1
|
Chen Y, Man-Tak Chu J, Liu JX, Duan YJ, Liang ZK, Zou X, Wei M, Xin W, Xu T, Tin-Chun Wong G, Feng X. Double negative T cells promote surgery-induced neuroinflammation, microglial engulfment and cognitive dysfunction via the IL-17/CEBPβ/C3 pathway in adult mice. Brain Behav Immun 2024:S0889-1591(24)00670-6. [PMID: 39491565 DOI: 10.1016/j.bbi.2024.10.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 10/11/2024] [Accepted: 10/26/2024] [Indexed: 11/05/2024] Open
Abstract
CD3(+) CD4(-) CD8(-) double negative T cells (DNTs) manifest themselves in autoimmune diseases and associated inflammation. In the central nervous system, the increased presence of DNTs is associated with the progression of neurological conditions and brain injury. Active DNTs that produce IL-17 have been regarded as a pro-inflammatory phenotype. The IL-17 signaling pathway mediates neuroinflammatory responses by inducing glial activation and producing inflammatory factors. Neuroinflammation is considered integral to the pathogenesis of perioperative neurocognitive disorders (PNDs), commonly developed after surgery in susceptible patients. We and others have demonstrated a significant role for complement C3 in surgery-induced neuroinflammation and cognitive impairment but the regulatory mechanisms for this remain unexplored. We hypothesized that surgery induces DNT infiltration into the CNS that in turn upregulate complement C3 expression and this causes changes that contribute to cognitive impairment. Using an adult murine abdominal surgery model, we investigated perioperative changes in cognitive performance, quantifying the presence of T cell subsets and phenotype, IL-17 signaling pathway activation, glial cell activation and C3 expression in the brain. Postoperative IL-17 specific inhibitor GSK2981278 administration or preoperatively conditional CEBPβ knock-down by AAV9 viral vector were then applied to evaluate the effect of inhibiting IL-17 signaling pathway on postoperative C3 expression and cognitive performance. The results showed an increased hippocampus infiltration of DNTs with augmented IL-17 production, along with C3 upregulation and cognitive impairment. Both inhibition of IL-17 or knock-down of CEBPβ significantly suppressed C3 expression, synaptic engulfment by microglia and attenuated cognitive impairment. These findings indicate that DNTs promote postoperative neuroinflammation and cognitive impairment via the IL-17/CEBPβ/C3 pathway and targeting this IL-17 axis could be a potential therapeutic strategy to ameliorate postoperative neuroinflammation and cognitive impairment.
Collapse
Affiliation(s)
- Ying Chen
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - John Man-Tak Chu
- Department of Anaesthesiology, LKS Faculty of Medicine, Queen Mary Hospital, The University of Hong Kong, Room K424, 4Th Floor, K Block, 102 Pokfulam Road, Pokfulam, Hong Kong SAR, China
| | - Jia-Xin Liu
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Yu-Juan Duan
- Neuroscience Program, Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Zheng-Kai Liang
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Xin Zou
- Neuroscience Program, Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Ming Wei
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Wenjun Xin
- Neuroscience Program, Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Ting Xu
- Neuroscience Program, Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Gordon Tin-Chun Wong
- Department of Anaesthesiology, LKS Faculty of Medicine, Queen Mary Hospital, The University of Hong Kong, Room K424, 4Th Floor, K Block, 102 Pokfulam Road, Pokfulam, Hong Kong SAR, China
| | - Xia Feng
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China.
| |
Collapse
|
2
|
Becher B, Derfuss T, Liblau R. Targeting cytokine networks in neuroinflammatory diseases. Nat Rev Drug Discov 2024; 23:862-879. [PMID: 39261632 DOI: 10.1038/s41573-024-01026-y] [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: 08/02/2024] [Indexed: 09/13/2024]
Abstract
In neuroinflammatory diseases, systemic (blood-borne) leukocytes invade the central nervous system (CNS) and lead to tissue damage. A causal relationship between neuroinflammatory diseases and dysregulated cytokine networks is well established across several preclinical models. Cytokine dysregulation is also observed as an inadvertent effect of cancer immunotherapy, where it often leads to neuroinflammation. Neuroinflammatory diseases can be separated into those in which a pathogen is at the centre of the immune response and those of largely unknown aetiology. Here, we discuss the pathophysiology, cytokine networks and therapeutic landscape of 'sterile' neuroinflammatory diseases such as multiple sclerosis (MS), neuromyelitis optica spectrum disorder (NMOSD), neurosarcoidosis and immune effector cell-associated neurotoxicity syndrome (ICANS) triggered by cancer immunotherapy. Despite successes in targeting cytokine networks in preclinical models of neuroinflammation, the clinical translation of targeting cytokines and their receptors has shown mixed and often paradoxical responses.
Collapse
Affiliation(s)
- Burkhard Becher
- Institute of experimental Immunology, University of Zurich, Zurich, Switzerland.
| | - Tobias Derfuss
- Department of Neurology and Biomedicine, Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel, University of Basel, Basel, Switzerland.
| | - Roland Liblau
- Institute for inflammatory and infectious diseases, INSERM UMR1291 - CNRS UMR505, Toulouse, France.
| |
Collapse
|
3
|
Holland C, Dravecz N, Owens L, Benedetto A, Dias I, Gow A, Broughton S. Understanding exogenous factors and biological mechanisms for cognitive frailty: A multidisciplinary scoping review. Ageing Res Rev 2024; 101:102461. [PMID: 39278273 DOI: 10.1016/j.arr.2024.102461] [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: 12/21/2023] [Revised: 07/15/2024] [Accepted: 08/15/2024] [Indexed: 09/18/2024]
Abstract
Cognitive frailty (CF) is the conjunction of cognitive impairment without dementia and physical frailty. While predictors of each element are well-researched, mechanisms of their co-occurrence have not been integrated, particularly in terms of relationships between social, psychological, and biological factors. This interdisciplinary scoping review set out to categorise a heterogenous multidisciplinary literature to identify potential pathways and mechanisms of CF, and research gaps. Studies were included if they used the definition of CF OR focused on conjunction of cognitive impairment and frailty (by any measure), AND excluded studies on specific disease populations, interventions, epidemiology or prediction of mortality. Searches used Web of Science, PubMed and Science Direct. Search terms included "cognitive frailty" OR (("cognitive decline" OR "cognitive impairment") AND (frail*)), with terms to elicit mechanisms, predictors, causes, pathways and risk factors. To ensure inclusion of animal and cell models, keywords such as "behavioural" or "cognitive decline" or "senescence", were added. 206 papers were included. Descriptive analysis provided high-level categorisation of determinants from social and environmental through psychological to biological. Patterns distinguishing CF from Alzheimer's disease were identified and social and psychological moderators and mediators of underlying biological and physiological changes and of trajectories of CF development were suggested as foci for further research.
Collapse
Affiliation(s)
- Carol Holland
- Division of Health Research, Health Innovation One, Sir John Fisher Drive, Lancaster University, Lancaster LA1 4YW, UK.
| | - Nikolett Dravecz
- Division of Health Research, Health Innovation One, Sir John Fisher Drive, Lancaster University, Lancaster LA1 4YW, UK.
| | - Lauren Owens
- Division of Biomedical and Life Sciences, Furness College, Lancaster University, LA1 4YG, UK.
| | - Alexandre Benedetto
- Division of Biomedical and Life Sciences, Furness College, Lancaster University, LA1 4YG, UK.
| | - Irundika Dias
- Aston University Medical School, Aston University, Birmingham B4 7ET, UK.
| | - Alan Gow
- Centre for Applied Behavioural Sciences, Department of Psychology, School of Social Sciences, Heriot-Watt University, Edinburgh EH14 4AS, UK.
| | - Susan Broughton
- Division of Biomedical and Life Sciences, Furness College, Lancaster University, LA1 4YG, UK.
| |
Collapse
|
4
|
Fan X, Chen H, He W, Zhang J. Emerging microglial biology highlights potential therapeutic targets for Alzheimer's disease. Ageing Res Rev 2024; 101:102471. [PMID: 39218078 DOI: 10.1016/j.arr.2024.102471] [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: 07/05/2024] [Revised: 08/21/2024] [Accepted: 08/24/2024] [Indexed: 09/04/2024]
Abstract
Alzheimer's disease is a chronic degenerative disease of the central nervous system, which primarily affects elderly people and accounts for 70-80 % of dementia cases. The current prevailing amyloid cascade hypothesis suggests that Alzheimer's disease begins with the deposition of amyloid β (Aβ) in the brain. Major therapeutic strategies target Aβ production, aggregation, and clearance, although many clinical trials have shown that these therapeutic strategies are not sufficient to completely improve cognitive deficits in AD patients. Recent genome-wide association studies have identified that multiple important regulators are the most significant genetic risk factors for Alzheimer's disease, especially in the innate immune pathways. These genetic risk factors suggest a critical role for microglia, highlighting their therapeutic potential in treating neurodegenerative diseases. In this review, we discuss how these recently documented AD risk genes affect microglial function and AD pathology and how they can be further targeted to regulate microglial states and slow AD progression, especially the highly anticipated APOE and TREM2 targets. We focused on recent findings that modulation of innate and adaptive neuroimmune microenvironment crosstalk reverses cognitive deficits in AD patients. We also considered novel strategies for microglia in AD patients.
Collapse
Affiliation(s)
- Xi Fan
- Department of Immunology, CAMS Key laboratory T cell and Cancer Immunotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, State Key Laboratory of Common Mechanism Research for Major Diseases, Beijing 100005, China; Research Unit of Diagnosis and Treatment of Chronic Nasal Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Hui Chen
- Department of Immunology, CAMS Key laboratory T cell and Cancer Immunotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, State Key Laboratory of Common Mechanism Research for Major Diseases, Beijing 100005, China.
| | - Wei He
- Department of Immunology, CAMS Key laboratory T cell and Cancer Immunotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, State Key Laboratory of Common Mechanism Research for Major Diseases, Beijing 100005, China.
| | - Jianmin Zhang
- Department of Immunology, CAMS Key laboratory T cell and Cancer Immunotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, State Key Laboratory of Common Mechanism Research for Major Diseases, Beijing 100005, China; Changzhou Xitaihu Institute for Frontier Technology of Cell Therapy, Changzhou, Jiangsu 213000, China.
| |
Collapse
|
5
|
Wang X, Campbell B, Bodogai M, McDevitt RA, Patrikeev A, Gusev F, Ragonnaud E, Kumaraswami K, Shirenova S, Vardy K, Alameh MG, Weissman D, Ishikawa-Ankerhold H, Okun E, Rogaev E, Biragyn A. CD8 + T cells exacerbate AD-like symptoms in mouse model of amyloidosis. Brain Behav Immun 2024; 122:444-455. [PMID: 39191349 PMCID: PMC11409913 DOI: 10.1016/j.bbi.2024.08.045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 08/01/2024] [Accepted: 08/22/2024] [Indexed: 08/29/2024] Open
Abstract
Alzheimer's disease (AD) is linked to toxic Aβ plaques in the brain and activation of innate responses. Recent findings however suggest that the disease may also depend on the adaptive immunity, as B cells exacerbate and CD8+ T cells limit AD-like pathology in mouse models of amyloidosis. Here, by artificially blocking or augmenting CD8+ T cells in the brain of 5xFAD mice, we provide evidence that AD-like pathology is promoted by pathogenic, proinflammatory cytokines and exhaustion markers expressing CXCR6+ CD39+CD73+/- CD8+ TRM-like cells. The CD8+ T cells appear to act by targeting disease associated microglia (DAM), as we find them in tight complexes with microglia around Aβ plaques in the brain of mice and humans with AD. We also report that these CD8+ T cells are induced by B cells in the periphery, further underscoring the pathogenic importance of the adaptive immunity in AD. We propose that CD8+ T cells and B cells should be considered as therapeutic targets for control of AD, as their ablation at the onset of AD is sufficient to decrease CD8+ T cells in the brain and block the amyloidosis-linked neurodegeneration.
Collapse
Affiliation(s)
- Xin Wang
- Immunoregulation Section, Laboratory of Molecular Biology and Immunolgy, USA
| | - Britney Campbell
- Immunoregulation Section, Laboratory of Molecular Biology and Immunolgy, USA
| | - Monica Bodogai
- Immunoregulation Section, Laboratory of Molecular Biology and Immunolgy, USA
| | - Ross A McDevitt
- Mouse Phenotyping Unit, Comparative Medicine Section, National Institute on Aging, Baltimore, MD, USA
| | - Anton Patrikeev
- Department of Psychiatry, University of Massachusetts Medical School, Worcester, MA, USA
| | - Fedor Gusev
- Department of Psychiatry, University of Massachusetts Medical School, Worcester, MA, USA
| | - Emeline Ragonnaud
- Immunoregulation Section, Laboratory of Molecular Biology and Immunolgy, USA
| | - Konda Kumaraswami
- Immunoregulation Section, Laboratory of Molecular Biology and Immunolgy, USA
| | - Sophie Shirenova
- The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Israel; The Mina and Everard Goodman Faculty of Life Sciences, Israel; The Paul Feder Laboratory on Alzheimer's Disease Research, Bar-Ilan University, Ramat Gan, Israel
| | - Karin Vardy
- The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Israel; The Mina and Everard Goodman Faculty of Life Sciences, Israel; The Paul Feder Laboratory on Alzheimer's Disease Research, Bar-Ilan University, Ramat Gan, Israel
| | | | - Drew Weissman
- Institute of RNA Innovation, University of Pennsylvania, Philadelphia, PA, USA
| | - Hellen Ishikawa-Ankerhold
- Department of Medicine I, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany; Institute of Surgical Research at the Walter Brendel Centre of Experimental Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Eitan Okun
- The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Israel; The Mina and Everard Goodman Faculty of Life Sciences, Israel; The Paul Feder Laboratory on Alzheimer's Disease Research, Bar-Ilan University, Ramat Gan, Israel
| | - Evgeny Rogaev
- Department of Psychiatry, University of Massachusetts Medical School, Worcester, MA, USA
| | - Arya Biragyn
- Immunoregulation Section, Laboratory of Molecular Biology and Immunolgy, USA.
| |
Collapse
|
6
|
Huang Y, Zhu S, Yao S, Zhai H, Liu C, Han JDJ. Unraveling aging from transcriptomics. Trends Genet 2024:S0168-9525(24)00214-2. [PMID: 39424502 DOI: 10.1016/j.tig.2024.09.006] [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: 07/31/2024] [Revised: 09/19/2024] [Accepted: 09/19/2024] [Indexed: 10/21/2024]
Abstract
Research into aging constitutes a pivotal endeavor aimed at elucidating the underlying biological mechanisms governing aging and age-associated diseases, as well as promoting healthy longevity. Recent advances in transcriptomic technologies, such as bulk RNA sequencing (RNA-seq), single-cell transcriptomics, and spatial transcriptomics, have revolutionized our ability to study aging at unprecedented resolution and scale. These technologies present novel opportunities for the discovery of biomarkers, elucidation of molecular pathways, and development of targeted therapeutic strategies for age-related disorders. This review surveys recent breakthroughs in different types of transcripts on aging, such as mRNA, long noncoding (lnc)RNA, tRNA, and miRNA, highlighting key findings and discussing their potential implications for future studies in this field.
Collapse
Affiliation(s)
- Yuanfang Huang
- Peking-Tsinghua Center for Life Sciences, Center for Quantitative Biology (CQB), Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Shouxuan Zhu
- Peking-Tsinghua Center for Life Sciences, Center for Quantitative Biology (CQB), Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Shuai Yao
- Peking-Tsinghua Center for Life Sciences, Center for Quantitative Biology (CQB), Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Haotian Zhai
- Peking-Tsinghua Center for Life Sciences, Center for Quantitative Biology (CQB), Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Chenyang Liu
- Peking-Tsinghua Center for Life Sciences, Center for Quantitative Biology (CQB), Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Jing-Dong J Han
- Peking-Tsinghua Center for Life Sciences, Center for Quantitative Biology (CQB), Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China; Peking University Chengdu Academy for Advanced Interdisciplinary Biotechnologies, Chengdu, China.
| |
Collapse
|
7
|
Gorelov R, Hochedlinger K. A cellular identity crisis? Plasticity changes during aging and rejuvenation. Genes Dev 2024; 38:823-842. [PMID: 39293862 DOI: 10.1101/gad.351728.124] [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] [Indexed: 09/20/2024]
Abstract
Cellular plasticity in adult multicellular organisms is a protective mechanism that allows certain tissues to regenerate in response to injury. Considering that aging involves exposure to repeated injuries over a lifetime, it is conceivable that cell identity itself is more malleable-and potentially erroneous-with age. In this review, we summarize and critically discuss the available evidence that cells undergo age-related shifts in identity, with an emphasis on those that contribute to age-associated pathologies, including neurodegeneration and cancer. Specifically, we focus on reported instances of programs associated with dedifferentiation, biased differentiation, acquisition of features from alternative lineages, and entry into a preneoplastic state. As some of the most promising approaches to rejuvenate cells reportedly also elicit transient changes to cell identity, we further discuss whether cell state change and rejuvenation can be uncoupled to yield more tractable therapeutic strategies.
Collapse
Affiliation(s)
- Rebecca Gorelov
- Department of Molecular Biology, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
- Cancer Center, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts 02138, USA
- Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Konrad Hochedlinger
- Department of Molecular Biology, Massachusetts General Hospital, Boston, Massachusetts 02114, USA;
- Cancer Center, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts 02138, USA
- Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
8
|
Ruetz TJ, Pogson AN, Kashiwagi CM, Gagnon SD, Morton B, Sun ED, Na J, Yeo RW, Leeman DS, Morgens DW, Tsui CK, Li A, Bassik MC, Brunet A. CRISPR-Cas9 screens reveal regulators of ageing in neural stem cells. Nature 2024; 634:1150-1159. [PMID: 39358505 PMCID: PMC11525198 DOI: 10.1038/s41586-024-07972-2] [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: 11/23/2021] [Accepted: 08/20/2024] [Indexed: 10/04/2024]
Abstract
Ageing impairs the ability of neural stem cells (NSCs) to transition from quiescence to proliferation in the adult mammalian brain. Functional decline of NSCs results in the decreased production of new neurons and defective regeneration following injury during ageing1-4. Several genetic interventions have been found to ameliorate old brain function5-8, but systematic functional testing of genes in old NSCs-and more generally in old cells-has not been done. Here we develop in vitro and in vivo high-throughput CRISPR-Cas9 screening platforms to systematically uncover gene knockouts that boost NSC activation in old mice. Our genome-wide screens in primary cultures of young and old NSCs uncovered more than 300 gene knockouts that specifically restore the activation of old NSCs. The top gene knockouts are involved in cilium organization and glucose import. We also establish a scalable CRISPR-Cas9 screening platform in vivo, which identified 24 gene knockouts that boost NSC activation and the production of new neurons in old brains. Notably, the knockout of Slc2a4, which encodes the GLUT4 glucose transporter, is a top intervention that improves the function of old NSCs. Glucose uptake increases in NSCs during ageing, and transient glucose starvation restores the ability of old NSCs to activate. Thus, an increase in glucose uptake may contribute to the decline in NSC activation with age. Our work provides scalable platforms to systematically identify genetic interventions that boost the function of old NSCs, including in vivo, with important implications for countering regenerative decline during ageing.
Collapse
Affiliation(s)
- Tyson J Ruetz
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Angela N Pogson
- Department of Genetics, Stanford University, Stanford, CA, USA
- Developmental Biology Graduate Program, Stanford University, Stanford, CA, USA
| | | | | | - Bhek Morton
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Eric D Sun
- Department of Genetics, Stanford University, Stanford, CA, USA
- Biomedical Informatics Graduate Program, Stanford University, Stanford, CA, USA
| | - Jeeyoon Na
- Department of Genetics, Stanford University, Stanford, CA, USA
- Stem Cell Biology & Regenerative Medicine Graduate Program, Stanford University, Stanford, CA, USA
| | - Robin W Yeo
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Dena S Leeman
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - David W Morgens
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - C Kimberly Tsui
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Amy Li
- Department of Genetics, Stanford University, Stanford, CA, USA
| | | | - Anne Brunet
- Department of Genetics, Stanford University, Stanford, CA, USA.
- Glenn Center for the Biology of Aging, Stanford University, Stanford, CA, USA.
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
9
|
Chaker Z, Makarouni E, Doetsch F. The Organism as the Niche: Physiological States Crack the Code of Adult Neural Stem Cell Heterogeneity. Annu Rev Cell Dev Biol 2024; 40:381-406. [PMID: 38985883 DOI: 10.1146/annurev-cellbio-120320-040213] [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] [Indexed: 07/12/2024]
Abstract
Neural stem cells (NSCs) persist in the adult mammalian brain and are able to give rise to new neurons and glia throughout life. The largest stem cell niche in the adult mouse brain is the ventricular-subventricular zone (V-SVZ) lining the lateral ventricles. Adult NSCs in the V-SVZ coexist in quiescent and actively proliferating states, and they exhibit a regionalized molecular identity. The importance of such spatial diversity is just emerging, as depending on their position within the niche, adult NSCs give rise to distinct subtypes of olfactory bulb interneurons and different types of glia. However, the functional relevance of stem cell heterogeneity in the V-SVZ is still poorly understood. Here, we put into perspective findings highlighting the importance of adult NSC diversity for brain plasticity, and how the body signals to brain stem cells in different physiological states to regulate their behavior.
Collapse
Affiliation(s)
- Zayna Chaker
- Biozentrum, University of Basel, Basel, Switzerland; , ,
| | | | - Fiona Doetsch
- Biozentrum, University of Basel, Basel, Switzerland; , ,
| |
Collapse
|
10
|
Krsek A, Ostojic L, Zivalj D, Baticic L. Navigating the Neuroimmunomodulation Frontier: Pioneering Approaches and Promising Horizons-A Comprehensive Review. Int J Mol Sci 2024; 25:9695. [PMID: 39273641 PMCID: PMC11396210 DOI: 10.3390/ijms25179695] [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: 07/31/2024] [Revised: 08/30/2024] [Accepted: 09/03/2024] [Indexed: 09/15/2024] Open
Abstract
The research in neuroimmunomodulation aims to shed light on the complex relationships that exist between the immune and neurological systems and how they affect the human body. This multidisciplinary field focuses on the way immune responses are influenced by brain activity and how neural function is impacted by immunological signaling. This provides important insights into a range of medical disorders. Targeting both brain and immunological pathways, neuroimmunomodulatory approaches are used in clinical pain management to address chronic pain. Pharmacological therapies aim to modulate neuroimmune interactions and reduce inflammation. Furthermore, bioelectronic techniques like vagus nerve stimulation offer non-invasive control of these systems, while neuromodulation techniques like transcranial magnetic stimulation modify immunological and neuronal responses to reduce pain. Within the context of aging, neuroimmunomodulation analyzes the ways in which immunological and neurological alterations brought on by aging contribute to cognitive decline and neurodegenerative illnesses. Restoring neuroimmune homeostasis through strategies shows promise in reducing age-related cognitive decline. Research into mood disorders focuses on how immunological dysregulation relates to illnesses including anxiety and depression. Immune system fluctuations are increasingly recognized for their impact on brain function, leading to novel treatments that target these interactions. This review emphasizes how interdisciplinary cooperation and continuous research are necessary to better understand the complex relationship between the neurological and immune systems.
Collapse
Affiliation(s)
- Antea Krsek
- Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Leona Ostojic
- Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Dorotea Zivalj
- Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Lara Baticic
- Department of Medical Chemistry, Biochemistry and Clinical Chemistry, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| |
Collapse
|
11
|
Feng H, Li J, Wang H, Wei Z, Feng S. Senescence- and Immunity-Related Changes in the Central Nervous System: A Comprehensive Review. Aging Dis 2024:AD.2024.0755. [PMID: 39325939 DOI: 10.14336/ad.2024.0755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 08/26/2024] [Indexed: 09/28/2024] Open
Abstract
Senescence is a cellular state characterized by an irreversible halt in the cell cycle, accompanied by alterations in cell morphology, function, and secretion. Senescent cells release a plethora of inflammatory and growth factors, extracellular matrix proteins, and other bioactive substances, collectively known as the senescence-associated secretory phenotype (SASP). These excreted substances serve as crucial mediators of senescent tissues, while the secretion of SASP by senescent neurons and glial cells in the central nervous system modulates the activity of immune cells. Senescent immune cells also influence the physiological activities of various cells in the central nervous system. Further, the interaction between cellular senescence and immune regulation collectively affects the physiological and pathological processes of the central nervous system. Herein, we explore the role of senescence in the physiological and pathological processes underlying embryonic development, aging, degeneration, and injury of the central nervous system, through the immune response. Further, we elucidate the role of senescence in the physiological and pathological processes of the central nervous system, proposing a new theoretical foundation for treating central nervous system diseases.
Collapse
Affiliation(s)
- Haiwen Feng
- Tianjin Key Laboratory of Spine and Spinal Cord, International Science and Technology Cooperation Base of Spinal Cord Injury, Department of Orthopedics, International Chinese Musculoskeletal Research Society Collaborating Center for Spinal Cord Injury, Tianjin Medical University General Hospital, Tianjin 300070, China
| | - Junjin Li
- Tianjin Key Laboratory of Spine and Spinal Cord, International Science and Technology Cooperation Base of Spinal Cord Injury, Department of Orthopedics, International Chinese Musculoskeletal Research Society Collaborating Center for Spinal Cord Injury, Tianjin Medical University General Hospital, Tianjin 300070, China
| | - Hongda Wang
- Tianjin Key Laboratory of Spine and Spinal Cord, International Science and Technology Cooperation Base of Spinal Cord Injury, Department of Orthopedics, International Chinese Musculoskeletal Research Society Collaborating Center for Spinal Cord Injury, Tianjin Medical University General Hospital, Tianjin 300070, China
| | - Zhijian Wei
- Orthopedic Research Center of Shandong University and Department of Orthopedics, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Shiqing Feng
- Tianjin Key Laboratory of Spine and Spinal Cord, International Science and Technology Cooperation Base of Spinal Cord Injury, Department of Orthopedics, International Chinese Musculoskeletal Research Society Collaborating Center for Spinal Cord Injury, Tianjin Medical University General Hospital, Tianjin 300070, China
- Orthopedic Research Center of Shandong University and Department of Orthopedics, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| |
Collapse
|
12
|
Chen S, Tan Y, Tian L. Immunophenotypes in psychosis: is it a premature inflamm-aging disorder? Mol Psychiatry 2024; 29:2834-2848. [PMID: 38532012 PMCID: PMC11420084 DOI: 10.1038/s41380-024-02539-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 03/15/2024] [Accepted: 03/18/2024] [Indexed: 03/28/2024]
Abstract
Immunopsychiatric field has rapidly accumulated evidence demonstrating the involvement of both innate and adaptive immune components in psychotic disorders such as schizophrenia. Nevertheless, researchers are facing dilemmas of discrepant findings of immunophenotypes both outside and inside the brains of psychotic patients, as discovered by recent meta-analyses. These discrepancies make interpretations and interrogations on their roles in psychosis remain vague and even controversial, regarding whether certain immune cells are more activated or less so, and whether they are causal or consequential, or beneficial or harmful for psychosis. Addressing these issues for psychosis is not at all trivial, as immune cells either outside or inside the brain are an enormously heterogeneous and plastic cell population, falling into a vast range of lineages and subgroups, and functioning differently and malleably in context-dependent manners. This review aims to overview the currently known immunophenotypes of patients with psychosis, and provocatively suggest the premature immune "burnout" or inflamm-aging initiated since organ development as a potential primary mechanism behind these immunophenotypes and the pathogenesis of psychotic disorders.
Collapse
Affiliation(s)
- Song Chen
- Peking University HuiLongGuan Clinical Medical School, Beijing Huilongguan Hospital, Beijing, PR China
| | - Yunlong Tan
- Peking University HuiLongGuan Clinical Medical School, Beijing Huilongguan Hospital, Beijing, PR China
| | - Li Tian
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
13
|
Greenberg EF, Voorbach MJ, Smith A, Reuter DR, Zhuang Y, Wang JQ, Wooten DW, Asque E, Hu M, Hoft C, Duggan R, Townsend M, Orsi K, Dalecki K, Amberg W, Duggan L, Knight H, Spina JS, He Y, Marsh K, Zhao V, Ybarra S, Mollon J, Fang Y, Vasanthakumar A, Westmoreland S, Droescher M, Finnema SJ, Florian H. Navitoclax safety, tolerability, and effect on biomarkers of senescence and neurodegeneration in aged nonhuman primates. Heliyon 2024; 10:e36483. [PMID: 39253182 PMCID: PMC11382177 DOI: 10.1016/j.heliyon.2024.e36483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 08/01/2024] [Accepted: 08/16/2024] [Indexed: 09/11/2024] Open
Abstract
Alzheimer's disease (AD) is the most common global dementia and is universally fatal. Most late-stage AD disease-modifying therapies are intravenous and target amyloid beta (Aβ), with only modest effects on disease progression: there remains a high unmet need for convenient, safe, and effective therapeutics. Senescent cells (SC) and the senescence-associated secretory phenotype (SASP) drive AD pathology and increase with AD severity. Preclinical senolytic studies have shown improvements in neuroinflammation, tau, Aβ, and CNS damage; most were conducted in transgenic rodent models with uncertain human translational relevance. In this study, aged cynomolgus monkeys had significant elevation of biomarkers of senescence, SASP, and neurological damage. Intermittent treatment with the senolytic navitoclax induced modest reversible thrombocytopenia; no serious drug-related toxicity was noted. Navitoclax reduced several senescence and SASP biomarkers, with CSF concentrations sufficient for senolysis. Finally, navitoclax reduced TSPO-PET frontal cortex binding and showed trends of improvement in CSF biomarkers of neuroinflammation, neuronal damage, and synaptic dysfunction. Overall, navitoclax administration was safe and well tolerated in aged monkeys, inducing trends of biomarker changes relevant to human neurodegenerative disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Min Hu
- AbbVie Inc., North Chicago, IL, United States
| | - Carolin Hoft
- AbbVie Deutschland GmbH & Co. KG, Neuroscience Research, Knollstrasse, 67061, Ludwigshafen, Germany
| | - Ryan Duggan
- AbbVie Inc., North Chicago, IL, United States
| | - Matthew Townsend
- AbbVie, Cambridge Research Center, 200 Sidney Street, Cambridge, MA, 02139, United States
| | - Karin Orsi
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA, 01605, United States
| | | | - Willi Amberg
- AbbVie Deutschland GmbH & Co. KG, Neuroscience Research, Knollstrasse, 67061, Ludwigshafen, Germany
| | - Lori Duggan
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA, 01605, United States
| | - Heather Knight
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA, 01605, United States
| | - Joseph S Spina
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA, 01605, United States
| | - Yupeng He
- AbbVie Inc., North Chicago, IL, United States
| | | | - Vivian Zhao
- AbbVie Bay Area, 1000 Gateway Boulevard, South San Francisco, CA, 94080, United States
| | - Suzanne Ybarra
- AbbVie Bay Area, 1000 Gateway Boulevard, South San Francisco, CA, 94080, United States
| | - Jennifer Mollon
- AbbVie Deutschland GmbH & Co. KG, Statistical Sciences and Analytics, Knollstrasse, 67061, Ludwigshafen, Germany
| | - Yuni Fang
- AbbVie Bay Area, 1000 Gateway Boulevard, South San Francisco, CA, 94080, United States
| | | | - Susan Westmoreland
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA, 01605, United States
| | - Mathias Droescher
- AbbVie Deutschland GmbH & Co. KG, Neuroscience Research, Knollstrasse, 67061, Ludwigshafen, Germany
| | | | | |
Collapse
|
14
|
Ma YZ, Cao JX, Zhang YS, Su XM, Jing YH, Gao LP. T Cells Trafficking into the Brain in Aging and Alzheimer's Disease. J Neuroimmune Pharmacol 2024; 19:47. [PMID: 39180590 DOI: 10.1007/s11481-024-10147-5] [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: 10/20/2022] [Accepted: 08/05/2024] [Indexed: 08/26/2024]
Abstract
The meninges, choroid plexus (CP) and blood-brain barrier (BBB) are recognized as important gateways for peripheral immune cell trafficking into the central nervous system (CNS). Accumulation of peripheral immune cells in brain parenchyma can be observed during aging and Alzheimer's disease (AD). However, the mechanisms by which peripheral immune cells enter the CNS through these three pathways and how they interact with resident cells within the CNS to cause brain injury are not fully understood. In this paper, we review recent research on T cells recruitment in the brain during aging and AD. This review focuses on the possible pathways through which T cells infiltrate the brain, the evidence that T cells are recruited to the brain, and how infiltrating T cells interact with the resident cells in the CNS during aging and AD. Unraveling these issues will contribute to a better understanding of the mechanisms of aging and AD from the perspective of immunity, and hopefully develop new therapeutic strategies for brain aging and AD.
Collapse
Affiliation(s)
- Yue-Zhang Ma
- Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Jia-Xin Cao
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Yi-Shu Zhang
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Xiao-Mei Su
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Yu-Hong Jing
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.
| | - Li-Ping Gao
- Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.
| |
Collapse
|
15
|
Weng R, Liu J, Yu Q, Yuan H, Qiu Y, Liu H, Wang L, Mei Z, Zhu F. The disparity of platelet factor 4 and platelets in individuals of different ages. Heliyon 2024; 10:e34923. [PMID: 39145023 PMCID: PMC11320319 DOI: 10.1016/j.heliyon.2024.e34923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 07/10/2024] [Accepted: 07/18/2024] [Indexed: 08/16/2024] Open
Abstract
The aging process profoundly impacts the systemic milieu, with specific blood-borne factors playing critical roles in its regulation. Platelet Factor 4 (PF4), released by platelets, has emerged as a novel blood-borne factor that contributes to the rejuvenation of aging brains in rodents. However, the age-related disparity in PF4 levels in humans remains poorly understood. To explore the relationship between PF4 and the natural aging process in humans, we collected peripheral blood (PB) samples from young (23.40 ± 2.13 years, n = 15) and elderly (75.23 ± 4.19 years, n = 13) individuals, along with cord blood (CB) samples (n = 15). ELISA analysis revealed higher PF4 levels in platelet-rich plasma lysate from young PB compared with that from elderly PB. Consistent with this, qPCR results demonstrated the highest PF4 expression in young PB among the three groups. In addition, FACS analysis showed increased expression of CXCR3 in mononuclear cells of young PB, indicating a greater responsiveness to PF4. Finally, our RNA-sequencing analysis corroborated platelets as a sensitive element during the natural aging process, and indicated platelets play a pivotal role in antioxidant response during aging, as evidenced by significant enrichment of several age-related pathways. These findings reveal that, alongside PF4 levels, platelets undergo substantial alterations during aging. Taken together, our data identified age-related disparities in platelets and PF4-related elements during natural aging and underscored the potential of targeting platelet modulation as an intervention in the aging process.
Collapse
Affiliation(s)
- Ruopeng Weng
- Department of Obstetrics, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiaqing Liu
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Qingtan Yu
- Department of Laboratory, Qingdao Special Servicemen Recuperation Center of PLA Navy, China
| | - Haitao Yuan
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Yun Qiu
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Huicong Liu
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Lingna Wang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Zijie Mei
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Fangfang Zhu
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
16
|
Han J, Wang Y, Wei P, Lu D, Shan Y. Unveiling the hidden connection: the blood-brain barrier's role in epilepsy. Front Neurol 2024; 15:1413023. [PMID: 39206290 PMCID: PMC11349696 DOI: 10.3389/fneur.2024.1413023] [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: 04/06/2024] [Accepted: 07/18/2024] [Indexed: 09/04/2024] Open
Abstract
Epilepsy is characterized by abnormal synchronous electrical activity of neurons in the brain. The blood-brain barrier, which is mainly composed of endothelial cells, pericytes, astrocytes and other cell types and is formed by connections between a variety of cells, is the key physiological structure connecting the blood and brain tissue and is critical for maintaining the microenvironment in the brain. Physiologically, the blood-brain barrier controls the microenvironment in the brain mainly by regulating the passage of various substances. Disruption of the blood-brain barrier and increased leakage of specific substances, which ultimately leading to weakened cell junctions and abnormal regulation of ion concentrations, have been observed during the development and progression of epilepsy in both clinical studies and animal models. In addition, disruption of the blood-brain barrier increases drug resistance through interference with drug trafficking mechanisms. The changes in the blood-brain barrier in epilepsy mainly affect molecular pathways associated with angiogenesis, inflammation, and oxidative stress. Further research on biomarkers is a promising direction for the development of new therapeutic strategies.
Collapse
Affiliation(s)
| | | | | | | | - Yongzhi Shan
- Department of Neurosurgery, Xuanwu Hospital Capital Medical University, Beijing, China
| |
Collapse
|
17
|
Fritze J, Lang S, Sommarin M, Soneji S, Ahlenius H. Single-cell RNA sequencing of aging neural progenitors reveals loss of excitatory neuron potential and a population with transcriptional immune response. Front Neurosci 2024; 18:1400963. [PMID: 39184324 PMCID: PMC11341460 DOI: 10.3389/fnins.2024.1400963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 07/08/2024] [Indexed: 08/27/2024] Open
Abstract
In the adult murine brain, neural stem cells (NSCs) can be found in two main niches: the dentate gyrus (DG) and the subventricular zone (SVZ). In the DG, NSCs produce intermediate progenitors (IPs) that differentiate into excitatory neurons, while progenitors in the SVZ migrate to the olfactory bulb (OB), where they mainly differentiate into inhibitory interneurons. Neurogenesis, the process of generating new neurons, persists throughout life but decreases dramatically with aging, concomitantly with increased inflammation. Although many cell types, including microglia, undergo significant transcriptional changes, few such changes have been detected in neural progenitors. Furthermore, transcriptional profiles in progenitors from different neurogenic regions have not been compared on a single-cell level, and little is known about how they are affected by aging-related inflammation. We have generated a single cell RNA sequencing dataset enriched for IPs, which revealed that most aged neural progenitors only acquire minor transcriptional changes. However, progenitors set to become excitatory neurons decrease faster than others. In addition, a population in the aged SVZ, not detected in the OB, acquired major transcriptional activation related to immune responses. This suggests that differences in age related neurogenic decline between regions is not due to tissue differences but rather cell type specific intrinsic transcriptional programs, and that subset of neuroblasts in the SVZ react strongly to age related inflammatory cues.
Collapse
Affiliation(s)
- Jonas Fritze
- Stem Cells, Aging and Neurodegeneration Group, Faculty of Medicine, Department of Experimental Medical Science, Lund University, Lund, Sweden
- Lund Stem Cell Center, Lund, Sweden
| | - Stefan Lang
- Lund Stem Cell Center, Lund, Sweden
- Computational Genomics Group, Faculty of Medicine, Division of Molecular Hematology, Lund University, Lund, Sweden
| | - Mikael Sommarin
- Lund Stem Cell Center, Lund, Sweden
- Stem Cells and Leukemia Group, Faculty of Medicine, Division of Molecular Hematology, Lund University, Lund, Sweden
| | - Shamit Soneji
- Lund Stem Cell Center, Lund, Sweden
- Computational Genomics Group, Faculty of Medicine, Division of Molecular Hematology, Lund University, Lund, Sweden
| | - Henrik Ahlenius
- Stem Cells, Aging and Neurodegeneration Group, Faculty of Medicine, Department of Experimental Medical Science, Lund University, Lund, Sweden
- Lund Stem Cell Center, Lund, Sweden
| |
Collapse
|
18
|
Norton ES, Whaley LA, Jones VK, Brooks MM, Russo MN, Morderer D, Jessen E, Schiapparelli P, Ramos-Fresnedo A, Zarco N, Carrano A, Rossoll W, Asmann YW, Lam TT, Chaichana KL, Anastasiadis PZ, Quiñones-Hinojosa A, Guerrero-Cázares H. Cell-specific cross-talk proteomics reveals cathepsin B signaling as a driver of glioblastoma malignancy near the subventricular zone. SCIENCE ADVANCES 2024; 10:eadn1607. [PMID: 39110807 PMCID: PMC11305394 DOI: 10.1126/sciadv.adn1607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 06/28/2024] [Indexed: 08/10/2024]
Abstract
Glioblastoma (GBM) is the most prevalent and aggressive malignant primary brain tumor. GBM proximal to the lateral ventricles (LVs) is more aggressive, potentially because of subventricular zone contact. Despite this, cross-talk between GBM and neural stem/progenitor cells (NSC/NPCs) is not well understood. Using cell-specific proteomics, we show that LV-proximal GBM prevents neuronal maturation of NSCs through induction of senescence. In addition, GBM brain tumor-initiating cells (BTICs) increase expression of cathepsin B (CTSB) upon interaction with NPCs. Lentiviral knockdown and recombinant protein experiments reveal that both cell-intrinsic and soluble CTSB promote malignancy-associated phenotypes in BTICs. Soluble CTSB stalls neuronal maturation in NPCs while promoting senescence, providing a link between LV-tumor proximity and neurogenesis disruption. Last, we show LV-proximal CTSB up-regulation in patients, showing the relevance of this cross-talk in human GBM biology. These results demonstrate the value of proteomic analysis in tumor microenvironment research and provide direction for new therapeutic strategies in GBM.
Collapse
Affiliation(s)
- Emily S. Norton
- Department of Neurosurgery, Mayo Clinic, Jacksonville, FL 32224, USA
- Neuroscience Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Jacksonville, FL 32224, USA
- Regenerative Sciences Training Program, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Lauren A. Whaley
- Department of Neurosurgery, Mayo Clinic, Jacksonville, FL 32224, USA
- Department of Biology, University of North Florida, Jacksonville, FL 32224, USA
| | - Vanessa K. Jones
- Department of Neurosurgery, Mayo Clinic, Jacksonville, FL 32224, USA
- Department of Biology, University of North Florida, Jacksonville, FL 32224, USA
| | - Mieu M. Brooks
- Department of Neurosurgery, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Marissa N. Russo
- Department of Neurosurgery, Mayo Clinic, Jacksonville, FL 32224, USA
- Neuroscience Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Dmytro Morderer
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Erik Jessen
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Jacksonville, FL 32224, USA
| | | | | | - Natanael Zarco
- Department of Neurosurgery, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Anna Carrano
- Department of Neurosurgery, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Wilfried Rossoll
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Yan W. Asmann
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Jacksonville, FL 32224, USA
| | - TuKiet T. Lam
- Keck MS and Proteomics Resource, Yale School of Medicine, New Haven, CT 06510, USA
- Department of Molecular Biophysics and Biochemistry, Yale School of Medicine, New Haven, CT 06510, USA
| | | | | | | | | |
Collapse
|
19
|
Liu H, Dong J, Duan Z, Xia F, Willner I, Huang F. Light-activated CRISPR-Cas12a for amplified imaging of microRNA in cell cycle phases at single-cell levels. SCIENCE ADVANCES 2024; 10:eadp6166. [PMID: 39047109 PMCID: PMC11268419 DOI: 10.1126/sciadv.adp6166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 06/20/2024] [Indexed: 07/27/2024]
Abstract
An ortho-nitrobenzyl phosphate ester-caged nucleic acid hairpin structure coupled to the CRISPR-Cas12a complex is introduced as a functional reaction module for the light-induced activation of the CRISPR-Cas12a (LAC12a) machinery toward the amplified fluorescence detection of microRNA-21 (miRNA-21). The LAC12a machinery is applied for the selective, in vitro sensing of miRNA-21 and for the intracellular imaging of miRNA-21 in different cell lines. The LAC12a system is used to image miRNA-21 in different cell cycle phases of MCF-7 cells. Moreover, the LAC12a machinery integrated in cells enables the two-photon laser confocal microscopy-assisted, light-stimulated spatiotemporal, selective activation of the CRISPR-Cas12a miRNA-21 imaging machinery at the single-cell level and the evaluation of relative expression levels of miRNA-21 at distinct cell cycle phases. The method is implemented to map the distribution of cell cycle phases in an array of single cells.
Collapse
Affiliation(s)
- Hong Liu
- State Key Laboratory of Biogeology and Environmental Geology, Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, P.R. China
| | - Jiantong Dong
- Institute of Chemistry and Center for Nanoscience and Nanotechnology, The Hebrew University of Jerusalem, Jerusalem 91904, Israel
| | - Zhijuan Duan
- State Key Laboratory of Biogeology and Environmental Geology, Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, P.R. China
| | - Fan Xia
- State Key Laboratory of Biogeology and Environmental Geology, Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, P.R. China
| | - Itamar Willner
- Institute of Chemistry and Center for Nanoscience and Nanotechnology, The Hebrew University of Jerusalem, Jerusalem 91904, Israel
| | - Fujian Huang
- State Key Laboratory of Biogeology and Environmental Geology, Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, P.R. China
| |
Collapse
|
20
|
Sun ED, Zhou OY, Hauptschein M, Rappoport N, Xu L, Navarro Negredo P, Liu L, Rando TA, Zou J, Brunet A. Spatiotemporal transcriptomic profiling and modeling of mouse brain at single-cell resolution reveals cell proximity effects of aging and rejuvenation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.16.603809. [PMID: 39071282 PMCID: PMC11275735 DOI: 10.1101/2024.07.16.603809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Old age is associated with a decline in cognitive function and an increase in neurodegenerative disease risk1. Brain aging is complex and accompanied by many cellular changes2-20. However, the influence that aged cells have on neighboring cells and how this contributes to tissue decline is unknown. More generally, the tools to systematically address this question in aging tissues have not yet been developed. Here, we generate spatiotemporal data at single-cell resolution for the mouse brain across lifespan, and we develop the first machine learning models based on spatial transcriptomics ('spatial aging clocks') to reveal cell proximity effects during brain aging and rejuvenation. We collect a single-cell spatial transcriptomics brain atlas of 4.2 million cells from 20 distinct ages and across two rejuvenating interventions-exercise and partial reprogramming. We identify spatial and cell type-specific transcriptomic fingerprints of aging, rejuvenation, and disease, including for rare cell types. Using spatial aging clocks and deep learning models, we find that T cells, which infiltrate the brain with age, have a striking pro-aging proximity effect on neighboring cells. Surprisingly, neural stem cells have a strong pro-rejuvenating effect on neighboring cells. By developing computational tools to identify mediators of these proximity effects, we find that pro-aging T cells trigger a local inflammatory response likely via interferon-γ whereas pro-rejuvenating neural stem cells impact the metabolism of neighboring cells possibly via growth factors (e.g. vascular endothelial growth factor) and extracellular vesicles, and we experimentally validate some of these predictions. These results suggest that rare cells can have a drastic influence on their neighbors and could be targeted to counter tissue aging. We anticipate that these spatial aging clocks will not only allow scalable assessment of the efficacy of interventions for aging and disease but also represent a new tool for studying cell-cell interactions in many spatial contexts.
Collapse
Affiliation(s)
- Eric D. Sun
- Department of Biomedical Data Science, Stanford University, CA, USA
- Department of Genetics, Stanford University, CA, USA
| | - Olivia Y. Zhou
- Department of Genetics, Stanford University, CA, USA
- Stanford Biophysics Program, Stanford University, CA, USA
- Stanford Medical Scientist Training Program, Stanford University, CA, USA
| | | | | | - Lucy Xu
- Department of Genetics, Stanford University, CA, USA
- Department of Biology, Stanford University, CA, USA
| | | | - Ling Liu
- Department of Neurology, Stanford University, CA, USA
- Department of Neurology, UCLA, Los Angeles, CA, USA
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Biology, UCLA, Los Angeles, CA, USA
| | - Thomas A. Rando
- Department of Neurology, Stanford University, CA, USA
- Department of Neurology, UCLA, Los Angeles, CA, USA
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Biology, UCLA, Los Angeles, CA, USA
| | - James Zou
- Department of Biomedical Data Science, Stanford University, CA, USA
- These authors contributed equally: James Zou, Anne Brunet
| | - Anne Brunet
- Department of Genetics, Stanford University, CA, USA
- Glenn Center for the Biology of Aging, Stanford University, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, CA, USA
- These authors contributed equally: James Zou, Anne Brunet
| |
Collapse
|
21
|
Remsik J, Boire A. The path to leptomeningeal metastasis. Nat Rev Cancer 2024; 24:448-460. [PMID: 38871881 PMCID: PMC11404355 DOI: 10.1038/s41568-024-00700-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/29/2024] [Indexed: 06/15/2024]
Abstract
The leptomeninges, the cerebrospinal-fluid-filled tissues surrounding the central nervous system, play host to various pathologies including infection, neuroinflammation and malignancy. Spread of systemic cancer into this space, termed leptomeningeal metastasis, occurs in 5-10% of patients with solid tumours and portends a bleak clinical prognosis. Previous, predominantly descriptive, clinical studies have provided few insights. Recent development of preclinical leptomeningeal metastasis models, alongside genomic, transcriptomic and proteomic sequencing efforts, has provided groundwork for mechanistic understanding and identification of long-needed therapeutic targets. Although previously understood as an anatomically isolated compartment, the leptomeninges are increasingly appreciated as a major conduit of communication between the systemic circulation and the central nervous system. Despite the unique nature of the leptomeningeal microenvironment, the general principles of metastasis hold true: cells metastasizing to the leptomeninges must gain access to the new environment, survive within the space and evade the immune system. The study of leptomeningeal metastasis has the potential to uncover novel site-specific metastatic principles and illuminate the physiology of the leptomeningeal space. In this Review, we provide a biology-focused overview of how metastatic cells reach the leptomeninges, thrive in this nutritionally sparse environment and evade the detection of the omnipresent immune system.
Collapse
Affiliation(s)
- Jan Remsik
- Human Oncology & Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Laboratory for Immunology of Metastatic Ecosystems, Center for Cancer Biology, VIB, Leuven, Belgium
- Department of Oncology, KU Leuven, Leuven, Belgium
| | - Adrienne Boire
- Human Oncology & Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Brain Tumour Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
22
|
Shi CL, Han XL, Chen JC, Pan QF, Gao YC, Guo PY, Min XL, Gao YJ. Single-nucleus transcriptome unveils the role of ferroptosis in ischemic stroke. Heliyon 2024; 10:e32727. [PMID: 38994078 PMCID: PMC11237950 DOI: 10.1016/j.heliyon.2024.e32727] [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: 03/17/2024] [Revised: 06/06/2024] [Accepted: 06/07/2024] [Indexed: 07/13/2024] Open
Abstract
Multiple cell death pathways are involved in neuronal death in ischemic stroke (IS). However, the role of different cell death pathways in different cell types has not been elucidated. By analyzing three single-nucleus RNA sequencing (snRNA-seq) data of IS, we first found that a variety of programmed cell death (PCD) -related genes were significantly changed in different cell types. Based on machine learning and virtual gene knockout, we found that ferroptosis related genes, ferritin heavy chain 1 (Fth1) and ferritin light chain (Ftl1), play a key role in IS. Ftl1 and Fth1 can promote microglia activation, as well as the production of inflammatory factors and chemokines. Cell communication analysis showed that activated microglia could enhance chemotactic peripheral leukocyte infiltration, such as macrophages and neutrophils, through Spp1-Cd44 and App-Cd74 signaling, thereby aggravating brain tissue damage. Furthermore, real-time quantitative polymerase chain reaction (RT-qPCR) showed that P2ry12 and Mef2c were significantly decreased in oxygen-glucose deprivation (OGD) group, while Ftl1, Fth1, Apoe, Ctsb, Cd44 and Cd74 were significantly increased in OGD group. Collectively, our findings suggested targeted therapy against microglia Ftl1 and Fth1 might improve the state of microglia, reduce the infiltration of peripheral immune cells and tissue inflammation, and then improve the ischemic brain injury in mouse.
Collapse
Affiliation(s)
- Cheng-Long Shi
- Department of Neurosurgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Xiu-Li Han
- Department of Stomatology, Kunming Children's Hospital, Kunming, 650100, China
| | - Jing-Ce Chen
- Department of Orthopedics, The First People's Hospital of Yunnan Province, Kunming, 650100, China
| | - Qian-Fan Pan
- Department of Neurosurgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Yong-Chao Gao
- Department of Neurosurgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Peng-Yan Guo
- Department of Neurosurgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Xiao-Li Min
- Department of Neurosurgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Yong-Jun Gao
- Department of Neurosurgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| |
Collapse
|
23
|
Wu W, Alexander JS, Booth JL, Miller CA, Metcalf JP, Drevets DA. Influenza virus infection exacerbates gene expression related to neurocognitive dysfunction in brains of old mice. Immun Ageing 2024; 21:39. [PMID: 38907247 PMCID: PMC11191167 DOI: 10.1186/s12979-024-00447-y] [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: 02/18/2024] [Accepted: 06/11/2024] [Indexed: 06/23/2024]
Abstract
BACKGROUND Age > 65 years is a key risk factor for poor outcomes after human influenza infection. Specifically, in addition to respiratory disease, non-neurotropic influenza A virus (IAV) causes neuro-cognitive complications, e.g. new onset depression and increases the risk of dementia after hospitalization. This study aimed to identify potential mechanisms of these effects by determining differences between young and old mice in brain gene expression in a mouse model of non-neurotropic IAV infection. METHODS Young (12 weeks) and old (70 weeks) C57Bl/6J mice were inoculated intranasally with 200 PFU H1N1 A/PR/34/8 (PR8) or sterile PBS (mock). Gene expression in lung and brain was measured by qRT-PCR and normalized to β-actin. Findings were confirmed using the nCounter Mouse Neuroinflammation Array (NanoString) and analyzed with nSolver 4.0 and Ingenuity Pathway Analysis (IPA, Qiagen). RESULTS IAV PR8 did not invade the central nervous system. Young and old mice differed significantly in brain gene expression at baseline and during non-neurotropic IAV infection. Expression of brain Ifnl, Irf7, and Tnf mRNAs was upregulated over baseline control at 3 days post-infection (p.i.) only in young mice, but old mice expressed more Ifnl than young mice 7 days p.i. Gene arrays showed down-regulation of the Epigenetic Regulation, Insulin Signaling, and Neurons and Neurotransmission pathways in old mice 3 days p.i. while young mice demonstrated no change or induction of these pathways at the same time point. IPA revealed marked baseline differences between old and young mice. Gene expression related to Cognitive Impairment, Memory Deficits and Learning worsened in old mice relative to young mice during IAV infection. Aged mice demonstrate more severe changes in gene expression related to memory loss and cognitive dysfunction by IPA. CONCLUSIONS These data suggest the genes and pathways related to learning and cognitive performance that were worse at baseline in old mice were further worsened by IAV infection, similar to old patients. Early events in the brain triggered by IAV infection portend downstream neurocognitive pathology in old adults.
Collapse
Affiliation(s)
- Wenxin Wu
- Pulmonary, Critical Care & Sleep Medicine, Department of Medicine, University of Oklahoma Health Sciences Center, Room 425, RP1 800 N. Research Pkwy, Oklahoma City, OK, 73104, USA
| | - Jeremy S Alexander
- Pulmonary, Critical Care & Sleep Medicine, Department of Medicine, University of Oklahoma Health Sciences Center, Room 425, RP1 800 N. Research Pkwy, Oklahoma City, OK, 73104, USA
| | - J Leland Booth
- Pulmonary, Critical Care & Sleep Medicine, Department of Medicine, University of Oklahoma Health Sciences Center, Room 425, RP1 800 N. Research Pkwy, Oklahoma City, OK, 73104, USA
| | - Craig A Miller
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Oklahoma State University, Stillwater, OK, USA
| | - Jordan P Metcalf
- Pulmonary, Critical Care & Sleep Medicine, Department of Medicine, University of Oklahoma Health Sciences Center, Room 425, RP1 800 N. Research Pkwy, Oklahoma City, OK, 73104, USA.
- Veterans Affairs Medical Center, Oklahoma City, OK, USA.
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| | - Douglas A Drevets
- Infectious Diseases, Department of Medicine, University of Oklahoma Health Sciences Center, 800 Stanton L. Young, Suite 7300, Oklahoma City, OK, 73104, USA.
| |
Collapse
|
24
|
Marin-Rodero M, Reyes EC, Walker AJ, Jayewickreme T, Pinho-Ribeiro FA, Richardson Q, Jackson R, Chiu IM, Benoist C, Stevens B, Trejo JL, Mathis D. The meninges host a unique compartment of regulatory T cells that bulwarks adult hippocampal neurogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.17.599387. [PMID: 38948783 PMCID: PMC11212874 DOI: 10.1101/2024.06.17.599387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Our knowledge about the meningeal immune system has recently burgeoned, particularly our understanding of how innate and adaptive effector cells are mobilized to meet brain challenges. However, information on how meningeal immunocytes guard brain homeostasis in healthy individuals remains sparse. This study highlights the heterogeneous and polyfunctional regulatory-T (Treg) cell compartment in the meninges. A Treg subtype specialized in controlling Th1-cell responses and another known to control responses in B-cell follicles were substantial components of this compartment, foretelling that punctual Treg-cell ablation rapidly unleashed interferon-gamma production by meningeal lymphocytes, unlocked their access to the brain parenchyma, and altered meningeal B-cell profiles. Distally, the hippocampus assumed a reactive state, with morphological and transcriptional changes in multiple glial-cell types; within the dentate gyrus, neural stem cells showed exacerbated death and desisted from further differentiation, associated with inhibition of spatial-reference memory. Thus, meningeal Treg cells are a multifaceted bulwark to brain homeostasis at steady-state. One sentence summary A distinct population of regulatory T cells in the murine meninges safeguards homeostasis by keeping local interferon-γ-producing lymphocytes in check, thereby preventing their invasion of the parenchyma, activation of hippocampal glial cells, death of neural stem cells, and memory decay.
Collapse
|
25
|
Zhang W, Sun HS, Wang X, Dumont AS, Liu Q. Cellular senescence, DNA damage, and neuroinflammation in the aging brain. Trends Neurosci 2024; 47:461-474. [PMID: 38729785 DOI: 10.1016/j.tins.2024.04.003] [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: 02/05/2024] [Revised: 04/10/2024] [Accepted: 04/15/2024] [Indexed: 05/12/2024]
Abstract
Aging may lead to low-level chronic inflammation that increases the susceptibility to age-related conditions, including memory impairment and progressive loss of brain volume. As brain health is essential to promoting healthspan and lifespan, it is vital to understand age-related changes in the immune system and central nervous system (CNS) that drive normal brain aging. However, the relative importance, mechanistic interrelationships, and hierarchical order of such changes and their impact on normal brain aging remain to be clarified. Here, we synthesize accumulating evidence that age-related DNA damage and cellular senescence in the immune system and CNS contribute to the escalation of neuroinflammation and cognitive decline during normal brain aging. Targeting cellular senescence and immune modulation may provide a logical rationale for developing new treatment options to restore immune homeostasis and counteract age-related brain dysfunction and diseases.
Collapse
Affiliation(s)
- Wenyan Zhang
- Department of Neurology, Tianjin Neurological Institute, Tianjin Institute of Immunology, State Key Laboratory of Experimental Hematology, International Joint Laboratory of Ocular Diseases, Ministry of Education, Haihe Laboratory of Cell Ecosystem, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Hong-Shuo Sun
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Xiaoying Wang
- Tulane Center for Clinical Neurosciences, Department of Neurosurgery and Neurology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Aaron S Dumont
- Tulane Center for Clinical Neurosciences, Department of Neurosurgery and Neurology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Qiang Liu
- Department of Neurology, Tianjin Neurological Institute, Tianjin Institute of Immunology, State Key Laboratory of Experimental Hematology, International Joint Laboratory of Ocular Diseases, Ministry of Education, Haihe Laboratory of Cell Ecosystem, Tianjin Medical University General Hospital, Tianjin 300052, China.
| |
Collapse
|
26
|
Caruso MG, Nicolas S, Lucassen PJ, Mul JD, O’Leary OF, Nolan YM. Ageing, Cognitive Decline, and Effects of Physical Exercise: Complexities, and Considerations from Animal Models. Brain Plast 2024; 9:43-73. [PMID: 38993577 PMCID: PMC11234681 DOI: 10.3233/bpl-230157] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/18/2024] [Indexed: 07/13/2024] Open
Abstract
In our ageing global population, the cognitive decline associated with dementia and neurodegenerative diseases represents a major healthcare problem. To date, there are no effective treatments for age-related cognitive impairment, thus preventative strategies are urgently required. Physical exercise is gaining traction as a non-pharmacological approach to promote brain health. Adult hippocampal neurogenesis (AHN), a unique form of brain plasticity which is necessary for certain cognitive functions declines with age and is enhanced in response to exercise. Accumulating evidence from research in rodents suggests that physical exercise has beneficial effects on cognition through its proneurogenic capabilities. Given ethical and technical limitations in human studies, preclinical research in rodents is crucial for a better understanding of such exercise-induced brain and behavioural changes. In this review, exercise paradigms used in preclinical research are compared. We provide an overview of the effects of different exercise paradigms on age-related cognitive decline from middle-age until older-age. We discuss the relationship between the age-related decrease in AHN and the potential impact of exercise on mitigating this decline. We highlight the emerging literature on the impact of exercise on gut microbiota during ageing and consider the role of the gut-brain axis as a future possible strategy to optimize exercise-enhanced cognitive function. Finally, we propose a guideline for designing optimal exercise protocols in rodent studies, which would inform clinical research and contribute to developing preventative strategies for age-related cognitive decline.
Collapse
Affiliation(s)
- Maria Giovanna Caruso
- Department of Anatomy and Neuroscience, University College Cork, Ireland
- APC Microbiome Ireland, University College Cork, Ireland
| | - Sarah Nicolas
- Department of Anatomy and Neuroscience, University College Cork, Ireland
- APC Microbiome Ireland, University College Cork, Ireland
| | - Paul J. Lucassen
- Brain Plasticity group, Swammerdam Institute for Life Sciences, University of Amsterdam, The Netherlands
- Center for Urban Mental Health, University of Amsterdam, Amsterdam, The Netherlands
| | - Joram D. Mul
- Brain Plasticity group, Swammerdam Institute for Life Sciences, University of Amsterdam, The Netherlands
- Center for Urban Mental Health, University of Amsterdam, Amsterdam, The Netherlands
| | - Olivia F. O’Leary
- Department of Anatomy and Neuroscience, University College Cork, Ireland
- APC Microbiome Ireland, University College Cork, Ireland
| | - Yvonne M. Nolan
- Department of Anatomy and Neuroscience, University College Cork, Ireland
- APC Microbiome Ireland, University College Cork, Ireland
| |
Collapse
|
27
|
Wang Y, Shen Y, Liang J, Wang S, Huang Y, Zhu Q, Zhang X, Yu K, Tong G, Yang C, Li Y, Wang J, Zhao Y. Neurons upregulate PD-L1 via IFN/STAT1/IRF1 to alleviate damage by CD8 + T cells in cerebral malaria. J Neuroinflammation 2024; 21:119. [PMID: 38715061 PMCID: PMC11077882 DOI: 10.1186/s12974-024-03114-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 04/29/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Cerebral malaria (CM) is the most lethal complication of malaria, and survivors usually endure neurological sequelae. Notably, the cytotoxic effect of infiltrating Plasmodium-activated CD8+ T cells on cerebral microvasculature endothelial cells is a prominent feature of the experimental CM (ECM) model with blood-brain barrier disruption. However, the damage effect of CD8+ T cells infiltrating the brain parenchyma on neurons remains unclear. Based on the immunosuppressive effect of the PD-1/PD-L1 pathway on T cells, our previous study demonstrated that the systemic upregulation of PD-L1 to inhibit CD8+ T cell function could effectively alleviate the symptoms of ECM mice. However, it has not been reported whether neurons can suppress the pathogenic effect of CD8+ T cells through the PD-1/PD-L1 negative immunomodulatory pathway. As the important inflammatory factor of CM, interferons can induce the expression of PD-L1 via different molecular mechanisms according to the neuro-immune microenvironment. Therefore, this study aimed to investigate the direct interaction between CD8+ T cells and neurons, as well as the mechanism of neurons to alleviate the pathogenic effect of CD8+ T cells through up-regulating PD-L1 induced by IFNs. METHODS Using the ECM model of C57BL/6J mice infected with Plasmodium berghei ANKA (PbA), morphological observations were conducted in vivo by electron microscope and IF staining. The interaction between the ECM CD8+ T cells (immune magnetic bead sorting from spleen of ECM mice) and primary cultured cortical neurons in vitro was observed by IF staining and time-lapse photography. RNA-seq was performed to analyze the signaling pathway of PD-L1 upregulation in neurons induced by IFNβ or IFNγ, and verified through q-PCR, WB, IF staining, and flow cytometry both in vitro and in vivo using IFNAR or IFNGR gene knockout mice. The protective effect of adenovirus-mediated PD-L1 IgGFc fusion protein expression was verified in ECM mice with brain stereotaxic injection in vivo and in primary cultured neurons via viral infection in vitro. RESULTS In vivo, ECM mice showed infiltration of activated CD8+ T cells and neuronal injury in the brain parenchyma. In vitro, ECM CD8+ T cells were in direct contact with neurons and induced axonal damage, as an active behavior. The PD-L1 protein level was elevated in neurons of ECM mice and in primary cultured neurons induced by IFNβ, IFNγ, or ECM CD8+ T cells in vitro. Furthermore, the IFNβ or IFNγ induced neuronal expression of PD-L1 was mediated by increasing STAT1/IRF1 pathway via IFN receptors. The increase of PD-L1 expression in neurons during PbA infection was weakened after deleting the IFNAR or IFNGR. Increased PD-L1 expression by adenovirus partially protected neurons from CD8+ T cell-mediated damage both in vitro and in vivo. CONCLUSION Our study demonstrates that both type I and type II IFNs can induce neurons to upregulate PD-L1 via the STAT1/IRF1 pathway mediated by IFN receptors to protect against activated CD8+ T cell-mediated damage, providing a targeted pathway to alleviate neuroinflammation during ECM.
Collapse
Affiliation(s)
- Yi Wang
- Department of Medical Microbiology and Parasitology, Air Force Medical University, 169# Changle West Road, Xi'an, 710032, China
| | - Yan Shen
- Department of Medical Microbiology and Parasitology, Air Force Medical University, 169# Changle West Road, Xi'an, 710032, China
| | - Jiao Liang
- Department of Medical Microbiology and Parasitology, Air Force Medical University, 169# Changle West Road, Xi'an, 710032, China
| | - Shubiao Wang
- Grade 2020 Clinical Medicine (Five-Year Program), Basic Medical College, Air Force Medical University, Xi'an, 710032, Shaanxi, China
| | - Yuxiao Huang
- Department of Medical Microbiology and Parasitology, Air Force Medical University, 169# Changle West Road, Xi'an, 710032, China
| | - Qinghao Zhu
- Department of Medical Microbiology and Parasitology, Air Force Medical University, 169# Changle West Road, Xi'an, 710032, China
| | - Xizhi Zhang
- Grade 2019 Clinical Medicine (Five-Year Program), Basic Medical College, Air Force Medical University, Xi'an, 710032, Shaanxi, China
| | - Kangjie Yu
- Department of Pathology, Air Force Hospital of Eastern Theater, Nanjing, Jiangsu, China
| | - Guodong Tong
- College of Life Sciences, Northwest University, Xi'an, 710069, Shaanxi, China
| | - Chao Yang
- College of Life Sciences, Northwest University, Xi'an, 710069, Shaanxi, China
| | - Yinghui Li
- Department of Medical Microbiology and Parasitology, Air Force Medical University, 169# Changle West Road, Xi'an, 710032, China.
| | - Jun Wang
- Department of Medical Microbiology and Parasitology, Air Force Medical University, 169# Changle West Road, Xi'an, 710032, China.
| | - Ya Zhao
- Department of Medical Microbiology and Parasitology, Air Force Medical University, 169# Changle West Road, Xi'an, 710032, China.
| |
Collapse
|
28
|
Ramnauth AD, Tippani M, Divecha HR, Papariello AR, Miller RA, Nelson ED, Pattie EA, Kleinman JE, Maynard KR, Collado-Torres L, Hyde TM, Martinowich K, Hicks SC, Page SC. Spatiotemporal analysis of gene expression in the human dentate gyrus reveals age-associated changes in cellular maturation and neuroinflammation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.11.20.567883. [PMID: 38045413 PMCID: PMC10690172 DOI: 10.1101/2023.11.20.567883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
The dentate gyrus of the hippocampus is important for many cognitive functions, including learning, memory, and mood. Here, we investigated age-associated changes in transcriptome-wide spatial gene expression in the human dentate gyrus across the lifespan. Genes associated with neurogenesis and the extracellular matrix were enriched in infants, while gene markers of inhibitory neurons and cell proliferation showed increases and decreases in post-infancy, respectively. While we did not find evidence for neural proliferation post-infancy, we did identify molecular signatures supporting protracted maturation of granule cells. We also identified a wide-spread hippocampal aging signature and an age-associated increase in genes related to neuroinflammation. Our findings suggest major changes to the putative neurogenic niche after infancy and identify molecular foci of brain aging in glial and neuropil enriched tissue.
Collapse
|
29
|
Dobner S, Tóth F, de Rooij LPMH. A high-resolution view of the heterogeneous aging endothelium. Angiogenesis 2024; 27:129-145. [PMID: 38324119 PMCID: PMC11021252 DOI: 10.1007/s10456-023-09904-6] [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: 08/21/2023] [Accepted: 12/28/2023] [Indexed: 02/08/2024]
Abstract
Vascular endothelial cell (EC) aging has a strong impact on tissue perfusion and overall cardiovascular health. While studies confined to the investigation of aging-associated vascular readouts in one or a few tissues have already drastically expanded our understanding of EC aging, single-cell omics and other high-resolution profiling technologies have started to illuminate the intricate molecular changes underlying endothelial aging across diverse tissues and vascular beds at scale. In this review, we provide an overview of recent insights into the heterogeneous adaptations of the aging vascular endothelium. We address critical questions regarding tissue-specific and universal responses of the endothelium to the aging process, EC turnover dynamics throughout lifespan, and the differential susceptibility of ECs to acquiring aging-associated traits. In doing so, we underscore the transformative potential of single-cell approaches in advancing our comprehension of endothelial aging, essential to foster the development of future innovative therapeutic strategies for aging-associated vascular conditions.
Collapse
Affiliation(s)
- Sarah Dobner
- The CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Fanni Tóth
- The CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Laura P M H de Rooij
- The CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria.
| |
Collapse
|
30
|
Kedlian VR, Wang Y, Liu T, Chen X, Bolt L, Tudor C, Shen Z, Fasouli ES, Prigmore E, Kleshchevnikov V, Pett JP, Li T, Lawrence JEG, Perera S, Prete M, Huang N, Guo Q, Zeng X, Yang L, Polański K, Chipampe NJ, Dabrowska M, Li X, Bayraktar OA, Patel M, Kumasaka N, Mahbubani KT, Xiang AP, Meyer KB, Saeb-Parsy K, Teichmann SA, Zhang H. Human skeletal muscle aging atlas. NATURE AGING 2024; 4:727-744. [PMID: 38622407 PMCID: PMC11108788 DOI: 10.1038/s43587-024-00613-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 03/19/2024] [Indexed: 04/17/2024]
Abstract
Skeletal muscle aging is a key contributor to age-related frailty and sarcopenia with substantial implications for global health. Here we profiled 90,902 single cells and 92,259 single nuclei from 17 donors to map the aging process in the adult human intercostal muscle, identifying cellular changes in each muscle compartment. We found that distinct subsets of muscle stem cells exhibit decreased ribosome biogenesis genes and increased CCL2 expression, causing different aging phenotypes. Our atlas also highlights an expansion of nuclei associated with the neuromuscular junction, which may reflect re-innervation, and outlines how the loss of fast-twitch myofibers is mitigated through regeneration and upregulation of fast-type markers in slow-twitch myofibers with age. Furthermore, we document the function of aging muscle microenvironment in immune cell attraction. Overall, we present a comprehensive human skeletal muscle aging resource ( https://www.muscleageingcellatlas.org/ ) together with an in-house mouse muscle atlas to study common features of muscle aging across species.
Collapse
Affiliation(s)
- Veronika R Kedlian
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Yaning Wang
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Tianliang Liu
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xiaoping Chen
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Liam Bolt
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Catherine Tudor
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Zhuojian Shen
- Department of Thoracic Surgery, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
| | - Eirini S Fasouli
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Elena Prigmore
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | | | - Jan Patrick Pett
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Tong Li
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - John E G Lawrence
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Shani Perera
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Martin Prete
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Ni Huang
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Qin Guo
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xinrui Zeng
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Lu Yang
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Krzysztof Polański
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Nana-Jane Chipampe
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Monika Dabrowska
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Xiaobo Li
- Core Facilities for Medical Science, Sun Yat-sen University, Guangzhou, China
| | - Omer Ali Bayraktar
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Minal Patel
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Natsuhiko Kumasaka
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Krishnaa T Mahbubani
- Department of Surgery, University of Cambridge, Cambridge, UK
- Collaborative Biorepository for Translational Medicine (CBTM), NIHR Cambridge Biomedical Research Centre, Cambridge, UK
| | - Andy Peng Xiang
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Kerstin B Meyer
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Kourosh Saeb-Parsy
- Department of Surgery, University of Cambridge, Cambridge, UK.
- Collaborative Biorepository for Translational Medicine (CBTM), NIHR Cambridge Biomedical Research Centre, Cambridge, UK.
| | - Sarah A Teichmann
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK.
- Cavendish Laboratory, University of Cambridge, Cambridge, UK.
| | - Hongbo Zhang
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
31
|
van Olst L, Kamermans A, Halters S, van der Pol SMA, Rodriguez E, Verberk IMW, Verberk SGS, Wessels DWR, Rodriguez-Mogeda C, Verhoeff J, Wouters D, Van den Bossche J, Garcia-Vallejo JJ, Lemstra AW, Witte ME, van der Flier WM, Teunissen CE, de Vries HE. Adaptive immune changes associate with clinical progression of Alzheimer's disease. Mol Neurodegener 2024; 19:38. [PMID: 38658964 PMCID: PMC11044380 DOI: 10.1186/s13024-024-00726-8] [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: 07/25/2023] [Accepted: 03/29/2024] [Indexed: 04/26/2024] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is the most frequent cause of dementia. Recent evidence suggests the involvement of peripheral immune cells in the disease, but the underlying mechanisms remain unclear. METHODS We comprehensively mapped peripheral immune changes in AD patients with mild cognitive impairment (MCI) or dementia compared to controls, using cytometry by time-of-flight (CyTOF). RESULTS We found an adaptive immune signature in AD, and specifically highlight the accumulation of PD1+ CD57+ CD8+ T effector memory cells re-expressing CD45RA in the MCI stage of AD. In addition, several innate and adaptive immune cell subsets correlated to cerebrospinal fluid (CSF) biomarkers of AD neuropathology and measures for cognitive decline. Intriguingly, subsets of memory T and B cells were negatively associated with CSF biomarkers for tau pathology, neurodegeneration and neuroinflammation in AD patients. Lastly, we established the influence of the APOE ε4 allele on peripheral immunity. CONCLUSIONS Our findings illustrate significant peripheral immune alterations associated with both early and late clinical stages of AD, emphasizing the necessity for further investigation into how these changes influence underlying brain pathology.
Collapse
Affiliation(s)
- Lynn van Olst
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, the Netherlands.
- Amsterdam Neuroscience, Neuroinfection & -Inflammation, Amsterdam, the Netherlands.
- Present address: The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| | - Alwin Kamermans
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Neuroinfection & -Inflammation, Amsterdam, the Netherlands
| | - Sem Halters
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Neuroinfection & -Inflammation, Amsterdam, the Netherlands
| | - Susanne M A van der Pol
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Neuroinfection & -Inflammation, Amsterdam, the Netherlands
| | - Ernesto Rodriguez
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, the Netherlands
- Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, the Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, the Netherlands
| | - Inge M W Verberk
- Amsterdam Neuroscience, Neuroinfection & -Inflammation, Amsterdam, the Netherlands
- Department of Laboratory Medicine, Neurochemistry Laboratory, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, the Netherlands
| | - Sanne G S Verberk
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, the Netherlands
- Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischemic Syndromes, Amsterdam, the Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, the Netherlands
| | - Danielle W R Wessels
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, the Netherlands
| | - Carla Rodriguez-Mogeda
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Neuroinfection & -Inflammation, Amsterdam, the Netherlands
| | - Jan Verhoeff
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, the Netherlands
- Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, the Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, the Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, the Netherlands
| | - Dorine Wouters
- Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischemic Syndromes, Amsterdam, the Netherlands
| | - Jan Van den Bossche
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, the Netherlands
- Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischemic Syndromes, Amsterdam, the Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, the Netherlands
| | - Juan J Garcia-Vallejo
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, the Netherlands
- Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, the Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, the Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, the Netherlands
| | - Afina W Lemstra
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, the Netherlands
- Department of Neurology, Amsterdam UMC Location VUmc, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Maarten E Witte
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Neuroinfection & -Inflammation, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, the Netherlands
- Amsterdam Institute for Infection and Immunity, Inflammatory Diseases, Amsterdam, the Netherlands
| | - Wiesje M van der Flier
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, the Netherlands
- Department of Neurology, Amsterdam UMC Location VUmc, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Department of Epidemiology & Data Science, Amsterdam UMC Location VUmc, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Charlotte E Teunissen
- Amsterdam Neuroscience, Neuroinfection & -Inflammation, Amsterdam, the Netherlands
- Department of Laboratory Medicine, Neurochemistry Laboratory, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, the Netherlands
| | - Helga E de Vries
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, the Netherlands
- Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, the Netherlands
- Amsterdam Cardiovascular Sciences, Microcirculation, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Neurovascular Disorders, Amsterdam, the Netherlands
| |
Collapse
|
32
|
Katdare KA, Kjar A, O’Brown NM, Neal EH, Sorets AG, Shostak A, Romero-Fernandez W, Kwiatkowski AJ, Mlouk K, Kim H, Cowell RP, Schwensen KR, Horner KB, Wilson JT, Schrag MS, Megason SG, Lippmann ES. IQGAP2 regulates blood-brain barrier immune dynamics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.02.07.527394. [PMID: 38645082 PMCID: PMC11030232 DOI: 10.1101/2023.02.07.527394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Brain endothelial cells (BECs) play an important role in maintaining central nervous system (CNS) homeostasis through blood-brain barrier (BBB) functions. BECs express low baseline levels of adhesion receptors, which limits entry of leukocytes. However, the molecular mediators governing this phenotype remain mostly unclear. Here, we explored how infiltration of immune cells across the BBB is influenced by the scaffold protein IQ motif containing GTPase activating protein 2 (IQGAP2). In mice and zebrafish, we demonstrate that loss of Iqgap2 increases infiltration of peripheral leukocytes into the CNS under homeostatic and inflammatory conditions. Using single-cell RNA sequencing and immunohistology, we further show that BECs from mice lacking Iqgap2 exhibit a profound inflammatory signature, including extensive upregulation of adhesion receptors and antigen-processing machinery. Human tissue analyses also reveal that Alzheimer's disease is associated with reduced hippocampal IQGAP2. Overall, our results implicate IQGAP2 as an essential regulator of BBB immune privilege and immune cell entry into the CNS.
Collapse
Affiliation(s)
- Ketaki A. Katdare
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA
| | - Andrew Kjar
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | | | - Emma H. Neal
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Alexander G. Sorets
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Alena Shostak
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | | | - Kate Mlouk
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Hyosung Kim
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Rebecca P. Cowell
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Katrina R. Schwensen
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Kensley B. Horner
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - John T. Wilson
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Matthew S. Schrag
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sean G. Megason
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Ethan S. Lippmann
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
33
|
Xu L, Ramirez-Matias J, Hauptschein M, Sun ED, Lunger JC, Buckley MT, Brunet A. Restoration of neuronal progenitors by partial reprogramming in the aged neurogenic niche. NATURE AGING 2024; 4:546-567. [PMID: 38553564 DOI: 10.1038/s43587-024-00594-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 02/13/2024] [Indexed: 04/21/2024]
Abstract
Partial reprogramming (pulsed expression of reprogramming transcription factors) improves the function of several tissues in old mice. However, it remains largely unknown how partial reprogramming impacts the old brain. Here we use single-cell transcriptomics to systematically examine how partial reprogramming influences the subventricular zone neurogenic niche in aged mouse brains. Whole-body partial reprogramming mainly improves neuroblasts (cells committed to give rise to new neurons) in the old neurogenic niche, restoring neuroblast proportion to more youthful levels. Interestingly, targeting partial reprogramming specifically to the neurogenic niche also boosts the proportion of neuroblasts and their precursors (neural stem cells) in old mice and improves several molecular signatures of aging, suggesting that the beneficial effects of reprogramming are niche intrinsic. In old neural stem cell cultures, partial reprogramming cell autonomously restores the proportion of neuroblasts during differentiation and blunts some age-related transcriptomic changes. Importantly, partial reprogramming improves the production of new neurons in vitro and in old brains. Our work suggests that partial reprogramming could be used to rejuvenate the neurogenic niche and counter brain decline in old individuals.
Collapse
Affiliation(s)
- Lucy Xu
- Department of Genetics, Stanford University, Stanford, CA, USA
- Department of Biology, Stanford University, Stanford, CA, USA
| | | | - Max Hauptschein
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Eric D Sun
- Department of Genetics, Stanford University, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Judith C Lunger
- Department of Genetics, Stanford University, Stanford, CA, USA
| | | | - Anne Brunet
- Department of Genetics, Stanford University, Stanford, CA, USA.
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA.
- Glenn Center for the Biology of Aging, Stanford University, Stanford, CA, USA.
| |
Collapse
|
34
|
Kallal N, Hugues S, Garnier L. Regulation of autoimmune-mediated neuroinflammation by endothelial cells. Eur J Immunol 2024; 54:e2350482. [PMID: 38335316 DOI: 10.1002/eji.202350482] [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: 07/14/2023] [Revised: 01/26/2024] [Accepted: 01/29/2024] [Indexed: 02/12/2024]
Abstract
The CNS has traditionally been considered an immune-privileged organ, but recent studies have identified a plethora of immune cells in the choroid plexus, meninges, perivascular spaces, and cribriform plate. Although those immune cells are crucial for the maintenance of CNS homeostasis and for neural protection against infections, they can lead to neuroinflammation in some circumstances. The blood and the lymphatic vasculatures exhibit distinct structural and molecular features depending on their location in the CNS, greatly influencing the compartmentalization and the nature of CNS immune responses. In this review, we discuss how endothelial cells regulate the migration and the functions of T cells in the CNS both at steady-state and in murine models of neuroinflammation, with a special focus on the anatomical, cellular, and molecular mechanisms implicated in EAE.
Collapse
Affiliation(s)
- Neil Kallal
- Department of Pathology and Immunology, Geneva Medical School, Geneva, Switzerland
| | - Stephanie Hugues
- Department of Pathology and Immunology, Geneva Medical School, Geneva, Switzerland
| | - Laure Garnier
- Department of Pathology and Immunology, Geneva Medical School, Geneva, Switzerland
| |
Collapse
|
35
|
de la Fuente AG, Dittmer M, Heesbeen EJ, de la Vega Gallardo N, White JA, Young A, McColgan T, Dashwood A, Mayne K, Cabeza-Fernández S, Falconer J, Rodriguez-Baena FJ, McMurran CE, Inayatullah M, Rawji KS, Franklin RJM, Dooley J, Liston A, Ingram RJ, Tiwari VK, Penalva R, Dombrowski Y, Fitzgerald DC. Ageing impairs the regenerative capacity of regulatory T cells in mouse central nervous system remyelination. Nat Commun 2024; 15:1870. [PMID: 38467607 PMCID: PMC10928230 DOI: 10.1038/s41467-024-45742-w] [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: 01/15/2023] [Accepted: 01/31/2024] [Indexed: 03/13/2024] Open
Abstract
Myelin regeneration (remyelination) is essential to prevent neurodegeneration in demyelinating diseases such as Multiple Sclerosis, however, its efficiency declines with age. Regulatory T cells (Treg) recently emerged as critical players in tissue regeneration, including remyelination. However, the effect of ageing on Treg-mediated regenerative processes is poorly understood. Here, we show that expansion of aged Treg does not rescue age-associated remyelination impairment due to an intrinsically diminished capacity of aged Treg to promote oligodendrocyte differentiation and myelination in male and female mice. This decline in regenerative Treg functions can be rescued by a young environment. We identified Melanoma Cell Adhesion Molecule 1 (MCAM1) and Integrin alpha 2 (ITGA2) as candidates of Treg-mediated oligodendrocyte differentiation that decrease with age. Our findings demonstrate that ageing limits the neuroregenerative capacity of Treg, likely limiting their remyelinating therapeutic potential in aged patients, and describe two mechanisms implicated in Treg-driven remyelination that may be targetable to overcome this limitation.
Collapse
Affiliation(s)
- Alerie Guzman de la Fuente
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, BT9 7BL, Northern Ireland, UK.
- Institute for Health and Biomedical Sciences of Alicante (ISABIAL), Alicante, 03010, Spain.
- Instituto de Neurosciencias CSIC-UMH, San Juan de Alicante, Alicante, 03550, Spain.
| | - Marie Dittmer
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, BT9 7BL, Northern Ireland, UK
| | - Elise J Heesbeen
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, BT9 7BL, Northern Ireland, UK
- Division of Pharmacology, Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Nira de la Vega Gallardo
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, BT9 7BL, Northern Ireland, UK
| | - Jessica A White
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, BT9 7BL, Northern Ireland, UK
| | - Andrew Young
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, BT9 7BL, Northern Ireland, UK
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, BT9 7BL, Northern Ireland, UK
| | - Tiree McColgan
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, BT9 7BL, Northern Ireland, UK
| | - Amy Dashwood
- Department of Pathology, University of Cambridge, CB2 1QP, Cambridge, UK
- Babraham Institute, CB22 3AT, Cambridge, UK
| | - Katie Mayne
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, BT9 7BL, Northern Ireland, UK
| | - Sonia Cabeza-Fernández
- Institute for Health and Biomedical Sciences of Alicante (ISABIAL), Alicante, 03010, Spain
- Instituto de Neurosciencias CSIC-UMH, San Juan de Alicante, Alicante, 03550, Spain
| | - John Falconer
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, BT9 7BL, Northern Ireland, UK
- CRUK Beatson Institute, G61 1BD, Glasgow, UK
| | | | - Christopher E McMurran
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
| | - Mohammed Inayatullah
- Institute of Molecular Medicine, University of Southern Denmark, 5000, Odense, Denmark
- Danish Institute for Advanced Study (DIAS), 5230, Odense, Denmark
| | - Khalil S Rawji
- Altos Labs - Cambridge Institute of Science, Granta Park, Cambridge, CB21 6GP, UK
| | - Robin J M Franklin
- Altos Labs - Cambridge Institute of Science, Granta Park, Cambridge, CB21 6GP, UK
| | - James Dooley
- Department of Pathology, University of Cambridge, CB2 1QP, Cambridge, UK
| | - Adrian Liston
- Department of Pathology, University of Cambridge, CB2 1QP, Cambridge, UK
| | - Rebecca J Ingram
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, BT9 7BL, Northern Ireland, UK
| | - Vijay K Tiwari
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, BT9 7BL, Northern Ireland, UK
- Institute of Molecular Medicine, University of Southern Denmark, 5000, Odense, Denmark
- Danish Institute for Advanced Study (DIAS), 5230, Odense, Denmark
- Department of Clinical Genetics, Odense University Hospital, 5000, Odense, Denmark
| | - Rosana Penalva
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, BT9 7BL, Northern Ireland, UK
| | - Yvonne Dombrowski
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, BT9 7BL, Northern Ireland, UK
| | - Denise C Fitzgerald
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, BT9 7BL, Northern Ireland, UK.
| |
Collapse
|
36
|
Chen BR, Wu T, Chen TH, Wang Y. Neuroimmune interactions and their roles in neurodegenerative diseases. FUNDAMENTAL RESEARCH 2024; 4:251-261. [PMID: 38933502 PMCID: PMC11197660 DOI: 10.1016/j.fmre.2023.04.002] [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: 07/21/2022] [Revised: 02/10/2023] [Accepted: 04/03/2023] [Indexed: 06/28/2024] Open
Abstract
The nervous system possesses bidirectional, sophisticated and delicate communications with the immune system. These neuroimmune interactions play a vitally important role in the initiation and development of many disorders, especially neurodegenerative diseases. Although scientific advancements have made tremendous progress in this field during the last few years, neuroimmune communications are still far from being elucidated. By organizing recent research, in this review, we discuss the local and intersystem neuroimmune interactions and their roles in Alzheimer's disease, Parkinson's disease and amyotrophic lateral sclerosis. Unveiling these will help us gain a better understanding of the process of interplay inside the body and how the organism maintains homeostasis. It will also facilitate a view of the diseases from a holistic, pluralistic and interconnected perspective, thus providing a basis of developing novel and effective methods to diagnose, intervene and treat diseases.
Collapse
Affiliation(s)
- Bai-Rong Chen
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100083, China
| | - Ting Wu
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100083, China
| | - Ting-Hui Chen
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100083, China
| | - Yun Wang
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100083, China
- PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| |
Collapse
|
37
|
Kwok AJ, Lu J, Huang J, Ip BY, Mok VCT, Lai HM, Ko H. High-resolution omics of vascular ageing and inflammatory pathways in neurodegeneration. Semin Cell Dev Biol 2024; 155:30-49. [PMID: 37380595 DOI: 10.1016/j.semcdb.2023.06.005] [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: 04/29/2023] [Accepted: 06/07/2023] [Indexed: 06/30/2023]
Abstract
High-resolution omics, particularly single-cell and spatial transcriptomic profiling, are rapidly enhancing our comprehension of the normal molecular diversity of gliovascular cells, as well as their age-related changes that contribute to neurodegeneration. With more omic profiling studies being conducted, it is becoming increasingly essential to synthesise valuable information from the rapidly accumulating findings. In this review, we present an overview of the molecular features of neurovascular and glial cells that have been recently discovered through omic profiling, with a focus on those that have potentially significant functional implications and/or show cross-species differences between human and mouse, and that are linked to vascular deficits and inflammatory pathways in ageing and neurodegenerative disorders. Additionally, we highlight the translational applications of omic profiling, and discuss omic-based strategies to accelerate biomarker discovery and facilitate disease course-modifying therapeutics development for neurodegenerative conditions.
Collapse
Affiliation(s)
- Andrew J Kwok
- Division of Neurology, Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Margaret K. L. Cheung Research Centre for Management of Parkinsonism, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Lau Tat-chuen Research Centre of Brain Degenerative Diseases in Chinese, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Gerald Choa Neuroscience Institute, The Chinese University of Hong Kong, Hong Kong SAR, China.
| | - Jianning Lu
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Department of Psychiatry, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Gerald Choa Neuroscience Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Junzhe Huang
- Division of Neurology, Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Margaret K. L. Cheung Research Centre for Management of Parkinsonism, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Lau Tat-chuen Research Centre of Brain Degenerative Diseases in Chinese, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Gerald Choa Neuroscience Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Bonaventure Y Ip
- Division of Neurology, Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Margaret K. L. Cheung Research Centre for Management of Parkinsonism, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Lau Tat-chuen Research Centre of Brain Degenerative Diseases in Chinese, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Gerald Choa Neuroscience Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Vincent C T Mok
- Division of Neurology, Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Margaret K. L. Cheung Research Centre for Management of Parkinsonism, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Lau Tat-chuen Research Centre of Brain Degenerative Diseases in Chinese, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Gerald Choa Neuroscience Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Hei Ming Lai
- Division of Neurology, Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Margaret K. L. Cheung Research Centre for Management of Parkinsonism, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Department of Psychiatry, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Gerald Choa Neuroscience Institute, The Chinese University of Hong Kong, Hong Kong SAR, China.
| | - Ho Ko
- Division of Neurology, Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Margaret K. L. Cheung Research Centre for Management of Parkinsonism, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Lau Tat-chuen Research Centre of Brain Degenerative Diseases in Chinese, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Department of Psychiatry, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Gerald Choa Neuroscience Institute, The Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
38
|
Ruiz-Fernández I, Sánchez-Díaz R, Ortega-Sollero E, Martín P. Update on the role of T cells in cognitive impairment. Br J Pharmacol 2024; 181:799-815. [PMID: 37559406 DOI: 10.1111/bph.16214] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 07/03/2023] [Accepted: 08/03/2023] [Indexed: 08/11/2023] Open
Abstract
The central nervous system (CNS) has long been considered an immune-privileged site, with minimal interaction between immune cells, particularly of the adaptive immune system. Previously, the presence of immune cells in this organ was primarily linked to events involving disruption of the blood-brain barrier (BBB) or inflammation. However, current research has shown that immune cells are found patrolling CNS under homeostatic conditions. Specifically, T cells of the adaptive immune system are able to cross the BBB and are associated with ageing and cognitive impairment. In addition, T-cell infiltration has been observed in pathological conditions, where inflammation correlates with poor prognosis. Despite ongoing research, the role of this population in the ageing brain under both physiological and pathological conditions is not yet fully understood. In this review, we provide an overview of the interactions between T cells and other immune and CNS parenchymal cells, and examine the molecular mechanisms by which these interactions may contribute to normal brain function and the scenarios in which disruption of these connections lead to cognitive impairment. A comprehensive understanding of the role of T cells in the ageing brain and the underlying molecular pathways under normal conditions could pave the way for new research to better understand brain disorders. LINKED ARTICLES: This article is part of a themed issue From Alzheimer's Disease to Vascular Dementia: Different Roads Leading to Cognitive Decline. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v181.6/issuetoc.
Collapse
Affiliation(s)
| | - Raquel Sánchez-Díaz
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBER-CV), Madrid, Spain
| | | | - Pilar Martín
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBER-CV), Madrid, Spain
| |
Collapse
|
39
|
Sun ED, Ma R, Navarro Negredo P, Brunet A, Zou J. TISSUE: uncertainty-calibrated prediction of single-cell spatial transcriptomics improves downstream analyses. Nat Methods 2024; 21:444-454. [PMID: 38347138 DOI: 10.1038/s41592-024-02184-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 01/12/2024] [Indexed: 02/27/2024]
Abstract
Whole-transcriptome spatial profiling of genes at single-cell resolution remains a challenge. To address this limitation, spatial gene expression prediction methods have been developed to infer the spatial expression of unmeasured transcripts, but the quality of these predictions can vary greatly. Here we present Transcript Imputation with Spatial Single-cell Uncertainty Estimation (TISSUE) as a general framework for estimating uncertainty for spatial gene expression predictions and providing uncertainty-aware methods for downstream inference. Leveraging conformal inference, TISSUE provides well-calibrated prediction intervals for predicted expression values across 11 benchmark datasets. Moreover, it consistently reduces the false discovery rate for differential gene expression analysis, improves clustering and visualization of predicted spatial transcriptomics and improves the performance of supervised learning models trained on predicted gene expression profiles. Applying TISSUE to a MERFISH spatial transcriptomics dataset of the adult mouse subventricular zone, we identified subtypes within the neural stem cell lineage and developed subtype-specific regional classifiers.
Collapse
Affiliation(s)
- Eric D Sun
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Rong Ma
- Department of Statistics, Stanford University, Stanford, CA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | | | - Anne Brunet
- Department of Genetics, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
- Glenn Center for the Biology of Aging, Stanford University, Stanford, CA, USA
| | - James Zou
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA.
| |
Collapse
|
40
|
Fang W, Liu X, Maiga M, Cao W, Mu Y, Yan Q, Zhu Q. Digital PCR for Single-Cell Analysis. BIOSENSORS 2024; 14:64. [PMID: 38391982 PMCID: PMC10886679 DOI: 10.3390/bios14020064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 01/18/2024] [Accepted: 01/22/2024] [Indexed: 02/24/2024]
Abstract
Single-cell analysis provides an overwhelming strategy for revealing cellular heterogeneity and new perspectives for understanding the biological function and disease mechanism. Moreover, it promotes the basic and clinical research in many fields at a single-cell resolution. A digital polymerase chain reaction (dPCR) is an absolute quantitative analysis technology with high sensitivity and precision for DNA/RNA or protein. With the development of microfluidic technology, digital PCR has been used to achieve absolute quantification of single-cell gene expression and single-cell proteins. For single-cell specific-gene or -protein detection, digital PCR has shown great advantages. So, this review will introduce the significance and process of single-cell analysis, including single-cell isolation, single-cell lysis, and single-cell detection methods, mainly focusing on the microfluidic single-cell digital PCR technology and its biological application at a single-cell level. The challenges and opportunities for the development of single-cell digital PCR are also discussed.
Collapse
Affiliation(s)
- Weibo Fang
- Research Center for Analytical Instrumentation, Institute of Cyber-Systems and Control, College of Control Science and Engineering, State Key Laboratory of Industrial Control Technology, Zhejiang University, Hangzhou 310027, China; (W.F.); (X.L.); (M.M.); (W.C.); (Y.M.)
| | - Xudong Liu
- Research Center for Analytical Instrumentation, Institute of Cyber-Systems and Control, College of Control Science and Engineering, State Key Laboratory of Industrial Control Technology, Zhejiang University, Hangzhou 310027, China; (W.F.); (X.L.); (M.M.); (W.C.); (Y.M.)
| | - Mariam Maiga
- Research Center for Analytical Instrumentation, Institute of Cyber-Systems and Control, College of Control Science and Engineering, State Key Laboratory of Industrial Control Technology, Zhejiang University, Hangzhou 310027, China; (W.F.); (X.L.); (M.M.); (W.C.); (Y.M.)
| | - Wenjian Cao
- Research Center for Analytical Instrumentation, Institute of Cyber-Systems and Control, College of Control Science and Engineering, State Key Laboratory of Industrial Control Technology, Zhejiang University, Hangzhou 310027, China; (W.F.); (X.L.); (M.M.); (W.C.); (Y.M.)
| | - Ying Mu
- Research Center for Analytical Instrumentation, Institute of Cyber-Systems and Control, College of Control Science and Engineering, State Key Laboratory of Industrial Control Technology, Zhejiang University, Hangzhou 310027, China; (W.F.); (X.L.); (M.M.); (W.C.); (Y.M.)
| | - Qiang Yan
- Department of Hepatobiliary and Pancreatic Surgery, Huzhou Central Hospital, Huzhou Key Laboratory of Intelligent and Digital Precision Surgery, Department of General Surgery, Affiliated Huzhou Hospital, School of Medicine, Zhejiang University, Huzhou 313000, China
| | - Qiangyuan Zhu
- Research Center for Analytical Instrumentation, Institute of Cyber-Systems and Control, College of Control Science and Engineering, State Key Laboratory of Industrial Control Technology, Zhejiang University, Hangzhou 310027, China; (W.F.); (X.L.); (M.M.); (W.C.); (Y.M.)
- Huzhou Institute of Zhejiang University, Huzhou 313002, China
| |
Collapse
|
41
|
Liu Y, Tan Y, Zhang Z, Yi M, Zhu L, Peng W. The interaction between ageing and Alzheimer's disease: insights from the hallmarks of ageing. Transl Neurodegener 2024; 13:7. [PMID: 38254235 PMCID: PMC10804662 DOI: 10.1186/s40035-024-00397-x] [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: 09/13/2023] [Revised: 12/31/2023] [Accepted: 01/02/2024] [Indexed: 01/24/2024] Open
Abstract
Ageing is a crucial risk factor for Alzheimer's disease (AD) and is characterised by systemic changes in both intracellular and extracellular microenvironments that affect the entire body instead of a single organ. Understanding the specific mechanisms underlying the role of ageing in disease development can facilitate the treatment of ageing-related diseases, such as AD. Signs of brain ageing have been observed in both AD patients and animal models. Alleviating the pathological changes caused by brain ageing can dramatically ameliorate the amyloid beta- and tau-induced neuropathological and memory impairments, indicating that ageing plays a crucial role in the pathophysiological process of AD. In this review, we summarize the impact of several age-related factors on AD and propose that preventing pathological changes caused by brain ageing is a promising strategy for improving cognitive health.
Collapse
Affiliation(s)
- Yuqing Liu
- Department of Integrated Traditional Chinese and Western Medicine, The Second Xiangya Hospital, Central South University, No.139 Middle Renmin Road, Changsha, 410011, Hunan, People's Republic of China
- National Clinical Research Center for Metabolic Diseases, Changsha, 410011, People's Republic of China
| | - Yejun Tan
- School of Mathematics, University of Minnesota Twin Cities, Minneapolis, MN, 55455, USA
| | - Zheyu Zhang
- Department of Integrated Traditional Chinese and Western Medicine, The Second Xiangya Hospital, Central South University, No.139 Middle Renmin Road, Changsha, 410011, Hunan, People's Republic of China
- National Clinical Research Center for Metabolic Diseases, Changsha, 410011, People's Republic of China
| | - Min Yi
- Department of Integrated Traditional Chinese and Western Medicine, The Second Xiangya Hospital, Central South University, No.139 Middle Renmin Road, Changsha, 410011, Hunan, People's Republic of China
- National Clinical Research Center for Metabolic Diseases, Changsha, 410011, People's Republic of China
| | - Lemei Zhu
- Academician Workstation, Changsha Medical University, Changsha, 410219, People's Republic of China
| | - Weijun Peng
- Department of Integrated Traditional Chinese and Western Medicine, The Second Xiangya Hospital, Central South University, No.139 Middle Renmin Road, Changsha, 410011, Hunan, People's Republic of China.
- National Clinical Research Center for Metabolic Diseases, Changsha, 410011, People's Republic of China.
| |
Collapse
|
42
|
Flotho M, Amand J, Hirsch P, Grandke F, Wyss-Coray T, Keller A, Kern F. ZEBRA: a hierarchically integrated gene expression atlas of the murine and human brain at single-cell resolution. Nucleic Acids Res 2024; 52:D1089-D1096. [PMID: 37941147 PMCID: PMC10767845 DOI: 10.1093/nar/gkad990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 10/02/2023] [Accepted: 10/16/2023] [Indexed: 11/10/2023] Open
Abstract
The molecular causes and mechanisms of neurodegenerative diseases remain poorly understood. A growing number of single-cell studies have implicated various neural, glial, and immune cell subtypes to affect the mammalian central nervous system in many age-related disorders. Integrating this body of transcriptomic evidence into a comprehensive and reproducible framework poses several computational challenges. Here, we introduce ZEBRA, a large single-cell and single-nucleus RNA-seq database. ZEBRA integrates and normalizes gene expression and metadata from 33 studies, encompassing 4.2 million human and mouse brain cells sampled from 39 brain regions. It incorporates samples from patients with neurodegenerative diseases like Alzheimer's disease, Parkinson's disease, and Multiple sclerosis, as well as samples from relevant mouse models. We employed scVI, a deep probabilistic auto-encoder model, to integrate the samples and curated both cell and sample metadata for downstream analysis. ZEBRA allows for cell-type and disease-specific markers to be explored and compared between sample conditions and brain regions, a cell composition analysis, and gene-wise feature mappings. Our comprehensive molecular database facilitates the generation of data-driven hypotheses, enhancing our understanding of mammalian brain function during aging and disease. The data sets, along with an interactive database are freely available at https://www.ccb.uni-saarland.de/zebra.
Collapse
Affiliation(s)
- Matthias Flotho
- Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, Saarland University Campus, 66123 Saarbrücken, Germany
- Clinical Bioinformatics, Center for Bioinformatics, Saarland University, 66123 Saarbrücken, Germany
| | - Jérémy Amand
- Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, Saarland University Campus, 66123 Saarbrücken, Germany
- Clinical Bioinformatics, Center for Bioinformatics, Saarland University, 66123 Saarbrücken, Germany
| | - Pascal Hirsch
- Clinical Bioinformatics, Center for Bioinformatics, Saarland University, 66123 Saarbrücken, Germany
| | - Friederike Grandke
- Clinical Bioinformatics, Center for Bioinformatics, Saarland University, 66123 Saarbrücken, Germany
| | - Tony Wyss-Coray
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
- The Phil and Penny Knight Initiative for Brain Resilience, Stanford University, Stanford, CA, USA
| | - Andreas Keller
- Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, Saarland University Campus, 66123 Saarbrücken, Germany
- Clinical Bioinformatics, Center for Bioinformatics, Saarland University, 66123 Saarbrücken, Germany
| | - Fabian Kern
- Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, Saarland University Campus, 66123 Saarbrücken, Germany
- Clinical Bioinformatics, Center for Bioinformatics, Saarland University, 66123 Saarbrücken, Germany
| |
Collapse
|
43
|
Zhao Y, Ma J, Ding G, Wang Y, Yu H, Cheng X. Astragalus polysaccharides promote neural stem cells-derived oligodendrogenesis through attenuating CD8 +T cell infiltration in experimental autoimmune encephalomyelitis. Int Immunopharmacol 2024; 126:111303. [PMID: 38043269 DOI: 10.1016/j.intimp.2023.111303] [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: 07/25/2023] [Revised: 10/31/2023] [Accepted: 11/24/2023] [Indexed: 12/05/2023]
Abstract
Endogenous neural stem cells (NSCs) have the potential to generate remyelinating oligodendrocytes, which play an important role in multiple sclerosis (MS). However, the differentiation of NSCs into oligodendrocytes is insufficient, which is considered a major cause of remyelination failure. Our previous work reported that Astragalus polysaccharides (APS) had a neuroprotective effect on experimental autoimmune encephalomyelitis (EAE) mice. However, it remains unclear whether APS regulate NSCs differentiation in EAE mice. In this study, our data illustrated that APS administration could promote NSCs in the subventricular zone (SVZ) to differentiate into oligodendrocytes. Furthermore, we found that APS significantly improved neuroinflammation and inhibited CD8+T cell infiltration into SVZ of EAE mice. We also found that MOG35-55-specific CD8+T cells suppressed NSCs differentiation into oligodendrocytes by secreting IFN-γ, and APS facilitated the differentiation of NSCs into oligodendrocytes which was related to decreased IFN-γ secretion. In addition, APS treatment did not show a better effect on the NSCs-derived oligodendrogenesis after CD8+T cell depletion. This present study demonstrated that APS alleviated neuroinflammation and CD8+T cell infiltration into SVZ to induce oligodendroglial differentiation, and thus exerted neuroprotective effect. Our findings revealed that reducing the infiltration of CD8+T cells might contribute to enhancing NSCs-derived neurogenesis. And APS might be a promising drug candidate to treat MS.
Collapse
Affiliation(s)
- Yan Zhao
- Institute of Clinical Immunology, Yue-yang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Jinyun Ma
- Institute of Clinical Immunology, Yue-yang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Guiqing Ding
- Institute of Clinical Immunology, Yue-yang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Yuanhua Wang
- Institute of Clinical Immunology, Yue-yang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Hua Yu
- Institute of Clinical Immunology, Yue-yang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Xiaodong Cheng
- Institute of Clinical Immunology, Yue-yang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China.
| |
Collapse
|
44
|
Kent SA, Miron VE. Microglia regulation of central nervous system myelin health and regeneration. Nat Rev Immunol 2024; 24:49-63. [PMID: 37452201 DOI: 10.1038/s41577-023-00907-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/13/2023] [Indexed: 07/18/2023]
Abstract
Microglia are resident macrophages of the central nervous system that have key functions in its development, homeostasis and response to damage and infection. Although microglia have been increasingly implicated in contributing to the pathology that underpins neurological dysfunction and disease, they also have crucial roles in neurological homeostasis and regeneration. This includes regulation of the maintenance and regeneration of myelin, the membrane that surrounds neuronal axons, which is required for axonal health and function. Myelin is damaged with normal ageing and in several neurodegenerative diseases, such as multiple sclerosis and Alzheimer disease. Given the lack of approved therapies targeting myelin maintenance or regeneration, it is imperative to understand the mechanisms by which microglia support and restore myelin health to identify potential therapeutic approaches. However, the mechanisms by which microglia regulate myelin loss or integrity are still being uncovered. In this Review, we discuss recent work that reveals the changes in white matter with ageing and neurodegenerative disease, how this relates to microglia dynamics during myelin damage and regeneration, and factors that influence the regenerative functions of microglia.
Collapse
Affiliation(s)
- Sarah A Kent
- UK Dementia Research Institute at The University of Edinburgh, Edinburgh, UK
- Centre for Discovery Brain Sciences, Chancellor's Building, The University of Edinburgh, Edinburgh, UK
| | - Veronique E Miron
- UK Dementia Research Institute at The University of Edinburgh, Edinburgh, UK.
- Centre for Discovery Brain Sciences, Chancellor's Building, The University of Edinburgh, Edinburgh, UK.
- Barlo Multiple Sclerosis Centre, St Michael's Hospital, Toronto, Ontario, Canada.
- Keenan Research Centre for Biomedical Science, St Michael's Hospital, Toronto, Ontario, Canada.
- Department of Immunology, The University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
45
|
Chen Y, Yang S, Yu K, Zhang J, Wu M, Zheng Y, Zhu Y, Dai J, Wang C, Zhu X, Dai Y, Sun Y, Wu T, Wang S. Spatial omics: An innovative frontier in aging research. Ageing Res Rev 2024; 93:102158. [PMID: 38056503 DOI: 10.1016/j.arr.2023.102158] [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: 08/28/2023] [Revised: 11/25/2023] [Accepted: 12/01/2023] [Indexed: 12/08/2023]
Abstract
Disentangling the impact of aging on health and disease has become critical as population aging progresses rapidly. Studying aging at the molecular level is complicated by the diverse aging profiles and dynamics. However, the examination of cellular states within aging tissues in situ is hampered by the lack of high-resolution spatial data. Emerging spatial omics technologies facilitate molecular and spatial analysis of tissues, providing direct access to precise information on various functional regions and serving as a favorable tool for unraveling the heterogeneity of aging. In this review, we summarize the recent advances in spatial omics application in multi-organ aging research, which has enhanced the understanding of aging mechanisms from multiple standpoints. We also discuss the main challenges in spatial omics research to date, the opportunities for further developing the technology, and the potential applications of spatial omics in aging and aging-related diseases.
Collapse
Affiliation(s)
- Ying Chen
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China; Ministry of Education, Key Laboratory of Cancer Invasion and Metastasis, Wuhan, China
| | - Shuhao Yang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China; Ministry of Education, Key Laboratory of Cancer Invasion and Metastasis, Wuhan, China
| | - Kaixu Yu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinjin Zhang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China; Ministry of Education, Key Laboratory of Cancer Invasion and Metastasis, Wuhan, China
| | - Meng Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China; Ministry of Education, Key Laboratory of Cancer Invasion and Metastasis, Wuhan, China
| | - Yongqiang Zheng
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Centre, Sun Yat-sen University, Guangzhou, China
| | - Yun Zhu
- Department of Internal Medicine, Southern Illinois University School of Medicine, 801 N. Rutledge, P.O. Box 19628, Springfield, IL 62702, USA
| | - Jun Dai
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China; Ministry of Education, Key Laboratory of Cancer Invasion and Metastasis, Wuhan, China
| | - Chunyan Wang
- College of Science & Engineering Jinan University, Guangzhou, China
| | - Xiaoran Zhu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China; Ministry of Education, Key Laboratory of Cancer Invasion and Metastasis, Wuhan, China
| | - Yun Dai
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China; Ministry of Education, Key Laboratory of Cancer Invasion and Metastasis, Wuhan, China
| | - Yunhong Sun
- Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tong Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China; Ministry of Education, Key Laboratory of Cancer Invasion and Metastasis, Wuhan, China.
| | - Shixuan Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China; Ministry of Education, Key Laboratory of Cancer Invasion and Metastasis, Wuhan, China.
| |
Collapse
|
46
|
Šimončičová E, Henderson Pekarik K, Vecchiarelli HA, Lauro C, Maggi L, Tremblay MÈ. Adult Neurogenesis, Learning and Memory. ADVANCES IN NEUROBIOLOGY 2024; 37:221-242. [PMID: 39207695 DOI: 10.1007/978-3-031-55529-9_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Neural plasticity can be defined as the ability of neural circuits to be shaped by external and internal factors. It provides the brain with a capacity for functional and morphological remodelling, with many lines of evidence indicating that these changes are vital for learning and memory formation. The basis of this brain plasticity resides in activity- and experience-driven modifications of synaptic strength, including synaptic formation, elimination or weakening, as well as of modulation of neuronal population, which drive the structural reorganization of neural networks. Recent evidence indicates that brain-resident glial cells actively participate in these processes, suggesting that mechanisms underlying plasticity in the brain are multifaceted. Establishing the 'tripartite' synapse, the role of astrocytes in modulating synaptic transmission in response to neuronal activity was recognized first. Further redefinition of the synapse as 'quad-partite' followed to acknowledge the contribution of microglia which were revealed to affect numerous brain functions via dynamic interactions with synapses, acting as 'synaptic sensors' that respond to neuronal activity and neurotransmitter release, as well as crosstalk with astrocytes. Early studies identified microglial ability to dynamically survey their local brain environment and established their integral role in the active interfacing of environmental stimuli (both internal and external), with brain plasticity and remodelling. Following the introduction to neurogenesis, this chapter details the role that microglia play in regulating neurogenesis in adulthood, specifically as it relates to learning and memory, as well as factors involved in modulation of microglia. Further, a microglial perspective is introduced for the context of environmental enrichment impact on neurogenesis, learning and memory across states of stress, ageing, disease and injury.
Collapse
Affiliation(s)
- Eva Šimončičová
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | | | | | - Clotilde Lauro
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Laura Maggi
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada.
| |
Collapse
|
47
|
Kimura K, Nishigori R, Hamatani M, Sawamura M, Ashida S, Fujii C, Takata M, Lin Y, Sato W, Okamoto T, Kuzuya A, Takahashi R, Yamamura T, Kondo T. Resident Memory-like CD8 + T Cells Are Involved in Chronic Inflammatory and Neurodegenerative Diseases in the CNS. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2024; 11:e200172. [PMID: 37949669 PMCID: PMC10691221 DOI: 10.1212/nxi.0000000000200172] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 08/29/2023] [Indexed: 11/12/2023]
Abstract
BACKGROUND AND OBJECTIVES Resident memory T (Trm) cells are a unique population that can survive and function in a compartmentalized tissue with inflammatory potential. We aim to investigate the alteration of Trm population in acute/chronic inflammatory and neurodegenerative diseases in the CNS. METHODS The frequencies of CD4+ and CD8+ T cells expressing both CD69 and CD103, the markers for Trm cells, were quantified in the peripheral blood and CSF (n = 80 and 44, respectively) in a cross-sectional manner. The transcriptional profile of Trm-like population in the CSF was further analyzed using a public single-cell dataset. RESULTS The frequency of CD69+CD103+CD8+ T cells was strikingly higher in the CSF than in the peripheral blood (among memory fraction, 13.5% vs 0.11%, difference (mean [SE]): 13.4% [2.9]). This CD69+CD103+CD8+ T-cell population was increased in the CSF from patients with chronic inflammatory diseases including multiple sclerosis and with neurodegenerative diseases such as Parkinson disease and Alzheimer disease compared with controls (11.5%, 13.0%, 8.1% vs 2.9%, respectively). By contrast, the frequency was not altered in acute inflammatory conditions in the CNS (4.0%). Single-cell RNAseq analysis confirmed Trm signature in CD69+CD103+CD8+ T cells in the CSF, supporting their Trm-like phenotype, which was not clear in controls. DISCUSSION Collectively, an increase in CD69+CD103+CD8+ Trm-like population in the CSF is related with both chronic neuroinflammatory and some neurodegenerative diseases in the CNS, suggesting a partially shared pathology in these diseases.
Collapse
Affiliation(s)
- Kimitoshi Kimura
- From the Department of Neurology (K.K., R.N., M.H., M.S., M.T., A.K., R.T.), Kyoto University Graduate School of Medicine; Department of Immunology (K.K., Y.L., W.S., T.O., T.Y.), National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira; Institute for the Advanced Study of Human Biology (M.H.), Kyoto University; Department of Neurology (S.A., C.F.), Graduate School of Medical Science, Kyoto Prefectural University of Medicine; and Department of Neurology (C.F., T.K.), Kansai Medical University Medical Center, Moriguchi, Japan.
| | - Ryusei Nishigori
- From the Department of Neurology (K.K., R.N., M.H., M.S., M.T., A.K., R.T.), Kyoto University Graduate School of Medicine; Department of Immunology (K.K., Y.L., W.S., T.O., T.Y.), National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira; Institute for the Advanced Study of Human Biology (M.H.), Kyoto University; Department of Neurology (S.A., C.F.), Graduate School of Medical Science, Kyoto Prefectural University of Medicine; and Department of Neurology (C.F., T.K.), Kansai Medical University Medical Center, Moriguchi, Japan
| | - Mio Hamatani
- From the Department of Neurology (K.K., R.N., M.H., M.S., M.T., A.K., R.T.), Kyoto University Graduate School of Medicine; Department of Immunology (K.K., Y.L., W.S., T.O., T.Y.), National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira; Institute for the Advanced Study of Human Biology (M.H.), Kyoto University; Department of Neurology (S.A., C.F.), Graduate School of Medical Science, Kyoto Prefectural University of Medicine; and Department of Neurology (C.F., T.K.), Kansai Medical University Medical Center, Moriguchi, Japan
| | - Masanori Sawamura
- From the Department of Neurology (K.K., R.N., M.H., M.S., M.T., A.K., R.T.), Kyoto University Graduate School of Medicine; Department of Immunology (K.K., Y.L., W.S., T.O., T.Y.), National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira; Institute for the Advanced Study of Human Biology (M.H.), Kyoto University; Department of Neurology (S.A., C.F.), Graduate School of Medical Science, Kyoto Prefectural University of Medicine; and Department of Neurology (C.F., T.K.), Kansai Medical University Medical Center, Moriguchi, Japan
| | - Shinji Ashida
- From the Department of Neurology (K.K., R.N., M.H., M.S., M.T., A.K., R.T.), Kyoto University Graduate School of Medicine; Department of Immunology (K.K., Y.L., W.S., T.O., T.Y.), National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira; Institute for the Advanced Study of Human Biology (M.H.), Kyoto University; Department of Neurology (S.A., C.F.), Graduate School of Medical Science, Kyoto Prefectural University of Medicine; and Department of Neurology (C.F., T.K.), Kansai Medical University Medical Center, Moriguchi, Japan
| | - Chihiro Fujii
- From the Department of Neurology (K.K., R.N., M.H., M.S., M.T., A.K., R.T.), Kyoto University Graduate School of Medicine; Department of Immunology (K.K., Y.L., W.S., T.O., T.Y.), National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira; Institute for the Advanced Study of Human Biology (M.H.), Kyoto University; Department of Neurology (S.A., C.F.), Graduate School of Medical Science, Kyoto Prefectural University of Medicine; and Department of Neurology (C.F., T.K.), Kansai Medical University Medical Center, Moriguchi, Japan
| | - Masaki Takata
- From the Department of Neurology (K.K., R.N., M.H., M.S., M.T., A.K., R.T.), Kyoto University Graduate School of Medicine; Department of Immunology (K.K., Y.L., W.S., T.O., T.Y.), National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira; Institute for the Advanced Study of Human Biology (M.H.), Kyoto University; Department of Neurology (S.A., C.F.), Graduate School of Medical Science, Kyoto Prefectural University of Medicine; and Department of Neurology (C.F., T.K.), Kansai Medical University Medical Center, Moriguchi, Japan
| | - Youwei Lin
- From the Department of Neurology (K.K., R.N., M.H., M.S., M.T., A.K., R.T.), Kyoto University Graduate School of Medicine; Department of Immunology (K.K., Y.L., W.S., T.O., T.Y.), National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira; Institute for the Advanced Study of Human Biology (M.H.), Kyoto University; Department of Neurology (S.A., C.F.), Graduate School of Medical Science, Kyoto Prefectural University of Medicine; and Department of Neurology (C.F., T.K.), Kansai Medical University Medical Center, Moriguchi, Japan
| | - Wakiro Sato
- From the Department of Neurology (K.K., R.N., M.H., M.S., M.T., A.K., R.T.), Kyoto University Graduate School of Medicine; Department of Immunology (K.K., Y.L., W.S., T.O., T.Y.), National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira; Institute for the Advanced Study of Human Biology (M.H.), Kyoto University; Department of Neurology (S.A., C.F.), Graduate School of Medical Science, Kyoto Prefectural University of Medicine; and Department of Neurology (C.F., T.K.), Kansai Medical University Medical Center, Moriguchi, Japan
| | - Tomoko Okamoto
- From the Department of Neurology (K.K., R.N., M.H., M.S., M.T., A.K., R.T.), Kyoto University Graduate School of Medicine; Department of Immunology (K.K., Y.L., W.S., T.O., T.Y.), National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira; Institute for the Advanced Study of Human Biology (M.H.), Kyoto University; Department of Neurology (S.A., C.F.), Graduate School of Medical Science, Kyoto Prefectural University of Medicine; and Department of Neurology (C.F., T.K.), Kansai Medical University Medical Center, Moriguchi, Japan
| | - Akira Kuzuya
- From the Department of Neurology (K.K., R.N., M.H., M.S., M.T., A.K., R.T.), Kyoto University Graduate School of Medicine; Department of Immunology (K.K., Y.L., W.S., T.O., T.Y.), National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira; Institute for the Advanced Study of Human Biology (M.H.), Kyoto University; Department of Neurology (S.A., C.F.), Graduate School of Medical Science, Kyoto Prefectural University of Medicine; and Department of Neurology (C.F., T.K.), Kansai Medical University Medical Center, Moriguchi, Japan
| | - Ryosuke Takahashi
- From the Department of Neurology (K.K., R.N., M.H., M.S., M.T., A.K., R.T.), Kyoto University Graduate School of Medicine; Department of Immunology (K.K., Y.L., W.S., T.O., T.Y.), National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira; Institute for the Advanced Study of Human Biology (M.H.), Kyoto University; Department of Neurology (S.A., C.F.), Graduate School of Medical Science, Kyoto Prefectural University of Medicine; and Department of Neurology (C.F., T.K.), Kansai Medical University Medical Center, Moriguchi, Japan
| | - Takashi Yamamura
- From the Department of Neurology (K.K., R.N., M.H., M.S., M.T., A.K., R.T.), Kyoto University Graduate School of Medicine; Department of Immunology (K.K., Y.L., W.S., T.O., T.Y.), National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira; Institute for the Advanced Study of Human Biology (M.H.), Kyoto University; Department of Neurology (S.A., C.F.), Graduate School of Medical Science, Kyoto Prefectural University of Medicine; and Department of Neurology (C.F., T.K.), Kansai Medical University Medical Center, Moriguchi, Japan
| | - Takayuki Kondo
- From the Department of Neurology (K.K., R.N., M.H., M.S., M.T., A.K., R.T.), Kyoto University Graduate School of Medicine; Department of Immunology (K.K., Y.L., W.S., T.O., T.Y.), National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira; Institute for the Advanced Study of Human Biology (M.H.), Kyoto University; Department of Neurology (S.A., C.F.), Graduate School of Medical Science, Kyoto Prefectural University of Medicine; and Department of Neurology (C.F., T.K.), Kansai Medical University Medical Center, Moriguchi, Japan
| |
Collapse
|
48
|
Church KA, Cardona AE, Hopp SC. Roles in Innate Immunity. ADVANCES IN NEUROBIOLOGY 2024; 37:263-286. [PMID: 39207697 DOI: 10.1007/978-3-031-55529-9_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Microglia are best known as the resident phagocytes of the central nervous system (CNS). As a resident brain immune cell population, microglia play key roles during the initiation, propagation, and resolution of inflammation. The discovery of resident adaptive immune cells in the CNS has unveiled a relationship between microglia and adaptive immune cells for CNS immune-surveillance during health and disease. The interaction of microglia with elements of the peripheral immune system and other CNS resident cells mediates a fine balance between neuroprotection and tissue damage. In this chapter, we highlight the innate immune properties of microglia, with a focus on how pattern recognition receptors, inflammatory signaling cascades, phagocytosis, and the interaction between microglia and adaptive immune cells regulate events that initiate an inflammatory or neuroprotective response within the CNS that modulates immune-mediated disease exacerbation or resolution.
Collapse
Affiliation(s)
- Kaira A Church
- Department of Molecular Microbiology & Immunology, The University of Texas at San Antonio, San Antonio, TX, USA
- South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX, USA
| | - Astrid E Cardona
- Department of Molecular Microbiology & Immunology, The University of Texas at San Antonio, San Antonio, TX, USA
- South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX, USA
| | - Sarah C Hopp
- Department of Pharmacology, Biggs Institute for Alzheimer's and Neurodegenerative Disease, The University of Texas Health Science Center San Antonio, San Antonio, TX, USA.
| |
Collapse
|
49
|
Chen Z, Ford K, Islam M, Iamsawat S, Davis B, Weiss C, Schwulst S. ANTI-CD49D ANTIBODY TREATMENT IMPROVES SURVIVAL AND ATTENUATES NEUROCOGNITIVE DEFICITS AFTER TRAUMATIC BRAIN INJURY IN AGED MICE. Shock 2024; 61:112-119. [PMID: 38010092 PMCID: PMC10841910 DOI: 10.1097/shk.0000000000002256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
ABSTRACT Patients 65 years and older account for an increasing proportion of traumatic brain injury (TBI) patients. Aged TBI patients experience increased morbidity and mortality compared with young TBI patients. We previously demonstrated a marked accumulation of CD8 + T-cells within the brains of aged TBI mice compared with young TBI mice. Therefore, we hypothesized that blocking peripheral T-cell infiltration into the injured brain would improve neurocognitive outcomes in aged mice after TBI. Young and aged male C57BL/6 mice underwent TBI via controlled cortical impact versus sham injury. Two hours after injuries, mice received an anti-CD49d antibody (aCD49d Ab) to block peripheral lymphocyte infiltration or its isotype control. Dosing was repeated every 2 weeks. Mortality was tracked. Neurocognitive testing for anxiety, associative learning, and memory was assessed. Motor function was evaluated. Plasma was collected for cytokine analysis. Flow cytometry was used to phenotype different immune cells within the brains. Consequently, aCD49d Ab treatment significantly improved post-TBI survival, anxiety level, associative learning, memory, and motor function in aged mice 2 months after TBI compared with isotype control treated mice. aCD49d Ab treatment augmented T H 2 response in the plasma of aged mice 2 months after TBI compared with isotype control-treated mice. Notably, aCD49d Ab treatment significantly reduced activated CD8 + cytotoxic T-cells within aged mouse brains after TBI. Contrastingly, no difference was detected in young mice after aCD49d Ab treatment. Collectively, aCD49 Ab treatment reduced T-cells in the injured brain, improved survival, and attenuated neurocognitive and gait deficits. Hence, aCD49d Ab may be a promising therapeutic intervention in aged TBI subjects-a population often excluded in TBI clinical trials.
Collapse
Affiliation(s)
- Zhangying Chen
- Department of Surgery, Division of Trauma and Critical
Care, Northwestern University Feinberg School of Medicine, Chicago IL, USA
- Driskill Graduate Program in Life Sciences, Northwestern
University Feinberg School of Medicine, Chicago IL, USA
| | - Kacie Ford
- Department of Surgery, Division of Trauma and Critical
Care, Northwestern University Feinberg School of Medicine, Chicago IL, USA
| | - Mecca Islam
- Department of Surgery, Division of Trauma and Critical
Care, Northwestern University Feinberg School of Medicine, Chicago IL, USA
| | - Supinya Iamsawat
- Department of Microbiology and Immunology, Northwestern
University Feinberg School of Medicine, Chicago, IL
| | - Booker Davis
- Department of Surgery, Division of Trauma and Critical
Care, Northwestern University Feinberg School of Medicine, Chicago IL, USA
| | - Craig Weiss
- Department of Neuroscience, Feinberg School of Medicine,
Northwestern University, Chicago, Illinois, USA
| | - Steven Schwulst
- Department of Surgery, Division of Trauma and Critical
Care, Northwestern University Feinberg School of Medicine, Chicago IL, USA
| |
Collapse
|
50
|
Yu D, Li M, Linghu G, Hu Y, Hajdarovic KH, Wang A, Singh R, Webb AE. CellBiAge: Improved single-cell age classification using data binarization. Cell Rep 2023; 42:113500. [PMID: 38032797 PMCID: PMC10791072 DOI: 10.1016/j.celrep.2023.113500] [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: 05/04/2023] [Revised: 10/20/2023] [Accepted: 11/13/2023] [Indexed: 12/02/2023] Open
Abstract
Aging is a major risk factor for many diseases. Accurate methods for predicting age in specific cell types are essential to understand the heterogeneity of aging and to assess rejuvenation strategies. However, classifying organismal age at single-cell resolution using transcriptomics is challenging due to sparsity and noise. Here, we developed CellBiAge, a robust and easy-to-implement machine learning pipeline, to classify the age of single cells in the mouse brain using single-cell transcriptomics. We show that binarization of gene expression values for the top highly variable genes significantly improved test performance across different models, techniques, sexes, and brain regions, with potential age-related genes identified for model prediction. Additionally, we demonstrate CellBiAge's ability to capture exercise-induced rejuvenation in neural stem cells. This study provides a broadly applicable approach for robust classification of organismal age of single cells in the mouse brain, which may aid in understanding the aging process and evaluating rejuvenation methods.
Collapse
Affiliation(s)
- Doudou Yu
- Molecular Biology, Cell Biology, and Biochemistry Graduate Program, Brown University, Providence, RI 02912, USA; Data Science Institute, Brown University, Providence, RI 02912, USA
| | - Manlin Li
- Data Science Institute, Brown University, Providence, RI 02912, USA
| | - Guanjie Linghu
- Data Science Institute, Brown University, Providence, RI 02912, USA
| | - Yihuan Hu
- Data Science Institute, Brown University, Providence, RI 02912, USA
| | | | - An Wang
- Department of Applied Mathematics & Statistics, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Ritambhara Singh
- Department of Computer Science, Brown University, Providence, RI 02912, USA; Center for Computational Molecular Biology, Brown University, Providence, RI 02912, USA.
| | - Ashley E Webb
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI 02912, USA; Center on the Biology of Aging, Brown University, Providence, RI 02912, USA; Carney Institute for Brain Science, Brown University, Providence, RI 02912, USA; Center for Translational Neuroscience, Brown University, Providence, RI 02912, USA.
| |
Collapse
|