251
|
Shimizu T, Maruyama K, Kawamura T, Urade Y, Wada Y. PERK participates in cardiac valve development via fatty acid oxidation and endocardial-mesenchymal transformation. Sci Rep 2020; 10:20094. [PMID: 33208886 PMCID: PMC7674462 DOI: 10.1038/s41598-020-77199-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 11/09/2020] [Indexed: 12/31/2022] Open
Abstract
Protein kinase R-like endoplasmic reticulum kinase (PERK) is one of the endoplasmic reticulum (ER) stress sensors. PERK loss-of-function mutations are known to cause Wolcott–Rallison syndrome. This disease is characterized by early-onset diabetes mellitus, skeletal dysplasia, and cardiac valve malformation. To understand the role of PERK in valve formation in vivo, we used an endothelial-specific PERK conditional knockout mice as well as in vitro PERK inhibition assays. We used ProteoStat dyes to visualize the accumulation of misfolded proteins in the endocardial cushion and valve mesenchymal cells (VMCs). Then, VMCs were isolated from E12.5 fetal mice, by fluorescence assisted cell sorting. Proteomic analysis of PERK-deleted VMCs identified the suppression of proteins related to fatty acid oxidation (FAO), especially carnitine palmitoyltransferase II (CPT2). CPT2 is a critical regulator of endocardial-mesenchymal transformation (EndoMT); however how TGF-β downstream signaling controls CPT2 expression remains unclear. Here, we showed that PERK inhibition suppressed, not only EndoMT but also CPT2 protein expression in human umbilical vein endothelial cells (HUVECs) under TGF-β1 stimulation. As a result, PERK inhibition suppressed mitochondrial metabolic activity. Taken together, these results demonstrate that PERK signaling is required for cardiac valve formation via FAO and EndoMT.
Collapse
Affiliation(s)
- Takashi Shimizu
- Isotope Science Center, The University of Tokyo, Tokyo, 113-0032, Japan. .,Department of Cardiovascular Medicine, The University of Tokyo, Graduate School of Medicine, Tokyo, 113-8655, Japan.
| | - Kazuaki Maruyama
- Isotope Science Center, The University of Tokyo, Tokyo, 113-0032, Japan
| | - Takeshi Kawamura
- Isotope Science Center, The University of Tokyo, Tokyo, 113-0032, Japan
| | - Yoshihiro Urade
- Isotope Science Center, The University of Tokyo, Tokyo, 113-0032, Japan
| | - Youichiro Wada
- Isotope Science Center, The University of Tokyo, Tokyo, 113-0032, Japan
| |
Collapse
|
252
|
Yan P, Ren J, Zhang W, Qu J, Liu GH. Protein quality control of cell stemness. CELL REGENERATION (LONDON, ENGLAND) 2020; 9:22. [PMID: 33179756 PMCID: PMC7658286 DOI: 10.1186/s13619-020-00064-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 09/14/2020] [Indexed: 02/07/2023]
Abstract
Protein quality control (PQC) systems play essential roles in the recognition, refolding and clearance of aberrant proteins, thus ensuring cellular protein homeostasis, or proteostasis. Especially, continued proliferation and differentiation of stem cells require a high rate of translation; therefore, accurate PQC systems are essential to maintain stem cell function. Growing evidence suggested crucial roles of PQC systems in regulating the stemness and differentiation of stem cells. This review focuses on current knowledge regarding the components of the proteostasis network in stem cells, and the importance of proteostasis in maintaining stem cell identity and regenerative functions. A complete understanding of this process might uncover potential applications in aging intervention and aging-related diseases.
Collapse
Affiliation(s)
- Pengze Yan
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jie Ren
- University of Chinese Academy of Sciences, Beijing, 100049, China
- China National Center for Bioinformation, Beijing, 100101, China
- CAS Key Laboratory of Genomic and Precision Medicine, Collaborative Innovation Center of Genetics and Development, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, China
- Institute for Stem cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
| | - Weiqi Zhang
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- China National Center for Bioinformation, Beijing, 100101, China.
- CAS Key Laboratory of Genomic and Precision Medicine, Collaborative Innovation Center of Genetics and Development, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, China.
- Institute for Stem cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Jing Qu
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Institute for Stem cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Guang-Hui Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Institute for Stem cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Brain Disorders, Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, 100053, China.
| |
Collapse
|
253
|
Abstract
Autophagy is an adaptive catabolic process functioning to promote cell survival in the event of inappropriate living conditions such as nutrient shortage and to cope with diverse cytotoxic insults. It is regarded as one of the key survival mechanisms of living organisms. Cells undergo autophagy to accomplish the lysosomal digestion of intracellular materials including damaged proteins, organelles, and foreign bodies, in a bulk, non-selective or a cargo-specific manner. Studies in the past decades have shed light on the association of autophagy pathways with various diseases and also highlighted the therapeutic value of autophagy modulation. Hence, it is crucial to develop effective approaches for monitoring intracellular autophagy dynamics, as a comprehensive account of methodology establishment is far from complete. In this review, we aim to provide an overview of the major current fluorescence-based techniques utilized for visualizing, sensing or measuring autophagic activities in cells or tissues, which are categorized firstly by targets detected and further by the types of fluorescence tools. We will mainly focus on the working mechanisms of these techniques, put emphasis on the insight into their roles in biomedical science and provide perspectives on the challenges and future opportunities in this field.
Collapse
Affiliation(s)
- Siyang Ding
- Department of Chemistry and Physics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne Victoria 3086, Australia.
| | | |
Collapse
|
254
|
Goodman JS, King GA, Ünal E. Cellular quality control during gametogenesis. Exp Cell Res 2020; 396:112247. [PMID: 32882217 PMCID: PMC7572901 DOI: 10.1016/j.yexcr.2020.112247] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 08/18/2020] [Accepted: 08/23/2020] [Indexed: 12/12/2022]
Abstract
A hallmark of aging is the progressive accumulation of cellular damage. Age-induced damage arises due to a decrease in organelle function along with a decline in protein quality control. Although somatic tissues deteriorate with age, the germline must maintain cellular homeostasis in order to ensure the production of healthy progeny. While germline quality control has been primarily studied in multicellular organisms, recent evidence suggests the existence of gametogenesis-specific quality control mechanisms in unicellular eukaryotes, highlighting the evolutionary conservation of meiotic events beyond chromosome morphogenesis. Notably, budding yeast eliminates age-induced damage during meiotic differentiation, employing novel organelle and protein quality control mechanisms to produce young and healthy gametes. Similarly, organelle and protein quality control is present in metazoan gametogenesis; however, whether and how these mechanisms contribute to cellular rejuvenation requires further investigation. Here, we summarize recent findings that describe organelle and protein quality control in budding yeast gametogenesis, examine similar quality control mechanisms in metazoan development, and identify research directions that will improve our understanding of meiotic cellular rejuvenation.
Collapse
Affiliation(s)
- Jay S Goodman
- Department of Molecular and Cell Biology, University of California Berkeley, 94720, USA
| | - Grant A King
- Department of Molecular and Cell Biology, University of California Berkeley, 94720, USA
| | - Elçin Ünal
- Department of Molecular and Cell Biology, University of California Berkeley, 94720, USA.
| |
Collapse
|
255
|
Stroobants S, D'Hooge R, Damme M. Aged Tmem106b knockout mice display gait deficits in coincidence with Purkinje cell loss and only limited signs of non-motor dysfunction. Brain Pathol 2020; 31:223-238. [PMID: 33016371 PMCID: PMC8018119 DOI: 10.1111/bpa.12903] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 09/14/2020] [Accepted: 09/28/2020] [Indexed: 12/13/2022] Open
Abstract
Genetic variants in TMEM106B are a major risk factor for several neurodegenerative diseases including frontotemporal degeneration, limbic‐predominant age‐related TDP‐43 encephalopathy, Parkinson's disease, late‐onset‐Alzheimer's disease and constitute a genetic determinant of differential aging. TMEM106B encodes an integral lysosomal membrane protein but its precise physiological function in the central nervous system remains enigmatic. Presently, we aimed to increase understanding of TMEM106B contribution to general brain function and aging. We analyzed an aged cohort of Tmem106b knockout‐, heterozygote and wild‐type mice in a behavioral test battery including assessments of motor function as well as, social, emotional and cognitive function. Aged Tmem106b knockout (KO) mice displayed diverse behavioral deficits including motor impairment, gait defects and reduced startle reactivity. In contrast, no prominent deficits were observed in social, emotional or cognitive behaviors. Histologically, we observed late‐onset loss of Purkinje cells followed by reactive gliosis in the cerebellum, which likely contributed to progressive decline in motor function and gait defects in particular. Reactive gliosis was not restricted to the cerebellum but observed in different areas of the brain including the brain stem and parts of the cerebral cortex. Surviving Purkinje cells showed vacuolated lysosomes in the axon initial segment, implicating TMEM106B‐dependent lysosomal trafficking defects as the underlying cause of axonal and more general neuronal dysfunction contributing to behavioral impairments. Our experiments help to elucidate how TMEM106B affects spatial neuronal homeostasis and exemplifies a critical role of TMEM106B in neuronal cells for survival.
Collapse
Affiliation(s)
- Stijn Stroobants
- Laboratory of Biological Psychology, KU Leuven, Tiensestraat 102, Leuven, 3000, Belgium
| | - Rudi D'Hooge
- Laboratory of Biological Psychology, KU Leuven, Tiensestraat 102, Leuven, 3000, Belgium
| | - Markus Damme
- Institut für Biochemie, Christian-Albrechts-Universität zu Kiel, Kiel, 24098, Germany
| |
Collapse
|
256
|
Paniagua-Herranz L, Menéndez-Méndez A, Gómez-Villafuertes R, Olivos-Oré LA, Biscaia M, Gualix J, Pérez-Sen R, Delicado EG, Artalejo AR, Miras-Portugal MT, Ortega F. Live Imaging Reveals Cerebellar Neural Stem Cell Dynamics and the Role of VNUT in Lineage Progression. Stem Cell Reports 2020; 15:1080-1094. [PMID: 33065045 PMCID: PMC7663791 DOI: 10.1016/j.stemcr.2020.09.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 09/20/2020] [Accepted: 09/21/2020] [Indexed: 11/04/2022] Open
Abstract
Little is known about the intrinsic specification of postnatal cerebellar neural stem cells (NSCs) and to what extent they depend on information from their local niche. Here, we have used an adapted cell preparation of isolated postnatal NSCs and live imaging to demonstrate that cerebellar progenitors maintain their neurogenic nature by displaying hallmarks of NSCs. Furthermore, by using this preparation, all the cell types produced postnatally in the cerebellum, in similar relative proportions to those observed in vivo, can be monitored. The fact that neurogenesis occurs in such organized manner in the absence of signals from the local environment, suggests that cerebellar lineage progression is to an important extent governed by cell-intrinsic or pre-programmed events. Finally, we took advantage of the absence of the niche to assay the influence of the vesicular nucleotide transporter inhibition, which dramatically reduced the number of NSCs in vitro by promoting their progression toward neurogenesis. We present a preparation that allows monitoring the behavior of cerebellar NSCs Isolated NSCs maintain their neurogenic nature in absence of niche factors The model enables monitoring the three postnatal cerebellar niches simultaneously VNUT influences the balance between quiescence and activation of cerebellar NSCs
Collapse
Affiliation(s)
- Lucía Paniagua-Herranz
- Departament of Biochemistry and Molecular Biology, Faculty of Veterinary, Universidad Complutense de Madrid (UCM), Madrid, Spain; Instituto Universitario de Investigación en Neuroquímica (IUIN), Madrid, Spain; Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid, Spain
| | - Aida Menéndez-Méndez
- Departament of Biochemistry and Molecular Biology, Faculty of Veterinary, Universidad Complutense de Madrid (UCM), Madrid, Spain; Instituto Universitario de Investigación en Neuroquímica (IUIN), Madrid, Spain; Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid, Spain
| | - Rosa Gómez-Villafuertes
- Departament of Biochemistry and Molecular Biology, Faculty of Veterinary, Universidad Complutense de Madrid (UCM), Madrid, Spain; Instituto Universitario de Investigación en Neuroquímica (IUIN), Madrid, Spain; Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid, Spain
| | - Luis A Olivos-Oré
- Instituto Universitario de Investigación en Neuroquímica (IUIN), Madrid, Spain; Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid, Spain; Department of Pharmacology and Toxicology, Faculty of Veterinary, Universidad Complutense de Madrid, Madrid, Spain
| | - Miguel Biscaia
- Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, Madrid, Spain
| | - Javier Gualix
- Departament of Biochemistry and Molecular Biology, Faculty of Veterinary, Universidad Complutense de Madrid (UCM), Madrid, Spain; Instituto Universitario de Investigación en Neuroquímica (IUIN), Madrid, Spain; Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid, Spain
| | - Raquel Pérez-Sen
- Departament of Biochemistry and Molecular Biology, Faculty of Veterinary, Universidad Complutense de Madrid (UCM), Madrid, Spain; Instituto Universitario de Investigación en Neuroquímica (IUIN), Madrid, Spain; Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid, Spain
| | - Esmerilda G Delicado
- Departament of Biochemistry and Molecular Biology, Faculty of Veterinary, Universidad Complutense de Madrid (UCM), Madrid, Spain; Instituto Universitario de Investigación en Neuroquímica (IUIN), Madrid, Spain; Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid, Spain
| | - Antonio R Artalejo
- Instituto Universitario de Investigación en Neuroquímica (IUIN), Madrid, Spain; Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid, Spain; Department of Pharmacology and Toxicology, Faculty of Veterinary, Universidad Complutense de Madrid, Madrid, Spain
| | - María Teresa Miras-Portugal
- Departament of Biochemistry and Molecular Biology, Faculty of Veterinary, Universidad Complutense de Madrid (UCM), Madrid, Spain; Instituto Universitario de Investigación en Neuroquímica (IUIN), Madrid, Spain; Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid, Spain
| | - Felipe Ortega
- Departament of Biochemistry and Molecular Biology, Faculty of Veterinary, Universidad Complutense de Madrid (UCM), Madrid, Spain; Instituto Universitario de Investigación en Neuroquímica (IUIN), Madrid, Spain; Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid, Spain.
| |
Collapse
|
257
|
Haidurov A, Budanov AV. Sestrin family - the stem controlling healthy ageing. Mech Ageing Dev 2020; 192:111379. [PMID: 33022334 DOI: 10.1016/j.mad.2020.111379] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 09/04/2020] [Accepted: 09/29/2020] [Indexed: 01/18/2023]
Abstract
Sestrins are a family of stress-responsive antioxidant proteins responsible for regulation of cell viability and metabolism. The best known Sestrin targets are mTORC1 and mTORC2 kinases that control different cellular processes including growth, viability, autophagy, and mitochondrial metabolism. Inactivation of the single Sestrin gene in invertebrates has an adverse impact on their healthspan and longevity, whereas each of the three Sestrin genes in mammals and other vertebrate organisms has a different impact on maintenance of a particular tissue, affecting its stress tolerance, function and regenerative capability. As a result, Sestrins attenuate ageing and suppress development of many age-related diseases including myocardial infarction, muscle atrophy, diabetes, and immune dysfunction, but exacerbate development of chronic obstructive pulmonary disease. Moreover, Sestrins play opposite roles in carcinogenesis in different tissues. Stem cells support tissue remodelling that influences ageing, and Sestrins might suppress ageing and age-related pathologies through control of stem cell biology. In this review, we will discuss the potential link between Sestrins, stem cells, and ageing.
Collapse
Affiliation(s)
- Alexander Haidurov
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Pearse Street, Dublin 2, Ireland; Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Andrei V Budanov
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Pearse Street, Dublin 2, Ireland; Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.
| |
Collapse
|
258
|
Liu B, Shen Y, Huang H, Croce KD, Wu M, Fan Y, Liu Y, Xu J, Yao G. Curcumin derivative C212 inhibits Hsp90 and eliminates both growing and quiescent leukemia cells in deep dormancy. Cell Commun Signal 2020; 18:159. [PMID: 32993709 PMCID: PMC7523331 DOI: 10.1186/s12964-020-00652-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 08/26/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Relapsed leukemia following initial therapeutic response and remission is difficult to treat and causes high patient mortality. Leukemia relapse is due to residual quiescent leukemia cells that escape conventional therapies and later reemerge. Eliminating not only growing but quiescent leukemia cells is critical to effectively treating leukemia and preventing its recurrence. Such dual targeting therapeutic agents, however, are lacking in the clinic. To start tackling this problem, encouraged by the promising anticancer effects of a set of curcumin derivatives in our earlier studies, we examined in this work the effects of a 4-arylmethyl curcumin derivative (C212) in eliminating both growing and quiescent leukemia cells. METHODS We analyzed the effects of C212 on the growth and viability of growing and quiescent leukemia cells using MTS, apoptosis, cell cycle and cell tracking assays. The effects of C212 on the quiescence depth of leukemia cells were measured using EdU incorporation assay upon growth stimulation. The mechanisms of C212-induced apoptosis and deep dormancy, particularly associated with its inhibition of Hsp90 activity, were studied using molecular docking, protein aggregation assay, and Western blot of client proteins. RESULTS C212, on the one hand, inhibits growing leukemia cells at a higher efficacy than curcumin by inducing apoptosis and G2/M accumulation; it, on the other hand, eliminates quiescent leukemia cells that are resistant to conventional treatments. Furthermore, C212 drives leukemia cells into and kills them at deep quiescence. Lastly, we show that C212 induces apoptosis and drives cells into deep dormancy at least partially by binding to and inhibiting Hsp90, leading to client protein degradation and protein aggregation. CONCLUSION C212 effectively eliminates both growing and quiescent leukemia cells by inhibiting Hsp90. The property of C212 to kill quiescent leukemia cells in deep dormancy avoids the risk associated with awaking therapy-resistant subpopulation of quiescent leukemia cells during treatments, which may lead to the development of novel therapies against leukemia relapse. Video abstract.
Collapse
Affiliation(s)
- Bi Liu
- School of Pharmacy, Fujian Provincial Key Laboratory of Natural Medicine Pharmacology, Fujian Medical University, Fuzhou, 350122 China
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ 85721 USA
| | - Yunzhu Shen
- School of Pharmacy, Fujian Provincial Key Laboratory of Natural Medicine Pharmacology, Fujian Medical University, Fuzhou, 350122 China
- The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000 Fujian China
| | - Huafang Huang
- School of Pharmacy, Fujian Provincial Key Laboratory of Natural Medicine Pharmacology, Fujian Medical University, Fuzhou, 350122 China
| | - Kimiko Della Croce
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ 85721 USA
| | - Min Wu
- School of Pharmacy, Fujian Provincial Key Laboratory of Natural Medicine Pharmacology, Fujian Medical University, Fuzhou, 350122 China
| | - Yingjuan Fan
- School of Pharmacy, Fujian Provincial Key Laboratory of Natural Medicine Pharmacology, Fujian Medical University, Fuzhou, 350122 China
| | - Yang Liu
- School of Pharmacy, Fujian Provincial Key Laboratory of Natural Medicine Pharmacology, Fujian Medical University, Fuzhou, 350122 China
| | - Jianhua Xu
- School of Pharmacy, Fujian Provincial Key Laboratory of Natural Medicine Pharmacology, Fujian Medical University, Fuzhou, 350122 China
| | - Guang Yao
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ 85721 USA
- Arizona Cancer Center, University of Arizona, Tucson, AZ 85719 USA
| |
Collapse
|
259
|
Mata-Cabana A, Romero-Expósito FJ, Olmedo M. Aging during C. elegans L1 quiescence. Aging (Albany NY) 2020; 12:17756-17758. [PMID: 32986012 PMCID: PMC7585126 DOI: 10.18632/aging.104027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 08/24/2020] [Indexed: 01/24/2023]
Affiliation(s)
- Alejandro Mata-Cabana
- Departamento de Genética, Facultad de Biología, Universidad de Sevilla, Sevilla 41012, Spain
| | | | - María Olmedo
- Departamento de Genética, Facultad de Biología, Universidad de Sevilla, Sevilla 41012, Spain
| |
Collapse
|
260
|
Kobayashi T, Kageyama R. Lysosomes and signaling pathways for maintenance of quiescence in adult neural stem cells. FEBS J 2020; 288:3082-3093. [PMID: 32902139 PMCID: PMC8246936 DOI: 10.1111/febs.15555] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 08/18/2020] [Accepted: 09/01/2020] [Indexed: 12/28/2022]
Abstract
Quiescence is a cellular strategy for maintaining somatic stem cells in a specific niche in a low metabolic state without senescence for a long period of time. During development, neural stem cells (NSCs) actively proliferate and self-renew, and their progeny differentiate into both neurons and glial cells to form mature brain tissues. On the other hand, most NSCs in the adult brain are quiescent and arrested in G0/G1 phase of the cell cycle. Quiescence is essential in order to avoid the precocious exhaustion of NSCs, ensuring a sustainable source of available stem cells in the brain throughout the lifespan. After receiving activation signals, quiescent NSCs reenter the cell cycle and generate new neurons. This switching between quiescence and proliferation is tightly regulated by diverse signaling pathways. Recent studies suggest significant involvement of cellular proteostasis (homeostasis of the proteome) in the quiescent state of NSCs. Proteostasis is the result of integrated regulation of protein synthesis, folding, and degradation. In this review, we discuss regulation of quiescence by multiple signaling pathways, especially bone morphogenetic protein and Notch signaling, and focus on the functional involvement of the lysosome, an organelle governing cellular degradation, in quiescence of adult NSCs.
Collapse
Affiliation(s)
- Taeko Kobayashi
- Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan.,Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Ryoichiro Kageyama
- Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan.,Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Graduate School of Biostudies, Kyoto University, Kyoto, Japan.,Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University, Kyoto, Japan
| |
Collapse
|
261
|
Bjedov I, Rallis C. The Target of Rapamycin Signalling Pathway in Ageing and Lifespan Regulation. Genes (Basel) 2020; 11:E1043. [PMID: 32899412 PMCID: PMC7565554 DOI: 10.3390/genes11091043] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 08/28/2020] [Accepted: 08/30/2020] [Indexed: 12/11/2022] Open
Abstract
Ageing is a complex trait controlled by genes and the environment. The highly conserved mechanistic target of rapamycin signalling pathway (mTOR) is a major regulator of lifespan in all eukaryotes and is thought to be mediating some of the effects of dietary restriction. mTOR is a rheostat of energy sensing diverse inputs such as amino acids, oxygen, hormones, and stress and regulates lifespan by tuning cellular functions such as gene expression, ribosome biogenesis, proteostasis, and mitochondrial metabolism. Deregulation of the mTOR signalling pathway is implicated in multiple age-related diseases such as cancer, neurodegeneration, and auto-immunity. In this review, we briefly summarise some of the workings of mTOR in lifespan and ageing through the processes of transcription, translation, autophagy, and metabolism. A good understanding of the pathway's outputs and connectivity is paramount towards our ability for genetic and pharmacological interventions for healthy ageing and amelioration of age-related disease.
Collapse
Affiliation(s)
- Ivana Bjedov
- UCL Cancer Institute, Paul O’Gorman Building, 72 Huntley Street, London WC1E 6DD, UK
| | - Charalampos Rallis
- School of Life Sciences, University of Essex, Wivenhoe Park, Colchester CO4 3SQ, UK
| |
Collapse
|
262
|
The intrinsic proteostasis network of stem cells. Curr Opin Cell Biol 2020; 67:46-55. [PMID: 32890906 DOI: 10.1016/j.ceb.2020.08.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 07/31/2020] [Accepted: 08/03/2020] [Indexed: 01/03/2023]
Abstract
The proteostasis network adjusts protein composition and maintains protein integrity, which are essential processes for cell function and viability. Current efforts, given their intrinsic characteristics, regenerative potential and fundamental biological functions, have been directed to define proteostasis of stem cells. These insights demonstrate that embryonic stem cells and induced pluripotent stem cells exhibit an endogenous proteostasis network that not only modulates their pluripotency and differentiation but also provides a striking ability to suppress aggregation of disease-related proteins. Moreover, recent findings establish a central role of enhanced proteostasis to prevent the aging of somatic stem cells in adult organisms. Notably, proteostasis is also required for the biological purpose of adult germline stem cells, that is to be passed from one generation to the next. Beyond these links between proteostasis and stem cell function, we also discuss the implications of these findings for disease, aging, and reproduction.
Collapse
|
263
|
Schüler SC, Gebert N, Ori A. Stem cell aging: The upcoming era of proteins and metabolites. Mech Ageing Dev 2020; 190:111288. [DOI: 10.1016/j.mad.2020.111288] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 06/04/2020] [Accepted: 06/16/2020] [Indexed: 02/07/2023]
|
264
|
Audesse AJ, Webb AE. Mechanisms of enhanced quiescence in neural stem cell aging. Mech Ageing Dev 2020; 191:111323. [PMID: 32781077 DOI: 10.1016/j.mad.2020.111323] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 07/20/2020] [Accepted: 07/23/2020] [Indexed: 12/24/2022]
Abstract
The maintenance of neural stem cell function is vital to ensure neurogenesis throughout adulthood. During aging, there is a significant reduction in adult neurogenesis that correlates with a decline in cognitive function. Although recent studies have revealed novel extrinsic and intrinsic mechanisms that regulate the adult neural stem cell (NSC) pool and lineage progression, the precise molecular mechanisms that drive dysregulation of adult neurogenesis in the context of aging are only beginning to emerge. Recent studies have shed light on mechanisms that regulate the earliest step of adult neurogenesis, the activation of quiescent NSCs. Interestingly, the ability of NSCs to enter the cell cycle in the aged brain significantly declines suggesting a deepend state of quiescence. Given the likely contribution of adult neurogenesis to supporting cognitive function in humans, enhancing neurogenesis may be a strategy to combat age-related cognitive decline. This review highlights the mechanisms that regulate the NSC pool throughout adulthood and discusses how dysregulation of these processes may contribute to the decline in neurogenesis and cognitive function throughout aging.
Collapse
Affiliation(s)
- Amanda J Audesse
- Graduate Program in Neuroscience, Brown University, USA; Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI 02912, USA
| | - Ashley E Webb
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI 02912, USA; Carney Institute for Brain Science, Brown University, Providence, RI 02912, USA; Center on the Biology of Aging, Brown University, Providence, RI 02912, USA.
| |
Collapse
|
265
|
Nicaise AM, Willis CM, Crocker SJ, Pluchino S. Stem Cells of the Aging Brain. Front Aging Neurosci 2020; 12:247. [PMID: 32848716 PMCID: PMC7426063 DOI: 10.3389/fnagi.2020.00247] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 07/20/2020] [Indexed: 12/12/2022] Open
Abstract
The adult central nervous system (CNS) contains resident stem cells within specific niches that maintain a self-renewal and proliferative capacity to generate new neurons, astrocytes, and oligodendrocytes throughout adulthood. Physiological aging is associated with a progressive loss of function and a decline in the self-renewal and regenerative capacities of CNS stem cells. Also, the biggest risk factor for neurodegenerative diseases is age, and current in vivo and in vitro models of neurodegenerative diseases rarely consider this. Therefore, combining both aging research and appropriate interrogation of animal disease models towards the understanding of the disease and age-related stem cell failure is imperative to the discovery of new therapies. This review article will highlight the main intrinsic and extrinsic regulators of neural stem cell (NSC) aging and discuss how these factors impact normal homeostatic functions within the adult brain. We will consider established in vivo animal and in vitro human disease model systems, and then discuss the current and future trajectories of novel senotherapeutics that target aging NSCs to ameliorate brain disease.
Collapse
Affiliation(s)
- Alexandra M Nicaise
- Department of Clinical Neurosciences and NIHR Biomedical Research Centre, University of Cambridge, Cambridge, United Kingdom
| | - Cory M Willis
- Department of Clinical Neurosciences and NIHR Biomedical Research Centre, University of Cambridge, Cambridge, United Kingdom
| | - Stephen J Crocker
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT, United States
| | - Stefano Pluchino
- Department of Clinical Neurosciences and NIHR Biomedical Research Centre, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
266
|
Jacobs KA, Maghe C, Gavard J. Lysosomes in glioblastoma: pump up the volume. Cell Cycle 2020; 19:2094-2104. [PMID: 32723137 DOI: 10.1080/15384101.2020.1796016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Lysosomes are acidic, dynamic organelles that supervise catabolism, integrate signaling cascades, and tune cellular trafficking. Moreover, the loss of their integrity may jeopardize cell viability. In cancer cells, lysosomes are qualitatively and quantitatively modified for the tumor's own benefit. For all these reasons, these organelles emerge as appealing intracellular targets to manipulate non-oncogene addiction. This is of particular interest for brain diseases, including neurodegenerative disorders and cancer, in which stem cells are exhausted and transformed, respectively. Recent publications had demonstrated that stem cells displayed disarmed lysosomes in terms of number and functions during aging and oncogenic progression. Likewise, our laboratory identified that the arginine protease MALT1, normally dedicated to the assembly of proper NF-kB activation and processing a number of substrates, arbitrates lysosome biogenesis and mTOR signaling in glioblastoma stem-like cells. Indeed, blocking either the expression or the activity of this enzyme leads to an aberrant increase of lysosomes, alongside of the down-regulation of the mTOR signaling. This surge of lysosomes eradicates glioblastoma stem-like cells. Targeting lysosomes might thus inspire the design of new strategies to face this devastating human cancer. Here, we provide an overview of the functions of the lysosome as well as its role as a cell death initiator, to highlight the potential of lysosomal drugs for glioblastoma therapy.
Collapse
Affiliation(s)
- Kathryn A Jacobs
- Team SOAP, CRCINA, Inserm, CNRS, Université De Nantes, Université d'Angers , Nantes, France
| | - Clément Maghe
- Team SOAP, CRCINA, Inserm, CNRS, Université De Nantes, Université d'Angers , Nantes, France
| | - Julie Gavard
- Team SOAP, CRCINA, Inserm, CNRS, Université De Nantes, Université d'Angers , Nantes, France.,Integrated Center for Oncology, ICO , St. Herblain, France
| |
Collapse
|
267
|
Navarro Negredo P, Yeo RW, Brunet A. Aging and Rejuvenation of Neural Stem Cells and Their Niches. Cell Stem Cell 2020; 27:202-223. [PMID: 32726579 DOI: 10.1016/j.stem.2020.07.002] [Citation(s) in RCA: 132] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Aging has a profound and devastating effect on the brain. Old age is accompanied by declining cognitive function and enhanced risk of brain diseases, including cancer and neurodegenerative disorders. A key question is whether cells with regenerative potential contribute to brain health and even brain "rejuvenation." This review discusses mechanisms that regulate neural stem cells (NSCs) during aging, focusing on the effect of metabolism, genetic regulation, and the surrounding niche. We also explore emerging rejuvenating strategies for old NSCs. Finally, we consider how new technologies may help harness NSCs' potential to restore healthy brain function during physiological and pathological aging.
Collapse
Affiliation(s)
| | - Robin W Yeo
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Anne Brunet
- Department of Genetics, Stanford University, Stanford, CA 94305, USA; Glenn Laboratories for the Biology of Aging, Stanford, CA 94305, USA.
| |
Collapse
|
268
|
Guo J, Zheng HJ, Zhang W, Lou W, Xia C, Han XT, Huang WJ, Zhang F, Wang Y, Liu WJ. Accelerated Kidney Aging in Diabetes Mellitus. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:1234059. [PMID: 32774664 PMCID: PMC7407029 DOI: 10.1155/2020/1234059] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 05/25/2020] [Accepted: 06/25/2020] [Indexed: 02/07/2023]
Abstract
With aging, the kidney undergoes inexorable and progressive changes in structural and functional performance. These aging-related alterations are more obvious and serious in diabetes mellitus (DM). Renal accelerated aging under DM conditions is associated with multiple stresses such as accumulation of advanced glycation end products (AGEs), hypertension, oxidative stress, and inflammation. The main hallmarks of cellular senescence in diabetic kidneys include cyclin-dependent kinase inhibitors, telomere shortening, and diabetic nephropathy-associated secretory phenotype. Lysosome-dependent autophagy and antiaging proteins Klotho and Sirt1 play a fundamental role in the accelerated aging of kidneys in DM, among which the autophagy-lysosome system is the convergent mechanism of the multiple antiaging pathways involved in renal aging under DM conditions. Metformin and the inhibitor of sodium-glucose cotransporter 2 are recommended due to their antiaging effects independent of antihyperglycemia, besides angiotensin-converting enzyme inhibitors/angiotensin receptor blockers. Additionally, diet intervention including low protein and low AGEs with antioxidants are suggested for patients with diabetic nephropathy (DN). However, their long-term benefits still need further study. Exploring the interactive relationships among antiaging protein Klotho, Sirt1, and autophagy-lysosome system may provide insight into better satisfying the urgent medical needs of elderly patients with aging-related DN.
Collapse
Affiliation(s)
- Jing Guo
- Renal Research Institution; Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
| | - Hui Juan Zheng
- Renal Research Institution; Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
| | - Wenting Zhang
- Renal Research Institution; Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
| | - Wenjiao Lou
- Renal Research Institution; Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
| | - Chenhui Xia
- Renal Research Institution; Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
| | - Xue Ting Han
- Renal Research Institution; Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
| | - Wei Jun Huang
- Renal Research Institution; Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
| | - Fan Zhang
- Renal Research Institution; Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
| | - Yaoxian Wang
- Renal Research Institution; Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
| | - Wei Jing Liu
- Renal Research Institution; Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
- Institute of Nephrology, and Zhanjiang Key Laboratory of Prevention and Management of Chronic Kidney Disease, Guangdong Medical University, No. 57th South Renmin Road, Zhanjiang, Guangdong 524001, China
| |
Collapse
|
269
|
Zhang Z, Liu Z, Tian Y. A DNA-Based FLIM Reporter for Simultaneous Quantification of Lysosomal pH and Ca 2+ during Autophagy Regulation. iScience 2020; 23:101344. [PMID: 32688287 PMCID: PMC7369617 DOI: 10.1016/j.isci.2020.101344] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 04/24/2020] [Accepted: 07/01/2020] [Indexed: 01/18/2023] Open
Abstract
pH and Ca2+ play important roles in regulating lysosomal activity and lysosome-mediated physiological and pathological processes. However, effective methods for simultaneous determination of pH and Ca2+ is the bottleneck. Herein, a single DNA-based FLIM reporter was developed for real-time imaging and simultaneous quantification of pH and Ca2+ in lysosomes with high affinity, in which a specific probe for recognition of Ca2+ was assembled onto a DNA nanostructure together with pH-responsive and lysosome-targeted molecules. The developed DNA reporter showed excellent biocompatibility and long-term stability up to ∼56 h in lysosomes. Using this powerful tool, it was discovered that pH was closely related to Ca2+ concentration in lysosome, whereas autophagy can be regulated by lysosomal pH and Ca2+. Furthermore, Aβ-induced neuronal death resulted from autophagy abnormal through lysosomal pH and Ca2+ changes. In addition, lysosomal pH and Ca2+ were found to regulate the transformation of NSCs, resulting in Rapamycin-induced antiaging. A DNA-based FLIM reporter was developed for tracking lysosomal pH and Ca2+ It was found that autophagy could be induced by lysosomal pH and Ca2+ Aβ-induced neuronal death was due to pHly- and [Ca2+]ly-mediated autophagy abnormal Antiaging-related transformation of qNSCs can be regulated by pHly and [Ca2+]ly
Collapse
Affiliation(s)
- Zhonghui Zhang
- State Key Laboratory of Precision Spectroscopy, East China Normal University, Dongchuan Road 500, Shanghai 200241, China; Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, East China Normal University, Dongchuan Road 500, Shanghai 200241, China
| | - Zhichao Liu
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, East China Normal University, Dongchuan Road 500, Shanghai 200241, China.
| | - Yang Tian
- State Key Laboratory of Precision Spectroscopy, East China Normal University, Dongchuan Road 500, Shanghai 200241, China; Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, East China Normal University, Dongchuan Road 500, Shanghai 200241, China.
| |
Collapse
|
270
|
Julian LM, Stanford WL. Organelle Cooperation in Stem Cell Fate: Lysosomes as Emerging Regulators of Cell Identity. Front Cell Dev Biol 2020; 8:591. [PMID: 32733892 PMCID: PMC7358313 DOI: 10.3389/fcell.2020.00591] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 06/17/2020] [Indexed: 12/26/2022] Open
Abstract
Regulation of stem cell fate is best understood at the level of gene and protein regulatory networks, though it is now clear that multiple cellular organelles also have critical impacts. A growing appreciation for the functional interconnectedness of organelles suggests that an orchestration of integrated biological networks functions to drive stem cell fate decisions and regulate metabolism. Metabolic signaling itself has emerged as an integral regulator of cell fate including the determination of identity, activation state, survival, and differentiation potential of many developmental, adult, disease, and cancer-associated stem cell populations and their progeny. As the primary adenosine triphosphate-generating organelles, mitochondria are well-known regulators of stem cell fate decisions, yet it is now becoming apparent that additional organelles such as the lysosome are important players in mediating these dynamic decisions. In this review, we will focus on the emerging role of organelles, in particular lysosomes, in the reprogramming of both metabolic networks and stem cell fate decisions, especially those that impact the determination of cell identity. We will discuss the inter-organelle interactions, cell signaling pathways, and transcriptional regulatory mechanisms with which lysosomes engage and how these activities impact metabolic signaling. We will further review recent data that position lysosomes as critical regulators of cell identity determination programs and discuss the known or putative biological mechanisms. Finally, we will briefly highlight the potential impact of elucidating mechanisms by which lysosomes regulate stem cell identity on our understanding of disease pathogenesis, as well as the development of refined regenerative medicine, biomarker, and therapeutic strategies.
Collapse
Affiliation(s)
- Lisa M. Julian
- Department of Biological Sciences, Simon Fraser University, Burnaby, BC, Canada
| | - William L. Stanford
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
- Ottawa Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
271
|
Kramarski L, Arbely E. Translational read-through promotes aggregation and shapes stop codon identity. Nucleic Acids Res 2020; 48:3747-3760. [PMID: 32128584 PMCID: PMC7144920 DOI: 10.1093/nar/gkaa136] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 02/07/2020] [Accepted: 02/22/2020] [Indexed: 12/14/2022] Open
Abstract
Faithful translation of genetic information depends on the ability of the translational machinery to decode stop codons as termination signals. Although termination of protein synthesis is highly efficient, errors in decoding of stop codons may lead to the synthesis of C-terminally extended proteins. It was found that in eukaryotes such elongated proteins do not accumulate in cells. However, the mechanism for sequestration of C-terminally extended proteins is still unknown. Here we show that 3′-UTR-encoded polypeptides promote aggregation of the C-terminally extended proteins, and targeting to lysosomes. We demonstrate that 3′-UTR-encoded polypeptides can promote different levels of protein aggregation, similar to random sequences. We also show that aggregation of endogenous proteins can be induced by aminoglycoside antibiotics that promote stop codon read-through, by UAG suppressor tRNA, or by knokcdown of release factor 1. Furthermore, we find correlation between the fidelity of termination signals, and the predicted propensity of downstream 3′-UTR-encoded polypeptides to form intrinsically disordered regions. Our data highlight a new quality control mechanism for elimination of C-terminally elongated proteins.
Collapse
Affiliation(s)
- Lior Kramarski
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Eyal Arbely
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel.,Department of Chemistry and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| |
Collapse
|
272
|
Kase Y, Shimazaki T, Okano H. Current understanding of adult neurogenesis in the mammalian brain: how does adult neurogenesis decrease with age? Inflamm Regen 2020; 40:10. [PMID: 32566044 PMCID: PMC7302355 DOI: 10.1186/s41232-020-00122-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 06/08/2020] [Indexed: 02/08/2023] Open
Abstract
Adult neurogenesis occurs throughout life in restricted brain regions in mammals. However, the number of neural stem cells (NSCs) that generate new neurons steadily decreases with age, resulting in a decrease in neurogenesis. Transplantation of mesenchymal cells or cultured NSCs has been studied as a promising treatment in models of several brain injuries including cerebral infarction and cerebral contusion. Considering the problems of host-versus-graft reactions and the tumorigenicity of transplanted cells, the mobilization of endogenous adult NSCs should be more feasible for the treatment of these brain injuries. However, the number of adult NSCs in the adult brain is limited, and their mitotic potential is low. Here, we outline what we know to date about why the number of NSCs and adult neurogenesis decrease with age. We also discuss issues applicable to regenerative medicine.
Collapse
Affiliation(s)
- Yoshitaka Kase
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582 Japan.,Department of Geriatric Medicine, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, 113-8655 Japan
| | - Takuya Shimazaki
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582 Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582 Japan
| |
Collapse
|
273
|
The Antihistamine Deptropine Induces Hepatoma Cell Death through Blocking Autophagosome-Lysosome Fusion. Cancers (Basel) 2020; 12:cancers12061610. [PMID: 32570749 PMCID: PMC7352610 DOI: 10.3390/cancers12061610] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 06/14/2020] [Accepted: 06/16/2020] [Indexed: 02/07/2023] Open
Abstract
Some antihistamines have exhibited significant antitumor activity alone or in combination with other therapies in in vitro and clinical studies. However, the underlying mechanisms of how antihistamines inhibit hepatocellular carcinoma proliferation are still unknown. We first screened the antiproliferation activity of 12 benzocycloheptene structural-analogue drugs, and results showed that deptropine was the most potent inhibitor of both Hep3B and HepG2 human hepatoma cells. Deptropine significantly increased light chain 3B-II (LC3B-II) expression but did not induce sequestosome 1 (SQSTM1/p62) degradation in either cell line. Interestingly, other autophagy-related proteins, such as autophagy-related 7 (ATG7), vacuolar protein sorting 34 (VPS34), phosphorylated adenosine 5'-monophosphate-activated protein kinase (AMPK), and phosphorylated protein kinase B (PKB, also known as Akt), exhibited no significant change in either deptropine-treated cell line. Deptropine also inhibited the processing of cathepsin L from its precursor form to its mature form. Immunofluorescence microscopy showed an increase of autophagosomes in deptropine-treated cells, but deptropine blocked the fusion between autophagosomes and lysosomes. In a xenograft nude mice model, 2.5 mg/kg deptropine showed a great inhibitory effect on Hep3B tumor growth. These results suggest that deptropine can induce in vitro and in vivo hepatoma cell death, and the underlying mechanisms might be mediated through inhibiting autophagy by blocking autophagosome-lysosome fusion.
Collapse
|
274
|
Maghe C, Jacobs KA, Bidère N, Gavard J. [MALT1 in glioblastoma: the Flowers of Evil]. Med Sci (Paris) 2020; 36:452-454. [PMID: 32452364 DOI: 10.1051/medsci/2020073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Clément Maghe
- Équipe SOAP (Signalisation en oncogenèse, angiogenèse et perméabilité), CRCINA (centre de recherche en cancérologie et immunologie Nantes Angers), Inserm, CNRS, Université de Nantes, Université d'Angers, IRSUN (Institut de recherche en santé de l'université de Nantes), 8 quai Moncousu, 44000 Nantes, France
| | - Kathryn A Jacobs
- Équipe SOAP (Signalisation en oncogenèse, angiogenèse et perméabilité), CRCINA (centre de recherche en cancérologie et immunologie Nantes Angers), Inserm, CNRS, Université de Nantes, Université d'Angers, IRSUN (Institut de recherche en santé de l'université de Nantes), 8 quai Moncousu, 44000 Nantes, France
| | - Nicolas Bidère
- Équipe SOAP (Signalisation en oncogenèse, angiogenèse et perméabilité), CRCINA (centre de recherche en cancérologie et immunologie Nantes Angers), Inserm, CNRS, Université de Nantes, Université d'Angers, IRSUN (Institut de recherche en santé de l'université de Nantes), 8 quai Moncousu, 44000 Nantes, France
| | - Julie Gavard
- Équipe SOAP (Signalisation en oncogenèse, angiogenèse et perméabilité), CRCINA (centre de recherche en cancérologie et immunologie Nantes Angers), Inserm, CNRS, Université de Nantes, Université d'Angers, IRSUN (Institut de recherche en santé de l'université de Nantes), 8 quai Moncousu, 44000 Nantes, France. - Institut de cancérologie de l'Ouest (ICO), Saint-Herblain, France
| |
Collapse
|
275
|
Pastore N, Huynh T, Herz NJ, Calcagni' A, Klisch TJ, Brunetti L, Kim KH, De Giorgi M, Hurley A, Carissimo A, Mutarelli M, Aleksieva N, D'Orsi L, Lagor WR, Moore DD, Settembre C, Finegold MJ, Forbes SJ, Ballabio A. TFEB regulates murine liver cell fate during development and regeneration. Nat Commun 2020; 11:2461. [PMID: 32424153 PMCID: PMC7235048 DOI: 10.1038/s41467-020-16300-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 04/20/2020] [Indexed: 12/29/2022] Open
Abstract
It is well established that pluripotent stem cells in fetal and postnatal liver (LPCs) can differentiate into both hepatocytes and cholangiocytes. However, the signaling pathways implicated in the differentiation of LPCs are still incompletely understood. Transcription Factor EB (TFEB), a master regulator of lysosomal biogenesis and autophagy, is known to be involved in osteoblast and myeloid differentiation, but its role in lineage commitment in the liver has not been investigated. Here we show that during development and upon regeneration TFEB drives the differentiation status of murine LPCs into the progenitor/cholangiocyte lineage while inhibiting hepatocyte differentiation. Genetic interaction studies show that Sox9, a marker of precursor and biliary cells, is a direct transcriptional target of TFEB and a primary mediator of its effects on liver cell fate. In summary, our findings identify an unexplored pathway that controls liver cell lineage commitment and whose dysregulation may play a role in biliary cancer.
Collapse
Affiliation(s)
- Nunzia Pastore
- Jan and Dan Duncan Neurological Research Institute, Texas Children Hospital, Houston, TX, 77030, USA.
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA.
| | - Tuong Huynh
- Jan and Dan Duncan Neurological Research Institute, Texas Children Hospital, Houston, TX, 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Niculin J Herz
- Jan and Dan Duncan Neurological Research Institute, Texas Children Hospital, Houston, TX, 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Alessia Calcagni'
- Jan and Dan Duncan Neurological Research Institute, Texas Children Hospital, Houston, TX, 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Tiemo J Klisch
- Jan and Dan Duncan Neurological Research Institute, Texas Children Hospital, Houston, TX, 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Lorenzo Brunetti
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, 77030, USA
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Kangho Ho Kim
- Department of Molecular and Cell Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Marco De Giorgi
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Ayrea Hurley
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Annamaria Carissimo
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, NA, 80078, Italy
| | | | - Niya Aleksieva
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, EH16 4UU, UK
| | - Luca D'Orsi
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, NA, 80078, Italy
| | - William R Lagor
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - David D Moore
- Department of Molecular and Cell Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Carmine Settembre
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, NA, 80078, Italy
- Department of Translational Medicine, Medical Genetics, Federico II University, Naples, 80131, Italy
| | - Milton J Finegold
- Department of Pathology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Stuart J Forbes
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, EH16 4UU, UK
| | - Andrea Ballabio
- Jan and Dan Duncan Neurological Research Institute, Texas Children Hospital, Houston, TX, 77030, USA.
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA.
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, NA, 80078, Italy.
- Department of Translational Medicine, Medical Genetics, Federico II University, Naples, 80131, Italy.
| |
Collapse
|
276
|
Durso W, Martins M, Marchetti L, Cremisi F, Luin S, Cardarelli F. Lysosome Dynamic Properties during Neuronal Stem Cell Differentiation Studied by Spatiotemporal Fluctuation Spectroscopy and Organelle Tracking. Int J Mol Sci 2020; 21:ijms21093397. [PMID: 32403391 PMCID: PMC7247004 DOI: 10.3390/ijms21093397] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 05/08/2020] [Accepted: 05/09/2020] [Indexed: 01/20/2023] Open
Abstract
We investigated lysosome dynamics during neuronal stem cell (NSC) differentiation by two quantitative and complementary biophysical methods based on fluorescence: imaging-derived mean square displacement (iMSD) and single-particle tracking (SPT). The former extracts the average dynamics and size of the whole population of moving lysosomes directly from imaging, with no need to calculate single trajectories; the latter resolves the finest heterogeneities and dynamic features at the single-lysosome level, which are lost in the iMSD analysis. In brief, iMSD analysis reveals that, from a structural point of view, lysosomes decrement in size during NSC differentiation, from 1 μm average diameter in the embryonic cells to approximately 500 nm diameter in the fully differentiated cells. Concomitantly, iMSD analysis highlights modification of key dynamic parameters, such as the average local organelle diffusivity and anomalous coefficient, which may parallel cytoskeleton remodeling during the differentiation process. From average to local, SPT allows mapping heterogeneous dynamic responses of single lysosomes in different districts of the cells. For instance, a dramatic decrease of lysosomal transport in the soma is followed by a rapid increase of transport in the projections at specific time points during neuronal differentiation, an observation compatible with the hypothesis that lysosomal active mobilization shifts from the soma to the newborn projections. Our combined results provide new insight into the lysosome size and dynamics regulation throughout NSC differentiation, supporting new functions proposed for this organelle.
Collapse
Affiliation(s)
- William Durso
- NEST Laboratory—Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy;
| | - Manuella Martins
- Bio@SNS Laboratory—Scuola Normale Superiore, via G. Moruzzi, 1, 56126 Pisa, Italy; (M.M.); (F.C.)
| | - Laura Marchetti
- Center for Nanotechnology Innovation@NEST (CNI@NEST), Piazza San Silvestro 12, 56126 Pisa, Italy;
| | - Federico Cremisi
- Bio@SNS Laboratory—Scuola Normale Superiore, via G. Moruzzi, 1, 56126 Pisa, Italy; (M.M.); (F.C.)
| | - Stefano Luin
- NEST Laboratory—Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy;
- NEST, Istituto Nanoscienze, CNR, Piazza San Silvestro 12, 56127 Pisa, Italy
- Correspondence: (S.L.); (F.C.)
| | - Francesco Cardarelli
- NEST Laboratory—Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy;
- Correspondence: (S.L.); (F.C.)
| |
Collapse
|
277
|
Hu G, Xia Y, Zhang J, Chen Y, Yuan J, Niu X, Zhao B, Li Q, Wang Y, Deng Z. ESC-sEVs Rejuvenate Senescent Hippocampal NSCs by Activating Lysosomes to Improve Cognitive Dysfunction in Vascular Dementia. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:1903330. [PMID: 32440476 PMCID: PMC7237844 DOI: 10.1002/advs.201903330] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 02/20/2020] [Accepted: 02/24/2020] [Indexed: 05/05/2023]
Abstract
Vascular dementia (VD) is one of the most common types of dementia, however, the intrinsic mechanism is unclear and there is still lack of effective medications. In this study, the VD rats exhibit a progressive cognitive impairment, as well as a time-related increasing in hippocampal neural stem cells (H-NSCs) senescence, lost and neurogenesis decline. Then, embryonic stem cell-derived small extracellular vesicles (ESC-sEVs) are intravenously injected into VD rats. ESC-sEVs treatment significantly alleviates H-NSCs senescence, recovers compromised proliferation and neuron differentiation capacity, and reverses cognitive impairment. By microarray analysis and RT-qPCR it is identified that several miRNAs including miR-17-5p, miR-18a-5p, miR-21-5p, miR-29a-3p, and let-7a-5p, that can inhibit mTORC1 activation, exist in ESC-sEVs. ESC-sEVs rejuvenate H-NSCs senescence partly by transferring these miRNAs to inhibit mTORC1 activation, promote transcription factor EB (TFEB) nuclear translocation and lysosome resumption. Taken together, these data indicate that H-NSCs senescence cause cell depletion, neurogenesis reduction, and cognitive impairment in VD. ESC-sEVs treatment ameliorates H-NSCs senescence by inhibiting mTORC1 activation, and promoting TFEB nuclear translocation and lysosome resumption, thereby reversing senescence-related neurogenesis dysfunction and cognitive impairment in VD. The application of ESC-sEVs may be a novel cell-free therapeutic tool for patients with VD, as well as other aging-related diseases.
Collapse
Affiliation(s)
- Guowen Hu
- Department of NeurosurgeryShanghai Jiaotong University Affiliated Sixth People's HospitalShanghai200233China
| | - Yuguo Xia
- Department of NeurosurgeryShanghai Jiaotong University Affiliated Sixth People's HospitalShanghai200233China
| | - Juntao Zhang
- Institute of Microsurgery on ExtremitiesShanghai Jiaotong University Affiliated Sixth People's HospitalShanghai200233China
| | - Yu Chen
- Institute of Microsurgery on ExtremitiesShanghai Jiaotong University Affiliated Sixth People's HospitalShanghai200233China
| | - Ji Yuan
- Institute of Microsurgery on ExtremitiesShanghai Jiaotong University Affiliated Sixth People's HospitalShanghai200233China
| | - Xin Niu
- Institute of Microsurgery on ExtremitiesShanghai Jiaotong University Affiliated Sixth People's HospitalShanghai200233China
| | - Bizeng Zhao
- Department of Orthopedic SurgeryShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233China
| | - Qing Li
- Institute of Microsurgery on ExtremitiesShanghai Jiaotong University Affiliated Sixth People's HospitalShanghai200233China
| | - Yang Wang
- Institute of Microsurgery on ExtremitiesShanghai Jiaotong University Affiliated Sixth People's HospitalShanghai200233China
| | - Zhifeng Deng
- Department of NeurosurgeryShanghai Jiaotong University Affiliated Sixth People's HospitalShanghai200233China
| |
Collapse
|
278
|
Zhang DY, Gao T, Xu RJ, Sun L, Zhang CF, Bai L, Chen W, Liu KY, Zhou Y, Jiao X, Zhang GH, Guo RL, Li JX, Gao Y, Jiao WJ, Tian H. SIRT3 Transfection of Aged Human Bone Marrow-Derived Mesenchymal Stem Cells Improves Cell Therapy-Mediated Myocardial Repair. Rejuvenation Res 2020; 23:453-464. [PMID: 32228121 DOI: 10.1089/rej.2019.2260] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Sirtuin 3 (SIRT3) is a deacetylase important for antioxidant protection, cell longevity, and aging. We hypothesized that SIRT3 improve oxidative resistance of aged cells and improve cell therapy in aged patients. In vitro, the proliferation and oxidative resistance of human mesenchymal stem cells (hMSCs) significantly declined with age. The expression and activity of antioxidant enzymes, including catalase (CAT) and manganese superoxide dismutase (MnSOD), increased after transfection of SIRT3 in hMSCs from older donors (O-hMSCs). The protein level of Forkhead box O3a (FOXO3a) in nucleus increased after SIRT3 overexpression. The antioxidant capacity of O-hMSCs increased after SIRT3 overexpression. 3-Amino-1,2,4-triazole (3-AT, CAT inhibitor) or diethyldithiocarbamate (DETC, SOD inhibitor) that was used to inhibit CAT or SOD activity significantly blocked the antioxidant function of SIRT3. When two inhibitors were used together, the antioxidant function of SIRT3 almost disappeared. Following myocardial infarction and intramyocardial injections of O-hMSCs in rats in vivo, the survival rate of O-hMSCs increased by SIRT3 transfection. The cardiac function of rats was improved after SIRT3-overexpressed O-hMSC transplantation. The infarct size, collagen content, and expression levels of matrix metalloproteinase 2 (MMP2) and MMP9 decreased. Besides, the protein level of vascular endothelial growth factor A and vascular density increased after cell transplantation with SIRT3-modified O-hMSCs. These results indicate that damage resistance of hMSCs decline with age and SIRT3 might protect O-hMSCs against oxidative damage by activating CAT and MnSOD through transferring FOXO3a into nucleus. Meanwhile, the therapeutic effect of aged hMSC transplantation can be improved by SIRT3 overexpression.
Collapse
Affiliation(s)
- Dong-Yang Zhang
- Department of Thoracic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China.,Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Tong Gao
- Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Department of Cardiovascular Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Rong-Jian Xu
- Department of Thoracic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Lu Sun
- Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Department of Cardiovascular Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chun-Feng Zhang
- Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Department of Cardiovascular Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Long Bai
- Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Department of Cardiovascular Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wei Chen
- Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Department of Cardiovascular Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Kai-Yu Liu
- Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Department of Cardiovascular Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yang Zhou
- Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Department of Cardiovascular Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xuan Jiao
- Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Department of Cardiovascular Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Gui-Huan Zhang
- Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Department of Cardiovascular Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Rui-Lin Guo
- The Second Clinical College of Harbin Medical University, Harbin, China
| | - Jing-Xuan Li
- The Second Clinical College of Harbin Medical University, Harbin, China
| | - Ying Gao
- The Second Clinical College of Harbin Medical University, Harbin, China
| | - Wen-Jie Jiao
- Department of Thoracic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hai Tian
- Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Department of Cardiovascular Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
279
|
Isaev NK, Stelmashook EV, Genrikhs EE. Neurogenesis and brain aging. Rev Neurosci 2020; 30:573-580. [PMID: 30763272 DOI: 10.1515/revneuro-2018-0084] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 11/18/2018] [Indexed: 12/13/2022]
Abstract
Human aging affects the entire organism, but aging of the brain must undoubtedly be different from that of all other organs, as neurons are highly differentiated postmitotic cells, for the majority of which the lifespan in the postnatal period is equal to the lifespan of the entire organism. In this work, we examine the distinctive features of brain aging and neurogenesis during normal aging, pathological aging (Alzheimer's disease), and accelerated aging (Hutchinson-Gilford progeria syndrome and Werner syndrome).
Collapse
Affiliation(s)
- Nickolay K Isaev
- M.V. Lomonosov Moscow State University, N.A. Belozersky Institute of Physico-Chemical Biology, Biological Faculty, Moscow 119991, Russia.,Research Center of Neurology, Moscow 125367, Russia
| | | | | |
Collapse
|
280
|
Vonk WIM, Rainbolt TK, Dolan PT, Webb AE, Brunet A, Frydman J. Differentiation Drives Widespread Rewiring of the Neural Stem Cell Chaperone Network. Mol Cell 2020; 78:329-345.e9. [PMID: 32268122 PMCID: PMC7288733 DOI: 10.1016/j.molcel.2020.03.009] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 11/26/2019] [Accepted: 03/08/2020] [Indexed: 12/15/2022]
Abstract
Neural stem and progenitor cells (NSPCs) are critical for continued cellular replacement in the adult brain. Lifelong maintenance of a functional NSPC pool necessitates stringent mechanisms to preserve a pristine proteome. We find that the NSPC chaperone network robustly maintains misfolded protein solubility and stress resilience through high levels of the ATP-dependent chaperonin TRiC/CCT. Strikingly, NSPC differentiation rewires the cellular chaperone network, reducing TRiC/CCT levels and inducing those of the ATP-independent small heat shock proteins (sHSPs). This switches the proteostasis strategy in neural progeny cells to promote sequestration of misfolded proteins into protective inclusions. The chaperone network of NSPCs is more effective than that of differentiated cells, leading to improved management of proteotoxic stress and amyloidogenic proteins. However, NSPC proteostasis is impaired by brain aging. The less efficient chaperone network of differentiated neural progeny may contribute to their enhanced susceptibility to neurodegenerative diseases characterized by aberrant protein misfolding and aggregation.
Collapse
Affiliation(s)
| | - T Kelly Rainbolt
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Patrick T Dolan
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Ashley E Webb
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI 02912, USA; Carney Institute for Brain Science, Brown University, Providence, RI 02912, USA; Center on the Biology of Aging, Brown University, Providence, RI 02912, USA
| | - Anne Brunet
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Judith Frydman
- Department of Biology, Stanford University, Stanford, CA 94305, USA; Department of Genetics, Stanford University, Stanford, CA 94305, USA; Chan Zuckerberg Biohub, San Francisco, CA 94158, USA.
| |
Collapse
|
281
|
Gerisch B, Tharyan RG, Mak J, Denzel SI, Popkes-van Oepen T, Henn N, Antebi A. HLH-30/TFEB Is a Master Regulator of Reproductive Quiescence. Dev Cell 2020; 53:316-329.e5. [PMID: 32302543 DOI: 10.1016/j.devcel.2020.03.014] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 01/28/2020] [Accepted: 03/15/2020] [Indexed: 12/20/2022]
Abstract
All animals have evolved the ability to survive nutrient deprivation, and nutrient signaling pathways are conserved modulators of health and disease. In C. elegans, late-larval starvation provokes the adult reproductive diapause (ARD), a long-lived quiescent state that enables survival for months without food, yet underlying molecular mechanisms remain unknown. Here, we show that ARD is distinct from other forms of diapause, showing little requirement for canonical longevity pathways, autophagy, and fat metabolism. Instead it requires the HLH-30/TFEB transcription factor to promote the morphological and physiological remodeling involved in ARD entry, survival, and recovery, suggesting that HLH-30 is a master regulator of reproductive quiescence. HLH-30 transcriptome and genetic analyses reveal that Max-like HLH factors, AMP-kinase, mTOR, protein synthesis, and mitochondrial fusion are target processes that promote ARD longevity. ARD thus rewires metabolism to ensure long-term survival and may illuminate similar mechanisms acting in stem cell quiescence and long-term fasting.
Collapse
Affiliation(s)
- Birgit Gerisch
- Department of Molecular Genetics of Ageing, Max Planck Institute for Biology of Ageing, Cologne 50931, Germany
| | - Rebecca George Tharyan
- Department of Molecular Genetics of Ageing, Max Planck Institute for Biology of Ageing, Cologne 50931, Germany
| | - Jennifer Mak
- Department of Molecular Genetics of Ageing, Max Planck Institute for Biology of Ageing, Cologne 50931, Germany
| | - Sarah I Denzel
- Department of Molecular Genetics of Ageing, Max Planck Institute for Biology of Ageing, Cologne 50931, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases, University of Cologne, Cologne 50931, Germany
| | - Till Popkes-van Oepen
- Department of Molecular Genetics of Ageing, Max Planck Institute for Biology of Ageing, Cologne 50931, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases, University of Cologne, Cologne 50931, Germany
| | - Nadine Henn
- Department of Molecular Genetics of Ageing, Max Planck Institute for Biology of Ageing, Cologne 50931, Germany
| | - Adam Antebi
- Department of Molecular Genetics of Ageing, Max Planck Institute for Biology of Ageing, Cologne 50931, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases, University of Cologne, Cologne 50931, Germany.
| |
Collapse
|
282
|
Thiruvalluvan A, de Mattos EP, Brunsting JF, Bakels R, Serlidaki D, Barazzuol L, Conforti P, Fatima A, Koyuncu S, Cattaneo E, Vilchez D, Bergink S, Boddeke EHWG, Copray S, Kampinga HH. DNAJB6, a Key Factor in Neuronal Sensitivity to Amyloidogenesis. Mol Cell 2020; 78:346-358.e9. [PMID: 32268123 DOI: 10.1016/j.molcel.2020.02.022] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 12/31/2019] [Accepted: 02/25/2020] [Indexed: 01/09/2023]
Abstract
CAG-repeat expansions in at least eight different genes cause neurodegeneration. The length of the extended polyglutamine stretches in the corresponding proteins is proportionally related to their aggregation propensity. Although these proteins are ubiquitously expressed, they predominantly cause toxicity to neurons. To understand this neuronal hypersensitivity, we generated induced pluripotent stem cell (iPSC) lines of spinocerebellar ataxia type 3 and Huntington's disease patients. iPSC generation and neuronal differentiation are unaffected by polyglutamine proteins and show no spontaneous aggregate formation. However, upon glutamate treatment, aggregates form in neurons but not in patient-derived neural progenitors. During differentiation, the chaperone network is drastically rewired, including loss of expression of the anti-amyloidogenic chaperone DNAJB6. Upregulation of DNAJB6 in neurons antagonizes glutamate-induced aggregation, while knockdown of DNAJB6 in progenitors results in spontaneous polyglutamine aggregation. Loss of DNAJB6 expression upon differentiation is confirmed in vivo, explaining why stem cells are intrinsically protected against amyloidogenesis and protein aggregates are dominantly present in neurons.
Collapse
Affiliation(s)
- Arun Thiruvalluvan
- Department of Biomedical Sciences of Cells & Systems, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Eduardo P de Mattos
- Department of Biomedical Sciences of Cells & Systems, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Jeanette F Brunsting
- Department of Biomedical Sciences of Cells & Systems, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Rob Bakels
- Department of Biomedical Sciences of Cells & Systems, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Despina Serlidaki
- Department of Biomedical Sciences of Cells & Systems, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Lara Barazzuol
- Department of Biomedical Sciences of Cells & Systems, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Paola Conforti
- Department of Biosciences, University of Milan, Milan, Italy; Istituto Nazionale di Genetica Molecolare, Romeo ed Enrica Invernizzi, Milan, Italy
| | - Azra Fatima
- Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Seda Koyuncu
- Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Elena Cattaneo
- Department of Biosciences, University of Milan, Milan, Italy; Istituto Nazionale di Genetica Molecolare, Romeo ed Enrica Invernizzi, Milan, Italy
| | - David Vilchez
- Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Steven Bergink
- Department of Biomedical Sciences of Cells & Systems, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Erik H W G Boddeke
- Department of Biomedical Sciences of Cells & Systems, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Sjef Copray
- Department of Biomedical Sciences of Cells & Systems, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Harm H Kampinga
- Department of Biomedical Sciences of Cells & Systems, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
| |
Collapse
|
283
|
Gao J, Liao Y, Qiu M, Shen W. Wnt/β-Catenin Signaling in Neural Stem Cell Homeostasis and Neurological Diseases. Neuroscientist 2020; 27:58-72. [PMID: 32242761 DOI: 10.1177/1073858420914509] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Neural stem/progenitor cells (NSCs) maintain the ability of self-renewal and differentiation and compose the complex nervous system. Wnt signaling is thought to control the balance of NSC proliferation and differentiation via the transcriptional coactivator β-catenin during brain development and adult tissue homeostasis. Disruption of Wnt signaling may result in developmental defects and neurological diseases. Here, we summarize recent findings of the roles of Wnt/β-catenin signaling components in NSC homeostasis for the regulation of functional brain circuits. We also suggest that the potential role of Wnt/β-catenin signaling might lead to new therapeutic strategies for neurological diseases, including, but not limited to, spinal cord injury, Alzheimer's disease, Parkinson's disease, and depression.
Collapse
Affiliation(s)
- Juanmei Gao
- Zhejiang Key Laboratory of Organ Development and Regeneration, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, China.,College of Life and Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yuan Liao
- Zhejiang Key Laboratory of Organ Development and Regeneration, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Mengsheng Qiu
- Zhejiang Key Laboratory of Organ Development and Regeneration, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, China.,College of Life and Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Wanhua Shen
- Zhejiang Key Laboratory of Organ Development and Regeneration, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, China
| |
Collapse
|
284
|
Maybury-Lewis SY, Webb AE. Taking Prisoners: Vimentin Cages Capture Proteasomes during NSC Activation. Cell Stem Cell 2020; 26:473-475. [PMID: 32243805 DOI: 10.1016/j.stem.2020.03.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Asymmetric partitioning of damaged proteins is thought to play a key role in preserving stem cell function with age. In this issue of Cell Stem Cell, Morrow et al. (2020) show that vimentin recruits proteasome machinery to aggresomes to control NSC proteostasis during quiescence exit.
Collapse
Affiliation(s)
- Sun Y Maybury-Lewis
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI 02912, USA
| | - Ashley E Webb
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI 02912, USA.
| |
Collapse
|
285
|
Zhuang J, Yang H, Li Y, Wang B, Li N, Zhao N. Efficient photosensitizers with aggregation-induced emission characteristics for lysosome- and Gram-positive bacteria-targeted photodynamic therapy. Chem Commun (Camb) 2020; 56:2630-2633. [PMID: 32016259 DOI: 10.1039/d0cc00394h] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Two efficient photosensitizers (PSs) with aggregation-induced emission characteristics were designed and synthesized for specific lysosome-targeted photodynamic therapy (PDT). Both PSs efficiently discriminated Gram-positive bacteria from Gram-negative bacteria and killed Gram-positive bacteria through the PDT effect.
Collapse
Affiliation(s)
- Jiabao Zhuang
- Key Laboratory of Macromolecular Science of Shaanxi Province, Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, School of Chemistry & Chemical Engineering, Shaanxi Normal University, 710119 Xi'an, China.
| | - Hanxiao Yang
- Key Laboratory of Macromolecular Science of Shaanxi Province, Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, School of Chemistry & Chemical Engineering, Shaanxi Normal University, 710119 Xi'an, China.
| | - Yue Li
- Key Laboratory of Macromolecular Science of Shaanxi Province, Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, School of Chemistry & Chemical Engineering, Shaanxi Normal University, 710119 Xi'an, China.
| | - Bing Wang
- Key Laboratory of Macromolecular Science of Shaanxi Province, Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, School of Chemistry & Chemical Engineering, Shaanxi Normal University, 710119 Xi'an, China.
| | - Nan Li
- Key Laboratory of Macromolecular Science of Shaanxi Province, Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, School of Chemistry & Chemical Engineering, Shaanxi Normal University, 710119 Xi'an, China.
| | - Na Zhao
- Key Laboratory of Macromolecular Science of Shaanxi Province, Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, School of Chemistry & Chemical Engineering, Shaanxi Normal University, 710119 Xi'an, China.
| |
Collapse
|
286
|
Qu A, Wu X, Li S, Sun M, Xu L, Kuang H, Xu C. An NIR-Responsive DNA-Mediated Nanotetrahedron Enhances the Clearance of Senescent Cells. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e2000184. [PMID: 32100405 DOI: 10.1002/adma.202000184] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 01/21/2020] [Indexed: 06/10/2023]
Abstract
Senescence is a state of stable cell cycle arrest that can escape apoptosis and lead to aging and numerous age-related diseases. In this study, an upconversion-nanoparticle (UCNP)-centered Au20 -Au30 nanoparticles tetrahedron (UAuTe) is prepared by DNA hybridization, which can selectively accelerate the clearance of senescent cells. When the beta-2-microglobulin antibody (anti-B2MG) on the Au NPs recognizes senescent cells, the application of near-infrared (NIR) light induces the disassembly of the UAuTe by breaking the boronic ester linkage. Subsequently, the Granzyme B exposed on the UCNPs induces apoptosis in senescent cells, which can then be tracked by changes in fluorescence. It is found that, as compared to single Granzyme B, the UAuTe can not only control the Granzyme B delivery by NIR-responsivity, but also synergistically target and activate the Granzyme B in the senescent cell without the need of perforin. Moreover, this tool is applied successfully in vivo; the results demonstrate that the NIR-responsive tetrahedron can restore renal function, tissue homeostasis, fur density, and athletic ability in a mouse model of senescence after 30 d of treatment. The NIR-induced tetrahedron provides a practical strategy for clinical diagnosis and therapy, particularly for aging and age-related diseases.
Collapse
Affiliation(s)
- Aihua Qu
- Key Lab of Synthetic and Biological Colloids, Ministry of Education, International Joint Research Laboratory for Biointerface and Biodetection, State Key Lab of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, P. R. China
| | - Xiaoling Wu
- Key Lab of Synthetic and Biological Colloids, Ministry of Education, International Joint Research Laboratory for Biointerface and Biodetection, State Key Lab of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, P. R. China
| | - Si Li
- Key Lab of Synthetic and Biological Colloids, Ministry of Education, International Joint Research Laboratory for Biointerface and Biodetection, State Key Lab of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, P. R. China
| | - Maozhong Sun
- Key Lab of Synthetic and Biological Colloids, Ministry of Education, International Joint Research Laboratory for Biointerface and Biodetection, State Key Lab of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, P. R. China
| | - Liguang Xu
- Key Lab of Synthetic and Biological Colloids, Ministry of Education, International Joint Research Laboratory for Biointerface and Biodetection, State Key Lab of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, P. R. China
| | - Hua Kuang
- Key Lab of Synthetic and Biological Colloids, Ministry of Education, International Joint Research Laboratory for Biointerface and Biodetection, State Key Lab of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, P. R. China
| | - Chuanlai Xu
- Key Lab of Synthetic and Biological Colloids, Ministry of Education, International Joint Research Laboratory for Biointerface and Biodetection, State Key Lab of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, P. R. China
| |
Collapse
|
287
|
Schmukler E, Pinkas‐Kramarski R. Autophagy induction in the treatment of Alzheimer's disease. Drug Dev Res 2020; 81:184-193. [DOI: 10.1002/ddr.21605] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 09/05/2019] [Accepted: 09/10/2019] [Indexed: 02/06/2023]
Affiliation(s)
- Eran Schmukler
- Department of NeurobiologySchool of Neurobiology, Biochemistry and Biophysics, the George S. Wise Faculty of Life Sciences, Tel‐Aviv University Ramat‐Aviv Israel
| | - Ronit Pinkas‐Kramarski
- Department of NeurobiologySchool of Neurobiology, Biochemistry and Biophysics, the George S. Wise Faculty of Life Sciences, Tel‐Aviv University Ramat‐Aviv Israel
| |
Collapse
|
288
|
Chang NC. Autophagy and Stem Cells: Self-Eating for Self-Renewal. Front Cell Dev Biol 2020; 8:138. [PMID: 32195258 PMCID: PMC7065261 DOI: 10.3389/fcell.2020.00138] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Accepted: 02/19/2020] [Indexed: 12/22/2022] Open
Abstract
Autophagy is a fundamental cell survival mechanism that allows cells to adapt to metabolic stress through the degradation and recycling of intracellular components to generate macromolecular precursors and produce energy. The autophagy pathway is critical for development, maintaining cellular and tissue homeostasis, as well as immunity and prevention of human disease. Defects in autophagy have been attributed to cancer, neurodegeneration, muscle and heart disease, infectious disease, as well as aging. While autophagy has classically been viewed as a passive quality control and general house-keeping mechanism, emerging evidence demonstrates that autophagy is an active process that regulates the metabolic status of the cell. Adult stem cells, which are long-lived cells that possess the unique ability to self-renew and differentiate into specialized cells throughout the body, have distinct metabolic requirements. Research in a variety of stem cell types have established that autophagy plays critical roles in stem cell quiescence, activation, differentiation, and self-renewal. Here, we will review the evidence demonstrating that autophagy is a key regulator of stem cell function and how defective stem cell autophagy contributes to degenerative disease, aging and the generation of cancer stem cells. Moreover, we will discuss the merits of targeting autophagy as a regenerative medicine strategy to promote stem cell function and improve stem cell-based therapies.
Collapse
Affiliation(s)
- Natasha C Chang
- Department of Biochemistry, Faculty of Medicine, McGill University, Montreal, QC, Canada
| |
Collapse
|
289
|
He M, Chiang HH, Luo H, Zheng Z, Qiao Q, Wang L, Tan M, Ohkubo R, Mu WC, Zhao S, Wu H, Chen D. An Acetylation Switch of the NLRP3 Inflammasome Regulates Aging-Associated Chronic Inflammation and Insulin Resistance. Cell Metab 2020; 31:580-591.e5. [PMID: 32032542 PMCID: PMC7104778 DOI: 10.1016/j.cmet.2020.01.009] [Citation(s) in RCA: 246] [Impact Index Per Article: 49.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 06/13/2019] [Accepted: 01/13/2020] [Indexed: 12/20/2022]
Abstract
It is well documented that the rate of aging can be slowed, but it remains unclear to which extent aging-associated conditions can be reversed. How the interface of immunity and metabolism impinges upon the diabetes pandemic is largely unknown. Here, we show that NLRP3, a pattern recognition receptor, is modified by acetylation in macrophages and is deacetylated by SIRT2, an NAD+-dependent deacetylase and a metabolic sensor. We have developed a cell-based system that models aging-associated inflammation, a defined co-culture system that simulates the effects of inflammatory milieu on insulin resistance in metabolic tissues during aging, and aging mouse models; and demonstrate that SIRT2 and NLRP3 deacetylation prevent, and can be targeted to reverse, aging-associated inflammation and insulin resistance. These results establish the dysregulation of the acetylation switch of the NLRP3 inflammasome as an origin of aging-associated chronic inflammation and highlight the reversibility of aging-associated chronic inflammation and insulin resistance.
Collapse
Affiliation(s)
- Ming He
- Program in Metabolic Biology, Nutritional Sciences & Toxicology, University of California, Berkeley, CA 94720, USA
| | - Hou-Hsien Chiang
- Program in Metabolic Biology, Nutritional Sciences & Toxicology, University of California, Berkeley, CA 94720, USA
| | - Hanzhi Luo
- Program in Metabolic Biology, Nutritional Sciences & Toxicology, University of California, Berkeley, CA 94720, USA
| | - Zhifang Zheng
- Program in Metabolic Biology, Nutritional Sciences & Toxicology, University of California, Berkeley, CA 94720, USA
| | - Qi Qiao
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA; Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA
| | - Li Wang
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA; Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA
| | - Mingdian Tan
- Program in Metabolic Biology, Nutritional Sciences & Toxicology, University of California, Berkeley, CA 94720, USA
| | - Rika Ohkubo
- Program in Metabolic Biology, Nutritional Sciences & Toxicology, University of California, Berkeley, CA 94720, USA
| | - Wei-Chieh Mu
- Program in Metabolic Biology, Nutritional Sciences & Toxicology, University of California, Berkeley, CA 94720, USA
| | - Shimin Zhao
- School of Life Sciences, Fudan University, Shanghai, China
| | - Hao Wu
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA; Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA
| | - Danica Chen
- Program in Metabolic Biology, Nutritional Sciences & Toxicology, University of California, Berkeley, CA 94720, USA.
| |
Collapse
|
290
|
Abstract
Ageing is a major risk factor for the development of many diseases, prominently including neurodegenerative disorders such as Alzheimer disease and Parkinson disease. A hallmark of many age-related diseases is the dysfunction in protein homeostasis (proteostasis), leading to the accumulation of protein aggregates. In healthy cells, a complex proteostasis network, comprising molecular chaperones and proteolytic machineries and their regulators, operates to ensure the maintenance of proteostasis. These factors coordinate protein synthesis with polypeptide folding, the conservation of protein conformation and protein degradation. However, sustaining proteome balance is a challenging task in the face of various external and endogenous stresses that accumulate during ageing. These stresses lead to the decline of proteostasis network capacity and proteome integrity. The resulting accumulation of misfolded and aggregated proteins affects, in particular, postmitotic cell types such as neurons, manifesting in disease. Recent analyses of proteome-wide changes that occur during ageing inform strategies to improve proteostasis. The possibilities of pharmacological augmentation of the capacity of proteostasis networks hold great promise for delaying the onset of age-related pathologies associated with proteome deterioration and for extending healthspan.
Collapse
|
291
|
Restraining Lysosomal Activity Preserves Hematopoietic Stem Cell Quiescence and Potency. Cell Stem Cell 2020; 26:359-376.e7. [PMID: 32109377 DOI: 10.1016/j.stem.2020.01.013] [Citation(s) in RCA: 161] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 10/17/2019] [Accepted: 01/17/2020] [Indexed: 12/18/2022]
Abstract
Quiescence is a fundamental property that maintains hematopoietic stem cell (HSC) potency throughout life. Quiescent HSCs are thought to rely on glycolysis for their energy, but the overall metabolic properties of HSCs remain elusive. Using combined approaches, including single-cell RNA sequencing (RNA-seq), we show that mitochondrial membrane potential (MMP) distinguishes quiescent from cycling-primed HSCs. We found that primed, but not quiescent, HSCs relied readily on glycolysis. Notably, in vivo inhibition of glycolysis enhanced the competitive repopulation ability of primed HSCs. We further show that HSC quiescence is maintained by an abundance of large lysosomes. Repression of lysosomal activation in HSCs led to further enlargement of lysosomes while suppressing glucose uptake. This also induced increased lysosomal sequestration of mitochondria and enhanced the competitive repopulation ability of primed HSCs by over 90-fold in vivo. These findings show that restraining lysosomal activity preserves HSC quiescence and potency and may be therapeutically relevant.
Collapse
|
292
|
Morrow CS, Porter TJ, Xu N, Arndt ZP, Ako-Asare K, Heo HJ, Thompson EAN, Moore DL. Vimentin Coordinates Protein Turnover at the Aggresome during Neural Stem Cell Quiescence Exit. Cell Stem Cell 2020; 26:558-568.e9. [PMID: 32109376 DOI: 10.1016/j.stem.2020.01.018] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 10/31/2019] [Accepted: 01/23/2020] [Indexed: 01/08/2023]
Abstract
Maintaining a healthy proteome throughout life is critical for proper somatic stem cell function, but the complexities of the stem cell response to increases in damaged or aggregated proteins remain unclear. Here we demonstrate that adult neural stem cells (NSCs) utilize aggresomes to recover from disrupted proteostasis and describe a novel function for the intermediate filament vimentin in proteostasis as a spatial coordinator of proteasomes to the aggresome. In the absence of vimentin, NSCs have a reduced capacity to exit quiescence, a time when NSCs are required to clear a wave of aggregated proteins, and demonstrate an early age-dependent decline in proliferation and neurogenesis. Taken together, these data reveal a significant role of vimentin and aggresomes in the regulation of proteostasis during quiescent NSC activation.
Collapse
Affiliation(s)
- Christopher S Morrow
- Department of Neuroscience, University of Wisconsin - Madison, Madison, WI 53705, USA
| | - Tiaira J Porter
- Department of Neuroscience, University of Wisconsin - Madison, Madison, WI 53705, USA
| | - Nan Xu
- Department of Neuroscience, University of Wisconsin - Madison, Madison, WI 53705, USA
| | - Zachary P Arndt
- Department of Neuroscience, University of Wisconsin - Madison, Madison, WI 53705, USA
| | - Kayla Ako-Asare
- Department of Neuroscience, University of Wisconsin - Madison, Madison, WI 53705, USA
| | - Helen J Heo
- Department of Neuroscience, University of Wisconsin - Madison, Madison, WI 53705, USA
| | | | - Darcie L Moore
- Department of Neuroscience, University of Wisconsin - Madison, Madison, WI 53705, USA.
| |
Collapse
|
293
|
Liang WJ, Gustafsson ÅB. The Aging Heart: Mitophagy at the Center of Rejuvenation. Front Cardiovasc Med 2020; 7:18. [PMID: 32140472 PMCID: PMC7042393 DOI: 10.3389/fcvm.2020.00018] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 02/03/2020] [Indexed: 12/22/2022] Open
Abstract
Aging is associated with structural and functional changes in the heart and is a major risk factor in developing cardiovascular disease. Many recent studies have focused on increasing our understanding of the basis of aging at the cellular and molecular levels in various tissues, including the heart. It is known that there is an age-related decline in cellular quality control pathways such as autophagy and mitophagy, which leads to accumulation of potentially harmful cellular components in cardiac myocytes. There is evidence that diminished autophagy and mitophagy accelerate the aging process, while enhancement preserves cardiac homeostasis and extends life span. Here, we review the current knowledge of autophagy and mitophagy in aging and discuss how age-associated alterations in these processes contribute to cardiac aging and age-related cardiovascular diseases.
Collapse
Affiliation(s)
- Wenjing J Liang
- Department of Pharmacology, Department of Medicine, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, CA, United States
| | - Åsa B Gustafsson
- Department of Pharmacology, Department of Medicine, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, CA, United States
| |
Collapse
|
294
|
Zambusi A, Pelin Burhan Ö, Di Giaimo R, Schmid B, Ninkovic J. Granulins Regulate Aging Kinetics in the Adult Zebrafish Telencephalon. Cells 2020; 9:E350. [PMID: 32028681 PMCID: PMC7072227 DOI: 10.3390/cells9020350] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 01/31/2020] [Accepted: 01/31/2020] [Indexed: 12/26/2022] Open
Abstract
Granulins (GRN) are secreted factors that promote neuronal survival and regulate inflammation in various pathological conditions. However, their roles in physiological conditions in the brain remain poorly understood. To address this knowledge gap, we analysed the telencephalon in Grn-deficient zebrafish and identified morphological and transcriptional changes in microglial cells, indicative of a pro-inflammatory phenotype in the absence of any insult. Unexpectedly, activated mutant microglia shared part of their transcriptional signature with aged human microglia. Furthermore, transcriptome profiles of the entire telencephali isolated from young Grn-deficient animals showed remarkable similarities with the profiles of the telencephali isolated from aged wildtype animals. Additionally, 50% of differentially regulated genes during aging were regulated in the telencephalon of young Grn-deficient animals compared to their wildtype littermates. Importantly, the telencephalon transcriptome in young Grn-deficent animals changed only mildly with aging, further suggesting premature aging of Grn-deficient brain. Indeed, Grn loss led to decreased neurogenesis and oligodendrogenesis, and to shortening of telomeres at young ages, to an extent comparable to that observed during aging. Altogether, our data demonstrate a role of Grn in regulating aging kinetics in the zebrafish telencephalon, thus providing a valuable tool for the development of new therapeutic approaches to treat age-associated pathologies.
Collapse
Affiliation(s)
- Alessandro Zambusi
- Institute of Stem Cell Research, Helmholtz Center Munich, 85764 Neuherberg, Germany
- Graduate School of Systemic Neuroscience; Biomedical Center, Faculty of Medicine, LMU Munich, 82152 Planegg, Germany
| | - Özge Pelin Burhan
- German Center for Neurodegenerative Diseases (DZNE), 81377 München, Germany; (Ö.P.B.); (B.S.)
| | - Rossella Di Giaimo
- Department of Biology, University of Naples Federico II, 80134 Naples, Italy;
| | - Bettina Schmid
- German Center for Neurodegenerative Diseases (DZNE), 81377 München, Germany; (Ö.P.B.); (B.S.)
| | - Jovica Ninkovic
- Institute of Stem Cell Research, Helmholtz Center Munich, 85764 Neuherberg, Germany
- Graduate School of Systemic Neuroscience; Biomedical Center, Faculty of Medicine, LMU Munich, 82152 Planegg, Germany
| |
Collapse
|
295
|
Dai Y, He F, Ji H, Zhao X, Misal S, Qi Z. Dual-Functional NIR AIEgens for High-Fidelity Imaging of Lysosomes in Cells and Photodynamic Therapy. ACS Sens 2020; 5:225-233. [PMID: 31854187 DOI: 10.1021/acssensors.9b02090] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Design and synthesis of water-soluble near-infrared (NIR) emissive fluorescent molecules with aggregation-induced emission (AIE) characteristics, perfect signal-to-noise ratio for imaging of organelle, and photodynamic therapy (PDT) functions has received enormous attention. However, the dual-functional NIR AIEgens of high-fidelity tracking lysosome and ablation cancer cells was rarely reported. Herein, a series of AIE luminogens (AIEgens) with a typical AIE effect, good biocompatibility, superior photostability, high brightness, and excellent reactive oxygen species (ROS) generation ability were developed, which had different electronic push-pull strength and conjugate system size in the molecular structure. These AIEgens could specifically "light up" and dynamically long-term track the lysosomes in living cells and zebrafish with ultrahigh colocalization imaging Pearson's correlation coefficients (Rr: 0.9687) and overlap coefficient (R: 0.9967). Additionally, the MPAT of NIR luminescence as a photosensitizer was used for photodynamic ablation of cancer cells, owing to prompt generation of the ROS under green light irradiation (495-530 nm, 10 mW cm-2). Hence, this research not only expands the application range of NIR AIEgens but also provides useful insights into design of split-new method for the treatment of cancer.
Collapse
Affiliation(s)
- Yanpeng Dai
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, School of Chemistry and Chemical Engineering, Southeast University, Nanjing, Jiangsu 211189, PR China
| | - Fangru He
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, School of Chemistry and Chemical Engineering, Southeast University, Nanjing, Jiangsu 211189, PR China
| | - Hefang Ji
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, School of Chemistry and Chemical Engineering, Southeast University, Nanjing, Jiangsu 211189, PR China
| | - Xinxin Zhao
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, School of Chemistry and Chemical Engineering, Southeast University, Nanjing, Jiangsu 211189, PR China
| | - Saima Misal
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, School of Chemistry and Chemical Engineering, Southeast University, Nanjing, Jiangsu 211189, PR China
| | - Zhengjian Qi
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, School of Chemistry and Chemical Engineering, Southeast University, Nanjing, Jiangsu 211189, PR China
| |
Collapse
|
296
|
Hidalgo San Jose L, Sunshine MJ, Dillingham CH, Chua BA, Kruta M, Hong Y, Hatters DM, Signer RAJ. Modest Declines in Proteome Quality Impair Hematopoietic Stem Cell Self-Renewal. Cell Rep 2020; 30:69-80.e6. [PMID: 31914399 PMCID: PMC7004491 DOI: 10.1016/j.celrep.2019.12.003] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 11/02/2019] [Accepted: 11/27/2019] [Indexed: 02/06/2023] Open
Abstract
Low protein synthesis is a feature of somatic stem cells that promotes regeneration in multiple tissues. Modest increases in protein synthesis impair stem cell function, but the mechanisms by which this occurs are largely unknown. We determine that low protein synthesis within hematopoietic stem cells (HSCs) is associated with elevated proteome quality in vivo. HSCs contain less misfolded and unfolded proteins than myeloid progenitors. Increases in protein synthesis cause HSCs to accumulate misfolded and unfolded proteins. To test how proteome quality affects HSCs, we examine Aarssti/sti mice that harbor a tRNA editing defect that increases amino acid misincorporation. Aarssti/sti mice exhibit reduced HSC numbers, increased proliferation, and diminished serial reconstituting activity. Misfolded proteins overwhelm the proteasome within Aarssti/sti HSCs, which is associated with increased c-Myc abundance. Deletion of one Myc allele partially rescues serial reconstitution defects in Aarssti/sti HSCs. Thus, HSCs are dependent on low protein synthesis to maintain proteostasis, which promotes their self-renewal.
Collapse
Affiliation(s)
- Lorena Hidalgo San Jose
- Division of Regenerative Medicine, Department of Medicine, Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Mary Jean Sunshine
- Division of Regenerative Medicine, Department of Medicine, Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Christopher H Dillingham
- Division of Regenerative Medicine, Department of Medicine, Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Bernadette A Chua
- Division of Regenerative Medicine, Department of Medicine, Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Miriama Kruta
- Division of Regenerative Medicine, Department of Medicine, Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Yuning Hong
- Department of Chemistry and Physics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3083, Australia
| | - Danny M Hatters
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Robert A J Signer
- Division of Regenerative Medicine, Department of Medicine, Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
297
|
Poiana G, Gioia R, Sineri S, Cardarelli S, Lupo G, Cacci E. Transcriptional regulation of adult neural stem/progenitor cells: tales from the subventricular zone. Neural Regen Res 2020; 15:1773-1783. [PMID: 32246617 PMCID: PMC7513981 DOI: 10.4103/1673-5374.280301] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
In rodents, well characterized neurogenic niches of the adult brain, such as the subventricular zone of the lateral ventricles and the subgranular zone of the hippocampus, support the maintenance of neural/stem progenitor cells (NSPCs) and the production of new neurons throughout the lifespan. The adult neurogenic process is dependent on the intrinsic gene expression signatures of NSPCs that make them competent for self-renewal and neuronal differentiation. At the same time, it is receptive to regulation by various extracellular signals that allow the modulation of neuronal production and integration into brain circuitries by various physiological stimuli. A drawback of this plasticity is the sensitivity of adult neurogenesis to alterations of the niche environment that can occur due to aging, injury or disease. At the core of the molecular mechanisms regulating neurogenesis, several transcription factors have been identified that maintain NSPC identity and mediate NSPC response to extrinsic cues. Here, we focus on REST, Egr1 and Dbx2 and their roles in adult neurogenesis, especially in the subventricular zone. We review recent work from our and other laboratories implicating these transcription factors in the control of NSPC proliferation and differentiation and in the response of NSPCs to extrinsic influences from the niche. We also discuss how their altered regulation may affect the neurogenic process in the aged and in the diseased brain. Finally, we highlight key open questions that need to be addressed to foster our understanding of the transcriptional mechanisms controlling adult neurogenesis.
Collapse
Affiliation(s)
- Giancarlo Poiana
- Department of Biology and Biotechnology "C. Darwin", Sapienza University of Rome, Rome, Italy
| | - Roberta Gioia
- Department of Biology and Biotechnology "C. Darwin", Sapienza University of Rome, Rome, Italy
| | - Serena Sineri
- Department of Biology and Biotechnology "C. Darwin", Sapienza University of Rome, Rome, Italy
| | - Silvia Cardarelli
- Department of Biology and Biotechnology "C. Darwin", Sapienza University of Rome, Rome, Italy
| | - Giuseppe Lupo
- Department of Biology and Biotechnology "C. Darwin", Sapienza University of Rome, Rome, Italy
| | - Emanuele Cacci
- Department of Biology and Biotechnology "C. Darwin", Sapienza University of Rome, Rome, Italy
| |
Collapse
|
298
|
Bacigaluppi M, Sferruzza G, Butti E, Ottoboni L, Martino G. Endogenous neural precursor cells in health and disease. Brain Res 2019; 1730:146619. [PMID: 31874148 DOI: 10.1016/j.brainres.2019.146619] [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: 08/23/2019] [Revised: 11/25/2019] [Accepted: 12/19/2019] [Indexed: 12/15/2022]
Abstract
Neurogenesis persists in the adult brain of mammals in the subventricular zone (SVZ) of the lateral ventricles and in the subgranular zone (SGZ) of the dentate gyrus (DG). The complex interactions between intrinsic and extrinsic signals provided by cells in the niche but also from distant sources regulate the fate of neural stem/progenitor cells (NPCs) in these sites. This fine regulation is perturbed in aging and in pathological conditions leading to a different NPC behavior, tailored to the specific physio-pathological features. Indeed, NPCs exert in physiological and pathological conditions important neurogenic and non-neurogenic regulatory functions and participate in maintaining and protecting brain tissue homeostasis. In this review, we discuss intrinsic and extrinsic signals that regulate NPC activation and NPC functional role in various homeostatic and non-homeostatic conditions.
Collapse
Affiliation(s)
- Marco Bacigaluppi
- Neuroimmunology Unit and Department of Neurology, Institute of Experimental Neurology, San Raffaele Hospital and Università Vita- Salute San Raffaele, Via Olgettina 60, 20132 Milano, Italy.
| | - Giacomo Sferruzza
- Neuroimmunology Unit and Department of Neurology, Institute of Experimental Neurology, San Raffaele Hospital and Università Vita- Salute San Raffaele, Via Olgettina 60, 20132 Milano, Italy
| | - Erica Butti
- Neuroimmunology Unit and Department of Neurology, Institute of Experimental Neurology, San Raffaele Hospital and Università Vita- Salute San Raffaele, Via Olgettina 60, 20132 Milano, Italy
| | - Linda Ottoboni
- Neuroimmunology Unit and Department of Neurology, Institute of Experimental Neurology, San Raffaele Hospital and Università Vita- Salute San Raffaele, Via Olgettina 60, 20132 Milano, Italy
| | - Gianvito Martino
- Neuroimmunology Unit and Department of Neurology, Institute of Experimental Neurology, San Raffaele Hospital and Università Vita- Salute San Raffaele, Via Olgettina 60, 20132 Milano, Italy
| |
Collapse
|
299
|
Abstract
The molecular basis for the neural stem cell quiescence-to-activation transition has become an important focus in the study of adult neurogenesis. Recently in Cell, Kalamakis et al. (2019) show that aged neural stem cells face greater barriers to exiting quiescence, imposed by the niche through inflammation and altered Wnt signaling.
Collapse
|
300
|
Quiescence of Adult Mammalian Neural Stem Cells: A Highly Regulated Rest. Neuron 2019; 104:834-848. [DOI: 10.1016/j.neuron.2019.09.026] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 09/05/2019] [Accepted: 09/17/2019] [Indexed: 12/14/2022]
|