1
|
Yuasa-Kawada J, Kinoshita-Kawada M, Hiramoto M, Yamagishi S, Mishima T, Yasunaga S, Tsuboi Y, Hattori N, Wu JY. Neuronal guidance signaling in neurodegenerative diseases: Key regulators that function at neuron-glia and neuroimmune interfaces. Neural Regen Res 2026; 21:612-635. [PMID: 39995079 DOI: 10.4103/nrr.nrr-d-24-01330] [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: 10/31/2024] [Accepted: 01/27/2025] [Indexed: 02/26/2025] Open
Abstract
The nervous system processes a vast amount of information, performing computations that underlie perception, cognition, and behavior. During development, neuronal guidance genes, which encode extracellular cues, their receptors, and downstream signal transducers, organize neural wiring to generate the complex architecture of the nervous system. It is now evident that many of these neuroguidance cues and their receptors are active during development and are also expressed in the adult nervous system. This suggests that neuronal guidance pathways are critical not only for neural wiring but also for ongoing function and maintenance of the mature nervous system. Supporting this view, these pathways continue to regulate synaptic connectivity, plasticity, and remodeling, and overall brain homeostasis throughout adulthood. Genetic and transcriptomic analyses have further revealed many neuronal guidance genes to be associated with a wide range of neurodegenerative and neuropsychiatric disorders. Although the precise mechanisms by which aberrant neuronal guidance signaling drives the pathogenesis of these diseases remain to be clarified, emerging evidence points to several common themes, including dysfunction in neurons, microglia, astrocytes, and endothelial cells, along with dysregulation of neuron-microglia-astrocyte, neuroimmune, and neurovascular interactions. In this review, we explore recent advances in understanding the molecular and cellular mechanisms by which aberrant neuronal guidance signaling contributes to disease pathogenesis through altered cell-cell interactions. For instance, recent studies have unveiled two distinct semaphorin-plexin signaling pathways that affect microglial activation and neuroinflammation. We discuss the challenges ahead, along with the therapeutic potentials of targeting neuronal guidance pathways for treating neurodegenerative diseases. Particular focus is placed on how neuronal guidance mechanisms control neuron-glia and neuroimmune interactions and modulate microglial function under physiological and pathological conditions. Specifically, we examine the crosstalk between neuronal guidance signaling and TREM2, a master regulator of microglial function, in the context of pathogenic protein aggregates. It is well-established that age is a major risk factor for neurodegeneration. Future research should address how aging and neuronal guidance signaling interact to influence an individual's susceptibility to various late-onset neurological diseases and how the progression of these diseases could be therapeutically blocked by targeting neuronal guidance pathways.
Collapse
Affiliation(s)
| | | | | | - Satoru Yamagishi
- Department of Optical Neuroanatomy, Institute of Photonics Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Takayasu Mishima
- Division of Neurology, Department of Internal Medicine, Sakura Medical Center, Toho University, Sakura, Japan
| | - Shin'ichiro Yasunaga
- Department of Biochemistry, Fukuoka University Faculty of Medicine, Fukuoka, Japan
| | - Yoshio Tsuboi
- Department of Neurology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Jane Y Wu
- Department of Neurology, Center for Genetic Medicine, Lurie Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
2
|
Rees KA, McCamy KM, Danao CI, Winzer-Serhan UH. Augmented hippocampal up-regulation of immune modulators following a peripheral immune challenge in a hemizygous mouse model of the 15q13.3 microdeletion. Cytokine 2025; 191:156951. [PMID: 40300236 DOI: 10.1016/j.cyto.2025.156951] [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: 01/07/2025] [Revised: 04/04/2025] [Accepted: 04/22/2025] [Indexed: 05/01/2025]
Abstract
The strongest known genetic risk factor for generalized epilepsy is the human hemizygous 15q13.3 microdeletion (MD). This 1.5 Mb MD encompasses six genes, including CHRNA7 encoding the alpha7 subunit that forms the homo-pentameric nicotinic acetylcholine receptor, a known regulator of the immune system. In the CNS, hyper activation of neuroimmune responses contributes to increased seizure susceptibility. In a mouse model with a hemizygous deletion of the orthologous region (Df(h15q13)/+) (Het), we previously demonstrated increased hippocampal expression of inflammatory cytokines compared to wildtype (WT) mice following a mild peripheral immune challenge. To further characterize neuroimmune responses, hippocampal mRNA expression of the chemokines CXCL2 and CXCL10, and the Gap junction protein connexin 43 (GJA1), all of which are implicated in neuronal hyperexcitability, were determined along with additional immune related targets. Three hours after a lipopolysaccharide (LPS, 0.1 mg/kg) or polyinosinic:polycytidylic acid (Poly(I:C), 5 mg/kg) injection (i.p.), hippocampi were collected, mRNA extracted, and cDNA prepared for qPCR. The results demonstrate extensive upregulation of CXCL2 and CXCL10 expression by LPS and Poly(I:C) (up to 200-fold CXCL2, up to 600-fold CXCL10) (p < 0.0001) with genotype x treatment interactions for CXCL2 by LPS (p < 0.007). Responses to treatment were far smaller in magnitude for all other targets. LPS and Poly(I:C) induced statistically similar increases for Toll-like receptor (TLR)2, TLR4, HMGB1, and C3, but Poly(I:C) had stronger effects on GJA1, TLR3, C1qA and MARCO expression. Remarkably, TLR3 was the only target with significant downregulation of expression after Poly(I:C) (p < 0.0001). In addition, genotype x treatment interactions were detected for TLR3, TLR4, HMGB1, and C1qA (p < 0.02). Thus, a peripheral immune challenge caused extensive increases for CXCL2 and CXCL10, and the genotype-treatment interactions that was seen for several targets, underscored the augmented neuroinflammatory response in mice carrying the MD. Of note is the dramatic upregulation of CXCL10 by low dose Poly(I:C). CXCL10 causes hyperexcitability via neuronal CXCR3 activation. Thus, even an asymptomatic viral infection may increase seizure susceptibility. In summary, a peripheral immune challenge causes strong upregulation of hippocampal inflammatory mediators implicated in neuronal excitability which is particularly detrimental for individuals with high seizure susceptibility, such as carriers of the 15q13.3 MD.
Collapse
Affiliation(s)
- Katherine A Rees
- Department of Neuroscience & Experimental Therapeutics, College of Medicine, Texas A&M Health Science Center, Bryan, TX 77807, USA
| | - Kristin M McCamy
- Department of Neuroscience & Experimental Therapeutics, College of Medicine, Texas A&M Health Science Center, Bryan, TX 77807, USA
| | - Conner I Danao
- Department of Neuroscience & Experimental Therapeutics, College of Medicine, Texas A&M Health Science Center, Bryan, TX 77807, USA
| | - Ursula H Winzer-Serhan
- Department of Neuroscience & Experimental Therapeutics, College of Medicine, Texas A&M Health Science Center, Bryan, TX 77807, USA.
| |
Collapse
|
3
|
Brunswick CA, Carpenter CM, Dennis NA, Kwapis JL. Not the same as it ever was: A review of memory modification, updating, and distortion in humans and rodents. Neurosci Biobehav Rev 2025; 174:106195. [PMID: 40324709 DOI: 10.1016/j.neubiorev.2025.106195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 04/16/2025] [Accepted: 05/01/2025] [Indexed: 05/07/2025]
Abstract
Memory is a reconstructive and continuous process that enables existing information to be modified in response to a changing environment. Being able to dynamically update outdated memories is critical to an organism's survival. Memory modifications have been extensively studied in both rodents and humans, and prior work has revealed many regional, cellular, neurotransmitter, and subcellular molecular mechanisms underlying this process. However, these diverse bodies of literature have not yet been fully integrated into a comprehensive cross-species review. Integrating the finding across rodent and human work is important for furthering our understanding of memory modifications and the underlying neural mechanisms that support memory modification in both species. Here, we discuss advances in our understanding of adaptive and maladaptive memory modifications in terms of both underlying mechanisms (regional, cellular, and molecular) and behavioral outcomes. By emphasizing findings from both humans and rodents, the two major model systems in which memory modifications have been studied, we are able to highlight converging mechanisms and point to open questions in the field. Specifically, we discuss the major findings from several memory paradigms including declarative, aversive and procedural memory designs and highlight paradigms and models that have been readily translated between rodent and human models. Ultimately, this review identifies key parallels underlying memory updating across species, paradigms, tasks, and models.
Collapse
Affiliation(s)
- Chad A Brunswick
- Department of Biology, The Pennsylvania State University, 208 Life Sciences Building, 432 Science Drive, University Park, PA 16802, USA
| | - Catherine M Carpenter
- Department of Psychology, The Pennsylvania State University, 441 Moore Building, 138 Fischer Road, University Park, PA 16802, USA
| | - Nancy A Dennis
- Department of Psychology, The Pennsylvania State University, 441 Moore Building, 138 Fischer Road, University Park, PA 16802, USA
| | - Janine L Kwapis
- Department of Biology, The Pennsylvania State University, 208 Life Sciences Building, 432 Science Drive, University Park, PA 16802, USA.
| |
Collapse
|
4
|
Kolibius LD, Josselyn SA, Hanslmayr S. On the origin of memory neurons in the human hippocampus. Trends Cogn Sci 2025; 29:421-433. [PMID: 40037964 DOI: 10.1016/j.tics.2025.01.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 01/22/2025] [Accepted: 01/27/2025] [Indexed: 03/06/2025]
Abstract
The hippocampus is essential for episodic memory, yet its coding mechanism remains debated. In humans, two main theories have been proposed: one suggests that concept neurons represent specific elements of an episode, while another posits a conjunctive code, where index neurons code the entire episode. Here, we integrate new findings of index neurons in humans and other animals with the concept-specific memory framework, proposing that concept neurons evolve from index neurons through overlapping memories. This process is supported by engram literature, which posits that neurons are allocated to a memory trace based on excitability and that reactivation induces excitability. By integrating these insights, we connect two historically disparate fields of neuroscience: engram research and human single neuron episodic memory research.
Collapse
Affiliation(s)
- Luca D Kolibius
- Department of Biomedical Engineering, Columbia University, New York City, NY, USA.
| | - Sheena A Josselyn
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Physiology, University of Toronto, Toronto, Ontario, Canada; Department of Psychology, University of Toronto, Toronto, Ontario, Canada; Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Simon Hanslmayr
- School of Psychology and Neuroscience and Centre for Neurotechnology, University of Glasgow, Glasgow, UK; Centre for Neurotechnology, University of Glasgow, Glasgow, UK.
| |
Collapse
|
5
|
Chung EN, Lee J, Polonio CM, Choi J, Akl CF, Kilian M, Weiß WM, Gunner G, Ye M, Heo TH, Drake SS, Yang L, d'Eca CRGL, Lee JH, Deng L, Farrenkopf D, Schüle AM, Lee HG, Afolabi O, Ghaznavi S, Smirnakis SM, Chiu IM, Kuchroo VK, Quintana FJ, Wheeler MA. Psychedelic control of neuroimmune interactions governing fear. Nature 2025:10.1038/s41586-025-08880-9. [PMID: 40269152 DOI: 10.1038/s41586-025-08880-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 03/11/2025] [Indexed: 04/25/2025]
Abstract
Neuroimmune interactions-signals transmitted between immune and brain cells-regulate many aspects of tissue physiology1, including responses to psychological stress2-5, which can predispose individuals to develop neuropsychiatric diseases6-9. Still, the interactions between haematopoietic and brain-resident cells that influence complex behaviours are poorly understood. Here, we use a combination of genomic and behavioural screens to show that astrocytes in the amygdala limit stress-induced fear behaviour through epidermal growth factor receptor (EGFR). Mechanistically, EGFR expression in amygdala astrocytes inhibits a stress-induced, pro-inflammatory signal-transduction cascade that facilitates neuron-glial crosstalk and stress-induced fear behaviour through the orphan nuclear receptor NR2F2 in amygdala neurons. In turn, decreased EGFR signalling and fear behaviour are associated with the recruitment of meningeal monocytes during chronic stress. This set of neuroimmune interactions is therapeutically targetable through the administration of psychedelic compounds, which reversed the accumulation of monocytes in the brain meninges along with fear behaviour. Together with validation in clinical samples, these data suggest that psychedelics can be used to target neuroimmune interactions relevant to neuropsychiatric disorders and potentially other inflammatory diseases.
Collapse
Affiliation(s)
- Elizabeth N Chung
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Jinsu Lee
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Carolina M Polonio
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Joshua Choi
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Camilo Faust Akl
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Michael Kilian
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Wiebke M Weiß
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Georgia Gunner
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Mingyu Ye
- Department of Neurology, Brigham and Women's Hospital and Jamaica Plain Veterans Administration Hospital, Harvard Medical School, Boston, MA, USA
| | - Tae Hyun Heo
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Sienna S Drake
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Liu Yang
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Catarina R G L d'Eca
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Joon-Hyuk Lee
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Liwen Deng
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Daniel Farrenkopf
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Anton M Schüle
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Hong-Gyun Lee
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Oreoluwa Afolabi
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Sharmin Ghaznavi
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Center for the Neuroscience of Psychedelics, Massachusetts General Hospital, Boston, MA, USA
| | - Stelios M Smirnakis
- Department of Neurology, Brigham and Women's Hospital and Jamaica Plain Veterans Administration Hospital, Harvard Medical School, Boston, MA, USA
| | - Isaac M Chiu
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Vijay K Kuchroo
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Francisco J Quintana
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Michael A Wheeler
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA.
| |
Collapse
|
6
|
Liu C, Liu Z, Liu K, Zhang T, Wang G, Xie H, Guan JS. Hippocampus alters visual representation to encode new memory. Cell Rep 2025; 44:115594. [PMID: 40244845 DOI: 10.1016/j.celrep.2025.115594] [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: 10/06/2024] [Revised: 01/27/2025] [Accepted: 03/30/2025] [Indexed: 04/19/2025] Open
Abstract
Hippocampal-cortical interaction is crucial for episodic memory encoding and retaining, but when and how this interaction occurs remains elusive. Here, within the trace eyeblink conditioning paradigm, we found a neuronal ensemble in layer II of mouse visual cortex (VIS) that responded to the paired stimulus of light flash (cue, or conditioned stimulus [CS]) and air puff (unconditioned stimulus [US]), but not discrete stimuli, resembling an associative event during learning. This neuronal representation in VIS is dependent on the hippocampus and contributes to encoding the association. Optogenetic activation of hippocampus can promote the emerging representation to allow the association of separated cues. Mechanistically, fos+ engram cells, modulated by VIP+ neurons, are hubs of the association-activated ensemble in VIS. The hippocampus-modulated memory ensemble emerging in VIS at an early stage implicates the structural organization of the memory network to encode new memory.
Collapse
Affiliation(s)
- Chenhui Liu
- School of Life Science and Technology & State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China
| | - Zhen Liu
- School of Life Science and Technology & State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China
| | - Kaiyuan Liu
- Institute of Photonic Chips, School of Artificial Intelligence Science and Technology, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Tianfu Zhang
- School of Life Science and Technology & State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China
| | - Guangyu Wang
- School of Life Science and Technology & State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China
| | - Hong Xie
- Institute of Photonic Chips, School of Artificial Intelligence Science and Technology, University of Shanghai for Science and Technology, Shanghai 200093, China.
| | - Ji-Song Guan
- School of Life Science and Technology & State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China.
| |
Collapse
|
7
|
Kinsky NR, Orlin DJ, Ruesch EA, Kim B, Coello S, Diba K, Ramirez S. Erasable hippocampal neural signatures predict memory discrimination. Cell Rep 2025; 44:115391. [PMID: 40057951 DOI: 10.1016/j.celrep.2025.115391] [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: 10/27/2023] [Revised: 11/23/2024] [Accepted: 02/12/2025] [Indexed: 03/29/2025] Open
Abstract
Memories involving the hippocampus can take several days to consolidate, challenging efforts to uncover the neuronal signatures underlying this process. Here, we use calcium imaging in freely moving mice to track the hippocampal dynamics underlying memory consolidation across a 10-day contextual fear conditioning task. We find two neural signatures that emerge following learning and predict memory performance: context-specific place field remapping and coordinated neural activity prior to memory recall (freezing). To test whether these signatures support memory consolidation, we pharmacologically induced amnesia in separate mice by administering anisomycin, a protein synthesis inhibitor, immediately following learning. We find that anisomycin paradoxically accelerates cell turnover. Anisomycin also arrests learning-related remapping and blocks coordinated activity predictive of memory-related freezing behavior, effects that are likewise absent in untreated mice that exhibit poor memory expression. We conclude that context-specific place field remapping and the development of coordinated ensemble activity underlie contextual memory consolidation.
Collapse
Affiliation(s)
- Nathaniel R Kinsky
- Department of Anesthesiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| | - Daniel J Orlin
- Neuroscience Graduate Program, Oregon Health & Science University, Portland, OR 97239, USA
| | - Evan A Ruesch
- Brain and Cognitive Sciences Department, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Brian Kim
- Department of Anesthesiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI 48109, USA
| | - Siria Coello
- Center for Systems Neuroscience, Boston University, Boston, MA 02451, USA
| | - Kamran Diba
- Department of Anesthesiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI 48109, USA
| | - Steve Ramirez
- Center for Systems Neuroscience, Boston University, Boston, MA 02451, USA.
| |
Collapse
|
8
|
Eom K, Kim D, Hyun JH. Engram and behavior: How memory is stored in the brain. Neurobiol Learn Mem 2025; 219:108047. [PMID: 40074071 DOI: 10.1016/j.nlm.2025.108047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 03/04/2025] [Accepted: 03/09/2025] [Indexed: 03/14/2025]
Abstract
During the processing of information in humans, activated neurons behave in a specific way. The activity of these neurons leaves traces on the neurons, such as changes in synaptic or intrinsic properties. Formation of the memory traces is associated with molecular changes in the neurons. Hence, monitoring collective neural activities and following the trace of neural activities are important to neuroscience research. This collective or group of neurons is described as a 'neural ensemble', while the neural trace is described as a 'neural engram'. Both terms have been used and studied by neuroscientists for a long time. In this article, we discuss the development of these concepts, current research methods, and future areas of development.
Collapse
Affiliation(s)
- Kisang Eom
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea
| | - Donguk Kim
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea
| | - Jung Ho Hyun
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea; Biomedical Sciences & Engineering Major of Interdisciplinary Studies, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea; Center for Synapse Diversity and Specificity, DGIST, Daegu 42988, Republic of Korea.
| |
Collapse
|
9
|
Sehgal M, Filho DA, Kastellakis G, Kim S, Lee J, Shen Y, Huang S, Lavi A, Fernandes G, Davila Mejia I, Martin SS, Pekcan A, Wu MS, Heo WD, Poirazi P, Trachtenberg JT, Silva AJ. Compartmentalized dendritic plasticity in the mouse retrosplenial cortex links contextual memories formed close in time. Nat Neurosci 2025; 28:602-615. [PMID: 39962274 PMCID: PMC11893454 DOI: 10.1038/s41593-025-01876-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 12/20/2024] [Indexed: 03/12/2025]
Abstract
Events occurring close in time are often linked in memory, and recent studies suggest that such memories are encoded by overlapping neuronal ensembles. However, the role of dendritic plasticity mechanisms in linking memories is unknown. Here we show that memory linking is dependent not only on neuronal ensemble overlap in the mouse retrosplenial cortex, but also on branch-specific dendritic allocation mechanisms. The same dendritic segments are preferentially activated by two linked (but not independent) contextual memories, and spine clusters added after each of two linked (but not independent) contextual memories are allocated to the same dendritic segments. Importantly, we show that the reactivation of dendrites activated during the first context exploration is sufficient to link two contextual memories. Our results demonstrate a critical role for localized dendritic plasticity in memory integration and reveal rules governing how linked and independent memories are allocated to dendritic compartments.
Collapse
Affiliation(s)
- Megha Sehgal
- Departments of Neurobiology, Psychiatry and Psychology & Integrative Center for Learning and Memory, University of California, Los Angeles, Los Angeles, CA, USA.
- Department of Psychology, The Ohio State University, Columbus, OH, USA.
| | - Daniel Almeida Filho
- Departments of Neurobiology, Psychiatry and Psychology & Integrative Center for Learning and Memory, University of California, Los Angeles, Los Angeles, CA, USA
- SENAI Institute of Innovation in Advanced Health Systems, University Center SENAI CIMATEC, Salvador, Brazil
| | - George Kastellakis
- Institute of Molecular Biology and Biotechnology (IMBB), Foundation for Research and Technology, Hellas (FORTH), Heraklion, Greece
| | - Sungsoo Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Jinsu Lee
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Yang Shen
- Departments of Neurobiology, Psychiatry and Psychology & Integrative Center for Learning and Memory, University of California, Los Angeles, Los Angeles, CA, USA
| | - Shan Huang
- Departments of Neurobiology, Psychiatry and Psychology & Integrative Center for Learning and Memory, University of California, Los Angeles, Los Angeles, CA, USA
| | - Ayal Lavi
- Departments of Neurobiology, Psychiatry and Psychology & Integrative Center for Learning and Memory, University of California, Los Angeles, Los Angeles, CA, USA
| | - Giselle Fernandes
- Departments of Neurobiology, Psychiatry and Psychology & Integrative Center for Learning and Memory, University of California, Los Angeles, Los Angeles, CA, USA
| | - Irene Davila Mejia
- Departments of Neurobiology, Psychiatry and Psychology & Integrative Center for Learning and Memory, University of California, Los Angeles, Los Angeles, CA, USA
| | - Sunaina Soans Martin
- Departments of Neurobiology, Psychiatry and Psychology & Integrative Center for Learning and Memory, University of California, Los Angeles, Los Angeles, CA, USA
| | - Asli Pekcan
- Departments of Neurobiology, Psychiatry and Psychology & Integrative Center for Learning and Memory, University of California, Los Angeles, Los Angeles, CA, USA
| | - Melody Shana Wu
- Departments of Neurobiology, Psychiatry and Psychology & Integrative Center for Learning and Memory, University of California, Los Angeles, Los Angeles, CA, USA
| | - Won Do Heo
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Panayiota Poirazi
- Institute of Molecular Biology and Biotechnology (IMBB), Foundation for Research and Technology, Hellas (FORTH), Heraklion, Greece.
| | - Joshua T Trachtenberg
- Departments of Neurobiology, Psychiatry and Psychology & Integrative Center for Learning and Memory, University of California, Los Angeles, Los Angeles, CA, USA
| | - Alcino J Silva
- Departments of Neurobiology, Psychiatry and Psychology & Integrative Center for Learning and Memory, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
10
|
An Y, Yao Y, Liu H, Xi Y, Pi M, Xu R, Huang Y, Li S, Gu X. The role of the CCL5-CCR5 axis in microglial activation leading to postoperative cognitive dysfunction. Exp Neurol 2025; 385:115114. [PMID: 39667654 DOI: 10.1016/j.expneurol.2024.115114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 11/29/2024] [Accepted: 12/09/2024] [Indexed: 12/14/2024]
Abstract
Postoperative cognitive dysfunction (POCD) is a common complication following surgeries involving general anesthesia. Although the CCL5-CCR5 axis is implicated in various neurological conditions, its role in POCD remains unclear. In our POCD model, we observed an increase in CCL5 and CCR5 levels concurrent with microglial activation and significant upregulation of inflammatory cytokines IL-6 and IL-1β. Administration of MVC, a CCR5 antagonist, alleviated neuroinflammation, prevented dendritic spine loss, and improved cognitive deficits by inhibiting the CCR5/CREB/NLRP1 pathway. However, the cognitive benefits of MVC were reversed by the CREB inhibitor 666-15. Our findings highlight the potential of targeting the CCL5-CCR5 axis as a therapeutic strategy for preventing and treating POCD.
Collapse
Affiliation(s)
- Yuanyuan An
- Department of Anesthesiology, Nanjing Drum Tower Clinical College of Xuzhou Medical University, Nanjing, China; Department of Anesthesiology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Yu Yao
- Department of Anesthesiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Huan Liu
- Department of Anesthesiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yuqing Xi
- Department of Anesthesiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Mengying Pi
- Department of Anesthesiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Rui Xu
- Department of Anesthesiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yulin Huang
- Department of Anesthesiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Shuming Li
- Department of Anesthesiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| | - Xiaoping Gu
- Department of Anesthesiology, Nanjing Drum Tower Clinical College of Xuzhou Medical University, Nanjing, China; Department of Anesthesiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| |
Collapse
|
11
|
Gao Z, Gao Y, Li Y, Zhou J, Li G, Xie S, Jia R, Wang L, Jiang Z, Liang M, Du C, Chen Y, Liu Y, Du L, Wang C, Dou S, Lv Z, Wang L, Wang R, Shen B, Wang Z, Li Y, Han G. 5-HT 7R enhances neuroimmune resilience and alleviates meningitis by promoting CCR5 ubiquitination. J Adv Res 2025; 68:317-330. [PMID: 38432392 PMCID: PMC11785565 DOI: 10.1016/j.jare.2024.02.017] [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: 01/23/2024] [Revised: 02/22/2024] [Accepted: 02/22/2024] [Indexed: 03/05/2024] Open
Abstract
INTRODUCTION Excessive immune activation induces tissue damage during infection. Compared to external strategies to reconstruct immune homeostasis, host balancing ways remain largely unclear. OBJECTIVES Here we found a neuroimmune way that prevents infection-induced tissue damage. METHODS By FACS and histopathology analysis of brain Streptococcus pneumonia meningitis infection model and behavioral testing. Western blot, co-immunoprecipitation, and ubiquitination analyze the Fluoxetine initiate 5-HT7R-STUB1-CCR5 K48-linked ubiquitination degradation. RESULTS Fluoxetine, a selective serotonin reuptake inhibitor, or the agonist of serotonin receptor 5-HT7R, protects mice from meningitis by inhibiting CCR5-mediated excessive immune response and tissue damage. Mechanistically, the Fluoxetine-5-HT7R axis induces proteasome-dependent degradation of CCR5 via mTOR signaling, and then recruits STUB1, an E3 ubiquitin ligase, to initiate K48-linked polyubiquitination of CCR5 at K138 and K322, promotes its proteasomal degradation. STUB1 deficiency blocks 5-HT7R-mediated CCR5 degradation. CONCLUSION Our results reveal a neuroimmune pathway that balances anti-infection immunity via happiness neurotransmitter receptor and suggest the 5-HT7R-CCR5 axis as a potential target to promote neuroimmune resilience.
Collapse
Affiliation(s)
- Zhenfang Gao
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Yang Gao
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Yuxiang Li
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Jie Zhou
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Ge Li
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Shun Xie
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Ruiyan Jia
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Lanying Wang
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Ziying Jiang
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Meng Liang
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Chunxiao Du
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Yaqiong Chen
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Yinji Liu
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Lin Du
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Cong Wang
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Shuaijie Dou
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Zhonglin Lv
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Lubin Wang
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Renxi Wang
- Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Beifen Shen
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Zhiding Wang
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China.
| | - Yunfeng Li
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China.
| | - Gencheng Han
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China.
| |
Collapse
|
12
|
Kovacs M, Dominguez-Belloso A, Ali-Moussa S, Deczkowska A. Immune control of brain physiology. Nat Rev Immunol 2025:10.1038/s41577-025-01129-6. [PMID: 39890999 DOI: 10.1038/s41577-025-01129-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/02/2025] [Indexed: 02/03/2025]
Abstract
The peripheral immune system communicates with the brain through complex anatomical routes involving the skull, the brain borders, circumventricular organs and peripheral nerves. These immune-brain communication pathways were classically considered to be dormant under physiological conditions and active only in cases of infection or damage. Yet, peripheral immune cells and signals are key in brain development, function and maintenance. In this Perspective, we propose an alternative framework for understanding the mechanisms of immune-brain communication. During brain development and in homeostasis, these anatomical structures allow selected elements of the peripheral immune system to affect the brain directly or indirectly, within physiological limits. By contrast, in ageing and pathological settings, detrimental peripheral immune signals hijack the existing communication routes or alter their structure. We discuss why a diversity of communication channels is needed and how they work in relation to one another to maintain homeostasis of the brain.
Collapse
Affiliation(s)
- Mariángeles Kovacs
- Brain-Immune Communication Lab, Institut Pasteur, Université Paris Cité, Inserm U1224, Paris, France
| | - Amaia Dominguez-Belloso
- Brain-Immune Communication Lab, Institut Pasteur, Université Paris Cité, Inserm U1224, Paris, France
| | - Samir Ali-Moussa
- Brain-Immune Communication Lab, Institut Pasteur, Université Paris Cité, Inserm U1224, Paris, France
| | - Aleksandra Deczkowska
- Brain-Immune Communication Lab, Institut Pasteur, Université Paris Cité, Inserm U1224, Paris, France.
| |
Collapse
|
13
|
Xu X, Jané P, Taelman V, Jané E, Dumont RA, Garama Y, Kim F, Del Val Gómez M, Gariani K, Walter MA. The Theranostic Genome. Nat Commun 2024; 15:10904. [PMID: 39738156 PMCID: PMC11686231 DOI: 10.1038/s41467-024-55291-x] [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: 01/25/2024] [Accepted: 12/05/2024] [Indexed: 01/01/2025] Open
Abstract
Theranostic drugs represent an emerging path to deliver on the promise of precision medicine. However, bottlenecks remain in characterizing theranostic targets, identifying theranostic lead compounds, and tailoring theranostic drugs. To overcome these bottlenecks, we present the Theranostic Genome, the part of the human genome whose expression can be utilized to combine therapeutic and diagnostic applications. Using a deep learning-based hybrid human-AI pipeline that cross-references PubMed, the Gene Expression Omnibus, DisGeNET, The Cancer Genome Atlas and the NIH Molecular Imaging and Contrast Agent Database, we bridge individual genes in human cancers with respective theranostic compounds. Cross-referencing the Theranostic Genome with RNAseq data from over 17'000 human tissues identifies theranostic targets and lead compounds for various human cancers, and allows tailoring targeted theranostics to relevant cancer subpopulations. We expect the Theranostic Genome to facilitate the development of new targeted theranostics to better diagnose, understand, treat, and monitor a variety of human cancers.
Collapse
Affiliation(s)
- Xiaoying Xu
- University of Lucerne, Lucerne, LU, Switzerland
| | - Pablo Jané
- University of Geneva, Geneva, GE, Switzerland
- Nuclear Medicine and Molecular Imaging Division, Geneva University Hospitals, Geneva, GE, Switzerland
| | | | - Eduardo Jané
- Departamento de Matemática Aplicada a la Ingeniería Aeroespacial - ETSIAE, Universidad Politécnica de Madrid, 28040, Madrid, Spain
| | | | | | | | - María Del Val Gómez
- Servicio de Medicina Nuclear, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Karim Gariani
- Division of Endocrinology, Diabetes, Nutrition and Patient Therapeutic Education, Geneva University Hospitals, Geneva, GE, Switzerland
| | - Martin A Walter
- University of Lucerne, Lucerne, LU, Switzerland.
- St. Anna Hospital, University of Lucerne, Lucerne, LU, Switzerland.
| |
Collapse
|
14
|
Zheng Z, Liu Y, Mu R, Guo X, Feng Y, Guo C, Yang L, Qiu W, Zhang Q, Yang W, Dong Z, Qiu S, Dong Y, Cui Y. A small population of stress-responsive neurons in the hypothalamus-habenula circuit mediates development of depression-like behavior in mice. Neuron 2024; 112:3924-3939.e5. [PMID: 39389052 DOI: 10.1016/j.neuron.2024.09.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 06/25/2024] [Accepted: 09/13/2024] [Indexed: 10/12/2024]
Abstract
Accumulating evidence has shown that various brain functions are associated with experience-activated neuronal ensembles. However, whether such neuronal ensembles are engaged in the pathogenesis of stress-induced depression remains elusive. Utilizing activity-dependent viral strategies in mice, we identified a small population of stress-responsive neurons, primarily located in the middle part of the lateral hypothalamus (mLH) and the medial part of the lateral habenula (LHbM). These neurons serve as "starter cells" to transmit stress-related information and mediate the development of depression-like behaviors during chronic stress. Starter cells in the mLH and LHbM form dominant connections, which are selectively potentiated by chronic stress. Silencing these connections during chronic stress prevents the development of depression-like behaviors, whereas activating these connections directly elicits depression-like behaviors without stress experience. Collectively, our findings dissect a core functional unit within the LH-LHb circuit that mediates the development of depression-like behaviors in mice.
Collapse
Affiliation(s)
- Zhiwei Zheng
- Department of Psychiatry of Sir Run Shaw Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Department of Neurology and International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu 322000, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou 310058, China
| | - Yiqin Liu
- Department of Psychiatry of Sir Run Shaw Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Department of Neurology and International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu 322000, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou 310058, China
| | - Ruiqi Mu
- Department of Psychiatry of Sir Run Shaw Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou 310058, China
| | - Xiaonan Guo
- Department of Psychiatry of Sir Run Shaw Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou 310058, China
| | - Yirong Feng
- Department of Psychiatry of Sir Run Shaw Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou 310058, China
| | - Chen Guo
- Department of Psychiatry of Sir Run Shaw Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou 310058, China
| | - Liang Yang
- Department of Psychiatry of Sir Run Shaw Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou 310058, China
| | - Wenxi Qiu
- Department of Psychiatry of Sir Run Shaw Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou 310058, China
| | - Qi Zhang
- Shanghai Key Laboratory of Brain Functional Genomics (Ministry of Education), School of Psychology and Cognitive Science, East China Normal University, Shanghai 200062, China
| | - Wei Yang
- Department of Biophysics and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Zhaoqi Dong
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Shuang Qiu
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou 310058, China
| | - Yiyan Dong
- Department of Psychiatry of Sir Run Shaw Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Department of Neurology and International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu 322000, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou 310058, China.
| | - Yihui Cui
- Department of Psychiatry of Sir Run Shaw Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Department of Neurology and International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu 322000, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
15
|
Zaki Y, Cai DJ. Memory engram stability and flexibility. Neuropsychopharmacology 2024; 50:285-293. [PMID: 39300271 PMCID: PMC11525749 DOI: 10.1038/s41386-024-01979-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 08/11/2024] [Accepted: 08/12/2024] [Indexed: 09/22/2024]
Abstract
Many studies have shown that memories are encoded in sparse neural ensembles distributed across the brain. During the post-encoding period, often during sleep, many of the cells that were active during encoding are reactivated, supporting consolidation of this memory. During memory recall, many of the same cells that were active during encoding and reactivated during consolidation are reactivated during recall. These ensembles of cells have been referred to as the memory engram cells, stably representing a specific memory. However, recent studies question the rigidity of the "stable memory engram." Here we review the past literature of how episodic-like memories are encoded, consolidated, and recalled. We also highlight more recent studies (as well as some older literature) that suggest that these stable memories and their representations are much more dynamic and flexible than previously thought. We highlight some of these processes, including memory updating, reconsolidation, forgetting, schema learning, memory-linking, and representational drift.
Collapse
Affiliation(s)
- Yosif Zaki
- Nash Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Denise J Cai
- Nash Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
16
|
Becher B, Derfuss T, Liblau R. Targeting cytokine networks in neuroinflammatory diseases. Nat Rev Drug Discov 2024; 23:862-879. [PMID: 39261632 DOI: 10.1038/s41573-024-01026-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/02/2024] [Indexed: 09/13/2024]
Abstract
In neuroinflammatory diseases, systemic (blood-borne) leukocytes invade the central nervous system (CNS) and lead to tissue damage. A causal relationship between neuroinflammatory diseases and dysregulated cytokine networks is well established across several preclinical models. Cytokine dysregulation is also observed as an inadvertent effect of cancer immunotherapy, where it often leads to neuroinflammation. Neuroinflammatory diseases can be separated into those in which a pathogen is at the centre of the immune response and those of largely unknown aetiology. Here, we discuss the pathophysiology, cytokine networks and therapeutic landscape of 'sterile' neuroinflammatory diseases such as multiple sclerosis (MS), neuromyelitis optica spectrum disorder (NMOSD), neurosarcoidosis and immune effector cell-associated neurotoxicity syndrome (ICANS) triggered by cancer immunotherapy. Despite successes in targeting cytokine networks in preclinical models of neuroinflammation, the clinical translation of targeting cytokines and their receptors has shown mixed and often paradoxical responses.
Collapse
Affiliation(s)
- Burkhard Becher
- Institute of experimental Immunology, University of Zurich, Zurich, Switzerland.
| | - Tobias Derfuss
- Department of Neurology and Biomedicine, Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel, University of Basel, Basel, Switzerland.
| | - Roland Liblau
- Institute for inflammatory and infectious diseases, INSERM UMR1291 - CNRS UMR505, Toulouse, France.
| |
Collapse
|
17
|
Miao J, Zhang B, Sun H, Zhang P, Shen H, Wang J, Jia J, Zhang K, Zheng Z, Zhu P. CCR5 mediates rheumatoid arthritis progression by promoting the activation and proliferation of non-classical Th1 cells. Int J Rheum Dis 2024; 27:e15370. [PMID: 39558608 DOI: 10.1111/1756-185x.15370] [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: 08/01/2024] [Revised: 09/12/2024] [Accepted: 09/25/2024] [Indexed: 11/20/2024]
Abstract
AIM Rheumatoid arthritis (RA) is a prevalent autoimmune disease characterized by immune dysegulation, including an immune imbalance due to abnormal activation of non-classical Th1 cells (CD161+ Th1). This study investigated the effects of CCR5 on the activation and proliferation of CD161+ Th1 and their pathogenicity in patients with RA. METHODS The study was conducted on 53 patients with RA and 32 age- and sex-matched healthy controls (HC). The cell phenotype was assessed by flow cytometry and the cytokine levels in the supernatant were detected by ELISA. RESULTS We demonstrate a marked increase in CD161+ Th1 cells in the synovial fluid of RA patients. These cells exhibit a hyperactivated and hyperproliferative state alongside elevated CCR5 expression. Furthermore, the levels of CD161+ Th1 cells, CD25, and CCR5 in RA synovial fluid show a positive correlation with the disease activity. Additionally, our study reveals that CCR5 facilitates the activation, proliferation, and cytokine production of CD161+ Th1 cells through the pZAP70/NFAT signaling pathway. CONCLUSION These findings contribute to a deeper understanding of RA pathogenesis and uncover a novel mechanism that regulates non-classical CD161+ Th1 responses in RA, which may provide a potential therapeutic target.
Collapse
Affiliation(s)
- Jinlin Miao
- Department of Clinical Immunology, Xijing Hospital, and National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Bei Zhang
- Department of Clinical Immunology, Xijing Hospital, and National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Haoyang Sun
- Department of Clinical Immunology, Xijing Hospital, and National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Peiyan Zhang
- Department of Clinical Immunology, Xijing Hospital, and National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Haomiao Shen
- Department of Clinical Immunology, Xijing Hospital, and National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Jiawei Wang
- Department of Clinical Immunology, Xijing Hospital, and National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Junfeng Jia
- Department of Clinical Immunology, Xijing Hospital, and National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Kui Zhang
- Department of Clinical Immunology, Xijing Hospital, and National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Zhaohui Zheng
- Department of Clinical Immunology, Xijing Hospital, and National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Ping Zhu
- Department of Clinical Immunology, Xijing Hospital, and National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
| |
Collapse
|
18
|
Lian N, Li F, Zhou C, Yin Y, Kang Y, Luo K, Lui S, Li T, Lu P. Protein phosphatase 2Cm-regulated branched-chain amino acid catabolic defect in dorsal root ganglion neurons drives pain sensitization. Acta Neuropathol Commun 2024; 12:147. [PMID: 39256776 PMCID: PMC11385486 DOI: 10.1186/s40478-024-01856-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 08/31/2024] [Indexed: 09/12/2024] Open
Abstract
Maladaptive changes of metabolic patterns in the lumbar dorsal root ganglion (DRG) are critical for nociceptive hypersensitivity genesis. The accumulation of branched-chain amino acids (BCAAs) in DRG has been implicated in mechanical allodynia and thermal hyperalgesia, but the exact mechanism is not fully understood. This study aimed to explore how BCAA catabolism in DRG modulates pain sensitization. Wildtype male mice were fed a high-fat diet (HFD) for 8 weeks. Adult PP2Cmfl/fl mice of both sexes were intrathecally injected with pAAV9-hSyn-Cre to delete the mitochondrial targeted 2 C-type serine/threonine protein phosphatase (PP2Cm) in DRG neurons. Here, we reported that BCAA catabolism was impaired in the lumbar 4-5 (L4-L5) DRGs of mice fed a high-fat diet (HFD). Conditional deletion of PP2Cm in DRG neurons led to mechanical allodynia, heat and cold hyperalgesia. Mechanistically, the genetic knockout of PP2Cm resulted in the upregulation of C-C chemokine ligand 5/C-C chemokine receptor 5 (CCL5/CCR5) axis and an increase in transient receptor potential ankyrin 1 (TRPA1) expression. Blocking the CCL5/CCR5 signaling or TRPA1 alleviated pain behaviors induced by PP2Cm deletion. Thus, targeting BCAA catabolism in DRG neurons may be a potential management strategy for pain sensitization.
Collapse
Affiliation(s)
- Nan Lian
- Laboratory of Mitochondria and Metabolism, West China Hospital of Sichuan University, Chengdu, 610041, China
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Fangzhou Li
- Department of Anesthesiology, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, 610041, China
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Cheng Zhou
- Department of Anesthesiology, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Yan Yin
- Department of Pain Management, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Yi Kang
- Department of Anesthesiology, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Kaiteng Luo
- Laboratory of Mitochondria and Metabolism, West China Hospital of Sichuan University, Chengdu, 610041, China
- Department of Anesthesiology, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, 610041, China
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Su Lui
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Tao Li
- Laboratory of Mitochondria and Metabolism, West China Hospital of Sichuan University, Chengdu, 610041, China.
- Department of Anesthesiology, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, 610041, China.
- West China Hospital, Sichuan University, No 37 Wainan Guoxue Road, Chengdu, 610041, Sichuan, China.
| | - Peilin Lu
- Laboratory of Mitochondria and Metabolism, West China Hospital of Sichuan University, Chengdu, 610041, China.
- Department of Anesthesiology, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, 610041, China.
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, 610041, China.
- West China Hospital, Sichuan University, No 37 Wainan Guoxue Road, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
19
|
Vicidomini C, Goode TD, McAvoy KM, Yu R, Beveridge CH, Iyer SN, Victor MB, Leary N, Evans L, Steinbaugh MJ, Lai ZW, Lyon MC, Silvestre MRFS, Bonilla G, Sadreyev RI, Walther TC, Sui SH, Saido T, Yamamoto K, Murakami M, Tsai LH, Chopra G, Sahay A. An aging-sensitive compensatory secretory phospholipase that confers neuroprotection and cognitive resilience. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.26.605338. [PMID: 39211220 PMCID: PMC11361190 DOI: 10.1101/2024.07.26.605338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Breakdown of lipid homeostasis is thought to contribute to pathological aging, the largest risk factor for neurodegenerative disorders such as Alzheimer's Disease (AD). Cognitive reserve theory posits a role for compensatory mechanisms in the aging brain in preserving neuronal circuit functions, staving off cognitive decline, and mitigating risk for AD. However, the identities of such mechanisms have remained elusive. A screen for hippocampal dentate granule cell (DGC) synapse loss-induced factors identified a secreted phospholipase, Pla2g2f, whose expression increases in DGCs during aging. Pla2g2f deletion in DGCs exacerbates aging-associated pathophysiological changes including synapse loss, inflammatory microglia, reactive astrogliosis, impaired neurogenesis, lipid dysregulation and hippocampal-dependent memory loss. Conversely, boosting Pla2g2f in DGCs during aging is sufficient to preserve synapses, reduce inflammatory microglia and reactive gliosis, prevent hippocampal-dependent memory impairment and modify trajectory of cognitive decline. Ex vivo, neuronal-PLA2G2F mediates intercellular signaling to decrease lipid droplet burden in microglia. Boosting Pla2g2f expression in DGCs of an aging-sensitive AD model reduces amyloid load and improves memory. Our findings implicate PLA2G2F as a compensatory neuroprotective factor that maintains lipid homeostasis to counteract aging-associated cognitive decline.
Collapse
Affiliation(s)
- Cinzia Vicidomini
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- BROAD Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Travis D Goode
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- BROAD Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Kathleen M McAvoy
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- BROAD Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Ruilin Yu
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Conor H Beveridge
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Sanjay N Iyer
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Matheus B Victor
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Noelle Leary
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Liam Evans
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- BROAD Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Michael J Steinbaugh
- Harvard Chan Bioinformatics Core, Harvard School of Public Health, Harvard University, Boston, Massachusetts, USA
| | - Zon Weng Lai
- Harvard Chan Advanced Multi-omics Platform, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA
| | - Marina C Lyon
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- BROAD Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Manuel Rico F S Silvestre
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- BROAD Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Gracia Bonilla
- Department of Molecular Biology. Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ruslan I Sadreyev
- Department of Molecular Biology. Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Tobias C Walther
- Harvard Chan Advanced Multi-omics Platform, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA
- Howard Hughes Medical Institute, Boston, Massachusetts, USA
| | - Shannan Ho Sui
- Harvard Chan Bioinformatics Core, Harvard School of Public Health, Harvard University, Boston, Massachusetts, USA
| | - Takaomi Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Saitama 351-0198 Japan
| | - Kei Yamamoto
- Graduate School of Technology, Industrial and Social Sciences, Tokushima University, 2-1 Minami-jyosanjima, Tokushima 770-8513, Japan
| | - Makoto Murakami
- Laboratory of Microenvironmental and Metabolic Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Li-Huei Tsai
- BROAD Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Gaurav Chopra
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47907, USA
- Purdue Institute for Drug Discovery, Purdue University, West Lafayette, IN 47907, USA
- Purdue Center for Cancer Research, Purdue University, West Lafayette, IN 47907, USA
- Regenstrief Center for Healthcare Engineering, Purdue University, West Lafayette, IN 47907, USA
| | - Amar Sahay
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- BROAD Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| |
Collapse
|
20
|
Joy MT, Carmichael ST. Activity-dependent transcriptional programs in memory regulate motor recovery after stroke. Commun Biol 2024; 7:1048. [PMID: 39183218 PMCID: PMC11345429 DOI: 10.1038/s42003-024-06723-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 08/12/2024] [Indexed: 08/27/2024] Open
Abstract
Stroke causes death of brain tissue leading to long-term deficits. Behavioral evidence from neurorehabilitative therapies suggest learning-induced neuroplasticity can lead to beneficial outcomes. However, molecular and cellular mechanisms that link learning and stroke recovery are unknown. We show that in a mouse model of stroke, which exhibits enhanced recovery of function due to genetic perturbations of learning and memory genes, animals display activity-dependent transcriptional programs that are normally active during formation or storage of new memories. The expression of neuronal activity-dependent genes are predictive of recovery and occupy a molecular latent space unique to motor recovery. With motor recovery, networks of activity-dependent genes are co-expressed with their transcription factor targets forming gene regulatory networks that support activity-dependent transcription, that are normally diminished after stroke. Neuronal activity-dependent changes at the circuit level are influenced by interactions with microglia. At the molecular level, we show that enrichment of activity-dependent programs in neurons lead to transcriptional changes in microglia where they differentially interact to support intercellular signaling pathways for axon guidance, growth and synaptogenesis. Together, these studies identify activity-dependent transcriptional programs as a fundamental mechanism for neural repair post-stroke.
Collapse
Affiliation(s)
- Mary T Joy
- The Jackson Laboratory, Bar Harbor, ME, 04609, USA.
| | - S Thomas Carmichael
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA
| |
Collapse
|
21
|
Suzuki H, Tanaka T, Li G, Ouchida T, Kaneko MK, Kato Y. Development of a Sensitive Anti-Mouse CCR5 Monoclonal Antibody for Flow Cytometry. Monoclon Antib Immunodiagn Immunother 2024; 43:96-100. [PMID: 38836505 DOI: 10.1089/mab.2024.0004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2024] Open
Abstract
C-C chemokine receptor 5 (CCR5), a member of the G protein-coupled receptor family, is the most common coreceptor for the human immunodeficiency virus type 1. CCR5 is also involved in the pathogenesis of tumors and inflammatory diseases. The CCR5 antagonists including monoclonal antibodies (mAbs) have been developed and evaluated in clinical trials. In this study, we developed novel mAbs for mouse CCR5 (mCCR5) using the Cell-Based Immunization and Screening (CBIS) method. One of the established anti-mCCR5 mAbs, C5Mab-2 (rat IgG2b, kappa), reacted with mCCR5-overexpressed Chinese hamster ovary-K1 (CHO/mCCR5) and an endogenously mCCR5-expressing cell line (L1210) by flow cytometry. Using flow cytometry, the dissociation constant (KD) of C5Mab-2 for CHO/mCCR5 was determined as 4.3 × 10-8 M. These results indicated that C5Mab-2 is useful for the detection of mCCR5 in flow cytometry and may be applicable to obtain the proof of concept in preclinical studies.
Collapse
Affiliation(s)
- Hiroyuki Suzuki
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tomohiro Tanaka
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Guanjie Li
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tsunenori Ouchida
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Mika K Kaneko
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yukinari Kato
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
22
|
Ubukata R, Suzuki H, Tanaka T, Li G, Kaneko MK, Kato Y. Development of Sensitive Anti-Mouse CCR5 Monoclonal Antibodies Using the N-Terminal Peptide Immunization. Monoclon Antib Immunodiagn Immunother 2024; 43:112-118. [PMID: 38868984 DOI: 10.1089/mab.2024.0009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2024] Open
Abstract
One of the G protein-coupled receptors, C-C chemokine receptor 5 (CCR5), is an important regulator for the activation of T and B lymphocytes, dendritic cells, natural killer cells, and macrophages. Upon binding to its ligands, CCR5 activates downstream signaling, which is an important regulator in the innate and adaptive immune response through the promotion of lymphocyte migration and the secretion of proinflammatory cytokines. Anti-CCR5 monoclonal antibodies (mAbs) have been developed and evaluated in clinical trials for tumors and inflammatory diseases. In this study, we developed novel mAbs for mouse CCR5 (mCCR5) using the N-terminal peptide immunization. Among the established anti-mCCR5 mAbs, C5Mab-4 (rat IgG2a, kappa) and C5Mab-8 (rat IgG1, kappa), recognized mCCR5-overexpressing Chinese hamster ovary-K1 (CHO/mCCR5) and an endogenously mCCR5-expressing cell line (L1210) by flow cytometry. The dissociation constant (KD) values of C5Mab-4 and C5Mab-8 for CHO/mCCR5 were determined as 3.5 × 10-8 M and 7.3 × 10-9 M, respectively. Furthermore, both C5Mab-4 and C5Mab-8 could detect mCCR5 by western blotting. These results indicated that C5Mab-4 and C5Mab-8 are useful for detecting mCCR5 by flow cytometry and western blotting and provide a possibility to obtain the proof of concept in preclinical studies.
Collapse
Affiliation(s)
| | | | - Tomohiro Tanaka
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Guanjie Li
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Mika K Kaneko
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yukinari Kato
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
23
|
Guo X, Hong P, Xiong S, Yan Y, Xie H, Guan JS. Kdm4a is an activity downregulated barrier to generate engrams for memory separation. Nat Commun 2024; 15:5887. [PMID: 39003305 PMCID: PMC11246488 DOI: 10.1038/s41467-024-50218-y] [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: 12/20/2023] [Accepted: 07/01/2024] [Indexed: 07/15/2024] Open
Abstract
Memory engrams are a subset of learning activated neurons critical for memory recall, consolidation, extinction and separation. While the transcriptional profile of engrams after learning suggests profound neural changes underlying plasticity and memory formation, little is known about how memory engrams are selected and allocated. As epigenetic factors suppress memory formation, we developed a CRISPR screening in the hippocampus to search for factors controlling engram formation. We identified histone lysine-specific demethylase 4a (Kdm4a) as a negative regulator for engram formation. Kdm4a is downregulated after neural activation and controls the volume of mossy fiber boutons. Mechanistically, Kdm4a anchors to the exonic region of Trpm7 gene loci, causing the stalling of nascent RNAs and allowing burst transcription of Trpm7 upon the dismissal of Kdm4a. Furthermore, the YTH domain containing protein 2 (Ythdc2) recruits Kdm4a to the Trpm7 gene and stabilizes nascent RNAs. Reducing the expression of Kdm4a in the hippocampus via genetic manipulation or artificial neural activation facilitated the ability of pattern separation in rodents. Our work indicates that Kdm4a is a negative regulator of engram formation and suggests a priming state to generate a separate memory.
Collapse
Affiliation(s)
- Xiuxian Guo
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Pengfei Hong
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Songhai Xiong
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Yuze Yan
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Hong Xie
- Institute of Photonic Chips, School of Artificial Intelligence Science and Technology, University of Shanghai for Science and Technology, Shanghai, China.
| | - Ji-Song Guan
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
- State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai, China.
| |
Collapse
|
24
|
Liu Y, Ye S, Li XN, Li WG. Memory Trace for Fear Extinction: Fragile yet Reinforceable. Neurosci Bull 2024; 40:777-794. [PMID: 37812300 PMCID: PMC11178705 DOI: 10.1007/s12264-023-01129-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 06/08/2023] [Indexed: 10/10/2023] Open
Abstract
Fear extinction is a biological process in which learned fear behavior diminishes without anticipated reinforcement, allowing the organism to re-adapt to ever-changing situations. Based on the behavioral hypothesis that extinction is new learning and forms an extinction memory, this new memory is more readily forgettable than the original fear memory. The brain's cellular and synaptic traces underpinning this inherently fragile yet reinforceable extinction memory remain unclear. Intriguing questions are about the whereabouts of the engram neurons that emerged during extinction learning and how they constitute a dynamically evolving functional construct that works in concert to store and express the extinction memory. In this review, we discuss recent advances in the engram circuits and their neural connectivity plasticity for fear extinction, aiming to establish a conceptual framework for understanding the dynamic competition between fear and extinction memories in adaptive control of conditioned fear responses.
Collapse
Affiliation(s)
- Ying Liu
- Department of Rehabilitation Medicine, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Huashan Hospital, Institute for Translational Brain Research, Fudan University, Shanghai, 200032, China
| | - Shuai Ye
- Department of Rehabilitation Medicine, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Huashan Hospital, Institute for Translational Brain Research, Fudan University, Shanghai, 200032, China
| | - Xin-Ni Li
- Department of Rehabilitation Medicine, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Huashan Hospital, Institute for Translational Brain Research, Fudan University, Shanghai, 200032, China
| | - Wei-Guang Li
- Department of Rehabilitation Medicine, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Huashan Hospital, Institute for Translational Brain Research, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
25
|
Choucry A, Nomoto M, Inokuchi K. Engram mechanisms of memory linking and identity. Nat Rev Neurosci 2024; 25:375-392. [PMID: 38664582 DOI: 10.1038/s41583-024-00814-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/25/2024] [Indexed: 05/25/2024]
Abstract
Memories are thought to be stored in neuronal ensembles referred to as engrams. Studies have suggested that when two memories occur in quick succession, a proportion of their engrams overlap and the memories become linked (in a process known as prospective linking) while maintaining their individual identities. In this Review, we summarize the key principles of memory linking through engram overlap, as revealed by experimental and modelling studies. We describe evidence of the involvement of synaptic memory substrates, spine clustering and non-linear neuronal capacities in prospective linking, and suggest a dynamic somato-synaptic model, in which memories are shared between neurons yet remain separable through distinct dendritic and synaptic allocation patterns. We also bring into focus retrospective linking, in which memories become associated after encoding via offline reactivation, and discuss key temporal and mechanistic differences between prospective and retrospective linking, as well as the potential differences in their cognitive outcomes.
Collapse
Affiliation(s)
- Ali Choucry
- Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
- Research Center for Idling Brain Science, University of Toyama, Toyama, Japan
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Masanori Nomoto
- Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
- Research Center for Idling Brain Science, University of Toyama, Toyama, Japan
- CREST, Japan Science and Technology Agency (JST), University of Toyama, Toyama, Japan
- Japan Agency for Medical Research and Development (AMED), Tokyo, Japan
| | - Kaoru Inokuchi
- Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan.
- Research Center for Idling Brain Science, University of Toyama, Toyama, Japan.
- CREST, Japan Science and Technology Agency (JST), University of Toyama, Toyama, Japan.
| |
Collapse
|
26
|
Delamare G, Tomé DF, Clopath C. Intrinsic Neural Excitability Biases Allocation and Overlap of Memory Engrams. J Neurosci 2024; 44:e0846232024. [PMID: 38561228 PMCID: PMC11112642 DOI: 10.1523/jneurosci.0846-23.2024] [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: 05/05/2023] [Revised: 03/25/2024] [Accepted: 03/25/2024] [Indexed: 04/04/2024] Open
Abstract
Memories are thought to be stored in neural ensembles known as engrams that are specifically reactivated during memory recall. Recent studies have found that memory engrams of two events that happened close in time tend to overlap in the hippocampus and the amygdala, and these overlaps have been shown to support memory linking. It has been hypothesized that engram overlaps arise from the mechanisms that regulate memory allocation itself, involving neural excitability, but the exact process remains unclear. Indeed, most theoretical studies focus on synaptic plasticity and little is known about the role of intrinsic plasticity, which could be mediated by neural excitability and serve as a complementary mechanism for forming memory engrams. Here, we developed a rate-based recurrent neural network that includes both synaptic plasticity and neural excitability. We obtained structural and functional overlap of memory engrams for contexts that are presented close in time, consistent with experimental and computational studies. We then investigated the role of excitability in memory allocation at the network level and unveiled competitive mechanisms driven by inhibition. This work suggests mechanisms underlying the role of intrinsic excitability in memory allocation and linking, and yields predictions regarding the formation and the overlap of memory engrams.
Collapse
Affiliation(s)
- Geoffroy Delamare
- Department of Bioengineering, Imperial College London, London SW7 2AZ, UK
| | - Douglas Feitosa Tomé
- Department of Bioengineering, Imperial College London, London SW7 2AZ, UK
- Institute of Science and Technology Austria, Klosterneuburg 3400, Austria
| | - Claudia Clopath
- Department of Bioengineering, Imperial College London, London SW7 2AZ, UK
| |
Collapse
|
27
|
Tian M, Kawaguchi R, Shen Y, Machnicki M, Villegas NG, Cooper DR, Montgomery N, Haring J, Lan R, Yuan AH, Williams CK, Magaki S, Vinters HV, Zhang Y, De Biase LM, Silva AJ, Carmichael ST. Intercellular Signaling Pathways as Therapeutic Targets for Vascular Dementia Repair. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.24.585301. [PMID: 38585718 PMCID: PMC10996514 DOI: 10.1101/2024.03.24.585301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Vascular dementia (VaD) is a white matter ischemic disease and the second-leading cause of dementia, with no direct therapy. Within the lesion site, cell-cell interactions dictate the trajectory towards disease progression or repair. To elucidate the underlying intercellular signaling pathways, a VaD mouse model was developed for transcriptomic and functional studies. The mouse VaD transcriptome was integrated with a human VaD snRNA-Seq dataset. A custom-made database encompassing 4053 human and 2032 mouse ligand-receptor (L-R) interactions identified significantly altered pathways shared between human and mouse VaD. Two intercellular L-R systems, Serpine2-Lrp1 and CD39-A3AR, were selected for mechanistic study as both the ligand and receptor were dysregulated in VaD. Decreased Seprine2 expression enhances OPC differentiation in VaD repair. A clinically relevant drug that reverses the loss of CD39-A3AR function promotes tissue and behavioral recovery in the VaD model. This study presents novel intercellular signaling targets and may open new avenues for VaD therapies.
Collapse
|
28
|
Baghdassarian HM, Lewis NE. Resource allocation in mammalian systems. Biotechnol Adv 2024; 71:108305. [PMID: 38215956 PMCID: PMC11182366 DOI: 10.1016/j.biotechadv.2023.108305] [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: 08/03/2023] [Revised: 12/17/2023] [Accepted: 12/18/2023] [Indexed: 01/14/2024]
Abstract
Cells execute biological functions to support phenotypes such as growth, migration, and secretion. Complementarily, each function of a cell has resource costs that constrain phenotype. Resource allocation by a cell allows it to manage these costs and optimize their phenotypes. In fact, the management of resource constraints (e.g., nutrient availability, bioenergetic capacity, and macromolecular machinery production) shape activity and ultimately impact phenotype. In mammalian systems, quantification of resource allocation provides important insights into higher-order multicellular functions; it shapes intercellular interactions and relays environmental cues for tissues to coordinate individual cells to overcome resource constraints and achieve population-level behavior. Furthermore, these constraints, objectives, and phenotypes are context-dependent, with cells adapting their behavior according to their microenvironment, resulting in distinct steady-states. This review will highlight the biological insights gained from probing resource allocation in mammalian cells and tissues.
Collapse
Affiliation(s)
- Hratch M Baghdassarian
- Bioinformatics and Systems Biology Graduate Program, University of California, San Diego, La Jolla, CA 92093, USA; Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Nathan E Lewis
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA; Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
29
|
Ellwanger JH, Chies JAB. Toxicogenomics of the C-C chemokine receptor type 5 (CCR5): Exploring the potential impacts of chemical-CCR5 interactions on inflammation and human health. Food Chem Toxicol 2024; 186:114511. [PMID: 38360389 DOI: 10.1016/j.fct.2024.114511] [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: 05/22/2023] [Revised: 02/01/2024] [Accepted: 02/07/2024] [Indexed: 02/17/2024]
Abstract
This article explores the impact of environmental chemicals on CCR5 expression and related inflammatory responses based on curated data from the Comparative Toxicogenomics Database (CTD). A total of 143 CCR5-interacting chemicals was found, with 229 chemical interactions. Of note, 67 (29.3%) out of 229 interactions resulted in "increased expression" of CCR5 mRNA or CCR5 protein, and 42 (18.3%) chemical interactions resulted in "decreased expression". The top-5 CCR5-interacting chemicals were "Tetrachlorodibenzodioxin", "Lipopolysaccharides", "Benzo(a)pyrene", "Drugs, Chinese Herbal", and "Ethinyl Estradiol". Based on the number of interactions and importance as environmental contaminant, we then focused our analysis on Tetrachlorodibenzodioxin and Benzo(a)pyrene. There is some consistency in the data supporting an increase in CCR5 expression triggered by Tetrachlorodibenzodioxin; although data concerning CCR5-Benzo(a)pyrene interactions is limited. Considering the high linkage disequilibrium between CCR5 and CCR2 genes, we also search for chemicals that interact with both genes, which resulted in 72 interacting chemicals, representing 50.3% of the 143 CCR5-interacting chemicals and 37.5% of the 192 CCR2-interacting chemicals. In conclusion, CTD data showed that environmental contaminants indeed affect CCR5 expression, with a tendency towards increased expression. The interaction of environmental contaminants with other chemokine receptor genes may potentialize their toxic effects on the chemokine system, favoring inflammation.
Collapse
Affiliation(s)
- Joel Henrique Ellwanger
- Laboratory of Immunobiology and Immunogenetics, Postgraduate Program in Genetics and Molecular Biology (PPGBM), Department of Genetics, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, 91501-970, Brazil.
| | - José Artur Bogo Chies
- Laboratory of Immunobiology and Immunogenetics, Postgraduate Program in Genetics and Molecular Biology (PPGBM), Department of Genetics, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, 91501-970, Brazil
| |
Collapse
|
30
|
Huang HY, Salinas S, Cornell J, Udoh IB, Shen Y, Zhou M. CCR5 regulates Aβ 1-42-induced learning and memory deficits in mice. Neurobiol Learn Mem 2024; 208:107890. [PMID: 38215963 DOI: 10.1016/j.nlm.2024.107890] [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: 03/28/2023] [Revised: 10/02/2023] [Accepted: 01/09/2024] [Indexed: 01/14/2024]
Abstract
C-C chemokine receptor 5 (CCR5) is a chemokine receptor involved in immune responses and a co-receptor for HIV infection. Recently, CCR5 has also been reported to play a role in synaptic plasticity, learning and memory, and cognitive deficits associated with normal aging, traumatic brain injury (TBI), and HIV-associated neurocognitive disorder (HAND). In contrast, the role of CCR5 in cognitive deficits associated with other disorders, including Alzheimer's disease (AD), is much less understood. Studies have reported an increase in expression of CCR5 or its ligands in both AD patients and AD rodent models, suggesting a correlation between AD and CCR5 expression. However, whether blocking CCR5 in specific brain regions, such as the hippocampus, could improve memory deficits in AD mouse models is unknown. To study the potential causal role of CCR5 in cognitive deficits in AD, we injected soluble Aβ1-42 or a control (Aβ42-1) oligomers in the dorsal CA1 region of the hippocampus and found that Aβ1-42 injection resulted in severe memory impairment in the object place recognition (OPR) and novel object recognition (NOR) tests. Aβ1-42 injection caused an increase in Ccr5, Ccl3, and Ccl4 in the dorsal hippocampus, and the expression levels of CCR5 and its ligands remained elevated at 2 weeks after Aβ1-42 injection. Knocking down Ccr5 in the CA1 region of dorsal hippocampus reversed the increase in microglia number and size in dorsal CA1 and rescued memory deficits. These results indicate that CCR5 plays an important role in modulating Aβ1-42-induced learning and memory deficits, and suggest that CCR5 antagonists may serve as a potential treatment to improve cognitive deficits associated with AD.
Collapse
Affiliation(s)
- Hou-Yuan Huang
- College of Dental Medicine, Western University of Health Sciences, Pomona, CA, USA
| | - Shelbi Salinas
- College of Dental Medicine, Western University of Health Sciences, Pomona, CA, USA
| | - Jessica Cornell
- College of Dental Medicine, Western University of Health Sciences, Pomona, CA, USA
| | - Iquo-Bella Udoh
- College of Dental Medicine, Western University of Health Sciences, Pomona, CA, USA
| | - Yang Shen
- Neurobiology, Psychiatry and Psychology Departments & Integrative Center for Learning and Memory, UCLA, Los Angeles, CA, USA
| | - Miou Zhou
- College of Dental Medicine, Western University of Health Sciences, Pomona, CA, USA.
| |
Collapse
|
31
|
Dong H, Dai X, Zhou Y, Shi C, Bhuiyan P, Sun Z, Li N, Jin W. Enhanced meningeal lymphatic drainage ameliorates lipopolysaccharide-induced brain injury in aged mice. J Neuroinflammation 2024; 21:36. [PMID: 38287311 PMCID: PMC10826026 DOI: 10.1186/s12974-024-03028-4] [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: 09/16/2023] [Accepted: 01/22/2024] [Indexed: 01/31/2024] Open
Abstract
BACKGROUND Sepsis-associated encephalopathy (SAE) is an acute cerebral dysfunction caused by sepsis. Neuroinflammation induced by sepsis is considered a potential mechanism of SAE; however, very little is known about the role of the meningeal lymphatic system in SAE. METHODS Sepsis was established in male C57BL/6J mice by intraperitoneal injection of 5 mg/kg lipopolysaccharide, and the function of meningeal lymphatic drainage was assessed. Adeno-associated virus 1-vascular endothelial growth factor C (AAV1-VEGF-C) was injected into the cisterna magna to induce meningeal lymphangiogenesis. Ligation of deep cervical lymph nodes (dCLNs) was performed to induce pre-existing meningeal lymphatic dysfunction. Cognitive function was evaluated by a fear conditioning test, and inflammatory factors were detected by enzyme-linked immunosorbent assay. RESULTS The aged mice with SAE showed a significant decrease in the drainage of OVA-647 into the dCLNs and the coverage of the Lyve-1 in the meningeal lymphatic, indicating that sepsis impaired meningeal lymphatic drainage and morphology. The meningeal lymphatic function of aged mice was more vulnerable to sepsis in comparison to young mice. Sepsis also decreased the protein levels of caspase-3 and PSD95, which was accompanied by reductions in the activity of hippocampal neurons. Microglia were significantly activated in the hippocampus of SAE mice, which was accompanied by an increase in neuroinflammation, as indicated by increases in interleukin-1 beta, interleukin-6 and Iba1 expression. Cognitive function was impaired in aged mice with SAE. However, the injection of AAV1-VEGF-C significantly increased coverage in the lymphatic system and tracer dye uptake in dCLNs, suggesting that AAV1-VEGF-C promotes meningeal lymphangiogenesis and drainage. Furthermore, AAV1-VEGF-C reduced microglial activation and neuroinflammation and improved cognitive dysfunction. Improvement of meningeal lymphatics also reduced sepsis-induced expression of disease-associated genes in aged mice. Pre-existing lymphatic dysfunction by ligating bilateral dCLNs aggravated sepsis-induced neuroinflammation and cognitive impairment. CONCLUSION The meningeal lymphatic drainage is damaged in sepsis, and pre-existing defects in this drainage system exacerbate SAE-induced neuroinflammation and cognitive dysfunction. Promoting meningeal lymphatic drainage improves SAE. Manipulation of meningeal lymphangiogenesis could be a new strategy for the treatment of SAE.
Collapse
Affiliation(s)
- Hongquan Dong
- Department of Anesthesiology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Xiaonan Dai
- Department of Obstetrics, Nanjing Women and Children's Healthcare Hospital, Women's Hospital of Nanjing Medical University, Nanjing, 210004, China
| | - Yin Zhou
- Department of Anesthesiology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Chonglong Shi
- Department of Anesthesiology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Piplu Bhuiyan
- Department of Anesthesiology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Zhaochu Sun
- Department of Anesthesiology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Nana Li
- Department of Anesthesiology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Wenjie Jin
- Department of Anesthesiology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
32
|
Jung JH, Wang Y, Rashid AJ, Zhang T, Frankland PW, Josselyn SA. Examining memory linking and generalization using scFLARE2, a temporally precise neuronal activity tagging system. Cell Rep 2023; 42:113592. [PMID: 38103203 PMCID: PMC10842737 DOI: 10.1016/j.celrep.2023.113592] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 10/26/2023] [Accepted: 11/30/2023] [Indexed: 12/18/2023] Open
Abstract
How memories are organized in the brain influences whether they are remembered discretely versus linked with other experiences or whether generalized information is applied to entirely novel situations. Here, we used scFLARE2 (single-chain fast light- and activity-regulated expression 2), a temporally precise tagging system, to manipulate mouse lateral amygdala neurons active during one of two 3 min threat experiences occurring close (3 h) or further apart (27 h) in time. Silencing scFLARE2-tagged neurons showed that two threat experiences occurring at distal times are dis-allocated to orthogonal engram ensembles and remembered discretely, whereas the same two threat experiences occurring in close temporal proximity are linked via co-allocation to overlapping engram ensembles. Moreover, we found that co-allocation mediates memory generalization applied to a completely novel stimulus. These results indicate that endogenous temporal evolution of engram ensemble neuronal excitability determines how memories are organized and remembered and that this would not be possible using conventional immediate-early gene-based tagging methods.
Collapse
Affiliation(s)
- Jung Hoon Jung
- Program in Neurosciences & Mental Health, Hospital for Sick Children, 555 University Avenue, Toronto, ON M5G 1X8, Canada
| | - Ying Wang
- Program in Neurosciences & Mental Health, Hospital for Sick Children, 555 University Avenue, Toronto, ON M5G 1X8, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Asim J Rashid
- Program in Neurosciences & Mental Health, Hospital for Sick Children, 555 University Avenue, Toronto, ON M5G 1X8, Canada
| | - Tao Zhang
- Program in Neurosciences & Mental Health, Hospital for Sick Children, 555 University Avenue, Toronto, ON M5G 1X8, Canada
| | - Paul W Frankland
- Program in Neurosciences & Mental Health, Hospital for Sick Children, 555 University Avenue, Toronto, ON M5G 1X8, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Psychology, University of Toronto, Toronto, ON M5S 3G3, Canada; Child & Brain Development Program, Canadian Institute for Advanced Research (CIFAR), Toronto, ON M5G 1M1, Canada
| | - Sheena A Josselyn
- Program in Neurosciences & Mental Health, Hospital for Sick Children, 555 University Avenue, Toronto, ON M5G 1X8, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Psychology, University of Toronto, Toronto, ON M5S 3G3, Canada.
| |
Collapse
|
33
|
Berdugo‐Vega G, Dhingra S, Calegari F. Sharpening the blades of the dentate gyrus: how adult-born neurons differentially modulate diverse aspects of hippocampal learning and memory. EMBO J 2023; 42:e113524. [PMID: 37743770 PMCID: PMC11059975 DOI: 10.15252/embj.2023113524] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 06/19/2023] [Accepted: 08/18/2023] [Indexed: 09/26/2023] Open
Abstract
For decades, the mammalian hippocampus has been the focus of cellular, anatomical, behavioral, and computational studies aimed at understanding the fundamental mechanisms underlying cognition. Long recognized as the brain's seat for learning and memory, a wealth of knowledge has been accumulated on how the hippocampus processes sensory input, builds complex associations between objects, events, and space, and stores this information in the form of memories to be retrieved later in life. However, despite major efforts, our understanding of hippocampal cognitive function remains fragmentary, and models trying to explain it are continually revisited. Here, we review the literature across all above-mentioned domains and offer a new perspective by bringing attention to the most distinctive, and generally neglected, feature of the mammalian hippocampal formation, namely, the structural separability of the two blades of the dentate gyrus into "supra-pyramidal" and "infra-pyramidal". Next, we discuss recent reports supporting differential effects of adult neurogenesis in the regulation of mature granule cell activity in these two blades. We propose a model for how differences in connectivity and adult neurogenesis in the two blades can potentially provide a substrate for subtly different cognitive functions.
Collapse
Affiliation(s)
- Gabriel Berdugo‐Vega
- CRTD‐Center for Regenerative Therapies DresdenTechnische Universität DresdenDresdenGermany
- Present address:
Laboratory of Neuroepigenetics, Brain Mind Institute, School of Life Sciences, École Polytechnique Fédérale Lausanne (EPFL)LausanneSwitzerland
| | - Shonali Dhingra
- CRTD‐Center for Regenerative Therapies DresdenTechnische Universität DresdenDresdenGermany
| | - Federico Calegari
- CRTD‐Center for Regenerative Therapies DresdenTechnische Universität DresdenDresdenGermany
| |
Collapse
|
34
|
Campos B, Choi H, DeMarco AT, Seydell-Greenwald A, Hussain SJ, Joy MT, Turkeltaub PE, Zeiger W. Rethinking Remapping: Circuit Mechanisms of Recovery after Stroke. J Neurosci 2023; 43:7489-7500. [PMID: 37940595 PMCID: PMC10634578 DOI: 10.1523/jneurosci.1425-23.2023] [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: 07/27/2023] [Revised: 08/21/2023] [Accepted: 08/21/2023] [Indexed: 11/10/2023] Open
Abstract
Stroke is one of the most common causes of disability, and there are few treatments that can improve recovery after stroke. Therapeutic development has been hindered because of a lack of understanding of precisely how neural circuits are affected by stroke, and how these circuits change to mediate recovery. Indeed, some of the hypotheses for how the CNS changes to mediate recovery, including remapping, redundancy, and diaschisis, date to more than a century ago. Recent technological advances have enabled the interrogation of neural circuits with ever greater temporal and spatial resolution. These techniques are increasingly being applied across animal models of stroke and to human stroke survivors, and are shedding light on the molecular, structural, and functional changes that neural circuits undergo after stroke. Here we review these studies and highlight important mechanisms that underlie impairment and recovery after stroke. We begin by summarizing knowledge about changes in neural activity that occur in the peri-infarct cortex, specifically considering evidence for the functional remapping hypothesis of recovery. Next, we describe the importance of neural population dynamics, disruptions in these dynamics after stroke, and how allocation of neurons into spared circuits can restore functionality. On a more global scale, we then discuss how effects on long-range pathways, including interhemispheric interactions and corticospinal tract transmission, contribute to post-stroke impairments. Finally, we look forward and consider how a deeper understanding of neural circuit mechanisms of recovery may lead to novel treatments to reduce disability and improve recovery after stroke.
Collapse
Affiliation(s)
- Baruc Campos
- Department of Neurology, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, California 90095
| | - Hoseok Choi
- Department of Neurology, Weill Institute for Neuroscience, University of California-San Francisco, San Francisco, California 94158
| | - Andrew T DeMarco
- Center for Brain Plasticity and Recovery, Georgetown University Medical Center, Georgetown University, Washington, DC 20057
- Department of Rehabilitation Medicine, Georgetown University Medical Center, Georgetown University, Washington, DC 20057
| | - Anna Seydell-Greenwald
- Center for Brain Plasticity and Recovery, Georgetown University Medical Center, Georgetown University, Washington, DC 20057
- MedStar National Rehabilitation Hospital, Washington, DC 20010
| | - Sara J Hussain
- Movement and Cognitive Rehabilitation Science Program, Department of Kinesiology and Health Education, University of Texas at Austin, Austin, Texas 78712
| | - Mary T Joy
- The Jackson Laboratory, Bar Harbor, Maine 04609
| | - Peter E Turkeltaub
- Center for Brain Plasticity and Recovery, Georgetown University Medical Center, Georgetown University, Washington, DC 20057
- MedStar National Rehabilitation Hospital, Washington, DC 20010
| | - William Zeiger
- Department of Neurology, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, California 90095
| |
Collapse
|
35
|
Huang Y, Liu B, Sinha SC, Amin S, Gan L. Mechanism and therapeutic potential of targeting cGAS-STING signaling in neurological disorders. Mol Neurodegener 2023; 18:79. [PMID: 37941028 PMCID: PMC10634099 DOI: 10.1186/s13024-023-00672-x] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 10/25/2023] [Indexed: 11/10/2023] Open
Abstract
DNA sensing is a pivotal component of the innate immune system that is responsible for detecting mislocalized DNA and triggering downstream inflammatory pathways. Among the DNA sensors, cyclic GMP-AMP synthase (cGAS) is a primary player in detecting cytosolic DNA, including foreign DNA from pathogens and self-DNA released during cellular damage, culminating in a type I interferon (IFN-I) response through stimulator of interferon genes (STING) activation. IFN-I cytokines are essential in mediating neuroinflammation, which is widely observed in CNS injury, neurodegeneration, and aging, suggesting an upstream role for the cGAS DNA sensing pathway. In this review, we summarize the latest developments on the cGAS-STING DNA-driven immune response in various neurological diseases and conditions. Our review covers the current understanding of the molecular mechanisms of cGAS activation and highlights cGAS-STING signaling in various cell types of central and peripheral nervous systems, such as resident brain immune cells, neurons, and glial cells. We then discuss the role of cGAS-STING signaling in different neurodegenerative conditions, including tauopathies, Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis, as well as aging and senescence. Finally, we lay out the current advancements in research and development of cGAS inhibitors and assess the prospects of targeting cGAS and STING as therapeutic strategies for a wide spectrum of neurological diseases.
Collapse
Affiliation(s)
- Yige Huang
- Helen and Robert Appel Alzheimer Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
- Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, USA
| | - Bangyan Liu
- Helen and Robert Appel Alzheimer Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
- Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, USA
| | - Subhash C Sinha
- Helen and Robert Appel Alzheimer Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Sadaf Amin
- Helen and Robert Appel Alzheimer Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Li Gan
- Helen and Robert Appel Alzheimer Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA.
- Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
36
|
Wang Z, Zheng D, Tan YS, Yuan Q, Yuan F, Zhang S. Enabling Survival of Transplanted Neural Precursor Cells in the Ischemic Brain. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2302527. [PMID: 37867250 PMCID: PMC10667812 DOI: 10.1002/advs.202302527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 07/24/2023] [Indexed: 10/24/2023]
Abstract
There is no effective therapy for ischemic stroke following the acute stage. Neural transplantation offers a potential option for repairing the ischemic lesion. However, this strategy is hindered by the poor survival of the neural precursor cells (NPCs) that are transplanted into the inflammatory ischemic core. Here, a chemical cocktail consisting of fibrinogen and maraviroc is developed to promote the survival of the transplanted NPCs in the ischemic core of the mouse cerebral cortex. The grafted NPCs survive in the presence of the cocktail but not fibrinogen or maraviroc alone at day 7. The surviving NPCs divide and differentiate to mature neurons by day 30, reconstituting the infarct cortex with vascularization. Molecular analysis in vivo and in vitro shows that blocking the activation of CCR5 on the NPCs protects the NPCs from apoptosis induced by pro-inflammatory factors, revealing the underlying protective effect of the cocktail for NPCs. The findings open an avenue to enable survival of the transplanted NPCs under the inflammatory neurological conditions like stroke.
Collapse
Affiliation(s)
- Zhifu Wang
- Program in Neuroscience & Behavioral Disorders, GK Goh Centre for NeuroscienceDuke‐NUS Medical SchoolSingapore169857Singapore
| | - Danyi Zheng
- Program in Neuroscience & Behavioral Disorders, GK Goh Centre for NeuroscienceDuke‐NUS Medical SchoolSingapore169857Singapore
| | - Ye Sing Tan
- Program in Neuroscience & Behavioral Disorders, GK Goh Centre for NeuroscienceDuke‐NUS Medical SchoolSingapore169857Singapore
| | - Qiang Yuan
- Program in Neuroscience & Behavioral Disorders, GK Goh Centre for NeuroscienceDuke‐NUS Medical SchoolSingapore169857Singapore
| | - Fang Yuan
- Program in Neuroscience & Behavioral Disorders, GK Goh Centre for NeuroscienceDuke‐NUS Medical SchoolSingapore169857Singapore
| | - Su‐Chun Zhang
- Program in Neuroscience & Behavioral Disorders, GK Goh Centre for NeuroscienceDuke‐NUS Medical SchoolSingapore169857Singapore
- Department of NeuroscienceDepartment of NeurologyWaisman CenterUniversity of Wisconsin‐MadisonMadisonWI53705USA
| |
Collapse
|
37
|
Ma W, Liu A, Wu X, Gao L, Chen J, Wu H, Liu M, Fan Y, Peng L, Yang J, Kong J, Li B, Ji Z, Dong Y, Luo S, Song J, Bao F. The intricate role of CCL5/CCR5 axis in Alzheimer disease. J Neuropathol Exp Neurol 2023; 82:894-900. [PMID: 37769321 PMCID: PMC10587995 DOI: 10.1093/jnen/nlad071] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/30/2023] Open
Abstract
The morbidity and mortality associated with Alzheimer disease (AD), one of the most common neurodegenerative diseases, are increasing each year. Although both amyloid β and tau proteins are known to be involved in AD pathology, their detailed functions in the pathogenesis of the disease are not fully understood. There is increasing evidence that neuroinflammation contributes to the development and progression of AD, with astrocytes, microglia, and the cytokines and chemokines they secrete acting coordinately in these processes. Signaling involving chemokine (C-C motif) ligand 5 (CCL5) and its main receptor C-C chemokine receptor 5 (CCR5) plays an important role in normal physiologic processes as well as pathologic conditions such as neurodegeneration. In recent years, many studies have shown that the CCL5/CCR5 axis plays a major effect in the pathogenesis of AD, but there are also a few studies that contradict this. In short, the role of CCL5/CCR5 axis in the pathogenesis of AD is still intricate. This review summarizes the structure, distribution, physiologic functions of the CCL5/CCR5 axis, and the progress in understanding its involvement in the pathogenesis of AD.
Collapse
Affiliation(s)
- Weijiang Ma
- Evidence-Based Medicine Team, Faculty of Basic Medical Sciences, The Institute for Tropical Medicine, Kunming Medical University, Kunming, Yunnan, China
| | - Aihua Liu
- Evidence-Based Medicine Team, Faculty of Basic Medical Sciences, The Institute for Tropical Medicine, Kunming Medical University, Kunming, Yunnan, China
- Yunnan Province Key Laboratory of Children’s Major Diseases Research, The Affiliated Children Hospital, Kunming Medical University, Kunming, Yunnan, China
| | - Xinya Wu
- Evidence-Based Medicine Team, Faculty of Basic Medical Sciences, The Institute for Tropical Medicine, Kunming Medical University, Kunming, Yunnan, China
| | - Li Gao
- Evidence-Based Medicine Team, Faculty of Basic Medical Sciences, The Institute for Tropical Medicine, Kunming Medical University, Kunming, Yunnan, China
| | - Jingjing Chen
- Evidence-Based Medicine Team, Faculty of Basic Medical Sciences, The Institute for Tropical Medicine, Kunming Medical University, Kunming, Yunnan, China
| | - Hanxin Wu
- Evidence-Based Medicine Team, Faculty of Basic Medical Sciences, The Institute for Tropical Medicine, Kunming Medical University, Kunming, Yunnan, China
| | - Meixiao Liu
- Evidence-Based Medicine Team, Faculty of Basic Medical Sciences, The Institute for Tropical Medicine, Kunming Medical University, Kunming, Yunnan, China
| | - Yuxin Fan
- Evidence-Based Medicine Team, Faculty of Basic Medical Sciences, The Institute for Tropical Medicine, Kunming Medical University, Kunming, Yunnan, China
| | - Li Peng
- Evidence-Based Medicine Team, Faculty of Basic Medical Sciences, The Institute for Tropical Medicine, Kunming Medical University, Kunming, Yunnan, China
| | - Jiaru Yang
- Evidence-Based Medicine Team, Faculty of Basic Medical Sciences, The Institute for Tropical Medicine, Kunming Medical University, Kunming, Yunnan, China
| | - Jing Kong
- Evidence-Based Medicine Team, Faculty of Basic Medical Sciences, The Institute for Tropical Medicine, Kunming Medical University, Kunming, Yunnan, China
| | - Bingxue Li
- Evidence-Based Medicine Team, Faculty of Basic Medical Sciences, The Institute for Tropical Medicine, Kunming Medical University, Kunming, Yunnan, China
| | - Zhenhua Ji
- Evidence-Based Medicine Team, Faculty of Basic Medical Sciences, The Institute for Tropical Medicine, Kunming Medical University, Kunming, Yunnan, China
| | - Yan Dong
- Evidence-Based Medicine Team, Faculty of Basic Medical Sciences, The Institute for Tropical Medicine, Kunming Medical University, Kunming, Yunnan, China
| | - Suyi Luo
- Evidence-Based Medicine Team, Faculty of Basic Medical Sciences, The Institute for Tropical Medicine, Kunming Medical University, Kunming, Yunnan, China
| | - Jieqin Song
- Evidence-Based Medicine Team, Faculty of Basic Medical Sciences, The Institute for Tropical Medicine, Kunming Medical University, Kunming, Yunnan, China
| | - Fukai Bao
- Evidence-Based Medicine Team, Faculty of Basic Medical Sciences, The Institute for Tropical Medicine, Kunming Medical University, Kunming, Yunnan, China
- Yunnan Province Key Laboratory of Children’s Major Diseases Research, The Affiliated Children Hospital, Kunming Medical University, Kunming, Yunnan, China
| |
Collapse
|
38
|
Sehgal M, Ehlers VE, Moyer JR. Synaptic and intrinsic plasticity within overlapping lateral amygdala ensembles following fear conditioning. Front Cell Neurosci 2023; 17:1221176. [PMID: 37876914 PMCID: PMC10590925 DOI: 10.3389/fncel.2023.1221176] [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: 05/11/2023] [Accepted: 09/20/2023] [Indexed: 10/26/2023] Open
Abstract
Introduction New learning results in modulation of intrinsic plasticity in the underlying brain regions. Such changes in intrinsic plasticity can influence allocation and encoding of future memories such that new memories encoded during the period of enhanced excitability are linked to the original memory. The temporal window during which the two memories interact depends upon the time course of intrinsic plasticity following new learning. Methods Using the well-characterized lateral amygdala-dependent auditory fear conditioning as a behavioral paradigm, we investigated the time course of changes in intrinsic excitability within lateral amygdala neurons. Results We found transient changes in the intrinsic excitability of amygdala neurons. Neuronal excitability was increased immediately following fear conditioning and persisted for up to 4 days post-learning but was back to naïve levels 10 days following fear conditioning. We also determined the relationship between learning-induced intrinsic and synaptic plasticity. Synaptic plasticity following fear conditioning was evident for up to 24 h but not 4 days later. Importantly, we demonstrated that the enhanced neuronal intrinsic excitability was evident in many of the same neurons that had undergone synaptic plasticity immediately following fear conditioning. Interestingly, such a correlation between synaptic and intrinsic plasticity following fear conditioning was no longer present 24 h post-learning. Discussion These data demonstrate that intrinsic and synaptic changes following fear conditioning are transient and co-localized to the same neurons. Since intrinsic plasticity following fear conditioning is an important determinant for the allocation and consolidation of future amygdala-dependent memories, these findings establish a time course during which fear memories may influence each other.
Collapse
Affiliation(s)
- Megha Sehgal
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, United States
| | - Vanessa E. Ehlers
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, United States
| | - James R. Moyer
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, United States
- Department of Biological Sciences, University of Wisconsin-Milwaukee, Milwaukee, WI, United States
| |
Collapse
|
39
|
Wu QL, Cui LY, Ma WY, Wang SS, Zhang Z, Feng ZP, Sun HS, Chu SF, He WB, Chen NH. A novel small-molecular CCR5 antagonist promotes neural repair after stroke. Acta Pharmacol Sin 2023; 44:1935-1947. [PMID: 37198412 PMCID: PMC10545720 DOI: 10.1038/s41401-023-01100-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 04/25/2023] [Indexed: 05/19/2023]
Abstract
Chemokine receptor 5 (CCR5) is one of the main co-receptors of HIV-1, and has been found to be a potential therapeutic target for stroke. Maraviroc is a classic CCR5 antagonist, which is undergoing clinical trials against stroke. As maraviroc shows poor blood-brain barrier (BBB) permeability, it is of interest to find novel CCR5 antagonists suitable for neurological medication. In this study we characterized the therapeutic potential of a novel CCR5 antagonist A14 in treating ischemic stroke mice. A14 was discovered in screening millions compounds in the Chemdiv library based on the molecular docking diagram of CCR5 and maraviroc. We found that A14 dose-dependently inhibited the CCR5 activity with an IC50 value of 4.29 μM. Pharmacodynamic studies showed that A14 treatment exerted protective effects against neuronal ischemic injury both in vitro and vivo. In a SH-SY5Y cell line overexpressing CCR5, A14 (0.1, 1 μM) significantly alleviated OGD/R-induced cell injury. We found that the expression of CCR5 and its ligand CKLF1 was significantly upregulated during both acute and recovery period in focal cortical stroke mice; oral administration of A14 (20 mg·kg-1·d-1, for 1 week) produced sustained protective effect against motor impairment. A14 treatment had earlier onset time, lower onset dosage and much better BBB permeability compared to maraviroc. MRI analysis also showed that A14 treatment significantly reduced the infarction volume after 1 week of treatment. We further revealed that A14 treatment blocked the protein-protein interaction between CCR5 and CKLF1, increasing the activity of CREB signaling pathway in neurons, thereby improving axonal sprouting and synaptic density after stroke. In addition, A14 treatment remarkably inhibited the reactive proliferation of glial cells after stroke and reduced the infiltration of peripheral immune cells. These results demonstrate that A14 is a promising novel CCR5 antagonist for promoting neuronal repair after ischemic stroke. A14 blocked the protein-protein interaction between CKLF1 and CCR5 after stroke by binding with CCR5 stably, improved the infarct area and promoted motor recovery through reversing the CREB/pCREB signaling which was inhibited by activated CCR5 Gαi pathway, and benefited to the dendritic spines and axons sprouting.
Collapse
Affiliation(s)
- Qing-Lin Wu
- Clinical Pharmacology Institute, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Li-Yuan Cui
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Wen-Yu Ma
- Clinical Pharmacology Institute, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Sha-Sha Wang
- Clinical Pharmacology Institute, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Zhao Zhang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Zhong-Ping Feng
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
| | - Hong-Shuo Sun
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
| | - Shi-Feng Chu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| | - Wen-Bin He
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Shanxi University of Chinese Medicine, Taiyuan, 030024, China.
- National International Joint Research Center for Molecular Chinese Medicine, Shanxi University of Chinese Medicine, Taiyuan, 030024, China.
| | - Nai-Hong Chen
- Clinical Pharmacology Institute, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| |
Collapse
|
40
|
Lin HY, Cathomas F, Li L, Cuttoli RDD, Guevara C, Bayrak CS, Wang Q, Gupta S, Chan KL, Shimo Y, Parise LF, Yuan C, Aubry AV, Chen F, Wong J, Morel C, Huntley GW, Zhang B, Russo SJ, Wang J. Chemokine receptor 5 signaling in PFC mediates stress susceptibility in female mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.18.553789. [PMID: 37662400 PMCID: PMC10473611 DOI: 10.1101/2023.08.18.553789] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Chronic stress induces changes in the periphery and the central nervous system (CNS) that contribute to neuropathology and behavioral abnormalities associated with psychiatric disorders. In this study, we examined the impact of peripheral and central inflammation during chronic social defeat stress (CSDS) in female mice. Compared to male mice, we found that female mice exhibited heightened peripheral inflammatory response and identified C-C motif chemokine ligand 5 (CCL5), as a stress-susceptibility marker in females. Blocking CCL5 signaling in the periphery promoted resilience to CSDS. In the brain, stress-susceptible mice displayed increased expression of C-C chemokine receptor 5 (CCR5), a receptor for CCL5, in microglia in the prefrontal cortex (PFC). This upregulation was associated with microglia morphological changes, their increased migration to the blood vessels, and enhanced phagocytosis of synaptic components and vascular material. These changes coincided with neurophysiological alterations and impaired blood-brain barrier (BBB) integrity. By blocking CCR5 signaling specifically in the PFC were able to prevent stress-induced physiological changes and rescue social avoidance behavior. Our findings are the first to demonstrate that stress-mediated dysregulation of the CCL5-CCR5 axis triggers excessive phagocytosis of synaptic materials and neurovascular components by microglia, resulting in disruptions in neurotransmission, reduced BBB integrity, and increased stress susceptibility. Our study provides new insights into the role of cortical microglia in female stress susceptibility and suggests that the CCL5-CCR5 axis may serve as a novel sex-specific therapeutic target for treating psychiatric disorders in females.
Collapse
|
41
|
Fang X, Alsbury-Nealy B, Wang Y, Frankland PW, Josselyn SA, Schlichting ML, Duncan KD. Time separating spatial memories does not influence their integration in humans. PLoS One 2023; 18:e0289649. [PMID: 37561677 PMCID: PMC10414573 DOI: 10.1371/journal.pone.0289649] [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: 12/24/2022] [Accepted: 07/23/2023] [Indexed: 08/12/2023] Open
Abstract
Humans can navigate through similar environments-like grocery stores-by integrating across their memories to extract commonalities or by differentiating between each to find idiosyncratic locations. Here, we investigate one factor that might impact whether two related spatial memories are integrated or differentiated: Namely, the temporal delay between experiences. Rodents have been shown to integrate memories more often when they are formed within 6 hours of each other. To test if this effect influences how humans spontaneously integrate spatial memories, we had 131 participants search for rewards in two similar virtual environments. We separated these learning experiences by either 30 minutes, 3 hours, or 27 hours. Memory integration was assessed three days later. Participants were able to integrate and simultaneously differentiate related memories across experiences. However, neither memory integration nor differentiation was modulated by temporal delay, in contrast to previous work. We further showed that both the levels of initial memory reactivation during the second experience and memory generalization to novel environments were comparable across conditions. Moreover, perseveration toward the initial reward locations during the second experience was related positively to integration and negatively to differentiation-but again, these associations did not vary by delay. Our findings identify important boundary conditions on the translation of rodent memory mechanisms to humans, motivating more research to characterize how even fundamental memory mechanisms are conserved and diverge across species.
Collapse
Affiliation(s)
- Xiaoping Fang
- Department of Psychology, University of Toronto, Toronto, Canada
- School of Psychology, Beijing Language and Culture University, Beijing, China
| | | | - Ying Wang
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, Canada
- Department of Physiology, University of Toronto, Toronto, Canada
| | - Paul W. Frankland
- Department of Psychology, University of Toronto, Toronto, Canada
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, Canada
- Department of Physiology, University of Toronto, Toronto, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
- Child & Brain Development Program, Canadian Institute for Advanced Research (CIFAR), Toronto, Canada
| | - Sheena A. Josselyn
- Department of Psychology, University of Toronto, Toronto, Canada
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, Canada
- Department of Physiology, University of Toronto, Toronto, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | | | | |
Collapse
|
42
|
Norasi E, Rastegar M, Hosseini SD, Aghcheli B, Tahamtan A. Prevalence of CCR5 Delta 32 Genetic Variant in the Turkmen Population of Golestan Province, Northeast of Iran. BIOMED RESEARCH INTERNATIONAL 2023; 2023:8823863. [PMID: 37388364 PMCID: PMC10307026 DOI: 10.1155/2023/8823863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 05/23/2023] [Accepted: 06/14/2023] [Indexed: 07/01/2023]
Abstract
The 32 bp deletion in the chemokine receptor (C-C motif) 5 gene (CCR5Δ32) is a natural loss of function polymorphism that prevents the protein from locating on the cell surface. This genetic variation acts as a double-edge sword in the pathogenesis/defense mechanism of different health conditions, such as viral infections, autoimmune diseases, and cancers. Here, we evaluated the prevalence of the CCR5Δ32 polymorphism in the Turkmen population of Golestan province, northeast of Iran. Blood samples were collected from 400 randomly selected Turkmen populations (199 women and 201 men), and genomic DNA was extracted. Characterization of CCR5Δ32 genotypes was performed by PCR using primers flanking the 32-nucleotide deletion in the CCR5 gene. The amplified DNA fragments were visualized on 2% agarose gel electrophoresis with cybergreen staining under UV light. All individuals were of Turkmen ethnicity and lived in the Golestan province, northeast of Iran. The mean age of all participants was 35.46 years, with a 20-45 year range. All the studied subjects were healthy without any severe conditions such as autoimmune disease and viral infections. All individuals had no history of HIV infection. The PCR product visualization showed that all the samples are at the 330 bp size, which means the CCR5Δ32 allele was utterly absent from the study population. The presence of the CCR5Δ32 allele among Turkmens may be attributed to the admixture with European descent people. We conclude that the CCR5Δ32 polymorphism may be absent in the Iranian Turkmen population, and further studies with a large population are needed.
Collapse
Affiliation(s)
- Elmira Norasi
- School of International, Golestan University of Medical Sciences, Gorgan, Iran
| | - Mostafa Rastegar
- Department of Microbiology, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | | | - Bahman Aghcheli
- Department of Microbiology, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Alireza Tahamtan
- Department of Microbiology, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
- Infectious Diseases Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| |
Collapse
|
43
|
Jing K, Chen F, Shi X, Guo J, Liu X. Dual effect of C-C motif chemokine receptor 5 on ischemic stroke: More harm than benefit? Eur J Pharmacol 2023:175857. [PMID: 37321471 DOI: 10.1016/j.ejphar.2023.175857] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 06/02/2023] [Accepted: 06/13/2023] [Indexed: 06/17/2023]
Abstract
Ischemic stroke involves a series of complex pathological mechanisms, of which neuroinflammation is currently the most widely recognized. C-C motif chemokine receptor 5 (CCR5) has recently been shown to be upregulated after cerebral ischemia. Notably, CCR5 is not only involved in neuroinflammation, but also in the blood-brain barrier, neural structures, and connections. Accumulating experimental studies indicate that CCR5 has a dual effect on ischemic stroke. In the acute phase after cerebral ischemia, the pro-inflammatory and disruptive effect of CCR5 on the blood-brain barrier predominates. However, in the chronic phase, the effect of CCR5 on the repair of neural structures and connections is thought to be cell-type dependent. Interestingly, clinical evidence has shown that CCR5 might be harmful rather than beneficial. CCR5-Δ32 mutation or CCR5 antagonist exerts a neuroprotective effect in patients with ischemic stroke. Considering CCR5 as an attractive potential target, we introduce the current research progress of the entangled relationships between CCR5 and ischemic stroke. Clinical data are still needed to determine the efficacy of activating or inactivating CCR5 in the treatment of ischemic stroke, especially for potential phase- or cell type-dependent treatments in the future.
Collapse
Affiliation(s)
- Kai Jing
- Department of Clinical Pharmacy, School of Pharmacy, Naval Medical University, Shanghai, China
| | - Feng Chen
- Department of Clinical Pharmacy, School of Pharmacy, Naval Medical University, Shanghai, China
| | - Xiaofei Shi
- Department of Clinical Pharmacy, School of Pharmacy, Naval Medical University, Shanghai, China
| | - Jinmin Guo
- Department of Clinical Pharmacy, 960th Hospital of Joint Logistic Support Force, Shandong, Jinan, China.
| | - Xia Liu
- Department of Clinical Pharmacy, School of Pharmacy, Naval Medical University, Shanghai, China.
| |
Collapse
|
44
|
Speidell A, Walton S, Campbell LA, Tomassoni-Ardori F, Tessarollo L, Corbo C, Taraballi F, Mocchetti I. Mice deficient for G-protein-coupled receptor 75 display altered presynaptic structural protein expression and disrupted fear conditioning recall. J Neurochem 2023; 165:827-841. [PMID: 36978267 PMCID: PMC10330141 DOI: 10.1111/jnc.15818] [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: 10/11/2022] [Revised: 03/23/2023] [Accepted: 03/26/2023] [Indexed: 03/30/2023]
Abstract
There are a number of G-protein-coupled receptors (GPCRs) that are considered "orphan receptors" because the information on their known ligands is incomplete. Yet, these receptors are important targets to characterize, as the discovery of their ligands may lead to potential new therapies. GPR75 was recently deorphanized because at least two ligands appear to bind to it, the chemokine CCL5 and the eicosanoid 20-Hydroxyeicosatetraenoic acid. Recent reports suggest that GPR75 may play a role in regulating insulin secretion and obesity. However, little is known about the function of this receptor in the brain. To study the function of GPR75, we have generated a knockout (KO) mouse model of this receptor and we evaluated the role that this receptor plays in the adult hippocampus by an array of histological, proteomic, and behavioral endpoints. Using RNAscope® technology, we identified GPR75 puncta in several Rbfox3-/NeuN-positive cells in the hippocampus, suggesting that this receptor has a neuronal expression. Proteomic analysis of the hippocampus in 3-month-old GPR75 KO animals revealed that several markers of synapses, including synapsin I and II are downregulated compared with wild type (WT). To examine the functional consequence of this down-regulation, WT and GPR75 KO mice were tested on a hippocampal-dependent behavioral task. Both contextual memory and anxiety-like behaviors were significantly altered in GPR75 KO, suggesting that GPR75 plays a role in hippocampal activity.
Collapse
Affiliation(s)
- Andrew Speidell
- Laboratory of Preclinical Neurobiology, Department of Neuroscience, Washington, DC
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC
| | - Sofia Walton
- Laboratory of Preclinical Neurobiology, Department of Neuroscience, Washington, DC
| | - Lee A Campbell
- Laboratory of Preclinical Neurobiology, Department of Neuroscience, Washington, DC
| | | | | | - Claudia Corbo
- School of Medicine and Surgery Nanomedicine Center, University of Milano-Bicocca, Milan, Italy
- IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| | - Francesca Taraballi
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute
- Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX
| | - Italo Mocchetti
- Laboratory of Preclinical Neurobiology, Department of Neuroscience, Washington, DC
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC
| |
Collapse
|
45
|
Abstract
Research in the last century has provided insight into the systems, cellular, and molecular processes involved in the formation, storage, recall, and update of memory engrams - the physical manifestation of the long sought-after philosophical and psychological concept of memory traces. Recent technologies allow scientists to visualize the key molecular players involved in segregating, ordering, and linking memories close in time, for future treatment of "disorders of the engram" where memory linking is deficient (e.g., cognitive aging or Alzheimer's) or excessive (e.g., PTSD).
Collapse
Affiliation(s)
- Catalin V Buhusi
- Department of Psychology, Interdisciplinary Program in Neuroscience, USTAR BioInnovations Center, Utah State University, Logan, UT, 84322, USA.
| | - Mona Buhusi
- Department of Psychology, Interdisciplinary Program in Neuroscience, USTAR BioInnovations Center, Utah State University, Logan, UT, 84322, USA
| |
Collapse
|
46
|
Zipp F, Bittner S, Schafer DP. Cytokines as emerging regulators of central nervous system synapses. Immunity 2023; 56:914-925. [PMID: 37163992 PMCID: PMC10233069 DOI: 10.1016/j.immuni.2023.04.011] [Citation(s) in RCA: 69] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 04/05/2023] [Accepted: 04/13/2023] [Indexed: 05/12/2023]
Abstract
Cytokines are key messengers by which immune cells communicate, and they drive many physiological processes, including immune and inflammatory responses. Early discoveries demonstrated that cytokines, such as the interleukin family members and TNF-α, regulate synaptic scaling and plasticity. Still, we continue to learn more about how these traditional immune system cytokines affect neuronal structure and function. Different cytokines shape synaptic function on multiple levels ranging from fine-tuning neurotransmission, to regulating synapse number, to impacting global neuronal networks and complex behavior. These recent findings have cultivated an exciting and growing field centered on the importance of immune system cytokines for regulating synapse and neural network structure and function. Here, we highlight the latest findings related to cytokines in the central nervous system and their regulation of synapse structure and function. Moreover, we explore how these mechanisms are becoming increasingly important to consider in diseases-especially those with a large neuroinflammatory component.
Collapse
Affiliation(s)
- Frauke Zipp
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine-Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany.
| | - Stefan Bittner
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine-Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Dorothy P Schafer
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
47
|
Greco GA, Rock M, Amontree M, Lanfranco MF, Korthas H, Hong SH, Turner RS, Rebeck GW, Conant K. CCR5 deficiency normalizes TIMP levels, working memory, and gamma oscillation power in APOE4 targeted replacement mice. Neurobiol Dis 2023; 179:106057. [PMID: 36878326 PMCID: PMC10291850 DOI: 10.1016/j.nbd.2023.106057] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 02/16/2023] [Accepted: 02/26/2023] [Indexed: 03/07/2023] Open
Abstract
The APOE4 allele increases the risk for Alzheimer's disease (AD) in a dose-dependent manner and is also associated with cognitive decline in non-demented elderly controls. In mice with targeted gene replacement (TR) of murine APOE with human APOE3 or APOE4, the latter show reduced neuronal dendritic complexity and impaired learning. APOE4 TR mice also show reduced gamma oscillation power, a neuronal population activity which is important to learning and memory. Published work has shown that brain extracellular matrix (ECM) can reduce neuroplasticity as well as gamma power, while attenuation of ECM can instead enhance this endpoint. In the present study we examine human cerebrospinal fluid (CSF) samples from APOE3 and APOE4 individuals and brain lysates from APOE3 and APOE4 TR mice for levels of ECM effectors that can increase matrix deposition and restrict neuroplasticity. We find that CCL5, a molecule linked to ECM deposition in liver and kidney, is increased in CSF samples from APOE4 individuals. Levels of tissue inhibitor of metalloproteinases (TIMPs), which inhibit the activity of ECM-degrading enzymes, are also increased in APOE4 CSF as well as astrocyte supernatants brain lysates from APOE4 TR mice. Importantly, as compared to APOE4/wild-type heterozygotes, APOE4/CCR5 knockout heterozygotes show reduced TIMP levels and enhanced EEG gamma power. The latter also show improved learning and memory, suggesting that the CCR5/CCL5 axis could represent a therapeutic target for APOE4 individuals.
Collapse
Affiliation(s)
- Griffin A Greco
- Georgetown University School of Medicine (GUMC), Department of Pharmacology, United States of America
| | | | - Matthew Amontree
- GUMC, United States of America; Interdisciplinary Program in Neuroscience, United States of America
| | | | - Holly Korthas
- Interdisciplinary Program in Neuroscience, United States of America
| | - Sung Hyeok Hong
- GUMC, Department of Biochemistry and Molecular & Cellular Biology, United States of America
| | | | - G William Rebeck
- Interdisciplinary Program in Neuroscience, United States of America; GUMC, Department of Neuroscience, United States of America
| | - Katherine Conant
- Interdisciplinary Program in Neuroscience, United States of America; GUMC, Department of Neuroscience, United States of America.
| |
Collapse
|
48
|
Yang L, Huh JR, Choi GB. One messenger shared by two systems: How cytokines directly modulate neurons. Curr Opin Neurobiol 2023; 80:102708. [PMID: 36947942 DOI: 10.1016/j.conb.2023.102708] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 02/16/2023] [Accepted: 02/19/2023] [Indexed: 03/24/2023]
Abstract
Cytokines are small, secreted proteins that are known for their roles in the immune system. An accumulating body of evidence indicates that cytokines also work as neuromodulators in the central nervous system (CNS). Cytokines can access the CNS through multiple routes to directly impact neurons. The neuromodulatory effects of cytokines maintain the overall homeostasis of neural networks. In addition, cytokines regulate a diverse repertoire of behaviors both at a steady state and in inflammatory conditions by acting on discrete brain regions and neural networks. In this review, we discuss recent findings that provide insight into how combinatorial codes of cytokines might mediate neuro-immune communications to orchestrate functional responses of the brain to changes in immunological milieus.
Collapse
Affiliation(s)
- Liu Yang
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jun R Huh
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Gloria B Choi
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
49
|
Aery Jones EA, Giocomo LM. Neural ensembles in navigation: From single cells to population codes. Curr Opin Neurobiol 2023; 78:102665. [PMID: 36542882 PMCID: PMC9845194 DOI: 10.1016/j.conb.2022.102665] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 10/27/2022] [Accepted: 11/21/2022] [Indexed: 12/23/2022]
Abstract
The brain can represent behaviorally relevant information through the firing of individual neurons as well as the coordinated firing of ensembles of neurons. Neurons in the hippocampus and associated cortical regions participate in a variety of types of ensembles to support navigation. These ensemble types include single cell codes, population codes, time-compressed sequences, behavioral sequences, and engrams. We present the physiological basis and behavioral relevance of ensemble firing. We discuss how these traditional definitions of ensembles can constrain or expand potential analyses due to the underlying assumptions and abstractions made. We highlight how coding can change at the ensemble level while underlying single cell codes remain intact. Finally, we present how ensemble definitions could be broadened to better understand the full complexity of the brain.
Collapse
Affiliation(s)
- Emily A Aery Jones
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| | - Lisa M Giocomo
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| |
Collapse
|
50
|
Hauser KF, Ohene-Nyako M, Knapp PE. Accelerated brain aging with opioid misuse and HIV: New insights on the role of glially derived pro-inflammation mediators and neuronal chloride homeostasis. Curr Opin Neurobiol 2023; 78:102653. [PMID: 36584655 PMCID: PMC9933139 DOI: 10.1016/j.conb.2022.102653] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 10/26/2022] [Accepted: 11/10/2022] [Indexed: 12/29/2022]
Abstract
Opioid use disorder (OUD) has become a national crisis and contributes to the spread of human immunodeficiency virus (HIV) infection. Emerging evidence and advances in experimental models, methodology, and our understanding of disease processes at the molecular and cellular levels reveal that opioids per se can directly exacerbate the pathophysiology of neuroHIV. Despite substantial inroads, the impact of OUD on the severity, development, and prognosis of neuroHIV and HIV-associated neurocognitive disorders is not fully understood. In this review, we explore current evidence that OUD and neuroHIV interact to accelerate cognitive deficits and enhance the neurodegenerative changes typically seen with aging, through their effects on neuroinflammation. We suggest new thoughts on the processes that may underlie accelerated brain aging, including dysregulation of neuronal inhibition, and highlight findings suggesting that opioids, through actions at the μ-opioid receptor, interact with HIV in the central nervous system to promote unique structural and functional comorbid deficits not seen in either OUD or neuroHIV alone.
Collapse
Affiliation(s)
- Kurt F Hauser
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298-0613, USA; Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298-0709, USA; Institute for Drug and Alcohol Studies, Virginia Commonwealth University, 203 East Cary Street, Richmond, Virginia 23298-0059, USA
| | - Michael Ohene-Nyako
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298-0613, USA
| | - Pamela E Knapp
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298-0613, USA; Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298-0709, USA; Institute for Drug and Alcohol Studies, Virginia Commonwealth University, 203 East Cary Street, Richmond, Virginia 23298-0059, USA.
| |
Collapse
|