61 results on '"Rustenhoven J"'
Search Results
2. P04.23 Pericytes contribute to tumour immune system evasion in glioblastoma multiforme through the under-expression of ICAM-1, VCAM-1 and MCP-1.
- Author
-
Joret, M O, primary, Park, T I, additional, Macapagal, J M, additional, Rustenhoven, J, additional, Kim, B J, additional, Correia, J, additional, Mee, E, additional, Faull, R L M, additional, Schweder, P, additional, and Dragunow, M, additional
- Published
- 2018
- Full Text
- View/download PDF
3. A role for human brain pericytes in neuroinflammation (vol 11, 104, 2014)
- Author
-
Jansson, D, Rustenhoven, J, Feng, S, Hurley, D, Oldfield, RL, Bergin, PS, Mee, EW, Faull, RLM, Dragunow, M, Jansson, D, Rustenhoven, J, Feng, S, Hurley, D, Oldfield, RL, Bergin, PS, Mee, EW, Faull, RLM, and Dragunow, M
- Published
- 2015
4. ChemInform Abstract: Experiments Directed Towards the Synthesis of Anthracyclinones. Part 28. Titanium(IV)- and Tin(IV)-Mediated Cyclizations of ortho-Methallyl- Substituted Homochiral Dioxans.
- Author
-
CAMBIE, R. C., primary, HIGGS, K. C., additional, RUSTENHOVEN, J. J., additional, and RUTLEDGE, P. S., additional
- Published
- 2010
- Full Text
- View/download PDF
5. ChemInform Abstract: Total Synthesis of 15‐Deoxoclerocidin.
- Author
-
Kende, A. S., primary, Rustenhoven, J. J., additional, and Zimmermann, K., additional
- Published
- 2000
- Full Text
- View/download PDF
6. Experiments Directed Towards the Synthesis of Anthracyclinones. XXXVI Asymmetric Dihydroxylations of Alkylidene Anthracyclinones
- Author
-
Cambie, Richard C., primary, Clark, Russell B., additional, Rutledge, Peter S., additional, and Rustenhoven, J. J., additional
- Published
- 1999
- Full Text
- View/download PDF
7. ChemInform Abstract: Experiments Directed Towards the Synthesis of Anthracyclinones. Part 29. Fluorosubstituted Tetracycles.
- Author
-
CAMBIE, R. C., primary, HIGGS, K. C., additional, RUSTENHOVEN, J. J., additional, and RUTLEDGE, P. S., additional
- Published
- 1996
- Full Text
- View/download PDF
8. ChemInform Abstract: Stereoselective Titanium‐Mediated Aldol Reactions of Menthyl Acetate Enolates, and Allylations with Benzaldehyde.
- Author
-
CAMBIE, R. C., primary, CODDINGTON, J. M., additional, MILBANK, J. B. J., additional, PAUSLER, M. G., additional, RUSTENHOVEN, J. J., additional, RUTLEDGE, P. S., additional, SHAW, G. L., additional, and SINKOVICH, P. I., additional
- Published
- 1993
- Full Text
- View/download PDF
9. ChemInform Abstract: Experiments Directed Towards the Synthesis of Anthracyclinones. Part 28. Titanium(IV)- and Tin(IV)-Mediated Cyclizations of ortho-Methallyl- Substituted Homochiral Dioxans.
- Author
-
CAMBIE, R. C., HIGGS, K. C., RUSTENHOVEN, J. J., and RUTLEDGE, P. S.
- Published
- 1996
- Full Text
- View/download PDF
10. Engineered T cell therapy for central nervous system injury.
- Author
-
Gao W, Kim MW, Dykstra T, Du S, Boskovic P, Lichti CF, Ruiz-Cardozo MA, Gu X, Weizman Shapira T, Rustenhoven J, Molina C, Smirnov I, Merbl Y, Ray WZ, and Kipnis J
- Subjects
- Animals, Female, Humans, Male, Mice, CD4-Positive T-Lymphocytes immunology, CD4-Positive T-Lymphocytes cytology, Clone Cells cytology, Clone Cells immunology, Disease Models, Animal, Interferon-gamma immunology, Mice, Inbred C57BL, Myelin Sheath immunology, Myeloid Cells immunology, Receptors, Antigen, T-Cell immunology, Receptors, Antigen, T-Cell metabolism, Receptors, Antigen, T-Cell genetics, Single-Cell Gene Expression Analysis, Nerve Tissue Proteins immunology, Autoimmunity, Cell Engineering methods, Cell- and Tissue-Based Therapy methods, Central Nervous System immunology, Central Nervous System injuries, Neuroprotection, Spinal Cord Injuries therapy, Spinal Cord Injuries immunology, T-Lymphocytes immunology, T-Lymphocytes transplantation
- Abstract
Traumatic injuries to the central nervous system (CNS) afflict millions of individuals worldwide
1 , yet an effective treatment remains elusive. Following such injuries, the site is populated by a multitude of peripheral immune cells, including T cells, but a comprehensive understanding of the roles and antigen specificity of these endogenous T cells at the injury site has been lacking. This gap has impeded the development of immune-mediated cellular therapies for CNS injuries. Here, using single-cell RNA sequencing, we demonstrated the clonal expansion of mouse and human spinal cord injury-associated T cells and identified that CD4+ T cell clones in mice exhibit antigen specificity towards self-peptides of myelin and neuronal proteins. Leveraging mRNA-based T cell receptor (TCR) reconstitution, a strategy aimed to minimize potential adverse effects from prolonged activation of self-reactive T cells, we generated engineered transiently autoimmune T cells. These cells demonstrated notable neuroprotective efficacy in CNS injury models, in part by modulating myeloid cells via IFNγ. Our findings elucidate mechanistic insight underlying the neuroprotective function of injury-responsive T cells and pave the way for the future development of T cell therapies for CNS injuries., (© 2024. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2024
- Full Text
- View/download PDF
11. Brain-Engrafted Monocyte-derived Macrophages from Blood and Skull-Bone Marrow Exhibit Distinct Identities from Microglia.
- Author
-
Du S, Drieu A, Cheng Y, Storck SE, Rustenhoven J, Mamuladze T, Bhattarai B, Brioschi S, Nguyen K, Ou F, Cao J, Rodrigues PF, Smirnov I, DeNardo D, Ginhoux F, Cella M, Colonna M, and Kipnis J
- Abstract
Microglia are thought to originate exclusively from primitive macrophage progenitors in the yolk sac (YS) and to persist throughout life without much contribution from definitive hematopoiesis. Here, using lineage tracing, pharmacological manipulation, and RNA-sequencing, we elucidated the presence and characteristics of monocyte-derived macrophages (MDMs) in the brain parenchyma at baseline and during microglia repopulation, and defined the core transcriptional signatures of brain-engrafted MDMs. Lineage tracing mouse models revealed that MDMs transiently express CD206 during brain engraftment as CD206
+ microglia precursors in the YS. We found that brain-engrafted MDMs exhibit transcriptional and epigenetic characteristics akin to meningeal macrophages, likely due to environmental imprinting within the meningeal space. Utilizing parabiosis and skull transplantation, we demonstrated that monocytes from both peripheral blood and skull bone marrow can repopulate microglia-depleted brains. Our results reveal the heterogeneous origins and cellular dynamics of brain parenchymal macrophages at baseline and in models of microglia depletion.- Published
- 2024
- Full Text
- View/download PDF
12. An inducible genetic tool to track and manipulate specific microglial states reveals their plasticity and roles in remyelination.
- Author
-
Barclay KM, Abduljawad N, Cheng Z, Kim MW, Zhou L, Yang J, Rustenhoven J, Mazzitelli JA, Smyth LCD, Kapadia D, Brioschi S, Beatty W, Hou J, Saligrama N, Colonna M, Yu G, Kipnis J, and Li Q
- Subjects
- Animals, Mice, Demyelinating Diseases genetics, Mice, Inbred C57BL, Mice, Transgenic, Disease Models, Animal, Brain, Myelin Sheath metabolism, White Matter pathology, Microglia physiology, Remyelination, Cell Plasticity genetics
- Abstract
Recent single-cell RNA sequencing studies have revealed distinct microglial states in development and disease. These include proliferative-region-associated microglia (PAMs) in developing white matter and disease-associated microglia (DAMs) prevalent in various neurodegenerative conditions. PAMs and DAMs share a similar core gene signature. However, the extent of the dynamism and plasticity of these microglial states, as well as their functional significance, remains elusive, partly due to the lack of specific tools. Here, we generated an inducible Cre driver line, Clec7a-CreER
T2 , that targets PAMs and DAMs in the brain parenchyma. Utilizing this tool, we profiled labeled cells during development and in several disease models, uncovering convergence and context-dependent differences in PAM and DAM gene expression. Through long-term tracking, we demonstrated microglial state plasticity. Lastly, we specifically depleted DAMs in demyelination, revealing their roles in disease recovery. Together, we provide a versatile genetic tool to characterize microglial states in CNS development and disease., Competing Interests: Declaration of interests The authors declare no competing interest., (Copyright © 2024 The Author(s). Published by Elsevier Inc. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
13. Identification of direct connections between the dura and the brain.
- Author
-
Smyth LCD, Xu D, Okar SV, Dykstra T, Rustenhoven J, Papadopoulos Z, Bhasiin K, Kim MW, Drieu A, Mamuladze T, Blackburn S, Gu X, Gaitán MI, Nair G, Storck SE, Du S, White MA, Bayguinov P, Smirnov I, Dikranian K, Reich DS, and Kipnis J
- Subjects
- Animals, Humans, Mice, Biological Transport, Encephalomyelitis, Autoimmune, Experimental immunology, Encephalomyelitis, Autoimmune, Experimental metabolism, Gene Expression Profiling, Magnetic Resonance Imaging, Mice, Transgenic, Subarachnoid Space anatomy & histology, Subarachnoid Space blood supply, Subarachnoid Space immunology, Subarachnoid Space metabolism, Cerebrospinal Fluid metabolism, Veins metabolism, Arachnoid anatomy & histology, Arachnoid blood supply, Arachnoid immunology, Arachnoid metabolism, Brain anatomy & histology, Brain blood supply, Brain immunology, Brain metabolism, Dura Mater anatomy & histology, Dura Mater blood supply, Dura Mater immunology, Dura Mater metabolism
- Abstract
The arachnoid barrier delineates the border between the central nervous system and dura mater. Although the arachnoid barrier creates a partition, communication between the central nervous system and the dura mater is crucial for waste clearance and immune surveillance
1,2 . How the arachnoid barrier balances separation and communication is poorly understood. Here, using transcriptomic data, we developed transgenic mice to examine specific anatomical structures that function as routes across the arachnoid barrier. Bridging veins create discontinuities where they cross the arachnoid barrier, forming structures that we termed arachnoid cuff exit (ACE) points. The openings that ACE points create allow the exchange of fluids and molecules between the subarachnoid space and the dura, enabling the drainage of cerebrospinal fluid and limited entry of molecules from the dura to the subarachnoid space. In healthy human volunteers, magnetic resonance imaging tracers transit along bridging veins in a similar manner to access the subarachnoid space. Notably, in neuroinflammatory conditions such as experimental autoimmune encephalomyelitis, ACE points also enable cellular trafficking, representing a route for immune cells to directly enter the subarachnoid space from the dura mater. Collectively, our results indicate that ACE points are a critical part of the anatomy of neuroimmune communication in both mice and humans that link the central nervous system with the dura and its immunological diversity and waste clearance systems., (© 2024. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2024
- Full Text
- View/download PDF
14. An inducible genetic tool for tracking and manipulating specific microglial states in development and disease.
- Author
-
Barclay KM, Abduljawad N, Cheng Z, Kim MW, Zhou L, Yang J, Rustenhoven J, Perez JM, Smyth L, Beatty W, Hou J, Saligrama N, Colonna M, Yu G, Kipnis J, and Li Q
- Abstract
Recent single-cell RNA sequencing studies have revealed distinct microglial states in development and disease. These include proliferative region-associated microglia (PAM) in developing white matter and disease-associated microglia (DAM) prevalent in various neurodegenerative conditions. PAM and DAM share a similar core gene signature and other functional properties. However, the extent of the dynamism and plasticity of these microglial states, as well as their functional significance, remains elusive, partly due to the lack of specific tools. Here, we report the generation of an inducible Cre driver line, Clec7a-CreER
T2 , designed to target PAM and DAM in the brain parenchyma. Utilizing this tool, we profile labeled cells during development and in several disease models, uncovering convergence and context-dependent differences in PAM/DAM gene expression. Through long-term tracking, we demonstrate surprising levels of plasticity in these microglial states. Lastly, we specifically depleted DAM in cuprizone-induced demyelination, revealing their roles in disease progression and recovery., Competing Interests: DECLARATION OF INTERESTS The authors declare no competing interest- Published
- 2023
- Full Text
- View/download PDF
15. Skull bone marrow channels as immune gateways to the central nervous system.
- Author
-
Mazzitelli JA, Pulous FE, Smyth LCD, Kaya Z, Rustenhoven J, Moskowitz MA, Kipnis J, and Nahrendorf M
- Subjects
- Meninges, Skull, Head, Bone Marrow, Central Nervous System
- Abstract
Decades of research have characterized diverse immune cells surveilling the CNS. More recently, the discovery of osseous channels (so-called 'skull channels') connecting the meninges with the skull and vertebral bone marrow has revealed a new layer of complexity in our understanding of neuroimmune interactions. Here we discuss our current understanding of skull and vertebral bone marrow anatomy, its contribution of leukocytes to the meninges, and its surveillance of the CNS. We explore the role of this hematopoietic output on CNS health, focusing on the supply of immune cells during health and disease., (© 2023. Springer Nature America, Inc.)
- Published
- 2023
- Full Text
- View/download PDF
16. Meningeal lymphatics stem cognitive decline in craniosynostosis.
- Author
-
Stevenson TJ, Hitpass Romero K, and Rustenhoven J
- Subjects
- Humans, Skull surgery, Cranial Sutures, Lymphatic System, Craniosynostoses surgery, Cognitive Dysfunction
- Abstract
Craniosynostosis is a congenital craniofacial disorder where premature fusion of cranial sutures causes elevated intracranial pressure and neurological deficits. In this issue of Cell Stem Cell, Ma et al. demonstrate that replenishing skull progenitor cells alleviates intracranial pressure elevations in craniosynostosis by restoring the meningeal lymphatic system, improving neurocognitive function., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2023 Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
17. A novel immune modulator IM33 mediates a glia-gut-neuronal axis that controls lifespan.
- Author
-
Xu W, Rustenhoven J, Nelson CA, Dykstra T, Ferreiro A, Papadopoulos Z, Burnham CD, Dantas G, Fremont DH, and Kipnis J
- Subjects
- Animals, Mice, Drosophila physiology, Dysbiosis, Neuroglia metabolism, Drosophila Proteins metabolism, Longevity, Secretory Leukocyte Peptidase Inhibitor metabolism
- Abstract
Aging is a complex process involving various systems and behavioral changes. Altered immune regulation, dysbiosis, oxidative stress, and sleep decline are common features of aging, but their interconnection is poorly understood. Using Drosophila, we discover that IM33, a novel immune modulator, and its mammalian homolog, secretory leukocyte protease inhibitor (SLPI), are upregulated in old flies and old mice, respectively. Knockdown of IM33 in glia elevates the gut reactive oxygen species (ROS) level and alters gut microbiota composition, including increased Lactiplantibacillus plantarum abundance, leading to a shortened lifespan. Additionally, dysbiosis induces sleep fragmentation through the activation of insulin-producing cells in the brain, which is mediated by the binding of Lactiplantibacillus plantarum-produced DAP-type peptidoglycan to the peptidoglycan recognition protein LE (PGRP-LE) receptor. Therefore, IM33 plays a role in the glia-microbiota-neuronal axis, connecting neuroinflammation, dysbiosis, and sleep decline during aging. Identifying molecular mediators of these processes could lead to the development of innovative strategies for extending lifespan., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2023 Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
18. "Bloody" good factors for keeping the brain young.
- Author
-
Stevenson TJ, Vinnell L, and Rustenhoven J
- Subjects
- Animals, Mice, Brain, Cognition
- Abstract
The increasing burden in dementia-related disorders has necessitated improved understanding of cognitive decline. In a recent issue of Nature, Schroer et al. demonstrate that platelet factor 4 in young blood reduces age-related hippocampal dysfunction and improves cognition in aged mice., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2023 Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
19. Age-dependent immune and lymphatic responses after spinal cord injury.
- Author
-
Salvador AFM, Dykstra T, Rustenhoven J, Gao W, Blackburn SM, Bhasiin K, Dong MQ, Guimarães RM, Gonuguntla S, Smirnov I, Kipnis J, and Herz J
- Subjects
- Mice, Animals, Spinal Cord pathology, Microglia pathology, Myeloid Cells, Mammals, Endothelial Cells pathology, Spinal Cord Injuries pathology
- Abstract
Spinal cord injury (SCI) causes lifelong debilitating conditions. Previous works demonstrated the essential role of the immune system in recovery after SCI. Here, we explored the temporal changes of the response after SCI in young and aged mice in order to characterize multiple immune populations within the mammalian spinal cord. We revealed substantial infiltration of myeloid cells to the spinal cord in young animals, accompanied by changes in the activation state of microglia. In contrast, both processes were blunted in aged mice. Interestingly, we discovered the formation of meningeal lymphatic structures above the lesion site, and their role has not been examined after contusive injury. Our transcriptomic data predicted lymphangiogenic signaling between myeloid cells in the spinal cord and lymphatic endothelial cells (LECs) in the meninges after SCI. Together, our findings delineate how aging affects the immune response following SCI and highlight the participation of the spinal cord meninges in supporting vascular repair., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2023 Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
20. Age-related alterations in meningeal immunity drive impaired CNS lymphatic drainage.
- Author
-
Rustenhoven J, Pavlou G, Storck SE, Dykstra T, Du S, Wan Z, Quintero D, Scallan JP, Smirnov I, Kamm RD, and Kipnis J
- Subjects
- Mice, Animals, Endothelial Cells, Central Nervous System, Meninges, Lymphatic System, Brain physiology, Lymphatic Vessels, Alzheimer Disease
- Abstract
The meningeal lymphatic network enables the drainage of cerebrospinal fluid (CSF) and facilitates the removal of central nervous system (CNS) waste. During aging and in Alzheimer's disease, impaired meningeal lymphatic drainage promotes the buildup of toxic misfolded proteins in the CNS. Reversing this age-related dysfunction represents a promising strategy to augment CNS waste clearance; however, the mechanisms underlying this decline remain elusive. Here, we demonstrate that age-related alterations in meningeal immunity underlie this lymphatic impairment. Single-cell RNA sequencing of meningeal lymphatic endothelial cells from aged mice revealed their response to IFNγ, which was increased in the aged meninges due to T cell accumulation. Chronic elevation of meningeal IFNγ in young mice via AAV-mediated overexpression attenuated CSF drainage-comparable to the deficits observed in aged mice. Therapeutically, IFNγ neutralization alleviated age-related impairments in meningeal lymphatic function. These data suggest manipulation of meningeal immunity as a viable approach to normalize CSF drainage and alleviate the neurological deficits associated with impaired waste removal., (© 2023 Rustenhoven et al.)
- Published
- 2023
- Full Text
- View/download PDF
21. Brain borders at the central stage of neuroimmunology.
- Author
-
Rustenhoven J and Kipnis J
- Subjects
- Spinal Cord immunology, Spinal Cord physiology, Spinal Cord physiopathology, Humans, Nervous System Diseases immunology, Nervous System Diseases physiopathology, Nervous System Diseases psychology, Brain immunology, Brain physiology, Brain physiopathology, Immune System immunology, Immune System physiology, Immune System physiopathology, Neuroimmunomodulation immunology, Neuroimmunomodulation physiology
- Abstract
The concept of immune privilege suggests that the central nervous system is isolated from the immune system. However, recent studies have highlighted the borders of the central nervous system as central sites of neuro-immune interactions. Although the nervous and immune systems both function to maintain homeostasis, under rare circumstances, they can develop pathological interactions that lead to neurological or psychiatric diseases. Here we discuss recent findings that dissect the key anatomical, cellular and molecular mechanisms that enable neuro-immune responses at the borders of the brain and spinal cord and the implications of these interactions for diseases of the central nervous system., (© 2022. Springer Nature Limited.)
- Published
- 2022
- Full Text
- View/download PDF
22. The amyotrophic lateral sclerosis-linked protein TDP-43 regulates interleukin-6 cytokine production by human brain pericytes.
- Author
-
Scotter EL, Cao MC, Jansson D, Rustenhoven J, Smyth LCD, Aalderink MC, Siemens A, Fan V, Wu J, Mee EW, Faull RLM, and Dragunow M
- Subjects
- Humans, Brain metabolism, Cytokines metabolism, Gene Expression, Spinal Cord metabolism, Amyotrophic Lateral Sclerosis metabolism, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, Interleukin-6 metabolism, Pericytes metabolism, Pericytes pathology
- Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal movement disorder involving degeneration of motor neurons through dysfunction of the RNA-binding protein TDP-43. Pericytes, the perivascular cells of the blood-brain, blood-spinal cord, and blood-CSF barriers also degenerate in ALS. Indeed, pericytes are among the earliest cell types to show gene expression changes in pre-symptomatic animal models of ALS. This suggests that pericyte degeneration precedes neurodegeneration and may involve pericyte cell-autonomous TDP-43 dysfunction. Here we determined the effect of TDP-43 dysfunction in human brain pericytes on interleukin 6 (IL-6), a critical secreted inflammatory mediator reported to be regulated by TDP 43. Primary human brain pericytes were cultured from biopsy tissue from epilepsy surgeries and TDP-43 was silenced using siRNA. TDP-43 silencing of pericytes stimulated with pro-inflammatory cytokines, interleukin-1β or tumour necrosis factor alpha, robustly suppressed the induction of IL-6 transcript and protein. IL-6 regulation by TDP-43 did not involve the assembly of TDP-43 nuclear splicing bodies, and did not occur via altered splicing of IL6. Instead, transcriptome-wide analysis by RNA-Sequencing identified a poison exon in the IL6 destabilising factor HNRNPD (AUF1) as a splicing target of TDP-43. Our data support a model whereby TDP-43 silencing favours destabilisation of IL6 mRNA, via enhanced AU-rich element-mediated decay by HNRNP/AUF1. This suggests that cell-autonomous deficits in TDP-43 function in human brain pericytes would suppress their production of IL-6. Given the importance of the blood-brain and blood-spinal cord barriers in maintaining motor neuron health, TDP-43 in human brain pericytes may represent a cellular target for ALS therapeutics., Competing Interests: Declaration of competing interest None., (Copyright © 2022 Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
23. Parenchymal border macrophages regulate the flow dynamics of the cerebrospinal fluid.
- Author
-
Drieu A, Du S, Storck SE, Rustenhoven J, Papadopoulos Z, Dykstra T, Zhong F, Kim K, Blackburn S, Mamuladze T, Harari O, Karch CM, Bateman RJ, Perrin R, Farlow M, Chhatwal J, Hu S, Randolph GJ, Smirnov I, and Kipnis J
- Subjects
- Animals, Mice, Alzheimer Disease metabolism, Brain metabolism, Meninges cytology, Rheology, Extracellular Matrix Proteins metabolism, Aging metabolism, Phagocytosis, Endocytosis, Interferon-gamma metabolism, Humans, Central Nervous System cytology, Central Nervous System metabolism, Cerebrospinal Fluid metabolism, Macrophages physiology, Parenchymal Tissue cytology
- Abstract
Macrophages are important players in the maintenance of tissue homeostasis
1 . Perivascular and leptomeningeal macrophages reside near the central nervous system (CNS) parenchyma2 , and their role in CNS physiology has not been sufficiently well studied. Given their continuous interaction with the cerebrospinal fluid (CSF) and strategic positioning, we refer to these cells collectively as parenchymal border macrophages (PBMs). Here we demonstrate that PBMs regulate CSF flow dynamics. We identify a subpopulation of PBMs that express high levels of CD163 and LYVE1 (scavenger receptor proteins), closely associated with the brain arterial tree, and show that LYVE1+ PBMs regulate arterial motion that drives CSF flow. Pharmacological or genetic depletion of PBMs led to accumulation of extracellular matrix proteins, obstructing CSF access to perivascular spaces and impairing CNS perfusion and clearance. Ageing-associated alterations in PBMs and impairment of CSF dynamics were restored after intracisternal injection of macrophage colony-stimulating factor. Single-nucleus RNA sequencing data obtained from patients with Alzheimer's disease (AD) and from non-AD individuals point to changes in phagocytosis, endocytosis and interferon-γ signalling on PBMs, pathways that are corroborated in a mouse model of AD. Collectively, our results identify PBMs as new cellular regulators of CSF flow dynamics, which could be targeted pharmacologically to alleviate brain clearance deficits associated with ageing and AD., (© 2022. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2022
- Full Text
- View/download PDF
24. Pericytes take up and degrade α-synuclein but succumb to apoptosis under cellular stress.
- Author
-
Stevenson TJ, Johnson RH, Savistchenko J, Rustenhoven J, Woolf Z, Smyth LCD, Murray HC, Faull RLM, Correia J, Schweder P, Heppner P, Turner C, Melki R, Dieriks BV, Curtis MA, and Dragunow M
- Subjects
- Apoptosis, Cytokines metabolism, Humans, Pericytes metabolism, Proteasome Endopeptidase Complex metabolism, Proteasome Inhibitors metabolism, Proteome metabolism, Reactive Oxygen Species metabolism, Ubiquitin metabolism, Parkinson Disease metabolism, alpha-Synuclein metabolism
- Abstract
Parkinson's disease (PD) is characterised by the progressive loss of midbrain dopaminergic neurons and the presence of aggregated α-synuclein (α-syn). Pericytes and microglia, two non-neuronal cells contain α-syn in the human brain, however, their role in disease processes is poorly understood. Pericytes, found surrounding the capillaries in the brain are important for maintaining the blood-brain barrier, controlling blood flow and mediating inflammation. In this study, primary human brain pericytes and microglia were exposed to two different α-synuclein aggregates. Inflammatory responses were assessed using immunocytochemistry, cytometric bead arrays and proteome profiler cytokine array kits. Fixed flow cytometry was used to investigate the uptake and subsequent degradation of α-syn in pericytes. We found that the two α-syn aggregates are devoid of inflammatory and cytotoxic actions on human brain derived pericytes and microglia. Although α-syn did not induce an inflammatory response, pericytes efficiently take up and degrade α-syn through the lysosomal pathway but not the ubiquitin-proteasome system. Furthermore, when pericytes were exposed the ubiquitin proteasome inhibitor-MG132 and α-syn aggregates, there was profound cytotoxicity through the production of reactive oxygen species resulting in apoptosis. These results suggest that the observed accumulation of α-syn in pericytes in human PD brains likely plays a role in PD pathogenesis, perhaps by causing cerebrovascular instability, under conditions of cellular stress., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
25. Cerebrospinal fluid regulates skull bone marrow niches via direct access through dural channels.
- Author
-
Mazzitelli JA, Smyth LCD, Cross KA, Dykstra T, Sun J, Du S, Mamuladze T, Smirnov I, Rustenhoven J, and Kipnis J
- Subjects
- Cerebrospinal Fluid, Head, Meninges, Myeloid Cells metabolism, Bone Marrow physiology, Skull
- Abstract
It remains unclear how immune cells from skull bone marrow niches are recruited to the meninges. Here we report that cerebrospinal fluid (CSF) accesses skull bone marrow via dura-skull channels, and CSF proteins signal onto diverse cell types within the niches. After spinal cord injury, CSF-borne cues promote myelopoiesis and egress of myeloid cells into meninges. This reveals a mechanism of CNS-to-bone-marrow communication via CSF that regulates CNS immune responses., (© 2022. The Author(s), under exclusive licence to Springer Nature America, Inc.)
- Published
- 2022
- Full Text
- View/download PDF
26. Characterisation of PDGF-BB:PDGFRβ signalling pathways in human brain pericytes: evidence of disruption in Alzheimer's disease.
- Author
-
Smyth LCD, Highet B, Jansson D, Wu J, Rustenhoven J, Aalderink M, Tan A, Li S, Johnson R, Coppieters N, Handley R, Narayan P, Singh-Bains MK, Schweder P, Turner C, Mee EW, Heppner P, Correia J, Park TI, Curtis MA, Faull RLM, and Dragunow M
- Subjects
- Becaplermin metabolism, Becaplermin pharmacology, Brain metabolism, Humans, Receptor, Platelet-Derived Growth Factor beta metabolism, Receptor, Platelet-Derived Growth Factor beta pharmacology, Alzheimer Disease metabolism, Pericytes
- Abstract
Platelet-derived growth factor-BB (PDGF-BB):PDGF receptor-β (PDGFRβ) signalling in brain pericytes is critical to the development, maintenance and function of a healthy blood-brain barrier (BBB). Furthermore, BBB impairment and pericyte loss in Alzheimer's disease (AD) is well documented. We found that PDGF-BB:PDGFRβ signalling components were altered in human AD brains, with a marked reduction in vascular PDGFB. We hypothesised that reduced PDGF-BB:PDGFRβ signalling in pericytes may impact on the BBB. We therefore tested the effects of PDGF-BB on primary human brain pericytes in vitro to define pathways related to BBB function. Using pharmacological inhibitors, we dissected distinct aspects of the PDGF-BB response that are controlled by extracellular signal-regulated kinase (ERK) and Akt pathways. PDGF-BB promotes the proliferation of pericytes and protection from apoptosis through ERK signalling. In contrast, PDGF-BB:PDGFRβ signalling through Akt augments pericyte-derived inflammatory secretions. It may therefore be possible to supplement PDGF-BB signalling to stabilise the cerebrovasculature in AD., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
27. Routine culture and study of adult human brain cells from neurosurgical specimens.
- Author
-
Park TI, Smyth LCD, Aalderink M, Woolf ZR, Rustenhoven J, Lee K, Jansson D, Smith A, Feng S, Correia J, Heppner P, Schweder P, Mee E, and Dragunow M
- Subjects
- Humans, Neurons cytology, Adult, Cells, Cultured, Neurosurgical Procedures methods, Brain cytology, Cell Culture Techniques methods
- Abstract
When modeling disease in the laboratory, it is important to use clinically relevant models. Patient-derived human brain cells grown in vitro to study and test potential treatments provide such a model. Here, we present simple, highly reproducible coordinated procedures that can be used to routinely culture most cell types found in the human brain from single neurosurgically excised brain specimens. The cell types that can be cultured include dissociated cultures of neurons, astrocytes, microglia, pericytes and brain endothelial and neural precursor cells, as well as explant cultures of the leptomeninges, cortical slice cultures and brain tumor cells. The initial setup of cultures takes ~2 h, and the cells are ready for further experiments within days to weeks. The resulting cells can be studied as purified or mixed population cultures, slice cultures and explant-derived cultures. This protocol therefore enables the investigation of human brain cells to facilitate translation of neuroscience research to the clinic., (© 2022. The Author(s), under exclusive licence to Springer Nature Limited.)
- Published
- 2022
- Full Text
- View/download PDF
28. A privileged brain.
- Author
-
Rustenhoven J
- Subjects
- Animals, Antigen-Presenting Cells immunology, Bone Marrow Cells immunology, Bone Marrow Cells physiology, Brain physiology, Cerebrospinal Fluid immunology, Dura Mater immunology, Humans, Immune Privilege, Leukocytes immunology, Mice, T-Lymphocytes immunology, Brain immunology, Immunologic Surveillance, Neuroimmunomodulation
- Abstract
Local neuroimmune collaboration safeguards the central nervous system.
- Published
- 2021
- Full Text
- View/download PDF
29. Profiling sensory neuron microenvironment after peripheral and central axon injury reveals key pathways for neural repair.
- Author
-
Avraham O, Feng R, Ewan EE, Rustenhoven J, Zhao G, and Cavalli V
- Subjects
- Animals, Axons, Biomarkers metabolism, Cell Proliferation, Cellular Microenvironment, Fenofibrate administration & dosage, Ganglia, Spinal metabolism, Macrophages cytology, Mice, PPAR alpha metabolism, Sensory Receptor Cells cytology, Sensory Receptor Cells metabolism, Single-Cell Analysis, Spinal Cord Injuries pathology, Spinal Cord Injuries physiopathology, Ganglia, Spinal cytology, Sensory Receptor Cells physiology
- Abstract
Sensory neurons with cell bodies in dorsal root ganglia (DRG) represent a useful model to study axon regeneration. Whereas regeneration and functional recovery occurs after peripheral nerve injury, spinal cord injury or dorsal root injury is not followed by regenerative outcomes. Regeneration of sensory axons in peripheral nerves is not entirely cell autonomous. Whether the DRG microenvironment influences the different regenerative capacities after injury to peripheral or central axons remains largely unknown. To answer this question, we performed a single-cell transcriptional profiling of mouse DRG in response to peripheral (sciatic nerve crush) and central axon injuries (dorsal root crush and spinal cord injury). Each cell type responded differently to the three types of injuries. All injuries increased the proportion of a cell type that shares features of both immune cells and glial cells. A distinct subset of satellite glial cells (SGC) appeared specifically in response to peripheral nerve injury. Activation of the PPARα signaling pathway in SGC, which promotes axon regeneration after peripheral nerve injury, failed to occur after central axon injuries. Treatment with the FDA-approved PPARα agonist fenofibrate increased axon regeneration after dorsal root injury. This study provides a map of the distinct DRG microenvironment responses to peripheral and central injuries at the single-cell level and highlights that manipulating non-neuronal cells could lead to avenues to promote functional recovery after CNS injuries or disease., Competing Interests: OA, RF, EE, JR, GZ, VC No competing interests declared, (© 2021, Avraham et al.)
- Published
- 2021
- Full Text
- View/download PDF
30. Skull and vertebral bone marrow are myeloid cell reservoirs for the meninges and CNS parenchyma.
- Author
-
Cugurra A, Mamuladze T, Rustenhoven J, Dykstra T, Beroshvili G, Greenberg ZJ, Baker W, Papadopoulos Z, Drieu A, Blackburn S, Kanamori M, Brioschi S, Herz J, Schuettpelz LG, Colonna M, Smirnov I, and Kipnis J
- Subjects
- Animals, Bone Marrow physiology, Brain cytology, Brain immunology, Brain physiology, Cell Movement, Central Nervous System cytology, Central Nervous System Diseases pathology, Dura Mater cytology, Dura Mater immunology, Dura Mater physiology, Encephalomyelitis, Autoimmune, Experimental immunology, Encephalomyelitis, Autoimmune, Experimental pathology, Homeostasis, Meninges cytology, Meninges physiology, Mice, Monocytes physiology, Neutrophils physiology, Spinal Cord cytology, Spinal Cord immunology, Spinal Cord physiology, Spinal Cord Injuries immunology, Spinal Cord Injuries pathology, Bone Marrow Cells physiology, Central Nervous System immunology, Central Nervous System Diseases immunology, Meninges immunology, Myeloid Cells physiology, Skull anatomy & histology, Spine anatomy & histology
- Abstract
The meninges are a membranous structure enveloping the central nervous system (CNS) that host a rich repertoire of immune cells mediating CNS immune surveillance. Here, we report that the mouse meninges contain a pool of monocytes and neutrophils supplied not from the blood but by adjacent skull and vertebral bone marrow. Under pathological conditions, including spinal cord injury and neuroinflammation, CNS-infiltrating myeloid cells can originate from brain borders and display transcriptional signatures distinct from their blood-derived counterparts. Thus, CNS borders are populated by myeloid cells from adjacent bone marrow niches, strategically placed to supply innate immune cells under homeostatic and pathological conditions. These findings call for a reinterpretation of immune-cell infiltration into the CNS during injury and autoimmunity and may inform future therapeutic approaches that harness meningeal immune cells., (Copyright © 2021 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2021
- Full Text
- View/download PDF
31. Cerebrovascular Anomalies: Perspectives From Immunology and Cerebrospinal Fluid Flow.
- Author
-
Rustenhoven J, Tanumihardja C, and Kipnis J
- Subjects
- Animals, Cerebral Arteries immunology, Cerebral Arteries metabolism, Cerebral Veins immunology, Cerebral Veins metabolism, Genetic Predisposition to Disease, Heredity, Humans, Phenotype, Risk Factors, Adaptive Immunity, Brain blood supply, Central Nervous System Vascular Malformations cerebrospinal fluid, Central Nervous System Vascular Malformations genetics, Central Nervous System Vascular Malformations immunology, Central Nervous System Vascular Malformations therapy, Cerebral Arteries abnormalities, Cerebral Veins abnormalities, Immunity, Innate
- Abstract
Appropriate vascular function is essential for the maintenance of central nervous system homeostasis and is achieved through virtue of the blood-brain barrier; a specialized structure consisting of endothelial, mural, and astrocytic interactions. While appropriate blood-brain barrier function is typically achieved, the central nervous system vasculature is not infallible and cerebrovascular anomalies, a collective terminology for diverse vascular lesions, are present in meningeal and cerebral vasculature supplying and draining the brain. These conditions, including aneurysmal formation and rupture, arteriovenous malformations, dural arteriovenous fistulas, and cerebral cavernous malformations, and their associated neurological sequelae, are typically managed with neurosurgical or pharmacological approaches. However, increasing evidence implicates interacting roles for inflammatory responses and disrupted central nervous system fluid flow with respect to vascular perturbations. Here, we discuss cerebrovascular anomalies from an immunologic angle and fluid flow perspective. We describe immune contributions, both common and distinct, to the formation and progression of diverse cerebrovascular anomalies. Next, we summarize how cerebrovascular anomalies precipitate diverse neurological sequelae, including seizures, hydrocephalus, and cognitive effects and possible contributions through the recently identified lymphatic and glymphatic systems. Finally, we speculate on and provide testable hypotheses for novel nonsurgical therapeutic approaches for alleviating neurological impairments arising from cerebrovascular anomalies, with a particular emphasis on the normalization of fluid flow and alleviation of inflammation through manipulations of the lymphatic and glymphatic central nervous system clearance pathways.
- Published
- 2021
- Full Text
- View/download PDF
32. Cardiac glycosides target barrier inflammation of the vasculature, meninges and choroid plexus.
- Author
-
Jansson D, Dieriks VB, Rustenhoven J, Smyth LCD, Scotter E, Aalderink M, Feng S, Johnson R, Schweder P, Mee E, Heppner P, Turner C, Curtis M, Faull R, and Dragunow M
- Subjects
- Blood-Brain Barrier metabolism, Blood-Brain Barrier pathology, Cells, Cultured, Choroid Plexus metabolism, Choroid Plexus pathology, Drug Evaluation, Preclinical, Endothelial Cells metabolism, Endothelial Cells pathology, High-Throughput Screening Assays, Humans, Inflammation metabolism, Inflammation pathology, Inflammation Mediators metabolism, Meninges metabolism, Meninges pathology, Pericytes metabolism, Pericytes pathology, Tissue Culture Techniques, Anti-Inflammatory Agents pharmacology, Blood-Brain Barrier drug effects, Choroid Plexus drug effects, Digoxin pharmacology, Endothelial Cells drug effects, Inflammation drug therapy, Lanatosides pharmacology, Meninges drug effects, Pericytes drug effects
- Abstract
Neuroinflammation is a key component of virtually all neurodegenerative diseases, preceding neuronal loss and associating directly with cognitive impairment. Neuroinflammatory signals can originate and be amplified at barrier tissues such as brain vasculature, surrounding meninges and the choroid plexus. We designed a high content screening system to target inflammation in human brain-derived cells of the blood-brain barrier (pericytes and endothelial cells) to identify inflammatory modifiers. Screening an FDA-approved drug library we identify digoxin and lanatoside C, members of the cardiac glycoside family, as inflammatory-modulating drugs that work in blood-brain barrier cells. An ex vivo assay of leptomeningeal and choroid plexus explants confirm that these drugs maintain their function in 3D cultures of brain border tissues. These results suggest that cardiac glycosides may be useful in targeting inflammation at border regions of the brain and offer new options for drug discovery approaches for neuroinflammatory driven degeneration.
- Published
- 2021
- Full Text
- View/download PDF
33. Functional characterization of the dural sinuses as a neuroimmune interface.
- Author
-
Rustenhoven J, Drieu A, Mamuladze T, de Lima KA, Dykstra T, Wall M, Papadopoulos Z, Kanamori M, Salvador AF, Baker W, Lemieux M, Da Mesquita S, Cugurra A, Fitzpatrick J, Sviben S, Kossina R, Bayguinov P, Townsend RR, Zhang Q, Erdmann-Gilmore P, Smirnov I, Lopes MB, Herz J, and Kipnis J
- Subjects
- Animals, Antigen Presentation immunology, Antigen-Presenting Cells metabolism, Antigens cerebrospinal fluid, Cellular Senescence, Chemokine CXCL12 pharmacology, Dura Mater blood supply, Female, Homeostasis, Humans, Immunity, Male, Mice, Inbred C57BL, Phenotype, Stromal Cells cytology, T-Lymphocytes cytology, Mice, Cranial Sinuses immunology, Cranial Sinuses physiology, Dura Mater immunology, Dura Mater physiology
- Abstract
Despite the established dogma of central nervous system (CNS) immune privilege, neuroimmune interactions play an active role in diverse neurological disorders. However, the precise mechanisms underlying CNS immune surveillance remain elusive; particularly, the anatomical sites where peripheral adaptive immunity can sample CNS-derived antigens and the cellular and molecular mediators orchestrating this surveillance. Here, we demonstrate that CNS-derived antigens in the cerebrospinal fluid (CSF) accumulate around the dural sinuses, are captured by local antigen-presenting cells, and are presented to patrolling T cells. This surveillance is enabled by endothelial and mural cells forming the sinus stromal niche. T cell recognition of CSF-derived antigens at this site promoted tissue resident phenotypes and effector functions within the dural meninges. These findings highlight the critical role of dural sinuses as a neuroimmune interface, where brain antigens are surveyed under steady-state conditions, and shed light on age-related dysfunction and neuroinflammatory attack in animal models of multiple sclerosis., Competing Interests: Declaration of interests J.K. is a shareholder and a member of the scientific advisory group for PureTech., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
34. Meningeal γδ T cells regulate anxiety-like behavior via IL-17a signaling in neurons.
- Author
-
Alves de Lima K, Rustenhoven J, Da Mesquita S, Wall M, Salvador AF, Smirnov I, Martelossi Cebinelli G, Mamuladze T, Baker W, Papadopoulos Z, Lopes MB, Cao WS, Xie XS, Herz J, and Kipnis J
- Subjects
- Animals, Anxiety psychology, Behavior, Animal, Cell Proliferation, Cerebral Cortex metabolism, Cerebral Cortex physiopathology, Disease Models, Animal, Dura Mater, Gene Expression Profiling, Gene Expression Regulation, Interleukin-17 genetics, Meninges immunology, Meninges metabolism, Mice, Mice, Knockout, Receptors, Antigen, T-Cell, gamma-delta genetics, Signal Transduction, Transcriptome, Anxiety etiology, Anxiety metabolism, Interleukin-17 metabolism, Neurons immunology, Neurons metabolism, Receptors, Antigen, T-Cell, gamma-delta metabolism, T-Lymphocyte Subsets immunology, T-Lymphocyte Subsets metabolism
- Abstract
Interleukin (IL)-17a has been highly conserved during evolution of the vertebrate immune system and widely studied in contexts of infection and autoimmunity. Studies suggest that IL-17a promotes behavioral changes in experimental models of autism and aggregation behavior in worms. Here, through a cellular and molecular characterization of meningeal γδ17 T cells, we defined the nearest central nervous system-associated source of IL-17a under homeostasis. Meningeal γδ T cells express high levels of the chemokine receptor CXCR6 and seed meninges shortly after birth. Physiological release of IL-17a by these cells was correlated with anxiety-like behavior in mice and was partially dependent on T cell receptor engagement and commensal-derived signals. IL-17a receptor was expressed in cortical glutamatergic neurons under steady state and its genetic deletion decreased anxiety-like behavior in mice. Our findings suggest that IL-17a production by meningeal γδ17 T cells represents an evolutionary bridge between this conserved anti-pathogen molecule and survival behavioral traits in vertebrates.
- Published
- 2020
- Full Text
- View/download PDF
35. Isolation and culture of functional adult human neurons from neurosurgical brain specimens.
- Author
-
Park TI, Schweder P, Lee K, Dieriks BV, Jung Y, Smyth L, Rustenhoven J, Mee E, Heppner P, Turner C, Curtis MA, Faull RLM, Montgomery JM, and Dragunow M
- Abstract
The ability to characterize and study primary neurons isolated directly from the adult human brain would greatly advance neuroscience research. However, significant challenges such as accessibility of human brain tissue and the lack of a robust neuronal cell culture protocol have hampered its progress. Here, we describe a simple and reproducible method for the isolation and culture of functional adult human neurons from neurosurgical brain specimens . In vitro , adult human neurons form a dense network and express a plethora of mature neuronal and synaptic markers. Most importantly, for the first time, we demonstrate the re-establishment of mature neurophysiological properties in vitro , such as repetitive fast-spiking action potentials, and spontaneous and evoked synaptic activity. Together, our dissociated and slice culture systems enable studies of adult human neurophysiology and gene expression under normal and pathological conditions and provide a high-throughput platform for drug testing on brain cells directly isolated from the adult human brain., (© The Author(s) (2020). Published by Oxford University Press on behalf of the Guarantors of Brain.)
- Published
- 2020
- Full Text
- View/download PDF
36. Meningeal Immunity and Its Function in Maintenance of the Central Nervous System in Health and Disease.
- Author
-
Alves de Lima K, Rustenhoven J, and Kipnis J
- Subjects
- Animals, Humans, Lymphatic Vessels immunology, Lymphatic Vessels metabolism, Neuroimmunomodulation, T-Lymphocyte Subsets immunology, T-Lymphocyte Subsets metabolism, Central Nervous System immunology, Central Nervous System metabolism, Disease Susceptibility, Homeostasis, Immunity, Meninges physiology
- Abstract
Neuroimmunology, albeit a relatively established discipline, has recently sparked numerous exciting findings on microglia, the resident macrophages of the central nervous system (CNS). This review addresses meningeal immunity, a less-studied aspect of neuroimmune interactions. The meninges, a triple layer of membranes-the pia mater, arachnoid mater, and dura mater-surround the CNS, encompassing the cerebrospinal fluid produced by the choroid plexus epithelium. Unlike the adjacent brain parenchyma, the meninges contain a wide repertoire of immune cells. These constitute meningeal immunity, which is primarily concerned with immune surveillance of the CNS, and-according to recent evidence-also participates in postinjury CNS recovery, chronic neurodegenerative conditions, and even higher brain function. Meningeal immunity has recently come under the spotlight owing to the characterization of meningeal lymphatic vessels draining the CNS. Here, we review the current state of our understanding of meningeal immunity and its effects on healthy and diseased brains.
- Published
- 2020
- Full Text
- View/download PDF
37. Bypassing the blood-brain barrier.
- Author
-
Rustenhoven J and Kipnis J
- Subjects
- Animals, Homeostasis, Humans, Immunologic Surveillance, Blood-Brain Barrier immunology, Meninges immunology
- Published
- 2019
- Full Text
- View/download PDF
38. Smelling Danger: Olfactory Stem Cells Control Immune Defense during Chronic Inflammation.
- Author
-
Rustenhoven J and Kipnis J
- Subjects
- Humans, Inflammation, Neurogenesis, Stem Cells, Nerve Regeneration, Olfactory Receptor Neurons
- Abstract
Neurogenesis is critical to continuously replacie olfactory neurons but is impaired during chronic inflammatory rhinosinusitis. In this issue of Cell Stem Cell, Chen et al. (2019) describe the inflammation-induced switching of olfactory stem cells from a regenerative phenotype to one participating in immune defense; this process contributes to deficient replacement of olfactory sensory neurons., (Copyright © 2019 Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
39. Old T Cells Interfer(on) with Neurogenesis.
- Author
-
Rustenhoven J, Herz J, and Kipnis J
- Subjects
- Adult, Brain, Humans, Neurogenesis, Single-Cell Analysis, T-Lymphocytes
- Abstract
Adult neurogenesis plays an important role in brain function and declines with aging. A recent report demonstrates clonal T cell expansion within neurogenic niches of the aged brain, impairing neurogenesis through IFNγ signaling (Dulken et al.,Nature, 2019). These results highlight T cells as important contributors to and potential therapeutic targets for age-related brain dysfunction., (Copyright © 2019 Elsevier Ltd. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
40. Markers for human brain pericytes and smooth muscle cells.
- Author
-
Smyth LCD, Rustenhoven J, Scotter EL, Schweder P, Faull RLM, Park TIH, and Dragunow M
- Subjects
- Actins metabolism, Biomarkers metabolism, Blood-Brain Barrier cytology, Brain cytology, CD13 Antigens metabolism, CD146 Antigen metabolism, Desmin metabolism, Humans, Myocytes, Smooth Muscle cytology, Pericytes cytology, Receptor, Platelet-Derived Growth Factor beta metabolism, Blood-Brain Barrier metabolism, Brain metabolism, Myocytes, Smooth Muscle metabolism, Pericytes metabolism
- Abstract
Brain pericytes and vascular smooth muscle cells (vSMCs) are a critical component of the neurovascular unit and are important in regulating cerebral blood flow and blood-brain barrier integrity. Identification of subtypes of mural cells in tissue and in vitro is important to any study of their function, therefore we identified distinct mural cell morphologies in neurologically normal post-mortem human brain. Further, the distribution of mural cell markers platelet-derived growth factor receptor-β (PDGFRβ), α-smooth muscle actin (αSMA), CD13, neural/glial antigen-2 (NG2), CD146 and desmin was examined. We determined that PDGFRβ, NG2, CD13, and CD146 were expressed in capillary-associated pericytes. NG2, and CD13 were also present on vSMCs in large vessels, however abundant CD146 and desmin staining was also detected in vSMCs on large vessels, co-labelling with αSMA. To determine whether cultures recapitulated observations from tissue, primary human brain pericytes derived from neurologically normal autopsies were analysed for the presence of pericyte markers by immunocytochemistry, western blotting and qPCR. The proteins observed in brain pericytes in tissue (PDGFRβ, αSMA, desmin, CD146, CD13, and NG2) were present in vitro, validating a panel of proteins that can be used to label brain pericytes and vSMCs in tissue and in vitro. Finally, we showed that the proteins CD146 and desmin that are expressed on large vessels in situ, are also selective markers of a smooth muscle cell phenotype in vitro., (Copyright © 2018 Elsevier B.V. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
41. PU.1 regulates Alzheimer's disease-associated genes in primary human microglia.
- Author
-
Rustenhoven J, Smith AM, Smyth LC, Jansson D, Scotter EL, Swanson MEV, Aalderink M, Coppieters N, Narayan P, Handley R, Overall C, Park TIH, Schweder P, Heppner P, Curtis MA, Faull RLM, and Dragunow M
- Subjects
- Alzheimer Disease genetics, Alzheimer Disease pathology, Gene Expression Regulation drug effects, Histone Deacetylase Inhibitors pharmacology, Humans, Microglia drug effects, Vorinostat pharmacology, Alzheimer Disease metabolism, Gene Expression Regulation physiology, Microglia metabolism, Proto-Oncogene Proteins metabolism, Trans-Activators metabolism
- Abstract
Background: Microglia play critical roles in the brain during homeostasis and pathological conditions. Understanding the molecular events underpinning microglial functions and activation states will further enable us to target these cells for the treatment of neurological disorders. The transcription factor PU.1 is critical in the development of myeloid cells and a major regulator of microglial gene expression. In the brain, PU.1 is specifically expressed in microglia and recent evidence from genome-wide association studies suggests that reductions in PU.1 contribute to a delayed onset of Alzheimer's disease (AD), possibly through limiting neuroinflammatory responses., Methods: To investigate how PU.1 contributes to immune activation in human microglia, microarray analysis was performed on primary human mixed glial cultures subjected to siRNA-mediated knockdown of PU.1. Microarray hits were confirmed by qRT-PCR and immunocytochemistry in both mixed glial cultures and isolated microglia following PU.1 knockdown. To identify attenuators of PU.1 expression in microglia, high throughput drug screening was undertaken using a compound library containing FDA-approved drugs. NanoString and immunohistochemistry was utilised to investigate the expression of PU.1 itself and PU.1-regulated mediators in primary human brain tissue derived from neurologically normal and clinically and pathologically confirmed cases of AD., Results: Bioinformatic analysis of gene expression upon PU.1 silencing in mixed glial cultures revealed a network of modified AD-associated microglial genes involved in the innate and adaptive immune systems, particularly those involved in antigen presentation and phagocytosis. These gene changes were confirmed using isolated microglial cultures. Utilising high throughput screening of FDA-approved compounds in mixed glial cultures we identified the histone deacetylase inhibitor vorinostat as an effective attenuator of PU.1 expression in human microglia. Further characterisation of vorinostat in isolated microglial cultures revealed gene and protein changes partially recapitulating those seen following siRNA-mediated PU.1 knockdown. Lastly, we demonstrate that several of these PU.1-regulated genes are expressed by microglia in the human AD brain in situ., Conclusions: Collectively, these results suggest that attenuating PU.1 may be a valid therapeutic approach to limit microglial-mediated inflammatory responses in AD and demonstrate utility of vorinostat for this purpose.
- Published
- 2018
- Full Text
- View/download PDF
42. Unique and shared inflammatory profiles of human brain endothelia and pericytes.
- Author
-
Smyth LCD, Rustenhoven J, Park TI, Schweder P, Jansson D, Heppner PA, O'Carroll SJ, Mee EW, Faull RLM, Curtis M, and Dragunow M
- Subjects
- Blood-Brain Barrier cytology, Blood-Brain Barrier drug effects, Brain cytology, Brain drug effects, Cells, Cultured, Coculture Techniques, Endothelial Cells drug effects, Humans, Inflammation chemically induced, Inflammation metabolism, Inflammation pathology, Inflammation Mediators pharmacology, Pericytes drug effects, Blood-Brain Barrier metabolism, Brain metabolism, Endothelial Cells metabolism, Inflammation Mediators metabolism, Pericytes metabolism
- Abstract
Background: Pericytes and endothelial cells are critical cellular components of the blood-brain barrier (BBB) and play an important role in neuroinflammation. To date, the majority of inflammation-related studies in endothelia and pericytes have been carried out using immortalised cell lines or non-human-derived cells. Whether these are representative of primary human cells is unclear and systematic comparisons of the inflammatory responses of primary human brain-derived pericytes and endothelia has yet to be performed., Methods: To study the effects of neuroinflammation at the BBB, primary brain endothelial cells and pericytes were isolated from human biopsy tissue. Culture purity was examined using qPCR and immunocytochemistry. Electrical cell-substrate impedance sensing (ECIS) was used to determine the barrier properties of endothelial and pericyte cultures. Using immunocytochemistry, cytometric bead array, and ECIS, we compared the responses of endothelia and pericytes to a panel of inflammatory stimuli (IL-1β, TNFα, LPS, IFN-γ, TGF-β
1 , IL-6, and IL-4). Secretome analysis was performed to identify unique secretions of endothelia and pericytes in response to IL-1β., Results: Endothelial cells were pure, moderately proliferative, retained the expression of BBB-related junctional proteins and transporters, and generated robust TEER. Both endothelia and pericytes have the same pattern of transcription factor activation in response to inflammatory stimuli but respond differently at the secretion level. Secretome analysis confirmed that endothelia and pericytes have overlapping but distinct secretome profiles in response to IL-1β. We identified several cell-type specific responses, including G-CSF and GM-CSF (endothelial-specific), and IGFBP2 and IGFBP3 (pericyte-specific). Finally, we demonstrated that direct addition of IL-1β, TNFα, LPS, and IL-4 contributed to the loss of endothelial barrier integrity in vitro., Conclusions: Here, we identify important cell-type differences in the inflammatory response of brain pericytes and endothelia and provide, for the first time, a comprehensive profile of the secretions of primary human brain endothelia and pericytes which has implications for understanding how inflammation affects the cerebrovasculature.- Published
- 2018
- Full Text
- View/download PDF
43. Modelling physiological and pathological conditions to study pericyte biology in brain function and dysfunction.
- Author
-
Rustenhoven J, Smyth LC, Jansson D, Schweder P, Aalderink M, Scotter EL, Mee EW, Faull RLM, Park TI, and Dragunow M
- Subjects
- Blood-Brain Barrier pathology, Brain pathology, Cells, Cultured, Cytokines metabolism, Fibronectins metabolism, Humans, Interleukin-1beta metabolism, Blood-Brain Barrier physiology, Brain physiology, Gene Expression Regulation physiology, Pericytes pathology, Pericytes physiology
- Abstract
Background: Brain pericytes ensheathe the endothelium and contribute to formation and maintenance of the blood-brain-barrier. Additionally, pericytes are involved in several aspects of the CNS immune response including scarring, adhesion molecule expression, chemokine secretion, and phagocytosis. In vitro cultures are routinely used to investigate these functions of brain pericytes, however, these are highly plastic cells and can display differing phenotypes and functional responses depending on their culture conditions. Here we sought to investigate how two commonly used culture media, high serum containing DMEM/F12 and low serum containing Pericyte Medium (ScienCell), altered the phenotype of human brain pericytes and neuroinflammatory responses., Methods: Pericytes were isolated from adult human brain biopsy tissue and cultured in DMEM/F12 (D-pericytes) or Pericyte Medium (P-pericytes). Immunocytochemistry, qRT-PCR, and EdU incorporation were used to determine how this altered their basal phenotype, including the expression of pericyte markers, proliferation, and cell morphology. To determine whether culture media altered the inflammatory response in human brain pericytes, immunocytochemistry, qRT-PCR, cytometric bead arrays, and flow cytometry were used to investigate transcription factor induction, chemokine secretion, adhesion molecule expression, migration, phagocytosis, and response to inflammatory-related growth factors., Results: P-pericytes displayed elevated proliferation and a distinct bipolar morphology compared to D-pericytes. Additionally, P-pericytes displayed lower expression of pericyte-associated markers NG2, PDGFRβ, and fibronectin, with notably lower αSMA, CD146, P4H and desmin, and higher Col-IV expression. Nuclear NF-kB translocation in response to IL-1β stimulation was observed in both cultures, however, P-pericytes displayed elevated expression of the transcription factor C/EBPδ, and lower expression of the adhesion molecule ICAM-1. P-pericytes displayed elevated phagocytic and migratory ability. Both cultures responded similarly to stimulation by the growth factors TGFβ
1 and PDGF-BB., Conclusions: Despite differences in their phenotype and magnitude of response, both P-pericytes and D-pericytes responded similarly to all examined functions, indicating that the neuroinflammatory phenotype of these cells is robust to culture conditions.- Published
- 2018
- Full Text
- View/download PDF
44. Brain Pericytes As Mediators of Neuroinflammation.
- Author
-
Rustenhoven J, Jansson D, Smyth LC, and Dragunow M
- Subjects
- Animals, Brain immunology, Encephalitis drug therapy, Encephalitis immunology, Humans, Pericytes immunology, Brain pathology, Encephalitis pathology, Pericytes pathology
- Abstract
Brain pericytes are perivascular cells that regulate capillary function, and this localization puts them in a pivotal position for the regulation of central nervous system (CNS) inflammatory responses at the neurovascular unit. Neuroinflammation, driven by microglia and astrocytes or resulting from peripheral leukocyte infiltration, has both homeostatic and detrimental consequences for brain function and is present in nearly every neurological disorder. More recently, brain pericytes have been shown to have many properties of immune regulating cells, including responding to and expressing a plethora of inflammatory molecules, presenting antigen, and displaying phagocytic ability. In this review we highlight the emerging role of pericytes in neuroinflammation and discuss pericyte-mediated neuroinflammation as a potential therapeutic target for the treatment of a range of devastating brain disorders., (Copyright © 2016 Elsevier Ltd. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
45. Interferon-γ blocks signalling through PDGFRβ in human brain pericytes.
- Author
-
Jansson D, Scotter EL, Rustenhoven J, Coppieters N, Smyth LC, Oldfield RL, Bergin PS, Mee EW, Graham ES, Faull RL, and Dragunow M
- Abstract
Background: Neuroinflammation and blood-brain barrier (BBB) disruption are common features of many brain disorders, including Alzheimer's disease, epilepsy, and motor neuron disease. Inflammation is thought to be a driver of BBB breakdown, but the underlying mechanisms for this are unclear. Brain pericytes are critical cells for maintaining the BBB and are immunologically active. We sought to test the hypothesis that inflammation regulates the BBB by altering pericyte biology., Methods: We exposed primary adult human brain pericytes to chronic interferon-gamma (IFNγ) for 4 days and measured associated functional aspects of pericyte biology. Specifically, we examined the influence of inflammation on platelet-derived growth factor receptor-beta (PDGFRβ) expression and signalling, as well as pericyte proliferation and migration by qRT-PCR, immunocytochemistry, flow cytometry, and western blotting., Results: Chronic IFNγ treatment had marked effects on pericyte biology most notably through the PDGFRβ, by enhancing agonist (PDGF-BB)-induced receptor phosphorylation, internalization, and subsequent degradation. Functionally, chronic IFNγ prevented PDGF-BB-mediated pericyte proliferation and migration., Conclusions: Because PDGFRβ is critical for pericyte function and its removal leads to BBB leakage, our results pinpoint a mechanism linking chronic brain inflammation to BBB dysfunction.
- Published
- 2016
- Full Text
- View/download PDF
46. Cultured pericytes from human brain show phenotypic and functional differences associated with differential CD90 expression.
- Author
-
Park TI, Feisst V, Brooks AE, Rustenhoven J, Monzo HJ, Feng SX, Mee EW, Bergin PS, Oldfield R, Graham ES, Curtis MA, Faull RL, Dunbar PR, and Dragunow M
- Subjects
- Adult, Biomarkers metabolism, Blood-Brain Barrier cytology, Blood-Brain Barrier metabolism, Brain metabolism, Cell Proliferation, Cells, Cultured, Female, Flow Cytometry, Gene Expression Regulation, Humans, Male, Pericytes metabolism, Phenotype, Young Adult, Brain cytology, Pericytes cytology, Thy-1 Antigens metabolism
- Abstract
The human brain is a highly vascular organ in which the blood-brain barrier (BBB) tightly regulates molecules entering the brain. Pericytes are an integral cell type of the BBB, regulating vascular integrity, neuroinflammation, angiogenesis and wound repair. Despite their importance, identifying pericytes amongst other perivascular cell types and deciphering their specific role in the neurovasculature remains a challenge. Using primary adult human brain cultures and fluorescent-activated cell sorting, we identified two CD73(+)CD45(-) mesenchymal populations that showed either high or low CD90 expression. CD90 is known to be present on neurons in the brain and peripheral blood vessels. We found in the human brain, that CD90 immunostaining localised to the neurovasculature and often associated with pericytes. In vitro, CD90(+) cells exhibited higher basal proliferation, lower expression of markers αSMA and CD140b, produced less extracellular matrix (ECM) proteins, and exhibited lesser pro-inflammatory responses when compared to the CD90(-) population. Thus, CD90 distinguishes two interrelated, yet functionally distinct pericyte populations in the adult human brain that may have discrete roles in neurovascular function, immune response and scar formation.
- Published
- 2016
- Full Text
- View/download PDF
47. Erratum to: Studying Human Brain Inflammation in Leptomeningeal and Choroid Plexus Explant Cultures.
- Author
-
Dragunow M, Feng S, Rustenhoven J, Curtis M, and Faull R
- Published
- 2016
- Full Text
- View/download PDF
48. Studying Human Brain Inflammation in Leptomeningeal and Choroid Plexus Explant Cultures.
- Author
-
Dragunow M, Feng S, Rustenhoven J, Curtis M, and Faull R
- Subjects
- Cell Movement, Cell Proliferation, Cicatrix pathology, Cytokines pharmacology, Fibroblasts cytology, Humans, Macrophages cytology, Microglia cytology, Pericytes cytology, Tissue Culture Techniques, Choroid Plexus cytology, Encephalitis pathology, Meninges cytology
- Abstract
The meninges (dura, pia and arachnoid) are critical membranes encasing and protecting the brain within the skull. The leptomeninges, which comprise the arachnoid and pia, have many functions beyond brain protection including roles in neurogenesis, fibrotic scar formation and brain inflammation. Similarly, the choroid plexus plays important roles in normal brain function but is also involved in brain inflammation. We have begun studying the role of human leptomeninges and choroid plexus in brain inflammation and leptomeninges in fibrotic scar formation, using human brain derived explant cultures. To study the composition of the cells generated in these explants we undertook immunocytochemical characterisation. Cells, mainly pericytes and meningeal macrophages, emerge from leptomeningeal explants (LME's) and respond to inflammatory mediators by producing inflammatory molecules. LME-derived cells also respond to mechanical injury and cytokines, providing an in vitro human brain model of fibrotic scar formation. Choroid plexus explants (CPE's) generate epithelial cells, pericytes and microglia/macrophages. CPE-derived cells also respond to inflammatory mediators. LME and CPE explants survive and generate cells for many months in vitro and provide a remarkable opportunity to study basic mechanisms of human brain inflammation and fibrosis and to test human-active anti-inflammatory and anti-scarring treatments.
- Published
- 2016
- Full Text
- View/download PDF
49. TGF-beta1 regulates human brain pericyte inflammatory processes involved in neurovasculature function.
- Author
-
Rustenhoven J, Aalderink M, Scotter EL, Oldfield RL, Bergin PS, Mee EW, Graham ES, Faull RL, Curtis MA, Park TI, and Dragunow M
- Subjects
- Cell Proliferation drug effects, Cell Survival drug effects, Culture Media, Conditioned pharmacology, Cyclooxygenase 2 metabolism, Humans, Interleukin-1beta pharmacology, Matrix Metalloproteinase 2 metabolism, NADPH Oxidase 4, NADPH Oxidases metabolism, NF-kappa B metabolism, Receptors, Scavenger genetics, Receptors, Scavenger metabolism, Signal Transduction drug effects, Smad2 Protein metabolism, Time Factors, Vascular Cell Adhesion Molecule-1 metabolism, Brain cytology, Cytokines metabolism, Gene Expression Regulation drug effects, Pericytes drug effects, Phagocytes drug effects, Transforming Growth Factor beta1 pharmacology
- Abstract
Background: Transforming growth factor beta 1 (TGFβ1) is strongly induced following brain injury and polarises microglia to an anti-inflammatory phenotype. Augmentation of TGFβ1 responses may therefore be beneficial in preventing inflammation in neurological disorders including stroke and neurodegenerative diseases. However, several other cell types display immunogenic potential and identifying the effect of TGFβ1 on these cells is required to more fully understand its effects on brain inflammation. Pericytes are multifunctional cells which ensheath the brain vasculature and have garnered recent attention with respect to their immunomodulatory potential. Here, we sought to investigate the inflammatory phenotype adopted by TGFβ1-stimulated human brain pericytes., Methods: Microarray analysis was performed to examine transcriptome-wide changes in TGFβ1-stimulated pericytes, and results were validated by qRT-PCR and cytometric bead arrays. Flow cytometry, immunocytochemistry and LDH/Alamar Blue® viability assays were utilised to examine phagocytic capacity of human brain pericytes, transcription factor modulation and pericyte health., Results: TGFβ1 treatment of primary human brain pericytes induced the expression of several inflammatory-related genes (NOX4, COX2, IL6 and MMP2) and attenuated others (IL8, CX3CL1, MCP1 and VCAM1). A synergistic induction of IL-6 was seen with IL-1β/TGFβ1 treatment whilst TGFβ1 attenuated the IL-1β-induced expression of CX3CL1, MCP-1 and sVCAM-1. TGFβ1 was found to signal through SMAD2/3 transcription factors but did not modify nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kB) translocation. Furthermore, TGFβ1 attenuated the phagocytic ability of pericytes, possibly through downregulation of the scavenger receptors CD36, CD47 and CD68. Whilst TGFβ did decrease pericyte number, this was due to a reduction in proliferation, not apoptotic death or compromised cell viability., Conclusions: TGFβ1 attenuated pericyte expression of key chemokines and adhesion molecules involved in CNS leukocyte trafficking and the modulation of microglial function, as well as reduced the phagocytic ability of pericytes. However, TGFβ1 also enhanced the expression of classical pro-inflammatory cytokines and enzymes which can disrupt BBB functioning, suggesting that pericytes adopt a phenotype which is neither solely pro- nor anti-inflammatory. Whilst the effects of pericyte modulation by TGFβ1 in vivo are difficult to infer, the reduction in pericyte proliferation together with the elevated IL-6, MMP-2 and NOX4 and reduced phagocytosis suggests a detrimental action of TGFβ1 on neurovasculature.
- Published
- 2016
- Full Text
- View/download PDF
50. Isolation of highly enriched primary human microglia for functional studies.
- Author
-
Rustenhoven J, Park TI, Schweder P, Scotter J, Correia J, Smith AM, Gibbons HM, Oldfield RL, Bergin PS, Mee EW, Faull RL, Curtis MA, Scott Graham E, and Dragunow M
- Subjects
- Biomarkers, Cells, Cultured, Chemokines biosynthesis, Cytokines biosynthesis, Gene Expression Regulation, Humans, Immunohistochemistry, Interleukin-1beta metabolism, NF-kappa B metabolism, Phagocytosis, Phenotype, Protein Transport, Cell Separation methods, Microglia cytology, Microglia physiology
- Abstract
Microglia, the resident macrophages of the central nervous system play vital roles in brain homeostasis through clearance of pathogenic material. Microglia are also implicated in neurological disorders through uncontrolled activation and inflammatory responses. To date, the vast majority of microglial studies have been performed using rodent models. Human microglia differ from rodent counterparts in several aspects including their response to pharmacological substances and their inflammatory secretions. Such differences highlight the need for studies on primary adult human brain microglia and methods to isolate them are therefore required. Our procedure generates microglial cultures of >95% purity from both biopsy and autopsy human brain tissue using a very simple media-based culture procedure that takes advantage of the adherent properties of these cells. Microglia obtained in this manner can be utilised for research within a week. Isolated microglia demonstrate phagocytic ability and respond to inflammatory stimuli and their purity makes them suitable for numerous other forms of in vitro studies, including secretome and transcriptome analysis. Furthermore, this protocol allows for the simultaneous isolation of neural precursor cells during the microglial isolation procedure. As human brain tissue is such a precious and valuable resource the simultaneous isolation of multiple cell types is highly beneficial.
- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.