151
|
Zhang L, Chen D, Chen Z, Moeckel GW. Hypertonicity-induced mitochondrial membrane permeability in renal medullary interstitial cells: protective role of osmolytes. Cell Physiol Biochem 2010; 25:753-60. [PMID: 20511721 DOI: 10.1159/000315095] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/06/2010] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Hyperosmotic stress causes cell death through activation of apoptotic pathways if the protective osmolyte response is impaired. In this study we attempt to elucidate the molecular mechanisms of hypertonicity-induced apoptosis and the effect of major organic osmolytes upon those. METHODS Hypertonicity-induced changes in Bcl2-family protein abundance and the presence of cytochrome c and apoptosis inducing factor (AIF) in the cytoplasm, were measured using western blot and immunofluorescence labeling. To determine dissipation of mitochondrial membrane potential (Delta Psi) though the permeability transition pore (PTP), the lipophilic cationic carbocyanine fluorescence probe JC-1 and TMRM fluorescence probes were used. RESULTS Hypertonic culture conditions increase the abundance of proapoptotic Bax and the concentration of cytochrome c and apoptosis inducing factor (AIF) in the cytoplasm. These changes are associated with a dissipation of Delta Psi and increased permeability of the PTP. We further show that organic osmolytes stabilize the Delta Psi and decrease the concentration of cytochrome c and AIF in the cytoplasm. CONCLUSION Our study shows that organic osmolytes prevent hypertonicity-induced apoptosis by preventing dissipation of Delta Psi through stabilization of the PTP. These findings further support the important role of organic osmolytes in preventing hypertonicity-mediated cell death in medullary kidney cells.
Collapse
Affiliation(s)
- Li Zhang
- Renal Pathology and Electron Microscopy Laboratory, Department of Pathology, Yale University School of Medicine, New Haven, CT 06520-8023, USA
| | | | | | | |
Collapse
|
152
|
Song R, Harris LD, Pettaway CA. Downmodulation of Bcl-2 sensitizes metastatic LNCaP-LN3 cells to undergo apoptosis via the intrinsic pathway. Prostate 2010; 70:571-83. [PMID: 19938012 DOI: 10.1002/pros.21091] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
BACKGROUND We explored the mechanisms of apoptosis after Bcl-2 protein downmodulation in metastatic LNCaP-LN3 cells (LN3). METHODS LNCaP, LNCaP-Pro5 (Pro5) and LN3 cells were cultured in 5% charcoal-stripped serum (CSS) or in R1881 (synthetic androgen) and bicalutamide (synthetic anti-androgen) and growth inhibition was assessed. Expression levels of androgen receptor (AR) and Bcl-2 were determined. LN3 cells were transfected with small interfering RNA Bcl-2 (siRNA Bcl-2) or control siRNA oligonucleotides. Rates of apoptosis and proliferation were obtained. Cytochrome c localization in treated and control cells was assessed +/- cyclosporine A (CsA). Caspases 9, 3, and poly (ADP-ribose) polymerase cleavage (PARP) were measured upon downmodulation of Bcl-2; and cell growth inhibition in vitro after Bcl-2 modulation combined with docetaxel chemotherapy was determined. RESULTS LN3 cells maintained growth under castrate conditions in vitro. AR protein amplification did not explain castrate-resistant LN3 cell growth. Bcl-2 protein levels in LN3 cells were significantly higher than in Pro5 cells, and were effectively downmodulated by siRNA Bcl-2. Subsequently increased apoptosis and decreased proliferation mediated by cytochrome c was noted and this was reversed by CsA. siRNA Bcl-2-transfected LN3 cells exhibited elevated levels of caspases 9, 3, and PARP cleavage. Exposure of LN3 cells to docetaxel led to increased apoptosis, and simultaneous downmodulation of Bcl-2 substantially enhanced this effect. CONCLUSIONS Downmodulation of Bcl-2 in metastatic castrate-resistant LNCaP-LN3 cells led to apoptosis via a cytochrome c-dependent pathway that was enhanced with docetaxel treatment.
Collapse
Affiliation(s)
- Renduo Song
- Department of Urology, University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030, USA
| | | | | |
Collapse
|
153
|
Pradelli LA, Bénéteau M, Ricci JE. Mitochondrial control of caspase-dependent and -independent cell death. Cell Mol Life Sci 2010; 67:1589-97. [PMID: 20151314 PMCID: PMC11115767 DOI: 10.1007/s00018-010-0285-y] [Citation(s) in RCA: 210] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2010] [Accepted: 01/20/2010] [Indexed: 12/12/2022]
Abstract
Mitochondria control whether a cell lives or dies. The role mitochondria play in deciding the fate of a cell was first identified in the mid-1990s, because mitochondria-enriched fractions were found to be necessary for activation of death proteases, the caspases, in a cell-free model of apoptotic cell death. Mitochondrial involvement in apoptosis was subsequently shown to be regulated by Bcl-2, a protein that was known to contribute to cancer in specific circumstances. The important role of mitochondria in promoting caspase activation has therefore been a major focus of apoptosis research; however, it is also clear that mitochondria contribute to cell death by caspase-independent mechanisms. In this review, we will highlight recent findings and discuss the mechanism underlying the mitochondrial control of apoptosis and caspase-independent cell death.
Collapse
Affiliation(s)
- Ludivine A. Pradelli
- Inserm, U895, Centre Méditerranéen de Médecine Moléculaire (C3M), équipe 3 AVENIR, 06204 Nice Cedex 3, France
- Université de Nice-Sophia-Antipolis, Faculté de Médecine, 06107 Nice Cedex 2, France
| | - Marie Bénéteau
- Inserm, U895, Centre Méditerranéen de Médecine Moléculaire (C3M), équipe 3 AVENIR, 06204 Nice Cedex 3, France
- Université de Nice-Sophia-Antipolis, Faculté de Médecine, 06107 Nice Cedex 2, France
| | - Jean-Ehrland Ricci
- Inserm, U895, Centre Méditerranéen de Médecine Moléculaire (C3M), équipe 3 AVENIR, 06204 Nice Cedex 3, France
- Université de Nice-Sophia-Antipolis, Faculté de Médecine, 06107 Nice Cedex 2, France
- Centre Hospitalier Universitaire de Nice, Département d’Anesthésie Réanimation, 06202 Nice Cedex 3, France
| |
Collapse
|
154
|
Ke B, Shen XD, Gao F, Qiao B, Ji H, Busuttil RW, Volk HD, Kupiec-Weglinski JW. Small interfering RNA targeting heme oxygenase-1 (HO-1) reinforces liver apoptosis induced by ischemia-reperfusion injury in mice: HO-1 is necessary for cytoprotection. Hum Gene Ther 2010; 20:1133-42. [PMID: 19534599 DOI: 10.1089/hum.2009.049] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
We have shown that overexpression of heme oxygenase-1 (HO-1) prevents the liver inflammation response leading to ischemia and reperfusion injury (IRI). This study was designed to explore the precise function and mechanism of HO-1 cytoprotection in liver IRI by employing a small interfering RNA (siRNA) that effectively suppresses HO-1 expression both in vitro and in vivo. Using a partial lobar liver warm ischemia model, mice were injected with HO-1 siRNA/nonspecific control siRNA or Ad-HO-1/Ad-beta-gal. Those treated with HO-1 siRNA showed increased serum glutamic-oxaloacetic transaminase levels, significant liver edema, sinusoidal congestion/cytoplasmic vacuolization, and severe hepatocellular necrosis. In contrast, Ad-HO-1-pretreated animals revealed only minimal sinusoidal congestion without edema/vacuolization or necrosis. Administration of HO-1 siRNA significantly increased local neutrophil accumulation and the frequency of apoptotic cells. Mice treated with HO-1 siRNA were characterized by increased caspase-3 activity and reduced HO-1 expression, whereas those given Ad-HO-1 showed decreased caspase-3 activity and increased HO-1/Bcl-2/Bcl-x(L), data confirmed by use of an in vitro cell culture system. Thus, by using an siRNA approach this study confirms that HO-1 provides potent cytoprotection against hepatic IRI and regulates liver apoptosis. Indeed, siRNA provides a powerful tool with which to study gene function in a wide range of liver diseases.
Collapse
Affiliation(s)
- Bibo Ke
- Dumont-UCLA Transplant Center, Department of Surgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | | | | | | | | | | | | | | |
Collapse
|
155
|
Jou MJ, Peng TI, Hsu LF, Jou SB, Reiter RJ, Yang CM, Chiao CC, Lin YF, Chen CC. Visualization of melatonin's multiple mitochondrial levels of protection against mitochondrial Ca(2+)-mediated permeability transition and beyond in rat brain astrocytes. J Pineal Res 2010; 48:20-38. [PMID: 19925580 DOI: 10.1111/j.1600-079x.2009.00721.x] [Citation(s) in RCA: 136] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Melatonin protects cells against various types of oxidative stress-induced apoptosis due primarily to its ability to effectively scavenge pathological and disease condition-augmented generation of mitochondrial reactive oxygen species (mROS). Once produced, mROS indiscriminately damage mitochondrial components and more importantly they crucially activate directly the mitochondrial permeability transition (MPT), one of the critical mechanisms for initiating post mitochondrial apoptotic signaling. Whether or not melatonin targets directly the MPT, however, remains inconclusive, particularly during oxidative stress. This study, thus, investigated this possibility of an 'oxidation free Ca(2+) stress' in the presence of vitamin E after ionomycin exposure as a sole Ca(2+)-mediated MPT in order to exclude melatonin's primary antioxidative effects as well as Ca(2+)-mediated oxidative stress. The studies were carried out using cultured rat brain astrocytes RBA-1. With the application of laser scanning multiple fluorescence imaging microscopy, we visualized for the first time multiple mitochondrial protective effects provided by melatonin during Ca(2+) stress. First, melatonin, due to its primary antioxidative actions, completely prevented mCa(2+)-induced mROS formation during ionomycin exposure. Secondly, when melatonin(')s antioxidative effects were prevented due to the addition of vitamin E, melatonin significantly prevented mCa(2+)-mediated MPT and apoptosis suggesting its direct targeting of the MPT. Surprisingly, in the presence of cyclosporin A, a MPT inhibitor, melatonin reduced further mCa(2+)-mediated apoptosis during ionomycin exposure also suggesting its targeting beyond the MPT. As astrocytes are actively involve in regulating synaptic transmission and neurovascular coupling in the CNS, these multiple mitochondrial layers of protection provided by melatonin against mCa(2+)-and/or mROS-mediated apoptosis in astrocytes may be crucial for future therapeutic prevention and treatment of astrocyte-mediated neurodegenerative diseases in the CNS.
Collapse
Affiliation(s)
- Mei-Jie Jou
- Department of Physiology and Pharmacology, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan.
| | | | | | | | | | | | | | | | | |
Collapse
|
156
|
Activation of mitochondrial ERK protects cancer cells from death through inhibition of the permeability transition. Proc Natl Acad Sci U S A 2009; 107:726-31. [PMID: 20080742 DOI: 10.1073/pnas.0912742107] [Citation(s) in RCA: 187] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
We studied human cancer cell models in which we detected constitutive activation of ERK. A fraction of active ERK was found to be located in mitochondria in RWPE-2 cells, obtained by v-Ki-Ras transformation of the epithelial prostate RWPE-1 cell line; in metastatic prostate cancer DU145 cells; and in osteosarcoma SAOS-2 cells. All these tumor cells displayed marked resistance to death caused by apoptotic stimuli like arachidonic acid and the BH3 mimetic EM20-25, which cause cell death through the mitochondrial permeability transition pore (PTP). PTP desensitization and the ensuing resistance to cell death induced by arachidonic acid or EM20-25 could be ablated by inhibiting ERK with the drug PD98059 or with a selective ERK activation inhibitor peptide. ERK inhibition enhanced glycogen synthase kinase-3 (GSK-3)-dependent phosphorylation of the pore regulator cyclophilin D, whereas GSK-3 inhibition protected from PTP opening. Neither active ERK in mitochondria nor pore desensitization was observed in non-transformed RWPE-1 cells. Thus, in tumor cells mitochondrial ERK activation desensitizes the PTP through a signaling axis that involves GSK-3 and cyclophilin D, a finding that provides a mechanistic basis for increased resistance to apoptosis of neoplastic cells.
Collapse
|
157
|
Abstract
Mitochondria play key roles in activating apoptosis in mammalian cells. Bcl-2 family members regulate the release of proteins from the space between the mitochondrial inner and outer membrane that, once in the cytosol, activate caspase proteases that dismantle cells and signal efficient phagocytosis of cell corpses. Here we review the extensive literature on proteins released from the intermembrane space and consider genetic evidence for and against their roles in apoptosis activation. We also compare and contrast apoptosis pathways in Caenorhabditis elegans, Drosophila melanogaster, and mammals that indicate major mysteries remaining to be solved.
Collapse
Affiliation(s)
- Chunxin Wang
- Biochemistry Section, Surgical Neurology Branch, NINDS, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | |
Collapse
|
158
|
Scovassi AI, Soldani C, Veneroni P, Bottone MG, Pellicciari C. Changes of mitochondria and relocation of the apoptosis-inducing factor during apoptosis. Ann N Y Acad Sci 2009; 1171:12-7. [PMID: 19723032 DOI: 10.1111/j.1749-6632.2009.04707.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
During apoptosis, apoptosis-inducing factor (AIF) is released from the mitochondrial intermembrane space to the cytosol and to the nucleus. We analyzed AIF in HeLa cells driven to apoptosis by either etoposide or actinomycin D, and we observed changes in the structure and function of mitochondria as well as the translocation of cytochrome c and AIF from mitochondria to the nucleus in early apoptosis. In cells with fragmented chromatin (i.e., in late apoptosis), the immunolabeling for AIF appeared to be distinct from chromatin, being mainly confined to mitochondria.
Collapse
Affiliation(s)
- A Ivana Scovassi
- Istituto di Genetica Molecolare, Consiglio Nazionale delle Ricerche, Pavia, Italy.
| | | | | | | | | |
Collapse
|
159
|
Wang Q, Liu M, Li X, Chen L, Tang H. Kazrin F is involved in apoptosis and interacts with BAX and ARC. Acta Biochim Biophys Sin (Shanghai) 2009; 41:763-72. [PMID: 19727525 DOI: 10.1093/abbs/gmp065] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Kazrin has recently been identified as a functional protein that is involved in cell-cell junctions and in signal transduction. Here, we identified a new isoform, Kazrin F, which is 518 aa in length and has 97 aa unique at the N-terminus. Knockdown of Kazrin F using siRNA caused cell apoptosis and a marked decrease in cell viability measured by MTT and TUNEL assays. Co-immunoprecipitation analysis revealed that Kazrin F interacts with ARC (apoptosis repressor with caspase recruitment domain) and Bax (Bcl-2-associated X protein). Co-localization of Kazrin F with ARC and Bax in the cytoplasm was determined by immunofluorescence analysis. These results suggested that Kazrin F might play an important role in regulating cellular apoptosis by interacting with ARC and Bax.
Collapse
Affiliation(s)
- Qiong Wang
- Tianjin Life Science Research Center, Tianjin Medical University, Tianjin 300070, China
| | | | | | | | | |
Collapse
|
160
|
Chinopoulos C, Adam-Vizi V. Mitochondria as ATP consumers in cellular pathology. Biochim Biophys Acta Mol Basis Dis 2009; 1802:221-7. [PMID: 19715757 DOI: 10.1016/j.bbadis.2009.08.008] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2009] [Revised: 07/17/2009] [Accepted: 08/17/2009] [Indexed: 12/21/2022]
Abstract
ATP provided by oxidative phosphorylation supports highly complex and energetically expensive cellular processes. Yet, in several pathological settings, mitochondria could revert to ATP consumption, aggravating an existing cellular pathology. Here we review (i) the pathological conditions leading to ATP hydrolysis by the reverse operation of the mitochondrial F(o)F(1)-ATPase, (ii) molecular and thermodynamic factors influencing the directionality of the F(o)F(1)-ATPase, (iii) the role of the adenine nucleotide translocase as the intermediary adenine nucleotide flux pathway between the cytosol and the mitochondrial matrix when mitochondria become ATP consumers, (iv) the role of the permeability transition pore in bypassing the ANT, thereby allowing the flux of ATP directly to the hydrolyzing F(o)F(1)-ATPase, (v) the impact of the permeability transition pore on glycolytic ATP production, and (vi) endogenous and exogenous interventions for limiting ATP hydrolysis by the mitochondrial F(o)F(1)-ATPase.
Collapse
Affiliation(s)
- Christos Chinopoulos
- Department of Medical Biochemistry, Semmelweis University, Neurobiochemical Group, Hungarian Academy of Sciences, Budapest, Hungary
| | | |
Collapse
|
161
|
Joza N, Pospisilik JA, Hangen E, Hanada T, Modjtahedi N, Penninger JM, Kroemer G. AIF: Not Just an Apoptosis-Inducing Factor. Ann N Y Acad Sci 2009; 1171:2-11. [DOI: 10.1111/j.1749-6632.2009.04681.x] [Citation(s) in RCA: 133] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
162
|
|
163
|
Varecha M, Zimmermann M, Amrichová J, Ulman V, Matula P, Kozubek M. Prediction of localization and interactions of apoptotic proteins. J Biomed Sci 2009; 16:59. [PMID: 19580669 PMCID: PMC2714591 DOI: 10.1186/1423-0127-16-59] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2009] [Accepted: 07/06/2009] [Indexed: 01/14/2023] Open
Abstract
During apoptosis several mitochondrial proteins are released. Some of them participate in caspase-independent nuclear DNA degradation, especially apoptosis-inducing factor (AIF) and endonuclease G (endoG). Another interesting protein, which was expected to act similarly as AIF due to the high sequence homology with AIF is AIF-homologous mitochondrion-associated inducer of death (AMID). We studied the structure, cellular localization, and interactions of several proteins in silico and also in cells using fluorescent microscopy. We found the AMID protein to be cytoplasmic, most probably incorporated into the cytoplasmic side of the lipid membranes. Bioinformatic predictions were conducted to analyze the interactions of the studied proteins with each other and with other possible partners. We conducted molecular modeling of proteins with unknown 3D structures. These models were then refined by MolProbity server and employed in molecular docking simulations of interactions. Our results show data acquired using a combination of modern in silico methods and image analysis to understand the localization, interactions and functions of proteins AMID, AIF, endonuclease G, and other apoptosis-related proteins.
Collapse
Affiliation(s)
- Miroslav Varecha
- Centre for Biomedical Image Analysis, Faculty of Informatics, Masaryk University, Botanická 68a, Brno 60200, Czech Republic.
| | | | | | | | | | | |
Collapse
|
164
|
Zhang F, Zhang T, Jiang T, Zhang R, Teng ZH, Li C, Gu ZP, Mei Q. Wortmannin potentiates roscovitine-induced growth inhibition in human solid tumor cells by repressing PI3K/Akt pathway. Cancer Lett 2009; 286:232-9. [PMID: 19541408 DOI: 10.1016/j.canlet.2009.05.039] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2008] [Revised: 03/03/2009] [Accepted: 05/27/2009] [Indexed: 11/18/2022]
Abstract
Roscovitine has been reported to have anti-tumor effects in some cancer cell lines. The phosphatidylinositol-3-kinase (PI3K) signaling, which activates protein kinase B (PKB)/Akt, is known to mediate cell survival. The current study examined the role of wortmannin, a PI3K inhibitor, as a chemosensitizer for roscovitine and its proposed mechanism of action. The results showed that wortmannin significantly chemosensitized three human tumor cell lines (A549, HCT116 and HeLa cells). In A549 cells, wortmannin increased roscovitine-induced apoptosis in a dose-dependent manner, which was correlated with the inhibition of phosphorylated PKB/Akt level. Wortmannin enhanced the effects of roscovitine by causing pronounced reduction of mitochondrial transmembrane potential (MMP) and increases of cytochrome c release and active caspase-3, as well as enhanced activation of Bax and Bad, including Bax oligomerization and mitochondrial translocation of Bax and Bad. Taken together, these results provide evidence for the potential application of roscovitine/wormannin combination in clinical treatment for solid tumors.
Collapse
Affiliation(s)
- Feng Zhang
- Department of Pharmacology, The Fourth Military Medical University, Xi'an 710032, China
| | | | | | | | | | | | | | | |
Collapse
|
165
|
Synaptic activity-mediated suppression of p53 and induction of nuclear calcium-regulated neuroprotective genes promote survival through inhibition of mitochondrial permeability transition. J Neurosci 2009; 29:4420-9. [PMID: 19357269 DOI: 10.1523/jneurosci.0802-09.2009] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Cellular stress caused by genetic or environmental factors are considered to be the major inducers of cell death under pathological conditions. Induction of the apoptotic function of the tumor suppressor p53 is a common cellular response to severe genotoxic and oxidative stresses. In the nervous system, accumulation of p53 and increased p53 activity are associated with neuronal loss in acute and chronic neurodegenerative disorders. Here, we show that regulation of the p53 gene (trp53) is an integral part of a synaptic activity-controlled, calcium-dependent neuroprotective transcriptional program. Action potential (AP) bursting suppresses trp53 expression and downregulates key proapoptotic p53 target genes, apaf1 and bbc3 (puma). At the same time, AP bursting activates the nuclear calcium-induced neuroprotective gene, btg2. Depletion of endogenous p53 levels using RNA interference or expression of Btg2 renders neurons more resistant against excitotoxicity-induced mitochondrial permeability transitions and promotes neuronal survival under severe cellular stresses. We propose that suppression of p53 functions together with nuclear calcium-regulated neuroprotective genes in a coordinate and synergistic manner to promote neuronal survival through the stabilization of mitochondria against cellular stresses.
Collapse
|
166
|
Samadder P, Bittman R, Byun HS, Arthur G. A glycosylated antitumor ether lipid kills cells via paraptosis-like cell death. Biochem Cell Biol 2009; 87:401-14. [DOI: 10.1139/o08-147] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Glycosylated antitumor ether lipids (GAELs) have superior anticancer properties relative to the alkyllysophospholipid class, but there have been no studies of the mechanisms of these compounds. The prototype GAEL, 1-O-hexadecyl-2-O-methyl-3-O-(2′-amino-2′-deoxy-β-d-glucopyranosyl)-sn-glycerol (Gln), effectively killed mouse embryonic fibroblasts (MEFs) lacking key molecules involved in caspase-dependent apoptosis, and cell death was not prevented by caspase inhibitors. Gln did not cause a loss of mitochondrial membrane potential, even in rounded-up dying cells. Gln stimulated the appearance and accumulation of LC3-II, a protein marker for autophagy, in a variety of cells, including wild-type MEFs, but not in MEFs lacking ATG5, a key protein required for autophagy. Gln induced LC3 puncta formation in Chinese hamster ovary cells stably expressing a LC3–green fluorescent protein fusion protein. Thus, Gln appears to induce autophagy. Autophagy was mTOR-independent and was not inhibited by 3-methyladenine or wortmannin. Although Gln is toxic, cellular ability to undergo autophagy was not essential for its toxicity. Furthermore, the GAEL analog 2-deoxy-C-Glc induced LC3 puncta formation but did not kill the cells. Gln, but not 2-deoxy-C-Glc, caused the accumulation of cytoplasmic acidic vacuoles in the cells. Our data suggest that GAELs may activate autophagy; however, GAELs do not kill cells by apoptosis or autophagy but rather by a paraptosis-like cell death mechanism.
Collapse
Affiliation(s)
- Pranati Samadder
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB R3E 0W3, Canada
- Department of Chemistry and Biochemistry, Queens College of the City University of New York, Flushing, NY 11367, USA
| | - Robert Bittman
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB R3E 0W3, Canada
- Department of Chemistry and Biochemistry, Queens College of the City University of New York, Flushing, NY 11367, USA
| | - Hoe-Sup Byun
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB R3E 0W3, Canada
- Department of Chemistry and Biochemistry, Queens College of the City University of New York, Flushing, NY 11367, USA
| | - Gilbert Arthur
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB R3E 0W3, Canada
- Department of Chemistry and Biochemistry, Queens College of the City University of New York, Flushing, NY 11367, USA
| |
Collapse
|
167
|
Tavian D, Colombo R, Misiti F, Ena P, Ena L, Sampaolese B, Giardina B, Clementi ME. Fibroblast apoptosis in a patient affected by lamellar ichthyosis. J Cutan Pathol 2009; 36:417-24. [PMID: 19278426 DOI: 10.1111/j.1600-0560.2008.01078.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND Lamellar ichthyosis (LI) is a congenital recessive skin disorder characterized by generalized scaling and hyperkeratosis. The pathology may be caused by mutations in transglutaminase 1 (TGM1) gene that encodes an enzyme critical for terminally differentiating keratinocytes. Because of evidences that transglutaminase enzymes are involved in programmed cell death, we investigated morphological and biochemical apoptotic parameters in cultured skin fibroblasts from a patient with a severe LI and homozygous for the TGM1 R142H mutation. METHOD The principle apoptotic signals (mitochondrial membrane potential, analysis of oxygen consumption, DNA fragmentation and Bax/Bcl-2 gene expression) were analyzed in cultured fibroblasts from a LI patient, his mother (TGM1 mutation carrier) and a control subject. RESULTS LI fibroblasts showing a reduction of fibronectin expression evidenced a strong inhibition of oxygen consumption, a dramatic drop in the mitochondrial membrane potential (Delta psi(m)), and a higher apoptotic index. CONCLUSION The present results suggest a possible connection between the alterations in the keratinization process leading to LI and the observed increased fibroblast apoptosis.
Collapse
Affiliation(s)
- Daniela Tavian
- Laboratory of Human Molecular Biology and Genetics, Catholic University of the Sacred Heart, Milan, Italy
| | | | | | | | | | | | | | | |
Collapse
|
168
|
Overexpression of CHMP6 induces cellular oncosis and apoptosis in HeLa cells. Biosci Biotechnol Biochem 2009; 73:494-501. [PMID: 19270365 DOI: 10.1271/bbb.80458] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Cell death can proceed via at least two distinct pathways, apoptosis and oncosis. Apoptosis is an energy-dependent process characterized morphologically by cell shrinkage, whereas oncosis is defined as a prelethal pathway leading to cell death associated with cellular swelling, organelle swelling, and increased membrane permeability. In this study, we found that overexpression of chromatin modifying protein 6 (CHMP6) induced cell death by a series of experiments, including morphological observation, intracellular ATP determination, caspase-3 activity, and flow cytometry. Typical morphological characteristics consistent with oncosis were observed by transmission electron microscopy. Simultaneously, we obtained some results that indicated apoptosis, but the anti-apoptotic gene Bcl-xL and caspase family inhibitor Z-VAD-FMK had little effect on CHMP6-induced cell death. These results suggest that CHMP6 overexpression can cause cell death, predominantly via oncosis and to a certain extent via apoptosis, and that CHMP6 might be a novel regulator involved in both oncosis and apoptosis.
Collapse
|
169
|
Zhang FJ, Yang JY, Mou YH, Sun BS, Ping YF, Wang JM, Bian XW, Wu CF. Inhibition of U-87 human glioblastoma cell proliferation and formyl peptide receptor function by oligomer procyanidins (F2) isolated from grape seeds. Chem Biol Interact 2009; 179:419-29. [PMID: 19167369 DOI: 10.1016/j.cbi.2008.12.017] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2008] [Revised: 12/14/2008] [Accepted: 12/29/2008] [Indexed: 10/21/2022]
Abstract
Gliomas are the most common and lethal tumor type in the brain. The present study investigated the effect of oligomer procyanidins (F2) (F2, degree of polymerization 2-15), a natural fraction isolated from grape seeds on the biological behavior of glioblastoma cells. We found that F2 significantly inhibited the glioblastoma growth, with little cytotoxicity on normal cells, induced G2/M arrest and decreased mitochondrial membrane potential in U-87 cells. It also induced a non-apoptotic cell death phenotype resembling paraptosis in U-87 cells. In addition, it was found for the first time that F2 in non-cytotoxic concentrations selectively inhibited U-87 cell chemotaxis mediated by a G-protein coupled receptor formyl peptide receptor FPR, which is implicated in tumor cell invasion and metastasis. Further experiments indicated that F2 inhibited fMLF-induced U-87 cell calcium mobilization and MAP kinases ERK1/2 phosphorylation. Moreover, F2 attenuated the glioblastoma FPR expression, a new molecular target for glioma therapeutics, which has been shown to play important roles in glioma cells chemotaxis, proliferation and angiogenesis in addition to its promotion to tumor progression, but did not affect FPR mRNA expression in U-87 cells. Taken together, our results suggest that F2 may be a promising candidate for the development of novel anti-tumor therapeutics.
Collapse
Affiliation(s)
- Feng-Jiao Zhang
- Department of Pharmacology, Shenyang Pharmaceutical University, 103# Wenhua Road, Shenhe District, 110016 Shenyang, PR China
| | | | | | | | | | | | | | | |
Collapse
|
170
|
David KK, Andrabi SA, Dawson TM, Dawson VL. Parthanatos, a messenger of death. Front Biosci (Landmark Ed) 2009; 14:1116-28. [PMID: 19273119 DOI: 10.2741/3297] [Citation(s) in RCA: 329] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Poly-ADP-ribose polymerase-1 (PARP-1)'s roles in the cell span from maintaining life to inducing death. The processes PARP-1 is involved in include DNA repair, DNA transcription, mitosis, and cell death. Of PARP-1's different cellular functions, its role in cell death is of particular interest to designing therapies for diseases. Genetic deletion of PARP-1 revealed that PARP-1 overactivation underlies cell death in models of stroke, diabetes, inflammation and neurodegeneration. Since interfering with PARP-1 mediated cell death will be clinically beneficial, great effort has been invested into understanding mechanisms downstream of PARP-1 overactivation. Recent evidence shows that poly-ADP ribose (PAR) polymer itself can act as a cell death effector downstream of PARP-1. We coined the term parthanatos after Thanatos, the personification of death in Greek mythology, to refer to PAR-mediated cell death. In this review, we will present evidence and questions raised by these recent findings, and summarize the proposed mechanisms by which PARP-1 overactivation kills. It is evident that further understanding of parthanatos opens up new avenues for therapy in ameliorating diseases related to PARP-1 overactivation.
Collapse
Affiliation(s)
- Karen Kate David
- Institute for Cell Engineering, The Johns Hopkins University School of Medicine, 733 North Broadway St., Suite 711, Baltimore, MD 21205, USA
| | | | | | | |
Collapse
|
171
|
Kumar S, Sitasawad SL. N-acetylcysteine prevents glucose/glucose oxidase-induced oxidative stress, mitochondrial damage and apoptosis in H9c2 cells. Life Sci 2008; 84:328-36. [PMID: 19159629 DOI: 10.1016/j.lfs.2008.12.016] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2008] [Revised: 12/10/2008] [Accepted: 12/16/2008] [Indexed: 11/16/2022]
Abstract
AIMS High blood glucose may auto-oxidize and generate free radicals, which are proposed to induce apoptosis in cardiac cells. The aim of the present study was to investigate the cell damage induced by glucose/glucose oxidase-dependent oxidative stress and the protective effect of N-acetylcysteine (NAC) on H9c2 cardiac muscle cells. MAIN METHODS H9c2 cells were exposed to 33 mM glucose (G)+1.6 milliunits (mU) of glucose oxidase (GO) and termed G/GO. Cell apoptosis, generation of reactive oxygen species (ROS-super oxide anion and hydrogen peroxide) and reactive nitrogen species (RNS-peroxinitrite), and the change in mitochondrial membrane potential (DeltaPsim) was studied using flow cytometry and confocal microscopy, and cytochrome c release was measured using confocal microscopy. The expression of Bcl-2, Bax and the activation of procaspase-9 was studied by western blot. KEY FINDINGS Exposure of H9c2 cells to G/GO resulted in a significant increase in cellular apoptosis (P<0.05) and the generation of ROS and RNS (P<0.001). Further, G/GO treatment led to a decrease in DeltaPsim, release of cytochrome c, decrease in Bcl-2, increase in Bax expression and the activation of procaspase-9. Treatment with NAC significantly decreased apoptosis (P<0.05) and reduced the levels of ROS and RNS (P<0.001). NAC was also able to normalize DeltaPsim, inhibit cytochrome c release, increase Bcl-2 and decrease Bax expression and procaspase-9 activation. SIGNIFICANCE Our studies suggest that NAC has antioxidative and antiapoptotic activity against G/GO-induced oxidative stress through the inhibition of mitochondrial damage in H9c2 cells.
Collapse
Affiliation(s)
- Santosh Kumar
- National Centre for Cell Science, NCCS Complex, Ganeshkhind, Pune 411007, India
| | | |
Collapse
|
172
|
Activity-dependent neuroprotective protein-derived peptide, NAP, preventing alcohol-induced apoptosis in fetal brain of C57BL/6 mouse. Neuroscience 2008; 158:1426-35. [PMID: 19073235 DOI: 10.1016/j.neuroscience.2008.11.021] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2008] [Revised: 11/05/2008] [Accepted: 11/10/2008] [Indexed: 11/22/2022]
Abstract
Possible prevention of the effects of prenatal alcohol exposure has been investigated using peptides that were previously shown to be involved in neuroprotection both in vitro and in vivo. I focused in this study on investigating the neuroprotective effects of one of these peptides with regard to the determination of the downstream signaling pathways involved in neuroprotection. This peptide with the sequence NAPVSIPQ, known as NAP, a fragment of activity-dependent neuroprotective protein, demonstrated a potent protective effect against oxidative stress associated with alcohol exposure. On embryonic day 7 (E7), weight-matched C57BL/6 pregnant females were assigned the following groups: (1) Ethanol liquid diet group (ALC) 25% (4.49%, v/v) ethano-derived calories, (2) Pair-fed (PF) control group (3) Chow control group, (4) treatment groups with alcohol alongside i.p. injections of d-NAP (ALC/d-NAP, 20 or 30 microg/20 g body weight), (5) PF/d-NAP control group. On E13, fetal brains were collected and assayed for TdT-mediated dUTP nick end labeling (TUNEL) staining, caspase-3 colorimetric assay and ELISA for cytochrome c detection. My results show that NAP significantly prevented alcohol-induced weight reduction of the fetal brain. Apoptosis was determined by TUNEL staining; NAP administration significantly prevented alcohol-induced increases in TUNEL-positive cells in primordium cingulate cortex and basal ganglia eminence. The investigation of downstream signaling pathways involving NAP neuroprotection revealed that this peptide significantly prevented alcohol-induced increase in the concentrations of caspase-3 in E13 fetal brains. Moreover, ELISA for cytochrome c shows that NAP significantly prevented both alcohol-induced increases in the level of cytosolic cytochrome c and alcohol-induced decreases in the level of mitochondrial cytochrome c. These data provide an understanding of NAP intracellular target, and the downstream mechanisms of action that will pave a path toward potential therapeutics against alcohol intoxication during prenatal stages.
Collapse
|
173
|
Abdin AA, Hamouda HE. Mechanism of the neuroprotective role of coenzyme Q10 with or without L-dopa in rotenone-induced parkinsonism. Neuropharmacology 2008; 55:1340-6. [PMID: 18817789 DOI: 10.1016/j.neuropharm.2008.08.033] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2008] [Revised: 07/16/2008] [Accepted: 08/19/2008] [Indexed: 12/21/2022]
Abstract
Current treatment options for parkinsonism as a neurodegenerative disease are limited and still mainly symptomatic and lack significant disease-modifying effect. Understanding its molecular pathology and finding the cause of dopaminergic cell loss will lead to exploring therapies that could prevent and cure the disease. Mitochondrial dysfunction was found to stimulate releasing of reactive oxygen species (ROS) with subsequent induction of apoptotic neuronal cell death. The aim of the present study was to throw the light on the role of coenzyme Q10 with or without L-dopa in an experimental model of parkinsonism induced by rotenone in rats. The present work showed that rotenone (2.5 mg/kg/day i.p. for 60 days) induced a model of parkinsonism (group II) resembling the basic findings in human characterized by bradykinesia and rigidity manifested as an increase in catalepsy score (detected after 20 days with bad prognosis after 60 days) with marked decrease in striatal dopamine levels. This model confirmed the implication of mitochondrial-apoptotic pathway in the pathogenesis of parkinsonism as there was a decrease in levels of striatal complex I activity and ATP as well as extreme overexpression of the antiapoptotic protein Bcl-2, and also exhibited the role of coenzyme Q10 where its plasma and striatal levels were found to be decreased in comparison to the normal control rats (group I). This proposed pathogenesis was evidenced by the significant correlation between catalepsy score and the neurochemical parameters obtained in the current work. The treated groups started to receive the drug(s) after 20 days from induction of parkinsonism and continued to complete for 60 days. Oral administration of Co Q10 in a low dose 200 mg/kg/day (group III) or a high dose 600 mg/kg/day (group IV), resulted in amelioration of the mitochondrial induced apoptosis by dose-dependent restoration of striatal complex I activity, ATP levels with temperate increase in expression of Bcl-2 as well as decrease in catalepsy score. Although both low and high doses of Co Q10 resulted in significant increase in its plasma and striatal levels, but only the high dose was shown to reach the recommended therapeutic levels. As a current replacement therapy, oral administration of levodopa 10 mg/kg/day (group V), caused symptomatic improvement in the form of reduction of catalepsy score with restoration of striatal dopamine levels, but it did not show any significant effects on either striatal complex I activity, ATP levels or the expression of Bcl-2, pointing to the lack of its disease-modifying role. On the other hand, its administration with high dose of coenzyme Q10 caused the most marked symptomatic improvement in catalepsy score when compared to its administration with low dose of coenzyme Q10, or when compared to either coenzyme Q10 high dose or L-dopa, respectively. Moreover, administration of high dose coenzyme Q10 with L-dopa provided a significant increase in striatal complex I activity, ATP levels and Bcl-2 expression in comparison to group administered coenzyme Q10 low dose with L-dopa, in addition to the significant restoration of striatal dopamine levels and both plasma and striatal Co Q10 levels. Regarding that L-dopa is viewed as a replacement therapy in parkinsonism, it could be concluded that addition of coenzyme Q10 in a high dose in early parkinson's disease could be recommended based on its proved disease-modifying role on several levels of the proposed mechanisms, including improvement of respiratory chain activity and intervention with neuronal apoptosis. A further research to investigate other apoptosis-targeted compounds will open a new era in the treatment of parkinsonism.
Collapse
Affiliation(s)
- Amany A Abdin
- Department of Pharmacology, Faculty of Medicine, Tanta University, AL-Geish Street, Tanta, Egypt.
| | | |
Collapse
|
174
|
Paranjpe A, Cacalano NA, Hume WR, Jewett A. Mechanisms of N-acetyl cysteine-mediated protection from 2-hydroxyethyl methacrylate-induced apoptosis. J Endod 2008; 34:1191-7. [PMID: 18793918 DOI: 10.1016/j.joen.2008.06.011] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2008] [Revised: 06/13/2008] [Accepted: 06/23/2008] [Indexed: 01/07/2023]
Abstract
Resin-based materials are now commonly used in dentistry in restorative materials as well as in endodontic sealers. These materials have been shown to be cytotoxic. The mechanisms by which resin-based materials mediate their adverse effects have not been completely elucidated. Here we show that 2-hydroxyethyl methacrylate (HEMA) induces apoptotic cell death in oral keratinocytes and immune cells through the intrinsic cell death pathway. Functional loss and cell death induced by HEMA was significantly inhibited in the presence of N-acetyl cysteine (NAC) treatment. In addition, HEMA induced a decrease in mitochondrial membrane potential, and an increase in cleaved caspases was potently inhibited in the presence of NAC treatment. Overall, the results reported in this article indicate that NAC is an effective chemoprotectant that can safely be used to protect the pulp and the surrounding tissues from adverse effects of dental restorative and endodontic materials.
Collapse
Affiliation(s)
- Avina Paranjpe
- The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, The Jonsson Comprehensive Cancer Center (JCCC), Dental Research Institute, Division of Oral Biology and Medicine, Los Angeles, CA, USA
| | | | | | | |
Collapse
|
175
|
Mercanoglu G, Safran N, Gungor M, Pamukcu B, Uzun H, Sezgin C, Mercanoglu F, Fici F. The effects of nebivolol on apoptosis in a rat infarct model. Circ J 2008; 72:660-70. [PMID: 18362441 DOI: 10.1253/circj.72.660] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND In the present study, nitric oxide (NO) was investigated to see if it mediated effects of nebivolol on apoptosis in the rat myocardial infarction (MI) model. METHODS AND RESULTS Rats were divided into 3 groups: sham operated (sham-control), MI-induced (MI-control) and nebivolol treated (MI-nebivolol). The initial dose of nebivolol was administrated intravenously (iv) within 10 min of post-MI reperfusion and continued orally for 28 days. NO mediated effects of nebivolol were assessed either in the early (2(nd) day) or sub-acute (28(th) day) period of MI by histologic, hemodynamic and biologic studies. Left ventricular (LV) pressure changes were prevented with nebivolol (the increase in LV end-diastolic pressure and the decrease in maximum rise and fall rate of LV pressure (+dp/dt and -dp/dt) was significantly less in MI-nebivolol). Total and regional apoptotic indexes were significantly lower in the MI-nebivolol group (10.2 vs 7.1%, respectively on the 2(nd) day; p=0.004). Although plasma nitrite/nitrate, cyclic guanylate cyclase and peroxynitrite concentrations were high both in MI-control and MI-nebivolol groups on the 2(nd) day, these concentrations were decreased to the basal value on the 28(th) day in the MI-nebivolol group. CONCLUSION As a result, nebivolol treatment (initially by iv within 10 min of reperfusion and continued orally) reduced the myocardial apoptosis after MI. This beneficial effect of nebivolol is mediated by NO regulation.
Collapse
Affiliation(s)
- Guldem Mercanoglu
- Department of Pharmacology, Faculty of Pharmacy, Yeditepe University, Kayisdagi, Istanbul, Turkey.
| | | | | | | | | | | | | | | |
Collapse
|
176
|
Ferraro E, Pulicati A, Cencioni MT, Cozzolino M, Navoni F, di Martino S, Nardacci R, Carrì MT, Cecconi F. Apoptosome-deficient cells lose cytochrome c through proteasomal degradation but survive by autophagy-dependent glycolysis. Mol Biol Cell 2008; 19:3576-88. [PMID: 18550800 DOI: 10.1091/mbc.e07-09-0858] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Cytochrome c release from mitochondria promotes apoptosome formation and caspase activation. The question as to whether mitochondrial permeabilization kills cells via a caspase-independent pathway when caspase activation is prevented is still open. Here we report that proneural cells of embryonic origin, when induced to die but rescued by apoptosome inactivation are deprived of cytosolic cytochrome c through proteasomal degradation. We also show that, in this context, those cells keep generating ATP by glycolysis for a long period of time and that they keep their mitochondria in a depolarized state that can be reverted. Moreover, under these conditions, such apoptosome-deficient cells activate a Beclin 1-dependent autophagy pathway to sustain glycolytic-dependent ATP production. Our findings contribute to elucidating what the point-of-no-return in apoptosis is. They also help in clarifying the issue of survival of apoptosome-deficient proneural cells under stress conditions. Unraveling this issue could be highly relevant for pharmacological intervention and for therapies based on neural stem cell transfer in the treatment of neurological disorders.
Collapse
Affiliation(s)
- Elisabetta Ferraro
- Laboratory of Molecular Neuroembryology, IRCCS Fondazione Santa Lucia, 00143, Rome, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
177
|
Lazarou M, Thorburn DR, Ryan MT, McKenzie M. Assembly of mitochondrial complex I and defects in disease. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2008; 1793:78-88. [PMID: 18501715 DOI: 10.1016/j.bbamcr.2008.04.015] [Citation(s) in RCA: 155] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2008] [Revised: 04/15/2008] [Accepted: 04/25/2008] [Indexed: 12/19/2022]
Abstract
Isolated complex I deficiency is the most common cause of respiratory chain dysfunction. Defects in human complex I result in energy generation disorders and they are also implicated in neurodegenerative disease and altered apoptotic signaling. Complex I dysfunction often occurs as a result of its impaired assembly. The assembly process of complex I is poorly understood, complicated by the fact that in mammals, it is composed of 45 different subunits and is regulated by both nuclear and mitochondrial genomes. However, in recent years we have gained new insights into complex I biogenesis and a number of assembly factors involved in this process have also been identified. In most cases, these factors have been discovered through their gene mutations that lead to specific complex I defects and result in mitochondrial disease. Here we review how complex I is assembled and the factors required to mediate this process.
Collapse
Affiliation(s)
- Michael Lazarou
- Department of Biochemistry, La Trobe University, 3086 Melbourne, Australia
| | | | | | | |
Collapse
|
178
|
Adhikari S, Bhatia M. H2S-induced pancreatic acinar cell apoptosis is mediated via JNK and p38 MAP kinase. J Cell Mol Med 2008; 12:1374-83. [PMID: 18373739 PMCID: PMC3865679 DOI: 10.1111/j.1582-4934.2008.00318.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Treatment of pancreatic acinar cells by hydrogen sulphide has been shown to induce apoptosis. However, a potential role of mitogen-activated protein kinases (MAPKs) in this apoptotic pathway remains unknown. The present study examined the role of MAPKs in H2S-induced apoptosis in mouse pancreatic acinar cells. Pancreatic acinar cells were treated with 10 μM NaHS (a donor of H2S) for 3 hrs. For the evaluation of the role of MAPKs, PD98059, SP600125 and SB203580 were used as MAPKs inhibitors for ERK1/2, JNK1/2 and p38 MAPK, respectively. We observed activation of ERK1/2, JNK1/2 and p38 when pancreatic acini were exposed to H2S. Moreover, H2S-induced ERK1/2, JNK1/2 and p38 activation were blocked by pre-treatment with their corresponding inhibitor in a dose-dependent manner. H2S-induced apoptosis led to an increase in caspase 3 activity and this activity was attenuated when caspase 3 inhibitor were used. Also, the cleavage of caspase 3 correlated with that of poly-(ADP-ribose)-polymerase (PARP) cleavage. H2S treatment induced the release of cytochrome c, smac from mitochondria into the cytoplasm, translocation of Bax into mitochondria and decreased the protein level of Bcl-2. Inhibition of ERK1/2 using PD98059 caused further enhancement of apoptosis as evidenced by annexin V staining, while SP600125 and SB203580 abrogated H2S-induced apoptosis. Taken together, the data suggest that activation of ERKs promotes cell survival, whereas activation of JNKs and p38 MAP kinase leads to H2S-induced apoptosis.
Collapse
Affiliation(s)
- Sharmila Adhikari
- Department of Pharmacology, National University of Singapore, Singapore
| | | |
Collapse
|
179
|
Van Linthout S, Spillmann F, Riad A, Trimpert C, Lievens J, Meloni M, Escher F, Filenberg E, Demir O, Li J, Shakibaei M, Schimke I, Staudt A, Felix SB, Schultheiss HP, De Geest B, Tschöpe C. Human Apolipoprotein A-I Gene Transfer Reduces the Development of Experimental Diabetic Cardiomyopathy. Circulation 2008; 117:1563-73. [DOI: 10.1161/circulationaha.107.710830] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background—
The hallmarks of diabetic cardiomyopathy are cardiac oxidative stress, intramyocardial inflammation, cardiac fibrosis, and cardiac apoptosis. Given the antioxidative, antiinflammatory, and antiapoptotic potential of high-density lipoprotein (HDL), we evaluated the hypothesis that increased HDL via gene transfer (GT) with human apolipoprotein (apo) A-I, the principal apolipoprotein of HDL, may reduce the development of diabetic cardiomyopathy.
Methods and Results—
Intravenous GT with 3×10
12
particles/kg of the E1E3E4-deleted vector
Ad.hapoA-I
, expressing human apoA-I, or
Ad.Null
, containing no expression cassette, was performed 5 days after streptozotocin (STZ) injection. Six weeks after apoA-I GT, HDL cholesterol levels were increased by 1.6-fold (
P
<0.001) compared with diabetic controls injected with the
Ad.Null
vector (STZ-
Ad.Null
). ApoA-I GT and HDL improved LV contractility in vivo and cardiomyocyte contractility ex vivo, respectively. Moreover, apoA-I GT was associated with decreased cardiac oxidative stress and reduced intramyocardial inflammation. In addition, compared with STZ-
Ad.Null
rats, cardiac fibrosis and glycogen accumulation were reduced by 1.7-fold and 3.1-fold, respectively (
P
<0.05). Caspase 3/7 activity was decreased 1.2-fold (
P
<0.05), and the ratio of Bcl-2 to Bax was upregulated 1.9-fold (
P
<0.005), translating to 2.1-fold (
P
<0.05) reduced total number of cardiomyocytes with apoptotic characteristics and 3.0-fold (
P
<0.005) reduced damaged endothelial cells compared with STZ-
Ad.Null
rats. HDL supplementation ex vivo reduced hyperglycemia-induced cardiomyocyte apoptosis by 3.4-fold (
P
<0.005). The apoA-I GT-mediated protection was associated with a 1.6-, 1.6-, and 2.4-fold induction of diabetes-downregulated phospho to Akt, endothelial nitric oxide synthase, and glycogen synthase kinase ratio, respectively (
P
<0.005).
Conclusion—
ApoA-I GT reduced the development of streptozotocin-induced diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Sophie Van Linthout
- From Abteilung für Kardiologie und Pneumologie, Charité-Universitätsklinikum Berlin, Campus Benjamin Franklin, Berlin, Germany (S.V.L., F.S., A.R., M.M., F.E., E.F., O.D., H.-P.S., C. Tschöpe); Klinik für Innere Medizin B, Ernst-Moritz-Arndt-Universität, Greifswald, Germany (C. Trimpert, A.S., S.B.F.); Center for Molecular and Vascular Biology, University of Leuven, Leuven, Belgium (J. Lievens, B.D.G.); Charité University Medicine Berlin, Campus Mitte, Center for Cardiovascular Research (J
| | - Frank Spillmann
- From Abteilung für Kardiologie und Pneumologie, Charité-Universitätsklinikum Berlin, Campus Benjamin Franklin, Berlin, Germany (S.V.L., F.S., A.R., M.M., F.E., E.F., O.D., H.-P.S., C. Tschöpe); Klinik für Innere Medizin B, Ernst-Moritz-Arndt-Universität, Greifswald, Germany (C. Trimpert, A.S., S.B.F.); Center for Molecular and Vascular Biology, University of Leuven, Leuven, Belgium (J. Lievens, B.D.G.); Charité University Medicine Berlin, Campus Mitte, Center for Cardiovascular Research (J
| | - Alexander Riad
- From Abteilung für Kardiologie und Pneumologie, Charité-Universitätsklinikum Berlin, Campus Benjamin Franklin, Berlin, Germany (S.V.L., F.S., A.R., M.M., F.E., E.F., O.D., H.-P.S., C. Tschöpe); Klinik für Innere Medizin B, Ernst-Moritz-Arndt-Universität, Greifswald, Germany (C. Trimpert, A.S., S.B.F.); Center for Molecular and Vascular Biology, University of Leuven, Leuven, Belgium (J. Lievens, B.D.G.); Charité University Medicine Berlin, Campus Mitte, Center for Cardiovascular Research (J
| | - Christiane Trimpert
- From Abteilung für Kardiologie und Pneumologie, Charité-Universitätsklinikum Berlin, Campus Benjamin Franklin, Berlin, Germany (S.V.L., F.S., A.R., M.M., F.E., E.F., O.D., H.-P.S., C. Tschöpe); Klinik für Innere Medizin B, Ernst-Moritz-Arndt-Universität, Greifswald, Germany (C. Trimpert, A.S., S.B.F.); Center for Molecular and Vascular Biology, University of Leuven, Leuven, Belgium (J. Lievens, B.D.G.); Charité University Medicine Berlin, Campus Mitte, Center for Cardiovascular Research (J
| | - Joke Lievens
- From Abteilung für Kardiologie und Pneumologie, Charité-Universitätsklinikum Berlin, Campus Benjamin Franklin, Berlin, Germany (S.V.L., F.S., A.R., M.M., F.E., E.F., O.D., H.-P.S., C. Tschöpe); Klinik für Innere Medizin B, Ernst-Moritz-Arndt-Universität, Greifswald, Germany (C. Trimpert, A.S., S.B.F.); Center for Molecular and Vascular Biology, University of Leuven, Leuven, Belgium (J. Lievens, B.D.G.); Charité University Medicine Berlin, Campus Mitte, Center for Cardiovascular Research (J
| | - Marco Meloni
- From Abteilung für Kardiologie und Pneumologie, Charité-Universitätsklinikum Berlin, Campus Benjamin Franklin, Berlin, Germany (S.V.L., F.S., A.R., M.M., F.E., E.F., O.D., H.-P.S., C. Tschöpe); Klinik für Innere Medizin B, Ernst-Moritz-Arndt-Universität, Greifswald, Germany (C. Trimpert, A.S., S.B.F.); Center for Molecular and Vascular Biology, University of Leuven, Leuven, Belgium (J. Lievens, B.D.G.); Charité University Medicine Berlin, Campus Mitte, Center for Cardiovascular Research (J
| | - Felicitas Escher
- From Abteilung für Kardiologie und Pneumologie, Charité-Universitätsklinikum Berlin, Campus Benjamin Franklin, Berlin, Germany (S.V.L., F.S., A.R., M.M., F.E., E.F., O.D., H.-P.S., C. Tschöpe); Klinik für Innere Medizin B, Ernst-Moritz-Arndt-Universität, Greifswald, Germany (C. Trimpert, A.S., S.B.F.); Center for Molecular and Vascular Biology, University of Leuven, Leuven, Belgium (J. Lievens, B.D.G.); Charité University Medicine Berlin, Campus Mitte, Center for Cardiovascular Research (J
| | - Elena Filenberg
- From Abteilung für Kardiologie und Pneumologie, Charité-Universitätsklinikum Berlin, Campus Benjamin Franklin, Berlin, Germany (S.V.L., F.S., A.R., M.M., F.E., E.F., O.D., H.-P.S., C. Tschöpe); Klinik für Innere Medizin B, Ernst-Moritz-Arndt-Universität, Greifswald, Germany (C. Trimpert, A.S., S.B.F.); Center for Molecular and Vascular Biology, University of Leuven, Leuven, Belgium (J. Lievens, B.D.G.); Charité University Medicine Berlin, Campus Mitte, Center for Cardiovascular Research (J
| | - Okan Demir
- From Abteilung für Kardiologie und Pneumologie, Charité-Universitätsklinikum Berlin, Campus Benjamin Franklin, Berlin, Germany (S.V.L., F.S., A.R., M.M., F.E., E.F., O.D., H.-P.S., C. Tschöpe); Klinik für Innere Medizin B, Ernst-Moritz-Arndt-Universität, Greifswald, Germany (C. Trimpert, A.S., S.B.F.); Center for Molecular and Vascular Biology, University of Leuven, Leuven, Belgium (J. Lievens, B.D.G.); Charité University Medicine Berlin, Campus Mitte, Center for Cardiovascular Research (J
| | - Jun Li
- From Abteilung für Kardiologie und Pneumologie, Charité-Universitätsklinikum Berlin, Campus Benjamin Franklin, Berlin, Germany (S.V.L., F.S., A.R., M.M., F.E., E.F., O.D., H.-P.S., C. Tschöpe); Klinik für Innere Medizin B, Ernst-Moritz-Arndt-Universität, Greifswald, Germany (C. Trimpert, A.S., S.B.F.); Center for Molecular and Vascular Biology, University of Leuven, Leuven, Belgium (J. Lievens, B.D.G.); Charité University Medicine Berlin, Campus Mitte, Center for Cardiovascular Research (J
| | - Mehdi Shakibaei
- From Abteilung für Kardiologie und Pneumologie, Charité-Universitätsklinikum Berlin, Campus Benjamin Franklin, Berlin, Germany (S.V.L., F.S., A.R., M.M., F.E., E.F., O.D., H.-P.S., C. Tschöpe); Klinik für Innere Medizin B, Ernst-Moritz-Arndt-Universität, Greifswald, Germany (C. Trimpert, A.S., S.B.F.); Center for Molecular and Vascular Biology, University of Leuven, Leuven, Belgium (J. Lievens, B.D.G.); Charité University Medicine Berlin, Campus Mitte, Center for Cardiovascular Research (J
| | - Ingolf Schimke
- From Abteilung für Kardiologie und Pneumologie, Charité-Universitätsklinikum Berlin, Campus Benjamin Franklin, Berlin, Germany (S.V.L., F.S., A.R., M.M., F.E., E.F., O.D., H.-P.S., C. Tschöpe); Klinik für Innere Medizin B, Ernst-Moritz-Arndt-Universität, Greifswald, Germany (C. Trimpert, A.S., S.B.F.); Center for Molecular and Vascular Biology, University of Leuven, Leuven, Belgium (J. Lievens, B.D.G.); Charité University Medicine Berlin, Campus Mitte, Center for Cardiovascular Research (J
| | - Alexander Staudt
- From Abteilung für Kardiologie und Pneumologie, Charité-Universitätsklinikum Berlin, Campus Benjamin Franklin, Berlin, Germany (S.V.L., F.S., A.R., M.M., F.E., E.F., O.D., H.-P.S., C. Tschöpe); Klinik für Innere Medizin B, Ernst-Moritz-Arndt-Universität, Greifswald, Germany (C. Trimpert, A.S., S.B.F.); Center for Molecular and Vascular Biology, University of Leuven, Leuven, Belgium (J. Lievens, B.D.G.); Charité University Medicine Berlin, Campus Mitte, Center for Cardiovascular Research (J
| | - Stephan B. Felix
- From Abteilung für Kardiologie und Pneumologie, Charité-Universitätsklinikum Berlin, Campus Benjamin Franklin, Berlin, Germany (S.V.L., F.S., A.R., M.M., F.E., E.F., O.D., H.-P.S., C. Tschöpe); Klinik für Innere Medizin B, Ernst-Moritz-Arndt-Universität, Greifswald, Germany (C. Trimpert, A.S., S.B.F.); Center for Molecular and Vascular Biology, University of Leuven, Leuven, Belgium (J. Lievens, B.D.G.); Charité University Medicine Berlin, Campus Mitte, Center for Cardiovascular Research (J
| | - Heinz-Peter Schultheiss
- From Abteilung für Kardiologie und Pneumologie, Charité-Universitätsklinikum Berlin, Campus Benjamin Franklin, Berlin, Germany (S.V.L., F.S., A.R., M.M., F.E., E.F., O.D., H.-P.S., C. Tschöpe); Klinik für Innere Medizin B, Ernst-Moritz-Arndt-Universität, Greifswald, Germany (C. Trimpert, A.S., S.B.F.); Center for Molecular and Vascular Biology, University of Leuven, Leuven, Belgium (J. Lievens, B.D.G.); Charité University Medicine Berlin, Campus Mitte, Center for Cardiovascular Research (J
| | - Bart De Geest
- From Abteilung für Kardiologie und Pneumologie, Charité-Universitätsklinikum Berlin, Campus Benjamin Franklin, Berlin, Germany (S.V.L., F.S., A.R., M.M., F.E., E.F., O.D., H.-P.S., C. Tschöpe); Klinik für Innere Medizin B, Ernst-Moritz-Arndt-Universität, Greifswald, Germany (C. Trimpert, A.S., S.B.F.); Center for Molecular and Vascular Biology, University of Leuven, Leuven, Belgium (J. Lievens, B.D.G.); Charité University Medicine Berlin, Campus Mitte, Center for Cardiovascular Research (J
| | - Carsten Tschöpe
- From Abteilung für Kardiologie und Pneumologie, Charité-Universitätsklinikum Berlin, Campus Benjamin Franklin, Berlin, Germany (S.V.L., F.S., A.R., M.M., F.E., E.F., O.D., H.-P.S., C. Tschöpe); Klinik für Innere Medizin B, Ernst-Moritz-Arndt-Universität, Greifswald, Germany (C. Trimpert, A.S., S.B.F.); Center for Molecular and Vascular Biology, University of Leuven, Leuven, Belgium (J. Lievens, B.D.G.); Charité University Medicine Berlin, Campus Mitte, Center for Cardiovascular Research (J
| |
Collapse
|
180
|
PTD-mediated delivery of anti-cell death proteins/peptides and therapeutic enzymes. Adv Drug Deliv Rev 2008; 60:499-516. [PMID: 18093693 DOI: 10.1016/j.addr.2007.09.011] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2007] [Accepted: 09/12/2007] [Indexed: 01/28/2023]
Abstract
Millions of unnecessary cells are removed from our body everyday by apoptosis to ensure our survivals. Apoptosis is a highly coordinated process. Failure in apoptotic regulation results in disease. A large number of studies have demonstrated that accelerated apoptosis is involved in degenerative diseases, ischemic injuries, immunodeficiency and infertility. These studies have also revealed the molecular mechanisms of apoptosis signal transduction to provide therapeutic targets. On the other hand, protein transduction technology has been developed to deliver full-length proteins to various tissues including the brain. So far, many studies have shown that in vivo delivery of therapeutic proteins/peptides, including anti-apoptotic proteins, an anti-oxidant enzyme, a neuroprotectant, enzymes involved in purine or tyrosine metabolism, caspase inhibitors, c-Jun N-terminal kinase inhibitors and an NF-kappaB inhibitor, by protein transduction technology mitigates various diseases in animal models.
Collapse
|
181
|
Contribution of calpain activation to early stages of hippocampal damage during oxygen–glucose deprivation. Brain Res 2008; 1196:121-30. [DOI: 10.1016/j.brainres.2007.12.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2007] [Revised: 11/30/2007] [Accepted: 12/01/2007] [Indexed: 01/19/2023]
|
182
|
Abstract
Mitochondria have multiple functions in eukaryotic cells and are organized into dynamic tubular networks that continuously undergo changes through coordinated fusion and fission and migration through the cytosol. Mitochondria integrate cell-signaling networks, especially those involving the intracellular messenger Ca(2+), into the regulation of metabolic pathways. Recently, it has become clear that mitochondria are central to the three main cell death pathways, namely necrosis, apoptosis, and autophagic cell death. This article discusses the role of mitochondria in drug-induced cholestatic injury to the liver. The role of mitochondria in the cellular adaptation against the toxic effects of bile acids is discussed also.
Collapse
Affiliation(s)
- George E N Kass
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH, UK.
| | | |
Collapse
|
183
|
Maulik N, Das DK. Emerging potential of thioredoxin and thioredoxin interacting proteins in various disease conditions. Biochim Biophys Acta Gen Subj 2008; 1780:1368-82. [PMID: 18206121 DOI: 10.1016/j.bbagen.2007.12.008] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2007] [Revised: 12/12/2007] [Accepted: 12/17/2007] [Indexed: 12/16/2022]
Abstract
Reactive oxygen species (ROS) are known to be mediators of intracellular signaling pathways. However the excessive production of ROS may be detrimental to the cell as a result of the increased oxidative stress and loss of cell function. Hence, well tuned, balanced and responsive antioxidant systems are vital for proper regulation of the redox status of the cell. The cells are normally able to defend themselves against the oxidative stress induced damage through the use of several antioxidant systems. Even though the free radical scavenging enzymes such as superoxide dismutase (SOD) and catalase can handle huge amounts of reactive oxygen species, should these systems fail some reactive molecules will evade the detoxification process and damage potential targets. In such a scenario, cells recruit certain small molecules and proteins as 'rescue specialists' in case the 'bodyguards' fail to protect potential targets from oxidative damage. The thioredoxin (Trx) system thus plays a vital role in the maintenance of a reduced intracellular redox state which is essential for the proper functioning of each individual cell. Trx alterations have been implicated in many diseases such as cataract formation, ischemic heart diseases, cancers, AIDS, complications of diabetes, hypertension etc. The interactions of Trx with many different proteins and different metabolic and signaling pathways as well as the significant species differences make it an attractive target for therapeutic intervention in many fields of medical science. In this review, we present, the critical roles that thioredoxins play in limiting oxidant stress through either its direct effect as an antioxidant or through its interactions with other key signaling proteins (thioredoxin interacting proteins) and its implications in various disease models.
Collapse
Affiliation(s)
- Nilanjana Maulik
- Cardiovascular Research Center, University of Connecticut Health Center, Farmington, Connecticut, 06030 USA
| | | |
Collapse
|
184
|
Rasola A, Bernardi P. The mitochondrial permeability transition pore and its involvement in cell death and in disease pathogenesis. Apoptosis 2008; 12:815-33. [PMID: 17294078 DOI: 10.1007/s10495-007-0723-y] [Citation(s) in RCA: 389] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Current research on the mitochondrial permeability transition pore (PTP) and its role in cell death faces a paradox. Initially considered as an in vitro artifact of little pathophysiological relevance, in recent years the PTP has received considerable attention as a potential mechanism for the execution of cell death. The recent successful use of PTP desensitizers in several disease paradigms leaves little doubt about its relevance in pathophysiology; and emerging findings that link the PTP to key cellular signalling pathways are increasing the interest on the pore as a pharmacological target. Yet, recent genetic data have challenged popular views on the molecular nature of the PTP, and called into question many early conclusions about its structure. Here we review basic concepts about PTP structure, function and regulation within the framework of intracellular death signalling, and its role in disease pathogenesis.
Collapse
Affiliation(s)
- Andrea Rasola
- CNR Institute of Neuroscience and Department of Biomedical Sciences, University of Padova, Viale Giuseppe Colombo 3, I-35121 Padua, Italy.
| | | |
Collapse
|
185
|
Abstract
The crucial step in the intrinsic, or mitochondrial, apoptotic pathway is permeabilization of the mitochondrial outer membrane. Permeabilization triggers release of apoptogenic factors, such as cytochrome c, from the mitochondrial intermembrane space into the cytosol where these factors ensure propagation of the apoptotic cascade and execution of cell death. However, the mechanism(s) underlying permeabilization of the outer membrane remain controversial. Two mechanisms, involving opening of two different mitochondrial channels, have been proposed to be responsible for the permeabilization; the permeability transition pore (PTP) in the inner membrane and the mitochondrial apoptosis-induced channel (MAC) in the outer membrane. Opening of PTP would lead to matrix swelling, subsequent rupture of the outer membrane, and an unspecific release of intermembrane proteins into the cytosol. However, many believe PTP opening is a consequence of apoptosis and this channel is thought to principally play a role in necrosis, not apoptosis. Activation of MAC is exquisitely regulated by Bcl-2 family proteins, which are the sentinels of apoptosis. MAC provides specific pores in the outer membrane for the passage of intermembrane proteins, in particular cytochrome c, to the cytosol. The electrophysiological characteristics of MAC are very similar to Bax channels and depletion of Bax significantly diminishes MAC activity, suggesting that Bax is an essential constituent of MAC in some systems. The characteristics of various mitochondrial channels and Bax are compared. The involvement of MAC and PTP activities in apoptosis of disease and their pharmacology are discussed.
Collapse
Affiliation(s)
- Kathleen W Kinnally
- Department of Basic Sciences, New York University College of Dentistry, 345 East 24th Street, New York, NY 10010, USA.
| | | |
Collapse
|
186
|
Lorenzo HK, Susin SA. Therapeutic potential of AIF-mediated caspase-independent programmed cell death. Drug Resist Updat 2008; 10:235-55. [PMID: 18180198 DOI: 10.1016/j.drup.2007.11.001] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2007] [Accepted: 11/01/2007] [Indexed: 01/15/2023]
Abstract
Resistance to anticancer drugs is often related to deficient cell death execution pathways in cancer cells. Apoptosis, which denotes a form of cell death executed by caspases, was traditionally considered as the only physiological and programmed form of cell death. However, recent evidence indicates that programmed cell death (PCD) can occur in complete absence of caspase activation. Indeed, a large number of caspase-independent models are now defined and a key protein implicated in this type of PCD, apoptosis-inducing factor (AIF), has been identified. AIF is a mitochondrial protein with two faces looking in opposite life/death directions. Recently, the identification of five different isoforms allowed a better characterization of AIFs life/mitochondrial versus death/nuclear functions, as well as definition of its pro-apoptotic region and some of its nuclear partners. Importantly, much work on caspase-independent PCD has revealed that AIF participates in more PCD systems than initially thought. A wider molecular knowledge of AIF, and of the caspase-independent PCDs in which it is involved, are key to provide new insights into the role of PCD. There is no doubt that these insights will lead to the development of more selective and efficient drugs against cancer, degenerative diseases, and other pathological disorders implicating AIF.
Collapse
Affiliation(s)
- Hans K Lorenzo
- INSERM U542, Institut André Lwoff, Lavoisier Building, 94803 Villejuif, France.
| | | |
Collapse
|
187
|
Zhang WH, Wang H, Wang X, Narayanan MV, Stavrovskaya IG, Kristal BS, Friedlander RM. Nortriptyline protects mitochondria and reduces cerebral ischemia/hypoxia injury. Stroke 2008; 39:455-62. [PMID: 18174477 DOI: 10.1161/strokeaha.107.496810] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
BACKGROUND AND PURPOSE Nortriptyline, an antidepressant, was identified as a strong inhibitor of mitochondrial permeability transition by our screening of a library of 1040 drugs. Because mitochondrial permeability transition and consequent mitochondrial dysfunction have been implicated in acute neuronal death, we proposed to investigate the possible neuroprotective effects of nortriptyline in cerebral ischemia. METHODS The effects of nortriptyline were first studied in oxygen/glucose deprivation-induced death of primary cerebrocortical neurons, a cellular model of cerebral ischemia. Mitochondrial membrane potential, mitochondrial factor release, and caspase 3 activation were evaluated after its treatment. Nortriptyline was also studied in a mouse model, which was established by occlusion of the middle cerebral artery. The infarct volume, neurological function, and biochemical events were examined in the absence or the presence of nortriptyline. RESULTS Nortriptyline inhibits oxygen/glucose deprivation-induced cell death, loss of mitochondrial membrane potential, downstream release of mitochondrial factors, and activation of caspase 3 in primary cerebrocortical neurons. Furthermore, it decreases infarct size and improves neurological scores after middle cerebral artery occlusion in mice. CONCLUSIONS The ability of nortriptyline to inhibit mitochondrial factor release and caspase activation and further protect the animals correlates to its inhibitory effect on mitochondrial permeability transition in isolated mitochondria. This study indicated that nortriptyline is neuroprotective against cerebral ischemia. It also suggested mitochondrial permeability transition might be a valuable therapeutic target for acute neurodegeneration.
Collapse
Affiliation(s)
- Wen-hua Zhang
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, 221 Longwood Avenue, LMRC 113, Boston, MA 02115, USA
| | | | | | | | | | | | | |
Collapse
|
188
|
Joubert V, Bourthoumieu S, Leveque P, Yardin C. Apoptosis is Induced by Radiofrequency Fields through the Caspase-Independent Mitochondrial Pathway in Cortical Neurons. Radiat Res 2008; 169:38-45. [DOI: 10.1667/rr1077.1] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2007] [Accepted: 08/29/2007] [Indexed: 11/03/2022]
|
189
|
|
190
|
Lee WYW, Chiu LCM, Yeung JHK. Cytotoxicity of major tanshinones isolated from Danshen (Salvia miltiorrhiza) on HepG2 cells in relation to glutathione perturbation. Food Chem Toxicol 2008; 46:328-38. [PMID: 17892911 DOI: 10.1016/j.fct.2007.08.013] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2006] [Revised: 08/07/2007] [Accepted: 08/12/2007] [Indexed: 11/23/2022]
Abstract
Tanshinones are abietane type-diterpene quinones isolated from the roots of Radix Salvia miltiorrhiza (Danshen), a well-known traditional Chinese medicine in the treatment of cardiovascular diseases. Among the major diterpenes isolated, including cryptotanshinone, tanshinone I, tanshinone IIA and dihydrotanshinone, tanshinone IIA had been shown to posses various pharmacological activities including antioxidant, protection/prevention from angina pectoris and myocardial infarction, and anticancer properties. Tanshinone IIA, usually the most abundant tanshinone present in the herb, has been the focus of studies in its clinical potential, among which its ability to inhibit the proliferation of cancer cell lines. The aim of this study was to study the cytotoxicity of the tanshinones on human HepG2 cells in vitro in relation to intracellular glutathione perturbation (reduced glutathione, GSH and oxidized glutathione, GSSG). Studies using MTT assay showed that all tanshinones decreased cell viability of HepG2 cells in a concentration-dependent manner, with the cell viability decreased to 60% and 35% after 24 h and 48 h treatment, respectively. Assessment of apoptotic cells with fragmented DNA by flow cytometry indicated that only tanshinone IIA (12.5 and 25 microM) induced apoptosis in the cancer cells. Tanshinone IIA and cryptotanshinone caused significant decreases in G(1) cells by 23% and 13%, respectively, after 24 h treatment. The declines in G(1) cells were compensated by increases in G(2)/M (15% for tanshinone IIA) and S cells (8% and 13% for tanshinone IIA and cryptotanshinone, respectively). All the tanshinones studied, except tanshinone IIA, elevated GSH/GSSG ratio at low concentrations (1.56 and 3.13 microM), but the ratio decreased, indicating oxidative stress at high concentrations (6.25-25 microM). Taken together, tanshinone IIA caused HepG2 cytotoxicity through apoptosis without influencing oxidative stress, while the other tanshinones showed lower efficacy in inducing apoptosis in the HepG2 cells.
Collapse
Affiliation(s)
- W Y W Lee
- Department of Pharmacology, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | | | | |
Collapse
|
191
|
Ferlini C, Scambia G. Assay for apoptosis using the mitochondrial probes, Rhodamine123 and 10-N-nonyl acridine orange. Nat Protoc 2007; 2:3111-4. [DOI: 10.1038/nprot.2007.397] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
192
|
Yasuhara S, Asai A, Sahani ND, Martyn JAJ. Mitochondria, endoplasmic reticulum, and alternative pathways of cell death in critical illness. Crit Care Med 2007; 35:S488-95. [PMID: 17713398 DOI: 10.1097/01.ccm.0000278045.91575.30] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Dying cells are distinguished by their biochemical and morphologic traits and categorized into three subtypes: apoptosis, oncosis (necrosis), and cell death with autophagy. Each of these types of cell death plays critical roles in tissue morphogenesis during normal development and in the pathogenesis of human diseases. Given that tissue homeostasis is controlled by the intricate balance between degeneration and regeneration, it is essential to understand the mechanisms of different forms of cell death to establish and improve therapeutic interventions for prevention and rescue of these cell death-related disorders. Critical illness, including sepsis, trauma, and burn injury, is often complicated by multiple organ dysfunction syndrome and is accompanied by increased cell death in parenchymal and nonparenchymal tissues. Accumulating evidence suggests that augmented cell death plays an important role in the organ failure in critical illness. We discuss possible therapeutic approaches for prevention of cell death, particularly apoptotic cell death.
Collapse
Affiliation(s)
- Shingo Yasuhara
- Department of Anesthesiology and Critical Care, Massachusetts General Hospital, Shriners Hospital for Children, and Harvard Medical School, Boston, MA, USA.
| | | | | | | |
Collapse
|
193
|
George J, Gondi CS, Dinh DH, Gujrati M, Rao JS. Restoration of tissue factor pathway inhibitor-2 in a human glioblastoma cell line triggers caspase-mediated pathway and apoptosis. Clin Cancer Res 2007; 13:3507-17. [PMID: 17575213 PMCID: PMC1905856 DOI: 10.1158/1078-0432.ccr-06-3023] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The induction of apoptotic pathways in cancer cells offers a novel and potentially useful approach to improve patient responses to conventional chemotherapy. Tissue factor pathway inhibitor-2 (TFPI-2) is a protease inhibitor that is abundant in the extracellular matrix and highly expressed in noninvasive cells but absent or undetectable in highly invasive human glioblastoma cells. EXPERIMENTAL DESIGN Using a recombinant adeno-associated viral vector carrying human TFPI-2 cDNA, we stably expressed TFPI-2 in U-251 cells, a highly invasive human glioblastoma cell line. Our previous studies showed that restoration of TFPI-2 in glioblastomas effectively prevents cell proliferation, angiogenesis, and tumor invasion. In this study, we determined whether TFPI-2 restoration could induce apoptosis through the caspase-mediated signaling pathway. RESULTS The results from nuclear chromatin staining, terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling assay, and fluorescence-activated cell sorting analysis showed increased apoptosis in U-251 cells after restoration of TFPI-2. Caspase-9 and caspase-3 activity assays showed increased activity, indicating enhanced apoptosis. Immunofluorescence for cleaved caspase-9 and caspase-3 depicted increased expression and colocalization of both molecules. Western blot analysis showed increased transcriptional activities of Fas ligand, tumor necrosis factor-alpha, Bax, Fas-associated death domain, and tumor necrosis factor receptor 1-associated death domain as well as elevated levels of cleaved caspases and poly(ADP-ribose) polymerase. Semiquantitative reverse transcription-PCR depicted increased expression of tumor necrosis factor-alpha and Fas ligand and the related death domains tumor necrosis factor receptor 1-associated death domain and Fas-associated death domain. CONCLUSIONS Taken together, these results show that restoration of TFPI-2 activates both intrinsic and extrinsic caspase-mediated, proapoptotic signaling pathways and induces apoptosis in U-251 cells. Furthermore, our study suggests that recombinant adeno-associated viral vector-mediated gene expression offers a novel tool for cancer gene therapy.
Collapse
Affiliation(s)
- Joseph George
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, IL 61605, USA
| | - Christopher S. Gondi
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, IL 61605, USA
| | - Dzung H. Dinh
- Department of Neurosurgery, University of Illinois at Chicago, College of Medicine at Peoria, Peoria, IL 61605, USA
| | - Meena Gujrati
- Department of Pathology, University of Illinois at Chicago, College of Medicine at Peoria, Peoria, IL 61605, USA
| | - Jasti S. Rao
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, IL 61605, USA
- Department of Neurosurgery, University of Illinois at Chicago, College of Medicine at Peoria, Peoria, IL 61605, USA
- *Address for correspondence: Jasti S. Rao, Ph.D., Department of Cancer Biology and Pharmacology, University of Illinois, College of Medicine at Peoria, Peoria, IL 61605, USA, Phone: 309-671-3445, Fax: 309-671-3442, E-mail:
| |
Collapse
|
194
|
Kim JY, Chung JY, Park JE, Lee SG, Kim YJ, Cha MS, Han MS, Lee HJ, Yoo YH, Kim JM. Benzo[a]pyrene induces apoptosis in RL95-2 human endometrial cancer cells by cytochrome P450 1A1 activation. Endocrinology 2007; 148:5112-22. [PMID: 17640999 DOI: 10.1210/en.2007-0096] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Benzo[a]pyrene (B[a]P) has been shown to be an inducer of apoptosis in some cell types. To date, due to the lack of an appropriate model system, studies of the cellular and biochemical mechanism(s) by which B[a]P induces apoptosis have been focused on Hepa1c1c7 cells. Moreover, the precise relationship between the bioactivation of B[a]P by CYP1A1 or CYP1B1 and the occurrence of cytotoxicity-mediated apoptosis requires further elucidation. In the present study, we showed that B[a]P-induced apoptosis in RL95-2 cells is accompanied by the activation of caspases. In addition, the mitochondrial changes, including the decrease of mitochondrial potential and the release of mitochondrial cytochrome c and second mitochondria-derived activator of caspases/direct inhibitor of apoptosis protein binding protein with low PI (Smac/DIABLO) into the cytosol, support the suggestion that the mitochondrial pathway is robustly associated with B[a]P-evoked apoptosis. This study showed the involvement of the nuclear translocation of mitochondrial apoptosis-inducing factor in B[a]P-induced apoptosis of RL95-2 cells. Exposure to B[a]P up-regulates aryl hydrocarbon receptor, heat-shock protein 90, cytochrome P450 1A1 (CYP1A1), cytochrome P450 1B1 (CYP1B1), and epoxide hydrolase significantly, which might be prerequisites for the conversion of B[a]P to B[a]P-7,8-dihydroxy-9,10-epoxide. Although both CYP1A1 and CYP1B1 proteins were up-regulated significantly by B[a]P, only CYP1A1 exhibited activity. Thus, CYP1A1 is believed to be a central oxidative enzyme that is ultimately required for formation of B[a]P-7,8-dihydroxy-9,10-epoxide from B[a]P in RL95-2 cells. Altogether, our data showed that RL95-2 cells are susceptible to apoptosis by exposure to B[a]P and that B[a]P-evoked apoptosis is mediated predominantly by the activation of CYP1A1. Here we suggest that RL95-2 cells are an excellent model for the investigation of xenobiotic mechanisms associated with CYP1A1 as well as CYP1B1.
Collapse
Affiliation(s)
- Ji Young Kim
- Department of Anatomy and Cell Biology, College of Medicine, Dong-A University, Dongdaeshin-dong 3-1, Seo-gu, Busan 602-714, Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
195
|
San-Miguel B, Alvarez M, Culebras JM, González-Gallego J, Tuñón MJ. N-acetyl-cysteine protects liver from apoptotic death in an animal model of fulminant hepatic failure. Apoptosis 2007; 11:1945-57. [PMID: 17021698 DOI: 10.1007/s10495-006-0090-0] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND This work was undertaken to investigate whether treatment with N-acetyl-cysteine (NAC) prevents oxidative stress and inhibits the apoptotic pathways in an animal model of fulminant hepatic failure. METHODS Rabbits were experimentally infected with 2x10(4) hemagglutination units of a rabbit hemorrhagic disease virus isolate. RESULTS The spontaneous mortality rate of infected animals was 67% at 36 h post infection (pi) and 90% at 48 h pi. This percentage decreased significantly in animals receiving an i.p. injection of NAC (150 mg/kg body way/daily), for 7 days prior to infection. From 36 h pi marked increases were detected in blood levels of transaminases, lactate dehydrogenase, bilirubin and the oxidised/reduced glutathione ratio. All these effects were significantly prevented by NAC treatment. The Bax to Bcl-2 relative expression, the expression of FasL, cytochrome c and PARP-1, and the activity of caspase 3 were significantly increased at 36 and 48 h pi in infected animals. These changes were markedly reduced in animals treated with NAC, with the exception of FasL. CONCLUSION Our results suggest a potential hepatoprotective role of NAC in fulminant hepatic failure, mediated partially through the modulation of the intrinsic pathway of apoptosis.
Collapse
Affiliation(s)
- B San-Miguel
- Department of Physiology, University of León, 24071, León, Spain
| | | | | | | | | |
Collapse
|
196
|
Hamacher-Brady A, Brady NR, Gottlieb RA. The interplay between pro-death and pro-survival signaling pathways in myocardial ischemia/reperfusion injury: apoptosis meets autophagy. Cardiovasc Drugs Ther 2007; 20:445-62. [PMID: 17149555 DOI: 10.1007/s10557-006-0583-7] [Citation(s) in RCA: 117] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Programmed cell death of cardiac myocytes occurs following a bout of ischemia/reperfusion (I/R), which results in reduced function of the heart. Numerous studies, including in vivo, have shown that cell death occurs via necrosis and apoptosis following I/R. Recently, autophagy has emerged as a powerful mediator of programmed cell death, either opposing or enhancing apoptosis, or acting as an alternative form of programmed cell death distinct from apoptosis. AIM Here we review the apoptotic and autophagic signaling pathways, their influences on each other, and we discuss the relevance of autophagy in the heart.
Collapse
Affiliation(s)
- Anne Hamacher-Brady
- Department of Molecular and Experimental Medicine MEM-220, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, San Diego, CA 92037, USA
| | | | | |
Collapse
|
197
|
Bras M, Yuste VJ, Roué G, Barbier S, Sancho P, Virely C, Rubio M, Baudet S, Esquerda JE, Merle-Béral H, Sarfati M, Susin SA. Drp1 mediates caspase-independent type III cell death in normal and leukemic cells. Mol Cell Biol 2007; 27:7073-88. [PMID: 17682056 PMCID: PMC2168919 DOI: 10.1128/mcb.02116-06] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Ligation of CD47 triggers caspase-independent programmed cell death (PCD) in normal and leukemic cells. Here, we characterize the morphological and biochemical features of this type of death and show that it displays the hallmarks of type III PCD. A molecular and biochemical approach has led us to identify a key mediator of this type of death, dynamin-related protein 1 (Drp1). CD47 ligation induces Drp1 translocation from cytosol to mitochondria, a process controlled by chymotrypsin-like serine proteases. Once in mitochondria, Drp1 provokes an impairment of the mitochondrial electron transport chain, which results in dissipation of mitochondrial transmembrane potential, reactive oxygen species generation, and a drop in ATP levels. Surprisingly, neither the activation of the most representative proapoptotic members of the Bcl-2 family, such as Bax or Bak, nor the release of apoptogenic proteins AIF (apoptosis-inducing factor), cytochrome c, endonuclease G (EndoG), Omi/HtrA2, or Smac/DIABLO from mitochondria to cytosol is observed. Responsiveness of cells to CD47 ligation increases following Drp1 overexpression, while Drp1 downregulation confers resistance to CD47-mediated death. Importantly, in B-cell chronic lymphocytic leukemia cells, mRNA levels of Drp1 strongly correlate with death sensitivity. Thus, this previously unknown mechanism controlling caspase-independent type III PCD may provide the basis for novel therapeutic approaches to overcome apoptotic avoidance in malignant cells.
Collapse
Affiliation(s)
- Marlène Bras
- Apoptose et Système Immunitaire, CNRS-URA 1961, Institut Pasteur, 25 rue du Dr. Roux, 75015 Paris, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
198
|
Michod D, Widmann C. DNA-damage sensitizers: Potential new therapeutical tools to improve chemotherapy. Crit Rev Oncol Hematol 2007; 63:160-71. [PMID: 17544289 DOI: 10.1016/j.critrevonc.2007.04.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2006] [Revised: 04/12/2007] [Accepted: 04/24/2007] [Indexed: 01/01/2023] Open
Abstract
Agents that induce DNA damage in cells--the so-called genotoxins--have successfully been used for decades to treat patients with tumors. Genotoxins alter the DNA of cells, which is detected by DNA damage sensors and which leads to the activation of p53. Activation of p53 can lead to the death of cancer cells. The efficacy of genotoxins in humans is however limited by their toxicity to normal tissues. Specific sensitization of tumor cells to the action of genotoxins would reduce the efficacious doses of genotoxins to be used in patients, diminishing the detrimental side-effects of the drugs on normal tissues. A series of compounds able to sensitize cancer cells to DNA-damaging drugs have recently been identified that have the potential to increase the efficacy of currently used anti-cancer treatments.
Collapse
Affiliation(s)
- David Michod
- Department of Physiology and Department of Cell Biology and Morphology, Biology and Medicine Faculty, Lausanne University, Switzerland
| | | |
Collapse
|
199
|
Moon DO, Kim MO, Lee JD, Choi YH, Lee MK, Kim GY. Molecular mechanisms of ZD1839 (Iressa)-induced apoptosis in human leukemic U937 cells. Acta Pharmacol Sin 2007; 28:1205-14. [PMID: 17640484 DOI: 10.1111/j.1745-7254.2007.00615.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
AIM To investigate the molecular mechanisms of ZD1839-induced apoptosis in human leukemic U937 cells. METHODS The inhibition of human leukemic U937 cell growth was assessed by 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphnyl-2H-tetrazolim bromide (MTT) assays, lactate dehydrogenase (LDH) release, and cell cycle distribution. The expression of anti- and pro-apoptotic proteins was detected by Western blot analysis. RESULTS This study demonstrated that ZD1839 induced apoptosis in leukemic U937 cells by the downregulation of Bcl-2, caspase activation and subsequent apoptotic features. Cotreatment with ZD1839 and the caspase-3 inhibitor z-DEVD-fmk blocked apoptosis, indicating that caspase-3 activation is at least partially responsible for ZD1839-induced apoptosis. The ectopic expression of Bcl-2 attenuated caspase-3 activation, PARP cleavage, and subsequent indicators of apoptosis, including sub-G1 DNA content and LDH release. These results indicate that the downregulation of Bcl-2 plays a major role in the initiation of ZD1839-induced apoptosis, and that the activation of a caspase cascade is involved in the execution of apoptosis. Furthermore, ZD1839 treatment triggered the activation of p38 mitogen-activated protein kinase (MAPK) and the down-regulation of c-Jun-N-terminal kinase (JNK), extracellular signal-regulated kinase (ERK) and phosphatidyl inositol 3-kinase (PI3K)/Akt. The inhibition of the ERK and PI3K/Akt pathways also significantly increased cellular death. CONCLUSION ZD1839 activated caspase-3 and the inhibited Bcl-2 in human leukemic U937 cells through the downregulation of the ERK and PI3K/Akt pathways.
Collapse
Affiliation(s)
- Dong-oh Moon
- Faculty of Applied Marine Science, Cheju National University, Jeju Special Self-Governing Province, Republic of Korea
| | | | | | | | | | | |
Collapse
|
200
|
Schmitt E, Paquet C, Beauchemin M, Bertrand R. DNA-damage response network at the crossroads of cell-cycle checkpoints, cellular senescence and apoptosis. J Zhejiang Univ Sci B 2007; 8:377-97. [PMID: 17565509 PMCID: PMC1879163 DOI: 10.1631/jzus.2007.b0377] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Tissue homeostasis requires a carefully-orchestrated balance between cell proliferation, cellular senescence and cell death. Cells proliferate through a cell cycle that is tightly regulated by cyclin-dependent kinase activities. Cellular senescence is a safeguard program limiting the proliferative competence of cells in living organisms. Apoptosis eliminates unwanted cells by the coordinated activity of gene products that regulate and effect cell death. The intimate link between the cell cycle, cellular senescence, apoptosis regulation, cancer development and tumor responses to cancer treatment has become eminently apparent. Extensive research on tumor suppressor genes, oncogenes, the cell cycle and apoptosis regulatory genes has revealed how the DNA damage-sensing and -signaling pathways, referred to as the DNA-damage response network, are tied to cell proliferation, cell-cycle arrest, cellular senescence and apoptosis. DNA-damage responses are complex, involving "sensor" proteins that sense the damage, and transmit signals to "transducer" proteins, which, in turn, convey the signals to numerous "effector" proteins implicated in specific cellular pathways, including DNA repair mechanisms, cell-cycle checkpoints, cellular senescence and apoptosis. The Bcl-2 family of proteins stands among the most crucial regulators of apoptosis and performs vital functions in deciding whether a cell will live or die after cancer chemotherapy and irradiation. In addition, several studies have now revealed that members of the Bcl-2 family also interface with the cell cycle, DNA repair/recombination and cellular senescence, effects that are generally distinct from their function in apoptosis. In this review, we report progress in understanding the molecular networks that regulate cell-cycle checkpoints, cellular senescence and apoptosis after DNA damage, and discuss the influence of some Bcl-2 family members on cell-cycle checkpoint regulation.
Collapse
Affiliation(s)
- Estelle Schmitt
- Notre Dame Hospital and Montreal Cancer Institute, Research Centre of University of Montreal Hospital Centre (CRCHUM), Montreal (Que) H2L 4M1, Canada
| | - Claudie Paquet
- Notre Dame Hospital and Montreal Cancer Institute, Research Centre of University of Montreal Hospital Centre (CRCHUM), Montreal (Que) H2L 4M1, Canada
| | - Myriam Beauchemin
- Notre Dame Hospital and Montreal Cancer Institute, Research Centre of University of Montreal Hospital Centre (CRCHUM), Montreal (Que) H2L 4M1, Canada
| | - Richard Bertrand
- Notre Dame Hospital and Montreal Cancer Institute, Research Centre of University of Montreal Hospital Centre (CRCHUM), Montreal (Que) H2L 4M1, Canada
- Medicine Department, University of Montreal, Montreal (Que) H3C 3J7, Canada
- †E-mail:
| |
Collapse
|