1
|
Simpson MA. Impacts of Hyaluronan on Extracellular Vesicle Production and Signaling. Cells 2025; 14:139. [PMID: 39851567 PMCID: PMC11763598 DOI: 10.3390/cells14020139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 01/14/2025] [Accepted: 01/15/2025] [Indexed: 01/26/2025] Open
Abstract
Hyaluronan (HA) is a critical component of cell and tissue matrices and an important signaling molecule. The enzymes that synthesize and process HA, as well as the HA receptors through which the signaling properties of HA are transmitted, have been identified in extracellular vesicles and implicated in context-specific processes associated with health and disease. The goal of this review is to present a comprehensive summary of the research on HA and its related receptors and enzymes in extracellular vesicle biogenesis and the cellular responses to vesicles bearing these extracellular matrix modulators. When present in extracellular vesicles, HA is assumed to be on the outside of the vesicle and is sometimes found associated with CD44 or the HAS enzyme itself. Hyaluronidases may be inside the vesicles or present on the vesicle surface via a transmembrane domain or GPI linkage. The implication of presenting these signals in extracellular vesicles is that there is a greater range of systemic distribution and more complex delivery media than previously thought for secreted HA or hyaluronidase alone. Understanding the context for these HA signals offers new diagnostic and therapeutic insight.
Collapse
Affiliation(s)
- Melanie A Simpson
- Department of Molecular and Structural Biochemistry, North Carolina State University, Raleigh, NC 27695-7622, USA
| |
Collapse
|
2
|
Sterin I, Niazi A, Kim J, Park J, Park S. Dynamic Organization of Neuronal Extracellular Matrix Revealed by HaloTag-HAPLN1. J Neurosci 2024; 44:e0666242024. [PMID: 39251350 PMCID: PMC11502233 DOI: 10.1523/jneurosci.0666-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 08/04/2024] [Accepted: 08/30/2024] [Indexed: 09/11/2024] Open
Abstract
The brain's extracellular matrix (ECM) regulates neuronal plasticity and animal behavior. ECM staining shows a net-like structure around a subset of neurons, a ring-like structure at the nodes of Ranvier, and diffuse staining in the interstitial matrix. However, understanding the structural features of ECM deposition across various neuronal types and subcellular compartments remains limited. To visualize the organization pattern and assembly process of the hyaluronan-scaffolded ECM in the brain, we fused a HaloTag to hyaluronan proteoglycan link protein 1, which links hyaluronan and proteoglycans. Expression or application of the probe in primary rat neuronal cultures enables us to identify spatial and temporal regulation of ECM deposition and heterogeneity in ECM aggregation among neuronal populations. Dual-color birthdating shows the ECM assembly process in culture and in vivo. Sparse expression in mouse brains of either sex reveals detailed ECM architectures around excitatory neurons and developmentally regulated dendritic ECM. Our study uncovers extensive structural features of the brain's ECM, suggesting diverse roles in regulating neuronal plasticity.
Collapse
Affiliation(s)
- Igal Sterin
- Department of Neurobiology, University of Utah, Salt Lake City, Utah 84112
| | - Ava Niazi
- Department of Neurobiology, University of Utah, Salt Lake City, Utah 84112
- Neuroscience Program, University of Utah, Salt Lake City, Utah 84112
| | - Jennifer Kim
- Department of Neurobiology, University of Utah, Salt Lake City, Utah 84112
| | - Joosang Park
- Department of Neurobiology, University of Utah, Salt Lake City, Utah 84112
| | - Sungjin Park
- Department of Neurobiology, University of Utah, Salt Lake City, Utah 84112
| |
Collapse
|
3
|
Zhang N, Song B, Bai P, Du L, Chen L, Xu Y, Zeng T. Perineuronal nets' role in metabolism. Am J Physiol Endocrinol Metab 2024; 327:E411-E421. [PMID: 39140971 DOI: 10.1152/ajpendo.00154.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 07/31/2024] [Accepted: 08/05/2024] [Indexed: 08/15/2024]
Abstract
Perineuronal nets (PNNs), specialized extracellular matrix (ECM) structures that envelop neurons, have recently been recognized as key players in the regulation of metabolism. This review explores the growing body of knowledge concerning PNNs and their role in metabolic control, drawing insights from recent research and relevant studies. The pivotal role of PNNs in the context of energy balance and whole body blood glucose is examined. This review also highlights novel findings, including the effects of astroglia, microglia, sex and gonadal hormones, nutritional regulation, circadian rhythms, and age on PNNs dynamics. These findings illuminate the complex and multifaceted role of PNNs in metabolic health.
Collapse
Affiliation(s)
- Nan Zhang
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Diabetes and Metabolic Disease Clinical Research Center of Hubei Province, Wuhan, People's Republic of China
- Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Hubei Branch of National Center for Clinical Medical Research of Metabolic Diseases, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Beite Song
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Diabetes and Metabolic Disease Clinical Research Center of Hubei Province, Wuhan, People's Republic of China
- Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Hubei Branch of National Center for Clinical Medical Research of Metabolic Diseases, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Peng Bai
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Li Du
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Lulu Chen
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Diabetes and Metabolic Disease Clinical Research Center of Hubei Province, Wuhan, People's Republic of China
- Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Hubei Branch of National Center for Clinical Medical Research of Metabolic Diseases, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Yong Xu
- Department of Pediatrics, USDA/ARS Children's Nutrition Research Center, Baylor College of Medicine, Houston, Texas, United States
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, United States
- Department of Medicine, Baylor College of Medicine, Houston, Texas, United States
| | - Tianshu Zeng
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Diabetes and Metabolic Disease Clinical Research Center of Hubei Province, Wuhan, People's Republic of China
- Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Hubei Branch of National Center for Clinical Medical Research of Metabolic Diseases, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| |
Collapse
|
4
|
Lisboa JRF, Costa O, Pakes GH, Colodete DAE, Gomes FV. Perineuronal net density in schizophrenia: A systematic review of postmortem brain studies. Schizophr Res 2024; 271:100-109. [PMID: 39018984 DOI: 10.1016/j.schres.2024.07.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 05/07/2024] [Accepted: 07/07/2024] [Indexed: 07/19/2024]
Abstract
BACKGROUND The onset of schizophrenia is concurrent with multiple key processes of brain development, such as the maturation of inhibitory networks. Some of these processes are proposed to depend on the development of perineuronal nets (PNNs), a specialized extracellular matrix structure that surrounds preferentially parvalbumin-containing GABAergic interneurons (PVIs). PNNs are fundamental to the postnatal experience-dependent maturation of inhibitory brain circuits. PNN abnormalities have been proposed as a core pathophysiological finding in SCZ, being linked to widespread consequences on circuit disruptions underlying SCZ symptoms. OBJECTIVE Here, we systematically evaluate PNN density in postmortem brain studies of subjects with SCZ. METHODS A systematic search in 3 online databases (PubMed, Embase, and Scopus) and qualitative review analysis of case-control studies reporting on PNN density in the postmortem brain of subjects with SCZ were performed. RESULTS Results consisted of 7 studies that were included in the final analysis. The specific brain regions investigated in the studies varied, with most attention given to the dorsolateral prefrontal cortex (DLPFC; 3 studies) and amygdala (2 studies). Findings were mostly positive for reduced PNN density in SCZ, with 6 of the 7 studies reporting significant reductions and one reporting a tendency towards reduced PNN density. Overall, tissue processing methodologies were heterogeneous. CONCLUSIONS Despite few studies, PNN density was consistently reduced in SCZ across different brain regions. These findings support evidence that implicates deficits in PNN density in the pathophysiology of SCZ. However, more studies, preferably using similar methodological approaches as well as replication of findings, are needed.
Collapse
Affiliation(s)
- João Roberto F Lisboa
- Department of Neuroscience and Behavior, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.
| | - Olga Costa
- Department of Neuroscience and Behavior, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Gustavo Henrique Pakes
- Department of Neuroscience and Behavior, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Debora Akemi E Colodete
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Felipe V Gomes
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
5
|
Vo GV, Rao KM, Chung I, Ha CS, An SSA, Yun YH. Derivatization of Hyaluronan to Target Neuroblastoma and Neuroglioma Expressing CD44. Pharmaceutics 2024; 16:836. [PMID: 38931956 PMCID: PMC11207210 DOI: 10.3390/pharmaceutics16060836] [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: 04/26/2024] [Revised: 05/18/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024] Open
Abstract
Therapeutics for actively targeting over-expressed receptors are of great interest because the majority of diseased tissues originate from normal cells and do not possess a unique receptor from which they can be differentiated. One such receptor is CD44, which has been shown to be highly overexpressed in many breast cancers and other types of cancer cells. While CD44 has been documented to express low levels in normal adult neurons, astrocytes, and microglia, this receptor may be overexpressed by neuroblastoma and neuroglioma. If differential expression exists between normal and cancerous cells, hyaluronan (HA) could be a useful carrier that targets carcinomas. Thus, HA was conjugated with resveratrol (HA-R), and its efficacy was tested on cortical-neuroblastoma hybrid, neuroblastoma, and neuroglioma cells. Confocal and flow cytometry showed these cells express CD44 and are able to bind and uptake HA-R. The toxicity of HA-R correlated well with CD44 expression in this study. Therefore, conjugating resveratrol and other chemotherapeutics to HA could minimize the side effects for normal cells within the brain and nervous system and could be a viable strategy for developing targeted therapies.
Collapse
Affiliation(s)
- Giau Van Vo
- Department of Bionano Technology, Gachon Bionano Research Institute, Gachon University, 1342 Seongnam-daero, Sujeong-gu, Seongnam-si 13120, Gyeonggi-do, Republic of Korea;
- Degenerative Diseases Program, Sanford Burnham Prebys Medical Discovery Institute, San Diego, CA 92037, USA
| | - Kummara Madhusudana Rao
- School of Chemical Engineering, Yeungnam University, Gyeongsan-si 38541, Gyeongbuk-do, Republic of Korea;
- Department of Polymer Science and Engineering, Pusan National University, Busan 46241, Gyeongsangnam-do, Republic of Korea; (I.C.); (C.-S.H.)
| | - Ildoo Chung
- Department of Polymer Science and Engineering, Pusan National University, Busan 46241, Gyeongsangnam-do, Republic of Korea; (I.C.); (C.-S.H.)
| | - Chang-Sik Ha
- Department of Polymer Science and Engineering, Pusan National University, Busan 46241, Gyeongsangnam-do, Republic of Korea; (I.C.); (C.-S.H.)
| | - Seong Soo A. An
- Degenerative Diseases Program, Sanford Burnham Prebys Medical Discovery Institute, San Diego, CA 92037, USA
| | - Yang H. Yun
- Department of Biomedical Engineering, The University of Akron, Akron, OH 44325-0302, USA
| |
Collapse
|
6
|
Sterin I, Niazi A, Kim J, Park J, Park S. Novel extracellular matrix architecture on excitatory neurons revealed by HaloTag-HAPLN1. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.29.587384. [PMID: 38585814 PMCID: PMC10996768 DOI: 10.1101/2024.03.29.587384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
The brain's extracellular matrix (ECM) regulates neuronal plasticity and animal behavior. ECM staining shows an aggregated pattern in a net-like structure around a subset of neurons and diffuse staining in the interstitial matrix. However, understanding the structural features of ECM deposition across various neuronal types and subcellular compartments remains limited. To visualize the organization pattern and assembly process of the hyaluronan-scaffolded ECM in the brain, we fused a HaloTag to HAPLN1, which links hyaluronan and proteoglycans. Expression or application of the probe enables us to identify spatial and temporal regulation of ECM deposition and heterogeneity in ECM aggregation among neuronal populations. Dual-color birthdating shows the ECM assembly process in culture and in vivo. Sparse expression in vivo reveals novel forms of ECM architecture around excitatory neurons and developmentally regulated dendritic ECM. Overall, our study uncovers extensive structural features of the brain' ECM, suggesting diverse roles in regulating neuronal plasticity.
Collapse
Affiliation(s)
- Igal Sterin
- Department of Neurobiology, University of Utah, Salt Lake City, Utah, USA
| | - Ava Niazi
- Department of Neurobiology, University of Utah, Salt Lake City, Utah, USA
- Neuroscience Program, University of Utah, Salt Lake City, Utah, USA
| | - Jennifer Kim
- Department of Neurobiology, University of Utah, Salt Lake City, Utah, USA
| | - Joosang Park
- Department of Neurobiology, University of Utah, Salt Lake City, Utah, USA
| | - Sungjin Park
- Department of Neurobiology, University of Utah, Salt Lake City, Utah, USA
| |
Collapse
|
7
|
Zhang YN, Wu Q, Zhang NN, Chen HS. Ischemic Preconditioning Alleviates Cerebral Ischemia-Reperfusion Injury by Interfering With Glycocalyx. Transl Stroke Res 2023; 14:929-940. [PMID: 36168082 DOI: 10.1007/s12975-022-01081-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 08/08/2022] [Accepted: 09/12/2022] [Indexed: 10/14/2022]
Abstract
Ischemic preconditioning (IPC) could protect the blood-brain barrier (BBB), but the underlying mechanism is not well understood. This preclinical study aimed to investigate whether glycocalyx could be involved in the neuroprotective effect of IPC on cerebral ischemia-reperfusion injury (IRI) and the possible mechanism in rat middle cerebral artery occlusion/reperfusion (MCAO/R) model. Neurological deficit scores, infarct volume, and brain edema were measured to assess the neuroprotection of IPC. Several serum biomarkers related to glycocalyx damage, such as hyaluronic acid (HA), heparan sulfate (HS), and syndecan-1 (SYND1), were evaluated, and their changes were normalized to the ratio of postoperative/preoperative concentration. Western blot and immunofluorescence were used to evaluate the content and cellular location of HA-related metabolic enzymes. This study found that (1) IPC improved brain infarction and edema, neurological impairment, and BBB disruption in IRI rats; (2) IPC significantly up-regulated HA ratio and down-regulated HS ratio, but did not affect SYND1 ratio compared with the IRI group. Moreover, the increased HA ratio was negatively related to brain edema and neurological deficit score. (3) IPC affected HA metabolism by up-regulating hyaluronate synthase-1 and matrix metalloproteinase-2, and down-regulating hyaluronidase-1 in brain tissue. Together, this is the first report that the neuroprotective effect of IPC on IRI may be mediated through interfering with glycocalyx in the MCAO/R model.
Collapse
Affiliation(s)
- Yi-Na Zhang
- Department of Neurology, General Hospital of Northern Theater Command, 83 Wen Hua Road, Shenyang, 110016, China
- Department of Neurology, General Hospital of Northern Theater Command of China Medical University, Shenyang, 110013, China
| | - Qiong Wu
- Department of Neurology, General Hospital of Northern Theater Command, 83 Wen Hua Road, Shenyang, 110016, China
| | - Nan-Nan Zhang
- Department of Neurology, General Hospital of Northern Theater Command, 83 Wen Hua Road, Shenyang, 110016, China
| | - Hui-Sheng Chen
- Department of Neurology, General Hospital of Northern Theater Command, 83 Wen Hua Road, Shenyang, 110016, China.
| |
Collapse
|
8
|
Lemieux SP, Lev-Ram V, Tsien RY, Ellisman MH. Perineuronal nets and the neuronal extracellular matrix can be imaged by genetically encoded labeling of HAPLN1 in vitro and in vivo. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.29.569151. [PMID: 38076839 PMCID: PMC10705503 DOI: 10.1101/2023.11.29.569151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2023]
Abstract
Neuronal extracellular matrix (ECM) and a specific form of ECM called the perineuronal net (PNN) are important structures for central nervous system (CNS) integrity and synaptic plasticity. PNNs are distinctive, dense extracellular structures that surround parvalbumin (PV)-positive inhibitory interneurons with openings at mature synapses. Enzyme-mediated PNN disruption can erase established memories and re-open critical periods in animals, suggesting that PNNs are important for memory stabilization and conservation. Here, we characterized the structure and distribution of several ECM/PNN molecules around neurons in culture, brain slice, and whole mouse brain. While specific lectins are well-established as PNN markers and label a distinct, fenestrated structure around PV neurons, we show that other CNS neurons possess similar extracellular structures assembled around hyaluronic acid, suggesting a PNN-like structure of different composition that is more widespread. We additionally report that genetically encoded labeling of hyaluronan and proteoglycan link protein 1 (HAPLN1) reveals a PNN-like structure around many neurons in vitro and in vivo. Our findings add to our understanding of neuronal extracellular structures and describe a new mouse model for monitoring live ECM dynamics.
Collapse
Affiliation(s)
- Sakina P. Lemieux
- Department of Neurosciences, University of California, San Diego, La Jolla CA 92093-0647
| | - Varda Lev-Ram
- Department of Neurosciences, University of California, San Diego, La Jolla CA 92093-0647
| | - Roger Y. Tsien
- Department of Neurosciences, University of California, San Diego, La Jolla CA 92093-0647
- Department of Pharmacology, University of California, San Diego, La Jolla CA 92093-0647
- National Center for Microscopy and Imaging Research, Center for Biological Systems, University of California, San Diego, La Jolla CA 92093
- Department of Chemistry & Biochemistry, University of California, San Diego, La Jolla CA 92093
| | - Mark H. Ellisman
- Department of Neurosciences, University of California, San Diego, La Jolla CA 92093-0647
- Howard Hughes Medical Institute, University of California, San Diego, La Jolla CA 92093-0647
| |
Collapse
|
9
|
Benbenishty A, Peled-Hajaj S, Krishnaswamy VR, Har-Gil H, Havusha-Laufer S, Ruggiero A, Slutsky I, Blinder P, Sagi I. Longitudinal in vivo imaging of perineuronal nets. NEUROPHOTONICS 2023; 10:015008. [PMID: 36970015 PMCID: PMC10037344 DOI: 10.1117/1.nph.10.1.015008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 02/23/2023] [Indexed: 06/18/2023]
Abstract
SIGNIFICANCE Perineuronal nets (PNNs) are extracellular matrix structures implicated in learning, memory, information processing, synaptic plasticity, and neuroprotection. However, our understanding of mechanisms governing the evidently important contribution of PNNs to central nervous system function is lacking. A primary cause for this gap of knowledge is the absence of direct experimental tools to study their role in vivo. AIM We introduce a robust approach for quantitative longitudinal imaging of PNNs in brains of awake mice at subcellular resolution. APPROACH We label PNNs in vivo with commercially available compounds and monitor their dynamics with two-photon imaging. RESULTS Using our approach, we show that it is possible to longitudinally follow the same PNNs in vivo while monitoring degradation and reconstitution of PNNs. We demonstrate the compatibility of our method to simultaneously monitor neuronal calcium dynamics in vivo and compare the activity of neurons with and without PNNs. CONCLUSION Our approach is tailored for studying the intricate role of PNNs in vivo, while paving the road for elucidating their role in different neuropathological conditions.
Collapse
Affiliation(s)
- Amit Benbenishty
- The Weizmann Institute of Science, Department of Immunology and Regenerative Biology, Rehovot, Israel
| | - Shany Peled-Hajaj
- Tel Aviv University, Neurobiology, Biochemistry, and Biophysics School, Tel Aviv, Israel
- Tel Aviv University, Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv, Israel
| | | | - Hagai Har-Gil
- Tel Aviv University, Neurobiology, Biochemistry, and Biophysics School, Tel Aviv, Israel
- Tel Aviv University, Sagol School of Neuroscience, Tel Aviv, Israel
| | - Sapir Havusha-Laufer
- The Weizmann Institute of Science, Department of Immunology and Regenerative Biology, Rehovot, Israel
| | - Antonella Ruggiero
- Tel Aviv University, Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv, Israel
| | - Inna Slutsky
- Tel Aviv University, Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv, Israel
- Tel Aviv University, Sagol School of Neuroscience, Tel Aviv, Israel
| | - Pablo Blinder
- Tel Aviv University, Neurobiology, Biochemistry, and Biophysics School, Tel Aviv, Israel
- Tel Aviv University, Sagol School of Neuroscience, Tel Aviv, Israel
| | - Irit Sagi
- The Weizmann Institute of Science, Department of Immunology and Regenerative Biology, Rehovot, Israel
| |
Collapse
|
10
|
Kozlov MM, Taraska JW. Generation of nanoscopic membrane curvature for membrane trafficking. Nat Rev Mol Cell Biol 2023; 24:63-78. [PMID: 35918535 DOI: 10.1038/s41580-022-00511-9] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/20/2022] [Indexed: 11/09/2022]
Abstract
Curved membranes are key features of intracellular organelles, and their generation involves dynamic protein complexes. Here we describe the fundamental mechanisms such as the hydrophobic insertion, scaffolding and crowding mechanisms these proteins use to produce membrane curvatures and complex shapes required to form intracellular organelles and vesicular structures involved in endocytosis and secretion. For each mechanism, we discuss its cellular functions as well as the underlying physical principles and the specific membrane properties required for the mechanism to be feasible. We propose that the integration of individual mechanisms into a highly controlled, robust process of curvature generation often relies on the assembly of proteins into coats. How cells unify and organize the curvature-generating factors at the nanoscale is presented for three ubiquitous coats central for membrane trafficking in eukaryotes: clathrin-coated pits, caveolae, and COPI and COPII coats. The emerging theme is that these coats arrange and coordinate curvature-generating factors in time and space to dynamically shape membranes to accomplish membrane trafficking within cells.
Collapse
Affiliation(s)
- Michael M Kozlov
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Justin W Taraska
- Biochemistry and Biophysics Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
11
|
Anwar MM, Özkan E, Gürsoy-Özdemir Y. The role of extracellular matrix alterations in mediating astrocyte damage and pericyte dysfunction in Alzheimer's disease: A comprehensive review. Eur J Neurosci 2022; 56:5453-5475. [PMID: 34182602 DOI: 10.1111/ejn.15372] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/19/2021] [Accepted: 06/21/2021] [Indexed: 12/14/2022]
Abstract
The brain is a highly vascularized tissue protected by the blood-brain barrier (BBB), a complex structure allowing only necessary substances to pass through into the brain while limiting the entrance of harmful toxins. The BBB comprises several components, and the most prominent features are tight junctions between endothelial cells (ECs), which are further wrapped in a layer of pericytes. Pericytes are multitasked cells embedded in a thick basement membrane (BM) that consists of a fibrous extracellular matrix (ECM) and are surrounded by astrocytic endfeet. The primary function of astrocytes and pericytes is to provide essential blood supply and vital nutrients to the brain. In Alzheimer's disease (AD), long-term neuroinflammatory cascades associated with infiltration of harmful neurotoxic proteins may lead to BBB dysfunction and altered ECM components resulting in brain homeostatic imbalance, synaptic damage, and declined cognitive functions. Moreover, BBB structure and functional integrity may be lost due to induced ECM alterations, astrocyte damage, and pericytes dysfunction, leading to amyloid-beta (Aβ) hallmarks deposition in different brain regions. Herein, we highlight how BBB, ECM, astrocytes, and pericytes dysfunction can play a leading role in AD's pathogenesis and discuss their impact on brain functions.
Collapse
Affiliation(s)
- Mai M Anwar
- Neuroscience Research Lab, Research Center for Translational Medicine (KUTTAM), Koç University, Istanbul, Turkey.,Department of Biochemistry, National Organization for Drug Control and Research (NODCAR)/Egyptian Drug Authority, Cairo, Egypt
| | - Esra Özkan
- Neuroscience Research Lab, Research Center for Translational Medicine (KUTTAM), Koç University, Istanbul, Turkey
| | - Yasemin Gürsoy-Özdemir
- Neuroscience Research Lab, Research Center for Translational Medicine (KUTTAM), Koç University, Istanbul, Turkey.,Department of Neurology, School of Medicine, Koç University, Istanbul, Turkey
| |
Collapse
|
12
|
Enzymatic Degradation of Cortical Perineuronal Nets Reverses GABAergic Interneuron Maturation. Mol Neurobiol 2022; 59:2874-2893. [PMID: 35233718 PMCID: PMC9016038 DOI: 10.1007/s12035-022-02772-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 02/16/2022] [Indexed: 12/03/2022]
Abstract
Perineuronal nets (PNNs) are specialised extracellular matrix structures which preferentially enwrap fast-spiking (FS) parvalbumin interneurons and have diverse roles in the cortex. PNN maturation coincides with closure of the critical period of cortical plasticity. We have previously demonstrated that BDNF accelerates interneuron development in a c-Jun-NH2-terminal kinase (JNK)–dependent manner, which may involve upstream thousand-and-one amino acid kinase 2 (TAOK2). Chondroitinase-ABC (ChABC) enzymatic digestion of PNNs reportedly reactivates ‘juvenile-like’ plasticity in the adult CNS. However, the mechanisms involved are unclear. We show that ChABC produces an immature molecular phenotype in cultured cortical neurons, corresponding to the phenotype prior to critical period closure. ChABC produced different patterns of PNN-related, GABAergic and immediate early (IE) gene expression than well-characterised modulators of mature plasticity and network activity (GABAA-R antagonist, bicuculline, and sodium-channel blocker, tetrodotoxin (TTX)). ChABC downregulated JNK activity, while this was upregulated by bicuculline. Bicuculline, but not ChABC, upregulated Bdnf expression and ERK activity. Furthermore, we found that BDNF upregulation of semaphorin-3A and IE genes was TAOK mediated. Our data suggest that ChABC heightens structural flexibility and network disinhibition, potentially contributing to ‘juvenile-like’ plasticity. The molecular phenotype appears to be distinct from heightened mature synaptic plasticity and could relate to JNK signalling. Finally, we highlight that BDNF regulation of plasticity and PNNs involves TAOK signalling.
Collapse
|
13
|
Eleftheriadou D, Evans RE, Atkinson E, Abdalla A, Gavins FKH, Boyd AS, Williams GR, Knowles JC, Roberton VH, Phillips JB. An alginate-based encapsulation system for delivery of therapeutic cells to the CNS. RSC Adv 2022; 12:4005-4015. [PMID: 35425456 PMCID: PMC8981497 DOI: 10.1039/d1ra08563h] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 01/22/2022] [Indexed: 12/21/2022] Open
Abstract
Treatment options for neurodegenerative conditions such as Parkinson's disease have included the delivery of cells which release dopamine or neurotrophic factors to the brain. Here, we report the development of a novel approach for protecting cells after implantation into the central nervous system (CNS), by developing dual-layer alginate beads that encapsulate therapeutic cells and release an immunomodulatory compound in a sustained manner. An optimal alginate formulation was selected with a view to providing a sustained physical barrier between engrafted cells and host tissue, enabling exchange of small molecules while blocking components of the host immune response. In addition, a potent immunosuppressant, FK506, was incorporated into the outer layer of alginate beads using electrosprayed poly-ε-caprolactone core–shell nanoparticles with prolonged release profiles. The stiffness, porosity, stability and ability of the alginate beads to support and protect encapsulated SH-SY5Y cells was demonstrated, and the release profile of FK506 and its effect on T-cell proliferation in vitro was characterized. Collectively, our results indicate this multi-layer encapsulation technology has the potential to be suitable for use in CNS cell delivery, to protect implanted cells from host immune responses whilst providing permeability to nutrients and released therapeutic molecules. Novel composite cell encapsulation system: dual-layer, micro-scale beads maintain cell survival while releasing immunomodulatory FK506 in a sustained manner. This biotechnology platform could be applicable for treatment of CNS and other disorders.![]()
Collapse
Affiliation(s)
- Despoina Eleftheriadou
- UCL Centre for Nerve Engineering, University College London London UK.,UCL School of Pharmacy, University College London London WC1N 1AX UK
| | - Rachael E Evans
- UCL Centre for Nerve Engineering, University College London London UK.,UCL School of Pharmacy, University College London London WC1N 1AX UK
| | - Emily Atkinson
- UCL Centre for Nerve Engineering, University College London London UK.,UCL School of Pharmacy, University College London London WC1N 1AX UK
| | - Ahmed Abdalla
- UCL Centre for Nerve Engineering, University College London London UK.,UCL School of Pharmacy, University College London London WC1N 1AX UK
| | - Francesca K H Gavins
- UCL Centre for Nerve Engineering, University College London London UK.,UCL School of Pharmacy, University College London London WC1N 1AX UK
| | - Ashleigh S Boyd
- UCL Institute of Immunity and Transplantation, Royal Free Hospital London UK
| | - Gareth R Williams
- UCL School of Pharmacy, University College London London WC1N 1AX UK
| | - Jonathan C Knowles
- Biomaterials & Tissue Engineering, UCL Eastman Dental Institute London UK
| | - Victoria H Roberton
- UCL Centre for Nerve Engineering, University College London London UK.,UCL School of Pharmacy, University College London London WC1N 1AX UK
| | - James B Phillips
- UCL Centre for Nerve Engineering, University College London London UK.,UCL School of Pharmacy, University College London London WC1N 1AX UK
| |
Collapse
|
14
|
Zakusilo FT, Kerry O'Banion M, Gelbard HA, Seluanov A, Gorbunova V. Matters of size: Roles of hyaluronan in CNS aging and disease. Ageing Res Rev 2021; 72:101485. [PMID: 34634492 PMCID: PMC8903057 DOI: 10.1016/j.arr.2021.101485] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 10/05/2021] [Accepted: 10/06/2021] [Indexed: 12/29/2022]
Abstract
Involvement of extracellular matrix (ECM) components in aging and age-related neurodegeneration is not well understood. The role of hyaluronan (HA), a major extracellular matrix glycosaminoglycan, in malignancy and inflammation is gaining new understanding. In particular, the differential biological effects of high molecular weight (HMW-HA) and low molecular weight hyaluronan (LMW-HA), and the mechanism behind such differences are being uncovered. Tightly regulated in the brain, HA can have diverse effects on cellular development, growth and degeneration. In this review, we summarize the homeostasis and signaling of HA in healthy tissue, discuss its distribution and ontogeny in the central nervous system (CNS), summarize evidence for its involvement in age-related neurodegeneration and Alzheimer Disease (AD), and assess the potential of HA as a therapeutic target in the CNS.
Collapse
Affiliation(s)
- Frances Tolibzoda Zakusilo
- Department of Neuroscience, University of Rochester Medical Center, Rochester, NY, USA; Departments of Biology and Medicine, University of Rochester, Rochester, NY, USA
| | - M Kerry O'Banion
- Department of Neuroscience, University of Rochester Medical Center, Rochester, NY, USA; Department of Neurology, University of Rochester Medical Center, Rochester, NY, USA
| | - Harris A Gelbard
- Department of Neurology, University of Rochester Medical Center, Rochester, NY, USA; Center for Neurotherapeutics Discovery, University of Rochester Medical Center, Rochester, NY, USA
| | - Andrei Seluanov
- Departments of Biology and Medicine, University of Rochester, Rochester, NY, USA
| | - Vera Gorbunova
- Departments of Biology and Medicine, University of Rochester, Rochester, NY, USA
| |
Collapse
|
15
|
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: 83] [Impact Index Per Article: 20.8] [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
|
16
|
Wilson ES, Litwa K. Synaptic Hyaluronan Synthesis and CD44-Mediated Signaling Coordinate Neural Circuit Development. Cells 2021; 10:2574. [PMID: 34685554 PMCID: PMC8533746 DOI: 10.3390/cells10102574] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 09/17/2021] [Accepted: 09/21/2021] [Indexed: 12/20/2022] Open
Abstract
The hyaluronan-based extracellular matrix is expressed throughout nervous system development and is well-known for the formation of perineuronal nets around inhibitory interneurons. Since perineuronal nets form postnatally, the role of hyaluronan in the initial formation of neural circuits remains unclear. Neural circuits emerge from the coordinated electrochemical signaling of excitatory and inhibitory synapses. Hyaluronan localizes to the synaptic cleft of developing excitatory synapses in both human cortical spheroids and the neonatal mouse brain and is diminished in the adult mouse brain. Given this developmental-specific synaptic localization, we sought to determine the mechanisms that regulate hyaluronan synthesis and signaling during synapse formation. We demonstrate that hyaluronan synthase-2, HAS2, is sufficient to increase hyaluronan levels in developing neural circuits of human cortical spheroids. This increased hyaluronan production reduces excitatory synaptogenesis, promotes inhibitory synaptogenesis, and suppresses action potential formation. The hyaluronan receptor, CD44, promotes hyaluronan retention and suppresses excitatory synaptogenesis through regulation of RhoGTPase signaling. Our results reveal mechanisms of hyaluronan synthesis, retention, and signaling in developing neural circuits, shedding light on how disease-associated hyaluronan alterations can contribute to synaptic defects.
Collapse
Affiliation(s)
| | - Karen Litwa
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA;
| |
Collapse
|
17
|
Bucher EA, Collins JM, King AE, Vickers JC, Kirkcaldie MTK. Coherence and cognition in the cortex: the fundamental role of parvalbumin, myelin, and the perineuronal net. Brain Struct Funct 2021; 226:2041-2055. [PMID: 34175994 DOI: 10.1007/s00429-021-02327-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 06/17/2021] [Indexed: 11/28/2022]
Abstract
The calcium binding protein parvalbumin is expressed in interneurons of two main morphologies, the basket and chandelier cells, which target perisomatic domains on principal cells and are extensively interconnected in laminar networks by synapses and gap junctions. Beyond its utility as a convenient cellular marker, parvalbumin is an unambiguous identifier of the key role that these interneurons play in the fundamental functions of the cortex. They provide a temporal framework for principal cell activity by propagating gamma oscillation, providing coherence for cortical information processing and the basis for timing-dependent plasticity processes. As these parvalbumin networks mature, they are physically and functionally stabilised by axonal myelination and development of the extracellular matrix structure termed the perineuronal net. This maturation correlates with the emergence of high-speed, highly energetic activity and provides a coherent foundation for the unique ability of the cortex to cross-correlate activity across sensory modes and internal representations.
Collapse
Affiliation(s)
- Ellie A Bucher
- Wicking Dementia Research and Education Centre, University of Tasmania, Private Bag 143, Hobart, TAS, 7001, Australia
| | - Jessica M Collins
- Wicking Dementia Research and Education Centre, University of Tasmania, Private Bag 143, Hobart, TAS, 7001, Australia
| | - Anna E King
- Wicking Dementia Research and Education Centre, University of Tasmania, Private Bag 143, Hobart, TAS, 7001, Australia
| | - James C Vickers
- Wicking Dementia Research and Education Centre, University of Tasmania, Private Bag 143, Hobart, TAS, 7001, Australia
| | - Matthew T K Kirkcaldie
- Wicking Dementia Research and Education Centre, University of Tasmania, Private Bag 143, Hobart, TAS, 7001, Australia.
| |
Collapse
|
18
|
Structural and Functional Modulation of Perineuronal Nets: In Search of Important Players with Highlight on Tenascins. Cells 2021; 10:cells10061345. [PMID: 34072323 PMCID: PMC8230358 DOI: 10.3390/cells10061345] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/18/2021] [Accepted: 05/26/2021] [Indexed: 12/30/2022] Open
Abstract
The extracellular matrix (ECM) of the brain plays a crucial role in providing optimal conditions for neuronal function. Interactions between neurons and a specialized form of ECM, perineuronal nets (PNN), are considered a key mechanism for the regulation of brain plasticity. Such an assembly of interconnected structural and regulatory molecules has a prominent role in the control of synaptic plasticity. In this review, we discuss novel ways of studying the interplay between PNN and its regulatory components, particularly tenascins, in the processes of synaptic plasticity, mechanotransduction, and neurogenesis. Since enhanced neuronal activity promotes PNN degradation, it is possible to study PNN remodeling as a dynamical change in the expression and organization of its constituents that is reflected in its ultrastructure. The discovery of these subtle modifications is enabled by the development of super-resolution microscopy and advanced methods of image analysis.
Collapse
|
19
|
An Extracellular Perspective on CNS Maturation: Perineuronal Nets and the Control of Plasticity. Int J Mol Sci 2021; 22:ijms22052434. [PMID: 33670945 PMCID: PMC7957817 DOI: 10.3390/ijms22052434] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/24/2021] [Accepted: 02/24/2021] [Indexed: 02/07/2023] Open
Abstract
During restricted time windows of postnatal life, called critical periods, neural circuits are highly plastic and are shaped by environmental stimuli. In several mammalian brain areas, from the cerebral cortex to the hippocampus and amygdala, the closure of the critical period is dependent on the formation of perineuronal nets. Perineuronal nets are a condensed form of an extracellular matrix, which surrounds the soma and proximal dendrites of subsets of neurons, enwrapping synaptic terminals. Experimentally disrupting perineuronal nets in adult animals induces the reactivation of critical period plasticity, pointing to a role of the perineuronal net as a molecular brake on plasticity as the critical period closes. Interestingly, in the adult brain, the expression of perineuronal nets is remarkably dynamic, changing its plasticity-associated conditions, including memory processes. In this review, we aimed to address how perineuronal nets contribute to the maturation of brain circuits and the regulation of adult brain plasticity and memory processes in physiological and pathological conditions.
Collapse
|
20
|
Katarzyna Greda A, Nowicka D. Hyaluronidase inhibition accelerates functional recovery from stroke in the mouse brain. J Neurochem 2021; 157:781-801. [PMID: 33345310 DOI: 10.1111/jnc.15279] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 11/30/2020] [Accepted: 12/17/2020] [Indexed: 12/15/2022]
Abstract
Perineuronal nets (PNNs) are presumed to limit plasticity in adult animals. Ischaemic stroke results in the massive breakdown of PNNs resulting in rejuvenating states of neuronal plasticity, but the mechanisms of this phenomenon are largely unknown. As hyaluronic acid (HA) is the structural backbone of PNNs, we hypothesized that these changes are a consequence of the altered expression of HA metabolism enzymes. Additionally, we investigated whether early hyaluronidase inhibition interferes with post-stroke PNN reduction and behavioural recovery. We investigated the mRNA/protein expression of these enzymes in the perilesional, remote and contralateral cortical regions in mice at different time points after photothrombosis, using quantitative real-time polymerase chain reaction and immunofluorescence. A skilled reaching test was employed to test hyaluronidase inhibitor L-ascorbic acid 6-hexadecanoate influence on post-stroke recovery. We found the simultaneous up-regulation of mRNA of HA synthesizing and degrading enzymes in the perilesional area early after stroke, suggesting an acceleration of HA turnover in ischaemic animals. Immunostaining revealed differential cellular localization of enzymes, with hyaluronidase 1 in astrocytes and hyaluronan synthase 2 in astrocytes and neurons, and post-stroke up-regulation of both of them in astrocytes. β-glucuronidase was observed in neurons but post-stroke up-regulation occurred in microglia. Inhibition of hyaluronidase activity early after stroke resulted in improved performance in skilled reaching test, without affecting the numbers of PNNs. These results suggest that after stroke, a substantial reorganization of polysaccharide content occurs, and interfering with this process at early time has a beneficial effect on recovery.
Collapse
Affiliation(s)
- Anna Katarzyna Greda
- Nencki Institute of Experimental Biology PAS, Laboratory of Epileptogenesis, Warsaw, Poland
| | - Dorota Nowicka
- Nencki Institute of Experimental Biology PAS, Laboratory of Epileptogenesis, Warsaw, Poland
| |
Collapse
|
21
|
Affiliation(s)
- Dong Gil Jang
- School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Korea
| | - Hyo Jung Sim
- School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Korea
| | - Eun Kyung Song
- School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Korea
| | - Taejoon Kwon
- School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Korea
- Center for Genomic Integrity, Institute for Basic Science (IBS), Ulsan 44919, Korea
| | - Tae Joo Park
- School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Korea
- Center for Genomic Integrity, Institute for Basic Science (IBS), Ulsan 44919, Korea
| |
Collapse
|
22
|
Willis A, Pratt JA, Morris BJ. BDNF and JNK Signaling Modulate Cortical Interneuron and Perineuronal Net Development: Implications for Schizophrenia-Linked 16p11.2 Duplication Syndrome. Schizophr Bull 2020; 47:812-826. [PMID: 33067994 PMCID: PMC8084442 DOI: 10.1093/schbul/sbaa139] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Schizophrenia (SZ) is a neurodevelopmental disorder caused by the interaction of genetic and environmental risk factors. One of the strongest genetic risk variants is duplication (DUP) of chr.16p11.2. SZ is characterized by cortical gamma-amino-butyric acid (GABA)ergic interneuron dysfunction and disruption to surrounding extracellular matrix structures, perineuronal nets (PNNs). Developmental maturation of GABAergic interneurons, and also the resulting closure of the critical period of cortical plasticity, is regulated by brain-derived neurotrophic factor (BDNF), although the mechanisms involved are unknown. Here, we show that BDNF promotes GABAergic interneuron and PNN maturation through JNK signaling. In mice reproducing the 16p11.2 DUP, where the JNK upstream activator Taok2 is overexpressed, we find that JNK is overactive and there are developmental abnormalities in PNNs, which persist into adulthood. Prefrontal cortex parvalbumin (PVB) expression is reduced, while PNN intensity is increased. Additionally, we report a unique role for TAOK2 signaling in the regulation of PVB interneurons. Our work implicates TAOK2-JNK signaling in cortical interneuron and PNN development, and in the responses to BDNF. It also demonstrates that over-activation of this pathway in conditions associated with SZ risk causes long-lasting disruption in cortical interneurons.
Collapse
Affiliation(s)
- Ashleigh Willis
- Institute of Neuroscience and Psychology, University of Glasgow, Glasgow, Scotland, UK
| | - Judith A Pratt
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, Scotland, UK
| | - Brian J Morris
- Institute of Neuroscience and Psychology, University of Glasgow, Glasgow, Scotland, UK,To whom correspondence should be addressed; Institute of Neuroscience and Psychology, University of Glasgow, G12 8QQ, Glasgow, Scotland, UK; tel: 0044-141-330-5361, fax: 0044-141-330-5659, e-mail:
| |
Collapse
|
23
|
Wilson E, Knudson W, Newell-Litwa K. Hyaluronan regulates synapse formation and function in developing neural networks. Sci Rep 2020; 10:16459. [PMID: 33020512 PMCID: PMC7536407 DOI: 10.1038/s41598-020-73177-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 09/07/2020] [Indexed: 02/08/2023] Open
Abstract
Neurodevelopmental disorders present with synaptic alterations that disrupt the balance between excitatory and inhibitory signaling. For example, hyperexcitability of cortical neurons is associated with both epilepsy and autism spectrum disorders. However, the mechanisms that initially establish the balance between excitatory and inhibitory signaling in brain development are not well understood. Here, we sought to determine how the extracellular matrix directs synapse formation and regulates synaptic function in a model of human cortical brain development. The extracellular matrix, making up twenty percent of brain volume, is largely comprised of hyaluronan. Hyaluronan acts as both a scaffold of the extracellular matrix and a space-filling molecule. Hyaluronan is present from the onset of brain development, beginning with neural crest cell migration. Through acute perturbation of hyaluronan levels during synaptogenesis, we sought to determine how hyaluronan impacts the ratio of excitatory to inhibitory synapse formation and the resulting neural activity. We used 3-D cortical spheroids derived from human induced pluripotent stem cells to replicate this neurodevelopmental window. Our results demonstrate that hyaluronan preferentially surrounds nascent excitatory synapses. Removal of hyaluronan increases the expression of excitatory synapse markers and results in a corresponding increase in the formation of excitatory synapses, while also decreasing inhibitory synapse formation. This increased excitatory synapse formation elevates network activity, as demonstrated by microelectrode array analysis. In contrast, the addition of purified hyaluronan suppresses excitatory synapse formation. These results establish that the hyaluronan extracellular matrix surrounds developing excitatory synapses, where it critically regulates synapse formation and the resulting balance between excitatory to inhibitory signaling.
Collapse
Affiliation(s)
- Emily Wilson
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University, Greenville, NC, USA
| | - Warren Knudson
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University, Greenville, NC, USA
| | - Karen Newell-Litwa
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University, Greenville, NC, USA.
| |
Collapse
|
24
|
Pong S, Karmacharya R, Sofman M, Bishop JR, Lizano P. The Role of Brain Microvascular Endothelial Cell and Blood-Brain Barrier Dysfunction in Schizophrenia. Complex Psychiatry 2020; 6:30-46. [PMID: 34883503 PMCID: PMC7673590 DOI: 10.1159/000511552] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 09/10/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Despite decades of research, little clarity exists regarding pathogenic mechanisms related to schizophrenia. Investigations on the disease biology of schizophrenia have primarily focused on neuronal alterations. However, there is substantial evidence pointing to a significant role for the brain's microvasculature in mediating neuroinflammation in schizophrenia. SUMMARY Brain microvascular endothelial cells (BMEC) are a central element of the microvasculature that forms the blood-brain barrier (BBB) and shields the brain against toxins and immune cells via paracellular, transcellular, transporter, and extracellular matrix proteins. While evidence for BBB dysfunction exists in brain disorders, including schizophrenia, it is not known if BMEC themselves are functionally compromised and lead to BBB dysfunction. KEY MESSAGES Genome-wide association studies, postmortem investigations, and gene expression analyses have provided some insights into the role of the BBB in schizophrenia pathophysiology. However, there is a significant gap in our understanding of the role that BMEC play in BBB dysfunction. Recent advances differentiating human BMEC from induced pluripotent stem cells (iPSC) provide new avenues to examine the role of BMEC in BBB dysfunction in schizophrenia.
Collapse
Affiliation(s)
- Sovannarath Pong
- Department of Psychiatry, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Rakesh Karmacharya
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts, USA
| | - Marianna Sofman
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Jeffrey R. Bishop
- Departments of Clinical and Experimental Pharmacology and Psychiatry, University of Minnesota, Minneapolis, Minnesota, USA
| | - Paulo Lizano
- Department of Psychiatry, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
25
|
Diverse Roles for Hyaluronan and Hyaluronan Receptors in the Developing and Adult Nervous System. Int J Mol Sci 2020; 21:ijms21175988. [PMID: 32825309 PMCID: PMC7504301 DOI: 10.3390/ijms21175988] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 08/17/2020] [Accepted: 08/18/2020] [Indexed: 02/07/2023] Open
Abstract
Hyaluronic acid (HA) plays a vital role in the extracellular matrix of neural tissues. Originally thought to hydrate tissues and provide mechanical support, it is now clear that HA is also a complex signaling molecule that can regulate cell processes in the developing and adult nervous systems. Signaling properties are determined by molecular weight, bound proteins, and signal transduction through specific receptors. HA signaling regulates processes such as proliferation, differentiation, migration, and process extension in a variety of cell types including neural stem cells, neurons, astrocytes, microglia, and oligodendrocyte progenitors. The synthesis and catabolism of HA and the expression of HA receptors are altered in disease and influence neuroinflammation and disease pathogenesis. This review discusses the roles of HA, its synthesis and breakdown, as well as receptor expression in neurodevelopment, nervous system function and disease.
Collapse
|
26
|
Crapser JD, Ochaba J, Soni N, Reidling JC, Thompson LM, Green KN. Microglial depletion prevents extracellular matrix changes and striatal volume reduction in a model of Huntington's disease. Brain 2020; 143:266-288. [PMID: 31848580 DOI: 10.1093/brain/awz363] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 08/22/2019] [Accepted: 10/01/2019] [Indexed: 12/12/2022] Open
Abstract
Huntington's disease is associated with a reactive microglial response and consequent inflammation. To address the role of these cells in disease pathogenesis, we depleted microglia from R6/2 mice, a rapidly progressing model of Huntington's disease marked by behavioural impairment, mutant huntingtin (mHTT) accumulation, and early death, through colony-stimulating factor 1 receptor inhibition (CSF1Ri) with pexidartinib (PLX3397) for the duration of disease. Although we observed an interferon gene signature in addition to downregulated neuritogenic and synaptic gene pathways with disease, overt inflammation was not evident by microglial morphology or cytokine transcript levels in R6/2 mice. Nonetheless, CSF1Ri-induced microglial elimination reduced or prevented disease-related grip strength and object recognition deficits, mHTT accumulation, astrogliosis, and striatal volume loss, the latter of which was not associated with reductions in cell number but with the extracellular accumulation of chondroitin sulphate proteoglycans (CSPGs)-a primary component of glial scars. A concurrent loss of proteoglycan-containing perineuronal nets was also evident in R6/2 mice, and microglial elimination not only prevented this but also strikingly increased perineuronal nets in the brains of naïve littermates, suggesting a new role for microglia as homeostatic regulators of perineuronal net formation and integrity.
Collapse
Affiliation(s)
- Joshua D Crapser
- Department of Neurobiology and Behavior, University of California, Irvine (UCI), Irvine, CA, USA.,Institute of Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA
| | - Joseph Ochaba
- Department of Neurobiology and Behavior, University of California, Irvine (UCI), Irvine, CA, USA.,Institute of Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA
| | - Neelakshi Soni
- Department of Neurobiology and Behavior, University of California, Irvine (UCI), Irvine, CA, USA.,Institute of Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA
| | - Jack C Reidling
- Department of Neurobiology and Behavior, University of California, Irvine (UCI), Irvine, CA, USA.,Institute of Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA
| | - Leslie M Thompson
- Department of Neurobiology and Behavior, University of California, Irvine (UCI), Irvine, CA, USA.,Institute of Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA.,Department of Psychiatry and Human Behavior, University of California, Irvine, CA, USA
| | - Kim N Green
- Department of Neurobiology and Behavior, University of California, Irvine (UCI), Irvine, CA, USA.,Institute of Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA
| |
Collapse
|
27
|
Takechi M, Oshima K, Nadano D, Kitagawa H, Matsuda T, Miyata S. A pericellular hyaluronan matrix is required for the morphological maturation of cortical neurons. Biochim Biophys Acta Gen Subj 2020; 1864:129679. [PMID: 32623025 DOI: 10.1016/j.bbagen.2020.129679] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/25/2020] [Accepted: 06/29/2020] [Indexed: 11/19/2022]
Abstract
BACKGROUND Hyaluronan (HA) is a major component of the extracellular matrix (ECM) and is involved in many cellular functions. In the adult brain, HA forms macromolecular aggregates around synapses and plays important roles in neural plasticity. In contrast to the well-characterized function of HA in the adult brain, its roles in the developing brain remain largely unknown. METHODS Biochemical and histochemical analyses were performed to analyze the amount, solubility, and localization of HA in the developing mouse brain. By combining in utero labeling, cell isolation, and in vitro cultures, we examined the expression of hyaluronan synthase (HAS) and morphological maturation of cortical neurons. RESULTS The amount of HA increased during perinatal development and decreased in the adult. HA existed as a soluble form in the early stages; however, its solubility markedly decreased during postnatal development. HA localized in cell-sparse regions in the embryonic stages, but was broadly distributed during the postnatal development of the cerebral cortex. Developing cortical neurons expressed both Has2 and Has3, but not Has1, suggesting the autonomous production of HA by neurons themselves. HA formed a pericellular matrix around the cell bodies and neurites of developing cortical neurons, and the inhibition of HA synthesis reduced neurite outgrowth. CONCLUSION The formation of the pericellular HA matrix is essential for the proper morphological maturation of developing neurons. GENERAL SIGNIFICANCE This study provides new insights into the roles of hyaluronan in the brain. DEVELOPMENT
Collapse
Affiliation(s)
- Mina Takechi
- Graduate School of Bioagricultural Sciences, Nagoya University, Furocho, Chikusa-Ku, Nagoya 464-8601, Japan
| | - Kenzi Oshima
- Graduate School of Bioagricultural Sciences, Nagoya University, Furocho, Chikusa-Ku, Nagoya 464-8601, Japan
| | - Daita Nadano
- Graduate School of Bioagricultural Sciences, Nagoya University, Furocho, Chikusa-Ku, Nagoya 464-8601, Japan
| | - Hiroshi Kitagawa
- Laboratory of Biochemistry, Kobe Pharmaceutical University, 4-19-1 Motoyamakitamachi, Higashinada-Ku, Kobe 658-8558, Japan
| | - Tsukasa Matsuda
- Graduate School of Bioagricultural Sciences, Nagoya University, Furocho, Chikusa-Ku, Nagoya 464-8601, Japan; Faculty of Food and Agricultural Sciences, Fukushima University, Kanayagawa 1, Fukushima 960-1296, Japan
| | - Shinji Miyata
- Graduate School of Bioagricultural Sciences, Nagoya University, Furocho, Chikusa-Ku, Nagoya 464-8601, Japan; Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwaicho, Fuchu, Tokyo 183-8509, Japan.
| |
Collapse
|
28
|
Perera TH, Howell SM, Smith Callahan LA. Manipulation of Extracellular Matrix Remodeling and Neurite Extension by Mouse Embryonic Stem Cells Using IKVAV and LRE Peptide Tethering in Hyaluronic Acid Matrices. Biomacromolecules 2019; 20:3009-3020. [PMID: 31306008 DOI: 10.1021/acs.biomac.9b00578] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Cellular remodeling of the matrix has recently emerged as a key factor in promoting neural differentiation. Most strategies to manipulate matrix remodeling focus on proteolytically cleavable cross-linkers, leading to changes in tethered biochemical signaling and matrix properties. Using peptides that are not the direct target of enzymatic degradation will likely reduce changes in the matrix and improve control of biological behavior. In this study, laminin-derived peptides, IKVAV and LRE, tethered to independent sites in hyaluronic acid matrices using Michael addition and strain-promoted azide-alkyne cycloaddition are sufficient to manipulate hyaluronic acid degradation, gelatinase expression, and protease expression, while promoting neurite extension through matrix metalloprotease-dependent mechanisms in mouse embryonic stem cells encapsulated in hyaluronic acid matrices using an oxidation-reduction reaction initiated gelation. This study provides the foundation for a new strategy to stimulate matrix remodeling that is not dependent on enzymatic cleavage targets.
Collapse
Affiliation(s)
- T Hiran Perera
- Vivian L. Smith Department of Neurosurgery , McGovern Medical School at the University of Texas Health Science Center at Houston McGovern Medical School , Houston , Texas 77030 , United States.,Center for Stem Cell and Regenerative Medicine, Brown Foundation Institute of Molecular Medicine , McGovern Medical School at the University of Texas Health Science Center at Houston , Houston , Texas 77030 , United States
| | - Skyler M Howell
- Vivian L. Smith Department of Neurosurgery , McGovern Medical School at the University of Texas Health Science Center at Houston McGovern Medical School , Houston , Texas 77030 , United States.,Center for Stem Cell and Regenerative Medicine, Brown Foundation Institute of Molecular Medicine , McGovern Medical School at the University of Texas Health Science Center at Houston , Houston , Texas 77030 , United States
| | - Laura A Smith Callahan
- Vivian L. Smith Department of Neurosurgery , McGovern Medical School at the University of Texas Health Science Center at Houston McGovern Medical School , Houston , Texas 77030 , United States.,Center for Stem Cell and Regenerative Medicine, Brown Foundation Institute of Molecular Medicine , McGovern Medical School at the University of Texas Health Science Center at Houston , Houston , Texas 77030 , United States.,Graduate School of Biomedical Sciences , MD Anderson Cancer Center UTHealth , Houston , Texas 77030 , United States
| |
Collapse
|
29
|
Rauti R, Renous N, Maoz BM. Mimicking the Brain Extracellular Matrix
in Vitro
: A Review of Current Methodologies and Challenges. Isr J Chem 2019. [DOI: 10.1002/ijch.201900052] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Rossana Rauti
- Department of Biomedical Engineering Tel Aviv University Israel
| | - Noa Renous
- Department of Biomedical Engineering Tel Aviv University Israel
| | - Ben M. Maoz
- Department of Biomedical Engineering Tel Aviv University Israel
- Sagol School of Neuroscience Tel Aviv University Tel Aviv Israel
- The Center for Nanoscience and Nanotechnology Tel Aviv University Tel Aviv 69978 Israel
| |
Collapse
|
30
|
Hayashi MK, Nishioka T, Shimizu H, Takahashi K, Kakegawa W, Mikami T, Hirayama Y, Koizumi S, Yoshida S, Yuzaki M, Tammi M, Sekino Y, Kaibuchi K, Shigemoto-Mogami Y, Yasui M, Sato K. Hyaluronan synthesis supports glutamate transporter activity. J Neurochem 2019; 150:249-263. [PMID: 31188471 DOI: 10.1111/jnc.14791] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 05/29/2019] [Accepted: 06/06/2019] [Indexed: 11/28/2022]
Abstract
Hyaluronan is synthesized, secreted, and anchored by hyaluronan synthases (HAS) at the plasma membrane and comprises the backbone of perineuronal nets around neuronal soma and dendrites. However, the molecular targets of hyaluronan to regulate synaptic transmission in the central nervous system have not been fully identified. Here, we report that hyaluronan is a negative regulator of excitatory signals. At excitatory synapses, glutamate is removed by glutamate transporters to turn off the signal and prevent excitotoxicity. Hyaluronan synthesized by HAS supports the activity of glial glutamate transporter 1 (GLT1). GLT1 also retracted from cellular processes of cultured astrocytes after hyaluronidase treatment and hyaluronan synthesis inhibition. A serial knockout study showed that all three HAS subtypes recruit GLT1 to cellular processes. Furthermore, hyaluronidase treatment activated neurons in a dissociated rat hippocampal culture and caused neuronal damage due to excitotoxicity. Our findings reveal that hyaluronan helps to turn off excitatory signals by supporting glutamate clearance. Cover Image for this issue: doi: 10.1111/jnc.14516.
Collapse
Affiliation(s)
- Mariko Kato Hayashi
- Medical School, International University of Health and Welfare, Narita, Chiba, Japan.,Division of Pharmacology, Laboratory of Neuropharmacology, National Institute of Health Sciences, Kawasaki, Kanagawa, Japan.,Department of Pharmacology, Keio University School of Medicine, Tokyo, Japan.,Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Tomoki Nishioka
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Hideo Shimizu
- Division of Pharmacology, Laboratory of Neuropharmacology, National Institute of Health Sciences, Kawasaki, Kanagawa, Japan
| | - Kanako Takahashi
- Division of Pharmacology, Laboratory of Neuropharmacology, National Institute of Health Sciences, Kawasaki, Kanagawa, Japan
| | - Wataru Kakegawa
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Tetsuri Mikami
- Department of Environmental and Life Sciences, Toyohashi University of Technology, Toyohashi, Aichi, Japan
| | - Yuri Hirayama
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Schuichi Koizumi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Sachiko Yoshida
- Department of Environmental and Life Sciences, Toyohashi University of Technology, Toyohashi, Aichi, Japan
| | - Michisuke Yuzaki
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Markku Tammi
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Yuko Sekino
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo, Japan
| | - Kozo Kaibuchi
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Yukari Shigemoto-Mogami
- Division of Pharmacology, Laboratory of Neuropharmacology, National Institute of Health Sciences, Kawasaki, Kanagawa, Japan
| | - Masato Yasui
- Department of Pharmacology, Keio University School of Medicine, Tokyo, Japan
| | - Kaoru Sato
- Division of Pharmacology, Laboratory of Neuropharmacology, National Institute of Health Sciences, Kawasaki, Kanagawa, Japan
| |
Collapse
|
31
|
Shurer CR, Kuo JCH, Roberts LM, Gandhi JG, Colville MJ, Enoki TA, Pan H, Su J, Noble JM, Hollander MJ, O'Donnell JP, Yin R, Pedram K, Möckl L, Kourkoutis LF, Moerner WE, Bertozzi CR, Feigenson GW, Reesink HL, Paszek MJ. Physical Principles of Membrane Shape Regulation by the Glycocalyx. Cell 2019; 177:1757-1770.e21. [PMID: 31056282 PMCID: PMC6768631 DOI: 10.1016/j.cell.2019.04.017] [Citation(s) in RCA: 178] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 02/19/2019] [Accepted: 04/09/2019] [Indexed: 12/12/2022]
Abstract
Cells bend their plasma membranes into highly curved forms to interact with the local environment, but how shape generation is regulated is not fully resolved. Here, we report a synergy between shape-generating processes in the cell interior and the external organization and composition of the cell-surface glycocalyx. Mucin biopolymers and long-chain polysaccharides within the glycocalyx can generate entropic forces that favor or disfavor the projection of spherical and finger-like extensions from the cell surface. A polymer brush model of the glycocalyx successfully predicts the effects of polymer size and cell-surface density on membrane morphologies. Specific glycocalyx compositions can also induce plasma membrane instabilities to generate more exotic undulating and pearled membrane structures and drive secretion of extracellular vesicles. Together, our results suggest a fundamental role for the glycocalyx in regulating curved membrane features that serve in communication between cells and with the extracellular matrix.
Collapse
Affiliation(s)
- Carolyn R Shurer
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Joe Chin-Hun Kuo
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY 14853, USA
| | | | - Jay G Gandhi
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY 14853, USA
| | | | - Thais A Enoki
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA
| | - Hao Pan
- Field of Biophysics, Cornell University, Ithaca, NY 14853, USA
| | - Jin Su
- Department of Clinical Sciences, Cornell University, Ithaca, NY 14853, USA
| | - Jade M Noble
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Michael J Hollander
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY 14853, USA
| | - John P O'Donnell
- Department of Molecular Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Rose Yin
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Kayvon Pedram
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - Leonhard Möckl
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - Lena F Kourkoutis
- School of Applied and Engineering Physics, Cornell University, Ithaca, NY 14853, USA; Kavli Institute at Cornell for Nanoscale Science, Ithaca, NY 14853, USA
| | - W E Moerner
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - Carolyn R Bertozzi
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Gerald W Feigenson
- Field of Biophysics, Cornell University, Ithaca, NY 14853, USA; Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA
| | - Heidi L Reesink
- Department of Clinical Sciences, Cornell University, Ithaca, NY 14853, USA
| | - Matthew J Paszek
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY 14853, USA; Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA; Field of Biophysics, Cornell University, Ithaca, NY 14853, USA; Kavli Institute at Cornell for Nanoscale Science, Ithaca, NY 14853, USA.
| |
Collapse
|
32
|
Wang H, Morales RTT, Cui X, Huang J, Qian W, Tong J, Chen W. A Photoresponsive Hyaluronan Hydrogel Nanocomposite for Dynamic Macrophage Immunomodulation. Adv Healthc Mater 2019; 8:e1801234. [PMID: 30537061 PMCID: PMC6392032 DOI: 10.1002/adhm.201801234] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Indexed: 12/22/2022]
Abstract
Macrophages are a predominant immune cell population that drive inflammatory responses and exhibit transitions in phenotype and function during tissue remodeling in disease and repair. Thus, engineering an immunomodulatory biomaterial has significant implications for resolving inflammation. Here, a biomimetic and photoresponsive hyaluronan (HA) hydrogel nanocomposite with tunable 3D extracellular matrix (ECM) adhesion sites for dynamic macrophage immunomodulation is engineered. Photodegradative alkoxylphenacyl-based polycarbonate (APP) nanocomposites are exploited to permit user-controlled Arg-Gly-Asp (RGD) adhesive peptide release and conjugation to a HA-based ECM for real-time integrin activation of macrophages encapsulated in 3D HA-APP nanocomposite hydrogels. It is demonstrated that photocontrolled 3D ECM-RGD peptide conjugation can activate αvβ3 integrin of macrophages, and periodic αvβ3 integrin activation can enhance anti-inflammatory M2 macrophage polarization. Altogether, an emerging use of biomimetic, photoresponsive, and bioactive HA-APP nanocomposite hydrogel is highlighted to command 3D cell-ECM interactions for modulating macrophage polarization, which may shed light on cell-ECM interactions in innate immunity and inspire new biomaterial-based immunomodulatory therapies.
Collapse
Affiliation(s)
- Haoyu Wang
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, NY, 11201, USA
| | - Renee-Tyler Tan Morales
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, NY, 11201, USA
| | - Xin Cui
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, NY, 11201, USA
| | - Jiongxian Huang
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, NY, 11201, USA
| | - Weiyi Qian
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, NY, 11201, USA
| | - Jie Tong
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, NY, 11201, USA
| | - Weiqiang Chen
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, NY, 11201, USA
- Department of Biomedical Engineering, New York University, Brooklyn, NY, 11201, USA
| |
Collapse
|
33
|
Melrose J. Keratan sulfate (KS)-proteoglycans and neuronal regulation in health and disease: the importance of KS-glycodynamics and interactive capability with neuroregulatory ligands. J Neurochem 2019; 149:170-194. [PMID: 30578672 DOI: 10.1111/jnc.14652] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Revised: 11/26/2018] [Accepted: 12/13/2018] [Indexed: 12/18/2022]
Abstract
Compared to the other classes of glycosaminoglycans (GAGs), that is, chondroitin/dermatan sulfate, heparin/heparan sulfate and hyaluronan, keratan sulfate (KS), have the least known of its interactive properties. In the human body, the cornea and the brain are the two most abundant tissue sources of KS. Embryonic KS is synthesized as a linear poly-N-acetyllactosamine chain of d-galactose-GlcNAc repeat disaccharides which become progressively sulfated with development, sulfation of GlcNAc is more predominant than galactose. KS contains multi-sulfated high-charge density, monosulfated and non-sulfated poly-N-acetyllactosamine regions and thus is a heterogeneous molecule in terms of chain length and charge distribution. A recent proteomics study on corneal KS demonstrated its interactivity with members of the Slit-Robbo and Ephrin-Ephrin receptor families and proteins which regulate Rho GTPase signaling and actin polymerization/depolymerization in neural development and differentiation. KS decorates a number of peripheral nervous system/CNS proteoglycan (PG) core proteins. The astrocyte KS-PG abakan defines functional margins of the brain and is up-regulated following trauma. The chondroitin sulfate/KS PG aggrecan forms perineuronal nets which are dynamic neuroprotective structures with anti-oxidant properties and roles in neural differentiation, development and synaptic plasticity. Brain phosphacan a chondroitin sulfate, KS, HNK-1 PG have roles in neural development and repair. The intracellular microtubule and synaptic vesicle KS-PGs MAP1B and SV2 have roles in metabolite transport, storage, and export of neurotransmitters and cytoskeletal assembly. MAP1B has binding sites for tubulin and actin through which it promotes cytoskeletal development in growth cones and is highly expressed during neurite extension. The interactive capability of KS with neuroregulatory ligands indicate varied roles for KS-PGs in development and regenerative neural processes.
Collapse
Affiliation(s)
- James Melrose
- Raymond Purves Bone and Joint Research Laboratory, Kolling Institute, 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 Campus, Royal North Shore Hospital, The University of Sydney, New South Wales, Australia.,Faculty of Medicine and Health, Royal North Shore Hospital, The University of Sydney, St. Leonards, New South Wales, Australia
| |
Collapse
|
34
|
Bejoy J, Wang Z, Bijonowski B, Yang M, Ma T, Sang QX, Li Y. Differential Effects of Heparin and Hyaluronic Acid on Neural Patterning of Human Induced Pluripotent Stem Cells. ACS Biomater Sci Eng 2018; 4:4354-4366. [PMID: 31572767 PMCID: PMC6768405 DOI: 10.1021/acsbiomaterials.8b01142] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
A lack of well-established animal models that can efficiently represent human brain pathology has led to the development of human induced pluripotent stem cell (hiPSC)-derived brain tissues. Brain organoids have enhanced our ability to understand the developing human brain and brain disorders (e.g., Schizophrenia, microcephaly), but the organoids still do not accurately recapitulate the anatomical organization of the human brain. Therefore, it is important to evaluate and optimize induction and signaling factors in order to engineer the next generation of brain organoids. In this study, the impact of hyaluronic acid (HA), a major brain extracellular matrix (ECM) component that interacts with cells through ligand-binding receptors, on the patterning of brain organoids from hiPSCs was evaluated. To mediate HA- binding capacity of signaling molecules, heparin was added in addition to HA or conjugated to HA to form hydrogels (with two different moduli). The neural cortical spheroids derived from hiPSCs were treated with either HA or heparin plus HA (Hep- HA) and were analyzed for ECM impacts on neural patterning. The results indicate that Hep-HA has a caudalizing effect on hiPSC-derived neural spheroids, in particular for stiff Hep-HA hydrogels. Wnt and Hippo/Yes-associated protein (YAP) signaling was modulated (using Wnt inhibitor IWP4 or actin disruption agent Cytochalasin D respectively) to understand the underlying mechanism. IWP4 and cytochalasin D promote forebrain identity. The results from this study should enhance the understanding of influence of biomimetic ECM factors for brain organoid generation.
Collapse
Affiliation(s)
- Julie Bejoy
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida, United States
| | - Zhe Wang
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, Florida, United States
| | - Brent Bijonowski
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida, United States
| | - Mo Yang
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, Florida, United States
| | - Teng Ma
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida, United States
| | - Qing-Xiang Sang
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, Florida, United States
- Institute of Molecular Biophysics, Florida State University, Tallahassee, Florida, United States
| | - Yan Li
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida, United States
- Institute of Molecular Biophysics, Florida State University, Tallahassee, Florida, United States
| |
Collapse
|
35
|
Hyaluronic acid is present on specific perineuronal nets in the mouse cerebral cortex. Brain Res 2018; 1698:139-150. [PMID: 30099038 DOI: 10.1016/j.brainres.2018.08.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 08/04/2018] [Accepted: 08/06/2018] [Indexed: 11/23/2022]
Abstract
In the central nervous system (CNS), extracellular matrix (ECM) molecules comprise more than 20% of the volume and are involved in neuronal plasticity, synaptic transmission, and differentiation. Perineuronal nets (PNNs) are ECM molecules that highly accumulate around the soma of neurons. The components of the ECM in the CNS include proteins, proteoglycans, and glycosaminoglycans. Although hyaluronic acid (HA) is considered a constituent element of PNNs, the distribution of HA in the cortex has not been clarified. To elucidate the cortical region-specific distribution of HA, we quantitatively analyzed HA binding protein (HABP)-positive PNNs in the mature mouse cerebral cortex. Our findings revealed that HABP-positive PNNs are present throughout the mouse cortex. The distribution of many HABP-positive PNNs differed from that of Wisteria floribunda agglutinin-positive PNNs. Furthermore, we observed granular-like HABP-positive PNNs in layer 1 of the cortex. These findings indicate that PNNs in the mouse cortex show region-dependent differences in composition. HABP-positive PNNs in layer 1 of the cortex may have different functions such as neuronal differentiation, proliferation, and migration unlike what has been reported for PNNs so far.
Collapse
|
36
|
Bennet L, Dhillon S, Lear CA, van den Heuij L, King V, Dean JM, Wassink G, Davidson JO, Gunn AJ. Chronic inflammation and impaired development of the preterm brain. J Reprod Immunol 2018; 125:45-55. [DOI: 10.1016/j.jri.2017.11.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 11/13/2017] [Accepted: 11/24/2017] [Indexed: 12/17/2022]
|
37
|
Filpa V, Bistoletti M, Caon I, Moro E, Grimaldi A, Moretto P, Baj A, Giron MC, Karousou E, Viola M, Crema F, Frigo G, Passi A, Giaroni C, Vigetti D. Changes in hyaluronan deposition in the rat myenteric plexus after experimentally-induced colitis. Sci Rep 2017; 7:17644. [PMID: 29247178 PMCID: PMC5732300 DOI: 10.1038/s41598-017-18020-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 12/05/2017] [Indexed: 12/27/2022] Open
Abstract
Myenteric plexus alterations hamper gastrointestinal motor function during intestinal inflammation. Hyaluronan (HA), an extracellular matrix glycosaminoglycan involved in inflammatory responses, may play a role in this process. In the colon of control rats, HA-binding protein (HABP), was detected in myenteric neuron soma, perineuronal space and ganglia surfaces. Prominent hyaluronan synthase 2 (HAS2) staining was found in myenteric neuron cytoplasm, suggesting that myenteric neurons produce HA. In the myenteric plexus of rats with 2, 4-dinitrobenzene sulfonic (DNBS)-induced colitis HABP staining was altered in the perineuronal space, while both HABP staining and HA levels increased in the muscularis propria. HAS2 immunopositive myenteric neurons and HAS2 mRNA and protein levels also increased. Overall, these observations suggest that inflammation alters HA distribution and levels in the gut neuromuscular compartment. Such changes may contribute to alterations in the myenteric plexus.
Collapse
Affiliation(s)
- Viviana Filpa
- Department of Medicine and Surgery, University of Insubria, via H. Dunant 5, Varese, Italy
| | - Michela Bistoletti
- Department of Medicine and Surgery, University of Insubria, via H. Dunant 5, Varese, Italy
| | - Ilaria Caon
- Department of Medicine and Surgery, University of Insubria, via H. Dunant 5, Varese, Italy
| | - Elisabetta Moro
- Department of Internal Medicine and Therapeutics, Section of Pharmacology, University of Pavia, Pavia, Italy
| | - Annalisa Grimaldi
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - Paola Moretto
- Department of Medicine and Surgery, University of Insubria, via H. Dunant 5, Varese, Italy
| | - Andreina Baj
- Department of Medicine and Surgery, University of Insubria, via H. Dunant 5, Varese, Italy
| | - Maria Cecilia Giron
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Evgenia Karousou
- Department of Medicine and Surgery, University of Insubria, via H. Dunant 5, Varese, Italy
| | - Manuela Viola
- Department of Medicine and Surgery, University of Insubria, via H. Dunant 5, Varese, Italy
| | - Francesca Crema
- Department of Internal Medicine and Therapeutics, Section of Pharmacology, University of Pavia, Pavia, Italy
| | - Gianmario Frigo
- Department of Internal Medicine and Therapeutics, Section of Pharmacology, University of Pavia, Pavia, Italy
| | - Alberto Passi
- Department of Medicine and Surgery, University of Insubria, via H. Dunant 5, Varese, Italy
| | - Cristina Giaroni
- Department of Medicine and Surgery, University of Insubria, via H. Dunant 5, Varese, Italy.
| | - Davide Vigetti
- Department of Medicine and Surgery, University of Insubria, via H. Dunant 5, Varese, Italy
| |
Collapse
|