Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Nat Commun ; 8: 14837, 2017 03 23.
Article in English | MEDLINE | ID: mdl-28332616

ABSTRACT

Protein drugs that neutralize vascular endothelial growth factor (VEGF), such as aflibercept or ranibizumab, rescue vision in patients with retinal vascular diseases. Nonetheless, optimal visual outcomes require intraocular injections as frequently as every month. Here we report a method to extend the intravitreal half-life of protein drugs as an alternative to either encapsulation or chemical modifications with polymers. We combine a 97-amino-acid peptide of human origin that binds hyaluronan, a major macromolecular component of the eye's vitreous, with therapeutic antibodies and proteins. When administered to rabbit and monkey eyes, the half-life of the modified proteins is increased ∼3-4-fold relative to unmodified proteins. We further show that prototype long-acting anti-VEGF drugs (LAVAs) that include this peptide attenuate VEGF-induced retinal changes in animal models of neovascular retinal disease ∼3-4-fold longer than unmodified drugs. This approach has the potential to reduce the dosing frequency associated with retinal disease treatments.


Subject(s)
Bevacizumab/administration & dosage , Ranibizumab/administration & dosage , Receptors, Vascular Endothelial Growth Factor/administration & dosage , Recombinant Fusion Proteins/administration & dosage , Retinal Diseases/drug therapy , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/pharmacokinetics , Animals , Bevacizumab/chemistry , Bevacizumab/pharmacokinetics , Disease Models, Animal , Female , Half-Life , Humans , Hyaluronic Acid/chemistry , Intravitreal Injections , Macaca fascicularis , Male , Rabbits , Ranibizumab/chemistry , Ranibizumab/pharmacokinetics , Receptors, Vascular Endothelial Growth Factor/chemistry , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/pharmacokinetics , Retinal Diseases/metabolism
2.
J Allergy Clin Immunol ; 137(2): 452-461.e9, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26531865

ABSTRACT

BACKGROUND: Allergen immunotherapy is currently the only disease-modifying treatment available for allergic rhinitis and allergic asthma. OBJECTIVES: We sought to evaluate the induction of sustained tolerance to allergen when anti-IL-4 was combined with a suboptimal course of grass pollen subcutaneous immunotherapy (SCIT) using the allergen-induced skin late-phase response (LPR) and exploratory immune monitoring as surrogate markers of therapeutic response. METHODS: In this randomized, double-blind, 3-group parallel design trial, 37 participants with seasonal allergic rhinitis received suboptimal SCIT (30,000 standardized quality units) in combination with anti-IL-4 (VAK694) and suboptimal SCIT (30,000 standardized quality units) plus placebo antibody or double placebo (placebo SCIT and placebo antibody) restricted to 13 weeks before the grass pollen season. The primary end point was the size of the LPR at 12 months. Exploratory end points included measures of the immunomodulatory activity of treatment by using IL-4 and IL-10 FluoroSpot assays, flow cytometry of T cells, and measurement of IgE, IgG4, and facilitated antigen binding. RESULTS: Both active treatment arms led to a substantial and sustained reduction of the LPR with no additional suppression with addition of anti-IL-4. Treatment with anti-IL-4 and SCIT compared with SCIT alone led to a sustained reduction in allergen-specific IL-4-producing cell counts (P < .01). Both active treatment arms led to induction of dual IL-4/IL-10-producing cells during the pollen season. CONCLUSION: The combination of anti-IL-4 with SCIT provided no additional benefit over SCIT alone in suppressing the allergen-induced skin LPR. A larger trial is needed to assess whether the observed ex vivo downregulation of TH2 responses might translate into clinical benefit.


Subject(s)
Allergens/immunology , Antibodies, Monoclonal/therapeutic use , Desensitization, Immunologic , Poaceae/adverse effects , Pollen/immunology , Rhinitis, Allergic, Seasonal/immunology , Rhinitis, Allergic, Seasonal/therapy , Adult , Allergens/administration & dosage , Antibodies, Monoclonal/pharmacology , Biomarkers , Combined Modality Therapy , Desensitization, Immunologic/adverse effects , Desensitization, Immunologic/methods , Female , Humans , Immunoglobulin E/blood , Immunoglobulin E/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Injections, Subcutaneous , Interleukin-10/metabolism , Interleukin-4/antagonists & inhibitors , Male , Middle Aged , Rhinitis, Allergic, Seasonal/diagnosis , Rhinitis, Allergic, Seasonal/drug therapy , Risk Factors , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Treatment Outcome , Young Adult
3.
Immunity ; 37(6): 1037-49, 2012 Dec 14.
Article in English | MEDLINE | ID: mdl-23159440

ABSTRACT

The regulation of actin dynamics is pivotal for cellular processes such as cell adhesion, migration, and phagocytosis and thus is crucial for neutrophils to fulfill their roles in innate immunity. Many factors have been implicated in signal-induced actin polymerization, but the essential nature of the potential negative modulators are still poorly understood. Here we report that NADPH oxidase-dependent physiologically generated reactive oxygen species (ROS) negatively regulate actin polymerization in stimulated neutrophils via driving reversible actin glutathionylation. Disruption of glutaredoxin 1 (Grx1), an enzyme that catalyzes actin deglutathionylation, increased actin glutathionylation, attenuated actin polymerization, and consequently impaired neutrophil polarization, chemotaxis, adhesion, and phagocytosis. Consistently, Grx1-deficient murine neutrophils showed impaired in vivo recruitment to sites of inflammation and reduced bactericidal capability. Together, these results present a physiological role for glutaredoxin and ROS- induced reversible actin glutathionylation in regulation of actin dynamics in neutrophils.


Subject(s)
Actins/metabolism , Neutrophils/immunology , Neutrophils/metabolism , Reactive Oxygen Species/metabolism , Animals , Bacterial Infections/genetics , Bacterial Infections/immunology , Cells, Cultured , Chemotaxis/immunology , Glutaredoxins/genetics , Glutaredoxins/immunology , Humans , Mice , Mice, Knockout , NADPH Oxidases/metabolism , Protein Binding , Pseudopodia/metabolism
4.
Mol Biol Cell ; 23(7): 1219-30, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22323291

ABSTRACT

The second messenger phosphatidylinositol(3,4,5)P(3) (PtdIns(3,4,5)P(3)) is formed by stimulation of various receptors, including G protein-coupled receptors and integrins. The lipid phosphatases PTEN and SHIP1 are critical in regulating the level of PtdIns(3,4,5)P(3) during chemotaxis. Observations that loss of PTEN had minor and loss of SHIP1 resulted in a severe chemotaxis defect in neutrophils led to the belief that SHIP1 rather than PTEN acts as a predominant phospholipid phosphatase in establishing a PtdIns(3,4,5)P(3) compass. In this study, we show that SHIP1 regulates PtdIns(3,4,5)P(3) production in response to cell adhesion and plays a limited role when cells are in suspension. SHIP1((-)/(-)) neutrophils lose their polarity upon cell adhesion and are extremely adherent, which impairs chemotaxis. However, chemo-taxis can be restored by reducing adhesion. Loss of SHIP1 elevates Akt activation following cell adhesion due to increased PtdIns(3,4,5)P(3) production. From our observations, we conclude that SHIP1 prevents formation of top-down PtdIns(3,4,5)P(3) polarity to facilitate proper cell attachment and detachment during chemotaxis.


Subject(s)
Cell Adhesion/physiology , Cell Movement/physiology , PTEN Phosphohydrolase/metabolism , Phosphoric Monoester Hydrolases/metabolism , Animals , Cell Polarity/physiology , Chemotaxis, Leukocyte/physiology , In Vitro Techniques , Inositol Polyphosphate 5-Phosphatases , Mice , Mice, Knockout , Models, Biological , Neutrophils/physiology , PTEN Phosphohydrolase/deficiency , PTEN Phosphohydrolase/genetics , Phosphatidylinositol Phosphates/biosynthesis , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases , Phosphoric Monoester Hydrolases/chemistry , Phosphoric Monoester Hydrolases/deficiency , Phosphoric Monoester Hydrolases/genetics , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , Second Messenger Systems , Tyrosine/chemistry
5.
Commun Integr Biol ; 3(3): 278-81, 2010 May.
Article in English | MEDLINE | ID: mdl-20714413

ABSTRACT

Neutrophil chemotaxis is a critical component in innate immunity. Recently, using a small-molecule functional screening, we identified NADPHoxidase- dependent Reactive Oxygen Species (ROS) as key regulators of neutrophil chemotactic migration. Neutrophils depleted of ROS form more frequent multiple pseudopodia and lost their directionality as they migrate up a chemoattractant concentration gradient. Here, we further studied the role of ROS in neutrophil chemotaxis and found that multiple pseudopodia formation induced by NADPH inhibitor diphenyleneiodonium chloride (DPI) was more prominent in relatively shallow chemoattractant gradient. It was reported that, in shallow chemoattractant gradients, new pseudopods are usually generated when existing ones bifurcate. Directional sensing is mediated by maintaining the most accurate existing pseudopod, and destroying pseudopods facing the wrong direction by actin depolymerization. We propose that NADPH-mediated ROS production may be critical for disruption of misoriented pseudopods in chemotaxing neutrophils. Thus, inhibition of ROS production will lead to formation of multiple pseudopodia.

6.
J Immunol ; 184(9): 5325-32, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20363972

ABSTRACT

We generated Fas-activated serine threonine phosphoprotein (FAST)-deficient mice (FAST(-/-)) to study the in vivo role of FAST in immune system function. In a model of house dust mite-induced allergic pulmonary inflammation, wild type mice develop a mixed cellular infiltrate composed of eosinophils, lymphocytes, and neutrophils. FAST(-/-) mice develop airway inflammation that is distinguished by the near absence of neutrophils. Similarly, LPS-induced alveolar neutrophil recruitment is markedly reduced in FAST(-/-) mice compared with wild type controls. This is accompanied by reduced concentrations of cytokines (TNF-alpha and IL-6 and -23) and chemoattractants (MIP-2 and keratinocyte chemoattractant) in bronchoalveolar lavage fluids. Because FAST(-/-) neutrophils exhibit normal chemotaxis and survival, impaired neutrophil recruitment is likely to be due to reduced production of chemoattractants within the pulmonary parenchyma. Studies using bone marrow chimeras implicate lung resident hematopoietic cells (e.g., pulmonary dendritic cells and/or alveolar macrophages) in this process. In conclusion, our results introduce FAST as a proinflammatory factor that modulates the function of lung resident hematopoietic cells to promote neutrophil recruitment and pulmonary inflammation.


Subject(s)
Mitochondrial Proteins/physiology , Phosphoproteins/physiology , Protein Serine-Threonine Kinases/physiology , Respiratory Hypersensitivity/immunology , Respiratory Hypersensitivity/pathology , fas Receptor/physiology , Acute Lung Injury/genetics , Acute Lung Injury/immunology , Acute Lung Injury/pathology , Allergens/administration & dosage , Allergens/immunology , Animals , Cells, Cultured , Chemotaxis, Leukocyte/genetics , Chemotaxis, Leukocyte/immunology , Dermatophagoides pteronyssinus/immunology , Dust/immunology , Female , Hematopoiesis/genetics , Hematopoiesis/immunology , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Mice, Transgenic , Mitochondrial Proteins/deficiency , Mitochondrial Proteins/genetics , Neutrophil Infiltration/genetics , Neutrophil Infiltration/immunology , Phosphoproteins/deficiency , Phosphoproteins/genetics , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , RNA-Binding Proteins/physiology , Respiratory Hypersensitivity/genetics , T-Cell Intracellular Antigen-1
7.
Proc Natl Acad Sci U S A ; 107(8): 3546-51, 2010 Feb 23.
Article in English | MEDLINE | ID: mdl-20142487

ABSTRACT

Neutrophil chemotaxis plays an essential role in innate immunity, but the underlying cellular mechanism is still not fully characterized. Here, using a small-molecule functional screening, we identified NADPH oxidase-dependent reactive oxygen species as key regulators of neutrophil chemotactic migration. Neutrophils with pharmacologically inhibited oxidase, or isolated from chronic granulomatous disease (CGD) patients and mice, formed more frequent multiple pseudopodia and lost their directionality as they migrated up a chemoattractant concentration gradient. Knocking down NADPH oxidase in differentiated neutrophil-like HL60 cells also led to defective chemotaxis. Consistent with the in vitro results, adoptively transferred CGD murine neutrophils showed impaired in vivo recruitment to sites of inflammation. Together, these results present a physiological role for reactive oxygen species in regulating neutrophil functions and shed light on the pathogenesis of CGD.


Subject(s)
Chemotaxis , Granulomatous Disease, Chronic/immunology , Neutrophils/immunology , Reactive Oxygen Species/metabolism , Small Molecule Libraries , Animals , Drug Evaluation, Preclinical , Gene Knockdown Techniques , Granulomatous Disease, Chronic/enzymology , HL-60 Cells , Humans , Mice , Mice, Inbred Strains , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Neutrophils/drug effects , Neutrophils/enzymology
8.
J Immunol ; 183(2): 1032-43, 2009 Jul 15.
Article in English | MEDLINE | ID: mdl-19561112

ABSTRACT

Various neutrophil functions such as phagocytosis, superoxide production, and survival are regulated by integrin signaling. Despite the essential role of focal adhesion kinase (FAK) in mediating this signaling pathway, its exact function in neutrophils is ill defined. In this study, we investigated the role of FAK in neutrophils using a myeloid-specific conditional FAK knockout mouse. As reported in many other cell types, FAK is required for regulation of focal adhesion dynamics when neutrophils adhere to fibronectin or ICAM-1. Adhesion on VCAM-1-coated surfaces and chemotaxis after adhesion were not altered in FAK null neutrophils. In addition, we observed significant reduction in NADPH oxidase-mediated superoxide production and complement-mediated phagocytosis in FAK null neutrophils. As a result, these neutrophils displayed decreased pathogen killing capability both in vitro and in vivo in a mouse peritonitis model. In adherent cells, the defects associated with FAK deficiency are likely due to suppression of phosphatidylinositol (3,4,5)-trisphosphate (PtdIns(3,4,5)P3) signaling and chemoattractant-elicited calcium signaling. Disruption of FAK also reduced chemoattractant-elicited superoxide production in suspended neutrophils in the absence of cell adhesion. This may be solely caused by suppression of PtdIns(3,4,5)P3 signaling in these cells, because the fMLP-elicited calcium signal was not altered. Consistent with decreased PtdIns(3,4,5)P3/Akt signaling in FAK null neutrophils, we also observed accelerated spontaneous death in these cells. Taken together, our results revealed previously unrecognized roles of FAK in neutrophil function and provided a potential therapeutic target for treatment of a variety of infectious and inflammatory diseases.


Subject(s)
Cell Adhesion/immunology , Focal Adhesion Protein-Tyrosine Kinases/physiology , Neutrophils/immunology , Phagocytosis , Signal Transduction/immunology , Animals , Calcium Signaling , Cell Adhesion Molecules/metabolism , Cell Death , Chemotaxis , Focal Adhesion Protein-Tyrosine Kinases/deficiency , Mice , Mice, Knockout , Neutrophils/cytology , Peritonitis/immunology , Phosphatidylinositol Phosphates/metabolism
9.
Mol Cell Biol ; 28(13): 4285-99, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18458059

ABSTRACT

Cancer cells in their respective microenvironments must endure various growth-constraining stresses. Under these conditions, the cancer cell-derived factors are thought to modulate the signaling pathways between cell growth and dormancy. Here, we describe a cancer cell-derived regulatory system that modulates the phosphatidylinositol 3'-kinase (PI3K)-Akt pathway under serum deprivation stress. Through the use of biochemical purification, we reveal that cancer cell-secreted insulin-like growth factor 1 (IGF-1) and clusterin, an extracellular stress protein, constitute this regulatory system. We show that secreted clusterin associates with IGF-1 and inhibits its binding to the IGF-1 receptor and hence negatively regulates the PI3K-Akt pathway during serum deprivation. This inhibitory function of clusterin appears to prefer IGF-1, as it fails to exert any effects on epidermal growth factor signaling. We demonstrate furthermore that the constitutive activation of oncogenic signaling downstream of IGF-1 confers insensitivity to the inhibitory effects of clusterin. Thus, the interplay between cancer cell-derived clusterin and IGF-1 may dictate the outcome of cell growth and dormancy during tumorigenic progression.


Subject(s)
Clusterin/metabolism , Insulin-Like Growth Factor I/metabolism , Neoplasms/enzymology , Neoplasms/pathology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Amino Acid Sequence , Animals , Cell Line , Cell Proliferation/drug effects , Clusterin/chemistry , Clusterin/isolation & purification , Culture Media, Serum-Free , Enzyme Activation/drug effects , Epidermal Growth Factor/pharmacology , Humans , Mice , Molecular Sequence Data , Protein Binding/drug effects , Receptor, IGF Type 1/metabolism , Signal Transduction/drug effects , Zebrafish
10.
Immunity ; 27(3): 453-67, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17825589

ABSTRACT

Many neutrophil functions are regulated by phosphatidylinositol-3,4,5-trisphosphate (PtdIns(3,4,5)P3) that mediates protein membrane translocation via binding to pleckstrin homolog (PH) domains within target proteins. Here we show that inositol 1,3,4,5-tetrakisphosphate (Ins(1,3,4,5)P4), a cytosolic small molecule, bound the same PH domain of target proteins and competed for binding to PtdIns(3,4,5)P3. In neutrophils, chemoattractant stimulation triggered rapid elevation in Ins(1,3,4,5)P4 concentration. Depletion of Ins(1,3,4,5)P4 by deleting the gene encoding InsP3KB, which converts Ins(1,4,5)P3 to Ins(1,3,4,5)P4, enhanced membrane translocation of the PtdIns(3,4,5)P3-specific PH domain. This led to enhanced sensitivity to chemoattractant stimulation, elevated superoxide production, and enhanced neutrophil recruitment to inflamed peritoneal cavity. On the contrary, augmentation of intracellular Ins(1,3,4,5)P4 concentration blocked PH domain-mediated membrane translocation of target proteins and dramatically decreased the sensitivity of neutrophils to chemoattractant stimulation. These findings establish a role for Ins(1,3,4,5)P4 in cellular signal transduction pathways and provide another mechanism for modulating PtdIns(3,4,5)P3 signaling in neutrophils.


Subject(s)
Inositol Phosphates/metabolism , Neutrophils/immunology , Neutrophils/metabolism , Phosphatidylinositol Phosphates/metabolism , Signal Transduction/immunology , Animals , Chromatography, High Pressure Liquid , Humans , Immunoprecipitation , Mice , Protein Transport/immunology
11.
Blood ; 110(2): 640-50, 2007 Jul 15.
Article in English | MEDLINE | ID: mdl-17420285

ABSTRACT

All-trans retinoic acid (ATRA) has been widely used in differentiation therapy for acute promyelocytic leukemia (APL). ATRA binds to retinoic acid receptor (RAR) and triggers the formation of the transcription coactivator complex, which leads to changes in gene expression, APL cell-cycle arrest and differentiation, and clinical remission. The mechanisms responsible for ATRA's beneficial effects are still ill-defined. Here, we conducted a large-scale, unbiased short hairpin RNA (shRNA) screen aiming to identify mediators of ATRA-induced differentiation and growth arrest of APL cells. Twenty-six proteins were identified. They cover a wide range of cellular functions, including gene expression, intracellular signaling, cell death control, stress responses, and metabolic regulation, indicating the complexity of ATRA-induced cell growth control and differentiation in APL. One of these proteins, the ubiquitin-conjugating enzyme UBE2D3, is up-regulated in ATRA-treated acute promyelocytic NB4 cells. UBE2D3 is physically associated with cyclin D1 and mediates ATRA-induced cyclin D1 degradation. Knocking down UBE2D3 by RNA interference (RNAi) leads to blockage of ATRA-induced cyclin D1 degradation and cell-cycle arrest. Thus, our results highlight the involvement of the ubiquitin-mediated proteolysis pathway in ATRA-induced cell-cycle arrest and provide a novel strategy for modulating ATRA-elicited cellular effects.


Subject(s)
Cell Division/drug effects , Leukemia, Promyelocytic, Acute/genetics , RNA Interference , RNA, Small Interfering/genetics , Tretinoin/pharmacology , Ubiquitin-Conjugating Enzymes/genetics , Cell Cycle , Cell Line, Tumor , Drug Resistance, Neoplasm , Humans , Leukemia, Promyelocytic, Acute/pathology , Oligonucleotide Array Sequence Analysis , Signal Transduction
12.
Blood ; 109(9): 4028-37, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17202315

ABSTRACT

The recruitment and activation of neutrophils at infected tissues is essential for host defense against invading microorganisms. However, excessive neutrophil recruitment or activation can also damage the surrounding tissues and cause unwanted inflammation. Hence, the responsiveness of neutrophils needs to be tightly regulated. In this study, we have investigated the functional role of tumor suppressor PTEN in neutrophils by using a mouse line in which PTEN is disrupted only in myeloid-derived cells. Chemoattractant-stimulated PTEN(-/-) neutrophils displayed significantly higher Akt phosphorylation and actin polymerization. A larger fraction of these neutrophils displayed membrane ruffles in response to chemoattractant stimulation. In addition, chemoattractant-induced transwell migration and superoxide production were also augmented. Single-cell chemotaxis assays showed that PTEN(-/-) neutrophils have a small (yet statistically significant) defect in directionality. However, these neutrophils also showed an increase in cell speed. As a result, overall chemotaxis, which depends on speed and directionality, was not affected. Consistent with the increased responsiveness of PTEN(-/-) neutrophils, the in vivo recruitment of these cells to the inflamed peritoneal cavity was significantly enhanced. Thus, as a physiologic-negative regulator, PTEN should be a promising therapeutic target for modulating neutrophil functions in various infectious and inflammatory diseases.


Subject(s)
Chemotaxis, Leukocyte/physiology , Neutrophil Infiltration/physiology , Neutrophils/metabolism , PTEN Phosphohydrolase/metabolism , Animals , Cell Line , Chemotactic Factors/pharmacology , Chemotaxis, Leukocyte/drug effects , Inflammation/genetics , Inflammation/metabolism , Mice , Mice, Knockout , Neutrophil Infiltration/drug effects , Neutrophils/cytology , PTEN Phosphohydrolase/deficiency , Proto-Oncogene Proteins c-akt/metabolism
13.
Proc Natl Acad Sci U S A ; 103(40): 14836-41, 2006 Oct 03.
Article in English | MEDLINE | ID: mdl-16988010

ABSTRACT

Neutrophil spontaneous death plays essential roles in neutrophil homeostasis and resolution of inflammation, whereas the underlying molecular mechanisms are still ill-defined. Neutrophils die because of programmed cell death or apoptosis. However, treatment with inhibitor of caspases, which are responsible for the majority of apoptotic cell deaths, does not prevent the spontaneous death of neutrophils. PKB/Akt possesses prosurvival and antiapoptotic activities in a variety of cells. In this study, we show that Akt activity decreases dramatically during the course of neutrophil death. Both phosphatidylinositol 3-kinase and Akt inhibitors enhance neutrophil death. Conditions delaying neutrophil death, such as treatment with granulocyte-macrophage colony-stimulating factor, granulocyte colony-stimulating factor, or IFN-gamma, restore Akt activity. Finally, we demonstrate that neutrophils depleted of PTEN, a phosphatidylinositol 3'-phosphatase that negatively regulates Akt activity, live much longer than WT neutrophils. Thus, we establish Akt deactivation as a causal mediator of neutrophil spontaneous death.


Subject(s)
Neutrophils/cytology , Phosphatidylinositol Phosphates/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Animals , Caspase Inhibitors , Cell Death , Down-Regulation/genetics , Enzyme Activation , Humans , Mice , PTEN Phosphohydrolase/deficiency
SELECTION OF CITATIONS
SEARCH DETAIL
...