1
|
Wu Z, Qu J, Liu GH. Roles of chromatin and genome instability in cellular senescence and their relevance to ageing and related diseases. Nat Rev Mol Cell Biol 2024; 25:979-1000. [PMID: 39363000 DOI: 10.1038/s41580-024-00775-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/12/2024] [Indexed: 10/05/2024]
Abstract
Ageing is a complex biological process in which a gradual decline in physiological fitness increases susceptibility to diseases such as neurodegenerative disorders and cancer. Cellular senescence, a state of irreversible cell-growth arrest accompanied by functional deterioration, has emerged as a pivotal driver of ageing. In this Review, we discuss how heterochromatin loss, telomere attrition and DNA damage contribute to cellular senescence, ageing and age-related diseases by eliciting genome instability, innate immunity and inflammation. We also discuss how emerging therapeutic strategies could restore heterochromatin stability, maintain telomere integrity and boost the DNA repair capacity, and thus counteract cellular senescence and ageing-associated pathologies. Finally, we outline current research challenges and future directions aimed at better comprehending and delaying ageing.
Collapse
Affiliation(s)
- Zeming Wu
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Jing Qu
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
- Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
- Aging Biomarker Consortium, Beijing, China.
| | - Guang-Hui Liu
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
- Aging Biomarker Consortium, Beijing, China.
- Aging Translational Medicine Center, International Center for Aging and Cancer, Xuanwu Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
2
|
Harley J, Santosa MM, Ng CY, Grinchuk OV, Hor JH, Liang Y, Lim VJ, Tee WW, Ong DST, Ng SY. Telomere shortening induces aging-associated phenotypes in hiPSC-derived neurons and astrocytes. Biogerontology 2024; 25:341-360. [PMID: 37987889 PMCID: PMC10998800 DOI: 10.1007/s10522-023-10076-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 10/13/2023] [Indexed: 11/22/2023]
Abstract
Telomere shortening is a well-established hallmark of cellular aging. Telomerase reverse transcriptase (TERT) plays a crucial role in maintaining the length of telomeres, which are specialised protective caps at the end of chromosomes. The lack of in vitro aging models, particularly for the central nervous system (CNS), has impeded progress in understanding aging and age-associated neurodegenerative diseases. In this study, we aimed to explore the possibility of inducing aging-associated features in cell types of the CNS using hiPSC (human induced pluripotent stem cell) technology. To achieve this, we utilised CRISPR/Cas9 to generate hiPSCs with a loss of telomerase function and shortened telomeres. Through directed differentiation, we generated motor neurons and astrocytes to investigate whether telomere shortening could lead to age-associated phenotypes. Our findings revealed that shortened telomeres induced age-associated characteristics in both motor neurons and astrocytes including increased cellular senescence, heightened inflammation, and elevated DNA damage. We also observed cell-type specific age-related morphology changes. Additionally, our study highlighted the fundamental role of TERT and telomere shortening in neural progenitor cell (NPC) proliferation and neuronal differentiation. This study serves as a proof of concept that telomere shortening can effectively induce aging-associated phenotypes, thereby providing a valuable tool to investigate age-related decline and neurodegenerative diseases.
Collapse
Affiliation(s)
- Jasmine Harley
- Institute of Molecular and Cell Biology, A*STAR Research Entities, Singapore, 138673, Singapore
| | - Munirah Mohamad Santosa
- Institute of Molecular and Cell Biology, A*STAR Research Entities, Singapore, 138673, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117593, Singapore
| | - Chong Yi Ng
- Institute of Molecular and Cell Biology, A*STAR Research Entities, Singapore, 138673, Singapore
| | - Oleg V Grinchuk
- Institute of Molecular and Cell Biology, A*STAR Research Entities, Singapore, 138673, Singapore
| | - Jin-Hui Hor
- Institute of Molecular and Cell Biology, A*STAR Research Entities, Singapore, 138673, Singapore
| | - Yajing Liang
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117593, Singapore
| | - Valerie Jingwen Lim
- Institute of Molecular and Cell Biology, A*STAR Research Entities, Singapore, 138673, Singapore
| | - Wee Wei Tee
- Institute of Molecular and Cell Biology, A*STAR Research Entities, Singapore, 138673, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117593, Singapore
| | - Derrick Sek Tong Ong
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117593, Singapore
| | - Shi-Yan Ng
- Institute of Molecular and Cell Biology, A*STAR Research Entities, Singapore, 138673, Singapore.
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117593, Singapore.
| |
Collapse
|
3
|
Zhong Y, Wang G, Yang S, Zhang Y, Wang X. The role of DNA damage in neural stem cells ageing. J Cell Physiol 2024; 239:e31187. [PMID: 38219047 DOI: 10.1002/jcp.31187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 11/17/2023] [Accepted: 12/20/2023] [Indexed: 01/15/2024]
Abstract
Neural stem cells (NSCs) are pluripotent stem cells with the potential to differentiate into a variety of nerve cells. NSCs are susceptible to both intracellular and extracellular insults, thus causing DNA damage. Extracellular insults include ultraviolet, ionizing radiation, base analogs, modifiers, alkyl agents and others, while intracellular factors include Reactive oxygen species (ROS) radicals produced by mitochondria, mismatches that occur during DNA replication, deamination of bases, loss of bases, and more. When encountered with DNA damage, cells typically employ three coping strategies: DNA repair, damage tolerance, and apoptosis. NSCs, like many other stem cells, have the ability to divide, differentiate, and repair DNA damage to prevent mutations from being passed down to the next generation. However, when DNA damage accumulates over time, it will lead to a series of alterations in the metabolism of cells, which will cause cellular ageing. The ageing and exhaustion of neural stem cell will have serious effects on the body, such as neurodegenerative diseases. The purpose of this review is to examine the processes by which DNA damage leads to NSCs ageing and the mechanisms of DNA repair in NSCs.
Collapse
Affiliation(s)
- Yiming Zhong
- School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Guangming Wang
- School of Medicine, Postdoctoral Station of Clinical Medicine, Shanghai Tongji Hospital, Tongji University, Shanghai, China
| | - Shangzhi Yang
- School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yang Zhang
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Xianli Wang
- School of Public Health, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
4
|
Carver AJ, Hing B, Elser BA, Lussier SJ, Yamanashi T, Howard MA, Kawasaki H, Shinozaki G, Stevens HE. Correlation of telomere length in brain tissue with peripheral tissues in living human subjects. Front Mol Neurosci 2024; 17:1303974. [PMID: 38516039 PMCID: PMC10954899 DOI: 10.3389/fnmol.2024.1303974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 02/26/2024] [Indexed: 03/23/2024] Open
Abstract
Telomeres are important to chromosomal stability, and changes in their length correlate with disease, potentially relevant to brain disorders. Assessing telomere length in human brain is invasive, but whether peripheral tissue telomere length correlates with that in brain is not known. Saliva, buccal, blood, and brain samples were collected at time points before, during, and after subjects undergoing neurosurgery (n = 35) for intractable epilepsy. DNA was isolated from samples and average telomere length assessed by qPCR. Correlations of telomere length between tissue samples were calculated across subjects. When data were stratified by sex, saliva telomere length correlated with brain telomere length in males only. Buccal telomere length correlated with brain telomere length when males and females were combined. These findings indicate that in living subjects, telomere length in peripheral tissues variably correlates with that in brain and may be dependent on sex. Peripheral tissue telomere length may provide insight into brain telomere length, relevant to assessment of brain disorder pathophysiology.
Collapse
Affiliation(s)
- Annemarie J. Carver
- Department of Psychiatry, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
- Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa City, IA, United States
| | - Benjamin Hing
- Department of Psychiatry, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Benjamin A. Elser
- Department of Psychiatry, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
- Interdisciplinary Graduate Program in Human Toxicology, University of Iowa, Iowa City, IA, United States
| | - Stephanie J. Lussier
- Department of Psychiatry, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
- Biostatistics Graduate Program, College of Public Health, University of Iowa, Iowa City, IA, United States
| | - Takehiko Yamanashi
- Department of Psychiatry, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Standford University, Stanford, CA, United States
- Division of Neuropsychiatry, Tottori University, Tottori, Japan
| | - Matthew A. Howard
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
- Department of Neurosurgery, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Hiroto Kawasaki
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
- Department of Neurosurgery, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Gen Shinozaki
- Department of Psychiatry, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Standford University, Stanford, CA, United States
| | - Hanna E. Stevens
- Department of Psychiatry, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
- Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa City, IA, United States
- Interdisciplinary Graduate Program in Human Toxicology, University of Iowa, Iowa City, IA, United States
- Hawk-Intellectual and Developmental Disabilities Research Center, University of Iowa, Iowa City, IA, United States
| |
Collapse
|
5
|
Liu M, Fan Y, Ni N, Yu T, Mao Z, Huang H, Zhang J, Tang Y, He H, Meng F, You Y, Zhou Q. TERT mediates the U-shape of glucocorticoids effects in modulation of hippocampal neural stem cells and associated brain function. CNS Neurosci Ther 2024; 30:e14577. [PMID: 38421107 PMCID: PMC10850922 DOI: 10.1111/cns.14577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 10/17/2023] [Accepted: 12/07/2023] [Indexed: 03/02/2024] Open
Abstract
BACKGROUND Glucocorticoids (GCs) are steroidal hormones produced by the adrenal cortex. A physiological-level GCs have a crucial function in maintaining many cognitive processes, like cognition, memory, and mood, however, both insufficient and excessive GCs impair these functions. Although this phenomenon could be explained by the U-shape of GC effects, the underlying mechanisms are still not clear. Therefore, understanding the underlying mechanisms of GCs may provide insight into the treatments for cognitive and mood-related disorders. METHODS Consecutive administration of corticosterone (CORT, 10 mg/kg, i.g.) proceeded for 28 days to mimic excessive GCs condition. Adrenalectomy (ADX) surgery was performed to ablate endogenous GCs in mice. Microinjection of 1 μL of Ad-mTERT-GFP virus into mouse hippocampus dentate gyrus (DG) and behavioral alterations in mice were observed 4 weeks later. RESULTS Different concentrations of GCs were shown to affect the cell growth and development of neural stem cells (NSCs) in a U-shaped manner. The physiological level of GCs (0.01 μM) promoted NSC proliferation in vitro, while the stress level of GCs (10 μM) inhibited it. The glucocorticoid synthesis blocker metyrapone (100 mg/kg, i.p.) and ADX surgery both decreased the quantity and morphological development of doublecortin (DCX)-positive immature cells in the DG. The physiological level of GCs activated mineralocorticoid receptor and then promoted the production of telomerase reverse transcriptase (TERT); in contrast, the stress level of GCs activated glucocorticoid receptor and then reduced the expression of TERT. Overexpression of TERT by AD-mTERT-GFP reversed both chronic stresses- and ADX-induced deficiency of TERT and the proliferation and development of NSCs, chronic stresses-associated depressive symptoms, and ADX-associated learning and memory impairment. CONCLUSION The bidirectional regulation of TERT by different GCs concentrations is a key mechanism mediating the U-shape of GC effects in modulation of hippocampal NSCs and associated brain function. Replenishment of TERT could be a common treatment strategy for GC dysfunction-associated diseases.
Collapse
Affiliation(s)
- Meng‐Ying Liu
- Department of Pharmacy, Nanjing Drum Tower HospitalAffiliated Hospital of Medical School, Nanjing UniversityNanjingChina
- State Key Laboratory of Reproductive Medicine, Department of Clinical Pharmacology, School of PharmacyNanjing Medical UniversityNanjingChina
| | - Yixin Fan
- State Key Laboratory of Reproductive Medicine, Department of Clinical Pharmacology, School of PharmacyNanjing Medical UniversityNanjingChina
- Department of Pharmacy, Sir Run Run HospitalNanjing Medical UniversityNanjingChina
| | - Ningjie Ni
- State Key Laboratory of Reproductive Medicine, Department of Clinical Pharmacology, School of PharmacyNanjing Medical UniversityNanjingChina
- Department of NeurosurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Tao Yu
- School of PharmacyNanjing Medical UniversityNanjingChina
| | - Zhiyuan Mao
- Key Laboratory for Aging & Disease, The State Key Laboratory of Reproductive Medicine, Department of Human Anatomy, Research Centre for Bone and Stem CellsNanjing Medical UniversityNanjingChina
| | - Hanyu Huang
- State Key Laboratory of Reproductive Medicine, Department of Clinical Pharmacology, School of PharmacyNanjing Medical UniversityNanjingChina
- Department of NeurosurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Jing Zhang
- Department of Clinical Pharmacology, School of PharmacyNanjing Medical UniversityNanjingChina
| | - Yulin Tang
- Department of Pharmacy, Sir Run Run HospitalNanjing Medical UniversityNanjingChina
| | - Hongliang He
- Department of Pharmacy, Sir Run Run HospitalNanjing Medical UniversityNanjingChina
| | - Fan Meng
- Department of Clinical Pharmacology, School of PharmacyNanjing Medical UniversityNanjingChina
| | - Yongping You
- Department of NeurosurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Qi‐Gang Zhou
- State Key Laboratory of Reproductive Medicine, Department of Clinical Pharmacology, School of PharmacyNanjing Medical UniversityNanjingChina
- Department of Pharmacy, Sir Run Run HospitalNanjing Medical UniversityNanjingChina
- Department of Clinical Pharmacology, School of PharmacyNanjing Medical UniversityNanjingChina
| |
Collapse
|
6
|
Cai C, Li H, Tian Z, Liang Q, Shen R, Wu Z, Liu B, Yang Y. HGF secreted by hUC-MSCs mitigates neuronal apoptosis to repair the injured spinal cord via phosphorylation of Akt/FoxO3a pathway. Biochem Biophys Res Commun 2024; 692:149321. [PMID: 38056156 DOI: 10.1016/j.bbrc.2023.149321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 11/16/2023] [Accepted: 11/22/2023] [Indexed: 12/08/2023]
Abstract
Spinal cord injury (SCI) can cause severe and permanent neurological damage, and neuronal apoptosis could inhibit functional recovery of damaged spinal cord greatly. Human umbilical cord mesenchymal stem cells (hUC-MSCs) have great potential to repair SCI because of a series of advantages, including inhibition of neuronal apoptosis and multiple differentiation. The former may play an important role. However, the detailed regulatory mechanism associated with the inhibition of neuronal apoptosis after hUC-MSCs administration has not been elucidated. In this study, proteomics analysis of precious human cerebrospinal fluid (CSF) samples collected from SCI subjects receiving hUC-MSCs delivery indicated that hepatocyte growth factor (HGF) is largely involved in SCI repair. Furthermore, overexpression of HGF derived from hUC-MSCs could decrease reactive oxygen species to prevent neuron apoptosis to the maximum, and thus lead to significant recovery of spinal cord dysfunction. Moreover, HGF could promote phosphorylation of Akt/FoxO3a pathway to decrease reactive oxygen species to reduce neuron apoptosis. For the first time, our research revealed that HGF secreted by hUC-MSCs inhibits neuron apoptosis by phosphorylation of Akt/FoxO3a to repair SCI. This study provides important clues associated with drug selection for the effective treatment of SCI in humans.
Collapse
Affiliation(s)
- Chaoyang Cai
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-sen University, No. 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, China; National Medical Products Administration (NMPA), Key Laboratory for Quality Research and Evaluation of Cell Products, No. 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, China; Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, No. 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, China; Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, No. 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, China
| | - Hong Li
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-sen University, No. 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, China; National Medical Products Administration (NMPA), Key Laboratory for Quality Research and Evaluation of Cell Products, No. 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, China; Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, No. 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, China; Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, No. 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, China
| | - Zhenming Tian
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-sen University, No. 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, China; National Medical Products Administration (NMPA), Key Laboratory for Quality Research and Evaluation of Cell Products, No. 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, China; Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, No. 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, China; Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, No. 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, China
| | - Qian Liang
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-sen University, No. 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, China; National Medical Products Administration (NMPA), Key Laboratory for Quality Research and Evaluation of Cell Products, No. 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, China; Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, No. 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, China; Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, No. 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, China
| | - Ruoqi Shen
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-sen University, No. 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, China; National Medical Products Administration (NMPA), Key Laboratory for Quality Research and Evaluation of Cell Products, No. 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, China; Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, No. 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, China; Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, No. 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, China
| | - Zizhao Wu
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-sen University, No. 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, China; National Medical Products Administration (NMPA), Key Laboratory for Quality Research and Evaluation of Cell Products, No. 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, China; Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, No. 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, China; Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, No. 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, China.
| | - Bin Liu
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-sen University, No. 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, China; National Medical Products Administration (NMPA), Key Laboratory for Quality Research and Evaluation of Cell Products, No. 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, China; Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, No. 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, China; Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, No. 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, China.
| | - Yang Yang
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-sen University, No. 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, China; National Medical Products Administration (NMPA), Key Laboratory for Quality Research and Evaluation of Cell Products, No. 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, China; Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, No. 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, China; Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, No. 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, China.
| |
Collapse
|
7
|
Shi J, Zhang X, Chen J, Shen R, Cui H, Wu H. Acupuncture and moxibustion therapy for cognitive impairment: the microbiome-gut-brain axis and its role. Front Neurosci 2024; 17:1275860. [PMID: 38274501 PMCID: PMC10808604 DOI: 10.3389/fnins.2023.1275860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 12/18/2023] [Indexed: 01/27/2024] Open
Abstract
Cognitive impairment poses a significant burden on individuals, families, and society worldwide. Despite the lack of effective treatment strategies, emerging evidence suggests that the microbiome-gut-brain (MGB) axis may play a critical role in the pathogenesis of cognitive impairment. While targeted treatment is not yet comprehensive, recently, acupuncture and moxibustion therapy has participated increasingly in the treatment of degenerative diseases and has achieved a certain therapeutic effect. In this review, the possible mechanisms by which acupuncture and moxibustion therapy may improve cognitive impairment through the MGB axis are reviewed, including regulating gut microbial homeostasis, improving intestinal inflammation mediated by the neuroendocrine-immune system, and enhancing intestinal barrier function. We also discuss common acupoints and corresponding mechanism analysis to provide insights into further exploration of mechanisms that target the MGB axis and thereby intervene in cognitive impairment.
Collapse
Affiliation(s)
- Jiatian Shi
- Department of Acupuncture and Moxibustion, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xinyue Zhang
- Department of Acupuncture and Moxibustion, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jianhua Chen
- Department of Mental Health, Shanghai Mental Health Center, Shanghai, China
| | - Ruishi Shen
- Department of Acupuncture and Moxibustion, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Huashun Cui
- Department of Acupuncture and Moxibustion, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Huangan Wu
- Department of Acupuncture and Moxibustion, Yueyang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
8
|
Gurvich C, Thomas N, Hudaib AR, Van Rheenen TE, Thomas EHX, Tan EJ, Neill E, Carruthers SP, Sumner PJ, Romano-Silva M, Bozaoglu K, Kulkarni J, Rossell SL. The relationship between cognitive clusters and telomere length in bipolar-schizophrenia spectrum disorders. Psychol Med 2023; 53:5119-5126. [PMID: 35920237 DOI: 10.1017/s0033291722002148] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND Schizophrenia and bipolar disorder are complex mental illnesses that are associated with cognitive deficits. There is considerable cognitive heterogeneity that exists within both disorders. Studies that cluster schizophrenia and bipolar patients into subgroups based on their cognitive profile increasingly demonstrate that, relative to healthy controls, there is a severely compromised subgroup and a relatively intact subgroup. There is emerging evidence that telomere shortening, a marker of cellular senescence, may be associated with cognitive impairments. The aim of this study was to explore the relationship between cognitive subgroups in bipolar-schizophrenia spectrum disorders and telomere length against a healthy control sample. METHODS Participants included a transdiagnostic group diagnosed with bipolar, schizophrenia or schizoaffective disorder (n = 73) and healthy controls (n = 113). Cognitive clusters within the transdiagnostic patient group, were determined using K-means cluster analysis based on current cognitive functioning (MATRICS Consensus Cognitive Battery scores). Telomere length was determined using quantitative PCRs genomic DNA extracted from whole blood. Emergent clusters were then compared to the healthy control group on telomere length. RESULTS Two clusters emerged within the patient group that were deemed to reflect a relatively intact cognitive group and a cognitively impaired subgroup. Telomere length was significantly shorter in the severely impaired cognitive subgroup compared to the healthy control group. CONCLUSIONS This study replicates previous findings of transdiagnostic cognitive subgroups and associates shorter telomere length with the severely impaired cognitive subgroup. These findings support emerging literature associating cognitive impairments in psychiatric disorders to accelerated cellular aging as indexed by telomere length.
Collapse
Affiliation(s)
- Caroline Gurvich
- Department of Psychiatry, Central Clinical School, Monash University and the Alfred Hospital, Melbourne, VIC, Australia
| | - Natalie Thomas
- Department of Biochemistry & Pharmacology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne VIC, Australia
| | - Abdul-Rahman Hudaib
- Department of Psychiatry, Central Clinical School, Monash University and the Alfred Hospital, Melbourne, VIC, Australia
| | - Tamsyn E Van Rheenen
- Melbourne Neuropsychiatry Centre, University of Melbourne, Melbourne, VIC, Australia
- Centre for Mental Health, Faculty of Health, Arts and Design, Swinburne University of Technology, Melbourne, VIC, Australia
| | - Elizabeth H X Thomas
- Department of Psychiatry, Central Clinical School, Monash University and the Alfred Hospital, Melbourne, VIC, Australia
| | - Eric J Tan
- Centre for Mental Health, Faculty of Health, Arts and Design, Swinburne University of Technology, Melbourne, VIC, Australia
- Department of Mental Health, St Vincent's Hospital, Melbourne, VIC, Australia
| | - Erica Neill
- Centre for Mental Health, Faculty of Health, Arts and Design, Swinburne University of Technology, Melbourne, VIC, Australia
- Department of Mental Health, St Vincent's Hospital, Melbourne, VIC, Australia
| | - Sean P Carruthers
- Centre for Mental Health, Faculty of Health, Arts and Design, Swinburne University of Technology, Melbourne, VIC, Australia
| | - Philip J Sumner
- Centre for Mental Health, Faculty of Health, Arts and Design, Swinburne University of Technology, Melbourne, VIC, Australia
| | - Marco Romano-Silva
- Department Saude Mental, Faculdade de Medicina, UFMG, Belo Horizonte, Brazil
| | - Kiymet Bozaoglu
- Bruce Lefroy Centre for Genetic Health Research, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
- Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia
| | - Jayashri Kulkarni
- Department of Psychiatry, Central Clinical School, Monash University and the Alfred Hospital, Melbourne, VIC, Australia
| | - Susan L Rossell
- Centre for Mental Health, Faculty of Health, Arts and Design, Swinburne University of Technology, Melbourne, VIC, Australia
- Department of Mental Health, St Vincent's Hospital, Melbourne, VIC, Australia
| |
Collapse
|
9
|
Ren Q, Zhang G, Dong C, Li Z, Zhou D, Huang L, Li W, Huang G, Yan J. Parental Folate Deficiency Inhibits Proliferation and Increases Apoptosis of Neural Stem Cells in Rat Offspring: Aggravating Telomere Attrition as a Potential Mechanism. Nutrients 2023; 15:2843. [PMID: 37447170 DOI: 10.3390/nu15132843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 06/17/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
The effect of maternal folate status on the fetal central nervous system (CNS) is well recognized, while evidence is emerging that such an association also exists between fathers and offspring. The biological functions of telomeres and telomerase are also related to neural cell proliferation and apoptosis. The study aimed to investigate the effect of parental folate deficiency on the proliferation and apoptosis of neural stem cells (NSCs) in neonatal offspring and the role of telomeres in this effect. In this study, rats were divided into four groups: maternal folate-deficient and paternal folate-deficient diet (D-D) group; maternal folate-deficient and paternal folate-normal diet (D-N) group; maternal folate-normal and paternal folate-deficient diet (N-D) group; and the maternal folate-normal and paternal folate-normal diet (N-N) group. The offspring were sacrificed at postnatal day 0 (PND0), and NSCs were cultured from the hippocampus and striatum tissues of offspring for future assay. The results revealed that parental folate deficiency decreased folate levels, increased homocysteine (Hcy) levels of the offspring's brain tissue, inhibited proliferation, increased apoptosis, shortened telomere length, and aggravated telomere attrition of offspring NSCs in vivo and in vitro. In vitro experiments further showed that offspring NSCs telomerase activity was inhibited due to parental folate deficiency. In conclusion, parental folate deficiency inhibited the proliferation and increased apoptosis of offspring NSCs, maternal folate deficiency had more adverse effects than paternal, and the mechanisms may involve the telomere attrition of NSCs.
Collapse
Affiliation(s)
- Qinghan Ren
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin 300070, China
| | - Guoquan Zhang
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin 300070, China
| | - Cuixia Dong
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin 300070, China
| | - Zhenshu Li
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin 300070, China
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin 300070, China
| | - Dezheng Zhou
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin 300070, China
| | - Li Huang
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin 300070, China
| | - Wen Li
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin 300070, China
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin 300070, China
| | - Guowei Huang
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin 300070, China
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin 300070, China
| | - Jing Yan
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin 300070, China
- Department of Social Medicine and Health Administration, School of Public Health, Tianjin Medical University, Tianjin 300070, China
| |
Collapse
|
10
|
Rahman Z, Dandekar MP. Implication of Paraprobiotics in Age-Associated Gut Dysbiosis and Neurodegenerative Diseases. Neuromolecular Med 2023; 25:14-26. [PMID: 35879588 DOI: 10.1007/s12017-022-08722-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 07/13/2022] [Indexed: 10/16/2022]
Abstract
Neurodegenerative diseases, including Alzheimer's and Parkinson's disease, are major age-related concerns in elderly people. Since no drug fully addresses the progression of neurodegenerative diseases, advance treatment strategies are urgently needed. Several studies have noted the senescence of immune system and the perturbation of gut microbiota in the aged population. In recent years, the role of gut microbiota has been increasingly studied in the manifestation of age-related CNS disorders. In this context, prebiotics, probiotics, and paraprobiotics are reported to improve the behavioural and neurobiological abnormalities in elderly patients. As live microbiota, prescribed in the form of probiotics, shows some adverse effects like sepsis, translocation, and horizontal gene transfer, paraprobiotics could be a possible alternative strategy in designing microbiome-based therapeutics. This review describes the health-beneficial effects of paraprobiotics in age-associated neurodegenerative diseases.
Collapse
Affiliation(s)
- Ziaur Rahman
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, 500037, India
| | - Manoj P Dandekar
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, 500037, India.
| |
Collapse
|
11
|
Wang S, Zhao X, Yu Y, Tao F, Liu D, Sun Y. Prolonged Rather Than Early Childhood Parent-Child Separation Predicts Change in Molecular Markers of Cellular Aging: A Consideration of the Role of Adolescence. J Youth Adolesc 2023; 52:165-176. [PMID: 36149547 DOI: 10.1007/s10964-022-01681-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 09/09/2022] [Indexed: 01/07/2023]
Abstract
Early life adversity is a major risk factor for the onset of psychopathology, and cellular aging may be a mechanism underlying these associations. It is unknown whether and how the pattern (timing and duration) of parent-child separation is associated with accelerated cellular aging, particularly with respect to functional aging and replicative senescence, indexed by mitochondrial DNA copy number (mtDNAcn) elevation and telomere length (TL) attrition. This cohort study included 252 rural adolescents (mean age 11.62 years, SD: 1.56). Nearly one in five participants were persistently separated from both parents since birth. Compared with participants who never separated from their parents, adolescents with prolonged parent-child separation had higher acceleration both in functional aging and replicative senescence of cells. However, that was not the case in adolescents who experienced parent-child separation in early childhood but regained stable parental care during adolescence. These findings indicate that pubertal development reopens a window of opportunity for buffering the adverse biological effect based on significant improvements in the supportiveness of the caregiving environment relative to that in childhood. Translating such knowledge to inform intervention and prevention strategies for youths exposed to adversity is a critical goal for the field.
Collapse
Affiliation(s)
- Shihong Wang
- Department of Maternal, Child & Adolescent Health, School of Public Health, Anhui Medical University, Hefei, Anhui Province, China
| | - Xudong Zhao
- Department of Maternal, Child & Adolescent Health, School of Public Health, Anhui Medical University, Hefei, Anhui Province, China
| | - Yue Yu
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| | - Fangbiao Tao
- Department of Maternal, Child & Adolescent Health, School of Public Health, Anhui Medical University, Hefei, Anhui Province, China.,Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, 230032, Anhui, China.,Anhui Provincial Key Laboratory of Population Health & Aristogenics, Hefei, Anhui Province, China
| | - Deyun Liu
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China.
| | - Ying Sun
- Department of Maternal, Child & Adolescent Health, School of Public Health, Anhui Medical University, Hefei, Anhui Province, China. .,Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, 230032, Anhui, China. .,Anhui Provincial Key Laboratory of Population Health & Aristogenics, Hefei, Anhui Province, China.
| |
Collapse
|
12
|
Becker J, Sun B, Alammari F, Haerty W, Vance KW, Szele FG. What has single-cell transcriptomics taught us about long non-coding RNAs in the ventricular-subventricular zone? Stem Cell Reports 2022; 18:354-376. [PMID: 36525965 PMCID: PMC9860170 DOI: 10.1016/j.stemcr.2022.11.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 11/14/2022] [Accepted: 11/14/2022] [Indexed: 12/16/2022] Open
Abstract
Long non-coding RNA (lncRNA) function is mediated by the process of transcription or through transcript-dependent associations with proteins or nucleic acids to control gene regulatory networks. Many lncRNAs are transcribed in the ventricular-subventricular zone (V-SVZ), a postnatal neural stem cell niche. lncRNAs in the V-SVZ are implicated in neurodevelopmental disorders, cancer, and brain disease, but their functions are poorly understood. V-SVZ neurogenesis capacity declines with age due to stem cell depletion and resistance to neural stem cell activation. Here we analyzed V-SVZ transcriptomics by pooling current single-cell RNA-seq data. They showed consistent lncRNA expression during stem cell activation, lineage progression, and aging. In conjunction with epigenetic and genetic data, we predicted V-SVZ lncRNAs that regulate stem cell activation and differentiation. Some of the lncRNAs validate known epigenetic mechanisms, but most remain uninvestigated. Our analysis points to several lncRNAs that likely participate in key aspects of V-SVZ stem cell activation and neurogenesis in health and disease.
Collapse
Affiliation(s)
- Jemima Becker
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Bin Sun
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Farah Alammari
- Department of Blood and Cancer Research, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia,Clinical Laboratory Sciences Department, College of Applied Medical Sciences, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | | | - Keith W. Vance
- Department of Life Sciences, University of Bath, Bath, UK
| | - Francis George Szele
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK.
| |
Collapse
|
13
|
Ortiz-Álvarez G, Fortoul A, Srivastava A, Moreau MX, Bouloudi B, Mailhes-Hamon C, Delgehyr N, Faucourt M, Bahin M, Blugeon C, Breau M, Géli V, Causeret F, Meunier A, Spassky N. p53/p21 pathway activation contributes to the ependymal fate decision downstream of GemC1. Cell Rep 2022; 41:111810. [PMID: 36516767 DOI: 10.1016/j.celrep.2022.111810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 07/08/2022] [Accepted: 11/18/2022] [Indexed: 12/14/2022] Open
Abstract
Multiciliated ependymal cells and adult neural stem cells are components of the adult neurogenic niche, essential for brain homeostasis. These cells share a common glial cell lineage regulated by the Geminin family members Geminin and GemC1/Mcidas. Ependymal precursors require GemC1/Mcidas expression to massively amplify centrioles and become multiciliated cells. Here, we show that GemC1-dependent differentiation is initiated in actively cycling radial glial cells, in which a DNA damage response, including DNA replication-associated damage and dysfunctional telomeres, is induced, without affecting cell survival. Genotoxic stress is not sufficient by itself to induce ependymal cell differentiation, although the absence of p53 or p21 in progenitors hinders differentiation by maintaining cell division. Activation of the p53-p21 pathway downstream of GemC1 leads to cell-cycle slowdown/arrest, which permits timely onset of ependymal cell differentiation in progenitor cells.
Collapse
Affiliation(s)
- Gonzalo Ortiz-Álvarez
- Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Département de Biologie, Ecole Normale Supérieure, CNRS UMR8197, INSERM U1024, Université PSL, 75005 Paris, France
| | - Aurélien Fortoul
- Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Département de Biologie, Ecole Normale Supérieure, CNRS UMR8197, INSERM U1024, Université PSL, 75005 Paris, France
| | - Ayush Srivastava
- Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Département de Biologie, Ecole Normale Supérieure, CNRS UMR8197, INSERM U1024, Université PSL, 75005 Paris, France
| | - Matthieu X Moreau
- Université de Paris, Imagine Institute, Team Genetics and Development of the Cerebral Cortex, 75015 Paris, France; Université de Paris, Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, 75014 Paris, France
| | - Benoît Bouloudi
- Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Département de Biologie, Ecole Normale Supérieure, CNRS UMR8197, INSERM U1024, Université PSL, 75005 Paris, France
| | - Caroline Mailhes-Hamon
- Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Département de Biologie, Ecole Normale Supérieure, CNRS UMR8197, INSERM U1024, Université PSL, 75005 Paris, France
| | - Nathalie Delgehyr
- Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Département de Biologie, Ecole Normale Supérieure, CNRS UMR8197, INSERM U1024, Université PSL, 75005 Paris, France
| | - Marion Faucourt
- Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Département de Biologie, Ecole Normale Supérieure, CNRS UMR8197, INSERM U1024, Université PSL, 75005 Paris, France
| | - Mathieu Bahin
- Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Département de Biologie, Ecole Normale Supérieure, CNRS UMR8197, INSERM U1024, Université PSL, 75005 Paris, France
| | - Corinne Blugeon
- Genomics Core Facility, Institut de Biologie de l'ENS (IBENS), Département de Biologie, Ecole Normale Supérieure, CNRS UMR8197, INSERM U1024, Université PSL, 75005 Paris, France
| | - Marielle Breau
- Marseille Cancer Research Centre (CRCM), U1068 INSERM, UMR7258 CNRS, UM105 Aix-Marseille University, Institut Paoli-Calmettes, Ligue Nationale Contre le Cancer (Équipe Labellisée) Marseille, 13009 Marseille, France
| | - Vincent Géli
- Marseille Cancer Research Centre (CRCM), U1068 INSERM, UMR7258 CNRS, UM105 Aix-Marseille University, Institut Paoli-Calmettes, Ligue Nationale Contre le Cancer (Équipe Labellisée) Marseille, 13009 Marseille, France
| | - Frédéric Causeret
- Université de Paris, Imagine Institute, Team Genetics and Development of the Cerebral Cortex, 75015 Paris, France; Université de Paris, Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, 75014 Paris, France
| | - Alice Meunier
- Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Département de Biologie, Ecole Normale Supérieure, CNRS UMR8197, INSERM U1024, Université PSL, 75005 Paris, France
| | - Nathalie Spassky
- Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Département de Biologie, Ecole Normale Supérieure, CNRS UMR8197, INSERM U1024, Université PSL, 75005 Paris, France.
| |
Collapse
|
14
|
Lupatov AY, Yarygin KN. Telomeres and Telomerase in the Control of Stem Cells. Biomedicines 2022; 10:biomedicines10102335. [PMID: 36289597 PMCID: PMC9598777 DOI: 10.3390/biomedicines10102335] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 09/15/2022] [Accepted: 09/16/2022] [Indexed: 11/16/2022] Open
Abstract
Stem cells serve as a source of cellular material in embryogenesis and postnatal growth and regeneration. This requires significant proliferative potential ensured by sufficient telomere length. Telomere attrition in the stem cells and their niche cells can result in the exhaustion of the regenerative potential of high-turnover organs, causing or contributing to the onset of age-related diseases. In this review, stem cells are examined in the context of the current telomere-centric theory of cell aging, which assumes that telomere shortening depends not just on the number of cell doublings (mitotic clock) but also on the influence of various internal and external factors. The influence of the telomerase and telomere length on the functional activity of different stem cell types, as well as on their aging and prospects of use in cell therapy applications, is discussed.
Collapse
|
15
|
Yu EY, Cheung NKV, Lue NF. Connecting telomere maintenance and regulation to the developmental origin and differentiation states of neuroblastoma tumor cells. J Hematol Oncol 2022; 15:117. [PMID: 36030273 PMCID: PMC9420296 DOI: 10.1186/s13045-022-01337-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 07/28/2022] [Indexed: 11/18/2022] Open
Abstract
A cardinal feature that distinguishes clinically high-risk neuroblastoma from low-risk tumors is telomere maintenance. Specifically, neuroblastoma tumors with either active telomerase or alternative lengthening of telomeres exhibit aggressive growth characteristics that lead to poor outcomes, whereas tumors without telomere maintenance can be managed with observation or minimal treatment. Even though the need for cancer cells to maintain telomere DNA-in order to sustain cell proliferation-is well established, recent studies suggest that the neural crest origin of neuroblastoma may enforce unique relationships between telomeres and tumor malignancy. Specifically in neuroblastoma, telomere structure and telomerase activity are correlated with the adrenergic/mesenchymal differentiation states, and manipulating telomerase activity can trigger tumor cell differentiation. Both findings may reflect features of normal neural crest development. This review summarizes recent advances in the characterization of telomere structure and telomere maintenance mechanisms in neuroblastoma and discusses the findings in the context of relevant literature on telomeres during embryonic and neural development. Understanding the canonical and non-canonical roles of telomere maintenance in neuroblastoma could reveal vulnerabilities for telomere-directed therapies with potential applications to other pediatric malignancies.
Collapse
Affiliation(s)
- Eun Young Yu
- Department of Microbiology & Immunology, W. R. Hearst Microbiology Research Center, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA
| | - Nai-Kong V Cheung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Neal F Lue
- Department of Microbiology & Immunology, W. R. Hearst Microbiology Research Center, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA.
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA.
| |
Collapse
|
16
|
Zaman Q, Zhang D, Reddy OS, Wong WT, Lai WF. Roles and Mechanisms of Astragaloside IV in Combating Neuronal Aging. Aging Dis 2022; 13:1845-1861. [DOI: 10.14336/ad.2022.0126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Accepted: 01/26/2022] [Indexed: 11/18/2022] Open
|
17
|
Cai NN, Geng Q, Jiang Y, Zhu WQ, Yang R, Zhang BY, Xiao YF, Tang B, Zhang XM. Schisandrin A and B affect the proliferation and differentiation of neural stem cells. J Chem Neuroanat 2021; 119:102058. [PMID: 34896558 DOI: 10.1016/j.jchemneu.2021.102058] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 10/31/2021] [Accepted: 12/06/2021] [Indexed: 01/20/2023]
Abstract
Schisandrin A and B (Sch A and B) are the important components of Asian dietary supplement and phytomedicine Schisandra chinensis (S. chinensis). They can enhance adult neurogenesis in vivo; however, these effects still need to be verified. Here NE-4 C neural stem cells (NSCs) were employed as the in vitro model and treated with Sch A and B at 0.1 μg/mL. EdU (5-Ethynyl-2'-deoxyuridine) labeling showed that both Sch A and B treatments enhanced NSC proliferation. Real-time PCR analysis showed the mRNA abundances of telomerase gene Tert and cell cycle gene Cyclin D1 were significantly up-regulated after the treatments. During the neurosphere induction, Sch B enhanced the neurosphere formation and neuronal differentiation, and increased the neurosphere semidiameters. Detection of the neuron differentiation marker Mapt indicates that both Sch A and B, especially Sch B, benefits the induced neuronal differentiation. Sch B treatment also enhanced mRNA expressions of the neurosphere-specific adhesion molecule Cdh2 and Wnt pathway-related genes including Mmp9, Cyclin D1 and β-catenin. Together, Sch A especially Sch B, promotes the proliferation, affects the survival, differentiation and neurogenesis of NSCs, which is consistent with their in vivo effects. This study provides further clue on the potential neuropharmacological effects of S. chinensis.
Collapse
Affiliation(s)
- Ning-Ning Cai
- College of Veterinary Medicine, Jilin University, Changchun 130062, China; Hebei Key Lab of Laboratory Animal Science, Hebei Medical University, Shijiazhuang 050017, China.
| | - Qi Geng
- Hebei Key Lab of Laboratory Animal Science, Hebei Medical University, Shijiazhuang 050017, China
| | - Yu Jiang
- College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Wen-Qian Zhu
- College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Rui Yang
- College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Bo-Yang Zhang
- College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Yu-Feng Xiao
- College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Bo Tang
- College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Xue-Ming Zhang
- College of Veterinary Medicine, Jilin University, Changchun 130062, China.
| |
Collapse
|
18
|
Mustafin RN. Relationship of Peptides and Long Non-Coding RNAs with Aging. ADVANCES IN GERONTOLOGY 2021. [DOI: 10.1134/s2079057021040081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
19
|
Samoilova EM, Belopasov VV, Ekusheva EV, Zhang C, Troitskiy AV, Baklaushev VP. Epigenetic Clock and Circadian Rhythms in Stem Cell Aging and Rejuvenation. J Pers Med 2021; 11:1050. [PMID: 34834402 PMCID: PMC8620936 DOI: 10.3390/jpm11111050] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/12/2021] [Accepted: 10/14/2021] [Indexed: 12/12/2022] Open
Abstract
This review summarizes the current understanding of the interaction between circadian rhythms of gene expression and epigenetic clocks characterized by the specific profile of DNA methylation in CpG-islands which mirror the senescence of all somatic cells and stem cells in particular. Basic mechanisms of regulation for circadian genes CLOCK-BMAL1 as well as downstream clock-controlled genes (ССG) are also discussed here. It has been shown that circadian rhythms operate by the finely tuned regulation of transcription and rely on various epigenetic mechanisms including the activation of enhancers/suppressors, acetylation/deacetylation of histones and other proteins as well as DNA methylation. Overall, up to 20% of all genes expressed by the cell are subject to expression oscillations associated with circadian rhythms. Additionally included in the review is a brief list of genes involved in the regulation of circadian rhythms, along with genes important for cell aging, and oncogenesis. Eliminating some of them (for example, Sirt1) accelerates the aging process, while the overexpression of Sirt1, on the contrary, protects against age-related changes. Circadian regulators control a number of genes that activate the cell cycle (Wee1, c-Myc, p20, p21, and Cyclin D1) and regulate histone modification and DNA methylation. Approaches for determining the epigenetic age from methylation profiles across CpG islands in individual cells are described. DNA methylation, which characterizes the function of the epigenetic clock, appears to link together such key biological processes as regeneration and functioning of stem cells, aging and malignant transformation. Finally, the main features of adult stem cell aging in stem cell niches and current possibilities for modulating the epigenetic clock and stem cells rejuvenation as part of antiaging therapy are discussed.
Collapse
Affiliation(s)
- Ekaterina M. Samoilova
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies, FMBA of Russia, 115682 Moscow, Russia; (A.V.T.); (V.P.B.)
| | | | - Evgenia V. Ekusheva
- Academy of Postgraduate Education of the Federal Scientific and Clinical Center for Specialized Types of Medical Care and Medical Technologies, FMBA of Russia, 125371 Moscow, Russia;
| | - Chao Zhang
- Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China;
| | - Alexander V. Troitskiy
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies, FMBA of Russia, 115682 Moscow, Russia; (A.V.T.); (V.P.B.)
| | - Vladimir P. Baklaushev
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies, FMBA of Russia, 115682 Moscow, Russia; (A.V.T.); (V.P.B.)
| |
Collapse
|
20
|
Tsai YY, Su CH, Tarn WY. p53 Activation in Genetic Disorders: Different Routes to the Same Destination. Int J Mol Sci 2021; 22:9307. [PMID: 34502215 PMCID: PMC8430931 DOI: 10.3390/ijms22179307] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 08/23/2021] [Accepted: 08/25/2021] [Indexed: 12/18/2022] Open
Abstract
The tumor suppressor p53 is critical for preventing neoplastic transformation and tumor progression. Inappropriate activation of p53, however, has been observed in a number of human inherited disorders that most often affect development of the brain, craniofacial region, limb skeleton, and hematopoietic system. Genes related to these developmental disorders are essentially involved in transcriptional regulation/chromatin remodeling, rRNA metabolism, DNA damage-repair pathways, telomere maintenance, and centrosome biogenesis. Perturbation of these activities or cellular processes may result in p53 accumulation in cell cultures, animal models, and perhaps humans as well. Mouse models of several p53 activation-associated disorders essentially recapitulate human traits, and inactivation of p53 in these models can alleviate disorder-related phenotypes. In the present review, we focus on how dysfunction of the aforementioned biological processes causes developmental defects via excessive p53 activation. Notably, several disease-related genes exert a pleiotropic effect on those cellular processes, which may modulate the magnitude of p53 activation and establish or disrupt regulatory loops. Finally, we discuss potential therapeutic strategies for genetic disorders associated with p53 misactivation.
Collapse
|
21
|
Haase FD, Coorey B, Riley L, Cantrill LC, Tam PPL, Gold WA. Pre-clinical Investigation of Rett Syndrome Using Human Stem Cell-Based Disease Models. Front Neurosci 2021; 15:698812. [PMID: 34512241 PMCID: PMC8423999 DOI: 10.3389/fnins.2021.698812] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 07/19/2021] [Indexed: 01/02/2023] Open
Abstract
Rett syndrome (RTT) is an X-linked neurodevelopmental disorder, mostly caused by mutations in MECP2. The disorder mainly affects girls and it is associated with severe cognitive and physical disabilities. Modeling RTT in neural and glial cell cultures and brain organoids derived from patient- or mutation-specific human induced pluripotent stem cells (iPSCs) has advanced our understanding of the pathogenesis of RTT, such as disease-causing mechanisms, disease progression, and cellular and molecular pathology enabling the identification of actionable therapeutic targets. Brain organoid models that recapitulate much of the tissue architecture and the complexity of cell types in the developing brain, offer further unprecedented opportunity for elucidating human neural development, without resorting to conventional animal models and the limited resource of human neural tissues. This review focuses on the new knowledge of RTT that has been gleaned from the iPSC-based models as well as limitations of the models and strategies to refine organoid technology in the quest for clinically relevant disease models for RTT and the broader spectrum of neurodevelopmental disorders.
Collapse
Affiliation(s)
- Florencia D. Haase
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Kids Neuroscience Centre, Kids Research, Children’s Hospital at Westmead, Westmead, NSW, Australia
- Molecular Neurobiology Research Laboratory, Kids Research, Children’s Hospital at Westmead, and Children’s Medical Research Institute, Westmead, NSW, Australia
| | - Bronte Coorey
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Kids Neuroscience Centre, Kids Research, Children’s Hospital at Westmead, Westmead, NSW, Australia
- Molecular Neurobiology Research Laboratory, Kids Research, Children’s Hospital at Westmead, and Children’s Medical Research Institute, Westmead, NSW, Australia
| | - Lisa Riley
- Rare Diseases Functional Genomics Laboratory, Kids Research, Children’s Hospital at Westmead, and Children’s Medical Research Institute, Westmead, NSW, Australia
| | - Laurence C. Cantrill
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Kids Research, Children’s Hospital at Westmead, Westmead, NSW, Australia
| | - Patrick P. L. Tam
- Embryology Research Unit, Children’s Medical Research Institute, The University of Sydney, Sydney, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Wendy A. Gold
- Kids Neuroscience Centre, Kids Research, Children’s Hospital at Westmead, Westmead, NSW, Australia
- Molecular Neurobiology Research Laboratory, Kids Research, Children’s Hospital at Westmead, and Children’s Medical Research Institute, Westmead, NSW, Australia
- Rare Diseases Functional Genomics Laboratory, Kids Research, Children’s Hospital at Westmead, and Children’s Medical Research Institute, Westmead, NSW, Australia
| |
Collapse
|
22
|
Di Mattia M, Mauro A, Citeroni MR, Dufrusine B, Peserico A, Russo V, Berardinelli P, Dainese E, Cimini A, Barboni B. Insight into Hypoxia Stemness Control. Cells 2021; 10:cells10082161. [PMID: 34440930 PMCID: PMC8394199 DOI: 10.3390/cells10082161] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 08/16/2021] [Accepted: 08/19/2021] [Indexed: 01/10/2023] Open
Abstract
Recently, the research on stemness and multilineage differentiation mechanisms has greatly increased its value due to the potential therapeutic impact of stem cell-based approaches. Stem cells modulate their self-renewing and differentiation capacities in response to endogenous and/or extrinsic factors that can control stem cell fate. One key factor controlling stem cell phenotype is oxygen (O2). Several pieces of evidence demonstrated that the complexity of reproducing O2 physiological tensions and gradients in culture is responsible for defective stem cell behavior in vitro and after transplantation. This evidence is still worsened by considering that stem cells are conventionally incubated under non-physiological air O2 tension (21%). Therefore, the study of mechanisms and signaling activated at lower O2 tension, such as those existing under native microenvironments (referred to as hypoxia), represent an effective strategy to define if O2 is essential in preserving naïve stemness potential as well as in modulating their differentiation. Starting from this premise, the goal of the present review is to report the status of the art about the link existing between hypoxia and stemness providing insight into the factors/molecules involved, to design targeted strategies that, recapitulating naïve O2 signals, enable towards the therapeutic use of stem cell for tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Miriam Di Mattia
- Unit of Basic and Applied Biosciences, Faculty of Bioscience and Agro-Food and Environmental Technology, University of Teramo, 64100 Teramo, Italy; (M.D.M.); (M.R.C.); (A.P.); (V.R.); (P.B.); (E.D.); (B.B.)
| | - Annunziata Mauro
- Unit of Basic and Applied Biosciences, Faculty of Bioscience and Agro-Food and Environmental Technology, University of Teramo, 64100 Teramo, Italy; (M.D.M.); (M.R.C.); (A.P.); (V.R.); (P.B.); (E.D.); (B.B.)
- Correspondence: ; Tel.: +39-086-1426-6888; Fax: +39-08-6126-6860
| | - Maria Rita Citeroni
- Unit of Basic and Applied Biosciences, Faculty of Bioscience and Agro-Food and Environmental Technology, University of Teramo, 64100 Teramo, Italy; (M.D.M.); (M.R.C.); (A.P.); (V.R.); (P.B.); (E.D.); (B.B.)
| | - Beatrice Dufrusine
- Department of Innovative Technologies in Medicine & Dentistry, University of Chieti-Pescara, 66100 Chieti, Italy;
- Center of Advanced Studies and Technology (CAST), 66100 Chieti, Italy
| | - Alessia Peserico
- Unit of Basic and Applied Biosciences, Faculty of Bioscience and Agro-Food and Environmental Technology, University of Teramo, 64100 Teramo, Italy; (M.D.M.); (M.R.C.); (A.P.); (V.R.); (P.B.); (E.D.); (B.B.)
| | - Valentina Russo
- Unit of Basic and Applied Biosciences, Faculty of Bioscience and Agro-Food and Environmental Technology, University of Teramo, 64100 Teramo, Italy; (M.D.M.); (M.R.C.); (A.P.); (V.R.); (P.B.); (E.D.); (B.B.)
| | - Paolo Berardinelli
- Unit of Basic and Applied Biosciences, Faculty of Bioscience and Agro-Food and Environmental Technology, University of Teramo, 64100 Teramo, Italy; (M.D.M.); (M.R.C.); (A.P.); (V.R.); (P.B.); (E.D.); (B.B.)
| | - Enrico Dainese
- Unit of Basic and Applied Biosciences, Faculty of Bioscience and Agro-Food and Environmental Technology, University of Teramo, 64100 Teramo, Italy; (M.D.M.); (M.R.C.); (A.P.); (V.R.); (P.B.); (E.D.); (B.B.)
| | - Annamaria Cimini
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy;
- Sbarro Institute for Cancer Research and Molecular Medicine and Center for Biotechnology, Temple University, Philadelphia, PA 19122, USA
| | - Barbara Barboni
- Unit of Basic and Applied Biosciences, Faculty of Bioscience and Agro-Food and Environmental Technology, University of Teramo, 64100 Teramo, Italy; (M.D.M.); (M.R.C.); (A.P.); (V.R.); (P.B.); (E.D.); (B.B.)
| |
Collapse
|
23
|
Dolejs J, Homolková H. Why Does Child Mortality Decrease With Age? Modeling the Age-Associated Decrease in Mortality Rate Using WHO Metadata From 25 Countries. Front Pediatr 2021; 9:657298. [PMID: 34458207 PMCID: PMC8387124 DOI: 10.3389/fped.2021.657298] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 07/06/2021] [Indexed: 11/23/2022] Open
Abstract
Background: Our previous study analyzed the age trajectory of mortality (ATM) in 14 European countries, while this study aimed at investigating ATM in other continents and in countries with a higher level of mortality. Data from 11 Non-European countries were used. Methods: The number of deaths was extracted from the WHO mortality database. The Halley method was used to calculate the mortality rates in all possible calendar years and all countries combined. This method enables us to combine more countries and more calendar years in one hypothetical population. Results: The age trajectory of total mortality (ATTM) and also ATM due to specific groups of diseases were very similar in the 11 non-European countries and in the 14 European countries. The level of mortality did not affect the main results found in European countries. The inverse proportion was valid for ATTM in non-European countries with two exceptions. Slower or no mortality decrease with age was detected in the first year of life, while the inverse proportion model was valid for the age range (1, 10) years in most of the main chapters of ICD10. Conclusions: The decrease in child mortality with age may be explained as the result of the depletion of individuals with congenital impairment. The majority of deaths up to the age of 10 years were related to congenital impairments, and the decrease in child mortality rate with age was a demonstration of population heterogeneity. The congenital impairments were latent and may cause death even if no congenital impairment was detected.
Collapse
Affiliation(s)
- Josef Dolejs
- Department of Informatics and Quantitative Methods, University of Hradec Králové, Hradec Králové, Czechia
| | - Helena Homolková
- Division of Pediatric Neurosurgery, Department of Pediatric and Trauma Surgery, Thomayer's Teaching Hospital and Third Faculty of Medicine, Charles University in Prague, Prague, Czechia
| |
Collapse
|
24
|
Franke K, Bublak P, Hoyer D, Billiet T, Gaser C, Witte OW, Schwab M. In vivo biomarkers of structural and functional brain development and aging in humans. Neurosci Biobehav Rev 2021; 117:142-164. [PMID: 33308708 DOI: 10.1016/j.neubiorev.2017.11.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 11/01/2017] [Accepted: 11/03/2017] [Indexed: 12/25/2022]
Abstract
Brain aging is a major determinant of aging. Along with the aging population, prevalence of neurodegenerative diseases is increasing, therewith placing economic and social burden on individuals and society. Individual rates of brain aging are shaped by genetics, epigenetics, and prenatal environmental. Biomarkers of biological brain aging are needed to predict individual trajectories of aging and the risk for age-associated neurological impairments for developing early preventive and interventional measures. We review current advances of in vivo biomarkers predicting individual brain age. Telomere length and epigenetic clock, two important biomarkers that are closely related to the mechanistic aging process, have only poor deterministic and predictive accuracy regarding individual brain aging due to their high intra- and interindividual variability. Phenotype-related biomarkers of global cognitive function and brain structure provide a much closer correlation to age at the individual level. During fetal and perinatal life, autonomic activity is a unique functional marker of brain development. The cognitive and structural biomarkers also boast high diagnostic specificity for determining individual risks for neurodegenerative diseases.
Collapse
Affiliation(s)
- K Franke
- Department of Neurology, Jena University Hospital, Jena, Germany.
| | - P Bublak
- Department of Neurology, Jena University Hospital, Jena, Germany
| | - D Hoyer
- Department of Neurology, Jena University Hospital, Jena, Germany
| | | | - C Gaser
- Department of Neurology, Jena University Hospital, Jena, Germany; Department of Psychiatry, Jena University Hospital, Jena, Germany
| | - O W Witte
- Department of Neurology, Jena University Hospital, Jena, Germany
| | - M Schwab
- Department of Neurology, Jena University Hospital, Jena, Germany
| |
Collapse
|
25
|
Abstract
This protocol entails a simple method for isolation, culturing, and in vitro differentiation of adult neural stem cells from the dentate gyrus in the hippocampus and the subventricular zone of adult mice. Cultured adult neural stem cells are an important in vitro model to investigate stem cell properties such as proliferation and differentiation and to expand the understanding of plasticity in the adult brain. For complete details on the use and execution of this protocol, please refer to Isaksen et al. (2020). A simple protocol for mouse adult neural stem cell isolation and culture Targeted differentiation into various neural cells Potential use of derived cells for studying brain plasticity
Collapse
Affiliation(s)
- Ahmed K M A Ahmed
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, 2-2, Yamadaoka, Suita, Osaka 565-0871, Japan.,WPI Immunology Frontier Research Center, Osaka University, 3-1, Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Toke Jost Isaksen
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, 2-2, Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Toshihide Yamashita
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, 2-2, Yamadaoka, Suita, Osaka 565-0871, Japan.,WPI Immunology Frontier Research Center, Osaka University, 3-1, Yamadaoka, Suita, Osaka 565-0871, Japan.,Graduate School of Frontier Bioscience, Osaka University, 2-2, Yamadaoka, Suita, Osaka 565-0871, Japan.,Department of Neuro-Medical Science, Graduate School of Medicine, Osaka University, 2-2, Yamadaoka, Suita, Osaka 565-0871, Japan
| |
Collapse
|
26
|
Rojas-Vázquez S, Blasco-Chamarro L, López-Fabuel I, Martínez-Máñez R, Fariñas I. Vascular Senescence: A Potential Bridge Between Physiological Aging and Neurogenic Decline. Front Neurosci 2021; 15:666881. [PMID: 33958987 PMCID: PMC8093510 DOI: 10.3389/fnins.2021.666881] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 03/25/2021] [Indexed: 01/25/2023] Open
Abstract
The adult mammalian brain contains distinct neurogenic niches harboring populations of neural stem cells (NSCs) with the capacity to sustain the generation of specific subtypes of neurons during the lifetime. However, their ability to produce new progeny declines with age. The microenvironment of these specialized niches provides multiple cellular and molecular signals that condition NSC behavior and potential. Among the different niche components, vasculature has gained increasing interest over the years due to its undeniable role in NSC regulation and its therapeutic potential for neurogenesis enhancement. NSCs are uniquely positioned to receive both locally secreted factors and adhesion-mediated signals derived from vascular elements. Furthermore, studies of parabiosis indicate that NSCs are also exposed to blood-borne factors, sensing and responding to the systemic circulation. Both structural and functional alterations occur in vasculature with age at the cellular level that can affect the proper extrinsic regulation of NSCs. Additionally, blood exchange experiments in heterochronic parabionts have revealed that age-associated changes in blood composition also contribute to adult neurogenesis impairment in the elderly. Although the mechanisms of vascular- or blood-derived signaling in aging are still not fully understood, a general feature of organismal aging is the accumulation of senescent cells, which act as sources of inflammatory and other detrimental signals that can negatively impact on neighboring cells. This review focuses on the interactions between vascular senescence, circulating pro-senescence factors and the decrease in NSC potential during aging. Understanding the mechanisms of NSC dynamics in the aging brain could lead to new therapeutic approaches, potentially include senolysis, to target age-dependent brain decline.
Collapse
Affiliation(s)
- Sara Rojas-Vázquez
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València-Universitat de València, Valencia, Spain.,Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Valencia, Spain.,Departamento de Biología Celular, Biología Funcional y Antropología Física, Universitat de València, Valencia, Spain
| | - Laura Blasco-Chamarro
- Departamento de Biología Celular, Biología Funcional y Antropología Física, Universitat de València, Valencia, Spain.,Instituto de Biotecnología y Biomedicina (BioTecMed), Universitat de València, Valencia, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Irene López-Fabuel
- Departamento de Biología Celular, Biología Funcional y Antropología Física, Universitat de València, Valencia, Spain.,Instituto de Biotecnología y Biomedicina (BioTecMed), Universitat de València, Valencia, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Ramón Martínez-Máñez
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València-Universitat de València, Valencia, Spain.,Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Valencia, Spain.,Unidad Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades y Nanomedicina, Universitat Politècnica de València, Centro de Investigación Príncipe Felipe, Valencia, Spain.,Unidad Mixta de Investigación en Nanomedicina y Sensores, Universitat Politècnica de València, IIS La Fe, Valencia, Spain
| | - Isabel Fariñas
- Departamento de Biología Celular, Biología Funcional y Antropología Física, Universitat de València, Valencia, Spain.,Instituto de Biotecnología y Biomedicina (BioTecMed), Universitat de València, Valencia, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| |
Collapse
|
27
|
Pathak GA, Wendt FR, Levey DF, Mecca AP, van Dyck CH, Gelernter J, Polimanti R. Pleiotropic effects of telomere length loci with brain morphology and brain tissue expression. Hum Mol Genet 2021; 30:1360-1370. [PMID: 33831179 PMCID: PMC8255129 DOI: 10.1093/hmg/ddab102] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 02/09/2021] [Accepted: 03/29/2021] [Indexed: 12/21/2022] Open
Abstract
Several studies have reported association between leukocyte telomere length (LTL) and neuropsychiatric disorders. Although telomere length is affected by environmental factors, genetic variants in certain loci are strongly associated with LTL. Thus, we aimed to identify the genomic relationship between genetic variants of LTL with brain-based regulatory changes and brain volume. We tested genetic colocalization of seven and nine LTL loci in two ancestry groups, European (EUR) and East-Asian (EAS), respectively, with brain morphology measures for 101 T1-magnetic resonance imaging-based region of interests (n = 21 821). The posterior probability (>90%) was observed for 'fourth ventricle', 'gray matter' and 'cerebellar vermal lobules I-IV' volumes. We then tested causal relationship using LTL loci for gene and methylation expression. We found causal pleiotropy for gene (EAS = four genes; EUR = five genes) and methylation expression (EUR = 17 probes; EAS = 4 probes) of brain tissues (P ≤ 2.47 × 10-6). Integrating chromatin profiles with LTL-single nucleotide polymorphisms identified 45 genes (EUR) and 79 genes (EAS) (P ≤ 9.78×10-7). We found additional 38 LTL-genes using chromatin-based gene mapping for EUR ancestry population. Gene variants in three LTL-genes-GPR37, OBFC1 and RTEL1/RTEL1-TNFRSF6B-show convergent evidence of pleiotropy with brain morphology, gene and methylation expression and chromatin association. Mapping gene functions to drug-gene interactions, we identified process 'transmission across chemical synapses' (P < 2.78 × 10-4). This study provides evidence that genetic variants of LTL have pleiotropic roles with brain-based effects that could explain the phenotypic association of LTL with several neuropsychiatric traits.
Collapse
Affiliation(s)
- Gita A Pathak
- Department of Psychiatry, Yale School of Medicine, Yale University, New Haven, CT 06551, USA,Veteran Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Frank R Wendt
- Department of Psychiatry, Yale School of Medicine, Yale University, New Haven, CT 06551, USA,Veteran Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Daniel F Levey
- Department of Psychiatry, Yale School of Medicine, Yale University, New Haven, CT 06551, USA,Veteran Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Adam P Mecca
- Department of Psychiatry, Yale School of Medicine, Yale University, New Haven, CT 06551, USA,Alzheimer’s Disease Research Unit, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Christopher H van Dyck
- Department of Psychiatry, Yale School of Medicine, Yale University, New Haven, CT 06551, USA,Alzheimer’s Disease Research Unit, Yale University School of Medicine, New Haven, CT 06511, USA,Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06511, USA,Department of Neurology, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Joel Gelernter
- Department of Psychiatry, Yale School of Medicine, Yale University, New Haven, CT 06551, USA,Veteran Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Renato Polimanti
- To whom correspondence should be addressed at: VA CT 116A2, 950 Campbell Avenue, West Haven, CT 06516, USA. Tel: +1 2039375711 ext. 5745; Fax: +1 2039373897;
| |
Collapse
|
28
|
Bakhshinyan D, Savage N, Salim SK, Venugopal C, Singh SK. The Strange Case of Jekyll and Hyde: Parallels Between Neural Stem Cells and Glioblastoma-Initiating Cells. Front Oncol 2021; 10:603738. [PMID: 33489908 PMCID: PMC7820896 DOI: 10.3389/fonc.2020.603738] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 11/24/2020] [Indexed: 12/15/2022] Open
Abstract
During embryonic development, radial glial precursor cells give rise to neural lineages, and a small proportion persist in the adult mammalian brain to contribute to long-term neuroplasticity. Neural stem cells (NSCs) reside in two neurogenic niches of the adult brain, the hippocampus and the subventricular zone (SVZ). NSCs in the SVZ are endowed with the defining stem cell properties of self-renewal and multipotent differentiation, which are maintained by intrinsic cellular programs, and extrinsic cellular and niche-specific interactions. In glioblastoma, the most aggressive primary malignant brain cancer, a subpopulation of cells termed glioblastoma stem cells (GSCs) exhibit similar stem-like properties. While there is an extensive overlap between NSCs and GSCs in function, distinct genetic profiles, transcriptional programs, and external environmental cues influence their divergent behavior. This review highlights the similarities and differences between GSCs and SVZ NSCs in terms of their gene expression, regulatory molecular pathways, niche organization, metabolic programs, and current therapies designed to exploit these differences.
Collapse
Affiliation(s)
- David Bakhshinyan
- Department of Biochemistry and Biomedical Sciences, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | - Neil Savage
- Department of Biochemistry and Biomedical Sciences, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | - Sabra Khalid Salim
- Department of Biochemistry and Biomedical Sciences, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | - Chitra Venugopal
- Department of Surgery, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | - Sheila K. Singh
- Department of Biochemistry and Biomedical Sciences, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
- Department of Surgery, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
29
|
Markiewicz-Potoczny M, Lobanova A, Loeb AM, Kirak O, Olbrich T, Ruiz S, Lazzerini Denchi E. TRF2-mediated telomere protection is dispensable in pluripotent stem cells. Nature 2021; 589:110-115. [PMID: 33239785 PMCID: PMC9161009 DOI: 10.1038/s41586-020-2959-4] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 09/14/2020] [Indexed: 12/21/2022]
Abstract
In mammals, telomere protection is mediated by the essential protein TRF2, which binds chromosome ends and ensures genome integrity1,2. TRF2 depletion results in end-to-end chromosome fusions in all cell types that have been tested so far. Here we find that TRF2 is dispensable for the proliferation and survival of mouse embryonic stem (ES) cells. Trf2-/- (also known as Terf2) ES cells do not exhibit telomere fusions and can be expanded indefinitely. In response to the deletion of TRF2, ES cells exhibit a muted DNA damage response that is characterized by the recruitment of γH2AX-but not 53BP1-to telomeres. To define the mechanisms that control this unique DNA damage response in ES cells, we performed a CRISPR-Cas9-knockout screen. We found a strong dependency of TRF2-null ES cells on the telomere-associated protein POT1B and on the chromatin remodelling factor BRD2. Co-depletion of POT1B or BRD2 with TRF2 restores a canonical DNA damage response at telomeres, resulting in frequent telomere fusions. We found that TRF2 depletion in ES cells activates a totipotent-like two-cell-stage transcriptional program that includes high levels of ZSCAN4. We show that the upregulation of ZSCAN4 contributes to telomere protection in the absence of TRF2. Together, our results uncover a unique response to telomere deprotection during early development.
Collapse
Affiliation(s)
| | | | - Anisha M Loeb
- Laboratory of Genome Integrity, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Oktay Kirak
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Teresa Olbrich
- Laboratory of Genome Integrity, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Sergio Ruiz
- Laboratory of Genome Integrity, National Cancer Institute, NIH, Bethesda, MD, USA
| | | |
Collapse
|
30
|
Pudas S, Josefsson M, Nordin Adolfsson A, Landfors M, Kauppi K, Veng-Taasti LM, Hultdin M, Adolfsson R, Degerman S. Short Leukocyte Telomeres, But Not Telomere Attrition Rates, Predict Memory Decline in the 20-Year Longitudinal Betula Study. J Gerontol A Biol Sci Med Sci 2020; 76:955-963. [PMID: 33367599 PMCID: PMC8140048 DOI: 10.1093/gerona/glaa322] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Indexed: 12/17/2022] Open
Abstract
Leukocyte telomere length (LTL) is a proposed biomarker for aging-related disorders, including cognitive decline and dementia. Long-term longitudinal studies measuring intra-individual changes in both LTL and cognitive outcomes are scarce, precluding strong conclusions about a potential aging-related relationship between LTL shortening and cognitive decline. This study investigated associations between baseline levels and longitudinal changes in LTL and memory performance across an up to 20-year follow-up in 880 dementia-free participants from a population-based study (mean baseline age: 56.8 years, range: 40–80; 52% female). Shorter baseline LTL significantly predicted subsequent memory decline (r = .34, 95% confidence interval: 0.06, 0.82), controlling for age, sex, and other relevant covariates. No significant associations were however observed between intra-individual changes in LTL and memory, neither concurrently nor with a 5-year time-lag between LTL shortening and memory decline. These results support the notion of short LTL as a predictive factor for aging-related memory decline, but suggest that LTL dynamics in adulthood and older age may be less informative of cognitive outcomes in aging. Furthermore, the results highlight the importance of long-term longitudinal evaluation of outcomes in biomarker research.
Collapse
Affiliation(s)
- Sara Pudas
- Department of Integrative Medical Biology, Umeå University, Sweden
| | - Maria Josefsson
- Center for Ageing and Demographic Research, Umeå University, Sweden
| | | | - Mattias Landfors
- Department of Medical Biosciences, Pathology, Umeå University, Sweden
| | - Karolina Kauppi
- Department of Integrative Medical Biology, Umeå University, Sweden.,Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
| | | | - Magnus Hultdin
- Department of Medical Biosciences, Pathology, Umeå University, Sweden
| | - Rolf Adolfsson
- Department of Clinical Sciences, Umeå University, Sweden
| | - Sofie Degerman
- Department of Medical Biosciences, Pathology, Umeå University, Sweden.,Department of Clinical Microbiology, Umeå University, Sweden
| |
Collapse
|
31
|
Ortiz-Álvarez G, Spassky N. One progenitor to generate them all: new evidence for multi-fated neural progenitors. Curr Opin Neurobiol 2020; 66:186-194. [PMID: 33276241 DOI: 10.1016/j.conb.2020.10.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 10/13/2020] [Accepted: 10/21/2020] [Indexed: 12/30/2022]
Abstract
The past two decades have left behind the old conception of early fate-restricted neural progenitors. The new paradigm is that of a more plastic brain, in which the cellular potential of multi-fated progenitors is progressively restricted. This is observed in the switch from neurogenesis to gliogenesis, but also in the generation of different types of glial cells and neurons at later stages. The mechanisms that establish brain cell diversity or heterogeneity within a single population are starting to be elucidated. The role of cell cycle regulators and dynamics and the asymmetric distribution of cell cargoes during cell division are attracting more attention. Understanding these mechanisms could open the way for new treatments against brain pathologies such as brain tumors or neurodegenerative disorders.
Collapse
Affiliation(s)
- Gonzalo Ortiz-Álvarez
- Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Ecole Normale Supérieure, CNRS, UMR8197, INSERM U1024, PSL Université Paris, 75005 Paris, France
| | - Nathalie Spassky
- Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Ecole Normale Supérieure, CNRS, UMR8197, INSERM U1024, PSL Université Paris, 75005 Paris, France.
| |
Collapse
|
32
|
Palmos AB, Duarte RRR, Smeeth DM, Hedges EC, Nixon DF, Thuret S, Powell TR. Telomere length and human hippocampal neurogenesis. Neuropsychopharmacology 2020; 45:2239-2247. [PMID: 32920596 PMCID: PMC7784985 DOI: 10.1038/s41386-020-00863-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 08/13/2020] [Accepted: 08/17/2020] [Indexed: 12/18/2022]
Abstract
Short telomere length is a risk factor for age-related disease, but it is also associated with reduced hippocampal volumes, age-related cognitive decline and psychiatric disorder risk. The current study explored whether telomere shortening might have an influence on cognitive function and psychiatric disorder pathophysiology, via its hypothesised effects on adult hippocampal neurogenesis. We modelled telomere shortening in human hippocampal progenitor cells in vitro using a serial passaging protocol that mimics the end-replication problem. Serially passaged progenitors demonstrated shorter telomeres (P ≤ 0.05), and reduced rates of cell proliferation (P ≤ 0.001), with no changes in the ability of cells to differentiate into neurons or glia. RNA-sequencing and gene-set enrichment analyses revealed an effect of cell ageing on gene networks related to neurogenesis, telomere maintenance, cell senescence and cytokine production. Downregulated transcripts in our model showed a significant overlap with genes regulating cognitive function (P ≤ 1 × 10-5), and risk for schizophrenia (P ≤ 1 × 10-10) and bipolar disorder (P ≤ 0.005). Collectively, our results suggest that telomere shortening could represent a mechanism that moderates the proliferative capacity of human hippocampal progenitors, which may subsequently impact on human cognitive function and psychiatric disorder pathophysiology.
Collapse
Affiliation(s)
- Alish B. Palmos
- grid.13097.3c0000 0001 2322 6764Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
| | - Rodrigo R. R. Duarte
- grid.13097.3c0000 0001 2322 6764Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK ,grid.5386.8000000041936877XDivision of Infectious Diseases, Weill Cornell Medicine, Cornell University, New York, NY USA
| | - Demelza M. Smeeth
- grid.13097.3c0000 0001 2322 6764Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
| | - Erin C. Hedges
- grid.13097.3c0000 0001 2322 6764Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
| | - Douglas F. Nixon
- grid.5386.8000000041936877XDivision of Infectious Diseases, Weill Cornell Medicine, Cornell University, New York, NY USA
| | - Sandrine Thuret
- grid.13097.3c0000 0001 2322 6764Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK ,Department of Neurology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Timothy R. Powell
- grid.13097.3c0000 0001 2322 6764Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK ,grid.5386.8000000041936877XDivision of Infectious Diseases, Weill Cornell Medicine, Cornell University, New York, NY USA
| |
Collapse
|
33
|
Olatz C, Patricia GG, Jon L, Iker B, Carmen DLH, Fernando U, Gaskon I, Ramon PJ. Is There Such a Thing as a Genuine Cancer Stem Cell Marker? Perspectives from the Gut, the Brain and the Dental Pulp. BIOLOGY 2020; 9:biology9120426. [PMID: 33260962 PMCID: PMC7760753 DOI: 10.3390/biology9120426] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/24/2020] [Accepted: 11/26/2020] [Indexed: 12/24/2022]
Abstract
The conversion of healthy stem cells into cancer stem cells (CSCs) is believed to underlie tumor relapse after surgical removal and fuel tumor growth and invasiveness. CSCs often arise from the malignant transformation of resident multipotent stem cells, which are present in most human tissues. Some organs, such as the gut and the brain, can give rise to very aggressive types of cancers, contrary to the dental pulp, which is a tissue with a very remarkable resistance to oncogenesis. In this review, we focus on the similarities and differences between gut, brain and dental pulp stem cells and their related CSCs, placing a particular emphasis on both their shared and distinctive cell markers, including the expression of pluripotency core factors. We discuss some of their similarities and differences with regard to oncogenic signaling, telomerase activity and their intrinsic propensity to degenerate to CSCs. We also explore the characteristics of the events and mutations leading to malignant transformation in each case. Importantly, healthy dental pulp stem cells (DPSCs) share a great deal of features with many of the so far reported CSC phenotypes found in malignant neoplasms. However, there exist literally no reports about the contribution of DPSCs to malignant tumors. This raises the question about the particularities of the dental pulp and what specific barriers to malignancy might be present in the case of this tissue. These notable differences warrant further research to decipher the singular properties of DPSCs that make them resistant to transformation, and to unravel new therapeutic targets to treat deadly tumors.
Collapse
Affiliation(s)
- Crende Olatz
- Department of Cell Biology and Histology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain; (C.O.); (G.-G.P.); (L.J.); (B.I.); (d.l.H.C.); (U.F.)
| | - García-Gallastegui Patricia
- Department of Cell Biology and Histology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain; (C.O.); (G.-G.P.); (L.J.); (B.I.); (d.l.H.C.); (U.F.)
| | - Luzuriaga Jon
- Department of Cell Biology and Histology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain; (C.O.); (G.-G.P.); (L.J.); (B.I.); (d.l.H.C.); (U.F.)
| | - Badiola Iker
- Department of Cell Biology and Histology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain; (C.O.); (G.-G.P.); (L.J.); (B.I.); (d.l.H.C.); (U.F.)
| | - de la Hoz Carmen
- Department of Cell Biology and Histology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain; (C.O.); (G.-G.P.); (L.J.); (B.I.); (d.l.H.C.); (U.F.)
| | - Unda Fernando
- Department of Cell Biology and Histology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain; (C.O.); (G.-G.P.); (L.J.); (B.I.); (d.l.H.C.); (U.F.)
| | - Ibarretxe Gaskon
- Department of Cell Biology and Histology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain; (C.O.); (G.-G.P.); (L.J.); (B.I.); (d.l.H.C.); (U.F.)
- Correspondence: (I.G.); (P.J.R.); Tel.: +34-946-013-218 (I.G.); +34-946-012-426 (P.J.R.)
| | - Pineda Jose Ramon
- Department of Cell Biology and Histology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain; (C.O.); (G.-G.P.); (L.J.); (B.I.); (d.l.H.C.); (U.F.)
- Achucarro Basque Center for Neuroscience Fundazioa, 48940 Leioa, Spain
- Correspondence: (I.G.); (P.J.R.); Tel.: +34-946-013-218 (I.G.); +34-946-012-426 (P.J.R.)
| |
Collapse
|
34
|
Dolejs J, Homolková H. Why Does Child Mortality Decrease With Age? Modeling the Age-Associated Decrease in Mortality Rate Using WHO Metadata From 14 European Countries. Front Pediatr 2020; 8:527811. [PMID: 33194882 PMCID: PMC7653179 DOI: 10.3389/fped.2020.527811] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 09/28/2020] [Indexed: 11/26/2022] Open
Abstract
Background: Mortality rate rapidly decreases with age after birth, and, simultaneously, the spectrum of death causes show remarkable changes with age. This study analyzed age-associated decreases in mortality rate from diseases of all main chapters of the 10th revision of the International Classification of Diseases. Methods: The number of deaths was extracted from the mortality database of the World Health Organization. As zero cases could be ascertained for a specific age category, the Halley method was used to calculate the mortality rates in all possible calendar years and in all countries combined. Results: All causes mortality from the 1st day of life to the age of 10 years can be represented by an inverse proportion model with a single parameter. High coefficients of determination were observed for total mortality in all populations (arithmetic mean = 0.9942 and standard deviation = 0.0039). Slower or no mortality decrease with age was detected in the 1st year of life, while the inverse proportion method was valid for the age range [1, 10) years in most of all main chapters with three exceptions. The decrease was faster for the chapter "Certain conditions originating in the perinatal period" (XVI).The inverse proportion was valid already from the 1st day for the chapter "Congenital malformations, deformations and chromosomal abnormalities" (XVII).The shape of the mortality decrease was very different for the chapter "Neoplasms" (II) and the rates of mortality from neoplasms were age-independent in the age range [1, 10) years in all populations. Conclusion: The theory of congenital individual risks of death is presented and can explain the results. If it is valid, latent congenital impairments may be present among all cases of death that are not related to congenital impairments. All results are based on published data, and the data are presented as a supplement.
Collapse
Affiliation(s)
- Josef Dolejs
- Department of Informatics and Quantitative Methods, University of Hradec Králové, Hradec Králové, Czechia
| | - Helena Homolková
- Division of Pediatric Neurosurgery, Department of Pediatric and Trauma Surgery, Thomayer's Teaching Hospital and Third Faculty of Medicine, Charles University in Prague, Prague, Czechia
| |
Collapse
|
35
|
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
|
36
|
Levstek T, Kozjek E, Dolžan V, Trebušak Podkrajšek K. Telomere Attrition in Neurodegenerative Disorders. Front Cell Neurosci 2020; 14:219. [PMID: 32760251 PMCID: PMC7373805 DOI: 10.3389/fncel.2020.00219] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 06/19/2020] [Indexed: 12/13/2022] Open
Abstract
Telomere attrition is increased in various disorders and is therefore a potential biomarker for diagnosis and/or prognosis of these disorders. The contribution of telomere attrition in the pathogenesis of neurodegenerative disorders is yet to be fully elucidated. We are reviewing the current knowledge regarding the telomere biology in two common neurodegenerative disorders, Alzheimer's disease (AD), and Parkinson's disease (PD). Furthermore, we are discussing future prospective of telomere research in these disorders. The majority of studies reported consistent evidence of the accelerated telomere attrition in AD patients, possibly in association with elevated oxidative stress levels. On the other hand in PD, various studies reported contradictory evidence regarding telomere attrition. Consequently, due to the low specificity and sensitivity, the clinical benefit of telomere length as a biomarker of neurodegenerative disease development and progression is not yet recognized. Nevertheless, longitudinal studies in large carefully selected cohorts might provide further elucidation of the complex involvement of the telomeres in the pathogenesis of neurodegenerative diseases. Telomere length maintenance is a complex process characterized by environmental, genetic, and epigenetic determinants. Thus, in addition to the selection of the study cohort, also the selection of analytical methods and types of biological samples for evaluation of the telomere attrition is of utmost importance.
Collapse
Affiliation(s)
- Tina Levstek
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Eva Kozjek
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Vita Dolžan
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Katarina Trebušak Podkrajšek
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Clinical Institute for Special Laboratory Diagnostics, University Children’s Hospital, University Medical Centre Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
37
|
Biohorology and biomarkers of aging: Current state-of-the-art, challenges and opportunities. Ageing Res Rev 2020; 60:101050. [PMID: 32272169 DOI: 10.1016/j.arr.2020.101050] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Revised: 02/06/2020] [Accepted: 03/22/2020] [Indexed: 02/08/2023]
Abstract
The aging process results in multiple traceable footprints, which can be quantified and used to estimate an organism's age. Examples of such aging biomarkers include epigenetic changes, telomere attrition, and alterations in gene expression and metabolite concentrations. More than a dozen aging clocks use molecular features to predict an organism's age, each of them utilizing different data types and training procedures. Here, we offer a detailed comparison of existing mouse and human aging clocks, discuss their technological limitations and the underlying machine learning algorithms. We also discuss promising future directions of research in biohorology - the science of measuring the passage of time in living systems. Overall, we expect deep learning, deep neural networks and generative approaches to be the next power tools in this timely and actively developing field.
Collapse
|
38
|
Papadopoulos D, Magliozzi R, Mitsikostas DD, Gorgoulis VG, Nicholas RS. Aging, Cellular Senescence, and Progressive Multiple Sclerosis. Front Cell Neurosci 2020; 14:178. [PMID: 32694983 PMCID: PMC7338849 DOI: 10.3389/fncel.2020.00178] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 05/22/2020] [Indexed: 12/12/2022] Open
Abstract
Aging is one of the most important risk factors for the development of several neurodegenerative diseases including progressive multiple sclerosis (MS). Cellular senescence (CS) is a key biological process underlying aging. Several stressors associated with aging and MS pathology, such as oxidative stress, mitochondrial dysfunction, cytokines and replicative exhaustion are known triggers of cellular senescence. Senescent cells exhibit stereotypical metabolic and functional changes, which include cell-cycle arrest and acquiring a pro-inflammatory phenotype secreting cytokines, growth factors, metalloproteinases and reactive oxygen species. They accumulate with aging and can convert neighboring cells to senescence in a paracrine manner. In MS, accelerated cellular senescence may drive disease progression by promoting chronic non-remitting inflammation, loss or altered immune, glial and neuronal function, failure of remyelination, impaired blood-brain barrier integrity and ultimately neurodegeneration. Here we discuss the evidence linking cellular senescence to the pathogenesis of MS and the putative role of senolytic and senomorphic agents as neuroprotective therapies in tackling disease progression.
Collapse
Affiliation(s)
- Dimitrios Papadopoulos
- Molecular Carcinogenesis Group, Laboratory of Histology and Embryology, School of Health Sciences, National and Kapodistrian University of Athens, Athens, Greece
| | - Roberta Magliozzi
- Department of Neuroscience, Biomedicine and Movement, University of Verona, Verona, Italy
| | - Dimos D Mitsikostas
- First Department of Neurology, Aeginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Vassilis G Gorgoulis
- Molecular Carcinogenesis Group, Laboratory of Histology and Embryology, School of Health Sciences, National and Kapodistrian University of Athens, Athens, Greece
| | - Richard S Nicholas
- Department of Neuroinflammation and Neurodegeneration, Faculty of Medicine, Imperial College London, London, United Kingdom.,Department of Visual Neuroscience, Faculty of Brain Sciences, Institute of Ophthalmology, University College London, London, United Kingdom
| |
Collapse
|
39
|
Cianflone E, Torella M, Biamonte F, De Angelis A, Urbanek K, Costanzo FS, Rota M, Ellison-Hughes GM, Torella D. Targeting Cardiac Stem Cell Senescence to Treat Cardiac Aging and Disease. Cells 2020; 9:E1558. [PMID: 32604861 PMCID: PMC7349658 DOI: 10.3390/cells9061558] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 06/19/2020] [Accepted: 06/25/2020] [Indexed: 12/13/2022] Open
Abstract
Adult stem/progenitor are a small population of cells that reside in tissue-specific niches and possess the potential to differentiate in all cell types of the organ in which they operate. Adult stem cells are implicated with the homeostasis, regeneration, and aging of all tissues. Tissue-specific adult stem cell senescence has emerged as an attractive theory for the decline in mammalian tissue and organ function during aging. Cardiac aging, in particular, manifests as functional tissue degeneration that leads to heart failure. Adult cardiac stem/progenitor cell (CSC) senescence has been accordingly associated with physiological and pathological processes encompassing both non-age and age-related decline in cardiac tissue repair and organ dysfunction and disease. Senescence is a highly active and dynamic cell process with a first classical hallmark represented by its replicative limit, which is the establishment of a stable growth arrest over time that is mainly secondary to DNA damage and reactive oxygen species (ROS) accumulation elicited by different intrinsic stimuli (like metabolism), as well as external stimuli and age. Replicative senescence is mainly executed by telomere shortening, the activation of the p53/p16INK4/Rb molecular pathways, and chromatin remodeling. In addition, senescent cells produce and secrete a complex mixture of molecules, commonly known as the senescence-associated secretory phenotype (SASP), that regulate most of their non-cell-autonomous effects. In this review, we discuss the molecular and cellular mechanisms regulating different characteristics of the senescence phenotype and their consequences for adult CSCs in particular. Because senescent cells contribute to the outcome of a variety of cardiac diseases, including age-related and unrelated cardiac diseases like diabetic cardiomyopathy and anthracycline cardiotoxicity, therapies that target senescent cell clearance are actively being explored. Moreover, the further understanding of the reversibility of the senescence phenotype will help to develop novel rational therapeutic strategies.
Collapse
Affiliation(s)
- Eleonora Cianflone
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy;
| | - Michele Torella
- Department of Translational Medical Sciences, AORN dei Colli/Monaldi Hospital, University of Campania “L. Vanvitelli”, Via Leonardo Bianchi, 80131 Naples, Italy;
| | - Flavia Biamonte
- Department of Experimental and Clinical Medicine and Interdepartmental Centre of Services (CIS), Magna Graecia University, 88100 Catanzaro, Italy; (F.B.); (F.S.C.)
| | - Antonella De Angelis
- Department of Experimental Medicine, Section of Pharmacology, University of Campania “L.Vanvitelli”, 80121 Naples, Italy;
| | - Konrad Urbanek
- Molecular and Cellular Cardiology, Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| | - Francesco S. Costanzo
- Department of Experimental and Clinical Medicine and Interdepartmental Centre of Services (CIS), Magna Graecia University, 88100 Catanzaro, Italy; (F.B.); (F.S.C.)
| | - Marcello Rota
- Department of Physiology, New York Medical College, Valhalla, NY 10595, USA;
| | - Georgina M. Ellison-Hughes
- Centre for Human and Applied Physiological Sciences and Centre for Stem Cells and Regenerative Medicine, School of Basic and Medical Biosciences, Faculty of Life Sciences & Medicine, King’s College London, Guys Campus-Great Maze Pond rd, London SE1 1UL, UK;
| | - Daniele Torella
- Molecular and Cellular Cardiology, Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| |
Collapse
|
40
|
Whittemore K, Derevyanko A, Martinez P, Serrano R, Pumarola M, Bosch F, Blasco MA. Telomerase gene therapy ameliorates the effects of neurodegeneration associated to short telomeres in mice. Aging (Albany NY) 2020; 11:2916-2948. [PMID: 31140977 PMCID: PMC6555470 DOI: 10.18632/aging.101982] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 05/17/2019] [Indexed: 12/26/2022]
Abstract
Neurodegenerative diseases associated with old age such as Alzheimer’s disease present major problems for society, and they currently have no cure. The telomere protective caps at the ends of chromosomes shorten with age, and when they become critically short, they can induce a persistent DNA damage response at chromosome ends, triggering secondary cellular responses such as cell death and cellular senescence. Mice and humans with very short telomeres owing to telomerase deficiencies have an earlier onset of pathologies associated with loss of the regenerative capacity of tissues. However, the effects of short telomeres in very low proliferative tissues such as the brain have not been thoroughly investigated. Here, we describe a mouse model of neurodegeneration owing to presence of short telomeres in the brain as the consequence of telomerase deficiency. Interestingly, we find similar signs of neurodegeneration in very old mice as the consequence of physiological mouse aging. Next, we demonstrate that delivery of telomerase gene therapy to the brain of these mice results in amelioration of some of these neurodegeneration phenotypes. These findings suggest that short telomeres contribute to neurodegeneration diseases with aging and that telomerase activation may have a therapeutic value in these diseases.
Collapse
Affiliation(s)
- Kurt Whittemore
- Telomeres and Telomerase Group, Molecular Oncology Program, Spanish National Cancer Centre (CNIO), Madrid 28029, Spain
| | - Aksinya Derevyanko
- Telomeres and Telomerase Group, Molecular Oncology Program, Spanish National Cancer Centre (CNIO), Madrid 28029, Spain
| | - Paula Martinez
- Telomeres and Telomerase Group, Molecular Oncology Program, Spanish National Cancer Centre (CNIO), Madrid 28029, Spain
| | - Rosa Serrano
- Telomeres and Telomerase Group, Molecular Oncology Program, Spanish National Cancer Centre (CNIO), Madrid 28029, Spain
| | - Martí Pumarola
- Unit of Murine and Comparative Pathology (UPMiC), Department of Animal Medicine and Surgery, Veterinary Faculty, Universitat Autònoma de Barcelona, 08193 Bellaterra (Cerdanyola del Vallès), Barcelona, Spain.,Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Universitat Autònoma de Barcelona, 08193 Bellaterra (Cerdanyola del Vallès), Barcelona, Spain
| | - Fàtima Bosch
- Center of Animal Biotechnology and Gene Therapy, Department of Animal Medicine and Surgery, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain.,Center of Animal Biotechnology and Gene Therapy, Department of Biochemistry and Molecular Biology, School of Veterinary Medicine, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
| | - Maria A Blasco
- Telomeres and Telomerase Group, Molecular Oncology Program, Spanish National Cancer Centre (CNIO), Madrid 28029, Spain
| |
Collapse
|
41
|
Long-lived post-mitotic cell aging: is a telomere clock at play? Mech Ageing Dev 2020; 189:111256. [PMID: 32380018 DOI: 10.1016/j.mad.2020.111256] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 04/25/2020] [Accepted: 04/27/2020] [Indexed: 12/27/2022]
Abstract
Senescence is a cellular response to stress for both dividing and post-mitotic cells. Noteworthy, long-lived post-mitotic cells (collectively named LLPMCs), which can live for decades in the organism, can exhibit a distinct type of cellular aging characterized by a progressive functional decline not associated to an overt senescence phenotype. The age-related drivers of senescence and aging in LLPMCs remain largely unknown. There is evidence that an increased production of reactive oxygen species (ROS) due to dysfunctional mitochondria, coupled with an inherent inability of cellular-degradation mechanisms to remove damaged molecules, is responsible for senescence and aging in LLPMC. Although telomeric DNA shortening, by nature linked to cell division, is generally not considered as a driver of LLPMC aging and senescence, we discuss recent reports revealing the existence of age-related telomere changes in LLPMC. These findings reveal unexpected roles for telomeres in LLPMC function and invite us to consider the hypothesis of a complex telomere clock involved in both dividing and non-dividing cell aging.
Collapse
|
42
|
Nemirovich-Danchenko NM, Khodanovich MY. Telomerase Gene Editing in the Neural Stem Cells in vivo as a Possible New Approach against Brain Aging. RUSS J GENET+ 2020. [DOI: 10.1134/s1022795420040092] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
43
|
Fossel M. A unified model of dementias and age-related neurodegeneration. Alzheimers Dement 2020; 16:365-383. [PMID: 31943780 DOI: 10.1002/alz.12012] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 11/09/2019] [Accepted: 11/25/2019] [Indexed: 12/14/2022]
|
44
|
Fan Y, Winanto, Ng SY. Replacing what's lost: a new era of stem cell therapy for Parkinson's disease. Transl Neurodegener 2020; 9:2. [PMID: 31911835 PMCID: PMC6945567 DOI: 10.1186/s40035-019-0180-x] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 11/26/2019] [Indexed: 12/31/2022] Open
Abstract
Background Stem cells hold tremendous promise for regenerative medicine because they can be expanded infinitely, giving rise to large numbers of differentiated cells required for transplantation. Stem cells can be derived from fetal sources, embryonic origins (embryonic stem cells or ESCs) or reprogrammed from adult cell types (induced pluripotent stem cells or iPSCs). One unique property of stem cells is their ability to be directed towards specific cell types of clinical interest, and can mature into functional cell types in vivo. While transplantations of fetal or ESC-derived tissues are known to illicit a host immunogenic response, autologous transplantations using cell types derived from one’s own iPSCs eliminate risks of tissue rejection and reduce the need for immunosuppressants. However, even with these benefits, cell therapy comes with significant hurdles that researchers are starting to overcome. In this review, we will discuss the various steps to ensure safety, efficacy and clinical practicality of cell replacement therapy in neurodegenerative diseases, in particular, Parkinson’s disease. Main body Parkinson’s disease (PD) results from a loss of dopaminergic neurons from the substantia nigra and is an ideal target for cell replacement therapy. Early trials using fetal midbrain material in the late 1980s have resulted in long term benefit for some patients, but there were multiple shortcomings including the non-standardization and quality control of the transplanted fetal material, and graft-induced dyskinesia that some patients experience as a result. On the other hand, pluripotent stem cells such as ESCs and iPSCs serve as an attractive source of cells because they can be indefinitely cultured and is an unlimited source of cells. Stem cell technologies and our understanding of the developmental potential of ESCs and iPSCs have deepened in recent years and a clinical trial for iPSC-derived dopaminergic cells is currently undergoing for PD patients in Japan. In this focused review, we will first provide a historical aspect of cell therapies in PD, and then discuss the various challenges pertaining to the safety and efficacy of stem cell-based cell transplantations, and how these hurdles were eventually overcome. Conclusion With the maturity of the iPSC technology, cell transplantation appears to be a safe and effective therapy. Grafts in non-human primates survive and remain functional for more than 2 years after transplantation, with no signs of tumorigenesis, indicating safety and efficacy of the treatment. However, immunosuppressants are still required because of the lack of “universal stem cells” that would not evoke an immune response. The results of ongoing and upcoming trials by a global consortium known as GForce-PD would be highly anticipated because the success of these trials would open up possibilities for using cell therapy for the treatment of PD and other degenerative diseases.
Collapse
Affiliation(s)
- Yong Fan
- 1The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150 China
| | - Winanto
- 2Institute of Molecular and Cell Biology, ASTAR Research Entities, Singapore, 138673 Singapore
| | - Shi-Yan Ng
- 1The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150 China.,2Institute of Molecular and Cell Biology, ASTAR Research Entities, Singapore, 138673 Singapore.,3Yong Loo Lin School of Medicine (Physiology), National University of Singapore, Singapore, 117456 Singapore.,4National Neuroscience Institute, Singapore, 308433 Singapore
| |
Collapse
|
45
|
Ullah M, Ng NN, Concepcion W, Thakor AS. Emerging role of stem cell-derived extracellular microRNAs in age-associated human diseases and in different therapies of longevity. Ageing Res Rev 2020; 57:100979. [PMID: 31704472 DOI: 10.1016/j.arr.2019.100979] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 10/17/2019] [Accepted: 10/28/2019] [Indexed: 12/15/2022]
Abstract
Organismal aging involves the progressive decline in organ function and increased susceptibility to age-associated diseases. This has been associated with the aging of stem cell populations within the body that decreases the capacity of stem cells to self-renew, differentiate, and regenerate damaged tissues and organs. This review aims to explore how aging is associated with the dysregulation of stem cell-derived extracellular vesicles (SCEVs) and their corresponding miRNA cargo (SCEV-miRNAs), which are short non-coding RNAs involved in post-transcriptional regulation of target genes. Recent evidence has suggested that in aging stem cells, SCEV-miRNAs may play a vital role regulating various processes that contribute to aging: cellular senescence, stem cell exhaustion, telomere length, and circadian rhythm. Hence, further clarifying the age-dependent molecular mechanisms through which SCEV-miRNAs exert their downstream effects may inform a greater understanding of the biology of aging, elucidate their role in stem cell function, and identify important targets for future regenerative therapies. Additionally, current studies evaluating therapeutic role of SCEVs and SCEV-miRNAs in treating several age-associated diseases are also discussed.
Collapse
|
46
|
Wade M, Fox NA, Zeanah CH, Nelson CA, Drury SS. Telomere Length and Psychopathology: Specificity and Direction of Effects Within the Bucharest Early Intervention Project. J Am Acad Child Adolesc Psychiatry 2020; 59:140-148.e3. [PMID: 30844465 PMCID: PMC8056885 DOI: 10.1016/j.jaac.2019.02.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 01/27/2019] [Accepted: 02/26/2019] [Indexed: 11/29/2022]
Abstract
OBJECTIVE Telomere length (TL) has been linked to several psychiatric conditions in children and adults. Telomere shortening is accelerated by early adversity, including maltreatment and psychosocial deprivation. These experiences also increase the risk of psychopathology in many domains. Two fundamental issues remain unresolved. The first concerns the specificity of the relations between TL and different dimensions of psychopathology; and the second relates to the direction of association between TL and psychopathology. METHOD This study addressed these shortcomings in a 2-fold manner. First, the association between TL and statistically independent general, internalizing, and externalizing psychopathology factors was examined to determine the specificity of this relation. Second, a 2-wave longitudinal cross-lagged model was used to explicitly examine the direction of the relation between TL and each psychopathology factor. Data were drawn from the Bucharest Early Intervention Project, a longitudinal study exploring the impact of severe psychosocial deprivation on child health and development (N = 195). At 8 to 10 and 12 to 14 years of age, buccal DNA was collected and teachers and/or caregivers reported on different domains of psychopathology. RESULTS Longitudinal path analyses showed that shorter TL was specifically associated with higher internalizing psychopathology at 8 to 10 years of age. In contrast, at 12 to 14 years, shorter TL was associated with higher general psychopathology. Most telling, internalizing psychopathology at 8 to 10 years predicted shorter TL at 12 to 14 years, with no reciprocal effects. CONCLUSION Results suggest that telomere erosion could be a consequence of distress-related psychopathology rather than a selection mechanism for later psychiatric problems. CLINICAL TRIAL REGISTRATION INFORMATION The Bucharest Early Intervention Project; https://clinicaltrials.gov/; NCT00747396.
Collapse
Affiliation(s)
- Mark Wade
- Boston Children's Hospital and Harvard Medical School, Boston, MA.
| | | | | | - Charles A Nelson
- Boston Children's Hospital and Harvard Medical School, Boston, MA; Harvard Graduate School of Education, Cambridge, MA
| | - Stacy S Drury
- Tulane University School of Medicine, New Orleans, LA
| |
Collapse
|
47
|
Wang X, Ma Y, Xu R, Ma J, Zhang H, Qi S, Xu J, Qin X, Zhang H, Liu C, Chen J, Li B, Yang H, Saijilafu. c‐Myc controls the fate of neural progenitor cells during cerebral cortex development. J Cell Physiol 2019; 235:4011-4021. [PMID: 31625158 DOI: 10.1002/jcp.29297] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Accepted: 09/30/2019] [Indexed: 01/03/2023]
Affiliation(s)
- Xiu‐Li Wang
- Department of Orthopaedics The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University Suzhou Jiangsu China
| | - Yan‐Xia Ma
- Department of Orthopaedics The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University Suzhou Jiangsu China
| | - Ren‐Jie Xu
- Department of Orthopaedics The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University Suzhou Jiangsu China
- Department of Orthopaedics Suzhou Municipal Hospital/The Affiliated Hospital of Nanjing Medical University Suzhou Jiangsu China
| | - Jin‐Jin Ma
- Department of Orthopaedics The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University Suzhou Jiangsu China
| | - Hong‐Cheng Zhang
- Department of Orthopaedics The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University Suzhou Jiangsu China
| | - Shi‐Bin Qi
- Department of Orthopaedics The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University Suzhou Jiangsu China
| | - Jin‐Hui Xu
- Department of Orthopaedics The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University Suzhou Jiangsu China
| | - Xu‐Zhen Qin
- Department of Orthopaedics The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University Suzhou Jiangsu China
| | - Hao‐Nan Zhang
- Department of Orthopaedics The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University Suzhou Jiangsu China
| | - Chang‐Mei Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology Chinese Academy of Science Beijing China
- Savaid Medical School University of Chinese Academy of Sciences Beijing China
| | - Jian‐Quan Chen
- Department of Orthopaedics The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University Suzhou Jiangsu China
| | - Bin Li
- Department of Orthopaedics The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University Suzhou Jiangsu China
| | - Hui‐Lin Yang
- Department of Orthopaedics The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University Suzhou Jiangsu China
| | - Saijilafu
- Department of Orthopaedics The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University Suzhou Jiangsu China
| |
Collapse
|
48
|
Ohashi M, Korsakova E, Allen D, Lee P, Fu K, Vargas BS, Cinkornpumin J, Salas C, Park JC, Germanguz I, Langerman J, Chronis C, Kuoy E, Tran S, Xiao X, Pellegrini M, Plath K, Lowry WE. Loss of MECP2 Leads to Activation of P53 and Neuronal Senescence. Stem Cell Reports 2019; 10:1453-1463. [PMID: 29742391 PMCID: PMC5995366 DOI: 10.1016/j.stemcr.2018.04.001] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 03/30/2018] [Accepted: 04/03/2018] [Indexed: 01/15/2023] Open
Abstract
To determine the role for mutations of MECP2 in Rett syndrome, we generated isogenic lines of human induced pluripotent stem cells, neural progenitor cells, and neurons from patient fibroblasts with and without MECP2 expression in an attempt to recapitulate disease phenotypes in vitro. Molecular profiling uncovered neuronal-specific gene expression changes, including induction of a senescence-associated secretory phenotype (SASP) program. Patient-derived neurons made without MECP2 showed signs of stress, including induction of P53, and senescence. The induction of P53 appeared to affect dendritic branching in Rett neurons, as P53 inhibition restored dendritic complexity. The induction of P53 targets was also detectable in analyses of human Rett patient brain, suggesting that this disease-in-a-dish model can provide relevant insights into the human disorder. Development of a patient-specific model of human Rett syndrome Loss of function of MECP2 leads to induction of p53 MECP2 null neurons show evidence of cellular senescence Inhibition of p53 can restore dendritic branching in MECP2 null neurons
Collapse
Affiliation(s)
- Minori Ohashi
- Department of Molecular Cell and Developmental Biology, UCLA, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center for Regenerative Medicine, UCLA, Los Angeles, CA 90095, USA
| | - Elena Korsakova
- Molecular Biology Institute, UCLA, Los Angeles, CA 90095, USA
| | - Denise Allen
- Department of Molecular Cell and Developmental Biology, UCLA, Los Angeles, CA 90095, USA
| | - Peiyee Lee
- Department of Molecular Cell and Developmental Biology, UCLA, Los Angeles, CA 90095, USA
| | - Kai Fu
- Department of Molecular Cell and Developmental Biology, UCLA, Los Angeles, CA 90095, USA
| | - Benni S Vargas
- Department of Molecular Cell and Developmental Biology, UCLA, Los Angeles, CA 90095, USA
| | - Jessica Cinkornpumin
- Department of Molecular Cell and Developmental Biology, UCLA, Los Angeles, CA 90095, USA
| | - Carlos Salas
- Department of Molecular Cell and Developmental Biology, UCLA, Los Angeles, CA 90095, USA
| | - Jenny C Park
- Department of Molecular Cell and Developmental Biology, UCLA, Los Angeles, CA 90095, USA
| | - Igal Germanguz
- Department of Molecular Cell and Developmental Biology, UCLA, Los Angeles, CA 90095, USA
| | - Justin Langerman
- Department of Biological Chemistry, UCLA, Los Angeles, CA 90095, USA
| | | | - Edward Kuoy
- Department of Biological Chemistry, UCLA, Los Angeles, CA 90095, USA
| | - Stephen Tran
- Department of Integrative Biology and Physiology, UCLA, Los Angeles, CA 90095, USA
| | - Xinshu Xiao
- Department of Integrative Biology and Physiology, UCLA, Los Angeles, CA 90095, USA
| | - Matteo Pellegrini
- Department of Molecular Cell and Developmental Biology, UCLA, Los Angeles, CA 90095, USA
| | - Kathrin Plath
- Department of Biological Chemistry, UCLA, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center for Regenerative Medicine, UCLA, Los Angeles, CA 90095, USA.
| | - William E Lowry
- Department of Molecular Cell and Developmental Biology, UCLA, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center for Regenerative Medicine, UCLA, Los Angeles, CA 90095, USA; Molecular Biology Institute, UCLA, Los Angeles, CA 90095, USA.
| |
Collapse
|
49
|
Gillis JC, Chang SC, Wang W, Simon NM, Normand SL, Rosner BA, Blacker D, DeVivo I, Okereke OI. The relation of telomere length at midlife to subsequent 20-year depression trajectories among women. Depress Anxiety 2019; 36:565-575. [PMID: 30958913 PMCID: PMC6548605 DOI: 10.1002/da.22892] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 02/18/2019] [Accepted: 03/13/2019] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND Telomeres cap and protect DNA but shorten with each somatic cell division. Aging and environmental and lifestyle factors contribute to the speed of telomere attrition. Current evidence suggests a link between relative telomere length (RTL) and depression but the directionality of the relationship remains unclear. We prospectively examined associations between RTL and subsequent depressive symptom trajectories. METHODS Among 8,801 women of the Nurses' Health Study, depressive symptoms were measured every 4 years from 1992 to 2012; group-based trajectories of symptoms were identified using latent class growth-curve analysis. Multinomial logistic models were used to relate midlife RTLs to the probabilities of assignment to subsequent depressive symptom trajectory groups. RESULTS We identified four depressive symptom trajectory groups: minimal depressive symptoms (62%), worsening depressive symptoms (14%), improving depressive symptoms (19%), and persistent-severe depressive symptoms (5%). Longer midlife RTLs were related to significantly lower odds of being in the worsening symptoms trajectory versus minimal trajectory but not to other trajectories. In comparison with being in the minimal symptoms group, the multivariable-adjusted odds ratio of being in the worsening depressive symptoms group was 0.78 (95% confidence interval, 0.62-0.97; p = 0.02), for every standard deviation increase in baseline RTL. CONCLUSIONS In this large prospective study of generally healthy women, longer telomeres at midlife were associated with significantly lower risk of a subsequent trajectory of worsening mood symptoms over 20 years. The results raise the possibility of telomere shortening as a novel contributing factor to late-life depression.
Collapse
Affiliation(s)
- J. Cai Gillis
- Harvard T.H. Chan School of Public Health, 677 Huntington Ave, Boston, MA 02115, USA,,Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, 181 Longwood Avenue, Boston, MA, 02115, USA
| | - Shun-Chiao Chang
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, 181 Longwood Avenue, Boston, MA, 02115, USA
| | - Wei Wang
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, 181 Longwood Avenue, Boston, MA, 02115, USA
| | - Naomi M. Simon
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, MA, 02114, USA,Department of Psychiatry, NYU School of Medicine, One Park Avenue, New York NY 10016
| | - Sharon-Lise Normand
- Harvard T.H. Chan School of Public Health, 677 Huntington Ave, Boston, MA 02115, USA,,Department of Health Care Policy, Harvard Medical School, 180 Longwood Avenue, Boston, MA, 02115, USA
| | - Bernard A. Rosner
- Harvard T.H. Chan School of Public Health, 677 Huntington Ave, Boston, MA 02115, USA,,Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, 181 Longwood Avenue, Boston, MA, 02115, USA
| | - Deborah Blacker
- Harvard T.H. Chan School of Public Health, 677 Huntington Ave, Boston, MA 02115, USA
| | - Immaculata DeVivo
- Harvard T.H. Chan School of Public Health, 677 Huntington Ave, Boston, MA 02115, USA
| | - Olivia I. Okereke
- Harvard T.H. Chan School of Public Health, 677 Huntington Ave, Boston, MA 02115, USA,,Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, 181 Longwood Avenue, Boston, MA, 02115, USA,,Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, MA, 02114, USA
| |
Collapse
|
50
|
Anitha A, Thanseem I, Vasu MM, Viswambharan V, Poovathinal SA. Telomeres in neurological disorders. Adv Clin Chem 2019; 90:81-132. [PMID: 31122612 DOI: 10.1016/bs.acc.2019.01.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Ever since their discovery, the telomeres and the telomerase have been topics of intensive research, first as a mechanism of cellular aging and later as an indicator of health and diseases in humans. By protecting the chromosome ends, the telomeres play a vital role in preserving the information in our genome. Telomeres shorten with age and the rate of telomere erosion provides insight into the proliferation history of cells. The pace of telomere attrition is known to increase at the onset of several pathological conditions. Telomere shortening has been emerging as a potential contributor in the pathogenesis of several neurological disorders including autism spectrum disorders (ASD), schizophrenia, Alzheimer's disease (AD), Parkinson's disease (PD) and depression. The rate of telomere attrition in the brain is slower than that of other tissues owing to the low rate of cell proliferation in brain. Telomere maintenance is crucial for the functioning of stem cells in brain. Taking together the studies on telomere attrition in various neurological disorders, an association between telomere shortening and disease status has been demonstrated in schizophrenia, AD and depression, in spite of a few negative reports. But, studies in ASD and PD have failed to produce conclusive results. The cause-effect relationship between TL and neurological disorders is yet to be elucidated. The factors responsible for telomere erosion, which have also been implicated in the pathogenesis of neurological disorders, need to be explored in detail. Telomerase activation is now being considered as a potential therapeutic strategy for neurological disorders.
Collapse
Affiliation(s)
- Ayyappan Anitha
- Institute for Communicative and Cognitive Neurosciences (ICCONS), Palakkad, Kerala, India.
| | - Ismail Thanseem
- Institute for Communicative and Cognitive Neurosciences (ICCONS), Palakkad, Kerala, India
| | - Mahesh Mundalil Vasu
- Institute for Communicative and Cognitive Neurosciences (ICCONS), Palakkad, Kerala, India
| | - Vijitha Viswambharan
- Institute for Communicative and Cognitive Neurosciences (ICCONS), Palakkad, Kerala, India
| | - Suresh A Poovathinal
- Institute for Communicative and Cognitive Neurosciences (ICCONS), Palakkad, Kerala, India
| |
Collapse
|