551
|
Mazzolini G, Sowa JP, Canbay A. Cell death mechanisms in human chronic liver diseases: a far cry from clinical applicability. Clin Sci (Lond) 2016; 130:2121-2138. [DOI: 10.1042/cs20160035] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
The liver is constantly exposed to a host of injurious stimuli. This results in hepatocellular death mainly by apoptosis and necrosis, but also due to autophagy, necroptosis, pyroptosis and in some cases by an intricately balanced combination thereof. Overwhelming and continuous cell death in the liver leads to inflammation, fibrosis, cirrhosis, and eventually hepatocellular carcinoma. Although data from various disease models may suggest a specific (predominant) cell death mode for different aetiologies, the clinical reality is not as clear cut. Reliable and non-invasive cell death markers are not available in general practice and assessment of cell death mode to absolute certainty from liver biopsies does not seem feasible, yet. Various aetiologies probably induce different predominant cell death modes within the liver, although the death modes involved may change during disease progression. Moreover, current methods applicable in patients are limited to surrogate markers for apoptosis (M30), and possibly for pyroptosis (IL-1 family) and necro(pto)sis (HMGB1). Although markers for some death modes are not available at all (autophagy), others may not be specific for a cell death mode or might not always definitely indicate dying cells. Physicians need to take care in asserting the presence of cell death. Still the serum-derived markers are valuable tools to assess severity of chronic liver diseases. This review gives a short overview of known hepatocellular cell death modes in various aetiologies of chronic liver disease. Also the limitations of current knowledge in human settings and utilization of surrogate markers for disease assessment are summarized.
Collapse
Affiliation(s)
- Guillermo Mazzolini
- Department for Gastroenterology and Hepatology, University Hospital, University Duisburg-Essen, 45147 Essen, Germany
- Gene Therapy Laboratory, Instituto de Investigaciones Medicas Aplicadas, Universidad Austral-CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas), Pilar Centro, Buenos Aires, Argentina
| | - Jan-Peter Sowa
- Department for Gastroenterology and Hepatology, University Hospital, University Duisburg-Essen, 45147 Essen, Germany
| | - Ali Canbay
- Department for Gastroenterology and Hepatology, University Hospital, University Duisburg-Essen, 45147 Essen, Germany
| |
Collapse
|
552
|
LaClair KD, Donde A, Ling JP, Jeong YH, Chhabra R, Martin LJ, Wong PC. Depletion of TDP-43 decreases fibril and plaque β-amyloid and exacerbates neurodegeneration in an Alzheimer's mouse model. Acta Neuropathol 2016; 132:859-873. [PMID: 27785573 PMCID: PMC5131701 DOI: 10.1007/s00401-016-1637-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 10/20/2016] [Accepted: 10/20/2016] [Indexed: 10/20/2022]
Abstract
TDP-43 proteinopathy, initially associated with ALS and FTD, is also found in 30-60% of Alzheimer's disease (AD) cases and correlates with worsened cognition and neurodegeneration. A major component of this proteinopathy is depletion of this RNA-binding protein from the nucleus, which compromises repression of non-conserved cryptic exons in neurodegenerative diseases. To test whether nuclear depletion of TDP-43 may contribute to the pathogenesis of AD cases with TDP-43 proteinopathy, we examined the impact of depletion of TDP-43 in populations of neurons vulnerable in AD, and on neurodegeneration in an AD-linked context. Here, we show that some populations of pyramidal neurons that are selectively vulnerable in AD are also vulnerable to TDP-43 depletion in mice, while other forebrain neurons appear spared. Moreover, TDP-43 depletion in forebrain neurons of an AD mouse model exacerbates neurodegeneration, and correlates with increased prefibrillar oligomeric Aβ and decreased Aβ plaque burden. These findings support a role for nuclear depletion of TDP-43 in the pathogenesis of AD and provide strong rationale for developing novel therapeutics to alleviate the depletion of TDP-43 and functional antemortem biomarkers associated with its nuclear loss.
Collapse
Affiliation(s)
- Katherine D LaClair
- Department of Pathology, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross 558, Baltimore, MD, 21205, USA
- Cellular and Molecular Medicine Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Aneesh Donde
- Department of Pathology, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross 558, Baltimore, MD, 21205, USA
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Jonathan P Ling
- Department of Pathology, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross 558, Baltimore, MD, 21205, USA
| | - Yun Ha Jeong
- Department of Pathology, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross 558, Baltimore, MD, 21205, USA
- Neural Development and Disease Department, Korea Brain Research Institute, Daegu, 701-300, Korea
| | - Resham Chhabra
- Department of Pathology, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross 558, Baltimore, MD, 21205, USA
| | - Lee J Martin
- Department of Pathology, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross 558, Baltimore, MD, 21205, USA
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Philip C Wong
- Department of Pathology, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross 558, Baltimore, MD, 21205, USA.
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
- Cellular and Molecular Medicine Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
553
|
Plasmodium falciparum exhibits markers of regulated cell death at high population density in vitro. Parasitol Int 2016; 65:715-727. [DOI: 10.1016/j.parint.2016.07.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 07/13/2016] [Accepted: 07/14/2016] [Indexed: 11/22/2022]
|
554
|
Wang H, Cai J. The role of microRNAs in heart failure. Biochim Biophys Acta Mol Basis Dis 2016; 1863:2019-2030. [PMID: 27916680 DOI: 10.1016/j.bbadis.2016.11.034] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 11/26/2016] [Accepted: 11/29/2016] [Indexed: 12/12/2022]
Abstract
MicroRNAs are small non-coding RNA molecules that regulate gene expression by inhibiting mRNA translation and/or inducing mRNA degradation. In the past decade, many in vitro and in vivo studies have explored the involvement of microRNAs in various cardiovascular diseases. In this paper, studies focused upon the target genes and functionality of miRNAs in the pathophysiological processes of heart failure are reviewed. The selected miRNAs are categorized according to the biological relevance of their target genes in relation to four cardiovascular pathologies, namely angiogenesis, cardiac hypertrophy, fibrosis and apoptosis. This review illustrates the involvement of miRNAs in different biological signaling pathways and provides an overview of current understanding of the roles of miRNAs in cardiovascular health and diseases. This article is part of a Special Issue entitled: Genetic and epigenetic control of heart failure - edited by Jun Ren & Megan Yingmei Zhang.
Collapse
Affiliation(s)
- Hongjiang Wang
- Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China.
| | - Jun Cai
- State Key Laboratory of Cardiovascular Disease of China, National Center for Cardiovascular Diseases of China, Chinese Academy of Medical Sciences and Peking Union Medical College, Hypertension Center, Fuwai Hospital, Xicheng District, North Lishi Road No. 167, Beijing 100037, China.
| |
Collapse
|
555
|
Assessment of Glycolytic Flux and Mitochondrial Respiration in the Course of Autophagic Responses. Methods Enzymol 2016; 588:155-170. [PMID: 28237099 DOI: 10.1016/bs.mie.2016.09.079] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Autophagy is an evolutionarily conserved process that mediates prominent homeostatic functions, both at the cellular and organismal level. Indeed, baseline autophagy not only ensures the disposal of cytoplasmic entities that may become cytotoxic upon accumulation, but also contributes to the maintenance of metabolic fitness in physiological conditions. Likewise, autophagy plays a fundamental role in the cellular and organismal adaptation to homeostatic perturbations of metabolic, physical, or chemical nature. Thus, the molecular machinery for autophagy is functionally regulated by a broad panel of sensors that detect indicators of metabolic homeostasis. Moreover, increases in autophagic flux have a direct impact on core metabolic circuitries including (but not limited to) glycolysis and mitochondrial respiration. Here, we detail a simple methodological approach to monitor these two processes in cultured cancer cells that mount a proficient autophagic response to stress.
Collapse
|
556
|
Song X, Xie Y, Kang R, Hou W, Sun X, Epperly MW, Greenberger JS, Tang D. FANCD2 protects against bone marrow injury from ferroptosis. Biochem Biophys Res Commun 2016; 480:443-449. [PMID: 27773819 PMCID: PMC6591579 DOI: 10.1016/j.bbrc.2016.10.068] [Citation(s) in RCA: 156] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 10/04/2016] [Accepted: 10/19/2016] [Indexed: 02/07/2023]
Abstract
Bone marrow injury remains a serious concern in traditional cancer treatment. Ferroptosis is an iron- and oxidative-dependent form of regulated cell death that has become part of an emerging strategy for chemotherapy. However, the key regulator of ferroptosis in bone marrow injury remains unknown. Here, we show that Fanconi anemia complementation group D2 (FANCD2), a nuclear protein involved in DNA damage repair, protects against ferroptosis-mediated injury in bone marrow stromal cells (BMSCs). The classical ferroptosis inducer erastin remarkably increased the levels of monoubiquitinated FANCD2, which in turn limited DNA damage in BMSCs. FANCD2-deficient BMSCs were more sensitive to erastin-induced ferroptosis (but not autophagy) than FANCD2 wild-type cells. Knockout of FANCD2 increased ferroptosis-associated biochemical events (e.g., ferrous iron accumulation, glutathione depletion, and malondialdehyde production). Mechanically, FANCD2 regulated genes and/or expression of proteins involved in iron metabolism (e.g., FTH1, TF, TFRC, HAMP, HSPB1, SLC40A1, and STEAP3) and lipid peroxidation (e.g., GPX4). Collectively, these findings indicate that FANCD2 plays a novel role in the negative regulation of ferroptosis. FANCD2 could represent an amenable target for the development of novel anticancer therapies aiming to reduce the side effects of ferroptosis inducers.
Collapse
Affiliation(s)
- Xinxin Song
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Yangchun Xie
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Rui Kang
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Wen Hou
- Department of Radiation Oncology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Xiaofang Sun
- The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510510, China
| | - Michael W Epperly
- Department of Radiation Oncology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Joel S Greenberger
- Department of Radiation Oncology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Daolin Tang
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA; The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510510, China.
| |
Collapse
|
557
|
Mervin LH, Cao Q, Barrett IP, Firth MA, Murray D, McWilliams L, Haddrick M, Wigglesworth M, Engkvist O, Bender A. Understanding Cytotoxicity and Cytostaticity in a High-Throughput Screening Collection. ACS Chem Biol 2016; 11:3007-3023. [PMID: 27571164 DOI: 10.1021/acschembio.6b00538] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
While mechanisms of cytotoxicity and cytostaticity have been studied extensively from the biological side, relatively little is currently understood regarding areas of chemical space leading to cytotoxicity and cytostasis in large compound collections. Predicting and rationalizing potential adverse mechanism-of-actions (MoAs) of small molecules is however crucial for screening library design, given the link of even low level cytotoxicity and adverse events observed in man. In this study, we analyzed results from a cell-based cytotoxicity screening cascade, comprising 296 970 nontoxic, 5784 cytotoxic and cytostatic, and 2327 cytostatic-only compounds evaluated on the THP-1 cell-line. We employed an in silico MoA analysis protocol, utilizing 9.5 million active and 602 million inactive bioactivity points to generate target predictions, annotate predicted targets with pathways, and calculate enrichment metrics to highlight targets and pathways. Predictions identify known mechanisms for the top ranking targets and pathways for both phenotypes after review and indicate that while processes involved in cytotoxicity versus cytostaticity seem to overlap, differences between both phenotypes seem to exist to some extent. Cytotoxic predictions highlight many kinases, including the potentially novel cytotoxicity-related target STK32C, while cytostatic predictions outline targets linked with response to DNA damage, metabolism, and cytoskeletal machinery. Fragment analysis was also employed to generate a library of toxicophores to improve general understanding of the chemical features driving toxicity. We highlight substructures with potential kinase-dependent and kinase-independent mechanisms of toxicity. We also trained a cytotoxic classification model on proprietary and public compound readouts, and prospectively validated these on 988 novel compounds comprising difficult and trivial testing instances, to establish the applicability domain of models. The proprietary model performed with precision and recall scores of 77.9% and 83.8%, respectively. The MoA results and top ranking substructures with accompanying MoA predictions are available as a platform to assess screening collections.
Collapse
Affiliation(s)
- Lewis H. Mervin
- Centre
for Molecular Informatics, Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
| | - Qing Cao
- Discovery Sciences, AstraZeneca R&D, Waltham, United States
| | - Ian P. Barrett
- Discovery Sciences, AstraZeneca R&D, Cambridge Science Park, Cambridge, United Kingdom
| | - Mike A. Firth
- Discovery Sciences, AstraZeneca R&D, Cambridge Science Park, Cambridge, United Kingdom
| | - David Murray
- Discovery Sciences, AstraZeneca R&D, Alderley Park, Macclesfield, United Kingdom
| | - Lisa McWilliams
- Discovery Sciences, AstraZeneca R&D, Alderley Park, Macclesfield, United Kingdom
| | - Malcolm Haddrick
- Discovery Sciences, AstraZeneca R&D, Alderley Park, Macclesfield, United Kingdom
| | - Mark Wigglesworth
- Discovery Sciences, AstraZeneca R&D, Alderley Park, Macclesfield, United Kingdom
| | - Ola Engkvist
- Discovery Sciences, AstraZeneca R&D, Mölndal, Sweden
| | - Andreas Bender
- Centre
for Molecular Informatics, Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
558
|
Activating autophagy to potentiate immunogenic chemotherapy and radiation therapy. Nat Rev Clin Oncol 2016; 14:247-258. [PMID: 27845767 DOI: 10.1038/nrclinonc.2016.183] [Citation(s) in RCA: 250] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Autophagy is fundamental to the maintenance of intracellular homeostasis in virtually all human cells. Accordingly, defective autophagy predisposes healthy cells to undergoing malignant transformation. By contrast, malignant cells are able to harness autophagy to thrive, despite adverse microenvironmental conditions, and to resist therapeutic challenges. Thus, inhibition of autophagy has been proposed as a strategy to kill cancer cells or sensitize them to therapy; however, autophagy is also critical for optimal immune function, and mediates cell-extrinsic homeostatic effects owing to its central role in danger signalling by neoplastic cells responding to immunogenic chemotherapy and/or radiation therapy. In this Perspective, we discuss accumulating preclinical and clinical evidence in support of the all-too-often dismissed possibility that activating autophagy might be a relevant clinical objective that enables an increase in the effectiveness of immunogenic chemotherapy and/or radiation therapy.
Collapse
|
559
|
Gordeev SA, Bykova TV, Zubova SG, Bystrova OA, Martynova MG, Pospelov VA, Pospelova TV. mTOR kinase inhibitor pp242 causes mitophagy terminated by apoptotic cell death in E1A-Ras transformed cells. Oncotarget 2016; 6:44905-26. [PMID: 26636543 PMCID: PMC4792600 DOI: 10.18632/oncotarget.6457] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 11/28/2015] [Indexed: 01/07/2023] Open
Abstract
mTOR is a critical target for controlling cell cycle progression, senescence and cell death in mammalian cancer cells. Here we studied the role of mTOR-dependent autophagy in implementating the antiprolifrative effect of mTORC1-specific inhibitor rapamycin and ATP-competitive mTOR kinase inhibitor pp242. We carried out a comprehensive analysis of pp242- and rapamycin-induced autophagy in ERas tumor cells. Rapamycin exerts cytostatic effect on ERas tumor cells, thus causing a temporary and reversible cell cycle arrest, activation of non-selective autophagy not accompanied by cell death. The rapamycin-treated cells are able to continue proliferation after drug removal. The ATP-competitive mTORC1/mTORC2 kinase inhibitor pp242 is highly cytotoxic by suppressing the function of mTORC1-4EBP1 axis and mTORC1-dependent phosphorylation of mTORC1 target--ULK1-Ser757 (Atg1). In contrast to rapamycin, pp242 activates the selective autophagy targeting mitochondria (mitophagy). The pp242-induced mitophagy is accompanied by accumulation of LC3 and conversion of LC3-I form to LC3-II. However reduced degradation of p62/SQSTM indicates abnormal flux of autophagic process. According to transmission electron microscopy data, short-term pp242-treated ERas cells exhibit numerous heavily damaged mitochondria, which are included in single membrane-bound autophagic/autolysophagic vacuoles (mitophagy). Despite the lack of typical for apoptosis features, ERas-treated cells with induced mitophagy revealed the activation of caspase 3, 9 and nucleosomal DNA fragmentation. Thus, pp242 activates autophagy with suppressed later stages, leading to impaired recycling and accumulation of dysfunctional mitochondria and cell death. Better understanding of how autophagy determines the fate of a cell--survival or cell death, can help to development of new strategy for cancer therapy.
Collapse
Affiliation(s)
- Serguei A Gordeev
- Institute of Cytology, Russian Academy of Sciences, St. Petersburg, Russia.,Saint Petersburg State University, St. Petersburg, Russia
| | - Tatiana V Bykova
- Institute of Cytology, Russian Academy of Sciences, St. Petersburg, Russia.,Saint Petersburg State University, St. Petersburg, Russia
| | - Svetlana G Zubova
- Institute of Cytology, Russian Academy of Sciences, St. Petersburg, Russia
| | - Olga A Bystrova
- Institute of Cytology, Russian Academy of Sciences, St. Petersburg, Russia
| | - Marina G Martynova
- Institute of Cytology, Russian Academy of Sciences, St. Petersburg, Russia
| | - Valery A Pospelov
- Institute of Cytology, Russian Academy of Sciences, St. Petersburg, Russia
| | | |
Collapse
|
560
|
Jemaà M, Manic G, Lledo G, Lissa D, Reynes C, Morin N, Chibon F, Sistigu A, Castedo M, Vitale I, Kroemer G, Abrieu A. Whole-genome duplication increases tumor cell sensitivity to MPS1 inhibition. Oncotarget 2016; 7:885-901. [PMID: 26637805 PMCID: PMC4808040 DOI: 10.18632/oncotarget.6432] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 11/18/2015] [Indexed: 12/31/2022] Open
Abstract
Several lines of evidence indicate that whole-genome duplication resulting in tetraploidy facilitates carcinogenesis by providing an intermediate and metastable state more prone to generate oncogenic aneuploidy. Here, we report a novel strategy to preferentially kill tetraploid cells based on the abrogation of the spindle assembly checkpoint (SAC) via the targeting of TTK protein kinase (better known as monopolar spindle 1, MPS1). The pharmacological inhibition as well as the knockdown of MPS1 kills more efficiently tetraploid cells than their diploid counterparts. By using time-lapse videomicroscopy, we show that tetraploid cells do not survive the aborted mitosis due to SAC abrogation upon MPS1 depletion. On the contrary diploid cells are able to survive up to at least two more cell cycles upon the same treatment. This effect might reflect the enhanced difficulty of cells with whole-genome doubling to tolerate a further increase in ploidy and/or an elevated level of chromosome instability in the absence of SAC functions. We further show that MPS1-inhibited tetraploid cells promote mitotic catastrophe executed by the intrinsic pathway of apoptosis, as indicated by the loss of mitochondrial potential, the release of the pro-apoptotic cytochrome c from mitochondria, and the activation of caspases. Altogether, our results suggest that MPS1 inhibition could be used as a therapeutic strategy for targeting tetraploid cancer cells.
Collapse
Affiliation(s)
- Mohamed Jemaà
- CRBM, CNRS UMR5237, Université de Montpellier, Montpellier, France
| | | | - Gwendaline Lledo
- CRBM, CNRS UMR5237, Université de Montpellier, Montpellier, France
| | - Delphine Lissa
- Université Paris-Sud/Paris XI, Le Kremlin-Bicêtre, France.,INSERM, UMRS1138, Paris, France.,Equipe 11 Labelisée par la Ligue Nationale Contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,Gustave Roussy Cancer Campus, Villejuif, France
| | - Christelle Reynes
- EA 2415, Laboratoire de Biostatistique, d'Epidémiologie et de Recherche Clinique, Université de Montpellier, Montpellier, France
| | - Nathalie Morin
- CRBM, CNRS UMR5237, Université de Montpellier, Montpellier, France
| | - Frédéric Chibon
- Department of Biopathology, Institut Bergonié, Comprehensive Cancer Centre, Bordeaux, France.,INSERM U916, Bordeaux, France
| | | | - Maria Castedo
- Université Paris-Sud/Paris XI, Le Kremlin-Bicêtre, France.,INSERM, UMRS1138, Paris, France.,Equipe 11 Labelisée par la Ligue Nationale Contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,Gustave Roussy Cancer Campus, Villejuif, France
| | - Ilio Vitale
- Regina Elena National Cancer Institute, Rome, Italy.,Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| | - Guido Kroemer
- INSERM, UMRS1138, Paris, France.,Equipe 11 Labelisée par la Ligue Nationale Contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,Université Pierre et Marie Curie/Paris VI, Paris, France.,Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Ariane Abrieu
- CRBM, CNRS UMR5237, Université de Montpellier, Montpellier, France
| |
Collapse
|
561
|
Zamyatnin AA. Plant Proteases Involved in Regulated Cell Death. BIOCHEMISTRY (MOSCOW) 2016; 80:1701-15. [PMID: 26878575 DOI: 10.1134/s0006297915130064] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Each plant genome encodes hundreds of proteolytic enzymes. These enzymes can be divided into five distinct classes: cysteine-, serine-, aspartic-, threonine-, and metalloproteinases. Despite the differences in their structural properties and activities, members of all of these classes in plants are involved in the processes of regulated cell death - a basic feature of eukaryotic organisms. Regulated cell death in plants is an indispensable mechanism supporting plant development, survival, stress responses, and defense against pathogens. This review summarizes recent advances in studies of plant proteolytic enzymes functioning in the initiation and execution of distinct types of regulated cell death.
Collapse
Affiliation(s)
- A A Zamyatnin
- Sechenov First Moscow State Medical University, Institute of Molecular Medicine, Moscow, 119991, Russia
| |
Collapse
|
562
|
Antiferroptotic activity of non-oxidative dopamine. Biochem Biophys Res Commun 2016; 480:602-607. [PMID: 27793671 DOI: 10.1016/j.bbrc.2016.10.099] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Accepted: 10/25/2016] [Indexed: 02/07/2023]
Abstract
Dopamine is a neurotransmitter that has many functions in the nervous and immune systems. Ferroptosis is a non-apoptotic form of regulated cell death that is involved in cancer and neurodegenerative diseases. However, the role of dopamine in ferroptosis remains unidentified. Here, we show that the non-oxidative form of dopamine is a strong inhibitor of ferroptotic cell death. Dopamine dose-dependently blocked ferroptosis in cancer (PANC1 and HEY) and non-cancer (MEF and HEK293) cells following treatment with erastin, a small molecule ferroptosis inducer. Notably, dopamine reduced erastin-induced ferrous iron accumulation, glutathione depletion, and malondialdehyde production. Mechanically, dopamine increased the protein stability of glutathione peroxidase 4, a phospholipid hydroperoxidase that protects cells against membrane lipid peroxidation. Moreover, dopamine suppressed dopamine receptor D4 protein degradation and promoted dopamine receptor D5 gene expression. Thus, our findings uncover a novel function of dopamine in cell death and provide new insight into the regulation of iron metabolism and lipid peroxidation by neurotransmitters.
Collapse
|
563
|
Autophagy and the invisible line between life and death. Eur J Cell Biol 2016; 95:598-610. [PMID: 28340912 DOI: 10.1016/j.ejcb.2016.10.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 10/24/2016] [Accepted: 10/24/2016] [Indexed: 02/07/2023] Open
Abstract
For a considerable time cell death has been considered to represent mutually exclusive states with cell death modalities that are governed by their inherent and unique mode of action involving specific molecular entities and have therefore been studied primarily in isolation. It is now, however, becoming increasingly clear that these modalities are regulated by similar pathways and share a number of initiator and effector molecules that control both cell death as well as cell survival mechanisms, demanding a newly aligned and integrative approach of cell death assessment. Frequently cell death is triggered through a dual action that incorporates signaling events associated with more than one death modality. Apoptosis and necrosis regularly co-operate in a tightly balanced interplay that involves autophagy to serve context dependently either as a pro-survival or a pro-death mechanism. In this review we will assess current cell death modalities and their molecular overlap with the goal of clarifying the controversial role of autophagy in the cell death response. By dissecting the key molecular pathways and their positioning within a network of regulatory signalling hubs and checkpoints we discuss a distinct approach that integrates autophagy with a resultant cell death manifestation. In doing so, former classifications of cell death modalities fade and reveal the intricate molecular proportions and complexities of the cell death response that may contribute towards an enhanced means of cell death control.
Collapse
|
564
|
Abstract
Immunogenicity depends on two key factors: antigenicity and adjuvanticity. The presence of exogenous or mutated antigens explains why infected cells and malignant cells can initiate an adaptive immune response provided that the cells also emit adjuvant signals as a consequence of cellular stress and death. Several infectious pathogens have devised strategies to control cell death and limit the emission of danger signals from dying cells, thereby avoiding immune recognition. Similarly, cancer cells often escape immunosurveillance owing to defects in the molecular machinery that underlies the release of endogenous adjuvants. Here, we review current knowledge on the mechanisms that underlie the activation of immune responses against dying cells and their pathophysiological relevance.
Collapse
|
565
|
Naji A, Suganuma N, Espagnolle N, Yagyu K, Baba N, Sensebé L, Deschaseaux F. Rationale for Determining the Functional Potency of Mesenchymal Stem Cells in Preventing Regulated Cell Death for Therapeutic Use. Stem Cells Transl Med 2016; 6:713-719. [PMID: 28297565 PMCID: PMC5442793 DOI: 10.5966/sctm.2016-0289] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 09/19/2016] [Indexed: 12/19/2022] Open
Abstract
Mesenchymal stem (stromal) cells (MSCs) are being investigated for treating degenerative and inflammatory disorders because of their reparative and immunomodulatory properties. Intricate mechanisms relate cell death processes with immune responses, which have implications for degenerative and inflammatory conditions. We review the therapeutic value of MSCs in terms of preventing regulated cell death (RCD). When cells identify an insult, specific intracellular pathways are elicited for execution of RCD processes, such as apoptosis, necroptosis, and pyroptosis. To some extent, exacerbated RCD can provoke an intense inflammatory response and vice versa. Emerging studies are focusing on the molecular mechanisms deployed by MSCs to ameliorate the survival, bioenergetics, and functions of unfit immune or nonimmune cells. Given these aspects, and in light of MSC actions in modulating cell death processes, we suggest the use of novel functional in vitro assays to ensure the potency of MSCs for preventing RCD. Such analyses should be associated with existing functional assays measuring the anti‐inflammatory capabilities of MSCs in vitro. MSCs selected on the basis of two in vitro functional criteria (i.e., prevention of inflammation and RCD) could possess optimal therapeutic efficacy in vivo. In addition, we underline the implications of these perspectives in clinical studies of MSC therapy, with particular focus on acute respiratory distress syndrome. Stem Cells Translational Medicine2017;6:713–719
Collapse
Affiliation(s)
- Abderrahim Naji
- Center for Innovative and Translational Medicine, Kochi Medical School, Kochi University, Kochi, Japan
- Department of Environmental Medicine, Kochi Medical School, Kochi University, Kochi, Japan
| | - Narufumi Suganuma
- Center for Innovative and Translational Medicine, Kochi Medical School, Kochi University, Kochi, Japan
- Department of Environmental Medicine, Kochi Medical School, Kochi University, Kochi, Japan
| | - Nicolas Espagnolle
- STROMALab, INSERM U1031, EFS Pyrénées‐Méditerranée, Université de Toulouse, Toulouse, France
| | - Ken‐ichi Yagyu
- Science Research Center, Division of Biological Research, Life Sciences and Functional Materials, Kochi Medical School, Kochi University, Kochi, Japan
| | - Nobuyasu Baba
- Center for Innovative and Translational Medicine, Kochi Medical School, Kochi University, Kochi, Japan
| | - Luc Sensebé
- STROMALab, INSERM U1031, EFS Pyrénées‐Méditerranée, Université de Toulouse, Toulouse, France
| | - Frédéric Deschaseaux
- STROMALab, INSERM U1031, EFS Pyrénées‐Méditerranée, Université de Toulouse, Toulouse, France
| |
Collapse
|
566
|
Furness S, Hare DL, Kourakis A, Turnley AM, Wookey PJ. A novel ligand of calcitonin receptor reveals a potential new sensor that modulates programmed cell death. Cell Death Discov 2016; 2:16062. [PMID: 27777788 PMCID: PMC5056446 DOI: 10.1038/cddiscovery.2016.62] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 06/14/2016] [Accepted: 07/14/2016] [Indexed: 11/24/2022] Open
Abstract
We have discovered that the accumulation of an anti-calcitonin receptor (anti-CTR) antibody conjugated to a fluorophore (mAb2C4:AF568) provides a robust signal for cells undergoing apoptotic programmed cell death (PCD). PCD is an absolute requirement for normal development of metazoan organisms. PCD is a hallmark of common diseases such as cardiovascular disease and tissue rejection in graft versus host pathologies, and chemotherapeutics work by increasing PCD. This robust signal or high fluorescent events were verified by confocal microscopy and flow cytometry in several cell lines and a primary culture in which PCD had been induced. In Jurkat cells, GBM-L2 and MG63 cells, the percentage undergoing PCD that were positive for both mAb2C4:AF568 and annexin V ranged between 70 and >90%. In MG63 cells induced for the preapoptotic cell stress response (PACSR), the normal expression of α-tubulin, a key structural component of the cytoskeleton, and accumulation of mAb2C4:AF568 were mutually exclusive. Our data support a model in which CTR is upregulated during PACSR and recycles to the plasma membrane with apoptosis. In cells committed to apoptosis (α-tubulin negative), there is accumulation of the CTR-ligand mAb2C4:AF568 generating a high fluorescent event. The reagent mAb2C4:AF568 effectively identifies a novel event linked to apoptosis.
Collapse
Affiliation(s)
- Sgb Furness
- Drug Discovery Biology Laboratory, Monash Institute of Pharmaceutical Science, 381 Royal Parade, Parkville, Victoria 3052, Australia; Department of Pharmacology, Monash University, Wellington Road, Clayton, Parkville, Victoria 3800, Australia
| | - D L Hare
- Department of Medicine, University of Melbourne, Austin Health , Level 7, Lance Townsend Building, Studley Road, Heidelberg, Victoria 3084, Australia
| | - A Kourakis
- Department of Medicine, University of Melbourne, Austin Health , Level 7, Lance Townsend Building, Studley Road, Heidelberg, Victoria 3084, Australia
| | - A M Turnley
- Department of Anatomy and Neuroscience, Melbourne Brain Centre, University of Melbourne , Royal Parade, Parkville, Victoria 3010, Australia
| | - P J Wookey
- Department of Medicine, University of Melbourne, Austin Health , Level 7, Lance Townsend Building, Studley Road, Heidelberg, Victoria 3084, Australia
| |
Collapse
|
567
|
Chernobrovkin AL, Zubarev RA. How well can morphology assess cell death modality? A proteomics study. Cell Death Discov 2016; 2:16068. [PMID: 27752363 PMCID: PMC5045961 DOI: 10.1038/cddiscovery.2016.68] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 07/19/2016] [Accepted: 07/26/2016] [Indexed: 11/16/2022] Open
Abstract
While the focus of attempts to classify cell death programs has finally shifted in 2010s from microscopy-based morphological characteristics to biochemical assays, more recent discoveries have put the underlying assumptions of many such assays under severe stress, mostly because of the limited specificity of the assays. On the other hand, proteomics can quantitatively measure the abundances of thousands of proteins in a single experiment. Thus proteomics could develop a modern alternative to both semiquantitative morphology assessment as well as single-molecule biochemical assays. Here we tested this hypothesis by analyzing the proteomes of cells dying after been treated with various chemical agents. The most striking finding is that, for a multivariate model based on the proteome changes in three cells lines, the regulation patterns of the 200–500 most abundant proteins typically attributed to household type more accurately reflect that of the proteins directly interacting with the drug than any other protein subset grouped by common function or biological process, including cell death. This is in broad agreement with the 'rigid cell death mechanics' model where drug action mechanism and morphological changes caused by it are bijectively linked. This finding, if confirmed, will open way for a broad use of proteomics in death modality assessment.
Collapse
Affiliation(s)
- Alexey L Chernobrovkin
- Division of Physiological Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institutet , Scheelesväg 2, SE-17 177 Stockholm, Sweden
| | - Roman A Zubarev
- Division of Physiological Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institutet , Scheelesväg 2, SE-17 177 Stockholm, Sweden
| |
Collapse
|
568
|
Biasutto L, Azzolini M, Szabò I, Zoratti M. The mitochondrial permeability transition pore in AD 2016: An update. BIOCHIMICA ET BIOPHYSICA ACTA 2016; 1863:2515-30. [PMID: 26902508 DOI: 10.1016/j.bbamcr.2016.02.012] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 02/04/2016] [Accepted: 02/05/2016] [Indexed: 12/13/2022]
Abstract
Over the past 30years the mitochondrial permeability transition - the permeabilization of the inner mitochondrial membrane due to the opening of a wide pore - has progressed from being considered a curious artifact induced in isolated mitochondria by Ca(2+) and phosphate to a key cell-death-inducing process in several major pathologies. Its relevance is by now universally acknowledged and a pharmacology targeting the phenomenon is being developed. The molecular nature of the pore remains to this day uncertain, but progress has recently been made with the identification of the FOF1 ATP synthase as the probable proteic substrate. Researchers sharing this conviction are however divided into two camps: these believing that only the ATP synthase dimers or oligomers can form the pore, presumably in the contact region between monomers, and those who consider that the ring-forming c subunits in the FO sector actually constitute the walls of the pore. The latest development is the emergence of a new candidate: Spastic Paraplegia 7 (SPG7), a mitochondrial AAA-type membrane protease which forms a 6-stave barrel. This review summarizes recent developments of research on the pathophysiological relevance and on the molecular nature of the mitochondrial permeability transition pore. This article is part of a Special Issue entitled: Mitochondrial Channels edited by Pierre Sonveaux, Pierre Maechler and Jean-Claude Martinou.
Collapse
Affiliation(s)
- Lucia Biasutto
- CNR Neuroscience Institute, Viale G. Colombo 3, 35121 Padova, Italy; University of Padova, Department of Biomedical Sciences, Viale G. Colombo 3, 35121 Padova, Italy
| | - Michele Azzolini
- CNR Neuroscience Institute, Viale G. Colombo 3, 35121 Padova, Italy; University of Padova, Department of Biomedical Sciences, Viale G. Colombo 3, 35121 Padova, Italy
| | - Ildikò Szabò
- CNR Neuroscience Institute, Viale G. Colombo 3, 35121 Padova, Italy; University of Padova, Department of Biology, Viale G. Colombo 3, 35121 Padova, Italy
| | - Mario Zoratti
- CNR Neuroscience Institute, Viale G. Colombo 3, 35121 Padova, Italy; University of Padova, Department of Biomedical Sciences, Viale G. Colombo 3, 35121 Padova, Italy.
| |
Collapse
|
569
|
Nauseef WM, Kubes P. Pondering neutrophil extracellular traps with healthy skepticism. Cell Microbiol 2016; 18:1349-57. [PMID: 27470975 PMCID: PMC5025378 DOI: 10.1111/cmi.12652] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 07/22/2016] [Accepted: 07/25/2016] [Indexed: 12/12/2022]
Abstract
The authors engage in a dialogue that evaluates critically the state of the study of neutrophil extracellular traps (NETs), a phenomenon currently the object of considerable interest, with the goal of identifying those aspects that merit clarification in order to assign the process its proper place in our current understanding of cell biology. Since the seminal observations in the Zychlinsky laboratory that described the extrusion of filaments of nuclear DNA associated with histones and granule proteins from neutrophils stimulated in vitro, many investigators have examined the phenomenon of NET formation in numerous and diverse settings. However, an overview of work in this rapidly growing field prompts several fundamental questions about NETs, including their precise composition, the mechanisms by which they arise, their clinical relevance, and the interrelationship of those observed in vitro and in vivo. In this discussion, the authors challenge interpretation of data from some experimental settings and provide recommendations for specific studies that would address the concerns raised, improve understanding of the biological relevance of NETs, and strengthen the field.
Collapse
Affiliation(s)
- William M Nauseef
- Inflammation Program and Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, and Veterans Administration Medical Center, Iowa City, Iowa, 52240, USA.
| | - Paul Kubes
- Department of Physiology and Pharmacology, Immunology Research Group, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, T2N 4N1, Canada.
| |
Collapse
|
570
|
Latrasse D, Benhamed M, Bergounioux C, Raynaud C, Delarue M. Plant programmed cell death from a chromatin point of view. JOURNAL OF EXPERIMENTAL BOTANY 2016; 67:5887-5900. [PMID: 27639093 DOI: 10.1093/jxb/erw329] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Programmed cell death (PCD) is a ubiquitous genetically regulated process consisting of the activation of finely controlled signalling pathways that lead to cellular suicide. PCD can be part of a developmental programme (dPCD) or be triggered by environmental conditions (ePCD). In plant cells, as in animal cells, extensive chromatin condensation and degradation of the nuclear DNA are among the most conspicuous features of cells undergoing PCD. Changes in chromatin condensation could either reflect the structural changes required for internucleosomal fragmentation of nuclear DNA or relate to large-scale chromatin rearrangements associated with a major transcriptional switch occurring during cell death. The aim of this review is to give an update on plant PCD processes from a chromatin point of view. The first part will be dedicated to chromatin conformational changes associated with cell death observed in various developmental and physiological conditions, whereas the second part will be devoted to histone dynamics and DNA modifications associated with critical changes in genome expression during the cell death process.
Collapse
Affiliation(s)
- D Latrasse
- Institute of Plant Sciences Paris-Saclay IPS2, CNRS, INRA, Université Paris-Sud, Université Evry, Université Paris-Saclay, Batiment 630, 91405 Orsay, France
- Institute of Plant Sciences Paris-Saclay IPS2, Paris Diderot, Sorbonne Paris-Cité, Bâtiment 630, 91405 Orsay, France
| | - M Benhamed
- Institute of Plant Sciences Paris-Saclay IPS2, CNRS, INRA, Université Paris-Sud, Université Evry, Université Paris-Saclay, Batiment 630, 91405 Orsay, France
- Institute of Plant Sciences Paris-Saclay IPS2, Paris Diderot, Sorbonne Paris-Cité, Bâtiment 630, 91405 Orsay, France
| | - C Bergounioux
- Institute of Plant Sciences Paris-Saclay IPS2, CNRS, INRA, Université Paris-Sud, Université Evry, Université Paris-Saclay, Batiment 630, 91405 Orsay, France
- Institute of Plant Sciences Paris-Saclay IPS2, Paris Diderot, Sorbonne Paris-Cité, Bâtiment 630, 91405 Orsay, France
| | - C Raynaud
- Institute of Plant Sciences Paris-Saclay IPS2, CNRS, INRA, Université Paris-Sud, Université Evry, Université Paris-Saclay, Batiment 630, 91405 Orsay, France
- Institute of Plant Sciences Paris-Saclay IPS2, Paris Diderot, Sorbonne Paris-Cité, Bâtiment 630, 91405 Orsay, France
| | - M Delarue
- Institute of Plant Sciences Paris-Saclay IPS2, CNRS, INRA, Université Paris-Sud, Université Evry, Université Paris-Saclay, Batiment 630, 91405 Orsay, France
- Institute of Plant Sciences Paris-Saclay IPS2, Paris Diderot, Sorbonne Paris-Cité, Bâtiment 630, 91405 Orsay, France
| |
Collapse
|
571
|
Timmons AK, Mondragon AA, Meehan TL, McCall K. Control of non-apoptotic nurse cell death by engulfment genes in Drosophila. Fly (Austin) 2016; 11:104-111. [PMID: 27686122 DOI: 10.1080/19336934.2016.1238993] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Programmed cell death occurs as a normal part of oocyte development in Drosophila. For each egg that is formed, 15 germline-derived nurse cells transfer their cytoplasmic contents into the oocyte and die. Disruption of apoptosis or autophagy only partially inhibits the death of the nurse cells, indicating that other mechanisms significantly contribute to nurse cell death. Recently, we demonstrated that the surrounding stretch follicle cells non-autonomously promote nurse cell death during late oogenesis and that phagocytosis genes including draper, ced-12, and the JNK pathway are crucial for this process. When phagocytosis genes are inhibited in the follicle cells, events specifically associated with death of the nurse cells are impaired. Death of the nurse cells is not completely blocked in draper mutants, suggesting that other engulfment receptors are involved. Indeed, we found that the integrin subunit, αPS3, is enriched on stretch follicle cells during late oogenesis and is required for elimination of the nurse cells. Moreover, double mutant analysis revealed that integrins act in parallel to draper. Death of nurse cells in the Drosophila ovary is a unique example of programmed cell death that is both non-apoptotic and non-cell autonomously controlled.
Collapse
Affiliation(s)
| | | | - Tracy L Meehan
- a Department of Biology , Boston University , Boston , MA
| | | |
Collapse
|
572
|
Heulot M, Chevalier N, Puyal J, Margue C, Michel S, Kreis S, Kulms D, Barras D, Nahimana A, Widmann C. The TAT-RasGAP317-326 anti-cancer peptide can kill in a caspase-, apoptosis-, and necroptosis-independent manner. Oncotarget 2016; 7:64342-64359. [PMID: 27602963 PMCID: PMC5325447 DOI: 10.18632/oncotarget.11841] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 08/24/2016] [Indexed: 01/07/2023] Open
Abstract
Tumor cell resistance to apoptosis, which is triggered by many anti-tumor therapies, remains a major clinical problem. Therefore, development of more efficient therapies is a priority to improve cancer prognosis. We have previously shown that a cell-permeable peptide derived from the p120 Ras GTPase-activating protein (RasGAP), called TAT-RasGAP317-326, bears anti-malignant activities in vitro and in vivo, such as inhibition of metastatic progression and tumor cell sensitization to cell death induced by various anti-cancer treatments. Recently, we discovered that this RasGAP-derived peptide possesses the ability to directly kill some cancer cells. TAT-RasGAP317-326 can cause cell death in a manner that can be either partially caspase-dependent or fully caspase-independent. Indeed, TAT-RasGAP317-326-induced toxicity was not or only partially prevented when apoptosis was inhibited. Moreover, blocking other forms of cell death, such as necroptosis, parthanatos, pyroptosis and autophagy did not hamper the killing activity of the peptide. The death induced by TAT-RasGAP317-326 can therefore proceed independently from these modes of death. Our finding has potentially interesting clinical relevance because activation of a death pathway that is distinct from apoptosis and necroptosis in tumor cells could lead to the generation of anti-cancer drugs that target pathways not yet considered for cancer treatment.
Collapse
Affiliation(s)
- Mathieu Heulot
- Department of Physiology, University of Lausanne, Lausanne, Switzerland
| | - Nadja Chevalier
- Department of Physiology, University of Lausanne, Lausanne, Switzerland
| | - Julien Puyal
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - Christiane Margue
- Signal Transduction Laboratory, Life Sciences Research Unit, University of Luxembourg, Luxembourg, Luxembourg
| | - Sébastien Michel
- Department of Physiology, University of Lausanne, Lausanne, Switzerland
| | - Stephanie Kreis
- Signal Transduction Laboratory, Life Sciences Research Unit, University of Luxembourg, Luxembourg, Luxembourg
| | - Dagmar Kulms
- Experimental Dermatology, Department of Dermatology, TU-Dresden, Dresden, Germany
- Center for Regenerative Therapies, TU-Dresden, Dresden, Germany
| | - David Barras
- Bioinformatics Core Facility, Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Aimable Nahimana
- Service and Central Laboratory of Hematology, University Hospital of Lausanne, Lausanne, Switzerland
| | - Christian Widmann
- Department of Physiology, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
573
|
Gebremeskel S, Johnston B. Concepts and mechanisms underlying chemotherapy induced immunogenic cell death: impact on clinical studies and considerations for combined therapies. Oncotarget 2016; 6:41600-19. [PMID: 26486085 PMCID: PMC4747176 DOI: 10.18632/oncotarget.6113] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 09/22/2015] [Indexed: 01/12/2023] Open
Abstract
Chemotherapy has historically been thought to induce cancer cell death in an immunogenically silent manner. However, recent studies have demonstrated that therapeutic outcomes with specific chemotherapeutic agents (e.g. anthracyclines) correlate strongly with their ability to induce a process of immunogenic cell death (ICD) in cancer cells. This process generates a series of signals that stimulate the immune system to recognize and clear tumor cells. Extensive studies have revealed that chemotherapy-induced ICD occurs via the exposure/release of calreticulin (CALR), ATP, chemokine (C–X–C motif) ligand 10 (CXCL10) and high mobility group box 1 (HMGB1). This review provides an in-depth look into the concepts and mechanisms underlying CALR exposure, activation of the Toll-like receptor 3/IFN/CXCL10 axis, and the release of ATP and HMGB1 from dying cancer cells. Factors that influence the impact of ICD in clinical studies and the design of therapies combining chemotherapy with immunotherapy are also discussed.
Collapse
Affiliation(s)
- Simon Gebremeskel
- Department of Microbiology & Immunology, Dalhousie University, Halifax, Nova Scotia, Canada.,Beatrice Hunter Cancer Research Institute, Halifax, Nova Scotia, Canada
| | - Brent Johnston
- Department of Microbiology & Immunology, Dalhousie University, Halifax, Nova Scotia, Canada.,Department of Pediatrics, Dalhousie University, Halifax, Nova Scotia, Canada.,Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada.,Beatrice Hunter Cancer Research Institute, Halifax, Nova Scotia, Canada
| |
Collapse
|
574
|
He Z, Agostini M, Liu H, Melino G, Simon HU. p73 regulates basal and starvation-induced liver metabolism in vivo. Oncotarget 2016; 6:33178-90. [PMID: 26375672 PMCID: PMC4741757 DOI: 10.18632/oncotarget.5090] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 08/26/2015] [Indexed: 12/30/2022] Open
Abstract
As a member of the p53 gene family, p73 regulates cell cycle arrest, apoptosis, neurogenesis, immunity and inflammation. Recently, p73 has been shown to transcriptionally regulate selective metabolic enzymes, such as cytochrome c oxidase subunit IV isoform 1, glucose 6-phosphate dehydrogenase and glutaminase-2, resulting in significant effects on metabolism, including hepatocellular lipid metabolism, glutathione homeostasis and the pentose phosphate pathway. In order to further investigate the metabolic effect of p73, here, we compared the global metabolic profile of livers from p73 knockout and wild-type mice under both control and starvation conditions. Our results show that the depletion of all p73 isoforms cause altered lysine metabolism and glycolysis, distinct patterns for glutathione synthesis and Krebs cycle, as well as an elevated pentose phosphate pathway and abnormal lipid accumulation. These results indicate that p73 regulates basal and starvation-induced fuel metabolism in the liver, a finding that is likely to be highly relevant for metabolism-associated disorders, such as diabetes and cancer.
Collapse
Affiliation(s)
- Zhaoyue He
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Massimiliano Agostini
- Medical Research Council, Toxicology Unit, Leicester, United Kingdom.,Department of Experimental Medicine and Surgery, University of Rome "Tor Vergata", Rome, Italy
| | - He Liu
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Gerry Melino
- Medical Research Council, Toxicology Unit, Leicester, United Kingdom.,Department of Experimental Medicine and Surgery, University of Rome "Tor Vergata", Rome, Italy
| | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| |
Collapse
|
575
|
Carmody M, Waszczak C, Idänheimo N, Saarinen T, Kangasjärvi J. ROS signalling in a destabilised world: A molecular understanding of climate change. JOURNAL OF PLANT PHYSIOLOGY 2016; 203:69-83. [PMID: 27364884 DOI: 10.1016/j.jplph.2016.06.008] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 06/13/2016] [Accepted: 06/14/2016] [Indexed: 05/29/2023]
Abstract
Climate change results in increased intensity and frequency of extreme abiotic and biotic stress events. In plants, reactive oxygen species (ROS) accumulate in proportion to the level of stress and are major signalling and regulatory metabolites coordinating growth, defence, acclimation and cell death. Our knowledge of ROS homeostasis, sensing, and signalling is therefore key to understanding the impacts of climate change at the molecular level. Current research is uncovering new insights into temporal-spatial, cell-to-cell and systemic ROS signalling pathways, particularly how these affect plant growth, defence, and more recently acclimation mechanisms behind stress priming and long term stress memory. Understanding the stabilising and destabilising factors of ROS homeostasis and signalling in plants exposed to extreme and fluctuating stress will concomitantly reveal how to address future climate change challenges in global food security and biodiversity management.
Collapse
Affiliation(s)
- Melanie Carmody
- Division of Plant Biology, Viikki Plant Science Centre, Department of Biosciences, University of Helsinki, 00014 Helsinki, Finland.
| | - Cezary Waszczak
- Division of Plant Biology, Viikki Plant Science Centre, Department of Biosciences, University of Helsinki, 00014 Helsinki, Finland.
| | - Niina Idänheimo
- Division of Plant Biology, Viikki Plant Science Centre, Department of Biosciences, University of Helsinki, 00014 Helsinki, Finland.
| | - Timo Saarinen
- Division of Plant Biology, Viikki Plant Science Centre, Department of Biosciences, University of Helsinki, 00014 Helsinki, Finland.
| | - Jaakko Kangasjärvi
- Division of Plant Biology, Viikki Plant Science Centre, Department of Biosciences, University of Helsinki, 00014 Helsinki, Finland; Distinguished Scientist Fellowship Program, College of Science, King Saud University, Riyadh, Saudi Arabia.
| |
Collapse
|
576
|
Garanina AS, Khashba LA, Onishchenko GE. Stages of Cell Cannibalism--Entosis--in Normal Human Keratinocyte Culture. BIOCHEMISTRY (MOSCOW) 2016; 80:1469-77. [PMID: 26615438 DOI: 10.1134/s0006297915110085] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Entosis is a type of cell cannibalism during which one cell penetrates into another cell and usually dies inside it. Researchers mainly pay attention to initial and final stages of entosis. Besides, tumor cells in suspension are the primary object of studies. In the present study, we investigated morphological changes of both cells-participants of entosis during this process. The substrate-dependent culture of human normal keratinocytes HaCaT was chosen for the work. A combination of light microscopy and scanning electron microscopy was used to prove that one cell was completely surrounded by the plasma membrane of another cell. We investigated such "cell-in-cell" structures and described the structural and functional changes of both cells during entosis. The outer cell nucleus localization and shape were changed. Gradual degradation of the inner cell nucleus and of the junctions between the inner and the outer cells was revealed. Moreover, repeated redistribution of the outer cell membrane organelles (Golgi apparatus, lysosomes, mitochondria, and autophagosomes), rearrangement of its cytoskeleton, and change in the lysosomal, autophagosomal, and mitochondrial state in both entotic cells were observed during entosis. On the basis of these data, we divided entosis into five stages that make it possible to systematize description of this type of cell death.
Collapse
Affiliation(s)
- A S Garanina
- Lomonosov Moscow State University, Biological Faculty, Moscow, 119991, Russia.
| | | | | |
Collapse
|
577
|
Papoudou-Bai A, Hatzimichael E, Barbouti A, Kanavaros P. Expression patterns of the activator protein-1 (AP-1) family members in lymphoid neoplasms. Clin Exp Med 2016; 17:291-304. [PMID: 27600282 DOI: 10.1007/s10238-016-0436-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2016] [Accepted: 08/23/2016] [Indexed: 12/22/2022]
Abstract
The activator protein-1 (AP-1) is a dimeric transcription factor composed of proteins belonging to the Jun (c-Jun, JunB and JunD), Fos (c-Fos, FosB, Fra1 and Fra2) and activating transcription factor protein families. AP-1 is involved in various cellular events including differentiation, proliferation, survival and apoptosis. Deregulated expression of AP-1 transcription factors is implicated in the pathogenesis of various lymphomas such as classical Hodgkin lymphomas, anaplastic large cell lymphomas, diffuse large B cell lymphomas and adult T cell leukemia/lymphoma. The main purpose of this review is the analysis of the expression patterns of AP-1 transcription factors in order to gain insight into the histophysiology of lymphoid tissues and the pathology of lymphoid malignancies.
Collapse
Affiliation(s)
| | | | - Alexandra Barbouti
- Department of Anatomy-Histology-Embryology, Faculty of Medicine, University of Ioannina, Ioannina, Greece
| | - Panagiotis Kanavaros
- Department of Anatomy-Histology-Embryology, Faculty of Medicine, University of Ioannina, Ioannina, Greece.
| |
Collapse
|
578
|
Denèfle T, Boullet H, Herbi L, Newton C, Martinez-Torres AC, Guez A, Pramil E, Quiney C, Pourcelot M, Levasseur MD, Lardé E, Moumné R, Ogi FX, Grondin P, Merle-Beral H, Lequin O, Susin SA, Karoyan P. Thrombospondin-1 Mimetic Agonist Peptides Induce Selective Death in Tumor Cells: Design, Synthesis, and Structure-Activity Relationship Studies. J Med Chem 2016; 59:8412-21. [PMID: 27526615 DOI: 10.1021/acs.jmedchem.6b00781] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Thrombospondin-1 (TSP-1) is a glycoprotein considered as a key actor within the tumor microenvironment. Its binding to CD47, a cell surface receptor, triggers programmed cell death. Previous studies allowed the identification of 4N1K decapeptide derived from the TSP-1/CD47 binding epitope. Here, we demonstrate that this peptide is able to induce selective apoptosis of various cancer cell lines while sparing normal cells. A structure-activity relationship study led to the design of the first serum stable TSP-1 mimetic agonist peptide able to trigger selective programmed cell death (PCD) of at least lung, breast, and colorectal cancer cells. Altogether, these results will be of valuable interest for further investigation in the design of potent CD47 agonist peptides, opening new perspectives for the development of original anticancer therapies.
Collapse
Affiliation(s)
- Thomas Denèfle
- Sorbonne Universités, UPMC Université Paris 06, Ecole Normale Supérieure, CNRS, Laboratoire des Biomolécules, 75005 Paris, France.,Département de Chimie, Ecole Normale Supérieure, PSL Research University, UPMC Université Paris 06, CNRS, Laboratoire des Biomolécules, 75005 Paris, France.,UPMC Université Paris 06, Laboratoire des BioMolécules, Site GSK, 25-27 Avenue du Québec, 91140 Les Ulis, France
| | - Héloise Boullet
- Sorbonne Universités, UPMC Université Paris 06, Ecole Normale Supérieure, CNRS, Laboratoire des Biomolécules, 75005 Paris, France.,Département de Chimie, Ecole Normale Supérieure, PSL Research University, UPMC Université Paris 06, CNRS, Laboratoire des Biomolécules, 75005 Paris, France
| | - Linda Herbi
- Cell Death and Drug Resistance in Lymphoproliferative Disorders Team, Centre de Recherche des Cordeliers, INSERM UMRS, 1138, Paris, France.,Sorbonne Universités, UPMC Université Paris 06, UMRS 1138, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, UMRS 1138, Paris, France
| | - Clara Newton
- Sorbonne Universités, UPMC Université Paris 06, Ecole Normale Supérieure, CNRS, Laboratoire des Biomolécules, 75005 Paris, France.,Département de Chimie, Ecole Normale Supérieure, PSL Research University, UPMC Université Paris 06, CNRS, Laboratoire des Biomolécules, 75005 Paris, France.,UPMC Université Paris 06, Laboratoire des BioMolécules, Site GSK, 25-27 Avenue du Québec, 91140 Les Ulis, France
| | - Ana-Carolina Martinez-Torres
- Cell Death and Drug Resistance in Lymphoproliferative Disorders Team, Centre de Recherche des Cordeliers, INSERM UMRS, 1138, Paris, France.,Sorbonne Universités, UPMC Université Paris 06, UMRS 1138, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, UMRS 1138, Paris, France
| | - Alexandre Guez
- Sorbonne Universités, UPMC Université Paris 06, Ecole Normale Supérieure, CNRS, Laboratoire des Biomolécules, 75005 Paris, France.,Département de Chimie, Ecole Normale Supérieure, PSL Research University, UPMC Université Paris 06, CNRS, Laboratoire des Biomolécules, 75005 Paris, France
| | - Elodie Pramil
- Sorbonne Universités, UPMC Université Paris 06, Ecole Normale Supérieure, CNRS, Laboratoire des Biomolécules, 75005 Paris, France.,Département de Chimie, Ecole Normale Supérieure, PSL Research University, UPMC Université Paris 06, CNRS, Laboratoire des Biomolécules, 75005 Paris, France.,Cell Death and Drug Resistance in Lymphoproliferative Disorders Team, Centre de Recherche des Cordeliers, INSERM UMRS, 1138, Paris, France.,Sorbonne Universités, UPMC Université Paris 06, UMRS 1138, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, UMRS 1138, Paris, France
| | - Claire Quiney
- Cell Death and Drug Resistance in Lymphoproliferative Disorders Team, Centre de Recherche des Cordeliers, INSERM UMRS, 1138, Paris, France.,Sorbonne Universités, UPMC Université Paris 06, UMRS 1138, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, UMRS 1138, Paris, France
| | - Marilyne Pourcelot
- Sorbonne Universités, UPMC Université Paris 06, Ecole Normale Supérieure, CNRS, Laboratoire des Biomolécules, 75005 Paris, France.,Département de Chimie, Ecole Normale Supérieure, PSL Research University, UPMC Université Paris 06, CNRS, Laboratoire des Biomolécules, 75005 Paris, France.,UPMC Université Paris 06, Laboratoire des BioMolécules, Site GSK, 25-27 Avenue du Québec, 91140 Les Ulis, France
| | - Mikail D Levasseur
- Sorbonne Universités, UPMC Université Paris 06, Ecole Normale Supérieure, CNRS, Laboratoire des Biomolécules, 75005 Paris, France.,Département de Chimie, Ecole Normale Supérieure, PSL Research University, UPMC Université Paris 06, CNRS, Laboratoire des Biomolécules, 75005 Paris, France.,UPMC Université Paris 06, Laboratoire des BioMolécules, Site GSK, 25-27 Avenue du Québec, 91140 Les Ulis, France
| | - Eva Lardé
- Sorbonne Universités, UPMC Université Paris 06, Ecole Normale Supérieure, CNRS, Laboratoire des Biomolécules, 75005 Paris, France.,Département de Chimie, Ecole Normale Supérieure, PSL Research University, UPMC Université Paris 06, CNRS, Laboratoire des Biomolécules, 75005 Paris, France.,UPMC Université Paris 06, Laboratoire des BioMolécules, Site GSK, 25-27 Avenue du Québec, 91140 Les Ulis, France
| | - Roba Moumné
- Sorbonne Universités, UPMC Université Paris 06, Ecole Normale Supérieure, CNRS, Laboratoire des Biomolécules, 75005 Paris, France.,Département de Chimie, Ecole Normale Supérieure, PSL Research University, UPMC Université Paris 06, CNRS, Laboratoire des Biomolécules, 75005 Paris, France
| | | | | | - Hélène Merle-Beral
- Cell Death and Drug Resistance in Lymphoproliferative Disorders Team, Centre de Recherche des Cordeliers, INSERM UMRS, 1138, Paris, France.,Sorbonne Universités, UPMC Université Paris 06, UMRS 1138, Paris, France.,AP-HP, GH Pitié-Salpêtrière, Service d'Hématologie Biologique, 75013 Paris, France
| | - Olivier Lequin
- Sorbonne Universités, UPMC Université Paris 06, Ecole Normale Supérieure, CNRS, Laboratoire des Biomolécules, 75005 Paris, France.,Département de Chimie, Ecole Normale Supérieure, PSL Research University, UPMC Université Paris 06, CNRS, Laboratoire des Biomolécules, 75005 Paris, France
| | - Santos A Susin
- Cell Death and Drug Resistance in Lymphoproliferative Disorders Team, Centre de Recherche des Cordeliers, INSERM UMRS, 1138, Paris, France.,Sorbonne Universités, UPMC Université Paris 06, UMRS 1138, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, UMRS 1138, Paris, France
| | - Philippe Karoyan
- Sorbonne Universités, UPMC Université Paris 06, Ecole Normale Supérieure, CNRS, Laboratoire des Biomolécules, 75005 Paris, France.,Département de Chimie, Ecole Normale Supérieure, PSL Research University, UPMC Université Paris 06, CNRS, Laboratoire des Biomolécules, 75005 Paris, France.,UPMC Université Paris 06, Laboratoire des BioMolécules, Site GSK, 25-27 Avenue du Québec, 91140 Les Ulis, France
| |
Collapse
|
579
|
Ilie A, Gao AYL, Reid J, Boucher A, McEwan C, Barrière H, Lukacs GL, McKinney RA, Orlowski J. A Christianson syndrome-linked deletion mutation (∆(287)ES(288)) in SLC9A6 disrupts recycling endosomal function and elicits neurodegeneration and cell death. Mol Neurodegener 2016; 11:63. [PMID: 27590723 PMCID: PMC5010692 DOI: 10.1186/s13024-016-0129-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 08/27/2016] [Indexed: 01/19/2023] Open
Abstract
Background Christianson Syndrome, a recently identified X-linked neurodevelopmental disorder, is caused by mutations in the human gene SLC9A6 encoding the recycling endosomal alkali cation/proton exchanger NHE6. The patients have pronounced limitations in cognitive ability, motor skills and adaptive behaviour. However, the mechanistic basis for this disorder is poorly understood as few of the more than 20 mutations identified thus far have been studied in detail. Methods Here, we examined the molecular and cellular consequences of a 6 base-pair deletion of amino acids Glu287 and Ser288 (∆ES) in the predicted seventh transmembrane helix of human NHE6 expressed in established cell lines (CHO/AP-1, HeLa and neuroblastoma SH-SY5Y) and primary cultures of mouse hippocampal neurons by measuring levels of protein expression, stability, membrane trafficking, endosomal function and cell viability. Results In the cell lines, immunoblot analyses showed that the nascent mutant protein was properly synthesized and assembled as a homodimer, but its oligosaccharide maturation and half-life were markedly reduced compared to wild-type (WT) and correlated with enhanced ubiquitination leading to both proteasomal and lysosomal degradation. Despite this instability, a measurable fraction of the transporter was correctly sorted to the plasma membrane. However, the rates of clathrin-mediated endocytosis of the ∆ES mutant as well as uptake of companion vesicular cargo, such as the ligand-bound transferrin receptor, were significantly reduced and correlated with excessive endosomal acidification. Notably, ectopic expression of ∆ES but not WT induced apoptosis when examined in AP-1 cells. Similarly, in transfected primary cultures of mouse hippocampal neurons, membrane trafficking of the ∆ES mutant was impaired and elicited marked reductions in total dendritic length, area and arborization, and triggered apoptotic cell death. Conclusions These results suggest that loss-of-function mutations in NHE6 disrupt recycling endosomal function and trafficking of cargo which ultimately leads to neuronal degeneration and cell death in Christianson Syndrome. Electronic supplementary material The online version of this article (doi:10.1186/s13024-016-0129-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Alina Ilie
- Department of Physiology, McGill University, Bellini Life Sciences Bldg., Rm, 166, 3649 Promenade Sir-William-Osler, Montreal, QC, H3G 0B1, Canada
| | - Andy Y L Gao
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada
| | - Jonathan Reid
- Department of Physiology, McGill University, Bellini Life Sciences Bldg., Rm, 166, 3649 Promenade Sir-William-Osler, Montreal, QC, H3G 0B1, Canada
| | - Annie Boucher
- Department of Physiology, McGill University, Bellini Life Sciences Bldg., Rm, 166, 3649 Promenade Sir-William-Osler, Montreal, QC, H3G 0B1, Canada
| | - Cassandra McEwan
- Department of Physiology, McGill University, Bellini Life Sciences Bldg., Rm, 166, 3649 Promenade Sir-William-Osler, Montreal, QC, H3G 0B1, Canada
| | - Hervé Barrière
- Department of Physiology, McGill University, Bellini Life Sciences Bldg., Rm, 166, 3649 Promenade Sir-William-Osler, Montreal, QC, H3G 0B1, Canada
| | - Gergely L Lukacs
- Department of Physiology, McGill University, Bellini Life Sciences Bldg., Rm, 166, 3649 Promenade Sir-William-Osler, Montreal, QC, H3G 0B1, Canada
| | - R Anne McKinney
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada
| | - John Orlowski
- Department of Physiology, McGill University, Bellini Life Sciences Bldg., Rm, 166, 3649 Promenade Sir-William-Osler, Montreal, QC, H3G 0B1, Canada.
| |
Collapse
|
580
|
Zhou H, Sauvat A, Gomes-da-Silva LC, Durand S, Forveille S, Iribarren K, Yamazaki T, Souquere S, Bezu L, Müller K, Leduc M, Liu P, Zhao L, Marabelle A, Zitvogel L, Rekdal Ø, Kepp O, Kroemer G. The oncolytic compound LTX-401 targets the Golgi apparatus. Cell Death Differ 2016; 23:2031-2041. [PMID: 27588704 DOI: 10.1038/cdd.2016.86] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 07/19/2016] [Accepted: 07/20/2016] [Indexed: 12/28/2022] Open
Abstract
LTX-401 is an oncolytic amino acid derivative with potential immunogenic properties. Here, we demonstrate that LTX-401 selectively destroys the structure of the Golgi apparatus, as determined by means of ultrastructural analyses and fluorescence microscopic observation of cells expressing Golgi-targeted GFP reporters. Subcellular fractionation followed by mass spectrometric detection revealed that LTX-401 selectively enriched in the Golgi rather than in mitochondria or in the cytosol. The Golgi-dissociating agent Brefeldin A (BFA) reduced cell killing by LTX-401 as it partially inhibited LTX-401-induced mitochondrial release of cytochrome c and the activation of BAX. The cytotoxic effect of LTX-401 was attenuated by the double knockout of BAX and BAK, as well as the mitophagy-enforced depletion of mitochondria, yet was refractory to caspase inhibition. LTX-401 induced all major hallmarks of immunogenic cell death detectable with biosensor cell lines including calreticulin exposure, ATP release, HMGB1 exodus and a type-1 interferon response. Moreover, LTX-401-treated tumors manifested a strong lymphoid infiltration. Altogether these results support the contention that LTX-401 can stimulate immunogenic cell death through a pathway in which Golgi-localized LTX-401 operates upstream of mitochondrial membrane permeabilization.
Collapse
Affiliation(s)
- Heng Zhou
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif 94805, France.,Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, INSERM U1138, Paris 75006, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris 75006, France.,Université Pierre et Marie Curie, 15 rue de l'Ecole de Médecine, Paris 75006, France.,University of Paris Sud XI, Le Kremlin-Bicêtre 94276, France
| | - Allan Sauvat
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif 94805, France.,Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, INSERM U1138, Paris 75006, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris 75006, France.,Université Pierre et Marie Curie, 15 rue de l'Ecole de Médecine, Paris 75006, France
| | - Lígia C Gomes-da-Silva
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif 94805, France.,Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, INSERM U1138, Paris 75006, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris 75006, France.,Université Pierre et Marie Curie, 15 rue de l'Ecole de Médecine, Paris 75006, France.,Chemistry Department, University of Coimbra, Coimbra 3004-535, Portugal
| | - Sylvère Durand
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif 94805, France.,Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, INSERM U1138, Paris 75006, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris 75006, France.,Université Pierre et Marie Curie, 15 rue de l'Ecole de Médecine, Paris 75006, France
| | - Sabrina Forveille
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif 94805, France.,Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, INSERM U1138, Paris 75006, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris 75006, France.,Université Pierre et Marie Curie, 15 rue de l'Ecole de Médecine, Paris 75006, France
| | - Kristina Iribarren
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif 94805, France.,Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, INSERM U1138, Paris 75006, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris 75006, France.,Université Pierre et Marie Curie, 15 rue de l'Ecole de Médecine, Paris 75006, France
| | - Takahiro Yamazaki
- University of Paris Sud XI, Le Kremlin-Bicêtre 94276, France.,Department of Immuno-Oncology, Institut de Cancérologie Gustave Roussy Cancer Campus, 114 rue Edouard Vaillant, Villejuif 94805, France.,Institut National de la Santé et de la Recherche Medicale (INSERM), U1015, Villejuif, France.,Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 507, Villejuif, France
| | - Sylvie Souquere
- Gustave Roussy Comprehensive Cancer Center, Villejuif, France CNRS, UMR9196, Villejuif, France
| | - Lucillia Bezu
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif 94805, France.,Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, INSERM U1138, Paris 75006, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris 75006, France.,Université Pierre et Marie Curie, 15 rue de l'Ecole de Médecine, Paris 75006, France.,University of Paris Sud XI, Le Kremlin-Bicêtre 94276, France
| | - Kevin Müller
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif 94805, France.,Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, INSERM U1138, Paris 75006, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris 75006, France.,Université Pierre et Marie Curie, 15 rue de l'Ecole de Médecine, Paris 75006, France.,University of Paris Sud XI, Le Kremlin-Bicêtre 94276, France
| | - Marion Leduc
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif 94805, France.,Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, INSERM U1138, Paris 75006, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris 75006, France.,Université Pierre et Marie Curie, 15 rue de l'Ecole de Médecine, Paris 75006, France
| | - Peng Liu
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif 94805, France.,Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, INSERM U1138, Paris 75006, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris 75006, France.,Université Pierre et Marie Curie, 15 rue de l'Ecole de Médecine, Paris 75006, France.,University of Paris Sud XI, Le Kremlin-Bicêtre 94276, France
| | - Liwei Zhao
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif 94805, France.,Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, INSERM U1138, Paris 75006, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris 75006, France.,Université Pierre et Marie Curie, 15 rue de l'Ecole de Médecine, Paris 75006, France.,University of Paris Sud XI, Le Kremlin-Bicêtre 94276, France
| | - Aurélien Marabelle
- Institut National de la Santé et de la Recherche Medicale (INSERM), U1015, Villejuif, France
| | - Laurence Zitvogel
- University of Paris Sud XI, Le Kremlin-Bicêtre 94276, France.,Department of Immuno-Oncology, Institut de Cancérologie Gustave Roussy Cancer Campus, 114 rue Edouard Vaillant, Villejuif 94805, France.,Institut National de la Santé et de la Recherche Medicale (INSERM), U1015, Villejuif, France.,Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 507, Villejuif, France
| | - Øystein Rekdal
- Lytix Biopharma AS, Oslo 0349, Norway.,Institute of Medical Biology, University of Tromsø, 9037 Tromsø, Norway
| | - Oliver Kepp
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif 94805, France.,Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, INSERM U1138, Paris 75006, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris 75006, France.,Université Pierre et Marie Curie, 15 rue de l'Ecole de Médecine, Paris 75006, France
| | - Guido Kroemer
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif 94805, France.,Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, INSERM U1138, Paris 75006, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris 75006, France.,Université Pierre et Marie Curie, 15 rue de l'Ecole de Médecine, Paris 75006, France.,Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris 75015, France.,Karolinska Institute, Department of Women's and Children's Health, Karolinska University Hospital, Stockholm 17176, Sweden
| |
Collapse
|
581
|
Ebrahim AS, Sabbagh H, Liddane A, Raufi A, Kandouz M, Al-Katib A. Hematologic malignancies: newer strategies to counter the BCL-2 protein. J Cancer Res Clin Oncol 2016; 142:2013-22. [PMID: 27043233 DOI: 10.1007/s00432-016-2144-1] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 03/09/2016] [Indexed: 12/23/2022]
Abstract
INTRODUCTION BCL-2 is the founding member of the BCL-2 family of apoptosis regulatory proteins that either induce (pro-apoptotic) or inhibit (anti-apoptotic) apoptosis. The anti-apoptotic BCL-2 is classified as an oncogene, as damage to the BCL-2 gene has been shown to cause a number of cancers, including lymphoma. Ongoing research has demonstrated that disruption of BCL-2 leads to cell death. BCL-2 is also known to be involved in the development of resistance to chemotherapeutic agents, further underscoring the importance of targeting the BCL-2 gene in cancer therapeutics. Thus, numerous approaches have been developed to block or modulate the production of BCL-2 at the RNA level using antisense oligonucleotides or at the protein level with BCL-2 inhibitors, such as the novel ABT737. METHODS In this article, we briefly review previous strategies to target the BCL-2 gene and focus on a new approach to silence DNA, DNA interference (DNAi). RESULTS AND CONCLUSION DNA interference is aimed at blocking BCL-2 gene transcription. Evaluations of this technology in preclinical and early clinical studies are very encouraging and strongly support further development of DNAi as cancer therapeutics. A pilot phase II clinical trial in patients with relapsed or refractory non-Hodgkin lymphoma, PNT2258 demonstrated clinical benefit in 11 of 13 patients with notable responses in diffuse large B cell lymphoma and follicular lymphoma. By targeting the DNA directly, the DNAi technology promises to be more effective compared with other gene-interference strategies that target the RNA or protein but leaves the dysregulated DNA functional.
Collapse
Affiliation(s)
- Abdul Shukkur Ebrahim
- Department of Internal Medicine-Lymphoma Research Lab, Wayne State University and School of Medicine, 8229 Scott Hall, 540 E. Canfield, Detroit, MI, 48201, USA
| | - Hussam Sabbagh
- Department of Internal Medicine-Lymphoma Research Lab, Wayne State University and School of Medicine, 8229 Scott Hall, 540 E. Canfield, Detroit, MI, 48201, USA
| | - Allison Liddane
- Department of Internal Medicine-Lymphoma Research Lab, Wayne State University and School of Medicine, 8229 Scott Hall, 540 E. Canfield, Detroit, MI, 48201, USA
| | - Ali Raufi
- Department of Internal Medicine-Lymphoma Research Lab, Wayne State University and School of Medicine, 8229 Scott Hall, 540 E. Canfield, Detroit, MI, 48201, USA
| | - Mustapha Kandouz
- Department of Pathology, Wayne State University, Detroit, MI, 48201, USA
| | - Ayad Al-Katib
- Department of Internal Medicine-Lymphoma Research Lab, Wayne State University and School of Medicine, 8229 Scott Hall, 540 E. Canfield, Detroit, MI, 48201, USA.
| |
Collapse
|
582
|
de Almeida SMV, da Silva LPBG, de Lima LRA, Longato GB, Padilha RJR, Alves LC, Brayner FA, Ruiz ALTG, de Carvalho JE, Beltrão EIC, de Lima MDCA, de Carvalho Júnior LB. Ultrastructural Assessment of 2-(acridin-9-ylmethylene)-N-phenylhydrazinecarbothioamide activity on human breast adenocarcinoma cells. Micron 2016; 90:114-122. [PMID: 27668344 DOI: 10.1016/j.micron.2016.08.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 07/18/2016] [Accepted: 08/28/2016] [Indexed: 12/31/2022]
Abstract
The aim of the present study was to investigate ultrastructural changes induced by (Z)-2-(acridin-9-ylmethylene)-N-phenylhydrazinecarbothioamide (APHCA) treatment on human breast adenocarcinoma cancer cells MCF-7, besides the evaluation of phosphatidylserine externalization and DNA fragmentation in treated cells. Cell viability analysis demonstrated concentration and time-manner cytotoxicity. Treated MCF-7 cells did not expose phosphatidylserine residues to the external plasma membrane surface and DNA fragmentation was not visualized by electrophoresis. Light microscopy showed compromised cell density and presence of vacuolization after APHCA treatment with 60μM. Scanning and transmission electron microscopies revealed hallmarks of autophagy, namely the presence of membrane bebbling and autophagosomes, besides shrunken cells and cell debris in treated MCF-7 cells. However, more specific tests such as the quantification of mammalian autophagy proteins are necessary to determine the kind of death that is trigged by APHCA.
Collapse
Affiliation(s)
- Sinara Mônica Vitalino de Almeida
- Laboratório de Imunopatologia Keizo Asami (LIKA) and Departamento de Bioquímica, Universidade Federal de Pernambuco (UFPE), Recife 50670-901, PE, Brazil; Faculdade de Ciências, Educação e Tecnologia de Garanhuns (FACETEG), Universidade de Pernambuco (UPE), Garanhuns 55290-000, PE, Brazil.
| | - Lúcia Patrícia Bezerra Gomes da Silva
- Laboratório de Imunopatologia Keizo Asami (LIKA) and Departamento de Bioquímica, Universidade Federal de Pernambuco (UFPE), Recife 50670-901, PE, Brazil
| | - Luiza Rayanna Amorim de Lima
- Laboratório de Imunopatologia Keizo Asami (LIKA) and Departamento de Bioquímica, Universidade Federal de Pernambuco (UFPE), Recife 50670-901, PE, Brazil
| | - Giovanna Barbarini Longato
- Laboratório de Pesquisa em Biologia Celular e Molecular de Tumores e Compostos Bioativos, Universidade São Francisco, Bragança Paulista 12916- 900, SP - Brazil
| | - Rafael José Ribeiro Padilha
- Laboratório de Imunopatologia Keizo Asami (LIKA) and Departamento de Bioquímica, Universidade Federal de Pernambuco (UFPE), Recife 50670-901, PE, Brazil
| | - Luiz Carlos Alves
- Laboratório de Imunopatologia Keizo Asami (LIKA) and Departamento de Bioquímica, Universidade Federal de Pernambuco (UFPE), Recife 50670-901, PE, Brazil; Fundação Oswaldo Cruz (CPqAM/FIOCRUZ), Centro de Pesquisas Aggeu Magalhães, Laboratório de Biologia Celular e Molecular. Avenida Professor Moraes Rêgo s/n, Cidade Universitária, Recife 50740-465, PE, Brazil; Universidade de Pernambuco (UPE), Instituto de Ciências Biológicas, Rua Arnóbio Marques 310, Recife 50100-130, Santo Amaro, PE, Brazil
| | - Fábio André Brayner
- Laboratório de Imunopatologia Keizo Asami (LIKA) and Departamento de Bioquímica, Universidade Federal de Pernambuco (UFPE), Recife 50670-901, PE, Brazil; Fundação Oswaldo Cruz (CPqAM/FIOCRUZ), Centro de Pesquisas Aggeu Magalhães, Laboratório de Biologia Celular e Molecular. Avenida Professor Moraes Rêgo s/n, Cidade Universitária, Recife 50740-465, PE, Brazil
| | - Ana Lucia Tasca Gois Ruiz
- Divisão de Farmacologia e Toxicologia, Centro Pluridisciplinar de Pesquisas Químicas, Biológicas e Agrícolas, Universidade Estadual de Campinas, Campinas 13083-970, SP, Brazil
| | - João Ernesto de Carvalho
- Divisão de Farmacologia e Toxicologia, Centro Pluridisciplinar de Pesquisas Químicas, Biológicas e Agrícolas, Universidade Estadual de Campinas, Campinas 13083-970, SP, Brazil; Faculdade de Ciências Farmacêuticas, Universidade Estadual de Campinas, Campinas 13083-859, SP, Brazil
| | - Eduardo Isidoro Carneiro Beltrão
- Laboratório de Imunopatologia Keizo Asami (LIKA) and Departamento de Bioquímica, Universidade Federal de Pernambuco (UFPE), Recife 50670-901, PE, Brazil
| | | | - Luiz Bezerra de Carvalho Júnior
- Laboratório de Imunopatologia Keizo Asami (LIKA) and Departamento de Bioquímica, Universidade Federal de Pernambuco (UFPE), Recife 50670-901, PE, Brazil
| |
Collapse
|
583
|
Forveille S, Zhou H, Sauvat A, Bezu L, Müller K, Liu P, Zitvogel L, Pierron G, Rekdal Ø, Kepp O, Kroemer G. The oncolytic peptide LTX-315 triggers necrotic cell death. Cell Cycle 2016; 14:3506-12. [PMID: 26566869 DOI: 10.1080/15384101.2015.1093710] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The oncolytic peptide LTX-315 has been designed for killing human cancer cells and turned out to stimulate anti-cancer immune responses when locally injected into tumors established in immunocompetent mice. Here, we investigated the question whether LTX-315 induces apoptosis or necrosis. Transmission electron microscopy or morphometric analysis of chromatin-stained tumor cells revealed that LTX-315 failed to induce apoptotic nuclear condensation and rather induced a necrotic phenotype. Accordingly, LTX-315 failed to stimulate the activation of caspase-3, and inhibition of caspases by means of Z-VAD-fmk was unable to reduce cell killing by LTX-315. In addition, 2 prominent inhibitors of regulated necrosis (necroptosis), namely, necrostatin-1 and cycosporin A, failed to reduce LTX-315-induced cell death. In conclusion, it appears that LTX-315 triggers unregulated necrosis, which may contribute to its pro-inflammatory and pro-immune effects.
Collapse
Affiliation(s)
- Sabrina Forveille
- a Metabolomics and Cell Biology Platforms; Gustave Roussy Comprehensive Cancer Institute ; Villejuif , France.,b Equipe 11 labellisée Ligue contre le Cancer; Center de Recherche des Cordeliers; INSERM U 1138 ; Paris , France.,c Université Paris Descartes; Sorbonne Paris Cité ; Paris , France.,d Université Pierre et Marie Curie ; Paris , France
| | - Heng Zhou
- a Metabolomics and Cell Biology Platforms; Gustave Roussy Comprehensive Cancer Institute ; Villejuif , France.,b Equipe 11 labellisée Ligue contre le Cancer; Center de Recherche des Cordeliers; INSERM U 1138 ; Paris , France.,c Université Paris Descartes; Sorbonne Paris Cité ; Paris , France.,d Université Pierre et Marie Curie ; Paris , France.,e University of Paris Sud XI; Kremlin Bicêtre , France
| | - Allan Sauvat
- a Metabolomics and Cell Biology Platforms; Gustave Roussy Comprehensive Cancer Institute ; Villejuif , France.,b Equipe 11 labellisée Ligue contre le Cancer; Center de Recherche des Cordeliers; INSERM U 1138 ; Paris , France.,c Université Paris Descartes; Sorbonne Paris Cité ; Paris , France.,d Université Pierre et Marie Curie ; Paris , France
| | - Lucillia Bezu
- a Metabolomics and Cell Biology Platforms; Gustave Roussy Comprehensive Cancer Institute ; Villejuif , France.,b Equipe 11 labellisée Ligue contre le Cancer; Center de Recherche des Cordeliers; INSERM U 1138 ; Paris , France.,c Université Paris Descartes; Sorbonne Paris Cité ; Paris , France.,d Université Pierre et Marie Curie ; Paris , France.,e University of Paris Sud XI; Kremlin Bicêtre , France
| | - Kevin Müller
- a Metabolomics and Cell Biology Platforms; Gustave Roussy Comprehensive Cancer Institute ; Villejuif , France.,b Equipe 11 labellisée Ligue contre le Cancer; Center de Recherche des Cordeliers; INSERM U 1138 ; Paris , France.,c Université Paris Descartes; Sorbonne Paris Cité ; Paris , France.,d Université Pierre et Marie Curie ; Paris , France.,e University of Paris Sud XI; Kremlin Bicêtre , France
| | - Peng Liu
- a Metabolomics and Cell Biology Platforms; Gustave Roussy Comprehensive Cancer Institute ; Villejuif , France.,b Equipe 11 labellisée Ligue contre le Cancer; Center de Recherche des Cordeliers; INSERM U 1138 ; Paris , France.,c Université Paris Descartes; Sorbonne Paris Cité ; Paris , France.,d Université Pierre et Marie Curie ; Paris , France.,e University of Paris Sud XI; Kremlin Bicêtre , France
| | - Laurence Zitvogel
- e University of Paris Sud XI; Kremlin Bicêtre , France.,f Department of Immuno-Oncology ; Institut de Cancérologie Gustave Roussy Cancer Campus ; Villejuif , France.,g Institut National de la Santé et de la Recherche Medicale (INSERM), U1015 ; Villejuif , France.,h Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 507 ; Villejuif , France
| | - Gérard Pierron
- i Gustave Roussy Comprehensive Cancer Center; Villejuif; France CNRS; UMR8122 , Villejuif , France
| | - Øystein Rekdal
- j University of Tromsø; Institute of Medical Biology ; Tromsø , Norway.,k Lytix Biopharma ; Oslo , Norway
| | - Oliver Kepp
- a Metabolomics and Cell Biology Platforms; Gustave Roussy Comprehensive Cancer Institute ; Villejuif , France.,b Equipe 11 labellisée Ligue contre le Cancer; Center de Recherche des Cordeliers; INSERM U 1138 ; Paris , France.,c Université Paris Descartes; Sorbonne Paris Cité ; Paris , France.,d Université Pierre et Marie Curie ; Paris , France
| | - Guido Kroemer
- a Metabolomics and Cell Biology Platforms; Gustave Roussy Comprehensive Cancer Institute ; Villejuif , France.,b Equipe 11 labellisée Ligue contre le Cancer; Center de Recherche des Cordeliers; INSERM U 1138 ; Paris , France.,c Université Paris Descartes; Sorbonne Paris Cité ; Paris , France.,d Université Pierre et Marie Curie ; Paris , France.,l Pôle de Biologie; Hôpital Européen Georges Pompidou; AP-HP ; Paris , France.,m Karolinska Institute ; Department of Women's and Children's Health; Karolinska University Hospital ; Stockholm , Sweden
| |
Collapse
|
584
|
Petitjean M, Teste MA, Léger-Silvestre I, François JM, Parrou JL. RETRACTED:A new function for the yeast trehalose-6P synthase (Tps1) protein, as key pro-survival factor during growth, chronological ageing, and apoptotic stress. Mech Ageing Dev 2016; 161:234-246. [PMID: 27507670 DOI: 10.1016/j.mad.2016.07.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 07/20/2016] [Accepted: 07/25/2016] [Indexed: 12/20/2022]
Abstract
This article has been retracted: please see Elsevier Policy on Article Withdrawal (https://www.elsevier.com/about/our-business/policies/article-withdrawal).
This article has been retracted at the request of Marie-Ange Teste, Isabelle Léger-Silvestre, Jean M François and Jean-Luc Parrou. Marjorie Petitjean could not be reached.
The corresponding author identified major issues and brought them to the attention of the Journal.
These issues span from significant errors in the Material and Methods section of the article and major flaws in cytometry data analysis to data fabrication on the part of one of the authors.
Given these errors, the retracting authors state that the only responsible course of action would be to retract the article, to respect scientific integrity and maintain the standards and rigor of literature from the retracting authors' group as well as the Journal.
The retracting authors sincerely apologize to the readers and editors.
Collapse
Affiliation(s)
| | - Marie-Ange Teste
- LISBP, Université de Toulouse, CNRS, INRA, INSA, Toulouse, France
| | - Isabelle Léger-Silvestre
- Laboratoire de Biologie Moléculaire Eucaryote, CNRS, Université de Toulouse, 118 route de Narbonne, F-31000 Toulouse, France
| | - Jean M François
- LISBP, Université de Toulouse, CNRS, INRA, INSA, Toulouse, France
| | - Jean-Luc Parrou
- LISBP, Université de Toulouse, CNRS, INRA, INSA, Toulouse, France.
| |
Collapse
|
585
|
Smethurst DGJ, Cooper KF. ER fatalities-The role of ER-mitochondrial contact sites in yeast life and death decisions. Mech Ageing Dev 2016; 161:225-233. [PMID: 27507669 DOI: 10.1016/j.mad.2016.07.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 06/22/2016] [Accepted: 07/19/2016] [Indexed: 12/22/2022]
Abstract
Following extracellular stress signals, all eukaryotic cells choose whether to elicit a pro-survival or pro-death response. The decision over which path to take is governed by the severity and duration of the damage. In response to mild stress, pro-survival programs are initiated (unfolded protein response, autophagy, mitophagy) whereas severe or chronic stress forces the cell to abandon these adaptive programs and shift towards regulated cell death to remove irreversibly damaged cells. Both pro-survival and pro-death programs involve regulated communication between the endoplasmic reticulum (ER) and mitochondria. In yeast, recent data suggest this inter-organelle contact is facilitated by the endoplasmic reticulum mitochondria encounter structure (ERMES). These membrane contacts are not only important for the exchange of cellular signals, but also play a role in mitochondrial tethering during mitophagy, mitochondrial fission and mitochondrial inheritance. This review focuses on recent findings in yeast that shed light on how ER-mitochondrial communication mediates critical cell fate decisions.
Collapse
Affiliation(s)
- Daniel G J Smethurst
- Department of Molecular Biology, Rowan University School of Osteopathic Medicine, Stratford, NJ, 08055 USA
| | - Katrina F Cooper
- Department of Molecular Biology, Rowan University School of Osteopathic Medicine, Stratford, NJ, 08055 USA.
| |
Collapse
|
586
|
Tumour-cell-induced endothelial cell necroptosis via death receptor 6 promotes metastasis. Nature 2016; 536:215-8. [DOI: 10.1038/nature19076] [Citation(s) in RCA: 379] [Impact Index Per Article: 42.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 07/04/2016] [Indexed: 02/08/2023]
|
587
|
Editorial overview: Cancer. Curr Opin Pharmacol 2016; 29:v-vii. [PMID: 27476753 DOI: 10.1016/j.coph.2016.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
588
|
Marchi S, Pinton P. Alterations of calcium homeostasis in cancer cells. Curr Opin Pharmacol 2016; 29:1-6. [DOI: 10.1016/j.coph.2016.03.002] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 03/04/2016] [Accepted: 03/16/2016] [Indexed: 02/01/2023]
|
589
|
Vacchelli E, Bloy N, Aranda F, Buqué A, Cremer I, Demaria S, Eggermont A, Formenti SC, Fridman WH, Fucikova J, Galon J, Spisek R, Tartour E, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: Immunotherapy plus radiation therapy for oncological indications. Oncoimmunology 2016; 5:e1214790. [PMID: 27757313 DOI: 10.1080/2162402x.2016.1214790] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 07/15/2016] [Indexed: 02/08/2023] Open
Abstract
Malignant cells succumbing to some forms of radiation therapy are particularly immunogenic and hence can initiate a therapeutically relevant adaptive immune response. This reflects the intrinsic antigenicity of malignant cells (which often synthesize a high number of potentially reactive neo-antigens) coupled with the ability of radiation therapy to boost the adjuvanticity of cell death as it stimulates the release of endogenous adjuvants from dying cells. Thus, radiation therapy has been intensively investigated for its capacity to improve the therapeutic profile of several anticancer immunotherapies, including (but not limited to) checkpoint blockers, anticancer vaccines, oncolytic viruses, Toll-like receptor (TLR) agonists, cytokines, and several small molecules with immunostimulatory effects. Here, we summarize recent preclinical and clinical advances in this field of investigation.
Collapse
Affiliation(s)
- Erika Vacchelli
- INSERM, U1138, Paris, France; Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France; Université Pierre et Marie Curie/Paris VI, Paris, France; Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers, Paris, France; Gustave Roussy Cancer Campus, Villejuif, France
| | - Norma Bloy
- INSERM, U1138, Paris, France; Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France; Université Pierre et Marie Curie/Paris VI, Paris, France; Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers, Paris, France; Gustave Roussy Cancer Campus, Villejuif, France
| | - Fernando Aranda
- Group of Immune receptors of the Innate and Adaptive System, Institut d'Investigacions Biomédiques August Pi i Sunyer (IDIBAPS) , Barcelona, Spain
| | - Aitziber Buqué
- INSERM, U1138, Paris, France; Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France; Université Pierre et Marie Curie/Paris VI, Paris, France; Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers, Paris, France; Gustave Roussy Cancer Campus, Villejuif, France
| | - Isabelle Cremer
- INSERM, U1138, Paris, France; Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France; Université Pierre et Marie Curie/Paris VI, Paris, France; Equipe 13, Center de Recherche des Cordeliers, Paris, France
| | - Sandra Demaria
- Department of Radiation Oncology, Weill Cornell Medical College , New York, NY, USA
| | | | | | - Wolf Hervé Fridman
- INSERM, U1138, Paris, France; Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France; Université Pierre et Marie Curie/Paris VI, Paris, France; Equipe 13, Center de Recherche des Cordeliers, Paris, France
| | - Jitka Fucikova
- Sotio, Prague, Czech Republic; Department of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University, Prague, Czech Republic
| | - Jérôme Galon
- INSERM, U1138, Paris, France; Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France; Université Pierre et Marie Curie/Paris VI, Paris, France; Laboratory of Integrative Cancer Immunology, Center de Recherche des Cordeliers, Paris, France
| | - Radek Spisek
- Sotio, Prague, Czech Republic; Department of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University, Prague, Czech Republic
| | - Eric Tartour
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France; INSERM, U970, Paris, France; Paris-Cardiovascular Research Center (PARCC), Paris, France; Service d'Immunologie Biologique, Hôpital Européen Georges Pompidou (HEGP), AP-HP, Paris, France
| | - Laurence Zitvogel
- Gustave Roussy Cancer Campus, Villejuif, France; INSERM, U1015, CICBT1428, Villejuif, France
| | - Guido Kroemer
- INSERM, U1138, Paris, France; Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France; Université Pierre et Marie Curie/Paris VI, Paris, France; Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers, Paris, France; Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France; Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France; Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
| | - Lorenzo Galluzzi
- INSERM, U1138, Paris, France; Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France; Université Pierre et Marie Curie/Paris VI, Paris, France; Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers, Paris, France; Gustave Roussy Cancer Campus, Villejuif, France; Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
590
|
Abstract
This review embarks upon a cell death journey from the discovery of apoptosis and necrosis through to the coalescence of these: necroptosis. The mechanisms of 2 emerging necrotic cell death pathways, pyroptosis and ferroptosis, will be explored before delving into apoptotic and necroptotic signaling cascades, highlighting the complex interplay between molecular players. The involvement of the ripoptosome, interferon signaling and DNA damage in necroptosis will be discussed briefly. The major focus is on necroptosis initiation by tumor necrosis factor-α (TNFα) and its cognate receptor TNFR1, caspase-independent RIP1/RIP3/MLKL necrosome activation and cell death propagation by damage-associated molecular pattern (DAMP) release. Finally, the implications of a complex cell death signaling network will be revealed in the context of cancer biology and therapy. The clinical contribution of the discovery of necroptosis as an unequivocally new way of dying is monumental and could drastically alter cancer therapy strategies in the future.
Collapse
Affiliation(s)
- Britt Hanson
- a Department of Molecular Medicine and Haematology, Faculty of Health Sciences , University of the Witwatersrand , Johannesburg , South Africa
| |
Collapse
|
591
|
Di Paolo NC, Shayakhmetov DM. Interleukin 1α and the inflammatory process. Nat Immunol 2016; 17:906-13. [PMID: 27434011 PMCID: PMC5152572 DOI: 10.1038/ni.3503] [Citation(s) in RCA: 411] [Impact Index Per Article: 45.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 05/31/2016] [Indexed: 02/08/2023]
Abstract
Inflammation occurs after disruption of tissue homeostasis by cell stress, injury or infection and ultimately involves the recruitment and retention of cells of hematopoietic origin, which arrive at the affected sites to resolve damage and initiate repair. Interleukin 1α (IL-1α) and IL-1β are equally potent inflammatory cytokines that activate the inflammatory process, and their deregulated signaling causes devastating diseases manifested by severe acute or chronic inflammation. Although much attention has been given to understanding the biogenesis of IL-1β, the biogenesis of IL-1α and its distinctive role in the inflammatory process remain poorly defined. In this review we examine key aspects of IL-1α biology and regulation and discuss its emerging importance in the initiation and maintenance of inflammation that underlie the pathology of many human diseases.
Collapse
Affiliation(s)
- Nelson C Di Paolo
- Lowance Center for Human Immunology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Dmitry M Shayakhmetov
- Lowance Center for Human Immunology, Emory University School of Medicine, Atlanta, Georgia, USA
- Center for Transplantation and Immune-mediated Disorders, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
- Emory Vaccine Center, Departments of Pediatrics and Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
592
|
Blagonravov ML, Korshunova AY, Azova MM, Bryk AA, Frolov VA. Expression of Bax Protein and Morphological Changes in the Myocardium in Experimental Acute Pressure Overload of the Left Ventricle. Bull Exp Biol Med 2016; 161:312-5. [PMID: 27383165 DOI: 10.1007/s10517-016-3403-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Indexed: 01/11/2023]
Abstract
The expression of Bax protein, marker of intracellular pathway of apoptosis initiation, in viable left ventricular cardiomyocytes and morphological changes in the myocardium in acute pressure overload of the left ventricle were studied in experiment on male rabbits. The content of Bax protein in the cardiomyocyte cytoplasm decreased, this indicating that the mitochondrial pathway was not involved in the realization of the apoptotic program. This decrease was associated with manifest destructive changes in the left ventricular myocardium.
Collapse
Affiliation(s)
- M L Blagonravov
- Department of Pathology and Pathological Physiology, Moscow, Russia
| | - A Yu Korshunova
- Department of Pathology and Pathological Physiology, Moscow, Russia
| | - M M Azova
- Department of Biology and Common Genetics, Medical Institute, Russian, University of Peoples' Friendship, Moscow, Russia.
| | - A A Bryk
- Department of Pathology and Pathological Physiology, Moscow, Russia
| | - V A Frolov
- Department of Pathology and Pathological Physiology, Moscow, Russia
| |
Collapse
|
593
|
Salvesen GS, Hempel A, Coll NS. Protease signaling in animal and plant-regulated cell death. FEBS J 2016; 283:2577-98. [PMID: 26648190 PMCID: PMC5606204 DOI: 10.1111/febs.13616] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 11/23/2015] [Accepted: 11/30/2015] [Indexed: 12/26/2022]
Abstract
This review aims to highlight the proteases required for regulated cell death mechanisms in animals and plants. The aim is to be incisive, and not inclusive of all the animal proteases that have been implicated in various publications. The review also aims to focus on instances when several publications from disparate groups have demonstrated the involvement of an animal protease, and also when there is substantial biochemical, mechanistic and genetic evidence. In doing so, the literature can be culled to a handful of proteases, covering most of the known regulated cell death mechanisms: apoptosis, regulated necrosis, necroptosis, pyroptosis and NETosis in animals. In plants, the literature is younger and not as extensive as for mammals, although the molecular drivers of vacuolar death, necrosis and the hypersensitive response in plants are becoming clearer. Each of these death mechanisms has at least one proteolytic component that plays a major role in controlling the pathway, and sometimes they combine in networks to regulate cell death/survival decision nodes. Some similarities are found among animal and plant cell death proteases but, overall, the pathways that they govern are kingdom-specific with very little overlap.
Collapse
Affiliation(s)
- Guy S. Salvesen
- Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Anne Hempel
- Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Nuria Sanchez Coll
- Centre for Research in Agricultural Genomics, Campus UAB, Edifici CRAG, Bellaterra 08193, Barcelona, Spain
| |
Collapse
|
594
|
Ke B, Tian M, Li J, Liu B, He G. Targeting Programmed Cell Death Using Small-Molecule Compounds to Improve Potential Cancer Therapy. Med Res Rev 2016; 36:983-1035. [PMID: 27357603 DOI: 10.1002/med.21398] [Citation(s) in RCA: 134] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 05/04/2016] [Accepted: 05/28/2016] [Indexed: 02/05/2023]
Affiliation(s)
- Bowen Ke
- Department of Anesthesiology, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy; West China Hospital, Sichuan University; Chengdu 610041 China
| | - Mao Tian
- Department of Anesthesiology, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy; West China Hospital, Sichuan University; Chengdu 610041 China
| | - Jingjing Li
- Department of Anesthesiology, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy; West China Hospital, Sichuan University; Chengdu 610041 China
| | - Bo Liu
- Department of Anesthesiology, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy; West China Hospital, Sichuan University; Chengdu 610041 China
| | - Gu He
- Department of Anesthesiology, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy; West China Hospital, Sichuan University; Chengdu 610041 China
| |
Collapse
|
595
|
Hou L, Liu K, Li Y, Ma S, Ji X, Liu L. Necrotic pyknosis is a morphologically and biochemically distinct event from apoptotic pyknosis. J Cell Sci 2016; 129:3084-90. [PMID: 27358477 DOI: 10.1242/jcs.184374] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 06/23/2016] [Indexed: 12/17/2022] Open
Abstract
Classification of apoptosis and necrosis by morphological differences has been widely used for decades. However, this usefulness of this method has been seriously questioned in recent years, mainly due to a lack of functional and biochemical evidence to interpret the morphology changes. To address this matter, we devised genetic manipulations in Drosophila to study pyknosis, a process of nuclear shrinkage and chromatin condensation that occurs in apoptosis and necrosis. By following the progression of necrotic pyknosis, we surprisingly observed a transient state of chromatin detachment from the nuclear envelope, followed by the nuclear envelope completely collapsing onto chromatin. This phenomenon led us to discover that phosphorylation of barrier-to-autointegration factor (BAF) mediates this initial separation of nuclear envelope from chromatin. Functionally, inhibition of BAF phosphorylation suppressed necrosis in both Drosophila and human cells, suggesting that necrotic pyknosis is conserved in the propagation of necrosis. In contrast, during apoptotic pyknosis the chromatin did not detach from the nuclear envelope and inhibition of BAF phosphorylation had no effect on apoptotic pyknosis and apoptosis. Our research provides the first genetic evidence supporting a morphological classification of apoptosis and necrosis through different forms of pyknosis.
Collapse
Affiliation(s)
- Lin Hou
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing 100871, China Aging and Disease Laboratory of Xuanwu Hospital and Center of Stroke, Beijing Institute for Brain Disorders, Capital Medical University, Youanmen, Beijing 100069, China
| | - Kai Liu
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing 100871, China
| | - Yuhong Li
- Aging and Disease Laboratory of Xuanwu Hospital and Center of Stroke, Beijing Institute for Brain Disorders, Capital Medical University, Youanmen, Beijing 100069, China
| | - Shuang Ma
- Aging and Disease Laboratory of Xuanwu Hospital and Center of Stroke, Beijing Institute for Brain Disorders, Capital Medical University, Youanmen, Beijing 100069, China
| | - Xunming Ji
- Aging and Disease Laboratory of Xuanwu Hospital and Center of Stroke, Beijing Institute for Brain Disorders, Capital Medical University, Youanmen, Beijing 100069, China
| | - Lei Liu
- Aging and Disease Laboratory of Xuanwu Hospital and Center of Stroke, Beijing Institute for Brain Disorders, Capital Medical University, Youanmen, Beijing 100069, China
| |
Collapse
|
596
|
Fulda S. Redox regulation of Smac mimetic-induced cell death. Mol Cell Oncol 2016; 2:e1000697. [PMID: 27308489 PMCID: PMC4905326 DOI: 10.1080/23723556.2014.1000697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 12/17/2014] [Accepted: 12/17/2014] [Indexed: 11/17/2022]
Abstract
Cell death and survival programs are controlled by the cellular redox state, which is typically dysregulated during oncogenesis. A recent study reports that the inhibition of antioxidant defenses resulting from glutathione depletion can prime acute lymphoblastic leukemia cells for death induced by Smac mimetics.
Collapse
Affiliation(s)
- Simone Fulda
- Institute for Experimental Cancer Research in Pediatrics; Goethe-University ; Frankfurt, Germany
| |
Collapse
|
597
|
High-content screening identifies kinase inhibitors that overcome venetoclax resistance in activated CLL cells. Blood 2016; 128:934-47. [PMID: 27297795 DOI: 10.1182/blood-2015-12-687814] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Accepted: 06/08/2016] [Indexed: 12/12/2022] Open
Abstract
Novel agents such as the Bcl-2 inhibitor venetoclax (ABT-199) are changing treatment paradigms for chronic lymphocytic leukemia (CLL) but important problems remain. Although some patients exhibit deep and durable responses to venetoclax as a single agent, other patients harbor subpopulations of resistant leukemia cells that mediate disease recurrence. One hypothesis for the origin of resistance to venetoclax is by kinase-mediated survival signals encountered in proliferation centers that may be unique for individual patients. An in vitro microenvironment model was developed with primary CLL cells that could be incorporated into an automated high-content microscopy-based screen of kinase inhibitors (KIs) to identify agents that may improve venetoclax therapy in a personalized manner. Marked interpatient variability was noted for which KIs were effective; nevertheless, sunitinib was identified as the most common clinically available KI effective in overcoming venetoclax resistance. Examination of the underlying mechanisms indicated that venetoclax resistance may be induced by microenvironmental signals that upregulate antiapoptotic Bcl-xl, Mcl-1, and A1, which can be counteracted more efficiently by sunitinib than by ibrutinib or idelalisib. Although patient-specific drug responses are common, for many patients, combination therapy with sunitinib may significantly improve the therapeutic efficacy of venetoclax.
Collapse
|
598
|
Murphy E, Ardehali H, Balaban RS, DiLisa F, Dorn GW, Kitsis RN, Otsu K, Ping P, Rizzuto R, Sack MN, Wallace D, Youle RJ. Mitochondrial Function, Biology, and Role in Disease: A Scientific Statement From the American Heart Association. Circ Res 2016; 118:1960-91. [PMID: 27126807 PMCID: PMC6398603 DOI: 10.1161/res.0000000000000104] [Citation(s) in RCA: 336] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Cardiovascular disease is a major leading cause of morbidity and mortality in the United States and elsewhere. Alterations in mitochondrial function are increasingly being recognized as a contributing factor in myocardial infarction and in patients presenting with cardiomyopathy. Recent understanding of the complex interaction of the mitochondria in regulating metabolism and cell death can provide novel insight and therapeutic targets. The purpose of this statement is to better define the potential role of mitochondria in the genesis of cardiovascular disease such as ischemia and heart failure. To accomplish this, we will define the key mitochondrial processes that play a role in cardiovascular disease that are potential targets for novel therapeutic interventions. This is an exciting time in mitochondrial research. The past decade has provided novel insight into the role of mitochondria function and their importance in complex diseases. This statement will define the key roles that mitochondria play in cardiovascular physiology and disease and provide insight into how mitochondrial defects can contribute to cardiovascular disease; it will also discuss potential biomarkers of mitochondrial disease and suggest potential novel therapeutic approaches.
Collapse
|
599
|
Shutinoski B, Alturki NA, Rijal D, Bertin J, Gough PJ, Schlossmacher MG, Sad S. K45A mutation of RIPK1 results in poor necroptosis and cytokine signaling in macrophages, which impacts inflammatory responses in vivo. Cell Death Differ 2016; 23:1628-37. [PMID: 27258786 DOI: 10.1038/cdd.2016.51] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Revised: 04/28/2016] [Accepted: 05/05/2016] [Indexed: 02/06/2023] Open
Abstract
Receptor interacting protein kinase 1 (RIPK1) participates in several cell signaling complexes that promote cell activation and cell death. Stimulation of RIPK1 in the absence of caspase signaling induces regulated necrosis (necroptosis), which promotes an inflammatory response. Understanding of the mechanisms through which RIPK1 promotes inflammation has been unclear. Herein we have evaluated the impact of a K45A mutation of RIPK1 on necroptosis of macrophages and the activation of inflammatory response. We show that K45A mutation of RIPK1 results in attenuated necroptosis of macrophages in response to stimulation with LPS, TNFα and IFNβ in the absence of caspase signaling. Impairment in necroptosis correlated with poor phosphorylation of RIPK1, RIPK3 and reduced trimerization of MLKL. Furthermore, K45A mutation of RIPK1 resulted in poor STAT1 phosphorylation (at S727) and expression of RANTES and MIP-1α following TNF-R engagement in the absence of caspase activation. Our results further indicate that in the absence of stimulation by pathogen-associated molecular patterns (PAMPs), cellular inhibitors of apoptotic proteins (cIAPs) prevent the K45-dependent auto-phosphorylation of RIPK1, leading to resistance against necroptosis. Finally, RIPK1(K45A) mice displayed attenuated inflammatory response in vivo as they were significantly resistant against endotoxin shock, but highly susceptible against a challenge with Salmonella typhimurium. This correlated with reduced expression of IL-1β and ROS, and poor processing of caspase 8 by RIPK1(K45A) macrophages. Overall, these results indicate that K45 mediated kinase activity of RIPK1 is not only important for necroptosis but it also has a key role in promoting cytokine signaling and host response to inflammatory stimuli.
Collapse
Affiliation(s)
- B Shutinoski
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada.,Program in Neuroscience and Division of Neurology, The Ottawa Hospital, Ottawa, ON, Canada
| | - N A Alturki
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - D Rijal
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - J Bertin
- Pattern Recognition Receptor Discovery Performance Unit, Immuno-Inflammation Therapeutic Area, GlaxoSmithKline, Collegeville, PA 19426, USA
| | - P J Gough
- Pattern Recognition Receptor Discovery Performance Unit, Immuno-Inflammation Therapeutic Area, GlaxoSmithKline, Collegeville, PA 19426, USA
| | - M G Schlossmacher
- Program in Neuroscience and Division of Neurology, The Ottawa Hospital, Ottawa, ON, Canada
| | - S Sad
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| |
Collapse
|
600
|
Galluzzi L, Bravo-San Pedro JM, Blomgren K, Kroemer G. Autophagy in acute brain injury. Nat Rev Neurosci 2016; 17:467-84. [PMID: 27256553 DOI: 10.1038/nrn.2016.51] [Citation(s) in RCA: 166] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Autophagy is an evolutionarily ancient mechanism that ensures the lysosomal degradation of old, supernumerary or ectopic cytoplasmic entities. Most eukaryotic cells, including neurons, rely on proficient autophagic responses for the maintenance of homeostasis in response to stress. Accordingly, autophagy mediates neuroprotective effects following some forms of acute brain damage, including methamphetamine intoxication, spinal cord injury and subarachnoid haemorrhage. In some other circumstances, however, the autophagic machinery precipitates a peculiar form of cell death (known as autosis) that contributes to the aetiology of other types of acute brain damage, such as neonatal asphyxia. Here, we dissect the context-specific impact of autophagy on non-infectious acute brain injury, emphasizing the possible therapeutic application of pharmacological activators and inhibitors of this catabolic process for neuroprotection.
Collapse
Affiliation(s)
- Lorenzo Galluzzi
- Equipe 11 Labellisée Ligue Contre le Cancer, Centre de Recherche des Cordeliers, 75006 Paris, France.,INSERM, U1138, 75006 Paris, France.,Université Paris Descartes/Paris V, Sorbonne Paris Cité, 75006 Paris, France.,Université Pierre et Marie Curie/Paris VI, 75006 Paris, France.,Gustave Roussy Comprehensive Cancer Institute, 94805 Villejuif, France
| | - José Manuel Bravo-San Pedro
- Equipe 11 Labellisée Ligue Contre le Cancer, Centre de Recherche des Cordeliers, 75006 Paris, France.,INSERM, U1138, 75006 Paris, France.,Université Paris Descartes/Paris V, Sorbonne Paris Cité, 75006 Paris, France.,Université Pierre et Marie Curie/Paris VI, 75006 Paris, France.,Gustave Roussy Comprehensive Cancer Institute, 94805 Villejuif, France
| | - Klas Blomgren
- Karolinska Institute, Department of Women's and Children's Health, Karolinska University Hospital Q2:07, 17176 Stockholm, Sweden
| | - Guido Kroemer
- Equipe 11 Labellisée Ligue Contre le Cancer, Centre de Recherche des Cordeliers, 75006 Paris, France.,INSERM, U1138, 75006 Paris, France.,Université Paris Descartes/Paris V, Sorbonne Paris Cité, 75006 Paris, France.,Université Pierre et Marie Curie/Paris VI, 75006 Paris, France.,Karolinska Institute, Department of Women's and Children's Health, Karolinska University Hospital Q2:07, 17176 Stockholm, Sweden.,Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, 94805 Villejuif, France.,Pôle de Biologie, Hopitâl Européen George Pompidou, AP-HP, 75015 Paris, France
| |
Collapse
|