1
|
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: 7] [Impact Index Per Article: 7.0] [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
|
2
|
Manzanera Esteve IV, Wang F, Reed JL, Qi HX, Thayer W, Gore JC, Chen LM. Model-based parcellation of diffusion MRI of injured spinal cord predicts hand use impairment and recovery in squirrel monkeys. Behav Brain Res 2024; 459:114808. [PMID: 38081518 PMCID: PMC10865381 DOI: 10.1016/j.bbr.2023.114808] [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: 07/26/2023] [Revised: 11/30/2023] [Accepted: 12/07/2023] [Indexed: 12/17/2023]
Abstract
A mathematical model-based parcellation of magnetic resonance diffusion tensor images (DTI) has been developed to quantify progressive changes in three types of tissues - grey (GM), white matter (WM), and damaged spinal cord tissue, along with behavioral assessments over a 6 month period following targeted spinal cord injuries (SCI) in monkeys. Sigmoid Gompertz function based fittings of DTI metrics provide early indicators that correlate with, and predict, recovery of hand grasping behavior. Our three tissue pool model provided unbiased, data-driven segmentation of spinal cord images and identified DTI metrics that can serve as reliable biomarkers of severity of spinal cord injuries and predictors of behavioral outcomes.
Collapse
Affiliation(s)
- Isaac V Manzanera Esteve
- Department of Plastic Surgery, Vanderbilt University Medical Center, Nashville, TN, USA; Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Feng Wang
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jamie L Reed
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Hui Xin Qi
- Department of Psychology, Vanderbilt University, Nashville, TN, USA
| | - Wesley Thayer
- Department of Plastic Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - John C Gore
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Li Min Chen
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
3
|
Mechanosensing and the Hippo Pathway in Microglia: A Potential Link to Alzheimer's Disease Pathogenesis? Cells 2021; 10:cells10113144. [PMID: 34831369 PMCID: PMC8622675 DOI: 10.3390/cells10113144] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 10/27/2021] [Accepted: 10/29/2021] [Indexed: 01/01/2023] Open
Abstract
The activation of microglia, the inflammatory cells of the central nervous system (CNS), has been linked to the pathogenesis of Alzheimer’s disease and other neurodegenerative diseases. How microglia sense the changing brain environment, in order to respond appropriately, is still being elucidated. Microglia are able to sense and respond to the mechanical properties of their microenvironment, and the physical and molecular pathways underlying this mechanosensing/mechanotransduction in microglia have recently been investigated. The Hippo pathway functions through mechanosensing and subsequent protein kinase cascades, and is critical for neuronal development and many other cellular processes. In this review, we examine evidence for the potential involvement of Hippo pathway components specifically in microglia in the pathogenesis of Alzheimer’s disease. We suggest that the Hippo pathway is worth investigating as a mechanosensing pathway in microglia, and could be one potential therapeutic target pathway for preventing microglial-induced neurodegeneration in AD.
Collapse
|
4
|
Pang N, Huang X, Zhou H, Xia X, Liu X, Wang Y, Meng W, Bian T, Meng L, Xu L, Niu L. Transcranial Ultrasound Stimulation of Hypothalamus in Aging Mice. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2021; 68:29-37. [PMID: 31985418 DOI: 10.1109/tuffc.2020.2968479] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The hypothalamus plays an important role in the control of aging. Transcranial ultrasound stimulation (TUS) has been reported as a noninvasive method of neuromodulation. However, the effect of TUS of the hypothalamus on aging remains unclear. Therefore, the aim of this study is to verify whether TUS of the hypothalamus could affect the behaviors of aging mice and the expression level of apoptosis factors and inflammatory cytokines. TUS was delivered to the hypothalamus of mice ( n = 44 ) for 14 days (15 min/day) at a fundamental frequency of 1 MHz, pulse repetition frequency of 1 kHz (US1) or 10 Hz (US2), duty cycle of 10%, and acoustic pressure of 0.13 MPa. The effect of TUS on aging was evaluated by the behavioral tests or Western blotting in different stages. The behavioral results showed that mice in the US2 group improved their movement and learning. In addition, there was a significant improvement in the grip strength after TUS in the second behavioral tests (Sham: 0.0351 ± 0.0020 N/g; US1: 0.0340 ± 0.0023 N/g; US2: 0.0425 ± 0.0029 N/g, p = 0.034 ). Furthermore, the level of inflammation (TNF- α : Sham: 0.69 ± 0.084; US1: 0.39 ± 0.054; US2: 0.49 ± 0.1, p = 0.021 ) and apoptosis (Bax: Sham: 0.47 ± 0.049; US1: 0.42 ± 0.054; US2: 0.18 ± 0.055, p = 0.001 ) was significantly reduced after TUS in this stage. We did not see a long-lasting effect of TUS in the third behavioral tests. In addition, we found that TUS is safe according to hematoxylin and eosin (HE) staining. In conclusion, TUS could effectively modulate the hypothalamus, which may provide a new method for controlling aging.
Collapse
|
5
|
Piekarz KM, Bhaskaran S, Sataranatarajan K, Street K, Premkumar P, Saunders D, Zalles M, Gulej R, Khademi S, Laurin J, Peelor R, Miller BF, Towner R, Van Remmen H. Molecular changes associated with spinal cord aging. GeroScience 2020; 42:765-784. [PMID: 32144690 DOI: 10.1007/s11357-020-00172-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 02/21/2020] [Indexed: 12/25/2022] Open
Abstract
Age-related muscle weakness and loss of muscle mass (sarcopenia) is a universal problem in the elderly. Our previous studies indicate that alpha motor neurons (α-MNs) play a critical role in this process. The goal of the current study is to uncover changes in the aging spinal cord that contribute to loss of innervation and the downstream degenerative processes that occur in skeletal muscle. The number of α-MNs is decreased in the spinal cord of wildtype mice during aging, beginning in middle age and reaching a 41% loss by 27 months of age. There is evidence for age-related loss of myelin and mild inflammation, including astrocyte and microglia activation and an increase in levels of sICAM-1. We identified changes in metabolites consistent with compromised neuronal viability, such as reduced levels of N-acetyl-aspartate. Cleaved caspase-3 is more abundant in spinal cord from old mice, suggesting that apoptosis contributes to neuronal loss. RNA-seq analysis revealed changes in the expression of a number of genes in spinal cord from old mice, in particular genes encoding extracellular matrix components (ECM) and a 172-fold increase in MMP-12 expression. Furthermore, blood-spinal cord barrier (BSCB) permeability is increased in old mice, which may contribute to alterations in spinal cord homeostasis and exacerbate neuronal distress. Together, these data show for the first time that the spinal cord undergoes significant changes during aging, including progressive α-MNs loss that is associated with low-grade inflammation, apoptosis, changes in ECM, myelination, and vascular permeability.
Collapse
Affiliation(s)
- Katarzyna M Piekarz
- Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73117, USA.,Program in Aging and Metabolism, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
| | - Shylesh Bhaskaran
- Program in Aging and Metabolism, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
| | | | - Kaitlyn Street
- Program in Aging and Metabolism, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
| | - Pavithra Premkumar
- Program in Aging and Metabolism, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
| | - Debra Saunders
- Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
| | - Michelle Zalles
- Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73117, USA.,Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
| | - Rafal Gulej
- Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
| | - Shadi Khademi
- Program in Aging and Metabolism, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
| | - Jaime Laurin
- Program in Aging and Metabolism, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
| | - Rick Peelor
- Program in Aging and Metabolism, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
| | - Benjamin F Miller
- Program in Aging and Metabolism, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
| | - Rheal Towner
- Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73117, USA.,Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
| | - Holly Van Remmen
- Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73117, USA. .,Program in Aging and Metabolism, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA. .,Oklahoma City VA Medical Center, Oklahoma City, OK, 73104, USA.
| |
Collapse
|
6
|
Kolos EA, Korzhevskii DE. Spinal Cord Microglia in Health and Disease. Acta Naturae 2020; 12:4-17. [PMID: 32477594 PMCID: PMC7245960 DOI: 10.32607/actanaturae.10934] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 11/09/2019] [Indexed: 12/11/2022] Open
Abstract
The review summarizes data of recent experimental studies on spinal microglia, the least explored cells of the spinal cord. It focuses on the origin and function of microglia in mammalian spinal cord embryogenesis. The main approaches to the classification of microgliocytes based on their structure, function, and immunophenotypic characteristics are analyzed. We discuss the results of studies conducted on experimental models of spinal cord diseases such as multiple sclerosis, amyotrophic lateral sclerosis, systemic inflammation, and some others, with special emphasis on the key role of microglia in the pathogenesis of these diseases. The review highlights the need to detect the new microglia-specific marker proteins expressed at all stages of ontogeny. New sensitive and selective microglial markers are necessary in order to improve identification of spinal cord microgliocytes in normal and pathological conditions. Possible morphometric methods to assess the functional activity of microglial cells are presented.
Collapse
Affiliation(s)
- E. A. Kolos
- Institute of Experimental Medicine, St. Petersburg, 197376 Russia
| | | |
Collapse
|