1
|
Sasuclark AR, Watanabe M, Roshto K, Kilonzo VW, Zhang Y, Pitts MW. Selenium deficiency impedes maturation of parvalbumin interneurons, perineuronal nets, and neural network activity. Redox Biol 2025; 81:103548. [PMID: 39983343 PMCID: PMC11893315 DOI: 10.1016/j.redox.2025.103548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 01/31/2025] [Accepted: 02/11/2025] [Indexed: 02/23/2025] Open
Abstract
Selenoproteins are fundamental players in redox signaling that are essential for proper brain development and function. They are indispensable for the vitality of GABAergic parvalbumin-expressing interneurons (PVIs), a cell type characterized by fast-spiking activity and heightened rates of metabolism. During development, PVIs are preferentially encapsulated by specialized extracellular matrix structures, termed perineuronal nets (PNNs), which serve to stabilize synaptic structure and act as protective barriers against redox insults. Consequently, alterations in PVIs and PNNs are well chronicled in neuropsychiatric disease, and evidence from animal models indicates that redox imbalance during adolescence impedes their maturation. Herein, we examined the influence of selenium on maturation of neural network structure and activity in primary cortical cultures. Cultures grown in selenium-deficient media exhibited reduced antioxidant activity, impaired PNN formation, and decreased synaptic input onto PVIs at 28 days in vitro, which coincided with increased oxidative stress. Parallel studies to monitor longitudinal maturation of in vitro electrophysiological activity were conducted using microelectrode arrays (MEA). Selenium content affected the electrophysiological profile of developing cultures, as selenium-deficient cultures exhibited impairments in long-term potentiation in conjunction with reduced spike counts for both network bursts and in response to stimulation. Finally, similar PNN deficits were observed in the cortex of mice raised on a selenium-deficient diet, providing corroborative evidence for the importance of selenium in PNN development. In sum, these findings show the vital role of selenium for the development of GABAergic inhibitory circuits.
Collapse
Affiliation(s)
- Alexandru R Sasuclark
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, 96813, USA
| | - Marissa Watanabe
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, 96813, USA
| | - Kai Roshto
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, 96813, USA
| | - Victor W Kilonzo
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, 96813, USA
| | - Yiqiang Zhang
- Department of Anatomy, Biochemistry, and Physiology, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, 96813, USA; Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, 96813, USA
| | - Matthew W Pitts
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, 96813, USA.
| |
Collapse
|
2
|
Vasquez LS, Stack S, Taylor WW, Dias BG. Intergenerational Effects of Stress - A Focus on Learning and Memory. Curr Top Behav Neurosci 2025. [PMID: 40119217 DOI: 10.1007/7854_2025_578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/24/2025]
Abstract
Stress is a ubiquitous facet of life. Ranging in form (e.g., psychosocial, physical, nutritional, economic) and longevity (e.g., acute, chronic), stressors affect the biology of those directly in their line of attack. As is becoming increasingly appreciated, the pernicious effects of stress echo across generations (Dias et al. 2015; Yehuda and Lehrner 2018; Jawaid et al. 2021; Dion et al. 2022; Zhou and Ryan 2023; Dias 2024). With a focus on learning and memory, this chapter addresses how stressors derail learning and memory in the generation directly exposed to them andin future generations. To do so, with a specific emphasis on associative fear conditioning in humans and rodents, we touch upon the relevance of extinction training in the aftermath of such conditioning and the recall of such extinction training as windows into normative and disrupted learning. Next, we briefly discuss underlying neuroanatomical substrates mediating these processes. We then draw attention to influences of postnatal, in utero, and pre-conceptional stress on learning and memory across generations. Finally, we briefly outline biological factors that underlie how learning and memory is derailed by these stressors.
Collapse
Affiliation(s)
- L S Vasquez
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, USA
- Developmental Neuroscience and Neurogenetics Program, The Saban Research Institute, Los Angeles, CA, USA
| | - S Stack
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, USA
- Developmental Neuroscience and Neurogenetics Program, The Saban Research Institute, Los Angeles, CA, USA
| | - W W Taylor
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, USA
- Developmental Neuroscience and Neurogenetics Program, The Saban Research Institute, Los Angeles, CA, USA
| | - B G Dias
- Developmental Neuroscience and Neurogenetics Program, The Saban Research Institute, Los Angeles, CA, USA.
- Division of Endocrinology, Diabetes and Metabolism, Children's Hospital Los Angeles, Los Angeles, CA, USA.
- Department of Pediatrics, Keck School of Medicine of USC, Los Angeles, CA, USA.
| |
Collapse
|
3
|
Syková E, Voříšek I, Starčuk Z, Kratochvíla J, Pavlova I, Ichikawa Y, Kwok JCF, Kmoníčková E, Myronchenko S, Hromádka T, Smolek T, Avila M, Basheer N, Žilka N. Disruption of Extracellular Matrix and Perineuronal Nets Modulates Extracellular Space Volume and Geometry. J Neurosci 2025; 45:e0517242024. [PMID: 39753300 PMCID: PMC11841756 DOI: 10.1523/jneurosci.0517-24.2024] [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: 03/18/2024] [Revised: 11/18/2024] [Accepted: 11/29/2024] [Indexed: 02/21/2025] Open
Abstract
Extracellular matrix (ECM) is a network of macromolecules which has two forms-perineuronal nets (PNNs) and a diffuse ECM (dECM)-both influence brain development, synapse formation, neuroplasticity, CNS injury and progression of neurodegenerative diseases. ECM remodeling can influence extrasynaptic transmission, mediated by diffusion of neuroactive substances in the extracellular space (ECS). In this study we analyzed how disrupted PNNs and dECM influence brain diffusibility. Two months after oral treatment of rats with 4-methylumbelliferone (4-MU), an inhibitor of hyaluronan (HA) synthesis, we found downregulated staining for PNNs, HA, chondroitin sulfate proteoglycans, and glial fibrillary acidic protein. These changes were enhanced after 4 and 6 months and were reversible after a normal diet. Morphometric analysis further indicated atrophy of astrocytes. Using real-time iontophoretic method dysregulation of ECM resulted in increased ECS volume fraction α in the somatosensory cortex by 35%, from α = 0.20 in control rats to α = 0.27 after the 4-MU diet. Diffusion-weighted magnetic resonance imaging revealed a decrease of mean diffusivity and fractional anisotropy (FA) in the cortex, hippocampus, thalamus, pallidum, and spinal cord. This study shows the increase in ECS volume, a loss of FA, and changes in astrocytes due to modulation of PNNs and dECM that could affect extrasynaptic transmission, cell-to-cell communication, and neural plasticity.
Collapse
Affiliation(s)
- Eva Syková
- Institute of Neuroimmunology, Slovak Academy of Science, Bratislava 84510, Slovakia
| | - Ivan Voříšek
- Institute of Neuroimmunology, Slovak Academy of Science, Bratislava 84510, Slovakia
| | - Zenon Starčuk
- Institute of Scientific Instruments, Czech Academy of Sciences, Brno 61200, Czech Republic
| | - Jiří Kratochvíla
- Institute of Scientific Instruments, Czech Academy of Sciences, Brno 61200, Czech Republic
| | - Iveta Pavlova
- Institute of Scientific Instruments, Czech Academy of Sciences, Brno 61200, Czech Republic
| | - Yuki Ichikawa
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Jessica C F Kwok
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Eva Kmoníčková
- Department of Pharmacology, Second Faculty of Medicine, Charles University, Prague 15000, Czech Republic
| | - Svitlana Myronchenko
- Department of Pharmacology, Second Faculty of Medicine, Charles University, Prague 15000, Czech Republic
| | - Tomáš Hromádka
- Institute of Neuroimmunology, Slovak Academy of Science, Bratislava 84510, Slovakia
| | - Tomáš Smolek
- Institute of Neuroimmunology, Slovak Academy of Science, Bratislava 84510, Slovakia
| | - Martin Avila
- Institute of Histology and Embryology of Mendoza, National University of Cuyo, National Scientific and Technical Research Council, Mendoza 5500, Argentina
| | - Neha Basheer
- Institute of Neuroimmunology, Slovak Academy of Science, Bratislava 84510, Slovakia
| | - Norbert Žilka
- Institute of Neuroimmunology, Slovak Academy of Science, Bratislava 84510, Slovakia
| |
Collapse
|
4
|
Magnard R, Cheng Y, Zhou J, Province H, Thiriet N, Janak PH, Vandaele Y. Sequence termination cues drive habits via dopamine-mediated credit assignment. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.10.16.618735. [PMID: 39463939 PMCID: PMC11507917 DOI: 10.1101/2024.10.16.618735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Mesolimbic dopamine (DA) neurons are central to sequence learning and habit formation. Yet, the mechanisms by which cue-induced DA neural activity drives goal-directed or habitual sequence execution remain unknown. We designed two novel tasks to investigate how sequence initiation and termination cues influence DA-driven behavioral strategies and learning. We found that sequence initiation and termination cues differentially affect reward expectation during action sequences, with only the termination cue contributing to greater outcome devaluation insensitivity, automaticity and behavioral chunking. Mesolimbic fiber photometry recording revealed that this habit-like behavior was associated with a rapid backpropagation in DA signals from the reward to the immediately preceding cue and with attenuated DA reward prediction error signals, which reflected greater behavioral inflexibility. Finally, in absence of external cues, brief optogenetic stimulation of VTA DA neurons at sequence termination was sufficient to drive automaticity and behavioral chunking. Our results highlight the critical role of cue-evoked DA signals at sequence termination in mediating credit assignment and driving the development of habitual action sequence execution.
Collapse
Affiliation(s)
- Robin Magnard
- Department of Psychological and Brain Sciences, Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, MD
| | - Yifeng Cheng
- Department of Psychological and Brain Sciences, Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, MD
- Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, MD
| | - Joanna Zhou
- Department of Psychological and Brain Sciences, Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, MD
| | - Haley Province
- Department of Psychological and Brain Sciences, Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, MD
| | - Nathalie Thiriet
- Université de Poitiers, INSERM, U-1084, Laboratoire des Neurosciences Expérimentales et Cliniques, Poitiers, France
| | - Patricia H. Janak
- Department of Psychological and Brain Sciences, Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, MD
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD
- Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, MD
| | - Youna Vandaele
- Department of Psychological and Brain Sciences, Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, MD
- Université de Poitiers, INSERM, U-1084, Laboratoire des Neurosciences Expérimentales et Cliniques, Poitiers, France
| |
Collapse
|
5
|
Ma YN, Yang CJ, Zhang CC, Sun YX, Yao XD, Liu X, Li XX, Wang HL, Wang H, Wang T, Wang XD, Zhang C, Su YA, Li JT, Si TM. Prefrontal parvalbumin interneurons mediate CRHR1-dependent early-life stress-induced cognitive deficits in adolescent male mice. Mol Psychiatry 2024:10.1038/s41380-024-02845-6. [PMID: 39578519 DOI: 10.1038/s41380-024-02845-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 11/04/2024] [Accepted: 11/13/2024] [Indexed: 11/24/2024]
Abstract
Cognitive impairment, a core symptom of psychiatric disorders, is frequently observed in adolescents exposed to early-life stress (ES). However, the underlying neural mechanisms are unclear, and therapeutic efficacy is limited. Targeting parvalbumin-expressing interneurons (PVIs) in the medial prefrontal cortex (mPFC), we report that ES reduces mPFC PVI activity, which causally mediated ES-induced cognitive deficits in adolescent male mice through chemogenetic and optogenetic experiments. To understand the possible causes of PVI activity reduction following ES, we then demonstrated that ES upregulated corticotropin-releasing hormone (CRH) receptor 1 [CRHR1, mainly expressed in pyramidal neurons (PNs)] and reduced activity of local pyramidal neurons (PNs) and their excitatory inputs to PVIs. The subsequent genetic manipulation experiments (CRHR1 knockout, CRH overexpression, and chemogenetics) highlight that ES-induced PVI activity reduction may result from CRHR1 upregulation and PN activity downregulation and that PVIs play indispensable roles in CRHR1- or PN-mediated cognitive deficits induced by ES. These results suggest that ES-induced cognitive deficits could be attributed to the prefrontal CRHR1-PN-PVI pathway. Finally, treatment with antalarmin (a CRHR1 antagonist) and environmental enrichment successfully restored the PVI activity and cognitive deficits induced by ES. These findings reveal the neurobiological mechanisms underlying ES-induced cognitive deficits in adolescent male mice and highlight the therapeutic potentials of PVIs in stress-related cognitive deficits in adolescent individuals.
Collapse
Affiliation(s)
- Yu-Nu Ma
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China
| | - Chao-Juan Yang
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, Beihang University, Beijing, China
| | - Chen-Chen Zhang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China
| | - Ya-Xin Sun
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China
| | - Xing-Duo Yao
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China
| | - Xiao Liu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China
| | - Xue-Xin Li
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China
| | - Hong-Li Wang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China
| | - Han Wang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China
| | - Ting Wang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China
| | - Xiao-Dong Wang
- Department of Neurobiology, Key Laboratory of Medical Neurobiology of Ministry of Health of China, Zhejiang Province Key Laboratory of Neurobiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Chen Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Yun-Ai Su
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China.
| | - Ji-Tao Li
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China.
| | - Tian-Mei Si
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China.
| |
Collapse
|
6
|
Riesgo VR, Zumaski T, Willing J. Tyrosine hydroxylase expression and neuronal loss in the male and female adolescent ventral tegmental area. Neurosci Lett 2024; 841:137961. [PMID: 39227004 DOI: 10.1016/j.neulet.2024.137961] [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: 05/16/2024] [Revised: 08/20/2024] [Accepted: 08/31/2024] [Indexed: 09/05/2024]
Abstract
Adolescence is a critical period of development characterized by numerous behavioral and neuroanatomical changes. While studies of adolescent neurodevelopment typically compare adolescent age groups with young adults, there are fewer studies that assess developmental trajectories within the adolescent period. In the adolescent prefrontal cortex, some maturational changes take place linearly/chronologically, while others are associated specifically with pubertal onset. The adolescent ventral tegmental area (VTA), a primary source of forebrain dopamine, is relatively understudied during this period. In the present study, dopamine neuron number, total neuron number and tyrosine hydroxylase expression are assessed in the male and female rat VTA at three timepoints: postnatal day(P) 30 (pre-pubertal), P40 (post-pubertal for females, pre-pubertal for males) and P60 (post-pubertal). There was a non-significant trend for a reduction in total VTA neuron number between P30 and P60, but there was a significant reduction in dopamine neuron number across age. The expression of tyrosine hydroxylase did not change with age. However, in a second cohort of subjects, brain tissue was collected pre-pubertal, from recently post-pubertal males and females, and young adults. In this cohort, there was a sex-specific and transient decrease in tyrosine hydroxylase expression in recently post-pubertal males. These results suggest a selective pruning of VTA dopamine cells between early adolescence and young adulthood, while pubertal onset may coincide with a rapid maturation of these neurons. These findings may have implications for psychiatric disorders associated with dopamine dysfunction that tend to manifest during adolescence.
Collapse
Affiliation(s)
- Victoria R Riesgo
- Department of Psychology: Neural and Cognitive Sciences Program, Bowling Green State University, Bowling Green, OH 43403, United States; John Paul Scott Center for Neuroscience, Mind and Behavior, Bowling Green State University, Bowling Green, OH 43403, United States
| | - Theador Zumaski
- Department of Psychology: Neural and Cognitive Sciences Program, Bowling Green State University, Bowling Green, OH 43403, United States
| | - Jari Willing
- Department of Psychology: Neural and Cognitive Sciences Program, Bowling Green State University, Bowling Green, OH 43403, United States; John Paul Scott Center for Neuroscience, Mind and Behavior, Bowling Green State University, Bowling Green, OH 43403, United States.
| |
Collapse
|
7
|
Wong AL, Eyssalenne AN, Carter L, Therrien AS. Different Sensory Information Is Used for State Estimation when Stationary or Moving. eNeuro 2024; 11:ENEURO.0357-23.2024. [PMID: 39147580 PMCID: PMC11376429 DOI: 10.1523/eneuro.0357-23.2024] [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: 09/14/2023] [Revised: 07/19/2024] [Accepted: 08/08/2024] [Indexed: 08/17/2024] Open
Abstract
The accurate estimation of limb state is necessary for movement planning and execution. While state estimation requires both feedforward and feedback information, we focus here on the latter. Prior literature has shown that integrating visual and proprioceptive feedback improves estimates of static limb position. However, differences in visual and proprioceptive feedback delays suggest that multisensory integration could be disadvantageous when the limb is moving. We formalized this hypothesis by modeling feedback-based state estimation using the long-standing maximum likelihood estimation model of multisensory integration, which we updated to account for sensory delays. Our model predicted that the benefit of multisensory integration was largely lost when the limb was passively moving. We tested this hypothesis in a series of experiments in human subjects that compared the degree of interference created by discrepant visual or proprioceptive feedback when estimating limb position either statically at the end of the movement or dynamically at movement midpoint. In the static case, we observed significant interference: discrepant feedback in one modality systematically biased sensory estimates based on the other modality. However, no interference was seen in the dynamic case: participants could ignore sensory feedback from one modality and accurately reproduce the motion indicated by the other modality. Together, these findings suggest that the sensory feedback used to compute a state estimate differs depending on whether the limb is stationary or moving. While the former may tend toward multimodal integration, the latter is more likely to be based on feedback from a single sensory modality.
Collapse
Affiliation(s)
- Aaron L Wong
- Moss Rehabilitation Research Institute, Thomas Jefferson University, Elkins Park, Pennsylvania 19027
- Department of Rehabilitation Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania 19107
| | - Alyssa N Eyssalenne
- Moss Rehabilitation Research Institute, Thomas Jefferson University, Elkins Park, Pennsylvania 19027
| | - Luke Carter
- Moss Rehabilitation Research Institute, Thomas Jefferson University, Elkins Park, Pennsylvania 19027
| | - Amanda S Therrien
- Moss Rehabilitation Research Institute, Thomas Jefferson University, Elkins Park, Pennsylvania 19027
- Department of Rehabilitation Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania 19107
| |
Collapse
|
8
|
van ‘t Spijker H, Richter JD. FMRP regulation of aggrecan mRNA translation controls perineuronal net development. J Neurochem 2024; 168:1909-1922. [PMID: 38225196 PMCID: PMC11247136 DOI: 10.1111/jnc.16048] [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: 09/23/2023] [Revised: 12/19/2023] [Accepted: 01/01/2024] [Indexed: 01/17/2024]
Abstract
Perineuronal nets (PNNs) are mesh-like structures on the surfaces of parvalbumin-expressing inhibitory and other neurons, and consist of proteoglycans such as aggrecan, brevican, and neurocan. PNNs regulate the Excitatory/Inhibitory (E/I) balance in the brain and are formed at the closure of critical periods of plasticity during development. PNN formation is disrupted in Fragile X Syndrome, which is caused by silencing of the fragile X messenger ribonucleoprotein 1 (Fmr1) gene and loss of its protein product FMRP. FXS is characterized by impaired synaptic plasticity resulting in neuronal hyperexcitability and E/I imbalance. Here, we investigate how PNN formation is altered in FXS. PNNs are reduced in Fmr1 KO mouse brain when examined by staining for the lectin Wisteria floribunda agglutin (WFA) and aggrecan. Examination of PNNs by WFA staining at P14 and P42 in the hippocampus, somatosensory cortex, and retrosplenial cortex shows that they were reduced in these brain regions at P14 but mostly less so at P42 in Fmr1 KO mice. However, some differential FMRP regulation of PNN development in these brain regions persists, perhaps caused by asynchrony in PNN development between brain regions in wild-type animals. During development, aggrecan PNN levels in the brain were reduced in all brain regions in Fmr1 KO mice. Aggrecan mRNA levels were unchanged at these times, suggesting that FMRP is normally an activator of aggrecan mRNA translation. This hypothesis is buttressed by the observations that FMRP binds aggrecan mRNA and that ribosome profiling data show that aggrecan mRNA is associated with reduced numbers of ribosomes in Fmr1 KO mouse brain, indicating reduced translational efficiency. Moreover, aggrecan mRNA poly(A) tail length is also reduced in Fmr1 KO mouse brain, suggesting a relationship between polyadenylation and translational control. We propose a model where FMRP modulates PNN formation through translational up-regulation of aggrecan mRNA polyadenylation and translation.
Collapse
Affiliation(s)
| | - Joel D. Richter
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester MA 01605 USA
| |
Collapse
|
9
|
da Silva MDV, Bacarin CC, Machado CCA, Franciosi A, Mendes JDDL, da Silva Watanabe P, Miqueloto CA, Fattori V, Albarracin OYE, Verri WA, Aktar R, Peiris M, Aziz Q, Blackshaw LA, de Almeida Araújo EJ. Descriptive study of perineuronal net in enteric nervous system of humans and mice. J Neurochem 2024; 168:1956-1972. [PMID: 38970456 DOI: 10.1111/jnc.16159] [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: 10/15/2023] [Revised: 05/17/2024] [Accepted: 06/11/2024] [Indexed: 07/08/2024]
Abstract
Perineuronal nets (PNN) are highly specialized structures of the extracellular matrix around specific groups of neurons in the central nervous system (CNS). They play functions related to optimizing physiological processes and protection neurons against harmful stimuli. Traditionally, their existence was only described in the CNS. However, there was no description of the presence and composition of PNN in the enteric nervous system (ENS) until now. Thus, our aim was to demonstrate the presence and characterize the components of the PNN in the enteric nervous system. Samples of intestinal tissue from mice and humans were analyzed by RT-PCR and immunofluorescence assays. We used a marker (Wisteria floribunda agglutinin) considered as standard for detecting the presence of PNN in the CNS and antibodies for labeling members of the four main PNN-related protein families in the CNS. Our results demonstrated the presence of components of PNN in the ENS of both species; however its molecular composition is species-specific.
Collapse
Affiliation(s)
- Matheus Deroco Veloso da Silva
- Laboratory of Enteric Neuroscience, Department of Histology, State University of Londrina, Londrina, Paraná, Brazil
- Laboratory of Pain, Inflammation, Neuropathy and Cancer, Department of Pathology, State University of Londrina, Londrina, Paraná, Brazil
| | - Cristiano Correia Bacarin
- Laboratory of Enteric Neuroscience, Department of Histology, State University of Londrina, Londrina, Paraná, Brazil
| | | | - Anelise Franciosi
- Laboratory of Enteric Neuroscience, Department of Histology, State University of Londrina, Londrina, Paraná, Brazil
- Laboratory of Pain, Inflammation, Neuropathy and Cancer, Department of Pathology, State University of Londrina, Londrina, Paraná, Brazil
| | - Joana Darc de Lima Mendes
- Laboratory of Enteric Neuroscience, Department of Histology, State University of Londrina, Londrina, Paraná, Brazil
| | - Paulo da Silva Watanabe
- Laboratory of Enteric Neuroscience, Department of Histology, State University of Londrina, Londrina, Paraná, Brazil
| | - Carlos Alberto Miqueloto
- Laboratory of Enteric Neuroscience, Department of Histology, State University of Londrina, Londrina, Paraná, Brazil
| | - Victor Fattori
- Laboratory of Pain, Inflammation, Neuropathy and Cancer, Department of Pathology, State University of Londrina, Londrina, Paraná, Brazil
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Waldiceu A Verri
- Laboratory of Pain, Inflammation, Neuropathy and Cancer, Department of Pathology, State University of Londrina, Londrina, Paraná, Brazil
| | - Rubina Aktar
- Wingate Institute for Neurogastroenterology, Queen Mary University of London, London, UK
| | - Madusha Peiris
- Wingate Institute for Neurogastroenterology, Queen Mary University of London, London, UK
| | - Qasim Aziz
- Wingate Institute for Neurogastroenterology, Queen Mary University of London, London, UK
| | - L Ashley Blackshaw
- Wingate Institute for Neurogastroenterology, Queen Mary University of London, London, UK
| | | |
Collapse
|
10
|
Rahimian R, Belliveau C, Simard S, Turecki G, Mechawar N. Perineuronal Net Alterations Following Early-Life Stress: Are Microglia Pulling Some Strings? Biomolecules 2024; 14:1087. [PMID: 39334854 PMCID: PMC11430691 DOI: 10.3390/biom14091087] [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: 07/31/2024] [Revised: 08/23/2024] [Accepted: 08/27/2024] [Indexed: 09/30/2024] Open
Abstract
The extracellular matrix plays a key role in synapse formation and in the modulation of synaptic function in the central nervous system. Recent investigations have revealed that microglia, the resident immune cells of the brain, are involved in extracellular matrix remodeling under both physiological and pathological conditions. Moreover, the dysregulation of both innate immune responses and the extracellular matrix has been documented in stress-related psychopathologies as well as in relation to early-life stress. However, the dynamics of microglial regulation of the ECM and how it can be impacted by early-life adversity have been understudied. This brief review provides an overview of the recent literature on this topic, drawing from both animal model and human post mortem studies. Direct and indirect mechanisms through which microglia may regulate the extracellular matrix-including perineuronal nets-are presented and discussed in light of the interactions with other cell types.
Collapse
Affiliation(s)
- Reza Rahimian
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Verdun, QC H4H 1R3, Canada; (R.R.); (C.B.); (S.S.); (G.T.)
- Department of Psychiatry, McGill University, Montreal, QC H3A 0G4, Canada
| | - Claudia Belliveau
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Verdun, QC H4H 1R3, Canada; (R.R.); (C.B.); (S.S.); (G.T.)
- Integrated Program in Neuroscience, McGill University, Montreal, QC H3A 0G4, Canada
| | - Sophie Simard
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Verdun, QC H4H 1R3, Canada; (R.R.); (C.B.); (S.S.); (G.T.)
- Integrated Program in Neuroscience, McGill University, Montreal, QC H3A 0G4, Canada
| | - Gustavo Turecki
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Verdun, QC H4H 1R3, Canada; (R.R.); (C.B.); (S.S.); (G.T.)
- Department of Psychiatry, McGill University, Montreal, QC H3A 0G4, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC H3A 0G4, Canada
| | - Naguib Mechawar
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Verdun, QC H4H 1R3, Canada; (R.R.); (C.B.); (S.S.); (G.T.)
- Department of Psychiatry, McGill University, Montreal, QC H3A 0G4, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC H3A 0G4, Canada
| |
Collapse
|
11
|
Carey SD, Conant K, Maguire-Zeiss KA. Short-term exposure to HIV Tat induces glial activation and changes in perineuronal nets. Eur J Neurosci 2024; 60:4303-4316. [PMID: 38844747 DOI: 10.1111/ejn.16427] [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: 12/29/2023] [Revised: 04/30/2024] [Accepted: 05/14/2024] [Indexed: 08/07/2024]
Abstract
Despite widespread use of combination antiretroviral therapy (cART), there remains a subset of individuals who display cognitive impairment broadly known as HIV-associated neurocognitive disorder (HAND). Interestingly, HIV-infected cells continuously release the HIV-1 protein Tat even in the presence of cART. Persistent exposure to Tat is proposed to increase both neuroinflammation and neurotoxicity. In vitro evidence shows that matrix metalloproteinases (MMPs) are among the neuroinflammatory molecules induced by Tat, which are known to disrupt specialized neuronal extracellular matrix structures called perineuronal nets (PNNs). PNNs predominantly surround parvalbumin interneurons and help to buffer these cells from oxidant stress and to independently increase their excitability. In order to better understand the link between short-term exposure to Tat, neuroinflammation, and PNNs, we explored the direct effects of Tat on glial cells and neurons. Herein, we report that in mixed glial cultures, Tat directly increases the expression of proinflammatory molecules, including MMP-9. Moreover, direct injection of Tat protein into mouse hippocampus increases the expression of astrocyte and microglia markers as well as MMP-9. The number of PNNs is decreased following Tat exposure, followed later by decreased numbers of hippocampal parvalbumin-expressing neurons. In older mice, Tat induced significant increases in the gene expression of proinflammatory molecules including markers of gliosis, MMPs and complement system proteins. Taken together, these data support a direct effect of Tat on glial-derived MMP expression subsequently affecting PNNs and neuronal health, with older mice more susceptible to Tat-induced inflammation.
Collapse
Affiliation(s)
- Sean D Carey
- Department of Biology, Georgetown University, Washington DC, United States
| | - Katherine Conant
- Department of Neuroscience, Georgetown University School of Medicine, Washington DC, United States
| | - Kathleen A Maguire-Zeiss
- Department of Neuroscience, Georgetown University School of Medicine, Washington DC, United States
| |
Collapse
|
12
|
Schreurs BG, O'Dell DE, Wang D. The Role of Cerebellar Intrinsic Neuronal Excitability, Synaptic Plasticity, and Perineuronal Nets in Eyeblink Conditioning. BIOLOGY 2024; 13:200. [PMID: 38534469 DOI: 10.3390/biology13030200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 02/29/2024] [Accepted: 03/19/2024] [Indexed: 03/28/2024]
Abstract
Evidence is strong that, in addition to fine motor control, there is an important role for the cerebellum in cognition and emotion. The deep nuclei of the mammalian cerebellum also contain the highest density of perineural nets-mesh-like structures that surround neurons-in the brain, and it appears there may be a connection between these nets and cognitive processes, particularly learning and memory. Here, we review how the cerebellum is involved in eyeblink conditioning-a particularly well-understood form of learning and memory-and focus on the role of perineuronal nets in intrinsic membrane excitability and synaptic plasticity that underlie eyeblink conditioning. We explore the development and role of perineuronal nets and the in vivo and in vitro evidence that manipulations of the perineuronal net in the deep cerebellar nuclei affect eyeblink conditioning. Together, these findings provide evidence of an important role for perineuronal net in learning and memory.
Collapse
Affiliation(s)
- Bernard G Schreurs
- Department of Neuroscience, West Virginia University, Morgantown, WV 26505, USA
| | - Deidre E O'Dell
- Department of Biology, Earth and Environmental Sciences, Pennsylvania Western (PennWest) University, California, PA 15419, USA
| | - Desheng Wang
- Department of Neuroscience, West Virginia University, Morgantown, WV 26505, USA
| |
Collapse
|
13
|
Li X, Wu X, Lu T, Kuang C, Si Y, Zheng W, Li Z, Xue Y. Perineuronal Nets in the CNS: Architects of Memory and Potential Therapeutic Target in Neuropsychiatric Disorders. Int J Mol Sci 2024; 25:3412. [PMID: 38542386 PMCID: PMC10970535 DOI: 10.3390/ijms25063412] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/08/2024] [Accepted: 03/12/2024] [Indexed: 11/11/2024] Open
Abstract
The extracellular matrix (ECM) within the brain possesses a distinctive composition and functionality, influencing a spectrum of physiological and pathological states. Among its constituents, perineuronal nets (PNNs) are unique ECM structures that wrap around the cell body of many neurons and extend along their dendrites within the central nervous system (CNS). PNNs are pivotal regulators of plasticity in CNS, both during development and adulthood stages. Characterized by their condensed glycosaminoglycan-rich structures and heterogeneous molecular composition, PNNs not only offer neuroprotection but also participate in signal transduction, orchestrating neuronal activity and plasticity. Interfering with the PNNs in adult animals induces the reactivation of critical period plasticity, permitting modifications in neuronal connections and promoting the recovery of neuroplasticity following spinal cord damage. Interestingly, in the adult brain, PNN expression is dynamic, potentially modulating plasticity-associated states. Given their multifaceted roles, PNNs have emerged as regulators in the domains of learning, memory, addiction behaviors, and other neuropsychiatric disorders. In this review, we aimed to address how PNNs contribute to the memory processes in physiological and pathological conditions.
Collapse
Affiliation(s)
- Xue Li
- National Institute on Drug Dependence, Peking University, Beijing 100191, China; (X.L.); (T.L.); (Y.S.); (Z.L.)
- School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Xianwen Wu
- Department of Laboratory Animal Sciences, Peking University Health Sciences Center, Beijing 100191, China;
| | - Tangsheng Lu
- National Institute on Drug Dependence, Peking University, Beijing 100191, China; (X.L.); (T.L.); (Y.S.); (Z.L.)
- School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Chenyan Kuang
- College of Forensic Medicine, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Medical University, Shijiazhuang 050017, China;
| | - Yue Si
- National Institute on Drug Dependence, Peking University, Beijing 100191, China; (X.L.); (T.L.); (Y.S.); (Z.L.)
- School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Wei Zheng
- Peking-Tsinghua Centre for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China;
| | - Zhonghao Li
- National Institute on Drug Dependence, Peking University, Beijing 100191, China; (X.L.); (T.L.); (Y.S.); (Z.L.)
- School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Yanxue Xue
- National Institute on Drug Dependence, Peking University, Beijing 100191, China; (X.L.); (T.L.); (Y.S.); (Z.L.)
| |
Collapse
|
14
|
Dong Y, Zhao K, Qin X, Du G, Gao L. The mechanisms of perineuronal net abnormalities in contributing aging and neurological diseases. Ageing Res Rev 2023; 92:102092. [PMID: 37839757 DOI: 10.1016/j.arr.2023.102092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 09/29/2023] [Accepted: 10/10/2023] [Indexed: 10/17/2023]
Abstract
The perineuronal net (PNN) is a highly latticed extracellular matrix in the central nervous system, which is composed of hyaluronic acid, proteoglycan, hyaluronan and proteoglycan link protein (Hapln), and tenascin. PNN is predominantly distributed in GABAergic interneurons expressing Parvalbumin (PV) and plays a critical role in synaptic function, learning and memory, oxidative stress, and inflammation. In addition, PNN's structure and function are also modulated by a variety of factors, including protein tyrosine phosphatase σ (PTPσ), orthodenticle homeo-box 2 (Otx2), and erb-b2 receptor tyrosine kinase 4 (ErbB4). Glycosaminoglycan (GAG), a component of proteoglycan, also influences PNN through its sulfate mode. PNN undergoes abnormal changes during aging and in various neurological diseases, such as Alzheimer's disease, Parkinson's disease, schizophrenia, autism spectrum disorder, and multiple sclerosis. Nevertheless, there is limited report on the relationship between PNN and aging or age-related neurological diseases. This review elaborates on the mechanisms governing PNN regulation and summarizes how PNN abnormalities contribute to aging and neurological diseases, offering insights for potential treatments.
Collapse
Affiliation(s)
- Yixiao Dong
- Modern Research Center for Traditional Chinese Medicine, the Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan, China; Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, Taiyuan, China
| | - Kunkun Zhao
- Modern Research Center for Traditional Chinese Medicine, the Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan, China; Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, Taiyuan, China
| | - Xuemei Qin
- Modern Research Center for Traditional Chinese Medicine, the Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan, China; Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, Taiyuan, China
| | - Guanhua Du
- Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Li Gao
- Modern Research Center for Traditional Chinese Medicine, the Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan, China; Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, Taiyuan, China.
| |
Collapse
|
15
|
Štepánková K, Chudíčková M, Šimková Z, Martinez-Varea N, Kubinová Š, Urdzíková LM, Jendelová P, Kwok JCF. Low oral dose of 4-methylumbelliferone reduces glial scar but is insufficient to induce functional recovery after spinal cord injury. Sci Rep 2023; 13:19183. [PMID: 37932336 PMCID: PMC10628150 DOI: 10.1038/s41598-023-46539-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Accepted: 11/02/2023] [Indexed: 11/08/2023] Open
Abstract
Spinal cord injury (SCI) induces the upregulation of chondroitin sulfate proteoglycans (CSPGs) at the glial scar and inhibits neuroregeneration. Under normal physiological condition, CSPGs interact with hyaluronan (HA) and other extracellular matrix on the neuronal surface forming a macromolecular structure called perineuronal nets (PNNs) which regulate neuroplasticity. 4-methylumbelliferone (4-MU) is a known inhibitor for HA synthesis but has not been tested in SCI. We first tested the effect of 4-MU in HA reduction in uninjured rats. After 8 weeks of 4-MU administration at a dose of 1.2 g/kg/day, we have not only observed a reduction of HA in the uninjured spinal cords but also a down-regulation of CS glycosaminoglycans (CS-GAGs). In order to assess the effect of 4-MU in chronic SCI, six weeks after Th8 spinal contusion injury, rats were fed with 4-MU or placebo for 8 weeks in combination with daily treadmill rehabilitation for 16 weeks to promote neuroplasticity. 4-MU treatment reduced the HA synthesis by astrocytes around the lesion site and increased sprouting of 5-hydroxytryptamine fibres into ventral horns. However, the current dose was not sufficient to suppress CS-GAG up-regulation induced by SCI. Further adjustment on the dosage will be required to benefit functional recovery after SCI.
Collapse
Affiliation(s)
- Kateřina Štepánková
- Institute of Experimental Medicine, Czech Academy of Sciences, Vídeňská, 1083, Prague, Czech Republic.
- Department of Neuroscience, Charles University, Second Faculty of Medicine, 15006, Prague, Czech Republic.
| | - Milada Chudíčková
- Institute of Experimental Medicine, Czech Academy of Sciences, Vídeňská, 1083, Prague, Czech Republic
| | - Zuzana Šimková
- Institute of Experimental Medicine, Czech Academy of Sciences, Vídeňská, 1083, Prague, Czech Republic
| | - Noelia Martinez-Varea
- Institute of Experimental Medicine, Czech Academy of Sciences, Vídeňská, 1083, Prague, Czech Republic
- Department of Neuroscience, Charles University, Second Faculty of Medicine, 15006, Prague, Czech Republic
| | - Šárka Kubinová
- Institute of Experimental Medicine, Czech Academy of Sciences, Vídeňská, 1083, Prague, Czech Republic
- Institute of Physics, Czech Academy of Sciences, 182 21, Prague, Czech Republic
| | - Lucia Machová Urdzíková
- Institute of Experimental Medicine, Czech Academy of Sciences, Vídeňská, 1083, Prague, Czech Republic.
- Department of Neuroscience, Charles University, Second Faculty of Medicine, 15006, Prague, Czech Republic.
| | - Pavla Jendelová
- Institute of Experimental Medicine, Czech Academy of Sciences, Vídeňská, 1083, Prague, Czech Republic.
- Department of Neuroscience, Charles University, Second Faculty of Medicine, 15006, Prague, Czech Republic.
| | - Jessica C F Kwok
- Institute of Experimental Medicine, Czech Academy of Sciences, Vídeňská, 1083, Prague, Czech Republic.
- Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK.
| |
Collapse
|
16
|
Melrose J. Hyaluronan hydrates and compartmentalises the CNS/PNS extracellular matrix and provides niche environments conducive to the optimisation of neuronal activity. J Neurochem 2023; 166:637-653. [PMID: 37492973 DOI: 10.1111/jnc.15915] [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: 05/26/2023] [Revised: 06/27/2023] [Accepted: 07/03/2023] [Indexed: 07/27/2023]
Abstract
The central nervous system/peripheral nervous system (CNS/PNS) extracellular matrix is a dynamic and highly interactive space-filling, cell-supportive, matrix-stabilising, hydrating entity that creates and maintains tissue compartments to facilitate regional ionic micro-environments and micro-gradients that promote optimal neural cellular activity. The CNS/PNS does not contain large supportive collagenous and elastic fibrillar networks but is dominated by a high glycosaminoglycan content, predominantly hyaluronan (HA) and collagen is restricted to the brain microvasculature, blood-brain barrier, neuromuscular junction and meninges dura, arachnoid and pia mater. Chondroitin sulphate-rich proteoglycans (lecticans) interactive with HA have stabilising roles in perineuronal nets and contribute to neural plasticity, memory and cognitive processes. Hyaluronan also interacts with sialoproteoglycan associated with cones and rods (SPACRCAN) to stabilise the interphotoreceptor matrix and has protective properties that ensure photoreceptor viability and function is maintained. HA also regulates myelination/re-myelination in neural networks. HA fragmentation has been observed in white matter injury, multiple sclerosis, and traumatic brain injury. HA fragments (2 × 105 Da) regulate oligodendrocyte precursor cell maturation, myelination/remyelination, and interact with TLR4 to initiate signalling cascades that mediate myelin basic protein transcription. HA and its fragments have regulatory roles over myelination which ensure high axonal neurotransduction rates are maintained in neural networks. Glioma is a particularly invasive brain tumour with extremely high mortality rates. HA, CD44 and RHAMM (receptor for HA-mediated motility) HA receptors are highly expressed in this tumour. Conventional anti-glioma drug treatments have been largely ineffective and surgical removal is normally not an option. CD44 and RHAMM glioma HA receptors can potentially be used to target gliomas with PEP-1, a cell-penetrating HA-binding peptide. PEP-1 can be conjugated to a therapeutic drug; such drug conjugates have successfully treated dense non-operative tumours in other tissues, therefore similar applications warrant exploration as potential anti-glioma treatments.
Collapse
Affiliation(s)
- James Melrose
- Raymond Purves Bone and Joint Research Laboratory, Kolling Institute, Northern Sydney Local Health District, St. Leonards, New South Wales, Australia
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, New South Wales, Australia
- Sydney Medical School, Northern, The University of Sydney, Camperdown, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Royal North Shore Hospital, St. Leonards, New South Wales, Australia
| |
Collapse
|
17
|
Engel K, Lee HN, Tewari BP, Lewkowicz AP, Ireland DDC, Manangeeswaran M, Verthelyi D. Neonatal Zika virus infection causes transient perineuronal net degradation. Front Cell Neurosci 2023; 17:1187425. [PMID: 37496706 PMCID: PMC10366369 DOI: 10.3389/fncel.2023.1187425] [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: 03/16/2023] [Accepted: 06/21/2023] [Indexed: 07/28/2023] Open
Abstract
Perineuronal nets (PNNs) form a specialized extracellular matrix that predominantly surrounds parvalbumin (PV)-expressing GABAergic inhibitory interneurons and help regulate neuronal activity. Their formation early in the postnatal period is regulated by neuronal signaling and glial activation raising concerns that part of the long-term effects ascribed to perinatal viral infections could be mediated by altered PNN formation. Previously, we developed a model of neonatal Zika virus (ZIKV) infection where mice have lifelong neurological sequelae that includes motor disfunction and reduced anxiety coupled with a persistent low-grade expression in proinflammatory markers despite resolving the acute infection. Here, we demonstrate that ZIKV infection to P1 neonatal mice results in a reduction of PNN formation during the acute disease with significant reduction in Wisteria floribunda agglutinin (WFA) staining at the peak of infection [15 days post infection (dpi)] that persisted after the symptoms resolved (30 dpi). At 60 dpi, when there is residual inflammation in the CNS, the number of WFA+ cells and the level of WFA staining as well as levels of aggrecan and brevican in the brains of convalescent mice were not different from those in uninfected controls, however, there was increased frequency of PNNs with an immature phenotype. Over time the impact of the perinatal infection became less evident and there were no clear differences in PNN morphology between the groups at 1 year post infection. Of note, the reduction in PNNs during acute ZIKV infection was not associated with decreased mRNA levels of aggrecan or brevican, but increased levels of degraded aggrecan and brevican indicating increased PNN degradation. These changes were associated with increased expression of matrix metalloproteinase 12 (MMP12) and MMP19, but not MMP9, a disintegrin and metalloproteinase with thrombospondin motifs 4 (ADAMTS4) or ADAMTS5. Together our findings indicate that infection at the time of PNN development interferes with PNN formation, but the nets can reform once the infection and inflammation subside.
Collapse
Affiliation(s)
- Kaliroi Engel
- Laboratory of Immunology, Center of Excellence in Infectious Disease and Inflammation, Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| | - Ha-Na Lee
- Laboratory of Immunology, Center of Excellence in Infectious Disease and Inflammation, Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| | - Bhanu P. Tewari
- Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA, United States
| | - Aaron P. Lewkowicz
- Laboratory of Immunology, Center of Excellence in Infectious Disease and Inflammation, Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| | - Derek D. C. Ireland
- Laboratory of Immunology, Center of Excellence in Infectious Disease and Inflammation, Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| | - Mohanraj Manangeeswaran
- Laboratory of Immunology, Center of Excellence in Infectious Disease and Inflammation, Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| | - Daniela Verthelyi
- Laboratory of Immunology, Center of Excellence in Infectious Disease and Inflammation, Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| |
Collapse
|
18
|
Ren J, Flamant F. Thyroid hormone as a temporal switch in mouse development. Eur Thyroid J 2023; 12:e220225. [PMID: 36715693 PMCID: PMC10083660 DOI: 10.1530/etj-22-0225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 01/30/2023] [Indexed: 01/31/2023] Open
Abstract
Thyroid hormones are known to trigger metamorphosis in an amphibian. This review discusses the hypothesis according to which they act in a similar manner to synchronize the post-natal development of mice, using brain, brown adipose tissue, and heart as examples.
Collapse
Affiliation(s)
- Juan Ren
- ENS de Lyon, INRAE, CNRS, Institut de Génomique Fonctionnelle de Lyon, Lyon, France
| | - Frédéric Flamant
- ENS de Lyon, INRAE, CNRS, Institut de Génomique Fonctionnelle de Lyon, Lyon, France
| |
Collapse
|
19
|
Hanssen KØ, Malthe-Sørenssen A. Perineuronal nets restrict transport near the neuron surface: A coarse-grained molecular dynamics study. Front Comput Neurosci 2022; 16:967735. [DOI: 10.3389/fncom.2022.967735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 11/02/2022] [Indexed: 11/18/2022] Open
Abstract
Perineuronal nets (PNNs) are mesh-like extracellular matrix structures that wrap around certain neurons in the central nervous system. They are hypothesized to stabilize memories in the brain and act as a barrier between cell and extracellular space. As a means to study the impact of PNNs on diffusion, the nets were approximated by negatively charged polymer brushes and simulated by coarse-grained molecular dynamics. Diffusion constants of single neutral and single charged particles were obtained in directions parallel and perpendicular to the brush substrate. The results for the neutral particle were compared to different theories of diffusion in a heuristic manner. Diffusion was found to be considerably reduced for brush spacings smaller than 10 nm, with a pronounced anisotropy for dense brushes. The exact dynamics of the chains was found to have a negligible impact on particle diffusion. The resistance of the brush proved small compared to typical values of the membrane resistance of a neuron, indicating that PNNs likely contribute little to the total resistance of an enwrapped neuron.
Collapse
|
20
|
Tewari BP, Chaunsali L, Prim CE, Sontheimer H. A glial perspective on the extracellular matrix and perineuronal net remodeling in the central nervous system. Front Cell Neurosci 2022; 16:1022754. [PMID: 36339816 PMCID: PMC9630365 DOI: 10.3389/fncel.2022.1022754] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 09/23/2022] [Indexed: 11/18/2022] Open
Abstract
A structural scaffold embedding brain cells and vasculature is known as extracellular matrix (ECM). The physical appearance of ECM in the central nervous system (CNS) ranges from a diffused, homogeneous, amorphous, and nearly omnipresent matrix to highly organized distinct morphologies such as basement membranes and perineuronal nets (PNNs). ECM changes its composition and organization during development, adulthood, aging, and in several CNS pathologies. This spatiotemporal dynamic nature of the ECM and PNNs brings a unique versatility to their functions spanning from neurogenesis, cell migration and differentiation, axonal growth, and pathfinding cues, etc., in the developing brain, to stabilizing synapses, neuromodulation, and being an active partner of tetrapartite synapses in the adult brain. The malleability of ECM and PNNs is governed by both intrinsic and extrinsic factors. Glial cells are among the major extrinsic factors that facilitate the remodeling of ECM and PNN, thereby acting as key regulators of diverse functions of ECM and PNN in health and diseases. In this review, we discuss recent advances in our understanding of PNNs and how glial cells are central to ECM and PNN remodeling in normal and pathological states of the CNS.
Collapse
|
21
|
Mueller-Buehl C, Reinhard J, Roll L, Bader V, Winklhofer KF, Faissner A. Brevican, Neurocan, Tenascin-C, and Tenascin-R Act as Important Regulators of the Interplay Between Perineuronal Nets, Synaptic Integrity, Inhibitory Interneurons, and Otx2. Front Cell Dev Biol 2022; 10:886527. [PMID: 35721494 PMCID: PMC9201762 DOI: 10.3389/fcell.2022.886527] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/11/2022] [Indexed: 11/13/2022] Open
Abstract
Fast-spiking parvalbumin interneurons are critical for the function of mature cortical inhibitory circuits. Most of these neurons are enwrapped by a specialized extracellular matrix (ECM) structure called perineuronal net (PNN), which can regulate their synaptic input. In this study, we investigated the relationship between PNNs, parvalbumin interneurons, and synaptic distribution on these cells in the adult primary visual cortex (V1) of quadruple knockout mice deficient for the ECM molecules brevican, neurocan, tenascin-C, and tenascin-R. We used super-resolution structured illumination microscopy (SIM) to analyze PNN structure and associated synapses. In addition, we examined parvalbumin and calretinin interneuron populations. We observed a reduction in the number of PNN-enwrapped cells and clear disorganization of the PNN structure in the quadruple knockout V1. This was accompanied by an imbalance of inhibitory and excitatory synapses with a reduction of inhibitory and an increase of excitatory synaptic elements along the PNNs. Furthermore, the number of parvalbumin interneurons was reduced in the quadruple knockout, while calretinin interneurons, which do not wear PNNs, did not display differences in number. Interestingly, we found the transcription factor Otx2 homeoprotein positive cell population also reduced. Otx2 is crucial for parvalbumin interneuron and PNN maturation, and a positive feedback loop between these parameters has been described. Collectively, these data indicate an important role of brevican, neurocan, tenascin-C, and tenascin-R in regulating the interplay between PNNs, inhibitory interneurons, synaptic distribution, and Otx2 in the V1.
Collapse
Affiliation(s)
- Cornelius Mueller-Buehl
- Department of Cell Morphology and Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr University Bochum, Bochum, Germany
| | - Jacqueline Reinhard
- Department of Cell Morphology and Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr University Bochum, Bochum, Germany
| | - Lars Roll
- Department of Cell Morphology and Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr University Bochum, Bochum, Germany
| | - Verian Bader
- Department of Molecular Cell Biology, Institute of Biochemistry and Pathobiochemistry, Ruhr University Bochum, Bochum, Germany
- Department of Biochemistry of Neurodegenerative Diseases, Institute of Biochemistry and Pathobiochemistry, Ruhr University Bochum, Bochum, Germany
| | - Konstanze F. Winklhofer
- Department of Molecular Cell Biology, Institute of Biochemistry and Pathobiochemistry, Ruhr University Bochum, Bochum, Germany
- Cluster of Excellence RESOLV, Ruhr University Bochum, Bochum, Germany
| | - Andreas Faissner
- Department of Cell Morphology and Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
22
|
Sánchez-Ventura J, Lane MA, Udina E. The Role and Modulation of Spinal Perineuronal Nets in the Healthy and Injured Spinal Cord. Front Cell Neurosci 2022; 16:893857. [PMID: 35669108 PMCID: PMC9163449 DOI: 10.3389/fncel.2022.893857] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 04/21/2022] [Indexed: 11/13/2022] Open
Abstract
Rather than being a stable scaffold, perineuronal nets (PNNs) are a dynamic and specialized extracellular matrix involved in plasticity modulation. They have been extensively studied in the brain and associated with neuroprotection, ionic buffering, and neural maturation. However, their biological function in the spinal cord and the effects of disrupting spinal PNNs remain elusive. The goal of this review is to summarize the current knowledge of spinal PNNs and their potential in pathological conditions such as traumatic spinal cord injury (SCI). We also highlighted interventions that have been used to modulate the extracellular matrix after SCI, targeting the glial scar and spinal PNNs, in an effort to promote regeneration and stabilization of the spinal circuits, respectively. These concepts are discussed in the framework of developmental and neuroplastic changes in PNNs, drawing similarities between immature and denervated neurons after an SCI, which may provide a useful context for future SCI research.
Collapse
Affiliation(s)
- Judith Sánchez-Ventura
- Department of Cell Biology, Physiology and Immunology, Institute of Neuroscience, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Bellaterra, Spain
| | - Michael A. Lane
- Department of Neurobiology and Anatomy, College of Medicine, Drexel University, Philadelphia, PA, United States
- The Marion Murray Spinal Cord Research Center, College of Medicine, Drexel University, Philadelphia, PA, United States
| | - Esther Udina
- Department of Cell Biology, Physiology and Immunology, Institute of Neuroscience, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Bellaterra, Spain
- *Correspondence: Esther Udina
| |
Collapse
|
23
|
Moretto E, Stuart S, Surana S, Vargas JNS, Schiavo G. The Role of Extracellular Matrix Components in the Spreading of Pathological Protein Aggregates. Front Cell Neurosci 2022; 16:844211. [PMID: 35573838 PMCID: PMC9100790 DOI: 10.3389/fncel.2022.844211] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 03/08/2022] [Indexed: 11/23/2022] Open
Abstract
Several neurodegenerative diseases are characterized by the accumulation of aggregated misfolded proteins. These pathological agents have been suggested to propagate in the brain via mechanisms similar to that observed for the prion protein, where a misfolded variant is transferred from an affected brain region to a healthy one, thereby inducing the misfolding and/or aggregation of correctly folded copies. This process has been characterized for several proteins, such as α-synuclein, tau, amyloid beta (Aβ) and less extensively for huntingtin and TDP-43. α-synuclein, tau, TDP-43 and huntingtin are intracellular proteins, and their aggregates are located in the cytosol or nucleus of neurons. They have been shown to spread between cells and this event occurs, at least partially, via secretion of these protein aggregates in the extracellular space followed by re-uptake. Conversely, Aβ aggregates are found mainly extracellularly, and their spreading occurs in the extracellular space between brain regions. Due to the inherent nature of their spreading modalities, these proteins are exposed to components of the extracellular matrix (ECM), including glycans, proteases and core matrix proteins. These ECM components can interact with or process pathological misfolded proteins, potentially changing their properties and thus regulating their spreading capabilities. Here, we present an overview of the documented roles of ECM components in the spreading of pathological protein aggregates in neurodegenerative diseases with the objective of identifying the current gaps in knowledge and stimulating further research in the field. This could potentially lead to the identification of druggable targets to slow down the spreading and/or progression of these pathologies.
Collapse
Affiliation(s)
- Edoardo Moretto
- Institute of Neuroscience, National Research Council, CNR, Milan, Italy
- UK Dementia Research Institute, University College London, London, United Kingdom
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, United Kingdom
- *Correspondence: Edoardo Moretto,
| | - Skye Stuart
- UK Dementia Research Institute, University College London, London, United Kingdom
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Sunaina Surana
- UK Dementia Research Institute, University College London, London, United Kingdom
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, United Kingdom
- UCL Queen Square Motor Neuron Disease Centre, University College London, London, United Kingdom
| | - Jose Norberto S. Vargas
- UK Dementia Research Institute, University College London, London, United Kingdom
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, United Kingdom
- UCL Queen Square Motor Neuron Disease Centre, University College London, London, United Kingdom
| | - Giampietro Schiavo
- UK Dementia Research Institute, University College London, London, United Kingdom
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, United Kingdom
- UCL Queen Square Motor Neuron Disease Centre, University College London, London, United Kingdom
- Giampietro Schiavo,
| |
Collapse
|
24
|
Abstract
The brain harbors a unique ability to, figuratively speaking, shift its gears. During wakefulness, the brain is geared fully toward processing information and behaving, while homeostatic functions predominate during sleep. The blood-brain barrier establishes a stable environment that is optimal for neuronal function, yet the barrier imposes a physiological problem; transcapillary filtration that forms extracellular fluid in other organs is reduced to a minimum in brain. Consequently, the brain depends on a special fluid [the cerebrospinal fluid (CSF)] that is flushed into brain along the unique perivascular spaces created by astrocytic vascular endfeet. We describe this pathway, coined the term glymphatic system, based on its dependency on astrocytic vascular endfeet and their adluminal expression of aquaporin-4 water channels facing toward CSF-filled perivascular spaces. Glymphatic clearance of potentially harmful metabolic or protein waste products, such as amyloid-β, is primarily active during sleep, when its physiological drivers, the cardiac cycle, respiration, and slow vasomotion, together efficiently propel CSF inflow along periarterial spaces. The brain's extracellular space contains an abundance of proteoglycans and hyaluronan, which provide a low-resistance hydraulic conduit that rapidly can expand and shrink during the sleep-wake cycle. We describe this unique fluid system of the brain, which meets the brain's requisites to maintain homeostasis similar to peripheral organs, considering the blood-brain-barrier and the paths for formation and egress of the CSF.
Collapse
Affiliation(s)
- Martin Kaag Rasmussen
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Humberto Mestre
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, New York
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, New York
| |
Collapse
|
25
|
Kumar A, Biswas A, Bojja SL, Kolathur KK, Volety SM. Emerging therapeutic role of chondroitinase (ChABC) in neurological disorders and cancer. CURRENT DRUG THERAPY 2022. [DOI: 10.2174/1574885517666220331151619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Abstract:
Proteoglycans are essential biomacromolecules that participate in matrix structure and organization, cell proliferation and migration, and cell surface signal transduction. However, their roles in physiology, particularly in CNS remain incompletely deciphered. Numerous studies highlight the elevated levels of chondroitin sulphate proteoglycans (CSPGs) in various diseases like cancers and neurological disorders like spinal cord injury (SCI), traumatic brain damage, neurodegenerative diseases, and are mainly implicated to hinder tissue repair. In such a context, chondroitinase ABC (ChABC), a therapeutic enzyme has shown immense hope to treat these diseases in several preclinical studies, primarily attributed to the digestion of the side chains of the proteoglycan chondroitin sulphate (CS) molecule. Despite extensive research, the progress in evolving the concept of therapeutic targeting of proteoglycans is still in its infancy. This review thus provides fresh insights into the emerging therapeutic applications of ChABC in various diseases apart from SCI and the underlying mechanisms.
Collapse
Affiliation(s)
- Akshara Kumar
- Department of Pharmaceutical Biotechnology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Aishi Biswas
- Department of Pharmaceutical Biotechnology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Sree Lalitha Bojja
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Kiran Kumar Kolathur
- Department of Pharmaceutical Biotechnology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Subrahmanyam M Volety
- Department of Pharmaceutical Biotechnology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| |
Collapse
|
26
|
Bosi A, Banfi D, Bistoletti M, Moretto P, Moro E, Crema F, Maggi F, Karousou E, Viola M, Passi A, Vigetti D, Giaroni C, Baj A. Hyaluronan: A Neuroimmune Modulator in the Microbiota-Gut Axis. Cells 2021; 11:cells11010126. [PMID: 35011688 PMCID: PMC8750446 DOI: 10.3390/cells11010126] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/22/2021] [Accepted: 12/28/2021] [Indexed: 12/22/2022] Open
Abstract
The commensal microbiota plays a fundamental role in maintaining host gut homeostasis by controlling several metabolic, neuronal and immune functions. Conversely, changes in the gut microenvironment may alter the saprophytic microbial community and function, hampering the positive relationship with the host. In this bidirectional interplay between the gut microbiota and the host, hyaluronan (HA), an unbranched glycosaminoglycan component of the extracellular matrix, has a multifaceted role. HA is fundamental for bacterial metabolism and influences bacterial adhesiveness to the mucosal layer and diffusion across the epithelial barrier. In the host, HA may be produced and distributed in different cellular components within the gut microenvironment, playing a role in the modulation of immune and neuronal responses. This review covers the more recent studies highlighting the relevance of HA as a putative modulator of the communication between luminal bacteria and the host gut neuro-immune axis both in health and disease conditions, such as inflammatory bowel disease and ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Annalisa Bosi
- Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy; (A.B.); (D.B.); (M.B.); (P.M.); (F.M.); (E.K.); (M.V.); (A.P.); (D.V.); (A.B.)
| | - Davide Banfi
- Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy; (A.B.); (D.B.); (M.B.); (P.M.); (F.M.); (E.K.); (M.V.); (A.P.); (D.V.); (A.B.)
| | - Michela Bistoletti
- Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy; (A.B.); (D.B.); (M.B.); (P.M.); (F.M.); (E.K.); (M.V.); (A.P.); (D.V.); (A.B.)
| | - Paola Moretto
- Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy; (A.B.); (D.B.); (M.B.); (P.M.); (F.M.); (E.K.); (M.V.); (A.P.); (D.V.); (A.B.)
| | - Elisabetta Moro
- Department of Internal Medicine and Therapeutics, University of Pavia, 27100 Pavia, Italy; (E.M.); (F.C.)
| | - Francesca Crema
- Department of Internal Medicine and Therapeutics, University of Pavia, 27100 Pavia, Italy; (E.M.); (F.C.)
| | - Fabrizio Maggi
- Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy; (A.B.); (D.B.); (M.B.); (P.M.); (F.M.); (E.K.); (M.V.); (A.P.); (D.V.); (A.B.)
| | - Evgenia Karousou
- Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy; (A.B.); (D.B.); (M.B.); (P.M.); (F.M.); (E.K.); (M.V.); (A.P.); (D.V.); (A.B.)
| | - Manuela Viola
- Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy; (A.B.); (D.B.); (M.B.); (P.M.); (F.M.); (E.K.); (M.V.); (A.P.); (D.V.); (A.B.)
| | - Alberto Passi
- Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy; (A.B.); (D.B.); (M.B.); (P.M.); (F.M.); (E.K.); (M.V.); (A.P.); (D.V.); (A.B.)
| | - Davide Vigetti
- Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy; (A.B.); (D.B.); (M.B.); (P.M.); (F.M.); (E.K.); (M.V.); (A.P.); (D.V.); (A.B.)
| | - Cristina Giaroni
- Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy; (A.B.); (D.B.); (M.B.); (P.M.); (F.M.); (E.K.); (M.V.); (A.P.); (D.V.); (A.B.)
- Centre of Neuroscience, University of Insubria, 21100 Varese, Italy
- Correspondence: ; Tel.: +39-0332-217412; Fax: +39-0332-217111
| | - Andreina Baj
- Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy; (A.B.); (D.B.); (M.B.); (P.M.); (F.M.); (E.K.); (M.V.); (A.P.); (D.V.); (A.B.)
| |
Collapse
|
27
|
Garr E, Padovan-Hernandez Y, Janak PH, Delamater AR. Maintained goal-directed control with overtraining on ratio schedules. Learn Mem 2021; 28:435-439. [PMID: 34782401 PMCID: PMC8600976 DOI: 10.1101/lm.053472.121] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 09/16/2021] [Indexed: 11/25/2022]
Abstract
It is thought that goal-directed control of actions weakens or becomes masked by habits over time. We tested the opposing hypothesis that goal-directed control becomes stronger over time, and that this growth is modulated by the overall action-outcome contiguity. Despite group differences in action-outcome contiguity early in training, rats trained under random and fixed ratio schedules showed equivalent goal-directed control of lever pressing that appeared to grow over time. We confirmed that goal-directed control was maintained after extended training under another type of ratio schedule-continuous reinforcement-using specific satiety and taste aversion devaluation methods. These results add to the growing literature showing that extensive training does not reliably weaken goal-directed control and that it may strengthen it, or at least maintain it.
Collapse
Affiliation(s)
- Eric Garr
- Department of Psychological and Brain Sciences, Johns Hopkins University, Baltimore, Maryland 21218, USA
| | - Yasmin Padovan-Hernandez
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, Maryland 21205, USA
| | - Patricia H Janak
- Department of Psychological and Brain Sciences, Johns Hopkins University, Baltimore, Maryland 21218, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, Maryland 21205, USA
| | - Andrew R Delamater
- Department of Psychology, Brooklyn College, City University of New York, New York 11210, USA
- Department of Psychology, Graduate Center, City University of New York, New York 10016, USA
| |
Collapse
|
28
|
Guadagno A, Belliveau C, Mechawar N, Walker CD. Effects of Early Life Stress on the Developing Basolateral Amygdala-Prefrontal Cortex Circuit: The Emerging Role of Local Inhibition and Perineuronal Nets. Front Hum Neurosci 2021; 15:669120. [PMID: 34512291 PMCID: PMC8426628 DOI: 10.3389/fnhum.2021.669120] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 07/29/2021] [Indexed: 01/10/2023] Open
Abstract
The links between early life stress (ELS) and the emergence of psychopathology such as increased anxiety and depression are now well established, although the specific neurobiological and developmental mechanisms that translate ELS into poor health outcomes are still unclear. The consequences of ELS are complex because they depend on the form and severity of early stress, duration, and age of exposure as well as co-occurrence with other forms of physical or psychological trauma. The long term effects of ELS on the corticolimbic circuit underlying emotional and social behavior are particularly salient because ELS occurs during critical developmental periods in the establishment of this circuit, its local balance of inhibition:excitation and its connections with other neuronal pathways. Using examples drawn from the human and rodent literature, we review some of the consequences of ELS on the development of the corticolimbic circuit and how it might impact fear regulation in a sex- and hemispheric-dependent manner in both humans and rodents. We explore the effects of ELS on local inhibitory neurons and the formation of perineuronal nets (PNNs) that terminate critical periods of plasticity and promote the formation of stable local networks. Overall, the bulk of ELS studies report transient and/or long lasting alterations in both glutamatergic circuits and local inhibitory interneurons (INs) and their associated PNNs. Since the activity of INs plays a key role in the maturation of cortical regions and the formation of local field potentials, alterations in these INs triggered by ELS might critically participate in the development of psychiatric disorders in adulthood, including impaired fear extinction and anxiety behavior.
Collapse
Affiliation(s)
- Angela Guadagno
- Douglas Mental Health University Institute, Montreal, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Claudia Belliveau
- Douglas Mental Health University Institute, Montreal, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Naguib Mechawar
- Douglas Mental Health University Institute, Montreal, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Claire-Dominique Walker
- Douglas Mental Health University Institute, Montreal, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| |
Collapse
|
29
|
Zaki Y, Cai DJ. Creating Space for Synaptic Formation-A New Role for Microglia in Synaptic Plasticity. Cell 2021; 182:265-267. [PMID: 32707091 DOI: 10.1016/j.cell.2020.06.042] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Memory formation is thought to occur in the brain through dynamic remodeling of the synaptic architecture between neurons. The cellular mechanisms underlying these dynamics remain unclear. In this issue, Nguyen et al. demonstrate a novel role for microglia in regulating synaptic formation by clearing extracellular matrix proteins that embed neurons.
Collapse
Affiliation(s)
- Yosif Zaki
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Denise J Cai
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
30
|
Li Y, Han H, Shi K, Cui D, Yang J, Alberts IL, Yuan L, Zhao G, Wang R, Cai X, Teng Z. The Mechanism of Downregulated Interstitial Fluid Drainage Following Neuronal Excitation. Aging Dis 2020; 11:1407-1422. [PMID: 33269097 PMCID: PMC7673848 DOI: 10.14336/ad.2020.0224] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 02/24/2020] [Indexed: 12/20/2022] Open
Abstract
The drainage of brain interstitial fluid (ISF) has been observed to slow down following neuronal excitation, although the mechanism underlying this phenomenon is yet to be elucidated. In searching for the changes in the brain extracellular space (ECS) induced by electrical pain stimuli in the rat thalamus, significantly decreased effective diffusion coefficient (DECS) and volume fraction (α) of the brain ECS were shown, accompanied by the slowdown of ISF drainage. The morphological basis for structural changes in the brain ECS was local spatial deformation of astrocyte foot processes following neuronal excitation. We further studied aquaporin-4 gene (APQ4) knockout rats in which the changes of the brain ECS structure were reversed and found that the slowed DECS and ISF drainage persisted, confirming that the down-regulation of ISF drainage following neuronal excitation was mainly attributable to the release of neurotransmitters rather than to structural changes of the brain ECS. Meanwhile, the dynamic changes in the DECS were synchronized with the release and elimination processes of neurotransmitters following neuronal excitation. In conclusion, the downregulation of ISF drainage following neuronal excitation was found to be caused by the restricted diffusion in the brain ECS, and DECS mapping may be used to track the neuronal activity in the deep brain.
Collapse
Affiliation(s)
- Yuanyuan Li
- Department of Radiology, Peking University Third Hospital, Beijing, China.
- Beijing Key Laboratory of Magnetic Resonance Imaging Equipment and Technique, Beijing, China.
| | - Hongbin Han
- Department of Radiology, Peking University Third Hospital, Beijing, China.
- Beijing Key Laboratory of Magnetic Resonance Imaging Equipment and Technique, Beijing, China.
- Institute of Medical Technology, Peking University Health Science Center, Beijing, China.
| | - Kuangyu Shi
- Department of Nuclear Medicine, University of Bern, 3010 Bern, Switzerland.
- Department of Informatics, Technical University of Munich, Garching 85748, Germany.
| | - Dehua Cui
- Beijing Key Laboratory of Magnetic Resonance Imaging Equipment and Technique, Beijing, China.
| | - Jun Yang
- Department of Radiology, Peking University Third Hospital, Beijing, China.
| | - Ian Leigh Alberts
- Department of Nuclear Medicine, University of Bern, 3010 Bern, Switzerland.
| | - Lan Yuan
- Peking University Medical and Health Analysis Center, Peking University Health Science Center, Beijing, China.
| | - Guomei Zhao
- Department of Radiology, Peking University Third Hospital, Beijing, China.
- Beijing Key Laboratory of Magnetic Resonance Imaging Equipment and Technique, Beijing, China.
| | - Rui Wang
- Department of Radiology, Peking University Third Hospital, Beijing, China.
- Beijing Key Laboratory of Magnetic Resonance Imaging Equipment and Technique, Beijing, China.
| | - Xianjie Cai
- Department of Radiology, Peking University Third Hospital, Beijing, China.
- Beijing Key Laboratory of Magnetic Resonance Imaging Equipment and Technique, Beijing, China.
| | - Ze Teng
- Department of Radiology, Cancer Hospital Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
31
|
Spijker S, Koskinen MK, Riga D. Incubation of depression: ECM assembly and parvalbumin interneurons after stress. Neurosci Biobehav Rev 2020; 118:65-79. [DOI: 10.1016/j.neubiorev.2020.07.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 07/06/2020] [Accepted: 07/15/2020] [Indexed: 02/06/2023]
|
32
|
Guadagno A, Verlezza S, Long H, Wong TP, Walker CD. It Is All in the Right Amygdala: Increased Synaptic Plasticity and Perineuronal Nets in Male, But Not Female, Juvenile Rat Pups after Exposure to Early-Life Stress. J Neurosci 2020; 40:8276-8291. [PMID: 32978287 PMCID: PMC7577595 DOI: 10.1523/jneurosci.1029-20.2020] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 09/13/2020] [Accepted: 09/16/2020] [Indexed: 01/09/2023] Open
Abstract
Early-life stress (ELS) is associated with increased vulnerability to mental disorders. The basolateral amygdala (BLA) plays a critical role in fear conditioning and is extremely sensitive to ELS. Using a naturalistic rodent model of ELS, the limited bedding paradigm (LB) between postnatal days 1-10, we previously documented that LB male, but not female preweaning rat pups display increased BLA neuron spine density paralleled with enhanced evoked synaptic responses and altered BLA functional connectivity. Since ELS effects are often sexually dimorphic and amygdala processes exhibit hemispheric asymmetry, we investigated changes in synaptic plasticity and neuronal excitability of BLA neurons in vitro in the left and right amygdala of postnatal days 22-28 male and female offspring from normal bedding or LB mothers. We report that LB conditions enhanced synaptic plasticity in the right, but not the left BLA of males exclusively. LB males also showed increased perineuronal net density, particularly around parvalbumin (PV) cells, and impaired fear-induced activity of PV interneurons only in the right BLA. Action potentials fired from right BLA neurons of LB females displayed slower maximal depolarization rates and decreased amplitudes compared with normal bedding females, concomitant with reduced NMDAR GluN1 subunit expression in the right BLA. In LB males, reduced GluA2 expression in the right BLA might contribute to the enhanced LTP. These findings suggest that LB differentially programs synaptic plasticity and PV/perineuronal net development in the left and right BLA. Furthermore, our study demonstrates that the effects of ELS exposure on BLA synaptic function are sexually dimorphic and possibly recruiting different mechanisms.SIGNIFICANCE STATEMENT Early-life stress (ELS) induces long-lasting consequences on stress responses and emotional regulation in humans, increasing vulnerability to the development of psychopathologies. The effects of ELS in a number of brain regions, including the amygdala, are often sexually dimorphic, and have been reproduced using the rodent limited bedding paradigm of early adversity. The present study examines sex differences in synaptic plasticity and cellular activation occurring in the developing left and right amygdala after limited bedding exposure, a phenomenon that could shape long-term emotional behavioral outcomes. Studying how ELS selectively produces effects in one amygdala hemisphere during a critical period of brain development could guide further investigation into sex-dependent mechanisms and allow for more targeted and improved treatment of stress-and emotionality-related disorders.
Collapse
Affiliation(s)
- Angela Guadagno
- Douglas Mental Health University Institute, Montreal, Quebec, H4H 1R3, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, Quebec, H3A 0G4, Canada
| | - Silvanna Verlezza
- Douglas Mental Health University Institute, Montreal, Quebec, H4H 1R3, Canada
| | - Hong Long
- Douglas Mental Health University Institute, Montreal, Quebec, H4H 1R3, Canada
| | - Tak Pan Wong
- Douglas Mental Health University Institute, Montreal, Quebec, H4H 1R3, Canada
- Department of Psychiatry, McGill University, Montreal, Quebec, H3A 0G4, Canada
| | - Claire-Dominique Walker
- Douglas Mental Health University Institute, Montreal, Quebec, H4H 1R3, Canada
- Department of Psychiatry, McGill University, Montreal, Quebec, H3A 0G4, Canada
| |
Collapse
|
33
|
Histochemical Characterization of the Vestibular Y-Group in Monkey. THE CEREBELLUM 2020; 20:701-716. [PMID: 33083961 PMCID: PMC8629908 DOI: 10.1007/s12311-020-01200-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 10/04/2020] [Indexed: 12/18/2022]
Abstract
The Y-group plays an important role in the generation of upward smooth pursuit eye movements and contributes to the adaptive properties of the vertical vestibulo-ocular reflex. Malfunction of this circuitry may cause eye movement disorders, such as downbeat nystagmus. To characterize the neuron populations in the Y-group, we performed immunostainings for cellular proteins related to firing characteristics and transmitters (calretinin, GABA-related proteins and ion channels) in brainstem sections of macaque monkeys that had received tracer injections into the oculomotor nucleus. Two histochemically different populations of premotor neurons were identified: The calretinin-positive population represents the excitatory projection to contralateral upgaze motoneurons, whereas the GABAergic population represents the inhibitory projection to ipsilateral downgaze motoneurons. Both populations receive a strong supply by GABAergic nerve endings most likely originating from floccular Purkinje cells. All premotor neurons express nonphosphorylated neurofilaments and are ensheathed by strong perineuronal nets. In addition, they contain the voltage-gated potassium channels Kv1.1 and Kv3.1b which suggests biophysical similarities to high-activity premotor neurons of vestibular and oculomotor systems. The premotor neurons of Y-group form a homogenous population with histochemical characteristics compatible with fast-firing projection neurons that can also undergo plasticity and contribute to motor learning as found for the adaptation of the vestibulo-ocular reflex in response to visual-vestibular mismatch stimulation. The histochemical characterization of premotor neurons in the Y-group allows the identification of the homologue cell groups in human, including their transmitter inputs and will serve as basis for correlated anatomical-neuropathological studies of clinical cases with downbeat nystagmus.
Collapse
|
34
|
Meurer YDSR, Brito RMDM, da Silva VP, Andade JMDA, Linhares SSG, Pereira Junior A, de Andrade-Neto VF, de Sá AL, Oliveira CBSD. Toxoplasma gondii infection damages the perineuronal nets in a murine model. Mem Inst Oswaldo Cruz 2020; 115:e200007. [PMID: 32935749 PMCID: PMC7491278 DOI: 10.1590/0074-02760200007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 08/10/2020] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Behavioral and neurochemical alterations associated with toxoplasmosis may be influenced by the persistence of tissue cysts and activation of an immune response in the brain of Toxoplasma gondii-infected hosts. The cerebral extracellular matrix is organised as perineuronal nets (PNNs) that are both released and ensheath by some neurons and glial cells. There is evidences to suggest that PNNs impairment is a pathophysiological mechanism associated with neuropsychiatric conditions. However, there is a lack of information regarding the impact of parasitic infections on the PNNs integrity and how this could affect the host’s behavior. OBJECTIVES In this context, we aimed to analyse the impact of T. gondii infection on cyst burden, PNNs integrity, and possible effects in the locomotor activity of chronically infected mice. METHODS We infected mice with T. gondii ME-49 strain. After thirty days, we assessed locomotor performance of animals using the open field test, followed by evaluation of cysts burden and PNNs integrity in four brain regions (primary and secondary motor cortices, prefrontal and somesthetic cortex) to assess the PNNs integrity using Wisteria floribunda agglutinin (WFA) labeling by immunohistochemical analyses. FINDINGS AND MAIN CONCLUSIONS Our findings revealed a random distribution of cysts in the brain, the disruption of PNNs surrounding neurons in four areas of the cerebral cortex and hyperlocomotor behavior in T. gondii-infected mice. These results can contribute to elucidate the link toxoplasmosis with the establishment of neuroinflammatory response in neuropsychiatric disorders and to raise a discussion about the mechanisms related to changes in brain connectivity, with possible behavioral repercussions during chronic T. gondii infection.
Collapse
Affiliation(s)
- Ywlliane da Silva Rodrigues Meurer
- Universidade Federal da Paraíba, Programa de Pós-Graduação em Neurociência Cognitiva e Comportamento, João Pessoa, PB, Brasil.,Universidade Federal do Rio Grande do Norte, Programa de Pós-Graduação em Psicobiologia, Natal, RN, Brasil
| | - Ramayana Morais de Medeiros Brito
- Universidade Federal do Rio Grande do Norte, Departamento de Microbiologia e Parasitologia, Laboratório de Biologia da Málaria e Toxoplasmose - LABMAT, Natal, RN, Brasil
| | - Valeria Palheta da Silva
- Universidade Federal do Rio Grande do Norte, Programa de Pós-Graduação em Psicobiologia, Natal, RN, Brasil
| | - Joelma Maria de Araujo Andade
- Universidade Federal do Rio Grande do Norte, Departamento de Microbiologia e Parasitologia, Laboratório de Biologia da Málaria e Toxoplasmose - LABMAT, Natal, RN, Brasil
| | | | - Antonio Pereira Junior
- Universidade Federal do Pará, Instituto de Ciências da Sáude, Laboratório de Neuroplasticidade, Belém, PA, Brasil
| | - Valter Ferreira de Andrade-Neto
- Universidade Federal do Rio Grande do Norte, Departamento de Microbiologia e Parasitologia, Laboratório de Biologia da Málaria e Toxoplasmose - LABMAT, Natal, RN, Brasil
| | - Andrea Lima de Sá
- Universidade Federal do Rio Grande do Norte, Departamento de Microbiologia e Parasitologia, Laboratório de Biologia da Málaria e Toxoplasmose - LABMAT, Natal, RN, Brasil
| | - Claudio Bruno Silva de Oliveira
- Universidade Federal do Rio Grande do Norte, Departamento de Microbiologia e Parasitologia, Laboratório de Biologia da Málaria e Toxoplasmose - LABMAT, Natal, RN, Brasil
| |
Collapse
|
35
|
Yang X. Chondroitin sulfate proteoglycans: key modulators of neuronal plasticity, long-term memory, neurodegenerative, and psychiatric disorders. Rev Neurosci 2020; 31:555-568. [PMID: 32126020 DOI: 10.1515/revneuro-2019-0117] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 01/02/2020] [Indexed: 12/19/2022]
Abstract
The chondroitin sulfate proteoglycans (CSPGs) are large groups of heterogenous proteoglycans that are mainly expressed by reactive astrocytes in the central nervous system (CNS). They share similar core proteins and are post-transcriptionally modified by chondroitin sulfate glycosaminoglycans. CSPGs are the major components of the perineuronal nets (PNN) that regulate the opening and closure of the critical period. Mounting reports have documented the crucial roles of CSPGs in restricting neuronal plasticity, axonal growth, and pathfinding during development as well as axonal regeneration after CNS injury. Moreover, CSPGs and PNNs modulate long-term memory, which impairments frequently happened in several neurodegenerative and psychiatric disorders. This review will shortly introduce the expression patterns of CSPGs during development and after injury, the PNNs constitutions, the roles of CSPGs and PNNs in axonal regrowth, discuss the most recently identified roles of CSPGs and PNNs in mediating long-term memory and their correlation with brain disorders, and finally, propose a short perspective of future investigations. Hopefully, further explorations may validate the therapeutic potentials of PNNs and CSPGs.
Collapse
Affiliation(s)
- Xin Yang
- Division of Life Science, Hong Kong University of Science and Technology, Hong Kong, P.R. China
| |
Collapse
|
36
|
Abstract
In the adult mammalian hippocampus, new neurons arise from stem and progenitor cell division, in a process known as adult neurogenesis. Adult-generated neurons are sensitive to experience and may participate in hippocampal functions, including learning and memory, anxiety and stress regulation, and social behavior. Increasing evidence emphasizes the importance of new neuron connectivity within hippocampal circuitry for understanding the impact of adult neurogenesis on brain function. In this Review, we discuss how the functional consequences of new neurons arise from the collective interactions of presynaptic and postsynaptic neurons, glial cells, and the extracellular matrix, which together form the "tetrapartite synapse."
Collapse
Affiliation(s)
- Elise C Cope
- Princeton Neuroscience Institute and Department of Psychology, Princeton University, Princeton, NJ 08544, USA
| | - Elizabeth Gould
- Princeton Neuroscience Institute and Department of Psychology, Princeton University, Princeton, NJ 08544, USA.
| |
Collapse
|
37
|
Bistoletti M, Bosi A, Caon I, Chiaravalli AM, Moretto P, Genoni A, Moro E, Karousou E, Viola M, Crema F, Baj A, Passi A, Vigetti D, Giaroni C. Involvement of hyaluronan in the adaptive changes of the rat small intestine neuromuscular function after ischemia/reperfusion injury. Sci Rep 2020; 10:11521. [PMID: 32661417 PMCID: PMC7359366 DOI: 10.1038/s41598-020-67876-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 04/30/2020] [Indexed: 12/12/2022] Open
Abstract
Intestinal ischemia/reperfusion (I/R) injury has severe consequences on myenteric neurons, which can be irreversibly compromised resulting in slowing of transit and hindered food digestion. Myenteric neurons synthesize hyaluronan (HA) to form a well-structured perineuronal net, which undergoes derangement when myenteric ganglia homeostasis is perturbed, i.e. during inflammation. In this study we evaluated HA involvement in rat small intestine myenteric plexus after in vivo I/R injury induced by clamping a branch of the superior mesenteric artery for 60 min, followed by 24 h of reperfusion. In some experiments, 4-methylumbelliferone (4-MU, 25 mg/kg), a HA synthesis inhibitor, was intraperitoneally administered to normal (CTR), sham-operated (SH) and I/R animals for 24 h. In longitudinal muscle myenteric plexus (LMMP) whole-mount preparations, HA binding protein staining as well as HA levels were significantly higher in the I/R group, and were reduced after 4-MU treatment. HA synthase 1 and 2 (HAS1 and HAS2) labelled myenteric neurons and mRNA levels in LMMPs increased in the I/R group with respect to CTR, and were reduced by 4-MU. The efficiency of the gastrointestinal transit was significantly reduced in I/R and 4-MU-treated I/R groups with respect to CTR and SH groups. In the 4-MU-treated I/R group gastric emptying was reduced with respect to the CTR, SH and I/R groups. Carbachol (CCh) and electrical field (EFS, 0.1–40 Hz) stimulated contractions and EFS-induced (10 Hz) NANC relaxations were reduced in the I/R group with respect to both CTR and SH groups. After I/R, 4-MU treatment increased EFS contractions towards control values, but did not affect CCh-induced contractions. NANC on-relaxations after I/R were not influenced by 4-MU treatment. Main alterations in the neurochemical coding of both excitatory (tachykinergic) and inhibitory pathways (iNOS, VIPergic) were also observed after I/R, and were influenced by 4-MU administration. Overall, our data suggest that, after an intestinal I/R damage, changes of HA homeostasis in specific myenteric neuron populations may influence the efficiency of the gastrointestinal transit. We cannot exclude that modulation of HA synthesis in these conditions may ameliorate derangement of the enteric motor function preventing, at least in part, the development of dysmotility.
Collapse
Affiliation(s)
- Michela Bistoletti
- Department of Medicine and Surgery, University of Insubria, via H. Dunant 5, Varese, Italy
| | - Annalisa Bosi
- Department of Medicine and Surgery, University of Insubria, via H. Dunant 5, Varese, Italy
| | - Ilaria Caon
- Department of Medicine and Surgery, University of Insubria, via H. Dunant 5, Varese, Italy
| | - Anna Maria Chiaravalli
- Department of Pathology, ASST-Sette Laghi, Ospedale di Circolo Viale L. Borri 57, 21100, Varese, Italy
| | - Paola Moretto
- Department of Medicine and Surgery, University of Insubria, via H. Dunant 5, Varese, Italy
| | - Angelo Genoni
- Department of Medicine and Surgery, University of Insubria, via H. Dunant 5, Varese, Italy
| | - Elisabetta Moro
- Department of Internal Medicine and Therapeutics, Section of Pharmacology, University of Pavia, Pavia, Italy
| | - Evgenia Karousou
- Department of Medicine and Surgery, University of Insubria, via H. Dunant 5, Varese, Italy
| | - Manuela Viola
- Department of Medicine and Surgery, University of Insubria, via H. Dunant 5, Varese, Italy
| | - Francesca Crema
- Department of Internal Medicine and Therapeutics, Section of Pharmacology, University of Pavia, Pavia, Italy
| | - Andreina Baj
- Department of Medicine and Surgery, University of Insubria, via H. Dunant 5, Varese, Italy
| | - Alberto Passi
- Department of Medicine and Surgery, University of Insubria, via H. Dunant 5, Varese, Italy
| | - Davide Vigetti
- Department of Medicine and Surgery, University of Insubria, via H. Dunant 5, Varese, Italy.
| | - Cristina Giaroni
- Department of Medicine and Surgery, University of Insubria, via H. Dunant 5, Varese, Italy.
| |
Collapse
|
38
|
Collins LN, Brunjes PC. The mouse olfactory peduncle 4: Development of synapses, perineuronal nets, and capillaries. J Comp Neurol 2020; 528:637-649. [PMID: 31571216 PMCID: PMC6944759 DOI: 10.1002/cne.24778] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 09/03/2019] [Accepted: 09/04/2019] [Indexed: 11/09/2022]
Abstract
Olfaction is critical for survival in neonatal mammals. However, little is known about the neural substrate for this ability as few studies of synaptic development in several olfactory processing regions have been reported. Odor information detected in the nasal cavity is first processed by the olfactory bulb and then sent via the lateral olfactory tract to a series of olfactory cortical areas. The first of these, the anterior olfactory nucleus pars principalis (AONpP), is a simple, two layered cortex with an outer plexiform and inner cell zone (Layers 1 and 2, respectively). Five sets of studies examined age-related changes in the AONpP. First, immunocytochemistry for glutamatergic (VGlut1 and VGlut2) and GABAergic (VGAT) synapses demonstrated that overall synaptic patterns remained uniform with age. The second set quantified synaptic development with electron microscopy and found different developmental patterns between Layers 1 and 2. As many of the interhemispheric connections in the olfactory system arise from AONpP, the third set examined the development of crossed projections using anterograde tracers and electron microscopy to explore the maturation of this pathway. A fourth study examined ontogenetic changes in immunostaining for the proteoglycans aggrecan and brevican, markers of mesh-like extracellular structures known as perineuronal nets whose maturation is associated with the end of early critical periods of synaptogenesis. A final study found no age-related changes in the density of vasculature in the peduncle from P5 to P30. This work is among the first to examine early postnatal changes in this initial cortical region of the olfactory system.
Collapse
Affiliation(s)
- Lindsay N. Collins
- Department Psychology, University of Virginia, Charlottesville, Virginia 22904 USA
| | - Peter C. Brunjes
- Department Psychology, University of Virginia, Charlottesville, Virginia 22904 USA
| |
Collapse
|
39
|
Cornez G, Shevchouk OT, Ghorbanpoor S, Ball GF, Cornil CA, Balthazart J. Testosterone stimulates perineuronal nets development around parvalbumin cells in the adult canary brain in parallel with song crystallization. Horm Behav 2020; 119:104643. [PMID: 31785283 PMCID: PMC7065963 DOI: 10.1016/j.yhbeh.2019.104643] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 11/18/2019] [Accepted: 11/19/2019] [Indexed: 10/25/2022]
Abstract
Perineuronal nets (PNN) of the extracellular matrix are dense aggregations of chondroitin-sulfate proteoglycans that usually surround fast-spiking parvalbumin-expressing inhibitory interneurons (PV). The development of PNN around PV appears specifically at the end of sensitive periods of visual learning and limits the synaptic plasticity in the visual cortex of mammals. Seasonal songbirds display a high level of adult neuroplasticity associated with vocal learning, which is regulated by fluctuations of circulating testosterone concentrations. Seasonal changes in testosterone concentrations and in neuroplasticity are associated with vocal changes between the non-breeding and breeding seasons. Increases in blood testosterone concentrations in the spring lead to the annual crystallization of song so that song becomes more stereotyped. Here we explore whether testosterone also regulates PNN expression in the song control system of male and female canaries. We show that, in both males and females, testosterone increases the number of PNN and of PV neurons in the three main telencephalic song control nuclei HVC, RA (nucleus robustus arcopallialis) and Area X and increases the PNN localization around PV interneurons. Singing activity was recorded in males and quantitative analyses demonstrated that testosterone also increased male singing rate, song duration and song energy while decreasing song entropy. Together, these data suggest that the development of PNN could provide the synaptic stability required to maintain the stability of the testosterone-induced crystallized song. This provides the new evidence for a role of PNN in the regulation of adult seasonal plasticity in seasonal songbirds.
Collapse
Affiliation(s)
- Gilles Cornez
- GIGA Neuroscience, University of Liege, Liege 4000, Belgium
| | | | | | - Gregory F Ball
- Department of Psychology, University of Maryland, College Park, MD 20742, USA
| | | | | |
Collapse
|
40
|
Paviolo C, Soria FN, Ferreira JS, Lee A, Groc L, Bezard E, Cognet L. Nanoscale exploration of the extracellular space in the live brain by combining single carbon nanotube tracking and super-resolution imaging analysis. Methods 2020; 174:91-99. [DOI: 10.1016/j.ymeth.2019.03.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 03/04/2019] [Accepted: 03/07/2019] [Indexed: 12/22/2022] Open
|
41
|
Chapman G, Shanmugalingam U, Smith PD. The Role of Neuronal Pentraxin 2 (NP2) in Regulating Glutamatergic Signaling and Neuropathology. Front Cell Neurosci 2020; 13:575. [PMID: 31969807 PMCID: PMC6960182 DOI: 10.3389/fncel.2019.00575] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Accepted: 12/12/2019] [Indexed: 01/30/2023] Open
Abstract
Pentraxins are a superfamily of evolutionarily conserved proteins that are characterized by their multimeric architecture and their calcium-dependent binding. They can be broadly grouped into two subfamilies: short pentraxins and long pentraxins. Pentraxins regulate many processes in the brain as well as the periphery. Neuronal pentraxin 2 (NP2/NPTX2), also known as neuronal activity-regulated pentraxin (Narp), is an immediate-early gene that has been shown to play a critical role in guiding synaptic plasticity. NP2 has been previously linked to excitatory neurotransmission, based on its ability to aggregate excitatory receptors in the central nervous system. The mechanisms mediating the effects of NP2 on excitatory neurotransmission remain unclear and warrants further investigation. This review article focuses on the biological features of NP2 and discusses the literature supporting a role for NP2 and other pentraxins in glutamatergic signaling. An analysis of evidence around the role of pentraxins in neuropathology is also reviewed.
Collapse
Affiliation(s)
- Georgina Chapman
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada
| | | | - Patrice D Smith
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada
| |
Collapse
|
42
|
Cornez G, Langro J, Cornil CA, Balthazart J, Lynch KS. Comparing perineuronal nets and parvalbumin development between blackbird species with differences in early developmental song exposure. ACTA ACUST UNITED AC 2020; 223:jeb.212910. [PMID: 31767738 DOI: 10.1242/jeb.212910] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 11/20/2019] [Indexed: 11/20/2022]
Abstract
Brood parasitic songbirds are a natural system in which developing birds are isolated from species-typical song and therefore present a unique opportunity to compare neural plasticity in song learners raised with and without conspecific tutors. We compared perineuronal nets (PNN) and parvalbumin (PV) in song control nuclei in juveniles and adults of two closely related icterid species (i.e. blackbirds): brown-headed cowbirds (Molothrus ater; brood parasite) and red-winged blackbirds (Agelaius phoeniceus; non-parasite). The number of PV cells per nucleus was significantly higher in adults compared with juveniles in the nucleus HVC and the robust nucleus of the arcopallium (RA), whereas no significant species difference appeared in any region of interest. The number of PNN per nuclei was significantly higher in adults compared with juveniles in HVC, RA and Area X, but only RA exhibited a significant difference between species. PV cells surrounded by PNN (PV+PNN) also exhibited age-related differences in HVC, RA and Area X, but RA was the only region in which PV+PNN exhibited significant species differences. Furthermore, a significant interaction existed in RA between age and species with respect to PNN and PV+PNN, revealing RA as a region displaying differing plasticity patterns across age and species. Additional comparisons of PNN and PV between adult male and female cowbirds revealed that males have greater numbers of all three measures in RA compared with females. Species-, sex- and age-related differences in RA suggest that species differences in neural plasticity are related to differences in song production rather than sensitivity to song learning, despite a stark contrast in early exposure to conspecific male tutors.
Collapse
Affiliation(s)
- Gilles Cornez
- Laboratory of Behavioral Neuroendocrinology, GIGA Neurosciences, University of Liege, 4000 Liege, Belgium
| | - Justin Langro
- Department of Biology, Hofstra University, Hempstead, NY 11549, USA
| | - Charlotte A Cornil
- Laboratory of Behavioral Neuroendocrinology, GIGA Neurosciences, University of Liege, 4000 Liege, Belgium
| | - Jacques Balthazart
- Laboratory of Behavioral Neuroendocrinology, GIGA Neurosciences, University of Liege, 4000 Liege, Belgium
| | - Kathleen S Lynch
- Department of Biology, Hofstra University, Hempstead, NY 11549, USA
| |
Collapse
|
43
|
Souter L, Kwok JCF. Visualization of Perineuronal Nets in Central Nervous System Tissue Sections. Methods Mol Biol 2020; 2043:251-260. [PMID: 31463917 DOI: 10.1007/978-1-4939-9698-8_20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The perineuronal net (PNN) is a specialized extracellular matrix structure that surrounds subpopulations of neurons in the central nervous system (CNS). The appearance of PNNs on the cell surface marks the closure of the critical period during development and has been observed to reduce synaptic plasticity. Perineuronal nets comprise hyaluronan, chondroitin sulfate proteoglycans (CSPGs), link proteins, tenascin-R, and other components, some of which are substrates for a disintegrin-like and metalloprotease domain with thrombospondin type 1 motifs (ADAMTS) proteases. There is a high heterogeneity of PNNs in the CNS. Depending on which part of the CNS is studied, the PNNs may be observed surrounding the soma, or both the soma and proximal dendrites. The most robust marker for PNN is a lectin called Wisteria floribunda agglutinin. Here, we describe a method for preparing tissue for visualization of PNNs in CNS.
Collapse
Affiliation(s)
- Luke Souter
- Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, UK.,School of Mechanical Engineering, Faculty of Engineering,, University of Leeds, Leeds, UK
| | - Jessica C F Kwok
- Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, UK. .,Institute of Experimental Medicine, Czech Academy of Science, Prague 4, Czech Republic.
| |
Collapse
|
44
|
Cornez G, Collignon C, Müller W, Ball GF, Cornil CA, Balthazart J. Seasonal changes of perineuronal nets and song learning in adult canaries (Serinus canaria). Behav Brain Res 2019; 380:112437. [PMID: 31857148 DOI: 10.1016/j.bbr.2019.112437] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 11/16/2019] [Accepted: 12/14/2019] [Indexed: 01/31/2023]
Abstract
Songbirds learn their song during a sensitive period of development associated with enhanced neural plasticity. In addition, in open-ended learners such as canaries, a sensitive period for sensorimotor vocal learning reopens each year in the fall and leads to song modifications between successive breeding seasons. The variability observed in song production across seasons in adult canaries correlates with seasonal fluctuations of testosterone concentrations and with morphological changes in nuclei of the song control system (SCS). The sensitive periods for song learning during ontogeny and then again in adulthood could be controlled by the development of perineuronal nets (PNN) around parvalbumin-expressing interneurones (PV) which limits learning-induced neuroplasticity. However, this relationship has never been investigated in the context of adult vocal learning in adult songbirds. Here we explored PNN and PV expression in the SCS of adult male Fife Fancy canaries in relation to the seasonal variations of their singing behaviour. We found a clear pattern of seasonal variation in testosterone concentrations and song production. Furthermore, PNN expression was significantly higher in two specific song control nuclei, the robust nucleus of the arcopallium (RA) and the Area X of the basal ganglia, during the breeding season and during the later stages of sensorimotor song development compared to birds in an earlier stage of sensorimotor development during the fall. These data provide the first evidence that changes in PNN expression could represent a mechanism regulating the closing-reopening of sensitive periods for vocal learning across seasons in adult songbirds.
Collapse
Affiliation(s)
- Gilles Cornez
- Laboratory of Behavioral Neuroendocrinology, GIGA Neurosciences, University of Liege, Belgium
| | - Clémentine Collignon
- Laboratory of Behavioral Neuroendocrinology, GIGA Neurosciences, University of Liege, Belgium
| | - Wendt Müller
- Behavioural Ecology and Ecophysiology Research Group, University of Antwerp, Belgium
| | - Gregory F Ball
- Department of Psychology, University of Maryland, College Park MD, USA
| | - Charlotte A Cornil
- Laboratory of Behavioral Neuroendocrinology, GIGA Neurosciences, University of Liege, Belgium
| | - Jacques Balthazart
- Laboratory of Behavioral Neuroendocrinology, GIGA Neurosciences, University of Liege, Belgium.
| |
Collapse
|
45
|
Neuronal Pentraxin 2 Binds PNNs and Enhances PNN Formation. Neural Plast 2019; 2019:6804575. [PMID: 31772567 PMCID: PMC6854953 DOI: 10.1155/2019/6804575] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 08/30/2019] [Accepted: 09/16/2019] [Indexed: 02/06/2023] Open
Abstract
The perineuronal net (PNN) is a mesh-like proteoglycan structure on the neuronal surface which is involved in regulating plasticity. The PNN regulates plasticity via multiple pathways, one of which is direct regulation of synapses through the control of AMPA receptor mobility. Since neuronal pentraxin 2 (Nptx2) is a known regulator of AMPA receptor mobility and Nptx2 can be removed from the neuronal surface by PNN removal, we investigated whether Nptx2 has a function in the PNN. We found that Nptx2 binds to the glycosaminoglycans hyaluronan and chondroitin sulphate E in the PNN. Furthermore, in primary cortical neuron cultures, the addition of NPTX2 to the culture medium enhances PNN formation during PNN development. These findings suggest Nptx2 as a novel PNN binding protein with a role in the mechanism of PNN formation.
Collapse
|
46
|
Quraishe S, Newman T, Anderson L. Auditory temporal acuity improves with age in the male mouse auditory thalamus: A role for perineuronal nets? J Neurosci Res 2019; 98:1780-1799. [PMID: 31562661 DOI: 10.1002/jnr.24537] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 09/13/2019] [Accepted: 09/15/2019] [Indexed: 11/09/2022]
Abstract
The ability to perceive and interpret environmental sound accurately is conserved across many species and is fundamental for understanding communication via vocalizations. Auditory acuity and temporally controlled neuronal firing underpin this ability. Deterioration in neuronal firing precision likely contributes to poorer hearing performance, yet the role of neural processing by key nuclei in the central auditory pathways is not fully understood. Here, we record from the auditory thalamus (medial geniculate body [MGB]) of young and middle-aged, normally hearing male CBA/Ca mice. We report changes in temporal processing of auditory stimuli, with neurons recorded from ventral and medial MGB subdivisions of older animals more likely to synchronize to rapid temporally varying stimuli. MGB subdivisions also showed increased probability of neuronal firing and shorter response latencies to clicks in older animals. Histological investigation of neuronal extracellular specializations, perineuronal nets (PNNs) and axonal coats, in the MGB identified greater organization of PNNs around MGB neurons and the presence of axonal coats within older animals. This supports the observation that neural responses recorded from ventral and medial MGB of older mice were more likely to synchronize to temporally varying stimuli presented at faster repetition rates than those recorded from young adult animals. These changes are observed in animals with normal hearing thresholds, confirming that neural processing differs between the MGB subdivisions and such processing is associated with age-related changes to PNNs. Understanding these age-related changes and how they occur have important implications for the design of effective therapeutic interventions to improve speech intelligibility into later life.
Collapse
Affiliation(s)
- Shmma Quraishe
- School of Biological Sciences, B85, University of Southampton, Southampton, UK
| | - Tracey Newman
- Clinical and Experimental Sciences, B85, University of Southampton, Southampton, UK
| | | |
Collapse
|
47
|
Duncan JA, Foster R, Kwok JC. The potential of memory enhancement through modulation of perineuronal nets. Br J Pharmacol 2019; 176:3611-3621. [PMID: 30924524 PMCID: PMC6715611 DOI: 10.1111/bph.14672] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 02/12/2019] [Accepted: 02/22/2019] [Indexed: 12/18/2022] Open
Abstract
With an increasingly aging global population, the incidence of neurological diseases such as dementia is set to increase to unmanageable levels, yet there are currently only symptomatic therapies available for treatment. The mechanisms underlying the development of some forms of dementia, such as Alzheimer's disease (AD), are not yet completely elucidated with several competing hypotheses existing. During the closure of the critical period in the brain, significant compositional changes occur to the neural extracellular matrix (ECM). Specifically, condensed mesh-like structures called perineuronal nets (PNNs) form around subsets of neurons and have a profound effect on axonal growth and limit neuronal plasticity. These PNNs act as a morphological checkpoint and can influence memory and cognition. Manipulating these important ECM structures may provide the key to reactivating plasticity and restoring memory, both of which are severely impaired in AD and other associated neurological diseases. This review explores the current understanding of how PNNs are manipulated and examines potential new methods for PNN modulation. LINKED ARTICLES: This article is part of a themed section on Therapeutics for Dementia and Alzheimer's Disease: New Directions for Precision Medicine. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.18/issuetoc.
Collapse
Affiliation(s)
| | - Richard Foster
- School of ChemistryUniversity of LeedsLeedsUK
- Astbury Centre for Structural Molecular BiologyUniversity of LeedsLeedsUK
| | - Jessica C.F. Kwok
- School of Biomedical SciencesUniversity of LeedsLeedsUK
- Institute of Experimental MedicineCzech Academy of SciencePragueCzechia
| |
Collapse
|
48
|
Krishnaswamy VR, Benbenishty A, Blinder P, Sagi I. Demystifying the extracellular matrix and its proteolytic remodeling in the brain: structural and functional insights. Cell Mol Life Sci 2019; 76:3229-3248. [PMID: 31197404 PMCID: PMC11105229 DOI: 10.1007/s00018-019-03182-6] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 05/29/2019] [Accepted: 05/31/2019] [Indexed: 12/29/2022]
Abstract
The extracellular matrix (ECM) plays diverse roles in several physiological and pathological conditions. In the brain, the ECM is unique both in its composition and in functions. Furthermore, almost all the cells in the central nervous system contribute to different aspects of this intricate structure. Brain ECM, enriched with proteoglycans and other small proteins, aggregate into distinct structures around neurons and oligodendrocytes. These special structures have cardinal functions in the normal functioning of the brain, such as learning, memory, and synapse regulation. In this review, we have compiled the current knowledge about the structure and function of important ECM molecules in the brain and their proteolytic remodeling by matrix metalloproteinases and other enzymes, highlighting the special structures they form. In particular, the proteoglycans in brain ECM, which are essential for several vital functions, are emphasized in detail.
Collapse
Affiliation(s)
| | - Amit Benbenishty
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Pablo Blinder
- Neurobiology, Biochemistry and Biophysics School, Tel Aviv University, Tel Aviv, Israel
- Sagol School for Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Irit Sagi
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
49
|
Bagamasbad PD, Espina JEC, Knoedler JR, Subramani A, Harden AJ, Denver RJ. Coordinated transcriptional regulation by thyroid hormone and glucocorticoid interaction in adult mouse hippocampus-derived neuronal cells. PLoS One 2019; 14:e0220378. [PMID: 31348800 PMCID: PMC6660079 DOI: 10.1371/journal.pone.0220378] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 07/15/2019] [Indexed: 12/04/2022] Open
Abstract
The hippocampus is a well-known target of thyroid hormone (TH; e.g., 3,5,3'-triiodothyronine-T3) and glucocorticoid (GC; e.g., corticosterone-CORT) action. Despite evidence that TH and GC play critical roles in neural development and function, few studies have identified genes and patterns of gene regulation influenced by the interaction of these hormones at a genome-wide scale. In this study we investigated gene regulation by T3, CORT, and T3 + CORT in the mouse hippocampus-derived cell line HT-22. We treated cells with T3, CORT, or T3 + CORT for 4 hr before cell harvest and RNA isolation for microarray analysis. We identified 9 genes regulated by T3, 432 genes by CORT, and 412 genes by T3 + CORT. Among the 432 CORT-regulated genes, there were 203 genes that exhibited an altered CORT response in the presence of T3, suggesting that T3 plays a significant role in modulating CORT-regulated genes. We also found 80 genes synergistically induced, and 73 genes synergistically repressed by T3 + CORT treatment. We performed in silico analysis using publicly available mouse neuronal chromatin immunoprecipitation-sequencing datasets and identified a considerable number of synergistically regulated genes with TH receptor and GC receptor peaks mapping within 1 kb of chromatin marks indicative of hormone-responsive enhancer regions. Functional annotation clustering of synergistically regulated genes reveal the relevance of proteasomal-dependent degradation, neuroprotective effect of growth hormones, and neuroinflammatory responses as key pathways to how TH and GC may coordinately influence learning and memory. Taken together, our transcriptome data represents a promising exploratory dataset for further study of common molecular mechanisms behind synergistic TH and GC gene regulation, and identify specific genes and their role in processes mediated by cross-talk between the thyroid and stress axes in a mammalian hippocampal model system.
Collapse
Affiliation(s)
- Pia D. Bagamasbad
- Department of Molecular, Cellular and Developmental Biology, The University of Michigan, Ann Arbor, Michigan, United States of America
- National Institute of Molecular Biology and Biotechnology, University of the Philippines Diliman, Quezon City, Philippines
| | - Jose Ezekiel C. Espina
- National Institute of Molecular Biology and Biotechnology, University of the Philippines Diliman, Quezon City, Philippines
| | - Joseph R. Knoedler
- Neuroscience Graduate Program, The University of Michigan, Ann Arbor, Michigan, United States of America
| | - Arasakumar Subramani
- Department of Molecular, Cellular and Developmental Biology, The University of Michigan, Ann Arbor, Michigan, United States of America
| | - Ariel J. Harden
- Department of Molecular, Cellular and Developmental Biology, The University of Michigan, Ann Arbor, Michigan, United States of America
| | - Robert J. Denver
- Department of Molecular, Cellular and Developmental Biology, The University of Michigan, Ann Arbor, Michigan, United States of America
| |
Collapse
|
50
|
Reichelt AC, Hare DJ, Bussey TJ, Saksida LM. Perineuronal Nets: Plasticity, Protection, and Therapeutic Potential. Trends Neurosci 2019; 42:458-470. [DOI: 10.1016/j.tins.2019.04.003] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 04/16/2019] [Accepted: 04/22/2019] [Indexed: 12/18/2022]
|