1
|
Chaves SR, Rego A, Santos-Pereira C, Sousa MJ, Côrte-Real M. Current and novel approaches in yeast cell death research. Cell Death Differ 2025; 32:207-218. [PMID: 38714881 PMCID: PMC11802841 DOI: 10.1038/s41418-024-01298-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 02/27/2024] [Accepted: 04/16/2024] [Indexed: 02/08/2025] Open
Abstract
The study of cell death mechanisms in fungi, particularly yeasts, has gained substantial interest in recent decades driven by the potential for biotechnological advancements and therapeutic interventions. Examples include the development of robust yeast strains for industrial fermentations and high-value compound production, novel food preservation strategies against spoilage yeasts, and the identification of targets for treating fungal infections in the clinic. In this review, we discuss a wide range of methods to characterize cellular alterations associated with yeast cell death, noting the advantages and limitations. We describe assays to monitor reversible events versus those that mark a commitment to cell death (point-of-no-return), as these distinctions are important to decipher the underlying regulatory mechanisms. Several well-known challenges remain, including the varied susceptibilities to death within a cell population and the delineation of detailed cell death mechanisms. The identification and characterization of morphologically distinct subsets of dying yeast cells within dynamic yeast populations provides opportunities to reveal novel vulnerabilities and survival mechanisms. Elucidating the intricacies of yeast regulated cell death (yRCD) will contribute to the advancement of scientific knowledge and foster breakthrough discoveries with broad-ranging implications.
Collapse
Affiliation(s)
- Susana R Chaves
- Centre of Molecular and Environmental Biology (CBMA), Department of Biology, University of Minho, Campus de Gualtar, Braga, Portugal.
| | - António Rego
- Centre of Biological Engineering (CEB), Department of Biological Engineering, University of Minho, Braga, Portugal
| | - Cátia Santos-Pereira
- Centre of Biological Engineering (CEB), Department of Biological Engineering, University of Minho, Braga, Portugal
| | - Maria João Sousa
- Centre of Molecular and Environmental Biology (CBMA), Department of Biology, University of Minho, Campus de Gualtar, Braga, Portugal
| | - Manuela Côrte-Real
- Centre of Molecular and Environmental Biology (CBMA), Department of Biology, University of Minho, Campus de Gualtar, Braga, Portugal.
| |
Collapse
|
2
|
Yin X, Dai F, Ran D, Zhang Y, Qu Z, Zheng S. Cysteine protease cathepsin B promotes lysosome integrity to extend the lifespan of alternative day fasting worms. Aging Cell 2024; 23:e14286. [PMID: 39046045 PMCID: PMC11561666 DOI: 10.1111/acel.14286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 06/30/2024] [Accepted: 07/10/2024] [Indexed: 07/25/2024] Open
Abstract
Alternative day fasting (ADF) has been shown to enhance the lifespan of animals. However, human trials evaluating the efficacy of ADF have only recently emerged, presenting challenges due to the extreme nature of this dietary regimen. To better understand the effects of ADF, we investigated its impact using Caenorhabditis elegans as a model organism. Our findings reveal that ADF extends the lifespan of worms nourished on animal-based protein source, while those fed with plant-based protein as the primary protein source do not experience such benefits. Remarkably, initiating ADF during midlife is sufficient to prolong lifespan, whereas implementation during youth results in developmental damage, and in older age, fails to provide additional extension effects. Furthermore, we discovered that midlife ADF up-regulates the expression of two cysteine protease cathepsin B genes, cpr-2 and cpr-5, which preserve lysosomal integrity and enhance its function in digesting aggregated proteins, as well as enhancing lipid metabolism and ameliorating neurodegenerative disease markers and phenomena during aging. This suggests that midlife ADF has long lasting anti-aging effects and may delay the onset of related diseases, specifically in animals consuming animal-based protein source. These findings offer valuable insights into the effects of ADF and provide guidance for future research and potential applications in individuals.
Collapse
Affiliation(s)
- Xue Yin
- School of Basic Medical SciencesHenan UniversityKaifengChina
| | - Fangzhou Dai
- School of Basic Medical SciencesHenan UniversityKaifengChina
| | - Dongyang Ran
- School of Basic Medical SciencesHenan UniversityKaifengChina
| | - Yutong Zhang
- School of Basic Medical SciencesHenan UniversityKaifengChina
| | - Zhi Qu
- School of Nursing and HealthHenan UniversityKaifengChina
| | - Shanqing Zheng
- School of Basic Medical SciencesHenan UniversityKaifengChina
- Laboratory of Cell Signal Transduction, Henan Provincial Engineering Centre for Tumor Molecular MedicineMedical School of Henan UniversityKaifengChina
- The Zhongzhou Laboratory for Integrative BiologyZhengzhouHenanChina
| |
Collapse
|
3
|
Wang Q, Zhang X, Du Z, Liu H, Xia Y, Xun L, Liu H. The Activity of YCA1 Metacaspase Is Regulated by Reactive Sulfane Sulfur via Persulfidation in Saccharomyces cerevisiae. Antioxidants (Basel) 2024; 13:589. [PMID: 38790694 PMCID: PMC11118234 DOI: 10.3390/antiox13050589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 05/06/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024] Open
Abstract
YCA1, the only metacaspase in Saccharomyces cerevisiae, plays important roles in the regulation of chronological lifespan, apoptosis, and cytokinesis. YCA1 has protein hydrolase activity and functions by cleaving itself and target proteins. However, there are few reports about the regulation of YCA1 activity. In this study, we observed that reactive sulfane sulfur (RSS) can inhibit the activity of YCA1. In vitro experiments demonstrated that RSS reacted with the Cys276 of YCA1, the residue central to its protein hydrolase activity, to form a persulfidation modification (protein-SSH). This modification inhibited both its self-cleavage and the cleavage of its substrate protein, BIR1. To investigate further, we constructed a low-endogenous-RSS mutant of S. cerevisiae, BY4742 Δcys3, in which the RSS-producing enzyme cystathionine-γ-lyase (CYS3) was knocked out. The activity of YCA1 was significantly increased by the deletion of CYS3. Moreover, increased YCA1 activity led to reduced chronological lifespan (CLS) and CLS-driven apoptosis. This study unveils the first endogenous factor that regulates YCA1 activity, introduces a novel mechanism of how yeast cells regulate chronological lifespan, and broadens our understanding of the multifaceted roles played by RSS.
Collapse
Affiliation(s)
- Qingda Wang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China; (Q.W.); (X.Z.); (Z.D.); (H.L.); (Y.X.); (L.X.)
| | - Xiaokun Zhang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China; (Q.W.); (X.Z.); (Z.D.); (H.L.); (Y.X.); (L.X.)
| | - Zhuang Du
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China; (Q.W.); (X.Z.); (Z.D.); (H.L.); (Y.X.); (L.X.)
| | - Honglei Liu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China; (Q.W.); (X.Z.); (Z.D.); (H.L.); (Y.X.); (L.X.)
| | - Yongzhen Xia
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China; (Q.W.); (X.Z.); (Z.D.); (H.L.); (Y.X.); (L.X.)
| | - Luying Xun
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China; (Q.W.); (X.Z.); (Z.D.); (H.L.); (Y.X.); (L.X.)
- Department of Chemistry, School of Molecular Biosciences, Washington State University, Pullman, WA 99164-4630, USA
| | - Huaiwei Liu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China; (Q.W.); (X.Z.); (Z.D.); (H.L.); (Y.X.); (L.X.)
| |
Collapse
|
4
|
Schulze A, Zimmermann A, Kainz K, Egger NB, Bauer MA, Madeo F, Carmona-Gutierrez D. Assessing chronological aging in Saccharomyces cerevisiae. Methods Cell Biol 2023; 181:87-108. [PMID: 38302246 DOI: 10.1016/bs.mcb.2022.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2024]
Abstract
Chronological age represents the time that passes between birth and a given date. To understand the complex network of factors contributing to chronological lifespan, a variety of model organisms have been implemented. One of the best studied organisms is the yeast Saccharomyces cerevisiae, which has greatly contributed toward identifying conserved biological mechanisms that act on longevity. Here, we discuss high- und low-throughput protocols to monitor and characterize chronological lifespan and chronological aging-associated cell death in S. cerevisiae. Included are propidium iodide staining with the possibility to quantitatively assess aging-associated cell death via flow cytometry or qualitative assessments via microscopy, cell viability assessment through plating and cell counting and cell death characterization via propidium iodide/AnnexinV staining and subsequent flow cytometric analysis or microscopy. Importantly, all of these methods combined give a clear picture of the chronological lifespan under different conditions or genetic backgrounds and represent a starting point for pharmacological or genetic interventions.
Collapse
Affiliation(s)
- Adina Schulze
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
| | - Andreas Zimmermann
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria; BioTechMed-Graz, Graz, Austria
| | - Katharina Kainz
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
| | - Nadine B Egger
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
| | - Maria A Bauer
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
| | - Frank Madeo
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria; BioTechMed-Graz, Graz, Austria; Field of Excellence BioHealth, University of Graz, Graz, Austria.
| | | |
Collapse
|
5
|
Nittari G, Tomassoni D, Roy P, Martinelli I, Tayebati SK, Amenta F. Batten disease through different in vivo and in vitro models: A review. J Neurosci Res 2023; 101:298-315. [PMID: 36434776 DOI: 10.1002/jnr.25147] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 10/25/2022] [Accepted: 11/09/2022] [Indexed: 11/27/2022]
Abstract
Batten disease consists of a family of primarily autosomal recessive, progressive neuropediatric disorders, also known as neuronal ceroid lipofuscinoses (NCLs). These pathologies are characterized by seizures and visual, cognitive and motor decline, and premature death. The pathophysiology of this rare disease is still unclear despite the years of trials and financial aids. This paper has reviewed advantages and limits of in vivo and in vitro models of Batten disease from murine and larger animal models to primitive unicellular models, until the most recently developed patient-derived induced pluripotent stem cells. For each model advantages, limits and applications were analyzed. The first prototypes investigated were murine models that due to their limits were replaced by larger animals. In vitro models gradually replaced animal models for practical, cost, and ethical reasons. Using induced pluripotent stem cells to study neurodegeneration is a new way of studying the disease, since they can be distinguished into differentiating elements like neurons, which are susceptible to neurodegeneration. In vivo and in vitro models have contributed to clarifying to some extent the pathophysiology of the disease. The collection and sharing of suitable human bio samples likely through biobanks can contribute to a better understanding, prevention, and to identify possible treatment strategies of Batten disease.
Collapse
Affiliation(s)
- Giulio Nittari
- School of Medicinal and Health Products Sciences, Clinical Research, Telemedicine and Telepharmacy Center, University of Camerino, Camerino, Italy
| | - Daniele Tomassoni
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Proshanta Roy
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Ilenia Martinelli
- School of Medicinal and Health Products Sciences, Clinical Research, Telemedicine and Telepharmacy Center, University of Camerino, Camerino, Italy
| | - Seyed Khosrow Tayebati
- School of Medicinal and Health Products Sciences, Clinical Research, Telemedicine and Telepharmacy Center, University of Camerino, Camerino, Italy
| | - Francesco Amenta
- School of Medicinal and Health Products Sciences, Clinical Research, Telemedicine and Telepharmacy Center, University of Camerino, Camerino, Italy
| |
Collapse
|
6
|
Gast V, Sandegren A, Dunås F, Ekblad S, Güler R, Thorén S, Tous Mohedano M, Molin M, Engqvist MKM, Siewers V. Engineering Saccharomyces cerevisiae for the production and secretion of Affibody molecules. Microb Cell Fact 2022; 21:36. [PMID: 35264156 PMCID: PMC8905840 DOI: 10.1186/s12934-022-01761-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 02/22/2022] [Indexed: 11/23/2022] Open
Abstract
Background Affibody molecules are synthetic peptides with a variety of therapeutic and diagnostic applications. To date, Affibody molecules have mainly been produced by the bacterial production host Escherichia coli. There is an interest in exploring alternative production hosts to identify potential improvements in terms of yield, ease of production and purification advantages. In this study, we evaluated the feasibility of Saccharomyces cerevisiae as a production chassis for this group of proteins. Results We examined the production of three different Affibody molecules in S. cerevisiae and found that these Affibody molecules were partially degraded. An albumin-binding domain, which may be attached to the Affibody molecules to increase their half-life, was identified to be a substrate for several S. cerevisiae proteases. We tested the removal of three vacuolar proteases, proteinase A, proteinase B and carboxypeptidase Y. Removal of one of these, proteinase A, resulted in intact secretion of one of the targeted Affibody molecules. Removal of either or both of the two additional proteases, carboxypeptidase Y and proteinase B, resulted in intact secretion of the two remaining Affibody molecules. The produced Affibody molecules were verified to bind their target, human HER3, as potently as the corresponding molecules produced in E. coli in an in vitro surface-plasmon resonance binding assay. Finally, we performed a fed-batch fermentation with one of the engineered protease-deficient S. cerevisiae strains and achieved a protein titer of 530 mg Affibody molecule/L. Conclusion This study shows that engineered S. cerevisiae has a great potential as a production host for recombinant Affibody molecules, reaching a high titer, and for proteins where endotoxin removal could be challenging, the use of S. cerevisiae obviates the need for endotoxin removal from protein produced in E. coli. Supplementary Information The online version contains supplementary material available at 10.1186/s12934-022-01761-0.
Collapse
Affiliation(s)
- Veronica Gast
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | | | | | | | | | | | - Marta Tous Mohedano
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Mikael Molin
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Martin K M Engqvist
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Verena Siewers
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden. .,Novo Nordisk Foundation Center for Biosustainability, Chalmers University of Technology, Gothenburg, Sweden.
| |
Collapse
|
7
|
Grosfeld EV, Bidiuk VA, Mitkevich OV, Ghazy ESMO, Kushnirov VV, Alexandrov AI. A Systematic Survey of Characteristic Features of Yeast Cell Death Triggered by External Factors. J Fungi (Basel) 2021; 7:886. [PMID: 34829175 PMCID: PMC8626022 DOI: 10.3390/jof7110886] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/15/2021] [Accepted: 10/17/2021] [Indexed: 12/20/2022] Open
Abstract
Cell death in response to distinct stimuli can manifest different morphological traits. It also depends on various cell death signaling pathways, extensively characterized in higher eukaryotes but less so in microorganisms. The study of cell death in yeast, and specifically Saccharomyces cerevisiae, can potentially be productive for understanding cell death, since numerous killing stimuli have been characterized for this organism. Here, we systematized the literature on external treatments that kill yeast, and which contains at least minimal data on cell death mechanisms. Data from 707 papers from the 7000 obtained using keyword searches were used to create a reference table for filtering types of cell death according to commonly assayed parameters. This table provides a resource for orientation within the literature; however, it also highlights that the common view of similarity between non-necrotic death in yeast and apoptosis in mammals has not provided sufficient progress to create a clear classification of cell death types. Differences in experimental setups also prevent direct comparison between different stimuli. Thus, side-by-side comparisons of various cell death-inducing stimuli under comparable conditions using existing and novel markers that can differentiate between types of cell death seem like a promising direction for future studies.
Collapse
Affiliation(s)
- Erika V. Grosfeld
- Moscow Institute of Physics and Technology, 9 Institutskiy per, Dolgoprudny, 141700 Moscow, Russia;
- Federal Research Center of Biotechnology of the RAS, Bach Institute of Biochemistry, 119071 Moscow, Russia; (V.A.B.); (O.V.M.); (E.S.M.O.G.); (V.V.K.)
| | - Victoria A. Bidiuk
- Federal Research Center of Biotechnology of the RAS, Bach Institute of Biochemistry, 119071 Moscow, Russia; (V.A.B.); (O.V.M.); (E.S.M.O.G.); (V.V.K.)
| | - Olga V. Mitkevich
- Federal Research Center of Biotechnology of the RAS, Bach Institute of Biochemistry, 119071 Moscow, Russia; (V.A.B.); (O.V.M.); (E.S.M.O.G.); (V.V.K.)
| | - Eslam S. M. O. Ghazy
- Federal Research Center of Biotechnology of the RAS, Bach Institute of Biochemistry, 119071 Moscow, Russia; (V.A.B.); (O.V.M.); (E.S.M.O.G.); (V.V.K.)
- Institute of Biochemical Technology and Nanotechnology, Peoples’ Friendship University of Russia (RUDN), 6 Miklukho-Maklaya Street, 117198 Moscow, Russia
- Department of Microbiology, Faculty of Pharmacy, Tanta University, Tanta 31111, Egypt
| | - Vitaliy V. Kushnirov
- Federal Research Center of Biotechnology of the RAS, Bach Institute of Biochemistry, 119071 Moscow, Russia; (V.A.B.); (O.V.M.); (E.S.M.O.G.); (V.V.K.)
| | - Alexander I. Alexandrov
- Federal Research Center of Biotechnology of the RAS, Bach Institute of Biochemistry, 119071 Moscow, Russia; (V.A.B.); (O.V.M.); (E.S.M.O.G.); (V.V.K.)
| |
Collapse
|
8
|
Zimmermann A, Tadic J, Kainz K, Hofer SJ, Bauer MA, Carmona-Gutierrez D, Madeo F. Transcriptional and epigenetic control of regulated cell death in yeast. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2020; 352:55-82. [PMID: 32334817 DOI: 10.1016/bs.ircmb.2019.12.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Unicellular organisms like yeast can undergo controlled demise in a manner that is partly reminiscent of mammalian cell death. This is true at the levels of both mechanistic and functional conservation. Yeast offers the combination of unparalleled genetic amenability and a comparatively simple biology to understand both the regulation and evolution of cell death. In this minireview, we address the capacity of the nucleus as a regulatory hub during yeast regulated cell death (RCD), which is becoming an increasingly central question in yeast RCD research. In particular, we explore and critically discuss the available data on stressors and signals that specifically impinge on the nucleus. Moreover, we also analyze the current knowledge on nuclear factors as well as on transcriptional control and epigenetic events that orchestrate yeast RCD. Altogether we conclude that the functional significance of the nucleus for yeast RCD in undisputable, but that further exploration beyond correlative work is necessary to disentangle the role of nuclear events in the regulatory network.
Collapse
Affiliation(s)
- Andreas Zimmermann
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
| | - Jelena Tadic
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria; Division of Immunology and Pathophysiology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Katharina Kainz
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
| | - Sebastian J Hofer
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
| | - Maria A Bauer
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
| | | | - Frank Madeo
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria; BioTechMed Graz, Graz, Austria.
| |
Collapse
|
9
|
Hu J, Dong Y, Wang W, Zhang W, Lou H, Chen Q. Deletion of Atg22 gene contributes to reduce programmed cell death induced by acetic acid stress in Saccharomyces cerevisiae. BIOTECHNOLOGY FOR BIOFUELS 2019; 12:298. [PMID: 31890026 PMCID: PMC6933646 DOI: 10.1186/s13068-019-1638-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 12/12/2019] [Indexed: 05/27/2023]
Abstract
BACKGROUND Programmed cell death (PCD) induced by acetic acid, the main by-product released during cellulosic hydrolysis, cast a cloud over lignocellulosic biofuel fermented by Saccharomyces cerevisiae and became a burning problem. Atg22p, an ignored integral membrane protein located in vacuole belongs to autophagy-related genes family; prior study recently reported that it is required for autophagic degradation and efflux of amino acids from vacuole to cytoplasm. It may alleviate the intracellular starvation of nutrition caused by Ac and increase cell tolerance. Therefore, we investigate the role of atg22 in cell death process induced by Ac in which attempt is made to discover new perspectives for better understanding of the mechanisms behind tolerance and more robust industrial strain construction. RESULTS In this study, we compared cell growth, physiological changes in the absence and presence of Atg22p under Ac exposure conditions. It is observed that disruption and overexpression of Atg22p delays and enhances acetic acid-induced PCD, respectively. The deletion of Atg22p in S. cerevisiae maintains cell wall integrity, and protects cytomembrane integrity, fluidity and permeability upon Ac stress by changing cytomembrane phospholipids, sterols and fatty acids. More interestingly, atg22 deletion increases intracellular amino acids to aid yeast cells for tackling amino acid starvation and intracellular acidification. Further, atg22 deletion upregulates series of stress response genes expression such as heat shock protein family, cell wall integrity and autophagy. CONCLUSIONS The findings show that Atg22p possessed the new function related to cell resistance to Ac. This may help us have a deeper understanding of PCD induced by Ac and provide a new strategy to improve Ac resistance in designing industrial yeast strains for bioethanol production during lignocellulosic biofuel fermentation.
Collapse
Affiliation(s)
- Jingjin Hu
- Department of Food Science and Nutrition, Key Laboratory for Food Microbial Technology of Zhejiang Province, Zhejiang University, Hangzhou, 310058 China
| | - Yachen Dong
- Department of Food Science and Nutrition, Key Laboratory for Food Microbial Technology of Zhejiang Province, Zhejiang University, Hangzhou, 310058 China
| | - Wei Wang
- Institute of Quality and Standard for Agriculture Products, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021 China
| | - Wei Zhang
- Department of Cardiovascular & Metabolic Sciences, The Lerner Research Institute, Cleveland Clinic, Cleveland, OH USA
| | - Hanghang Lou
- Department of Food Science and Nutrition, Key Laboratory for Food Microbial Technology of Zhejiang Province, Zhejiang University, Hangzhou, 310058 China
| | - Qihe Chen
- Department of Food Science and Nutrition, Key Laboratory for Food Microbial Technology of Zhejiang Province, Zhejiang University, Hangzhou, 310058 China
| |
Collapse
|
10
|
Porras-Agüera JA, Moreno-García J, Mauricio JC, Moreno J, García-Martínez T. First Proteomic Approach to Identify Cell Death Biomarkers in Wine Yeasts during Sparkling Wine Production. Microorganisms 2019; 7:microorganisms7110542. [PMID: 31717411 PMCID: PMC6920952 DOI: 10.3390/microorganisms7110542] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 10/22/2019] [Accepted: 11/07/2019] [Indexed: 12/22/2022] Open
Abstract
Apoptosis and later autolysis are biological processes which take place in Saccharomyces cerevisiae during industrial fermentation processes, which involve costly and time-consuming aging periods. Therefore, the identification of potential cell death biomarkers can contribute to the creation of a long-term strategy in order to improve and accelerate the winemaking process. Here, we performed a proteomic analysis based on the detection of possible apoptosis and autolysis protein biomarkers in two industrial yeast strains commonly used in post-fermentative processes (sparkling wine secondary fermentation and biological aging) under typical sparkling wine elaboration conditions. Pressure had a negatively effect on viability for flor yeast, whereas the sparkling wine strain seems to be more adapted to these conditions. Flor yeast strain experienced an increase in content of apoptosis-related proteins, glucanases and vacuolar proteases at the first month of aging. Significant correlations between viability and apoptosis proteins were established in both yeast strains. Multivariate analysis based on the proteome of each process allowed to distinguish among samples and strains. The proteomic profile obtained in this study could provide useful information on the selection of wine strains and yeast behavior during sparkling wine elaboration. Additionally, the use of flor yeasts for sparkling wine improvement and elaboration is proposed.
Collapse
Affiliation(s)
- Juan Antonio Porras-Agüera
- Department of Microbiology, Agrifood Campus of International Excellence ceiA3, University of Cordoba, 14014 Cordoba, Spain; (J.A.P.-A.); (J.M.-G.); (T.G.-M.)
| | - Jaime Moreno-García
- Department of Microbiology, Agrifood Campus of International Excellence ceiA3, University of Cordoba, 14014 Cordoba, Spain; (J.A.P.-A.); (J.M.-G.); (T.G.-M.)
| | - Juan Carlos Mauricio
- Department of Microbiology, Agrifood Campus of International Excellence ceiA3, University of Cordoba, 14014 Cordoba, Spain; (J.A.P.-A.); (J.M.-G.); (T.G.-M.)
- Correspondence: ; Tel.: +34-957-218640; Fax: +34-957-218650
| | - Juan Moreno
- Department of Agricultural Chemistry, Agrifood Campus of International Excellence ceiA3, University of Cordoba, 14014 Cordoba, Spain;
| | - Teresa García-Martínez
- Department of Microbiology, Agrifood Campus of International Excellence ceiA3, University of Cordoba, 14014 Cordoba, Spain; (J.A.P.-A.); (J.M.-G.); (T.G.-M.)
| |
Collapse
|
11
|
Lu H, Shu Q, Lou H, Chen Q. Mitochondria-Mediated Programmed Cell Death in Saccharomyces cerevisiae Induced by Betulinic Acid Is Accelerated by the Deletion of PEP4 Gene. Microorganisms 2019; 7:microorganisms7110538. [PMID: 31703462 PMCID: PMC6920885 DOI: 10.3390/microorganisms7110538] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Revised: 10/27/2019] [Accepted: 11/05/2019] [Indexed: 11/22/2022] Open
Abstract
In this work, using Saccharomyces cerevisiae as a model, we showed that BetA could inhibit cell proliferation and lead to lethal cytotoxicity accompanying programmed cell death (PCD). Interestingly, it was found that vacuolar protease Pep4p played a pivotal role in BetA-induced S. cerevisiae PCD. The presence of Pep4p reduced the damage of BetA-induced cells. This work implied that BetA may induce cell death of S. cerevisiae through mitochondria-mediated PCD, and the deletion of Pep4 gene possibly accelerated the effect of PCD. The present investigation provided the preliminary research for the complicated mechanism of BetA-induced cell PCD regulated by vacular protease Pep4p and lay the foundation for understanding of the Pep4p protein in an animal model.
Collapse
Affiliation(s)
| | | | | | - Qihe Chen
- Correspondence: ; Tel.: +86-0571-8698-4316
| |
Collapse
|
12
|
Cellular models of Batten disease. Biochim Biophys Acta Mol Basis Dis 2019; 1866:165559. [PMID: 31655107 PMCID: PMC7338907 DOI: 10.1016/j.bbadis.2019.165559] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 09/05/2019] [Accepted: 09/13/2019] [Indexed: 12/22/2022]
Abstract
The Neuronal Ceroid Lipofuscinoses (NCL), otherwise known as Batten disease, are a group of neurodegenerative diseases caused by mutations in 13 known genes. All except one NCL is autosomal recessive in inheritance, with similar aetiology and characterised by the accumulation of autofluorescent storage material in the lysosomes of cells. Age of onset and the rate of progression vary between the NCLs. They are collectively one of the most common lysosomal storage diseases, but the enigma remains of how genetically distinct diseases result in such remarkably similar pathogenesis. Much has been learnt from cellular studies about the function of the proteins encoded by the affected genes. Such research has utilised primitive unicellular models such as yeast and amoeba containing gene orthologues, cells derived from naturally occurring (sheep) and genetically engineered (mouse) animal models or patient-derived cells. Most recently, patient-derived induced pluripotent stem cell (iPSC) lines have been differentiated into neural cell-types to study molecular pathogenesis in the cells most profoundly affected by disease. Here, we review how cell models have informed much of the biochemical understanding of the NCLs and how more complex models are being used to further this understanding and potentially act as platforms for therapeutic efficacy studies in the future. Developments made in cellular models for neuronal ceroid lipofuscinosis (NCL) in basic biology and use as therapeutic platforms. Cellular models elucidating function of NCL proteins. NCL proteins implicated in the mTor signalling pathway. Patient-derived induced pluripotent stem cell (iPSC) lines have been differentiated into neural cell-types providing insights into the molecular pathogenesis of NCL.
Collapse
|
13
|
Hu J, Yu L, Shu Q, Chen Q. Identification of Down-Regulated Proteome in Saccharomyces cerevisiae with the Deletion of Yeast Cathepsin D in Response to Nitrogen Stress. Microorganisms 2019; 7:microorganisms7080214. [PMID: 31344930 PMCID: PMC6723583 DOI: 10.3390/microorganisms7080214] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 07/17/2019] [Accepted: 07/23/2019] [Indexed: 11/16/2022] Open
Abstract
Vacuolar proteinase A (Pep4p) is required for the post-translational precursor maturation of vacuolar proteinases in Saccharomyces cerevisiae, and important for protein turnover after oxidative damage. The presence of proteinase A in brewing yeast leads to the decline of beer foam stability, thus the deletion or inhibition of Pep4p is generally used. However, the influence of Pep4p deletion on cell metabolism in Saccharomyces cerevisiae is still unclear. Herein, we report the identification of differentially down-regulated metabolic proteins in the absence of Pep4p by a comparative proteomics approach. 2D-PAGE (two-dimensional polyacrylamide gel electrophoresis) presented that the number of significantly up-regulated spots (the Pep4p-deficient species versus the wild type) was 183, whereas the down-regulated spots numbered 111. Among them, 35 identified proteins were differentially down-regulated more than 10-fold in the Pep4p-deficient compared to the wild-type species. The data revealed that Pep4p was required for the synthesis and maturation of several glycolytic enzymes and stress proteins, including Eno2p, Fba1p, Pdc1p, Tpi1p, Ssa1, Hsp82p, and Trr1p. The transcription and post-translational modifications of glycolytic enzymes like Eno2p and Fba1p were sensitive to the absence of Pep4p; whereas the depletion of the pep4 gene had a negative impact on mitochondrial and other physiological functions. The finding of this study provides a systematic understanding that Pep4p may serve as a regulating factor for cell physiology and metabolic processes in S. cerevisiae under a nitrogen stress environment.
Collapse
Affiliation(s)
- Jingjin Hu
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou 310058, China
| | - Lingxiao Yu
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou 310058, China
| | - Qin Shu
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou 310058, China
| | - Qihe Chen
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
14
|
Aufschnaiter A, Büttner S. The vacuolar shapes of ageing: From function to morphology. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1866:957-970. [PMID: 30796938 DOI: 10.1016/j.bbamcr.2019.02.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 02/13/2019] [Accepted: 02/15/2019] [Indexed: 12/21/2022]
Abstract
Cellular ageing results in accumulating damage to various macromolecules and the progressive decline of organelle function. Yeast vacuoles as well as their counterpart in higher eukaryotes, the lysosomes, emerge as central organelles in lifespan determination. These acidic organelles integrate enzymatic breakdown and recycling of cellular waste with nutrient sensing, storage, signalling and mobilization. Establishing physical contact with virtually all other organelles, vacuoles serve as hubs of cellular homeostasis. Studies in Saccharomyces cerevisiae contributed substantially to our understanding of the ageing process per se and the multifaceted roles of vacuoles/lysosomes in the maintenance of cellular fitness with progressing age. Here, we discuss the multiple roles of the vacuole during ageing, ranging from vacuolar dynamics and acidification as determinants of lifespan to the function of this organelle as waste bin, recycling facility, nutrient reservoir and integrator of nutrient signalling.
Collapse
Affiliation(s)
- Andreas Aufschnaiter
- Institute of Molecular Biosciences, University of Graz, Humboldtstraße 50, 8010 Graz, Austria
| | - Sabrina Büttner
- Institute of Molecular Biosciences, University of Graz, Humboldtstraße 50, 8010 Graz, Austria; Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Svante Arrhenius väg 20C, 106 91 Stockholm, Sweden.
| |
Collapse
|
15
|
Carmona-Gutierrez D, Zimmermann A, Kainz K, Pietrocola F, Chen G, Maglioni S, Schiavi A, Nah J, Mertel S, Beuschel CB, Castoldi F, Sica V, Trausinger G, Raml R, Sommer C, Schroeder S, Hofer SJ, Bauer MA, Pendl T, Tadic J, Dammbrueck C, Hu Z, Ruckenstuhl C, Eisenberg T, Durand S, Bossut N, Aprahamian F, Abdellatif M, Sedej S, Enot DP, Wolinski H, Dengjel J, Kepp O, Magnes C, Sinner F, Pieber TR, Sadoshima J, Ventura N, Sigrist SJ, Kroemer G, Madeo F. The flavonoid 4,4'-dimethoxychalcone promotes autophagy-dependent longevity across species. Nat Commun 2019; 10:651. [PMID: 30783116 PMCID: PMC6381180 DOI: 10.1038/s41467-019-08555-w] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 01/11/2019] [Indexed: 01/08/2023] Open
Abstract
Ageing constitutes the most important risk factor for all major chronic ailments, including malignant, cardiovascular and neurodegenerative diseases. However, behavioural and pharmacological interventions with feasible potential to promote health upon ageing remain rare. Here we report the identification of the flavonoid 4,4'-dimethoxychalcone (DMC) as a natural compound with anti-ageing properties. External DMC administration extends the lifespan of yeast, worms and flies, decelerates senescence of human cell cultures, and protects mice from prolonged myocardial ischaemia. Concomitantly, DMC induces autophagy, which is essential for its cytoprotective effects from yeast to mice. This pro-autophagic response induces a conserved systemic change in metabolism, operates independently of TORC1 signalling and depends on specific GATA transcription factors. Notably, we identify DMC in the plant Angelica keiskei koidzumi, to which longevity- and health-promoting effects are ascribed in Asian traditional medicine. In summary, we have identified and mechanistically characterised the conserved longevity-promoting effects of a natural anti-ageing drug.
Collapse
Affiliation(s)
| | - Andreas Zimmermann
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, 8010, Austria
- Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, Graz, 8036, Austria
| | - Katharina Kainz
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, 8010, Austria
| | - Federico Pietrocola
- Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, INSERM U 1138, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Center, Villejuif, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
- Université Pierre et Marie Curie, Paris, France
| | - Guo Chen
- Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, INSERM U 1138, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Center, Villejuif, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
- Université Pierre et Marie Curie, Paris, France
| | - Silvia Maglioni
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, 40225, Germany
| | - Alfonso Schiavi
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, 40225, Germany
| | - Jihoon Nah
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Sara Mertel
- Institute for Biology/Genetics, Freie Universität Berlin, Berlin, 14195, Germany
| | - Christine B Beuschel
- Institute for Biology/Genetics, Freie Universität Berlin, Berlin, 14195, Germany
| | - Francesca Castoldi
- Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, INSERM U 1138, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Center, Villejuif, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
- Université Pierre et Marie Curie, Paris, France
- Sotio a.c, 17000, Prague, Czech Republic
| | - Valentina Sica
- Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, INSERM U 1138, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Center, Villejuif, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
- Université Pierre et Marie Curie, Paris, France
| | - Gert Trausinger
- Joanneum Research Forschungsgesellschaft m.b.H., HEALTH, Institute for Biomedicine and Health Sciences, Graz, 8010, Austria
| | - Reingard Raml
- Joanneum Research Forschungsgesellschaft m.b.H., HEALTH, Institute for Biomedicine and Health Sciences, Graz, 8010, Austria
| | - Cornelia Sommer
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, 8010, Austria
| | - Sabrina Schroeder
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, 8010, Austria
| | - Sebastian J Hofer
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, 8010, Austria
| | - Maria A Bauer
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, 8010, Austria
| | - Tobias Pendl
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, 8010, Austria
| | - Jelena Tadic
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, 8010, Austria
| | | | - Zehan Hu
- Department of Cardiology, Medical University of Graz, Graz, 8036, Austria
| | - Christoph Ruckenstuhl
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, 8010, Austria
| | - Tobias Eisenberg
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, 8010, Austria
| | - Sylvere Durand
- Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, INSERM U 1138, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Center, Villejuif, France
| | - Noélie Bossut
- Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, INSERM U 1138, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Center, Villejuif, France
| | - Fanny Aprahamian
- Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, INSERM U 1138, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Center, Villejuif, France
| | - Mahmoud Abdellatif
- Department of Cardiology, Medical University of Graz, Graz, 8036, Austria
| | - Simon Sedej
- Department of Cardiology, Medical University of Graz, Graz, 8036, Austria
- BioTechMed Graz, Graz, 8010, Austria
| | - David P Enot
- Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, INSERM U 1138, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Center, Villejuif, France
| | - Heimo Wolinski
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, 8010, Austria
| | - Jörn Dengjel
- Department of Biology, Université de Fribourg, Chemin du Musée 10, 1700, Fribourg, Switzerland
| | - Oliver Kepp
- Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, INSERM U 1138, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Center, Villejuif, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
- Université Pierre et Marie Curie, Paris, France
| | - Christoph Magnes
- Joanneum Research Forschungsgesellschaft m.b.H., HEALTH, Institute for Biomedicine and Health Sciences, Graz, 8010, Austria
| | - Frank Sinner
- Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, Graz, 8036, Austria
- Joanneum Research Forschungsgesellschaft m.b.H., HEALTH, Institute for Biomedicine and Health Sciences, Graz, 8010, Austria
| | - Thomas R Pieber
- Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, Graz, 8036, Austria
- Joanneum Research Forschungsgesellschaft m.b.H., HEALTH, Institute for Biomedicine and Health Sciences, Graz, 8010, Austria
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Natascia Ventura
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, 40225, Germany
- Institute for Clinical Chemistry and Laboratory Diagnostic, Medical Faculty of the Heinrich Heine University, Moorenstrasse 5, 40225, Düsseldorf, Germany
| | - Stephan J Sigrist
- Institute for Biology/Genetics, Freie Universität Berlin, Berlin, 14195, Germany
- NeuroCure, Charité, Berlin, 10117, Germany
| | - Guido Kroemer
- Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, INSERM U 1138, Paris, France.
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Center, Villejuif, France.
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France.
- Université Pierre et Marie Curie, Paris, France.
- Pôle de Biologie, Hôpital Européen Georges Pompidou, Paris, France.
- Karolinska Institute, Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden.
| | - Frank Madeo
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, 8010, Austria.
- BioTechMed Graz, Graz, 8010, Austria.
| |
Collapse
|
16
|
Kerstens W, Van Dijck P. A Cinderella story: how the vacuolar proteases Pep4 and Prb1 do more than cleaning up the cell's mass degradation processes. MICROBIAL CELL 2018; 5:438-443. [PMID: 30386788 PMCID: PMC6206407 DOI: 10.15698/mic2018.10.650] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Recently, several research groups have assigned non-vacuolar functions to the well-known Saccharomyces cerevisiae vacuolar proteases Pep4 and Prb1, which are also known as proteinases A and B. These non-vacuolar activities seem to be autophagy-independent and stress-induced and suggest an unexplored but possibly prominent role for the proteases outside the vacuole. The functions range from the involvement in programmed cell death, to protection from hazardous protein forms and regulation of gene expression. We propose that a deeper understanding of these molecular processes will provide new insights that will be important for both fungal biology as well as studies in mammalian cells, as they might open up perspectives in the search for novel drug targets. To illustrate this, we summarize the recent literature on non-vacuolar Pep4 and Prb1 functions in S. cerevisiae and review the current data on the protein homologs in pathogenic fungi.
Collapse
Affiliation(s)
- Winnie Kerstens
- VIB-KU Leuven Center for Microbiology, Kasteelpark Arenberg 31, B-3001 Leuven, Belgium.,Laboratory of Molecular Cell Biology, Institute of Botany and Microbiology, KU Leuven, Kasteelpark Arenberg 31, B-3001 Leuven, Belgium
| | - Patrick Van Dijck
- VIB-KU Leuven Center for Microbiology, Kasteelpark Arenberg 31, B-3001 Leuven, Belgium.,Laboratory of Molecular Cell Biology, Institute of Botany and Microbiology, KU Leuven, Kasteelpark Arenberg 31, B-3001 Leuven, Belgium
| |
Collapse
|
17
|
Juárez-Montiel M, Tesillo-Moreno P, Cruz-Angeles A, Soberanes-Gutiérrez V, Chávez-Camarillo G, Ibarra JA, Hernández-Rodríguez C, Villa-Tanaca L. Heterologous expression and characterization of the aspartic endoprotease Pep4um from Ustilago maydis, a homolog of the human Chatepsin D, an important breast cancer therapeutic target. Mol Biol Rep 2018; 45:1155-1163. [PMID: 30076522 DOI: 10.1007/s11033-018-4267-8] [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: 06/05/2018] [Accepted: 07/16/2018] [Indexed: 10/28/2022]
Abstract
The pep4um gene (um04926) of Ustilago maydis encodes a protein related to either vacuolar or lysosomal aspartic proteases. Bioinformatic analysis of the Pep4um protein revealed that it is a soluble protein with a signal peptide suggesting that it likely passes through the secretory pathway, and it has two probable self-activation sites, which are similar to those in Saccharomyces cerevisiae PrA. Moreover, the active site of the Pep4um has the two characteristic aspartic acid residues of aspartyl proteases. The pep4um gene was cloned, expressed in Pichia pastoris and a 54 kDa recombinant protein was observed. Pep4um-rec was confirmed to be an aspartic protease by specifically inhibiting its enzymatic activity with pepstatin A. Pep4um-rec enzymatic activity on acidic hemoglobin was optimal at pH 4.0 and at 40 °C. To the best of our knowledge this is the first report about the heterologous expression of an aspartic protease from a basidiomycete. An in-depth in silico analysis suggests that Pep4um is homolog of the human cathepsin D protein. Thus, the Pep4um-rec protein may be used to test inhibitors of human cathepsin D, an important breast cancer therapeutic target.
Collapse
Affiliation(s)
- Margarita Juárez-Montiel
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, DF, Mexico.,Laboratorio de Biología Molecular de Bacterias y Levaduras, Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas del Instituto Politécnico Nacional, Plan de Ayala y Prol. Carpio. Col. Casco de Santo Tomás, Mexico City, DF, CP 11340, Mexico
| | - Pedro Tesillo-Moreno
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, DF, Mexico.,Laboratorio de Biología Molecular de Bacterias y Levaduras, Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas del Instituto Politécnico Nacional, Plan de Ayala y Prol. Carpio. Col. Casco de Santo Tomás, Mexico City, DF, CP 11340, Mexico
| | - Ana Cruz-Angeles
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, DF, Mexico
| | - Valentina Soberanes-Gutiérrez
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, DF, Mexico
| | - Griselda Chávez-Camarillo
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, DF, Mexico
| | - J Antonio Ibarra
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, DF, Mexico
| | - César Hernández-Rodríguez
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, DF, Mexico.,Laboratorio de Biología Molecular de Bacterias y Levaduras, Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas del Instituto Politécnico Nacional, Plan de Ayala y Prol. Carpio. Col. Casco de Santo Tomás, Mexico City, DF, CP 11340, Mexico
| | - Lourdes Villa-Tanaca
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, DF, Mexico. .,Laboratorio de Biología Molecular de Bacterias y Levaduras, Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas del Instituto Politécnico Nacional, Plan de Ayala y Prol. Carpio. Col. Casco de Santo Tomás, Mexico City, DF, CP 11340, Mexico.
| |
Collapse
|
18
|
Meng MB, Wang HH, Cui YL, Wu ZQ, Shi YY, Zaorsky NG, Deng L, Yuan ZY, Lu Y, Wang P. Necroptosis in tumorigenesis, activation of anti-tumor immunity, and cancer therapy. Oncotarget 2018; 7:57391-57413. [PMID: 27429198 PMCID: PMC5302997 DOI: 10.18632/oncotarget.10548] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 06/20/2016] [Indexed: 02/05/2023] Open
Abstract
While the mechanisms underlying apoptosis and autophagy have been well characterized over recent decades, another regulated cell death event, necroptosis, remains poorly understood. Elucidating the signaling networks involved in the regulation of necroptosis may allow this form of regulated cell death to be exploited for diagnosis and treatment of cancer, and will contribute to the understanding of the complex tumor microenvironment. In this review, we have summarized the mechanisms and regulation of necroptosis, the converging and diverging features of necroptosis in tumorigenesis, activation of anti-tumor immunity, and cancer therapy, as well as attempts to exploit this newly gained knowledge to provide therapeutics for cancer.
Collapse
Affiliation(s)
- Mao-Bin Meng
- Department of Radiation Oncology, Tianjin's Clinical Research Center for Cancer and Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Huan-Huan Wang
- Department of Radiation Oncology, Tianjin's Clinical Research Center for Cancer and Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Yao-Li Cui
- Department of Lymphoma, Tianjin's Clinical Research Center for Cancer and Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Zhi-Qiang Wu
- Department of Radiation Oncology, Tianjin's Clinical Research Center for Cancer and Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Yang-Yang Shi
- Stanford University School of Medicine, Stanford, CA, United States of America
| | - Nicholas G Zaorsky
- Department of Radiation Oncology, Fox Chase Cancer Center, Philadelphia, PA, United States of America
| | - Lei Deng
- Department of Thoracic Cancer and Huaxi Student Society of Oncology Research, West China Hospital, West China School of Medicine, Sichuan University, Sichuan Province, China
| | - Zhi-Yong Yuan
- Department of Radiation Oncology, Tianjin's Clinical Research Center for Cancer and Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - You Lu
- Department of Thoracic Cancer and Huaxi Student Society of Oncology Research, West China Hospital, West China School of Medicine, Sichuan University, Sichuan Province, China
| | - Ping Wang
- Department of Radiation Oncology, Tianjin's Clinical Research Center for Cancer and Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China
| |
Collapse
|
19
|
Carmona-Gutierrez D, Bauer MA, Zimmermann A, Aguilera A, Austriaco N, Ayscough K, Balzan R, Bar-Nun S, Barrientos A, Belenky P, Blondel M, Braun RJ, Breitenbach M, Burhans WC, Büttner S, Cavalieri D, Chang M, Cooper KF, Côrte-Real M, Costa V, Cullin C, Dawes I, Dengjel J, Dickman MB, Eisenberg T, Fahrenkrog B, Fasel N, Fröhlich KU, Gargouri A, Giannattasio S, Goffrini P, Gourlay CW, Grant CM, Greenwood MT, Guaragnella N, Heger T, Heinisch J, Herker E, Herrmann JM, Hofer S, Jiménez-Ruiz A, Jungwirth H, Kainz K, Kontoyiannis DP, Ludovico P, Manon S, Martegani E, Mazzoni C, Megeney LA, Meisinger C, Nielsen J, Nyström T, Osiewacz HD, Outeiro TF, Park HO, Pendl T, Petranovic D, Picot S, Polčic P, Powers T, Ramsdale M, Rinnerthaler M, Rockenfeller P, Ruckenstuhl C, Schaffrath R, Segovia M, Severin FF, Sharon A, Sigrist SJ, Sommer-Ruck C, Sousa MJ, Thevelein JM, Thevissen K, Titorenko V, Toledano MB, Tuite M, Vögtle FN, Westermann B, Winderickx J, Wissing S, Wölfl S, Zhang ZJ, Zhao RY, Zhou B, Galluzzi L, Kroemer G, Madeo F. Guidelines and recommendations on yeast cell death nomenclature. MICROBIAL CELL (GRAZ, AUSTRIA) 2018; 5:4-31. [PMID: 29354647 PMCID: PMC5772036 DOI: 10.15698/mic2018.01.607] [Citation(s) in RCA: 130] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 12/29/2017] [Indexed: 12/18/2022]
Abstract
Elucidating the biology of yeast in its full complexity has major implications for science, medicine and industry. One of the most critical processes determining yeast life and physiology is cel-lular demise. However, the investigation of yeast cell death is a relatively young field, and a widely accepted set of concepts and terms is still missing. Here, we propose unified criteria for the defi-nition of accidental, regulated, and programmed forms of cell death in yeast based on a series of morphological and biochemical criteria. Specifically, we provide consensus guidelines on the differ-ential definition of terms including apoptosis, regulated necrosis, and autophagic cell death, as we refer to additional cell death rou-tines that are relevant for the biology of (at least some species of) yeast. As this area of investigation advances rapidly, changes and extensions to this set of recommendations will be implemented in the years to come. Nonetheless, we strongly encourage the au-thors, reviewers and editors of scientific articles to adopt these collective standards in order to establish an accurate framework for yeast cell death research and, ultimately, to accelerate the pro-gress of this vibrant field of research.
Collapse
Affiliation(s)
| | - Maria Anna Bauer
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
| | - Andreas Zimmermann
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
| | - Andrés Aguilera
- Centro Andaluz de Biología, Molecular y Medicina Regenerativa-CABIMER, Universidad de Sevilla, Sevilla, Spain
| | | | - Kathryn Ayscough
- Department of Biomedical Science, University of Sheffield, Sheffield, United Kingdom
| | - Rena Balzan
- Department of Physiology and Biochemistry, University of Malta, Msida, Malta
| | - Shoshana Bar-Nun
- Department of Biochemistry and Molecular Biology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Antonio Barrientos
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, USA
- Department of Neurology, University of Miami Miller School of Medi-cine, Miami, USA
| | - Peter Belenky
- Department of Molecular Microbiology and Immunology, Brown University, Providence, USA
| | - Marc Blondel
- Institut National de la Santé et de la Recherche Médicale UMR1078, Université de Bretagne Occidentale, Etablissement Français du Sang Bretagne, CHRU Brest, Hôpital Morvan, Laboratoire de Génétique Moléculaire, Brest, France
| | - Ralf J. Braun
- Institute of Cell Biology, University of Bayreuth, Bayreuth, Germany
| | | | - William C. Burhans
- Department of Molecular and Cellular Biology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Sabrina Büttner
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | | | - Michael Chang
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Katrina F. Cooper
- Dept. Molecular Biology, Graduate School of Biomedical Sciences, Rowan University, Stratford, USA
| | - Manuela Côrte-Real
- Center of Molecular and Environmental Biology, Department of Biology, University of Minho, Braga, Portugal
| | - Vítor Costa
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- Departamento de Biologia Molecular, Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | | | - Ian Dawes
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia
| | - Jörn Dengjel
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Martin B. Dickman
- Institute for Plant Genomics and Biotechnology, Texas A&M University, Texas, USA
| | - Tobias Eisenberg
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
| | - Birthe Fahrenkrog
- Laboratory Biology of the Nucleus, Institute for Molecular Biology and Medicine, Université Libre de Bruxelles, Charleroi, Belgium
| | - Nicolas Fasel
- Department of Biochemistry, University of Lausanne, Lausanne, Switzerland
| | - Kai-Uwe Fröhlich
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
| | - Ali Gargouri
- Laboratoire de Biotechnologie Moléculaire des Eucaryotes, Center de Biotechnologie de Sfax, Sfax, Tunisia
| | - Sergio Giannattasio
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Research Council, Bari, Italy
| | - Paola Goffrini
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
| | - Campbell W. Gourlay
- Kent Fungal Group, School of Biosciences, University of Kent, Canterbury, United Kingdom
| | - Chris M. Grant
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Michael T. Greenwood
- Department of Chemistry and Chemical Engineering, Royal Military College, Kingston, Ontario, Canada
| | - Nicoletta Guaragnella
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Research Council, Bari, Italy
| | | | - Jürgen Heinisch
- Department of Biology and Chemistry, University of Osnabrück, Osnabrück, Germany
| | - Eva Herker
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | | | - Sebastian Hofer
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
| | | | - Helmut Jungwirth
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
| | - Katharina Kainz
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
| | - Dimitrios P. Kontoyiannis
- Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Paula Ludovico
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Minho, Portugal
- ICVS/3B’s - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Stéphen Manon
- Institut de Biochimie et de Génétique Cellulaires, UMR5095, CNRS & Université de Bordeaux, Bordeaux, France
| | - Enzo Martegani
- Department of Biotechnolgy and Biosciences, University of Milano-Bicocca, Milano, Italy
| | - Cristina Mazzoni
- Instituto Pasteur-Fondazione Cenci Bolognetti - Department of Biology and Biotechnology "C. Darwin", La Sapienza University of Rome, Rome, Italy
| | - Lynn A. Megeney
- Sprott Center for Stem Cell Research, Ottawa Hospital Research Institute, The Ottawa Hospital, Ottawa, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada
- Department of Medicine, Division of Cardiology, University of Ottawa, Ottawa, Canada
| | - Chris Meisinger
- Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Jens Nielsen
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
- Novo Nordisk Foundation Center for Biosustainability, Chalmers University of Technology, Gothenburg, Sweden
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, DK2800 Lyngby, Denmark
| | - Thomas Nyström
- Institute for Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Heinz D. Osiewacz
- Institute for Molecular Biosciences, Goethe University, Frankfurt am Main, Germany
| | - Tiago F. Outeiro
- Department of Experimental Neurodegeneration, Center for Nanoscale Microscopy and Molecular Physiology of the Brain, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany
- Max Planck Institute for Experimental Medicine, Göttingen, Germany
- Institute of Neuroscience, The Medical School, Newcastle University, Framlington Place, Newcastle Upon Tyne, NE2 4HH, United Kingdom
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Hay-Oak Park
- Department of Molecular Genetics, The Ohio State University, Columbus, OH, USA
| | - Tobias Pendl
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
| | - Dina Petranovic
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
- Novo Nordisk Foundation Center for Biosustainability, Chalmers University of Technology, Gothenburg, Sweden
| | - Stephane Picot
- Malaria Research Unit, SMITh, ICBMS, UMR 5246 CNRS-INSA-CPE-University Lyon, Lyon, France
- Institut of Parasitology and Medical Mycology, Hospices Civils de Lyon, Lyon, France
| | - Peter Polčic
- Department of Biochemistry, Faculty of Natural Sciences, Comenius University in Bratislava, Bratislava, Slovak Republic
| | - Ted Powers
- Department of Molecular and Cellular Biology, College of Biological Sciences, UC Davis, Davis, California, USA
| | - Mark Ramsdale
- Biosciences, University of Exeter, Exeter, United Kingdom
| | - Mark Rinnerthaler
- Department of Cell Biology and Physiology, Division of Genetics, University of Salzburg, Salzburg, Austria
| | - Patrick Rockenfeller
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
- Kent Fungal Group, School of Biosciences, University of Kent, Canterbury, United Kingdom
| | | | - Raffael Schaffrath
- Institute of Biology, Division of Microbiology, University of Kassel, Kassel, Germany
| | - Maria Segovia
- Department of Ecology, Faculty of Sciences, University of Malaga, Malaga, Spain
| | - Fedor F. Severin
- A.N. Belozersky Institute of physico-chemical biology, Moscow State University, Moscow, Russia
| | - Amir Sharon
- School of Plant Sciences and Food Security, Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Stephan J. Sigrist
- Institute for Biology/Genetics, Freie Universität Berlin, Berlin, Germany
| | - Cornelia Sommer-Ruck
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
| | - Maria João Sousa
- Center of Molecular and Environmental Biology, Department of Biology, University of Minho, Braga, Portugal
| | - Johan M. Thevelein
- Laboratory of Molecular Cell Biology, Institute of Botany and Microbiology, KU Leuven, Leuven, Belgium
- Center for Microbiology, VIB, Leuven-Heverlee, Belgium
| | - Karin Thevissen
- Centre of Microbial and Plant Genetics, KU Leuven, Leuven, Belgium
| | | | - Michel B. Toledano
- Institute for Integrative Biology of the Cell (I2BC), SBIGEM, CEA-Saclay, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Mick Tuite
- Kent Fungal Group, School of Biosciences, University of Kent, Canterbury, United Kingdom
| | - F.-Nora Vögtle
- Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | | | - Joris Winderickx
- Department of Biology, Functional Biology, KU Leuven, Leuven-Heverlee, Belgium
| | | | - Stefan Wölfl
- Institute of Pharmacy and Molecu-lar Biotechnology, Heidelberg University, Heidelberg, Germany
| | - Zhaojie J. Zhang
- Department of Zoology and Physiology, University of Wyoming, Laramie, USA
| | - Richard Y. Zhao
- Department of Pathology, University of Maryland School of Medicine, Baltimore, USA
| | - Bing Zhou
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
- Université Paris Descartes/Paris V, Paris, France
| | - Guido Kroemer
- Université Paris Descartes/Paris V, Paris, France
- Equipe 11 Labellisée Ligue Contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- Cell Biology and Metabolomics Platforms, Gustave Roussy Comprehensive Cancer Center, Villejuif, France
- INSERM, U1138, Paris, France
- Université Pierre et Marie Curie/Paris VI, Paris, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou, Paris, France
- Institute, Department of Women’s and Children’s Health, Karolinska University Hospital, Stockholm, Sweden
| | - Frank Madeo
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
| |
Collapse
|
20
|
Quercetin Protects Yeast Saccharomyces cerevisiae pep4 Mutant from Oxidative and Apoptotic Stress and Extends Chronological Lifespan. Curr Microbiol 2017; 75:519-530. [PMID: 29224051 DOI: 10.1007/s00284-017-1412-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 12/01/2017] [Indexed: 02/06/2023]
|
21
|
Aufschnaiter A, Habernig L, Kohler V, Diessl J, Carmona-Gutierrez D, Eisenberg T, Keller W, Büttner S. The Coordinated Action of Calcineurin and Cathepsin D Protects Against α-Synuclein Toxicity. Front Mol Neurosci 2017; 10:207. [PMID: 28713240 PMCID: PMC5491553 DOI: 10.3389/fnmol.2017.00207] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 06/12/2017] [Indexed: 11/24/2022] Open
Abstract
The degeneration of dopaminergic neurons during Parkinson’s disease (PD) is intimately linked to malfunction of α-synuclein (αSyn), the main component of the proteinaceous intracellular inclusions characteristic for this pathology. The cytotoxicity of αSyn has been attributed to disturbances in several biological processes conserved from yeast to humans, including Ca2+ homeostasis, general lysosomal function and autophagy. However, the precise sequence of events that eventually results in cell death remains unclear. Here, we establish a connection between the major lysosomal protease cathepsin D (CatD) and the Ca2+/calmodulin-dependent phosphatase calcineurin. In a yeast model for PD, high levels of human αSyn triggered cytosolic acidification and reduced vacuolar hydrolytic capacity, finally leading to cell death. This could be counteracted by overexpression of yeast CatD (Pep4), which re-installed pH homeostasis and vacuolar proteolytic function, decreased αSyn oligomers and aggregates, and provided cytoprotection. Interestingly, these beneficial effects of Pep4 were independent of autophagy. Instead, they required functional calcineurin signaling, since deletion of calcineurin strongly reduced both the proteolytic activity of endogenous Pep4 and the cytoprotective capacity of overexpressed Pep4. Calcineurin contributed to proper endosomal targeting of Pep4 to the vacuole and the recycling of the Pep4 sorting receptor Pep1 from prevacuolar compartments back to the trans-Golgi network. Altogether, we demonstrate that stimulation of this novel calcineurin-Pep4 axis reduces αSyn cytotoxicity.
Collapse
Affiliation(s)
| | - Lukas Habernig
- Institute of Molecular Biosciences, University of GrazGraz, Austria.,Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm UniversityStockholm, Sweden
| | - Verena Kohler
- Institute of Molecular Biosciences, University of GrazGraz, Austria
| | - Jutta Diessl
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm UniversityStockholm, Sweden
| | | | - Tobias Eisenberg
- Institute of Molecular Biosciences, University of GrazGraz, Austria
| | - Walter Keller
- Institute of Molecular Biosciences, University of GrazGraz, Austria
| | - Sabrina Büttner
- Institute of Molecular Biosciences, University of GrazGraz, Austria.,Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm UniversityStockholm, Sweden
| |
Collapse
|
22
|
Stekovic S, Ruckenstuhl C, Royer P, Winkler-Hermaden C, Carmona-Gutierrez D, Fröhlich KU, Kroemer G, Madeo F. The neuroprotective steroid progesterone promotes mitochondrial uncoupling, reduces cytosolic calcium and augments stress resistance in yeast cells. MICROBIAL CELL (GRAZ, AUSTRIA) 2017; 4:191-199. [PMID: 28660203 PMCID: PMC5473691 DOI: 10.15698/mic2017.06.577] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 05/22/2017] [Indexed: 11/13/2022]
Abstract
The steroid hormone progesterone is not only a crucial sex hormone, but also serves as a neurosteroid, thus playing an important role in brain function. Epidemiological data suggest that progesterone improves the recovery of patients after traumatic brain injury. Brain injuries are often connected to elevated calcium spikes, reactive oxygen species (ROS) and programmed cell death affecting neurons. Here, we establish a yeast model to study progesterone-mediated cytoprotection. External supply of progesterone protected yeast cells from apoptosis-inducing stress stimuli and resulted in elevated mitochondrial oxygen uptake accompanied by a drop in ROS generation and ATP levels during chronological aging. In addition, cellular Ca2+ concentrations were reduced upon progesterone treatment, and this effect occurred independently of known Ca2+ transporters and mitochondrial respiration. All effects were also independent of Dap1, the yeast orthologue of the progesterone receptor. Altogether, our observations provide new insights into the cytoprotective effects of progesterone.
Collapse
Affiliation(s)
- Slaven Stekovic
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, 8010 Graz, Austria
| | - Christoph Ruckenstuhl
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, 8010 Graz, Austria
| | - Philipp Royer
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, 8010 Graz, Austria
| | | | | | - Kai-Uwe Fröhlich
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, 8010 Graz, Austria
| | - Guido Kroemer
- Equipe 11 labellisée par la Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- Karolinska Institute, Department of Women's and Children's Health, Karolinska University Hospital, 17176 Stockholm, Sweden
| | - Frank Madeo
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, 8010 Graz, Austria
- BioTechMed Graz, Austria
| |
Collapse
|
23
|
Dong Y, Hu J, Fan L, Chen Q. RNA-Seq-based transcriptomic and metabolomic analysis reveal stress responses and programmed cell death induced by acetic acid in Saccharomyces cerevisiae. Sci Rep 2017; 7:42659. [PMID: 28209995 PMCID: PMC5314350 DOI: 10.1038/srep42659] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 01/12/2017] [Indexed: 12/19/2022] Open
Abstract
As a typical harmful inhibitor in cellulosic hydrolyzates, acetic acid not only hinders bioethanol production, but also induces cell death in Saccharomyces cerevisiae. Herein, we conducted both transcriptomic and metabolomic analyses to investigate the global responses under acetic acid stress at different stages. There were 295 up-regulated and 427 down-regulated genes identified at more than two time points during acetic acid treatment (150 mM, pH 3.0). These differentially expressed genes (DEGs) were mainly involved in intracellular homeostasis, central metabolic pathway, transcription regulation, protein folding and stabilization, ubiquitin-dependent protein catabolic process, vesicle-mediated transport, protein synthesis, MAPK signaling pathways, cell cycle, programmed cell death, etc. The interaction network of all identified DEGs was constructed to speculate the potential regulatory genes and dominant pathways in response to acetic acid. The transcriptional changes were confirmed by metabolic profiles and phenotypic analysis. Acetic acid resulted in severe acidification in both cytosol and mitochondria, which was different from the effect of extracellular pH. Additionally, the imbalance of intracellular acetylation was shown to aggravate cell death under this stress. Overall, this work provides a novel and comprehensive understanding of stress responses and programmed cell death induced by acetic acid in yeast.
Collapse
Affiliation(s)
- Yachen Dong
- Department of Food Science and Nutrition, Key Laboratory for Food Microbial Technology of Zhejiang Province, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Jingjin Hu
- Department of Food Science and Nutrition, Key Laboratory for Food Microbial Technology of Zhejiang Province, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Linlin Fan
- Department of Food Science and Nutrition, Key Laboratory for Food Microbial Technology of Zhejiang Province, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Qihe Chen
- Department of Food Science and Nutrition, Key Laboratory for Food Microbial Technology of Zhejiang Province, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| |
Collapse
|
24
|
Carmona-Gutierrez D, Hughes AL, Madeo F, Ruckenstuhl C. The crucial impact of lysosomes in aging and longevity. Ageing Res Rev 2016; 32:2-12. [PMID: 27125853 DOI: 10.1016/j.arr.2016.04.009] [Citation(s) in RCA: 173] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Revised: 03/26/2016] [Accepted: 04/23/2016] [Indexed: 02/07/2023]
Abstract
Lysosomes are the main catabolic organelles of a cell and play a pivotal role in a plethora of cellular processes, including responses to nutrient availability and composition, stress resistance, programmed cell death, plasma membrane repair, development, and cell differentiation. In line with this pleiotropic importance for cellular and organismal life and death, lysosomal dysfunction is associated with many age-related pathologies like Parkinson's and Alzheimer's disease, as well as with a decline in lifespan. Conversely, targeting lysosomal functional capacity is emerging as a means to promote longevity. Here, we analyze the current knowledge on the prominent influence of lysosomes on aging-related processes, such as their executory and regulatory roles during general and selective macroautophagy, or their storage capacity for amino acids and ions. In addition, we review and discuss the roles of lysosomes as active players in the mechanisms underlying known lifespan-extending interventions like, for example, spermidine or rapamycin administration. In conclusion, this review aims at critically examining the nature and pliability of the different layers, in which lysosomes are involved as a control hub for aging and longevity.
Collapse
|
25
|
Kainz K, Tadic J, Zimmermann A, Pendl T, Carmona-Gutierrez D, Ruckenstuhl C, Eisenberg T, Madeo F. Methods to Assess Autophagy and Chronological Aging in Yeast. Methods Enzymol 2016; 588:367-394. [PMID: 28237110 DOI: 10.1016/bs.mie.2016.09.086] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Autophagy is a catabolic process that is crucial for cellular homeostasis and adaptive response to changing environments. Importantly, autophagy has been shown to be induced in many longevity-associated scenarios and to be required to maintain lifespan extension. Notably, autophagy is a highly conserved cellular process among eukaryotes, and the yeast Saccharomyces cerevisiae has become a universal model system for unraveling the molecular machinery underlying autophagic mechanisms. Here, we discuss different protocols to monitor survival and autophagy of yeast cells upon chronological aging. These include the use of propidium iodide to assess the loss of cell membrane integrity, as well as clonogenic assays to directly determine survival rates. Additionally, we describe methods to quantify autophagic flux, including the alkaline phosphatase activity or the GFP liberation assays, which measure the delivery of autophagosomal cargo to the vacuole. In sum, we have recapped established protocols used to evaluate a link between lifespan extension and autophagy in yeast.
Collapse
Affiliation(s)
- K Kainz
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
| | - J Tadic
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
| | - A Zimmermann
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
| | - T Pendl
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
| | - D Carmona-Gutierrez
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
| | - C Ruckenstuhl
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
| | - T Eisenberg
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria; BioTechMed-Graz, Graz, Austria
| | - F Madeo
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria; BioTechMed-Graz, Graz, Austria.
| |
Collapse
|
26
|
Michiels CF, Kurdi A, Timmermans JP, De Meyer GR, Martinet W. Spermidine reduces lipid accumulation and necrotic core formation in atherosclerotic plaques via induction of autophagy. Atherosclerosis 2016; 251:319-327. [DOI: 10.1016/j.atherosclerosis.2016.07.899] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 07/01/2016] [Accepted: 07/13/2016] [Indexed: 12/27/2022]
|
27
|
Falcone C, Mazzoni C. External and internal triggers of cell death in yeast. Cell Mol Life Sci 2016; 73:2237-50. [PMID: 27048816 PMCID: PMC4887522 DOI: 10.1007/s00018-016-2197-y] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 03/18/2016] [Indexed: 01/30/2023]
Abstract
In recent years, yeast was confirmed as a useful eukaryotic model system to decipher the complex mechanisms and networks occurring in higher eukaryotes, particularly in mammalian cells, in physiological as well in pathological conditions. This article focuses attention on the contribution of yeast in the study of a very complex scenario, because of the number and interconnection of pathways, represented by cell death. Yeast, although it is a unicellular organism, possesses the basal machinery of different kinds of cell death occurring in higher eukaryotes, i.e., apoptosis, regulated necrosis and autophagy. Here we report the current knowledge concerning the yeast orthologs of main mammalian cell death regulators and executors, the role of organelles and compartments, and the cellular phenotypes observed in the different forms of cell death in response to external and internal triggers. Thanks to the ease of genetic manipulation of this microorganism, yeast strains expressing human genes that promote or counteract cell death, onset of tumors and neurodegenerative diseases have been constructed. The effects on yeast cells of some of these genes are also presented.
Collapse
Affiliation(s)
- Claudio Falcone
- Pasteur Institute-Cenci Bolognetti Foundation; Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Cristina Mazzoni
- Pasteur Institute-Cenci Bolognetti Foundation; Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, Piazzale Aldo Moro 5, 00185, Rome, Italy.
| |
Collapse
|
28
|
Morrow G, Kim HJ, Pellerito O, Bourrelle-Langlois M, Le Pécheur M, Groebe K, Tanguay RM. Changes in Drosophila mitochondrial proteins following chaperone-mediated lifespan extension confirm a role of Hsp22 in mitochondrial UPR and reveal a mitochondrial localization for cathepsin D. Mech Ageing Dev 2016; 155:36-47. [PMID: 26930296 DOI: 10.1016/j.mad.2016.02.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 01/20/2016] [Accepted: 02/25/2016] [Indexed: 10/22/2022]
Abstract
Hsp22 is a small mitochondrial heat shock protein (sHSP) preferentially up-regulated during aging in Drosophila melanogaster. Its developmental expression is strictly regulated and it is rapidly induced in conditions of stress. Hsp22 is one of the few sHSP to be localized inside mitochondria, and is the first sHSP to be involved in the mitochondrial unfolding protein response (UPR(MT)) together with Hsp60, mitochondrial Hsp70 and TRAP1. The UPR(MT) is a pro-longevity mechanism, and interestingly Hsp22 over-expression by-itself increases lifespan and resistance to stress. To unveil the effect of Hsp22 on the mitochondrial proteome, comparative IEF/SDS polyacrylamide 2D gels were done on mitochondria from Hsp22+ flies and controls. Among the proteins influenced by Hsp22 expression were proteins from the electron transport chain (ETC), the TCA cycle and mitochondrial Hsp70. Hsp22 co-migrates with ETC components and its over-expression is associated with an increase in mitochondrial protease activity. Interestingly, the only protease that showed significant changes upon Hsp22 over-expression in the comparative IEF/SDS-PAGE analysis was cathepsin D, which is localized in mitochondria in addition to lysosome in D. melanogaster as evidenced by cellular fractionation. Together the results are consistent with a role of Hsp22 in the UPR(MT) and in mitochondrial proteostasis.
Collapse
Affiliation(s)
- Geneviève Morrow
- Laboratoire de Génétique Cellulaire et Développementale, Département de biologie moléculaire, biochimie médicale et pathologie, Institut de Biologie Intégrative et des Systèmes (IBIS) and PROTEO, Université Laval, Québec, G1V 0A6, Canada
| | - Hyun-Ju Kim
- Laboratoire de Génétique Cellulaire et Développementale, Département de biologie moléculaire, biochimie médicale et pathologie, Institut de Biologie Intégrative et des Systèmes (IBIS) and PROTEO, Université Laval, Québec, G1V 0A6, Canada
| | - Ornella Pellerito
- Laboratoire de Génétique Cellulaire et Développementale, Département de biologie moléculaire, biochimie médicale et pathologie, Institut de Biologie Intégrative et des Systèmes (IBIS) and PROTEO, Université Laval, Québec, G1V 0A6, Canada
| | - Maxime Bourrelle-Langlois
- Laboratoire de Génétique Cellulaire et Développementale, Département de biologie moléculaire, biochimie médicale et pathologie, Institut de Biologie Intégrative et des Systèmes (IBIS) and PROTEO, Université Laval, Québec, G1V 0A6, Canada
| | - Marie Le Pécheur
- Laboratoire de Génétique Cellulaire et Développementale, Département de biologie moléculaire, biochimie médicale et pathologie, Institut de Biologie Intégrative et des Systèmes (IBIS) and PROTEO, Université Laval, Québec, G1V 0A6, Canada
| | | | - Robert M Tanguay
- Laboratoire de Génétique Cellulaire et Développementale, Département de biologie moléculaire, biochimie médicale et pathologie, Institut de Biologie Intégrative et des Systèmes (IBIS) and PROTEO, Université Laval, Québec, G1V 0A6, Canada.
| |
Collapse
|
29
|
Tower J. Programmed cell death in aging. Ageing Res Rev 2015; 23:90-100. [PMID: 25862945 DOI: 10.1016/j.arr.2015.04.002] [Citation(s) in RCA: 288] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Revised: 03/15/2015] [Accepted: 04/01/2015] [Indexed: 02/08/2023]
Abstract
Programmed cell death (PCD) pathways, including apoptosis and regulated necrosis, are required for normal cell turnover and tissue homeostasis. Mis-regulation of PCD is increasingly implicated in aging and aging-related disease. During aging the cell turnover rate declines for several highly-mitotic tissues. Aging-associated disruptions in systemic and inter-cell signaling combined with cell-autonomous damage and mitochondrial malfunction result in increased PCD in some cell types, and decreased PCD in other cell types. Increased PCD during aging is implicated in immune system decline, skeletal muscle wasting (sarcopenia), loss of cells in the heart, and neurodegenerative disease. In contrast, cancer cells and senescent cells are resistant to PCD, enabling them to increase in abundance during aging. PCD pathways limit life span in fungi, but whether PCD pathways normally limit adult metazoan life span is not yet clear. PCD is regulated by a balance of negative and positive factors, including the mitochondria, which are particularly subject to aging-associated malfunction.
Collapse
|
30
|
Pereira H, Oliveira CSF, Castro L, Preto A, Chaves SR, Côrte-Real M. Yeast as a tool to explore cathepsin D function. MICROBIAL CELL 2015; 2:225-234. [PMID: 28357298 PMCID: PMC5349170 DOI: 10.15698/mic2015.07.212] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cathepsin D has garnered increased attention in recent years, mainly since it has been associated with several human pathologies. In particular, cathepsin D is often overexpressed and hypersecreted in cancer cells, implying it may constitute a therapeutic target. However, cathepsin D can have both anti- and pro-survival functions depending on its proteolytic activity, cellular context and stress stimulus. Therefore, a more detailed understanding of cathepsin D regulation and how to modulate its apoptotic functions is clearly needed. In this review, we provide an overview of the role of cathepsin D in physiological and pathological scenarios. We then focus on the opposing functions of cathepsin D in apoptosis, particularly relevant in cancer research. Emphasis is given to the role of the yeast protease Pep4p, the vacuolar counterpart of cathepsin D, in life and death. Finally, we discuss how insights from yeast cathepsin D and its role in regulated cell death can unveil novel functions of mammalian cathepsin D in apoptosis and cancer.
Collapse
Affiliation(s)
- H Pereira
- CBMA- Centre of Molecular and Environmental Biology. Department of Biology, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal
| | - C S F Oliveira
- CBMA- Centre of Molecular and Environmental Biology. Department of Biology, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal. ; ICBAS - Institute of Biomedical Sciences Abel Salazar, University of Porto, 4050-313, Porto, Portugal
| | - L Castro
- CBMA- Centre of Molecular and Environmental Biology. Department of Biology, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal
| | - A Preto
- CBMA- Centre of Molecular and Environmental Biology. Department of Biology, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal
| | - S R Chaves
- CBMA- Centre of Molecular and Environmental Biology. Department of Biology, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal
| | - M Côrte-Real
- CBMA- Centre of Molecular and Environmental Biology. Department of Biology, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal
| |
Collapse
|
31
|
Cathepsin D protects colorectal cancer cells from acetate-induced apoptosis through autophagy-independent degradation of damaged mitochondria. Cell Death Dis 2015; 6:e1788. [PMID: 26086961 PMCID: PMC4669836 DOI: 10.1038/cddis.2015.157] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Revised: 04/24/2015] [Accepted: 05/07/2015] [Indexed: 12/15/2022]
Abstract
Acetate is a short-chain fatty acid secreted by Propionibacteria from the human intestine, known to induce mitochondrial apoptotic death in colorectal cancer (CRC) cells. We previously established that acetate also induces lysosome membrane permeabilization in CRC cells, associated with release of the lysosomal protease cathepsin D (CatD), which has a well-established role in the mitochondrial apoptotic cascade. Unexpectedly, we showed that CatD has an antiapoptotic role in this process, as pepstatin A (a CatD inhibitor) increased acetate-induced apoptosis. These results mimicked our previous data in the yeast system showing that acetic acid activates a mitochondria-dependent apoptosis process associated with vacuolar membrane permeabilization and release of the vacuolar protease Pep4p, ortholog of mammalian CatD. Indeed, this protease was required for cell survival in a manner dependent on its catalytic activity and for efficient mitochondrial degradation independently of autophagy. In this study, we therefore assessed the role of CatD in acetate-induced mitochondrial alterations. We found that, similar to acetic acid in yeast, acetate-induced apoptosis is not associated with autophagy induction in CRC cells. Moreover, inhibition of CatD with small interfering RNA or pepstatin A enhanced apoptosis associated with higher mitochondrial dysfunction and increased mitochondrial mass. This effect seems to be specific, as inhibition of CatB and CatL with E-64d had no effect, nor were these proteases significantly released to the cytosol during acetate-induced apoptosis. Using yeast cells, we further show that the role of Pep4p in mitochondrial degradation depends on its protease activity and is complemented by CatD, indicating that this mechanism is conserved. In summary, the clues provided by the yeast model unveiled a novel CatD function in the degradation of damaged mitochondria when autophagy is impaired, which protects CRC cells from acetate-induced apoptosis. CatD inhibitors could therefore enhance acetate-mediated cancer cell death, presenting a novel strategy for prevention or therapy of CRC.
Collapse
|
32
|
Faller KME, Gutierrez-Quintana R, Mohammed A, Rahim AA, Tuxworth RI, Wager K, Bond M. The neuronal ceroid lipofuscinoses: Opportunities from model systems. Biochim Biophys Acta Mol Basis Dis 2015; 1852:2267-78. [PMID: 25937302 DOI: 10.1016/j.bbadis.2015.04.022] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 04/13/2015] [Accepted: 04/22/2015] [Indexed: 12/16/2022]
Abstract
The neuronal ceroid lipofuscinoses are a group of severe and progressive neurodegenerative disorders, generally with childhood onset. Despite the fact that these diseases remain fatal, significant breakthroughs have been made in our understanding of the genetics that underpin these conditions. This understanding has allowed the development of a broad range of models to study disease processes, and to develop new therapeutic approaches. Such models have contributed significantly to our knowledge of these conditions. In this review we will focus on the advantages of each individual model, describe some of the contributions the models have made to our understanding of the broader disease biology and highlight new techniques and approaches relevant to the study and potential treatment of the neuronal ceroid lipofuscinoses. This article is part of a Special Issue entitled: "Current Research on the Neuronal Ceroid Lipofuscinoses (Batten Disease)".
Collapse
Affiliation(s)
- Kiterie M E Faller
- School of Veterinary Medicine, College of Veterinary, Medical and Life Sciences, Bearsden Road, Glasgow G61 1QH, UK
| | - Rodrigo Gutierrez-Quintana
- School of Veterinary Medicine, College of Veterinary, Medical and Life Sciences, Bearsden Road, Glasgow G61 1QH, UK
| | - Alamin Mohammed
- College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Ahad A Rahim
- UCL School of Pharmacy, 29-39 Brunswick Square, London WC1N 1AX, UK
| | - Richard I Tuxworth
- College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Kim Wager
- Cardiff School of Biosciences, Cardiff University, The Sir Martin Evans Building, Museum Avenue, Cardiff CF10 3AX, UK
| | - Michael Bond
- MRC Laboratory for Molecular Cell Biology, University College of London, Gower Street, London WC1E 6BT, UK.
| |
Collapse
|
33
|
Minois N, Rockenfeller P, Smith TK, Carmona-Gutierrez D. Spermidine feeding decreases age-related locomotor activity loss and induces changes in lipid composition. PLoS One 2014; 9:e102435. [PMID: 25010732 PMCID: PMC4092136 DOI: 10.1371/journal.pone.0102435] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Accepted: 06/19/2014] [Indexed: 12/12/2022] Open
Abstract
Spermidine is a natural polyamine involved in many important cellular functions, whose supplementation in food or water increases life span and stress resistance in several model organisms. In this work, we expand spermidine's range of age-related beneficial effects by demonstrating that it is also able to improve locomotor performance in aged flies. Spermidine's mechanism of action on aging has been primarily related to general protein hypoacetylation that subsequently induces autophagy. Here, we suggest that the molecular targets of spermidine also include lipid metabolism: Spermidine-fed flies contain more triglycerides and show altered fatty acid and phospholipid profiles. We further determine that most of these metabolic changes are regulated through autophagy. Collectively, our data suggests an additional and novel lipid-mediated mechanism of action for spermidine-induced autophagy.
Collapse
Affiliation(s)
- Nadège Minois
- Biomedical Sciences Research Complex, University of St. Andrews, St. Andrews, United Kingdom
| | | | - Terry K. Smith
- Biomedical Sciences Research Complex, University of St. Andrews, St. Andrews, United Kingdom
| | | |
Collapse
|
34
|
Carmona-Gutierrez D, Büttner S. The many ways to age for a single yeast cell. Yeast 2014; 31:289-98. [PMID: 24842537 PMCID: PMC4140606 DOI: 10.1002/yea.3020] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Revised: 05/07/2014] [Accepted: 05/09/2014] [Indexed: 12/17/2022] Open
Abstract
The identification and characterization of the molecular determinants governing ageing represents the key to counteracting age-related diseases and eventually prolonging our health span. A large number of fundamental insights into the ageing process have been provided by research into the budding yeast Saccharomyces cerevisiae, which couples a wide array of technical advantages with a high degree of genetic, proteomic and mechanistic conservation. Indeed, this unicellular organism harbours regulatory pathways, such as those related to programmed cell death or nutrient signalling, that are crucial for ageing control and are reminiscent of other eukaryotes, including mammals. Here, we summarize and discuss three different paradigms of yeast ageing: replicative, chronological and colony ageing. We address their physiological relevance as well as the specific and common characteristics and regulators involved, providing an overview of the network underlying ageing in one of the most important eukaryotic model organisms.
Collapse
|
35
|
Moschou PN, Roubelakis-Angelakis KA. Polyamines and programmed cell death. JOURNAL OF EXPERIMENTAL BOTANY 2014; 65:1285-96. [PMID: 24218329 DOI: 10.1093/jxb/ert373] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Polyamines (PAs) have been considered as important molecules for survival. However, evidence reinforces that PAs are also implicated, directly or indirectly, in pathways regulating programmed cell death (PCD). Direct correlation of PAs with cell death refers to their association with particular biological processes, and their physical contact with molecules or structures involved in cell death. Indirectly, PAs regulate PCD through their metabolic derivatives, such as catabolic and interconversion products. Cytotoxic products of PA metabolism are involved in PCD cascades, whereas it remains largely elusive how PAs directly control pathways leading to PCD. In this review, we present and compare advances in PA-dependent PCD in animals and plants.
Collapse
Affiliation(s)
- Panagiotis N Moschou
- Department of Plant Biology and Forest Genetics, Uppsala BioCenter, Swedish University of Agricultural Sciences and Linnean Center for Plant Biology, 75007 Uppsala, Sweden
| | | |
Collapse
|
36
|
Minois N. Molecular Basis of the Anti-Aging' Effect of Spermidine and Other Natural Polyamines - A Mini-Review. Gerontology 2014; 60:319-26. [DOI: 10.1159/000356748] [Citation(s) in RCA: 113] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2013] [Accepted: 10/15/2013] [Indexed: 11/19/2022] Open
|
37
|
Lin SJ, Austriaco N. Aging and cell death in the other yeasts, Schizosaccharomyces pombe and Candida albicans. FEMS Yeast Res 2013; 14:119-35. [PMID: 24205865 DOI: 10.1111/1567-1364.12113] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Revised: 09/18/2013] [Accepted: 10/10/2013] [Indexed: 12/22/2022] Open
Abstract
How do cells age and die? For the past 20 years, the budding yeast, Saccharomyces cerevisiae, has been used as a model organism to uncover the genes that regulate lifespan and cell death. More recently, investigators have begun to interrogate the other yeasts, the fission yeast, Schizosaccharomyces pombe, and the human fungal pathogen, Candida albicans, to determine if similar longevity and cell death pathways exist in these organisms. After summarizing the longevity and cell death phenotypes in S. cerevisiae, this mini-review surveys the progress made in the study of both aging and programed cell death (PCD) in the yeast models, with a focus on the biology of S. pombe and C. albicans. Particular emphasis is placed on the similarities and differences between the two types of aging, replicative aging, and chronological aging, and between the three types of cell death, intrinsic apoptosis, autophagic cell death, and regulated necrosis, found in these yeasts. The development of the additional microbial models for aging and PCD in the other yeasts may help further elucidate the mechanisms of longevity and cell death regulation in eukaryotes.
Collapse
Affiliation(s)
- Su-Ju Lin
- Department of Microbiology and Molecular Genetics, College of Biological Sciences, University of California, Davis, CA, USA
| | | |
Collapse
|
38
|
Sheibani S, Richard VR, Beach A, Leonov A, Feldman R, Mattie S, Khelghatybana L, Piano A, Greenwood M, Vali H, Titorenko VI. Macromitophagy, neutral lipids synthesis, and peroxisomal fatty acid oxidation protect yeast from "liponecrosis", a previously unknown form of programmed cell death. Cell Cycle 2013; 13:138-47. [PMID: 24196447 DOI: 10.4161/cc.26885] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
We identified a form of cell death called "liponecrosis." It can be elicited by an exposure of the yeast Saccharomyces cerevisiae to exogenous palmitoleic acid (POA). Our data imply that liponecrosis is: (1) a programmed, regulated form of cell death rather than an accidental, unregulated cellular process and (2) an age-related form of cell death. Cells committed to liponecrotic death: (1) do not exhibit features characteristic of apoptotic cell death; (2) do not display plasma membrane rupture, a hallmark of programmed necrotic cell death; (3) akin to cells committed to necrotic cell death, exhibit an increased permeability of the plasma membrane for propidium iodide; (4) do not display excessive cytoplasmic vacuolization, a hallmark of autophagic cell death; (5) akin to cells committed to autophagic death, exhibit a non-selective en masse degradation of cellular organelles and require the cytosolic serine/threonine protein kinase Atg1p for executing the death program; and (6) display a hallmark feature that has not been reported for any of the currently known cell death modalities-namely, an excessive accumulation of lipid droplets where non-esterified fatty acids (including POA) are deposited in the form of neutral lipids. We therefore concluded that liponecrotic cell death subroutine differs from the currently known subroutines of programmed cell death. Our data suggest a hypothesis that liponecrosis is a cell death module dynamically integrated into a so-called programmed cell death network, which also includes the apoptotic, necrotic, and autophagic modules of programmed cell death. Based on our findings, we propose a mechanism underlying liponecrosis.
Collapse
Affiliation(s)
- Sara Sheibani
- Department of Anatomy and Cell Biology; McGill University; Montreal, Quebec, Canada; Department of Chemistry and Chemical Engineering; Royal Military College of Canada; Kingston, Ontario, Canada
| | - Vincent R Richard
- Department of Biology; Concordia University; Montreal, Quebec, Canada
| | - Adam Beach
- Department of Biology; Concordia University; Montreal, Quebec, Canada
| | - Anna Leonov
- Department of Biology; Concordia University; Montreal, Quebec, Canada
| | - Rachel Feldman
- Department of Biology; Concordia University; Montreal, Quebec, Canada
| | - Sevan Mattie
- Department of Biology; Concordia University; Montreal, Quebec, Canada
| | | | - Amanda Piano
- Department of Biology; Concordia University; Montreal, Quebec, Canada
| | - Michael Greenwood
- Department of Chemistry and Chemical Engineering; Royal Military College of Canada; Kingston, Ontario, Canada
| | - Hojatollah Vali
- Department of Anatomy and Cell Biology; McGill University; Montreal, Quebec, Canada
| | | |
Collapse
|
39
|
Carmona-Gutierrez D, Alavian-Ghavanini A, Habernig L, Bauer MA, Hammer A, Rossmann C, Zimmermann AS, Ruckenstuhl C, Büttner S, Eisenberg T, Sattler W, Malle E, Madeo F. The cell death protease Kex1p is essential for hypochlorite-induced apoptosis in yeast. Cell Cycle 2013; 12:1704-12. [PMID: 23656787 PMCID: PMC3713129 DOI: 10.4161/cc.24801] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Revised: 04/24/2013] [Accepted: 04/24/2013] [Indexed: 11/19/2022] Open
Abstract
Following microbial pathogen invasion, the human immune system of activated phagocytes generates and releases the potent oxidant hypochlorous acid (HOCl), which contributes to the killing of menacing microorganisms. Though tightly controlled, HOCl generation by the myeloperoxidase-hydrogen peroxide-chloride system of neutrophils/monocytes may occur in excess and lead to tissue damage. It is thus of marked importance to delineate the molecular pathways underlying HOCl cytotoxicity in both microbial and human cells. Here, we show that HOCl induces the generation of reactive oxygen species (ROS), apoptotic cell death and the formation of specific HOCl-modified epitopes in the budding yeast Saccharomyces cerevisiae. Interestingly, HOCl cytotoxicity can be prevented by treatment with ROS scavengers, suggesting oxidative stress to mediate the lethal effect. The executing pathway involves the pro-apoptotic protease Kex1p, since its absence diminishes HOCl-induced production of ROS, apoptosis and protein modification. By characterizing HOCl-induced cell death in yeast and identifying a corresponding central executor, these results pave the way for the use of Saccharomyces cerevisiae in HOCl research, not least given that it combines both being a microorganism as well as a model for programmed cell death in higher eukaryotes.
Collapse
Affiliation(s)
| | - Ali Alavian-Ghavanini
- Institute of Molecular Biology and Biochemistry; Center for Molecular Medicine; Medical University of Graz; Graz, Austria
| | - Lukas Habernig
- Institute of Molecular Biosciences; University of Graz; Graz, Austria
| | - Maria Anna Bauer
- Institute of Molecular Biosciences; University of Graz; Graz, Austria
| | - Astrid Hammer
- Institute of Cell Biology, Histology and Embryology; Center for Molecular Medicine; Medical University of Graz; Graz, Austria
| | - Christine Rossmann
- Institute of Molecular Biology and Biochemistry; Center for Molecular Medicine; Medical University of Graz; Graz, Austria
| | | | | | - Sabrina Büttner
- Institute of Molecular Biosciences; University of Graz; Graz, Austria
| | - Tobias Eisenberg
- Institute of Molecular Biosciences; University of Graz; Graz, Austria
| | - Wolfgang Sattler
- Institute of Molecular Biology and Biochemistry; Center for Molecular Medicine; Medical University of Graz; Graz, Austria
| | - Ernst Malle
- Institute of Molecular Biology and Biochemistry; Center for Molecular Medicine; Medical University of Graz; Graz, Austria
| | - Frank Madeo
- Institute of Molecular Biosciences; University of Graz; Graz, Austria
| |
Collapse
|
40
|
Marques C, Oliveira CSF, Alves S, Chaves SR, Coutinho OP, Côrte-Real M, Preto A. Acetate-induced apoptosis in colorectal carcinoma cells involves lysosomal membrane permeabilization and cathepsin D release. Cell Death Dis 2013; 4:e507. [PMID: 23429293 PMCID: PMC3734821 DOI: 10.1038/cddis.2013.29] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Colorectal carcinoma (CRC) is one of the most common causes of cancer-related mortality. Short-chain fatty acids secreted by dietary propionibacteria from the intestine, such as acetate, induce apoptosis in CRC cells and may therefore be relevant in CRC prevention and therapy. We previously reported that acetic acid-induced apoptosis in Saccharomyces cerevisiae cells involves partial vacuole permeabilization and release of Pep4p, the yeast cathepsin D (CatD), which has a protective role in this process. In cancer cells, lysosomes have emerged as key players in apoptosis through selective lysosomal membrane permeabilization (LMP) and release of cathepsins. However, the role of CatD in CRC survival is controversial and has not been assessed in response to acetate. We aimed to ascertain whether LMP and CatD are involved in acetate-induced apoptosis in CRC cells. We showed that acetate per se inhibits proliferation and induces apoptosis. More importantly, we uncovered that acetate triggers LMP and CatD release to the cytosol. Pepstatin A (a CatD inhibitor) but not E64d (a cathepsin B and L inhibitor) increased acetate-induced apoptosis of CRC cells, suggesting that CatD has a protective role in this process. Our data indicate that acetate induces LMP and subsequent release of CatD in CRC cells undergoing apoptosis, and suggest exploiting novel strategies using acetate as a prevention/therapeutic agent in CRC, through simultaneous treatment with CatD inhibitors.
Collapse
Affiliation(s)
- C Marques
- CBMA (Centre of Molecular and Environmental Biology), Department of Biology, University of Minho, Campus de Gualtar, Braga, Portugal
| | | | | | | | | | | | | |
Collapse
|
41
|
Use of model organisms for the study of neuronal ceroid lipofuscinosis. Biochim Biophys Acta Mol Basis Dis 2013; 1832:1842-65. [PMID: 23338040 DOI: 10.1016/j.bbadis.2013.01.009] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Revised: 01/07/2013] [Accepted: 01/08/2013] [Indexed: 12/26/2022]
Abstract
Neuronal ceroid lipofuscinoses are a group of fatal progressive neurodegenerative diseases predominantly affecting children. Identification of mutations that cause neuronal ceroid lipofuscinosis, and subsequent functional and pathological studies of the affected genes, underpins efforts to investigate disease mechanisms and identify and test potential therapeutic strategies. These functional studies and pre-clinical trials necessitate the use of model organisms in addition to cell and tissue culture models as they enable the study of protein function within a complex organ such as the brain and the testing of therapies on a whole organism. To this end, a large number of disease models and genetic tools have been identified or created in a variety of model organisms. In this review, we will discuss the ethical issues associated with experiments using model organisms, the factors underlying the choice of model organism, the disease models and genetic tools available, and the contributions of those disease models and tools to neuronal ceroid lipofuscinosis research. This article is part of a Special Issue entitled: The Neuronal Ceroid Lipofuscinoses or Batten Disease.
Collapse
|
42
|
Bauer MA, Carmona-Gutiérrez D, Ruckenstuhl C, Reisenbichler A, Megalou EV, Eisenberg T, Magnes C, Jungwirth H, Sinner FM, Pieber TR, Fröhlich KU, Kroemer G, Tavernarakis N, Madeo F. Spermidine promotes mating and fertilization efficiency in model organisms. Cell Cycle 2013; 12:346-52. [PMID: 23255134 PMCID: PMC3575463 DOI: 10.4161/cc.23199] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Spermidine is a naturally occurring polyamine involved in multiple biological processes, including DNA metabolism, autophagy and aging. Like other polyamines, spermidine is also indispensable for successful reproduction at several stages. However, a direct influence on the actual fertilization process, i.e., the fusion of an oocyte with a spermatocyte, remains uncertain. To explore this possibility, we established the mating process in the yeast Saccharomyces cerevisiae as a model for fertilization in higher eukaryotes. During human fertilization, the sperm capacitates and the acrosome reaction is necessary for penetration of the oocyte. Similarly, sexually active yeasts form a protrusion called "shmoo" as a prerequisite for mating. In this study, we demonstrate that pheromone-induced shmoo formation requires spermidine. In addition, we show that spermidine is essential for mating in yeast as well as for egg fertilization in the nematode Caenorhabditis elegans. In both cases, this occurs independently from autophagy. In synthesis, we identify spermidine as an important mating component in unicellular and multicellular model organisms, supporting an unprecedented evolutionary conservation of the mechanisms governing fertilization-related cellular fusion.
Collapse
Affiliation(s)
- Maria Anna Bauer
- Institute of Molecular Biosciences; Karl-Franzens University of Graz; Graz, Austria
| | | | | | - Angela Reisenbichler
- Institute of Molecular Biosciences; Karl-Franzens University of Graz; Graz, Austria
| | - Evgenia V. Megalou
- Institute of Molecular Biology and Biotechnology; Foundation for Research and Technology-Hellas; Heraklion, Greece
| | - Tobias Eisenberg
- Institute of Molecular Biosciences; Karl-Franzens University of Graz; Graz, Austria
| | - Christoph Magnes
- Institute of Medical Technologies and Health Management; Joanneum Research; Graz, Austria
- Department of Internal Medicine; Division of Diabetes and Metabolism; Medical University of Graz; Graz, Austria
| | - Helmut Jungwirth
- Institute of Molecular Biosciences; Karl-Franzens University of Graz; Graz, Austria
| | - Frank M. Sinner
- Institute of Medical Technologies and Health Management; Joanneum Research; Graz, Austria
- Department of Internal Medicine; Division of Diabetes and Metabolism; Medical University of Graz; Graz, Austria
| | - Thomas R. Pieber
- Institute of Medical Technologies and Health Management; Joanneum Research; Graz, Austria
- Department of Internal Medicine; Division of Diabetes and Metabolism; Medical University of Graz; Graz, Austria
| | - Kai-Uwe Fröhlich
- Institute of Molecular Biosciences; Karl-Franzens University of Graz; Graz, Austria
| | - Guido Kroemer
- INSERM; U848 and Institut Gustave Roussy; Villejuif, France
- Metabolomics Platform; Institut Gustave Roussy; Villejuif, France
- Centre de Recherche des Cordeliers; Paris, France
- Pôle de Biologie; Hôpital Européen Georges Pompidou; Assistance Publique–Hôpitaux de Paris (AP-HP); Paris, France
- Université Paris Descartes/Paris 5; Sorbonne Paris Cité; Paris, France
| | - Nektarios Tavernarakis
- Institute of Molecular Biology and Biotechnology; Foundation for Research and Technology-Hellas; Heraklion, Greece
| | - Frank Madeo
- Institute of Molecular Biosciences; Karl-Franzens University of Graz; Graz, Austria
| |
Collapse
|
43
|
Pereira H, Azevedo F, Rego A, Sousa MJ, Chaves SR, Côrte-Real M. The protective role of yeast Cathepsin D in acetic acid-induced apoptosis depends on ANT (Aac2p) but not on the voltage-dependent channel (Por1p). FEBS Lett 2012; 587:200-5. [DOI: 10.1016/j.febslet.2012.11.025] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2012] [Revised: 10/20/2012] [Accepted: 11/25/2012] [Indexed: 11/16/2022]
|
44
|
Ramsdale M. Programmed cell death in the cellular differentiation of microbial eukaryotes. Curr Opin Microbiol 2012; 15:646-52. [DOI: 10.1016/j.mib.2012.09.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2012] [Revised: 09/21/2012] [Accepted: 09/25/2012] [Indexed: 01/22/2023]
|
45
|
Wloch-Salamon D, Bem A. Types of cell death and methods of their detection in yeast Saccharomyces cerevisiae. J Appl Microbiol 2012; 114:287-98. [DOI: 10.1111/jam.12024] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Revised: 09/13/2012] [Accepted: 09/19/2012] [Indexed: 12/16/2022]
Affiliation(s)
- D.M. Wloch-Salamon
- Institute of Environmental Sciences; Jagiellonian University; Krakow Poland
| | - A.E. Bem
- Host-Microbe Interactomics; Wageningen University; Wageningen The Netherlands
| |
Collapse
|
46
|
Minois N, Carmona-Gutierrez D, Bauer MA, Rockenfeller P, Eisenberg T, Brandhorst S, Sigrist SJ, Kroemer G, Madeo F. Spermidine promotes stress resistance in Drosophila melanogaster through autophagy-dependent and -independent pathways. Cell Death Dis 2012; 3:e401. [PMID: 23059820 PMCID: PMC3481127 DOI: 10.1038/cddis.2012.139] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2012] [Revised: 07/31/2012] [Accepted: 09/03/2012] [Indexed: 01/05/2023]
Abstract
The naturally occurring polyamine spermidine (Spd) has recently been shown to promote longevity across species in an autophagy-dependent manner. Here, we demonstrate that Spd improves both survival and locomotor activity of the fruit fly Drosophila melanogaster upon exposure to the superoxide generator and neurotoxic agent paraquat. Although survival to a high paraquat concentration (20 mM) was specifically increased in female flies only, locomotor activity and survival could be rescued in both male and female animals when exposed to lower paraquat levels (5 mM). These effects are dependent on the autophagic machinery, as Spd failed to confer resistance to paraquat-induced toxicity and locomotor impairment in flies deleted for the essential autophagic regulator ATG7 (autophagy-related gene 7). Spd treatment did also protect against mild doses of another oxidative stressor, hydrogen peroxide, but in this case in an autophagy-independent manner. Altogether, this study establishes that the protective effects of Spd can be exerted through different pathways that depending on the oxidative stress scenario do or do not involve autophagy.
Collapse
Affiliation(s)
- N Minois
- School of Biology, Biomedical Sciences Research Complex, University of St. Andrews, St. Andrews, UK
| | | | - M A Bauer
- Institute for Molecular Biosciences, University of Graz, Graz, Austria
| | - P Rockenfeller
- Institute for Molecular Biosciences, University of Graz, Graz, Austria
| | - T Eisenberg
- Institute for Molecular Biosciences, University of Graz, Graz, Austria
| | - S Brandhorst
- Andrus Gerontology Center, Department of Biological Sciences and Norris Cancer Center, University of Southern California, Los Angeles, CA, USA
- Centre for Medical Biotechnology, Faculty of Biology, University Duisburg–Essen, Essen, Germany
| | - S J Sigrist
- Department of Genetics, Institute for Biology, Free University Berlin, Berlin, Germany
- Neurocure Cluster of Excellence, Charité Berlin, Berlin, Germany
| | - G Kroemer
- INSERM, U848, Institute Gustave Roussy, University Paris XI, Villejuif, France
- Metabolomics Platform, Institut Gustave Roussy, Villejuif, France
- Centre de Recherche des Cordeliers, Université Paris Descartes, Paris 5, Paris, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| | - F Madeo
- Institute for Molecular Biosciences, University of Graz, Graz, Austria
| |
Collapse
|
47
|
Abstract
Saccharomyces cerevisiae has directly or indirectly contributed to the identification of arguably more mammalian genes that affect aging than any other model organism. Aging in yeast is assayed primarily by measurement of replicative or chronological life span. Here, we review the genes and mechanisms implicated in these two aging model systems and key remaining issues that need to be addressed for their optimization. Because of its well-characterized genome that is remarkably amenable to genetic manipulation and high-throughput screening procedures, S. cerevisiae will continue to serve as a leading model organism for studying pathways relevant to human aging and disease.
Collapse
Affiliation(s)
- Valter D Longo
- Longevity Institute, School of Gerontology, Department of Biological Sciences, and Norris Cancer Center, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089-0191, USA.
| | | | | | | |
Collapse
|
48
|
Farrugia G, Balzan R. Oxidative stress and programmed cell death in yeast. Front Oncol 2012; 2:64. [PMID: 22737670 PMCID: PMC3380282 DOI: 10.3389/fonc.2012.00064] [Citation(s) in RCA: 200] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2012] [Accepted: 06/02/2012] [Indexed: 12/11/2022] Open
Abstract
Yeasts, such as Saccharomyces cerevisiae, have long served as useful models for the study of oxidative stress, an event associated with cell death and severe human pathologies. This review will discuss oxidative stress in yeast, in terms of sources of reactive oxygen species (ROS), their molecular targets, and the metabolic responses elicited by cellular ROS accumulation. Responses of yeast to accumulated ROS include upregulation of antioxidants mediated by complex transcriptional changes, activation of pro-survival pathways such as mitophagy, and programmed cell death (PCD) which, apart from apoptosis, includes pathways such as autophagy and necrosis, a form of cell death long considered accidental and uncoordinated. The role of ROS in yeast aging will also be discussed.
Collapse
Affiliation(s)
- Gianluca Farrugia
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of MaltaMsida, Malta
| | - Rena Balzan
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of MaltaMsida, Malta
| |
Collapse
|
49
|
Clapp C, Portt L, Khoury C, Sheibani S, Eid R, Greenwood M, Vali H, Mandato CA, Greenwood MT. Untangling the Roles of Anti-Apoptosis in Regulating Programmed Cell Death using Humanized Yeast Cells. Front Oncol 2012; 2:59. [PMID: 22708116 PMCID: PMC3374133 DOI: 10.3389/fonc.2012.00059] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2012] [Accepted: 05/24/2012] [Indexed: 11/13/2022] Open
Abstract
Genetically programmed cell death (PCD) mechanisms, including apoptosis, are important for the survival of metazoans since it allows, among things, the removal of damaged cells that interfere with normal function. Cell death due to PCD is observed in normal processes such as aging and in a number of pathophysiologies including hypoxia (common causes of heart attacks and strokes) and subsequent tissue reperfusion. Conversely, the loss of normal apoptotic responses is associated with the development of tumors. So far, limited success in preventing unwanted PCD has been reported with current therapeutic approaches despite the fact that inhibitors of key apoptotic inducers such as caspases have been developed. Alternative approaches have focused on mimicking anti-apoptotic processes observed in cells displaying increased resistance to apoptotic stimuli. Hormesis and pre-conditioning are commonly observed cellular strategies where sub-lethal levels of pro-apoptotic stimuli lead to increased resistance to higher or lethal levels of stress. Increased expression of anti-apoptotic sequences is a common mechanism mediating these protective effects. The relevance of the latter observation is exemplified by the observation that transgenic mice overexpressing anti-apoptotic genes show significant reductions in tissue damage following ischemia. Thus strategies aimed at increasing the levels of anti-apoptotic proteins, using gene therapy or cell penetrating recombinant proteins are being evaluated as novel therapeutics to decrease cell death following acute periods of cell death inducing stress. In spite of its functional and therapeutic importance, more is known regarding the processes involved in apoptosis than anti-apoptosis. The genetically tractable yeast Saccharomyces cerevisiae has emerged as an exceptional model to study multiple aspects of PCD including the mitochondrial mediated apoptosis observed in metazoans. To increase our knowledge of the process of anti-apoptosis, we screened a human heart cDNA expression library in yeast cells undergoing PCD due to the conditional expression of a mammalian pro-apoptotic Bax cDNA. Analysis of the multiple Bax suppressors identified revealed several previously known as well as a large number of clones representing potential novel anti-apoptotic sequences. The focus of this review is to report on recent achievements in the use of humanized yeast in genetic screens to identify novel stress-induced PCD suppressors, supporting the use of yeast as a unicellular model organism to elucidate anti-apoptotic and cell survival mechanisms.
Collapse
Affiliation(s)
- Caitlin Clapp
- Department of Chemistry and Chemical Engineering, Royal Military College Kingston, ON, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Autophagy and apoptosis interplay during follicular atresia in fish ovary: a morphological and immunocytochemical study. Cell Tissue Res 2012; 347:467-78. [DOI: 10.1007/s00441-012-1327-6] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2011] [Accepted: 01/11/2012] [Indexed: 11/30/2022]
|