1
|
Gómez-Virgilio L, Silva-Lucero MDC, Flores-Morelos DS, Gallardo-Nieto J, Lopez-Toledo G, Abarca-Fernandez AM, Zacapala-Gómez AE, Luna-Muñoz J, Montiel-Sosa F, Soto-Rojas LO, Pacheco-Herrero M, Cardenas-Aguayo MDC. Autophagy: A Key Regulator of Homeostasis and Disease: An Overview of Molecular Mechanisms and Modulators. Cells 2022; 11:cells11152262. [PMID: 35892559 PMCID: PMC9329718 DOI: 10.3390/cells11152262] [Citation(s) in RCA: 115] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 07/07/2022] [Accepted: 07/11/2022] [Indexed: 01/18/2023] Open
Abstract
Autophagy is a highly conserved lysosomal degradation pathway active at basal levels in all cells. However, under stress conditions, such as a lack of nutrients or trophic factors, it works as a survival mechanism that allows the generation of metabolic precursors for the proper functioning of the cells until the nutrients are available. Neurons, as post-mitotic cells, depend largely on autophagy to maintain cell homeostasis to get rid of damaged and/or old organelles and misfolded or aggregated proteins. Therefore, the dysfunction of this process contributes to the pathologies of many human diseases. Furthermore, autophagy is highly active during differentiation and development. In this review, we describe the current knowledge of the different pathways, molecular mechanisms, factors that induce it, and the regulation of mammalian autophagy. We also discuss its relevant role in development and disease. Finally, here we summarize several investigations demonstrating that autophagic abnormalities have been considered the underlying reasons for many human diseases, including liver disease, cardiovascular, cerebrovascular diseases, neurodegenerative diseases, neoplastic diseases, cancers, and, more recently, infectious diseases, such as SARS-CoV-2 caused COVID-19 disease.
Collapse
Affiliation(s)
- Laura Gómez-Virgilio
- Laboratory of Cellular Reprogramming, Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autonoma de Mexico, Mexico City 04510, Mexico; (L.G.-V.); (M.-d.-C.S.-L.); (D.-S.F.-M.); (J.G.-N.); (G.L.-T.); (A.-M.A.-F.)
| | - Maria-del-Carmen Silva-Lucero
- Laboratory of Cellular Reprogramming, Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autonoma de Mexico, Mexico City 04510, Mexico; (L.G.-V.); (M.-d.-C.S.-L.); (D.-S.F.-M.); (J.G.-N.); (G.L.-T.); (A.-M.A.-F.)
| | - Diego-Salvador Flores-Morelos
- Laboratory of Cellular Reprogramming, Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autonoma de Mexico, Mexico City 04510, Mexico; (L.G.-V.); (M.-d.-C.S.-L.); (D.-S.F.-M.); (J.G.-N.); (G.L.-T.); (A.-M.A.-F.)
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo de los Bravo 39070, Guerrero, Mexico;
| | - Jazmin Gallardo-Nieto
- Laboratory of Cellular Reprogramming, Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autonoma de Mexico, Mexico City 04510, Mexico; (L.G.-V.); (M.-d.-C.S.-L.); (D.-S.F.-M.); (J.G.-N.); (G.L.-T.); (A.-M.A.-F.)
- Biotechnology Engeniering, Universidad Politécnica de Quintana Roo, Cancún 77500, Quintana Roo, Mexico
| | - Gustavo Lopez-Toledo
- Laboratory of Cellular Reprogramming, Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autonoma de Mexico, Mexico City 04510, Mexico; (L.G.-V.); (M.-d.-C.S.-L.); (D.-S.F.-M.); (J.G.-N.); (G.L.-T.); (A.-M.A.-F.)
| | - Arminda-Mercedes Abarca-Fernandez
- Laboratory of Cellular Reprogramming, Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autonoma de Mexico, Mexico City 04510, Mexico; (L.G.-V.); (M.-d.-C.S.-L.); (D.-S.F.-M.); (J.G.-N.); (G.L.-T.); (A.-M.A.-F.)
- Biotechnology Engeniering, Universidad Politécnica de Quintana Roo, Cancún 77500, Quintana Roo, Mexico
| | - Ana-Elvira Zacapala-Gómez
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo de los Bravo 39070, Guerrero, Mexico;
| | - José Luna-Muñoz
- National Dementia BioBank, Ciencias Biológicas, Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México, Cuautitlan Izcalli 53150, Estado de México, Mexico; (J.L.-M.); (F.M.-S.)
- Banco Nacional de Cerebros-UNPHU, Universidad Nacional Pedro Henríquez Ureña, Santo Domingo 11805, Dominican Republic
| | - Francisco Montiel-Sosa
- National Dementia BioBank, Ciencias Biológicas, Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México, Cuautitlan Izcalli 53150, Estado de México, Mexico; (J.L.-M.); (F.M.-S.)
| | - Luis O. Soto-Rojas
- Laboratorio de Patogénesis Molecular, Laboratorio 4, Edificio A4, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Estado de México, Mexico;
- Red MEDICI, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Estado de México, Mexico
| | - Mar Pacheco-Herrero
- Neuroscience Research Laboratory, Faculty of Health Sciences, Pontificia Universidad Católica Madre y Maestra, Santiago de los Caballeros 51000, Dominican Republic;
| | - Maria-del-Carmen Cardenas-Aguayo
- Laboratory of Cellular Reprogramming, Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autonoma de Mexico, Mexico City 04510, Mexico; (L.G.-V.); (M.-d.-C.S.-L.); (D.-S.F.-M.); (J.G.-N.); (G.L.-T.); (A.-M.A.-F.)
- Correspondence: ; Tel.: +52-55-2907-0937
| |
Collapse
|
2
|
Lin Z, Liu H, Yang C, Zheng H, Zhang Y, Su W, Shang J. Curcumin mediates autophagy and apoptosis in granulosa cells: a study of integrated network pharmacology and molecular docking to elucidate toxicological mechanisms. Drug Chem Toxicol 2021; 45:2411-2423. [PMID: 34315305 DOI: 10.1080/01480545.2021.1956941] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Curcumin (Cur) is a flavonoid derived from Curcuma longa L. that has been shown to have a variety of biological activities, but some previous studies have described its non-negligible negative effects on female reproduction and embryo development. To further explore the toxic stress effect, this study investigated apoptosis and autophagy of healthy buffalo (Bubalus bubalis) derived granulosa cells (GCs) exposed to Cur and/or autophagy inhibitors. Results showed that Cur declined viability of GCs in a concentration-dependent manner. Apoptosis was observed in Cur-treated GCs from 3 h. Meanwhile, under Cur stress, autophagosomes accumulated in cells, and the expression levels of autophagy key proteins LC3 and Beclin 1 were up-regulated, suggesting that Cur could induce autophagy in GCs. Early autophagy inhibitor 3-methyladenine (3-MA) increased the apoptosis rate of Cur exposed GCs, but the autophagosome degradation inhibitor chloroquine (CQ) had no effect on the apoptosis rate. The network pharmacological and molecular docking analysis indicated that the perturbation of IKK/NF-κB might be the cause of Cur toxicity toward GCs. This study unveiled another side of Cur pharmacological effects that programmed cell death can be induced by Cur in GCs, suggesting that it should be prudent to use Cur as a clinical drug for its side effects on the female reproductive system.
Collapse
Affiliation(s)
- Zhen Lin
- Key Laboratory of Buffalo Genetics, Breeding and Reproduction Technology, Ministry of Agriculture and Guangxi Buffalo Research Institute, Chinese Academy of Agricultural Sciences, Nanning, China.,College of Chemistry & Environmental Science, Guangdong Ocean University, Zhanjiang, China
| | - Huazhong Liu
- College of Chemistry & Environmental Science, Guangdong Ocean University, Zhanjiang, China
| | - Chunyan Yang
- Key Laboratory of Buffalo Genetics, Breeding and Reproduction Technology, Ministry of Agriculture and Guangxi Buffalo Research Institute, Chinese Academy of Agricultural Sciences, Nanning, China
| | - Haiying Zheng
- Key Laboratory of Buffalo Genetics, Breeding and Reproduction Technology, Ministry of Agriculture and Guangxi Buffalo Research Institute, Chinese Academy of Agricultural Sciences, Nanning, China
| | - Yu Zhang
- Key Laboratory of Buffalo Genetics, Breeding and Reproduction Technology, Ministry of Agriculture and Guangxi Buffalo Research Institute, Chinese Academy of Agricultural Sciences, Nanning, China.,College of Chemistry & Environmental Science, Guangdong Ocean University, Zhanjiang, China
| | - Weiming Su
- College of Food Science and Technology, Guangdong Ocean University, Zhanjiang, China
| | - Jianghua Shang
- Key Laboratory of Buffalo Genetics, Breeding and Reproduction Technology, Ministry of Agriculture and Guangxi Buffalo Research Institute, Chinese Academy of Agricultural Sciences, Nanning, China
| |
Collapse
|
3
|
Booth LA, Roberts JL, Dent P. The role of cell signaling in the crosstalk between autophagy and apoptosis in the regulation of tumor cell survival in response to sorafenib and neratinib. Semin Cancer Biol 2020; 66:129-139. [PMID: 31644944 PMCID: PMC7167338 DOI: 10.1016/j.semcancer.2019.10.013] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 09/23/2019] [Accepted: 10/16/2019] [Indexed: 12/19/2022]
Abstract
The molecular mechanisms by which tumor cells survive or die following therapeutic interventions are complex. There are three broadly defined categories of cell death processes: apoptosis (Type I), autophagic cell death (Type II), and necrosis (Type III). In hematopoietic tumor cells, the majority of toxic stimuli cause these cells to undergo a death process called apoptosis; apoptosis specifically involves the cleavage of DNA into large defined pieces and their subsequent localization in vesicles. Thus, 'pure' apoptosis largely lacks inflammatory potential. In carcinomas, however, the mechanisms by which tumor cells ultimately die are considerably more complex. Although the machinery of apoptosis is engaged by toxic stimuli, other processes such as autophagy ("self-eating") and replicative cell death can lead to observations that do not simplistically correspond to any of the individual Type I-III formalized death categories. The 'hybrid' forms of cell death observed in carcinoma cells result in cellular materials being released into the extracellular space without packaging, which promotes inflammation, potentially leading to the accelerated re-growth of surviving tumor cells by macrophages. Drugs as single agents or in combinations can simultaneously initiate signaling via both apoptotic and autophagic pathways. Based on the tumor type and its oncogene drivers, as well as the drug(s) being used and the duration and intensity of the autophagosome signal, apoptosis and autophagy have the potential to act in concert to kill or alternatively that the actions of either pathway can act to suppress signaling by the other pathway. And, there also is evidence that autophagic flux, by causing lysosomal protease activation, with their subsequent release into the cytosol, can directly mediate killing. This review will discuss the interactive biology between apoptosis and autophagy in carcinoma cells. Finally, the molecular actions of the FDA-approved drugs neratinib and sorafenib, and how they enhance both apoptotic and toxic autophagic processes, alone or in combination with other agents, is discussed in a bench-to-bedside manner.
Collapse
Affiliation(s)
- Laurence A Booth
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, 401 College St, Richmond, VA 23298, United States
| | - Jane L Roberts
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, 401 College St, Richmond, VA 23298, United States
| | - Paul Dent
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, 401 College St, Richmond, VA 23298, United States.
| |
Collapse
|
4
|
Ferguson EL, Naseer S, Powell LC, Hardwicke J, Young FI, Zhu B, Liu Q, Song B, Thomas DW. Controlled release of dextrin-conjugated growth factors to support growth and differentiation of neural stem cells. Stem Cell Res 2018; 33:69-78. [PMID: 30321831 PMCID: PMC6288241 DOI: 10.1016/j.scr.2018.10.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 09/06/2018] [Accepted: 10/03/2018] [Indexed: 12/16/2022] Open
Abstract
An essential aspect of stem cell in vitro culture and in vivo therapy is achieving sustained levels of growth factors to support stem cell survival and expansion, while maintaining their multipotency and differentiation potential. This study investigated the ability of dextrin (~74,000 g/mol; 27.8 mol% succinoylation) conjugated to epidermal growth factor (EGF) and basic fibroblast growth factor (bFGF; or FGF-2) (3.9 and 6.7% w/w protein loading, respectively) to support the expansion and differentiation of stem cells in vitro via sustained, controllable growth factor release. Supplementation of mouse neural stem cells (mNSCs) with dextrin-growth factor conjugates led to greater and prolonged proliferation compared to unbound EGF/bFGF controls, with no detectable apoptosis after 7 days of treatment. Immunocytochemical detection of neural precursor (nestin) and differentiation (Olig2, MAP2, GFAP) markers verified that controlled release of dextrin-conjugated growth factors preserves stem cell properties of mNSCs for up to 7 days. These results show the potential of dextrin-growth factor conjugates for localized delivery of bioactive therapeutic agents to support stem cell expansion and differentiation, and as an adjunct to direct neuronal repair.
Collapse
Affiliation(s)
- Elaine L Ferguson
- Oral and Biomedical Sciences, School of Dentistry, College of Biomedical and Life Sciences, Cardiff University, Heath Park, Cardiff CF14 4XY, UK; Cardiff Institute of Tissue Engineering and Repair, Cardiff University, 10 Museum Place, Cardiff, South Glamorgan, CF10 3BG, UK.
| | - Sameza Naseer
- Oral and Biomedical Sciences, School of Dentistry, College of Biomedical and Life Sciences, Cardiff University, Heath Park, Cardiff CF14 4XY, UK
| | - Lydia C Powell
- Oral and Biomedical Sciences, School of Dentistry, College of Biomedical and Life Sciences, Cardiff University, Heath Park, Cardiff CF14 4XY, UK; Cardiff Institute of Tissue Engineering and Repair, Cardiff University, 10 Museum Place, Cardiff, South Glamorgan, CF10 3BG, UK
| | - Joseph Hardwicke
- Oral and Biomedical Sciences, School of Dentistry, College of Biomedical and Life Sciences, Cardiff University, Heath Park, Cardiff CF14 4XY, UK; Cardiff Institute of Tissue Engineering and Repair, Cardiff University, 10 Museum Place, Cardiff, South Glamorgan, CF10 3BG, UK
| | - Fraser I Young
- Oral and Biomedical Sciences, School of Dentistry, College of Biomedical and Life Sciences, Cardiff University, Heath Park, Cardiff CF14 4XY, UK; Cardiff Institute of Tissue Engineering and Repair, Cardiff University, 10 Museum Place, Cardiff, South Glamorgan, CF10 3BG, UK
| | - Bangfu Zhu
- Oral and Biomedical Sciences, School of Dentistry, College of Biomedical and Life Sciences, Cardiff University, Heath Park, Cardiff CF14 4XY, UK
| | - Qian Liu
- Oral and Biomedical Sciences, School of Dentistry, College of Biomedical and Life Sciences, Cardiff University, Heath Park, Cardiff CF14 4XY, UK
| | - Bing Song
- Oral and Biomedical Sciences, School of Dentistry, College of Biomedical and Life Sciences, Cardiff University, Heath Park, Cardiff CF14 4XY, UK; Cardiff Institute of Tissue Engineering and Repair, Cardiff University, 10 Museum Place, Cardiff, South Glamorgan, CF10 3BG, UK
| | - David W Thomas
- Oral and Biomedical Sciences, School of Dentistry, College of Biomedical and Life Sciences, Cardiff University, Heath Park, Cardiff CF14 4XY, UK; Cardiff Institute of Tissue Engineering and Repair, Cardiff University, 10 Museum Place, Cardiff, South Glamorgan, CF10 3BG, UK
| |
Collapse
|
5
|
Sanberg PR, Greene-Zavertnik CR. Article Commentary: Stem Cells and Development Publishes Neural Stem Cells Compendium. Cell Transplant 2017; 14:855-857. [DOI: 10.3727/000000005783982459] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Paul R. Sanberg
- University of South Florida College of Medicine, Tampa, FL, USA
| | | |
Collapse
|
6
|
Kazim SF, Cardenas-Aguayo MDC, Arif M, Blanchard J, Fayyaz F, Grundke-Iqbal I, Iqbal K. Sera from children with autism induce autistic features which can be rescued with a CNTF small peptide mimetic in rats. PLoS One 2015; 10:e0118627. [PMID: 25769033 PMCID: PMC4359103 DOI: 10.1371/journal.pone.0118627] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 01/21/2015] [Indexed: 12/29/2022] Open
Abstract
Autism is a neurodevelopmental disorder characterized clinically by impairments in social interaction and verbal and non-verbal communication skills as well as restricted interests and repetitive behavior. It has been hypothesized that altered brain environment including an imbalance in neurotrophic support during early development contributes to the pathophysiology of autism. Here we report that sera from children with autism which exhibited abnormal levels of various neurotrophic factors induced cell death and oxidative stress in mouse primary cultured cortical neurons. The effects of sera from autistic children were rescued by pre-treatment with a ciliary neurotrophic factor (CNTF) small peptide mimetic, Peptide 6 (P6), which was previously shown to exert its neuroprotective effect by modulating CNTF/JAK/STAT pathway and LIF signaling and by enhancing brain derived neurotrophic factor (BDNF) expression. Similar neurotoxic effects and neuroinflammation were observed in young Wistar rats injected intracerebroventricularly with autism sera within hours after birth. The autism sera injected rats demonstrated developmental delay and deficits in social communication, interaction, and novelty. Both the neurobiological changes and the behavioral autistic phenotype were ameliorated by P6 treatment. These findings implicate the involvement of neurotrophic imbalance during early brain development in the pathophysiology of autism and a proof of principle of P6 as a potential therapeutic strategy for autism.
Collapse
Affiliation(s)
- Syed Faraz Kazim
- Inge Grundke-Iqbal Research Floor, Department of Neurochemistry, New York State Institute for Basic Research in Developmental Disabilities (NYSIBR), Staten Island, New York, United States of America
- Neural and Behavioral Science Graduate Program, State University of New York (SUNY) Downstate Medical Center, Brooklyn, New York, United States of America
- SUNY Downstate/NYSIBR Center for Developmental Neuroscience (CDN), Staten Island, New York, United States of America
| | - Maria del Carmen Cardenas-Aguayo
- Inge Grundke-Iqbal Research Floor, Department of Neurochemistry, New York State Institute for Basic Research in Developmental Disabilities (NYSIBR), Staten Island, New York, United States of America
| | - Mohammad Arif
- Inge Grundke-Iqbal Research Floor, Department of Neurochemistry, New York State Institute for Basic Research in Developmental Disabilities (NYSIBR), Staten Island, New York, United States of America
| | - Julie Blanchard
- Inge Grundke-Iqbal Research Floor, Department of Neurochemistry, New York State Institute for Basic Research in Developmental Disabilities (NYSIBR), Staten Island, New York, United States of America
| | - Fatima Fayyaz
- Inge Grundke-Iqbal Research Floor, Department of Neurochemistry, New York State Institute for Basic Research in Developmental Disabilities (NYSIBR), Staten Island, New York, United States of America
| | - Inge Grundke-Iqbal
- Inge Grundke-Iqbal Research Floor, Department of Neurochemistry, New York State Institute for Basic Research in Developmental Disabilities (NYSIBR), Staten Island, New York, United States of America
| | - Khalid Iqbal
- Inge Grundke-Iqbal Research Floor, Department of Neurochemistry, New York State Institute for Basic Research in Developmental Disabilities (NYSIBR), Staten Island, New York, United States of America
- SUNY Downstate/NYSIBR Center for Developmental Neuroscience (CDN), Staten Island, New York, United States of America
- * E-mail:
| |
Collapse
|
7
|
Kim HS, Choi SI, Jeung EB, Yoo YM. Cyclosporine A induces apoptotic and autophagic cell death in rat pituitary GH3 cells. PLoS One 2014; 9:e108981. [PMID: 25299210 PMCID: PMC4191984 DOI: 10.1371/journal.pone.0108981] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Accepted: 09/02/2014] [Indexed: 12/13/2022] Open
Abstract
Cyclosporine A (CsA) is a powerful immunosuppressive drug with side effects including the development of chronic nephrotoxicity. In this study, we investigated CsA treatment induced apoptotic and autophagic cell death in pituitary GH3 cells. CsA treatment (0.1 to 10 µM) decreased survival of GH3 cells in a dose-dependent manner. Cell viability decreased significantly with increasing CsA concentrations largely due to an increase in apoptosis, while cell death rates due to autophagy altered only slightly. Several molecular and morphological features correlated with cell death through these distinct pathways. At concentrations ranging from 1.0 to 10 µM, CsA induced a dose-dependent increase in expression of the autophagy markers LC3-I and LC3-II. Immunofluorescence staining revealed markedly increased levels of both LC3 and lysosomal-associated membrane protein 2 (Lamp2), indicating increases in autophagosomes. At the same CsA doses, apoptotic cell death was apparent as indicated by nuclear and DNA fragmentation and increased p53 expression. In apoptotic or autophagic cells, p-ERK levels were highest at 1.0 µM CsA compared to control or other doses. In contrast, Bax levels in both types of cell death were increased in a dose-dependent manner, while Bcl-2 levels showed dose-dependent augmentation in autophagy and were decreased in apoptosis. Manganese superoxide dismutase (Mn-SOD) showed a similar dose-dependent reduction in cells undergoing apoptosis, while levels of the intracellular calcium ion exchange maker calbindin-D9k were decreased in apoptosis (1.0 to 5 µM CsA), but unchanged in autophagy. In conclusion, these results suggest that CsA induction of apoptotic or autophagic cell death in rat pituitary GH3 cells depends on the relative expression of factors and correlates with Bcl-2 and Mn-SOD levels.
Collapse
Affiliation(s)
- Han Sung Kim
- Department of Biomedical Engineering, College of Health Science, Yonsei University, Wonju, Gangwon-do, Republic of Korea
| | - Seung-Il Choi
- Cornea Dystrophy Research Institute and Department of Ophthalmology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Eui-Bae Jeung
- Laboratory of Veterinary Biochemistry and Molecular Biology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea
- * E-mail: (YMY); (EBJ)
| | - Yeong-Min Yoo
- Department of Biomedical Engineering, College of Health Science, Yonsei University, Wonju, Gangwon-do, Republic of Korea
- * E-mail: (YMY); (EBJ)
| |
Collapse
|
8
|
Disease modifying effect of chronic oral treatment with a neurotrophic peptidergic compound in a triple transgenic mouse model of Alzheimer's disease. Neurobiol Dis 2014; 71:110-30. [PMID: 25046994 DOI: 10.1016/j.nbd.2014.07.001] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2014] [Revised: 05/28/2014] [Accepted: 07/08/2014] [Indexed: 12/16/2022] Open
Abstract
Besides the presence of amyloid beta (Aβ) plaques and neurofibrillary tangles, neurogenesis and synaptic plasticity are markedly impaired in Alzheimer's disease (AD) possibly contributing to cognitive impairment. In this context, neurotrophic factors serve as a promising therapeutic approach via utilization of regenerative capacity of brain to shift the balance from neurodegeneration to neural regeneration. However, besides more conventional "bystander" effect, to what extent can neurotrophic compounds affect underlying AD pathology remains questionable. Here we investigated the effect of chronic oral treatment with a ciliary neurotrophic factor (CNTF) derived peptidergic compound, P021 (Ac-DGGL(A)G-NH2), on disease pathology both at moderate and severe stages in a transgenic mouse model of AD. 3xTg-AD and wild type female mice were treated for 12months with P021 or vehicle diet starting at 9-10months of age. A significant reduction in abnormal hyperphosphorylation and accumulation of tau at known major AD neurofibrillary pathology associated sites was observed. The effect of P021 on Aβ pathology was limited to a significant decrease in soluble Aβ levels and a trend towards reduction in Aβ plaque load in CA1 region of hippocampus, consistent with reduction in Aβ generation and not clearance. This disease modifying effect was probably via increased brain derived neurotrophic factor (BDNF) expression mediated decrease in glycogen synthase kinase-3-β (GSK3β) activity we found in P021 treated 3xTg-AD mice. P021 treatment also rescued deficits in cognition, neurogenesis, and synaptic plasticity in 3xTg-AD mice. These findings demonstrate the potential of the neurotrophic peptide mimetic as a disease modifying therapy for AD.
Collapse
|
9
|
Tiwari M, Sharma LK, Vanegas D, Callaway DA, Bai Y, Lechleiter JD, Herman B. A nonapoptotic role for CASP2/caspase 2: modulation of autophagy. Autophagy 2014. [PMID: 24879153 DOI: 10.4161/auto.28528.erratum.in:autophagy.2017;13(3):637] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023] Open
Abstract
CASP2/caspase 2 plays a role in aging, neurodegeneration, and cancer. The contributions of CASP2 have been attributed to its regulatory role in apoptotic and nonapoptotic processes including the cell cycle, DNA repair, lipid biosynthesis, and regulation of oxidant levels in the cells. Previously, our lab demonstrated CASP2-mediated modulation of autophagy during oxidative stress. Here we report the novel finding that CASP2 is an endogenous repressor of autophagy. Knockout or knockdown of CASP2 resulted in upregulation of autophagy in a variety of cell types and tissues. Reinsertion of Caspase-2 gene (Casp2) in mouse embryonic fibroblast (MEFs) lacking Casp2 (casp2(-/-)) suppresses autophagy, suggesting its role as a negative regulator of autophagy. Loss of CASP2-mediated autophagy involved AMP-activated protein kinase, mechanistic target of rapamycin, mitogen-activated protein kinase, and autophagy-related proteins, indicating the involvement of the canonical pathway of autophagy. The present study also demonstrates an important role for loss of CASP2-induced enhanced reactive oxygen species production as an upstream event in autophagy induction. Additionally, in response to a variety of stressors that induce CASP2-mediated apoptosis, casp2(-/-) cells demonstrate a further upregulation of autophagy compared with wild-type MEFs, and upregulated autophagy provides a survival advantage. In conclusion, we document a novel role for CASP2 as a negative regulator of autophagy, which may provide important insight into the role of CASP2 in various processes including aging, neurodegeneration, and cancer.
Collapse
Affiliation(s)
- Meenakshi Tiwari
- Department of Cellular and Structural Biology; University of Texas Health Science Center at San Antonio; South Texas Research Facility; San Antonio, TX USA; Department of Pathology and Laboratory Medicine; All India Institute of Medical Sciences; Patna, India
| | - Lokendra K Sharma
- Department of Cellular and Structural Biology; University of Texas Health Science Center at San Antonio; South Texas Research Facility; San Antonio, TX USA
| | - Difernando Vanegas
- Department of Cellular and Structural Biology; University of Texas Health Science Center at San Antonio; South Texas Research Facility; San Antonio, TX USA
| | - Danielle A Callaway
- Department of Cellular and Structural Biology; University of Texas Health Science Center at San Antonio; South Texas Research Facility; San Antonio, TX USA
| | - Yidong Bai
- Department of Cellular and Structural Biology; University of Texas Health Science Center at San Antonio; South Texas Research Facility; San Antonio, TX USA
| | - James D Lechleiter
- Department of Cellular and Structural Biology; University of Texas Health Science Center at San Antonio; South Texas Research Facility; San Antonio, TX USA
| | - Brian Herman
- Department of Cellular and Structural Biology; University of Texas Health Science Center at San Antonio; South Texas Research Facility; San Antonio, TX USA
| |
Collapse
|
10
|
Tiwari M, Sharma LK, Vanegas D, Callaway DA, Bai Y, Lechleiter JD, Herman B. A nonapoptotic role for CASP2/caspase 2: modulation of autophagy. Autophagy 2014; 10:1054-1070. [PMID: 24879153 PMCID: PMC4091168 DOI: 10.4161/auto.28528] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2013] [Revised: 03/09/2014] [Accepted: 03/13/2014] [Indexed: 12/19/2022] Open
Abstract
CASP2/caspase 2 plays a role in aging, neurodegeneration, and cancer. The contributions of CASP2 have been attributed to its regulatory role in apoptotic and nonapoptotic processes including the cell cycle, DNA repair, lipid biosynthesis, and regulation of oxidant levels in the cells. Previously, our lab demonstrated CASP2-mediated modulation of autophagy during oxidative stress. Here we report the novel finding that CASP2 is an endogenous repressor of autophagy. Knockout or knockdown of CASP2 resulted in upregulation of autophagy in a variety of cell types and tissues. Reinsertion of Caspase-2 gene (Casp2) in mouse embryonic fibroblast (MEFs) lacking Casp2 (casp2(-/-)) suppresses autophagy, suggesting its role as a negative regulator of autophagy. Loss of CASP2-mediated autophagy involved AMP-activated protein kinase, mechanistic target of rapamycin, mitogen-activated protein kinase, and autophagy-related proteins, indicating the involvement of the canonical pathway of autophagy. The present study also demonstrates an important role for loss of CASP2-induced enhanced reactive oxygen species production as an upstream event in autophagy induction. Additionally, in response to a variety of stressors that induce CASP2-mediated apoptosis, casp2(-/-) cells demonstrate a further upregulation of autophagy compared with wild-type MEFs, and upregulated autophagy provides a survival advantage. In conclusion, we document a novel role for CASP2 as a negative regulator of autophagy, which may provide important insight into the role of CASP2 in various processes including aging, neurodegeneration, and cancer.
Collapse
Affiliation(s)
- Meenakshi Tiwari
- Department of Cellular and Structural Biology; University of Texas Health Science Center at San Antonio; South Texas Research Facility; San Antonio, TX USA
- Department of Pathology and Laboratory Medicine; All India Institute of Medical Sciences; Patna, India
| | - Lokendra K Sharma
- Department of Cellular and Structural Biology; University of Texas Health Science Center at San Antonio; South Texas Research Facility; San Antonio, TX USA
| | - Difernando Vanegas
- Department of Cellular and Structural Biology; University of Texas Health Science Center at San Antonio; South Texas Research Facility; San Antonio, TX USA
| | - Danielle A Callaway
- Department of Cellular and Structural Biology; University of Texas Health Science Center at San Antonio; South Texas Research Facility; San Antonio, TX USA
| | - Yidong Bai
- Department of Cellular and Structural Biology; University of Texas Health Science Center at San Antonio; South Texas Research Facility; San Antonio, TX USA
| | - James D Lechleiter
- Department of Cellular and Structural Biology; University of Texas Health Science Center at San Antonio; South Texas Research Facility; San Antonio, TX USA
| | - Brian Herman
- Department of Cellular and Structural Biology; University of Texas Health Science Center at San Antonio; South Texas Research Facility; San Antonio, TX USA
| |
Collapse
|
11
|
Booth LA, Tavallai S, Hamed HA, Cruickshanks N, Dent P. The role of cell signalling in the crosstalk between autophagy and apoptosis. Cell Signal 2014; 26:549-55. [PMID: 24308968 PMCID: PMC4054685 DOI: 10.1016/j.cellsig.2013.11.028] [Citation(s) in RCA: 277] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2013] [Revised: 11/21/2013] [Accepted: 11/26/2013] [Indexed: 12/12/2022]
Abstract
Not surprisingly, the death of a cell is a complex and well controlled process. For several decades, apoptosis, the first genetically programmed death process to be identified has taken centre stage as the principal mechanism of programmed cell death (type I cell death) in mammalian tissues. Apoptosis has been extensively studied and its contribution to the pathogenesis of disease well documented. However, apoptosis does not function alone in determining the fate of a cell. More recently, autophagy, a process in which de novo formed membrane enclosed vesicles engulf and consume cellular components, has been shown to engage in complex interplay with apoptosis. As a result, cell death has been subdivided into the categories apoptosis (Type I), autophagic cell death (Type II), and necrosis (Type III). The boundary between Type I and II cell death is not completely clear and as we will discuss in this review and perhaps a discrete difference does not exist, due to intrinsic factors among different cell types and crosstalk among organelles within each cell type. Apoptosis may begin with autophagy and autophagy can often end with apoptosis, inhibition or a blockade of caspase activity may lead a cell to default into Type II cell death from Type I.
Collapse
Affiliation(s)
- Laurence A Booth
- Department of Neurosurgery, Virginia Commonwealth University, 401 College St, Richmond, VA 23298, United States.
| | - Seyedmehrad Tavallai
- Department of Neurosurgery, Virginia Commonwealth University, 401 College St, Richmond, VA 23298, United States
| | - Hossein A Hamed
- Department of Neurosurgery, Virginia Commonwealth University, 401 College St, Richmond, VA 23298, United States
| | - Nichola Cruickshanks
- Department of Neurosurgery, Virginia Commonwealth University, 401 College St, Richmond, VA 23298, United States
| | - Paul Dent
- Department of Neurosurgery, Virginia Commonwealth University, 401 College St, Richmond, VA 23298, United States; Virginia Institute of Molecular Medicine, Virginia Commonwealth University, 401 College St, Richmond, VA 23298, United States
| |
Collapse
|
12
|
Eom YW, Oh JE, Lee JI, Baik SK, Rhee KJ, Shin HC, Kim YM, Ahn CM, Kong JH, Kim HS, Shim KY. The role of growth factors in maintenance of stemness in bone marrow-derived mesenchymal stem cells. Biochem Biophys Res Commun 2014; 445:16-22. [PMID: 24491556 DOI: 10.1016/j.bbrc.2014.01.084] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Accepted: 01/18/2014] [Indexed: 01/13/2023]
Abstract
Mesenchymal stem cells (MSCs) are an active topic of research in regenerative medicine due to their ability to secrete a variety of growth factors and cytokines that promote healing of damaged tissues and organs. In addition, these secreted growth factors and cytokines have been shown to exert an autocrine effect by regulating MSC proliferation and differentiation. We found that expression of EGF, FGF-4 and HGF were down-regulated during serial passage of bone marrow-derived mesenchymal stem cells (BMSCs). Proliferation and differentiation potentials of BMSCs treated with these growth factors for 2 months were evaluated and compared to BMSCs treated with FGF-2, which increased proliferation of BMSCs. FGF-2 and -4 increased proliferation potentials at high levels, about 76- and 26-fold, respectively, for 2 months, while EGF and HGF increased proliferation of BMSCs by less than 2.8-fold. Interestingly, differentiation potential, especially adipogenesis, was maintained only by HGF treatment. Treatment with FGF-2 rapidly induced activation of AKT and later induced ERK activation. The basal level of phosphorylated ERK increased during serial passage of BMSCs treated with FGF-2. The expression of LC3-II, an autophagy marker, was gradually increased and the population of senescent cells was increased dramatically at passage 7 in non-treated controls. But FGF-2 and FGF-4 suppressed LC3-II expression and down-regulated senescent cells during long-term (i.e. 2month) cultures. Taken together, depletion of growth factors during serial passage could induce autophagy, senescence and down-regulation of stemness (proliferation via FGF-2/-4 and differentiation via HGF) through suppression of AKT and ERK signaling.
Collapse
Affiliation(s)
- Young Woo Eom
- Cell Therapy and Tissue Engineering Center, Wonju College of Medicine, Yonsei Univ., Wonju, Republic of Korea
| | - Ji-Eun Oh
- Cell Therapy and Tissue Engineering Center, Wonju College of Medicine, Yonsei Univ., Wonju, Republic of Korea
| | - Jong In Lee
- Department of Hematology-Oncology, Wonju College of Medicine, Yonsei Univ., Wonju, Republic of Korea
| | - Soon Koo Baik
- Cell Therapy and Tissue Engineering Center, Wonju College of Medicine, Yonsei Univ., Wonju, Republic of Korea; Department of Internal Medicine, Wonju College of Medicine, Yonsei Univ., Wonju, Republic of Korea
| | - Ki-Jong Rhee
- Department of Biomedical Laboratory Science, College of Health Sciences, Yonsei Univ., Wonju, Republic of Korea
| | | | - Yong Man Kim
- Pharmicell Co., Ltd., Sungnam, Republic of Korea
| | - Chan Mug Ahn
- Department of Basic Science, Wonju College of Medicine, Yonsei Univ., Wonju, Republic of Korea
| | - Jee Hyun Kong
- Department of Hematology-Oncology, Wonju College of Medicine, Yonsei Univ., Wonju, Republic of Korea
| | - Hyun Soo Kim
- Pharmicell Co., Ltd., Sungnam, Republic of Korea.
| | - Kwang Yong Shim
- Department of Hematology-Oncology, Wonju College of Medicine, Yonsei Univ., Wonju, Republic of Korea.
| |
Collapse
|
13
|
Cardenas-Aguayo MDC, Kazim SF, Grundke-Iqbal I, Iqbal K. Neurogenic and neurotrophic effects of BDNF peptides in mouse hippocampal primary neuronal cell cultures. PLoS One 2013; 8:e53596. [PMID: 23320097 PMCID: PMC3539976 DOI: 10.1371/journal.pone.0053596] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2012] [Accepted: 11/30/2012] [Indexed: 12/11/2022] Open
Abstract
The level of brain-derived neurotrophic factor (BDNF), a member of the neurotrophin family, is down regulated in Alzheimer’s disease (AD), Parkinson’s disease (PD), depression, stress, and anxiety; conversely the level of this neurotrophin is increased in autism spectrum disorders. Thus, modulating the level of BDNF can be a potential therapeutic approach for nervous system pathologies. In the present study, we designed five different tetra peptides (peptides B-1 to B-5) corresponding to different active regions of BDNF. These tetra peptides were found to be non-toxic, and they induced the expression of neuronal markers in mouse embryonic day 18 (E18) primary hippocampal neuronal cultures. Additionally, peptide B-5 induced the expression of BDNF and its receptor, TrkB, suggesting a positive feedback mechanism. The BDNF peptides induced only a moderate activation (phosphorylation at Tyr 706) of the TrkB receptor, which could be blocked by the Trk’s inhibitor, K252a. Peptide B-3, when combined with BDNF, potentiated the survival effect of this neurotrophin on H2O2-treated E18 hippocampal cells. Peptides B-3 and B-5 were found to work as partial agonists and as partial antagonists competing with BDNF to activate the TrkB receptor in a dose-dependent manner. Taken together, these results suggest that the described BDNF tetra peptides are neurotrophic, can modulate BDNF signaling in a partial agonist/antagonist way, and offer a novel therapeutic approach to neural pathologies where BDNF levels are dysregulated.
Collapse
Affiliation(s)
- Maria del Carmen Cardenas-Aguayo
- Department of Neurochemistry, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, New York, United States of America
| | - Syed Faraz Kazim
- Department of Neurochemistry, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, New York, United States of America
- Neural and Behavioral Science Graduate Program, State University of New York Downstate Medical Center, Brooklyn, New York, United States of America
| | - Inge Grundke-Iqbal
- Department of Neurochemistry, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, New York, United States of America
| | - Khalid Iqbal
- Department of Neurochemistry, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, New York, United States of America
- * E-mail:
| |
Collapse
|
14
|
Li H, Jin X, Zhang Z, Xing Y, Kong X. Inhibition of autophagy enhances apoptosis induced by the PI3K/AKT/mTor inhibitor NVP-BEZ235 in renal cell carcinoma cells. Cell Biochem Funct 2012; 31:427-33. [PMID: 23086777 DOI: 10.1002/cbf.2917] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2012] [Revised: 09/18/2012] [Accepted: 09/19/2012] [Indexed: 12/16/2022]
Affiliation(s)
- Hongyan Li
- Department of Urology, China-Japan Union Hospital; Jilin University; Changchun; Jilin; China
| | - Xuefei Jin
- Department of Urology, China-Japan Union Hospital; Jilin University; Changchun; Jilin; China
| | - Zhuo Zhang
- Department of Urology, China-Japan Union Hospital; Jilin University; Changchun; Jilin; China
| | - Yuanyuan Xing
- Department of Urology, China-Japan Union Hospital; Jilin University; Changchun; Jilin; China
| | - Xiangbo Kong
- Department of Urology, China-Japan Union Hospital; Jilin University; Changchun; Jilin; China
| |
Collapse
|
15
|
Abstract
We have analysed telomerase activity to determine whether it can be modified when BCL-2 is endogenously overexpressed in response to a mild oxidative stress treatment as part of a survival mechanism, in contrast with an exogenous bcl-2 overexpression due to a retroviral infection. Endogenous bcl-2 overexpression was induced after a low oxidative insult of H2O2 in mice primary lung fibroblasts and L929 cell, whereas bcl-2 exogenous overexpression was performed using a retroviral infection in L929 cells. Telomerase activity was quantified in Bcl-2 overexpressing cells by the TRAP assay. When the cells were treated with different H2O2 concentrations, only those exposed to 50 μM showed increased telomerase activity. This correlates with BCL-2 expression as part of the endogenous response to mild oxidative stress. Oxidative stress generated during the toxic mechanism of chemotherapeutic drugs might induce BCL-2 increment, enhancing telomerase activity and reactivating the oncogenic process. Clinical trials should take into consideration the possibility of telomerase activation following increased BCL-2 expression when treating patients with ROS (reactive oxygen species) generation by anti-cancer drugs.
Collapse
|
16
|
Ro SH, Liu D, Yeo H, Paik JH. FoxOs in neural stem cell fate decision. Arch Biochem Biophys 2012; 534:55-63. [PMID: 22902436 DOI: 10.1016/j.abb.2012.07.017] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2012] [Revised: 07/30/2012] [Accepted: 07/31/2012] [Indexed: 02/07/2023]
Abstract
Neural stem cells (NSCs) persist over the lifespan of mammals to give rise to committed progenitors and their differentiated cells in order to maintain the brain homeostasis. To this end, NSCs must be able to self-renew and otherwise maintain their quiescence. Suppression of aberrant proliferation or undesired differentiation is crucial to preclude either malignant growth or precocious depletion of NSCs. The PI3K-Akt-FoxO signaling pathway plays a central role in the regulation of multiple stem cells including one in the mammalian brain. In particular, members of FoxO family transcription factors are highly expressed in these stem cells. As an important downstream effector of growth, differentiation, and stress stimuli, mammalian FoxO transcription factor family controls cellular proliferation, oxidative stress response, homeostasis, and eventual maintenance of long-term repopulating potential. The review will focus on the current understanding of FoxO function in NSCs as well as discuss their biological activities that contribute to determining neural stem cell fate.
Collapse
Affiliation(s)
- Seung-Hyun Ro
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | | | | | | |
Collapse
|
17
|
Tabor-Godwin JM, Tsueng G, Sayen MR, Gottlieb RA, Feuer R. The role of autophagy during coxsackievirus infection of neural progenitor and stem cells. Autophagy 2012; 8:938-53. [PMID: 22751470 DOI: 10.4161/auto.19781] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Coxsackievirus B3 (CVB3) has previously been shown to utilize autophagy in an advantageous manner during the course of infection of the host cell. However, few studies have determined whether stem cells induce autophagy in a similar fashion, and whether virus-induced autophagy occurs following infection of stem cells. Therefore, we compared the induction of autophagy following CVB3 infection of neural progenitor and stem cells (NPSCs), which we have recently shown to be highly susceptible to CVB3 infection, to HL-1 cells, a transformed cardiomyocyte cell line. As previously demonstrated for other susceptible host cells, HL-1 cells showed an increase in the activity of autophagic signaling following infection with a CVB3 expressing dsRed protein (dsRed-CVB3). Furthermore, viral titers in HL-1 cells increased in the presence of an inducer of autophagy (CCPA), while viral titers decreased in the presence of an inhibitor of autophagy (3-MA). In contrast, no change in autophagic signaling was seen in NPSCs following infection with dsRed-CVB3. Also, basal levels of autophagy in NPSCs were found to be highly elevated in comparison to HL-1 cells. Autophagy could be induced in NPSCs in the presence of rapamycin without altering levels of dsRed-CVB3 replication. In differentiated NPSC precursors, autophagy was activated during the differentiation process, and a decrease in autophagic signaling was observed within all three CNS lineages following dsRed-CVB3 infection. Hence, we conclude that the role of autophagy in modulating CVB3 replication appears cell type-specific, and stem cells may uniquely regulate autophagy in response to infection.
Collapse
Affiliation(s)
- Jenna M Tabor-Godwin
- Cell & Molecular Biology Joint Doctoral Program, Department of Biology, San Diego State University, San Diego, CA, USA
| | | | | | | | | |
Collapse
|
18
|
Transduction of E13 murine neural precursor cells by non-immunogenic recombinant adeno-associated viruses induces major changes in neuronal phenotype. Neuroscience 2012; 210:82-98. [PMID: 22406416 DOI: 10.1016/j.neuroscience.2012.02.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2011] [Revised: 02/03/2012] [Accepted: 02/06/2012] [Indexed: 11/21/2022]
Abstract
Neural precursor cells (NPCs) provide a cellular model to compare transduction efficiency and toxicity for a series of recombinant adeno-associated viruses (rAAVs). Results led to the choice of rAAV9 as a preferred candidate to transduce NPCs for in vivo transplantation. Importantly, transduction promoted a neuronal phenotype characterized by neurofilament M (NFM) with a concomitant decrease in the embryonic marker, nestin, without significant change in glial fibrillary acidic protein (GFAP). In marked contrast to recent studies for induced pluripotent stem cells (iPSCs), exposure to rAAVs is non-immunogenic and these do not result in genetic abnormalities, thus bolstering the earlier use of NPCs such as those isolated from E13 murine cells for clinical applications. Mechanisms of cellular interactions were explored by treatment with genistein, a pan-specific inhibitor of protein receptor tyrosine kinases (PRTKs) that blocked the transduction and differentiation, thus implying a central role for this pathway for inducing infectivity along with observed phenotypic changes and as a method for drug design. Implantation of transduced NPCs into adult mouse hippocampus survived up to 28 days producing a time line for targeting or migration to dentate gyrus and CA3-1 compatible with future clinical applications. Furthermore, a majority showed commitment to highly differentiated neuronal phenotypes. Lack of toxicity and immune response of rAAVs plus ability for expansion of NPCs in vitro auger well for their isolation and suggest potential therapeutic applications in repair or replacement of diseased neurons in neurodegeneration.
Collapse
|
19
|
Abe A, Yamada H, Moriya S, Miyazawa K. The .BETA.-Carboline Alkaloid Harmol Induces Cell Death via Autophagy but Not Apoptosis in Human Non-small Cell Lung Cancer A549 Cells. Biol Pharm Bull 2011; 34:1264-72. [DOI: 10.1248/bpb.34.1264] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Akihisa Abe
- Department of Biochemistry, Tokyo Medical University
| | - Hiroyuki Yamada
- Bacteriology Division, Mycobacterium Reference Center, Research Institute of Tuberculosis, Japan Anti-Tuberculosis Association (JATA)
| | - Shota Moriya
- Department of Biochemistry, Tokyo Medical University
| | | |
Collapse
|
20
|
González-Puertos VY, Hernández-Pérez E, Nuño-Lámbarri N, Ventura-Gallegos JL, López-Diázguerrero NE, Robles-Díaz G, Gutiérrez-Ruiz MC, Konigsberg M. Bcl-2 overexpression in hepatic stellate cell line CFSC-2G, induces a pro-fibrotic state. J Gastroenterol Hepatol 2010; 25:1306-14. [PMID: 20594261 DOI: 10.1111/j.1440-1746.2009.06175.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
BACKGROUND AND AIM Development of hepatic fibrosis is a complex process that involves oxidative stress (OS) and an altered balance between pro- and anti-apoptotic molecules. Since Bcl-2 overexpression preserves viability against OS, our objective was to address the effect of Bcl-2 overexpression in the hepatic stellate cells (HSC) cell-line CFSC-2G under acetaldehyde and H(2)O(2) challenge, and explore if it protects these cells against OS, induces replicative senescence and/or modify extracellular matrix (ECM) remodeling potential. METHODS To induce Bcl-2 overexpression, HSC cell line CFSC-2G was transfected by lipofection technique. Green fluorescent protein-only CFSC-2G cells were used as a control. Cell survival after H(2)O(2) treatment and total protein oxidation were assessed. To determine cell cycle arrest, proliferation-rate, DNA synthesis and senescence were assessed. Matrix metalloproteinases (MMP), tissue-inhibitor of MMP (TIMP), transglutaminases (TG) and smooth muscle a-actin (alpha-SMA) were evaluated by western blot in response to acetaldehyde treatment as markers of ECM remodeling capacity in addition to transforming growth factor-beta (TGF-beta) mRNA. RESULTS Cells overexpressing Bcl-2 survived approximately 20% more than control cells when exposed to H(2)O(2) and approximately 35% proteins were protected from oxidation, but Bcl-2 did not slow proliferation or induced senescence. Bcl-2 overexpression did not change alpha-SMA levels, but it increased TIMP-1 (55%), tissue transglutaminases (tTG) (25%) and TGF-beta mRNA (49%), when exposed to acetaldehyde, while MMP-13 content decreased (47%). CONCLUSIONS Bcl-2 overexpression protected HSC against oxidative stress but it did not induce replicative senescence. It increased TIMP-1, tTG and TGF-beta mRNA levels and decreased MMP-13 content, suggesting that Bcl-2 overexpression may play a key role in the progression of fibrosis in chronic liver diseases.
Collapse
Affiliation(s)
- Viridiana Y González-Puertos
- Health and Sciences Department, Biological and Health Sciences Division, Metropolitan Independent University, México, DF, México
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Priault M, Hue E, Marhuenda F, Pilet P, Oliver L, Vallette FM. Differential dependence on Beclin 1 for the regulation of pro-survival autophagy by Bcl-2 and Bcl-xL in HCT116 colorectal cancer cells. PLoS One 2010; 5:e8755. [PMID: 20090905 PMCID: PMC2807451 DOI: 10.1371/journal.pone.0008755] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2009] [Accepted: 12/15/2009] [Indexed: 12/20/2022] Open
Abstract
Autophagy is described to be involved in homeostasis, development and disease, both as a survival and a death process. Its involvement in cell death proceeds from interrelationships with the apoptotic pathway. We focused on survival autophagy and investigated its interplays with the apoptotic machinery. We found that while Mcl-1 remained ineffective, Bcl-2 and Bcl-xL were required for starved cells to display a fully functional autophagic pathway as shown by proteolysis activity and detection of autophagic vesicles. Such pro-autophagic functions of Bcl-2 and Bcl-xL were independent of Bax. However they appeared to operate through non redundant mechanisms as Bcl-xL wielded a tighter control than Bcl-2 over the regulation of autophagy: unlike Bcl-2, Bcl-xL and Atg7 manipulation yielded identical phenotypes suggesting they could be components of the same signalling pathway; Bcl-xL subcellular localisation was modified upon starvation, and importantly Bcl-xL acted independently of Beclin 1. Still an intact BH3-binding site was required for Bcl-xL to stimulate a fully functional autophagic pathway. This study highlights that, in addition to their well-established anti-death function during apoptosis, Bcl-2 and Bcl-xL have a broader role in cell survival. Should Bcl-2 and Bcl-xL stand at the cross-roads between pro-survival and pro-death autophagy, this study introduces the new concept that the regulation of autophagy by Bcl-2 and Bcl-xL is adjusted according to its survival or death outcome.
Collapse
|
22
|
Kimura A, Ishida Y, Wada T, Hisaoka T, Morikawa Y, Sugaya T, Mukaida N, Kondo T. The absence of interleukin-6 enhanced arsenite-induced renal injury by promoting autophagy of tubular epithelial cells with aberrant extracellular signal-regulated kinase activation. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 176:40-50. [PMID: 20008137 PMCID: PMC2797868 DOI: 10.2353/ajpath.2010.090146] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 09/03/2009] [Indexed: 12/28/2022]
Abstract
Sodium arsenite (NaAs)-induced autophagic cell death (ACD) of a mouse renal tubular epithelial cell line (mProx24), which expresses enhanced levels of interleukin-6 (IL-6), was reduced by the suppression of autophagy by 3-methyladenine or Atg7 knockdown. The inhibition of the IL-6/signal transducer and activator of transcription 3 (STAT3) signal pathway by anti-IL-6 antibody or a Jak2 inhibitor (AG490) exaggerated ACD of mProx24 cells after NaAs challenge, attenuating STAT3 activation and reciprocally enhancing extracellular signal-regulated kinase (ERK) phosphorylation. In contrast, an ERK inhibitor, PD98059, reduced NaAs-induced ACD in mProx24 cells. Subcutaneous injection of NaAs (12.5 mg/kg) into BALB/c (wild-type) mice enhanced intrarenal expression of IL-6, mainly produced by tubular cells, and caused severe renal injury characterized by hemorrhages, acute tubular necrosis, cast formation, and brush border disappearance, with increases in serum urea nitrogen (blood urea nitrogen) and creatinine levels. In addition, IL-6-deficient (IL-6(-/-)) mice exhibited exaggerated histopathological changes with higher blood urea nitrogen and creatinine levels. Moreover, in IL-6(-/-) mice treated with NaAs, ACD in renal tubular cells was significantly augmented, along with diminished STAT3 activation and reciprocal enhancement of ERK signaling, compared with wild-type mice. Finally, the administration of exogenous IL-6 into wild-type mice significantly reduced NaAs-induced ACD along with diminished ERK activation and eventually alleviated acute renal dysfunction. Thus, IL-6/STAT3 signal pathway could inhibit ERK activation, a crucial step for ACD, eventually attenuating NaAs-induced renal dysfunction.
Collapse
Affiliation(s)
- Akihiko Kimura
- Department of Forensic Medicine, Wakayama Medical University, Wakayama, Japan
| | - Yuko Ishida
- Department of Forensic Medicine, Wakayama Medical University, Wakayama, Japan
| | - Takashi Wada
- Department of Laboratory Medicine, Division of Nephrology, Kanazawa University, Kanazawa, Japan
| | - Tomoko Hisaoka
- Departments of Anatomy and Neurobiology, Wakayama Medical University, Wakayama, Japan
| | - Yoshihiro Morikawa
- Departments of Anatomy and Neurobiology, Wakayama Medical University, Wakayama, Japan
| | - Takeshi Sugaya
- Discovery Research Laboratory, Tanabe Seiyaku Company Limited, Osaka, Japan
| | - Naofumi Mukaida
- Division of Molecular Bioregulation, Kanazawa University Cancer Research Institute, Kanazawa, Japan
| | - Toshikazu Kondo
- Department of Forensic Medicine, Wakayama Medical University, Wakayama, Japan
| |
Collapse
|
23
|
Jaeger PA, Wyss-Coray T. All-you-can-eat: autophagy in neurodegeneration and neuroprotection. Mol Neurodegener 2009; 4:16. [PMID: 19348680 PMCID: PMC2679749 DOI: 10.1186/1750-1326-4-16] [Citation(s) in RCA: 132] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2008] [Accepted: 04/06/2009] [Indexed: 12/12/2022] Open
Abstract
Autophagy is the major pathway involved in the degradation of proteins and organelles, cellular remodeling, and survival during nutrient starvation. Autophagosomal dysfunction has been implicated in an increasing number of diseases from cancer to bacterial and viral infections and more recently in neurodegeneration. While a decrease in autophagic activity appears to interfere with protein degradation and possibly organelle turnover, increased autophagy has been shown to facilitate the clearance of aggregation-prone proteins and promote neuronal survival in a number of disease models. On the other hand, too much autophagic activity can be detrimental as well and lead to cell death, suggesting the regulation of autophagy has an important role in cell fate decisions. An increasing number of model systems are now available to study the role of autophagy in the central nervous system and how it might be exploited to treat disease. We will review here the current knowledge of autophagy in the central nervous system and provide an overview of the various models that have been used to study acute and chronic neurodegeneration.
Collapse
Affiliation(s)
- Philipp A Jaeger
- Geriatric Research Education and Clinical Center, VA Palo Alto Health Care System, 3801 Miranda Ave, Palo Alto, California, USA.
| | | |
Collapse
|
24
|
Zhang S, Qi J, Sun L, Cheng B, Pan S, Zhou M, Sun X. Matrine induces programmed cell death and regulates expression of relevant genes based on PCR array analysis in C6 glioma cells. Mol Biol Rep 2009; 36:791-799. [PMID: 18392946 DOI: 10.1007/s11033-008-9247-y] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2008] [Accepted: 03/31/2008] [Indexed: 11/29/2022]
Abstract
Matrine, one of the main components extracted from Sophora flavescens Ait, has a wide range of pharmacological effects including anti-tumor activities on a number of cancer cell lines. This study has investigated whether matrine could also display anti-tumor action on rat C6 glioma cells. Exposure of C6 cells to matrine resulted in inhibition of proliferation and induction of apoptosis in a dose-dependent manner, as measured by the MTT assay and Flow cytometry. The Annexin V/PI staining further detected the apoptotic cells at both early and late phases of apoptosis. We used AO/EB staining to examine the programmed cell death of matrine-treated C6 cells, and showed that the death rate detected by AO/EB staining was higher than the apoptosis rate measured by Annexin V/PI staining, suggesting that autophagy, the Type II programmed cell death, may be involved in matrine-induced cell death, which was further confirmed by electronic microscopy. To explore the molecular mechanism, an apoptosis real-time PCR array was performed, which has demonstrated that 57 genes were at least 2-fold upregulated, and 11 genes were at least 2-fold downregulated in matrine-treated C6 cells, compared with untreated cells. However, the gene expression profiles could only partly and roughly explain molecular mechanisms of apoptosis and autophagy in matrine-treated C6 cells, thus further investigations are required to confirm the specific molecular pathways and related molecules responsible for the programmed cell death.
Collapse
Affiliation(s)
- Shujun Zhang
- Department of Pathology, The First Clinical Medical School of Harbin Medical University, Harbin 150012, China
| | | | | | | | | | | | | |
Collapse
|
25
|
Combination of adenoviral virotherapy and temozolomide chemotherapy eradicates malignant glioma through autophagic and apoptotic cell death in vivo. Br J Cancer 2009; 100:1154-64. [PMID: 19277041 PMCID: PMC2664399 DOI: 10.1038/sj.bjc.6604969] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Conditionally replicative adenoviruses (CRAds) represent a novel treatment strategy for malignant glioma. Recent studies suggest that the cytopathic effect elicited by these vectors is mediated through autophagy, a form of programmed cell death. Likewise, temozolomide (TMZ), a chemotherapeutic agent used for the treatment of malignant gliomas, also triggers autophagic cell death. In this study, we examined the potential to combine the two treatments in the setting of experimental glioma. In vitro, pretreatment with TMZ followed by CRAd-Surivin-pk7 enhanced cytotoxicity against a panel of glioma cell lines. Western blot analysis showed increased expression of BAX and p53, decreased expression of BCL2 and elevated level of APG5. Treatment with TMZ followed by CRAd-Survivin-pk7 (CRAd-S-pk7) led to a significant over-expression of autophagy markers, acidic vesicular organelles and light-chain 3 (LC3). These results were further evaluated in vivo, in which 90% of the mice with intracranial tumours were long-term survivors (>100 days) after treatment with TMZ and CRAd-S-pk7 (P<0.01). Analysis of tumours ex vivo showed expression of both LC3 and cleaved Caspase-3, proving that both autophagy and apoptosis are responsible for cell death in vivo. These results suggest that combination of chemovirotherapy offers a powerful tool against malignant glioma and should be further explored in the clinical setting.
Collapse
|
26
|
Qin AP, Zhang HL, Qin ZH. Mechanisms of lysosomal proteases participating in cerebral ischemia-induced neuronal death. Neurosci Bull 2008; 24:117-23. [PMID: 18369392 DOI: 10.1007/s12264-008-0117-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
There are three different types of cell death, including apoptosis (Type I), autophagic cell death (Type II), and necrosis (Type III). Ischemic neuronal death influences stroke development and progression. Lysosomes are important organelles having an acidic milieu to maintain cellular metabolism by degrading unneeded extra- and intracellular substances. Lysosomal enzymes, including cathepsins and some lipid hydrolases, when secreted following rupture of the lysosomal membrane, can be very harmful to their environment, which results in pathological destruction of cellular structures. Since lysosomes contain catalytic enzymes for degrading proteins, carbohydrates and lipids, it seems natural that they should participate in cellular death and dismantling. In this review, we discuss the recent developments in ischemic neuronal death, and present the possible molecular mechanisms that the lysosomal enzymes participate in the three different types of cell death in ischemic brain damage. Moreover, the research related to the selective cathepsin inhibitors may provide a novel therapeutic target for treating stroke and promoting recovery.
Collapse
Affiliation(s)
- Ai-Ping Qin
- Department of Pharmacology, Laboratory of Aging and Nervous Disease, Soochow University School of Medicine, Suzhou 215123, China
| | | | | |
Collapse
|
27
|
Bialik S, Kimchi A. Autophagy and tumor suppression: recent advances in understanding the link between autophagic cell death pathways and tumor development. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2008; 615:177-200. [PMID: 18437896 DOI: 10.1007/978-1-4020-6554-5_9] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Autophagy is a process by which the cell recycles its components through self-consumption of cellular organelles and bulk cytoplasm. In times of stress, it serves to generate much needed nutrients. When overactivated, however, the orderly destruction of organelles can lead to cell death. At times, autophagic cell death is used as an alternative to apoptosis to eliminate unwanted, damaged, or transformed cells. Consistent with this, tumorigenesis is associated with a downregulation in autophagy, and genes that mediate the execution of the process have been shown to be tumor suppressors. At the same time, basal autophagy has been harnessed by some tumor cells as a survival mechanism to protect against ischemia and signals that induce apoptosis. Thus, the relationship between autophagy and tumor development is complex. Here, we discuss the basic machinery of mammalian autophagy and its regulators, with specific emphasis on those genes that have been linked to cancer. Research supporting the divergent nature of autophagy in both tumor suppression and tumor progression is presented. We conclude with a survey of recent approaches to treating cancer with strategies that modulate autophagy.
Collapse
Affiliation(s)
- Shani Bialik
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | | |
Collapse
|
28
|
Luo S, Rubinsztein DC. Atg5 and Bcl-2 provide novel insights into the interplay between apoptosis and autophagy. Cell Death Differ 2007; 14:1247-50. [PMID: 17431417 DOI: 10.1038/sj.cdd.4402149] [Citation(s) in RCA: 115] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- S Luo
- Department of Medical Genetics, Cambridge Institute for Medical Research, Wellcome/MRC Building, Addenbrooke's Hospital, Hills Road, Cambridge CB2 2XY, UK
| | | |
Collapse
|
29
|
Ferraro E, Cecconi F. Autophagic and apoptotic response to stress signals in mammalian cells. Arch Biochem Biophys 2007; 462:210-9. [PMID: 17374522 DOI: 10.1016/j.abb.2007.02.006] [Citation(s) in RCA: 142] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2006] [Revised: 01/27/2007] [Accepted: 02/04/2007] [Indexed: 02/06/2023]
Abstract
Autophagy is a highly conserved catabolic programme for degrading proteins and organelles. This process has been shown to act as a pro-survival or pro-death mechanism in different physiological and pathological conditions. Several stress stimuli can induce autophagy, such as nutrient deprivation or critical steps in development of lower and higher eukaryotes. Apoptosis is an orchestrated form of cell death in which cells are actively involved in their own demise. Again, stress is a positive regulator of apoptosis and, in particular, of its apoptosome-mediated mitochondrial pathway. Besides discussing the individual roles played by the key molecules involved in autophagy in mammals in response to stress signals, we discuss here the interrelations between autophagy and apoptosis under these conditions.
Collapse
Affiliation(s)
- Elisabetta Ferraro
- Laboratory of Molecular Neuroembryology, IRCCS Fondazione Santa Lucia, Rome 00143, Italy
| | | |
Collapse
|
30
|
Abstract
Autophagy is a physiological and evolutionarily conserved phenomenon maintaining homeostatic functions like protein degradation and organelle turnover. It is rapidly upregulated under conditions leading to cellular stress, such as nutrient or growth factor deprivation, providing an alternative source of intracellular building blocks and substrates for energy generation to enable continuous cell survival. Yet accumulating data provide evidence that the autophagic machinery can be also recruited to kill cells under certain conditions generating a caspase-independent form of programed cell death (PCD), named autophagic cell death. Due to increasing interest in nonapoptotic PCD forms and the development of mammalian genetic tools to study autophagy, autophagic cell death has achieved major prominence, and is recognized now as a legitimate alternative death pathway to apoptosis. This chapter aims at summarizing the recent data in the field of autophagy signaling and autophagic cell death.
Collapse
Affiliation(s)
- Devrim Gozuacik
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel
| | | |
Collapse
|
31
|
López-Diazguerrero NE, López-Araiza H, Conde-Perezprina JC, Bucio L, Cárdenas-Aguayo MC, Ventura JL, Covarrubias L, Gutiérrez-Ruíz MC, Zentella A, Königsberg M. Bcl-2 protects against oxidative stress while inducing premature senescence. Free Radic Biol Med 2006; 40:1161-9. [PMID: 16545683 DOI: 10.1016/j.freeradbiomed.2005.11.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2005] [Revised: 10/28/2005] [Accepted: 11/03/2005] [Indexed: 01/15/2023]
Abstract
Replicative senescence is a cellular response to stress that has been postulated to serve as a tumor suppression mechanism and a contributor to aging. Numerous experimental studies have demonstrated that an apparently identical senescent state can also be prematurely induced in vitro in different cell types following sublethal oxidative stress stimuli. The former suggests a molecular link between cell cycle regulation and cell survival that could involve regulatory proteins such as Bcl-2. There is strong evidence that, in addition to its well-known effects on apoptosis, Bcl-2 is involved in antioxidant protection and regulation of cell cycle progression. The aim of this work was to determine if the protection against oxidative stress mediated by Bcl-2 could prevent or delay oxidative stress-induced senescence. Using a retroviral infection system, Bcl-2 was overexpressed in primary, nonembryonic mice fibroblasts obtained from lungs derived from 2-month-old CD1 mice. Fibroblasts overexpressing Bcl-2 were exposed to 75 microM H2O2 for 2 h to induce SIPS. The rate of proliferation and the increment of senescent cells were then determined. Our results indicate that overexpression of Bcl-2 protected primary fibroblasts against oxidative stress-mediated reduction in cell proliferation, but did not prevent premature senescence.
Collapse
Affiliation(s)
- Norma E López-Diazguerrero
- Departamento Ciencias de la Salud, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Iztapalapa. A.P. 55-535, C.P 09340, México D.F., México
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Matyja E, Taraszewska A, Nagańska E, Rafałowska J. Autophagic degeneration of motor neurons in a model of slow glutamate excitotoxicity in vitro. Ultrastruct Pathol 2005; 29:331-9. [PMID: 16257859 DOI: 10.1080/01913120500214333] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
There is increasing evidence that so-called "autophagic cell death" participates in cell degeneration in certain pathological conditions. Autophagy might be involved in some neurodegenerative processes, including lateral amyotrophic sclerosis (SLA). The exact mechanism leading to progressive motor neuron (MN) loss remains unclear, but glutamate-mediated mechanism is thought to be responsible. Previous ultrastructural studies by the authors performed on a model of SLA in vitro, based on chronic glutamate excitotoxicity, revealed a subset of morphological features characteristic to different modes of neuronal death, including autophagic degeneration. The contribution of this pathway of MNs death is evaluated in organotypic cultures of rat lumbar spinal cord chronically exposed to specific glutamate uptake blockers: DL-threo-beta-hydroxyaspartate (THA) and L-transpyrrolidine-2,4-dicarboxylate (PDC). The study documents the various steps of authophagy in slowly evolving process of MN neurodegeneration. The cells undergoing autophagy usually exhibited sequestration of some parts of cytoplasm with normal and/or degenerated organelles, whereas other parts of cytoplasm as well as neuronal nucleus remained unchanged. The advanced autophagic changes were often associated with other modes of MN death, especially with apoptosis. Numerous MNs revealed apoptotic nuclear features with typical peripheral margination of nuclear chromatin, accompanied by severe autophagic or autophagic-necrotic degeneration of the cytoplasm. These results support the opinion of unclear distinction between different modes of cell death and indicate the involvement of autophagey in MNs neurodegeneration in vitro.
Collapse
Affiliation(s)
- Ewa Matyja
- Department of Experimental and Clinical Neuropathology, Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | | | | | | |
Collapse
|
33
|
Abstract
The visualization of autophagosomes in dying cells has led to the belief that autophagy is a nonapoptotic form of programmed cell death. This concept has now been evaluated using cells and organisms deficient in autophagy genes. Most evidence indicates that, at least in cells with intact apoptotic machinery, autophagy is primarily a pro-survival rather than a pro-death mechanism. This review summarizes the evidence linking autophagy to cell survival and cell death, the complex interplay between autophagy and apoptosis pathways, and the role of autophagy-dependent survival and death pathways in clinical diseases.
Collapse
Affiliation(s)
- Beth Levine
- Division of Infectious Diseases, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9113, USA.
| | | |
Collapse
|
34
|
Abstract
Cell death has been subdivided into the categories apoptosis (Type I), autophagic cell death (Type II), and necrosis (Type III). The boundary between Type I and II has never been completely clear and perhaps does not exist due to intrinsic factors among different cell types and the crosstalk among organelles within each type. Apoptosis can begin with autophagy, autophagy can end with apoptosis, and blockage of caspase activity can cause a cell to default to Type II cell death from Type I. Furthermore, autophagy is a normal physiological process active in both homeostasis (organelle turnover) and atrophy. "Autophagic cell death" may be interpreted as the process of autophagy that, unlike other situations, does not terminate before the cell collapses. Since switching among the alternative pathways to death is relatively common, interpretations based on knockouts or inhibitors, and therapies directed at controlling apoptosis must include these considerations.
Collapse
Affiliation(s)
- Richard A Lockshin
- Department of Biological Sciences, St. John's University, 8000 Utopia Parkway, Jamaica, NY 11439, USA.
| | | |
Collapse
|
35
|
Naumann U, Wischhusen J, Weit S, Rieger J, Wolburg H, Massing U, Weller M. Alkylphosphocholine-induced glioma cell death is BCL-XL-sensitive, caspase-independent and characterized by massive cytoplasmic vacuole formation. Cell Death Differ 2004; 11:1326-41. [PMID: 15389288 DOI: 10.1038/sj.cdd.4401503] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Alkylphosphocholines (APC) are candidate anticancer agents. We here report that APC induce the formation of large vacuoles and typical features of apoptosis in human glioma cell lines, but not in immortalized astrocytes. APC promote caspase activation, poly(ADP-ribose)-polymerase (PARP) processing and cytochrome c release from mitochondria. Adenoviral X-linked inhibitor of apoptosis (XIAP) gene transfer, or exposure to the caspase inhibitor, benzyloxycarbonyl-Val-Ala-DL-Asp-fluoro-methylketone zVAD-fmk, blocks caspase-7 and PARP processing, but not cell death, whereas BCL-X(L) blocks not only caspase-7 and PARP processing but also cell death. APC induce changes in Delta Psi m in sensitive glioma cells, but not in resistant astrocytes. The changes in Delta Psi m are unaffected by crm-A (cowpox serpin-cytokine response modifier protein A), XIAP or zVAD-fmk, but blocked by BCL-X(L), and are thus a strong predictor of cell death in response to APC. Free radicals are induced, but not responsible for cell death. APC thus induce a characteristic morphological, BCL-X(L)-sensitive, apparently caspase-independent cell death involving mitochondrial alterations selectively in neoplastic astrocytic cells.
Collapse
Affiliation(s)
- U Naumann
- Laboratory of Molecular Neuro-Oncology, Department of General Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany.
| | | | | | | | | | | | | |
Collapse
|