Tài liệu Báo cáo khoa học: Mitochondrial chaperone tumour necrosis factor receptor-associated protein 1 protects cardiomyocytes from hypoxic injury by regulating mitochondrial permeability transition pore opening docx

10 507 0
Tài liệu Báo cáo khoa học: Mitochondrial chaperone tumour necrosis factor receptor-associated protein 1 protects cardiomyocytes from hypoxic injury by regulating mitochondrial permeability transition pore opening docx

Đang tải... (xem toàn văn)

Thông tin tài liệu

Mitochondrial chaperone tumour necrosis factor receptor-associated protein 1 protects cardiomyocytes from hypoxic injury by regulating mitochondrial permeability transition pore opening Fei Xiang, Yue-Sheng Huang, Xiao-Hua Shi and Qiong Zhang Institute of Burn Research, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University, Chongqing, China Introduction Hypoxia is one of the main causes of myocardial damage after the receipt of a burn. In the early stages after a severe burn, myocardial damage not only causes cardiac insufficiency, but also induces or aggravates burn shock, which can cause or aggravate ischaemic ⁄ hypoxic injury to other organs [1,2]. Hence, it is important to protect cardiomyocytes from hypoxic damage. Mitochondria are the primary target of hypoxic damage in cardiomyocytes. Several inter- related factors, including calcium overload, an increase in reactive oxygen species (ROS) and a decrease in adenine nucleotides, contribute to mitochondrial impairment during hypoxia and ischaemia [3]. Mito- chondrial dysfunction in cardiomyocytes can also Keywords cardiomyocytes; cell damage; hypoxia; mitochondrial permeability transition pore; tumour necrosis factor receptor-associated protein 1 Correspondence Y S. Huang, Institute of Burn Research, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University, Chongqing 400038, China Fax: +86 23 65461696 Tel: +86 23 65461696 E-mail: yshuang.tmmu@gmail.com (Received 3 December 2009, revised 3 February 2010, accepted 11 February 2010) doi:10.1111/j.1742-4658.2010.07615.x Tumour necrosis factor receptor-associated protein 1 (TRAP1) is a mito- chondrial chaperone that plays a role in maintaining mitochondrial func- tion and regulating cell apoptosis. The opening of the mitochondrial permeability transition pore (MPTP) is a key step in cell death after hypoxia. However, it is still unclear whether TRAP1 protects cardiomyo- cytes from hypoxic damage by regulating the opening of the pore. In the present study, primary cultured cardiomyocytes from neonatal rats were used to investigate changes in TRAP1 expression after hypoxia treatment as well as the mechanism and effect of TRAP1 on hypoxic damage. The results obtained showed that TRAP1 expression increased after 1 h of hypoxia and continued to increase for up to 12 h of treatment. Hypoxia caused an increase in cell death and decreased cell viability and mitochon- drial membrane potential; overexpressing TRAP1 prevented hypoxia- induced damage to cardiomyocytes. The silencing of TRAP1 induced an increase in cell death and decreased both cell viability and mitochondrial membrane potential in cardiomyocytes under normoxic and hypoxic condi- tions. Furthermore, cell damage induced by the silencing of TRAP1 was prevented by the mitochondrial permeability transition pore inhibitor, cyclosporin A. These data demonstrate that hypoxia induces an increase in TRAP1 expression in cardiomyocytes, and that TRAP1 plays a protective role by regulating the opening of the mitochondrial permeability transition pore. Abbreviations Ad-TRAP1, recombinant adenovirus vector for TRAP1 overexpression; CsA, cyclosporin A; CypD, cyclophilin D; GFP, green fluorescent protein; HSP, heat shock protein; MPTP, mitochondrial permeability transition pore; ROS, reactive oxygen species; siRNA, small interfering RNA; TRAP1, tumour necrosis factor receptor-associated protein 1. FEBS Journal 277 (2010) 1929–1938 ª 2010 The Authors Journal compilation ª 2010 FEBS 1929 directly lead to cell death after hypoxia. The mitochon- drial permeability transition pore (MPTP) is a nonspe- cific pore that opens during the time of calcium overload, oxidative stress, adenine nucleotide depletion and elevated phosphate levels. Many studies have dem- onstrated the role of MPTP opening during an ischae- mia ⁄ reperfusion injury to the heart and other organs [4–6]. We have also demonstrated that more MPTPs open in cardiomyocytes after hypoxia compared to normoxic conditions [7]. Once the pore opens, the membrane potential and pH gradient dissipate, pre- venting ATP generation by oxidative phosphorylation. Ultimately, these changes lead to cell death through the activation of phospholipases, nucleases and prote- ases [8]. Indeed, the irreversible mitochondrial injury caused by MPTP opening is the key step in cell death that occurs during hypoxia and other conditions [9]. Tumour necrosis factor receptor-associated protein 1 (TRAP1) localizes to the mitochondria and its targeting sequence, which is found in the N-terminus of the pro- tein, is for mitochondria matrix. An analysis of the cDNA sequences reveals that TRAP1 is identical to heart shock protein (HSP) 75, which is a member of the HSP90 family [10]. HSP90 comprises an important molecular chaperone that is involved in many cellular processes. After hypoxia treatment, HSP90 expression increases, and this plays a protective role against dam- age [11]. However, the changes in TRAP1 in cardiomyo- cytes under hypoxic conditions remain unclear. TRAP1 comprises a mitochondrial chaperone that is critical for importing proteins into the mitochondrial matrix [12]. A previous study showed that up-regulation of TRAP1 expression suppressed arsenite-induced apoptosis in lung epithelium cells [13]. Apoptogenic inducers, such as the protein-tyrosine kinase inhibitor b-hydroxyisovaler- ylshikonin or the topoisomerase II inhibitor VP16, can decrease TRAP1 expression [14]. At the same time, TRAP1 antagonizes ROS production and protects tumour cells from granzyme M-mediated apoptosis [15]. A recent study also demonstrated that TRAP1 over- expression preserves the mitochondrial membrane potential (Dw) and maintains ATP levels and cell viabil- ity during ischaemic-like injury in vivo [16]. These data suggest that TRAP1 may play an important role in maintaining mitochondrial function. As noted above, MPTP is recognized as a key player in cell death. How- ever, whether TRAP1 can protect cells from hypoxic damage by regulating MPTP opening in cardiomyocytes has remained unclear until now. The present study aimed to observe changes in TRAP1 expression after hypoxia treatment and to investigate the effect of TRAP1 on cell death and MPTP opening in primary cardiomyocytes. Results Hypoxia increases TRAP1 expression in cardiomyocytes Western blot analysis was used to investigate TRAP1 expression after hypoxia treatment in cardiomyocytes. TRAP1 content increased after 1 h of hypoxia and continued to increase until for up to 12 h compared to the normoxic group. At the same time, longer hypoxic treatments yielded higher TRPA1 expression (Fig. 1A,B). We then examined TRAP1 immunoreac- tivity with an immunofluorescence assay. After 1 h of hypoxia, TRAP1 fluorescence intensity was brighter in hypoxic cells than in normoxic cells, which meant that TRAP1 expression increased after 1 h of hypoxia (Fig. 1C,D). Furthermore, increases in TRAP1 fluores- cence intensity became greater with an extension of hypoxic treatment time (Figs 1E–G and 2I). The results obtained were similar to those observed with the western blot. TRAP1 overexpression decreases hypoxic damage to cardiomyocytes Because TRAP1 expression of cardiomyocytes was increased after hypoxia treatment, we performed exper- iments to determine whether the increase in TRAP1 expression plays a protective role in hypoxic cardio- myocytes. We constructed a recombinant adenovirus vector for TRAP1 overexpression (Ad-TRAP1) and transfected the cardiomyocytes. After 48 h of infection, infection efficiency was visualized by the expression of green fluorescent protein (GFP), and more than 90% of the cardiomyocytes were infected (Fig. 2A). Protein was then harvested and the results obtained by western blotting revealed that TRAP1 expression increased sig- nificantly in cardiomyocytes infected with Ad-TRAP1 compared to the expression in negative vector-trans- duced cardiomyocytes and to endogenous TRAP1 levels in normoxic cells (Fig. 2B). To evaluate the role of TRAP1 overexpression in cardiomyocytes under hypoxic conditions, we investi- gated cell viability, Dw and cell death. After 6 h of hypoxia, cell viability and Dw were significantly lower in the uninfected and vector-infected cardiomyocytes compared to normoxic cells. By contrast, TRAP1 overexpression increased hypoxic cell viability (Fig. 2C) and preserved Dw (Fig. 2D). Additionally, propidium iodide staining was used to investigate the effect of TRAP1 overexpression on cell death. As shown in Fig. 3, hypoxia treatment resulted in increased cell death, which was reduced by TRAP1 TRAP1 protects cells from hypoxic injury by MPTP F. Xiang et al. 1930 FEBS Journal 277 (2010) 1929–1938 ª 2010 The Authors Journal compilation ª 2010 FEBS overexpression. At the same time, infection with the negative vector had no effect on hypoxia-induced cell death. Silencing of TRAP1 expression induces cardiomyocyte damage After demonstrating that TRAP1 overexpression can prevent hypoxic damage in cardiomyocytes, we next examined whether silencing TRAP1 expression induced damage in cardiomyocytes. After infection with TRAP1-small interfering RNA (siRNA) or control vector adenovirus for 4 days, more than 90% of the cardiomyocytes were determined to be infected by observing GFP expression using a fluorescent micro- scope (Fig. 4A). The effective silencing of endogenous TRAP1 by TRAP1-siRNA adenovirus infection was also confirmed by western blotting (Fig. 4B). After TRAP1-siRNA infection, the viability of the cardiomyocytes was significantly decreased compared to that of normoxic cells and vector-infected cells (Fig. 4C). Furthermore, silencing TRAP1 expression induced a decrease in Dw of cardiomyocytes under normoxic conditions and aggravated Dw loss induced by hypoxia (Fig. 4D). As shown in Fig. 5, TRAP1 depletion also induced a significant increase in cardio- myocytes death, whereas there was very little cell death in the normoxic cardiomyocytes and vector-infected cardiomyocytes. In addition, we also observed the effect of silencing TRAP1 expression on cardiomyocyte damage under hypoxic conditions. It was found that hypoxia induced more injuries in cardiomyocytes in terms of both viability and cell death after TRAP1-siRNA infection (Fig. 6A,B). MPTP mediates the TRAP1 effect TRAP1 is a mitochondria chaperon and plays a role in maintaining mitochondrial homeostasis, whereas MPTP opening is a key step in the process of cell death. Therefore, we aimed to determine whether MPTP opening mediates TRAP1 behaviour. After cardiomyocytes were infected with TRAP1-siRNA or negative vector for 2 days, cyclosporin A (CsA; 2 lm), a selective inhibitor of MPTP opening, was added to the cardiomyocytes. Cells were then infected for an additional 2 days (4 days in total). Treatment with CsA prevented the decrease in cardiomyocyte viability and the increase in cell death induced by TRAP1-siRNA infection under normoxic conditions (Fig. 7). However, there were no differences between vector-infected cells and vector-infected cells after CsA treatment (Fig. 7). Because silencing TRAP1 expression aggravated hypoxic damage of cardiomyocytes, we next investi- gated the effect of CsA on cell viability and cell death A B I CDE FGH 40 30 20 10 fluorescence intensity (arbitrarty units) 0 Normoxia * * * * 13 6 H yp oxia treatment (h) 12 0.4 TRAP1 β-actin 0.3 0.2 0.1 TRAP1/β-actin 0 Normoxia * * * * 136 Hypoxia treatment (h) Hypoxia treatment (h) 12 Normoxia 13 612 75 kD a 43 kD a Fig. 1. Effects of hypoxia on the TRAP1 levels in primary cultured cardiomyocytes. (A) Western blots show TRAP1 immunoreactivity in normoxic or hypoxic cells at the indicated times. b-actin was used as an internal control. (B) TRAP1 levels were normalized with b-actin under normoxic or hypoxic conditions. (C–G) TRAP1 expres- sion detected by immunofluorescence under normoxic conditions (C) and hypoxic conditions for 1 h (D), 3 h (E), 6 h (F) and 12 h (G). TRAP1 primary antibody was omitted as a negative control (H). (I) Differences in fluorescence intensity of TRAP1 in normoxic or hypoxic cells. Data are the mean ± SEM. Scale bar = 25 lm. *P < 0.05 compared to the normoxic group. The experiment was repeated three times. F. Xiang et al. TRAP1 protects cells from hypoxic injury by MPTP FEBS Journal 277 (2010) 1929–1938 ª 2010 The Authors Journal compilation ª 2010 FEBS 1931 after TRAP1-siRNA infection under hypoxic condi- tions. After 6 h of hypoxia, treatment with CsA abol- ished cardiomyocyte damage induced both by hypoxia and silencing TRAP1 under hypoxic conditions (Fig. 6). On the basis of the results described above, we conclude that silencing TRAP1 expression induces MPTP opening in cardiomyocytes, resulting in cell injury. Furthermore, the up-regulation of TRAP1 expression may play a protective role in hypoxic cardiomyocytes by reducing MPTP opening. Discussion In the present study, we found that TRAP1 expression of cardiomyocytes increases after hypoxia and that TRAP1 overexpression protects cardiomyocytes from hypoxic damage. At the same time, silencing TRAP1 expression causes cell damage under normoxic and hypoxic conditions. Our data also indicate that TRAP1 plays a role in cardiomyocytes by regulating MPTP opening. TRAP1 was initially identified by the yeast two- hybrid system as a novel protein that interacted with the intercellular domain of the type 1 tumour necrosis factor receptor [17]. On the basis of the sequence of the homologue, TRAP1 was identified as a member of the HSP90 family. The ATPase activity of TRAP1 is inhibited by geldanamycin, which is a specific inhibitor of HSP90. Despite its ATP-binding activity, TRAP1 does not form a stable complex with the co-chaperones of HSP90, such as Hop and p23 [18]. Studies have shown that TRAP1 does not have a C-terminal EEVD sequence, which exists in HSP90 and is important for HSP90-Hop binding [19]. Thus, it appears that TRAP1 has specific functions that are different from those of other well-characterized HSP90 homologues. TRAP1 is up-regulated by glucose deprivation, oxidative stress and ultraviolet A irradiation, but cannot be induced A B C D Vector Vector Control Ad-TRAP1 Ad-TRAP1 75 kD a 36 kD a TRAP1 GAPDH 0.5 0.4 0.3 0.2 0.1 0 50 60 40 30 20 10 0 Normoxia Hypoxia Vector Ad-TRAP1 Ad-TRAP1 Normoxia Fluorescence intensity (arbitrary units) D 450 Vector H yp oxia Hypoxia Hypoxia # * * * * # Fig. 2. TRAP1 overexpression prevented the hypoxia-induced reductions in cell viabil- ity and Dw in primary cultured cardiomyo- cytes. (A) Cardiomyocytes were infected with negative vector or Ad-TRAP1 for 48 h and then observed under a fluorescence microscope to determine the infection efficiency by visualizing expression of the gene for GFP. Scale bar = 200 lm. (B) Expression of TRAP1 levels in the unin- fected control, negative vector-infected and Ad-TRAP1-infected cardiomyocytes as deter- mined by western blotting. (C, D) Cardio- myocytes were infected with vector or Ad-TRAP1 for 48 h, starved, and then treated for 6 h under hypoxic conditions; cell viability was determined with a cell counting kit (C) and Dw was determined with tetram- ethylrhodamine ethylester; and then one hundred cells from each group were randomly chosen to measure fluorescence intensity (D). Data are the mean ± SEM. *P < 0.05 compared to the normoxic group. #P < 0.05 compared with the hypoxic and hypoxia + vector groups. The experiment was repeated three times. TRAP1 protects cells from hypoxic injury by MPTP F. Xiang et al. 1932 FEBS Journal 277 (2010) 1929–1938 ª 2010 The Authors Journal compilation ª 2010 FEBS by heat [16,20,21]. Furthermore, deferoxamine, an iron chelator, decreases TRAP1 levels in a dose- and time- dependent manner and induces mitochondrial dysfunc- tion in human hepatocytes [22]. However, the changes induced in TRAP1 expression in cardiomyocytes after hypoxia treatment are still unclear. In the present study, we demonstrated that hypoxia treatment (for 1, 3, 6 and 12 h, respectively) induces a time-dependent increase in the levels of TRAP1 protein. Hypoxia is a common pathophysiological process in diseases such as shock, stroke and heart failure. Hypoxic damage of the myocardium is relevant not only to coronary artery diseases, but also to hyper- tensive and cardiomyopathic heart disease [23,24]. Mitochondria are the most susceptible organelles to hypoxic damage in cardiomyocytes. Although hypoxia induced TRAP1 expression increases in cardiomyocytes, the role of that TRAP1 increase remains unclear. The question remains as to whether the hypoxia-induced TRAP1 increase is a protective reaction in cardiomyocytes. Because TRAP1 is a mitochondrial chaperone, it has an important role in regulating cell apoptosis and maintaining mitochon- drial homeostasis and function. Silencing TRAP1 enhances cytochrome c release from the mitochondria and apoptosis induced by b-hydroxyisovalerylshikonin and VP16 [14]. TRAP1 depletion also sensitizes PC12 cells to oxidative stress-induced cytochrome c release and cell death, which means that TRAP1 play a role in the modulation of the mitochondrial apoptotic cas- cade [25]. Moreover, TRAP1 overexpression improves mitochondrial function after ischaemic injury in primary astrocytes in vitro [16]. In the present study, we found that TRAP1 overexpression abolishes the hypoxic damage in cardiomyocytes. Silencing TRAP1 expression not only induces cell damage under normoxic conditions, but it also aggravates hypoxic damage of cardiomyocytes. MPTP is a channel consisting of several proteins that is usually in a low permeability or closed state. Some models have proposed the presence of other molecular components of the pore, although there is still no consensus regarding the exact components. However, cyclophilin D (CypD) is generally accepted as a critical regulatory component of MPTP and plays an important role in regulating MPTP opening [8,26]. CsA, a selective MPTP inhibitor, prevents MPTP opening by inhibiting the activity of the pept- idyl-prolyl cis-trans isomerase of CypD [27,28]. The consequences of MPTP opening are cell necrosis and apoptosis and, even if MPTP opening is insufficient to cause necrosis, apoptosis can occur. After the MPTP opens, apoptogenic substrates (i.e. cytochrome c) are released into the cytoplasm and activate cas- pase-dependent apoptotic pathways. Because MPTP plays a critical role in cell necrosis and apoptosis, it is also involved in protecting cell against hypoxic and ischaemic damages [29,30]. MPTP not only contrib- utes to the early and delayed protective effects of ischaemic preconditioning in rat or rabbit heart, but it is also relevant to ischaemic post-conditioning [31]. We had also previously demonstrated that adenosine A1 receptor activation reduces hypoxic damage by preventing MPTP opening in rat cardiomyocytes [7]. Many studies have demonstrated that Dw loss is accompanied by an increase in MPTP opening [32– 34]. It is considered that Dw reflects the state of MPTP opening indirectly. In the present study, we found that silencing TRAP1 induces Dw loss in cardiomyocytes, and that overexpression of TRAP1 Fig. 3. TRAP1 overexpression decreased hypoxia-induced cell death in primary cultured cardiomyocytes. Cell death was deter- mined by incubating normoxic cells, hypoxic cells, vector-infected hypoxic cells and Ad-TRAP1-infected cells after 6 h of hypoxia with Hoechst 33342 (10 lgÆmL )1 , blue) and propidium iodide (PI) (10 lgÆmL )1 , red). Scale bar = 50 lm. Graphs show the quantifica- tion of cell death (mean ± SEM) and 200–300 cells were counted for each group. *P < 0.05 compared to the normoxic group. #P < 0.05 compared to the hypoxic and hypoxic + vector groups. The experiment was repeated three times. F. Xiang et al. TRAP1 protects cells from hypoxic injury by MPTP FEBS Journal 277 (2010) 1929–1938 ª 2010 The Authors Journal compilation ª 2010 FEBS 1933 suppresses Dw loss caused by hypoxia. Furthermore, our present data also show that CsA prevents the cell damage induced by TRAP1 depletion under normoxic and hypoxic conditions, which means that silencing TRAP1 expression can cause MPTP opening and lead to damage. Because the opening of MPTP increases after hypoxia treatment, and TRAP1 overexpression abolishes hypoxic damage, we therefore assume that TRAP1 overexpression may prevent MPTP opening and having a protective effect under hypoxic condi- tions in cardiomyocytes. In tumour cells, TRAP1 interacts with CypD, and the association of TRAP1 with CypD is prevented by CsA and not geldanamy- cin, suggesting that this association may be necessary for CypD activity [35]. Many factors are involved in inducing MPTP open- ing, especially calcium overload and oxidative stress [36,37]. ROS increases could lead to the MPTP open- ing persistently. However, TRAP1 also shows an important role in regulating ROS generation. ROS production is decreased by TRAP1 overexpression and promoted by silencing TRAP1 expression [15,16,38]. Because TRAP1 plays a role against cell damage by MPTP, further studies are needed to determine whether ROS are mediators between TRAP1 and MPTP in cardiomyocytes. In summary, hypoxia increases the level of TRAP1 in cardiomyocytes, which may protect cells from hypoxic damage by regulating MPTP opening. These results provide us with a deeper understanding of the protective role of TRAP1 in cardiomyocytes and offer new consid- erations for myocardial protection after burn shock. Materials and methods Cardiomyocyte culture and hypoxia treatment Primary cardiomyocyte cultures were prepared from the ventricles of neonatal Sprague-Dawley rats (days 1–3) and trypsinized as described previously [39] in accordance with A B C D Vector VectorControl TRAP1-siRNA TRAP1-siRNA 75 kDa 36 kDa TRAP1 GAPDH 0.5 0.4 0.3 0.2 0.1 0 50 40 30 20 10 0 Normoxia Normoxia Vector TRAP1-siRNA Normoxia Fluorescence intensity (arbitrary units) D 450 Normoxia Vector Vector Hypoxia HypoxiaTRAP1-siRNA TRAP1-siRNA * * * * # Fig. 4. Silencing TRAP1 expression induced cell viability and Dw in primary cultured cardiomyocytes. (A) Cardiomyocytes were infected with negative vector or TRAP1-siRNA for 4 days, and then a fluorescence microscope was used to observe the infection efficiency by visualizing expression of the gene for GFP. Scale bar = 200 lm. (B) Expression of TRAP1 levels in uninfected control, vector-infected and TRAP1-siRNA-infected cardiomyocytes as determined by western blotting. (C) Cardiomyocytes were infected with vector or TRAP1-siRNA for 4 days, starved, and then cell viability was determined under normoxic conditions. (D) Cardiomyocytes were infected with vector or TRAP1-siRNA for 4 days, starved, and then Dw was determined under normoxic conditions or after 6 h of hypoxia. The results are shown as the mean ± SEM. *P < 0.05 compared to the normoxic and normoxic + vector groups. #P < 0.05 compared to the hypoxic and hypoxic + vector groups. The experiment was repeated three times. TRAP1 protects cells from hypoxic injury by MPTP F. Xiang et al. 1934 FEBS Journal 277 (2010) 1929–1938 ª 2010 The Authors Journal compilation ª 2010 FEBS a protocol approved by the Animal Care and Use Commit- tee of the Third Military Medical University. The cultures were grown in a DMEM ⁄ F12 medium (Hyclone, Logan, UT, USA) with 10% (v ⁄ v) fetal bovine serum (Hyclone), 0.1 mm bromodeoxyuridine (Sigma-Aldrich, St Louis, MO, USA), 100 UÆmL )1 penicillin and 100 UÆmL )1 streptomy- cin. Cells were maintained in a 5% CO 2 incubator at 37 °C. Before hypoxia treatment, the cardiomyocytes were deprived of serum for 12 h. Hypoxic conditions were prepared by using an anaerobic jar (Mitsubishi, Tokyo, Japan) and a vacuum glove box (Chunlong, Lianyungang, China). Serum-free medium was placed in the vacuum glove box filled with a mixed gas con- taining 94% nitrogen, 5% CO 2 and 1% O 2 overnight and allowed to equilibrate with the hypoxic atmosphere. Cardiomyocytes were then subjected to hypoxic conditions by replacing the normoxic medium with hypoxic medium and placing the cultures in an anaerobic jar. All procedures were performed in vacuum glove box. Recombinant adenovirus vector for TRAP1 overexpression Ad-TRAP1 and a negative adenovirus vector were pro- duced by Shanghai GeneChem, Co. Ltd (Shanghai, China). The vectors encoded the GFP sequence, which served as a marker gene. A high titre adenovirus stock was made after several rounds of amplification in HEK293A (American Type Culture Collection, Manassas, VA, USA). All recombinant adenoviruses were tested for transgene expression in cardiomyocytes by western blot- ting. Cardiomyocytes were infected with Ad-TRAP1 or a negative vector at a multiplicity of infection of 10 for Fig. 5. Silencing TRAP1 expression induced cell death in primary cultured cardiomyocytes under normoxic conditions. Cell death was determined by incubating uninfected, vector-infected and TRAP1- siRNA-infected cardiomyocytes under normoxic conditions with Hoechst 33342 (10 lgÆmL )1 , blue) and propidium iodide (PI) (10 lgÆmL )1 , red). Scale bar = 50 lm. Graphs show the quantifica- tion of cell death (mean ± SEM) and 200–300 cells were counted for each group. *P < 0.05 compared to the normoxic and normoxic + vector groups. The experiment was repeated three times. A B Fig. 6. CsA prevented hypoxic damage after TRAP1-siRNA infec- tion in primary cardiomyocytes. CsA (2 l M) was added into vector- infected and TRAP1-siRNA-infected cardiomyocytes after 2 days of infection. The cells were then starved, and subjected to hypoxia for 6 h after 4 days of infection. (A) Effects of CsA on cell death in uninfected, vector-infected and TRAP1-siRNA-infected cells under hypoxic conditions. In each group, 200–300 cells were counted. (B) Effects of CsA on cell viability in uninfected, vector-infected and TRAP1-siRNA-infected cells under hypoxic conditions. *P < 0.05 compared to the normoxic group. #P < 0.05 compared to the hyp- oxic and hypoxic + vector groups. **P < 0.05 compared to the hyp- oxic and hypoxic + vector groups. ##P < 0.05 compared to the hypoxic + TRAP1-siRNA group (data are the mean ± SEM). The experiment was repeated three times. F. Xiang et al. TRAP1 protects cells from hypoxic injury by MPTP FEBS Journal 277 (2010) 1929–1938 ª 2010 The Authors Journal compilation ª 2010 FEBS 1935 48 h and then subjected to experiments after being deprived of serum for 12 h. Recombinant adenovirus vector for silencing of TRAP1 expression The recombinant adenovirus vector for silencing of TRAP1 expression (TRAP1-siRNA) was purchased from Shanghai GeneChem, Co. Ltd. The targeting sequence of the siRNA against rat TRAP1 was 5¢-CAACAGAGATTGATCAA AT-3¢. A negative control adenovirus vector containing nonspecific siRNA was constructed in the same way (non- specific vector, 5¢-TTCTCCGAACGTGTCACGT-3¢). All vectors contained the gene for GFP, which served as a mar- ker. Cardiomyocytes were infected with TRAP1-siRNA or control vector by the addition of adenovirus to the cell cul- ture at a multiplicity of infection of 10. After 4 days of infection, the cells were serum starved for 12 h and then treated. Preparation of cell lysates Cells were washed three times with ice-cold NaCl ⁄ P i at the appropriate time after treatment, and lysed in radioimmuno- precipitation assay (Sigma-Aldrich) lysis buffer that contained 2 lgÆmL )1 aprotinin, 2 lgÆmL )1 pepstatin, 2 lgÆmL )1 leupeptin and 100 lgÆmL )1 phenylmethanesulfo- nyl fluoride. Cells were then scraped, and the resulting lysate was ultrasonicated and centrifuged at 12 000 g for 20 min at 4 °C. The supernatant was subjected to western blot analysis. Western blot analysis Protein concentrations were determined by the RC DC assay (Bio-Rad, Hercules, CA, USA). Thirty micrograms of proteins were fractionated by 10% SDS-PAGE and then transferred to a poly(vinylidene difluoride) membrane (Roche, Rotkreuz, Switzerland). The membrane was blocked with 5% (w ⁄ v) skim milk in TBST [20 mm Tris- HCl (pH 8.0), 150 mm NaCl and 0.1% (v ⁄ v) Tween-20] for 2 h at room temperature. Next, the membrane was probed with a 1 : 500 dilution of primary anti-TRAP1 serum (BD Biosciences, San Jose, CA, USA) in blocking buffer at 4 °C overnight. The membrane was washed four times with TBST and incubated with a horseradish peroxidase-conju- gated antibody against mouse IgG for 1 h at room temper- ature. The membrane was then rinsed with TBST, and the protein bands were visualized with ECL Western Blotting Detection Reagents (GE Healthcare, Piscataway, NJ, USA). The images were analysed with quantity one 4.1 software (Bio-Rad). The experiment was repeated three times, and the same results were obtained. Immunofluorescence assay Cardiomyocytes were grown on coverslips. After hypoxia treatment, the cells were fixed with 4% (w ⁄ v) formaldehyde in NaCl ⁄ P i for 10 min and permeabilized with 0.2% (v ⁄ v) Triton X-100 for 15 min at room temperature. Nonspecific binding sites were blocked by incubating the coverslips with 10% (v ⁄ v) goat serum in NaCl ⁄ P i for 1 h. Cells were probed with primary anti-TRAP1 serum at a 1 : 100 dilution over- night at 4 °C, washed with NaCl ⁄ P i , and incubated in the dark at 37 °C for 1 h with fluorescein isothiocyanate-conju- gated IgG. The cells were then washed again with NaCl ⁄ P i and stained with 0.4 mgÆmL )1 4¢,6-diamidino-2-phenylindole (Sigma-Aldrich) for 10 min at room temperature. Micro- scopic images were acquired using a Leica Confocal Micro- scope (Leica Microsystems, Wetzlar, Germany). In the negative control, the primary antibody was omitted. Detection of cardiomyocyte viability Cardiomyocyte viability was determined with a cell counting kit (CCK-8, Dojindo Molecular Technologies, Kumamoto, A B Fig. 7. CsA prevented the cell damage induced by silencing TRAP1 in primary cardiomyocytes under normoxic conditions. CsA (2 l M) was added to vector-infected and TRAP1-siRNA-infected cardio- myocytes after 2 days of infection. The cells were then subjected to cell viability and cell death assay after 4 days of infection. (A) Effects of CsA on cell death in uninfected, vector-infected and TRAP1-siRNA-infected cells. In each group, 200–300 cells were counted. (B) Effects of CsA on cell viability in uninfected, vector- infected and TRAP1-siRNA-infected cells. *P < 0.05 compared to the normoxic and normoxic + vector groups. #P < 0.05 compared to the normoxic + TRAP1-siRNA group (data are the mean ± SEM). The experiment was repeated three times. TRAP1 protects cells from hypoxic injury by MPTP F. Xiang et al. 1936 FEBS Journal 277 (2010) 1929–1938 ª 2010 The Authors Journal compilation ª 2010 FEBS Japan). Cells were cultured in 96-well plates (10 000 cells per well) and the original medium was removed after 6 h of hypoxia. Then, 10 lL of CCK-8 solution and 100 lLof DMEM ⁄ F12 medium were added to each well, and the cells were incubated at 37 °C in the dark for 1 h in accordance with the manufacturer’s instructions. The value of D 450 was determined (n = 3) and the experiment was repeated three times. Cell death assays Cell death was quantified in Hoechst 33342 (10 lgÆmL )1 ; Sigma-Aldrich) and propidium iodide (10 lgÆmL )1 ; Sigma- Aldrich)-labelled cells. Propidium iodide readily penetrates cells with compromised plasma membranes (dead cells) but does not cross intact plasma membranes. Hoechst is a cell- permeable nucleic acid stain that labels both live and dead nuclei. Mitochondrial membrane potential Dw was monitored by tetramethylrhodamine ethylester (Sigma-Aldrich). Cells cultured in a serum-free medium were incubated in the dark with 200 nmolÆL )1 tetramethylrhod- amine ethylester at 37 °C for 15 min. Cells were then washed with NaCl ⁄ P i and observed using a laser scanning confocal microscope. The experiment was repeated three times. Statistical analysis All values were expressed as the mean ± SEM. spss, version 11.0 (SPSS Inc., Chicago, IL, USA) was used to conduct analyses of variance and Tukey’s tests. P < 0.05 was considered statistically significant. Acknowledgements This work was supported by the Key Project of China National Programs for Basic Research and Develop- ment (2005CB522601), the Key Program of National Natural Science Foundation of China (30430680), the Program for Changjiang Scholars, and the Innovative Research Team in University (IRT0712). We thank Sun Wei and Wang Li-ting (Central Library of The Third Military Medical University) for their technical assis- tance with the laser scanning confocal microscope. The authors declare that there are no conflicts of interest. References 1 Huang YS, Yang ZC, Yan BG, Yang JM, Chen FM, Crowther RS & Li A (1999) Pathogenesis of early cardiac myocyte damage after severe burns. J Trauma 46, 428–432. 2 Huang Y, Li Z & Yang Z (2003) Roles of ischemia and hypoxia and the molecular pathogenesis of post-burn cardiac shock. Burns 29, 828–833. 3 Baines CP (2009) The mitochondrial permeability tran- sition pore and ischemia-reperfusion injury. Basic Res Cardiol 104, 181–188. 4 Halestrap AP, Clarke SJ & Javadov SA (2004) Mito- chondrial permeability transition pore opening during myocardial reperfusion – a target for cardioprotection. Cardiovasc Res 61, 372–385. 5 Kim JS, He L, Qian T & Lemasters JJ (2003) Role of the mitochondrial permeability transition in apoptotic and necrotic death after ischemia ⁄ reperfusion injury to hepatocytes. Curr Mol Med 3, 527–535. 6 Matsumoto S, Friberg H, Ferrand-Drake M & Wieloch T (1999) Blockade of the mitochondrial permeability transition pore diminishes infarct size in the rat after transient middle cerebral artery occlusion. J Cereb Blood Flow Metab 19, 736–741. 7 Fei X, Yue-Sheng H, Dong-Xia Z, Zhi-Gang C, Jia-Ping Z & Qiong Z (2009) Adenosine A1 receptor activation reduces mitochondrial permeability transition pores opening in hypoxic cardiomyocytes. Clin Exp Pharmacol Physiol 37, 343–349. 8 Leung AW & Halestrap AP (2008) Recent progress in elucidating the molecular mechanism of the mitochon- drial permeability transition pore. Biochim Biophys Acta 1777, 946–952. 9 Weiss JN, Korge P, Honda HM & Ping P (2003) Role of the mitochondrial permeability transition in myocar- dial disease. Circ Res 93, 292–301. 10 Chen CF, Chen Y, Dai K, Chen PL, Riley DJ & Lee WH (1996) A new member of the hsp90 family of molecular chaperones interacts with the retinoblastoma protein during mitosis and after heat shock. Mol Cell Biol 16, 4691–4699. 11 Wu WC, Kao YH, Hu PS & Chen JH (2007) Geldana- mycin, a HSP90 inhibitor, attenuates the hypoxia- induced vascular endothelial growth factor expression in retinal pigment epithelium cells in vitro. Exp Eye Res 85, 721–731. 12 Kaul SC, Deocaris CC & Wadhwa R (2007) Three faces of mortalin: a housekeeper, guardian and killer. Exp Gerontol 42, 263–274. 13 Lau AT, He QY & Chiu JF (2004) A proteome analysis of the arsenite response in cultured lung cells: evidence for in vitro oxidative stress-induced apoptosis. Biochem J 382, 641–650. 14 Masuda Y, Shima G, Aiuchi T, Horie M, Hori K, Nakajo S, Kajimoto S, Shibayama-Imazu T & Nakaya K (2004) Involvement of tumor necrosis factor recep- tor-associated protein 1 (TRAP1) in apoptosis induced by beta-hydroxyisovalerylshikonin. J Biol Chem 279, 42503–42515. F. Xiang et al. TRAP1 protects cells from hypoxic injury by MPTP FEBS Journal 277 (2010) 1929–1938 ª 2010 The Authors Journal compilation ª 2010 FEBS 1937 15 Hua G, Zhang Q & Fan Z (2007) Heat shock protein 75 (TRAP1) antagonizes reactive oxygen species genera- tion and protects cells from granzyme M-mediated apoptosis. J Biol Chem 282, 20553–20560. 16 Voloboueva LA, Duan M, Ouyang Y, Emery JF, Stoy C & Giffard RG (2008) Overexpression of mitochon- drial Hsp70 ⁄ Hsp75 protects astrocytes against ischemic injury in vitro. J Cereb Blood Flow Metab 28, 1009– 1016. 17 Song HY, Dunbar JD, Zhang YX, Guo D & Donner DB (1995) Identification of a protein with homology to hsp90 that binds the type 1 tumor necrosis factor recep- tor. J Biol Chem 270, 3574–3581. 18 Felts SJ, Owen BA, Nguyen P, Trepel J, Donner DB & Toft DO (2000) The hsp90-related protein TRAP1 is a mitochondrial protein with distinct functional proper- ties. J Biol Chem 275, 3305–3312. 19 Chen B, Piel WH, Gui L, Bruford E & Monteiro A (2005) The HSP90 family of genes in the human genome: insights into their divergence and evolution. Genomics 86, 627–637. 20 Carette J, Lehnert S & Chow TY (2002) Implication of PBP74 ⁄ mortalin ⁄ GRP75 in the radio-adaptive response. Int J Radiat Biol 78, 183–190. 21 Lee AS (2001) The glucose-regulated proteins: stress induction and clinical applications. Trends Biochem Sci 26, 504–510. 22 Im CN, Lee JS, Zheng Y & Seo JS (2007) Iron chelation study in a normal human hepatocyte cell line suggests that tumor necrosis factor receptor-associated protein 1 (TRAP1) regulates production of reactive oxygen species. J Cell Biochem 100, 474–486. 23 Kyriakides ZS, Kremastinos DT, Michelakakis NA, Matsakas EP, Demovelis T & Toutouzas PK (1991) Coronary collateral circulation in coronary artery disease and systemic hypertension. Am J Cardiol 67, 687–690. 24 Horwitz LD, Fennessey PV, Shikes RH & Kong Y (1994) Marked reduction in myocardial infarct size due to prolonged infusion of an antioxidant during reperfu- sion. Circulation 89, 1792–1801. 25 Pridgeon JW, Olzmann JA, Chin LS & Li L (2007) PINK1 protects against oxidative stress by phosphorylat- ing mitochondrial chaperone TRAP1. PLoS Biol 5, e172. 26 Halestrap AP (2009) What is the mitochondrial perme- ability transition pore? J Mol Cell Cardiol 46, 821–831. 27 Halestrap AP & Davidson AM (1990) Inhibition of Ca2(+)-induced large-amplitude swelling of liver and heart mitochondria by cyclosporin is probably caused by the inhibitor binding to mitochondrial-matrix peptidyl-prolyl cis-trans isomerase and preventing it interacting with the adenine nucleotide translocase. Biochem J 268 , 153–160. 28 Connern CP & Halestrap AP (1992) Purification and N-terminal sequencing of peptidyl-prolyl cis-trans-isom- erase from rat liver mitochondrial matrix reveals the existence of a distinct mitochondrial cyclophilin. Bio- chem J 284, 381–385. 29 Zhong Z, Ramshesh VK, Rehman H, Currin RT, Sridharan V, Theruvath TP, Kim I, Wright GL & Lemasters JJ (2008) Activation of the oxygen-sensing signal cascade prevents mitochondrial injury after mouse liver ischemia-reperfusion. Am J Physiol Gastrointest Liver Physiol 295, G823–G832. 30 Shanmuganathan S, Hausenloy DJ, Duchen MR & Yellon DM (2005) Mitochondrial permeability transition pore as a target for cardioprotection in the human heart. Am J Physiol Heart Circ Physiol 289, H237–H242. 31 Hausenloy DJ, Ong SB & Yellon DM (2009) The mito- chondrial permeability transition pore as a target for preconditioning and postconditioning. Basic Res Cardiol 104, 189–202. 32 Sugrue MM, Wang Y, Rideout HJ, Chalmers-Redman RM & Tatton WG (1999) Reduced mitochondrial mem- brane potential and altered responsiveness of a mitochon- drial membrane megachannel in p53-induced senescence. Biochem Biophys Res Commun 261, 123–130. 33 Saotome M, Katoh H, Satoh H, Nagasaka S, Yoshi- hara S, Terada H & Hayashi H (2005) Mitochondrial membrane potential modulates regulation of mitochon- drial Ca 2+ in rat ventricular myocytes. Am J Physiol Heart Circ Physiol 288, H1820–H1828. 34 Lee CS, Park SY, Ko HH, Song JH, Shin YK & Han ES (2005) Inhibition of MPP+-induced mitochondrial damage and cell death by trifluoperazine and W-7 in PC12 cells. Neurochem Int 46, 169–178. 35 Kang BH, Plescia J, Dohi T, Rosa J, Doxsey SJ & Altieri DC (2007) Regulation of tumor cell mitochon- drial homeostasis by an organelle-specific Hsp90 chap- erone network. Cell 131, 257–270. 36 Zorov DB, Juhaszova M, Yaniv Y, Nuss HB, Wang S & Sollott SJ (2009) Regulation and pharmacology of the mitochondrial permeability transition pore. Cardio- vasc Res 83, 213–225. 37 Baumgartner HK, Gerasimenko JV, Thorne C, Ferdek P, Pozzan T, Tepikin AV, Petersen OH, Sutton R, Watson AJ & Gerasimenko OV (2009) Calcium eleva- tion in mitochondria is the main Ca 2+ requirement for mitochondrial permeability transition pore (mPTP) opening. J Biol Chem 284, 20796–20803. 38 Xu L, Voloboueva LA, Ouyang Y, Emery JF & Giffard RG (2009) Overexpression of mitochondrial Hsp70 ⁄ Hsp75 in rat brain protects mitochondria, reduces oxidative stress, and protects from focal ischemia. J Cereb Blood Flow Metab 29, 365–374. 39 Simpson P & Savion S (1982) Differentiation of rat myocytes in single cell cultures with and without prolif- erating nonmyocardial cells. Cross-striations, ultrastruc- ture, and chronotropic response to isoproterenol. Circ Res 50, 101–116. TRAP1 protects cells from hypoxic injury by MPTP F. Xiang et al. 1938 FEBS Journal 277 (2010) 1929–1938 ª 2010 The Authors Journal compilation ª 2010 FEBS . 2 010 , accepted 11 February 2 010 ) doi :10 .11 11/ j .17 42-4658.2 010 .07 615 .x Tumour necrosis factor receptor-associated protein 1 (TRAP1) is a mito- chondrial chaperone. Mitochondrial chaperone tumour necrosis factor receptor-associated protein 1 protects cardiomyocytes from hypoxic injury by regulating mitochondrial permeability

Ngày đăng: 16/02/2014, 14:20

Từ khóa liên quan

Tài liệu cùng người dùng

  • Đang cập nhật ...

Tài liệu liên quan