ArticlePublisher preview available

The cathepsin B inhibitor z-FA-CMK induces cell death in leukemic T cells via oxidative stress

Authors:
To read the full-text of this research, you can request a copy directly from the authors.

Abstract and Figures

The cathepsin B inhibitor benzyloxycarbonyl-phenylalanine-alanine-chloromethyl ketone (z-FA-CMK) was recently found to induce apoptosis at low concentrations in Jurkat T cells, while at higher concentrations, the cells die of necrosis. In the present study, we showed that z-FA-CMK readily depletes intracellular glutathione (GSH) with a concomitant increase in reactive oxygen species (ROS) generation. The toxicity of z-FA-CMK in Jurkat T cells was completely abrogated by N-acetylcysteine (NAC), suggesting that the toxicity mediated by z-FA-CMK is due to oxidative stress. We found that l-buthionine sulfoximine (BSO) which depletes intracellular GSH through the inhibition of GSH biosynthesis in Jurkat T cells did not promote ROS increase or induce cell death. However, NAC was still able to block z-FA-CMK toxicity in Jurkat T cells in the presence of BSO, indicating that the protective effect of NAC does not involve GSH biosynthesis. This is further corroborated by the protective effect of the non-metabolically active d-cysteine on z-FA-CMK toxicity. Furthermore, in BSO-treated cells, z-FA-CMK-induced ROS increased which remains unchanged, suggesting that the depletion of GSH and increase in ROS generation mediated by z-FA-CMK may be two separate events. Collectively, our results demonstrated that z-FA-CMK toxicity is mediated by oxidative stress through the increase in ROS generation.
This content is subject to copyright. Terms and conditions apply.
ORIGINAL ARTICLE
The cathepsin B inhibitor z-FA-CMK induces cell death
in leukemic T cells via oxidative stress
K. Y. Liow
1
&Sek C. Chow
1
Received: 2 May 2017 /Accepted: 20 October 2017 / Published online: 31 October 2017
#Springer-Verlag GmbH Germany 2017
Abstract The cathepsin B inhibitor benzyloxycarbonyl-
phenylalanine-alanine-chloromethyl ketone (z-FA-CMK)
was recently found to induce apoptosis at low concentrations
in Jurkat Tcells, while at higher concentrations, the cells die of
necrosis. In the present study, we showed that z-FA-CMK
readily depletes intracellular glutathione (GSH) with a con-
comitant increase in reactive oxygen species (ROS) genera-
tion. The toxicity of z-FA-CMK in Jurkat T cells was
completely abrogated by N-acetylcysteine (NAC), suggesting
that the toxicity mediated by z-FA-CMK is due to oxidative
stress. We found that L-buthionine sulfoximine (BSO) which
depletes intracellular GSH through the inhibition of GSH bio-
synthesis in Jurkat T cells did not promote ROS increase or
induce cell death. However, NAC was still able to block z-FA-
CMK toxicity in Jurkat T cells in the presence of BSO, indi-
cating that the protective effect of NAC does not involve GSH
biosynthesis. This is further corroborated by the protective
effect of the non-metabolically active D-cysteine on z-FA-
CMK toxicity. Furthermore, in BSO-treated cells, z-FA-
CMK-induced ROS increased which remains unchanged,
suggesting that the depletion of GSH and increase in ROS
generation mediated by z-FA-CMK may be two separate
events. Collectively, our results demonstrated that z-FA-
CMK toxicity is mediated by oxidative stress through the
increase in ROS generation.
Keywords Z-FA-CMK .Tcells .Glutathione .Reactive
oxygen species .Oxidative stress .N-Acetylcysteine .
L-Cysteine .D-Cysteine .L-Buthionine sulfoximine
Introduction
Peptidyl chloromethyl ketones (CMKs) are among the first
active site-directed irreversible protease inhibitors developed
to block serine and cysteine proteases (Schoellmann and Shaw
1962). They have since become valuable tools for the eluci-
dation of enzyme function, structure, and reaction mecha-
nisms in normal physiology as well as in pathological condi-
tions (Powers et al. 2002;Paceetal.2013). Although widely
used, many studies have now shown that peptidyl CMKs pos-
sess numerous off-target effects besides blocking serine pro-
teases (Weis et al. 1995; Gillibert et al. 2005; Perez-G et al.
2008;Haetal.2009). This presumably is due to the highly
reactive electrophile CMK which lacks specificity and alkyl-
ates non-target molecules or cellular nucleophiles indiscrimi-
nately in cells (Rauber et al. 1986). Despite lacking in speci-
ficity, many peptidyl CMKs were useful biochemical and
pharmacological tools in characterizing the role of serine pro-
teases in animal disease models (Powers et al. 2002).
To overcome the lack of specificity in peptidyl CMKs,
analogous inhibitor structures were developed with different
leaving groups to replace the chlorine atom. Some of these
include bromomethyl and iodomethyl ketones which turn out
to be more reactive and less stable in aqueous solution
(Rasnick 1985;Demuth1990;Powersetal.2002). Efforts to
replace the reactive chlorine atom led to the eventual synthesis
of peptidyl fluoromethyl ketones (FMKs) (Rasnick 1985).
*Sek C. Chow
chow.sek.chuen@monash.edu
1
School of Science, Monash University Malaysia, Jalan Lagoon
Selatan, 46150 Bandar Sunway, Selangor Darul Ehsan, Malaysia
Naunyn-Schmiedeberg's Arch Pharmacol (2018) 391:7182
https://doi.org/10.1007/s00210-017-1436-6
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
... Cathepsin B inhibitors are being actively investigated as potential anticancer agents due to their role in cancer cell invasion and migration. Targeting and inhibiting cathepsin B in cancer cells could have significant therapeutic implications [26,27]. Several small inhibitors of cathepsin B have been developed and are currently in various stages of preclinical and clinical evaluations [28,29]. ...
Article
Full-text available
Numerous cathepsin B inhibitors have been developed and are under investigation as potential cancer treatments. They have been evaluated for their ability to inhibit cathepsin B activity and reduce tumor growth. However, they have shown critical limitations, including low anticancer efficacy and high toxicity, due to their low selectivity and delivery problems. In this study, we developed a novel peptide and drug conjugate (PDC)-based cathepsin B inhibitor using cathepsin-B-specific peptide (RR) and bile acid (BA). Interestingly, this RR and BA conjugate (RR-BA) was able to self-assemble in an aqueous solution, and as a result, it formed stable nanoparticles. The nano-sized RR-BA conjugate showed significant cathepsin B inhibitory effects and anticancer effects against mouse colorectal cancer (CT26) cells. Its therapeutic effect and low toxicity were also confirmed in CT26 tumor-bearing mice after intravenous injection. Therefore, based on these results, the RR-BA conjugate could be developed as an effective anticancer drug candidate for inhibiting cathepsin B in anticancer therapy.
... Cathepsin B (CTSB) is a typical cysteine lysosomal protease correlated with apoptosis, autophagy, oxidative stress and inflammation (Liu et al., 2020). CTSB can aggravate cardiomyocyte apoptosis and oxidative stress in doxorubicin-induced cardiomyocyte inujury (Bai et al., 2018;Liow and Chow, 2018). Whether CTSB participates in regulating LPS-mediated cardiomyocyte damage has not been studied. ...
Article
Myocardial injury is a common complication of sepsis. MicroRNA (miRNA) miR-214-3p is protective against myocardial injury caused by sepsis, but its mechanism in lipopolysaccharide (LPS)- induced cardiomyocyte injury is still unclear. An AC16 cell injury model was induced by LPS treatment. Cell Counting Kit-8 and flow cytometry assay showed decreased cell viability and increased apoptosis in LPS-treated AC16 cells. The levels of caspase- 3, Bax, atrial natriuretic peptide (ANP), brain natriuretic peptide (BNP), myosin 6 (Myh6), myosin 7 (Myh7), reactive oxygen species (ROS), and malondialdehyde (MDA) were increased in LPS-treated AC16 cells, but the levels of Bcl-2 and superoxide dismutase (SOD) were decreased. MiR-214-3p was down-regulated and cathepsin B (CTSB) was upregulated in LPS-treated AC16 cells. At the same time, miR-214-3p could target CTSB and reduce its expression. We also found that a miR-214-3p mimic or CTSB silencing could significantly reduce LPSinduced apoptosis, decrease ROS, MDA, caspase-3, and Bax and increase SOD and Bcl-2. CTSB silencing could significantly reduce ANP, BNP, Myh6, and Myh7 in LPS-treated AC16 cells. The effects of CTSB silencing were reversed by a miR-214-3p inhibitor. In summary, miR-214-3p could inhibit LPSinduced myocardial injury by targeting CTSB, which provides a new idea for myocardial damage caused by sepsis.
... Consistent with these findings, benzyl-oxycarbonyl-phenylalanine-alanine-chloromethyl ketone (z-FA-CMK), a specific CTSB inhibitor has recently been shown to induce oxidative stress mediated apoptosis in leukemic T-cells at low concentrations, while at high concentrations, it induced necrotic cell death. 65,66 The putative roles of cysteine cathepsins in leukemia have been summarized in ►Fig. 2. ...
Article
Full-text available
Cysteine cathepsins are lysosomal proteases that require Cys-His ion pair in their catalytic site for enzymatic activity. While their aberrant expression and oncogenic functions have been widely reported in solid tumors, recent findings suggest that these proteases also play an important role in the pathogenesis of hematological malignancies. In this review, we summarize the potential clinical implications of cysteine cathepsins as diagnostic and prognostic markers in leukemia, and present evidences which supports the utility of these proteases as potential therapeutic targets in hematological malignancies. We also highlight the available information on the expression patterns, regulation, and potential functions of cysteine cathepsins in normal hematopoiesis and hematological malignancies. In hematopoiesis, cysteine cathepsins play a variety of physiological roles including regulation of hematopoietic stem cell adhesion in the bone marrow, trafficking, and maturation. They are also involved in several functions of immune cells which include the selection of lymphocytes in the thymus, antigen processing, and presentation. However, the expression of cysteine cathepsins is dysregulated in hematological malignancies where they have been shown to play diverse functions. Interestingly, several pieces of evidence over the past few years have demonstrated overexpression of cathepsins in leukemia and their association with worst survival outcomes in patients. Strategies aimed at altering the expression, activity, and subcellular localization of these cathepsins are emerging as potential therapeutic modalaties in the management of hematological malignancies. Recent findings also suggest the involvement of these proteases in modulating the immune response in leukemia and lymphomas.
... Moreover, cTSB is also expressed in cardiomyocytes, and increased cTSB expression levels and activity in the myocardium are reported to be induced by doX (7,8), angiotensin ii (9) and isoproterenol (10) and in patients with dilated cardiomyopathy (11). additionally, cTSB is associated with apoptosis (12,13) and oxidative stress (14,15), which serve key roles in the process of doX-induced myocardial injury. our previous study demonstrated that cTSB was upregulated in the heart following pressure overload, and functions as a modulator of the hypertrophic response via regulating the TnF-α/apoptosis signal-regulating kinase 1 (aSK1)/JnK pathway (9). ...
Article
Full-text available
Myocyte apoptosis and oxidative stress key critical roles in the process of doxorubicin (DOX)‑induced cardiotoxicity. However, how apoptosis and oxidative stress arise in DOX‑induced heart injury remains largely unknown. Cathepsin B (CTSB) is a typical lysosomal cysteine protease that is associated with apoptosis, inflammatory responses, oxidative stress and autophagy. The present study aimed to investigate the role of CTSB in DOX‑induced heart injury and its potential mechanism. H9C2 cells were infected with adenovirus or transfected with small interfering RNA to overexpress or knock down CTSB, respectively, and then stimulated with DOX. DOX induced increased CTSB expression levels in H9C2 cells. DOX‑induced cardiomyocyte apoptosis and oxidative stress were attenuated by CTSB knockdown but aggravated by CTSB overexpression in vitro. Mechanistically, the present study showed that CTSB activated the NF‑κB pathway in response to DOX. In summary, CTSB aggravated DOX‑induced H9C2 cell apoptosis and oxidative stress via NF‑κB signalling. CTSB constitutes a potential therapeutic target for the treatment of DOX‑induced cardiotoxicity.
... Another report demonstrated that inhibition of vacuolar H + -ATPases (V-AT-Pases) with concanamycin A decreased Cathepsin B activity in cell lysates of metastatic breast cancer cells [96]. Liow et al. and Chow et al. recently found that Cathepsin B inhibitor benzyloxycarbonyl-phenylalanine-alanine-chloromethyl ketone (z-FA-CMK) induces apoptosis at low concentrations and necrosis at higher concentrations in leukemic T cells via oxidative stress [97]. Most recently, Tang et al. reported a new autophagy inhibitor cepharanthine (CEP) with an anti-cancer effect on non-small lung cancer cells mediated by dacomitinib (DAC) via blockage of autophagosome-lysosome fusion and inhibition of lysosomal Cathepsin B and D maturation [98]. ...
Article
Clinical, biochemical and molecular biology studies have identified lysosome-encapsulated cellular proteases as critical risk factors for cancer progression. Cathepsins represent a group of such proteases aimed at maintenance of cellular homeostasis. Nevertheless, recent reports suggest that Cathepsin B executes other cellular programs such as controlling tumor growth, migration, invasion, angiogenesis, and metastases development. In fact, elevated levels of Cathepsins are found under different pathological conditions including inflammation, infection, neurodegenerative disease, and cancer. Furthermore, the discovery of Cathepsin B secretion and function as an extracellular matrix protein has broadened our appreciation for the impact of Cathepsin B on cancer progression. Underneath a façade of an intracellular protease with limited therapeutic potential hides a central role of cathepsins in extracellular functions. Moreover, this role is incredibly diverse from one condition to the next – from driving caspase-dependent apoptosis to facilitating tumor neovascularization and metastasis. Here we discuss the role of Cathepsin B in the oncogenic process and perspective the use of Cathepsin B for diagnostic and therapeutic applications.
... Another report demonstrated that inhibition of vacuolar H + -ATPases (V-AT-Pases) with concanamycin A decreased Cathepsin B activity in cell lysates of metastatic breast cancer cells [96]. Liow et al. and Chow et al. recently found that Cathepsin B inhibitor benzyloxycarbonyl-phenylalanine-alanine-chloromethyl ketone (z-FA-CMK) induces apoptosis at low concentrations and necrosis at higher concentrations in leukemic T cells via oxidative stress [97]. Most recently, Tang et al. reported a new autophagy inhibitor cepharanthine (CEP) with an anti-cancer effect on non-small lung cancer cells mediated by dacomitinib (DAC) via blockage of autophagosome-lysosome fusion and inhibition of lysosomal Cathepsin B and D maturation [98]. ...
Article
Full-text available
This study aimed to evaluate possible association between genotypes and alleles of two 17q12 polymorphisms (rs3760511 and rs7501939) and prostate cancer (PCa) risk and progression. Two hundred seventy-one patients with PCa, 261 patients with benign prostatic hyperplasia (BPH), and 171 controls were included in the study. Single nucleotide polymorphisms (SNPs) were genotyped by using PCR followed by restriction fragment length (PCR-RFLP) analysis. We conducted meta-analysis of published studies regarding association of these SNPs with PCa risk. Evidence of positive association between the AC genotype of the SNP rs3760511 and BPH risk for the best-fitting overdominant model of association (BPH vs. controls comparison, p = 0.026; odds ratio [OR] = 1.58; 95% confidence interval [95%CI] 1.05-2.36) were obtained. The association between T allele of rs7501939 and PCa risk was determined in PCa versus controls comparison (p = 0.0032; OR = 0.66, 95%CI 0.50-0.87) with the best-fitting model of inheritance being log-additive. This variant was also found to be associated with the risk of BPH (p = 0.0023; OR = 0.65, 95%CI 0.49-0.86). We found no association between parameters of PCa progression and the analyzed SNPs. Meta-analysis showed strong association between these variants and PCa risk. Our study shows association between SNPs at locus 17q12 and the risk of prostatic diseases in Serbian population. At the same time, results of meta-analysis suggest the association of these SNPs with PCa risk.
Article
Full-text available
A number of chloromethyl ketone protease inhibitors reversibly inhibit β-galactosidase induction, growth, RNA synthesis, and protein synthesis in Escherichia coli at concentrations (0.1 to 1.0 mm) which are not toxic to the cells. The competitive protease inhibitor pentamidine isothionate inhibits β-galactosidase induction at a concentration (0.01 mm) which has little effect on general macromolecular synthesis. The action of chloromethyl ketone compounds is specifically blocked by reduced glutathione. Neither oxidized glutathione, Cleland's reagent, cysteine, nor mercaptoethanol were able to replace glutathione. Although there is a structural similarity between chloromethyl ketones and iodoacetamide, our studies indicate that chloromethyl ketones do not act as general “sulfhydryl inhibitors.” While tosyl lysine chloromethyl ketone can react with glutathione under neutral or alkaline conditions it does not inactivate RNase, as does iodoacetamide. Furthermore, glutathione was able to reverse the inhibition caused by tosyl-lysine chloromethyl ketone even when added 15 min after inhibition has been established. Glutathione is not able to reverse the inhibition caused by iodoacetamide. In E. coli, the inhibition of macromolecular synthesis by chloromethyl ketones may be the result of reactions with intracellular glutathione.
Article
Full-text available
Incorporation of amino acids into proteins in HeLa cells, virus-transformed 3T3 mouse fibroblasts, and mouse plasmacytoma cells is inhibited after the addition of L-1-tosylamido-2-phenylethyl chloromethyl ketone, an alkylating agent and chymotrypsin-specific protease inhibitor. Addition of this drug to tissue culture cells at concentrations of 20 to 30 mug per ml results in an irreversible inhibition of the incorporation of amino acids into cellular proteins, and a rapid and complete breakdown of polyribosomes. A comparative study examining the effects of L-1-tosylamido-2-phenylethyl chloromethyl ketone and several known inhibitors of in vivo protein synthesis, with known mechanisms of action, revealed that an optimal concentration of L-1-tosylamido-2-phenylethyl chloromethyl ketone: (a) immediately and selectively inhibits initiation of protein synthesis, (b) does not significantly affect normal elongation rates, and (c) does not promote a premature release of nascent peptides. L-1-Tosylamido-2-phenylethyl chloromenthyl ketone may prove to be a useful tool in investigating the initiatior of protein synthesis in eukaryotic cells.
Article
Full-text available
During the cellular oxidation of fuels, electrons are used to power the proton pumps of the mitochondrial electron transport chain (ETC) and ultimately drive ATP synthesis and the reduction of molecular oxygen to water. During these oxidative processes, some electrons can 'spin off' during fuel oxidation and electron transport to univalently reduce O2, forming reactive oxygen species (ROS). In excess, ROS can be detrimental; however, at low concentrations oxyradicals are essential signaling molecules. Mitochondria thus use a battery of systems to finely control types and levels of ROS, including antioxidants. Several antioxidant systems depend on glutathione. Here, we review mitochondrial ROS homeostatic systems, including emerging knowledge about roles of glutathione in redox balance and the control of protein function by post-translational modification.
Article
Full-text available
In the present study we investigated the Fas-mediated cellular events using the human leukemic T cell line, JURKAT. Ligation of the Fas receptor with a monoclonal antibody (IgM) resulted in the rapid (within 3 h) induction of apoptosis and was characterized by a sequence of distinct morphological and biochemical events. Thus, plasma membrane blebbing, condensation of the chromatin, and formation of high-molecular-weight (HMW) DNA fragments were the earliest events observed (by 45 min). They were followed by cleavage of DNA into oligonucleosomal-length fragments (laddering pattern) and the formation of apoptotic bodies, and finally, rounding of the apoptotic cells and complete cleavage of DNA into oligonucleosomal-length fragments occurred. The mitochondria remained structurally intact up to the stage of oligonucleosomal-length DNA cleavage, and the ability of the cells to exclude trypan blue was not compromised throughout the time course of the experiments. In contrast to many other model systems, apoptosis in JURKAT cells after anti-Fas treatment did not require the presence of extracellular Ca2+ or Mg2+ and was only partially inhibited by Zn2+. In addition, Fas-mediated apoptosis was unaffected by the presence of free radical scavengers or inhibitors of protein phosphatases, protein kinases, and nitric oxide synthesis. However, the serine protease inhibitors, N-tosyl-L-phenylalanine chloromethyl ketone (TPCK) and 3,4-dichloroisocoumarin (DCI) prevented anti-Fas-induced apoptosis in JURKAT cells. Low concentrations of these inhibitors blocked oligonucleosomal-length, but not HMW, DNA fragmentation. The latter required a higher concentration of TPCK or DCI to block. In addition, low concentrations of DCI also prevented Fas-mediated plasma membrane blebbing. In summary, our results suggest that proteolysis plays a central role in Fas-mediated apoptosis and that distinct proteolytic enzymes are involved in HMW DNA fragmentation, and oligonucleosomal-length DNA fragmentation, as well as in plasma membrane blebbing.
Article
Full-text available
Superoxide (O(2) (-)) is an important reactive oxygen species (ROS), and has an essential role in physiology and pathophysiology. An accurate detection of O(2) (-) is needed to better understand numerous vascular pathologies. In this study, we performed a mechanistic study by using the xanthine oxidase (XOD)/hypoxanthine (HX) assay for O(2) (-) generation and a O(2) (-) sensitive fluorescent dye dihydroethidium (DHE) for O(2) (-) measurement. To quantify O(2) (-) and DHE interactions, we measured fluorescence using a microplate reader. We conducted a detailed reaction kinetic analysis for DHE-O(2) (-) interaction to understand the effect of O(2) (-) self-dismutation and to quantify DHE-O(2) (-) reaction rate. Fluorescence of DHE and 2-hydroethidium (EOH), a product of DHE and O(2) (-) interaction, were dependent on reaction conditions. Kinetic analysis resulted in a reaction rate constant of 2.169 ± 0.059 × 10(3) M(-1) s(-1) for DHE-O(2) (-) reaction that is ~100× slower than the reported value of 2.6 ± 0.6 × 10(5) M(-1) s(-1). In addition, the O(2) (-) self-dismutation has significant effect on DHE-O(2) (-) interaction. A slower reaction rate of DHE with O(2) (-) is more reasonable for O(2) (-) measurements. In this manner, the DHE is not competing with superoxide dismutase and NO for O(2) (-). Results suggest that an accurate measurement of O(2) (-) production rate may be difficult due to competitive interference for many factors; however O(2) (-) concentration may be quantified.
Chapter
Full-text available
Reactive oxygen and nitrogen species are thought to contribute to pathogenesis of many cardiovascular diseases including hypertension, atherosclerosis, restenosis, heart failure, and diabetic vascular complications. Some of these reactive oxygen species also play an important role in vascular signaling. In this chapter, we describe various techniques that we have successfully employed to reliably measure superoxide and hydrogen peroxide. Because reactive oxygen species are capable of rapidly inactivating nitric oxide and because endothelial function characterized by nitric oxide bioavailability is an important indicator of vascular health, we have also included novel techniques capable of directly measuring nitric oxide radical from vascular cells and tissues. KeywordsReactive oxygen species–Nitric oxide–Vascular smooth muscle cells–Methods
Article
The caspase inhibitor benzyloxycarbony (Cbz)-l-Val-Ala-Asp (OMe)-fluoromethylketone (z-VAD-FMK) has recently been shown to inhibit T cell proliferation without blocking caspase-8 and caspase-3 activation in primary T cells. We showed in this study that z-VAD-FMK treatment leads to a decrease in intracellular glutathione (GSH) with a concomitant increase in reactive oxygen species (ROS) levels in activated T cells. The inhibition of anti-CD3-mediated T cell proliferation induced by z-VAD-FMK was abolished by the presence of low molecular weight thiols such as GSH, N-acetylcysteine (NAC) and L-cysteine, whereas D-cysteine which cannot be metabolized to GSH has no effect. These results suggest that the depletion of intracellular GSH is the underlying cause of z-VAD-FMK-mediated inhibition of T cell activation and proliferation. The presence of exogenous GSH also attenuated the inhibition of anti-CD3-induced CD25 and CD69 expression mediated by z-VAD-FMK. However, none of the low molecular weight thiols were able to restore the caspase-inhibitory properties of z-VAD-FMK in activated T cells where caspase-8 and caspase-3 remain activated and processed into their respective subunits in the presence of the caspase inhibitor. This suggests that the inhibition of T cell proliferation can be uncoupled from the caspase-inhibitory properties of z-VAD-FMK. Taken together, the immunosuppressive effects in primary T cells mediated by z-VAD-FMK are due to oxidative stress via the depletion of GSH.
Article
The cathepsin B inhibitor, benzyloxycarbonyl-phenylalanine-alanine-chloromethylketone (z-FA-CMK) was found to be toxic and readily induce cell death in the human T cell line, Jurkat, whereas two other analogues benzyloxycarbonyl-phenylalanine-alanine-fluoromethylketone (z-FA-FMK) and benzyloxycarbonyl-phenylalanine-alanine-diazomethylketone (z-FA-DMK) were not toxic. The toxicity of z-FA-CMK requires not only the CMK group, but also the presence of alanine in the P1 position and the benzyloxycarbonyl group at the N-terminal. Dose-response studies showed that lower concentrations of z-FA-CMK induced apoptosis in Jurkat T cells whereas higher concentrations induced necrosis. In z-FA-CMK-induced apoptosis, both initiator caspases (-8 and -9) and effector caspases (-3, -6 and -7) were processed to their respective subunits in Jurkat T cells. However, only the pro-form of the initiator caspases were reduced in z-FA-CMK-induced necrosis and no respective subunits were apparent. The caspase inihibitor benzyloxycarbonyl-valine-alanine-aspartic acid-(O-methyl)-fluoromehylketone (z-VAD-FMK) inhibits apoptosis and caspase processing in Jurkat T cells treated with low concentration of z-FA-CMK but has no effect on z-FA-CMK-induced necrosis and the loss of initiator caspases. This suggests that the loss of initiator caspases in Jurkat T cells during z-FA-CMK-induced necrosis is not a caspase-dependent process. Taken together, we have demonstrated that z-FA-CMK is toxic to Jurkat T cells and induce apoptosis at low concentrations while at higher concentrations the cells die of necrosis.
Article
Abstract— Irradiation of the skin with ultraviolet-B (UVB) radiation causes a local and systemic suppression of T-cell-medi-ated immune responses. Recently, N-acetylcysteine (NAC) was found to protect against UVB-induced immunosuppression and several other types of UV damage. The protective effects appeared to be based on the ability of NAC to increase glutathione (GSH) levels by promoting GSH synthesis. In this study, it was investigated whether topical application of NAC was still effective against UVB-induced suppression of contact hypersensitivity if GSH synthesis was blocked. Mice were pretreated with buthionine sulfoximine (BSO), an inhibitor of GSH synthesis, which succeeded in blocking the increase in epidermal GSH after topical application of NAC. Whereas the non-BSO-treated animals showed an increase to around 155% of the control GSH level for all NAC doses tested, only a slight (nonsignificant) increase in epidermal GSH was observed in the BSO-treated animals. Surprisingly, the protective efficacy of NAC against UVB-induced immunosuppression was not affected by the BSO pretreatment. No significant difference between the protective efficacy of NAC in the two groups was observed. Apparently, the antioxidant effect of NAC itself was sufficient to provide protection against UVB-immunosuppression, independent of GSH synthesis.