1
|
Mitochondrial Dysfunction: The Hidden Player in the Pathogenesis of Atherosclerosis? Int J Mol Sci 2023; 24:ijms24021086. [PMID: 36674602 PMCID: PMC9861427 DOI: 10.3390/ijms24021086] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/29/2022] [Accepted: 01/04/2023] [Indexed: 01/09/2023] Open
Abstract
Atherosclerosis is a multifactorial inflammatory pathology that involves metabolic processes. Improvements in therapy have drastically reduced the prognosis of cardiovascular disease. Nevertheless, a significant residual risk is still relevant, and is related to unmet therapeutic targets. Endothelial dysfunction and lipid infiltration are the primary causes of atherosclerotic plaque progression. In this contest, mitochondrial dysfunction can affect arterial wall cells, in particular macrophages, smooth muscle cells, lymphocytes, and endothelial cells, causing an increase in reactive oxygen species (ROS), leading to oxidative stress, chronic inflammation, and intracellular lipid deposition. The detection and characterization of mitochondrial DNA (mtDNA) is crucial for assessing mitochondrial defects and should be considered the goal for new future therapeutic interventions. In this review, we will focus on a new idea, based on the analysis of data from many research groups, namely the link between mitochondrial impairment and endothelial dysfunction and, in particular, its effect on atherosclerosis and aging. Therefore, we discuss known and novel mitochondria-targeting therapies in the contest of atherosclerosis.
Collapse
|
2
|
Juncheng P, Joseph A, Lafarge A, Martins I, Obrist F, Pol J, Saavedra E, Li S, Sauvat A, Cerrato G, Lévesque S, Leduc M, Kepp O, Durand S, Aprahamian F, Nirmalathansan N, Michels J, Kroemer G, Castedo M. Cancer cell-autonomous overactivation of PARP1 compromises immunosurveillance in non-small cell lung cancer. J Immunother Cancer 2022; 10:jitc-2021-004280. [PMID: 35772809 PMCID: PMC9247697 DOI: 10.1136/jitc-2021-004280] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/25/2022] [Indexed: 12/26/2022] Open
Abstract
Background High activity of poly(ADP-ribose) polymerase-1 (PARP1) in non-small cell lung cancer (NSCLC) cells leads to an increase in immunohistochemically detectable PAR, correlating with poor prognosis in patients with NSCLC, as well as reduced tumor infiltration by cytotoxic T lymphocytes (CTLs). Intrigued by this observation, we decided to determine whether PARP1 activity in NSCLC cells may cause an alteration of anticancer immunosurveillance. Methods Continuous culture of mouse NSCLC cells in the presence of cisplatin led to the generation of cisplatin-resistant PARhigh clones. As compared with their parental controls, such PARhigh cells formed tumors that were less infiltrated by CTLs when they were injected into immunocompetent mice, suggesting a causative link between high PARP1 activity and compromised immunosurveillance. To confirm this cause-and-effect relationship, we used CRISPR/Cas9 technology to knock out PARP1 in two PARhigh NSCLC mouse cell lines (Lewis lung cancer [LLC] and tissue culture number one [TC1]), showing that the removal of PARP1 indeed restored cisplatin-induced cell death responses. Results PARP1 knockout (PARP1KO) cells became largely resistant to the PARP inhibitor niraparib, meaning that they exhibited less cell death induction, reduced DNA damage response, attenuated metabolic shifts and no induction of PD-L1 and MHC class-I molecules that may affect their immunogenicity. PARhigh tumors implanted in mice responded to niraparib irrespective of the presence or absence of T lymphocytes, suggesting that cancer cell-autonomous effects of niraparib dominate over its possible immunomodulatory action. While PARhigh NSCLC mouse cell lines proliferated similarly in immunocompetent and T cell-deficient mice, PARP1KO cells were strongly affected by the presence of T cells. PARP1KO LLC tumors grew more quickly in immunodeficient than in immunocompetent mice, and PARP1KO TC1 cells could only form tumors in T cell-deficient mice, not in immunocompetent controls. Importantly, as compared with PARhigh controls, the PARP1KO LLC tumors exhibited signs of T cell activation in the immune infiltrate such as higher inducible costimulator (ICOS) expression and lower PD-1 expression on CTLs. Conclusions These results prove at the genetic level that PARP1 activity within malignant cells modulates the tumor microenvironment.
Collapse
Affiliation(s)
- Pan Juncheng
- Equipe 11 labellisée par la Ligue contre le Cancer, Université de Paris Cité, Sorbonne Université, Centre de Recherche des Cordeliers, INSERM UMR1138, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.,Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicetre, France
| | - Adrien Joseph
- Equipe 11 labellisée par la Ligue contre le Cancer, Université de Paris Cité, Sorbonne Université, Centre de Recherche des Cordeliers, INSERM UMR1138, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.,Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicetre, France
| | - Antoine Lafarge
- Equipe 11 labellisée par la Ligue contre le Cancer, Université de Paris Cité, Sorbonne Université, Centre de Recherche des Cordeliers, INSERM UMR1138, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.,Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicetre, France
| | - Isabelle Martins
- Equipe 11 labellisée par la Ligue contre le Cancer, Université de Paris Cité, Sorbonne Université, Centre de Recherche des Cordeliers, INSERM UMR1138, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Florine Obrist
- Equipe 11 labellisée par la Ligue contre le Cancer, Université de Paris Cité, Sorbonne Université, Centre de Recherche des Cordeliers, INSERM UMR1138, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.,Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicetre, France
| | - Jonathan Pol
- Equipe 11 labellisée par la Ligue contre le Cancer, Université de Paris Cité, Sorbonne Université, Centre de Recherche des Cordeliers, INSERM UMR1138, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Ester Saavedra
- Equipe 11 labellisée par la Ligue contre le Cancer, Université de Paris Cité, Sorbonne Université, Centre de Recherche des Cordeliers, INSERM UMR1138, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Sijing Li
- Equipe 11 labellisée par la Ligue contre le Cancer, Université de Paris Cité, Sorbonne Université, Centre de Recherche des Cordeliers, INSERM UMR1138, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.,Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicetre, France
| | - Allan Sauvat
- Equipe 11 labellisée par la Ligue contre le Cancer, Université de Paris Cité, Sorbonne Université, Centre de Recherche des Cordeliers, INSERM UMR1138, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Giulia Cerrato
- Equipe 11 labellisée par la Ligue contre le Cancer, Université de Paris Cité, Sorbonne Université, Centre de Recherche des Cordeliers, INSERM UMR1138, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Sarah Lévesque
- Equipe 11 labellisée par la Ligue contre le Cancer, Université de Paris Cité, Sorbonne Université, Centre de Recherche des Cordeliers, INSERM UMR1138, Paris, France.,Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicetre, France
| | - Marion Leduc
- Equipe 11 labellisée par la Ligue contre le Cancer, Université de Paris Cité, Sorbonne Université, Centre de Recherche des Cordeliers, INSERM UMR1138, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Oliver Kepp
- Equipe 11 labellisée par la Ligue contre le Cancer, Université de Paris Cité, Sorbonne Université, Centre de Recherche des Cordeliers, INSERM UMR1138, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Sylvère Durand
- Equipe 11 labellisée par la Ligue contre le Cancer, Université de Paris Cité, Sorbonne Université, Centre de Recherche des Cordeliers, INSERM UMR1138, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Fanny Aprahamian
- Equipe 11 labellisée par la Ligue contre le Cancer, Université de Paris Cité, Sorbonne Université, Centre de Recherche des Cordeliers, INSERM UMR1138, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Nitharsshini Nirmalathansan
- Equipe 11 labellisée par la Ligue contre le Cancer, Université de Paris Cité, Sorbonne Université, Centre de Recherche des Cordeliers, INSERM UMR1138, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Judith Michels
- Département de Médecine Oncologique, Gustave Roussy Cancer Campus, F-94805, Villejuif, France
| | - Guido Kroemer
- Equipe 11 labellisée par la Ligue contre le Cancer, Université de Paris Cité, Sorbonne Université, Centre de Recherche des Cordeliers, INSERM UMR1138, Paris, France .,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.,Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, Assitance Publique-Hôpitaux de Paris, Paris, France
| | - Maria Castedo
- Equipe 11 labellisée par la Ligue contre le Cancer, Université de Paris Cité, Sorbonne Université, Centre de Recherche des Cordeliers, INSERM UMR1138, Paris, France .,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| |
Collapse
|
3
|
Álvarez-Abril B, Bloy N, Galassi C, Sato A, Jiménez-Cortegana C, Klapp V, Aretz A, Guilbaud E, Buqué A, Galluzzi L, Yamazaki T. Cytofluorometric assessment of acute cell death responses driven by radiation therapy. Methods Cell Biol 2022; 172:17-36. [DOI: 10.1016/bs.mcb.2022.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
4
|
The anti-carcinogenesis properties of erianin in the modulation of oxidative stress-mediated apoptosis and immune response in liver cancer. Aging (Albany NY) 2019; 11:10284-10300. [PMID: 31754081 PMCID: PMC6914393 DOI: 10.18632/aging.102456] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 11/07/2019] [Indexed: 12/25/2022]
Abstract
In this study, erianin was found to reduce the viability of cancer cells, inhibit their proliferation and migration, induce G2/M phase arrest, enhance cancer cell apoptosis, promote an increase in levels of intracellular reactive oxygen species and a decrease in mitochondrial membrane potential, and regulate the expression levels of anti- and pro-apoptosis-related proteins in HepG2 and SMMC-7721 cells. Erianin inhibited tumor growth in HepG2- and SMMC-7721-xenograft tumor nude mouse models, reduced the expression levels of anti-apoptosis proteins and enhanced the expression levels of pro-apoptosis proteins in tumor tissues. Erianin inhibited tumor growth in immunosuppressed BALB/c mice bearing heterotopic tumors. Among 111 types of cytokines detected in proteome profiling of tumor tissues, erianin substantially influenced levels of 38 types of cytokines in HepG2-xenografted tumors and of 15 types of cytokines in SMMC-7721-xenografted tumors, most of which are related to immune functions. Erianin strongly affected the serum levels of cytokines, and regulated the activation of nuclear factor-kappa B (NF-κB), and the expression levels of nuclear factor erythroid 2-related factor 2 (Nrf2) and its downstream proteins in spleen. The anti-liver cancer properties of erianin were found to be related mostly to its modulation of oxidative stress-mediated mitochondrial apoptosis and immune response.
Collapse
|
5
|
Bonora M, Wieckowski MR, Sinclair DA, Kroemer G, Pinton P, Galluzzi L. Targeting mitochondria for cardiovascular disorders: therapeutic potential and obstacles. Nat Rev Cardiol 2019; 16:33-55. [PMID: 30177752 DOI: 10.1038/s41569-018-0074-0] [Citation(s) in RCA: 209] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
A large body of evidence indicates that mitochondrial dysfunction has a major role in the pathogenesis of multiple cardiovascular disorders. Over the past 2 decades, extraordinary efforts have been focused on the development of agents that specifically target mitochondria for the treatment of cardiovascular disease. Despite such an intensive wave of investigation, no drugs specifically conceived to modulate mitochondrial functions are currently available for the clinical management of cardiovascular disease. In this Review, we discuss the therapeutic potential of targeting mitochondria in patients with cardiovascular disease, examine the obstacles that have restrained the development of mitochondria-targeting agents thus far, and identify strategies that might empower the full clinical potential of this approach.
Collapse
Affiliation(s)
- Massimo Bonora
- Ruth L. and David S. Gottesman Institute for Stem Cell, Regenerative Medicine Research, Department of Cell Biology and Stem Cell Institute, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Mariusz R Wieckowski
- Department of Biochemistry, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - David A Sinclair
- Department of Genetics, Paul F. Glenn Center for the Biology of Aging, Harvard Medical School, Boston, MA, USA.,Department of Pharmacology, School of Medical Sciences, The University of New South Wales, Sydney, New South Wales, Australia
| | - Guido Kroemer
- Equipe 11 labellisée Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,INSERM, U1138, Paris, France.,Université Paris Descartes/Paris V, Paris, France.,Université Pierre et Marie Curie, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Center, Villejuif, France.,Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.,Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
| | - Paolo Pinton
- Department of Morphology, Surgery, and Experimental Medicine, Section of Pathology, Oncology, and Experimental Biology, Laboratory for Technologies of Advanced Therapies, University of Ferrara, Ferrara, Italy. .,Maria Cecilia Hospital, GVM Care & Research, E.S. Health Science Foundation, Cotignola, Italy.
| | - Lorenzo Galluzzi
- Université Paris Descartes/Paris V, Paris, France. .,Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA. .,Sandra and Edward Meyer Cancer Center, New York, NY, USA.
| |
Collapse
|
6
|
Zhang Y, Sun D, Meng Q, Guo W, Chen Q, Zhang Y. Grifola frondosa polysaccharides induce breast cancer cell apoptosis via the mitochondrial-dependent apoptotic pathway. Int J Mol Med 2017; 40:1089-1095. [PMID: 28765878 PMCID: PMC5593468 DOI: 10.3892/ijmm.2017.3081] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 07/04/2017] [Indexed: 12/27/2022] Open
Abstract
Grifola frondosa, a type of food and medical fungus, has been shown to exhibit various pharmacological activities, including anticancer effects. As the most typical cancer diagnosed among female patients, breast cancer remains a huge concern threatening human health globally. In the present study, the anti-breast cancer effects of Grifola frondosa polysaccharides (GFPs) and the underlying mechanisms were investigated in MCF-7 and MDA-MB-231 cells, as well as in nude mice bearing MCF-7 tumor xenografts. GFPs exerted cytotoxic effects on the cells, as indicated by a decrease in cell viability, and an increase in the apoptototic rate, lactate dehydrogenase release and reactive oxygen species accumulation, inducing mitochondrial dysfunction. The increased expression of Bax, cleaved caspase-3 and caspase-8, and the reduced levels of B-cell lymphoma 2 (Bcl-2) and Bcl-extra large (Bcl-xL) were observed in the cells incubated with GFPs and in the tumor tissues of the mice treated with GFPs. Moreover, the GFPs significantly suppressed the phosphorylation of AKT/glycogen synthase kinase-3β and extracellular signal-regulated kinases in a time-dependent manner. Finally, the inhibition of MCF-7 tumor xenograft growth further confirmed the anti-breast cancer effects of GFPs. All these findings revealed that GFPs induced human breast cancer cell apoptosis via the mitochondrial-dependent apoptotic pathway, and provide experimental evidence to support the use of Grifola frondosa as a potential treatment for breast cancer.
Collapse
Affiliation(s)
- Yizhi Zhang
- Department of Neurology, The Second Hospital of Jilin University, Jilin University, Changchun, Jilin 130041, P.R. China
| | - Dejun Sun
- Department of Biomedicine, Institute for Regeneration Medicine, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Qingjin Meng
- Department of Neurology, Brain Hospital of Jilin Province, Siping, Jilin 136000, P.R. China
| | - Wanxu Guo
- Department of Neurology, The Second Hospital of Jilin University, Jilin University, Changchun, Jilin 130041, P.R. China
| | - Qiuhui Chen
- Department of Neurology, The Second Hospital of Jilin University, Jilin University, Changchun, Jilin 130041, P.R. China
| | - Ying Zhang
- Department of Neurology, The Second Hospital of Jilin University, Jilin University, Changchun, Jilin 130041, P.R. China
| |
Collapse
|
7
|
Song J, Wang Y, Teng M, Zhang S, Yin M, Lu J, Liu Y, Lee RJ, Wang D, Teng L. Cordyceps militaris induces tumor cell death via the caspase‑dependent mitochondrial pathway in HepG2 and MCF‑7 cells. Mol Med Rep 2016; 13:5132-40. [PMID: 27109250 PMCID: PMC4878560 DOI: 10.3892/mmr.2016.5175] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 03/30/2016] [Indexed: 01/07/2023] Open
Abstract
Cordyceps militaris (CM), an entomopathogenic fungus belonging to the class ascomycetes, possesses various pharmacological activities, including cytotoxic effects, on various types of human tumor cells. The present study investigated the anti-hepatocellular carcinoma (HCC) and anti-breast cancer effects of CM in in vitro and in vivo models. CM aqueous extract reduced cell viability, suppressed cell proliferation, inhibited cell migration ability, caused the over-release of lactate dehydrogenase, induced mitochondrial dysfunction and enhanced apoptotic rates in MCF-7 and HepG2 cells. The expression levels of cleaved poly (ADP ribose) polymerase and caspase-3, biomarkers of apoptosis, were increased following treatment with CM aqueous extract for 24 h. Furthermore, in the MCF-7 and HepG2 cells, enhanced levels of B cell-associated X protein and cleaved caspase-8 were observed in the CM-treated cells. Finally, the antitumor activities of CM in HCC and breast cancer were also confirmed in MCF-7- and HepG2-xengraft nude mice models. Collectively, the data obtained in the present study suggested that the cytotoxic effects of CM aqueous extract on HCC and breast cancer are associated with the caspase-dependent mitochondrial pathway.
Collapse
Affiliation(s)
- Jingjing Song
- School of Life Sciences, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Yingwu Wang
- School of Life Sciences, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Meiyu Teng
- School of Life Sciences, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Shiqiang Zhang
- School of Life Sciences, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Mengya Yin
- School of Life Sciences, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Jiahui Lu
- School of Life Sciences, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Yan Liu
- School of Life Sciences, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Robert J Lee
- School of Life Sciences, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Di Wang
- School of Life Sciences, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Lesheng Teng
- School of Life Sciences, Jilin University, Changchun, Jilin 130012, P.R. China
| |
Collapse
|
8
|
Harish G, Mahadevan A, Pruthi N, Sreenivasamurthy SK, Puttamallesh VN, Keshava Prasad TS, Shankar SK, Srinivas Bharath MM. Characterization of traumatic brain injury in human brains reveals distinct cellular and molecular changes in contusion and pericontusion. J Neurochem 2015; 134:156-72. [PMID: 25712633 DOI: 10.1111/jnc.13082] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2014] [Revised: 01/07/2015] [Accepted: 02/19/2015] [Indexed: 12/22/2022]
Abstract
Traumatic brain injury (TBI) contributes to fatalities and neurological disabilities worldwide. While primary injury causes immediate damage, secondary events contribute to long-term neurological defects. Contusions (Ct) are primary injuries correlated with poor clinical prognosis, and can expand leading to delayed neurological deterioration. Pericontusion (PC) (penumbra), the region surrounding Ct, can also expand with edema, increased intracranial pressure, ischemia, and poor clinical outcome. Analysis of Ct and PC can therefore assist in understanding the pathobiology of TBI and its management. This study on human TBI brains noted extensive neuronal, astroglial and inflammatory changes, alterations in mitochondrial, synaptic and oxidative markers, and associated proteomic profile, with distinct differences in Ct and PC. While Ct displayed petechial hemorrhages, thrombosis, inflammation, neuronal pyknosis, and astrogliosis, PC revealed edema, vacuolation of neuropil, axonal loss, and dystrophic changes. Proteomic analysis demonstrated altered immune response, synaptic, and mitochondrial dysfunction, among others, in Ct, while PC displayed altered regulation of neurogenesis and cytoskeletal architecture, among others. TBI brains displayed oxidative damage, glutathione depletion, mitochondrial dysfunction, and loss of synaptic proteins, with these changes being more profound in Ct. We suggest that analysis of markers specific to Ct and PC may be valuable in the evaluation of TBI pathobiology and therapeutics. We have characterized the primary injury in human traumatic brain injury (TBI). Contusions (Ct) - the injury core displayed hemorrhages, inflammation, and astrogliosis, while the surrounding pericontusion (PC) revealed edema, vacuolation, microglial activation, axonal loss, and dystrophy. Proteomic analysis demonstrated altered immune response, synaptic and mitochondrial dysfunction in Ct, and altered regulation of neurogenesis and cytoskeletal architecture in PC. Ct displayed more oxidative damage, mitochondrial, and synaptic dysfunction compared to PC.
Collapse
Affiliation(s)
- Gangadharappa Harish
- Department of Neurochemistry, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, Karnataka, India
| | - Anita Mahadevan
- Department of Neuropathology, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, Karnataka, India
| | - Nupur Pruthi
- Department of Neurosurgery, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, Karnataka, India
| | | | | | | | - Susarla Krishna Shankar
- Department of Neuropathology, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, Karnataka, India
| | | |
Collapse
|
9
|
Kepp O, Senovilla L, Vitale I, Vacchelli E, Adjemian S, Agostinis P, Apetoh L, Aranda F, Barnaba V, Bloy N, Bracci L, Breckpot K, Brough D, Buqué A, Castro MG, Cirone M, Colombo MI, Cremer I, Demaria S, Dini L, Eliopoulos AG, Faggioni A, Formenti SC, Fučíková J, Gabriele L, Gaipl US, Galon J, Garg A, Ghiringhelli F, Giese NA, Guo ZS, Hemminki A, Herrmann M, Hodge JW, Holdenrieder S, Honeychurch J, Hu HM, Huang X, Illidge TM, Kono K, Korbelik M, Krysko DV, Loi S, Lowenstein PR, Lugli E, Ma Y, Madeo F, Manfredi AA, Martins I, Mavilio D, Menger L, Merendino N, Michaud M, Mignot G, Mossman KL, Multhoff G, Oehler R, Palombo F, Panaretakis T, Pol J, Proietti E, Ricci JE, Riganti C, Rovere-Querini P, Rubartelli A, Sistigu A, Smyth MJ, Sonnemann J, Spisek R, Stagg J, Sukkurwala AQ, Tartour E, Thorburn A, Thorne SH, Vandenabeele P, Velotti F, Workenhe ST, Yang H, Zong WX, Zitvogel L, Kroemer G, Galluzzi L. Consensus guidelines for the detection of immunogenic cell death. Oncoimmunology 2014; 3:e955691. [PMID: 25941621 PMCID: PMC4292729 DOI: 10.4161/21624011.2014.955691] [Citation(s) in RCA: 643] [Impact Index Per Article: 58.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Accepted: 08/04/2014] [Indexed: 02/07/2023] Open
Abstract
Apoptotic cells have long been considered as intrinsically tolerogenic or unable to elicit immune responses specific for dead cell-associated antigens. However, multiple stimuli can trigger a functionally peculiar type of apoptotic demise that does not go unnoticed by the adaptive arm of the immune system, which we named "immunogenic cell death" (ICD). ICD is preceded or accompanied by the emission of a series of immunostimulatory damage-associated molecular patterns (DAMPs) in a precise spatiotemporal configuration. Several anticancer agents that have been successfully employed in the clinic for decades, including various chemotherapeutics and radiotherapy, can elicit ICD. Moreover, defects in the components that underlie the capacity of the immune system to perceive cell death as immunogenic negatively influence disease outcome among cancer patients treated with ICD inducers. Thus, ICD has profound clinical and therapeutic implications. Unfortunately, the gold-standard approach to detect ICD relies on vaccination experiments involving immunocompetent murine models and syngeneic cancer cells, an approach that is incompatible with large screening campaigns. Here, we outline strategies conceived to detect surrogate markers of ICD in vitro and to screen large chemical libraries for putative ICD inducers, based on a high-content, high-throughput platform that we recently developed. Such a platform allows for the detection of multiple DAMPs, like cell surface-exposed calreticulin, extracellular ATP and high mobility group box 1 (HMGB1), and/or the processes that underlie their emission, such as endoplasmic reticulum stress, autophagy and necrotic plasma membrane permeabilization. We surmise that this technology will facilitate the development of next-generation anticancer regimens, which kill malignant cells and simultaneously convert them into a cancer-specific therapeutic vaccine.
Collapse
Key Words
- APC, antigen-presenting cell
- ATF6, activating transcription factor 6
- ATP release
- BAK1, BCL2-antagonist/killer 1
- BAX, BCL2-associated X protein
- BCL2, B-cell CLL/lymphoma 2 protein
- CALR, calreticulin
- CTL, cytotoxic T lymphocyte
- DAMP, damage-associated molecular pattern
- DAPI, 4′,6-diamidino-2-phenylindole
- DiOC6(3), 3,3′-dihexyloxacarbocyanine iodide
- EIF2A, eukaryotic translation initiation factor 2A
- ER, endoplasmic reticulum
- FLT3LG, fms-related tyrosine kinase 3 ligand
- G3BP1, GTPase activating protein (SH3 domain) binding protein 1
- GFP, green fluorescent protein
- H2B, histone 2B
- HMGB1
- HMGB1, high mobility group box 1
- HSP, heat shock protein
- HSV-1, herpes simplex virus type I
- ICD, immunogenic cell death
- IFN, interferon
- IL, interleukin
- MOMP, mitochondrial outer membrane permeabilization
- PDIA3, protein disulfide isomerase family A
- PI, propidium iodide
- RFP, red fluorescent protein
- TLR, Toll-like receptor
- XBP1, X-box binding protein 1
- autophagy
- calreticulin
- endoplasmic reticulum stress
- immunotherapy
- member 3
- Δψm, mitochondrial transmembrane potential
Collapse
Affiliation(s)
- Oliver Kepp
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Center de Recherche des Cordeliers; Paris, France
- INSERM; U1138; Paris, France
- Metabolomics and Cell Biology Platforms; Gustave Roussy Cancer Campus; Villejuif, France
| | - Laura Senovilla
- INSERM; U1138; Paris, France
- Metabolomics and Cell Biology Platforms; Gustave Roussy Cancer Campus; Villejuif, France
- INSERM; U1015; Villejuif, France
| | - Ilio Vitale
- Regina Elena National Cancer Institute; Rome, Italy
| | - Erika Vacchelli
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Center de Recherche des Cordeliers; Paris, France
- INSERM; U1138; Paris, France
- Gustave Roussy Cancer Campus; Villejuif, France
| | - Sandy Adjemian
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Center de Recherche des Cordeliers; Paris, France
- INSERM; U1138; Paris, France
- Gustave Roussy Cancer Campus; Villejuif, France
- Molecular Cell Biology Laboratory; Department of Immunology; Institute of Biomedical Sciences; University of São Paulo; São Paulo, Brazil
| | - Patrizia Agostinis
- Cell Death Research and Therapy (CDRT) Laboratory; Department of Cellular and Molecular Medicine; University of Leuven; Leuven, Belgium
| | - Lionel Apetoh
- INSERM; UMR866; Dijon, France
- Centre Georges François Leclerc; Dijon, France
- Université de Bourgogne; Dijon, France
| | - Fernando Aranda
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Center de Recherche des Cordeliers; Paris, France
- INSERM; U1138; Paris, France
- Gustave Roussy Cancer Campus; Villejuif, France
| | - Vincenzo Barnaba
- Departement of Internal Medicine and Medical Sciences; University of Rome La Sapienza; Rome, Italy
- Istituto Pasteur; Fondazione Cenci Bolognetti; Rome, Italy
| | - Norma Bloy
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Center de Recherche des Cordeliers; Paris, France
- INSERM; U1138; Paris, France
- Gustave Roussy Cancer Campus; Villejuif, France
| | - Laura Bracci
- Department of Hematology; Oncology and Molecular Medicine; Istituto Superiore di Sanità (ISS); Rome, Italy
| | - Karine Breckpot
- Laboratory of Molecular and Cellular Therapy (LMCT); Department of Biomedical Sciences Medical School of the Free University of Brussels (VUB); Jette, Belgium
| | - David Brough
- Faculty of Life Sciences; University of Manchester; Manchester, UK
| | - Aitziber Buqué
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Center de Recherche des Cordeliers; Paris, France
- INSERM; U1138; Paris, France
- Gustave Roussy Cancer Campus; Villejuif, France
| | - Maria G. Castro
- Department of Neurosurgery and Cell and Developmental Biology; University of Michigan School of Medicine; Ann Arbor, MI USA
| | - Mara Cirone
- Department of Experimental Medicine; University of Rome La Sapienza; Rome, Italy
| | - Maria I. Colombo
- Laboratorio de Biología Celular y Molecular; Instituto de Histología y Embriología (IHEM); Facultad de Ciencias Médicas; Universidad Nacional de Cuyo; CONICET; Mendoza, Argentina
| | - Isabelle Cremer
- INSERM; U1138; Paris, France
- Université Pierre et Marie Curie/Paris VI; Paris, France
- Equipe 13; Center de Recherche des Cordeliers; Paris, France
| | - Sandra Demaria
- Department of Pathology; New York University School of Medicine; New York, NY USA
| | - Luciana Dini
- Department of Biological and Environmental Science and Technology (DiSTeBA); University of Salento; Lecce, Italy
| | - Aristides G. Eliopoulos
- Molecular and Cellular Biology Laboratory; Division of Basic Sciences; University of Crete Medical School; Heraklion, Greece
- Institute of Molecular Biology and Biotechnology; Foundation of Research and Technology - Hellas; Heraklion, Greece
| | - Alberto Faggioni
- Department of Experimental Medicine; University of Rome La Sapienza; Rome, Italy
| | - Silvia C. Formenti
- Department of Radiation Oncology; NewYork University School of Medicine and Langone Medical Center; New York, NY USA
| | - Jitka Fučíková
- Department of Immunology; 2 Faculty of Medicine and University Hospital Motol, Charles University; Prague, Czech Republic
- Sotio; Prague, Czech Republic
| | - Lucia Gabriele
- Department of Hematology; Oncology and Molecular Medicine; Istituto Superiore di Sanità (ISS); Rome, Italy
| | - Udo S. Gaipl
- Department of Radiation Oncology; University Hospital Erlangen; University of Erlangen-Nürnberg; Erlangen, Germany
| | - Jérôme Galon
- INSERM; U1138; Paris, France
- Université Pierre et Marie Curie/Paris VI; Paris, France
- Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France
- Laboratory of Integrative Cancer Immunology; Center de Recherche des Cordeliers; Paris, France
| | - Abhishek Garg
- Cell Death Research and Therapy (CDRT) Laboratory; Department of Cellular and Molecular Medicine; University of Leuven; Leuven, Belgium
| | - François Ghiringhelli
- INSERM; UMR866; Dijon, France
- Centre Georges François Leclerc; Dijon, France
- Université de Bourgogne; Dijon, France
| | - Nathalia A. Giese
- European Pancreas Center; Department of Surgery; University Hospital Heidelberg; Heidelberg, Germany
| | - Zong Sheng Guo
- Department of Surgery; University of Pittsburgh; Pittsburgh, PA USA
| | - Akseli Hemminki
- Cancer Gene Therapy Group; Transplantation laboratory; Haartman Institute; University of Helsinki; Helsinki, Finland
| | - Martin Herrmann
- Department of Internal Medicine 3; University of Erlangen-Nuremberg; Erlangen, Germany
| | - James W. Hodge
- Laboratory of Tumor Immunology and Biology; Center for Cancer Research; National Cancer Institute (NCI), National Institutes of Health (NIH); Bethesda, MD USA
| | - Stefan Holdenrieder
- Institute of Clinical Chemistry and Clinical Pharmacology; University Hospital Bonn; Bonn, Germany
| | - Jamie Honeychurch
- Faculty of Medical and Human Sciences, Institute of Cancer Studies; Manchester Academic Health Sciences Center; University of Manchester; Manchester, UK
| | - Hong-Min Hu
- Cancer Research and Biotherapy Center; Second Affiliated Hospital of Southeast University; Nanjing, China
- Laboratory of Cancer Immunobiology; Earle A. Chiles Research Institute; Providence Portland Medical Center; Portland, OR USA
| | - Xing Huang
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Center de Recherche des Cordeliers; Paris, France
- INSERM; U1138; Paris, France
- Metabolomics and Cell Biology Platforms; Gustave Roussy Cancer Campus; Villejuif, France
| | - Tim M. Illidge
- Faculty of Medical and Human Sciences, Institute of Cancer Studies; Manchester Academic Health Sciences Center; University of Manchester; Manchester, UK
| | - Koji Kono
- Department of Surgery; National University of Singapore; Singapore, Singapore
- Cancer Science Institute of Singapore; National University of Singapore; Singapore, Singapore
| | | | - Dmitri V. Krysko
- VIB Inflammation Research Center; Ghent, Belgium
- Department of Biomedical Molecular Biology; Ghent University; Ghent, Belgium
| | - Sherene Loi
- Division of Cancer Medicine and Division of Research; Peter MacCallum Cancer Center; East Melbourne; Victoria, Australia
| | - Pedro R. Lowenstein
- Department of Neurosurgery and Cell and Developmental Biology; University of Michigan School of Medicine; Ann Arbor, MI USA
| | - Enrico Lugli
- Unit of Clinical and Experimental Immunology; Humanitas Clinical and Research Center; Milan, Italy
- Department of Medical Biotechnologies and Translational Medicine, University of Milan; Rozzano, Italy
| | - Yuting Ma
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Center de Recherche des Cordeliers; Paris, France
- INSERM; U1138; Paris, France
- Gustave Roussy Cancer Campus; Villejuif, France
| | - Frank Madeo
- Institute of Molecular Biosciences; University of Graz; Graz, Austria
| | - Angelo A. Manfredi
- University Vita-Salute San Raffaele; Milano, Italy
- San Raffaele Scientific Institute; Milano, Italy
| | - Isabelle Martins
- Gustave Roussy Cancer Campus; Villejuif, France
- INSERM, U1030; Villejuif, France
- Faculté de Médecine; Université Paris-Sud/Paris XI; Kremlin-Bicêtre, France
| | - Domenico Mavilio
- Unit of Clinical and Experimental Immunology; Humanitas Clinical and Research Center; Milan, Italy
- Department of Medical Biotechnologies and Translational Medicine, University of Milan; Rozzano, Italy
| | - Laurie Menger
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Center de Recherche des Cordeliers; Paris, France
- INSERM; U1138; Paris, France
- Gustave Roussy Cancer Campus; Villejuif, France
- Cancer Immunology Unit, Research Department of Haematology; University College London (UCL) Cancer Institute; London, UK
| | - Nicolò Merendino
- Department of Ecological and Biological Sciences (DEB), Tuscia University; Viterbo, Italy
| | - Michael Michaud
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Center de Recherche des Cordeliers; Paris, France
- INSERM; U1138; Paris, France
- Gustave Roussy Cancer Campus; Villejuif, France
| | - Gregoire Mignot
- Cellular and Molecular Immunology and Endocrinology, Oniris; Nantes, France
| | - Karen L. Mossman
- Department of Pathology and Molecular Medicine; McMaster Immunology Research Center; Hamilton, Canada
- Institute for Infectious Disease Research; McMaster University; Hamilton, Canada
| | - Gabriele Multhoff
- Department of Radiation Oncology; Klinikum rechts der Isar; Technical University of Munich; Munich, Germany
| | - Rudolf Oehler
- Comprehensive Cancer Center; Medical University of Vienna; Vienna, Austria
| | - Fabio Palombo
- Departement of Internal Medicine and Medical Sciences; University of Rome La Sapienza; Rome, Italy
- Istituto Pasteur; Fondazione Cenci Bolognetti; Rome, Italy
| | | | - Jonathan Pol
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Center de Recherche des Cordeliers; Paris, France
- INSERM; U1138; Paris, France
- Gustave Roussy Cancer Campus; Villejuif, France
| | - Enrico Proietti
- Department of Hematology; Oncology and Molecular Medicine; Istituto Superiore di Sanità (ISS); Rome, Italy
| | - Jean-Ehrland Ricci
- INSERM; U1065; Nice, France
- Equipe “Contrôle Métabolique des Morts Cellulaires,” Center Méditerranéen de Médecine Moléculaire (C3M); Nice, France
- Faculté de Médecine; Université de Nice Sophia Antipolis; Nice, France
- Centre Hospitalier Universitaire de Nice; Nice, France
| | - Chiara Riganti
- Department of Oncology and Subalpine Center for Research and Experimental Medicine (CeRMS); University of Turin; Turin, Italy
| | - Patrizia Rovere-Querini
- University Vita-Salute San Raffaele; Milano, Italy
- San Raffaele Scientific Institute; Milano, Italy
| | - Anna Rubartelli
- Cell Biology Unit; Azienda Ospedaliera Universitaria San Martino; Istituto Nazionale per la Ricerca sul Cancro; Genova, Italy
| | | | - Mark J. Smyth
- Immunology in Cancer and Infection Laboratory; QIMR Berghofer Medical Research Institute; Herston, Australia
- School of Medicine, University of Queensland; Herston, Australia
| | - Juergen Sonnemann
- Department of Pediatric Haematology and Oncology; Jena University Hospital, Children's Clinic; Jena, Germany
| | - Radek Spisek
- Department of Immunology; 2 Faculty of Medicine and University Hospital Motol, Charles University; Prague, Czech Republic
- Sotio; Prague, Czech Republic
| | - John Stagg
- Centre de Recherche du Center Hospitalier de l’Université de Montréal; Faculté de Pharmacie, Université de Montréal; Montréal, Canada
| | - Abdul Qader Sukkurwala
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Center de Recherche des Cordeliers; Paris, France
- INSERM; U1138; Paris, France
- Gustave Roussy Cancer Campus; Villejuif, France
- Department of Pathology, Dow International Medical College; Dow University of Health Sciences; Karachi, Pakistan
| | - Eric Tartour
- INSERM; U970; Paris, France
- Pôle de Biologie; Hôpital Européen Georges Pompidou; AP-HP; Paris, France
| | - Andrew Thorburn
- Department of Pharmacology; University of Colorado School of Medicine; Aurora, CO USA
| | | | - Peter Vandenabeele
- VIB Inflammation Research Center; Ghent, Belgium
- Department of Biomedical Molecular Biology; Ghent University; Ghent, Belgium
- Methusalem Program; Ghent University; Ghent, Belgium
| | - Francesca Velotti
- Department of Ecological and Biological Sciences (DEB), Tuscia University; Viterbo, Italy
| | - Samuel T. Workenhe
- Department of Pathology and Molecular Medicine; McMaster Immunology Research Center; Hamilton, Canada
- Institute for Infectious Disease Research; McMaster University; Hamilton, Canada
| | - Haining Yang
- University of Hawaii Cancer Center; Honolulu, HI USA
| | - Wei-Xing Zong
- Department of Molecular Genetics and Microbiology; Stony Brook University; Stony Brook, NY USA
| | - Laurence Zitvogel
- INSERM; U1015; Villejuif, France
- Gustave Roussy Cancer Campus; Villejuif, France
- Centre d’Investigation Clinique Biothérapie 507 (CICBT507); Gustave Roussy Cancer Campus; Villejuif, France
| | - Guido Kroemer
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Center de Recherche des Cordeliers; Paris, France
- INSERM; U1138; Paris, France
- Metabolomics and Cell Biology Platforms; Gustave Roussy Cancer Campus; Villejuif, France
- Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France
- Pôle de Biologie; Hôpital Européen Georges Pompidou; AP-HP; Paris, France
| | - Lorenzo Galluzzi
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Center de Recherche des Cordeliers; Paris, France
- INSERM; U1138; Paris, France
- Gustave Roussy Cancer Campus; Villejuif, France
- Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France
| |
Collapse
|
10
|
Cheng G, Kong RH, Zhang LM, Zhang JN. Mitochondria in traumatic brain injury and mitochondrial-targeted multipotential therapeutic strategies. Br J Pharmacol 2013; 167:699-719. [PMID: 23003569 DOI: 10.1111/j.1476-5381.2012.02025.x] [Citation(s) in RCA: 256] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Traumatic brain injury (TBI) is a major health and socioeconomic problem throughout the world. It is a complicated pathological process that consists of primary insults and a secondary insult characterized by a set of biochemical cascades. The imbalance between a higher energy demand for repair of cell damage and decreased energy production led by mitochondrial dysfunction aggravates cell damage. At the cellular level, the main cause of the secondary deleterious cascades is cell damage that is centred in the mitochondria. Excitotoxicity, Ca(2+) overload, reactive oxygen species (ROS), Bcl-2 family, caspases and apoptosis inducing factor (AIF) are the main participants in mitochondria-centred cell damage following TBI. Some preclinical and clinical results of mitochondria-targeted therapy show promise. Mitochondria- targeted multipotential therapeutic strategies offer new hope for the successful treatment of TBI and other acute brain injuries.
Collapse
Affiliation(s)
- Gang Cheng
- Neurosurgical Department, PLA Navy General Hospital, Beijing, China
| | | | | | | |
Collapse
|
11
|
Galluzzi L, Vitale I, Senovilla L, Olaussen KA, Pinna G, Eisenberg T, Goubar A, Martins I, Michels J, Kratassiouk G, Carmona-Gutierrez D, Scoazec M, Vacchelli E, Schlemmer F, Kepp O, Shen S, Tailler M, Niso-Santano M, Morselli E, Criollo A, Adjemian S, Jemaà M, Chaba K, Pailleret C, Michaud M, Pietrocola F, Tajeddine N, de La Motte Rouge T, Araujo N, Morozova N, Robert T, Ripoche H, Commo F, Besse B, Validire P, Fouret P, Robin A, Dorvault N, Girard P, Gouy S, Pautier P, Jägemann N, Nickel AC, Marsili S, Paccard C, Servant N, Hupé P, Behrens C, Behnam-Motlagh P, Kohno K, Cremer I, Damotte D, Alifano M, Midttun O, Ueland PM, Lazar V, Dessen P, Zischka H, Chatelut E, Castedo M, Madeo F, Barillot E, Thomale J, Wistuba II, Sautès-Fridman C, Zitvogel L, Soria JC, Harel-Bellan A, Kroemer G. Prognostic impact of vitamin B6 metabolism in lung cancer. Cell Rep 2012; 2:257-69. [PMID: 22854025 DOI: 10.1016/j.celrep.2012.06.017] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2012] [Revised: 06/13/2012] [Accepted: 06/22/2012] [Indexed: 02/03/2023] Open
Abstract
Patients with non-small cell lung cancer (NSCLC) are routinely treated with cytotoxic agents such as cisplatin. Through a genome-wide siRNA-based screen, we identified vitamin B6 metabolism as a central regulator of cisplatin responses in vitro and in vivo. By aggravating a bioenergetic catastrophe that involves the depletion of intracellular glutathione, vitamin B6 exacerbates cisplatin-mediated DNA damage, thus sensitizing a large panel of cancer cell lines to apoptosis. Moreover, vitamin B6 sensitizes cancer cells to apoptosis induction by distinct types of physical and chemical stress, including multiple chemotherapeutics. This effect requires pyridoxal kinase (PDXK), the enzyme that generates the bioactive form of vitamin B6. In line with a general role of vitamin B6 in stress responses, low PDXK expression levels were found to be associated with poor disease outcome in two independent cohorts of patients with NSCLC. These results indicate that PDXK expression levels constitute a biomarker for risk stratification among patients with NSCLC.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Animals
- Antineoplastic Agents/administration & dosage
- Apoptosis/drug effects
- Apoptosis/genetics
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/metabolism
- Carcinoma, Non-Small-Cell Lung/mortality
- Carcinoma, Non-Small-Cell Lung/pathology
- Cisplatin/administration & dosage
- Cohort Studies
- Disease-Free Survival
- Female
- Gene Expression Regulation, Enzymologic/drug effects
- Gene Expression Regulation, Enzymologic/genetics
- Gene Expression Regulation, Neoplastic/drug effects
- Gene Expression Regulation, Neoplastic/genetics
- Genome-Wide Association Study
- Humans
- Lung Neoplasms/drug therapy
- Lung Neoplasms/genetics
- Lung Neoplasms/metabolism
- Lung Neoplasms/mortality
- Lung Neoplasms/pathology
- Male
- Mice
- Middle Aged
- Neoplasm Proteins/biosynthesis
- Neoplasm Proteins/genetics
- Pyridoxal Kinase/biosynthesis
- Pyridoxal Kinase/genetics
- Survival Rate
- Vitamin B 6/genetics
- Vitamin B 6/metabolism
Collapse
|
12
|
Huang T, Lin J, Cao J, Zhang P, Bai Y, Chen G, Chen K. An exopolysaccharide from Trichoderma pseudokoningii and its apoptotic activity on human leukemia K562 cells. Carbohydr Polym 2012; 89:701-8. [PMID: 24750776 DOI: 10.1016/j.carbpol.2012.03.079] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2011] [Revised: 03/22/2012] [Accepted: 03/24/2012] [Indexed: 11/30/2022]
Abstract
In this study, a novel exopolysaccharide (EPS) was isolated from the fermentation broth of Trichoderma pseudokoningii and its anticancer activities on human leukemia K562 cells were studied. EPS could significantly inhibited K562 cells proliferation in a time- and concentration-dependent manner. Meanwhile, characteristic of apoptosis, including apoptotic morphological features and the apoptosis rate were obtained. Sequentially, the dissipation of mitochondrial membrane potential, increase production of Reactive oxygen species (ROS), enhancement of the concentration of intracellular, up-regulation of Bax and p53 mRNA, down-regulation of Bcl-2 mRNA were also detected. The results indicate that the EPS could induce of K562 cells apoptosis, primarily in involved the mitochondrial pathways. The present studies suggest that EPS could be a new potential adjuvant chemotherapeutic and chemo preventive agent against human leukemia.
Collapse
Affiliation(s)
- Taotao Huang
- School of Life Sciences, Shandong University, Jinan 250100, PR China
| | - Jun Lin
- Department of Pharmacy, Wannan Medical college, Wuhu 241000, PR China
| | - Jianfeng Cao
- School of Life Sciences, Shandong University, Jinan 250100, PR China
| | - Pengying Zhang
- School of Life Sciences, Shandong University, Jinan 250100, PR China; National Glycoengineering Research Center, School of Life Science, Shandong University, Jinan 250100, PR China
| | - Yungui Bai
- School of Life Sciences, Shandong University, Jinan 250100, PR China
| | - Guochuang Chen
- School of Life Sciences, Shandong University, Jinan 250100, PR China
| | - Kaoshan Chen
- School of Life Sciences, Shandong University, Jinan 250100, PR China; Department of Pharmacy, Wannan Medical college, Wuhu 241000, PR China; National Glycoengineering Research Center, School of Life Science, Shandong University, Jinan 250100, PR China
| |
Collapse
|
13
|
Abstract
Mammalian target of rapamycin (mTOR) is an evolutionarily conserved kinase that integrates signals from nutrients and growth factors for the coordinate regulation of many cellular processes, including proliferation and cell death. Constitutive mTOR signaling characterizes multiple human malignancies, and pharmacological inhibitors of mTOR such as the immunosuppressant rapamycin and some of its nonimmunosuppressive derivatives not only have been ascribed with promising anticancer properties in vitro and in vivo but are also being extensively evaluated in clinical trials. mTOR inhibition rapidly leads to the activation of autophagy, which most often exerts prosurvival effects, although in some cases it accompanies cell death. Thus, depending on the specific experimental setting (cell type, concentration, stimulation time, and presence of concurrent stimuli), rapamycin can activate/favor a wide spectrum of cellular responses/phenotypes, ranging from adaptation to stress and survival to cell death. The (at least partial) overlap among the biochemical and morphological responses triggered by rapamycin considerably complicates the study of cell death-associated variables. Moreover, rapamycin presumably triggers acute cell death mainly via off-target mechanisms. Here, we describe a set of assays that can be employed for the routine quantification of rapamycin-induced cell death in vitro, as well as a set of guidelines that should be applied for their correct interpretation.
Collapse
|
14
|
Rello-Varona S, Kepp O, Vitale I, Michaud M, Senovilla L, Jemaà M, Joza N, Galluzzi L, Castedo M, Kroemer G. An automated fluorescence videomicroscopy assay for the detection of mitotic catastrophe. Cell Death Dis 2011; 1:e25. [PMID: 21364633 PMCID: PMC3032329 DOI: 10.1038/cddis.2010.6] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Mitotic catastrophe can be defined as a cell death mode that occurs during or shortly after a prolonged/aberrant mitosis, and can show apoptotic or necrotic features. However, conventional procedures for the detection of apoptosis or necrosis, including biochemical bulk assays and cytofluorometric techniques, cannot discriminate among pre-mitotic, mitotic and post-mitotic death, and hence are inappropriate to monitor mitotic catastrophe. To address this issue, we generated isogenic human colon carcinoma cell lines that differ in ploidy and p53 status, yet express similar amounts of fluorescent biosensors that allow for the visualization of chromatin (histone H2B coupled to green fluorescent protein (GFP)) and centrosomes (centrin coupled to the Discosoma striata red fluorescent protein (DsRed)). By combining high-resolution fluorescence videomicroscopy and automated image analysis, we established protocols and settings for the simultaneous assessment of ploidy, mitosis, centrosome number and cell death (which in our model system occurs mainly by apoptosis). Time-lapse videomicroscopy showed that this approach can be used for the high-throughput detection of mitotic catastrophe induced by three mechanistically distinct anti-mitotic agents (dimethylenastron (DIMEN), nocodazole (NDZ) and paclitaxel (PTX)), and – in this context – revealed an important role of p53 in the control of centrosome number.
Collapse
|
15
|
Caloric restriction and resveratrol promote longevity through the Sirtuin-1-dependent induction of autophagy. Cell Death Dis 2011; 1:e10. [PMID: 21364612 PMCID: PMC3032517 DOI: 10.1038/cddis.2009.8] [Citation(s) in RCA: 474] [Impact Index Per Article: 33.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Caloric restriction and autophagy-inducing pharmacological agents can prolong lifespan in model organisms including mice, flies, and nematodes. In this study, we show that transgenic expression of Sirtuin-1 induces autophagy in human cells in vitro and in Caenorhabditis elegans in vivo. The knockdown or knockout of Sirtuin-1 prevented the induction of autophagy by resveratrol and by nutrient deprivation in human cells as well as by dietary restriction in C. elegans. Conversely, Sirtuin-1 was not required for the induction of autophagy by rapamycin or p53 inhibition, neither in human cells nor in C. elegans. The knockdown or pharmacological inhibition of Sirtuin-1 enhanced the vulnerability of human cells to metabolic stress, unless they were stimulated to undergo autophagy by treatment with rapamycin or p53 inhibition. Along similar lines, resveratrol and dietary restriction only prolonged the lifespan of autophagy-proficient nematodes, whereas these beneficial effects on longevity were abolished by the knockdown of the essential autophagic modulator Beclin-1. We conclude that autophagy is universally required for the lifespan-prolonging effects of caloric restriction and pharmacological Sirtuin-1 activators.
Collapse
|
16
|
Abstract
Mitochondria are the cells' powerhouse, but also their suicidal weapon store. Dozens of lethal signal transduction pathways converge on mitochondria to cause the permeabilization of the mitochondrial outer membrane, leading to the cytosolic release of pro-apoptotic proteins and to the impairment of the bioenergetic functions of mitochondria. The mitochondrial metabolism of cancer cells is deregulated owing to the use of glycolytic intermediates, which are normally destined for oxidative phosphorylation, in anabolic reactions. Activation of the cell death machinery in cancer cells by inhibiting tumour-specific alterations of the mitochondrial metabolism or by stimulating mitochondrial membrane permeabilization could therefore be promising therapeutic approaches.
Collapse
Affiliation(s)
- Simone Fulda
- University Children's Hospital, Ulm University, Eythstrasse 24, D-89075 Ulm, Germany.
| | | | | |
Collapse
|
17
|
Galluzzi L, Morselli E, Vitale I, Kepp O, Senovilla L, Criollo A, Servant N, Paccard C, Hupé P, Robert T, Ripoche H, Lazar V, Harel-Bellan A, Dessen P, Barillot E, Kroemer G. miR-181a and miR-630 regulate cisplatin-induced cancer cell death. Cancer Res 2010; 70:1793-803. [PMID: 20145152 DOI: 10.1158/0008-5472.can-09-3112] [Citation(s) in RCA: 223] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
MicroRNAs (miRNA) are noncoding RNAs that regulate multiple cellular processes, including proliferation and apoptosis. We used microarray technology to identify miRNAs that were upregulated by non-small cell lung cancer (NSCLC) A549 cells in response to cisplatin (CDDP). The corresponding synthetic miRNA precursors (pre-miRNAs) per se were not lethal when transfected into A549 cells yet affected cell death induction by CDDP, C2-ceramide, cadmium, etoposide, and mitoxantrone in an inducer-specific fashion. Whereas synthetic miRNA inhibitors (anti-miRNAs) targeting miR-181a and miR-630 failed to modulate the response of A549 to CDDP, pre-miR-181a and pre-miR-630 enhanced and reduced CDDP-triggered cell death, respectively. Pre-miR-181a and pre-miR-630 consistently modulated mitochondrial/postmitochondrial steps of the intrinsic pathway of apoptosis, including Bax oligomerization, mitochondrial transmembrane potential dissipation, and the proteolytic maturation of caspase-9 and caspase-3. In addition, pre-miR-630 blocked early manifestations of the DNA damage response, including the phosphorylation of the ataxia-telangiectasia mutated (ATM) kinase and of two ATM substrates, histone H2AX and p53. Pharmacologic and genetic inhibition of p53 corroborated the hypothesis that pre-miR-630 (but not pre-miR-181a) blocks the upstream signaling pathways that are ignited by DNA damage and converge on p53 activation. Pre-miR-630 arrested A549 cells in the G0-G1 phase of the cell cycle, correlating with increased levels of the cell cycle inhibitor p27(Kip1) as well as with reduced proliferation rates and resulting in greatly diminished sensitivity of A549 cells to the late S-G2-M cell cycle arrest mediated by CDDP. Altogether, these results identify miR-181a and miR-630 as novel modulators of the CDDP response in NSCLC.
Collapse
Affiliation(s)
- Lorenzo Galluzzi
- Institut National de la Sante et de la Recherche Medicale, U848, Villejuif, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Galluzzi L, Aaronson SA, Abrams J, Alnemri ES, Andrews DW, Baehrecke EH, Bazan NG, Blagosklonny MV, Blomgren K, Borner C, Bredesen DE, Brenner C, Castedo M, Cidlowski JA, Ciechanover A, Cohen GM, De Laurenzi V, De Maria R, Deshmukh M, Dynlacht BD, El-Deiry WS, Flavell RA, Fulda S, Garrido C, Golstein P, Gougeon ML, Green DR, Gronemeyer H, Hajnόczky G, Hardwick JM, Hengartner MO, Ichijo H, Jäättelä M, Kepp O, Kimchi A, Klionsky DJ, Knight RA, Kornbluth S, Kumar S, Levine B, Lipton SA, Lugli E, Madeo F, Malorni W, Marine JCW, Martin SJ, Medema JP, Mehlen P, Melino G, Moll UM, Morselli E, Nagata S, Nicholson DW, Nicotera P, Nuñez G, Oren M, Penninger J, Pervaiz S, Peter ME, Piacentini M, Prehn JHM, Puthalakath H, Rabinovich GA, Rizzuto R, Rodrigues CMP, Rubinsztein DC, Rudel T, Scorrano L, Simon HU, Steller H, Tschopp J, Tsujimoto Y, Vandenabeele P, Vitale I, Vousden KH, Youle RJ, Yuan J, Zhivotovsky B, Kroemer G. Guidelines for the use and interpretation of assays for monitoring cell death in higher eukaryotes. Cell Death Differ 2009; 16:1093-107. [PMID: 19373242 PMCID: PMC2757140 DOI: 10.1038/cdd.2009.44] [Citation(s) in RCA: 519] [Impact Index Per Article: 32.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Cell death is essential for a plethora of physiological processes, and its deregulation characterizes numerous human diseases. Thus, the in-depth investigation of cell death and its mechanisms constitutes a formidable challenge for fundamental and applied biomedical research, and has tremendous implications for the development of novel therapeutic strategies. It is, therefore, of utmost importance to standardize the experimental procedures that identify dying and dead cells in cell cultures and/or in tissues, from model organisms and/or humans, in healthy and/or pathological scenarios. Thus far, dozens of methods have been proposed to quantify cell death-related parameters. However, no guidelines exist regarding their use and interpretation, and nobody has thoroughly annotated the experimental settings for which each of these techniques is most appropriate. Here, we provide a nonexhaustive comparison of methods to detect cell death with apoptotic or nonapoptotic morphologies, their advantages and pitfalls. These guidelines are intended for investigators who study cell death, as well as for reviewers who need to constructively critique scientific reports that deal with cellular demise. Given the difficulties in determining the exact number of cells that have passed the point-of-no-return of the signaling cascades leading to cell death, we emphasize the importance of performing multiple, methodologically unrelated assays to quantify dying and dead cells.
Collapse
Affiliation(s)
- L Galluzzi
- INSERM, U848, F-94805 Villejuif, France
- Institut Gustave Roussy, F-94805 Villejuif, France
- Université Paris Sud-XI, F-94805 Villejuif, France
| | - SA Aaronson
- Department of Oncological Sciences, Mount Sinai School of Medicine, New York, NY 10029, USA
| | - J Abrams
- Department of Cell Biology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - ES Alnemri
- Department of Biochemistry and Molecular Biology, Center for Apoptosis Research, Kimmel Cancer Institute, Thomas Jefferson University, Philadelphia, PA 19107-5587, USA
| | - DW Andrews
- Department of Biochemistry and Biomedical Sciences, McMaster University, L8N 3Z5 Hamilton, Canada
| | - EH Baehrecke
- Department of Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605-2324, USA
| | - NG Bazan
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | | | - K Blomgren
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, University of Gothenburg, SE-405 30 Gothenburg, Sweden
- Department of Pediatric Oncology, The Queen Silvia Children’s Hospital, SE-416 85 Gothenburg, Sweden
| | - C Borner
- Institute of Molecular Medicine and Cell Research (ZBMZ), Albert-Ludwigs-Universität Freiburg, 79104 Freiburg, Germany
| | - DE Bredesen
- Buck Institute for Age Research, Novato, CA 94945, USA
- University of California – San Francisco, San Francisco, CA 94143, USA
| | - C Brenner
- University of Versailles/St Quentin, 78035 Versailles, France
- CNRS, UMR8159, 78035 Versailles, France
| | - M Castedo
- INSERM, U848, F-94805 Villejuif, France
- Institut Gustave Roussy, F-94805 Villejuif, France
- Université Paris Sud-XI, F-94805 Villejuif, France
| | - JA Cidlowski
- National Institutes of Environmental Health Sciences, NIH, Duhram, NC 27709, USA
| | - A Ciechanover
- Vascular and Tumor Biology Research Center, The Rappaport Faculty of Medicine, Technion – Israel Institute of Technology, 31096 Haifa, Israel
| | - GM Cohen
- Medical Research Council, Toxicology Unit, Leicester University, Leicester LE1 9HN, UK
| | - V De Laurenzi
- Dipartimento di Scienze Biomediche, Università ‘G. d’Annunzio’ Chieti-Pescara, 66100 Chieti, Italy
| | - R De Maria
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy
- Mediterranean Institute of Oncology, 95030 Catania, Italy
| | - M Deshmukh
- Neuroscience Center, Department of Cell and Developmental Biology, University of North Carolina, Chapel Hill, NC 27599-7250, USA
| | - BD Dynlacht
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
| | - WS El-Deiry
- Hematology-Oncology Division, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - RA Flavell
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815-6789, USA
| | - S Fulda
- University Children’s Hospital, 89075 Ulm, Germany
| | - C Garrido
- INSERM, UMR866, 21049 Dijon, France
- Faculty of Medicine and Pharmacy, University of Burgundy, 21049 Dijon, France
| | - P Golstein
- INSERM, U631, 13288 Marseille, France
- CNRS, UMR6102, 13288 Marseille, France
- Centre d’Immunologie de Marseille-Luminy, Aix Marseille Université, 13288 Marseille, France
| | - M-L Gougeon
- Institut Pasteur, Antiviral Immunity, Biotherapy and Vaccine Unit, 75015 Paris, France
| | - DR Green
- Department of Immunology, St. Jude Children′s Research Hospital, Memphis, TN 38105, USA
| | - H Gronemeyer
- Department of Cancer Biology – Institut de Génétique et de Biologie Moléculaire et Cellulaire, 67404 Illkirch, France
- CNRS, UMR7104, 67404 Illkirch, France
- INSERM, U964, 67404 Illkirch, France
| | - G Hajnόczky
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - JM Hardwick
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University, Baltimore, MD 21205, USA
| | - MO Hengartner
- Institute of Molecular Biology, University of Zurich, 8057 Zurich, Switzerland
| | - H Ichijo
- Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo 113-0033, Japan
| | - M Jäättelä
- Danish Cancer Society, Department of Apoptosis, Institute of Cancer Biology, DK-2100 Copenhagen, Denmark
| | - O Kepp
- INSERM, U848, F-94805 Villejuif, France
- Institut Gustave Roussy, F-94805 Villejuif, France
- Université Paris Sud-XI, F-94805 Villejuif, France
| | - A Kimchi
- Department of Molecular Genetics, Weizmann Institute of Science, 76100 Rehovot, Israel
| | - DJ Klionsky
- Life Sciences Institute and Department of Molecular, Cellular, and Developmental Biology and Biological Chemistry, University of Michigan, Ann Arbor, MI 48109, USA
| | - RA Knight
- Institute of Child Health, University College London, London WC1N 1EH, UK
| | - S Kornbluth
- Duke University School of Medicine, Durham, NC 27710, USA
| | - S Kumar
- Centre for Cancer Biology, Hanson Institute, Adelaide, South Australia 5000, Australia
| | - B Levine
- Howard Hughes Medical Institute, Chevy Chase, MD 20815-6789, USA
- Southwestern Medical Center, University of Texas, Dallas, TX 75390, USA
| | - SA Lipton
- Burnham Institute for Medical Research, La Jolla, CA 92037, USA
- The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
- The Scripps Research Institute, La Jolla, CA 92037, USA
- Univerisity of California-San Diego, La Jolla, CA 92093, USA
| | - E Lugli
- Immunotechnology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
| | - F Madeo
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| | - W Malorni
- Department of Therapeutic Research and Medicines Evaluation, Section of Cell Aging and Degeneration, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - J-CW Marine
- Laboratory for Molecular Cancer Biology, VIB, 9052 Ghent, Belgium
- Department for Molecular Biology, Ghent University, 9052 Ghent, Belgium
| | - SJ Martin
- Department of Genetics, Trinity College, Dublin 2, Ireland
| | - JP Medema
- Center for Experimental and Molecular Medicine, Academic Medical Center, 1105 AZ Amsterdam, The Netherlands
- University of Amsterdam, 1012 ZA Amsterdam, The Netherlands
| | - P Mehlen
- Apoptosis, Cancer, and Development Laboratory, Centre Léon Berard, 69008 Lyon, France
- CNRS, UMR5238, 69008 Lyon, France
- Université de Lyon, 69008 Lyon, France
| | - G Melino
- Medical Research Council, Toxicology Unit, Leicester University, Leicester LE1 9HN, UK
- Department of Experimental Medicine and Biochemical Sciences, University of Rome ‘Tor Vergata’, 00133 Rome, Italy
| | - UM Moll
- Department of Pathology, Stony Brook University, Stony Brook, NY 11794-8691, USA
- Department of Molecular Oncology, Goöttingen Center of Molecular Biosciences, 37077 Göttingen, Germany
- Faculty of Medicine, University of Göttingen, 37077 Göttingen, Germany
| | - E Morselli
- INSERM, U848, F-94805 Villejuif, France
- Institut Gustave Roussy, F-94805 Villejuif, France
- Université Paris Sud-XI, F-94805 Villejuif, France
| | - S Nagata
- Department of Medical Chemistry, Graduate School of Medicine, University of Kyoto, Kyoto 606-8501, Japan
| | - DW Nicholson
- Merck Research Laboratories, Rahway, NJ 07065-0900, USA
| | - P Nicotera
- Medical Research Council, Toxicology Unit, Leicester University, Leicester LE1 9HN, UK
| | - G Nuñez
- University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - M Oren
- Department of Molecular Cell Biology, Weizmann Institute of Science, 76100 Rehovot, Israel
| | - J Penninger
- Institute of Molecular Biotechnology of the Austrian Academy of Science, 1030 Vienna, Austria
| | - S Pervaiz
- Department of Physiology, Yong Loo Lin School of Medicine, Graduate School for Integrative Sciences and Engineering, National University of Singapore, 117597 Singapore
- Singapore-MIT Alliance, National University of Singapore, 117576 Singapore
- Duke-NUS Graduate Medical School, 169547 Singapore
| | - ME Peter
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL 60637, USA
| | - M Piacentini
- Laboratory of Cell Biology, National Institute for Infectious Diseases IRCCS ‘L. Spallanzani’, 00149 Rome, Italy
- Department of Biology, University of Rome ‘Tor Vergata’, 00133 Rome, Italy
| | - JHM Prehn
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - H Puthalakath
- Department of Biochemistry, La Trobe University, 3086 Victoria, Australia
| | - GA Rabinovich
- Laboratorio de Inmunopatología, Instituto de Biología y Medicina Experimental (IBYME- CONICET), C1428 Buenos Aires, Argentina
| | - R Rizzuto
- Department of Biomedical Sciences, University of Padova, 35121 Padova, Italy
| | - CMP Rodrigues
- iMed.UL, Faculty of Pharmacy, University of Lisbon, 1649-003 Lisbon, Portugal
| | - DC Rubinsztein
- Cambridge Institute for Medical Research, Cambridge CB2 0XY, UK
| | - T Rudel
- Biocenter, University of Würzburg, 97074 Würzburg, Germany
| | - L Scorrano
- Department of Cell Physiology and Metabolism, University of Geneva Medical School, 1211 Geneva, Switzerland
- Dulbecco-Telethon Institute, Venetian Institute of Molecular Medicine, 35129 Padova, Italy
| | - H-U Simon
- Department of Pharmacology, University of Bern, 3010 Bern, Switzerland
| | - H Steller
- Howard Hughes Medical Institute, Chevy Chase, MD 20815-6789, USA
- Laboratory of Apoptosis and Cancer Biology, The Rockefeller University, New York, NY 10065, USA
| | - J Tschopp
- Department of Biochemistry, University of Lausanne, 1066 Epalinges, Switzerland
| | - Y Tsujimoto
- Department of Medical Genetics, Osaka University Medical School, Osaka 565-0871, Japan
| | - P Vandenabeele
- Department for Molecular Biology, Ghent University, 9052 Ghent, Belgium
- Department for Molecular Biomedical Research, VIB, 9052 Ghent, Belgium
| | - I Vitale
- INSERM, U848, F-94805 Villejuif, France
- Institut Gustave Roussy, F-94805 Villejuif, France
- Université Paris Sud-XI, F-94805 Villejuif, France
| | - KH Vousden
- The Beatson Institute for Cancer Research, Glasgow G61 1BD, UK
| | - RJ Youle
- Biochemistry Section, Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD 20892, USA
| | - J Yuan
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - B Zhivotovsky
- Institute of Environmental Medicine, Division of Toxicology, Karolinska Institute, SE- 171 77 Stockholm, Sweden
| | - G Kroemer
- INSERM, U848, F-94805 Villejuif, France
- Institut Gustave Roussy, F-94805 Villejuif, France
- Université Paris Sud-XI, F-94805 Villejuif, France
| |
Collapse
|
19
|
Hisatomi T, Ishibashi T, Miller JW, Kroemer G. Pharmacological inhibition of mitochondrial membrane permeabilization for neuroprotection. Exp Neurol 2009; 218:347-52. [PMID: 19303007 DOI: 10.1016/j.expneurol.2009.03.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2008] [Revised: 02/26/2009] [Accepted: 03/03/2009] [Indexed: 01/09/2023]
Abstract
Recent data have provided important clues about the molecular mechanisms underlying certain neurodegenerative diseases. Most cell death in vertebrates proceeds via the mitochondrial pathway of apoptosis. Mitochondria contain proapoptotic factors such as cytochrome c and AIF in their intermembrane space. Furthermore, mitochondrial membrane permeabilization (MMP) is a critical event during apoptosis, representing the "point of no return" of the lethal process. Modern medicine is developing an increasing number of drugs for neurodegenerative disease, but no neuroprotective treatment has yet been established. While current treatments temporarily alleviate symptoms, they do not halt disease progression. This paper briefly reviews the pharmacological inhibition of mitochondrial membrane permeabilization for neuroprotection.
Collapse
Affiliation(s)
- Toshio Hisatomi
- Department of Ophthalmology, Kyushu University, Fukuoka, Japan.
| | | | | | | |
Collapse
|