholipase C gamma/metabolism , Platelet Activation/genetics , Platelet Aggregation/drug effects , Platelet Aggregation/genetics , Platelet Count , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Thrombosis/metabolism
10.
J Exp Med ; 210(11): 2257-71, 2013 Oct 21.
Article in English | MEDLINE | ID: mdl-24127488

ABSTRACT

Myeloid-derived suppressor cells (MDSCs) favor tumor promotion, mainly by suppressing antitumor T cell responses in many cancers. Although the mechanism of T cell inhibition is established, the pathways leading to MDSC accumulation in bone marrow and secondary lymphoid organs of tumor-bearing hosts remain unclear. We demonstrate that down-regulation of PLCγ2 signaling in MDSCs is responsible for their aberrant expansion during tumor progression. PLCγ2(-/-) MDSCs show stronger immune-suppressive activity against CD8(+) T cells than WT MDSCs and potently promote tumor growth when adoptively transferred into WT mice. Mechanistically, PLCγ2(-/-) MDSCs display reduced ß-catenin levels, and restoration of ß-catenin expression decreases their expansion and tumor growth. Consistent with a negative role for ß-catenin in MDSCs, its deletion in the myeloid population leads to MDSC accumulation and supports tumor progression, whereas expression of ß-catenin constitutively active reduces MDSC numbers and protects from tumor growth. Further emphasizing the clinical relevance of these findings, MDSCs isolated from pancreatic cancer patients show reduced p-PLCγ2 and ß-catenin levels compared with healthy controls, similar to tumor-bearing mice. Thus, for the first time, we demonstrate that down-regulation of PLCγ2-ß-catenin pathway occurs in mice and humans and leads to MDSC-mediated tumor expansion, raising concerns about the efficacy of systemic ß-catenin blockade as anti-cancer therapy.


Subject(s)
Down-Regulation , Myeloid Cells/metabolism , Myeloid Cells/pathology , Neoplasms/metabolism , Neoplasms/pathology , Phospholipase C gamma/metabolism , beta Catenin/metabolism , Adoptive Transfer , Animals , Cell Differentiation , Cell Line, Tumor , Cell Proliferation , Disease Progression , Female , Humans , Immunosuppression Therapy , Mice , Mice, Inbred C57BL , Phospholipase C gamma/deficiency , Protein Stability , Signal Transduction
11.
J Immunol ; 189(5): 2326-32, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22837484

ABSTRACT

One of the important signaling events following TCR engagement is activation of phospholipase Cγ (PLCγ). PLCγ has two isoforms, PLCγ1 and PLCγ2. It is known that PLCγ1 is important for TCR signaling and TCR-mediated T cell selection and functions, whereas PLCγ2 is critical for BCR signal transduction and BCR-mediated B cell maturation and functions. In this study, we report that PLCγ2 was expressed in primary T cells, and became associated with linker for activated T cells and Src homology 2-domain containing leukocyte protein of 76 kDa and activated upon TCR stimulation. PLCγ1/PLCγ2 double-deficient T cells displayed further block from CD4 and CD8 double-positive to single-positive transition compared with PLCγ1 single-deficient T cells. TCR-mediated proliferation was further impaired in PLCγ1/PLCγ2 double-deficient T cells compared with PLCγ1 single-deficient T cells. TCR-mediated signal transduction, including Ca²âº mobilization and Erk activation, was further impaired in PLCγ1/PLCγ2 double-deficient relative to PLCγ1 single-deficient T cells. In addition, in HY TCR transgenic mouse model, thymic positive and negative selections were reduced in PLCγ1 heterozygous- and PLCγ2 homozygous-deficient (PLCγ1⁺/⁻PLCγ2⁻/⁻) relative to wild-type, PLCγ2 single-deficient (PLCγ2⁻/⁻), or PLCγ1 heterozygous-deficient (PLCγ1⁺/⁻) mice. Taken together, these data demonstrate that PLCγ2 participates in TCR signal transduction and plays a role in T cell selection.


Subject(s)
Phospholipase C gamma/physiology , Receptors, Antigen, T-Cell/physiology , Signal Transduction/immunology , Thymus Gland/enzymology , Thymus Gland/immunology , Animals , Cell Differentiation/genetics , Cell Differentiation/immunology , Cells, Cultured , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Phospholipase C gamma/deficiency , Phospholipase C gamma/genetics , Primary Cell Culture , Signal Transduction/genetics , Thymus Gland/cytology
12.
Cancer Res ; 71(14): 4799-808, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21602433

ABSTRACT

Blockade of osteoclast (OC) activity efficiently decreases tumor burden as well as associated bone erosion in immune-compromised animals bearing human osteolytic cancers. In this study, we showed that modulation of antitumor T-cell responses alters tumor growth in bone, regardless of OC status, by using genetic and pharmacologic models. PLCγ2(-/-) mice, with dysfunctional OCs and impaired dendritic cell (DC)-mediated T-cell activation, had increased bone tumor burden despite protection from bone loss. In contrast, Lyn(-/-) mice, with more numerous OCs and a hyperactive myeloid population leading to increased T-cell responses, had reduced tumor growth in bone despite enhanced osteolysis. The unexpected tumor/bone phenotype observed in PLCγ2(-/-) and Lyn(-/-) mice was transplantable, suggesting the involvement of an immune component. Consistent with this hypothesis, T-cell activation diminished skeletal metastasis whereas T-cell depletion enhanced it, even in the presence of zoledronic acid, a potent antiresorptive agent. Importantly, injection of antigen-specific wild-type cytotoxic CD8(+) T cells in PLCγ2(-/-) mice or CD8(+) T-cell depletion in Lyn(-/-) mice normalized tumor growth in bone. Our findings show the important contribution of CD8(+) T cells in the regulation of bone metastases regardless of OC status, thus including T cells as critical regulators of tumor growth in bone.


Subject(s)
Bone Neoplasms/immunology , Bone Resorption/immunology , CD8-Positive T-Lymphocytes/immunology , Osteoclasts/immunology , Animals , Bone Neoplasms/enzymology , Bone Neoplasms/pathology , Bone Neoplasms/therapy , Bone Resorption/enzymology , Bone Resorption/pathology , Cell Growth Processes/immunology , Female , Immunotherapy, Adoptive , Lymphocyte Activation , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Osteoclasts/enzymology , Osteoclasts/pathology , Phospholipase C gamma/deficiency , Phospholipase C gamma/immunology , src-Family Kinases/deficiency , src-Family Kinases/immunology
13.
Blood ; 116(5): 793-800, 2010 Aug 05.
Article in English | MEDLINE | ID: mdl-20457868

ABSTRACT

Migration of megakaryocytes (MKs) from the proliferative osteoblastic niche to the capillary-rich vascular niche is essential for proplatelet formation and platelet release. In this study, we explore the role of surface glycoprotein receptors and signaling proteins in regulating MK migration and platelet recovery after immune-induced thrombocytopenia. We show that spreading and migration of mouse primary bone marrow-derived MKs on a fibronectin matrix are abolished by the Src family kinases inhibitor PP1, the Syk kinase inhibitor R406 and the integrin alphaIIbbeta3 antagonist lotrafiban. We also demonstrate that these responses are inhibited in primary phospholipase C gamma2 (PLCgamma2)-deficient MKs. Conversely, MK spreading and migration were unaltered in the absence of the collagen receptor, the glycoprotein VI-FcRgamma-chain complex. We previously reported a correlation between a defect in MK migration and platelet recovery in the absence of platelet endothelial cell adhesion molecule-1 and the tyrosine phosphatase CD148. This correlation also holds for mice deficient in PLCgamma2. This study identifies a model in which integrin signaling via Src family kinases and Syk kinase to PLCgamma2 is required for MK spreading, migration, and platelet formation.


Subject(s)
Blood Platelets/cytology , Intracellular Signaling Peptides and Proteins/physiology , Megakaryocytes/cytology , Phospholipase C gamma/physiology , Protein-Tyrosine Kinases/physiology , Proto-Oncogene Proteins pp60(c-src)/physiology , Thrombopoiesis/physiology , Animals , Benzodiazepines/pharmacology , Cell Movement/drug effects , Cell Movement/physiology , Cell Shape , Cells, Cultured/cytology , Cells, Cultured/enzymology , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Megakaryocytes/enzymology , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxazines/pharmacology , Phospholipase C gamma/antagonists & inhibitors , Phospholipase C gamma/deficiency , Phosphorylation/drug effects , Piperidines/pharmacology , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , Platelet Glycoprotein GPIIb-IIIa Complex/physiology , Protein Kinase Inhibitors/pharmacology , Protein Processing, Post-Translational/drug effects , Protein-Tyrosine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins pp60(c-src)/antagonists & inhibitors , Purpura, Thrombocytopenic, Idiopathic/chemically induced , Purpura, Thrombocytopenic, Idiopathic/pathology , Pyrazoles/pharmacology , Pyridines/pharmacology , Pyrimidines/pharmacology , Signal Transduction/drug effects , Signal Transduction/physiology , Syk Kinase
14.
J Exp Med ; 207(2): 309-18, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-20123962

ABSTRACT

Phospholipase Cgamma1 (PLCgamma1) is an important signaling effector of T cell receptor (TCR). To investigate the role of PLCgamma1 in T cell biology, we generated and examined mice with T cell-specific deletion of PLCgamma1. We demonstrate that PLCgamma1 deficiency affects positive and negative selection, significantly reduces single-positive thymocytes and peripheral T cells, and impairs TCR-induced proliferation and cytokine production, and the activation of ERK, JNK, AP-1, NFAT, and NF-kappaB. Importantly, PLCgamma1 deficiency impairs the development and function of FoxP3(+) regulatory T cells, causing inflammatory/autoimmune symptoms. Therefore, PLCgamma1 is essential for T cell development, activation, and tolerance.


Subject(s)
Lymphocyte Activation , Phospholipase C gamma/immunology , T-Lymphocytes , Animals , Autoimmune Diseases/enzymology , Autoimmune Diseases/immunology , Autoimmunity , Cell Differentiation , Forkhead Transcription Factors/immunology , Forkhead Transcription Factors/metabolism , Inflammation/enzymology , Inflammation/immunology , MAP Kinase Kinase 4/immunology , MAP Kinase Kinase 4/metabolism , Mice , Mice, Transgenic , NF-kappa B/immunology , NF-kappa B/metabolism , NFATC Transcription Factors/immunology , NFATC Transcription Factors/metabolism , Phospholipase C gamma/deficiency , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocyte Subsets/enzymology , T-Lymphocyte Subsets/immunology , T-Lymphocytes/enzymology , T-Lymphocytes/immunology , Transcription Factor AP-1/immunology
15.
Blood ; 115(15): 3118-27, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20167705

ABSTRACT

Selectins mediate leukocyte rolling, trigger beta(2)-integrin activation, and promote leukocyte recruitment into inflamed tissue. E-selectin binding to P-selectin glycoprotein ligand 1 (PSGL-1) leads to activation of an immunoreceptor tyrosine-based activation motif (ITAM)-dependent pathway, which in turn activates the spleen tyrosine kinase (Syk). However, the signaling pathway linking Syk to integrin activation after E-selectin engagement is unknown. To identify the pathway, we used different gene-deficient mice in autoperfused flow chamber, intravital microscopy, peritonitis, and biochemical studies. We report here that the signaling pathway downstream of Syk divides into a phospholipase C (PLC) gamma2- and phosphoinositide 3-kinase (PI3K) gamma-dependent pathway. The Tec family kinase Bruton tyrosine kinase (Btk) is required for activating both pathways, generating inositol-3,4,5-trisphosphate (IP(3)), and inducing E-selectin-mediated slow rolling. Inhibition of this signal-transduction pathway diminished Galpha(i)-independent leukocyte adhesion to and transmigration through endothelial cells in inflamed postcapillary venules of the cremaster. Galpha(i)-independent neutrophil recruitment into the inflamed peritoneal cavity was reduced in Btk(-/-) and Plcg2(-/-) mice. Our data demonstrate the functional importance of this newly identified signaling pathway mediated by E-selectin engagement.


Subject(s)
CD18 Antigens/metabolism , E-Selectin/metabolism , Neutrophil Infiltration , Phosphatidylinositol 3-Kinases/metabolism , Phospholipase C gamma/metabolism , Protein-Tyrosine Kinases/metabolism , Signal Transduction , Agammaglobulinaemia Tyrosine Kinase , Animals , Cell Adhesion/drug effects , Chemokines/pharmacology , Enzyme Activation/drug effects , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Leukocyte Rolling/drug effects , Mice , Mice, Inbred C57BL , Neutrophils/cytology , Neutrophils/drug effects , Neutrophils/enzymology , Phospholipase C gamma/deficiency , Phosphorylation/drug effects , Protein-Tyrosine Kinases/deficiency , Signal Transduction/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
16.
Am J Physiol Cell Physiol ; 298(2): C274-82, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19923421

ABSTRACT

The Caenorhabditis elegans intestinal epithelium generates rhythmic inositol 1,4,5-trisphosphate (IP(3))-dependent Ca(2+) oscillations that control muscle contractions required for defecation. Two highly Ca(2+)-selective transient receptor potential (TRP) melastatin (TRPM) channels, GON-2 and GTL-1, function with PLCgamma in a common signaling pathway that regulates IP(3)-dependent intracellular Ca(2+) release. A second PLC, PLCbeta, is also required for IP(3)-dependent Ca(2+) oscillations, but functions in an independent signaling mechanism. PLCgamma generates IP(3) that regulates IP(3) receptor activity. We demonstrate here that PLCgamma via hydrolysis of phosphatidylinositol 4,5-bisphosphate (PIP(2)) also regulates GON-2/GTL-1 function. Knockdown of PLCgamma but not PLCbeta activity by RNA interference (RNAi) inhibits channel activity approximately 80%. Inhibition is fully reversed by agents that deplete PIP(2) levels. PIP(2) added to the patch pipette has no effect on channel activity in PLCgamma RNAi cells. However, in control cells, 10 microM PIP(2) inhibits whole cell current approximately 80%. Channel inhibition by phospholipids is selective for PIP(2) with an IC(50) value of 2.6 microM. Elevated PIP(2) levels have no effect on channel voltage and Ca(2+) sensitivity and likely inhibit by reducing channel open probability, single-channel conductance, and/or trafficking. We conclude that hydrolysis of PIP(2) by PLCgamma functions in the activation of both the IP(3) receptor and GON-2/GTL-1 channels. GON-2/GTL-1 functions as the major intestinal cell Ca(2+) influx pathway. Calcium influx through the channel feedback regulates its activity and likely functions to modulate IP(3) receptor function. PIP(2)-dependent regulation of GON-2/GTL-1 may provide a mechanism to coordinate plasma membrane Ca(2+) influx with PLCgamma and IP(3) receptor activity as well as intracellular Ca(2+) store depletion.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Calcium Signaling , Intestinal Mucosa/enzymology , Ion Channels/metabolism , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phospholipase C gamma/deficiency , Animals , Binding Sites , Caenorhabditis elegans , Caenorhabditis elegans Proteins/antagonists & inhibitors , Caenorhabditis elegans Proteins/genetics , Feedback, Physiological , GATA Transcription Factors/genetics , GATA Transcription Factors/metabolism , Hydrolysis , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Ion Channel Gating , Ion Channels/antagonists & inhibitors , Ion Channels/genetics , Kinetics , Membrane Potentials , Periodicity , Phospholipase C beta/genetics , Phospholipase C beta/metabolism , Phospholipase C gamma/genetics , Phospholipids/metabolism , Protein Transport , RNA Interference , Recombinant Fusion Proteins/metabolism
17.
Mol Brain ; 2: 16, 2009 Jun 08.
Article in English | MEDLINE | ID: mdl-19505325

ABSTRACT

BACKGROUND: Neural stem cells (NSCs) are present in the adult mammalian brain and sustain life-long adult neurogenesis in the dentate gyrus of the hippocampus. In culture, fibroblast growth factor-2 (FGF-2) is sufficient to maintain the self-renewal of adult NSCs derived from the adult rat hippocampus. The underlying signalling mechanism is not fully understood. RESULTS: In the established adult rat NSC culture, FGF-2 promotes self-renewal by increasing proliferation and inhibiting spontaneous differentiation of adult NSCs, accompanied with activation of MAPK and PLC pathways. Using a molecular genetic approach, we demonstrate that activation of FGF receptor 1 (FGFR1), largely through two key cytoplasmic amino acid residues that are linked to MAPK and PLC activation, suffices to promote adult NSC self-renewal. The canonical MAPK, Erk1/2 activation, is both required and sufficient for the NSC expansion and anti-differentiation effects of FGF-2. In contrast, PLC activation is integral to the maintenance of adult NSC characteristics, including the full capacity for neuronal and oligodendroglial differentiation. CONCLUSION: These studies reveal two amino acid residues in FGFR1 with linked downstream intracellular signal transduction pathways that are essential for maintaining adult NSC self-renewal. The findings provide novel insights into the molecular mechanism regulating adult NSC self-renewal, and pose implications for using these cells in potential therapeutic applications.


Subject(s)
Adult Stem Cells/cytology , Extracellular Signal-Regulated MAP Kinases/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/enzymology , Phospholipase C gamma/metabolism , Receptor, Fibroblast Growth Factor, Type 1/genetics , Signal Transduction/genetics , Animals , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Clone Cells , Enzyme Activation/drug effects , Fibroblast Growth Factor 2/pharmacology , Humans , Intracellular Space/drug effects , Intracellular Space/metabolism , Mice , Models, Biological , Mutation/genetics , Neural Stem Cells/drug effects , Oligodendroglia/cytology , Oligodendroglia/drug effects , Phospholipase C gamma/deficiency , Rats , Rats, Inbred F344 , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptor, trkA/metabolism , Signal Transduction/drug effects
18.
J Exp Med ; 206(3): 577-93, 2009 Mar 16.
Article in English | MEDLINE | ID: mdl-19273622

ABSTRACT

beta(2) integrins and Fcgamma receptors are critically involved in neutrophil activation at the site of inflammation. Both receptor types trigger a receptor-proximal tyrosine phosphorylation cascade through Src family kinases and Syk, but further downstream signaling events are poorly understood. We show that phospholipase C (PLC) gamma2 is phosphorylated downstream of Src family kinases and Syk during integrin or Fc receptor-mediated activation of neutrophils. PLCgamma2(-/-) neutrophils are completely defective in beta(2) integrin or Fcgamma receptor-mediated functional responses such as respiratory burst, degranulation, or cell spreading in vitro and show reduced adhesion/spreading in inflamed capillary venules in vivo. However, PLCgamma2(-/-) neutrophils respond normally to various other agonists, including chemokines, bacterial formyl peptides, Toll-like receptor ligands, or proinflammatory cytokines, and migrate normally both in vitro and in vivo. To confirm the in vivo relevance of these observations, the effect of the PLCgamma2(-/-) mutation was tested in the K/BxN serum transfer arthritis model, which is known to require beta(2) integrins, Fcgamma receptors, and neutrophils. PLCgamma2 deficiency completely protected mice from clinical signs and histological features of arthritis as well as from arthritis-induced loss of articular function. These results identify PLCgamma2 as a critical player of integrin and Fc receptor-mediated neutrophil functions and the neutrophil-mediated effector phase of autoimmune arthritis.


Subject(s)
Arthritis/enzymology , Autoimmune Diseases/enzymology , CD18 Antigens/metabolism , Neutrophils/cytology , Neutrophils/enzymology , Phospholipase C gamma/metabolism , Receptors, IgG/metabolism , Animals , Arthritis/complications , Arthritis/pathology , Arthritis/prevention & control , Autoimmune Diseases/complications , Autoimmune Diseases/pathology , Autoimmune Diseases/prevention & control , Biomarkers/metabolism , Bone Marrow/drug effects , Bone Marrow/enzymology , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Movement/drug effects , Chimera , Endothelial Cells/cytology , Endothelial Cells/drug effects , Intracellular Signaling Peptides and Proteins/metabolism , Isoenzymes/metabolism , Leukocytes/cytology , Leukocytes/drug effects , Mice , Muscles/drug effects , Muscles/metabolism , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Neutrophils/drug effects , Phospholipase C gamma/deficiency , Phosphorylation/drug effects , Protein-Tyrosine Kinases/metabolism , Receptors, Cytokine/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects , Syk Kinase , src-Family Kinases/metabolism
19.
J Exp Med ; 206(3): 681-9, 2009 Mar 16.
Article in English | MEDLINE | ID: mdl-19273623

ABSTRACT

Resting antigen-experienced memory B cells are thought to be responsible for the more rapid and robust antibody responses after antigen reencounter, which are the hallmark of memory humoral responses. The molecular basis for the development and survival of memory B cells remains largely unknown. We report that phospholipase C (PLC) gamma2 is required for efficient formation of germinal center (GC) and memory B cells. Moreover, memory B cell homeostasis is severely hampered by inducible loss of PLC-gamma2. Accordingly, mice with a conditional deletion of PLC-gamma2 in post-GC B cells had an almost complete abrogation of the secondary antibody response. Collectively, our data suggest that PLC-gamma2 conveys a survival signal to GC and memory B cells and that this signal is required for a productive secondary immune response.


Subject(s)
B-Lymphocytes/enzymology , B-Lymphocytes/immunology , Immunologic Memory/immunology , Phospholipase C gamma/metabolism , Animals , B-Lymphocytes/cytology , B-Lymphocytes/drug effects , Cell Cycle/drug effects , Cell Survival/drug effects , Gene Deletion , Germinal Center/cytology , Germinal Center/drug effects , Germinal Center/enzymology , Germinal Center/immunology , Immunization , Immunoglobulin G/immunology , Immunologic Memory/drug effects , Mice , Mice, Knockout , Nitrophenols/pharmacology , Phenylacetates/pharmacology , Phospholipase C gamma/deficiency
20.
Development ; 136(2): 191-5, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19056831

ABSTRACT

The lymphatic vasculature originates from the blood vasculature through a mechanism relying on Prox1 expression and VEGFC signalling, and is separated and kept separate from the blood vasculature in a Syk- and SLP76-dependent manner. However, the mechanism by which lymphatic vessels are separated from blood vessels is not known. To gain an understanding of the vascular partitioning, we searched for the affected gene in a spontaneous mouse mutant exhibiting blood-filled lymphatic vessels, and identified a null mutation of the Plcg2 gene, which encodes phospholipase Cgamma2 (PLCgamma2), by positional candidate cloning. The blood-lymph shunt observed in PLCgamma2-null mice was due to aberrant separation of blood and lymphatic vessels. A similar phenotype was observed in lethally irradiated wild-type mice reconstituted with PLCgamma2-null bone marrow cells. These findings indicate that PLCgamma2 plays an essential role in initiating and maintaining the separation of the blood and lymphatic vasculature.


Subject(s)
Blood Vessels/embryology , Blood Vessels/enzymology , Lymphatic Vessels/embryology , Lymphatic Vessels/enzymology , Phospholipase C gamma/physiology , Animals , Base Sequence , Blood Vessels/abnormalities , Bone Marrow Transplantation , Cells, Cultured , DNA Primers/genetics , Endothelial Cells/enzymology , Female , Humans , Lymphatic Vessels/abnormalities , Mice , Mice, Knockout , Mice, Mutant Strains , Mice, Transgenic , Phospholipase C gamma/deficiency , Phospholipase C gamma/genetics , Polymorphism, Genetic , Pregnancy , Radiation Chimera
SELECTION OF CITATIONS
SEARCH DETAIL
...