Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Cell Death Dis ; 12(6): 538, 2021 05 25.
Article in English | MEDLINE | ID: mdl-34035216

ABSTRACT

Removal of apoptotic cells by phagocytes (also called efferocytosis) is a crucial process for tissue homeostasis. Professional phagocytes express a plethora of surface receptors enabling them to sense and engulf apoptotic cells, thus avoiding persistence of dead cells and cellular debris and their consequent effects. Dysregulation of efferocytosis is thought to lead to secondary necrosis and associated inflammation and immune activation. Efferocytosis in primarily murine macrophages and dendritic cells has been shown to require TAM RTKs, with MERTK and AXL being critical for clearance of apoptotic cells. The functional role of human orthologs, especially the exact contribution of each individual receptor is less well studied. Here we show that human macrophages differentiated in vitro from iPSC-derived precursor cells express both AXL and MERTK and engulf apoptotic cells. TAM RTK agonism by the natural ligand growth-arrest specific 6 (GAS6) significantly enhanced such efferocytosis. Using a newly-developed mouse model of kinase-dead MERTK, we demonstrate that MERTK kinase activity is essential for efferocytosis in peritoneal macrophages in vivo. Moreover, human iPSC-derived macrophages treated in vitro with blocking antibodies or small molecule inhibitors recapitulated this observation. Hence, our results highlight a conserved MERTK function between mice and humans, and the critical role of its kinase activity in homeostatic efferocytosis.


Subject(s)
Macrophages/physiology , Phagocytosis/physiology , c-Mer Tyrosine Kinase/metabolism , Animals , Cell Differentiation , Cells, Cultured , HEK293 Cells , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/physiology , Intercellular Signaling Peptides and Proteins/physiology , Ligands , Macrophages/drug effects , Mice , Mice, Knockout , Phagocytosis/drug effects , Phagocytosis/genetics , Phosphatidylserines/pharmacology , c-Mer Tyrosine Kinase/agonists , c-Mer Tyrosine Kinase/genetics
2.
Int J Mol Sci ; 21(13)2020 Jul 07.
Article in English | MEDLINE | ID: mdl-32645954

ABSTRACT

Tissue-resident macrophages are key players in inflammatory processes, and their activation and functionality are crucial in health and disease. Numerous diseases are associated with alterations in homeostasis or dysregulation of the innate immune system, including allergic reactions, autoimmune diseases, and cancer. Macrophages are a prime target for drug discovery due to their major regulatory role in health and disease. Currently, the main sources of macrophages used for therapeutic compound screening are primary cells isolated from blood or tissue or immortalized or neoplastic cell lines (e.g., THP-1). Here, we describe an improved method to employ induced pluripotent stem cells (iPSCs) for the high-yield, large-scale production of cells resembling tissue-resident macrophages. For this, iPSC-derived macrophage-like cells are thoroughly characterized to confirm their cell identity and thus their suitability for drug screening purposes. These iPSC-derived macrophages show strong cellular identity with primary macrophages and recapitulate key functional characteristics, including cytokine release, phagocytosis, and chemotaxis. Furthermore, we demonstrate that genetic modifications can be readily introduced at the macrophage-like progenitor stage in order to interrogate drug target-relevant pathways. In summary, this novel method overcomes previous shortcomings with primary and leukemic cells and facilitates large-scale production of genetically modified iPSC-derived macrophages for drug screening applications.


Subject(s)
Induced Pluripotent Stem Cells/cytology , Macrophages/cytology , Cell Culture Techniques/methods , Cell Line , Chemotaxis/physiology , Cytokines/metabolism , Drug Evaluation, Preclinical/methods , Humans , Induced Pluripotent Stem Cells/metabolism , Macrophages/metabolism , Phagocytosis/physiology
3.
Nat Commun ; 8(1): 1101, 2017 10 24.
Article in English | MEDLINE | ID: mdl-29062109

ABSTRACT

While constantly rising, the prevalence of allergies is globally one of the highest among chronic diseases. Current treatments of allergic diseases include the application of anti-histamines, immunotherapy, steroids, and anti-immunoglobulin E (IgE) antibodies. Here we report mammalian cells engineered with a synthetic signaling cascade able to monitor extracellular pathophysiological levels of interleukin 4 and interleukin 13, two main cytokines orchestrating allergic inflammation. Upon activation of transgenic cells by these cytokines, designed ankyrin repeat protein (DARPin) E2_79, a non-immunogenic protein binding human IgE, is secreted in a precisely controlled and reversible manner. Using human whole blood cell culturing, we demonstrate that the mammalian dual T helper 2 cytokine sensor produces sufficient levels of DARPin E2_79 to dampen histamine release in allergic subjects exposed to allergens. Hence, therapeutic gene networks monitoring disease-associated cytokines coupled with in situ production, secretion and systemic delivery of immunomodulatory biologics may foster advances in the treatment of allergies.


Subject(s)
Hypersensitivity/genetics , Interleukin-13/immunology , Interleukin-4/immunology , Recombinant Fusion Proteins/genetics , Allergens/immunology , Cell Line , Genetic Engineering , Histamine/immunology , Histamine/metabolism , Humans , Hypersensitivity/immunology , Immunoglobulin E/immunology , Interleukin-13/genetics , Interleukin-4/genetics , Protein Binding , Recombinant Fusion Proteins/metabolism , Th2 Cells/immunology
4.
Cell Death Differ ; 23(12): 1961-1972, 2016 12.
Article in English | MEDLINE | ID: mdl-27740624

ABSTRACT

Eosinophils are frequently elevated in pathological conditions and can cause tissue damage and disease exacerbation. The number of eosinophils in the blood is largely regulated by factors controlling their production in the bone marrow. While several exogenous factors, such as interleukin-5, have been described to promote eosinophil differentiation, comparatively little is known about eosinophil-intrinsic factors that control their de novo generation. Here, we report that the small atypical GTPase RhoH is induced during human eosinophil differentiation, highly expressed in mature blood eosinophils and further upregulated in patients suffering from a hypereosinophilic syndrome. Overexpression of RhoH increases, in a Rho-associated protein kinase-dependent manner, the expression of GATA-2, a transcription factor involved in regulating eosinophil differentiation. In RhoH-/- mice, we observed reduced GATA-2 expression as well as accelerated eosinophil differentiation both in vitro and in vivo. Conversely, RhoH overexpression in bone marrow progenitors reduces eosinophil development in mixed bone marrow chimeras. These results highlight a novel negative regulatory role for RhoH in eosinophil differentiation, most likely in consequence of altered GATA-2 levels.


Subject(s)
Eosinophils/metabolism , Transcription Factors/metabolism , rho GTP-Binding Proteins/metabolism , Animals , Bone Marrow/metabolism , Cell Count , Cell Cycle , Cell Differentiation , Cell Movement , Cell Survival , Cells, Cultured , Chimera , Eosinophils/cytology , GATA2 Transcription Factor/metabolism , Humans , Immunophenotyping , Interleukin-5/metabolism , Longevity , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Receptors, Interleukin-5/metabolism , Up-Regulation
5.
Mol Cell Biol ; 36(1): 132-43, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26483415

ABSTRACT

Death-associated protein kinase 2 (DAPK2) is a Ca(2+)/calmodulin-dependent Ser/Thr kinase that possesses tumor-suppressive functions and regulates programmed cell death, autophagy, oxidative stress, hematopoiesis, and motility. As only few binding partners of DAPK2 have been determined, the molecular mechanisms governing these biological functions are largely unknown. We report the identification of 180 potential DAPK2 interaction partners by affinity purification-coupled mass spectrometry, 12 of which are known DAPK binding proteins. A small subset of established and potential binding proteins detected in this screen was further investigated by bimolecular fluorescence complementation (BiFC) assays, a method to visualize protein interactions in living cells. These experiments revealed that α-actinin-1 and 14-3-3-ß are novel DAPK2 binding partners. The interaction of DAPK2 with α-actinin-1 was localized at the plasma membrane, resulting in massive membrane blebbing and reduced cellular motility, whereas the interaction of DAPK2 with 14-3-3-ß was localized to the cytoplasm, with no impact on blebbing, motility, or viability. Our results therefore suggest that DAPK2 effector functions are influenced by the protein's subcellular localization and highlight the utility of combining mass spectrometry screening with bimolecular fluorescence complementation to identify and characterize novel protein-protein interactions.


Subject(s)
Apoptosis/physiology , Death-Associated Protein Kinases/metabolism , Fluorescence , Proteomics , Autophagy/physiology , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cells, Cultured , Humans , Protein Binding/physiology , Proteomics/methods
6.
Biotechnol Bioeng ; 113(3): 588-97, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26348251

ABSTRACT

Encapsulated designer cells implanted into mice are currently used to validate the efficacy of therapeutic gene networks for the diagnosis and treatment of various human diseases in preclinical research. Because many human conditions cannot be adequately replicated by animal models, complementary and alternative procedures to test future treatment strategies are required. Here we describe a novel approach utilizing an ex vivo human whole-blood culture system to validate synthetic biology-inspired designer cell-based treatment strategies. The viability and functionality of transgenic mammalian designer cells co-cultured with primary human immune cells were characterized. We demonstrated that transgenic mammalian designer cells required adequate insulation from the human blood microenvironment to maintain viability and functionality. The biomaterial alginate-(poly-l-lysine)-alginate used to encapsulate the transgenic designer cells did neither affect the viability of primary granulocytes and lymphocytes nor the functionality of lymphocytes. Additionally, alginate-encapsulated transgenic designer cells remained responsive to the release of the pro-inflammatory cytokine tumor necrosis factor (TNF) from the whole-blood culture upon exposure to bacterial lipopolysaccharide (LPS). TNF diffused into the alginate capsules, bound to the specific TNF receptors on the transgenic designer cells' surface and triggered the expression of the reporter gene SEAP (human placental secreted alkaline phosphatase) that was rewired to the TNF-specific signaling cascade. Human whole-blood culture systems can therefore be considered as valuable complementary assays to animal models for the validation of synthetic circuits in genetically modified mammalian cells and may speed up preclinical research in a world of personalized medicine.


Subject(s)
Alginates/metabolism , Biocompatible Materials/metabolism , Blood Cells , Cell Culture Techniques/methods , Cell Physiological Phenomena , Coculture Techniques/methods , Cytological Techniques/methods , Capsules , Cell Survival , Glucuronic Acid/metabolism , Hexuronic Acids/metabolism , Humans
7.
Sci Transl Med ; 7(318): 318ra201, 2015 Dec 16.
Article in English | MEDLINE | ID: mdl-26676608

ABSTRACT

Psoriasis is a chronic inflammatory skin disease characterized by a relapsing-remitting disease course and correlated with increased expression of proinflammatory cytokines, such as tumor necrosis factor (TNF) and interleukin 22 (IL22). Psoriasis is hard to treat because of the unpredictable and asymptomatic flare-up, which limits handling of skin lesions to symptomatic treatment. Synthetic biology-based gene circuits are uniquely suited for the treatment of diseases with complex dynamics, such as psoriasis, because they can autonomously couple the detection of disease biomarkers with the production of therapeutic proteins. We designed a mammalian cell synthetic cytokine converter that quantifies psoriasis-associated TNF and IL22 levels using serially linked receptor-based synthetic signaling cascades, processes the levels of these proinflammatory cytokines with AND-gate logic, and triggers the corresponding expression of therapeutic levels of the anti-inflammatory/psoriatic cytokines IL4 and IL10, which have been shown to be immunomodulatory in patients. Implants of microencapsulated cytokine converter transgenic designer cells were insensitive to simulated bacterial and viral infections as well as psoriatic-unrelated inflammation. The designer cells specifically prevented the onset of psoriatic flares, stopped acute psoriasis, improved psoriatic skin lesions and restored normal skin-tissue morphology in mice. The antipsoriatic designer cells were equally responsive to blood samples from psoriasis patients, suggesting that the synthetic cytokine converter captures the clinically relevant cytokine range. Implanted designer cells that dynamically interface with the patient's metabolism by detecting specific disease metabolites or biomarkers, processing their blood levels with synthetic circuits in real time, and coordinating immediate production and systemic delivery of protein therapeutics may advance personalized gene- and cell-based therapies.


Subject(s)
Cell Transplantation/methods , Genetic Engineering/methods , Genetic Therapy/methods , Interleukin-10/biosynthesis , Interleukin-4/biosynthesis , Logic , Psoriasis/therapy , Skin/metabolism , Aminoquinolines , Animals , CHO Cells , Cricetulus , Disease Models, Animal , Female , Gene Expression Regulation , HEK293 Cells , HeLa Cells , Humans , Imiquimod , Inflammation Mediators/metabolism , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-4/genetics , Interleukin-4/immunology , Interleukins/genetics , Interleukins/metabolism , Mice, Inbred C57BL , Psoriasis/chemically induced , Psoriasis/genetics , Psoriasis/immunology , Psoriasis/metabolism , Psoriasis/pathology , Reproducibility of Results , Signal Transduction , Skin/immunology , Skin/pathology , Time Factors , Transfection , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Interleukin-22
8.
Int J Biochem Cell Biol ; 65: 151-4, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26055515

ABSTRACT

Death-associated protein kinase 2 (DAPK2/DRP-1) belongs to a family of five related serine/threonine kinases that mediate a range of cellular processes, including membrane blebbing, apoptosis, and autophagy, and possess tumour suppressive functions. The three most conserved family members DAPK1/DAPK, DAPK2 and DAPK3/ZIPK share a high degree of homology in their catalytic domain, but differ significantly in their extra-catalytic structures and tissue-expression profiles. Hence, each orthologue binds to various unique interaction partners, localizes to different subcellular regions and controls some dissimilar cellular functions. In recent years, mechanistic studies have broadened our knowledge of the molecular mechanisms that activate DAPK2 and that execute DAPK2-mediated apoptosis, autophagy and inflammation. In this "molecules in focus" review on DAPK2, the structure, modes of regulation and various cellular functions of DAPK2 will be summarized and discussed.


Subject(s)
Death-Associated Protein Kinases/metabolism , Inflammation/enzymology , Animals , Apoptosis , Autophagy/physiology , Cell Differentiation , Humans
9.
Trends Biotechnol ; 33(2): 65-79, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25466879

ABSTRACT

Humans have manipulated the immune system to dampen or boost the immune response for thousands of years. As our understanding of fundamental immunology and biotechnological methodology accumulates, we can capitalize on this combined knowledge to engineer biological devices with the aim of rationally manipulating the immune response. We address therapeutic approaches based on the principles of synthetic immunology that either ameliorate disorders of the immune system by interfering with the immune response, or improve diverse pathogenic conditions by exploiting immune cell effector functions. We specifically highlight synthetic proteins investigated in preclinical and clinical trials, summarize studies that have used engineered immune cells, and finish with a discussion of possible future therapeutic concepts.


Subject(s)
Allergy and Immunology/trends , Antibodies, Monoclonal/biosynthesis , Immunomodulation/drug effects , Synthetic Biology/methods , Antibodies, Monoclonal/administration & dosage , Cell Engineering/methods , Cytotoxicity, Immunologic/drug effects , Dendritic Cells/cytology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Genetic Therapy/methods , Humans , Immune System/drug effects , Immunotherapy, Adoptive/methods , Lymphocytes/cytology , Lymphocytes/drug effects , Lymphocytes/immunology , Monocytes/cytology , Monocytes/drug effects , Monocytes/immunology , Recombinant Proteins/administration & dosage , Recombinant Proteins/biosynthesis , Synthetic Biology/trends
10.
Nat Commun ; 5: 4408, 2014 Aug 05.
Article in English | MEDLINE | ID: mdl-25093291

ABSTRACT

Allergic disorders are markedly increasing in industrialized countries. The identification of compounds that trigger the immunoglobulin E-dependent allergic reaction remains the key to limit patients' exposure to critical allergens and improve their quality of life. Here we use synthetic biology principles to design a mammalian cell-based allergy profiler that scores the allergen-triggered release of histamine from whole-blood-derived human basophils. A synthetic signalling cascade engineered within the allergy profiler rewires histamine input to the production of reporter protein, thereby integrating histamine levels in whole-blood samples with remarkable sensitivity and a wide dynamic range, allowing for rapid results or long-term storage of output, respectively. This approach provides non-intrusive allergy profiles for the personalized medicine era.


Subject(s)
Histamine Release/immunology , Histamine/chemistry , Hypersensitivity/immunology , Precision Medicine/methods , Synthetic Biology , Allergens/chemistry , Animals , Basophils/cytology , Basophils/metabolism , CHO Cells , Cricetulus , Gene Expression Regulation , Genes, Reporter , HEK293 Cells , Humans , Immunoglobulin E/immunology , Pollen/chemistry , Quality of Life , Transgenes
11.
J Leukoc Biol ; 95(2): 293-303, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24163421

ABSTRACT

The tight regulation of granulocyte chemotaxis is crucial for initiation and resolution of inflammation. Here, we show that DAPK2, a Ca(2+)/CaM-sensitive serine/threonine kinase known to modulate cell death in various cell types, is a novel regulator of migration in granulocytes. We demonstrate that human neutrophils and eosinophils express DAPK2 but unlike other leukocytes, no DAPK1 or DAPK3 protein. When DAPK activities were blocked by inhibitors, we found that neither granulocyte lifespan nor phagocytosis was affected. However, such pharmacological inactivation of DAPK activity abolished motility of granulocytes in response to intermediary but not end-target chemoattractants ex vivo. The defect in chemotaxis in DAPK2-inactive granulocytes is likely a result of reduced polarization of the cells, mediated by a lack of MLC phosphorylation, resulting in radial F-actin and pseudopod formation. As neutrophils treated with DAPKi also showed reduced recruitment to the site of inflammation in a mouse peritonitis model, DAPK2 may be a novel target for anti-inflammatory therapies.


Subject(s)
Cell Movement/drug effects , Chemotactic Factors/pharmacology , Death-Associated Protein Kinases/metabolism , Eosinophils/cytology , Neutrophils/cytology , Neutrophils/enzymology , Animals , Cell Adhesion/drug effects , Cell Polarity/drug effects , Cell Survival/drug effects , Death-Associated Protein Kinases/antagonists & inhibitors , Disease Models, Animal , Eosinophils/drug effects , Eosinophils/enzymology , Humans , Inflammation/pathology , Jurkat Cells , Mice , Myosin Light Chains/metabolism , Neutrophils/drug effects , Peritonitis/pathology , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Small Molecule Libraries/pharmacology
12.
Trends Immunol ; 34(8): 398-409, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23665135

ABSTRACT

Neutrophils, eosinophils, and basophils play essential roles during microbe-induced and sterile inflammation. The severity of such inflammatory processes is controlled, at least in part, by factors that regulate cell death and survival of granulocytes. In recent years, major progress has been made in understanding the molecular mechanisms of granulocyte cell death and in identifying novel damage- and pathogen-associated molecular patterns as well as regulatory cytokines impacting granulocyte viability. Furthermore, an increased interest in innate immunity has boosted our overall understanding of granulocyte biology. In this review, we describe and compare factors and mechanisms regulating neutrophil, eosinophil, and basophil lifespan. Because dysregulation of death pathways in granulocytes can contribute to inflammation-associated immunopathology, targeting granulocyte lifespan could be therapeutically promising.


Subject(s)
Basophils/cytology , Eosinophils/cytology , Inflammation/immunology , Neutrophils/cytology , Animals , Basophils/immunology , Cell Death/immunology , Eosinophils/immunology , Humans , Neutrophils/immunology
13.
J Immunol Methods ; 373(1-2): 209-18, 2011 Oct 28.
Article in English | MEDLINE | ID: mdl-21925181

ABSTRACT

Neutrophils are terminally differentiated cells with a short life-span due to constitutive apoptosis. Because of these characteristics, genetic manipulation of neutrophils has been difficult, although it is highly desired given the importance of neutrophils in the immune system. Here we demonstrate that transduction of primary human mature neutrophils with enhanced green fluorescent protein (eGFP)-encoding lentiviral particles results in GFP-containing cells as previously reported. Yet, our data further show that GFP expression in neutrophils upon transduction is largely due to protein transfer, a process called lentiviral pseudotransduction, and not due to bona fide transduction. Thus, inhibition of viral genome integration by the reverse transcriptase inhibitor 3'-azido-3'-deoxythymidine (AZT) or of protein biosynthesis by cycloheximide (CHX) did not abolish GFP levels in transduced neutrophils. Importantly, lentiviral pseudotransduction of the enzyme death-associated protein kinase 2 (DAPK2) into primary human mature neutrophils resulted in increased protein levels, but not enzymatic functionality. Based on our data and previous reports of unspecific viral effects on immune cells following lentiviral transduction, we discourage scientists to use lentiviral transduction methods to manipulate primary mature neutrophils.


Subject(s)
Lentivirus/genetics , Neutrophils/metabolism , Recombinant Fusion Proteins/metabolism , Transduction, Genetic/methods , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cells, Cultured , Cycloheximide/pharmacology , Death-Associated Protein Kinases , Flow Cytometry , Genetic Vectors/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Immunoblotting , Jurkat Cells , Mutation , Neutrophils/cytology , Neutrophils/drug effects , Phosphorylation , Protein Synthesis Inhibitors/pharmacology , Recombinant Fusion Proteins/genetics , Reverse Transcriptase Inhibitors/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic/genetics , Zidovudine/pharmacology
15.
Blood ; 117(22): 5953-62, 2011 Jun 02.
Article in English | MEDLINE | ID: mdl-21478427

ABSTRACT

The most common form of neutrophil death is apoptosis. In the present study, we report surprising differences in the molecular mechanisms used for caspase activation between FAS/CD95-stimulated and TNF receptor 1 (TNFR1)-stimulated neutrophils. Whereas FAS-induced apoptosis was followed by caspase-8 activation and required Bid to initiate the mitochondrial amplification loop, TNF-α-induced apoptosis involved class IA PI3Ks, which were activated by MAPK p38. TNF-α-induced PI3K activation resulted in the generation of reactive oxygen species, which activated caspase-3, a mechanism that did not operate in neutrophils without active NADPH oxidase. We conclude that in neutrophils, proapoptotic pathways after TNFR1 stimulation are initiated by p38 and PI3K, but not by caspase-8, a finding that should be considered in anti-inflammatory drug-development strategies.


Subject(s)
Apoptosis , Neutrophils/pathology , Phosphatidylinositol 3-Kinases/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism , Signal Transduction , Animals , BH3 Interacting Domain Death Agonist Protein/physiology , Caspase 3/metabolism , Caspase 8/metabolism , Cells, Cultured , Cytochromes c/metabolism , Humans , Membrane Potential, Mitochondrial , Mice , Mitochondria/metabolism , NADPH Oxidases/metabolism , Neutrophils/metabolism , Reactive Oxygen Species/metabolism , Tumor Necrosis Factor-alpha/pharmacology , fas Receptor/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism
16.
Eur J Immunol ; 40(2): 525-9, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19950172

ABSTRACT

RhoH is a member of the Rho (ras homologous) GTPase family, yet it lacks GTPase activity and thus remains in its active conformation. Unlike other Rho GTPases, the RhoH gene transcript is restricted to hematopoietic cells and RhoH was shown to be required for adequate T-cell activation through the TCR. Here, we demonstrate that both blood T and B cells, but not neutrophils or monocytes, express RhoH protein under physiological conditions. Upon TCR complex activation, RhoH was degraded in lysosomes of primary and Jurkat T cells. Pharmacologic activation of T cells distal to the TCR complex had no effect on RhoH protein levels suggesting that early events during T-cell activation are required for RhoH protein degradation. In contrast to T cells, activation of the BCR in blood B cells was not associated with changes in RhoH levels. These data suggest that RhoH function might be regulated by lysosomal degradation of RhoH protein following TCR complex but not BCR activation. This newly discovered regulatory pathway of RhoH expression might limit TCR signaling and subsequent T-cell activation upon Ag contact.


Subject(s)
Lysosomes/metabolism , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/metabolism , Transcription Factors/metabolism , rho GTP-Binding Proteins/metabolism , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , CD3 Complex/immunology , Cell Line , Cells, Cultured , Humans , Immunoblotting , Jurkat Cells , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Receptors, Antigen, B-Cell/metabolism , T-Lymphocytes/drug effects , Transcription Factors/genetics , rho GTP-Binding Proteins/genetics
17.
Trends Biochem Sci ; 34(3): 115-27, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19299143

ABSTRACT

Both the biology and the therapeutic potential of the phosphoinositide 3-kinase (PI3K) signalling axis have been the subject of intense investigation; however, little is known about the regulation of PI3K expression. Emerging evidence indicates that PI3K levels change in response to cellular stimulation with insulin and nuclear receptor ligands, and during various physiological and pathological processes including differentiation, regeneration, hypertension and cancer. Recently identified mechanisms that control PI3K production include increased gene copy number in cancer, and transcriptional regulation of the p110alpha PI3K gene by FOXO3a, NF-kappaB and p53, and of the PI3K regulatory subunits by STAT3, EBNA-2 and SREBP. In most instances, however, the impact of alterations in PI3K expression on PI3K signalling and disease remains to be established.


Subject(s)
Gene Expression Regulation, Neoplastic/physiology , Neoplasms/physiopathology , Phosphatidylinositol 3-Kinases/metabolism , Animals , Humans , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Isoenzymes/physiology , Phosphatidylinositol 3-Kinases/chemistry , Phosphatidylinositol 3-Kinases/genetics , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , Protein Subunits/physiology , Signal Transduction/physiology
18.
Shock ; 32(4): 386-92, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19295491

ABSTRACT

Sialic-acid-binding immunoglobulin-like lectin (Siglec) 9 mediates death signals in neutrophils. The objective of this study was to determine the heterogeneity of neutrophil death responses in septic shock patients and to analyze whether these ex vivo data are related to the severity and outcome of septic shock. In this prospective cohort study, blood samples of patients with septic shock (n = 26) in a medical-surgical intensive care unit (ICU) were taken within 24 h of starting the treatment of septic shock (phase A), after circulatory stabilization (phase B), and 10 days after admission or at ICU discharge if earlier (phase C). Neutrophil death was quantified in the presence and absence of an agonistic anti-Siglec-9 antibody after 24 h ex vivo. In phase A, two distinct patterns of Siglec-9-mediated neutrophil death were observed: resistance to neutrophil death (n = 14; Siglec-9 nonresponders) and increased neutrophil death (n = 12; Siglec-9 responders) after Siglec-9 ligation compared with neutrophils from normal donors. Experiments using a pharmacological pan-caspase-inhibitor provided evidence for caspase-independent neutrophil death in Siglec-9 responders upon Siglec-9 ligation. There were no differences between Siglec-9 responders and nonresponders in length of ICU or hospital stay of survivors or severity of organ dysfunction. Taken together, septic shock patients exhibit different ex vivo death responses of blood neutrophils after Siglec-9 ligation early in shock. Both the resistance and the increased susceptibility to Siglec-9-mediated neutrophil death tend to normalize within 72 h after shock. Further studies are required to understand the role of Siglec-9-mediated neutrophil death in septic shock.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antigens, CD/physiology , Cell Death/drug effects , Lectins/agonists , Lectins/physiology , Neutrophils/cytology , Neutrophils/metabolism , Shock, Septic/metabolism , Antibodies, Monoclonal/immunology , Antigens, CD/immunology , Cells, Cultured , Cytokines/blood , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Humans , Immunoblotting , Intensive Care Units , Lectins/immunology , Neutrophils/drug effects , Shock, Septic/blood , Shock, Septic/pathology , Sialic Acid Binding Immunoglobulin-like Lectins
19.
Proc Natl Acad Sci U S A ; 104(19): 7809-14, 2007 May 08.
Article in English | MEDLINE | ID: mdl-17470792

ABSTRACT

Class IA phosphoinositide 3-kinases (PI3Ks) signal downstream of tyrosine kinases and Ras and control a wide variety of biological responses. In mammals, these heterodimeric PI3Ks consist of a p110 catalytic subunit (p110alpha, p110beta, or p110delta) bound to any of five distinct regulatory subunits (p85alpha, p85beta, p55gamma, p55alpha, and p50alpha, collectively referred to as "p85s"). The relative expression levels of p85 and p110 have been invoked to explain key features of PI3K signaling. For example, free (i.e., non-p110-bound) p85alpha has been proposed to negatively regulate PI3K signaling by competition with p85/p110 for recruitment to phosphotyrosine docking sites. Using affinity and ion exchange chromatography and quantitative mass spectrometry, we demonstrate that the p85 and p110 subunits are present in equimolar amounts in mammalian cell lines and tissues. No evidence for free p85 or p110 subunits could be obtained. Cell lines contain 10,000-15,000 p85/p110 complexes per cell, with p110beta and p110delta being the most prevalent catalytic subunits in nonleukocytes and leukocytes, respectively. These results argue against a role of free p85 in PI3K signaling and provide insights into the nonredundant functions of the different class IA PI3K isoforms.


Subject(s)
Phosphatidylinositol 3-Kinases/chemistry , Animals , Catalytic Domain , Dimerization , Mass Spectrometry , Mice , Mice, Inbred C57BL , NIH 3T3 Cells , Phosphatidylinositol 3-Kinases/analysis , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/physiology , Protein Subunits , RNA, Messenger/analysis , Signal Transduction
20.
Mol Cell Proteomics ; 4(8): 1038-51, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15879432

ABSTRACT

Protein phosphorylation plays a critical role in normal cellular function and is often subverted in disease. Although major advances have recently been made in identification and quantitation of protein phosphorylation sites by MS, current methodological limitations still preclude routine, easily usable, and comprehensive quantitative analysis of protein phosphorylation. Here we report a simple LC-MS method to quantify gel-separated proteins and their sites of phosphorylation; in this approach, integrated chromatographic peak areas of peptide analytes from proteins under study are normalized to those of a non-isotopically labeled internal standard protein spiked into the excised gel samples just prior to in-gel digestion. The internal standard intensities correct for differences in enzymatic activities and sample losses that may occur during the processes of in-gel digestion and peptide extraction from the gel pieces. We used this method of peak area measurement with an internal standard to investigate the effects of pervanadate on protein phosphorylation in the WEHI-231 B cell lymphoma cell line and to assess the role of phosphoinositide 3-kinase (PI3K) in these phosphorylation events. Phosphoproteins, isolated from total cell lysates using IMAC or by immunoprecipitation using Tyr(P) antibodies, were analyzed using this method, leading to identification of >400 proteins, several of which were found at higher levels in phosphoprotein fractions after pervanadate treatment. Pretreatment of cells with the PI3K inhibitor wortmannin reduced the phosphorylation level of certain proteins (e.g. STAT1 and phospholipase Cgamma2) while increasing the phosphorylation of several others. Peak area measurement with an internal standard was also used to follow the dynamics of PI3K-dependent and -independent changes in the post-translational modification of both known and novel phospholipase Cgamma2 phosphorylation sites. Our results illustrate the capacity of this conceptually simple LC-MS method for quantification of gel-separated proteins and their phosphorylation sites and for quantitative profiling of biological systems.


Subject(s)
Lymphoma, B-Cell/metabolism , Neoplasm Proteins/metabolism , Phosphoproteins/metabolism , Androstadienes/pharmacology , Chromatography, Liquid , Electrophoresis, Polyacrylamide Gel , Humans , Neoplasm Proteins/isolation & purification , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Protein Processing, Post-Translational , Reference Standards , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tumor Cells, Cultured , Wortmannin
SELECTION OF CITATIONS
SEARCH DETAIL
...