1
|
Kim E, Tanzi RE, Choi SH. Therapeutic potential of exercise-hormone irisin in Alzheimer's disease. Neural Regen Res 2025; 20:1555-1564. [PMID: 38993140 DOI: 10.4103/nrr.nrr-d-24-00098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 06/04/2024] [Indexed: 07/13/2024] Open
Abstract
Irisin is a myokine that is generated by cleavage of the membrane protein fibronectin type III domain-containing protein 5 (FNDC5) in response to physical exercise. Studies reveal that irisin/FNDC5 has neuroprotective functions against Alzheimer's disease, the most common form of dementia in the elderly, by improving cognitive function and reducing amyloid-β and tau pathologies as well as neuroinflammation in cell culture or animal models of Alzheimer's disease. Although current and ongoing studies on irisin/FNDC5 show promising results, further mechanistic studies are required to clarify its potential as a meaningful therapeutic target for alleviating Alzheimer's disease. We recently found that irisin treatment reduces amyloid-β pathology by increasing the activity/levels of amyloid-β-degrading enzyme neprilysin secreted from astrocytes. Herein, we present an overview of irisin/FNDC5's protective roles and mechanisms against Alzheimer's disease.
Collapse
Affiliation(s)
- Eunhee Kim
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
- McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA, USA
| | - Rudolph E Tanzi
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
- McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA, USA
| | - Se Hoon Choi
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
- McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
2
|
Elroy-Stein O. Effective extraction of polyribosomes from astrocytes enables future discoveries on translation regulation. Neural Regen Res 2025; 20:1083-1084. [PMID: 38989942 PMCID: PMC11438352 DOI: 10.4103/nrr.nrr-d-24-00204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 04/18/2024] [Accepted: 04/30/2024] [Indexed: 07/12/2024] Open
Affiliation(s)
- Orna Elroy-Stein
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel; Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
3
|
Seiler S, Rudolf F, Gomes FR, Pavlovic A, Nebel J, Seidenbecher CI, Foo LC. Astrocyte-derived factors regulate CNS myelination. Glia 2024; 72:2038-2060. [PMID: 39092473 DOI: 10.1002/glia.24596] [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: 10/03/2023] [Revised: 06/20/2024] [Accepted: 07/10/2024] [Indexed: 08/04/2024]
Abstract
The role that astrocytes play in central nervous system (CNS) myelination is poorly understood. We investigated the contribution of astrocyte-derived factors to myelination and revealed a substantial overlap in the secretomes of human and rat astrocytes. Using in vitro myelinating co-cultures of primary retinal ganglion cells and cortical oligodendrocyte precursor cells, we discovered that factors secreted by resting astrocytes, but not reactive astrocytes, facilitated myelination. Soluble brevican emerged as a new enhancer of developmental myelination in vivo, CNS and its absence was linked to remyelination deficits following an immune-mediated damage in an EAE mouse model. The observed reduction of brevican expression in reactive astrocytes and human MS lesions suggested a potential link to the compromised remyelination characteristic of neurodegenerative diseases. Our findings suggested brevican's role in myelination may be mediated through interactions with binding partners such as contactin-1 and tenascin-R. Proteomic analysis of resting versus reactive astrocytes highlighted a shift in protein expression profiles, pinpointing candidates that either facilitate or impede CNS repair, suggesting that depending on their reactivity state, astrocytes play a dual role during myelination.
Collapse
Affiliation(s)
- Sybille Seiler
- F. Hoffmann-La Roche, pRED, Neuroscience, Discovery & Translational Area (NRD), Basel, Switzerland
- Biozentrum, University of Basel, Basel, Switzerland
| | - Franziska Rudolf
- F. Hoffmann-La Roche, pRED, Neuroscience, Discovery & Translational Area (NRD), Basel, Switzerland
| | - Filipa Ramilo Gomes
- F. Hoffmann-La Roche, pRED, Neuroscience, Discovery & Translational Area (NRD), Basel, Switzerland
| | - Anto Pavlovic
- F. Hoffmann-La Roche, pRED, Neuroscience, Discovery & Translational Area (NRD), Basel, Switzerland
| | - Jana Nebel
- Department Neurochemistry & Molecular Biology, Leibniz Institute for Neurobiology (LIN), Magdeburg, Germany
| | - Constanze I Seidenbecher
- Department Neurochemistry & Molecular Biology, Leibniz Institute for Neurobiology (LIN), Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
| | - Lynette C Foo
- F. Hoffmann-La Roche, pRED, Neuroscience, Discovery & Translational Area (NRD), Basel, Switzerland
| |
Collapse
|
4
|
Juráková V, Széky B, Zapletalová M, Fehér A, Zana M, Pandey S, Kučera R, Šerý O, Hudeček J, Dinnyés A, Lochman J. Assessment and Evaluation of Contemporary Approaches for Astrocyte Differentiation from hiPSCs: A Modeling Paradigm for Alzheimer's Disease. Biol Proced Online 2024; 26:30. [PMID: 39342077 PMCID: PMC11437813 DOI: 10.1186/s12575-024-00257-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 09/09/2024] [Indexed: 10/01/2024] Open
Abstract
BACKGROUND Astrocytes have recently gained attention as key players in the pathogenesis of neurodegenerative diseases, including Alzheimer's disease. Numerous differentiation protocols have been developed to study human astrocytes in vitro. However, the properties of the resulting glia are inconsistent, making it difficult to select an appropriate method for a given research question. Therefore, we compared three approaches for the generation of iPSC-derived astrocytes. We performed a detailed analysis using a widely used long serum-free (LSFP) and short serum-free (SSFP) protocol, as well as a TUSP protocol using serum for a limited time of differentiation. RESULTS We used RNA sequencing and immunochemistry to characterize the cultures. Astrocytes generated by the LSFP and SSFP methods differed significantly in their characteristics from those generated by the TUSP method using serum. The TUSP astrocytes had a less neuronal pattern, showed a higher degree of extracellular matrix formation, and were more mature. The short-term presence of FBS in the medium facilitated the induction of astroglia characteristics but did not result in reactive astrocytes. Data from cell-type deconvolution analysis applied to bulk transcriptomes from the cultures assessed their similarity to primary and fetal human astrocytes. CONCLUSIONS Overall, our analyses highlight the need to consider the advantages and disadvantages of a given differentiation protocol for solving specific research tasks or drug discovery studies with iPSC-derived astrocytes.
Collapse
Affiliation(s)
- Veronika Juráková
- Department of Biochemistry, Faculty of Science, Masaryk University, Brno, Czech Republic
| | | | - Martina Zapletalová
- Department of Biochemistry, Faculty of Science, Masaryk University, Brno, Czech Republic
| | | | | | - Shashank Pandey
- Department of Pharmacology and Toxicology, Faculty of Medicine in Pilsen, Charles University, Prague, Czech Republic
| | - Radek Kučera
- Department of Pharmacology and Toxicology, Faculty of Medicine in Pilsen, Charles University, Prague, Czech Republic
| | - Omar Šerý
- Department of Biochemistry, Faculty of Science, Masaryk University, Brno, Czech Republic
- Laboratory of Neurobiology and Pathological Physiology, Institute of Animal Physiology and Genetics, The Czech Academy of Science, Veveří 97, 60200, Brno, Czech Republic
| | - Jiří Hudeček
- Psychiatric Clinic, University Hospital and Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - András Dinnyés
- BioTalentum Ltd, Godollo, Hungary
- Department of Physiology and Animal Health, Institute of Physiology and Animal Nutrition, Hungarian University of Agriculture and Life Sciences, Godollo, Hungary
| | - Jan Lochman
- Department of Biochemistry, Faculty of Science, Masaryk University, Brno, Czech Republic.
- Laboratory of Neurobiology and Pathological Physiology, Institute of Animal Physiology and Genetics, The Czech Academy of Science, Veveří 97, 60200, Brno, Czech Republic.
| |
Collapse
|
5
|
Ishibashi K, Hirata E. Multifaceted interactions between cancer cells and glial cells in brain metastasis. Cancer Sci 2024; 115:2871-2878. [PMID: 38992968 PMCID: PMC11462981 DOI: 10.1111/cas.16241] [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: 04/17/2024] [Revised: 05/20/2024] [Accepted: 05/26/2024] [Indexed: 07/13/2024] Open
Abstract
Cancer brain metastasis has a poor prognosis, is commonly observed in clinical practice, and the number of cases is increasing as overall cancer survival improves. However, experiments in mouse models have shown that brain metastasis itself is an inefficient process. One reason for this inefficiency is the brain microenvironment, which differs significantly from that of other organs, making it difficult for cancer cells to adapt. The brain microenvironment consists of unique resident cell types such as neurons, oligodendrocytes, astrocytes, and microglia. Accumulating evidence over the past decades suggests that the interactions between cancer cells and glial cells can positively or negatively influence the development of brain metastasis. Nevertheless, elucidating the complex interactions between cancer cells and glial cells remains challenging, in part due to the limitations of existing experimental models for glial cell culture. In this review, we first provide an overview of glial cell culture methods and then examine recent discoveries regarding the interactions between brain metastatic cancer cells and the surrounding glial cells, with a special focus on astrocytes and microglia. Finally, we discuss future perspectives for understanding the multifaceted interactions between cancer cells and glial cells for the treatment of metastatic brain tumors.
Collapse
Affiliation(s)
- Kojiro Ishibashi
- Division of Tumor Cell Biology and BioimagingCancer Research Institute of Kanazawa UniversityKanazawaIshikawaJapan
| | - Eishu Hirata
- Division of Tumor Cell Biology and BioimagingCancer Research Institute of Kanazawa UniversityKanazawaIshikawaJapan
- WPI Nano Life Science Institute, Kanazawa UniversityKanazawaIshikawaJapan
| |
Collapse
|
6
|
Renz P, Steinfort M, Haesler V, Tscherrig V, Huang EJ, Chavali M, Liddelow S, Rowitch DH, Surbek D, Schoeberlein A, Brosius Lutz A. Neuroinflammatory reactive astrocyte formation correlates with adverse outcomes in perinatal white matter injury. Glia 2024; 72:1663-1673. [PMID: 38924630 DOI: 10.1002/glia.24575] [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: 09/07/2023] [Revised: 05/15/2024] [Accepted: 05/20/2024] [Indexed: 06/28/2024]
Abstract
Perinatal white matter injury (WMI) is the leading cause of long-term neurological morbidity in infants born preterm. Neuroinflammation during a critical window of early brain development plays a key role in WMI disease pathogenesis. The mechanisms linking inflammation with the long-term myelination failure that characterizes WMI, however, remain unknown. Here, we investigate the role of astrocyte reactivity in WMI. In an experimental mouse model of WMI, we demonstrate that WMI disease outcomes are improved in mutant mice lacking secretion of inflammatory molecules TNF-α, IL-1α, and C1q known, in addition to other roles, to induce the formation of a neuroinflammatory reactive astrocyte substate. We show that astrocytes express molecular signatures of the neuroinflammatory reactive astrocyte substate in both our WMI mouse model and human tissue affected by WMI, and that this gene expression pattern is dampened in injured mutant mice. Our data provide evidence that a neuroinflammatory reactive astrocyte substate correlates with adverse WMI disease outcomes, thus highlighting the need for further investigation of these cells as potential causal players in WMI pathology.
Collapse
Affiliation(s)
- Patricia Renz
- Department of Obstetrics and Gynecology, Division of Feto-Maternal Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Marel Steinfort
- Department of Obstetrics and Gynecology, Division of Feto-Maternal Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Valérie Haesler
- Department of Obstetrics and Gynecology, Division of Feto-Maternal Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Vera Tscherrig
- Department of Obstetrics and Gynecology, Division of Feto-Maternal Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Eric J Huang
- Department of Pathology, UCSF, San Francisco, California, USA
| | - Manideep Chavali
- Department of Pediatrics and Papé Family Pediatric Research Institute, Oregon Health and Science University, Oregon, USA
| | - Shane Liddelow
- Neuroscience Institute, NYU Grossman School of Medicine, New York, New York, USA
- Department of Neuroscience and Physiology, NYU Grossman School of Medicine, New York, New York, USA
- Department of Ophthalmology, NYU Grossman School of Medicine, New York, New York, USA
| | - David H Rowitch
- Department of Pediatrics, UCSF, San Francisco, California, USA
- Eli and Edythe Broad Institute for Stem Cell Research and Regeneration Medicine, UCSF, San Francisco, California, USA
- Newborn Brain Research Institute, UCSF, San Francisco, California, USA
- Department of Paediatrics and Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Daniel Surbek
- Department of Obstetrics and Gynecology, Division of Feto-Maternal Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Andreina Schoeberlein
- Department of Obstetrics and Gynecology, Division of Feto-Maternal Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Amanda Brosius Lutz
- Department of Obstetrics and Gynecology, Division of Feto-Maternal Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| |
Collapse
|
7
|
Chen K, Xu B, Qiu S, Long L, Zhao Q, Xu J, Wang H. Inhibition of phosphodiesterase 4 attenuates aquaporin 4 expression and astrocyte swelling following cerebral ischemia/reperfusion injury. Glia 2024; 72:1629-1645. [PMID: 38785370 DOI: 10.1002/glia.24572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 05/08/2024] [Accepted: 05/13/2024] [Indexed: 05/25/2024]
Abstract
We have previously shown that phosphodiesterase 4 (PDE4) inhibition protects against neuronal injury in rats following middle cerebral artery occlusion/reperfusion (MCAO/R). However, the effects of PDE4 on brain edema and astrocyte swelling are unknown. In this study, we showed that inhibition of PDE4 by Roflumilast (Roflu) reduced brain edema and brain water content in rats subjected to MCAO/R. Roflu decreased the expression of aquaporin 4 (AQP4), while the levels of phosphorylated protein kinase B (Akt) and forkhead box O3a (FoxO3a) were increased. In addition, Roflu reduced cell volume and the expression of AQP4 in primary astrocytes undergoing oxygen and glucose deprivation/reoxygenation (OGD/R). Consistently, PDE4B knockdown showed similar effects as PDE4 inhibition; and PDE4B overexpression rescued the inhibitory role of PDE4B knockdown on AQP4 expression. We then found that the effects of Roflu on the expression of AQP4 and cell volume were blocked by the Akt inhibitor MK2206. Since neuroinflammation and astrocyte activation are the common events that are observed in stroke, we treated primary astrocytes with interleukin-1β (IL-1β). Astrocytes treated with IL-1β showed decreased AQP4 and phosphorylated Akt and FoxO3a. Roflu significantly reduced AQP4 expression, which was accompanied by increased phosphorylation of Akt and FoxO3a. Furthermore, overexpression of FoxO3a partly reversed the effect of Roflu on AQP4 expression. Our findings suggest that PDE4 inhibition limits ischemia-induced brain edema and astrocyte swelling via the Akt/FoxO3a/AQP4 pathway. PDE4 is a promising target for the intervention of brain edema after cerebral ischemia.
Collapse
Affiliation(s)
- Kechun Chen
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Bingtian Xu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Shuqin Qiu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Lu Long
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Qian Zhao
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Jiangping Xu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
- Key Laboratory of Mental Health of the Ministry of Education, Southern Medical University, Guangzhou, China
- Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong-Macao Greater Bay Area, Guangzhou, China
| | - Haitao Wang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
- Key Laboratory of Mental Health of the Ministry of Education, Southern Medical University, Guangzhou, China
- Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong-Macao Greater Bay Area, Guangzhou, China
| |
Collapse
|
8
|
Liddelow SA, Olsen ML, Sofroniew MV. Reactive Astrocytes and Emerging Roles in Central Nervous System (CNS) Disorders. Cold Spring Harb Perspect Biol 2024; 16:a041356. [PMID: 38316554 PMCID: PMC11216178 DOI: 10.1101/cshperspect.a041356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2024]
Abstract
In addition to their many functions in the healthy central nervous system (CNS), astrocytes respond to CNS damage and disease through a process called "reactivity." Recent evidence reveals that astrocyte reactivity is a heterogeneous spectrum of potential changes that occur in a context-specific manner. These changes are determined by diverse signaling events and vary not only with the nature and severity of different CNS insults but also with location in the CNS, genetic predispositions, age, and potentially also with "molecular memory" of previous reactivity events. Astrocyte reactivity can be associated with both essential beneficial functions as well as with harmful effects. The available information is rapidly expanding and much has been learned about molecular diversity of astrocyte reactivity. Emerging functional associations point toward central roles for astrocyte reactivity in determining the outcome in CNS disorders.
Collapse
Affiliation(s)
- Shane A Liddelow
- Neuroscience Institute, NYU School of Medicine, New York, New York 10016, USA
- Department of Neuroscience and Physiology, NYU School of Medicine, New York, New York 10016, USA
- Department of Ophthalmology, NYU School of Medicine, New York, New York 10016, USA
| | - Michelle L Olsen
- School of Neuroscience, Virginia Tech, Blacksburg, Virginia 24061, USA
| | - Michael V Sofroniew
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, California 90095, USA
| |
Collapse
|
9
|
White KE, Bailey HL, Shaw BS, Geiszler PC, Mesquita-Ribeiro R, Scott D, Layfield R, Serres S. A convenient model of serum-induced reactivity of human astrocytes to investigate astrocyte-derived extracellular vesicles. Front Cell Neurosci 2024; 18:1414142. [PMID: 38915876 PMCID: PMC11195030 DOI: 10.3389/fncel.2024.1414142] [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: 04/08/2024] [Accepted: 05/17/2024] [Indexed: 06/26/2024] Open
Abstract
Extracellular vesicles (EVs) are secreted by all cells in the CNS, including neurons and astrocytes. EVs are lipid membrane enclosed particles loaded with various bioactive cargoes reflecting the dynamic activities of cells of origin. In contrast to neurons, the specific role of EVs released by astrocytes is less well understood, partly due to the difficulty in maintaining primary astrocyte cultures in a quiescent state. The aim of this study was to establish a human serum-free astrocyte culture system that maintains primary astrocytes in a quiescent state to study the morphology, function, and protein cargoes of astrocyte-derived EVs. Serum-free medium with G5 supplement and serum-supplemented medium with 2% FBS were compared for the culture of commercially available human primary fetal astrocytes. Serum-free astrocytes displayed morphologies similar to in vivo astrocytes, and surprisingly, higher levels of astrocyte markers compared to astrocytes chronically cultured in FBS. In contrast, astrocyte and inflammatory markers in serum-free astrocytes were upregulated 24 h after either acute 2% FBS or cytokine exposure, confirming their capacity to become reactive. Importantly, this suggests that distinct signaling pathways are involved in acute and chronic astrocyte reactivity. Despite having a similar morphology, chronically serum-cultured astrocyte-derived EVs (ADEVs) were smaller in size compared to serum-free ADEVs and could reactivate serum-free astrocytes. Proteomic analysis identified distinct protein datasets for both types of ADEVs with enrichment of complement and coagulation cascades for chronically serum-cultured astrocyte-derived EVs, offering insights into their roles in the CNS. Collectively, these results suggest that human primary astrocytes cultured in serum-free medium bear similarities with in vivo quiescent astrocytes and the addition of serum induces multiple morphological and transcriptional changes that are specific to human reactive astrocytes and their ADEVs. Thus, more emphasis should be made on using multiple structural, molecular, and functional parameters when evaluating ADEVs as biomarkers of astrocyte health.
Collapse
Affiliation(s)
- Katherine E. White
- School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Hannah L. Bailey
- School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Barry S. Shaw
- School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | | | | | - Daniel Scott
- School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Robert Layfield
- School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Sébastien Serres
- School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
- The David Greenfield Human Physiology Unit, University of Nottingham, Nottingham, United Kingdom
| |
Collapse
|
10
|
Milne SM, Lahiri A, Sanchez CL, Marshall MJ, Jahan I, Meares GP. Myelin oligodendrocyte glycoprotein reactive Th17 cells drive Janus Kinase 1 dependent transcriptional reprogramming in astrocytes and alter cell surface cytokine receptor profiles during experimental autoimmune encephalomyelitis. Sci Rep 2024; 14:13146. [PMID: 38849434 PMCID: PMC11161502 DOI: 10.1038/s41598-024-63877-0] [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: 02/05/2024] [Accepted: 06/03/2024] [Indexed: 06/09/2024] Open
Abstract
Multiple sclerosis (MS) is an autoimmune demyelinating disease affecting the central nervous system (CNS). T helper (Th) 17 cells are involved in the pathogenesis of MS and its animal model of experimental autoimmune encephalomyelitis (EAE) by infiltrating the CNS and producing effector molecules that engage resident glial cells. Among these glial cells, astrocytes have a central role in coordinating inflammatory processes by responding to cytokines and chemokines released by Th17 cells. In this study, we examined the impact of pathogenic Th17 cells on astrocytes in vitro and in vivo. We identified that Th17 cells reprogram astrocytes by driving transcriptomic changes partly through a Janus Kinase (JAK)1-dependent mechanism, which included increased chemokines, interferon-inducible genes, and cytokine receptors. In vivo, we observed a region-specific heterogeneity in the expression of cell surface cytokine receptors on astrocytes, including those for IFN-γ, IL-1, TNF-α, IL-17, TGFβ, and IL-10. Additionally, these receptors were dynamically regulated during EAE induced by adoptive transfer of myelin-reactive Th17 cells. This study overall provides evidence of Th17 cell reprogramming of astrocytes, which may drive changes in the astrocytic responsiveness to cytokines during autoimmune neuroinflammation.
Collapse
MESH Headings
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Animals
- Astrocytes/metabolism
- Th17 Cells/immunology
- Th17 Cells/metabolism
- Mice
- Myelin-Oligodendrocyte Glycoprotein
- Receptors, Cytokine/metabolism
- Receptors, Cytokine/genetics
- Janus Kinase 1/metabolism
- Mice, Inbred C57BL
- Cytokines/metabolism
- Cellular Reprogramming
- Female
- Cells, Cultured
Collapse
Affiliation(s)
- Sarah M Milne
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, 26506, USA
| | - Anirudhya Lahiri
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, 26506, USA
| | - Cristina L Sanchez
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, 26506, USA
| | - Micah J Marshall
- Department of Neurology, The Ohio State University College of Medicine, IBMR 415D, 460 Medical Center Drive, Columbus, OH, 43210, USA
| | - Ishrat Jahan
- Department of Neurology, The Ohio State University College of Medicine, IBMR 415D, 460 Medical Center Drive, Columbus, OH, 43210, USA
| | - Gordon P Meares
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, 26506, USA.
- Department of Neurology, The Ohio State University College of Medicine, IBMR 415D, 460 Medical Center Drive, Columbus, OH, 43210, USA.
- Department of Neuroscience, West Virginia University, Morgantown, WV, 26506, USA.
- Rockefeller Neuroscience Institute, Morgantown, WV, 26506, USA.
| |
Collapse
|
11
|
Jovanovic VM, Mesch KT, Tristan CA. hPSC-Derived Astrocytes at the Forefront of Translational Applications in Neurological Disorders. Cells 2024; 13:903. [PMID: 38891034 PMCID: PMC11172187 DOI: 10.3390/cells13110903] [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: 04/02/2024] [Revised: 05/17/2024] [Accepted: 05/23/2024] [Indexed: 06/20/2024] Open
Abstract
Astrocytes, the most abundant glial cell type in the brain, play crucial roles in maintaining homeostasis within the central nervous system (CNS). Impairment or abnormalities of typical astrocyte functions in the CNS serve as a causative or contributing factor in numerous neurodevelopmental, neurodegenerative, and neuropsychiatric disorders. Currently, disease-modeling and drug-screening approaches, primarily focused on human astrocytes, rely on human pluripotent stem cell (hPSC)-derived astrocytes. However, it is important to acknowledge that these hPSC-derived astrocytes exhibit notable differences across studies and when compared to their in vivo counterparts. These differences may potentially compromise translational outcomes if not carefully accounted for. This review aims to explore state-of-the-art in vitro models of human astrocyte development, focusing on the developmental processes, functional maturity, and technical aspects of various hPSC-derived astrocyte differentiation protocols. Additionally, it summarizes their successful application in modeling neurological disorders. The discussion extends to recent advancements in the large-scale production of human astrocytes and their application in developing high-throughput assays conducive to therapeutic drug discovery.
Collapse
Affiliation(s)
- Vukasin M. Jovanovic
- Stem Cell Translation Laboratory (SCTL), Division of Preclinical Innovation (DPI), National Center for Advancing Translational Sciences (NCATS), NIH, Rockville, MD 20850, USA (C.A.T.)
| | | | | |
Collapse
|
12
|
Calì C. Regulated exocytosis from astrocytes: a matter of vesicles? Front Neurosci 2024; 18:1393165. [PMID: 38800570 PMCID: PMC11116621 DOI: 10.3389/fnins.2024.1393165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 04/30/2024] [Indexed: 05/29/2024] Open
Affiliation(s)
- Corrado Calì
- Department of Neuroscience “Rita Levi Montalcini”, University of Turin, Turin, Italy
- Neuroscience Institute Cavalieri Ottolenghi, Orbassano, Italy
| |
Collapse
|
13
|
Hou SJ, Huang YR, Zhu J, Jia YB, Niu XY, Yang JJ, Yu XL, Du XY, Liang SY, Cui F, Li LJ, Tian C, Liu RT. Mouse serum albumin induces neuronal apoptosis and tauopathies. Acta Neuropathol Commun 2024; 12:66. [PMID: 38654316 PMCID: PMC11040793 DOI: 10.1186/s40478-024-01771-6] [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: 12/04/2023] [Accepted: 03/31/2024] [Indexed: 04/25/2024] Open
Abstract
The elderly frequently present impaired blood-brain barrier which is closely associated with various neurodegenerative diseases. However, how the albumin, the most abundant protein in the plasma, leaking through the disrupted BBB, contributes to the neuropathology remains poorly understood. We here demonstrated that mouse serum albumin-activated microglia induced astrocytes to A1 phenotype to remarkably increase levels of Elovl1, an astrocytic synthase for very long-chain saturated fatty acids, significantly promoting VLSFAs secretion and causing neuronal lippoapoptosis through endoplasmic reticulum stress response pathway. Moreover, MSA-activated microglia triggered remarkable tau phosphorylation at multiple sites through NLRP3 inflammasome pathway. Intracerebroventricular injection of MSA into the brains of C57BL/6J mice to a similar concentration as in patient brains induced neuronal apoptosis, neuroinflammation, increased tau phosphorylation, and decreased the spatial learning and memory abilities, while Elovl1 knockdown significantly prevented the deleterious effect of MSA. Overall, our study here revealed that MSA induced tau phosphorylation and neuron apoptosis based on MSA-activated microglia and astrocytes, respectively, showing the critical roles of MSA in initiating the occurrence of tauopathies and cognitive decline, and providing potential therapeutic targets for MSA-induced neuropathology in multiple neurodegenerative disorders.
Collapse
Affiliation(s)
- Sheng-Jie Hou
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Haidian District, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ya-Ru Huang
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Haidian District, Beijing, 100190, China
| | - Jie Zhu
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Haidian District, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ying-Bo Jia
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Haidian District, Beijing, 100190, China
| | - Xiao-Yun Niu
- Ningxia University, Yinchuan, 750021, Ningxia, China
| | - Jin-Ju Yang
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Haidian District, Beijing, 100190, China
| | - Xiao-Lin Yu
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Haidian District, Beijing, 100190, China
| | - Xiao-Yu Du
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Haidian District, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Shi-Yu Liang
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Haidian District, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Fang Cui
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Haidian District, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ling-Jie Li
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Haidian District, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Chen Tian
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Haidian District, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Rui-Tian Liu
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Haidian District, Beijing, 100190, China.
| |
Collapse
|
14
|
Clayton BLL, Kristell JD, Allan KC, Cohn EF, Karl M, Jerome AD, Garrison E, Maeno-Hikichi Y, Sturno AM, Kerr A, Shick HE, Sepeda JA, Freundt EC, Sas AR, Segal BM, Miller RH, Tesar PJ. A phenotypic screening platform for identifying chemical modulators of astrocyte reactivity. Nat Neurosci 2024; 27:656-665. [PMID: 38378993 PMCID: PMC11034956 DOI: 10.1038/s41593-024-01580-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 01/15/2024] [Indexed: 02/22/2024]
Abstract
Disease, injury and aging induce pathological reactive astrocyte states that contribute to neurodegeneration. Modulating reactive astrocytes therefore represent an attractive therapeutic strategy. Here we describe the development of an astrocyte phenotypic screening platform for identifying chemical modulators of astrocyte reactivity. Leveraging this platform for chemical screening, we identify histone deacetylase 3 (HDAC3) inhibitors as effective suppressors of pathological astrocyte reactivity. We demonstrate that HDAC3 inhibition reduces molecular and functional characteristics of reactive astrocytes in vitro. Transcriptional and chromatin mapping studies show that HDAC3 inhibition disarms pathological astrocyte gene expression and function while promoting the expression of genes associated with beneficial astrocytes. Administration of RGFP966, a small molecule HDAC3 inhibitor, blocks reactive astrocyte formation and promotes neuroprotection in vivo in mice. Collectively, these results establish a platform for discovering modulators of reactive astrocyte states, inform the mechanisms that control astrocyte reactivity and demonstrate the therapeutic benefits of modulating astrocyte reactivity for neurodegenerative diseases.
Collapse
Affiliation(s)
- Benjamin L L Clayton
- Institute for Glial Sciences, Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA.
| | - James D Kristell
- Institute for Glial Sciences, Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Kevin C Allan
- Institute for Glial Sciences, Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Erin F Cohn
- Institute for Glial Sciences, Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Molly Karl
- Department of Anatomy and Cell Biology, George Washington University School of Medicine, Washington, DC, USA
| | - Andrew D Jerome
- Department of Neurology, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Neuroscience Research Institute, The Ohio State University, Columbus, OH, USA
| | - Eric Garrison
- Department of Anatomy and Cell Biology, George Washington University School of Medicine, Washington, DC, USA
| | - Yuka Maeno-Hikichi
- Institute for Glial Sciences, Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Annalise M Sturno
- Institute for Glial Sciences, Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Alexis Kerr
- Institute for Glial Sciences, Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - H Elizabeth Shick
- Institute for Glial Sciences, Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Jesse A Sepeda
- Department of Neurology, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Neuroscience Research Institute, The Ohio State University, Columbus, OH, USA
| | - Eric C Freundt
- Department of Biology, The University of Tampa, Tampa, FL, USA
| | - Andrew R Sas
- Department of Neurology, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Neuroscience Research Institute, The Ohio State University, Columbus, OH, USA
| | - Benjamin M Segal
- Department of Neurology, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Neuroscience Research Institute, The Ohio State University, Columbus, OH, USA
| | - Robert H Miller
- Department of Anatomy and Cell Biology, George Washington University School of Medicine, Washington, DC, USA
| | - Paul J Tesar
- Institute for Glial Sciences, Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA.
| |
Collapse
|
15
|
Komai M, Noda Y, Ikeda A, Kaneshiro N, Kamikubo Y, Sakurai T, Uehara T, Takasugi N. Nuclear SphK2/S1P signaling is a key regulator of ApoE production and Aβ uptake in astrocytes. J Lipid Res 2024; 65:100510. [PMID: 38280459 PMCID: PMC10907773 DOI: 10.1016/j.jlr.2024.100510] [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: 10/24/2023] [Revised: 01/11/2024] [Accepted: 01/12/2024] [Indexed: 01/29/2024] Open
Abstract
The link between changes in astrocyte function and the pathological progression of Alzheimer's disease (AD) has attracted considerable attention. Interestingly, activated astrocytes in AD show abnormalities in their lipid content and metabolism. In particular, the expression of apolipoprotein E (ApoE), a lipid transporter, is decreased. Because ApoE has anti-inflammatory and amyloid β (Aβ)-metabolizing effects, the nuclear receptors, retinoid X receptor (RXR) and LXR, which are involved in ApoE expression, are considered promising therapeutic targets for AD. However, the therapeutic effects of agents targeting these receptors are limited or vary considerably among groups, indicating the involvement of an unknown pathological factor that modifies astrocyte and ApoE function. Here, we focused on the signaling lipid, sphingosine-1-phosphate (S1P), which is mainly produced by sphingosine kinase 2 (SphK2) in the brain. Using astrocyte models, we found that upregulation of SphK2/S1P signaling suppressed ApoE induction by both RXR and LXR agonists. We also found that SphK2 activation reduced RXR binding to the APOE promoter region in the nucleus, suggesting the nuclear function of SphK2/S1P. Intriguingly, suppression of SphK2 activity by RNA knockdown or specific inhibitors upregulated lipidated ApoE induction. Furthermore, the induced ApoE facilitates Aβ uptake in astrocytes. Together with our previous findings that SphK2 activity is upregulated in AD brain and promotes Aβ production in neurons, these results indicate that SphK2/S1P signaling is a promising multifunctional therapeutic target for AD that can modulate astrocyte function by stabilizing the effects of RXR and LXR agonists, and simultaneously regulate neuronal pathogenesis.
Collapse
Affiliation(s)
- Masato Komai
- Department of Medicinal Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita, Okayama, Japan; Research Fellow of Japan Society for the Promotion of Science, Chiyoda, Tokyo, Japan
| | - Yuka Noda
- Department of Medicinal Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita, Okayama, Japan
| | - Atsuya Ikeda
- Department of Medicinal Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita, Okayama, Japan
| | - Nanaka Kaneshiro
- Department of Medicinal Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita, Okayama, Japan
| | - Yuji Kamikubo
- Department of Cellular and Molecular Pharmacology, Juntendo University Graduate School of Medicine, Bunkyo, Tokyo, Japan
| | - Takashi Sakurai
- Department of Cellular and Molecular Pharmacology, Juntendo University Graduate School of Medicine, Bunkyo, Tokyo, Japan
| | - Takashi Uehara
- Department of Medicinal Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita, Okayama, Japan
| | - Nobumasa Takasugi
- Department of Medicinal Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita, Okayama, Japan; Department of Cellular and Molecular Pharmacology, Juntendo University Graduate School of Medicine, Bunkyo, Tokyo, Japan.
| |
Collapse
|
16
|
Linnerbauer M, Lößlein L, Vandrey O, Peter A, Han Y, Tsaktanis T, Wogram E, Needhamsen M, Kular L, Nagel L, Zissler J, Andert M, Meszaros L, Hanspach J, Zuber F, Naumann UJ, Diebold M, Wheeler MA, Beyer T, Nirschl L, Cirac A, Laun FB, Günther C, Winkler J, Bäuerle T, Jagodic M, Hemmer B, Prinz M, Quintana FJ, Rothhammer V. The astrocyte-produced growth factor HB-EGF limits autoimmune CNS pathology. Nat Immunol 2024; 25:432-447. [PMID: 38409259 PMCID: PMC10907300 DOI: 10.1038/s41590-024-01756-6] [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: 03/09/2023] [Accepted: 01/12/2024] [Indexed: 02/28/2024]
Abstract
Central nervous system (CNS)-resident cells such as microglia, oligodendrocytes and astrocytes are gaining increasing attention in respect to their contribution to CNS pathologies including multiple sclerosis (MS). Several studies have demonstrated the involvement of pro-inflammatory glial subsets in the pathogenesis and propagation of inflammatory events in MS and its animal models. However, it has only recently become clear that the underlying heterogeneity of astrocytes and microglia can not only drive inflammation, but also lead to its resolution through direct and indirect mechanisms. Failure of these tissue-protective mechanisms may potentiate disease and increase the risk of conversion to progressive stages of MS, for which currently available therapies are limited. Using proteomic analyses of cerebrospinal fluid specimens from patients with MS in combination with experimental studies, we here identify Heparin-binding EGF-like growth factor (HB-EGF) as a central mediator of tissue-protective and anti-inflammatory effects important for the recovery from acute inflammatory lesions in CNS autoimmunity. Hypoxic conditions drive the rapid upregulation of HB-EGF by astrocytes during early CNS inflammation, while pro-inflammatory conditions suppress trophic HB-EGF signaling through epigenetic modifications. Finally, we demonstrate both anti-inflammatory and tissue-protective effects of HB-EGF in a broad variety of cell types in vitro and use intranasal administration of HB-EGF in acute and post-acute stages of autoimmune neuroinflammation to attenuate disease in a preclinical mouse model of MS. Altogether, we identify astrocyte-derived HB-EGF and its epigenetic regulation as a modulator of autoimmune CNS inflammation and potential therapeutic target in MS.
Collapse
Affiliation(s)
- Mathias Linnerbauer
- Department of Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen Nuremberg, Erlangen, Germany
| | - Lena Lößlein
- Department of Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen Nuremberg, Erlangen, Germany
| | - Oliver Vandrey
- Department of Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen Nuremberg, Erlangen, Germany
| | - Anne Peter
- Department of Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen Nuremberg, Erlangen, Germany
| | - Yanan Han
- Department of Clinical Neuroscience, Karolinska Institutet, Karolinska University Hospital, Center for Molecular Medicine, Stockholm, Sweden
| | - Thanos Tsaktanis
- Department of Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen Nuremberg, Erlangen, Germany
| | - Emile Wogram
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Maria Needhamsen
- Department of Clinical Neuroscience, Karolinska Institutet, Karolinska University Hospital, Center for Molecular Medicine, Stockholm, Sweden
| | - Lara Kular
- Department of Clinical Neuroscience, Karolinska Institutet, Karolinska University Hospital, Center for Molecular Medicine, Stockholm, Sweden
| | - Lisa Nagel
- Institute of Radiology, University Hospital Erlangen, Friedrich-Alexander University Erlangen Nuremberg, Erlangen, Germany
| | - Julia Zissler
- Department of Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen Nuremberg, Erlangen, Germany
| | - Marie Andert
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen Nuremberg, Erlangen, Germany
| | - Lisa Meszaros
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen Nuremberg, Erlangen, Germany
| | - Jannis Hanspach
- Institute of Radiology, University Hospital Erlangen, Friedrich-Alexander University Erlangen Nuremberg, Erlangen, Germany
| | - Finnja Zuber
- Department of Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen Nuremberg, Erlangen, Germany
| | - Ulrike J Naumann
- Department of Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen Nuremberg, Erlangen, Germany
| | - Martin Diebold
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Michael A Wheeler
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- The Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Tobias Beyer
- Department of Neurology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Lucy Nirschl
- Department of Neurology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Ana Cirac
- Department of Neurology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Frederik B Laun
- Institute of Radiology, University Hospital Erlangen, Friedrich-Alexander University Erlangen Nuremberg, Erlangen, Germany
| | - Claudia Günther
- Department of Medicine 1, University Hospital Erlangen, Friedrich-Alexander University Erlangen Nuremberg, Erlangen, Germany
| | - Jürgen Winkler
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Tobias Bäuerle
- Institute of Radiology, University Hospital Erlangen, Friedrich-Alexander University Erlangen Nuremberg, Erlangen, Germany
| | - Maja Jagodic
- Department of Clinical Neuroscience, Karolinska Institutet, Karolinska University Hospital, Center for Molecular Medicine, Stockholm, Sweden
| | - Bernhard Hemmer
- Department of Neurology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Marco Prinz
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Francisco J Quintana
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- The Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Veit Rothhammer
- Department of Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen Nuremberg, Erlangen, Germany.
- Deutsches Zentrum Immuntherapie (DZI), University Hospital Erlangen, Erlangen, Germany.
| |
Collapse
|
17
|
Yadav B, Kaur S, Yadav A, Verma H, Kar S, Sahu BK, Pati KR, Sarkar B, Dhiman M, Mantha AK. Implications of organophosphate pesticides on brain cells and their contribution toward progression of Alzheimer's disease. J Biochem Mol Toxicol 2024; 38:e23660. [PMID: 38356323 DOI: 10.1002/jbt.23660] [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: 09/12/2023] [Revised: 01/04/2024] [Accepted: 01/18/2024] [Indexed: 02/16/2024]
Abstract
The most widespread neurodegenerative disorder, Alzheimer's disease (AD) is marked by severe behavioral abnormalities, cognitive and functional impairments. It is inextricably linked with the deposition of amyloid β (Aβ) plaques and tau protein in the brain. Loss of white matter, neurons, synapses, and reactive microgliosis are also frequently observed in patients of AD. Although the causative mechanisms behind the neuropathological alterations in AD are not fully understood, they are likely influenced by hereditary and environmental factors. The etiology and pathogenesis of AD are significantly influenced by the cells of the central nervous system, namely, glial cells and neurons, which are directly engaged in the transmission of electrical signals and the processing of information. Emerging evidence suggests that exposure to organophosphate pesticides (OPPs) can trigger inflammatory responses in glial cells, leading to various cascades of events that contribute to neuroinflammation, neuronal damage, and ultimately, AD pathogenesis. Furthermore, there are striking similarities between the biomarkers associated with AD and OPPs, including neuroinflammation, oxidative stress, dysregulation of microRNA, and accumulation of toxic protein aggregates, such as amyloid β. These shared markers suggest a potential mechanistic link between OPP exposure and AD pathology. In this review, we attempt to address the role of OPPs on altered cell physiology of the brain cells leading to neuroinflammation, mitochondrial dysfunction, and oxidative stress linked with AD pathogenesis.
Collapse
Affiliation(s)
- Bharti Yadav
- Department of Zoology, Central University of Punjab, Bathinda, Punjab, India
| | - Sharanjot Kaur
- Department of Microbiology, Central University of Punjab, Bathinda, Punjab, India
| | - Anuradha Yadav
- Department of Zoology, Central University of Punjab, Bathinda, Punjab, India
| | - Harkomal Verma
- Department of Zoology, Central University of Punjab, Bathinda, Punjab, India
| | - Swastitapa Kar
- Department of Zoology, Central University of Punjab, Bathinda, Punjab, India
| | - Binit Kumar Sahu
- Department of Zoology, Central University of Punjab, Bathinda, Punjab, India
| | - Kumari Riya Pati
- Department of Zoology, Central University of Punjab, Bathinda, Punjab, India
| | - Bibekanada Sarkar
- Department of Zoology, Central University of Punjab, Bathinda, Punjab, India
| | - Monisha Dhiman
- Department of Microbiology, Central University of Punjab, Bathinda, Punjab, India
| | - Anil Kumar Mantha
- Department of Zoology, Central University of Punjab, Bathinda, Punjab, India
| |
Collapse
|
18
|
Prakash P, Erdjument-Bromage H, O'Dea MR, Munson CN, Labib D, Fossati V, Neubert TA, Liddelow SA. Proteomic profiling of interferon-responsive reactive astrocytes in rodent and human. Glia 2024; 72:625-642. [PMID: 38031883 PMCID: PMC10843807 DOI: 10.1002/glia.24494] [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: 05/16/2023] [Revised: 11/07/2023] [Accepted: 11/14/2023] [Indexed: 12/01/2023]
Abstract
Astrocytes are a heterogeneous population of central nervous system glial cells that respond to pathological insults and injury by undergoing a transformation called "reactivity." Reactive astrocytes exhibit distinct and context-dependent cellular, molecular, and functional state changes that can either support or disturb tissue homeostasis. We recently identified a reactive astrocyte sub-state defined by interferon-responsive genes like Igtp, Ifit3, Mx1, and others, called interferon-responsive reactive astrocytes (IRRAs). To further this transcriptomic definition of IRRAs, we wanted to define the proteomic changes that occur in this reactive sub-state. We induced IRRAs in immunopanned rodent astrocytes and human iPSC-differentiated astrocytes using TNF, IL1α, C1Q, and IFNβ and characterized their proteomic profile (both cellular and secreted) using unbiased quantitative proteomics. We identified 2335 unique cellular proteins, including IFIT2/3, IFITM3, OASL1/2, MX1/2/3, and STAT1. We also report that rodent and human IRRAs secrete PAI1, a serine protease inhibitor which may influence reactive states and functions of nearby cells. Finally, we evaluated how IRRAs are distinct from neurotoxic reactive astrocytes (NRAs). While NRAs are described by expression of the complement protein C3, it was not upregulated in IRRAs. Instead, we found ~90 proteins unique to IRRAs not identified in NRAs, including OAS1A, IFIT3, and MX1. Interferon signaling in astrocytes is critical for the antiviral immune response and for regulating synaptic plasticity and glutamate transport mechanisms. How IRRAs contribute to these functions is unknown. This study provides the basis for future experiments to define the functional roles of IRRAs in the context of neurodegenerative disorders.
Collapse
Affiliation(s)
- Priya Prakash
- Neuroscience Institute, NYU Grossman School of Medicine, New York, New York, USA
| | - Hediye Erdjument-Bromage
- Neuroscience Institute, NYU Grossman School of Medicine, New York, New York, USA
- Department of Neuroscience and Physiology, NYU Grossman School of Medicine, New York, New York, USA
| | - Michael R O'Dea
- Neuroscience Institute, NYU Grossman School of Medicine, New York, New York, USA
| | - Christy N Munson
- Neuroscience Institute, NYU Grossman School of Medicine, New York, New York, USA
| | - David Labib
- The New York Stem Cell Foundation Research Institute, New York, New York, USA
| | - Valentina Fossati
- The New York Stem Cell Foundation Research Institute, New York, New York, USA
| | - Thomas A Neubert
- Neuroscience Institute, NYU Grossman School of Medicine, New York, New York, USA
- Department of Neuroscience and Physiology, NYU Grossman School of Medicine, New York, New York, USA
| | - Shane A Liddelow
- Neuroscience Institute, NYU Grossman School of Medicine, New York, New York, USA
- Department of Neuroscience and Physiology, NYU Grossman School of Medicine, New York, New York, USA
- Department of Ophthalmology, NYU Grossman School of Medicine, New York, New York, USA
- Parekh Center for Interdisciplinary Neurology, NYU Grossman School of Medicine, New York, New York, USA
| |
Collapse
|
19
|
Soto JS, Jami-Alahmadi Y, Wohlschlegel JA, Khakh BS. In vivo identification of astrocyte and neuron subproteomes by proximity-dependent biotinylation. Nat Protoc 2024; 19:896-927. [PMID: 38062165 DOI: 10.1038/s41596-023-00923-7] [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: 08/03/2023] [Accepted: 10/03/2023] [Indexed: 02/08/2024]
Abstract
The central nervous system (CNS) comprises diverse and morphologically complex cells. To understand the molecular basis of their physiology, it is crucial to assess proteins expressed within intact cells. Commonly used methods utilize cell dissociation and sorting to isolate specific cell types such as neurons and astrocytes, the major CNS cells. Proteins purified from isolated cells are identified by mass spectrometry-based proteomics. However, dissociation and cell-sorting methods lead to near total loss of cellular morphology, thereby losing proteins from key relevant subcompartments such as processes, end feet, dendrites and axons. Here we provide a systematic protocol for cell- and subcompartment-specific labeling and identification of proteins found within intact astrocytes and neurons in vivo. This protocol utilizes the proximity-dependent biotinylation system BioID2, selectively expressed in either astrocytes or neurons, to label proximal proteins in a cell-specific manner. BioID2 is targeted genetically to assess the subproteomes of subcellular compartments such as the plasma membrane and sites of cell-cell contacts. We describe in detail the expression methods (variable timing), stereotaxic surgeries for expression (1-2 d and then 3 weeks), in vivo protein labeling (7 d), protein isolation (2-3 d), protein identification methods (2-3 d) and data analysis (1 week). The protocol can be applied to any area of the CNS in mouse models of physiological processes and for disease-related research.
Collapse
Affiliation(s)
- Joselyn S Soto
- Department of Physiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
| | - Yasaman Jami-Alahmadi
- Department of Biological Chemistry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - James A Wohlschlegel
- Department of Biological Chemistry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Baljit S Khakh
- Department of Physiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
- Department of Neurobiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
20
|
Sun Z, Zhang X, Dong Y, Liu Y, Wang C, Li Y, Ma C, Xu G, Wang S, Yang C, Zhang G, Cong B. Norepinephrine-Activated p38 MAPK Pathway Mediates Stress-Induced Cytotoxic Edema of Basolateral Amygdala Astrocytes. Brain Sci 2024; 14:161. [PMID: 38391735 PMCID: PMC10887202 DOI: 10.3390/brainsci14020161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 01/31/2024] [Accepted: 02/01/2024] [Indexed: 02/24/2024] Open
Abstract
The amygdala is a core region in the limbic system that is highly sensitive to stress. Astrocytes are key players in stress disorders such as anxiety and depression. However, the effects of stress on the morphology and function of amygdala astrocytes and its potential mechanisms remain largely unknown. Hence, we performed in vivo and in vitro experiments using a restraint stress (RS) rat model and stress-induced astrocyte culture, respectively. Our data show that norepinephrine (NE) content increased, cytotoxic edema occurred, and aquaporin-4 (AQP4) expression was up-regulated in the basolateral amygdala (BLA) obtained from RS rats. Additionally, the p38 mitogen-activated protein kinase (MAPK) pathway was also observed to be significantly activated in the BLA of rats subjected to RS. The administration of NE to in vitro astrocytes increased the AQP4 level and induced cell edema. Furthermore, p38 MAPK signaling was activated. The NE inhibitor alpha-methyl-p-tyrosine (AMPT) alleviated cytotoxic edema in astrocytes, inhibited AQP4 expression, and inactivated the p38 MAPK pathway in RS rats. Meanwhile, in the in vitro experiment, the p38 MAPK signaling inhibitor SB203580 reversed NE-induced cytotoxic edema and down-regulated the expression of AQP4 in astrocytes. Briefly, NE-induced activation of the p38 MAPK pathway mediated cytotoxic edema in BLA astrocytes from RS rats. Thus, our data provide novel evidence that NE-induced p38 MAPK pathway activation may be one of the mechanisms leading to cytotoxic edema in BLA under stress conditions, which also could enable the development of an effective therapeutic strategy against cytotoxic edema in BLA under stress and provide new ideas for the treatment of neuropsychiatric diseases.
Collapse
Affiliation(s)
- Zhaoling Sun
- Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, College of Forensic Medicine, Hebei Medical University, Shijiazhuang 050017, China
| | - Xiaojing Zhang
- Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, College of Forensic Medicine, Hebei Medical University, Shijiazhuang 050017, China
| | - Yiming Dong
- Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, College of Forensic Medicine, Hebei Medical University, Shijiazhuang 050017, China
| | - Yichang Liu
- Department of Forensic Medicine, College of Medicine, Nantong University, Nantong 226000, China
| | - Chuan Wang
- Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, College of Forensic Medicine, Hebei Medical University, Shijiazhuang 050017, China
| | - Yingmin Li
- Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, College of Forensic Medicine, Hebei Medical University, Shijiazhuang 050017, China
| | - Chunling Ma
- Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, College of Forensic Medicine, Hebei Medical University, Shijiazhuang 050017, China
| | - Guangming Xu
- Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, College of Forensic Medicine, Hebei Medical University, Shijiazhuang 050017, China
| | - Songjun Wang
- Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, College of Forensic Medicine, Hebei Medical University, Shijiazhuang 050017, China
| | - Chenteng Yang
- Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, College of Forensic Medicine, Hebei Medical University, Shijiazhuang 050017, China
| | - Guozhong Zhang
- Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, College of Forensic Medicine, Hebei Medical University, Shijiazhuang 050017, China
- Hebei Province Laboratory of Experimental Animal, Shijiazhuang 050017, China
| | - Bin Cong
- Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, College of Forensic Medicine, Hebei Medical University, Shijiazhuang 050017, China
- Hainan Tropical Forensic Medicine Academician Workstation, Haikou 571199, China
| |
Collapse
|
21
|
Pio T, Hill EJ, Kebede N, Andersen J, Sloan SA. Neuron-Astrocyte Interactions: A Human Perspective. ADVANCES IN NEUROBIOLOGY 2024; 39:69-93. [PMID: 39190072 DOI: 10.1007/978-3-031-64839-7_4] [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: 08/28/2024]
Abstract
This chapter explores the intricate interactions between neurons and astrocytes within the nervous system with a particular emphasis on studies conducted in human tissue or with human cells. We specifically explore how neuron-astrocyte interactions relate to processes of cellular development, morphology, migration, synapse formation, and metabolism. These findings enrich our understanding of basic neurobiology and how disruptions in these processes are relevant to human diseases.The study of human neuron-astrocyte interactions is made possible because of transformative in vitro advancements that have facilitated the generation and sustained culture of human neural cells. In addition, the rise of techniques like sequencing at single-cell resolution has enabled the exploration of numerous human cell atlases and their comparisons to other animal model systems. Thus, the innovations outlined in this chapter illuminate the convergence and divergence of neuron-astrocyte interactions across species. As technologies progress, continually more sophisticated in vitro systems will increasingly reflect in vivo environments and deepen our command of neuron-glial interactions in human biology.
Collapse
Affiliation(s)
- Taylor Pio
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Emily J Hill
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Nardos Kebede
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Jimena Andersen
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Steven A Sloan
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
22
|
Rosenberg MF, Godoy MI, Wade SD, Paredes MF, Zhang Y, Molofsky AV. β-Adrenergic Signaling Promotes Morphological Maturation of Astrocytes in Female Mice. J Neurosci 2023; 43:8621-8636. [PMID: 37845031 PMCID: PMC10727121 DOI: 10.1523/jneurosci.0357-23.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 06/24/2023] [Accepted: 07/31/2023] [Indexed: 10/18/2023] Open
Abstract
Astrocytes play essential roles in the developing nervous system, including supporting synapse function. These astrocyte support functions emerge coincident with brain maturation and may be tailored in a region-specific manner. For example, gray matter astrocytes have elaborate synapse-associated processes and are morphologically and molecularly distinct from white matter astrocytes. This raises the question of whether there are unique environmental cues that promote gray matter astrocyte identity and synaptogenic function. We previously identified adrenergic receptors as preferentially enriched in developing gray versus white matter astrocytes, suggesting that noradrenergic signaling could be a cue that promotes the functional maturation of gray matter astrocytes. We first characterized noradrenergic projections during postnatal brain development in mouse and human, finding that process density was higher in the gray matter and increased concurrently with astrocyte maturation. RNA sequencing revealed that astrocytes in both species expressed α- and β-adrenergic receptors. We found that stimulation of β-adrenergic receptors increased primary branching of rodent astrocytes in vitro Conversely, astrocyte-conditional knockout of the β1-adrenergic receptor reduced the size of gray matter astrocytes and led to dysregulated sensorimotor integration in female mice. These studies suggest that adrenergic signaling to developing astrocytes impacts their morphology and has implications for adult behavior, particularly in female animals. More broadly, they demonstrate a mechanism through which environmental cues impact astrocyte development. Given the key roles of norepinephrine in brain states, such as arousal, stress, and learning, these findings could prompt further inquiry into how developmental stressors impact astrocyte development and adult brain function.SIGNIFICANCE STATEMENT This study demonstrates a role for noradrenergic signaling in the development of gray matter astrocytes. We provide new evidence that the β1-adrenergic receptor is robustly expressed by both mouse and human astrocytes, and that conditional KO of the β1-adrenergic receptor from female mouse astrocytes impairs gray matter astrocyte maturation. Moreover, female conditional KO mice exhibit behavioral deficits in two paradigms that test sensorimotor function. Given the emerging interest in moving beyond RNA sequencing to probe specific pathways that underlie astrocyte heterogeneity, this study provides a foundation for future investigation into the effect of noradrenergic signaling on astrocyte functions in conditions where noradrenergic signaling is altered, such as stress, arousal, and learning.
Collapse
Affiliation(s)
- Marci F Rosenberg
- Department of Psychiatry and Behavioral Sciences and Weill Institute of Neurosciences, University of California at San Francisco, San Francisco, California 94143
- Medical Scientist Training Program and Biomedical Sciences Graduate Program, University of California at San Francisco, San Francisco, California 94143
| | - Marlesa I Godoy
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, California 90095
| | - Sarah D Wade
- Department of Psychiatry and Behavioral Sciences and Weill Institute of Neurosciences, University of California at San Francisco, San Francisco, California 94143
- Neurosciences Graduate Program, University of California at San Francisco, San Francisco, California 94143
| | - Mercedes F Paredes
- Department of Neurology, Weill Institute of Neurosciences, University of California, San Francisco, San Francisco, California 94143
- Chan Zuckerberg Biohub-San Francisco, San Francisco, California 94158
| | - Ye Zhang
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, California 90095
| | - Anna V Molofsky
- Department of Psychiatry and Behavioral Sciences and Weill Institute of Neurosciences, University of California at San Francisco, San Francisco, California 94143
- Neurosciences Graduate Program, University of California at San Francisco, San Francisco, California 94143
- Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, California 94143
| |
Collapse
|
23
|
Qin L, Xiao G. Primary Astrocytes Purification and Immortalization. Curr Protoc 2023; 3:e964. [PMID: 38131300 DOI: 10.1002/cpz1.964] [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: 12/23/2023]
Abstract
Astrocytes, the most abundant cells in the central nervous system (CNS), are essential for neuronal development, network formation, and overall CNS homeostasis. Primary astrocyte culture has been successfully used as a tool to study astrocyte biology in vitro. In the present protocol, a modified immunopanning method was utilized to obtain and purify primary astrocytes from mouse cortex and spinal cord in a relatively quick and inexpensive way. Purified primary astrocytes were then immortalized through infection of lentivirus expressing the SV40 large T antigens. In addition, we provide protocols to determine the expression levels of astrocyte-specific markers and to perform functional studies measuring the ATP-induced calcium flux in the immortalized astrocytes. Following the described protocols assures that the immortalized astrocytes that one prepares mimic the cell biology of primary astrocytes in culture. Thus, the purification and immortalization protocols for primary astrocytes presented in here provide two models for the studies of astrocyte biology and may be useful for the immortalization of other types of primary cells. © 2023 Wiley Periodicals LLC. Basic Protocol 1: Primary astrocyte purification by a modified immunopanning method Support Protocol: Serum-free primary astrocyte culture Basic Protocol 2: Primary astrocyte immortalization Basic Protocol 3: Calcium transient detection in astrocytes.
Collapse
Affiliation(s)
- Lei Qin
- Department of Orthopedics, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China
| | - Guozhi Xiao
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
24
|
Kim E, Kim H, Jedrychowski MP, Bakiasi G, Park J, Kruskop J, Choi Y, Kwak SS, Quinti L, Kim DY, Wrann CD, Spiegelman BM, Tanzi RE, Choi SH. Irisin reduces amyloid-β by inducing the release of neprilysin from astrocytes following downregulation of ERK-STAT3 signaling. Neuron 2023; 111:3619-3633.e8. [PMID: 37689059 PMCID: PMC10840702 DOI: 10.1016/j.neuron.2023.08.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/09/2023] [Accepted: 08/11/2023] [Indexed: 09/11/2023]
Abstract
A pathological hallmark of Alzheimer's disease (AD) is the deposition of amyloid-β (Aβ) protein in the brain. Physical exercise has been shown to reduce Aβ burden in various AD mouse models, but the underlying mechanisms have not been elucidated. Irisin, an exercise-induced hormone, is the secreted form of fibronectin type-III-domain-containing 5 (FNDC5). Here, using a three-dimensional (3D) cell culture model of AD, we show that irisin significantly reduces Aβ pathology by increasing astrocytic release of the Aβ-degrading enzyme neprilysin (NEP). This is mediated by downregulation of ERK-STAT3 signaling. Finally, we show that integrin αV/β5 acts as the irisin receptor on astrocytes required for irisin-induced release of astrocytic NEP, leading to clearance of Aβ. Our findings reveal for the first time a cellular and molecular mechanism by which exercise-induced irisin attenuates Aβ pathology, suggesting a new target pathway for therapies aimed at the prevention and treatment of AD.
Collapse
Affiliation(s)
- Eunhee Kim
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Hyeonwoo Kim
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Department of Cell Biology, Harvard University Medical School, Boston, MA 02115, USA; Department of Biological Sciences, Korea Advanced Institute of Science & Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Mark P Jedrychowski
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Department of Cell Biology, Harvard University Medical School, Boston, MA 02115, USA
| | - Grisilda Bakiasi
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Joseph Park
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Jane Kruskop
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Younjung Choi
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Sang Su Kwak
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Luisa Quinti
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Doo Yeon Kim
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Christiane D Wrann
- McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA 02114, USA; Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Bruce M Spiegelman
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Department of Cell Biology, Harvard University Medical School, Boston, MA 02115, USA
| | - Rudolph E Tanzi
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA 02114, USA.
| | - Se Hoon Choi
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA 02114, USA.
| |
Collapse
|
25
|
Cho YJ, Park SH, Ryu KY. Mild Oxidative Stress Induced by Sodium Arsenite Reduces Lipocalin-2 Expression Levels in Cortical Glial Cells. Int J Mol Sci 2023; 24:15864. [PMID: 37958847 PMCID: PMC10649205 DOI: 10.3390/ijms242115864] [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: 10/06/2023] [Revised: 10/28/2023] [Accepted: 10/31/2023] [Indexed: 11/15/2023] Open
Abstract
Astrocytes and microglia, the most abundant glial cells in the central nervous system, are involved in maintaining homeostasis in the brain microenvironment and in the progression of various neurological disorders. Lipocalin-2 (LCN2) is a small secretory protein that can be transcriptionally upregulated via nuclear factor kappa B (NF-κB) signaling. It is synthesized and secreted by glial cells, resulting in either the restoration of damaged neural tissues or the induction of neuronal apoptosis in a context-dependent manner. It has recently been reported that when glial cells are under lipopolysaccharide-induced inflammatory stress, either reduced production or accelerated degradation of LCN2 can alleviate neurotoxicity. However, the regulatory mechanisms of LCN2 in glial cells are not yet fully understood. In this study, we used primary astroglial-enriched cells which produce LCN2 and found that the production of LCN2 could be reduced by sodium arsenite treatment. Surprisingly, the reduced LCN2 production was not due to the suppression of NF-κB signaling. Mild oxidative stress induced by sodium arsenite treatment activated antioxidant responses and downregulated Lcn2 expression without reducing the viability of astroglial-enriched cells. Intriguingly, reduced LCN2 production could not be achieved by simple activation of the nuclear factor erythroid-2-related factor 2 (Nrf2)-Kelch-like ECH-associated protein 1 (Keap1) pathway in astroglial-enriched cells. Thus, it appears that mild oxidative stress, occurring in an Nrf2-independent manner, is required for the downregulation of Lcn2 expression. Taken together, our findings provide new insights into the regulatory mechanisms of LCN2 and suggest that mild oxidative stress may alter LCN2 homeostasis, even under neuroinflammatory conditions.
Collapse
Affiliation(s)
| | | | - Kwon-Yul Ryu
- Department of Life Science, University of Seoul, Seoul 02504, Republic of Korea; (Y.-J.C.); (S.-H.P.)
| |
Collapse
|
26
|
Martinez-Lozada Z, Todd FW, Schober AL, Krizman E, Robinson MB, Murai KK. Cooperative and competitive regulation of the astrocytic transcriptome by neurons and endothelial cells: Impact on astrocyte maturation. J Neurochem 2023; 167:52-75. [PMID: 37525469 PMCID: PMC10543513 DOI: 10.1111/jnc.15908] [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: 03/01/2023] [Revised: 06/19/2023] [Accepted: 06/20/2023] [Indexed: 08/02/2023]
Abstract
Astrocytes have essential roles in central nervous system (CNS) health and disease. During development, immature astrocytes show complex interactions with neurons, endothelial cells, and other glial cell types. Our work and that of others have shown that these interactions are important for astrocytic maturation. However, whether and how these cells work together to control this process remains poorly understood. Here, we test the hypothesis that cooperative interactions of astrocytes with neurons and endothelial cells promote astrocytic maturation. Astrocytes were cultured alone, with neurons, endothelial cells, or a combination of both. This was followed by astrocyte sorting, RNA sequencing, and bioinformatic analysis to detect transcriptional changes. Across culture configurations, 7302 genes were differentially expressed by 4 or more fold and organized into 8 groups that demonstrate cooperative and antagonist effects of neurons and endothelia on astrocytes. We also discovered that neurons and endothelial cells caused splicing of 200 and 781 mRNAs, respectively. Changes in gene expression were validated using quantitative PCR, western blot (WB), and immunofluorescence analysis. We found that the transcriptomic data from the three-culture configurations correlated with protein expression of three representative targets (FAM107A, GAT3, and GLT1) in vivo. Alternative splicing results also correlated with cortical tissue isoform representation of a target (Fibronectin 1) at different developmental stages. By comparing our results to published transcriptomes of immature and mature astrocytes, we found that neurons or endothelia shift the astrocytic transcriptome toward a mature state and that the presence of both cell types has a greater effect on maturation than either cell alone. These results increase our understanding of cellular interactions/pathways that contribute to astrocytic maturation. They also provide insight into how alterations to neurons and/or endothelial cells may alter astrocytes with implications for astrocytic changes in CNS disorders and diseases.
Collapse
Affiliation(s)
- Zila Martinez-Lozada
- Departments of Pediatrics and Systems Pharmacology & Translational Therapeutics, The Children’s Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA, USA, 19104-4318
| | - Farmer W. Todd
- Centre for Research in Neuroscience, Department of Neurology & Neurosurgery, Brain Repair and Integrative Neuroscience Program, The Research Institute of the McGill University Health Centre, Montreal General Hospital, Montreal, Quebec, Canada H3G 1A4
| | - Alexandra L. Schober
- Centre for Research in Neuroscience, Department of Neurology & Neurosurgery, Brain Repair and Integrative Neuroscience Program, The Research Institute of the McGill University Health Centre, Montreal General Hospital, Montreal, Quebec, Canada H3G 1A4
| | - Elizabeth Krizman
- Departments of Pediatrics and Systems Pharmacology & Translational Therapeutics, The Children’s Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA, USA, 19104-4318
| | - Michael B. Robinson
- Departments of Pediatrics and Systems Pharmacology & Translational Therapeutics, The Children’s Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA, USA, 19104-4318
| | - Keith K. Murai
- Centre for Research in Neuroscience, Department of Neurology & Neurosurgery, Brain Repair and Integrative Neuroscience Program, The Research Institute of the McGill University Health Centre, Montreal General Hospital, Montreal, Quebec, Canada H3G 1A4
| |
Collapse
|
27
|
Cullen PF, Mazumder AG, Sun D, Flanagan JG. Rapid isolation of intact retinal astrocytes: a novel approach. Acta Neuropathol Commun 2023; 11:154. [PMID: 37749651 PMCID: PMC10521529 DOI: 10.1186/s40478-023-01641-7] [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: 07/07/2023] [Accepted: 08/23/2023] [Indexed: 09/27/2023] Open
Abstract
Astrocytes are a major category of glial support cell in the central nervous system and play a variety of essential roles in both health and disease. As our understanding of the diverse functions of these cells improves, the extent of heterogeneity between astrocyte populations has emerged as a key area of research. Retinal astrocytes, which form the direct cellular environment of retinal ganglion cells somas and axons, undergo a reactive response in both human glaucoma and animal models of the disease, yet their contributions to its pathology and progression remain relatively unknown. This gap in knowledge is largely a function of inadequate isolation techniques, driven in part by the sparseness of these cells and their similarities with the more abundant retinal Müller cells. Here, we present a novel method of isolating retinal astrocytes and enriching their RNA, tested in both normal and ocular hypertensive mice, a common model of experimental glaucoma. Our approach combines a novel enzyme assisted microdissection of retinal astrocytes with selective ribosome immunoprecipitation using the Ribotag method. Our microdissection method is rapid and preserves astrocyte morphology, resulting in a brief post-mortem interval and minimizing loss of RNA from distal regions of these cells. Both microdissection and Ribotag immunoprecipitation require a minimum of specialized equipment or reagents, and by using them in conjunction we are able to achieve > 100-fold enrichment of astrocyte RNA.
Collapse
Affiliation(s)
- Paul F Cullen
- Department of Ophthalmology, Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, 02114, USA
| | - Arpan G Mazumder
- Department of Ophthalmology, Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, 02114, USA
| | - Daniel Sun
- Department of Ophthalmology, Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, 02114, USA.
| | - John G Flanagan
- Herbert Wertheim School of Optometry and Vision Science, University of California at Berkeley, Berkeley, CA, USA
| |
Collapse
|
28
|
Clark DN, O'Neil SM, Xu L, Steppe JT, Savage JT, Raghunathan K, Filiano AJ. Prolonged STAT1 activation in neurons drives a pathological transcriptional response. J Neuroimmunol 2023; 382:578168. [PMID: 37556887 PMCID: PMC10527980 DOI: 10.1016/j.jneuroim.2023.578168] [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: 05/15/2023] [Revised: 07/21/2023] [Accepted: 07/31/2023] [Indexed: 08/11/2023]
Abstract
Neurons require physiological IFN-γ signaling to maintain central nervous system (CNS) homeostasis, however, pathological IFN-γ signaling can cause CNS pathologies. The downstream signaling mechanisms that cause these drastically different outcomes in neurons has not been well studied. We hypothesized that different levels of IFN-γ signaling in neurons results in differential activation of its downstream transcription factor, signal transducer and activator of transduction 1 (STAT1), causing varying outcomes. Using primary cortical neurons, we showed that physiological IFN-γ elicited brief and transient STAT1 activation, whereas pathological IFN-γ induced prolonged STAT1 activation, which primed the pathway to be more responsive to a subsequent IFN-γ challenge. This is an IFN-γ specific response, as other IFNs and cytokines did not elicit such STAT1 activation nor priming in neurons. Additionally, we did not see the same effect in microglia or astrocytes, suggesting this non-canonical IFN-γ/STAT1 signaling is unique to neurons. Prolonged STAT1 activation was facilitated by continuous janus kinase (JAK) activity, even in the absence of IFN-γ. Finally, although IFN-γ initially induced a canonical IFN-γ transcriptional response in neurons, pathological levels of IFN-γ caused long-term changes in synaptic pathway transcripts. Overall, these findings suggest that IFN-γ signaling occurs via non-canonical mechanisms in neurons, and differential STAT1 activation may explain how neurons have both homeostatic and pathological responses to IFN-γ signaling.
Collapse
Affiliation(s)
- Danielle N Clark
- Department of Integrative Immunobiology, Duke University, Durham, NC 27705, USA; Marcus Center for Cellular Cures, Duke University, Durham, NC 27705, USA
| | - Shane M O'Neil
- Marcus Center for Cellular Cures, Duke University, Durham, NC 27705, USA
| | - Li Xu
- Marcus Center for Cellular Cures, Duke University, Durham, NC 27705, USA
| | - Justin T Steppe
- Department of Pathology, Duke University, Durham, NC 27705, USA
| | - Justin T Savage
- Department of Neurobiology, Duke University, Durham, NC 27705, USA
| | | | - Anthony J Filiano
- Department of Integrative Immunobiology, Duke University, Durham, NC 27705, USA; Department of Pathology, Duke University, Durham, NC 27705, USA; Department of Neurosurgery, Duke University, Durham, NC 27705, USA; Marcus Center for Cellular Cures, Duke University, Durham, NC 27705, USA.
| |
Collapse
|
29
|
Byun YG, Kim NS, Kim G, Jeon YS, Choi JB, Park CW, Kim K, Jang H, Kim J, Kim E, Han YM, Yoon KJ, Lee SH, Chung WS. Stress induces behavioral abnormalities by increasing expression of phagocytic receptor MERTK in astrocytes to promote synapse phagocytosis. Immunity 2023; 56:2105-2120.e13. [PMID: 37527657 DOI: 10.1016/j.immuni.2023.07.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 05/09/2023] [Accepted: 07/06/2023] [Indexed: 08/03/2023]
Abstract
Childhood neglect and/or abuse can induce mental health conditions with unknown mechanisms. Here, we identified stress hormones as strong inducers of astrocyte-mediated synapse phagocytosis. Using in vitro, in vivo, and human brain organoid experiments, we showed that stress hormones increased the expression of the Mertk phagocytic receptor in astrocytes through glucocorticoid receptor (GR). In post-natal mice, exposure to early social deprivation (ESD) specifically activated the GR-MERTK pathway in astrocytes, but not in microglia. The excitatory post-synaptic density in cortical regions was reduced in ESD mice, and there was an increase in the astrocytic engulfment of these synapses. The loss of excitatory synapses, abnormal neuronal network activities, and behavioral abnormalities in ESD mice were largely prevented by ablating GR or MERTK in astrocytes. Our work reveals the critical roles of astrocytic GR-MERTK activation in evoking stress-induced abnormal behaviors in mice, suggesting GR-MERTK signaling as a therapeutic target for stress-induced mental health conditions.
Collapse
Affiliation(s)
- Youkyeong Gloria Byun
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea; KAIST Stem Cell Center, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Nam-Shik Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea; KAIST Stem Cell Center, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Gyuri Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea; KAIST Stem Cell Center, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Yi-Seon Jeon
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea; Center for Synaptic Brain Dysfunctions, Institute for Basic Science, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Jong Bin Choi
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea; KAIST Stem Cell Center, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea; Graduate School of Medical Science and Engineering, KAIST, Daejeon 34141, Republic of Korea
| | - Chan-Woo Park
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea; KAIST Stem Cell Center, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Kyungdeok Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea; Center for Synaptic Brain Dysfunctions, Institute for Basic Science, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Hyunsoo Jang
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea; KAIST Stem Cell Center, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Jinkyeong Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea; Center for Synaptic Brain Dysfunctions, Institute for Basic Science, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Eunjoon Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea; Center for Synaptic Brain Dysfunctions, Institute for Basic Science, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Yong-Mahn Han
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea; KAIST Stem Cell Center, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea; Graduate School of Medical Science and Engineering, KAIST, Daejeon 34141, Republic of Korea
| | - Ki-Jun Yoon
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea; KAIST Stem Cell Center, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Seung-Hee Lee
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea; Center for Synaptic Brain Dysfunctions, Institute for Basic Science, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Won-Suk Chung
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea; KAIST Stem Cell Center, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea.
| |
Collapse
|
30
|
Frazel PW, Prakash P. Transplanted Astrocytes Show Functional Flexibility in the Recipient Brain. J Neurosci 2023; 43:6228-6229. [PMID: 37673676 PMCID: PMC10490451 DOI: 10.1523/jneurosci.0802-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 07/20/2023] [Accepted: 07/20/2023] [Indexed: 09/08/2023] Open
Affiliation(s)
- Paul W Frazel
- Neuroscience Institute, New York University Grossman School of Medicine, New York, New York 10016
| | - Priya Prakash
- Neuroscience Institute, New York University Grossman School of Medicine, New York, New York 10016
| |
Collapse
|
31
|
Zelenka L, Jarek M, Pägelow D, Geffers R, van Vorst K, Fulde M. Crosstalk of Highly Purified Microglia and Astrocytes in the Frame of Toll-like Receptor (TLR)2/1 Activation. Neuroscience 2023; 526:256-266. [PMID: 37391121 DOI: 10.1016/j.neuroscience.2023.05.001] [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: 07/21/2022] [Revised: 04/26/2023] [Accepted: 05/02/2023] [Indexed: 07/02/2023]
Abstract
The major immune cells of the central nervous systems (CNS) are microglia and astrocytes, subsets of the glial cell population. The crosstalk between glia via soluble signaling molecules plays an indispensable role for neuropathologies, brain development as well as homeostasis. However, the investigation of the microglia-astrocyte crosstalk has been hampered due to the lack of suitable glial isolation methods. In this study, we investigated for the first time the crosstalk between highly purified Toll-like receptor (TLR)2-knock out (TLR2-KO) and wild-type (WT) microglia and astrocytes. We examined the crosstalk of TLR2-KO microglia and astrocytes in the presence of WT supernatants of the respective other glial cell type. Interestingly, we observed a significant TNF release by TLR2-KO astrocytes, which were activated with Pam3CSK4-stimulated WT microglial supernatants, strongly indicating a crosstalk between microglia and astrocytes after TLR2/1 activation. Furthermore, transcriptome analysis using RNA-seq revealed a wide range of significant up- and down-regulated genes such as Cd300, Tnfrsf9 or Lcn2, which might be involved in the molecular conversation between microglia and astrocytes. Finally, co-culturing microglia and astrocytes confirmed the prior results by demonstrating a significant TNF release by WT microglia co-cultured with TLR2-KO astrocytes. Our findings suggest a molecular TLR2/1-dependent conversation between highly pure activated microglia and astrocytes via signaling molecules. Furthermore, we demonstrate the first crosstalk experiments using ∼100% pure microglia and astrocyte mono-/co-cultures derived from mice with different genotypes highlighting the urgent need of efficient glial isolation protocols, which particularly holds true for astrocytes.
Collapse
Affiliation(s)
- Laura Zelenka
- Centre for Infection Medicine, Institute of Microbiology and Epizootics, Freie Universität Berlin, Berlin, Germany
| | - Michael Jarek
- Helmholtz Centre for Infection Research, Research Group Genome Analytics (GMAK), Braunschweig, Germany
| | - Dennis Pägelow
- Centre for Infection Medicine, Institute of Microbiology and Epizootics, Freie Universität Berlin, Berlin, Germany
| | - Robert Geffers
- Helmholtz Centre for Infection Research, Research Group Genome Analytics (GMAK), Braunschweig, Germany
| | - Kira van Vorst
- Centre for Infection Medicine, Institute of Microbiology and Epizootics, Freie Universität Berlin, Berlin, Germany
| | - Marcus Fulde
- Centre for Infection Medicine, Institute of Microbiology and Epizootics, Freie Universität Berlin, Berlin, Germany.
| |
Collapse
|
32
|
Nguyen TT, Camp CR, Doan JK, Traynelis SF, Sloan SA, Hall RA. GPR37L1 controls maturation and organization of cortical astrocytes during development. Glia 2023; 71:1921-1946. [PMID: 37029775 PMCID: PMC10315172 DOI: 10.1002/glia.24375] [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: 06/02/2022] [Revised: 02/24/2023] [Accepted: 03/24/2023] [Indexed: 04/09/2023]
Abstract
Astrocyte maturation is crucial to proper brain development and function. This maturation process includes the ramification of astrocytic morphology and the establishment of astrocytic domains. While this process has been well-studied, the mechanisms by which astrocyte maturation is initiated are not well understood. GPR37L1 is an astrocyte-specific G protein-coupled receptor (GPCR) that is predominantly expressed in mature astrocytes and has been linked to the modulation of seizure susceptibility in both humans and mice. To investigate the role of GPR37L1 in astrocyte biology, RNA-seq analyses were performed on astrocytes immunopanned from P7 Gpr37L1-/- knockout (L1KO) mouse cortex and compared to those from wild-type (WT) mouse cortex. These RNA-seq studies revealed that pathways involved in central nervous system development were altered and that L1KO cortical astrocytes express lower amounts of mature astrocytic genes compared to WT astrocytes. Immunohistochemical studies of astrocytes from L1KO mouse brain revealed that these astrocytes exhibit overall shorter total process length, and are also less complex and spaced further apart from each other in the mouse cortex. This work sheds light on how GPR37L1 regulates cellular processes involved in the control of astrocyte biology and maturation.
Collapse
Affiliation(s)
| | - Chad R. Camp
- Emory University School of Medicine, Department of Pharmacology and Chemical Biology
| | - Juleva K. Doan
- Emory University School of Medicine, Department of Pharmacology and Chemical Biology
| | - Stephen F. Traynelis
- Emory University School of Medicine, Department of Pharmacology and Chemical Biology
| | - Steven A. Sloan
- Emory University School of Medicine, Department of Human Genetics
| | - Randy A. Hall
- Emory University School of Medicine, Department of Pharmacology and Chemical Biology
| |
Collapse
|
33
|
Jung BK, Park Y, Yoon B, Bae JS, Han SW, Heo JE, Kim DE, Ryu KY. Reduced secretion of LCN2 (lipocalin 2) from reactive astrocytes through autophagic and proteasomal regulation alleviates inflammatory stress and neuronal damage. Autophagy 2023; 19:2296-2317. [PMID: 36781380 PMCID: PMC10351455 DOI: 10.1080/15548627.2023.2180202] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 02/06/2023] [Accepted: 02/09/2023] [Indexed: 02/15/2023] Open
Abstract
LCN2/neutrophil gelatinase-associated lipocalin/24p3 (lipocalin 2) is a secretory protein that acts as a mammalian bacteriostatic molecule. Under neuroinflammatory stress conditions, LCN2 is produced and secreted by activated microglia and reactive astrocytes, resulting in neuronal apoptosis. However, it remains largely unknown whether inflammatory stress and neuronal loss can be minimized by modulating LCN2 production and secretion. Here, we first demonstrated that LCN2 was secreted from reactive astrocytes, which were stimulated by treatment with lipopolysaccharide (LPS) as an inflammatory stressor. Notably, we found two effective conditions that led to the reduction of induced LCN2 levels in reactive astrocytes: proteasome inhibition and macroautophagic/autophagic flux activation. Mechanistically, proteasome inhibition suppresses NFKB/NF-κB activation through NFKBIA/IκBα stabilization in primary astrocytes, even under inflammatory stress conditions, resulting in the downregulation of Lcn2 expression. In contrast, autophagic flux activation via MTOR inhibition reduced the intracellular levels of LCN2 through its pre-secretory degradation. In addition, we demonstrated that the N-terminal signal peptide of LCN2 is critical for its secretion and degradation, suggesting that these two pathways may be mechanistically coupled. Finally, we observed that LPS-induced and secreted LCN2 levels were reduced in the astrocyte-cultured medium under the above-mentioned conditions, resulting in increased neuronal viability, even under inflammatory stress.Abbreviations: ACM, astrocyte-conditioned medium; ALP, autophagy-lysosome pathway; BAF, bafilomycin A1; BTZ, bortezomib; CHX, cycloheximide; CNS, central nervous system; ER, endoplasmic reticulum; GFAP, glial fibrillary acidic protein; GFP, green fluorescent protein; JAK, Janus kinase; KD, knockdown; LCN2, lipocalin 2; LPS, lipopolysaccharide; MACS, magnetic-activated cell sorting; MAP1LC3/LC3, microtubule-associated protein 1 light chain 3; MTOR, mechanistic target of rapamycin kinase; NFKB/NF-κB, nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105; NFKBIA/IκBα, nuclear factor of kappa light polypeptide gene enhancer in B cells inhibitor, alpha; OVEX, overexpression; SLC22A17, solute carrier family 22 member 17; SP, signal peptide; SQSTM1, sequestosome 1; STAT3, signal transducer and activator of transcription 3; TNF/TNF-α, tumor necrosis factor; TUBA, tubulin, alpha; TUBB3/β3-TUB, tubulin, beta 3 class III; UB, ubiquitin; UPS, ubiquitin-proteasome system.
Collapse
Affiliation(s)
- Byung-Kwon Jung
- Department of Life Science, University of Seoul, Seoul, Republic of Korea
| | - Yujin Park
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, Republic of Korea
| | - Boran Yoon
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, Republic of Korea
| | - Jin-Sil Bae
- Department of Life Science, University of Seoul, Seoul, Republic of Korea
| | - Seung-Woo Han
- Department of Life Science, University of Seoul, Seoul, Republic of Korea
| | - Ji-Eun Heo
- Department of Life Science, University of Seoul, Seoul, Republic of Korea
| | - Dong-Eun Kim
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, Republic of Korea
| | - Kwon-Yul Ryu
- Department of Life Science, University of Seoul, Seoul, Republic of Korea
| |
Collapse
|
34
|
Patani R, Hardingham GE, Liddelow SA. Functional roles of reactive astrocytes in neuroinflammation and neurodegeneration. Nat Rev Neurol 2023; 19:395-409. [PMID: 37308616 DOI: 10.1038/s41582-023-00822-1] [Citation(s) in RCA: 102] [Impact Index Per Article: 102.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/11/2023] [Indexed: 06/14/2023]
Abstract
Despite advances in uncovering the mechanisms that underlie neuroinflammation and neurodegenerative disease, therapies that prevent neuronal loss remain elusive. Targeting of disease-defining markers in conditions such as Alzheimer disease (amyloid-β and tau) or Parkinson disease (α-synuclein) has been met with limited success, suggesting that these proteins do not act in isolation but form part of a pathological network. This network could involve phenotypic alteration of multiple cell types in the CNS, including astrocytes, which have a major neurosupportive, homeostatic role in the healthy CNS but adopt reactive states under acute or chronic adverse conditions. Transcriptomic studies in human patients and disease models have revealed the co-existence of many putative reactive sub-states of astrocytes. Inter-disease and even intra-disease heterogeneity of reactive astrocytic sub-states are well established, but the extent to which specific sub-states are shared across different diseases is unclear. In this Review, we highlight how single-cell and single-nuclei RNA sequencing and other 'omics' technologies can enable the functional characterization of defined reactive astrocyte states in various pathological scenarios. We provide an integrated perspective, advocating cross-modal validation of key findings to define functionally important sub-states of astrocytes and their triggers as tractable therapeutic targets with cross-disease relevance.
Collapse
Affiliation(s)
- Rickie Patani
- Department of Neuromuscular Disease, UCL Queen Square Institute of Neurology, London, UK
- The Francis Crick Institute, Human Stem Cells and Neurodegeneration Laboratory, London, UK
| | - Giles E Hardingham
- Euan MacDonald Centre for MND, University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute at the University of Edinburgh, University of Edinburgh, Edinburgh, UK
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Shane A Liddelow
- Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, USA.
- Department of Neuroscience & Physiology, NYU Grossman School of Medicine, New York, NY, USA.
- Department of Ophthalmology, NYU Grossman School of Medicine, New York, NY, USA.
- Parekh Center for Interdisciplinary Neurology, NYU Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
35
|
Lindblad C, Neumann S, Kolbeinsdóttir S, Zachariadis V, Thelin EP, Enge M, Thams S, Brundin L, Svensson M. Stem cell-derived brainstem mouse astrocytes obtain a neurotoxic phenotype in vitro upon neuroinflammation. J Inflamm (Lond) 2023; 20:22. [PMID: 37370141 PMCID: PMC10303821 DOI: 10.1186/s12950-023-00349-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
BACKGROUND Astrocytes respond to injury and disease through a process known as reactive astrogliosis, of which inflammatory signaling is one subset. This inflammatory response is heterogeneous with respect to the inductive stimuli and the afflicted central nervous system region. This is of plausible importance in e.g. traumatic axonal injury (TAI), where lesions in the brainstem carries a particularly poor prognosis. In fact, astrogliotic forebrain astrocytes were recently suggested to cause neuronal death following axotomy. We therefore sought to assess if ventral brainstem- or rostroventral spinal astrocytes exert similar effects on motor neurons in vitro. METHODS We derived brainstem/rostroventral spinal astrocyte-like cells (ES-astrocytes) and motor neurons using directed differentiation of mouse embryonic stem cells (ES). We activated the ES-astrocytes using the neurotoxicity-eliciting cytokines interleukin- (IL-) 1α and tumor necrosis factor-(TNF-)α and clinically relevant inflammatory mediators. In co-cultures with reactive ES-astrocytes and motor neurons, we assessed neurotoxic ES-astrocyte activity, similarly to what has previously been shown for other central nervous system (CNS) regions. RESULTS We confirmed the brainstem/rostroventral ES-astrocyte identity using RNA-sequencing, immunocytochemistry, and by comparison with primary subventricular zone-astrocytes. Following cytokine stimulation, the c-Jun N-terminal kinase pathway down-stream product phosphorylated c-Jun was increased, thus demonstrating ES-astrocyte reactivity. These reactive ES-astrocytes conferred a contact-dependent neurotoxic effect upon co-culture with motor neurons. When exposed to IL-1β and IL-6, two neuroinflammatory cytokines found in the cerebrospinal fluid and serum proteome following human severe traumatic brain injury (TBI), ES-astrocytes exerted similar effects on motor neurons. Activation of ES-astrocytes by these cytokines was associated with pathways relating to endoplasmic reticulum stress and altered regulation of MYC. CONCLUSIONS Ventral brainstem and rostroventral spinal cord astrocytes differentiated from mouse ES can exert neurotoxic effects in vitro. This highlights how neuroinflammation following CNS lesions can exert region- and cell-specific effects. Our in vitro model system, which uniquely portrays astrocytes and neurons from one niche, allows for a detailed and translationally relevant model system for future studies on how to improve neuronal survival in particularly vulnerable CNS regions following e.g. TAI.
Collapse
Affiliation(s)
- Caroline Lindblad
- Department of Clinical Neuroscience, Karolinska Institutet, J5:20 Svensson Group, Karolinska Universitetssjukhuset Solna, SE-171 77, Stockholm, Sweden.
| | - Susanne Neumann
- Department of Clinical Neuroscience, Karolinska Institutet, J5:20 Svensson Group, Karolinska Universitetssjukhuset Solna, SE-171 77, Stockholm, Sweden
| | | | | | - Eric P Thelin
- Department of Clinical Neuroscience, Karolinska Institutet, J5:20 Svensson Group, Karolinska Universitetssjukhuset Solna, SE-171 77, Stockholm, Sweden
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden
| | - Martin Enge
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Sebastian Thams
- Department of Clinical Neuroscience, Karolinska Institutet, J5:20 Svensson Group, Karolinska Universitetssjukhuset Solna, SE-171 77, Stockholm, Sweden
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden
| | - Lou Brundin
- Department of Clinical Neuroscience, Karolinska Institutet, J5:20 Svensson Group, Karolinska Universitetssjukhuset Solna, SE-171 77, Stockholm, Sweden
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden
| | - Mikael Svensson
- Department of Clinical Neuroscience, Karolinska Institutet, J5:20 Svensson Group, Karolinska Universitetssjukhuset Solna, SE-171 77, Stockholm, Sweden
- Department of Neurosurgery, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
36
|
Zhao Q, Li H, Li H, Xie F, Zhang J. Research progress of neuroinflammation-related cells in traumatic brain injury: A review. Medicine (Baltimore) 2023; 102:e34009. [PMID: 37352020 PMCID: PMC10289497 DOI: 10.1097/md.0000000000034009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/24/2023] [Accepted: 05/25/2023] [Indexed: 06/25/2023] Open
Abstract
Neuroinflammation after traumatic brain injury (TBI) is related to chronic neurodegenerative diseases and is one of the causes of acute secondary injury after TBI. Therefore, it is particularly important to clarify the role of cellular mechanisms in the neuroinflammatory response after TBI. The objective of this article is to understand the involvement of cells during the TBI inflammatory response (for instance, astrocytes, microglia, and oligodendrocytes) and shed light on the recent progress in the stimulation and interaction of granulocytes and lymphocytes, to provide a novel approach for clinical research. We searched articles in PubMed published between 1950 and 2023, using the following keywords: TBI, neuroinflammation, inflammatory cells, neuroprotection, clinical. Articles for inclusion in this paper were finalized based on their novelty, representativeness, and relevance to the main arguments of this review. We found that the neuroinflammatory response after TBI includes the activation of glial cells, the release of inflammatory mediators in the brain, and the recruitment of peripheral immune cells. These inflammatory responses not only induce secondary brain damage, but also have a role in repairing the nervous system to some extent. However, not all of the mechanisms of cell-to-cell interactions have been well studied. After TBI, clinical treatment cannot simply suppress the inflammatory response, and the inflammatory phenotype of patients' needs to be defined according to their specific conditions after injury. Clinical trials of personalized inflammation regulation therapy for specific patients should be carried out in order to improve the prognosis of patients.
Collapse
Affiliation(s)
- Qinghui Zhao
- Institute of Physical Culture, Huanghuai University, Zhumadian, China
| | - Huige Li
- Institute of Physical Culture, Huanghuai University, Zhumadian, China
| | - Hongru Li
- Zhumadian Central Hospital, Zhumadian, China
| | - Fei Xie
- Faculty of Environment and Life, Beijing University of Technology, Beijing, China
| | - Jianhua Zhang
- Institute of Physical Culture, Huanghuai University, Zhumadian, China
| |
Collapse
|
37
|
Levy-Myers R, Daudelin D, Na CH, Sockanathan S. An independent regulator of global release pathways in astrocytes generates a subtype of extracellular vesicles required for postsynaptic function. SCIENCE ADVANCES 2023; 9:eadg2067. [PMID: 37352348 PMCID: PMC10289663 DOI: 10.1126/sciadv.adg2067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 05/18/2023] [Indexed: 06/25/2023]
Abstract
Extracellular vesicles (EVs) are heterogeneous in size, composition, and function. We show that the six-transmembrane protein glycerophosphodiester phosphodiesterase 3 (GDE3) regulates actin remodeling, a global EV biogenic pathway, to release an EV subtype with distinct functions. GDE3 is necessary and sufficient for releasing EVs containing annexin A1 and GDE3 from the plasma membrane via Wiskott-Aldrich syndrome protein family member 3 (WAVE3), a major regulator of actin dynamics. GDE3 is expressed in astrocytes but not neurons, yet mice lacking GDE3 [Gde3 knockout (KO)] have decreased miniature excitatory postsynaptic current (mEPSC) amplitudes in hippocampal CA1 neurons. EVs from cultured wild-type astrocytes restore mEPSC amplitudes in Gde3 KOs, while EVs from Gde3 KO astrocytes or astrocytes inhibited for WAVE3 actin branching activity do not. Thus, GDE3-WAVE3 is a nonredundant astrocytic pathway that remodels actin to release a functionally distinct EV subtype, supporting the concept that independent regulation of global EV release pathways differentially regulates EV signaling within the cellular EV landscape.
Collapse
Affiliation(s)
- Reuben Levy-Myers
- The Solomon Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, PCTB1004, 725 N. Wolfe Street, Baltimore, MD 21205, USA
| | - Daniel Daudelin
- The Solomon Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, PCTB1004, 725 N. Wolfe Street, Baltimore, MD 21205, USA
| | - Chan Hyun Na
- Department of Neurology, Institute for Cell Engineering, Johns Hopkins University School of Medicine, MRB 706, 733 N. Broadway, Baltimore, MD 21205, USA
| | - Shanthini Sockanathan
- The Solomon Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, PCTB1004, 725 N. Wolfe Street, Baltimore, MD 21205, USA
| |
Collapse
|
38
|
Dong X, Shu L, Zhang J, Yang X, Cheng X, Zhao X, Qu W, Zhu Q, Shou Y, Peng G, Sun B, Yi W, Shu Q, Li X. Ogt-mediated O-GlcNAcylation inhibits astrocytes activation through modulating NF-κB signaling pathway. J Neuroinflammation 2023; 20:146. [PMID: 37349834 DOI: 10.1186/s12974-023-02824-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 06/05/2023] [Indexed: 06/24/2023] Open
Abstract
Previous studies have shown that Ogt-mediated O-GlcNAcylation is essential for neuronal development and function. However, the function of O-GlcNAc transferase (Ogt) and O-GlcNAcylation in astrocytes remains largely unknown. Here we show that Ogt deficiency induces inflammatory activation of astrocytes in vivo and in vitro, and impairs cognitive function of mice. The restoration of O-GlcNAcylation via GlcNAc supplementation inhibits the activation of astrocytes, inflammation and improves the impaired cognitive function of Ogt deficient mice. Mechanistically, Ogt interacts with NF-κB p65 and catalyzes the O-GlcNAcylation of NF-κB p65 in astrocytes. Ogt deficiency induces the activation of NF-κB signaling pathway by promoting Gsk3β binding. Moreover, Ogt depletion induces the activation of astrocytes derived from human induced pluripotent stem cells. The restoration of O-GlcNAcylation inhibits the activation of astrocytes, inflammation and reduces Aβ plaque of AD mice in vitro and in vivo. Collectively, our study reveals a critical function of Ogt-mediated O-GlcNAcylation in astrocytes through regulating NF-κB signaling pathway.
Collapse
Affiliation(s)
- Xiaoxue Dong
- The Children's Hospital, National Clinical Research Center for Child Health, School of Medicine, Zhejiang University, Hangzhou, 310052, China
- The Institute of Translational Medicine, School of Medicine, Zhejiang University, Hangzhou, 310029, China
| | - Liqi Shu
- Department of Neurology, The Warren Alpert Medical School of Brown University, Providence, RI, 02908, USA
| | - Jinyu Zhang
- The Children's Hospital, National Clinical Research Center for Child Health, School of Medicine, Zhejiang University, Hangzhou, 310052, China
- The Institute of Translational Medicine, School of Medicine, Zhejiang University, Hangzhou, 310029, China
| | - Xu Yang
- The Children's Hospital, National Clinical Research Center for Child Health, School of Medicine, Zhejiang University, Hangzhou, 310052, China
- The Institute of Translational Medicine, School of Medicine, Zhejiang University, Hangzhou, 310029, China
| | - Xuejun Cheng
- The Children's Hospital, National Clinical Research Center for Child Health, School of Medicine, Zhejiang University, Hangzhou, 310052, China
| | - Xingsen Zhao
- The Children's Hospital, National Clinical Research Center for Child Health, School of Medicine, Zhejiang University, Hangzhou, 310052, China
- The Institute of Translational Medicine, School of Medicine, Zhejiang University, Hangzhou, 310029, China
| | - Wenzheng Qu
- The Children's Hospital, National Clinical Research Center for Child Health, School of Medicine, Zhejiang University, Hangzhou, 310052, China
| | - Qiang Zhu
- MOE Key Laboratory of Biosystems Homeostasis & Protection, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yikai Shou
- The Children's Hospital, National Clinical Research Center for Child Health, School of Medicine, Zhejiang University, Hangzhou, 310052, China
| | - Guoping Peng
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Binggui Sun
- The Children's Hospital, National Clinical Research Center for Child Health, School of Medicine, Zhejiang University, Hangzhou, 310052, China.
- NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, 310058, Zhejiang, China.
| | - Wen Yi
- MOE Key Laboratory of Biosystems Homeostasis & Protection, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China.
| | - Qiang Shu
- The Children's Hospital, National Clinical Research Center for Child Health, School of Medicine, Zhejiang University, Hangzhou, 310052, China.
| | - Xuekun Li
- The Children's Hospital, National Clinical Research Center for Child Health, School of Medicine, Zhejiang University, Hangzhou, 310052, China.
- The Institute of Translational Medicine, School of Medicine, Zhejiang University, Hangzhou, 310029, China.
- Zhejiang University Cancer Center, Zhejiang University, Hangzhou, 310029, China.
- Binjiang Institute of Zhejiang University, Hangzhou, 310053, China.
| |
Collapse
|
39
|
Kloske CM, Barnum CJ, Batista AF, Bradshaw EM, Brickman AM, Bu G, Dennison J, Gearon MD, Goate AM, Haass C, Heneka MT, Hu WT, Huggins LKL, Jones NS, Koldamova R, Lemere CA, Liddelow SA, Marcora E, Marsh SE, Nielsen HM, Petersen KK, Petersen M, Piña-Escudero SD, Qiu WQ, Quiroz YT, Reiman E, Sexton C, Tansey MG, Tcw J, Teunissen CE, Tijms BM, van der Kant R, Wallings R, Weninger SC, Wharton W, Wilcock DM, Wishard TJ, Worley SL, Zetterberg H, Carrillo MC. APOE and immunity: Research highlights. Alzheimers Dement 2023; 19:2677-2696. [PMID: 36975090 DOI: 10.1002/alz.13020] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 12/19/2022] [Indexed: 03/29/2023]
Abstract
INTRODUCTION At the Alzheimer's Association's APOE and Immunity virtual conference, held in October 2021, leading neuroscience experts shared recent research advances on and inspiring insights into the various roles that both the apolipoprotein E gene (APOE) and facets of immunity play in neurodegenerative diseases, including Alzheimer's disease and other dementias. METHODS The meeting brought together more than 1200 registered attendees from 62 different countries, representing the realms of academia and industry. RESULTS During the 4-day meeting, presenters illuminated aspects of the cross-talk between APOE and immunity, with a focus on the roles of microglia, triggering receptor expressed on myeloid cells 2 (TREM2), and components of inflammation (e.g., tumor necrosis factor α [TNFα]). DISCUSSION This manuscript emphasizes the importance of diversity in current and future research and presents an integrated view of innate immune functions in Alzheimer's disease as well as related promising directions in drug development.
Collapse
Affiliation(s)
| | | | - Andre F Batista
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Departments of Neurology, Harvard Medical School, Boston, Massachusetts, USA
| | - Elizabeth M Bradshaw
- Department of Neurology, Columbia University Irving Medical Center, New York, New York, USA
| | - Adam M Brickman
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, G.H. Sergievsky Center, and Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Guojun Bu
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | - Jessica Dennison
- Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Mary D Gearon
- Department of Physiology, University of Kentucky, Lexington, Kentucky, USA
| | - Alison M Goate
- Department of Genetics & Genomic Sciences, Ronald M. Loeb Center for Alzheimer's disease, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Christian Haass
- Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, LMU Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany 3 Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Michael T Heneka
- Luxembourg Centre for Systems Biomedicine (LCSB) University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - William T Hu
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School and Center for Healthy Aging, Rutgers Institute for Health, Health Care Policy, and Aging Research, New Brunswick, New Jersey, USA
| | - Lenique K L Huggins
- Department of Biology, Duke University, Durham, North Carolina, USA
- Yale School of Medicine, New Haven, Connecticut, USA
| | - Nahdia S Jones
- Interdisciplinary Program in Neuroscience, Georgetown University, Washington, District of Columbia, USA
| | - Radosveta Koldamova
- EOH, School of Public Health University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Cynthia A Lemere
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Departments of Neurology, Harvard Medical School, Boston, Massachusetts, USA
| | - Shane A Liddelow
- Neuroscience Institute and Departments of Neuroscience & Physiology and of Ophthalmology, NYU Grossman School of Medicine, New York, New York, USA
| | - Edoardo Marcora
- Ronald M. Loeb Center for Alzheimer's disease, Dept. of Genetics & Genomic Sciences, Dept. of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Samuel E Marsh
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Henrietta M Nielsen
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Kellen K Petersen
- The Saul R. Korey Department of Neurology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Melissa Petersen
- Department of Family Medicine, Institute of Translational Research, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - Stefanie D Piña-Escudero
- Global Brain Health Institute, Department of Neurology, University of California, San Francisco, California, USA
| | - Wei Qiao Qiu
- Boston University School of Medicine, Boston, Massachusetts, USA
| | - Yakeel T Quiroz
- Departments of Psychiatry and Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Eric Reiman
- Banner Alzheimer's Institute, Phoenix, Arizona, USA
- Banner Research, Phoenix, Arizona, USA
| | | | - Malú Gámez Tansey
- Department of Neuroscience and Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Julia Tcw
- Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Charlotte E Teunissen
- Neurochemistry Laboratory, Clinical Chemistry department, Amsterdam Neuroscience, Program Neurodegeneration, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Betty M Tijms
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
| | - Rik van der Kant
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research (CNCR), VU University Amsterdam, Amsterdam, The Netherlands
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Rebecca Wallings
- CTRND, Department of Neuroscience, University of Florida, Florida, USA
| | | | | | - Donna M Wilcock
- Sanders-Brown Center on Aging and Department of Physiology, University of Kentucky, Lexington, Kentucky, USA
| | - Tyler James Wishard
- Psychiatry & Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, California, USA
| | - Susan L Worley
- Independent science writer, Bryn Mawr, Pennsylvania, USA
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
| | | |
Collapse
|
40
|
Chen HJC, Mazzaferro S, Tian T, Mali I, Merkle FT. Differentiation, Transcriptomic Profiling, and Calcium Imaging of Human Hypothalamic Neurons. Curr Protoc 2023; 3:e786. [PMID: 37272700 PMCID: PMC7614736 DOI: 10.1002/cpz1.786] [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: 06/06/2023]
Abstract
Neurons in the hypothalamus orchestrate homeostatic physiological processes and behaviors essential for life. Human pluripotent stem cells (hPSCs) can be differentiated into many types of hypothalamic neurons, progenitors, and glia. This updated unit includes published studies and protocols with new advances in the differentiation, maturation, and interrogation by transcriptomic profiling and calcium imaging of human hypothalamic cell populations. Specifically, new methods to freeze and thaw hypothalamic progenitors after they have been patterned and before substantial neurogenesis has occurred are provided that will facilitate experimental flexibility and planning. Also included are updated recipes and protocols for neuronal maturation, with details on the equipment and methods for examining their transcriptomic response and cell-autonomous properties in culture in the presence of synaptic blockers. Together, these protocols facilitate the adoption and use of this model system for fundamental biological discovery and therapeutic translation to human diseases such as obesity, diabetes, sleep disorders, infertility, and chronic stress. © 2023 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: hPSC maintenance Basic Protocol 2: Hypothalamic neuron differentiation Support Protocol 1: Cortical neuron (control) differentiation Basic Protocol 3: Neuronal maturation Support Protocol 2: Cryopreservation and thawing of neuronal progenitors Support Protocol 3: Quality control: Confirmation of hypothalamic patterning and neurogenesis Support Protocol 4: Bulk RNA sequencing of hypothalamic cultures Basic Protocol 4: Calcium imaging of hypothalamic neurons using Fura-2 AM Alternate Protocol: Calcium imaging of green fluorescent hypothalamic neurons using Rhod-3 AM.
Collapse
Affiliation(s)
- Hsiao-Jou Cortina Chen
- Metabolic Research Laboratories, Wellcome Trust MRC Institute of Metabolic Science, University of Cambridge, Addenbrooke’s Hospital, Cambridge, UK
- Wellcome Trust–Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Simone Mazzaferro
- Metabolic Research Laboratories, Wellcome Trust MRC Institute of Metabolic Science, University of Cambridge, Addenbrooke’s Hospital, Cambridge, UK
- Wellcome Trust–Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Tian Tian
- Metabolic Research Laboratories, Wellcome Trust MRC Institute of Metabolic Science, University of Cambridge, Addenbrooke’s Hospital, Cambridge, UK
- Wellcome Trust–Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Iman Mali
- Metabolic Research Laboratories, Wellcome Trust MRC Institute of Metabolic Science, University of Cambridge, Addenbrooke’s Hospital, Cambridge, UK
| | - Florian T. Merkle
- Metabolic Research Laboratories, Wellcome Trust MRC Institute of Metabolic Science, University of Cambridge, Addenbrooke’s Hospital, Cambridge, UK
- Wellcome Trust–Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| |
Collapse
|
41
|
Gradisnik L, Velnar T. Astrocytes in the central nervous system and their functions in health and disease: A review. World J Clin Cases 2023; 11:3385-3394. [PMID: 37383914 PMCID: PMC10294192 DOI: 10.12998/wjcc.v11.i15.3385] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 02/19/2023] [Accepted: 04/14/2023] [Indexed: 05/25/2023] Open
Abstract
Astrocytes are key cells in the central nervous system. They are involved in many important functions under physiological and pathological conditions. As part of neuroglia, they have been recognised as cellular elements in their own right. The name astrocyte was first proposed by Mihaly von Lenhossek in 1895 because of the finely branched processes and star-like appearance of these particular cells. As early as the late 19th and early 20th centuries, Ramon y Cajal and Camillo Golgi had noted that although astrocytes have stellate features, their morphology is extremely diverse. Modern research has confirmed the morphological diversity of astrocytes both in vitro and in vivo and their complex, specific, and important roles in the central nervous system. In this review, the functions of astrocytes and their roles are described.
Collapse
Affiliation(s)
- Lidija Gradisnik
- Institute of Biomedical Sciences, Medical Faculty Maribor, Maribor 2000, Slovenia
| | - Tomaz Velnar
- Department of Neurosurgery, University Medical Centre Ljubljana, Ljubljana 1000, Slovenia
- AMEU ECM Maribor, Maribor 2000, Slovenia
| |
Collapse
|
42
|
Yi W, Yang D, Xu Z, Chen Z, Xiao G, Qin L. Immortalization of mouse primary astrocytes. Gene 2023; 865:147327. [PMID: 36870428 DOI: 10.1016/j.gene.2023.147327] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/08/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023]
Abstract
In cell culture studies, immortalized primary cells have become a useful tool to investigate the molecular and cellular functions of different types of cells. Several immortalization agents, such as human telomerase reverse transcriptase (hTERT) and Simian Virus 40 (SV40) T antigens, are commonly used for primary cell immortalization. Astrocytes, as the most abundant glial cell type in the central nervous system, are promising therapeutical targets for many neuronal disorders, such as Alzheimer's disease and Parkinson's disease. Immortalized primary astrocytes can provide useful information for astrocytes biology, astrocytes-neuron interactions, glial interactions and astrocytes-associated neuronal diseases. In this study, we successfully purified primary astrocytes with immuno-panning method and examined the astrocyte functions after immortalization through both hTERT and SV40 Large-T antigens. As expected, both immortalized astrocytes presented unlimited lifespan and highly expressed multiple astrocyte-specific markers. However, SV40 Large-T antigen, but not hTERT, immortalized astrocytes displayed fast ATP-induced calcium wave in culture. Hence, SV40 Large-T antigen could be a better choice for primary astrocyte immortalization, which closely mimics the cell biology of primary astrocytes in culture. In summary, the purification and immortalization of primary astrocytes presented in this study can be used for studying astrocyte biology under physiological and pathological conditions.
Collapse
Affiliation(s)
- Weihong Yi
- Department of Orthopedics, Huazhong University of Science and Technology Union Shenzhen Hospital, No. 89 Taoyuan Road, 518000 Shenzhen, China
| | - Dazhi Yang
- Department of Orthopedics, Huazhong University of Science and Technology Union Shenzhen Hospital, No. 89 Taoyuan Road, 518000 Shenzhen, China
| | - Zhen Xu
- Department of Orthopedics, Huazhong University of Science and Technology Union Shenzhen Hospital, No. 89 Taoyuan Road, 518000 Shenzhen, China
| | - Zecai Chen
- Department of Orthopedics, Huazhong University of Science and Technology Union Shenzhen Hospital, No. 89 Taoyuan Road, 518000 Shenzhen, China
| | - Guozhi Xiao
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, 518055 Shenzhen, China.
| | - Lei Qin
- Department of Orthopedics, Huazhong University of Science and Technology Union Shenzhen Hospital, No. 89 Taoyuan Road, 518000 Shenzhen, China.
| |
Collapse
|
43
|
Hasel P, Cooper ML, Marchildon AE, Rufen-Blanchette UA, Kim RD, Ma TC, Kang UJ, Chao MV, Liddelow SA. Defining the molecular identity and morphology of glia limitans superficialis astrocytes in mouse and human. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.06.535893. [PMID: 37066303 PMCID: PMC10104130 DOI: 10.1101/2023.04.06.535893] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Astrocytes are a highly abundant glial cell type that perform critical homeostatic functions in the central nervous system. Like neurons, astrocytes have many discrete heterogenous subtypes. The subtype identity and functions are, at least in part, associated with their anatomical location and can be highly restricted to strategically important anatomical domains. Here, we report that astrocytes forming the glia limitans superficialis, the outermost border of brain and spinal cord, are a highly specialized astrocyte subtype and can be identified by a single marker: Myocilin (Myoc). We show that Myoc+ astrocytes cover the entire brain and spinal cord surface, exhibit an atypical morphology, and are evolutionarily conserved from rodents to humans. Identification of this highly specialized astrocyte subtype will advance our understanding of CNS homeostasis and potentially be targeted for therapeutic intervention to combat peripheral inflammatory effects on the CNS.
Collapse
Affiliation(s)
- Philip Hasel
- Neuroscience Institute, NYU Grossman School of Medicine, New York, NY., USA
| | - Melissa L Cooper
- Neuroscience Institute, NYU Grossman School of Medicine, New York, NY., USA
| | - Anne E Marchildon
- Neuroscience Institute, NYU Grossman School of Medicine, New York, NY., USA
| | | | - Rachel D Kim
- Neuroscience Institute, NYU Grossman School of Medicine, New York, NY., USA
| | - Thong C Ma
- Fresco Institute for Parkinson’s and Movement Disorders, Department of Neurology, NYU Grossman School of Medicine, New York, NY., USA
- Parekh Center for Interdisciplinary Neurology, NYU Grossman School of Medicine, New York, NY., USA
- Department of Neuroscience and Physiology, NYU Grossman School of Medicine, New York, NY., USA
| | - Un Jung Kang
- Neuroscience Institute, NYU Grossman School of Medicine, New York, NY., USA
- Fresco Institute for Parkinson’s and Movement Disorders, Department of Neurology, NYU Grossman School of Medicine, New York, NY., USA
- Parekh Center for Interdisciplinary Neurology, NYU Grossman School of Medicine, New York, NY., USA
- Department of Neuroscience and Physiology, NYU Grossman School of Medicine, New York, NY., USA
| | - Moses V Chao
- Neuroscience Institute, NYU Grossman School of Medicine, New York, NY., USA
- Department of Neuroscience and Physiology, NYU Grossman School of Medicine, New York, NY., USA
- Department of Cell Biology, NYU Grossman School of Medicine, New York, NY., USA
- Department of Psychiatry, NYU Grossman School of Medicine, New York, NY., USA
| | - Shane A Liddelow
- Neuroscience Institute, NYU Grossman School of Medicine, New York, NY., USA
- Parekh Center for Interdisciplinary Neurology, NYU Grossman School of Medicine, New York, NY., USA
- Department of Neuroscience and Physiology, NYU Grossman School of Medicine, New York, NY., USA
- Department of Ophthalmology, NYU Grossman School of Medicine, New York, NY., USA
| |
Collapse
|
44
|
Dion-Albert L, Dudek KA, Russo SJ, Campbell M, Menard C. Neurovascular adaptations modulating cognition, mood, and stress responses. Trends Neurosci 2023; 46:276-292. [PMID: 36805768 DOI: 10.1016/j.tins.2023.01.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 01/11/2023] [Accepted: 01/25/2023] [Indexed: 02/19/2023]
Abstract
The neurovascular unit (NVU) is a dynamic center for substance exchange between the blood and the brain, making it an essential gatekeeper for central nervous system (CNS) homeostasis. Recent evidence supports a role for the NVU in modulating brain function and cognition. In addition, alterations in NVU processes are observed in response to stress, although the mechanisms via which they can affect mood and cognitive functions remain elusive. Here, we summarize recent studies of neurovascular regulation of emotional processes and cognitive function, including under stressful conditions. We also highlight relevant RNA-sequencing (RNA-seq) databases aiming to profile the NVU along with innovative tools to study and manipulate NVU function that can be exploited in the context of cognition and stress research throughout development, aging, or brain disorders.
Collapse
Affiliation(s)
- Laurence Dion-Albert
- Department of Psychiatry and Neuroscience, Faculty of Medicine and CERVO Brain Research Center, Université Laval, Quebec City, QC, Canada
| | - Katarzyna A Dudek
- Department of Psychiatry and Neuroscience, Faculty of Medicine and CERVO Brain Research Center, Université Laval, Quebec City, QC, Canada
| | - Scott J Russo
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai and Center for Affective Neuroscience, 1 Gustave L Levy Place, New York, NY, USA
| | - Matthew Campbell
- Smurfit Institute of Genetics, Trinity College Dublin, Lincoln Place Gate, Dublin 2, Ireland
| | - Caroline Menard
- Department of Psychiatry and Neuroscience, Faculty of Medicine and CERVO Brain Research Center, Université Laval, Quebec City, QC, Canada.
| |
Collapse
|
45
|
Hasel P, Aisenberg WH, Bennett FC, Liddelow SA. Molecular and metabolic heterogeneity of astrocytes and microglia. Cell Metab 2023; 35:555-570. [PMID: 36958329 DOI: 10.1016/j.cmet.2023.03.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 01/26/2023] [Accepted: 03/08/2023] [Indexed: 03/25/2023]
Abstract
Astrocytes and microglia are central players in a myriad of processes in the healthy and diseased brain, ranging from metabolism to immunity. The crosstalk between these two cell types contributes to pathology in many if not all neuroinflammatory and neurodegenerative diseases. Recent advancements in integrative multimodal sequencing techniques have begun to highlight how heterogeneous both cell types are and the importance of metabolism to their regulation. We discuss here the transcriptomic, metabolic, and functional heterogeneity of astrocytes and microglia and highlight their interaction in health and disease.
Collapse
Affiliation(s)
- Philip Hasel
- Neuroscience Institute, NYU Grossman School of Medicine, New York, NY 10016, USA.
| | - William H Aisenberg
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA.
| | - F Chris Bennett
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA.
| | - Shane A Liddelow
- Neuroscience Institute, NYU Grossman School of Medicine, New York, NY 10016, USA; Department of Neuroscience and Physiology, NYU Grossman School of Medicine, New York, NY 10016, USA; Department of Ophthalmology, NYU Grossman School of Medicine, New York, NY 10016, USA; Parekh Center for Interdisciplinary Neurology, NYU Grossman School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
46
|
Cellular Prion Protein Attenuates OGD/R-Induced Damage by Skewing Microglia toward an Anti-inflammatory State via Enhanced and Prolonged Activation of Autophagy. Mol Neurobiol 2023; 60:1297-1316. [PMID: 36441478 DOI: 10.1007/s12035-022-03099-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 10/21/2022] [Indexed: 11/29/2022]
Abstract
Modulation of microglial pro/anti-inflammatory states and autophagy are promising new therapies for ischemic stroke, but the underlying mechanisms remain largely unexplored. The objective of the study is to determine the intrinsic role of PrPC (cellular prion protein) in the regulation of microglial inflammatory states and autophagy in ischemic stroke. PrPC was expressed in murine microglia, and an in vitro oxygen-glucose deprivation/reperfusion (OGD/R) model was established in microglia of different PRNP genotypes. During reperfusion following OGD, wild-type (WT) microglia had significantly increased pro/anti-inflammatory microglial percentages and related cytokine [interleukin [IL]-6, IL-10, IL-4, tumor necrosis factor, and interferon-gamma] release at reperfusion after 48 or 72 h. WT microglia also showed greater accumulation of the autophagy markers LC3B-II/I (microtubule-associated protein B-light chain 3), but not of p62 or LAMP1 (lysosome-associated membrane protein) at reperfusion after 24 h and 48 h. Inhibition of autophagy using 3-methyladenine or bafilomycin A1 aggravated the OGD/R-induced pro-inflammatory state, and the effect of 3-methyladenine was significantly stronger than that of bafilomycin A1. Concomitantly, PRNP knockout shortened the accumulation of LC3B-II/I, suppressed microglial anti-inflammatory states, and further aggravated the pro-inflammatory states. Conversely, PRNP overexpression had the opposite effects. Bafilomycin A1 reversed the effect of PrPC on microglial inflammatory state transformation. Moreover, microglia with PRNP overexpression exhibited higher levels of LAMP1 expression in the control and OGD/R groups and delayed the OGD/R-induced decrease of LAMP1 to reperfusion after 48 h. PrPC attenuates OGD/R-induced damage by skewing microglia toward an anti-inflammatory state via enhanced and prolonged activation of autophagy.
Collapse
|
47
|
Wenzel TJ, Le J, He J, Alcorn J, Mousseau DD. Fundamental Neurochemistry Review: Incorporating a greater diversity of cell types, including microglia, in brain organoid cultures improves clinical translation. J Neurochem 2023; 164:560-582. [PMID: 36517959 DOI: 10.1111/jnc.15741] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 12/03/2022] [Accepted: 12/07/2022] [Indexed: 12/23/2022]
Abstract
Brain organoids have the potential to improve clinical translation, with the added benefit of reducing any extraneous use of experimental animals. As brain organoids are three-dimensional in vitro constructs that emulate the human brain, they bridge in vitro and in vivo studies more appropriately than monocultures. Although many factors contribute to the failure of extrapolating monoculture-based information to animal-based experiments and clinical trials, for the purpose of this review, we will focus on glia (non-neuronal brain cells), whose functions and transcriptome are particularly abnormal in monocultures. As discussed herein, glia require signals from-and contact with-other cell types to exist in their homeostatic state, which likely contributes to some of the differences between data derived from monocultures and data derived from brain organoids and even two-dimensional co-cultures. Furthermore, we highlight transcriptomic differences between humans and mice in regard to aging and Alzheimer's disease, emphasizing need for a model using the human genome-again, a benefit of brain organoids-to complement data derived from animals. We also identify an urgency for guidelines to improve the reporting and transparency of research using organoids. The lack of reporting standards creates challenges for the comparison and discussion of data from different articles. Importantly, brain organoids mark the first human model enabling the study of brain cytoarchitecture and development.
Collapse
Affiliation(s)
- Tyler J Wenzel
- Cell Signalling Laboratory, Department of Psychiatry, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada.,College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Jennifer Le
- Toxicology Centre, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Jim He
- Toxicology Centre, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Jane Alcorn
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Darrell D Mousseau
- Cell Signalling Laboratory, Department of Psychiatry, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| |
Collapse
|
48
|
Baudon A, Clauss Creusot E, Charlet A. [Emergent role of astrocytes in oxytocin-mediated modulatory control of neuronal circuits and brain functions]. Biol Aujourdhui 2023; 216:155-165. [PMID: 36744981 DOI: 10.1051/jbio/2022022] [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: 09/22/2022] [Indexed: 02/07/2023]
Abstract
The neuropeptide oxytocin has been in the focus of scientists for decades due to its profound and pleiotropic effects on physiology, activity of neuronal circuits and behaviors. Until recently, it was believed that oxytocinergic action exclusively occurs through direct activation of neuronal oxytocin receptors. However, several studies demonstrated the existence and functional relevance of astroglial oxytocin receptors in various brain regions in the mouse and rat brain. Astrocytic signaling and activity are critical for many important physiological processes including metabolism, neurotransmitter clearance from the synaptic cleft and integrated brain functions. While it can be speculated that oxytocinergic action on astrocytes predominantly facilitates neuromodulation via the release of gliotransmitters, the precise role of astrocytic oxytocin receptors remains elusive. In this review, we discuss the latest studies on the interaction between the oxytocinergic system and astrocytes, and give details of underlying intracellular cascades.
Collapse
Affiliation(s)
- Angel Baudon
- Centre National de la Recherche Scientifique et Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, 8 allée du Général Rouvillois, 67000 Strasbourg, France
| | - Etienne Clauss Creusot
- Centre National de la Recherche Scientifique et Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, 8 allée du Général Rouvillois, 67000 Strasbourg, France
| | - Alexandre Charlet
- Centre National de la Recherche Scientifique et Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, 8 allée du Général Rouvillois, 67000 Strasbourg, France
| |
Collapse
|
49
|
Preininger MK, Zaytseva D, Lin JM, Kaufer D. Blood-brain barrier dysfunction promotes astrocyte senescence through albumin-induced TGFβ signaling activation. Aging Cell 2023; 22:e13747. [PMID: 36606305 PMCID: PMC9924950 DOI: 10.1111/acel.13747] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 08/22/2022] [Accepted: 11/06/2022] [Indexed: 01/07/2023] Open
Abstract
Blood-brain barrier dysfunction (BBBD) and accumulation of senescent astrocytes occur during brain aging and contribute to neuroinflammation and disease. Here, we explored the relationship between these two age-related events, hypothesizing that chronic hippocampal exposure to the blood-borne protein serum albumin could induce stress-induced premature senescence (SIPS) in astrocytes via transforming growth factor beta 1 (TGFβ) signaling. We found that 1 week of albumin exposure significantly increased TGFβ signaling and senescence marker expression in cultured rat hippocampal astrocytes. These changes were preventable by pharmacological inhibition of the type I TGFβ receptor (TGFβR) ALK5. To study these effects in vivo, we utilized an animal model of BBBD in which albumin was continuously infused into the lateral ventricles of adult mice. Consistent with our in vitro results, 1 week of albumin infusion significantly increased TGFβ signaling activation and the burden of senescent astrocytes in hippocampal tissue. Pharmacological inhibition of ALK5 TGFβR or conditional genetic knockdown of astrocytic TGFβR prior to albumin infusion was sufficient to prevent albumin-induced astrocyte senescence. Together, these results establish a link between TGFβ signaling activation and astrocyte senescence and suggest that prolonged exposure to serum albumin due to BBBD can trigger these phenotypic changes.
Collapse
Affiliation(s)
- Marcela K. Preininger
- Department of Integrative BiologyUniversity of California, BerkeleyBerkeleyCaliforniaUSA
- Department of Molecular and Cell BiologyUniversity of California, BerkeleyBerkeleyCaliforniaUSA
| | - Dasha Zaytseva
- Department of Integrative BiologyUniversity of California, BerkeleyBerkeleyCaliforniaUSA
- Department of BiologySan Francisco State UniversitySan FranciscoCaliforniaUSA
| | - Jessica May Lin
- Department of Integrative BiologyUniversity of California, BerkeleyBerkeleyCaliforniaUSA
| | - Daniela Kaufer
- Department of Integrative BiologyUniversity of California, BerkeleyBerkeleyCaliforniaUSA
- Helen Wills Neuroscience InstituteUniversity of California, BerkeleyBerkeleyCaliforniaUSA
| |
Collapse
|
50
|
Freitag K, Eede P, Ivanov A, Sterczyk N, Schneeberger S, Borodina T, Sauer S, Beule D, Heppner FL. Diverse but unique astrocytic phenotypes during embryonic stem cell differentiation, culturing and development. Commun Biol 2023; 6:40. [PMID: 36639529 PMCID: PMC9839673 DOI: 10.1038/s42003-023-04410-3] [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: 02/15/2022] [Accepted: 01/03/2023] [Indexed: 01/14/2023] Open
Abstract
Astrocytes are resident glial cells of the central nervous system (CNS) that play complex and heterogeneous roles in brain development, homeostasis and disease. Since their vast involvement in health and disease is becoming increasingly recognized, suitable and reliable tools for studying these cells in vivo and in vitro are of utmost importance. One of the key challenges hereby is to adequately mimic their context-dependent in vivo phenotypes and functions in vitro. To better understand the spectrum of astrocytic variations in defined settings we performed a side-by-side-comparison of murine embryonic stem cell (ESC)-derived astrocytes as well as primary neonatal and adult astrocytes, revealing major differences on a functional and transcriptomic level, specifically on proliferation, migration, calcium signaling and cilium activity. Our results highlight the need to carefully consider the choice of astrocyte origin and phenotype with respect to age, isolation and culture protocols based on the respective biological question.
Collapse
Affiliation(s)
- Kiara Freitag
- grid.6363.00000 0001 2218 4662Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Neuropathology, Charitéplatz 1, 10117 Berlin, Germany ,grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE) within the Helmholtz Association, Berlin, Germany
| | - Pascale Eede
- grid.6363.00000 0001 2218 4662Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Neuropathology, Charitéplatz 1, 10117 Berlin, Germany ,Present Address: Apollo Health Ventures, Schlüterstr. 36, 10629 Berlin, Germany
| | - Andranik Ivanov
- grid.6363.00000 0001 2218 4662Core Unit Bioinformatics, Berlin Institute of Health, Charité - University Hospital Berlin, 10117 Berlin, Germany
| | - Nele Sterczyk
- grid.6363.00000 0001 2218 4662Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Neuropathology, Charitéplatz 1, 10117 Berlin, Germany
| | - Shirin Schneeberger
- grid.6363.00000 0001 2218 4662Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Neuropathology, Charitéplatz 1, 10117 Berlin, Germany ,grid.517316.7Cluster of Excellence, NeuroCure, Berlin, Germany ,Present Address: Apollo Health Ventures, Schlüterstr. 36, 10629 Berlin, Germany
| | - Tatiana Borodina
- grid.419491.00000 0001 1014 0849Scientific Genomics Platforms, Max Delbrück Center for Molecular Medicine (MDC) in the Helmholtz Society, Berlin, Germany and Berlin Institute of Health (BIH), Berlin, Germany
| | - Sascha Sauer
- grid.419491.00000 0001 1014 0849Scientific Genomics Platforms, Max Delbrück Center for Molecular Medicine (MDC) in the Helmholtz Society, Berlin, Germany and Berlin Institute of Health (BIH), Berlin, Germany
| | - Dieter Beule
- grid.6363.00000 0001 2218 4662Core Unit Bioinformatics, Berlin Institute of Health, Charité - University Hospital Berlin, 10117 Berlin, Germany
| | - Frank L. Heppner
- grid.6363.00000 0001 2218 4662Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Neuropathology, Charitéplatz 1, 10117 Berlin, Germany ,grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE) within the Helmholtz Association, Berlin, Germany ,grid.517316.7Cluster of Excellence, NeuroCure, Berlin, Germany
| |
Collapse
|