ABSTRACT
Forgetting has been thought to occur as a result of the natural decay of the neuronal changes induced by learning or because of interference from other cognitive functions. In this issue, Shuai et al. (2010) find that the small G protein Rac may function as a switch for remembering versus forgetting.
Subject(s)
Drosophila/physiology , rac GTP-Binding Proteins/physiology , Animals , Drosophila Proteins/physiology , Memory/physiologyABSTRACT
Initially acquired memory dissipates rapidly if not consolidated. Such memory decay is thought to result either from the inherently labile nature of newly acquired memories or from interference by subsequently attained information. Here we report that a small G protein Rac-dependent forgetting mechanism contributes to both passive memory decay and interference-induced forgetting in Drosophila. Inhibition of Rac activity leads to slower decay of early memory, extending it from a few hours to more than one day, and to blockade of interference-induced forgetting. Conversely, elevated Rac activity in mushroom body neurons accelerates memory decay. This forgetting mechanism does not affect memory acquisition and is independent of Rutabaga adenylyl cyclase-mediated memory formation mechanisms. Endogenous Rac activation is evoked on different time scales during gradual memory loss in passive decay and during acute memory removal in reversal learning. We suggest that Rac's role in actin cytoskeleton remodeling may contribute to memory erasure.
Subject(s)
Drosophila Proteins/physiology , Drosophila/physiology , rac GTP-Binding Proteins/physiology , Actin Depolymerizing Factors/genetics , Animals , Memory/physiology , Memory Disorders , Mushroom BodiesABSTRACT
Developmental programs often rely on parallel morphogenetic mechanisms that guarantee precise tissue architecture. While redundancy constitutes an obvious selective advantage, little is known on how novel morphogenetic mechanisms emerge during evolution. In zebrafish, rhombomeric boundaries behave as an elastic barrier, preventing cell intermingling between adjacent compartments. Here, we identify the fundamental role of the small-GTPase Rac3b in actomyosin cable assembly at hindbrain boundaries. We show that the novel rac3b/rfng/sgca regulatory cluster, which is specifically expressed at the boundaries, emerged in the Ostariophysi superorder by chromosomal rearrangement that generated new cis-regulatory interactions. By combining 4C-seq, ATAC-seq, transgenesis, and CRISPR-induced deletions, we characterized this regulatory domain, identifying hindbrain boundary-specific cis-regulatory elements. Our results suggest that the capacity of boundaries to act as an elastic mesh for segregating rhombomeric cells evolved by cooption of critical genes to a novel regulatory block, refining the mechanisms for hindbrain segmentation.
Subject(s)
Actomyosin/physiology , Gene Expression Regulation, Developmental , Rhombencephalon/embryology , Sarcoglycans/physiology , Zebrafish Proteins/physiology , Zebrafish/embryology , rac GTP-Binding Proteins/physiology , Animals , Body Patterning/genetics , CRISPR-Cas Systems , Cell Movement , Characidae/genetics , Characidae/physiology , Chromatin/genetics , Chromatin/ultrastructure , Evolution, Molecular , Fishes/classification , Fishes/genetics , Morphogenesis , Mutagenesis, Site-Directed , Neurogenesis , Phylogeny , Sarcoglycans/genetics , Species Specificity , Zebrafish/genetics , Zebrafish Proteins/genetics , rac GTP-Binding Proteins/geneticsABSTRACT
Rac signaling impacts a relatively large number of downstream targets; however, few studies have established an association between Rac pathways and pathological conditions. In the present study, we generated mice with double knockout of Rac1 and Rac3 (Atoh1-Cre;Rac1flox/flox;Rac3-/- ) in cerebellar granule neurons (CGNs). We observed impaired tangential migration at E16.5, as well as numerous apoptotic CGNs at the deepest layer of the external granule layer (EGL) in the medial cerebellum of Atoh1-Cre;Rac1flox/flox;Rac3-/- mice at P8. Atoh1-Cre;Rac1flox/flox;Rac3-/- CGNs differentiated normally until expression of p27kip1 and NeuN in the deep EGL at P5. Primary CGNs and cerebellar microexplants from Atoh1-Cre;Rac1flox/flox;Rac3-/- mice exhibited impaired neuritogenesis, which was more apparent in Map2-positive dendrites. Such findings suggest that impaired tangential migration and final differentiation of CGNs have resulted in decreased cerebellum size and agenesis of the medial internal granule layer, respectively. Furthermore, Rac depleted/deleted cells exhibited decreased levels of Mid1 and impaired mTORC1 signaling. Mid1 depletion in CGNs produced mild impairments in neuritogenesis and reductions in mTORC1 signaling. Thus, a novel Rac-signaling pathway (Rac1-Mid1-mTORC1) may be involved in medial cerebellar development.
Subject(s)
Cerebellum/embryology , Proteins/physiology , rac GTP-Binding Proteins/physiology , Animals , Cell Differentiation/genetics , Cells, Cultured , Cerebellum/metabolism , HEK293 Cells , Humans , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Knockout , Multiprotein Complexes/physiology , Neurogenesis/genetics , Organogenesis/genetics , Proteins/genetics , Signal Transduction/genetics , TOR Serine-Threonine Kinases/physiology , Ubiquitin-Protein Ligases , rac GTP-Binding Proteins/geneticsABSTRACT
Rac proteins are involved in a variety of cellular processes. Effector proteins that interact with active Rac convey the GTPase-generated signal to downstream developmental cascades and processes. Here we report on the analysis of the main effector and signal cascade downstream of BcRac, the Rac homolog of the grey mold fungus Botrytis cinerea. Several lines of evidence highlighted the p21-activated kinase Cla4 as an important effector of Rac in fungi. Analysis of Δbccla4 strains revealed that the BcCla4 protein was sufficient to mediate all of the examined BcRac-driven processes, including hyphal growth and morphogenesis, conidia production and pathogenicity. In addition, the Δbccla4 strains had altered nuclei content, a phenomenon that was previously observed in Δbcrac isolates, thus connecting the BcRac/BcCla4 module with cell cycle control. Further analyses revealed that BcRac/BcCla4 control mitotic entry through changes in phosphorylation status of the cyclin dependent kinase BcCdk1. The complete cascade includes the kinase BcWee1, which is downstream of BcCla4 and upstream of BcCdk1. These results provide a mechanistic insight on the connection of cell cycle, morphogenesis and pathogenicity in fungi, and position BcCla4 as the most essential effector and central regulator of all of these processes downstream of BcRac.
Subject(s)
Botrytis/physiology , Protein Serine-Threonine Kinases/physiology , rac GTP-Binding Proteins/physiology , Botrytis/enzymology , Botrytis/growth & development , Botrytis/metabolism , Cell Cycle/physiology , Cell Division/physiology , Fungal Proteins/metabolism , Morphogenesis , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Spores, Fungal/metabolism , p21-Activated Kinases/metabolism , rac GTP-Binding Proteins/metabolismABSTRACT
Rac-GTPases and their Rac-GEF activators play important roles in the recruitment and host defence functions of neutrophils. These proteins control the activation of adhesion molecules and the cytoskeletal dynamics that enable the adhesion, migration and tissue recruitment of neutrophils. They also regulate the effector functions that allow neutrophils to kill bacterial and fungal pathogens, and to clear debris. This review focuses on the roles of Rac-GTPases and Rac-GEFs in neutrophil adhesion, migration and recruitment.
Subject(s)
Neutrophils/physiology , Rho Guanine Nucleotide Exchange Factors/physiology , rac1 GTP-Binding Protein/physiology , Cell Adhesion/physiology , Cell Movement/physiology , Guanine Nucleotide Exchange Factors/physiology , Humans , Neutrophil Infiltration/physiology , Neutrophils/enzymology , Proto-Oncogene Proteins c-vav/physiology , Signal Transduction/physiology , T-Lymphoma Invasion and Metastasis-inducing Protein 1/physiology , rac GTP-Binding Proteins/physiologyABSTRACT
NET formation in mice (NETosis) is supported by reactive oxygen species (ROS) production by NADPH oxidase and histone hypercitrullination by peptidylarginine deiminase 4 (PAD4). Rac1 and Rac2, expressed in polymorphonuclear neutrophils (PMNs), regulate the cytoskeleton, cell shape, adhesion, and migration and are also essential components of the NADPH oxidase complex. We aimed to explore the role of the Rac signaling pathway including the upstream guanosine exchange factor (GEF) activator, Vav, and a downstream effector, the p21-activated kinase, Pak, on NETosis in PMNs using a previously described flow-cytometry-based assay. Rac2-/- PMNs showed reduced levels of citrullinated histone H3 (H3Cit)-positive cells and defective NETosis. Rac1Δ/Δ ; Rac2-/- PMNs demonstrated a further reduction in PMA-induced H3Cit levels and a more profound impairment of NETosis than deletion of Rac2 alone, suggesting an overlapping role of these two highly related proteins. Genetic knockouts of Vav1, or Vav2, did not impair H3Cit response to phorbol myristate ester (PMA) or NETosis. Combined, Vav1 and Vav3 deletions decreased H3Cit response and caused a modest but significant impairment of NETosis. Pharmacologic inhibition of Pak by two inhibitors with distinct mechanisms of action, led to reduced H3Cit levels after PMA stimulation, as well as significant inhibition of NETosis. We validated the importance of Pak using Pak2Δ/Δ PMNs, which demonstrated significantly impaired histone H3 citrullination and NETosis. These data confirm and more comprehensively define the key role of the Rac signaling pathway in PMN NETosis. The Rac signaling cascade may represent a valuable target for inhibition of NETosis and related pathological processes.
Subject(s)
Extracellular Traps/metabolism , Signal Transduction , p21-Activated Kinases/physiology , rac GTP-Binding Proteins/physiology , Animals , Citrullination , Histones/metabolism , Mice , NADPH Oxidases/metabolism , Reactive Oxygen Species/metabolism , p21-Activated Kinases/metabolism , rac GTP-Binding Proteins/metabolismABSTRACT
Small GTPases in the Rho family act as major nodes with functions beyond cytoskeletal rearrangements shaping the Caenorhabditis elegans embryo during development. These small GTPases are key signal transducers that integrate diverse developmental signals to produce a coordinated response in the cell. In C. elegans, the best studied members of these highly conserved Rho family small GTPases, RHO-1/RhoA, CED-10/Rac, and CDC-42, are crucial in several cellular processes dealing with cytoskeletal reorganization. In this review, we update the functions described for the Rho family small GTPases in spindle orientation and cell division, engulfment, and cellular movements during C. elegans embryogenesis, focusing on the Rho subfamily Rac. Please also see the video abstract here.
Subject(s)
Caenorhabditis elegans Proteins/physiology , Caenorhabditis elegans/embryology , Cell Cycle Proteins/physiology , Cytoskeleton/metabolism , GTP-Binding Proteins/physiology , rac GTP-Binding Proteins/physiology , rho GTP-Binding Proteins/physiology , Animals , Caenorhabditis elegans/metabolism , Signal TransductionABSTRACT
Nervous system injury or disease leads to activation of glia, which govern postinjury responses in the nervous system. Axonal injury in Drosophila results in transcriptional up-regulation of the glial engulfment receptor Draper; there is extension of glial membranes to the injury site (termed activation), and then axonal debris is internalized and degraded. Loss of the small GTPase Rac1 from glia completely suppresses glial responses to injury, but upstream activators remain poorly defined. Loss of the Rac guanine nucleotide exchange factor (GEF) Crk/myoblast city (Mbc)/dCed-12 has no effect on glial activation, but blocks internalization and degradation of debris. Here we show that the signaling molecules downstream of receptor kinase (DRK) and daughter of sevenless (DOS) (mammalian homologs, Grb2 and Gab2, respectively) and the GEF son of sevenless (SOS) (mammalian homolog, mSOS) are required for efficient activation of glia after axotomy and internalization/degradation of axonal debris. At the earliest steps of glial activation, DRK/DOS/SOS function in a partially redundant manner with Crk/Mbc/dCed-12, with blockade of both complexes strongly suppressing all glial responses, similar to loss of Rac1. This work identifies DRK/DOS/SOS as the upstream Rac GEF complex required for glial responses to axonal injury, and demonstrates a critical requirement for multiple GEFs in efficient glial activation after injury and internalization/degradation of axonal debris.
Subject(s)
Drosophila Proteins/physiology , Drosophila melanogaster/cytology , Drosophila melanogaster/physiology , Eye Proteins/physiology , Neuroglia/physiology , Son of Sevenless Protein, Drosophila/physiology , rac GTP-Binding Proteins/physiology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/physiology , Animals , Animals, Genetically Modified , Axons/physiology , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/physiology , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Eye Proteins/genetics , Genes, Insect , Mutation , Nerve Degeneration , Phagosomes/physiology , Proto-Oncogene Proteins c-crk/genetics , Proto-Oncogene Proteins c-crk/physiology , Son of Sevenless Protein, Drosophila/genetics , rac GTP-Binding Proteins/genetics , ras Proteins/genetics , ras Proteins/physiologyABSTRACT
Lung cancer is still the leading cause of malignant deaths in the world. It is of great importance to find novel functional genes for the tumorigenesis of lung cancer. We demonstrated that Rac3 could promote cell proliferation and inhibit apoptosis in lung adenocarcinoma cell line A549 previously. The aim of this study was to investigate the function and mechanism of Rac3 in lung adenocarcinoma cell lines. Immunohistochemistry staining was performed in 107 lung adenocarcinoma tissues and matched non-tumor tissues. Multivariate analysis and Kaplan-Meier analysis were used to investigate the correlation between Rac3 expression and the clinical outcomes. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, colony formation assay, and flow cytometry analysis were employed to determine the proliferative ability, cell cycle distribution, and apoptosis in H1299 and H1975 cell lines. Gene expression microarray and pathway analysis between the Rac3-siRNA group and the control group in A549 cells were performed to investigate the pathways and mechanism of Rac3 regulation. Rac3 was shown to be positively expressed in lung adenocarcinoma tissues, and the expression of Rac3 associates with longer survival in lung adenocarcinoma patients. Silencing of Rac3 significantly induced cell growth inhibition, colony formation decrease, cell cycle arrest, and apoptosis of lung adenocarcinoma cell lines, which accompanied by obvious downregulation of CCND1, MYC, and TFDP1 of cell cycle pathway involving in the tumorigenesis of lung adenocarcinoma based on the gene expression microarray. In conclusion, these findings suggest that Rac3 has the potential of being a therapeutic target for lung adenocarcinoma.
Subject(s)
Adenocarcinoma/pathology , Lung Neoplasms/pathology , rac GTP-Binding Proteins/physiology , Adenocarcinoma/mortality , Adenocarcinoma of Lung , Adult , Aged , Apoptosis , Cell Cycle Checkpoints , Cell Proliferation , Cyclin D1/genetics , Female , Humans , Lung Neoplasms/mortality , Male , Middle Aged , Prognosis , Proto-Oncogene Proteins c-myc/genetics , Transcription Factor DP1/genetics , rac GTP-Binding Proteins/analysisABSTRACT
During chemotaxis, cells sense extracellular chemical gradients and position Ras GTPase activation and phosphatidylinositol (3,4,5)-triphosphate (PIP3) production toward chemoattractants. These two major signaling events are visualized by biosensors in a crescent-like zone at the plasma membrane. Here, we show that a Dictyostelium Rho GTPase, RacE, and a guanine nucleotide exchange factor, GxcT, stabilize the orientation of Ras activation and PIP3 production in response to chemoattractant gradients, and this regulation occurred independently of the actin cytoskeleton and cell polarity. Cells lacking RacE or GxcT fail to persistently direct Ras activation and PIP3 production toward chemoattractants, leading to lateral pseudopod extension and impaired chemotaxis. Constitutively active forms of RacE and human RhoA are located on the portion of the plasma membrane that faces lower concentrations of chemoattractants, opposite of PIP3 production. Mechanisms that control the localization of the constitutively active form of RacE require its effector domain, but not PIP3. Our findings reveal a critical role for Rho GTPases in positioning Ras activation and thereby establishing the accuracy of directional sensing.
Subject(s)
Chemotactic Factors/metabolism , Chemotaxis/physiology , Phosphatidylinositol Phosphates/metabolism , Signal Transduction/physiology , rac GTP-Binding Proteins/metabolism , Blotting, Southern , Gene Knockout Techniques , Guanine Nucleotide Exchange Factors/metabolism , Microscopy, Confocal , rac GTP-Binding Proteins/physiologyABSTRACT
The p21-activated kinases (Paks) are serine/threonine kinases that are major effectors of the Rho guanosine 5'\x{2011}triphosphatase, Rac, and Cdc42. Rac and Cdc42 are known regulators of hematopoietic stem and progenitor cell (HSPC) function, however, a direct role for Paks in HSPCs has yet to be elucidated. Lin(-)Sca1(+)c-kit(+) (LSK) cells from wild-type mice were transduced with retrovirus expressing Pak inhibitory domain (PID), a well-characterized inhibitor of Pak activation. Defects in marrow homing and in vitro cell migration, assembly of the actin cytoskeleton, proliferation, and survival were associated with engraftment failure of PID-LSK. The PID-LSK demonstrated decreased phosphorylation of extracellular signal-regulated kinase (ERK), whereas constitutive activation of ERK in these cells led to rescue of hematopoietic progenitor cell proliferation in vitro and partial rescue of Pak-deficient HSPC homing and engraftment in vivo. Using conditional knock-out mice, we demonstrate that among group A Paks, Pak2(-/-) HSPC show reduced homing to the bone marrow and altered cell shape similar to PID-LSK cells in vitro and are completely defective in HSPC engraftment. These data demonstrate that Pak proteins are key components of multiple engraftment-associated HSPC functions and play a direct role in activation of ERK in HSPCs, and that Pak2 is specifically essential for HSPC engraftment.
Subject(s)
Cell Movement/genetics , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/physiology , p21-Activated Kinases/physiology , Animals , Cell Movement/physiology , Cell Proliferation , Cell Survival/genetics , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases/metabolism , Extracellular Signal-Regulated MAP Kinases/physiology , Hematopoietic Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , p21-Activated Kinases/genetics , rac GTP-Binding Proteins/metabolism , rac GTP-Binding Proteins/physiologyABSTRACT
Chronic myelogenous leukemia (CML) is a clonal myeloproliferative disease (MPD) initiated by expression of the p210-BCR-ABL fusion protein. We demonstrate in a murine model of p210-BCR-ABL-induced MPD that gene targeting of Rac1 and Rac2 significantly delays or abrogates disease development. Attenuation of the disease phenotype is associated with severely diminished p210-BCR-ABL-induced downstream signaling in primary hematopoietic cells. We utilize NSC23766, a small molecule antagonist of Rac activation, to validate biochemically and functionally Rac as a molecular target in both a relevant animal model and in primary human CML cells in vitro and in a xenograft model in vivo, including in Imatinib-resistant p210-BCR-ABL disease. These data demonstrate that Rac is an additional therapeutic target in p210-BCR-ABL-mediated MPD.
Subject(s)
Fusion Proteins, bcr-abl/metabolism , Gene Expression Regulation, Leukemic , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/metabolism , rac GTP-Binding Proteins/physiology , Aminoquinolines/pharmacology , Animals , Antigens, CD34/biosynthesis , Cell Line, Tumor , Dose-Response Relationship, Drug , Hematopoietic Stem Cells/metabolism , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy , Mice , Myeloproliferative Disorders/therapy , Neoplasm Transplantation , Phenotype , Pyrimidines/pharmacology , rac GTP-Binding Proteins/metabolism , rac1 GTP-Binding Protein/metabolism , RAC2 GTP-Binding ProteinABSTRACT
Hippocampus-dependent learning and memory relies on synaptic plasticity as well as network adaptations provided by the addition of adult-born neurons. We have previously shown that activity-induced intracellular signaling through the Rho family small GTPase Rac1 is necessary in forebrain projection neurons for normal synaptic plasticity in vivo, and here we show that selective loss of neuronal Rac1 also impairs the learning-evoked increase in neurogenesis in the adult mouse hippocampus. Earlier work has indicated that experience elevates the abundance of adult-born neurons in the hippocampus primarily by enhancing the survival of neurons produced just before the learning event. Loss of Rac1 in mature projection neurons did reduce learning-evoked neurogenesis but, contrary to our expectations, these effects were not mediated by altering the survival of young neurons in the hippocampus. Instead, loss of neuronal Rac1 activation selectively impaired a learning-evoked increase in the proliferation and accumulation of neural precursors generated during the learning event itself. This indicates that experience-induced alterations in neurogenesis can be mechanistically resolved into two effects: (1) the well documented but Rac1-independent signaling cascade that enhances the survival of young postmitotic neurons; and (2) a previously unrecognized Rac1-dependent signaling cascade that stimulates the proliferative production and retention of new neurons generated during learning itself.
Subject(s)
Adult Stem Cells/physiology , Maze Learning/physiology , Neural Stem Cells/physiology , Neurogenesis/physiology , Neurons/physiology , Neuropeptides/physiology , rac GTP-Binding Proteins/physiology , Adult Stem Cells/cytology , Animals , Antigens, Differentiation/genetics , Antigens, Differentiation/metabolism , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Cell Proliferation , Cell Survival/physiology , Green Fluorescent Proteins/genetics , Hippocampus/cytology , Hippocampus/physiology , Male , Memory, Long-Term/physiology , Mice , Mice, Knockout , Mitosis/physiology , Neural Stem Cells/cytology , Neuronal Plasticity/physiology , Neurons/cytology , Neuropeptides/genetics , Receptor, trkB/genetics , Receptor, trkB/metabolism , Receptors, AMPA/physiology , Space Perception/physiology , rac GTP-Binding Proteins/genetics , rac1 GTP-Binding ProteinABSTRACT
Activation of receptor tyrosine kinases leads to the formation of two different types of plasma membrane structures: peripheral ruffles and dorsal ruffles. Although the formation of both ruffle types requires activation of the small GTPase Rac, the difference in kinetics suggests that a distinct regulatory mechanism operates for their ruffle formation. DOCK1 and DOCK5 are atypical Rac activators and are both expressed in mouse embryonic fibroblasts (MEFs). We found that although PDGF-induced Rac activation and peripheral ruffle formation were coordinately regulated by DOCK1 and DOCK5 in MEFs, DOCK1 deficiency alone impaired dorsal ruffle formation in MEFs. Unlike DOCK5, DOCK1 bound to phosphatidic acid (PA) through the C-terminal polybasic amino acid cluster and was localized to dorsal ruffles. When this interaction was blocked, PDGF-induced dorsal ruffle formation was severely impaired. In addition, we show that phospholipase D, an enzyme that catalyzes PA synthesis, is required for PDGF-induced dorsal, but not peripheral, ruffle formation. These results indicate that the phospholipase D-PA axis selectively controls dorsal ruffle formation by regulating DOCK1 localization.
Subject(s)
Cell Membrane Structures/metabolism , Phosphatidic Acids/physiology , rac GTP-Binding Proteins/metabolism , Amino Acid Sequence , Animals , Cells, Cultured , Conserved Sequence , Enzyme Activation , Guanine Nucleotide Exchange Factors/metabolism , Guanine Nucleotide Exchange Factors/physiology , Mice , Mice, Transgenic , Microscopy, Fluorescence , Molecular Sequence Data , Phosphatidic Acids/metabolism , Phospholipase D/metabolism , Platelet-Derived Growth Factor/physiology , Protein Structure, Tertiary , Protein Transport , Protein-Tyrosine Kinases/metabolism , Signal Transduction , rac GTP-Binding Proteins/genetics , rac GTP-Binding Proteins/physiologyABSTRACT
Despite the introduction of tyrosine kinase inhibitor therapy, the prognosis for p190-BCR-ABL(+) acute lymphoblastic leukemia remains poor. In the present study, we present the cellular and molecular roles of the Rho GTPase guanine nucleotide exchange factor Vav in lymphoid leukemogenesis and explore the roles of Vav proteins in BCR-ABL-dependent signaling. We show that genetic deficiency of the guanine nucleotide exchange factor Vav3 delays leukemogenesis by p190-BCR-ABL and phenocopies the effect of Rac2 deficiency, a downstream effector of Vav3. Compensatory up-regulation of expression and activation of Vav3 in Vav1/Vav2-deficient B-cell progenitors increases the transformation ability of p190-BCR-ABL. Vav3 deficiency induces apoptosis of murine and human leukemic lymphoid progenitors, decreases the activation of Rho GTPase family members and p21-activated kinase, and is associated with increased Bad phosphorylation and up-regulation of Bax, Bak, and Bik. Finally, Vav3 activation only partly depends on ABL TK activity, and Vav3 deficiency collaborates with tyrosine kinase inhibitors to inhibit CrkL activation and impair leukemogenesis in vitro and in vivo. We conclude that Vav3 represents a novel specific molecular leukemic effector for multitarget therapy in p190-BCR-ABL-expressing acute lymphoblastic leukemia.
Subject(s)
B-Lymphocytes/pathology , Cell Transformation, Neoplastic/pathology , Fusion Proteins, bcr-abl/metabolism , Lymphoid Progenitor Cells/pathology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Proto-Oncogene Proteins c-vav/physiology , Animals , B-Lymphocytes/metabolism , Cell Transformation, Neoplastic/metabolism , Cells, Cultured , Female , Fetal Blood/cytology , Fetal Blood/metabolism , Humans , Lymphoid Progenitor Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Survival Rate , Tumor Stem Cell Assay , rac GTP-Binding Proteins/physiology , RAC2 GTP-Binding ProteinABSTRACT
Hematopoietic stem cells (HSCs) interact with osteoblastic, stromal, and vascular components of the BM hematopoietic microenvironment (HM) that are required for the maintenance of long-term self-renewal in vivo. Osteoblasts have been reported to be a critical cell type making up the HSC niche in vivo. Rac1 GTPase has been implicated in adhesion, spreading, and differentiation of osteoblast cell lines and is critical for HSC engraftment and retention. Recent data suggest a differential role of GTPases in endosteal/osteoblastic versus perivascular niche function. However, whether Rac signaling pathways are also necessary in the cell-extrinsic control of HSC function within the HM has not been examined. In the present study, genetic and inducible models of Rac deletion were used to demonstrate that Rac depletion causes impaired proliferation and induction of apoptosis in the OP9 cell line and in primary BM stromal cells. Deletion of Rac proteins caused reduced trabecular and cortical long bone growth in vivo. Surprisingly, HSC function and maintenance of hematopoiesis in vivo was preserved despite these substantial cell-extrinsic changes. These data have implications for therapeutic strategies to target Rac signaling in HSC mobilization and in the treatment of leukemia and provide clarification to our evolving concepts of HSC-HM interactions.
Subject(s)
Bone Development/physiology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Osteoblasts/metabolism , Signal Transduction , rac GTP-Binding Proteins/metabolism , Animals , Apoptosis , Blotting, Western , Bone Marrow Cells , Cell Communication , Cell Differentiation , Cell Movement , Cell Proliferation , Cells, Cultured , Flow Cytometry , Hematopoiesis , Immunoenzyme Techniques , Mice , Mice, Knockout , Neuropeptides/physiology , Osteoblasts/cytology , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Stromal Cells , X-Ray Microtomography , rac GTP-Binding Proteins/antagonists & inhibitors , rac GTP-Binding Proteins/genetics , rac GTP-Binding Proteins/physiology , rac1 GTP-Binding Protein , RAC2 GTP-Binding ProteinABSTRACT
To understand the role of cytoskeleton and membrane signaling molecules in erythroblast enucleation, we developed a novel analysis protocol of multiparameter high-speed cell imaging in flow. This protocol enabled us to observe F-actin and phosphorylated myosin regulatory light chain (pMRLC) assembled into a contractile actomyosin ring (CAR) between nascent reticulocyte and nucleus, in a population of enucleating erythroblasts. CAR formation and subsequent enucleation were not affected in murine erythroblasts with genetic deletion of Rac1 and Rac2 GTPases because of compensation by Rac3. Pharmacologic inhibition or genetic deletion of all Rac GTPases altered the distribution of F-actin and pMRLC and inhibited enucleation. Erythroblasts treated with NSC23766, cytochalasin-D, colchicine, ML7, or filipin that inhibited Rac activity, actin or tubulin polymerization, MRLC phosphorylation, or lipid raft assembly, respectively, exhibited decreased enucleation efficiency, as quantified by flow cytometry. As assessed by high-speed flow-imaging analysis, colchicine inhibited erythroblast polarization, implicating microtubules during the preparatory stage of enucleation, whereas NSC23766 led to absence of lipid raft assembly in the reticulocyte-pyrenocyte border. In conclusion, enucleation is a multistep process that resembles cytokinesis, requiring establishment of cell polarity through microtubule function, followed by formation of a contractile actomyosin ring, and coalescence of lipid rafts between reticulocyte and pyrenocyte.
Subject(s)
Cell Nucleus/metabolism , Cytoskeleton/physiology , Erythroblasts/physiology , Reticulocytes/physiology , Actins/metabolism , Animals , Biological Transport/genetics , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Nucleus/physiology , Cytoskeleton/chemistry , Cytoskeleton/metabolism , Cytoskeleton/ultrastructure , Erythroblasts/cytology , Erythroblasts/ultrastructure , Erythropoiesis/genetics , Erythropoiesis/physiology , Membrane Microdomains/metabolism , Membrane Microdomains/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microtubules/genetics , Microtubules/metabolism , Microtubules/physiology , Reticulocytes/cytology , Reticulocytes/metabolism , Reticulocytes/ultrastructure , Signal Transduction/genetics , Signal Transduction/physiology , rac GTP-Binding Proteins/genetics , rac GTP-Binding Proteins/metabolism , rac GTP-Binding Proteins/physiologyABSTRACT
Chronic myeloid leukemia in chronic phase (CML-CP) is induced by BCR-ABL1 oncogenic tyrosine kinase. Tyrosine kinase inhibitors eliminate the bulk of CML-CP cells, but fail to eradicate leukemia stem cells (LSCs) and leukemia progenitor cells (LPCs) displaying innate and acquired resistance, respectively. These cells may accumulate genomic instability, leading to disease relapse and/or malignant progression to a fatal blast phase. In the present study, we show that Rac2 GTPase alters mitochondrial membrane potential and electron flow through the mitochondrial respiratory chain complex III (MRC-cIII), thereby generating high levels of reactive oxygen species (ROS) in CML-CP LSCs and primitive LPCs. MRC-cIII-generated ROS promote oxidative DNA damage to trigger genomic instability, resulting in an accumulation of chromosomal aberrations and tyrosine kinase inhibitor-resistant BCR-ABL1 mutants. JAK2(V617F) and FLT3(ITD)-positive polycythemia vera cells and acute myeloid leukemia cells also produce ROS via MRC-cIII. In the present study, inhibition of Rac2 by genetic deletion or a small-molecule inhibitor and down-regulation of mitochondrial ROS by disruption of MRC-cIII, expression of mitochondria-targeted catalase, or addition of ROS-scavenging mitochondria-targeted peptide aptamer reduced genomic instability. We postulate that the Rac2-MRC-cIII pathway triggers ROS-mediated genomic instability in LSCs and primitive LPCs, which could be targeted to prevent the relapse and malignant progression of CML.
Subject(s)
Electron Transport Complex III/metabolism , Genomic Instability , Leukemia, Myeloid, Chronic-Phase/pathology , Neoplasm Proteins/physiology , Neoplastic Stem Cells/drug effects , Reactive Oxygen Species/metabolism , rac GTP-Binding Proteins/physiology , Animals , Catalase/metabolism , DNA Damage , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , Disease Progression , Electron Transport , Fusion Proteins, bcr-abl/genetics , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Membrane Potential, Mitochondrial , Methacrylates/pharmacology , Mice , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplastic Stem Cells/metabolism , Polycythemia Vera/metabolism , Polycythemia Vera/pathology , Recombinant Fusion Proteins/antagonists & inhibitors , Recombinant Fusion Proteins/physiology , Superoxide Dismutase/metabolism , Thiazoles/pharmacology , rac GTP-Binding Proteins/antagonists & inhibitors , rac GTP-Binding Proteins/genetics , RAC2 GTP-Binding ProteinABSTRACT
RNA receptors such as TLR3 and retinoid acid-inducible gene I/melanoma differentiation-associated gene 5 play essential roles in innate immunity to RNA viruses. However, how innate immunity to RNAs is controlled at the molecular level is not well understood. We describe in this study a new regulatory pathway of anti-RNA immunity that is composed of PI3K, its target GTPase Rac, and the newly described immune regulator TNF-α-induced protein 8 like-2 (TIPE2, or TNFAIP8L2). Polyinosinic-polycytidylic acid [Poly (I:C)], a dsRNA receptor ligand, activates Rac via its guanine nucleotide exchange factor Tiam; this leads to the activation of cytokine genes and, paradoxically, downregulation of the Tipe2 gene. TIPE2 is a negative regulator of immunity; its deficiency leads to hyperactivation of the PI3K-Rac pathway as exemplified by enhanced AKT, Rac, P21-activated kinase, and IFN regulatory factor 3 activities. As a consequence, TIPE2 knockout myeloid cells are hyperreactive to Poly (I:C) stimulation, and TIPE2 knockout mice are hypersensitive to Poly (I:C)-induced lethality. These results indicate that TIPE2 controls innate immunity to RNA by targeting the PI3K-Rac pathway. Therefore, manipulating TIPE2 or Rac functions can be effective for controlling RNA viral infections.