Overexpression of optic atrophy 1 protein increases cisplatin resistance via inactivation of caspase-dependent apoptosis in lung adenocarcinoma cells

Share Embed


Descrição do Produto

Human Pathology (2011) xx, xxx–xxx

www.elsevier.com/locate/humpath

Original contribution

Overexpression of optic atrophy 1 protein increases cisplatin resistance via inactivation of caspase-dependent apoptosis in lung adenocarcinoma cells☆,☆☆ Hsin-Yuan Fang MD a,1 , Chih-Yi Chen MD a,b,1 , Shiow-Her Chiou PhD d , Yu-Ting Wang MSc e , Tze-Yi Lin MD, PhD c , Hui-Wen Chang MD c , I-Ping Chiang MD c , Kuo-Jung Lan BSc e , Kuan-Chih Chow PhD e,⁎ a

Department of Surgery, China Medical University Hospital, China Medical University, Taichung, Taiwan 40452 Comprehensive Cancer Center, China Medical University Hospital, China Medical University, Taichung, Taiwan 40452 c Department of Pathology, China Medical University Hospital, China Medical University, Taichung, Taiwan 40452 d Graduate Institute of Microbiology and Public Health, National Chung Hsing University, Taichung, Taiwan 40227 e Graduate Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan 40227 b

Received 1 December 2010; revised 9 April 2011; accepted 13 April 2011

Keywords: Apoptosis-inducing factor; Caspase; Cytochrome c; Drug resistance; Mitochondrial membrane

Summary Optic atrophy 1 protein, a 112-kd guanosine triphosphatase, is involved in the mitochondrial inner membrane fusion and anticancer drug-mediated cytotoxicity, which implicate an association with disease progression of the cancer. In this study, we investigated the prognostic value of optic atrophy 1 expression in patients with lung adenocarcinoma. Using immunohistochemical staining, expression of optic atrophy 1 was determined in 286 lung adenocarcinoma patients. Expression of optic atrophy 1 was confirmed by immunoblotting. The relationship between optic atrophy 1 expression and clinicopathological parameters was analyzed statistically by comparing survival between different groups using the log-rank test. The results showed that optic atrophy 1 overexpression was detected in 219 (76.6%) of lung adenocarcinoma patients. A significant difference was found in cumulative survival between patients with high optic atrophy 1 levels and those with low optic atrophy 1 levels (P = .0016). In the in vitro experiments with cell lines, silencing of optic atrophy 1 expression reduced cisplatin resistance, which was further shown via increased release of cytochrome c and activation of caspase-dependent apoptotic pathway. In conclusion, optic atrophy 1 is highly expressed in lung adenocarcinoma and indicates poor prognosis. © 2011 Elsevier Inc. All rights reserved.

Abbreviations: ATAD3A, the ATPase family, AAA domain containing 3A; DRP1, dynamin-related protein 1; LADC, lung adenocarcinoma; Mfn-2, mitofusin-2; NTLT, nontumor lung tissues; OPA1, optic atrophy 1; RT-PCR, reverse transcription polymerase chain reaction.

☆ This study was supported in part by the Comprehensive Academic Promotion Projects, Department of Education, Taichung, Taiwan (NCHU 995002 to K. C. Chow), and in part by theTaiwan Department of Health, China Medical University Hospital, Cancer Research of Excellence, Taichung, Taiwan (DOH99-TDC-111-005 to C. Y. Chen). RNAi for silencing OPA1 expression was obtained from the National RNAi Core Facility in the Institute of Molecular Biology/ Genomic Research Centre, Academia Sinica, Taipei, Taiwan, supported by the National Research Program for Genomic Medicine Grants of NSC, Taipei, Taiwan (NSC 97-3112-B-001-016). ☆☆ The authors declare no conflict of interest. ⁎ Corresponding author. E-mail address: [email protected] (K. -C. Chow). 1 These are the joint first authors of this article.

0046-8177/$ – see front matter © 2011 Elsevier Inc. All rights reserved. doi:10.1016/j.humpath.2011.04.012

2

1. Introduction The mitochondrion is one of the major organelles responsible for intracellular energy supply and regulation of programmed cell death [1]. Because of the lack of DNA repair and the necessity to maintain integrity of the genome, which encodes only 13 vital proteins for ATP synthesis [2], mitochondria constantly exchange DNA with one another by fusion and fission [3]. Two proteins, dynamin-related protein 1 (DRP1) and human homolog of yeast mitochondria fission protein 1, are essential for mitochondrial fission [4]. Three proteins, mitofusin (Mfn)-1, Mfn-2, and optic atrophy 1 (OPA1), are vital for mitochondrial fusion [5]. Among them, Mfn-1 and Mfn-2 are imperative for fusion of the mitochondrial outer membrane (MOM), whereas OPA1, behaving like its yeast homolog Mgm1, is critical for fusion of the mitochondrial inner membrane [6]. OPA1 is a dynamin-related guanosine triphosphatase. The gene is located on chromosome 3q28-q29, comprising 31 exons [7]. The alternative splicing of exons 4, 4b, and 5b produces 8 transcript variants (http://www.ncbi.nlm.nih.gov/ nucleotide/GeneID: 4976). Depending upon the type of transcript variants, the molecular mass of OPA1 ranges from 107 to 118 kd (924∼1015 amino acid residues). Unlike the other members of dynamin family, OPA1 does not contain the pleckstrin homology region, the guanosine triphosphatase effector domain, or the proline-rich stretch at the carboxyl terminus [7-9]. Genetically, OPA1 mutations, which are frequently detected in patients with autosomal dominant optic atrophy, are correlated with progressive loss of visual acuity, color vision, and central vision field, as well as temporal discoloration of optic disc, ascending atrophy, and dysmyelination of optic nerve [7,8]. Silencing of OPA1 expression by siRNA induces alteration of mitochondrial morphology, mitochondrial fragmentation, disruption of mitochondrial cristae, as well as release of cytochrome c and induction of apoptosis, suggesting that OPA1 is an antiapoptotic factor [10-12]. Recently, Tondera et al [9] showed that OPA1 was indeed required for cell protection against apoptotic stress mediated by actinomycin D or UV irradiation. Moreover, remodeling of mitochondrial nucleoids, the replication complexes in mitochondrial matrix of which the structural integrity was maintained by OPA1, increased cell resistance to the anticancer drug doxorubicin [13,14]. Interestingly, remodeling of the mitochondrial nucleoid has also been suggested to be regulated by nuclear p53 and ataxia telangiectasia mutated protein, indicating that mitochondrial nucleoid and genomic DNA synchronously respond to DNA damage. However, OPA1 has not been studied in lung adenocarcinoma (LADC), of which the incidence and mortality have increased dramatically in the last 2 decades in Taiwan [15]. Although cigarette smoking has been associated with the disease progression and treatment failures [16], a portion of patients who are women and nonsmokers do not respond well to the radiation and

H. -Y. Fang et al. chemotherapy either [17]. The treatment failure, however, correlated with DNA repair– and hypoxia-induced drug and radiation resistance as well as rapid tumor cell growth and metastasis, which were closely associated with mitochondria-related gene expression [16-21]. In this study, we used immunohistochemistry and immunoblotting to determine the expression level of OPA1 in LADC cells and pathological specimens. We also evaluated the statistical correlation between the expression of OPA1 and the clinicopathological parameters as well as the prognostic significance of OPA1 expression in LADC patients. The effect of OPA1 expression on cisplatin sensitivity was characterized in vitro. Our results showed that expression of OPA1 protein could increase cisplatin resistance, and this effect could be through the inhibition of caspase-dependent cell death in LADC cells.

2. Materials and methods 2.1. Tissue specimens and LADC cell lines From January 2001 to December 2004, tissue specimens were collected from 286 patients with newly diagnosed LADC. Samples from all patients, for whom at least one follow-up examination or death was documented, were pathologically confirmed LADC. The stage of the disease was classified according to the new international staging system for lung cancer [22]. The protocols of these studies had been approved by the Medical Ethics Committee at China Medical University Hospital (DMR99-IRB-062). Nine NSCLC cell lines (H23, H125, H226, H838, H1437, H2009, H2087, A549, and H520) were used for the in vitro evaluation of gene expression. H23, H838, H1437, H2009, H2087, and A549 are LADC cells; and H125, H226, and H520 are epithelial type cells. Cells were grown at 37°C in a monolayer in RPMI 1640 supplemented with 10% fetal calf serum, 100 IU/mL penicillin, and 100 μg/mL streptomycin.

2.2. Immunoblotting analysis and immunostaining Immunoblotting and immunohistochemistry were performed as described previously [16]. Antibodies for β-actin were from Chemicon International (Temecula, CA). Antibodies to OPA1 were cultured in the laboratory. For immunocytochemical staining, the cells were grown overnight on slide and then fixed with cold methanol/acetone at 4°C for 10 minutes before staining. The immunological staining was performed following inactivation of endogenous peroxidase [16]. Antibodies for DRP1, ATAD3A, Mfn1, and Mfn-2 were cultured and characterized in our laboratory [20,21,23]. Antibodies to OPA1 were not added for the negative control group.

OPA1 in LADC

2.3. Slide evaluation In each pathological section, nontumor lung tissue served as internal negative control. Slides were evaluated by 2 independent pathologists blinded to the clinicopathological data. The ImmunoReactive Scoring system was adapted for this study [24]. Briefly, a specimen was considered having strong signals when more than 50% of cancer cells were positively stained; intermediate if 25% to 50% of the cells stained positive; weak if less than 25% or

3 more than 10% of the cells were positively stained; and negative if less than 10% of the cancer cells were stained. Cases with strong and intermediate signals were classified as OPA1+, and those with weak or negative signals were classified as OPA1− [16,20,21].

2.4. Statistical analysis Correlation of OPA1 level with clinicopathological factors was analyzed by either the χ2 test or the Fisher

Fig. 1 A, Expression of OPA1 mRNA was detected by RT-PCR in 1 HeLa and 8 LADC cell lines. Expression of β-actin was used as a monitoring standard for the relative expression ratio of OPA1 mRNA. B, Immunoblotting revealed that monoclonal antibodies raised against recombinant OPA1 recognized 2 protein bands of approximately 95 kd. Expression of the 82-kd OPA1 was detected in all 9 human lung cancer cell lines, high in H520; intermediate in H23, H125, H226, H1437, H2009, H2087, and A549 (relative to H838); and low in H838 cells. Expression of the 110-kd protein was detected in 8 human lung cancer cell lines, but not in H2009 cells. Cell lysate from H23 were precipitated by OPA1-specific monoclonal antibodies and protein G sepharose. Both 82- and 110-kd proteins were characterized by matrix-assisted laser desorption/ionization–time-of-flight mass spectrometry. Peptide mass fingerprints of 82- and 110-kd proteins matched that of OPA1: MS-Fit search (http://prospector.ucsf.edu/): EAW78064.1, OPA1 (autosomal dominant). C, Immunocytochemical staining showed that OPA1 was present as distinct granules in the cytoplasm of LADC cells, suggesting that OPA1 is located in the mitochondria. D, Immunoblotting of subcellular fractions, which were separated by sucrose gradient ultracentrifugation. Cytosol, cytosolic fraction; COX IV, cytochrome c oxidase IV of mitochondria. E, Confocal fluorescence immunocytochemical staining of LADC cells. OPA1 was detected by specific monoclonal antibodies labeled with FITC. Mitochondria were labeled with MitoTracker Red CMXRos dye. Nuclei were stained with fluorescent dye 4′,6diamidino-2-phenylindole (DAPI). A merged image of the first, second, and third columns is shown in the fourth column; and the magnification of a specific cell confirms that OPA1 is located in mitochondria. The white bar represents 10 μm.

4 exact test. Survival curves were plotted using the KaplanMeier estimator [25]. Statistical difference in survival between different groups was compared by the log-rank test [26]. Statistical analysis was performed using GraphPad Prism5 statistics software (San Diego, CA). Statistical significance was set at P b .05.

H. -Y. Fang et al.

3. Results 3.1. Expression of OPA1 in LADC cell lines Expression of OPA1 mRNA was examined by reverse transcription polymerase chain reaction(RT-PCR) in 9 lung

Fig. 2 Correlation between OPA1 expression and survival of LADC patients. A, Expression of OPA1 was determined by immunoblotting. Expression of β-actin was used as a monitoring standard for the relative expression of OPA1. Abbreviations: N, NTLT; T, tumor fraction of surgical resections. B, Representative examples of OPA1 expression in pathological specimens as detected by immunohistochemical staining (crimson precipitates). Expression of OPA1 was not detected in (B1) the NTLT, but only in the LADC tumor nests of (B2) acinar, (B3) papillary, (B4) solid, and (B5) mixed subtypes. OPA1 could also be detected in (B6) specimens from the bronchial brushing. C, Comparison of Kaplan-Meier product limit estimates of survival analysis in LADC patients. Patients were divided into 2 groups depending on the expression of OPA1. The difference in survival between the 2 groups was compared by the log-rank test, and the P value was .0016.

OPA1 in LADC cancer cell lines. OPA1 was detected in all cell lines (Fig. 1A). Following sequence analysis, which was performed using fluorescence-labeled dideoxy nucleotides (by Mission Biotech, Taipei, Taiwan), and DNA sequencing ladder read by an ABI PRISM 3700 DNA Analyzer (CD Genomics, Shirley, NY), nucleotide sequence homology of cDNA fragments from the 9 cell lines matched that of OPA1: BC075805, Homo sapiens OPA1, autosomal dominant (http://blast.ncbi.nlm.nih.gov/Blast.cgi). The protein level of OPA1 was determined by immunoblotting. Two protein bands (82 and 110 kd), corresponding to the anticipated molecular mass of OPA1, were recognized in all the cell lines (Fig. 1B). An 82-kd protein was highly expressed in A549, H520, and H2009 cells. To determine the identity of the proteins, cell lysate from H520 was precipitated with antibodies to OPA1; and the respective protein bands were subjected to analysis of matrix-assisted laser desorption/ionization–time-of-flight mass spectrometry. The peptide mass fingerprints of both 82- and 110-kd proteins matched that of OPA1: GenBank| EAW78064.1, OPA1 (autosomal dominant, H sapiens). The matched peptides covered 33.8% (325/960 amino acids) of the protein. These data indicated that both 82- and 110-kd proteins were OPA1 (EAW78064.1). Immunocytochemical staining showed that OPA1 was abundantly present in cytoplasm. The granular appearance of subcellular structures suggested that OPA1 could be present in mitochondria (Fig. 1C). An immunoblotting of sucrose gradient-separated organelle fractions confirmed that OPA1 was localized in the mitochondria (Fig. 1D). Using a MitoTracker Red CMXRos (Molecular Probes, Eugene, OR) uptake and confocal immunofluorescence microscopy (Fig. 1E), we further showed that OPA1 was mainly localized in the mitochondria. However, some of OPA1 signals did not colocalize with mitochondria; and the grainy appearance suggested that some of the OPA1 might be present in other organelles.

3.2. Pathological expression of OPA1 in LADC specimens Using immunoblotting, we detected that the major OPA1 expressed in LADC specimens was the 82-kd OPA1 (Fig. 2A). Immunohistochemistry identified OPA1 in 219 (76.6%) of the pathological specimens. The signals were not identified in the nearby lymphocytes or in the nontumor lung tissue (NTLT) (Fig. 2B1). They were predominantly detected in cancer cells, including acinar (Fig. 2B2), papillary (Fig. 2B3), solid (Fig. 2B4), and mixed subtypes (Fig. 2B5). OPA1 could also be detected in bronchial brushing specimens (Fig. 2B6) and in 61.27% (87/142) of metastatic lymph nodes (data not shown). Statistical analysis showed that overexpression of OPA1 correlated with patient's sex, cell differentiation, histopathological subtypes, tumor stage, and lymphovascular invasion, as well as expression of mitochondria-associated proteins, for example, DRP1, Mfn-1,

5 Table 1 Correlation of OPA1 expression with clinicopathological parameters in LADC patients Parameter Sex Male (n = 224) Female (n = 62) Cigarette smoking Smoker (n = 175) Nonsmoker (n = 111) Stage I (n = 89) II (n = 122) III (n = 75) Histopathological subtype Acinar (n = 37) Micropapillary (n = 6) Papillary (n = 14) Solid (n = 17) Bronchioloalveolar carcinoma (n = 15) Mixed subtype (n = 213) Undetermined (n = 4) Cell differentiation Well (n = 39) Moderate (n = 169) Poor (n = 78) Lymphovascular invasion Positive (n = 216) Negative (n = 70) Mitochondria-associated proteins DRP1nuc+ (n = 226) c DRP1nuc− (n = 60) c ATAD3A+ (n = 242) ATAD3A− (n = 44) Mfn-1+ (n = 203) Mfn-1− (n = 83) Mfn-2+ (n = 177) Mfn-2− (n = 109)

OPA1+ OPA1− P value/ (n = 219) (n = 67) correlation d 178 41

46 21

.041 a/0.13

142 77

33 34

.022 a/0.135

81 101 37

8 21 38

b.001 b/0.44

25 6 11 10 7

12 0 3 7 8

.003 b/−0.157

177 3

36 1

22 131 66

17 38 12

.003 b/−0.187

178 41

38 29

b.001 a/0.242

191 28 212 7 191 28 158 61

35 32 30 37 12 55 19 48

b.001 a/0.364 b.001 a/0.611 b.001 a/0.647 b.001 a/0.382

Two-sided P value determined by χ2 test. Two-sided P value determined by Fisher exact test. c Nuclear expression of DRP1 labeled as DRP1nuc+. Cells that did not express DRP1 or in which the DRP1 was in the cytoplasm were labeled as DRP1nuc−. d The value was determined by the Spearman correlation, which was based on normal approximation and not assuming the null hypothesis. a

b

and ATAD3A (Table 1), suggesting that OPA1 expression was associated with cell growth and metastatic potential [20,21]. These data further indicated that the pathological significance of OPA1 could be intertwined with the other mitochondriaassociated proteins. Interestingly, among the 219 OPA1+ patients, 97 (44.3%) patients had tumor recurrence during follow-up examination. Among the 67 OPA1− patients, 8 (11.9%) patients had recurrence. All 105 patients who had recurrence developed tumors within 18 months after operation. The risk of recurrence for OPA1+ patients was

6

H. -Y. Fang et al. 3.709-fold higher than that for OPA1− patients (P b .001). Survival of OPA1− patients was significantly better than that of OPA1+ patients (Fig. 2C). The hazard ratio between the 2 groups was 1.69, and the difference in cumulative survival was significant (P = .0016) by the log-rank test. Multivariate analysis revealed that the difference in OPA1 expression between the 2 groups was significant as well (P = .004). OPA1 as prognostic marker in survival of stage I patients was also evident (Fig. 2D).

3.3. OPA1 in LADC cells is phosphorylated by protein kinase C, and phosphorylation is essential for maintaining OPA1 stability Previously, we showed that phosphorylation of DRP1 and ATAD3A was critical for protein stability [20,21]. However, DRP1 was phosphorylated by hypoxia-related adenosine monophosphate–activating protein kinase, whereas ATAD3A was phosphorylated by protein kinase C (PKC). OPA1 phosphorylation and the prospective kinase, which were predicted by a NetPhosK program (http://www.cbs.dtu. dk/services/NetPhosK/), showed that, for OPA1 (variant 1), the most probable kinase was PKC at Thr503 , Ser708, Thr816, Thr893, and Thr929. LADC cell lysates were therefore treated with calf intestinal alkaline phosphatase (CIP) before immunoblotting. After CIP treatment, the amount of the 110-kd protein was reduced; but that of the 82-kd protein was increased (Fig. 3A), suggesting that the 110-kd OPA1 could be a phosphorylated form. LADC cells were then treated with a panel of kinase inhibitors. As shown in Fig. 3B, addition of calphostin C, a PKC inhibitor, reduced the intensity of both the 82- and 110-kd protein bands, but not that of β-actin protein, confirming that PKC was the kinase for OPA1 phosphorylation and that phosphorylation was essential for the OPA1 stability. Interestingly, because nicotine activates

Fig. 3 Expression and posttranslational modification of OPA1 in LADC cells. A, Two bands of OPA1 protein were detected in LADC cells and pathological specimens by immunoblotting. However, RT-PCR showed that only one mRNA was detected in these samples, suggesting that the 110-kd protein could be a posttranslationally modified OPA1. H23 cell lysate was treated with CIP before immunoblotting; the 110-kd protein band became less intense, whereas the 82-kd protein band became more intense. B, The effect of kinase inhibitor on OPA1 phosphorylation. The H23 cells were treated with a panel of kinase inhibitors before immunoblotting. Only treatment with 5 μmol/L calphostin C, a PKC inhibitor, for 2 to 4 hours reduced the intensity of the 110and 82-kd protein bands. The results suggested that the PKCmediated OPA1 phosphorylation was essential for maintaining protein stability. C, Addition of 10 μmol/L nicotine at 37°C for 4 hours induced OPA1 phosphorylation. D, Increase of phosphorylated OPA1 level increased cisplatin resistance. ◯, control H23 cells; ●, H23 cells treated with 10 μmol/L of nicotine before cisplatin challenge.

OPA1 in LADC

7 PKC activity, addition of 10 μmol/L nicotine is anticipated to increase levels of 110-kd OPA1 (Fig. 3C) and drug resistance (Fig. 3D).

3.4. Silencing of OPA1 expression increases drug sensitivity, caspase-dependent cell death, and abrasion of MOM Previous studies suggest that OPA1 plays an antiapoptotic role in response to cell stress [13,14]. As anticipated, silencing of OPA1 (OPA1kd) expression by siRNA (Fig. 4A) increased cisplatin sensitivity (Fig. 4B). Moreover, results of confocal fluorescence immunocytochemistry (Fig. 4C) and immunoblotting of sucrose gradient-separated organelle fractions (Fig. 4D) showed that, in OPA1kd cells, the cytoplasmic cytochrome c, but not cytoplasmic nor nuclear apoptosis-inducing factor (AIF), increased following staurosporine (STS) treatment, suggesting that OPA1 deficiency might increase caspase-dependent cell death. To further prove this notion, we examined caspase-3 profiles following STS treatment. As shown in Fig. 4E1, STS treatment increases proteolysis of caspase-3 and degradation of poly (ADP-ribose) polymerase (PARP) (Fig. 4E2) in OPA1kd cells, implicating that OPA1kd-related cell damage may be at

Fig. 4 A, Silencing of OPA1 expression by siRNAs (OPA1kd) for 72 hours reduced the protein level of OPA1 in LADC cells as determined by immunoblotting analysis. B, Silencing of OPA1 expression increased cisplatin sensitivity in H23 cells. ●, H23, wild-type; ◯, H23, OPA1kd. F test, P b .01. Influence of OPA1 expression on caspase-dependent and -independent cell death as shown by (C) confocal fluorescence immunocytochemistry and (D) immunoblotting of sucrose gradient-separated organelle fractions. Silencing of OPA1 (OPA1kd) expression increased cytoplasmic levels of cytochrome c, but not cytoplasmic AIF nor nuclear AIF, following treatment of cells with 0.25 μmol/L of STS for 4 hours. The results suggested that OPA1 deficiency increased caspasedependent cell death. For retaining AIF in the cytoplasm, we simultaneously suppressed expression of human homolog of yeast Rad23A (hHR23A) by siRNA. E, Effect of OPA1 silencing on caspase 3 activation and degradation of PARP. E1, Addition of 0.25 μmol/L STS for 4 hours induced very little effect on caspase 3 activation in wild-type A549 cells, but a greater effect in OPA1kd cells. E2, Addition of 0.1 or 0.25 μmol/L of STS did not induce degradation of PARP in wild-type A549 cells, but a marked breakdown of PARP in OPA1kd cells. An increase of nonspecific PARP degradation was detected in OPA1kd cells. F, Effect of OPA1 silencing on mitochondrial morphology. Compared with (F1) the wild-type cells, (F2) the evident swelling of cristae (black arrowheads) and abrasion (or erosion) of MOM (black arrows) were detected in OPA1kd cells. Moreover, small vesicles among dilated cristae (white arrow heads) within the mitochondria became more evident in OPA1kd cells. In some mitochondria, both MOM and mitochondrial inner membrane (MIM) were eroded; only silhouette of mitochondria (white arrows) and residual cristae remained. OPA1 silencing was carried out by siRNA treatment for 48 to 96 hours before cell harvest.

8

H. -Y. Fang et al.

}

Fig 4. (continued).

the mitochondrial membrane, which regulates the release of cytochrome c. Electron micrographs showed that, compared with the wild-type cells (Fig. 4F1), in addition to the evident swelling of cristae, the outer membrane of some mitochondria was abraded or disappeared in OPA1kd cells (Fig. 4F2). Small vesicles among dilated cristae, of which the pathological significance is not clear, are also identified in these mitochondria, confirming that OPA1 is indeed involved in the shaping of mitochondrial cristae.

4. Discussion Our results show that overexpression of OPA1 is frequently detected in LADC. Increased OPA1 expression in LADC patients significantly correlates with patient's sex,

tumor cell differentiation, tumor stage, histopathological subtypes, and higher incidence of tumor recurrence as well as reduced drug sensitivity, all of which lead to poor prognosis. As shown in the immunoblotting experiment, besides an 82-kDA protein, OPA1 signals of size between 82 and 110 kd were detected in pathological specimens and the LADC cell lines. After CIP treatment, the intensity of the protein bands greater than 82 kd was reduced and that of the 82-kd protein increased, indicating that the protein bands greater than 82-kd OPA1 was phosphorylated. However, the results that the 82-kd protein was also sensitive to CIP suggested that the 82-kd OPA1 was phosphorylated as well. Treatment with calphostin C, a pan-PKC inhibitor, reduced phosphorylation and protein level of 82- and 110-kd OPA1, further indicating that PKC could be the kinase that catalyzes OPA1 phosphorylation [27] and that posttranslational modification is essential for maintaining OPA1 stability.

OPA1 in LADC It is worth noting that elevated OPA1 expression increased drug resistance and that silencing of OPA1 expression reduced cisplatin resistance. These results clearly indicate that the OPA1 plays an important role in the drug sensitivity of LADC, which then reflects in patient's survival. Patients with low OPA1 level are more sensitive to cisplatin-based chemotherapy and have a better prognosis. In contrast, patients with high OPA1 level are more resistant to chemotherapy and, thus, have a poorer prognosis. The exact mechanism by which OPA1 contributes to drug resistance and why the highly expressed OPA1 is mostly detected in the early stage tumors, however, are yet to be determined. By showing that addition of nicotine increased both protein and phosphorylation levels of OPA1, our results supported the previous observations that cigarette smoking could induce overexpression of hepatocyte growth factor and that hepatocyte growth factor increased drug resistance as well as metastatic potential of lung cancer cells [16,28]. Our results also provided an explanation for the prior studies in which smoking was shown to deteriorate mitochondrial activities [29,30] and smoking cessation restored mitochondrial respiratory chain function [31]. Tang et al [32] revealed that OPA1 deficiency (OPA1+/−) induced mitochondrial dysfunction in Drosophila. However, addition of antioxidants delayed the onset of such processes, suggesting that OPA1 deficiency might increase mitochondrial leakage of reactive oxygen species. Using electron microscopy, Olichon et al [10] demonstrated that OPA1 deficiency could lead to perturbation of mitochondrial inner membrane, implying that the OPA1 was essential for maintaining the mitochondrial cristae integrity and for sequestering cytochrome c as well as preventing the aberrant cell death [10,11]. In fact, the mitochondrion has been shown to be one of the major targets for clinically important anticancer drugs, such as cisplatin and etoposide [33,34]. The current evidence suggests that, besides the nuclear objects, these drugs may directly target the mitochondria, which are imperative for sequestering the cytotoxic cytochrome c to avoid the accidental induction of caspase-dependent cell death. The present study sheds further light on the status of mitochondrial OPA1 in the cell cycle as well as in the nicotine-treated cells and how these affect drug sensitivity. The increased drug sensitivity in OPA1kd cells when compared with those that are logarithmically growing is consistent with the observations of others that, like DRP1 and ATAD3A, OPA1 could be an antiapoptotic factor [10,20,21]. Suppression of OPA1 expression increased deformation of mitochondria, bulging of the mitochondrial cristae, and abrasion and erosion of the MOM, indicating that, besides mitochondrial inner membrane fusion, OPA1 might have a role in maintaining MOM integrity, that is, by reducing reactive oxygen species–associated membrane damage [11,12] or by facilitating the phospholipid exchange between the ER and the mitochondria [21]. Without proper exchange of phospholipids, a biogenesis gap of MOM, which appears

9 as abrasion or erosion, may increase the release of cytochrome c (caspase-dependent cell death), but not AIF (caspase-independent cell death), into the cytosol when cells are further challenged with genotoxic stress, supporting the previous studies that suggest that OPA1 sequesters cytochrome c, but not AIF, in the cristae [9-11,13]. The mechanism of differential release of cytochrome c and AIF, however, remains to be resolved. In an ongoing study, we are investigating how OPA1 expression may affect the cristae morphology and MOM integrity. In conclusion, immunoblotting and immunohistochemical staining reveal abundant expression of OPA1 in the LADC cells. Pathological results indicate that OPA1 expression correlates with patient's sex, tumor stage, and expression of other essential mitochondrial proteins, the features that are closely associated with the increased drug resistance and metastatic potential as well as the poor prognosis. In the in vitro experiments using LADC cells, our results show that OPA1 expression is tightly regulated during cell cycle and that OPA1 is phosphorylated by PKC. Silencing of OPA1 expression increases the level of cisplatin sensitivity, possibly via caspase-dependent cell death pathway.

References [1] Aon MA, Cortassa S, Akar FG, O'Rourke B. Mitochondrial criticality: a new concept at the turning point of life or death. Biochim Biophys Acta 2006;1762:232-40. [2] Wallace DC. Mitochondrial diseases in man and mouse. Science 1999;283:1482-8. [3] Twig G, Hyde B, Shirihai OS. Mitochondrial fusion, fission and autophagy as a quality control axis: the bioenergetic view. Biochim Biophys Acta 2008;1777:1092-7. [4] Yoon Y, Krueger EW, Oswald BJ, McNiven MA. The mitochondrial protein hFis1 regulates mitochondrial fission in mammalian cells through an interaction with the dynamin-like protein DLP1. Mol Cell Biol 2003;23:5409-20. [5] Cipolat S, Martins de Brito O, Dal Zilio B, Scorrano L. OPA1 requires mitofusin 1 to promote mitochondrial fusion. Proc Natl Acad Sci USA 2004;101:15927-32. [6] Griparic L, van der Wel NN, Orozco IJ, Peters PJ, van der Bliek AM. Loss of the intermembrane space protein Mgm1/OPA1 induces swelling and localized constrictions along the lengths of mitochondria. J Biol Chem 2004;279:18792-8. [7] Davies V, Votruba M. Focus on molecules: the OPA1 protein. Exp Eye Res 2006;83:1003-4. [8] Chen H, Chan DC. Emerging functions of mammalian mitochondrial fusion and fission. Hum Mol Genet 2005;14:R283-9. [9] Tondera D, Grandemange S, Jourdain A, et al. SLP-2 is required for stress-induced mitochondrial hyperfusion. EMBO J 2009;28: 1589-600. [10] Olichon A, Baricault L, Gas N, et al. Loss of OPA1 perturbates the mitochondrial inner membrane structure and integrity leading to cytochrome c release and apoptosis. J Biol Chem 2003;278:7743-6. [11] Frezza C, Cipolat S, Martins de Brito O, et al. OPA1 controls apoptotic cristae remodeling independently from mitochondrial fusion. Cell 2006;126:177-89. [12] Ishihara N, Fujita Y, Oka T, Mihara K. Regulation of mitochondrial morphology through proteolytic cleavage of OPA1. EMBO J 2006;25: 2966-77.

10 [13] Garrido N, Griparic L, Jokitalo E, Wartiovaara J, van der Bliek AM, Spelbrink JN. Composition and dynamics of human mitochondrial nucleoids. Mol Biol Cell 2003;14:1583-96. [14] Ashley N, Poulton J. Anticancer DNA intercalators cause p53dependent mitochondrial DNA nucleoid re-modelling. Oncogene 2009;28:3880-91. [15] Annual reports of the Department of Health, the Executive Yuan, Republic of China, 2006. [16] Chen JT, Lin TS, Chow KC, et al. Cigarette smoking induces overexpression of HGF in type II pneumocytes and lung cancer cells. Am J Respir Cell Mol Biol 2006;34:264-73. [17] Toh CK, Gao F, Lim WT, et al. Never-smokers with lung cancer: epidemiologic evidence of a distinct disease entity. J Clin Oncol 2006;24:2245-51. [18] Shannon AM, Bouchier-Hayes DJ, Condron CM, Toomey D. Tumor hypoxia, chemotherapeutic resistance and hypoxia-related therapies. Cancer Treat Rev 2003;29:297-307. [19] Lai TC, Chow KC, Lin TY, et al. Expression of 53BP1 as a cisplatinresistant marker in patients with lung adenocarcinomas. Oncol Rep 2010;24:321-8. [20] Chiang YY, Chen SL, Hsiao YT, et al. Nuclear expression of dynaminrelated protein 1 in lung adenocarcinomas. Mod Pathol 2009;22: 1139-50. [21] Fang HY, Chang CL, Hsu SH, et al. ATPase family AAA domaincontaining 3A is a novel anti-apoptotic factor in lung adenocarcinoma cells. J Cell Sci 2010;123:1171-80. [22] Detterbeck FC, Boffa DJ, Tanoue LT. The new lung cancer staging system. Chest 2009;136:260-71. [23] Lin MY. Expression of mitofusins in non-small cell lung cancer. Master thesis, Graduate Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan, 2005. [24] Remmele W, Schicketanz KH. Immunohistochemical determination of estrogen and progesterone receptor content in human breast cancer.

H. -Y. Fang et al.

[25] [26]

[27]

[28]

[29]

[30]

[31]

[32]

[33]

[34]

Computer-assisted image analysis (QIC score) vs. subjective grading (IRS). Pathol Res Pract 1993;189:862-6. Kaplan EL, Meier P. Nonparametric estimation from incomplete observations. J Am Stat Assoc 1958;53:457-81. Mantel N. Evaluation of survival data and two new rank order statistics arising in its consideration. Cancer Chemother Rep 1966;50:163-70. Kobayashi E, Nakano H, Morimoto M, Tamaoki T. Calphostin C (UCN-1028C), a novel microbial compound, is a highly potent and specific inhibitor of protein kinase C. Biochem Biophys Res Commun 1989;159:548-53. Chen JT, Huang CY, Chiang YY, et al. HGF increases cisplatin resistance via down-regulation of AIF in lung cancer cells. Am J Respir Cell Mol Biol 2008;38:559-65. Bouhours-Nouet N, May-Panloup P, Coutant R, et al. Maternal smoking is associated with mitochondrial DNA depletion and respiratory chain complex III deficiency in placenta. Am J Physiol Endocrinol Metab 2005;288:E171-7. Masayesva BG, Mambo E, Taylor RJ, et al. Mitochondrial DNA content increase in response to cigarette smoking. Cancer Epidemiol Biomarkers Prev 2006;15:19-24. Cardellach F, Alonso JR, López S, Casademont J, Miró O. Effect of smoking cessation on mitochondrial respiratory chain function. J Toxicol Clin Toxicol 2003;41:223-8. Tang S, Le PK, Tse S, Wallace DC, Huang T. Heterozygous mutation of Opa1 in Drosophila shortens lifespan mediated through increased reactive oxygen species production. PLoS One 2009;4: e4492. Yang J, Liu X, Bhalla K, et al. Prevention of apoptosis by Bcl-2: release of cytochrome c from mitochondria blocked. Science 1997;275:1129-32. Park JH, Kim TH. Release of cytochrome c from isolated mitochondria by etoposide. J Biochem Mol Biol 2005;38:619-23.

Lihat lebih banyak...

Comentários

Copyright © 2017 DADOSPDF Inc.