8 research outputs found

    Tumor necrosis factor alpha induces a caspase-independent death pathway in human neutrophils

    No full text
    Tumor necrosis factor alpha (TNF-alpha) is a cytokine with multiple roles in the immune system, including the induction and potentiation of cellular functions in neutrophils (PMNs). TNF-alpha also induces apoptotic signals leading to the activation of several caspases, which are involved in different steps of the process of cell death. Inhibition of caspases usually increases cell survival. Here, we found that inhibition of caspases by the general caspase inhibitor zVAD-fmk did not prevent TNF-alpha-induced PMN death. After 6 hours of incubation, TNF-alpha alone caused PMN death with characteristic apoptotic features (typical morphologic changes, DNA laddering, external phosphatidyl serine [PS] exposure in the plasma membrane, Bax clustering and translocation to the mitochondria, and degradation of mitochondria), which coincided with activation of caspase-8 and caspase-3. However, in the presence of TNF-alpha, PMNs died even when caspases were completely inhibited. This type of cell death lacked nuclear features of apoptosis (le, no DNA laddering but aberrant hyperlobulated nuclei without typical chromatin condensation) and demonstrated no Bax redistribution, but it did show mitochondria clustering and plasma membrane PS exposure. In contrast, Fas-triggered PMN apoptosis was completely blocked by zVAD-fmk. Experiments with scavengers of reactive oxygen species (ROS) and with inhibitors of mitochondrial respiration, with PMN-derived cytoplasts (which lack mitochondria) and with PMNs from patients with chronic granulomatous disease (which have impaired nicotinamide adenine dinucleotide phosphate [NADPH] oxidase) indicated that TNF-alpha/zVAD-fmk-induced cell death depends on mitochondria-derived ROS. Thus, TNF-alpha can induce a "classical," caspase-dependent and a "nonclassical" caspase-independent cell deat

    Granulocyte colony-stimulating factor inhibits the mitochondria-dependent activation of caspase-3 in neutrophils

    No full text
    The exact mechanism of apoptosis in neutrophils (PMNs) and the explanation for the antiapoptotic effect of granulocyte colony-stimulating factor (G-CSF) in PMNs are unclear. Using specific fluorescent mitochondrial staining, immunofluorescent confocal microscopy, Western blotting, and flow cytometry, this study found that PMNs possess an unexpectedly large number of mitochondria, which are involved in apoptosis. Spontaneous PMN apoptosis was associated with translocation of the Bcl-2-like protein Bax to the mitochondria and subsequent caspase-3 activation, but not with changes in the expression of Bax. G-CSF delayed PMN apoptosis and prevented both associated events. These G-CSF effects were inhibited by cycloheximide. The general caspase inhibitor z-Val-Ala-DL-Asp-fluoromethylketone (zVAD-fmk) prevented caspase-3 activation and apoptosis In PMNs, but not Bax redistribution. PMN-derived cytoplasts, which lack a nucleus, granules, and mitochondria, spontaneously underwent caspase-3 activation and apoptosis (phosphatidylserine exposure), without Bax redistribution. zVAD-fmk inhibited both caspase-3 activation and phosphatidylserine exposure in cultured cytoplasts. Yet, G-CSF prevented neither caspase-3 activation nor apoptosis In cytoplasts, confirming the need for protein synthesis in the G-CSIF effects. These data demonstrate that (at least) 2 routes regulate PMN apoptosis: one via Bax-to-mitochondria translocation and a second mitochondria-independent pathway, both linked to caspase-3 activation. Moreover, G-CSF exerts its antiapoptotic effect In the first, that Is, mitochondria-dependent, route and has no Impact on the second. (Blood. 2002;99:672-679) (C) 2002 by The American Society of Hematolog

    Apoptotic neutrophils in the circulation of patients with glycogen storage disease type 1b (GSD1b)

    No full text
    Glycogen storage disease type 1b (GSD1b) is a rare autosomal recessive disorder characterized by hypoglycemia, hepatomegaly, and growth retardation, and associated-for unknown reasons-with neutropenia and neutrophil dysfunction. In 5 GSD1b patients in whom nicotinamide adenine dinucleotide phosphate-oxidase activity and chemotaxis were defective, we found that the majority of circulating granulocytes bound Annexin-V. The neutrophils showed signs of apoptosis with increased caspase activity, condensed nuclei, and perinuclear clustering of mitochondria to which the proapoptotic Bcl-2 member Bax had translocated already. Granulocyte colony-stimulating factor (G-CSF) addition to in vitro cultures did not rescue the GSD1b neutrophils from apoptosis as occurs with G-CSF-treated control neutrophils. Moreover, the 2 GSD1b patients on G-CSF treatment did not show significantly lower levels of apoptotic neutrophils in the bloodstream. Current understanding of neutrophil apoptosis and the accompanying functional demise suggests that GSD1b granulocytes are dysfunctional because they are apoptotic. (C) 2003 by The American Society of Hematolog

    Growth factors G-CSF and GM-CSF differentially preserve chemotaxis of neutrophils aging in vitro

    No full text
    OBJECTIVE: The ability of human neutrophils to migrate was studied during culture in vitro. METHODS: Neutrophils were isolated from human blood and cultured at 37 degrees C. Apoptosis was determined by Annexin-V fluorescein isothiocyanate binding. Receptor expression was measured by fluorescence in situ hybridization analysis with monoclonal antibodies. Migration was assessed with Transwell Fluoroblock inserts and calcein-stained neutrophils. Extracellular signal-regulated kinase 1/2 (ERK-1/2) activation was determined with monoclonal antibody against phosphorylated ERK-1/2. RESULTS: Upon culture, untreated neutrophils downregulated the chemotaxin receptors FPR, CXC chemokine receptor 1, and CXC chemokine receptor 2 and lost the ability to migrate to formyl-methionyl-leucyl-phenylalanin, interleukin 8 (IL-8), and C5a. In contrast, expression of CXCR4 was induced; this receptor was able to signal (increase in intracellular free calcium ions [Ca(2+)](i), ERK-1/2 activation) but was nonfunctional (no chemotaxis to stromal cell-derived factor-1alpha). The myeloid growth factors granulocyte colony-stimulating factor (G-CSF) and granulocyte-macrophage colony-stimulating factor (GM-CSF) retarded the process of functional decay during cell culture. However, while preserving chemotaxis of neutrophils toward formyl-methionyl-leucyl-phenylalanin or C5a, GM-CSF-in contrast to G-CSF-did not preserve chemotaxis toward IL-8, with a corresponding downregulation of the IL-8 receptors. The decay in neutrophil chemotaxis occurred prior to detectable phosphatidylserine (PS)-exposure. In contrast, the induction of [Ca(2+)](i) rises and ERK-1/2 activation correlated with chemotaxin receptor expression unless the cells were truly apoptotic. CONCLUSION: Neutrophils aging in vitro lose their chemotactic capacity. Functional decay starts prior to PS exposure and can be partially prevented by G-CSF and GM-CSF, in a differential fashion. These growth factors act by increasing the number of viable neutrophils, by altering the levels of chemotaxin receptor expression, and-independently-by affecting signaling cascade

    Neutrophils in Barth syndrome (BTHS) avidly bind annexin-V in the absence of apoptosis

    No full text
    Barth syndrome (BTHS) is a rare X-linked disease characterized by a triad of dilated cardiomyopathy, skeletal myopathy, and neutropenia. The disease is associated with mutations of the TAZ gene, resulting in defective cardiolipin (CL), an important inner mitochondrial membrane component. Untreated boys die in infancy or early childhood from septicemia or cardiac failure. To date, neutrophil function has never been studied. Directed motility and killing activity of neutrophils was investigated in 7 BTHS patients and found normal in those tested. The circulating neutrophils and eosinophils (but not monocytes or lymphocytes) showed annexin-V binding, suggesting phosphatidylserine (PS) exposure due to apoptosis. However, caspase activity was absent in fresh BTHS cells. Unexpectedly, the near absence of CL impacted neither the mitochondrial mass and shape in fresh BTHS neutrophils nor mitochondrial clustering and Bax translocation upon apoptosis. Annexin-V binding to BTHS neutrophils was not caused by phospholipid scrambling. Moreover, freshly purified BTHS neutrophils were not phagocytosed by macrophages. In sum, a massive number of circulating annexin-V-binding neutrophils in the absence of apoptosis can be demonstrated in BTHS. These neutrophils expose an alternative substrate for annexin-V different from PS and not recognized by macrophages, excluding early clearance as an explanation for the neutropeni
    corecore