首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 27 毫秒
1.
2.
Paneth cells are a secretory epithelial lineage that release dense core granules rich in host defense peptides and proteins from the base of small intestinal crypts. Enteric α-defensins, termed cryptdins (Crps) in mice, are highly abundant in Paneth cell secretions and inherently resistant to proteolysis. Accordingly, we tested the hypothesis that enteric α-defensins of Paneth cell origin persist in a functional state in the mouse large bowel lumen. To test this idea, putative Crps purified from mouse distal colonic lumen were characterized biochemically and assayed in vitro for bactericidal peptide activities. The peptides comigrated with cryptdin control peptides in acid-urea-PAGE and SDS-PAGE, providing identification as putative Crps. Matrix-assisted laser desorption ionization time-of-flight mass spectrometry experiments showed that the molecular masses of the putative α-defensins matched those of the six most abundant known Crps, as well as N-terminally truncated forms of each, and that the peptides contain six Cys residues, consistent with identities as α-defensins. N-terminal sequencing definitively revealed peptides with N termini corresponding to full-length, (des-Leu)-truncated, and (des-Leu-Arg)-truncated N termini of Crps 1–4 and 6. Crps from mouse large bowel lumen were bactericidal in the low micromolar range. Thus, Paneth cell α-defensins secreted into the small intestinal lumen persist as intact and functional forms throughout the intestinal tract, suggesting that the peptides may mediate enteric innate immunity in the colonic lumen, far from their upstream point of secretion in small intestinal crypts.Antimicrobial peptides (AMPs)2 are released by epithelial cells onto mucosal surfaces as effectors of innate immunity (15). In mammals, most AMPs derive from two major families, the cathelicidins and defensins (6). The defensins comprise the α-, β-, and θ-defensin subfamilies, which are defined by the presence of six cysteine residues paired in characteristic tridisulfide arrays (7). α-Defensins are highly abundant in two primary cell lineages: phagocytic leukocytes, primarily neutrophils, of myeloid origin and Paneth cells, which are secretory epithelial cells located at the base of the crypts of Lieberkühn in the small intestine (810). Neutrophil α-defensins are stored in azurophilic granules and contribute to non-oxidative microbial cell killing in phagolysosomes (11, 12), except in mice whose neutrophils lack defensins (13). In the small bowel, α-defensins and other host defense proteins (1418) are released apically as components of Paneth cell secretory granules in response to cholinergic stimulation and after exposure to bacterial antigens (19). Therefore, the release of Paneth cell products into the crypt lumen is inferred to protect mitotically active crypt cells from colonization by potential pathogens and confer protection against enteric infection (7, 20, 21).Under normal, homeostatic conditions, Paneth cells are not found outside the small bowel, although they may appear ectopically in response to local inflammation throughout the gastrointestinal tract (22, 23). Paneth cell numbers increase progressively throughout the small intestine, occurring at highest numbers in the distal ileum (24). Mouse Paneth cells express numerous α-defensin isoforms, termed cryptdins (Crps) (25), that have broad spectrum antimicrobial activities (6, 26). Collectively, α-defensins constitute approximately seventy percent of the bactericidal peptide activity in mouse Paneth cell secretions (19), selectively killing bacteria by membrane-disruptive mechanisms (2730). The role of Paneth cell α-defensins in gastrointestinal mucosal immunity is evident from studies of mice transgenic for human enteric α-defensin-5, HD-5, which are immune to infection by orally administered Salmonella enterica sv. typhimurium (S. typhimurium) (31).The biosynthesis of mature, bactericidal α-defensins from their inactive precursors requires activation by lineage-specific proteolytic convertases. In mouse Paneth cells, inactive ∼8.4-kDa Crp precursors are processed intracellularly into microbicidal ∼4-kDa Crps by specific cleavage events mediated by matrix metalloproteinase-7 (MMP-7) (32, 33). MMP-7 null mice exhibit increased susceptibility to systemic S. typhimurium infection and decreased clearance of orally administered non-invasive Escherichia coli (19, 32). Although the α-defensin proregions are sensitive to proteolysis, the mature, disulfide-stabilized peptides resist digestion by their converting enzymes in vitro, whether the convertase is MMP-7 (32), trypsin (34), or neutrophil serine proteinases (35). Because α-defensins resist proteolysis in vitro, we hypothesized that Paneth cell α-defensins resist degradation and remain in a functional state in the large bowel, a complex, hostile environment containing varied proteases of both host and microbial origin.Here, we report on the isolation and characterization of a population of enteric α-defensins from the mouse colonic lumen. Full-length and N-terminally truncated Paneth cell α-defensins were identified and are abundant in the distal large bowel lumen.  相似文献   

3.
The mechanisms of free fatty acid-induced lipoapoptosis are incompletely understood. Here we demonstrate that Mcl-1, an anti-apoptotic member of the Bcl-2 family, was rapidly degraded in hepatocytes in response to palmitate and stearate by a proteasome-dependent pathway. Overexpression of a ubiquitin-resistant Mcl-1 mutant in Huh-7 cells attenuated palmitate-mediated Mcl-1 loss and lipoapoptosis; conversely, short hairpin RNA-targeted knockdown of Mcl-1 sensitized these cells to lipoapoptosis. Palmitate-induced Mcl-1 degradation was attenuated by the novel protein kinase C (PKC) inhibitor rottlerin. Of the two human novel PKC isozymes, PKCδ and PKCθ, only activation of PKCθ was observed by phospho-immunoblot analysis. As compared with Jurkat cells, a smaller PKCθ polypeptide and mRNA were expressed in hepatocytes consistent with an alternative splice variant. Short hairpin RNA-mediated knockdown of PKCθ reduced Mcl-1 degradation and lipoapoptosis. Likewise, genetic deletion of Pkcθ also attenuated Mcl-1 degradation and cytotoxicity by palmitate in primary hepatocytes. During treatment with palmitate, rottlerin inhibited phosphorylation of Mcl-1 at Ser159, a phosphorylation site previously implicated in Mcl-1 turnover. Consistent with these results, an Mcl-1 S159A mutant was resistant to degradation and improved cell survival during palmitate treatment. Collectively, these results implicate PKCθ-dependent destabilization of Mcl-1 as a mechanism contributing to hepatocyte lipoapoptosis.Current evidence suggests that hepatic steatosis is present in up to 30% of the American population (1). A subset of these individuals develop severe hepatic lipotoxicity, a syndrome referred to as NASH2 (2), which can progress to cirrhosis and its chronic sequela (3, 4). A major risk factor for hepatic lipotoxicity is insulin resistance (57), resulting in excessive lipolysis within peripheral adipose tissue with release of high levels of free fatty acids (FFA) to the circulation. Circulating FFA are taken up by the liver via fatty acid transporter 5 and CD36 (810), and the bulk of hepatic neutral fat is derived from re-esterification of circulating FFA (8). Current concepts indicate that FFA, and not their esterified product (triglyceride), mediate hepatic lipotoxicity (11, 12). Elevated serum FFA correlate with liver disease severity (1315), and therapies that enhance insulin sensitivity ameliorate hepatic lipotoxicity, in part, by decreasing plasma FFA (16). Hepatic FFA also accumulate in experimental steatohepatitis, further supporting a role for these nutrients in hepatic lipotoxicity (17). Saturated FFA are more strongly implicated in hepatic lipotoxicity than unsaturated FFA (18, 19). Saturated FFA induce hepatocyte apoptosis (20, 21), a cardinal feature of nonalcoholic fatty liver disease (22), and serum biomarkers of apoptosis are useful for identifying hepatic lipotoxicity (23). Thus, FFA-mediated lipotoxicity occurs, in part, by apoptosis.Apoptosis is regulated by members of the Bcl-2 protein family (24). These proteins can be categorized into three subsets as follows: the guardians or anti-apoptotic members of this family, which include Bcl-2, A1, Mcl-1, Bcl-xL, and Bcl-w; the multidomain executioners or proapoptotic members of this family, which include Bax and Bak; and the messengers or biosensors of cell death, which share only the third Bcl-2 homology domain and are referred to as BH3-only proteins. This last group of proteins includes Bid, Bim, Bmf, Puma, Noxa, Hrk, Bad, and Bik. We have previously reported that cytotoxic FFA induce Bim expression by a FoxO3a-dependent mechanism that contributes, in part, to lipoapoptosis by activating Bax (20, 21). However, Bax activation can be held in check by anti-apoptotic members of the Bcl-2 family suggesting their function may also be dysregulated during FFA-mediated cytotoxicity.Bcl-2 is not expressed in hepatocytes at the protein level (25), whereas Bcl-w and Bfl-1/A1 knock-out mice have no liver phenotype (2628). However, both potent anti-apoptotic proteins Bcl-xL and Mcl-1 are expressed by hepatocytes and exhibit a liver phenotype in knock-out mice (29, 30), whereas up-regulation of Mcl-1 renders hepatocytes resistant to apoptosis (3133). It has also been posited that cellular elimination of Mcl-1 is a critical step in certain proapoptotic cascades (34, 35). Mcl-1 is unique among Bcl-2 proteins in that it has a short half-life, 30–120 min in most cell types, due to the presence of two sequences rich in proline, glutamic acid, serine, and threonine, which target the protein for rapid degradation by the proteasome (36). Proteasomal degradation of Mcl-1 is promoted by ubiquitination, which in turn is regulated by various kinase cascades (36). Despite its potential importance, a role for Mcl-1 in regulating hepatocyte FFA-mediated lipoapoptosis remains unexplored.Given that FFA induce insulin resistance (37), the kinases potentially regulating lipoapoptosis are likely those also identified in insulin resistance syndromes, especially the novel PKC isoforms PKCδ and PKCθ (38). The novel PKC isoforms are activated by diacylglycerol, which rises in the presence of FFA (3941), and diacylglycerol levels are significantly increased in NASH (42). A role for PKCδ in apoptosis has not been described. PKCθ has recently been shown to be activated by endoplasmic reticulum stress in liver cells (43) and lipids in vivo (44, 45). Furthermore, PKCθ has also been implicated in apoptosis of Jurkat cells, neuroblastoma cells, and myeloid leukemia cells (46, 47). However, neither its role in mediating lipoapoptosis nor modulating levels/activity of Bcl-2 proteins has been examined.This study addresses the role of Mcl-1 and PKCθ in FFA-induced lipoapoptosis. We identify a pathway that involves PKCθ-dependent proteasomal degradation of Mcl-1. Using inhibitors of various steps along this pathway, along with Mcl-1 mutants that are resistant to proteasomal degradation or Ser159 phosphorylation, our studies implicate Mcl-1 degradation via a PKCθ-dependent process as a critical step in lipoapoptosis.  相似文献   

4.
5.
A polyomavirus mutant (315YF) blocked in binding phosphatidylinositol 3-kinase (PI 3-kinase) has previously been shown to be partially deficient in transformation and to induce fewer tumors and with a significant delay compared to wild-type virus. The role of polyomavirus middle T antigen-activated PI 3-kinase in apoptosis was investigated as a possible cause of this behavior. When grown in medium containing 1d-3-deoxy-3-fluoro-myo-inositol to block formation of 3′-phosphorylated phosphatidylinositols, F111 rat fibroblasts transformed by wild-type polyomavirus (PyF), but not normal F111 cells, showed a marked loss of viability with evidence of apoptosis. Similarly, treatment with wortmannin, an inhibitor of PI 3-kinase, stimulated apoptosis in PyF cells but not in normal cells. Activation of Akt, a serine/threonine kinase whose activity has been correlated with regulation of apoptosis, was roughly twofold higher in F111 cells transformed by either wild-type virus or mutant 250YS blocked in binding Shc compared to cells transformed by mutant 315YF. In the same cells, levels of apoptosis were inversely correlated with Akt activity. Apoptosis induced by serum withdrawal in Rat-1 cells expressing a temperature-sensitive p53 was shown to be at least partially p53 independent. Expression of either wild-type or 250YS middle T antigen inhibited apoptosis in serum-starved Rat-1 cells at both permissive and restrictive temperatures for p53. Mutant 315YF middle T antigen was partially defective for inhibition of apoptosis in these cells. The results indicate that unlike other DNA tumor viruses which block apoptosis by inactivation of p53, polyomavirus achieves protection from apoptotic death through a middle T antigen–PI 3-kinase–Akt pathway that is at least partially p53 independent.Programmed cell death occurs during normal development and under certain pathological conditions. In mammalian cells, apoptosis can be induced by a variety of stimuli, including DNA damage (45), virus infection (54, 57), oncogene activation (25), and serum withdrawal (34, 37). Apoptosis can also be blocked by a number of factors, including adenovirus E1B 55- or 19-kDa proteins (9, 16), baculovirus p35 and iap genes (10), Bcl-2 (36, 61), and survival factors (12, 21). DNA tumor viruses have evolved mechanisms that both trigger and inhibit apoptosis. These frequently involve binding and inactivation of tumor suppressor proteins. E7 in some papillomaviruses (22), E1A in adenovirus (31, 43, 64), and large T antigen in simian virus 40 (SV40) (17) bind Rb and/or p300 and lead to upregulation of p53, which is thought to trigger apoptosis in virus-infected cells. The same viruses also inhibit apoptosis by inactivating p53 by various mechanisms (44, 63, 67). In contrast, the mechanism by which polyomavirus interacts with apoptotic pathways in the cell is not known; no direct interaction with p53 by any of the proteins encoded by this virus has been demonstrated (19, 62).The principal oncoprotein of polyomavirus is the middle T antigen. Neoplastic transformation by polyomavirus middle T antigen has as a central feature its association with and activation of members of the Src family of tyrosine kinases p60c-src (13) and p62c-yes (42). The major known consequence of these interactions is phosphorylation of middle T antigen on specific tyrosine residues creating binding sites for other signaling proteins. Phosphorylation at tyrosines 250, 315, and 322 promotes binding to Shc (18), the p85 regulatory subunit of phosphatidylinositol 3-kinase (PI 3-kinase) (59), and phospholipase Cγ-1 (58), respectively. Recognition of multiple signaling pathways emanating from middle T antigen has led to a keen interest in identifying their downstream biochemical effects, which collectively lead to the emergence of neoplastic transformation and presumably underlie the dramatic ability of the virus to induce many kinds of tumors in the mouse.Previous work has shown that the binding of PI 3-kinase to middle T antigen is essential for full transformation of rat fibroblasts in culture (8) and for rapid development of a broad spectrum of tumors in mice (30), for translocation of the GLUT1 transporter (68), and activation of p70 S6 kinase (14). While the mutant 315YF (blocked in PI 3-kinase activation) was able to induce some tumors, it did so at reduced frequencies and with an average latency three times longer than that of either the wild-type virus or a mutant, 250YS, blocked in binding Shc (4, 30). Recent studies have indicated a role of PI 3-kinase in blocking apoptosis in nonviral systems. Growth factor receptors acting through protein tyrosine kinases may prevent apoptosis by activating PI 3-kinase in PC12 cells, T lymphocytes, hematopoietic progenitors, and rat fibroblasts (7, 48, 56, 65, 66). The failure of mutant 315YF to induce full transformation of cells in culture and to induce the rapid development of tumors in mice could therefore be related, at least in part, to a failure to block apoptosis. In this study, we focus on the question of whether middle T antigen–PI 3-kinase interaction is involved in blocking apoptosis in cells transformed by polyomavirus.  相似文献   

6.
7.
Ligation of major histocompatability complex class I (MHC-I) molecules expressed on T cells leads to both growth arrest and apoptosis. The aim of the current study was to investigate the intracellular signal pathways that mediate these effects.MHC-I ligation of human Jurkat T cells induced a morphologically distinct form of apoptosis within 6 h. A specific caspase inhibitor, which inhibited Fas-induced apoptosis, did not affect apoptosis induced by MHC-I ligation. Furthermore, MHC-I–induced apoptosis did not involve cleavage and activation of the poly(ADP- ribose) polymerase (PARP) endonuclease or degradation of genomic DNA into the typical fragmentation ladder, both prominent events of Fas-induced apoptosis. These results suggest that MHC-I ligation of Jurkat T cells induce apoptosis through a signal pathway distinct from the Fas molecule.In our search for other signal pathways leading to apoptosis, we found that the regulatory 85-kD subunit of the phosphoinositide-3 kinase (PI-3) kinase was tyrosine phosphorylated after ligation of MHC-I and the PI-3 kinase inhibitor wortmannin selectively blocked MHC-I–, but not Fas-induced, apoptosis. As the c-Jun NH2-terminal kinase (JNK) can be activated by PI-3 kinase activity, and has been shown to be involved in apoptosis of lymphocytes, we examined JNK activation after MHC-I ligation. Strong JNK activity was observed after MHC-I ligation and the activity was completely blocked by wortmannin. Inhibition of JNK activity, by transfecting cells with a dominant-negative JNKK– MKK4 construct, led to a strong reduction of apoptosis after MHC-I ligation. These results suggest a critical engagement of PI-3 kinase–induced JNK activity in apoptosis induced by MHC-I ligation.Apoptosis is an active form of cell death associated with certain characteristic morphological changes of the cell. These include cell shrinkage, condensation of chromatin, and usually, but not always, fragmentation of genomic DNA into specific oligonucleosomal fragments, also referred to as apoptotic DNA ladder (21). In addition, a morphologically distinct form of apoptosis has been described in germinal centers, thymocyte suspensions, and certain tumors with characteristic features of type B dark cells (7, 34). The condensed chromatin in these cells is not smoothly redistributed into the characteristic eye seen in the nucleus of classical apoptosis; the cytoplasm is darkened and the mitochondria and endoplasmatic reticulum tend to be swollen (7, 34).The mammalian interleukin-1β–convertase enzyme (ICE)1 protease family (caspases) are known to be critically involved in Fas- and tumor necrosis factor α–induced apoptosis (12). All caspases share two features: (a) they are synthesized as proenzymes and they are activated by cleavage at specific aspartate residues, and (b) both have the same consensus sequence as their own protease activity (14). Thus, the caspases are presumed to be regulated within a hierarchy of auto- and trans-cleavage. In Fas- mediated apoptosis, a sequential activation of caspase 1 (ICE) and caspase 3 (CPP32) has been demonstrated (13). Recent evidence suggests that one subfamily of caspases (inhibitable by YVAD pseudo-substrate) works proximally in Fas-induced apoptosis, leading to release of cytochrome C and other factors from the mitochondria. The initially activated caspases, together with the released factors from the mitochondria, activate the effector caspases (inhibitable by DEVD pseudo-substrate) by a mechanism that is not well defined (22). One of the targets of the effector caspases is the nuclear enzyme poly(ADP-ribose) polymerase (PARP) involved in DNA damage sensing. PARP activity is thought to be critical for apoptosis induced through caspase 3 (5, 43). The PARP enzyme is a 116-kD protein that is cleaved into a 85-kD fragment upon activation (43).Aurintricarboxylic acid (ATA), an inhibitor of Ca2+- dependent endonuclease activity (23, 29), has been shown to inhibit apoptosis that occurs without DNA fragmentation (25, 32). The mechanism by which ATA inhibits apoptosis is not fully understood. However, inhibition of endonuclease activity may not be the only function of ATA; rather, inhibition of topoisomerase II that induces chromatin condensation during apoptosis seems to be important (6). ATA has also been shown to inhibit the Ca2+-activated enzyme calpain, which may be involved in apoptosis (33).A new member of the mitogen-activated protein kinase (MAPK) superfamily designated c-Jun NH2-terminal kinase (JNK), has recently been identified (16). A signal pathway functionally independent from extracellular signal-regulated kinase (ERK), which involves JNKK–MKK4, activates JNK (11, 49, 52). JNK is activated by dual phosphorylation of a Thr-Pro-Tyr motif during apoptosis induced by UV light, heat shock, and ligation of the Fas antigen (8, 17, 45, 50). Costimulation of T cells with T cell antigen receptor complex (TCR–CD3) and CD28 ligation, or CD40 ligation of B cells also results in activation of JNK (36, 41). Thus, JNK activity is involved in processes leading to both cell death and differentiation/activation. It has been demonstrated that apoptosis can be regulated through a balance between members of the MAPK superfamily; e.g., high JNK and low ERK activities may lead to apoptosis, whereas high JNK and ERK activities prevent apoptosis (17, 51).Stimulation of T cells through the major histocompatability complex class I (MHC-I) molecule initiates a cascade of biochemical changes that can lead to either activation and growth or cell cycle arrest and apoptosis (1, 4, 31, 40, 44). One of the critical events that occurs in both cases is the activation of tyrosine kinases, resulting in tyrosine phosphorylation of a variety of proteins including phospholipase C-γ1 (PLC-γ1) and ZAP70 (38, 39). We have recently shown that tyrosine kinase activity appears to be critical for growth inhibition and apoptosis induced by ligation of MHC-I molecules (4, 38). The functional outcome of MHC-I ligation is tightly linked to regulation of peripheral T cell activity and tolerance induction (37, 42).In the present study we have investigated the intracellular signal pathway leading to apoptosis after MHC-I ligation of T cells, in an attempt to find a cause-and-effect relationship between the biochemical and functional consequences of MHC-I ligation. We present evidence that MHC-I induces apoptosis through a distinct pathway involving phosphoinositide-3 (PI-3) kinase–induced JNK activity.  相似文献   

8.
9.
10.
11.
12.
13.
14.
Sphingolipids are important signaling molecules in many biological processes, but little is known regarding their physiological roles in the mitochondrion. We focused on the biochemical characters of a novel sphingomyelinase (SMase) and its function in mitochondrial cer a mide generation in zebrafish embryonic cells. The cloned SMase cDNA encoded a polypeptide of 545 amino acid residues (putative molecular weight, 61,300) containing a mitochondrial localization signal (MLS) and a predicted transmembrane domain. The mature endogenous enzyme was predicted to have a molecular weight of 57,000, and matrix-assisted laser de sorp tion ionization time-of-flight mass spectrometry analysis indicated that the N-terminal amino acid residue of the mature enzyme was Ala-36. The purified enzyme optimally hydrolyzed [14C]sphingomyelin in the presence of 10 mm Mg2+ at pH 7.5. In HEK293 cells that overexpressed SMase cDNA, the enzyme was localized to the mitochondrial fraction, whereas mutant proteins lacking MLS or both the MLS and the transmembrane domain were absent from the mitochondrial fraction. Endogenous SMase protein co-localized with a mitochondrial cytostaining marker. Using a protease protection assay, we found that SMase was distributed throughout the intermembrane space and/or the inner membrane of the mitochondrion. Furthermore, the overexpression of SMase in HEK293 cells induced cer a mide generation and sphingomyelin hydrolysis in the mitochondrial fraction. Antisense phosphorothioate oligonucleotide-induced knockdown repressed cer a mide generation and sphingomyelin hydrolysis in the mitochondrial fraction in zebrafish embryonic cells. These observations indicate that SMase catalyzes the hydrolysis of sphingomyelin and generates cer a mide in mitochondria in fish cells.Sphingomyelinase (SMase,2 sphingomyelin phosphodiesterase, EC 3.1.4.12) hydrolyzes sphingomyelin and produces ceramide and phosphocholine. Ceramide plays an important role as a signaling molecule in cell proliferation, apoptosis, cell cycle arrest, differentiation, and the stress response in animal cells (15). To date, three distinct classes of acid, neutral, and alkaline SMases have been identified according to optimum pH, cation dependence, amino acid sequence, and subcellular localization (3).The Mg2+-dependent neutral SMases have emerged as major candidates in the mediation of ceramide-induced cell signaling (6). Recent research has identified at least three distinct neutral SMases in human and mouse, designated as neutral SMase 1, SMase 2, and SMase 3 (79). Neutral SMase 1 was the first SMase identified in human and mouse. Although mammalian enzymes exhibited Mg2+-dependent neutral SMase activity in vitro (9), no significant biological functions in sphingomyelin and ceramide metabolism were identified in SMase 1-overexpressing cells (10) or neutral SMase 1 knock-out mice (11). In zebrafish embryos, Mg2+-dependent neutral SMase 1 produced ceramide and caused thalidomide-induced vascular defects (12). In addition, SMase 1 was found to mediate heat-induced ceramide generation and apoptosis (13).The neutral SMase 2 gene SMPD3, has also been identified based on its similarity to Bacillus cereus SMase DNA sequences (7). This gene encodes a membrane-bound protein expressed in the brain and liver that has two highly hydrophobic segments near the N-terminal region, both of which are thought to function as transmembrane domains. Unlike neutral SMase 1, neutral SMase 2 possesses Mg2+-dependent neutral SMase activity in vivo in MCF-7 cells (14). When overexpressed in the confluent phase of MCF-7 cells, mouse neutral SMase 2 was palmitoylated via thioester bonds and localized in the inner leaflet of the plasma membrane (15). In MCF-7 cells stably expressing neutral SMase 2, the enzyme inhibited cell growth and was required for cells to undergo confluence-induced cell cycle arrest (16). Interestingly, neutral SMase 2 was isolated as the confluent 3Y1 cell-associated 1 gene (cca1) in rat 3Y1 cells (17). Neutral SMase 2 has been implicated in signal transduction events in cell growth and the cellular response to cytokines (18, 19), oxidative stress (20), and amyloid β-peptide (21).Stoffel et al. (22) demonstrated that gene-targeted mice deficient for neutral SMase 2 developed a novel form of dwarfism and had delayed puberty as part of a hypothalamus-induced pituitary hormone deficiency. Strikingly, positional cloning of the recessive mutation fragilitas ossium in mice identified a deletion in the gene that encodes neutral SMase 2, leading to the complete loss of neutral SMase activity (23). The mutant fragilitas ossium mice develop severe osteogenesis and dentinogenesis imperfecta, with no collagen defect. Thus, mouse neutral SMase 2 is essential for late embryonic and postnatal development.Mitochondria contain small amounts of a variety of sphingolipids, including ceramide and sphingomyelin (2426), which may be derived from the endoplasmic reticulum via intimate membrane contacts or produced in response to apoptosis. For mitochondria isolated from HL-60 cells, treatment with ceramide inhibited the mitochondrial respiratory chain complex III (27). Birbes et al. (28) found that the selective hydrolysis of a mitochondrial pool of sphingomyelin induced apoptosis. They transfected MCF-7 cells with B. cereus SMase targeted to various subcellular organelles, but they observed cytochrome c release and apoptosis induction only when the enzyme was targeted to the mitochondria. Ceramide activated the mitochondrial protein phosphatase 2A, which dephosphorylated Bcl-2 and led to apoptosis (29). In MCF-7 cells, mitochondrial ceramide generation in response to tumor necrosis factor-α induced Bax translocation to mitochondria and subsequent cytochrome c release and apoptosis (30). The permeability of the mitochondrial outer membrane correlates directly with the level of ceramide in the membrane (31). The concentration of ceramide at which significant channel formation occurs is consistent with the level of mitochondrial ceramide that occurs during the induction phase of apoptosis (31). In isolated mitochondria, ceramide can also form membrane channels large enough to release cytochrome c and other small proteins (32). Ceramide-metabolizing enzymes, such as a bovine liver ceramide synthase (33) and human ceramidase (34), are localized to the mitochondrion. These observations suggest the existence of a mitochondrial pool of sphingomyelin and the function of a sphingomyelin-specific metabolic pathway in mitochondria. However, no SMase has been identified in mitochondria.We identified and examined the biochemical properties of a novel SMase localized to the zebrafish mitochondrion. The enzyme was cloned from a cDNA library of embryonic zebrafish cells. It was found to regulate mitochondrial ceramide levels.  相似文献   

15.
16.
Human immunodeficiency virus infection is characterized by a progressive decline in the number of peripheral blood CD4+ T lymphocytes, which finally leads to AIDS. This T-cell decline correlates with the degree of in vitro-induced lymphocyte apoptosis. However, such a correlation has not yet been described in feline AIDS, caused by feline immunodeficiency virus (FIV) infection. We therefore investigated the intensity of in vitro-induced apoptosis in peripheral blood lymphocytes from cats experimentally infected with a Swiss isolate of FIV for 1 year and for 6 years and from a number of long-term FIV-infected cats which were coinfected with feline leukemia virus. Purified peripheral blood lymphocytes were either cultured overnight under nonstimulating conditions or stimulated with phytohemagglutinin and interleukin-2 for 60 h. Under stimulating conditions, the isolates from the infected cats showed significantly higher relative counts of apoptotic cells than did those from noninfected controls (1-year-infected cats, P = 0.01; 6-year-infected cats, P = 0.006). The frequency of in vitro-induced apoptosis was inversely correlated with the CD4+ cell count (P = 0.002), bright CD8+ cell count (P = 0.009), and CD4/CD8 ratio (P = 0.01) and directly correlated with the percentage of bright major histocompatibility complex class II-positive peripheral blood lymphocytes (P = 0.004). However, we found no correlation between in vitro-induced apoptosis and the viral load in serum samples. Coinfection with feline leukemia virus enhanced the degree of in vitro-induced apoptosis compared with that in FIV monoinfected cats. We concluded that the degree of in vitro-induced apoptosis was closely related to FIV-mediated T-cell depletion and lymphocyte activation and could be used as an additional marker for disease progression in FIV infection.Feline immunodeficiency virus (FIV) infection is a naturally occurring infection, and disease progression in infected cats is associated with a decline in the number of CD4+ cells (2, 6, 22, 23, 36), a loss of bright CD8+ cells in the advanced stage of the disease (22), an increased number of activated T cells (39, 41), and a changed cytokine production, i.e., decreased production of interleukin-2 (IL-2) and concomitantly increased production of tumor necrosis factor alpha (TNF-α) (25, 26). The increased production of TNF-α has been reported to induce apoptosis in chronically FIV-infected cells (38). Apoptosis, a controlled mode of cell death (34), plays an important role in the regulation of immune responses (5, 14). As described for FIV (23), the hallmark of human immunodeficiency virus (HIV)-induced disease is the loss of T-helper cells (31, 43). Theoretically, cell loss can be caused by decreased production of cells, increased destruction, or a combination of the two mechanisms. Findings of an early infection of thymocytes followed by pathologic changes in the thymus support the model of decreased T-helper cell production triggered by HIV (13) and FIV (52). The destruction model is supported by findings of an increased number of peripheral blood T cells undergoing apoptosis upon HIV (20, 32) and FIV (11, 21, 33) infection. However, increased CD4+-T-cell turnover may not be the main cause of the observed T-helper cell decline in HIV-1 infection, as reviewed by others (44, 51). In addition, the degree of HIV-induced apoptosis correlates with the T-helper cell decline and disease progression (19, 40). However, such a relationship has not yet been described for FIV. It has been reported that cross-linking of CD4 molecules by HIV gp160 triggers apoptosis in noninfected CD4+ T cells (1). Investigation of this aspect in the feline system is especially interesting since FIV does not use the feline homologue of CD4 (50).The aim of the present study was to compare the degree of in vitro-induced lymphocyte apoptosis in FIV-infected cats with normal and decreased T-helper cell counts. We used two different culture conditions to trigger apoptosis in vitro: cells were either cultured overnight under nonstimulating conditions and in the absence of growth factors or cultured for 60 h in the presence of phytohemagglutinin, IL-2, and fetal calf serum. We additionally examined cats which were coinfected with the feline leukemia virus (FeLV). This coinfection is known to accelerate the progression toward feline AIDS (23) by an unknown mechanism (8).  相似文献   

17.
18.
Aldo-keto reductase family 1 member B10 (AKR1B10) is primarily expressed in the normal human colon and small intestine but overexpressed in liver and lung cancer. Our previous studies have shown that AKR1B10 mediates the ubiquitin-dependent degradation of acetyl-CoA carboxylase-α. In this study, we demonstrate that AKR1B10 is critical to cell survival. In human colon carcinoma cells (HCT-8) and lung carcinoma cells (NCI-H460), small-interfering RNA-induced AKR1B10 silencing resulted in caspase-3-mediated apoptosis. In these cells, the total and subspecies of cellular lipids, particularly of phospholipids, were decreased by more than 50%, concomitant with 2–3-fold increase in reactive oxygen species, mitochondrial cytochrome c efflux, and caspase-3 cleavage. AKR1B10 silencing also increased the levels of α,β-unsaturated carbonyls, leading to the 2–3-fold increase of cellular lipid peroxides. Supplementing the HCT-8 cells with palmitic acid (80 μm), the end product of fatty acid synthesis, partially rescued the apoptosis induced by AKR1B10 silencing, whereas exposing the HCT-8 cells to epalrestat, an AKR1B10 inhibitor, led to more than 2-fold elevation of the intracellular lipid peroxides, resulting in apoptosis. These data suggest that AKR1B10 affects cell survival through modulating lipid synthesis, mitochondrial function, and oxidative status, as well as carbonyl levels, being an important cell survival protein.Aldo-keto reductase family 1 member B10 (AKR1B10,2 also designated aldose reductase-like-1, ARL-1) is primarily expressed in the human colon, small intestine, and adrenal gland, with a low level in the liver (13). However, this protein is overexpressed in hepatocellular carcinoma, cervical cancer, lung squamous cell carcinoma, and lung adenocarcinoma in smokers, being a potential diagnostic and/or prognostic marker (1, 2, 46).The biological function of AKR1B10 in the intestine and adrenal gland, as well as its role in tumor development and progression, remains unclear. AKR1B10 is a monomeric enzyme that efficiently catalyzes the reduction to corresponding alcohols of a range of aromatic and aliphatic aldehydes and ketones, including highly electrophilic α,β-unsaturated carbonyls and antitumor drugs containing carbonyl groups, with NADPH as a co-enzyme (1, 712). The electrophilic carbonyls are constantly produced by lipid peroxidation, particularly in oxidative conditions, and are highly cytotoxic; through interaction with proteins, peptides, and DNA, the carbonyls cause protein dysfunction and DNA damage (breaks and mutations), resulting in mutagenesis, carcinogenesis, or apoptosis (10, 1319). AKR1B10 also shows strong enzymatic activity toward all-trans-retinal, 9-cis-retinal, and 13-cis-retinal, reducing them to the corresponding retinols, which may regulate the intracellular retinoic acid, a signaling molecule modulating cell proliferation and differentiation (6, 2023). In lung cancer, AKR1B10 expression is correlated with the patient smoking history and activates procarcinogens in cigarette smoke, such as polycyclic aromatic hydrocarbons, thus involved in lung tumorigenesis (2426).Recent studies have shown that in breast cancer cells, AKR1B10 associates with acetyl-CoA carboxylase-α (ACCA) and blocks its ubiquitination and proteasome degradation (27). ACCA is a rate-limiting enzyme of de novo synthesis of long chain fatty acids, catalyzing the ATP-dependent carboxylation of acetyl-CoA to form malonyl-CoA (28). Long chain fatty acids are the building blocks of biomembranes and the precursor of lipid second messengers, playing a critical role in cell growth and proliferation (29, 30). Therefore, ACCA activity is tightly regulated by both metabolite-mediated allosteric mechanisms and phosphorylation-dependent mechanisms; the latter are controlled by multiple hormones, such as insulin, glucagon, and growth factors (3133). ACCA activity is also regulated through physical protein-protein interaction. For instance, breast cancer 1 (BRCA1) protein associates with the ACCA and blocks its Ser79 residue from dephosphorylation (34, 35). The AKR1B10-mediated regulation on ACCA stability represents a novel regulatory mechanism, and this current study elucidated the biological significance of this regulation. The results show that AKR1B10 promotes cell survival via modulating lipid synthesis, mitochondrial function and oxidative stress, and carbonyl levels.  相似文献   

19.
20.
设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号