Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
J Alzheimers Dis ; 61(4): 1477-1483, 2018.
Article in English | MEDLINE | ID: mdl-29376863

ABSTRACT

The term frontotemporal lobar degeneration (FTLD) defines a group of heterogeneous conditions histologically characterized by neuronal degeneration, inclusions of various proteins, and synaptic loss. However, the molecular mechanisms contributing to these alterations are still unknown. As the Rho-GTPase family member Cell division cycle 42 (Cdc42) plays a key role in the regulation of actin cytoskeleton dynamics and spine formation, we investigated whether Cdc42 protein levels were altered in the disease. Cdc42 was increased in the frontal cortex of FTLD patients compared to age-matched controls, but also in Alzheimer's disease (AD) patients included in the data-set. On the other hand, the pool of circulating Cdc42 in the plasma was altered in FTLD but not in AD patients. Interestingly, the stratification of the FTLD patients according to the different clinical variants showed a specific decrease of Cdc42 expression in the behavioral subgroup. This data support a role of Cdc42 in FTLD and specifically in the behavioral variant.


Subject(s)
Alzheimer Disease/blood , Frontotemporal Lobar Degeneration/blood , cdc42 GTP-Binding Protein/blood , Aged , Aged, 80 and over , Brain/pathology , Case-Control Studies , Female , Frontotemporal Lobar Degeneration/pathology , Humans , Male , Middle Aged
2.
J Gerontol A Biol Sci Med Sci ; 72(9): 1196-1200, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-28498918

ABSTRACT

The small RhoGTPase Cdc42 is mechanistically linked to aging of multiple tissues and to rejuvenation of hematopoietic stem cells in mice. However, data validating Cdc42 activity and expression as biomarker for aging in humans are still missing. Here, we hypothesized that Cdc42 might serve as a novel biomarker of aging in older adults and therefore we determined Cdc42 activity and expression levels in peripheral blood cells from a cohort of 196 donors. We investigated the association of these parameters with both chronological and biological aging. We also tested in this cohort of older adults a recently published algorithm determining chronological age based on DNA methylation profiles. A positive correlation with chronological age was found for both the level of Cdc42 mRNA and the level of active Cdc42 protein (the GTP bound form). Notably, the level of Cdc42 mRNA as well as total protein showed a specific strong association to cardiovascular disease and Cdc42 mRNA levels also to a history of myocardial infarction. In summary, these data validate Cdc42 as a blood biomarker of both chronological aging as well as aging-associated diseases like cardiovascular disease and myocardial infarction.


Subject(s)
Cellular Senescence/physiology , Hematopoietic Stem Cells/metabolism , cdc42 GTP-Binding Protein/blood , Aged , Algorithms , Anthropometry , Biomarkers/blood , Cardiovascular Diseases/genetics , DNA Methylation , Female , Germany , Humans , Male , Phenotype , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction
3.
J Thromb Haemost ; 13(4): 619-30, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25556321

ABSTRACT

BACKGROUND: Platelet cytoskeletal reorganization is essential for platelet adhesion and thrombus formation in hemostasis and thrombosis. The Rho GTPases RhoA, Rac1 and Cdc42 are the main players in cytoskeletal dynamics of platelets and induce filopodia and lamellipodia formation and actin polymerization to strongly increase the platelet surface upon activation. Moreover, they are important for platelet secretion, integrin activation and arterial thrombus formation. OBJECTIVES: Rho GTPases are regulated by GTPase-activating proteins (GAPs) that stimulate their GTPase activity to terminate Rho signaling. The regulation of Rho GTPase activity in platelets is not well defined. Recently, we identified oligophrenin1 (OPHN1), a RhoGAP in platelets that exhibits strong GTPase-stimulating activity towards RhoA, Cdc42 and Rac1. RESULTS: In the present study we show for the first time, that deficiency of OPHN1 led to abnormal Rho activation and increased platelet cytoskeletal reorganization, including cell adhesion and lamellipodia formation on fibrinogen. Furthermore, platelets from ophn1(-/-) mice showed enhanced susceptibility to platelet activation with alterations in actin distribution and early release of granules. Platelet activation was enhanced following GPVI and PAR4 stimulation. This translated into elevated platelet thrombus formation and promoted arterial thrombosis under low shear conditions with altered hemostasis, as detected by tail bleeding time. CONCLUSIONS: The results of the present study identified OPHN1 as an important regulator of platelet cytoskeletal reorganization and demonstrate that abnormal regulation of Rho proteins leads to increased platelet adhesion and thrombus formation under low shear conditions in vitro and in vivo, suggesting a prothrombotic phenotype of mice critical for acute thrombotic occlusions.


Subject(s)
Blood Coagulation , Blood Platelets/enzymology , Cytoskeletal Proteins/deficiency , GTPase-Activating Proteins/deficiency , Nuclear Proteins/deficiency , Thrombosis/enzymology , rho GTP-Binding Proteins/blood , Animals , Cytoskeletal Proteins/genetics , Cytoskeleton/enzymology , Disease Models, Animal , Enzyme Activation , Female , GTPase-Activating Proteins/genetics , Male , Mice, Inbred C57BL , Mice, Knockout , Neuropeptides/blood , Nuclear Proteins/genetics , Platelet Activation , Pseudopodia/enzymology , Signal Transduction , Thrombosis/blood , Thrombosis/genetics , Time Factors , cdc42 GTP-Binding Protein/blood , rac1 GTP-Binding Protein/blood , rhoA GTP-Binding Protein
4.
J Thromb Haemost ; 11(1): 35-46, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23121917

ABSTRACT

The Rho family of GTP binding proteins, also commonly referred to as the Rho GTPases, are master regulators of the platelet cytoskeleton and platelet function. These low-molecular-weight or 'small' GTPases act as signaling switches in the spatial and temporal transduction, and amplification of signals from platelet cell surface receptors to the intracellular signaling pathways that drive platelet function. The Rho GTPase family members RhoA, Cdc42 and Rac1 have emerged as key regulators in the dynamics of the actin cytoskeleton in platelets and play key roles in platelet aggregation, secretion, spreading and thrombus formation. Rho GTPase regulators, including GEFs and GAPs and downstream effectors, such as the WASPs, formins and PAKs, may also regulate platelet activation and function. In this review, we provide an overview of Rho GTPase signaling in platelet physiology. Previous studies of Rho GTPases and platelets have had a shared history, as platelets have served as an ideal, non-transformed cellular model to characterize Rho function. Likewise, recent studies of the cell biology of Rho GTPase family members have helped to build an understanding of the molecular regulation of platelet function and will continue to do so through the further characterization of Rho GTPases as well as Rho GAPs, GEFs, RhoGDIs and Rho effectors in actin reorganization and other Rho-driven cellular processes.


Subject(s)
Blood Platelets/enzymology , Platelet Activation , Thrombosis/enzymology , rho GTP-Binding Proteins/blood , Actin Cytoskeleton/enzymology , Animals , Cell Shape , GTPase-Activating Proteins/blood , Guanine Nucleotide Exchange Factors/blood , Humans , Pseudopodia/enzymology , Signal Transduction , Thrombosis/blood , cdc42 GTP-Binding Protein/blood , p21-Activated Kinases/blood , rac GTP-Binding Proteins/blood , rho-Specific Guanine Nucleotide Dissociation Inhibitors/blood , rhoA GTP-Binding Protein/blood
5.
J Cell Physiol ; 205(3): 452-62, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16113997

ABSTRACT

The biological and pathophysiological significance of class II phosphoinositide 3-kinase enzyme expression currently remains unclear. Using an in vitro scrape wound assay and time-lapse video microscopy, we demonstrate that cell motility is increased in cultures expressing recombinant PI3K-C2beta enzyme. In addition, overexpression of PI3K-C2beta transiently decreased cell adhesion, stimulated the formation of cytoplasmic processes, and decreased the rate of cell proliferation. Consistent with these observations, expression of PI3K-C2beta also decreased expression of alpha4 beta1 integrin subunits. Using asynchronous cultures, we show that endogenous PI3K-C2beta is present in lamellipodia of motile cells. When cells expressing recombinant PI3K-C2beta were plated onto fibronectin, cortical actin staining increased markedly and actin rich lamellipodia and filopodia became evident. Overexpression of a 2xFYVE(Hrs) domain fusion protein abolished this response demonstrating that the effect of PI3K-C2beta on the reorganization of actin filaments is dependent upon PtdIns3P. Finally, overexpression of PI3K-C2beta increased GTP loading of Cdc42. Our data demonstrates for the first time, that PI3K-C2beta plays a regulatory role in cell motility and that the mechanism by which it reorganizes the actin cytoskeleton is dependent upon PtdIns3P production.


Subject(s)
Cell Movement/drug effects , Cell Movement/physiology , Phosphatidylinositol 3-Kinases/pharmacology , Phosphatidylinositol Phosphates/physiology , Actins/physiology , Animals , Cattle/blood , Cell Adhesion/drug effects , Cell Line , Cell Proliferation/drug effects , Cells/ultrastructure , Class II Phosphatidylinositol 3-Kinases , Cytoskeleton/drug effects , Fetal Blood , Guanosine Triphosphate/metabolism , Humans , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Recombinant Proteins/pharmacology , Tissue Distribution , cdc42 GTP-Binding Protein/blood , cdc42 GTP-Binding Protein/metabolism , rac GTP-Binding Proteins/metabolism
6.
J Immunol ; 170(5): 2670-9, 2003 Mar 01.
Article in English | MEDLINE | ID: mdl-12594296

ABSTRACT

Eosinophil respiratory burst is an important event in asthma and related inflammatory disorders. However, little is known concerning activation of the respiratory burst NADPH oxidase in human eosinophils. Conversely, neutrophils are known to assemble NADPH oxidase in intracellular and plasma membranes. We hypothesized that eosinophils and neutrophils translocate NADPH oxidase to distinct intracellular locations, consistent with their respective functions in O(2)(-)-mediated cytotoxicity. PMA-induced O(2)(-) release assayed by cytochrome c was 3.4-fold higher in atopic human eosinophils than in neutrophils, although membrane-permeable dihydrorhodamine-123 showed similar amounts of release. Eosinophil O(2)(-) release was dependent on Rac, in that it was 54% inhibited by Clostridium difficile toxin B (400-800 ng/ml). In eosinophils stimulated with PMA, a pronounced shift of cytosolic Rac to p22(phox)-positive plasma membrane was observed by confocal microscopy, whereas neutrophils directed Rac2 mainly to intracellular sites coexpressing p22(phox). Similarly, ex vivo sputum eosinophils from asthmatic subjects exhibited predominantly plasma membrane-associated immunoreactivity for Rac, whereas sputum neutrophils exhibited cytoplasmic Rac2 staining. Thus, activated sputum eosinophils, rather than neutrophils, may contribute significantly to the pathogenesis of asthma by extracellular release of tissue-damaging O(2)(-). Our findings suggest that the differential modes of NADPH oxidase assembly in these cells may have important implications for oxidant-mediated tissue injury.


Subject(s)
Eosinophils/metabolism , Hypersensitivity, Immediate/blood , Hypersensitivity, Immediate/metabolism , Membrane Transport Proteins , Neutrophils/metabolism , Respiratory Burst/physiology , Sputum/metabolism , Asthma/metabolism , Asthma/pathology , Cell Membrane/enzymology , Enzyme Activation/physiology , Eosinophils/enzymology , Eosinophils/pathology , Extracellular Space/metabolism , Humans , Hypersensitivity, Immediate/enzymology , Hypersensitivity, Immediate/pathology , Immunohistochemistry , Intracellular Fluid/metabolism , NADPH Dehydrogenase/blood , NADPH Dehydrogenase/metabolism , NADPH Oxidases/blood , NADPH Oxidases/metabolism , Neutrophils/pathology , Phosphoproteins/blood , Phosphoproteins/metabolism , Protein Processing, Post-Translational , Protein Transport , RNA, Messenger/biosynthesis , Sputum/cytology , Sputum/enzymology , Superoxides/blood , Superoxides/metabolism , cdc42 GTP-Binding Protein/biosynthesis , cdc42 GTP-Binding Protein/blood , cdc42 GTP-Binding Protein/genetics , rac GTP-Binding Proteins/biosynthesis , rac GTP-Binding Proteins/blood , rac GTP-Binding Proteins/genetics , rac GTP-Binding Proteins/metabolism , rac1 GTP-Binding Protein/biosynthesis , rac1 GTP-Binding Protein/blood , rac1 GTP-Binding Protein/genetics , rho GTP-Binding Proteins/blood , rho GTP-Binding Proteins/metabolism , RAC2 GTP-Binding Protein
7.
Blood ; 98(13): 3708-16, 2001 Dec 15.
Article in English | MEDLINE | ID: mdl-11739176

ABSTRACT

In this study, the receptors and signals involved in collagen-induced platelet spreading were examined. It was found that platelet spreading on collagen (presenting a polygon shape with a number of filopodialike projections) was inhibited by the anti-integrin alpha(2) antibody, suggesting the involvement of integrin alpha(2)beta(1) in this process. Studies with a glutathione-S-transferase fusion protein that binds specifically to activated Rac and in vitro p21-activated kinase (PAK) kinase assays revealed that Rac and PAK were activated during this collagen-activated process. Platelet spreading on collagen-coated surfaces was inhibited strongly by PP1 (a Src family kinase inhibitor) or weakly by wortmannin (a phosphatidylinositol 3-kinase [PI3-kinase] inhibitor) but not at all by Y-27632 (a Rho kinase inhibitor). The surfaces coated with anti-integrin alpha(2)beta(1) antibodies also induced platelet spreading (presenting an almost complete round shape) and activation of Rac and PAK, although more slowly than collagen-coated surfaces. The antibody-induced responses were strongly inhibited by PP1 or wortmannin but not by Y-27632. The same concentration of Y-27632 inhibited collagen-induced shape change of platelets in suspension. These findings suggest that Rac and/or PAK activation, but not Rho, may play certain roles in platelet spreading via integrin alpha(2)beta(1) and that Src family kinases and PI3-kinase participate in these processes. Furthermore, the difference between spreading on collagen and the anti-integrin antibody suggests the involvement of other receptor(s) (in addition to the integrin alpha(2)beta(1)) for collagen-induced spreading, the most likely candidate being glycoprotein VI.


Subject(s)
Blood Platelets/physiology , Collagen/pharmacology , Integrins/physiology , Protein Serine-Threonine Kinases/blood , rac GTP-Binding Proteins/blood , Amides/pharmacology , Androstadienes/pharmacology , Antibodies/pharmacology , Antigens, CD/immunology , Apyrase/pharmacology , Blood Platelets/drug effects , Enzyme Activation , Enzyme Inhibitors/pharmacology , Humans , Immunoglobulin Fab Fragments/pharmacology , Integrin alpha2 , Integrin beta1/immunology , Integrins/immunology , Phosphoinositide-3 Kinase Inhibitors , Platelet Adhesiveness , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pyrazoles/pharmacology , Pyridines/pharmacology , Pyrimidines/pharmacology , Receptors, Collagen , Wortmannin , cdc42 GTP-Binding Protein/blood , p21-Activated Kinases , rac GTP-Binding Proteins/antagonists & inhibitors
8.
J Cell Biol ; 150(4): 785-96, 2000 Aug 21.
Article in English | MEDLINE | ID: mdl-10953003

ABSTRACT

We developed a permeabilization method that retains coupling between N-formyl-methionyl-leucyl-phenylalanine tripeptide (FMLP) receptor stimulation, shape changes, and barbed-end actin nucleation in human neutrophils. Using GTP analogues, phosphoinositides, a phosphoinositide-binding peptide, constitutively active or inactive Rho GTPase mutants, and activating or inhibitory peptides derived from neural Wiskott-Aldrich syndrome family proteins (N-WASP), we identified signaling pathways leading from the FMLP receptor to actin nucleation that require Cdc42, but then diverge. One branch traverses the actin nucleation pathway involving N-WASP and the Arp2/3 complex, whereas the other operates through active Rac to promote actin nucleation. Both pathways depend on phosphoinositide expression. Since maximal inhibition of the Arp2/3 pathway leaves an N17Rac inhibitable alternate pathway intact, we conclude that this alternate involves phosphoinositide-mediated uncapping of actin filament barbed ends.


Subject(s)
Actins/blood , Neutrophils/physiology , Receptors, Immunologic/blood , Receptors, Peptide/blood , cdc42 GTP-Binding Protein/blood , Adult , Cell Membrane/drug effects , Cell Membrane/ultrastructure , Cell Membrane Permeability , Cell Size/drug effects , Glucosides/pharmacology , Guanosine Triphosphate/analogs & derivatives , Guanosine Triphosphate/pharmacology , Humans , In Vitro Techniques , Kinetics , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Nerve Tissue Proteins/chemistry , Neutrophils/cytology , Neutrophils/drug effects , Peptide Fragments/pharmacology , Receptors, Formyl Peptide , Signal Transduction/drug effects , Signal Transduction/physiology , Wiskott-Aldrich Syndrome , Wiskott-Aldrich Syndrome Protein, Neuronal
SELECTION OF CITATIONS
SEARCH DETAIL
...