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 37 毫秒
1.
Caspases, a unique family of cysteine proteases involved in cytokine activation and in the execution of apoptosis can be sub-grouped according to the length of their prodomain. Long prodomain caspases such as caspase-8 and caspase-9 are believed to act mainly as upstream caspases to cleave downstream short prodomain caspases such as caspases-3 and -7. We report here the identification of caspases as direct substrates of calcium-activated proteases, calpains. Calpains cleave caspase-7 at sites distinct from those of the upstream caspases, generating proteolytically inactive fragments. Caspase-8 and caspase-9 can also be directly cleaved by calpains. Two calpain cleavage sites in caspase-9 have been identified by N-terminal sequencing of the cleaved products. Cleavage of caspase-9 by calpain generates truncated caspase-9 that is unable to activate caspase-3 in cell lysates. Furthermore, direct cleavage of caspase-9 by calpain blocks dATP and cytochrome-c induced caspase-3 activation. Therefore our results suggest that calpains may act as negative regulators of caspase processing and apoptosis by effectively inactivating upstream caspases.  相似文献   

2.
Cleavage of structural proteins by caspases has been associated with the severe morphological changes occurring during the apoptotic process. One of the proteins regulating the connection of the actin filament with cadherins in a cell-cell adhesion complex is beta-catenin. During apoptosis, both an N-terminal and a small C-terminal part are removed from beta-catenin. Removal of the N-terminal part may result in a disconnection of the actin filament from a cadherin cell-cell adhesion complex. We demonstrate that caspase-8, -3 and -6 directly proteolyse beta-catenin in vitro. However, the beta-catenin cleavage products generated by caspase-8 were different from those generated by caspase-3 or caspase-6. Caspase-1, -2, -4/11 and -7 did not or only very inefficiently cleave beta-catenin. These data suggest that activation of procaspase-3, -6 or -8 by different stimuli in the cell might result in a differential proteolysis of beta-catenin.  相似文献   

3.
In the previous reports, we showed that the familial Alzheimer's disease (AD)-linked presenilin-1 (PS1) mutation induced the fragility to the endoplasmic reticulum (ER) stress and that caspase-4 mediates ER stress-induced- and beta-amyloid induced-apoptotic signaling in human cells. These results suggest the involvement of ER stress and caspase-4 in the cell death observed in AD. In this report, we studied the activation of caspase-4 in the familial AD-linked PS1 mutation (DeltaE9). Cleavage of caspase-4 under ER stress was enhanced by the overexpression of the familial AD-linked mutation (DeltaE9), showing that caspase-4 is a key caspase involved in the apoptotic signaling of AD. We also showed that the overexpression of caspase-4 induced cleavage of caspase-9 and caspase-3 without releasing cytochrome-c from the mitochondria. Thus, caspase-4 activates downstream caspases independently of mitochondrial apoptotic signaling and this might contribute to the pathogenesis of AD. To sum up our data, the familial AD-linked PS1 mutation accelerates the cleavage of caspase-4 under the ER stress and results in the activation of caspase-9 and caspase-3, apoptosis signal, without releasing cytochrome-c.  相似文献   

4.
Lesions in the parkin gene cause early onset Parkinson's disease by a loss of dopaminergic neurons, thus demonstrating a vital role for parkin in the survival of these neurons. Parkin is inactivated by caspase cleavage, and the major cleavage site is after Asp126. Caspases responsible for parkin cleavage were identified by several experimental paradigms. Transient coexpression of caspases and wild type parkin in HEK-293 cells identified caspase-1, -3, and -8 as efficient inducers of parkin cleavage whereas caspase-2, -7, -9, and -11 did not induce cleavage. A D126A parkin mutation abrogates cleavage induced by caspase-1 and -8, but not by caspase-3. In anti-Fas-treated Jurkat T cells, parkin cleavage was inhibited by caspase inhibitors hFlip and CrmA (but not by X-linked inhibitor of apoptosis (XIAP)), indicating that caspase-8 (but not caspase-3) is responsible for the parkin cleavage in this model. Moreover, induction of apoptosis in caspase-3-deficient MCF7 cells, either by caspase-1 or -8 overexpression or by tumor necrosis factor-alpha treatment, led to parkin cleavage. These results demonstrate that caspase-1 and -8 can directly cleave parkin and suggest that death receptor activation and inflammatory stress can cause loss of the ubiquitin ligase activity of parkin, thus causing accumulation of toxic parkin substrates and triggering dopaminergic cell death.  相似文献   

5.
In general, apoptotic stimuli lead to activation of caspases. Once activated, a caspase can induce intracellular signaling pathways involving proteolytic activation of other caspase family members. We report the in vitro processing of eight murine procaspases by their enzymatically active counterparts. Caspase-8 processed all procaspases examined. Caspase-1 and -11 processed the effector caspases procaspase-3 and -7, and to a lesser extent procaspase-6. However, vice versa, none of the caspase-1-like procaspases was activated by the effector caspases. This suggests that the caspase-1 subfamily members either act upstream of the apoptosis effector caspases or else are part of a totally separate activation pathway. Procaspase-2 was maturated by caspase-8 and -3, and to a lesser extent by caspase-7, while the active caspase-2 did not process any of the procaspases examined, except its own precursor. Hence, caspase-2 might not be able to initiate a wide proteolytic signaling cascade. Additionally, cleavage data reveal not only proteolytic amplification between caspase-3 and -8, caspase-6 and -3, and caspase-6 and -7, but also positive feedback loops involving multiple activated caspases. Our results suggest the existence of a hierarchic proteolytic procaspase activation network, which would lead to a dramatic increase in multiple caspase activities once key caspases are activated. The proteolytic procaspase activation network might allow that different apoptotic stimuli result in specific cleavage of substrates responsible for typical processes at the cell membrane, the cytosol, the organelles, and the nucleus, which characterize a cell dying by apoptosis.  相似文献   

6.
Caspases are proteases with a well-defined role in apoptosis. However, increasing evidence indicates multiple functions of caspases outside apoptosis. Caspase-1 and caspase-11 have roles in inflammation and mediating inflammatory cell death by pyroptosis. Similarly, caspase-8 has dual role in cell death, mediating both receptor-mediated apoptosis and in its absence, necroptosis. Caspase-8 also functions in maintenance and homeostasis of the adult T-cell population. Caspase-3 has important roles in tissue differentiation, regeneration and neural development in ways that are distinct and do not involve any apoptotic activity. Several other caspases have demonstrated anti-tumor roles. Notable among them are caspase-2, -8 and -14. However, increased caspase-2 and -8 expression in certain types of tumor has also been linked to promoting tumorigenesis. Increased levels of caspase-3 in tumor cells causes apoptosis and secretion of paracrine factors that promotes compensatory proliferation in surrounding normal tissues, tumor cell repopulation and presents a barrier for effective therapeutic strategies. Besides this caspase-2 has emerged as a unique caspase with potential roles in maintaining genomic stability, metabolism, autophagy and aging. The present review focuses on some of these less studied and emerging functions of mammalian caspases.  相似文献   

7.
Caspases orchestrate the controlled demise of a cell after an apoptotic signal through specific protease activity and cleavage of many substrates altering protein function and ensuring apoptosis proceeds efficiently. Comparing a variety of substrates of each apoptotic caspase (2, 3, 6, 7, 8, 9 and 10) showed that the cleavage sites had a general motif, sometimes specific for one caspase, but other times specific for several caspases. Using commercially available short peptide-based substrates and inhibitors the promiscuity for different cleavage motifs was indicated, with caspase-3 able to cleave most substrates more efficiently than those caspases to which the substrates are reportedly specific. In a cell-free system, immunodepletion of caspases before or after cytochrome c-dependent activation of the apoptosome indicated that the majority of activity on synthetic substrates was dependent on caspase-3, with minor roles played by caspases-6 and -7. Putative inhibitors of individual caspases were able to abolish all cytochrome c-induced caspase activity in a cell-free system and inhibit apoptosis in whole cells through the extrinsic and intrinsic pathways, raising issues regarding the use of such inhibitors to define relevant caspases and pathways. Finally, caspase activity in cells lacking caspase-9 displayed substrate cleavage activity of a putative caspase-9-specific substrate underlining the lack of selectivity of peptide-based substrates and inhibitors of caspases.  相似文献   

8.
Chang HC  Hsu C  Hsu HK  Yang RC 《IUBMB life》2003,55(7):403-407
We have previously shown that sphingosine increased caspase-3 activity and induced apoptosis in human hepatoma cells. Our data also suggest that other caspases may be involved in sphingosine-triggered apoptosis. In order to clarify this issue, we used different approaches to study the functional role of several initiator or executioner caspases in apoptosis induced by sphingosine. Activation of procaspases-2, -7, and -8, was clearly demonstrated during sphingosine-triggered apoptosis. Pretreatment with chemical inhibitors for caspase-7 and -8, attenuated apoptotic cell death induced by sphingosine. Conversely, pretreatment with specific caspase-2 inhibitor Z-VDVAD-FMK did not show any protective effect. In addition, enforced expression of constitutively activated AKT kinase which is known to inhibit apoptosis induced by sphingosine, potently suppressed activation of procaspases-7 and -8. In summary, these data suggest that in addition to caspases-3, caspase-7 and -8 are involved in the apoptosis induced by sphingosine.  相似文献   

9.
Inflammasomes are sensors that serve as activation platforms for caspase-1 — a mechanism that set the prevailing paradigm for inflammatory caspase activation. A recent Nature paper by Shi et al. upends this paradigm by describing an unprecedented model for caspase activation whereby caspase-4, -5, and -11 directly bind their agonist, cytosolic LPS, triggering auto-activation and subsequent pyroptotic cell death.The inflammatory caspases — among them caspase-1, murine caspase-11, and human caspase-4 and -5 (homologs of murine caspase-11) — are central to depriving infectious agents of intracellular replication niches. Upon responding to a given stimulus, they become catalytically active and initiate a form of programmed inflammatory cell death termed pyroptosis. By examining their homology and adjacent chromosomal arrangement in humans and other mammals, it is apparent that the inflammatory caspases originate from a series of gene duplications and subsequent divergences.A balanced caspase-1 response is critical to defense against a variety of infectious agents, whereas its aberrant activation underlies a number of immune pathologies. Less is known about caspase-11, -4, and -5 in infection; however, we have shown that one physiological role of caspase-11 is to detect and help clear cytosol invasive infections, such as those caused by Burkholderia thailandensis1. More recent work has shown that caspase-11 mediates resistance to DSS-induced colitis2 and clearance of Salmonella enterica serovar Typhimurium-infected cells in the intestinal epithelium3, perhaps limited to the times when these bacteria enter the cytosol. Likewise, caspase-4 responds to S. Typhimurium, enteropathogenic E. coli3, and Shigella flexneri4 infections in human intestinal epithelial cells. As with caspase-1, moderation of caspase-11 activity is key to limiting immune pathology: much of the lethality of bolus lipopolysaccharide (LPS) injection is mediated by caspase-115,6,7,8. Shedding light on the mechanisms underlying these observations, our lab and that of Dr Vishva Dixit independently determined that caspase-11 is activated in response to cytosolic LPS7,8; however, whether caspase-4 (and/or -5) functions similarly was not determined.Of the inflammatory caspases, the activation mechanism of caspase-1 is the best described. Via its N-terminal CARD domain and an adaptor protein called ASC, caspase-1 interacts with a family of cytosolic proteins, the inflammasomes, that detect signatures of infection (Figure 1). It then initiates pyroptosis and directs proinflammatory cytokine secretion. Inflammasomes thus follow the paradigm of apoptotic caspase activation, where apoptosis initiators caspase-2, -8 and -9 are recruited and activated by death domain family-containing upstream sensors: the piddosome, DISC, and apoptosome, respectively. Therefore, we and others assumed that the model of upstream sensor activating downstream caspase would hold for the other inflammatory caspases as well. For example, Kayagaki and colleagues coined the term ''noncanonical inflammasome pathway'' to describe activation of caspase-11 by a putative LPS sensor5,6. However, a recent elegant paper by Shi et al.9 proves this hypothesis wrong and describes an entirely novel paradigm of caspase activation. Moreover, the authors address many of the gaps in our understanding of caspase-11, -4, and -5 biology.Open in a separate windowFigure 1Schematic of canonical and noncanonical inflammasome pathways for inflammatory caspase activation. Left: Inflammasomes such as AIM2, NLRP3, and NLRC4 detect contamination of the cytosol with microbial ligands (e.g., DNA, flagellin, bacterial type 3 secretion system components) or certain cellular perturbations. Via the adaptor protein ASC, they subsequently activate caspase-1 (in the case of certain CARD-containing inflammasomes, such as NLRC4, direct interaction with caspase-1 can also occur), which initiates pyroptosis and secretion of the proinflammatory cytokines IL-1β and IL-18. Right: Caspase-4, -5, and -11 directly bind cytosolic LPS from Gram-negative bacteria. They subsequently oligomerize, activate, and initiate pyroptosis.Using electroporation to deliver bacterial components into the cytosol of cells, the authors first determined that caspase-4 responds to LPS in human monocytes by triggering pyroptosis. These findings extended to non-myeloid cells, where caspase-4 is constitutively expressed. The authors then demonstrated that caspase-4 and caspase-11 are functionally interchangable, supporting that they are homologs.Shi and colleagues next began identifying the molecule that actually binds LPS in the cytosol. They screened a number of NLRs and CARD domain-containing proteins, but no candidates emerged. In agreement with this, unpublished work from our lab also ruled out virtually all known CARD-containing proteins as the LPS sensor. Clues to the identity of the sensor arose from the following astute observations: First, Shi et al. noticed that both caspase-4 and caspase-11, when purified from E. coli, eluted from columns as large oligomers, suggesting activation, whereas they eluted as monomers when expressed in and purified from insect cells. Second, they found that the LPS contents of caspase-4 and -11 purified from E. coli were three orders of magnitude higher than what they typically observed when purifying bacterial proteins. Together, these results suggested that caspase-4 and -11 directly bind LPS. A series of pull-downs and surface plasmon resonance experiments confirmed this notion, revealing stable interaction of LPS with caspase-4 and -11 in cells transfected with LPS. Furthermore, the authors showed that caspase-5 similarly binds LPS. In all cases, LPS binding and caspase oligomerization was CARD domain dependent; indeed, purified caspase-4 and -11 CARD domains were sufficient to bind LPS and oligomerize. Three regions of basic residues in the caspase-11 CARD domain — mostly conserved in caspase-4 and -5, but not caspase-1 — were critical for LPS binding. Last, the authors determined that caspase-11 and -4 oligomerization stimulates activation, as measured by cleavage of a fluorogenic substrate. Interestingly, the known antagonists of caspase-11 activation Lipid IVa and atypical LPS from Rhodobacter sphaeroides bound caspase-4 and -11, but failed to induce oligomerization and activation.The Shi et al. paper brings to light a number of fascinating perspectives. First, binding of LPS by caspase-4, -5, and -11 establishes a new paradigm for caspase activation: Direct detection of a cell death-inducing ligand by a caspase. As the authors noted, this is analogous to horseshoe crab factors C and G, which bind LPS and β-(1,3)-D-glucan, respectively, and initiate coagulation cascades in haemolymphs.Second, the cell expression patterns of caspase-11, -4, and -5 may have important implications in future strategies for treating endotoxemia and Gram-negative sepsis. Caspase-11 expression is inducible in myeloid cells, where its basal expression is low; in contrast, caspase-4 appears to be constitutively expressed in human myeloid cells. Therefore, aberrant translocation of LPS into the cytosol of human myeoloid cells may not require priming to activate caspase-4 and initiate pyroptosis, perhaps sensitizing humans to the deleterious effects of LPS compared to mice. Investigating other cell type expression differences in this context will be informative.In answering so many questions about the biology of the inflammatory caspases, the work of Shi and colleagues raises many more. Among them: Why do antagonists of caspase-11 fail to induce oligomerization? How do the CARD domains of these caspases “see” LPS? During binding of LPS by MD2, the acyl chains of lipid A extend into the binding cleft of MD210 in a manner sensitive to acyl chain length; in contrast, caspase-11 detects very diverse lipid A acyl chain lengths and structures, such as those of Salmonella and Legionella species7,8,11, suggesting that the CARD domain may wrap around the lipid groups of LPS near the phosphate head groups of lipid A. Insights into these questions will surely come from crystal structures of caspase-11, -4, and -5 bound to various LPS structures.  相似文献   

10.
Myeloic cells express a peculiar surface receptor for extracellular ATP, called the P2Z/P2X7 purinoreceptor, which is involved in cell death signalling. Here, we investigated the role of caspases, a family of proteases implicated in apoptosis and the cytokine secretion. We observed that extracellular ATP induced the activation of multiple caspases including caspase-1, -3 and -8, and subsequent cleavage of the caspase substrates PARP and lamin B. Using caspase inhibitors, it was found that caspases were specifically involved in ATP-induced apoptotic damage such as chromatin condensation and DNA fragmentation. In contrast, inhibition of caspases only marginally affected necrotic alterations and cell death proceeded normally whether or not nuclear damage was blocked. Our results therefore suggest that the activation of caspases by the P2Z receptor is required for apoptotic but not necrotic alterations of ATP-induced cell death.  相似文献   

11.
In cells undergoing apoptosis, a 22-amino-acid presenilin-2-loop peptide (PS2-LP, amino acids 308-329 in presenilin-2) is generated through cleavage of the carboxyl-terminal fragment of presenilin-2 by caspase-3. The impact of PS2-LP on the progression of apoptosis, however, is not known. Here we show that PS2-LP is a potent inducer of the mitochondrial-dependent cell death pathway when transduced as a fusion protein with HIV-TAT. Biochemical and functional studies demonstrate that TAT-PS2-LP can interact with the inositol 1,4,5-trisphosphate receptor and activate Ca(2+) release from the endoplasmic reticulum. These results indicate that PS2-LP-mediated alteration of intracellular Ca(2+) homeostasis may be linked to the acceleration of apoptosis. Therefore, targeting the function of PS2-LP could provide a useful therapeutic tool for the treatment of cancer and degenerative diseases.  相似文献   

12.
13.
Inhibitor of apoptosis (IAP) gene products play an evolutionarily conserved role in regulating programmed cell death in diverse species ranging from insects to humans. Human XIAP, cIAP1 and cIAP2 are direct inhibitors of at least two members of the caspase family of cell death proteases: caspase-3 and caspase-7. Here we compared the mechanism by which IAPs interfere with activation of caspase-3 and other effector caspases in cytosolic extracts where caspase activation was initiated by caspase-8, a proximal protease activated by ligation of TNF-family receptors, or by cytochrome c, which is released from mitochondria into the cytosol during apoptosis. These studies demonstrate that XIAP, cIAP1 and cIAP2 can prevent the proteolytic processing of pro-caspases -3, -6 and -7 by blocking the cytochrome c-induced activation of pro-caspase-9. In contrast, these IAP family proteins did not prevent caspase-8-induced proteolytic activation of pro-caspase-3; however, they subsequently inhibited active caspase-3 directly, thus blocking downstream apoptotic events such as further activation of caspases. These findings demonstrate that IAPs can suppress different apoptotic pathways by inhibiting distinct caspases and identify pro-caspase-9 as a new target for IAP-mediated inhibition of apoptosis.  相似文献   

14.
Members of the caspase family of proteases play essential roles in the initiation and execution of apoptosis. These caspases are divided into two groups: the initiator caspases (caspase-2, -8, -9 and -10), which are the first to be activated in response to a signal, and the executioner caspases (caspase-3, -6, and -7) that carry out the demolition phase of apoptosis. Many conventional cancer therapies induce apoptosis to remove the cancer cell by engaging these caspases indirectly. Newer therapeutic applications have been designed, including those that specifically activate individual caspases using gene therapy approaches and small molecules that repress natural inhibitors of caspases already present in the cell. For such approaches to have maximal clinical efficacy, emerging insights into non-apoptotic roles of these caspases need to be considered. This review will discuss the roles of caspases as safeguards against cancer in the context of the advantages and potential limitations of targeting apoptotic caspases for the treatment of cancer.  相似文献   

15.
Phosphatidylinositol 3-kinase (PI3K) promotes cell survival and communication by activating its downstream effector Akt kinase. Here we show that PS1, a protein involved in familial Alzheimer's disease (FAD), promotes cell survival by activating the PI3K/Akt cell survival signaling. This function of PS1 is unaffected by gamma-secretase inhibitors. Pharmacological and genetic evidence indicates that PS1 acts upstream of Akt, at or before PI3K kinase. PS1 forms complexes with the p85 subunit of PI3K and promotes cadherin/PI3K association. Furthermore, conditions that inhibit this association prevent the PS1-induced PI3K/Akt activation, indicating that PS1 stimulates PI3K/Akt signaling by promoting cadherin/PI3K association. By activating PI3K/Akt signaling, PS1 promotes phosphorylation/inactivation of glycogen synthase kinase-3 (GSK-3), suppresses GSK-3-dependent phosphorylation of tau at residues overphosphorylated in AD and prevents apoptosis of confluent cells. PS1 FAD mutations inhibit the PS1-dependent PI3K/Akt activation, thus promoting GSK-3 activity and tau overphosphorylation at AD-related residues. Our data raise the possibility that PS1 may prevent development of AD pathology by activating the PI3K/Akt signaling pathway. In contrast, FAD mutations may promote AD pathology by inhibiting this pathway.  相似文献   

16.
Abeta42-lowering nonsteroidal anti-inflammatory drugs (NSAIDs) constitute the founding members of a new class of gamma-secretase modulators that avoid side effects of pan-gamma-secretase inhibitors on NOTCH processing and function, holding promise as potential disease-modifying agents for Alzheimer disease (AD). These modulators are active in cell-free gamma-secretase assays indicating that they directly target the gamma-secretase complex. Additional support for this hypothesis was provided by the observation that certain mutations in presenilin-1 (PS1) associated with early-onset familial AD (FAD) change the cellular drug response to Abeta42-lowering NSAIDs. Of particular interest is the PS1-DeltaExon9 mutation, which provokes a pathogenic increase in the Abeta42/Abeta40 ratio and dramatically reduces the cellular response to the Abeta42-lowering NSAID sulindac sulfide. This FAD PS1 mutant is unusual as a splice-site mutation results in deletion of amino acids Thr(291)-Ser(319) including the endoproteolytic cleavage site of PS1, and an additional amino acid exchange (S290C) at the exon 8/10 splice junction. By genetic dissection of the PS1-DeltaExon9 mutation, we now demonstrate that a synergistic effect of the S290C mutation and the lack of endoproteolytic cleavage is sufficient to elevate the Abeta42/Abeta40 ratio and that the attenuated response to sulindac sulfide results partially from the deficiency in endoproteolysis. Importantly, a wider screen revealed that a diminished response to Abeta42-lowering NSAIDs is common among aggressive FAD PS1 mutations. Surprisingly, these mutations were also partially unresponsive to gamma-secretase inhibitors of different structural classes. This was confirmed in a mouse model with transgenic expression of the PS1-L166P mutation, in which the potent gamma-secretase inhibitor LY-411575 failed to reduce brain levels of soluble Abeta42. In summary, these findings highlight the importance of genetic background in drug discovery efforts aimed at gamma-secretase, suggesting that certain AD mouse models harboring aggressive PS mutations may not be informative in assessing in vivo effects of gamma-secretase modulators and inhibitors.  相似文献   

17.
Interaction of influenza A virus M1 matrix protein with caspases   总被引:8,自引:0,他引:8  
In this investigation, an ability of influenza A virus M1 matrix protein to bind intracellular caspases, the key enzymes of cell apoptosis, has been examined. Protein–protein binding on polystyrene plates and polyvinyl pyrrolidone membrane was employed for this purpose. Under a comparative study of caspases-3, -6, -7, -8 influenza virus M1 protein specifically bound caspase-8 and weakly bound caspase-7. Using a computer analysis of the N-terminal region of M1 protein, a site similar to the anti-caspase site of baculovirus p35 protein, which inhibits caspases and displays antiapoptotic activity, was identified. These results are in good agreement with the supposition that influenza virus M1 protein is involved in a caspase-8-mediated apoptosis pathway in influenza virus infected cells.  相似文献   

18.
Hyperthermia during embryogenesis has been reported to induce deformities in Atlantic salmon (Salmo salar). To examine the involvement of executioner caspases in hyperthermia-induced cell-death in a poikilotherm vertebrate species, five genes encoding caspase-3,-6, and -7 were cloned from Atlantic salmon, and the expression was studied in thermal stressed salmon embryos. The salmon genome contained two genetically distinct variants of both salmon caspase-3 and caspase-6 that is likely the result of two independent chromosome or genome duplications. Whereas only partial caspase-3A encoding sequences were isolated, the full-length caspase-3B cDNA encodes the inactive proenzyme of 279 amino acids (aa) consisting of an N-terminal prodomain and the large and the small subunit. The salmon caspase-6A and caspase-6B proenzymes include an additional linker region between the two subunits. The deduced salmon caspase-7 consists of only 245 aa and lacks the prodomain and part of the large subunit similar to the predicted caspase-7 of the puffer fish Tetraodon sp.. Increased apoptotic activity as evidenced by cleavage of nuclear DNA was demonstrated in salmon embryos incubated at 18-20 degrees C for 84 h after acclimatization at 8 degrees C. Hyperthermia-induced activation of the executioner caspases was indicated by the increased mRNA levels of caspase-3B, caspase-6A/B and caspase-7 after 54 h heat exposure as quantified by real-time RT-PCR. The 2-2.5 fold increase in the mRNA expression of the heat shock protein Hsp70 gene coincided with the peak mRNA values of the executioner caspases. Whole-mount in situ hybridization of the salmon embryo identified caspase-7 mRNA in the lens exclusively, while caspase-3B and caspase-6A/B were expressed in multiple tissues of exposed and control embryos. Interestingly, cardiac expression of caspase-6A/B was only identified in heat stressed embryos. Altogether, these results shed light on evolutionary aspects of the executioner caspases in vertebrates and their expression in salmon embryos exposed to hyperthermia. In particular, the heat sensitive caspase-6 expression in the embryonic heart is of interest since cardiac malformations are an emergent problem in salmon aquaculture.  相似文献   

19.
Role of the executioner caspases during lens development   总被引:2,自引:0,他引:2  
  相似文献   

20.
Apoptosis is characterized by the proteolytic cleavage of hundreds of proteins. One of them, the type 1 inositol-1,4,5-trisphosphate receptor (IP(3) R-1), a multimeric receptor located on the endoplasmic reticulum (ER) membrane that is critical to calcium homeostasis, was reported to be cleaved during staurosporine (STS) induced-apoptosis in Jurkat cells. Because the reported cleavage site separates the IP(3) binding site from the channel moiety, its cleavage would shut down a critical signaling pathway that is common to several cellular processes. Here we show that IP(3) R-1 is not cleaved in 293 cells treated with STS, TNFα, Trail, or ultra-violet (UV) irradiation. Further, it is not cleaved in Hela or Jurkat cells induced to undergo apoptosis with Trail, TNFα, or UV. In accordance with previous reports, we demonstrate that it is cleaved in a Jurkat cell line treated with STS. However its cleavage occurs only after poly(ADP-ribose) polymerase (PARP), which cleavage is a hallmark of apoptosis, and p23, a poor caspase-7 substrate, are completely cleaved, suggesting that IP(3) R-1 is a relatively late substrate of caspases. Nevertheless, the receptor is fully accessible to proteolysis in cellulo by ectopically overexpressed caspase-7 or by the tobacco etch virus (TEV) protease. Finally, using recombinant caspase-3 and microsomal fractions enriched in IP(3) R-1, we show that the receptor is a poor caspase-3 substrate. Consequently, we conclude that IP(3) R-1 is not a key death substrate.  相似文献   

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