1
|
Melrose J. CNS/PNS proteoglycans functionalize neuronal and astrocyte niche microenvironments optimizing cellular activity by preserving membrane polarization dynamics, ionic microenvironments, ion fluxes, neuronal activation, and network neurotransductive capacity. J Neurosci Res 2024; 102:e25361. [PMID: 39034899 DOI: 10.1002/jnr.25361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 03/22/2024] [Accepted: 05/27/2024] [Indexed: 07/23/2024]
Abstract
Central and peripheral nervous system (CNS/PNS) proteoglycans (PGs) have diverse functional roles, this study examined how these control cellular behavior and tissue function. The CNS/PNS extracellular matrix (ECM) is a dynamic, responsive, highly interactive, space-filling, cell supportive, stabilizing structure maintaining tissue compartments, ionic microenvironments, and microgradients that regulate neuronal activity and maintain the neuron in an optimal ionic microenvironment. The CNS/PNS contains a high glycosaminoglycan content (60% hyaluronan, HA) and a diverse range of stabilizing PGs. Immobilization of HA in brain tissues by HA interactive hyalectan PGs preserves tissue hydration and neuronal activity, a paucity of HA in brain tissues results in a pro-convulsant epileptic phenotype. Diverse CS, KS, and HSPGs stabilize the blood-brain barrier and neurovascular unit, provide smart gel neurotransmitter neuron vesicle storage and delivery, organize the neuromuscular junction basement membrane, and provide motor neuron synaptic plasticity, and photoreceptor and neuron synaptic functions. PG-HA networks maintain ionic fluxes and microgradients and tissue compartments that contribute to membrane polarization dynamics essential to neuronal activation and neurotransduction. Hyalectans form neuroprotective perineuronal nets contributing to synaptic plasticity, memory, and cognitive learning. Sialoglycoprotein associated with cones and rods (SPACRCAN), an HA binding CSPG, stabilizes the inter-photoreceptor ECM. HSPGs pikachurin and eyes shut stabilize the photoreceptor synapse aiding in phototransduction and neurotransduction with retinal bipolar neurons crucial to visual acuity. This is achieved through Laminin G motifs in pikachurin, eyes shut, and neurexins that interact with the dystroglycan-cytoskeleton-ECM-stabilizing synaptic interconnections, neuronal interactive specificity, and co-ordination of regulatory action potentials in neural networks.
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 Faculty of Medicine and Health, Royal North Shore Hospital, St. Leonards, New South Wales, Australia
| |
Collapse
|
2
|
Rudolph S, Badura A, Lutzu S, Pathak SS, Thieme A, Verpeut JL, Wagner MJ, Yang YM, Fioravante D. Cognitive-Affective Functions of the Cerebellum. J Neurosci 2023; 43:7554-7564. [PMID: 37940582 PMCID: PMC10634583 DOI: 10.1523/jneurosci.1451-23.2023] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/21/2023] [Accepted: 08/22/2023] [Indexed: 11/10/2023] Open
Abstract
The cerebellum, traditionally associated with motor coordination and balance, also plays a crucial role in various aspects of higher-order function and dysfunction. Emerging research has shed light on the cerebellum's broader contributions to cognitive, emotional, and reward processes. The cerebellum's influence on autonomic function further highlights its significance in regulating motivational and emotional states. Perturbations in cerebellar development and function have been implicated in various neurodevelopmental disorders, including autism spectrum disorder and attention deficit hyperactivity disorder. An increasing appreciation for neuropsychiatric symptoms that arise from cerebellar dysfunction underscores the importance of elucidating the circuit mechanisms that underlie complex interactions between the cerebellum and other brain regions for a comprehensive understanding of complex behavior. By briefly discussing new advances in mapping cerebellar function in affective, cognitive, autonomic, and social processing and reviewing the role of the cerebellum in neuropathology beyond the motor domain, this Mini-Symposium review aims to provide a broad perspective of cerebellar intersections with the limbic brain in health and disease.
Collapse
Affiliation(s)
- Stephanie Rudolph
- Department of Neuroscience, Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, New York, New York 10461
| | - Aleksandra Badura
- Department of Neuroscience, Erasmus MC Rotterdam, Rotterdam, 3015 GD, The Netherlands
| | - Stefano Lutzu
- Department of Neuroscience, Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, New York, New York 10461
| | - Salil Saurav Pathak
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, Minnesota 55812
| | - Andreas Thieme
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, Essen, D-45147, Germany
| | - Jessica L Verpeut
- Department of Psychology, Arizona State University, Tempe, Arizona 85287
| | - Mark J Wagner
- National Institute of Neurological Disorders & Stroke, National Institutes of Health, Bethesda, Maryland 20814
| | - Yi-Mei Yang
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, Minnesota 55812
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455
| | - Diasynou Fioravante
- Center for Neuroscience, University of California-Davis, Davis, California 95618
- Department of Neurobiology, Physiology and Behavior, University of California-Davis, Davis, California 95618
| |
Collapse
|
3
|
Ju S, Shin Y, Han S, Kwon J, Choi TG, Kang I, Kim SS. The Gut-Brain Axis in Schizophrenia: The Implications of the Gut Microbiome and SCFA Production. Nutrients 2023; 15:4391. [PMID: 37892465 PMCID: PMC10610543 DOI: 10.3390/nu15204391] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 10/10/2023] [Accepted: 10/11/2023] [Indexed: 10/29/2023] Open
Abstract
Schizophrenia, a severe mental illness affecting about 1% of the population, manifests during young adulthood, leading to abnormal mental function and behavior. Its multifactorial etiology involves genetic factors, experiences of adversity, infection, and gene-environment interactions. Emerging research indicates that maternal infection or stress during pregnancy may also increase schizophrenia risk in offspring. Recent research on the gut-brain axis highlights the gut microbiome's potential influence on central nervous system (CNS) function and mental health, including schizophrenia. The gut microbiota, located in the digestive system, has a significant role to play in human physiology, affecting immune system development, vitamin synthesis, and protection against pathogenic bacteria. Disruptions to the gut microbiota, caused by diet, medication use, environmental pollutants, and stress, may lead to imbalances with far-reaching effects on CNS function and mental health. Of interest are short-chain fatty acids (SCFAs), metabolic byproducts produced by gut microbes during fermentation. SCFAs can cross the blood-brain barrier, influencing CNS activity, including microglia and cytokine modulation. The dysregulation of neurotransmitters produced by gut microbes may contribute to CNS disorders, including schizophrenia. This review explores the potential relationship between SCFAs, the gut microbiome, and schizophrenia. Our aim is to deepen the understanding of the gut-brain axis in schizophrenia and to elucidate its implications for future research and therapeutic approaches.
Collapse
Affiliation(s)
- Songhyun Ju
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (Y.S.); (S.H.); (J.K.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea;
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Yoonhwa Shin
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (Y.S.); (S.H.); (J.K.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea;
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Sunhee Han
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (Y.S.); (S.H.); (J.K.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea;
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Juhui Kwon
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (Y.S.); (S.H.); (J.K.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea;
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Tae Gyu Choi
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea;
| | - Insug Kang
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (Y.S.); (S.H.); (J.K.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea;
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Sung Soo Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (Y.S.); (S.H.); (J.K.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea;
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
4
|
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
|
5
|
Raj Shekhar B, Rupani K, Raghunath Parkar S, Sunil Nayak A, Vasant Kumbhar B, Khare SP, Menon S, Gawde H, Kumar Das D. Identifying Novel Risk Conferring Genes Involved in Glycosylation Processes with Familial Schizophrenia in an Indian Cohort: Prediction of ADAMTS9 gene Variant for Structural Stability. Gene 2023; 872:147443. [PMID: 37105505 DOI: 10.1016/j.gene.2023.147443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 04/10/2023] [Accepted: 04/21/2023] [Indexed: 04/29/2023]
Abstract
Schizophrenia is a complex neuropsychiatric disorder and heritability is as high as 80% making it the most heritable mental disorder. Although GWAS has identified numerous variants, the pathophysiology is still elusive. Here, an attempt was made to identify genetic risk factors in familial cases of schizophrenia that are associated with a common causative pathway. To achieve this objective, exome sequencing was done in 4 familial cases and identified six unique coding variants in five genes. Among these genes, PIGQ gene has two pathogenic variants, one nonsense and in-frame deletion. One missense variant in GALNT16 and one in GALNT5 have variable damaging score, however, the other variants, in ADAMTS9 and in LTBP4 have the highest damaging score. Further analysis showed that the variant of LTBP4 was not present in the functional domain. The other missense variant in the ADAMTS9 gene was found to be significant and was present in the thrombospondin repeat motif, one of the important motifs. Detailed molecular dynamics simulation study on this variant showed a damaging effect on structural stability. Since, all these genes culminated into the glycosylation process, it was evident that an aberrant glycosylation process may be one of the risk factors. Although, extracellular matrix formation through glycosylation have been shown to be associated, the involvement of ADAMTS9 and PIGQ gene mediated glycosylation has not been reported. In this paper, a novel link between ADAMTS9 and PIGQ gene with schizophrenia have been reported. Therefore, this novel observation has contributed immensely to the existing knowledge on risk factor of Schizophrenia.
Collapse
Affiliation(s)
- Bipin Raj Shekhar
- Genetic Research Centre, ICMR-National Institute for Research in Reproductive and Child Health, Parel, Mumbai, Maharashtra-400012, India; Stem Cell Biology, ICMR-National Institute for Research in Reproductive and Child Health, Parel, Mumbai, Maharashtra-400012, India
| | - Karishma Rupani
- Department of Psychiatry, Seth GS Medical College and KEM Hospital, Parel, Mumbai, Maharashtra-400012, India
| | - Shubhangi Raghunath Parkar
- Department of Psychiatry, Seth GS Medical College and KEM Hospital, Parel, Mumbai, Maharashtra-400012, India
| | - Ajita Sunil Nayak
- Department of Psychiatry, Seth GS Medical College and KEM Hospital, Parel, Mumbai, Maharashtra-400012, India
| | - Bajarang Vasant Kumbhar
- Department of Biological Sciences, Sunandan Divatia School of Science, NMIMS University (Deemed), Mumbai, Maharashtra-400012, India
| | - Satyajeet P Khare
- Symbiosis School of Biological Sciences, Symbiosis International University, Pune, Maharashtra-412115, India
| | - Shyla Menon
- Stem Cell Biology, ICMR-National Institute for Research in Reproductive and Child Health, Parel, Mumbai, Maharashtra-400012, India
| | - Harshavardhan Gawde
- Genetic Research Centre, ICMR-National Institute for Research in Reproductive and Child Health, Parel, Mumbai, Maharashtra-400012, India
| | - Dhanjit Kumar Das
- Stem Cell Biology, ICMR-National Institute for Research in Reproductive and Child Health, Parel, Mumbai, Maharashtra-400012, India.
| |
Collapse
|
6
|
Structural and Functional Deviations of the Hippocampus in Schizophrenia and Schizophrenia Animal Models. Int J Mol Sci 2022; 23:ijms23105482. [PMID: 35628292 PMCID: PMC9143100 DOI: 10.3390/ijms23105482] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/09/2022] [Accepted: 05/11/2022] [Indexed: 01/04/2023] Open
Abstract
Schizophrenia is a grave neuropsychiatric disease which frequently onsets between the end of adolescence and the beginning of adulthood. It is characterized by a variety of neuropsychiatric abnormalities which are categorized into positive, negative and cognitive symptoms. Most therapeutical strategies address the positive symptoms by antagonizing D2-dopamine-receptors (DR). However, negative and cognitive symptoms persist and highly impair the life quality of patients due to their disabling effects. Interestingly, hippocampal deviations are a hallmark of schizophrenia and can be observed in early as well as advanced phases of the disease progression. These alterations are commonly accompanied by a rise in neuronal activity. Therefore, hippocampal formation plays an important role in the manifestation of schizophrenia. Furthermore, studies with animal models revealed a link between environmental risk factors and morphological as well as electrophysiological abnormalities in the hippocampus. Here, we review recent findings on structural and functional hippocampal abnormalities in schizophrenic patients and in schizophrenia animal models, and we give an overview on current experimental approaches that especially target the hippocampus. A better understanding of hippocampal aberrations in schizophrenia might clarify their impact on the manifestation and on the outcome of this severe disease.
Collapse
|
7
|
Wu XL, Yan QJ, Zhu F. Abnormal synaptic plasticity and impaired cognition in schizophrenia. World J Psychiatry 2022; 12:541-557. [PMID: 35582335 PMCID: PMC9048451 DOI: 10.5498/wjp.v12.i4.541] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 07/28/2021] [Accepted: 03/27/2022] [Indexed: 02/06/2023] Open
Abstract
Schizophrenia (SCZ) is a severe mental illness that affects several brain domains with relation to cognition and behaviour. SCZ symptoms are typically classified into three categories, namely, positive, negative, and cognitive. The etiology of SCZ is thought to be multifactorial and poorly understood. Accumulating evidence has indicated abnormal synaptic plasticity and cognitive impairments in SCZ. Synaptic plasticity is thought to be induced at appropriate synapses during memory formation and has a critical role in the cognitive symptoms of SCZ. Many factors, including synaptic structure changes, aberrant expression of plasticity-related genes, and abnormal synaptic transmission, may influence synaptic plasticity and play vital roles in SCZ. In this article, we briefly summarize the morphology of the synapse, the neurobiology of synaptic plasticity, and the role of synaptic plasticity, and review potential mechanisms underlying abnormal synaptic plasticity in SCZ. These abnormalities involve dendritic spines, postsynaptic density, and long-term potentiation-like plasticity. We also focus on cognitive dysfunction, which reflects impaired connectivity in SCZ. Additionally, the potential targets for the treatment of SCZ are discussed in this article. Therefore, understanding abnormal synaptic plasticity and impaired cognition in SCZ has an essential role in drug therapy.
Collapse
Affiliation(s)
- Xiu-Lin Wu
- State Key Laboratory of Virology and Hubei Province Key Laboratory of Allergy and Immunology, Department of Medical Microbiology, School of Medicine, Wuhan University, Wuhan 430071, Hubei Province, China
| | - Qiu-Jin Yan
- State Key Laboratory of Virology and Hubei Province Key Laboratory of Allergy and Immunology, Department of Medical Microbiology, School of Medicine, Wuhan University, Wuhan 430071, Hubei Province, China
| | - Fan Zhu
- State Key Laboratory of Virology and Hubei Province Key Laboratory of Allergy and Immunology, Department of Medical Microbiology, School of Medicine, Wuhan University, Wuhan 430071, Hubei Province, China
| |
Collapse
|
8
|
Dankovich TM, Rizzoli SO. Extracellular Matrix Recycling as a Novel Plasticity Mechanism With a Potential Role in Disease. Front Cell Neurosci 2022; 16:854897. [PMID: 35431813 PMCID: PMC9008140 DOI: 10.3389/fncel.2022.854897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 02/09/2022] [Indexed: 11/13/2022] Open
Abstract
The extracellular matrix (ECM) stabilizes neural circuits and synapses in the healthy brain, while also retaining the ability to be remodeled, to allow synapses to be plastic. A well-described mechanism for ECM remodeling is through the regulated secretion of proteolytic enzymes at the synapse, together with the synthesis of new ECM molecules. The importance of this process is evidenced by the large number of brain disorders that are associated with a dysregulation of ECM-cleaving protease activity. While most of the brain ECM molecules are indeed stable for remarkable time periods, evidence in other cell types, as cancer cells, suggests that at least a proportion of the ECM molecules may be endocytosed regularly, and could even be recycled back to the ECM. In this review, we discuss the involvement of such a mechanism in the brain, under physiological activity conditions and in relation to synapse and brain disease.
Collapse
Affiliation(s)
- Tal M. Dankovich
- Institute of Neuro- and Sensory Physiology, University Medical Center Göttingen, Göttingen, Germany
- International Max Planck Research School for Neurosciences, Göttingen, Germany
- *Correspondence: Tal M. Dankovich,
| | - Silvio O. Rizzoli
- Institute of Neuro- and Sensory Physiology, University Medical Center Göttingen, Göttingen, Germany
- Biostructural Imaging of Neurodegeneration (BIN) Center & Multiscale Bioimaging Excellence Center, Göttingen, Germany
- Silvio O. Rizzoli,
| |
Collapse
|
9
|
Trnski S, Nikolić B, Ilic K, Drlje M, Bobic-Rasonja M, Darmopil S, Petanjek Z, Hranilovic D, Jovanov-Milosevic N. The Signature of Moderate Perinatal Hypoxia on Cortical Organization and Behavior: Altered PNN-Parvalbumin Interneuron Connectivity of the Cingulate Circuitries. Front Cell Dev Biol 2022; 10:810980. [PMID: 35295859 PMCID: PMC8919082 DOI: 10.3389/fcell.2022.810980] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 01/11/2022] [Indexed: 11/29/2022] Open
Abstract
This study was designed in a rat model to determine the hallmarks of possible permanent behavioral and structural brain alterations after a single moderate hypoxic insult. Eighty-two Wistar Han (RccHan: WIST) rats were randomly subjected to hypoxia (pO2 73 mmHg/2 h) or normoxia at the first postnatal day. The substantially increased blood lactate, a significantly decreased cytochrome-C-oxygenase expression in the brain, and depleted subventricular zone suggested a high vulnerability of subset of cell populations to oxidative stress and consequent tissue response even after a single, moderate, hypoxic event. The results of behavioral tests (open-field, hole-board, social-choice, and T-maze) applied at the 30–45th and 70–85th postnatal days revealed significant hyperactivity and a slower pace of learning in rats subjected to perinatal hypoxia. At 3.5 months after hypoxic insult, the histochemical examination demonstrated a significantly increased number of specific extracellular matrix—perineuronal nets and increased parvalbumin expression in a subpopulation of interneurons in the medial and retrosplenial cingulate cortex of these animals. Conclusively, moderate perinatal hypoxia in rats causes a long-lasting reorganization of the connectivity in the cingulate cortex and consequent alterations of related behavioral and cognitive abilities. This non-invasive hypoxia model in the rat successfully and complementarily models the moderate perinatal hypoxic injury in fetuses and prematurely born human babies and may enhance future research into new diagnostic and therapeutic strategies for perinatal medicine.
Collapse
Affiliation(s)
- Sara Trnski
- Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Barbara Nikolić
- Department of Biology, Faculty of Science, University of Zagreb, Zagreb, Croatia
| | - Katarina Ilic
- Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Neuroimaging, BRAIN Centre, Institute of Psychiatry, Psychology, and Neuroscience, King’s College London, London, United Kingdom
| | - Matea Drlje
- Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Mihaela Bobic-Rasonja
- Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Biology, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Sanja Darmopil
- Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Anatomy and Clinical Anatomy, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Zdravko Petanjek
- Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Anatomy and Clinical Anatomy, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Dubravka Hranilovic
- Department of Biology, Faculty of Science, University of Zagreb, Zagreb, Croatia
| | - Natasa Jovanov-Milosevic
- Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Biology, School of Medicine, University of Zagreb, Zagreb, Croatia
- *Correspondence: Natasa Jovanov-Milosevic,
| |
Collapse
|
10
|
Rahman T, Purves-Tyson T, Geddes AE, Huang XF, Newell KA, Weickert CS. N-Methyl-d-Aspartate receptor and inflammation in dorsolateral prefrontal cortex in schizophrenia. Schizophr Res 2022; 240:61-70. [PMID: 34952289 DOI: 10.1016/j.schres.2021.11.045] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 11/02/2021] [Accepted: 11/27/2021] [Indexed: 10/19/2022]
Abstract
Lower N-methyl-d-aspartate receptor (NMDAR) GluN1 subunit levels and heightened neuroinflammation are found in the cortex in schizophrenia. Since neuroinflammation can lead to changes in NMDAR function, it is possible that these observations are linked in schizophrenia. We aimed to extend our previous studies by measuring molecular indices of NMDARs that define key functional properties of this receptor - particularly the ratio of GluN2A and GluN2B subunits - in dorsolateral prefrontal cortex (DLPFC) from schizophrenia and control cases (37/37). We sought to test whether changes in these measures are specific to the subset of schizophrenia cases with high levels of inflammation-related mRNAs, defined as a high inflammatory subgroup. Quantitative autoradiography was used to detect 'functional' NMDARs ([3H]MK-801), GluN1-coupled-GluN2A subunits ([3H]CGP-39653), and GluN1-coupled-GluN2B subunits ([3H]Ifenprodil). Quantitative RT-PCR was used to measure NMDAR subunit transcripts (GRIN1, GRIN2A and GRIN2B). The ratios of GluN2A:GluN2B binding and GRIN2A:GRIN2B mRNAs were calculated as an index of putative NMDAR composition. We found: 1) GluN2A binding, and 2) the ratios of GluN2A:GluN2B binding and GRIN2A:GRIN2B mRNAs were lower in schizophrenia cases versus controls (p < 0.05), and 3) lower GluN2A:GluN2B binding and GRIN2A:GRIN2B mRNA ratios were exaggerated in the high inflammation/schizophrenia subgroup compared to the low inflammation/control subgroup (p < 0.05). No other NMDAR-related indices were significantly changed in the high inflammation/schizophrenia subgroup. This suggests that neuroinflammation may alter NMDAR stoichiometry rather than targeting total NMDAR levels overall, and future studies could aim to determine if anti-inflammatory treatment can alleviate this aspect of NMDAR-related pathology.
Collapse
Affiliation(s)
- Tasnim Rahman
- Neuroscience Research Australia (NeuRA), Sydney, NSW, Australia; School of Psychiatry, University of New South Wales, Sydney, NSW, Australia
| | - Tertia Purves-Tyson
- Neuroscience Research Australia (NeuRA), Sydney, NSW, Australia; School of Psychiatry, University of New South Wales, Sydney, NSW, Australia
| | - Amy E Geddes
- School of Medicine and Molecular Horizons, University of Wollongong, Wollongong, NSW, Australia; Illawarra Health and Medical Research Institute, Wollongong, Australia
| | - Xu-Feng Huang
- School of Medicine and Molecular Horizons, University of Wollongong, Wollongong, NSW, Australia; Illawarra Health and Medical Research Institute, Wollongong, Australia
| | - Kelly A Newell
- School of Medicine and Molecular Horizons, University of Wollongong, Wollongong, NSW, Australia; Illawarra Health and Medical Research Institute, Wollongong, Australia.
| | - Cynthia Shannon Weickert
- Neuroscience Research Australia (NeuRA), Sydney, NSW, Australia; School of Psychiatry, University of New South Wales, Sydney, NSW, Australia; Department of Neuroscience & Physiology, Upstate Medical University, Syracuse, NY, USA.
| |
Collapse
|
11
|
Peng H, Ouyang L, Li D, Li Z, Yuan L, Fan L, Liao A, Li J, Wei Y, Yang Z, Ma X, Chen X, He Y. Short-chain fatty acids in patients with schizophrenia and ultra-high risk population. Front Psychiatry 2022; 13:977538. [PMID: 36578297 PMCID: PMC9790925 DOI: 10.3389/fpsyt.2022.977538] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 11/21/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Individuals who experience the prodromal phase of schizophrenia (SCZ), a common and complex psychiatric disorder, are referred to as ultra-high-risk (UHR) individuals. Short-chain fatty acid (SCFA) is imperative in the microbiota-gut-brain axis and brain function. Accumulating amount of evidence shows the connections between psychiatric disorders and SCFAs. This study aims to explore the underlying roles SCFAs play in SCZ by investigating the association of alterations in SCFAs concentrations with common cognitive functions in both the SCZ and UHR populations. METHODS The study recruited 59 SCZ patients (including 15 participants converted from the UHR group), 51 UHR participants, and 40 healthy controls (HC) within a complete follow-up of 2 years. Results of cognitive functions, which were assessed by utilizing HVLT-R and TMT, and serum concentrations of SCFAs were obtained for all participants and for UHR individuals at the time of their conversion to SCZ. RESULTS Fifteen UHR participants converted to SCZ within a 2-year follow-up. Valeric acid concentration levels were lower in both the baseline of UHR individuals whom later converted to SCZ (p = 0.046) and SCZ patients (p = 0.036) than the HC group. Additionally, there were lower concentrations of caproic acid in the baseline of UHR individuals whom later transitioned to SCZ (p = 0.019) and the UHR group (p = 0.016) than the HC group. Furthermore, the caproic acid levels in the UHR group are significantly positively correlated with immediate memory (r = 0.355, p = 0.011) and negatively correlated with TMT-B (r = -0.366, p = 0.009). Significant differences in levels of acetic acid, butyric acid and isovaleric acid were absent among the three groups and in UHR individuals before and after transition to SCZ. CONCLUSION Our study suggests that alterations in concentrations of SCFAs may be associated with the pathogenesis and the cognitive impairment of schizophrenia. Further researches are warranted to explore this association. The clinical implications of our findings were discussed.
Collapse
Affiliation(s)
- Huiqing Peng
- National Clinical Research Center for Mental Disorders, Department of Psychiatry, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Psychiatry and Mental Health, China National Technology Institute on Mental Disorders, Institute of Mental Health, Hunan Medical Center for Mental Health, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Lijun Ouyang
- National Clinical Research Center for Mental Disorders, Department of Psychiatry, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Psychiatry and Mental Health, China National Technology Institute on Mental Disorders, Institute of Mental Health, Hunan Medical Center for Mental Health, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - David Li
- National Clinical Research Center for Mental Disorders, Department of Psychiatry, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Psychiatry and Mental Health, China National Technology Institute on Mental Disorders, Institute of Mental Health, Hunan Medical Center for Mental Health, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Zongchang Li
- National Clinical Research Center for Mental Disorders, Department of Psychiatry, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Psychiatry and Mental Health, China National Technology Institute on Mental Disorders, Institute of Mental Health, Hunan Medical Center for Mental Health, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Liu Yuan
- National Clinical Research Center for Mental Disorders, Department of Psychiatry, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Psychiatry and Mental Health, China National Technology Institute on Mental Disorders, Institute of Mental Health, Hunan Medical Center for Mental Health, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Lejia Fan
- National Clinical Research Center for Mental Disorders, Department of Psychiatry, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Psychiatry and Mental Health, China National Technology Institute on Mental Disorders, Institute of Mental Health, Hunan Medical Center for Mental Health, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Aijun Liao
- National Clinical Research Center for Mental Disorders, Department of Psychiatry, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Psychiatry and Mental Health, China National Technology Institute on Mental Disorders, Institute of Mental Health, Hunan Medical Center for Mental Health, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Jinguang Li
- National Clinical Research Center for Mental Disorders, Department of Psychiatry, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Psychiatry and Mental Health, China National Technology Institute on Mental Disorders, Institute of Mental Health, Hunan Medical Center for Mental Health, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Yisen Wei
- National Clinical Research Center for Mental Disorders, Department of Psychiatry, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Psychiatry and Mental Health, China National Technology Institute on Mental Disorders, Institute of Mental Health, Hunan Medical Center for Mental Health, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Zihao Yang
- National Clinical Research Center for Mental Disorders, Department of Psychiatry, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Psychiatry and Mental Health, China National Technology Institute on Mental Disorders, Institute of Mental Health, Hunan Medical Center for Mental Health, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Xiaoqian Ma
- National Clinical Research Center for Mental Disorders, Department of Psychiatry, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Psychiatry and Mental Health, China National Technology Institute on Mental Disorders, Institute of Mental Health, Hunan Medical Center for Mental Health, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Xiaogang Chen
- National Clinical Research Center for Mental Disorders, Department of Psychiatry, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Psychiatry and Mental Health, China National Technology Institute on Mental Disorders, Institute of Mental Health, Hunan Medical Center for Mental Health, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Ying He
- National Clinical Research Center for Mental Disorders, Department of Psychiatry, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Psychiatry and Mental Health, China National Technology Institute on Mental Disorders, Institute of Mental Health, Hunan Medical Center for Mental Health, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| |
Collapse
|
12
|
Milbocker KA, Campbell TS, Collins N, Kim S, Smith IF, Roth TL, Klintsova AY. Glia-Driven Brain Circuit Refinement Is Altered by Early-Life Adversity: Behavioral Outcomes. Front Behav Neurosci 2021; 15:786234. [PMID: 34924972 PMCID: PMC8678604 DOI: 10.3389/fnbeh.2021.786234] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/12/2021] [Indexed: 12/12/2022] Open
Abstract
Early-life adversity (ELA), often clinically referred to as "adverse childhood experiences (ACE)," is the exposure to stress-inducing events in childhood that can result in poor health outcomes. ELA negatively affects neurodevelopment in children and adolescents resulting in several behavioral deficits and increasing the risk of developing a myriad of neuropsychiatric disorders later in life. The neurobiological mechanisms by which ELA alters neurodevelopment in childhood have been the focus of numerous reviews. However, a comprehensive review of the mechanisms affecting adolescent neurodevelopment (i.e., synaptic pruning and myelination) is lacking. Synaptic pruning and myelination are glia-driven processes that are imperative for brain circuit refinement during the transition from adolescence to adulthood. Failure to optimize brain circuitry between key brain structures involved in learning and memory, such as the hippocampus and prefrontal cortex, leads to the emergence of maladaptive behaviors including increased anxiety or reduced executive function. As such, we review preclinical and clinical literature to explore the immediate and lasting effects of ELA on brain circuit development and refinement. Finally, we describe a number of therapeutic interventions best-suited to support adolescent neurodevelopment in children with a history of ELA.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Anna Y. Klintsova
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE, United States
| |
Collapse
|
13
|
Crapser JD, Arreola MA, Tsourmas KI, Green KN. Microglia as hackers of the matrix: sculpting synapses and the extracellular space. Cell Mol Immunol 2021; 18:2472-2488. [PMID: 34413489 PMCID: PMC8546068 DOI: 10.1038/s41423-021-00751-3] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 07/26/2021] [Indexed: 02/08/2023] Open
Abstract
Microglia shape the synaptic environment in health and disease, but synapses do not exist in a vacuum. Instead, pre- and postsynaptic terminals are surrounded by extracellular matrix (ECM), which together with glia comprise the four elements of the contemporary tetrapartite synapse model. While research in this area is still just beginning, accumulating evidence points toward a novel role for microglia in regulating the ECM during normal brain homeostasis, and such processes may, in turn, become dysfunctional in disease. As it relates to synapses, microglia are reported to modify the perisynaptic matrix, which is the diffuse matrix that surrounds dendritic and axonal terminals, as well as perineuronal nets (PNNs), specialized reticular formations of compact ECM that enwrap neuronal subsets and stabilize proximal synapses. The interconnected relationship between synapses and the ECM in which they are embedded suggests that alterations in one structure necessarily affect the dynamics of the other, and microglia may need to sculpt the matrix to modify the synapses within. Here, we provide an overview of the microglial regulation of synapses, perisynaptic matrix, and PNNs, propose candidate mechanisms by which these structures may be modified, and present the implications of such modifications in normal brain homeostasis and in disease.
Collapse
Affiliation(s)
- Joshua D. Crapser
- grid.266093.80000 0001 0668 7243Department of Neurobiology and Behavior, University of California, Irvine, CA USA
| | - Miguel A. Arreola
- grid.266093.80000 0001 0668 7243Department of Neurobiology and Behavior, University of California, Irvine, CA USA
| | - Kate I. Tsourmas
- grid.266093.80000 0001 0668 7243Department of Neurobiology and Behavior, University of California, Irvine, CA USA
| | - Kim N. Green
- grid.266093.80000 0001 0668 7243Department of Neurobiology and Behavior, University of California, Irvine, CA USA
| |
Collapse
|
14
|
Perineuronal Nets and Metal Cation Concentrations in the Microenvironments of Fast-Spiking, Parvalbumin-Expressing GABAergic Interneurons: Relevance to Neurodevelopment and Neurodevelopmental Disorders. Biomolecules 2021; 11:biom11081235. [PMID: 34439901 PMCID: PMC8391699 DOI: 10.3390/biom11081235] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/11/2021] [Accepted: 08/14/2021] [Indexed: 12/31/2022] Open
Abstract
Because of their abilities to catalyze generation of toxic free radical species, free concentrations of the redox reactive metals iron and copper are highly regulated. Importantly, desired neurobiological effects of these redox reactive metal cations occur within very narrow ranges of their local concentrations. For example, synaptic release of free copper acts locally to modulate NMDA receptor-mediated neurotransmission. Moreover, within the developing brain, iron is critical to hippocampal maturation and the differentiation of parvalbumin-expressing neurons, whose soma and dendrites are surrounded by perineuronal nets (PNNs). The PNNs are a specialized component of brain extracellular matrix, whose polyanionic character supports the fast-spiking electrophysiological properties of these parvalbumin-expressing GABAergic interneurons. In addition to binding cations and creation of the Donnan equilibrium that support the fast-spiking properties of this subset of interneurons, the complex architecture of PNNs also binds metal cations, which may serve a protective function against oxidative damage, especially of these fast-spiking neurons. Data suggest that pathological disturbance of the population of fast-spiking, parvalbumin-expressing GABAergic inhibitory interneurons occur in at least some clinical presentations, which leads to disruption of the synchronous oscillatory output of assemblies of pyramidal neurons. Increased expression of the GluN2A NMDA receptor subunit on parvalbumin-expressing interneurons is linked to functional maturation of both these neurons and the perineuronal nets that surround them. Disruption of GluN2A expression shows increased susceptibility to oxidative stress, reflected in redox dysregulation and delayed maturation of PNNs. This may be especially relevant to neurodevelopmental disorders, including autism spectrum disorder. Conceivably, binding of metal redox reactive cations by the perineuronal net helps to maintain safe local concentrations, and also serves as a reservoir buffering against second-to-second fluctuations in their concentrations outside of a narrow physiological range.
Collapse
|
15
|
Patrono E, Svoboda J, Stuchlík A. Schizophrenia, the gut microbiota, and new opportunities from optogenetic manipulations of the gut-brain axis. Behav Brain Funct 2021; 17:7. [PMID: 34158061 PMCID: PMC8218443 DOI: 10.1186/s12993-021-00180-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 06/01/2021] [Indexed: 12/18/2022] Open
Abstract
Schizophrenia research arose in the twentieth century and is currently rapidly developing, focusing on many parallel research pathways and evaluating various concepts of disease etiology. Today, we have relatively good knowledge about the generation of positive and negative symptoms in patients with schizophrenia. However, the neural basis and pathophysiology of schizophrenia, especially cognitive symptoms, are still poorly understood. Finding new methods to uncover the physiological basis of the mental inabilities related to schizophrenia is an urgent task for modern neuroscience because of the lack of specific therapies for cognitive deficits in the disease. Researchers have begun investigating functional crosstalk between NMDARs and GABAergic neurons associated with schizophrenia at different resolutions. In another direction, the gut microbiota is getting increasing interest from neuroscientists. Recent findings have highlighted the role of a gut-brain axis, with the gut microbiota playing a crucial role in several psychopathologies, including schizophrenia and autism. There have also been investigations into potential therapies aimed at normalizing altered microbiota signaling to the enteric nervous system (ENS) and the central nervous system (CNS). Probiotics diets and fecal microbiota transplantation (FMT) are currently the most common therapies. Interestingly, in rodent models of binge feeding, optogenetic applications have been shown to affect gut colony sensitivity, thus increasing colonic transit. Here, we review recent findings on the gut microbiota–schizophrenia relationship using in vivo optogenetics. Moreover, we evaluate if manipulating actors in either the brain or the gut might improve potential treatment research. Such research and techniques will increase our knowledge of how the gut microbiota can manipulate GABA production, and therefore accompany changes in CNS GABAergic activity.
Collapse
Affiliation(s)
- Enrico Patrono
- Institute of Physiology of the Czech Academy of Sciences, Videnska, 1830, Prague, 142 20, Czech Republic.
| | - Jan Svoboda
- Institute of Physiology of the Czech Academy of Sciences, Videnska, 1830, Prague, 142 20, Czech Republic
| | - Aleš Stuchlík
- Institute of Physiology of the Czech Academy of Sciences, Videnska, 1830, Prague, 142 20, Czech Republic.
| |
Collapse
|
16
|
Sharmin S, Pradhan J, Zhang Z, Bellingham M, Simmons D, Piper M. Perineuronal net abnormalities in Slc13a4 +/- mice are rescued by postnatal administration of N-acetylcysteine. Exp Neurol 2021; 342:113734. [PMID: 33945789 DOI: 10.1016/j.expneurol.2021.113734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 03/30/2021] [Accepted: 04/29/2021] [Indexed: 10/21/2022]
Abstract
Disruptions to either sulfate supply or sulfation enzymes can affect brain development and have long-lasting effects on brain function, yet our understanding of the molecular mechanisms governing this are incomplete. Perineuronal nets (PNNs) are highly sulfated, specialized extracellular matrix structures that regulate the maturation of synaptic connections and neuronal plasticity. We have previously shown that mice heterozygous for the brain sulfate transporter Slc13a4 have abnormal social interactions, memory, exploratory behaviors, stress and anxiety of postnatal origin, pointing to potential deficits in PNN biology, and implicate SLC13A4 as a critical factor required for regulating normal synaptic connectivity and function. Here, we sought to investigate aberrant PNN formation as a potential mechanism contributing to the functional deficits displayed by Slc13a4+/- mice. Following social interactions, we reveal reduced neuronal activation in the somatosensory cortex of Slc13a4+/- mice, and altered inhibitory and excitatory postsynaptic currents. In line with this, we found a reduction in parvalbumin-expressing neurons decorated with PNNs, as well as reduced expression of markers for PNN maturation. Finally, we reveal that postnatal administration of N-acetylcysteine prevented PNN abnormalities from manifesting in Slc13a4+/- adult animals. Collectively, these data highlight a central role for postnatal SLC13A4 in normal PNN formation, circuit function and subsequent animal behavior.
Collapse
Affiliation(s)
- Sazia Sharmin
- The School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Jonu Pradhan
- The School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Zhe Zhang
- The School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia.
| | - Mark Bellingham
- The School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - David Simmons
- The School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia.
| | - Michael Piper
- The School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia; Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia.
| |
Collapse
|
17
|
Wegrzyn D, Freund N, Faissner A, Juckel G. Poly I:C Activated Microglia Disrupt Perineuronal Nets and Modulate Synaptic Balance in Primary Hippocampal Neurons in vitro. Front Synaptic Neurosci 2021; 13:637549. [PMID: 33708102 PMCID: PMC7940526 DOI: 10.3389/fnsyn.2021.637549] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 02/03/2021] [Indexed: 12/31/2022] Open
Abstract
Perineuronal nets (PNNs) are specialized, reticular structures of the extracellular matrix (ECM) that can be found covering the soma and proximal dendrites of a neuronal subpopulation. Recent studies have shown that PNNs can highly influence synaptic plasticity and are disrupted in different neuropsychiatric disorders like schizophrenia. Interestingly, there is a growing evidence that microglia can promote the loss of PNNs and contribute to neuropsychiatric disorders. Based on this knowledge, we analyzed the impact of activated microglia on hippocampal neuronal networks in vitro. Therefore, primary cortical microglia were cultured and stimulated via polyinosinic-polycytidylic acid (Poly I:C; 50 μg/ml) administration. The Poly I:C treatment induced the expression and secretion of different cytokines belonging to the CCL- and CXCL-motif chemokine family as well as interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α). In addition, the expression of matrix metalloproteinases (MMPs) could be verified via RT-PCR analysis. Embryonic hippocampal neurons were then cultured for 12 days in vitro (DIV) and treated for 24 h with microglial conditioned medium. Interestingly, immunocytochemical staining of the PNN component Aggrecan revealed a clear disruption of PNNs accompanied by a significant increase of glutamatergic and a decrease of γ-aminobutyric acid-(GABA)ergic synapse numbers on PNN wearing neurons. In contrast, PNN negative neurons showed a significant reduction in both, glutamatergic and GABAergic synapses. Electrophysiological recordings were performed via multielectrode array (MEA) technology and unraveled a significantly increased spontaneous network activity that sustained also 24 and 48 h after the administration of microglia conditioned medium. Taken together, we could observe a strong impact of microglial secreted factors on PNN integrity, synaptic plasticity and electrophysiological properties of cultured neurons. Our observations might enhance the understanding of neuron-microglia interactions considering the ECM.
Collapse
Affiliation(s)
- David Wegrzyn
- Department of Cell Morphology and Molecular Neurobiology, Ruhr-University Bochum, Bochum, Germany
| | - Nadja Freund
- Division of Experimental and Molecular Psychiatry, Department of Psychiatry, Psychotherapy and Preventive Medicine, LWL University Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Andreas Faissner
- Department of Cell Morphology and Molecular Neurobiology, Ruhr-University Bochum, Bochum, Germany
| | - Georg Juckel
- Division of Experimental and Molecular Psychiatry, Department of Psychiatry, Psychotherapy and Preventive Medicine, LWL University Hospital, Ruhr-University Bochum, Bochum, Germany
| |
Collapse
|
18
|
Holter MC, Hewitt LT, Nishimura KJ, Knowles SJ, Bjorklund GR, Shah S, Fry NR, Rees KP, Gupta TA, Daniels CW, Li G, Marsh S, Treiman DM, Olive MF, Anderson TR, Sanabria F, Snider WD, Newbern JM. Hyperactive MEK1 Signaling in Cortical GABAergic Neurons Promotes Embryonic Parvalbumin Neuron Loss and Defects in Behavioral Inhibition. Cereb Cortex 2021; 31:3064-3081. [PMID: 33570093 DOI: 10.1093/cercor/bhaa413] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 12/22/2020] [Accepted: 12/23/2020] [Indexed: 12/20/2022] Open
Abstract
Many developmental syndromes have been linked to genetic mutations that cause abnormal ERK/MAPK activity; however, the neuropathological effects of hyperactive signaling are not fully understood. Here, we examined whether hyperactivation of MEK1 modifies the development of GABAergic cortical interneurons (CINs), a heterogeneous population of inhibitory neurons necessary for cortical function. We show that GABAergic-neuron specific MEK1 hyperactivation in vivo leads to increased cleaved caspase-3 labeling in a subpopulation of immature neurons in the embryonic subpallial mantle zone. Adult mutants displayed a significant loss of parvalbumin (PV), but not somatostatin, expressing CINs and a reduction in perisomatic inhibitory synapses on excitatory neurons. Surviving mutant PV-CINs maintained a typical fast-spiking phenotype but showed signs of decreased intrinsic excitability that coincided with an increased risk of seizure-like phenotypes. In contrast to other mouse models of PV-CIN loss, we discovered a robust increase in the accumulation of perineuronal nets, an extracellular structure thought to restrict plasticity. Indeed, we found that mutants exhibited a significant impairment in the acquisition of behavioral response inhibition capacity. Overall, our data suggest PV-CIN development is particularly sensitive to hyperactive MEK1 signaling, which may underlie certain neurological deficits frequently observed in ERK/MAPK-linked syndromes.
Collapse
Affiliation(s)
- Michael C Holter
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - Lauren T Hewitt
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA.,Interdepartmental Neuroscience Graduate Program, University of Texas, Austin, TX 78712, USA
| | - Kenji J Nishimura
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA.,Interdepartmental Neuroscience Graduate Program, University of Texas, Austin, TX 78712, USA
| | - Sara J Knowles
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | | | - Shiv Shah
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - Noah R Fry
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - Katherina P Rees
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - Tanya A Gupta
- Department of Psychology, Arizona State University, Tempe, AZ 85287, USA
| | - Carter W Daniels
- Department of Psychology, Arizona State University, Tempe, AZ 85287, USA.,Department of Psychiatry, Columbia University, New York, NY 10032, USA
| | - Guohui Li
- College of Medicine, University of Arizona, Phoenix, AZ 85004, USA
| | - Steven Marsh
- Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | | | | | - Trent R Anderson
- College of Medicine, University of Arizona, Phoenix, AZ 85004, USA
| | - Federico Sanabria
- Department of Psychology, Arizona State University, Tempe, AZ 85287, USA
| | - William D Snider
- University of North Carolina Neuroscience Center, The University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Jason M Newbern
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| |
Collapse
|
19
|
Zhu X, Grace AA. Prepubertal Environmental Enrichment Prevents Dopamine Dysregulation and Hippocampal Hyperactivity in MAM Schizophrenia Model Rats. Biol Psychiatry 2021; 89:298-307. [PMID: 33357630 PMCID: PMC7927755 DOI: 10.1016/j.biopsych.2020.09.023] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 09/07/2020] [Accepted: 09/20/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Schizophrenia (SCZ) is a neurodevelopmental disorder with a progressive, prolonged course. Early prevention for SCZ is promising but overall lacks support from preclinical evidence. Previous studies have tested environmental enrichment (EE) in certain models of SCZ and discovered a broadly beneficial effect in preventing behavioral abnormalities relevant, yet not specific, to the disorder. Nonetheless, whether EE can prevent dopamine (DA) dysregulation, a hallmark of psychosis and SCZ, had not been tested. METHODS Using the MAM (methylazoxymethanol acetate) rat model of schizophrenia and saline-treated control animals, we investigated the long-term electrophysiological effects of prepubertal (postnatal day 21-40) EE on DA neurons, pyramidal neurons in the ventral hippocampus, and projection neurons in the basolateral amygdala. Anxiety-related behaviors in the elevated plus maze and locomotor responses to amphetamine were also analyzed. RESULTS Prepubertal EE prevented the increased population activity of DA neurons and the associated increase in locomotor response to amphetamine. Prepubertal EE also prevented hyperactivity in the ventral hippocampus but did not prevent hyperactivity in the basolateral amygdala. Anxiety-like behaviors in MAM rats were not ameliorated by prepubertal exposure to EE. CONCLUSIONS Twenty-day prepubertal EE is sufficient to prevent DA hyperresponsivity in the MAM model, measured by electrophysiological recordings and locomotor response to amphetamine. This effect is potentially mediated by normalizing excessive firing in the ventral hippocampus without affecting anxiety-like behaviors and basolateral amygdala firing. This study identified EE as a useful preventative approach that may protect against the pathophysiological development of SCZ.
Collapse
Affiliation(s)
- Xiyu Zhu
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania.
| | - Anthony A Grace
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Psychology, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
20
|
Datta D, Enwright JF, Arion D, Paspalas CD, Morozov YM, Lewis DA, Arnsten AFT. Mapping Phosphodiesterase 4D (PDE4D) in Macaque Dorsolateral Prefrontal Cortex: Postsynaptic Compartmentalization in Layer III Pyramidal Cell Circuits. Front Neuroanat 2020; 14:578483. [PMID: 33328902 PMCID: PMC7714912 DOI: 10.3389/fnana.2020.578483] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 10/06/2020] [Indexed: 12/20/2022] Open
Abstract
cAMP signaling has powerful, negative effects on cognitive functions of the primate dorsolateral prefrontal cortex (dlPFC), opening potassium channels to reduce firing and impair working memory, and increasing tau phosphorylation in aging neurons. This contrasts with cAMP actions in classic circuits, where it enhances plasticity and transmitter release. PDE4 isozymes regulate cAMP actions, and thus have been a focus of research and drug discovery. Previous work has focused on the localization of PDE4A and PDE4B in dlPFC, but PDE4D is also of great interest, as it is the predominant PDE4 isoform in primate association cortex, and PDE4D expression decreases with aging in human dlPFC. Here we used laser-capture microdissection transcriptomics and found that PDE4D message is enriched in pyramidal cells compared to GABAergic PV-interneurons in layer III of the human dlPFC. A parallel study in rhesus macaques using high-spatial resolution immunoelectron microscopy revealed the ultrastructural locations of PDE4D in primate dlPFC with clarity not possible in human post-mortem tissue. PDE4D was especially prominent in dendrites associated with microtubules, mitochondria, and likely smooth endoplasmic reticulum (SER). There was substantial postsynaptic labeling in dendritic spines, associated with the SER spine-apparatus near glutamatergic-like axospinous synapses, but sparse labeling in axon terminals. We also observed dense PDE4D labeling perisynaptically in astroglial leaflets ensheathing glutamatergic connections. These data suggest that PDE4D is strategically positioned to regulate cAMP signaling in dlPFC glutamatergic synapses and circuits, especially in postsynaptic compartments where it is localized to influence cAMP actions on intracellular trafficking, mitochondrial physiology, and internal calcium release.
Collapse
Affiliation(s)
- Dibyadeep Datta
- Department of Neuroscience, Yale School of Medicine, Yale University, New Haven, CT, United States
| | - John F. Enwright
- Department of Psychiatry, Translational Neuroscience Program, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Dominique Arion
- Department of Psychiatry, Translational Neuroscience Program, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Constantinos D. Paspalas
- Department of Neuroscience, Yale School of Medicine, Yale University, New Haven, CT, United States
| | - Yury M. Morozov
- Department of Neuroscience, Yale School of Medicine, Yale University, New Haven, CT, United States
| | - David A. Lewis
- Department of Psychiatry, Translational Neuroscience Program, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Amy F. T. Arnsten
- Department of Neuroscience, Yale School of Medicine, Yale University, New Haven, CT, United States
| |
Collapse
|
21
|
Alonge KM, Mirzadeh Z, Scarlett JM, Logsdon AF, Brown JM, Cabrales E, Chan CK, Kaiyala KJ, Bentsen MA, Banks WA, Guttman M, Wight TN, Morton GJ, Schwartz MW. Hypothalamic perineuronal net assembly is required for sustained diabetes remission induced by fibroblast growth factor 1 in rats. Nat Metab 2020; 2:1025-1033. [PMID: 32895577 PMCID: PMC7572652 DOI: 10.1038/s42255-020-00275-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 08/07/2020] [Indexed: 11/29/2022]
Abstract
We recently showed that perineuronal nets (PNNs) enmesh glucoregulatory neurons in the arcuate nucleus (Arc) of the mediobasal hypothalamus (MBH)1, but whether these PNNs play a role in either the pathogenesis of type 2 diabetes (T2D) or its treatment remains unclear. Here we show that PNN abundance within the Arc is markedly reduced in the Zucker diabetic fatty (ZDF) rat model of T2D, compared with normoglycaemic rats, correlating with altered PNN-associated sulfation patterns of chondroitin sulfate glycosaminoglycans in the MBH. Each of these PNN-associated changes is reversed following a single intracerebroventricular (icv) injection of fibroblast growth factor 1 (FGF1) at a dose that induces sustained diabetes remission in male ZDF rats. Combined with previous work localizing this FGF1 effect to the Arc area2-4, our finding that enzymatic digestion of Arc PNNs markedly shortens the duration of diabetes remission following icv FGF1 injection in these animals identifies these extracellular matrix structures as previously unrecognized participants in the mechanism underlying diabetes remission induced by the central action of FGF1.
Collapse
Affiliation(s)
- Kimberly M Alonge
- University of Washington Medicine Diabetes Institute, University of Washington, Seattle, WA, USA
| | - Zaman Mirzadeh
- Department of Neurosurgery, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Jarrad M Scarlett
- University of Washington Medicine Diabetes Institute, University of Washington, Seattle, WA, USA
- Department of Pediatric Gastroenterology and Hepatology, Seattle Children's Hospital, Seattle, WA, USA
| | - Aric F Logsdon
- Department of Geriatric Research Education and Clinical Center (GRECC), Veterans Affairs Puget Sound Health Care System, University of Washington, Seattle, WA, USA
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Jenny M Brown
- University of Washington Medicine Diabetes Institute, University of Washington, Seattle, WA, USA
| | - Elaine Cabrales
- Department of Neurosurgery, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Christina K Chan
- Matrix Biology Program, Benaroya Research Institute, Seattle, WA, USA
| | - Karl J Kaiyala
- Department of Oral Health Sciences, School of Dentistry, University of Washington, Seattle, WA, USA
| | - Marie A Bentsen
- University of Washington Medicine Diabetes Institute, University of Washington, Seattle, WA, USA
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - William A Banks
- Department of Geriatric Research Education and Clinical Center (GRECC), Veterans Affairs Puget Sound Health Care System, University of Washington, Seattle, WA, USA
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Miklos Guttman
- Department of Medicinal Chemistry, University of Washington, Seattle, WA, USA
| | - Thomas N Wight
- Matrix Biology Program, Benaroya Research Institute, Seattle, WA, USA
| | - Gregory J Morton
- University of Washington Medicine Diabetes Institute, University of Washington, Seattle, WA, USA
| | - Michael W Schwartz
- University of Washington Medicine Diabetes Institute, University of Washington, Seattle, WA, USA.
| |
Collapse
|
22
|
Wegrzyn D, Manitz MP, Kostka M, Freund N, Juckel G, Faissner A. Poly I:C-induced maternal immune challenge reduces perineuronal net area and raises spontaneous network activity of hippocampal neurons in vitro. Eur J Neurosci 2020; 53:3920-3941. [PMID: 32757397 DOI: 10.1111/ejn.14934] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 07/08/2020] [Accepted: 07/23/2020] [Indexed: 12/17/2022]
Abstract
Activation of the maternal immune system (MIA) during gestation is linked to neuropsychiatric diseases like schizophrenia. While many studies address behavioural aspects, less is known about underlying cellular mechanisms. In the following study, BALB/c mice received intraperitoneal injections of polyinosinic-polycytidylic acid (Poly I:C) (20 µg/ml) or saline (0.9%) at gestation day (GD) 9.5 before hippocampal neurons were isolated and cultured from embryonic mice for further analysis. Interestingly, strongest effects were observed when the perineuronal net (PNN) wearing subpopulation of neurons was analysed. Here, a significant reduction of aggrecan staining intensity, area and soma size could be detected. Alterations of PNNs are often linked to neuropsychiatric diseases, changes in synaptic plasticity and in electrophysiology. Utilizing multielectrode array analysis (MEA), we observed a remarkable increase of the spontaneous network activity in neuronal networks after 21 days in vitro (DIV) when mother mice suffered a prenatal immune challenge. As PNNs are associated with GABAergic interneurons, our data indicate that this neuronal subtype might be stronger affected by a prenatal MIA. Degradation or damage of this subtype might cause the hyperexcitability observed in the whole network. In addition, embryonic neurons of the Poly I:C condition developed significantly shorter axons after five days in culture, while dendritic parameters and apoptosis rate remained unchanged. Structural analysis of synapse numbers revealed an increase of postsynaptic density 95 (PSD-95) puncta after 14 DIV and an increase of presynaptic vesicular glutamate transporter (vGlut) puncta after 21 DIV, while inhibitory synaptic proteins were not altered.
Collapse
Affiliation(s)
- David Wegrzyn
- Department of Cell Morphology and Molecular Neurobiology, Ruhr-University Bochum, Bochum, Germany
| | - Marie-Pierre Manitz
- Division of Experimental and Molecular Psychiatry, Department of Psychiatry, Psychotherapy and Preventive Medicine, LWL University Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Michael Kostka
- Department of Cell Morphology and Molecular Neurobiology, Ruhr-University Bochum, Bochum, Germany
| | - Nadja Freund
- Division of Experimental and Molecular Psychiatry, Department of Psychiatry, Psychotherapy and Preventive Medicine, LWL University Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Georg Juckel
- Department of Psychiatry, LWL University Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Andreas Faissner
- Department of Cell Morphology and Molecular Neurobiology, Ruhr-University Bochum, Bochum, Germany
| |
Collapse
|
23
|
Kaushik R, Lipachev N, Matuszko G, Kochneva A, Dvoeglazova A, Becker A, Paveliev M, Dityatev A. Fine structure analysis of perineuronal nets in the ketamine model of schizophrenia. Eur J Neurosci 2020; 53:3988-4004. [PMID: 32510674 DOI: 10.1111/ejn.14853] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 05/12/2020] [Accepted: 05/30/2020] [Indexed: 12/12/2022]
Abstract
Perineuronal nets (PNNs) represent a highly condensed specialized form of brain extracellular matrix (ECM) enwrapping mostly parvalbumin-positive interneurons in the brain in a mesh-like fashion. PNNs not only regulate the onset and completion of the critical period during postnatal brain development, control cell excitability, and synaptic transmission but are also implicated in several brain disorders including schizophrenia. Holes in the perineuronal nets, harboring the synaptic contacts, along with hole-surrounding ECM barrier can be viewed as PNN compartmentalization units that might determine the properties of synapses and heterosynaptic communication. In this study, we developed a novel open-source script for Fiji (ImageJ) to semi-automatically quantify structural alterations of PNNs such as the number of PNN units, area, mean intensity of PNN marker expression in 2D and 3D, shape parameters of PNN units in the ketamine-treated Sprague-Dawley rat model of schizophrenia using high-resolution confocal microscopic images. We discovered that the mean intensity of ECM within PNN units is inversely correlated with the area and the perimeter of the PNN holes. The intensity, size, and shape of PNN units proved to be three major principal factors to describe their variability. Ketamine-treated rats had more numerous but smaller and less circular PNN units than control rats. These parameters allowed to correctly classify individual PNNs as derived from control or ketamine-treated groups with ≈85% reliability. Thus, the proposed multidimensional analysis of PNN units provided a robust and comprehensive morphometric fingerprinting of fine ECM structure abnormalities in the experimental model of schizophrenia.
Collapse
Affiliation(s)
- Rahul Kaushik
- Molecular Neuroplasticity, German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany.,Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
| | - Nikita Lipachev
- Molecular Neuroplasticity, German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany.,Institute of Physics, Kazan Federal University, Kazan, Russia
| | - Gabriela Matuszko
- Molecular Neuroplasticity, German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Anastasia Kochneva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Anastasia Dvoeglazova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Axel Becker
- Institute of Pharmacology and Toxicology, Faculty of Medicine, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Mikhail Paveliev
- Danish Research Institute of Translational Neuroscience, Aarhus University, Aarhus, Denmark.,Neuroscience Center, University of Helsinki, Helsinki, Finland
| | - Alexander Dityatev
- Molecular Neuroplasticity, German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany.,Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany.,Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| |
Collapse
|
24
|
Yu Z, Chen N, Hu D, Chen W, Yuan Y, Meng S, Zhang W, Lu L, Han Y, Shi J. Decreased Density of Perineuronal Net in Prelimbic Cortex Is Linked to Depressive-Like Behavior in Young-Aged Rats. Front Mol Neurosci 2020; 13:4. [PMID: 32116542 PMCID: PMC7025547 DOI: 10.3389/fnmol.2020.00004] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 01/08/2020] [Indexed: 11/13/2022] Open
Abstract
Perineuronal nets (PNNs) are condensed extracellular matrix (ECM) structures regulating developmental plasticity and protecting neurons against oxidative stress. PNN abnormalities have been observed in various psychiatric disorders such as schizophrenia and bipolar disorder, but the relationship between PNN density and depression still remains unclear. In the present study, we examined the density and components of PNNs including aggrecan, neurocan and Tenascin-R in the prelimbic cortex (PrL) after chronic unpredictable mild stress (CUMS). We found that depressive-like behaviors were induced after 30 days of CUMS accompanied by decreases in PNN+ cell density and aggrecan expression in the PrL. In addition, rats subjected to 20 days of CUMS were separated into vulnerable and resilient subpopulations that differ along several behavioral domains. Consistently, the density of PNNs and the expression level of neurocan in the vulnerable group were decreased compared to control and resilient groups. Finally, we examined individual differences based on locomotion in a novel context and classified rats as high responding (HR) and low responding (LR) phenotypes. The density of PNNs and the expression level of neurocan in the LR group were lower than the HR group. Moreover, the LR rats were more susceptible to depressive-like behaviors compared with HR rats. Altogether, these results suggest that the density of PNNs in the PrL is associated with depressive-like behaviors in young-aged rats, and it may serve as a potential endophenotype or therapeutic target for depression.
Collapse
Affiliation(s)
- Zhoulong Yu
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, China.,Department of Pharmacology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Na Chen
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, China.,Department of Pharmacology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Die Hu
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, China.,Department of Pharmacology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Wenxi Chen
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, China.,Department of Pharmacology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Yi Yuan
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, China.,Department of Pharmacology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Shiqiu Meng
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, China
| | - Wen Zhang
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, China
| | - Lin Lu
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, China.,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.,Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China
| | - Ying Han
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, China
| | - Jie Shi
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, China.,The State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, China.,The Key Laboratory for Neuroscience of the Ministry of Education and Health, Peking University, Beijing, China
| |
Collapse
|
25
|
Goodwill HL, Manzano-Nieves G, LaChance P, Teramoto S, Lin S, Lopez C, Stevenson RJ, Theyel BB, Moore CI, Connors BW, Bath KG. Early Life Stress Drives Sex-Selective Impairment in Reversal Learning by Affecting Parvalbumin Interneurons in Orbitofrontal Cortex of Mice. Cell Rep 2019; 25:2299-2307.e4. [PMID: 30485800 PMCID: PMC6310486 DOI: 10.1016/j.celrep.2018.11.010] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 09/18/2018] [Accepted: 10/31/2018] [Indexed: 01/25/2023] Open
Abstract
Poverty, displacement, and parental stress represent potent sources of early life stress (ELS). Stress disproportionately affects females, who are at increased risk for stress-related pathologies associated with cognitive impairment. Mechanisms underlying stress-associated cognitive impairment and enhanced risk of females remain unknown. Here, ELS is associated with impaired rule-reversal (RR) learning in females, but not males. Impaired performance was associated with decreased expression and density of interneurons expressing parvalbumin (PV+) in orbitofrontal cortex (OFC), but not other inter-neuron subtypes. Optogenetic silencing of PV+ inter-neuron activity in OFC of control mice phenocopied RR learning deficits observed in ELS females. Localization of reversal learning deficits to PV+ interneurons in OFC was confirmed by optogenetic studies in which neurons in medial prefrontal cortex (mPFC) were silenced and associated with select deficits in rule-shift learning. Sex-, cell-, and region-specific effects show altered PV+ interneuron development can be a driver of sex differences in cognitive dysfunction. Goodwill et al. investigate the effect of early life stress (ELS) on cognitive development in a mouse model. Using a combination of genetic, histological, optogenetic, and behavioral techniques, they find that ELS leads to female-selective impairments in the ability to engage in rule reversal, but not other forms of attentional learning. Impairments are associated with diminished parvalbumin (PV) expression and a decreased density of PV+ interneurons in the orbitofrontal cortex (OFC).
Collapse
Affiliation(s)
- Haley L Goodwill
- Department of Neuroscience, Brown University, Providence, RI 02912, USA
| | | | - Patrick LaChance
- Department of Neuroscience, Brown University, Providence, RI 02912, USA
| | - Sana Teramoto
- Department of Neuroscience, Brown University, Providence, RI 02912, USA
| | - Shirley Lin
- Department of Neuroscience, Brown University, Providence, RI 02912, USA
| | - Chelsea Lopez
- Department of Neuroscience, Brown University, Providence, RI 02912, USA
| | - Rachel J Stevenson
- Department of Molecular Pharmacology, Physiology, and Biotechnology, Brown University, Providence, RI 02912, USA
| | - Brian B Theyel
- Department of Psychiatry and Human Behavior, Brown University, Providence, RI 02912, USA
| | | | - Barry W Connors
- Department of Neuroscience, Brown University, Providence, RI 02912, USA
| | - Kevin G Bath
- Department of Cognitive, Linguistic and Psychological Sciences, Brown University, Providence, RI 02912, USA.
| |
Collapse
|
26
|
Domínguez S, Rey CC, Therreau L, Fanton A, Massotte D, Verret L, Piskorowski RA, Chevaleyre V. Maturation of PNN and ErbB4 Signaling in Area CA2 during Adolescence Underlies the Emergence of PV Interneuron Plasticity and Social Memory. Cell Rep 2019; 29:1099-1112.e4. [DOI: 10.1016/j.celrep.2019.09.044] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 07/31/2019] [Accepted: 09/13/2019] [Indexed: 12/28/2022] Open
|
27
|
Stolp HB, Fleiss B, Arai Y, Supramaniam V, Vontell R, Birtles S, Yates AG, Baburamani AA, Thornton C, Rutherford M, Edwards AD, Gressens P. Interneuron Development Is Disrupted in Preterm Brains With Diffuse White Matter Injury: Observations in Mouse and Human. Front Physiol 2019; 10:955. [PMID: 31417418 PMCID: PMC6683859 DOI: 10.3389/fphys.2019.00955] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 07/09/2019] [Indexed: 12/18/2022] Open
Abstract
Preterm brain injury, occurring in approximately 30% of infants born <32 weeks gestational age, is associated with an increased risk of neurodevelopmental disorders, such as autism spectrum disorder (ASD) and attention deficit hyperactivity disorder (ADHD). The mechanism of gray matter injury in preterm born children is unclear and likely to be multifactorial; however, inflammation, a high predictor of poor outcome in preterm infants, has been associated with disrupted interneuron maturation in a number of animal models. Interneurons are important for regulating normal brain development, and disruption in interneuron development, and the downstream effects of this, has been implicated in the etiology of neurodevelopmental disorders. Here, we utilize postmortem tissue from human preterm cases with or without diffuse white matter injury (WMI; PMA range: 23+2 to 28+1 for non-WMI group, 26+6 to 30+0 for WMI group, p = 0.002) and a model of inflammation-induced preterm diffuse white matter injury (i.p. IL-1β, b.d., 10 μg/kg/injection in male CD1 mice from P1–5). Data from human preterm infants show deficits in interneuron numbers in the cortex and delayed growth of neuronal arbors at this early stage of development. In the mouse, significant reduction in the number of parvalbumin-positive interneurons was observed from postnatal day (P) 10. This decrease in parvalbumin neuron number was largely rectified by P40, though there was a significantly smaller number of parvalbumin positive cells associated with perineuronal nets in the upper cortical layers. Together, these data suggest that inflammation in the preterm brain may be a contributor to injury of specific interneuron in the cortical gray matter. This may represent a potential target for postnatal therapy to reduce the incidence and/or severity of neurodevelopmental disorders in preterm infants.
Collapse
Affiliation(s)
- Helen B Stolp
- Department for Comparative Biomedical Sciences, Royal Veterinary College, London, United Kingdom.,Department of Perinatal Imaging & Health, Centre for the Developing Brain, School of Biomedical Engineering and Imaging Science, King's College London, London, United Kingdom
| | - Bobbi Fleiss
- Department of Perinatal Imaging & Health, Centre for the Developing Brain, School of Biomedical Engineering and Imaging Science, King's College London, London, United Kingdom.,Université de Paris, NeuroDiderot, Inserm, Paris, France.,School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
| | - Yoko Arai
- Université de Paris, NeuroDiderot, Inserm, Paris, France
| | - Veena Supramaniam
- Department of Perinatal Imaging & Health, Centre for the Developing Brain, School of Biomedical Engineering and Imaging Science, King's College London, London, United Kingdom
| | - Regina Vontell
- Department of Perinatal Imaging & Health, Centre for the Developing Brain, School of Biomedical Engineering and Imaging Science, King's College London, London, United Kingdom.,Department of Neurology, University of Miami, Miller School of Medicine, Miami, FL, United States
| | - Sebastian Birtles
- Department of Perinatal Imaging & Health, Centre for the Developing Brain, School of Biomedical Engineering and Imaging Science, King's College London, London, United Kingdom
| | - Abi G Yates
- Department of Perinatal Imaging & Health, Centre for the Developing Brain, School of Biomedical Engineering and Imaging Science, King's College London, London, United Kingdom.,Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Ana A Baburamani
- Department of Perinatal Imaging & Health, Centre for the Developing Brain, School of Biomedical Engineering and Imaging Science, King's College London, London, United Kingdom
| | - Claire Thornton
- Department for Comparative Biomedical Sciences, Royal Veterinary College, London, United Kingdom.,Department of Perinatal Imaging & Health, Centre for the Developing Brain, School of Biomedical Engineering and Imaging Science, King's College London, London, United Kingdom
| | - Mary Rutherford
- Department of Perinatal Imaging & Health, Centre for the Developing Brain, School of Biomedical Engineering and Imaging Science, King's College London, London, United Kingdom
| | - A David Edwards
- Department of Perinatal Imaging & Health, Centre for the Developing Brain, School of Biomedical Engineering and Imaging Science, King's College London, London, United Kingdom
| | - Pierre Gressens
- Department of Perinatal Imaging & Health, Centre for the Developing Brain, School of Biomedical Engineering and Imaging Science, King's College London, London, United Kingdom.,Université de Paris, NeuroDiderot, Inserm, Paris, France
| |
Collapse
|
28
|
Yukawa T, Iwakura Y, Takei N, Saito M, Watanabe Y, Toyooka K, Igarashi M, Niizato K, Oshima K, Kunii Y, Yabe H, Matsumoto J, Wada A, Hino M, Iritani S, Niwa SI, Takeuchi R, Takahashi H, Kakita A, Someya T, Nawa H. Pathological alterations of chondroitin sulfate moiety in postmortem hippocampus of patients with schizophrenia. Psychiatry Res 2018; 270:940-946. [PMID: 30551347 DOI: 10.1016/j.psychres.2018.10.062] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Revised: 08/27/2018] [Accepted: 10/23/2018] [Indexed: 12/20/2022]
Abstract
Perineuronal nets comprise chondroitin sulfate moieties and their core proteins, and their neuropathological alterations have been implicated in schizophrenia. To explore the molecular mechanism of the perineuronal net impairments in schizophrenia, we measured the immunoreactivity of chondroitin sulfate moieties, major components of perineuronal nets, in three brain regions (postmortem dorsolateral prefrontal cortex, caudate nucleus, and hippocampus) of schizophrenia patients and control subjects. Immunoblotting for chondroitin 4-sulfate and chondroitin 6-sulfate moieties revealed a significant increase in intensity of a 180 kD band of chondroitin 4-sulfate immunoreactivity in the hippocampus of patients, although we detected no significant alteration in their immunoreactivities with any other molecular sizes or in other brain regions. The levels of immunoreactivity were not correlated with postmortem interval, age, or storage time. We failed to find such an increase in a similar molecular range of the chondroitin 4-sulfate immunoreactivity in the hippocampus of the rats chronically treated with haloperidol. These results suggest that the level alteration of the chondroitin 4-sulfate moiety might contribute to the perineuronal net abnormality found in patients with schizophrenia.
Collapse
Affiliation(s)
- Takayuki Yukawa
- Department of Molecular Neurobiology, Brain Research Institute, Niigata University, 1-757, Asahimachi-dori, Chuo-ku Niigata, Niigata 951-8585, Japan; Department of Psychiatry, Graduate School of Medical and Dental Sciences, Niigata University, 1-757, Asahimachi-dori, Chuo-ku Niigata, Niigata 951-8510, Japan
| | - Yuriko Iwakura
- Department of Molecular Neurobiology, Brain Research Institute, Niigata University, 1-757, Asahimachi-dori, Chuo-ku Niigata, Niigata 951-8585, Japan
| | - Nobuyuki Takei
- Department of Molecular Neurobiology, Brain Research Institute, Niigata University, 1-757, Asahimachi-dori, Chuo-ku Niigata, Niigata 951-8585, Japan
| | - Mami Saito
- Department of Molecular Neurobiology, Brain Research Institute, Niigata University, 1-757, Asahimachi-dori, Chuo-ku Niigata, Niigata 951-8585, Japan; Department of Psychiatry, Graduate School of Medical and Dental Sciences, Niigata University, 1-757, Asahimachi-dori, Chuo-ku Niigata, Niigata 951-8510, Japan
| | - Yuichiro Watanabe
- Department of Psychiatry, Graduate School of Medical and Dental Sciences, Niigata University, 1-757, Asahimachi-dori, Chuo-ku Niigata, Niigata 951-8510, Japan
| | - Kazuhiko Toyooka
- Minamihama Hospital, 4540, Shimami-cho, Kita-ku Niigata, Niigata 950-3102, Japan
| | - Michihiro Igarashi
- Department of Neurochemistry and Molecular Cell Biology, Graduate School of Medical and Dental Sciences and Trans-disciplinary Research Program, Niigata University, 1-757, Asahimachi-dori, Chuo-ku Niigata, Niigata 951-8510, Japan
| | - Kazuhiro Niizato
- Tokyo Metropolitan Matsuzawa Hospital, 2-1-1, Kamikitazawa, Setagaya-ku, Tokyo 156-0057, Japan
| | - Kenichi Oshima
- Tokyo Metropolitan Matsuzawa Hospital, 2-1-1, Kamikitazawa, Setagaya-ku, Tokyo 156-0057, Japan
| | - Yasuto Kunii
- Department of Neuropsychiatry, Fukushima Medical University School of Medicine, 1- Hikarigaoka, Fukushima, Fukushima 960-1295, Japan
| | - Hirooki Yabe
- Department of Neuropsychiatry, Fukushima Medical University School of Medicine, 1- Hikarigaoka, Fukushima, Fukushima 960-1295, Japan
| | - Junya Matsumoto
- Department of Neuropsychiatry, Fukushima Medical University School of Medicine, 1- Hikarigaoka, Fukushima, Fukushima 960-1295, Japan
| | - Akira Wada
- Department of Neuropsychiatry, Fukushima Medical University School of Medicine, 1- Hikarigaoka, Fukushima, Fukushima 960-1295, Japan
| | - Mizuki Hino
- Department of Neuropsychiatry, Fukushima Medical University School of Medicine, 1- Hikarigaoka, Fukushima, Fukushima 960-1295, Japan
| | - Shuji Iritani
- Tokyo Metropolitan Matsuzawa Hospital, 2-1-1, Kamikitazawa, Setagaya-ku, Tokyo 156-0057, Japan; Department of Mental Health, Nagoya University Graduate School of Medicine, 65, Tsurumai-cho, Showa-ku, Nagoya, Aichi 466-8550, Japan
| | - Shin-Ichi Niwa
- Department of Neuropsychiatry, Fukushima Medical University School of Medicine, 1- Hikarigaoka, Fukushima, Fukushima 960-1295, Japan
| | - Ryoko Takeuchi
- Pathology and Brain Disease Research Center, Brain Research Institute, Niigata University, 1-757, Asahimachi-dori, Chuo-ku Niigata, Niigata 951-8585, Japan
| | - Hitoshi Takahashi
- Pathology and Brain Disease Research Center, Brain Research Institute, Niigata University, 1-757, Asahimachi-dori, Chuo-ku Niigata, Niigata 951-8585, Japan
| | - Akiyoshi Kakita
- Pathology and Brain Disease Research Center, Brain Research Institute, Niigata University, 1-757, Asahimachi-dori, Chuo-ku Niigata, Niigata 951-8585, Japan
| | - Toshiyuki Someya
- Department of Psychiatry, Graduate School of Medical and Dental Sciences, Niigata University, 1-757, Asahimachi-dori, Chuo-ku Niigata, Niigata 951-8510, Japan
| | - Hiroyuki Nawa
- Department of Molecular Neurobiology, Brain Research Institute, Niigata University, 1-757, Asahimachi-dori, Chuo-ku Niigata, Niigata 951-8585, Japan.
| |
Collapse
|
29
|
Paylor JW, Wendlandt E, Freeman TS, Greba Q, Marks WN, Howland JG, Winship IR. Impaired Cognitive Function after Perineuronal Net Degradation in the Medial Prefrontal Cortex. eNeuro 2018; 5:ENEURO.0253-18.2018. [PMID: 30627657 PMCID: PMC6325561 DOI: 10.1523/eneuro.0253-18.2018] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 11/09/2018] [Accepted: 11/29/2018] [Indexed: 11/21/2022] Open
Abstract
Perineuronal nets (PNNs) are highly organized components of the extracellular matrix that surround a subset of mature neurons in the CNS. These structures play a critical role in regulating neuronal plasticity, particularly during neurodevelopment. Consistent with this role, their presence is associated with functional and structural stability of the neurons they ensheath. A loss of PNNs in the prefrontal cortex (PFC) has been suggested to contribute to cognitive impairment in disorders such as schizophrenia. However, the direct consequences of PNN loss in medial PFC (mPFC) on cognition has not been demonstrated. Here, we examined behavior after disruption of PNNs in mPFC of Long-Evans rats following injection of the enzyme chondroitinase ABC (ChABC). Our data show that ChABC-treated animals were impaired on tests of object oddity perception. Performance in the cross-modal object recognition (CMOR) task was not significantly different for ChABC-treated rats, although ChABC-treated rats were not able to perform above chance levels whereas control rats were. ChABC-treated animals were not significantly different from controls on tests of prepulse inhibition (PPI), set-shifting (SS), reversal learning, or tactile and visual object recognition memory. Posthumous immunohistochemistry confirmed significantly reduced PNNs in mPFC due to ChABC treatment. Moreover, PNN density in the mPFC predicted performance on the oddity task, where higher PNN density was associated with better performance. These findings suggest that PNN loss within the mPFC impairs some aspects of object oddity perception and recognition and that PNNs contribute to cognitive function in young adulthood.
Collapse
Affiliation(s)
- John W. Paylor
- Neurochemical Research Unit, Department of Psychiatry, University of Alberta, Edmonton, T6G 2R3 Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, T6G 2E1 Canada
| | - Eszter Wendlandt
- Neurochemical Research Unit, Department of Psychiatry, University of Alberta, Edmonton, T6G 2R3 Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, T6G 2E1 Canada
| | - Tara S. Freeman
- Neurochemical Research Unit, Department of Psychiatry, University of Alberta, Edmonton, T6G 2R3 Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, T6G 2E1 Canada
| | - Quentin Greba
- Department of Physiology, University of Saskatchewan, Saskatoon, S7N 5E Canada
| | - Wendie N. Marks
- Department of Physiology, University of Saskatchewan, Saskatoon, S7N 5E Canada
| | - John G. Howland
- Department of Physiology, University of Saskatchewan, Saskatoon, S7N 5E Canada
| | - Ian R. Winship
- Neurochemical Research Unit, Department of Psychiatry, University of Alberta, Edmonton, T6G 2R3 Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, T6G 2E1 Canada
| |
Collapse
|
30
|
Tamura H, Shiosaka S, Morikawa S. Trophic modulation of gamma oscillations: The key role of processing protease for Neuregulin-1 and BDNF precursors. Neurochem Int 2018; 119:2-10. [DOI: 10.1016/j.neuint.2017.12.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 11/11/2017] [Accepted: 12/08/2017] [Indexed: 12/26/2022]
|
31
|
Abush H, Ghose S, Van Enkevort EA, Clementz BA, Pearlson GD, Sweeney JA, Keshavan MS, Tamminga CA, Ivleva EI. Associations between adolescent cannabis use and brain structure in psychosis. Psychiatry Res Neuroimaging 2018; 276:53-64. [PMID: 29628270 PMCID: PMC5959798 DOI: 10.1016/j.pscychresns.2018.03.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 03/20/2018] [Accepted: 03/21/2018] [Indexed: 01/28/2023]
Abstract
Associations between cannabis use and psychotic disorders suggest that cannabis may be a contributory risk factor in the neurobiology of psychosis. In this study, we examined brain structure characteristics, total and regional gray matter density (GMD), using Voxel Based Morphometry, in psychotic individuals, stratified by history of cannabis use (total n = 109). We also contrasted GMD estimates in individual diagnostic groups (schizophrenia/bipolar I disorder) with and without history of adolescent cannabis use (ACU). Individuals with psychosis as a whole, both with and without history of ACU, had lower total and regional GMD, compared to healthy controls. ACU was associated with attenuated GMD reductions, compared to non-users, especially in the schizophrenia cases, who showed robust GMD reductions in fronto-temporal and parietal cortex, as well as subcortical regions. Notably, total and regional GMD estimates in individuals with psychosis and ACU were not different from controls with no ACU. These data indicate that the history of ACU in psychotic individuals is associated with attenuated GMD abnormalities. Future investigations targeting potential unique etiological and risk factors associated with psychosis in individuals with ACU may help in understanding of the neurobiology of psychotic disorders and novel treatment options for these individuals.
Collapse
Affiliation(s)
- Hila Abush
- Department of Psychiatry, UT Southwestern Medical Center, Dallas TX, USA
| | - Subroto Ghose
- Department of Psychiatry, UT Southwestern Medical Center, Dallas TX, USA
| | | | - Brett A Clementz
- Departments of Psychology and Neuroscience, BioImaging Research Center, University of Georgia, Athens GA, USA
| | - Godfrey D Pearlson
- Department of Psychiatry, Yale University School of Medicine, New Haven CT, USA; Institute of Living, Hartford Hospital, Hartford CT, USA
| | - John A Sweeney
- Department of Psychiatry, UT Southwestern Medical Center, Dallas TX, USA
| | | | - Carol A Tamminga
- Department of Psychiatry, UT Southwestern Medical Center, Dallas TX, USA
| | - Elena I Ivleva
- Department of Psychiatry, UT Southwestern Medical Center, Dallas TX, USA.
| |
Collapse
|
32
|
Abstract
Background The claustrum (CLA) has been discussed as central to integrated conscious percepts, although recent evidence has emphasized a role in detecting sensory novelty or in amplifying correlated cortical inputs. Objective We report that many neurons in the macaque CLA are ensheathed in perineuronal nets (PNNs), which contribute to synaptic stability and enhance neuronal excitability, among other properties. Design We visualized PNNs by wisteria floribunda agglutinin (WFA) immunohistochemistry, and quantified these in comparison these to parvalbumin+ (PV) subsets and total neurons. Results PNNs ensheath about 11% of the total neurons. These are a range of large, medium, and small neurons, likely corresponding to PV+ and/or other inhibitory interneurons. The PNNs were themselves heterogeneous, consisting of lattice-like, weakly labeled, and diffuse subtypes, and showed some regional preference for the medial CLA. Conclusion The abundant neuronal labeling by PNNs in the CLA suggests an important and nuanced role for inhibition, consistent with recent physiological studies of claustrocortical circuitry. For comparison, diversified inhibition in the reticular nucleus of the thalamus (a pan-inhibitory nucleus, with extensive cortical input) exerts a spectrum of control at different local and global spatiotemporal scales. Further investigation of PNN+ neurons in the macaque CLA offers a potentially important new approach to CLA function, relevant to the human brain both in normal and diseased conditions.
Collapse
Affiliation(s)
- Mihovil Pletikos
- Department of Anatomy and Neurobiology, Boston University School of Medicine, 72 East Concord St., Boston, MA. 02118
| | - Kathleen S Rockland
- Department of Anatomy and Neurobiology, Boston University School of Medicine, 72 East Concord St., Boston, MA. 02118
| |
Collapse
|
33
|
Yoshioka N, Miyata S, Tamada A, Watanabe Y, Kawasaki A, Kitagawa H, Takao K, Miyakawa T, Takeuchi K, Igarashi M. Abnormalities in perineuronal nets and behavior in mice lacking CSGalNAcT1, a key enzyme in chondroitin sulfate synthesis. Mol Brain 2017; 10:47. [PMID: 28982363 PMCID: PMC5629790 DOI: 10.1186/s13041-017-0328-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 09/26/2017] [Indexed: 11/10/2022] Open
Abstract
Chondroitin sulfate (CS) is an important glycosaminoglycan and is mainly found in the extracellular matrix as CS proteoglycans. In the brain, CS proteoglycans are highly concentrated in perineuronal nets (PNNs), which surround synapses and modulate their functions. To investigate the importance of CS, we produced and precisely examined mice that were deficient in the CS synthesizing enzyme, CSGalNAcT1 (T1KO). Biochemical analysis of T1KO revealed that loss of this enzyme reduced the amount of CS by approximately 50% in various brain regions. The amount of CS in PNNs was also diminished in T1KO compared to wild-type mice, although the amount of a major CS proteoglycan core protein, aggrecan, was not changed. In T1KO, we observed abnormalities in several behavioral tests, including the open-field test, acoustic startle response, and social preference. These results suggest that T1 is important for plasticity, probably due to regulation of CS-dependent PNNs, and that T1KO is a good model for investigation of PNNs.
Collapse
Affiliation(s)
- Nozomu Yoshioka
- Department of Neurochemistry and Molecular Cell Biology, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi, Chuo-ku, Niigata, 951-8510, Japan.,Transdiciplinary Research Program, Niigata University, Asahi-machi, Niigata, 951-8510, Japan.,Present address: Divisions of Neurobiology and Anatomy, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Shinji Miyata
- Department of Biochemistry, Kobe Pharmaceutical University, Motoyamakita-machi, Kobe, 658-8558, Japan.,Institute for Advanced Research, Nagoya University, Furo-cho, Nagoya, 464-8601, Japan
| | - Atsushi Tamada
- Department of Neurochemistry and Molecular Cell Biology, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi, Chuo-ku, Niigata, 951-8510, Japan.,Transdiciplinary Research Program, Niigata University, Asahi-machi, Niigata, 951-8510, Japan.,PRESTO, Japan Science and Technology Agency (JST), Chiyoda-ku, Tokyo, 102-0075, Japan
| | - Yumi Watanabe
- Department of Neurochemistry and Molecular Cell Biology, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi, Chuo-ku, Niigata, 951-8510, Japan.,Present address: Divisions of Preventive Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Asami Kawasaki
- Department of Neurochemistry and Molecular Cell Biology, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi, Chuo-ku, Niigata, 951-8510, Japan.,Transdiciplinary Research Program, Niigata University, Asahi-machi, Niigata, 951-8510, Japan
| | - Hiroshi Kitagawa
- Department of Biochemistry, Kobe Pharmaceutical University, Motoyamakita-machi, Kobe, 658-8558, Japan
| | - Keizo Takao
- Section of Behavior Patterns, National Institute of Physiological Sciences, Okazaki, Aichi, 444-8787, Japan.,Division of Experimental Animal Resource and Development, Life Science Research Center, Toyama University, Toyama, 930-0194, Japan
| | - Tsuyoshi Miyakawa
- Section of Behavior Patterns, National Institute of Physiological Sciences, Okazaki, Aichi, 444-8787, Japan.,Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Aichi, 470-1192, Japan
| | - Kosei Takeuchi
- Department of Neurochemistry and Molecular Cell Biology, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi, Chuo-ku, Niigata, 951-8510, Japan.,Department of Medical Biology, School of Medicine, Aichi Medical University, Nagakute, Aichi, 480-1103, Japan
| | - Michihiro Igarashi
- Department of Neurochemistry and Molecular Cell Biology, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi, Chuo-ku, Niigata, 951-8510, Japan. .,Transdiciplinary Research Program, Niigata University, Asahi-machi, Niigata, 951-8510, Japan.
| |
Collapse
|
34
|
Smolyakov G, Dague E, Roux C, Seguelas MH, Galés C, Senard JM, Arvanitis DN. Nanoscale structural mapping as a measure of maturation in the murine frontal cortex. Brain Struct Funct 2017; 223:255-265. [PMID: 28779306 DOI: 10.1007/s00429-017-1486-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 07/26/2017] [Indexed: 01/01/2023]
Abstract
Atomic force microscopy (AFM) is emerging as an innovative tool to phenotype the brain. This study demonstrates the utility of AFM to determine nanomechanical and nanostructural features of the murine dorsolateral frontal cortex from weaning to adulthood. We found an increase in tissue stiffness of the primary somatosensory cortex with age, along with an increased cortical mechanical heterogeneity. To characterize the features potentially responsible for this heterogeneity, we applied AFM scan mode to directly image the topography of thin sections of the primary somatosensory cortical layers II/III, IV and V/VI. Topographical mapping of the cortical layers at successive ages showed progressive smoothing of the surface. Topographical images were also compared with histochemically derived morphological information, which demonstrated the deposition of perineuronal nets, important extracellular components and markers of maturity. Our work demonstrates that high-resolution AFM images can be used to determine the nanostructural properties of cortical maturation, well beyond embryonic and postnatal development. Furthermore, it may offer a new method for brain phenotyping and screening to uncover topographical changes in early stages of neurodegenerative diseases.
Collapse
Affiliation(s)
- G Smolyakov
- LAAS-CNRS, Université de Toulouse, CNRS, Toulouse, France
- ITAV-CNRS, Université de Toulouse, CNRS, Toulouse, France
| | - E Dague
- LAAS-CNRS, Université de Toulouse, CNRS, Toulouse, France.
- ITAV-CNRS, Université de Toulouse, CNRS, Toulouse, France.
| | - C Roux
- LAAS-CNRS, Université de Toulouse, CNRS, Toulouse, France
- ITAV-CNRS, Université de Toulouse, CNRS, Toulouse, France
- Laboratoire Des IMRCP, Université de Toulouse, CNRS UMR 5623, Université de Toulouse, 118 Route de Narbonne, 31062, Toulouse Cedex 9, France
- Institut Des Maladies Métaboliques Et Cardiovasculaires, INSERM, UMR1048, Université de Toulouse, 118 Route de Narbonne, 31062, Toulouse Cedex 9, France
| | - M H Seguelas
- Institut Des Maladies Métaboliques Et Cardiovasculaires, INSERM, UMR1048, Université de Toulouse, 118 Route de Narbonne, 31062, Toulouse Cedex 9, France
| | - C Galés
- ITAV-CNRS, Université de Toulouse, CNRS, Toulouse, France
- Institut Des Maladies Métaboliques Et Cardiovasculaires, INSERM, UMR1048, Université de Toulouse, 118 Route de Narbonne, 31062, Toulouse Cedex 9, France
| | - J M Senard
- Institut Des Maladies Métaboliques Et Cardiovasculaires, INSERM, UMR1048, Université de Toulouse, 118 Route de Narbonne, 31062, Toulouse Cedex 9, France
| | - D N Arvanitis
- Institut Des Maladies Métaboliques Et Cardiovasculaires, INSERM, UMR1048, Université de Toulouse, 118 Route de Narbonne, 31062, Toulouse Cedex 9, France.
| |
Collapse
|
35
|
Wu YT, Adnan A. Effect of Shock-Induced Cavitation Bubble Collapse on the damage in the Simulated Perineuronal Net of the Brain. Sci Rep 2017; 7:5323. [PMID: 28706307 PMCID: PMC5509702 DOI: 10.1038/s41598-017-05790-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 05/23/2017] [Indexed: 01/13/2023] Open
Abstract
The purpose of this study is to conduct modeling and simulation to understand the effect of shock-induced mechanical loading, in the form of cavitation bubble collapse, on damage to the brain's perineuronal nets (PNNs). It is known that high-energy implosion due to cavitation collapse is responsible for corrosion or surface damage in many mechanical devices. In this case, cavitation refers to the bubble created by pressure drop. The presence of a similar damage mechanism in biophysical systems has long being suspected but not well-explored. In this paper, we use reactive molecular dynamics (MD) to simulate the scenario of a shock wave induced cavitation collapse within the perineuronal net (PNN), which is the near-neuron domain of a brain's extracellular matrix (ECM). Our model is focused on the damage in hyaluronan (HA), which is the main structural component of PNN. We have investigated the roles of cavitation bubble location, shockwave intensity and the size of a cavitation bubble on the structural evolution of PNN. Simulation results show that the localized supersonic water hammer created by an asymmetrical bubble collapse may break the hyaluronan. As such, the current study advances current knowledge and understanding of the connection between PNN damage and neurodegenerative disorders.
Collapse
Affiliation(s)
- Yuan-Ting Wu
- Mechanical and Aerospace Engineering, the University of Texas at Arlington, Arlington, 76010, USA
| | - Ashfaq Adnan
- Mechanical and Aerospace Engineering, the University of Texas at Arlington, Arlington, 76010, USA.
| |
Collapse
|
36
|
Sugarcoated Perineuronal Nets Regulate “GABAergic” Transmission: Bittersweet Hypothesis in Autism Spectrum Disorder. Clin Neuropharmacol 2017; 40:120-130. [DOI: 10.1097/wnf.0000000000000209] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
37
|
Morikawa S, Ikegaya Y, Narita M, Tamura H. Activation of perineuronal net-expressing excitatory neurons during associative memory encoding and retrieval. Sci Rep 2017; 7:46024. [PMID: 28378772 PMCID: PMC5380958 DOI: 10.1038/srep46024] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 03/09/2017] [Indexed: 12/21/2022] Open
Abstract
Perineuronal nets (PNNs), proteoglycan-rich extracellular matrix structures, are thought to be expressed around inhibitory neurons and contribute to critical periods of brain function and synaptic plasticity. However, in some specific brain regions such as the amygdala, PNNs were predominantly expressed around excitatory neurons. These neurons were recruited during auditory fear conditioning and memory retrieval. Indeed, the activation of PNN-expressing excitatory neurons predicted cognitive performance.
Collapse
Affiliation(s)
- Shota Morikawa
- Laboratory of Gene Regulation Research, Graduate School of Biological Sciences, Nara Institute of Science and Technology (NAIST), 8916-5, Takayama, Ikoma, Nara 630-0192, Japan.,Life Science Tokyo Advanced Research Center (L-StaR), Hoshi University School of Pharmacy and Pharmaceutical Sciences, 2-4-41, Ebara, Shinagawa-ku, Tokyo 142-8501, Japan
| | - Yuji Ikegaya
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo 113-0033, Japan.,Center for Information and Neural Networks, National Institute of Information and Communications Technology, Suita City, Osaka, 565-0871, Japan
| | - Minoru Narita
- Department of Pharmacology, Hoshi University School of Pharmacy and Pharmaceutical Sciences, 2-4-41, Ebara, Shinagawa-ku, Tokyo 142-8501, Japan
| | - Hideki Tamura
- Life Science Tokyo Advanced Research Center (L-StaR), Hoshi University School of Pharmacy and Pharmaceutical Sciences, 2-4-41, Ebara, Shinagawa-ku, Tokyo 142-8501, Japan
| |
Collapse
|
38
|
Paylor JW, Lins BR, Greba Q, Moen N, de Moraes RS, Howland JG, Winship IR. Developmental disruption of perineuronal nets in the medial prefrontal cortex after maternal immune activation. Sci Rep 2016; 6:37580. [PMID: 27876866 PMCID: PMC5120325 DOI: 10.1038/srep37580] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 11/01/2016] [Indexed: 02/06/2023] Open
Abstract
Maternal infection during pregnancy increases the risk of offspring developing schizophrenia later in life. Similarly, animal models of maternal immune activation (MIA) induce behavioural and anatomical disturbances consistent with a schizophrenia-like phenotype in offspring. Notably, cognitive impairments in tasks dependent on the prefrontal cortex (PFC) are observed in humans with schizophrenia and in offspring after MIA during pregnancy. Recent studies of post-mortem tissue from individuals with schizophrenia revealed deficits in extracellular matrix structures called perineuronal nets (PNNs), particularly in PFC. Given these findings, we examined PNNs over the course of development in a well-characterized rat model of MIA using polyinosinic-polycytidylic acid (polyI:C). We found selective reductions of PNNs in the PFC of polyI:C offspring which did not manifest until early adulthood. These deficits were not associated with changes in parvalbumin cell density, but a decrease in the percentage of parvalbumin cells surrounded by a PNN. Developmental expression of PNNs was also significantly altered in the amygdala of polyI:C offspring. Our results indicate MIA causes region specific developmental abnormalities in PNNs in the PFC of offspring. These findings confirm the polyI:C model replicates neuropathological alterations associated with schizophrenia and may identify novel mechanisms for cognitive and emotional dysfunction in the disorder.
Collapse
Affiliation(s)
- John W Paylor
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, T6G 2E1, AB, Canada.,Neurochemical Research Unit, Department of Psychiatry, University of Alberta, Edmonton, T6G 2B7, AB, Canada
| | - Brittney R Lins
- Department of Physiology, University of Saskatchewan, Saskatoon, S7N 5E5, SK, Canada
| | - Quentin Greba
- Department of Physiology, University of Saskatchewan, Saskatoon, S7N 5E5, SK, Canada
| | - Nicholas Moen
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, T6G 2E1, AB, Canada
| | | | - John G Howland
- Department of Physiology, University of Saskatchewan, Saskatoon, S7N 5E5, SK, Canada
| | - Ian R Winship
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, T6G 2E1, AB, Canada.,Neurochemical Research Unit, Department of Psychiatry, University of Alberta, Edmonton, T6G 2B7, AB, Canada
| |
Collapse
|
39
|
Gottschling C, Geissler M, Springer G, Wolf R, Juckel G, Faissner A. First and second generation antipsychotics differentially affect structural and functional properties of rat hippocampal neuron synapses. Neuroscience 2016; 337:117-130. [DOI: 10.1016/j.neuroscience.2016.08.055] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 08/25/2016] [Accepted: 08/31/2016] [Indexed: 01/23/2023]
|
40
|
Bitanihirwe BKY, Mauney SA, Woo TUW. Weaving a Net of Neurobiological Mechanisms in Schizophrenia and Unraveling the Underlying Pathophysiology. Biol Psychiatry 2016; 80:589-98. [PMID: 27113498 PMCID: PMC5017894 DOI: 10.1016/j.biopsych.2016.03.1047] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 02/14/2016] [Accepted: 03/03/2016] [Indexed: 12/26/2022]
Abstract
Perineuronal nets (PNNs) are enigmatic structures composed of extracellular matrix molecules that encapsulate the soma, dendrites, and axon segments of neurons in a lattice-like fashion. Although most PNNs condense around parvalbumin-expressing gamma-aminobutyric acidergic interneurons, some glutamatergic pyramidal cells in the brain are also surrounded by PNNs. Experimental findings suggest pivotal roles of PNNs in the regulation of synaptic formation and function. Also, an increasing body of evidence links PNN abnormalities to schizophrenia. The number of PNNs progressively increases during postnatal development until plateauing around the period of late adolescence and early adulthood, which temporally coincides with the age of onset of schizophrenia. Given the established role of PNNs in modulating developmental plasticity, the PNN represents a possible candidate for altering the onset and progression of schizophrenia. Similarly, the reported function of PNNs in regulating the trafficking of glutamate receptors places them in a critical position to modulate synaptic pathology, considered a cardinal feature of schizophrenia. We discuss the physiologic role of PNNs in neural function, synaptic assembly, and plasticity as well as how they interface with circuit/system mechanisms of cognition. An integrated understanding of these neurobiological processes should provide a better basis to elucidate how PNN abnormalities influence brain function and contribute to the pathogenesis of neurodevelopmental disorders such as schizophrenia.
Collapse
Affiliation(s)
- Byron K Y Bitanihirwe
- Department of International Health, The Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland
| | - Sarah A Mauney
- Program in Cellular Neuropathology, McLean Hospital, Belmont
| | - Tsung-Ung W Woo
- Program in Cellular Neuropathology, McLean Hospital, Belmont; Department of Psychiatry, Beth Israel Deaconess Medical Center, Boston, Massachusetts; Department of Psychiatry, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
41
|
Trumbore CN. Shear-Induced Amyloid Formation in the Brain: I. Potential Vascular and Parenchymal Processes. J Alzheimers Dis 2016; 54:457-70. [PMID: 27567812 PMCID: PMC5026135 DOI: 10.3233/jad-160027] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/14/2016] [Indexed: 01/05/2023]
Abstract
Shear distortion of amyloid-beta (Aβ) solutions accelerates amyloid cascade reactions that may yield different toxic oligomers than those formed in quiescent solutions. Recent experiments indicate that cerebrospinal fluid (CSF) and interstitial fluid (ISF) containing Aβ flow through narrow brain perivascular pathways and brain parenchyma. This paper suggests that such flow causes shear distortion of Aβ molecules involving conformation changes that may be one of the initiating events in the etiology of Alzheimer's disease. Aβ shearing can occur in or around brain arteries and arterioles and is suggested as the origin of cerebral amyloid angiopathy deposits in cerebrovascular walls. Comparatively low flow rates of ISF within the narrow extracellular spaces (ECS) of the brain parenchyma are suggested as a possible initiating factor in both the formation of neurotoxic Aβ42 oligomers and amyloid fibrils. Aβ42 in slow-flowing ISF can gain significant shear energy at or near the walls of tortuous brain ECS flow paths, promoting the formation of a shear-distorted, excited state hydrophobic Aβ42* conformation. This Aβ42* molecule could possibly be involved in one of two paths, one involving rapid adsorption to a brain membrane surface, ultimately forming neurotoxic oligomers on membranes, and the other ultimately forming plaque within the ECS flow pathways. Rising Aβ concentrations combined with shear at or near critical brain membranes are proposed as contributing factors to Alzheimer's disease neurotoxicity. These hypotheses may be applicable in other neurodegenerative diseases, including tauopathies and alpha-synucleinopathies, in which shear-distorted proteins also may form in the brain ECS.
Collapse
|
42
|
Reduced Labeling of Parvalbumin Neurons and Perineuronal Nets in the Dorsolateral Prefrontal Cortex of Subjects with Schizophrenia. Neuropsychopharmacology 2016; 41:2206-14. [PMID: 26868058 PMCID: PMC4946056 DOI: 10.1038/npp.2016.24] [Citation(s) in RCA: 165] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Revised: 01/06/2016] [Accepted: 02/08/2016] [Indexed: 12/14/2022]
Abstract
Alterations in cortical parvalbumin (PV)-containing neurons, including a reduced density of detectable neurons and lower PV levels, have frequently been reported in the dorsolateral prefrontal cortex (DLPFC) of schizophrenia subjects. Most PV neurons are surrounded by perineuronal nets (PNNs) and the density of PNNs, as detected by Wisteria floribunda agglutinin (WFA) labeling, has been reported to be lower in schizophrenia. However, the nature of these PNN alterations, and their relationship to disease-related changes in PV neurons, has not been assessed. Using confocal microscopy, we quantified the densities and fluorescence intensities of PV neurons and PNNs labeled with WFA or immunoreactive for the major PNN protein, aggrecan, in the DLPFC from schizophrenia and matched comparison subjects. In schizophrenia, the densities of PV cells and of PNNs were not altered; however, the fluorescence intensities of PV immunoreactivity in cell bodies and of WFA labeling and aggrecan immunoreactivity in individual PNNs around PV cells were lower. These findings indicate that the normal complements of PV cells and PNNs are preserved in schizophrenia, but the levels of PV protein and of individual PNN components, especially the carbohydrate moieties on proteoglycans to which WFA binds, are lower. Given the roles of PV neurons in regulating DLPFC microcircuits and of PNNs in regulating PV cellular physiology, the identified alterations in PV neurons and their PNNs could contribute to DLPFC dysfunction in schizophrenia.
Collapse
|
43
|
Filice F, Vörckel KJ, Sungur AÖ, Wöhr M, Schwaller B. Reduction in parvalbumin expression not loss of the parvalbumin-expressing GABA interneuron subpopulation in genetic parvalbumin and shank mouse models of autism. Mol Brain 2016; 9:10. [PMID: 26819149 PMCID: PMC4729132 DOI: 10.1186/s13041-016-0192-8] [Citation(s) in RCA: 161] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 01/20/2016] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND A reduction of the number of parvalbumin (PV)-immunoreactive (PV(+)) GABAergic interneurons or a decrease in PV immunoreactivity was reported in several mouse models of autism spectrum disorders (ASD). This includes Shank mutant mice, with SHANK being one of the most important gene families mutated in human ASD. Similar findings were obtained in heterozygous (PV+/-) mice for the Pvalb gene, which display a robust ASD-like phenotype. Here, we addressed the question whether the observed reduction in PV immunoreactivity was the result of a decrease in PV expression levels and/or loss of the PV-expressing GABA interneuron subpopulation hereafter called "Pvalb neurons". The two alternatives have important implications as they likely result in opposing effects on the excitation/inhibition balance, with decreased PV expression resulting in enhanced inhibition, but loss of the Pvalb neuron subpopulation in reduced inhibition. METHODS Stereology was used to determine the number of Pvalb neurons in ASD-associated brain regions including the medial prefrontal cortex, somatosensory cortex and striatum of PV-/-, PV+/-, Shank1-/- and Shank3B-/- mice. As a second marker for the identification of Pvalb neurons, we used Vicia Villosa Agglutinin (VVA), a lectin recognizing the specific extracellular matrix enwrapping Pvalb neurons. PV protein and Pvalb mRNA levels were determined quantitatively by Western blot analyses and qRT-PCR, respectively. RESULTS Our analyses of total cell numbers in different brain regions indicated that the observed "reduction of PV(+) neurons" was in all cases, i.e., in PV+/-, Shank1-/- and Shank3B-/- mice, due to a reduction in Pvalb mRNA and PV protein, without any indication of neuronal cell decrease/loss of Pvalb neurons evidenced by the unaltered numbers of VVA(+) neurons. CONCLUSIONS Our findings suggest that the PV system might represent a convergent downstream endpoint for some forms of ASD, with the excitation/inhibition balance shifted towards enhanced inhibition due to the down-regulation of PV being a promising target for future pharmacological interventions. Testing whether approaches aimed at restoring normal PV protein expression levels and/or Pvalb neuron function might reverse ASD-relevant phenotypes in mice appears therefore warranted and may pave the way for novel therapeutic treatment strategies.
Collapse
Affiliation(s)
- Federica Filice
- Anatomy, Department of Medicine, University of Fribourg, Route Albert-Gockel 1, CH-1700, Fribourg, Switzerland.
| | - Karl Jakob Vörckel
- Behavioral Neuroscience, Faculty of Psychology, Philipps-University of Marburg, Gutenbergstraβe 18, D-35032, Marburg, Germany.
| | - Ayse Özge Sungur
- Behavioral Neuroscience, Faculty of Psychology, Philipps-University of Marburg, Gutenbergstraβe 18, D-35032, Marburg, Germany.
| | - Markus Wöhr
- Behavioral Neuroscience, Faculty of Psychology, Philipps-University of Marburg, Gutenbergstraβe 18, D-35032, Marburg, Germany.
| | - Beat Schwaller
- Anatomy, Department of Medicine, University of Fribourg, Route Albert-Gockel 1, CH-1700, Fribourg, Switzerland.
| |
Collapse
|
44
|
Morris SE, Vaidyanathan U, Cuthbert BN. Changing the Diagnostic Concept of Schizophrenia: The NIMH Research Domain Criteria Initiative. NEBRASKA SYMPOSIUM ON MOTIVATION. NEBRASKA SYMPOSIUM ON MOTIVATION 2016; 63:225-52. [PMID: 27627829 DOI: 10.1007/978-3-319-30596-7_8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
45
|
In Sickness and in Health: Perineuronal Nets and Synaptic Plasticity in Psychiatric Disorders. Neural Plast 2015; 2016:9847696. [PMID: 26839720 PMCID: PMC4709762 DOI: 10.1155/2016/9847696] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 09/27/2015] [Indexed: 12/25/2022] Open
Abstract
Rapidly emerging evidence implicates perineuronal nets (PNNs) and extracellular matrix (ECM) molecules that compose or interact with PNNs, in the pathophysiology of several psychiatric disorders. Studies on schizophrenia, autism spectrum disorders, mood disorders, Alzheimer's disease, and epilepsy point to the involvement of ECM molecules such as chondroitin sulfate proteoglycans, Reelin, and matrix metalloproteases, as well as their cell surface receptors. In many of these disorders, PNN abnormalities have also been reported. In the context of the “quadripartite” synapse concept, that is, the functional unit composed of the pre- and postsynaptic terminals, glial processes, and ECM, and of the role that PNNs and ECM molecules play in regulating synaptic functions and plasticity, these findings resonate with one of the most well-replicated aspects of the pathology of psychiatric disorders, that is, synaptic abnormalities. Here we review the evidence for PNN/ECM-related pathology in these disorders, with particular emphasis on schizophrenia, and discuss the hypothesis that such pathology may significantly contribute to synaptic dysfunction.
Collapse
|
46
|
Bertero T, Cottrill KA, Annis S, Bhat B, Gochuico BR, Osorio JC, Rosas I, Haley KJ, Corey KE, Chung RT, Nelson Chau B, Chan SY. A YAP/TAZ-miR-130/301 molecular circuit exerts systems-level control of fibrosis in a network of human diseases and physiologic conditions. Sci Rep 2015; 5:18277. [PMID: 26667495 PMCID: PMC4678880 DOI: 10.1038/srep18277] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 10/08/2015] [Indexed: 01/18/2023] Open
Abstract
The molecular origins of fibrosis affecting multiple tissue beds remain incompletely defined. Previously, we delineated the critical role of the control of extracellular matrix (ECM) stiffening by the mechanosensitive microRNA-130/301 family, as activated by the YAP/TAZ co-transcription factors, in promoting pulmonary hypertension (PH). We hypothesized that similar mechanisms may dictate fibrosis in other tissue beds beyond the pulmonary vasculature. Employing an in silico combination of microRNA target prediction, transcriptomic analysis of 137 human diseases and physiologic states, and advanced gene network modeling, we predicted the microRNA-130/301 family as a master regulator of fibrotic pathways across a cohort of seemingly disparate diseases and conditions. In two such diseases (pulmonary fibrosis and liver fibrosis), inhibition of microRNA-130/301 prevented the induction of ECM modification, YAP/TAZ, and downstream tissue fibrosis. Thus, mechanical forces act through a central feedback circuit between microRNA-130/301 and YAP/TAZ to sustain a common fibrotic phenotype across a network of human physiologic and pathophysiologic states. Such re-conceptualization of interconnections based on shared systems of disease and non-disease gene networks may have broad implications for future convergent diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Thomas Bertero
- Divisions of Cardiovascular and Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA
| | - Katherine A. Cottrill
- Divisions of Cardiovascular and Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA
| | - Sofia Annis
- Divisions of Cardiovascular and Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA
| | | | | | - Juan C. Osorio
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Ivan Rosas
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Kathleen J. Haley
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Kathleen E. Corey
- Liver Center and Gastrointestinal Division, Massachusetts General Hospital, Boston, MA, USA
| | - Raymond T. Chung
- Liver Center and Gastrointestinal Division, Massachusetts General Hospital, Boston, MA, USA
| | | | - Stephen Y. Chan
- Divisions of Cardiovascular and Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA
| |
Collapse
|
47
|
Athanas K, Mauney SL, Woo TUW. Increased extracellular clusterin in the prefrontal cortex in schizophrenia. Schizophr Res 2015; 169:381-385. [PMID: 26482819 PMCID: PMC4681675 DOI: 10.1016/j.schres.2015.10.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 09/29/2015] [Accepted: 10/03/2015] [Indexed: 11/29/2022]
Abstract
The expression of the gene that encodes clusterin, a glycoprotein that has been implicated in the regulation of many cellular processes, has previously been found in gene expression profiling studies to be among the most significantly differentially expressed genes in pyramidal and parvalbumin-containing inhibitory neurons in the cerebral cortex in subjects with schizophrenia. In this study, we investigated whether clusterin may also be dysregulated at the protein level in schizophrenia subjects. We found that, although the intracellular amount of clusterin may be unchanged, the level of extracellular, secreted clusterin appears to be significantly increased in schizophrenia subjects. It is speculated that this finding may represent a neuroprotective response to pathophysiological events that underlie schizophrenia.
Collapse
Affiliation(s)
- Katina Athanas
- Laboratory of Cellular Neuropathology, McLean Hospital Belmont, MA 02478
| | - Sarah L. Mauney
- Laboratory of Cellular Neuropathology, McLean Hospital Belmont, MA 02478
| | - Tsung-Ung W. Woo
- Laboratory of Cellular Neuropathology, McLean Hospital Belmont, MA 02478,Department of Psychiatry, Beth Israel Deaconess Medical Center, Boston, MA 02215,Department of Psychiatry, Harvard Medical School, Boston, MA 02215
| |
Collapse
|
48
|
Ashbrook DG, Williams RW, Lu L, Hager R. A cross-species genetic analysis identifies candidate genes for mouse anxiety and human bipolar disorder. Front Behav Neurosci 2015; 9:171. [PMID: 26190982 PMCID: PMC4486840 DOI: 10.3389/fnbeh.2015.00171] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 06/18/2015] [Indexed: 12/21/2022] Open
Abstract
Bipolar disorder (BD) is a significant neuropsychiatric disorder with a lifetime prevalence of ~1%. To identify genetic variants underlying BD genome-wide association studies (GWAS) have been carried out. While many variants of small effect associated with BD have been identified few have yet been confirmed, partly because of the low power of GWAS due to multiple comparisons being made. Complementary mapping studies using murine models have identified genetic variants for behavioral traits linked to BD, often with high power, but these identified regions often contain too many genes for clear identification of candidate genes. In the current study we have aligned human BD GWAS results and mouse linkage studies to help define and evaluate candidate genes linked to BD, seeking to use the power of the mouse mapping with the precision of GWAS. We use quantitative trait mapping for open field test and elevated zero maze data in the largest mammalian model system, the BXD recombinant inbred mouse population, to identify genomic regions associated with these BD-like phenotypes. We then investigate these regions in whole genome data from the Psychiatric Genomics Consortium's bipolar disorder GWAS to identify candidate genes associated with BD. Finally we establish the biological relevance and pathways of these genes in a comprehensive systems genetics analysis. We identify four genes associated with both mouse anxiety and human BD. While TNR is a novel candidate for BD, we can confirm previously suggested associations with CMYA5, MCTP1, and RXRG. A cross-species, systems genetics analysis shows that MCTP1, RXRG, and TNR coexpress with genes linked to psychiatric disorders and identify the striatum as a potential site of action. CMYA5, MCTP1, RXRG, and TNR are associated with mouse anxiety and human BD. We hypothesize that MCTP1, RXRG, and TNR influence intercellular signaling in the striatum.
Collapse
Affiliation(s)
- David G Ashbrook
- Computational and Evolutionary Biology, Faculty of Life Sciences, University of Manchester Manchester, UK
| | - Robert W Williams
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, University of Tennessee Memphis, TN, USA
| | - Lu Lu
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, University of Tennessee Memphis, TN, USA ; Jiangsu Key Laboratory of Neuroregeneration, Nantong University Nantong, China
| | - Reinmar Hager
- Computational and Evolutionary Biology, Faculty of Life Sciences, University of Manchester Manchester, UK
| |
Collapse
|
49
|
Keshavan MS, Mehta UM, Padmanabhan JL, Shah JL. Dysplasticity, metaplasticity, and schizophrenia: Implications for risk, illness, and novel interventions. Dev Psychopathol 2015; 27:615-35. [PMID: 25997775 PMCID: PMC6283269 DOI: 10.1017/s095457941500019x] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
In this paper, we review the history of the concept of neuroplasticity as it relates to the understanding of neuropsychiatric disorders, using schizophrenia as a case in point. We briefly review the myriad meanings of the term neuroplasticity, and its neuroscientific basis. We then review the evidence for aberrant neuroplasticity and metaplasticity associated with schizophrenia as well as the risk for developing this illness, and discuss the implications of such understanding for prevention and therapeutic interventions. We argue that the failure and/or altered timing of plasticity of critical brain circuits might underlie cognitive and deficit symptoms, and may also lead to aberrant plastic reorganization in other circuits, leading to affective dysregulation and eventually psychosis. This "dysplastic" model of schizophrenia can suggest testable etiology and treatment-relevant questions for the future.
Collapse
Affiliation(s)
- Matcheri S. Keshavan
- Department of Psychiatry, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, United States
| | - Urvakhsh Meherwan Mehta
- Department of Psychiatry, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, United States
- Department of Psychiatry, National Institute of Mental Health and Neurosciences, Bangalore, India
| | - Jaya L. Padmanabhan
- Department of Psychiatry, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, United States
| | - Jai L. Shah
- Douglas Hospital Research Center and Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
50
|
Bitanihirwe BKY, Woo TUW. Transcriptional dysregulation of γ-aminobutyric acid transporter in parvalbumin-containing inhibitory neurons in the prefrontal cortex in schizophrenia. Psychiatry Res 2014; 220:1155-9. [PMID: 25312391 PMCID: PMC4447488 DOI: 10.1016/j.psychres.2014.09.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Revised: 08/15/2014] [Accepted: 09/23/2014] [Indexed: 12/18/2022]
Abstract
Parvalbumin (PV)-containing neurons are functionally compromised in schizophrenia. Using double in situ hybridization in postmortem human prefrontal cortex, we found that the messenger RNA (mRNA) for the γ-aminobutyric acid (GABA) transporter GAT-1 was undetectable in 22-41% of PV neurons in layers 3-4 in schizophrenia. In the remaining PV neurons with detectable GAT-1 mRNA, transcript expression was decreased by 26% in layer 3. Hence, the dysfunction of PV neurons involves the molecular dysregulation of presynaptic GABA reuptake.
Collapse
Affiliation(s)
- Byron K. Y. Bitanihirwe
- System and Cell Biology of Neurodegeneration, University of Zürich, Zürich, Switzerland,Program in Cellular Neuropathology, McLean Hospital, Belmont, Massachusetts, USA
| | - Tsung-Ung W. Woo
- Program in Cellular Neuropathology, McLean Hospital, Belmont, Massachusetts, USA,Department of Psychiatry, Harvard Medical School, Boston, Massachusetts, USA,Department of Psychiatry, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| |
Collapse
|