1
|
Pandya VA, Patani R. The role of glial cells in amyotrophic lateral sclerosis. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2024; 176:381-450. [PMID: 38802179 DOI: 10.1016/bs.irn.2024.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) has traditionally been considered a neuron-centric disease. This view is now outdated, with increasing recognition of cell autonomous and non-cell autonomous contributions of central and peripheral nervous system glia to ALS pathomechanisms. With glial research rapidly accelerating, we comprehensively interrogate the roles of astrocytes, microglia, oligodendrocytes, ependymal cells, Schwann cells and satellite glia in nervous system physiology and ALS-associated pathology. Moreover, we highlight the inter-glial, glial-neuronal and inter-system polylogue which constitutes the healthy nervous system and destabilises in disease. We also propose classification based on function for complex glial reactive phenotypes and discuss the pre-requisite for integrative modelling to advance translation. Given the paucity of life-enhancing therapies currently available for ALS patients, we discuss the promising potential of harnessing glia in driving ALS therapeutic discovery.
Collapse
Affiliation(s)
- Virenkumar A Pandya
- University College London Medical School, London, United Kingdom; The Francis Crick Institute, London, United Kingdom.
| | - Rickie Patani
- The Francis Crick Institute, London, United Kingdom; Department of Neuromuscular Diseases, University College London Queen Square Institute of Neurology, Queen Square, London, United Kingdom.
| |
Collapse
|
2
|
Gilchrist JM, Yang ND, Jiang V, Moyer BD. Pharmacologic Characterization of LTGO-33, a Selective Small Molecule Inhibitor of the Voltage-Gated Sodium Channel Na V1.8 with a Unique Mechanism of Action. Mol Pharmacol 2024; 105:233-249. [PMID: 38195157 DOI: 10.1124/molpharm.123.000789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 11/28/2023] [Accepted: 01/02/2024] [Indexed: 01/11/2024] Open
Abstract
Discovery and development of new molecules directed against validated pain targets is required to advance the treatment of pain disorders. Voltage-gated sodium channels (NaVs) are responsible for action potential initiation and transmission of pain signals. NaV1.8 is specifically expressed in peripheral nociceptors and has been genetically and pharmacologically validated as a human pain target. Selective inhibition of NaV1.8 can ameliorate pain while minimizing effects on other NaV isoforms essential for cardiac, respiratory, and central nervous system physiology. Here we present the pharmacology, interaction site, and mechanism of action of LTGO-33, a novel NaV1.8 small molecule inhibitor. LTGO-33 inhibited NaV1.8 in the nM potency range and exhibited over 600-fold selectivity against human NaV1.1-NaV1.7 and NaV1.9. Unlike prior reported NaV1.8 inhibitors that preferentially interacted with an inactivated state via the pore region, LTGO-33 was state-independent with similar potencies against closed and inactivated channels. LTGO-33 displayed species specificity for primate NaV1.8 over dog and rodent NaV1.8 and inhibited action potential firing in human dorsal root ganglia neurons. Using chimeras combined with mutagenesis, the extracellular cleft of the second voltage-sensing domain was identified as the key site required for channel inhibition. Biophysical mechanism of action studies demonstrated that LTGO-33 inhibition was relieved by membrane depolarization, suggesting the molecule stabilized the deactivated state to prevent channel opening. LTGO-33 equally inhibited wild-type and multiple NaV1.8 variants associated with human pain disorders. These collective results illustrate LTGO-33 inhibition via both a novel interaction site and mechanism of action previously undescribed in NaV1.8 small molecule pharmacologic space. SIGNIFICANCE STATEMENT: NaV1.8 sodium channels primarily expressed in peripheral pain-sensing neurons represent a validated target for the development of novel analgesics. Here we present the selective small molecule NaV1.8 inhibitor LTGO-33 that interdicts a distinct site in a voltage-sensor domain to inhibit channel opening. These results inform the development of new analgesics for pain disorders.
Collapse
Affiliation(s)
| | - Nien-Du Yang
- Latigo Biotherapeutics, Inc., Thousand Oaks, California
| | | | - Bryan D Moyer
- Latigo Biotherapeutics, Inc., Thousand Oaks, California
| |
Collapse
|
3
|
Bennet BM, Pardo ID, Assaf BT, Buza E, Cramer SD, Crawford LK, Engelhardt JA, Galbreath EJ, Grubor B, Morrison JP, Osborne TS, Sharma AK, Bolon B. Scientific and Regulatory Policy Committee Technical Review: Biology and Pathology of Ganglia in Animal Species Used for Nonclinical Safety Testing. Toxicol Pathol 2023; 51:278-305. [PMID: 38047294 DOI: 10.1177/01926233231213851] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Dorsal root ganglia (DRG), trigeminal ganglia (TG), other sensory ganglia, and autonomic ganglia may be injured by some test article classes, including anti-neoplastic chemotherapeutics, adeno-associated virus-based gene therapies, antisense oligonucleotides, nerve growth factor inhibitors, and aminoglycoside antibiotics. This article reviews ganglion anatomy, cytology, and pathology (emphasizing sensory ganglia) among common nonclinical species used in assessing product safety for such test articles (TAs). Principal histopathologic findings associated with sensory ganglion injury include neuron degeneration, necrosis, and/or loss; increased satellite glial cell and/or Schwann cell numbers; and leukocyte infiltration and/or inflammation. Secondary nerve fiber degeneration and/or glial reactions may occur in nerves, dorsal spinal nerve roots, spinal cord (dorsal and occasionally lateral funiculi), and sometimes the brainstem. Ganglion findings related to TA administration may result from TA exposure and/or trauma related to direct TA delivery into the central nervous system or ganglia. In some cases, TA-related effects may need to be differentiated from a spectrum of artifactual and/or spontaneous background changes.
Collapse
Affiliation(s)
| | | | | | - Elizabeth Buza
- University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | | | | | | | | | - James P Morrison
- Charles River Laboratories, Inc., Shrewsbury, Massachusetts, USA
| | | | | | | |
Collapse
|
4
|
Wannemacher R, Reiß A, Rohn K, Lühder F, Flügel A, Baumgärtner W, Hülskötter K. Ovalbumin-specific CD4 + and CD8 + T cells contribute to different susceptibility for Theiler's murine encephalomyelitis virus persistence. Front Immunol 2023; 14:1194842. [PMID: 37292191 PMCID: PMC10244668 DOI: 10.3389/fimmu.2023.1194842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 05/12/2023] [Indexed: 06/10/2023] Open
Abstract
Theiler's murine encephalomyelitis virus (TMEV) is the causative agent of TMEV-induced demyelinating disease (TMEV-IDD); a well-established animal model for the chronic progressive form of human multiple sclerosis (MS). In susceptible mice with an inadequate immune response, TMEV-IDD is triggered by virus persistence and maintained by a T cell mediated immunopathology. OT-mice are bred on a TMEV-resistant C57BL/6 background and own predominantly chicken ovalbumin (OVA)-specific populations of CD8+ T cells (OT-I) or CD4+ T cells (OT-II), respectively. It is hypothesized that the lack of antigen specific T cell populations increases susceptibility for a TMEV-infection in OT-mice on a TMEV-resistant C57BL/6 background. OT-I, OT-II, and C57BL/6 control mice were infected intracerebrally with the TMEV-BeAn strain. Mice were scored weekly for clinical disease and after necropsy, histological and immunohistochemical evaluation was performed. OT-I mice started to develop progressive motor dysfunction between 7 and 21 days post infection (dpi), leading up to hind limb paresis and critical weight loss, which resulted in euthanasia for humane reasons between 14 and 35 dpi. OT-I mice displayed a high cerebral virus load, an almost complete absence of CD8+ T cells from the central nervous system (CNS) and a significantly diminished CD4+ T cell response. Contrarily, only 60% (12 of 20) of infected OT-II mice developed clinical disease characterized by mild ataxia. 25% of clinically affected OT-II mice (3 of 12) made a full recovery. 5 of 12 OT-II mice with clinical disease developed severe motor dysfunction similar to OT-I mice and were euthanized for humane reasons between 13 and 37 dpi. OT-II mice displayed only low virus-immunoreactivity, but clinical disease correlated well with severely reduced infiltration of CD8+ T cells and the increased presence of CD4+ T cells in the brains of OT-II mice. Though further studies are needed to reveal the underlying pathomechanisms following TMEV infection in OT mice, findings indicate an immunopathological process as a main contributor to clinical disease in OT-II mice, while a direct virus-associated pathology may be the main contributor to clinical disease in TMEV-infected OT-I mice.
Collapse
Affiliation(s)
- Rouven Wannemacher
- Department of Pathology, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Anna Reiß
- Department of Pathology, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
- Center for Systems Neuroscience, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Karl Rohn
- Department of Biometry, Epidemiology and Data Processing, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Fred Lühder
- Institute of Neuroimmunology and Multiple Sclerosis Research, University Medical Center Göttingen, Göttingen, Germany
| | - Alexander Flügel
- Center for Systems Neuroscience, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
- Institute of Neuroimmunology and Multiple Sclerosis Research, University Medical Center Göttingen, Göttingen, Germany
| | - Wolfgang Baumgärtner
- Department of Pathology, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
- Center for Systems Neuroscience, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Kirsten Hülskötter
- Department of Pathology, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| |
Collapse
|
5
|
Haberberger RV, Kuramatilake J, Barry CM, Matusica D. Ultrastructure of dorsal root ganglia. Cell Tissue Res 2023:10.1007/s00441-023-03770-w. [PMID: 37079097 PMCID: PMC10115609 DOI: 10.1007/s00441-023-03770-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 03/28/2023] [Indexed: 04/21/2023]
Abstract
Dorsal root ganglia (DRG) contains thousands of sensory neurons that transmit information about our external and internal environment to the central nervous system. This includes signals related to proprioception, temperature, and nociception. Our understanding of DRG has increased tremendously over the last 50 years and has established the DRG as an active participant in peripheral processes. This includes interactions between neurons and non-neuronal cells such as satellite glia cells and macrophages that contribute to an increasingly complex cellular environment that modulates neuronal function. Early ultrastructural investigations of the DRG have described subtypes of sensory neurons based on differences in the arrangement of organelles such as the Golgi apparatus and the endoplasmic reticulum. The neuron-satellite cell complex and the composition of the axon hillock in DRG have also been investigated, but, apart from basic descriptions of Schwann cells, ultrastructural investigations of other cell types in DRG are limited. Furthermore, detailed descriptions of key components of DRG, such as blood vessels and the capsule that sits at the intersection of the meninges and the connective tissue covering the peripheral nervous system, are lacking to date. With rising interest in DRG as potential therapeutic targets for aberrant signalling associated with chronic pain conditions, gaining further insights into DRG ultrastructure will be fundamental to understanding cell-cell interactions that modulate DRG function. In this review, we aim to provide a synopsis of the current state of knowledge on the ultrastructure of the DRG and its components, as well as to identify areas of interest for future studies.
Collapse
Affiliation(s)
- Rainer Viktor Haberberger
- Division of Anatomy and Pathology, School of Biomedicine, The University of Adelaide, Adelaide, Australia.
| | - Jaliya Kuramatilake
- Division of Anatomy and Pathology, School of Biomedicine, The University of Adelaide, Adelaide, Australia
| | - Christine M Barry
- Anatomy, Histology & Pathology, College of Medicine and Public Health, Flinders University, Adelaide, Australia
| | - Dusan Matusica
- Anatomy, Histology & Pathology, College of Medicine and Public Health, Flinders University, Adelaide, Australia
| |
Collapse
|
6
|
Klein I, Boenert J, Lange F, Christensen B, Wassermann MK, Wiesen MHJ, Olschewski DN, Rabenstein M, Müller C, Lehmann HC, Fink GR, Schroeter M, Rueger MA, Vay SU. Glia from the central and peripheral nervous system are differentially affected by paclitaxel chemotherapy via modulating their neuroinflammatory and neuroregenerative properties. Front Pharmacol 2022; 13:1038285. [PMID: 36408236 PMCID: PMC9666700 DOI: 10.3389/fphar.2022.1038285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 10/06/2022] [Indexed: 11/07/2022] Open
Abstract
Glia are critical players in defining synaptic contacts and maintaining neuronal homeostasis. Both astrocytes as glia of the central nervous system (CNS), as well as satellite glial cells (SGC) as glia of the peripheral nervous system (PNS), intimately interact with microglia, especially under pathological conditions when glia regulate degenerative as well as regenerative processes. The chemotherapeutic agent paclitaxel evokes peripheral neuropathy and cognitive deficits; however, the mechanisms underlying these diverse clinical side effects are unclear. We aimed to elucidate the direct effects of paclitaxel on the function of astrocytes, microglia, and SGCs, and their glia-glia and neuronal-glia interactions. After intravenous application, paclitaxel was present in the dorsal root ganglia of the PNS and the CNS of rodents. In vitro, SGC enhanced the expression of pro-inflammatory factors and reduced the expression of neurotrophic factor NT-3 upon exposure to paclitaxel, resulting in predominantly neurotoxic effects. Likewise, paclitaxel induced a switch towards a pro-inflammatory phenotype in microglia, exerting neurotoxicity. In contrast, astrocytes expressed neuroprotective markers and increasingly expressed S100A10 after paclitaxel exposure. Astrocytes, and to a lesser extent SGCs, had regulatory effects on microglia independent of paclitaxel exposure. Data suggest that paclitaxel differentially modulates glia cells regarding their (neuro-) inflammatory and (neuro-) regenerative properties and also affects their interaction. By elucidating those processes, our data contribute to the understanding of the mechanistic pathways of paclitaxel-induced side effects in CNS and PNS.
Collapse
Affiliation(s)
- Ines Klein
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Neurology, Cologne, Germany
| | - Janne Boenert
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Neurology, Cologne, Germany
| | - Felix Lange
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Neurology, Cologne, Germany
| | - Britt Christensen
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Neurology, Cologne, Germany
| | - Meike K. Wassermann
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Neurology, Cologne, Germany
| | - Martin H. J. Wiesen
- Center of Pharmacology, Therapeutic Drug Monitoring, University Hospital of Cologne, Cologne, Germany
| | - Daniel Navin Olschewski
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Neurology, Cologne, Germany
| | - Monika Rabenstein
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Neurology, Cologne, Germany
| | - Carsten Müller
- Center of Pharmacology, Therapeutic Drug Monitoring, University Hospital of Cologne, Cologne, Germany
| | - Helmar C. Lehmann
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Neurology, Cologne, Germany
| | - Gereon Rudolf Fink
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Neurology, Cologne, Germany
- Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Centre Juelich, Juelich, Germany
| | - Michael Schroeter
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Neurology, Cologne, Germany
- Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Centre Juelich, Juelich, Germany
| | - Maria Adele Rueger
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Neurology, Cologne, Germany
- Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Centre Juelich, Juelich, Germany
| | - Sabine Ulrike Vay
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Neurology, Cologne, Germany
- *Correspondence: Sabine Ulrike Vay,
| |
Collapse
|
7
|
Zdora I, Jubran L, Allnoch L, Hansmann F, Baumgärtner W, Leitzen E. Morphological and phenotypical characteristics of porcine satellite glial cells of the dorsal root ganglia. Front Neuroanat 2022; 16:1015281. [PMID: 36337140 PMCID: PMC9626980 DOI: 10.3389/fnana.2022.1015281] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 10/04/2022] [Indexed: 10/23/2023] Open
Abstract
Satellite glial cells (SGCs) of the dorsal root ganglia (DRG) ensure homeostasis and proportional excitability of sensory neurons and gained interest in the field of development and maintenance of neuropathic pain. Pigs represent a suitable species for translational medicine with a more similar anatomy and physiology to humans compared to rodents, and are used in research regarding treatment of neuropathic pain. Knowledge of anatomical and physiological features of porcine SGCs is prerequisite for interpreting potential alterations. However, state of knowledge is still limited. In the present study, light microscopy, ultrastructural analysis and immunofluorescence staining was performed. SGCs tightly surround DRG neurons with little vascularized connective tissue between SGC-neuron units, containing, among others, axons and Schwann cells. DRG were mainly composed of large sized neurons (∼59%), accompanied by fewer medium sized (∼36%) and small sized sensory neurons (∼6%). An increase of neuronal body size was concomitant with an increased number of surrounding SGCs. The majority of porcine SGCs expressed glutamine synthetase and inwardly rectifying potassium channel Kir 4.1, known as SGC-specific markers in other species. Similar to canine SGCs, marked numbers of porcine SGCs were immunopositive for glial fibrillary acidic protein, 2',3'-cyclic-nucleotide 3'-phosphodiesterase and the transcription factor Sox2. Low to moderate numbers of SGCs showed aquaporin 4-immunoreactivity (AQP4) as described for murine SGCs. AQP4-immunoreactivity was primarily found in SGCs ensheathing small and medium sized neuronal somata. Low numbers of SGCs were immunopositive for ionized calcium-binding adapter molecule 1, indicating a potential immune cell character. No immunoreactivity for common leukocyte antigen CD45 nor neural/glial antigen 2 was detected. The present study provides essential insights into the characteristic features of non-activated porcine SGCs, contributing to a better understanding of this cell population and its functional aspects. This will help to interpret possible changes that might occur under activating conditions such as pain.
Collapse
Affiliation(s)
- Isabel Zdora
- Department of Pathology, University of Veterinary Medicine, Hanover, Germany
- Center of Systems Neuroscience, Hanover Graduate School for Neurosciences, Infection Medicine, and Veterinary Sciences (HGNI), Hanover, Germany
| | - Lorna Jubran
- Department of Pathology, University of Veterinary Medicine, Hanover, Germany
- Center of Systems Neuroscience, Hanover Graduate School for Neurosciences, Infection Medicine, and Veterinary Sciences (HGNI), Hanover, Germany
| | - Lisa Allnoch
- Department of Pathology, University of Veterinary Medicine, Hanover, Germany
| | - Florian Hansmann
- Department of Pathology, University of Veterinary Medicine, Hanover, Germany
| | - Wolfgang Baumgärtner
- Department of Pathology, University of Veterinary Medicine, Hanover, Germany
- Center of Systems Neuroscience, Hanover Graduate School for Neurosciences, Infection Medicine, and Veterinary Sciences (HGNI), Hanover, Germany
| | - Eva Leitzen
- Department of Pathology, University of Veterinary Medicine, Hanover, Germany
| |
Collapse
|
8
|
Glutamine Maintains Satellite Glial Cells Growth and Survival in Culture. Neurochem Res 2022; 47:3635-3646. [PMID: 35522367 DOI: 10.1007/s11064-022-03614-z] [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: 01/08/2022] [Revised: 04/10/2022] [Accepted: 04/19/2022] [Indexed: 10/18/2022]
Abstract
Satellite glial cells (SGCs) tightly surround neurons and modulate sensory transmission in dorsal root ganglion (DRG). At present, the biological property of primary SGCs in culture deserves further investigation. To reveal the key factor for SGCs growth and survival, we examined the effects of different culture supplementations containing Dulbecco's Modified Eagle Medium (DMEM)/F12, DMEM high glucose (HG) or Neurobasal-A (NB). CCK-8 proliferation assay showed an increased proliferation of SGCs in DMEM/F12 and DMEM/HG, but not in NB medium. Bax, AnnexinV, and propidium iodide (PI) staining results showed that NB medium caused cell death and apoptosis. We showed that glutamine was over 2.5 mM in DMEM/F12 and DMEM/HG, whereas it was absence in NB medium. Interestingly, exogenous glutamine application significantly reversed the poor proliferation and cell death of SGCs in NB medium. These findings demonstrated that DMEM/F12 medium was optimal to get high-purity SGCs. Glutamine was the key molecule to maintain SGCs growth and survival in culture. Here, we provided a novel approach to get high-purity SGCs by changing the key component of culture medium. Our study shed a new light on understanding the biological property and modulation of glial cells of primary sensory ganglia.
Collapse
|
9
|
Tukov FF, Mansfield K, Milton M, Meseck E, Penraat K, Chand D, Hartmann A. Single-Dose Intrathecal Dorsal Root Ganglia Toxicity of Onasemnogene Abeparvovec in Cynomolgus Monkeys. Hum Gene Ther 2022; 33:740-756. [PMID: 35331006 PMCID: PMC9347375 DOI: 10.1089/hum.2021.255] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Intravenous onasemnogene abeparvovec is approved for the treatment of spinal muscular atrophy in children < 2 years. For later-onset patients, intrathecal onasemnogene abeparvovec may be advantageous over intravenous administration. Recently, microscopic dorsal root ganglion (DRG) changes were observed in nonhuman primates (NHPs) following intrathecal onasemnogene abeparvovec administration. To characterize these DRG findings, two NHP studies evaluating intrathecal onasemnogene abeparvovec administration were conducted: a 12-month study with a 6-week interim cohort and a 13-week study with a 2-week interim cohort. The latter investigated the potential impact of prednisolone or rituximab plus everolimus on DRG toxicity. An additional 6-month, single-dose, intravenous NHP study conducted in parallel evaluated onasemnogene abeparvovec safety (including DRG toxicity) with or without prednisolone coadministration. Intrathecal onasemnogene abeparvovec administration was well tolerated and not associated with clinical observations. Microscopic onasemnogene abeparvovec-related changes were observed in the DRG and trigeminal ganglion (TG) and included mononuclear cell inflammation and/or neuronal degeneration, which was colocalized with high vector transcript expression at 6 weeks postdose. Incidence and severity of DRG changes were generally decreased after 52 weeks compared with 6 weeks postdose. Other onasemnogene abeparvovec-related microscopic findings of axonal degeneration, mononuclear cell infiltrates and/or gliosis in the spinal cord, dorsal spinal nerve root/spinal nerves, and/or peripheral nerves were absent or found at decreased incidences and/or severities after 52 weeks. DRG and/or TG microscopic findings following intravenous onasemnogene abeparvovec dosing included minimal to slight neuronal degeneration and mononuclear cell inflammation at 6 weeks and 6 months postdose. Nervous system microscopic findings following intrathecal onasemnogene abeparvovec (≥1.2 × 1013 vg/animal) trended toward resolution after 52 weeks, supporting nonprogression of changes, including in the DRG. Onasemnogene abeparvovec-related DRG findings were not associated with electrophysiology changes and were not ameliorated by prednisolone or rituximab plus everolimus coadministration. The pathogenesis is possibly a consequence of increased vector genome transduction and/or transgene expression.
Collapse
Affiliation(s)
| | - Keith Mansfield
- Novartis Institutes for BioMedical Research Inc, 33413, Cambridge, Massachusetts, United States;
| | - Mark Milton
- Novartis Institutes for BioMedical Research Inc, 33413, Cambridge, Massachusetts, United States;
| | - Emily Meseck
- Novartis Pharmaceuticals Corp, 33412, East Hanover, New Jersey, United States;
| | - Kelley Penraat
- Novartis Institutes for BioMedical Research Inc, 33413, Cambridge, Massachusetts, United States;
| | - Deepa Chand
- Novartis Gene Therapies, Inc., Bannockburn, United States.,Washington University School of Medicine in Saint Louis, 12275, St Louis, Missouri, United States;
| | | |
Collapse
|
10
|
Shen S, Tiwari N, Madar J, Mehta P, Qiao LY. Beta 2-adrenergic receptor mediates noradrenergic action to induce cyclic adenosine monophosphate response element-binding protein phosphorylation in satellite glial cells of dorsal root ganglia to regulate visceral hypersensitivity. Pain 2022; 163:180-192. [PMID: 33941754 PMCID: PMC8556417 DOI: 10.1097/j.pain.0000000000002330] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 04/28/2021] [Indexed: 01/03/2023]
Abstract
ABSTRACT Sympathoneuronal outflow into dorsal root ganglia (DRG) is suggested to be involved in sympathetically maintained chronic pain, which is mediated by norepinephrine (NE) action on DRG cells. This study combined in vitro and in vivo approaches to identify the cell types of DRG that received NE action and examined cell type-specific expression of adrenergic receptors (ARs) in DRG. Using DRG explants, we identified that NE acted on satellite glial cells (SGCs) to induce the phosphorylation of cAMP response element-binding protein (CREB). Using primarily cultured SGCs, we identified that beta (β)2-adrenergic receptor but not alpha (α)adrenergic receptor nor other βAR isoforms mediated NE-induced CREB phosphorylation and CRE-promoted luciferase transcriptional activity. Using fluorescence in situ hybridization and affinity purification of mRNA from specific cell types, we identified that β2AR was expressed by SGCs but not DRG neurons. We further examined β2AR expression and CREB phosphorylation in vivo in a model of colitis in which sympathetic nerve sprouting in DRG was observed. We found that β2AR expression and CREB phosphorylation were increased in SGCs of thoracolumbar DRG on day 7 after colitis induction. Inhibition but not augmentation of β2AR reduced colitis-induced calcitonin gene-related peptide release into the spinal cord dorsal horn and colonic pain responses to colorectal distention. Prolonged activation of β2AR in naive DRG increased calcitonin gene-related peptide expression in DRG neurons. These findings provide molecular basis of sympathetic modulation of sensory activity and chronic pain that involves β2AR-mediated signaling in SGCs of DRG.
Collapse
Affiliation(s)
- Shanwei Shen
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | | | | | | | | |
Collapse
|
11
|
Huang B, Zdora I, de Buhr N, Eikelberg D, Baumgärtner W, Leitzen E. Phenotypical changes of satellite glial cells in a murine model of G M1 -gangliosidosis. J Cell Mol Med 2021; 26:527-539. [PMID: 34877779 PMCID: PMC8743646 DOI: 10.1111/jcmm.17113] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 11/12/2021] [Accepted: 11/25/2021] [Indexed: 12/14/2022] Open
Abstract
Satellite glial cells (SGCs) of dorsal root ganglia (DRG) react in response to various injuries in the nervous system. This study investigates reactive changes within SGCs in a murine model for GM1‐gangliosidosis (GM1). DRG of homozygous β‐galactosidase‐knockout mice and homozygous C57BL/6 wild‐type mice were investigated performing immunostaining on formalin‐fixed, paraffin‐embedded tissue. A marked upregulation of glial fibrillary acidic protein (GFAP), the progenitor marker nestin and Ki67 within SGCs of diseased mice, starting after 4 months at the earliest GFAP, along with intracytoplasmic accumulation of ganglioside within neurons and deterioration of clinical signs was identified. Interestingly, nestin‐positive SGCs were detected after 8 months only. No changes regarding inwardly rectifying potassium channel 4.1, 2, 3‐cyclic nucleotide 3‐phosphodiesterase, Sox2, doublecortin, periaxin and caspase3 were observed in SGCs. Iba1 was only detected in close vicinity of SGCs indicating infiltrating or tissue‐resident macrophages. These results indicate that SGCs of DRG show phenotypical changes during the course of GM1, characterized by GFAP upregulation, proliferation and expression of a neural progenitor marker at a late time point. This points towards an important role of SGCs during neurodegenerative disorders and supports that SGCs represent a multipotent glial precursor cell line with high plasticity and functionality.
Collapse
Affiliation(s)
- Bei Huang
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany.,Center of Systems Neuroscience, Hannover, Germany
| | - Isabel Zdora
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany.,Center of Systems Neuroscience, Hannover, Germany
| | - Nicole de Buhr
- Department of Biochemistry, University of Veterinary Medicine Hannover, Hannover, Germany.,Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, Hannover, Germany
| | - Deborah Eikelberg
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Wolfgang Baumgärtner
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany.,Center of Systems Neuroscience, Hannover, Germany
| | - Eva Leitzen
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany
| |
Collapse
|
12
|
Discrepancy in the Usage of GFAP as a Marker of Satellite Glial Cell Reactivity. Biomedicines 2021; 9:biomedicines9081022. [PMID: 34440226 PMCID: PMC8391720 DOI: 10.3390/biomedicines9081022] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/04/2021] [Accepted: 08/11/2021] [Indexed: 12/13/2022] Open
Abstract
Satellite glial cells (SGCs) surrounding the neuronal somas in peripheral sensory ganglia are sensitive to neuronal stressors, which induce their reactive state. It is believed that such induced gliosis affects the signaling properties of the primary sensory neurons and is an important component of the neuropathic phenotype leading to pain and other sensory disturbances. Efforts to understand and manipulate such gliosis relies on reliable markers to confirm induced SGC reactivity and ultimately the efficacy of targeted intervention. Glial fibrillary acidic protein (GFAP) is currently the only widely used marker for such analyses. However, we have previously described the lack of SGC upregulation of GFAP in a mouse model of sciatic nerve injury, suggesting that GFAP may not be a universally suitable marker of SGC gliosis across species and experimental models. To further explore this, we here investigate the regulation of GFAP in two different experimental models in both rats and mice. We found that whereas GFAP was upregulated in both rodent species in the applied inflammation model, only the rat demonstrated increased GFAP in SGCs following sciatic nerve injury; we did not observe any such GFAP upregulation in the mouse model at either protein or mRNA levels. Our results demonstrate an important discrepancy between species and experimental models that prevents the usage of GFAP as a universal marker for SGC reactivity.
Collapse
|
13
|
Schwarz S, Mathes K, Wohlsein P. Rhabdomyosarcoma on the Forelimb of a Common Musk Turtle (Sternotherus odoratus). J Comp Pathol 2021; 186:73-76. [PMID: 34340808 DOI: 10.1016/j.jcpa.2021.06.002] [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: 03/23/2021] [Revised: 04/22/2021] [Accepted: 06/14/2021] [Indexed: 10/20/2022]
Abstract
Malignant mesenchymal tumours are only rarely reported in turtles. In the present case, an 8-year-old female common musk turtle (Sternotherus odoratus) was presented with a solid, reddened, non-mobile mass on the right forelimb. The mass had a thin, membranous lining and a grey-white cut surface. Histological examination revealed a cell-rich, focally infiltrative neoplasm consisting of spindloid tumour cells arranged in bundles and streams. Rarely, indistinct cytoplasmic cross-striations were seen in the neoplastic cells. Transmission electron microscopy revealed cytoplasmic, disorganized muscle fibrils and haphazardly arranged, attenuated Z-lines in the neoplastic cells. The histological, histochemical and ultrastructural findings led to the diagnosis of a rhabdomyosarcoma. This is the first description of rhabdomyosarcoma in a freshwater turtle.
Collapse
Affiliation(s)
- Sarah Schwarz
- Department of Pathology, University of Veterinary Medicine, Hannover, Germany
| | - Karina Mathes
- Clinic of Small Mammals, Reptiles and Birds, University of Veterinary Medicine, Hannover, Germany
| | - Peter Wohlsein
- Department of Pathology, University of Veterinary Medicine, Hannover, Germany.
| |
Collapse
|
14
|
Huang B, Zdora I, de Buhr N, Lehmbecker A, Baumgärtner W, Leitzen E. Phenotypical peculiarities and species-specific differences of canine and murine satellite glial cells of spinal ganglia. J Cell Mol Med 2021; 25:6909-6924. [PMID: 34096171 PMCID: PMC8278083 DOI: 10.1111/jcmm.16701] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 05/11/2021] [Accepted: 05/13/2021] [Indexed: 12/16/2022] Open
Abstract
Satellite glial cells (SGCs) are located in the spinal ganglia (SG) of the peripheral nervous system and tightly envelop each neuron. They preserve tissue homeostasis, protect neurons and react in response to injury. This study comparatively characterizes the phenotype of murine (mSGCs) and canine SGCs (cSGCs). Immunohistochemistry and immunofluorescence as well as 2D and 3D imaging techniques were performed to describe a SGC-specific marker panel, identify potential functional subsets and other phenotypical, species-specific peculiarities. Glutamine synthetase (GS) and the potassium channel Kir 4.1 are SGC-specific markers in murine and canine SG. Furthermore, a subset of mSGCs showed CD45 immunoreactivity and the majority of mSGCs were immunopositive for neural/glial antigen 2 (NG2), indicating an immune and a progenitor cell character. The majority of cSGCs were immunopositive for glial fibrillary acidic protein (GFAP), 2',3'-cyclic-nucleotide 3'-phosphodiesterase (CNPase) and Sox2. Therefore, cSGCs resemble central nervous system glial cells and progenitor cells. SGCs lacked expression of macrophage markers CD107b, Iba1 and CD204. Double labelling with GS/Kir 4.1 highlights the unique anatomy of SGC-neuron units and emphasizes the indispensability of further staining and imaging techniques for closer insights into the specific distribution of markers and potential colocalizations.
Collapse
Affiliation(s)
- Bei Huang
- Department of Pathology, University of Veterinary Medicine, Hannover, Germany.,Center of Systems Neuroscience, Hannover, Germany
| | - Isabel Zdora
- Department of Pathology, University of Veterinary Medicine, Hannover, Germany.,Center of Systems Neuroscience, Hannover, Germany
| | - Nicole de Buhr
- Department of Biochemistry, University of Veterinary Medicine, Hannover, Germany.,Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine, Hannover, Germany
| | - Annika Lehmbecker
- Department of Pathology, University of Veterinary Medicine, Hannover, Germany
| | - Wolfgang Baumgärtner
- Department of Pathology, University of Veterinary Medicine, Hannover, Germany.,Center of Systems Neuroscience, Hannover, Germany
| | - Eva Leitzen
- Department of Pathology, University of Veterinary Medicine, Hannover, Germany
| |
Collapse
|
15
|
Harm TA, Hostetter SJ, Nenninger AS, Valentine BN, Ellinwood NM, Smith JD. Temporospatial Development of Neuropathologic Findings in a Canine Model of Mucopolysaccharidosis IIIB. Vet Pathol 2020; 58:205-222. [PMID: 33205707 DOI: 10.1177/0300985820960128] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Mucopolysaccharidosis (MPS) IIIB is a neuropathic lysosomal storage disease characterized by the deficient activity of a lysosomal enzyme obligate for the degradation of the glycosaminoglycan (GAG) heparan sulfate (HS). The pathogenesis of neurodegeneration in MPS IIIB is incompletely understood. Large animal models are attractive for pathogenesis and therapeutic studies due to their larger size, outbred genetics, longer lifespan, and naturally occurring MPS IIIB disease. However, the temporospatial development of neuropathologic changes has not been reported for canine MPS IIIB. Here we describe lesions in 8 brain regions, cervical spinal cord, and dorsal root ganglion (DRG) in a canine model of MPS IIIB that includes dogs aged from 2 to 26 months of age. Pathological changes in the brain included early microscopic vacuolation of glial cells initially observed at 2 months, and vacuolation of neurons initially observed at 10 months. Inclusions within affected cells variably stained positively with PAS and LFB stains. Quantitative immunohistochemistry demonstrated increased glial expression of GFAP and Iba1 in dogs with MPS IIIB compared to age-matched controls at all time points, suggesting neuroinflammation occurs early in disease. Loss of Purkinje cells was initially observed at 10 months and was pronounced in 18- and 26-month-old dogs with MPS IIIB. Our results support the dog as a replicative model of MPS IIIB neurologic lesions and detail the pathologic and neuroinflammatory changes in the spinal cord and DRG of MPS IIIB-affected dogs.
Collapse
Affiliation(s)
| | - Shannon J Hostetter
- 70724Iowa State University, Ames, IA, USA.,Current address: Department of Veterinary Pathology, University of Georgia, Athens, GA, USA
| | | | | | | | | |
Collapse
|
16
|
Horalskyi LP, Kolesnik NL, Sokulskiy IM, Tsekhmistrenko SI, Dunaievska OF, Goralska IY. Morphology of spinal ganglia of different segmentary levels in the domestic dog. REGULATORY MECHANISMS IN BIOSYSTEMS 2020. [DOI: 10.15421/022076] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
The spinal ganglia, which perform the function of the first link on the afferent impulses’ way from the receptors to the central nervous system, recognize internal and external irritations, and are the first to transform them into a nervous impulse. As the representatives of the peripheral nervous system, they are some of the main objects of the studies in contemporary neuromorphology. Based on the results of anatomic, neurohistological, histochemical, morphometric and statistical methods of the studies, we conducted a complex survey, revealing the morphology of spinal ganglia of different segmental levels in the domestic dog. In particular, we determined the differences in the microscopic structure and morphometric parameters of cervical, thoracic, lumbar and sacral spinal ganglia and the ganglia of the cervical and lumbar enlargements in mature domestic dogs. The study showed that the spinal ganglia of domestic dogs can have different skeletotopy, different shape and sizes due to their species peculiarity. Also, the surveyed animals, according to the results of our studies, had the cervical and thoracic spinal ganglia of oval, while the lumbar and sacral – spindle-like shapes. According to the results of morphometry, the area of the spinal ganglia in lengthwise section differed: the smallest area belonged to the thoracic, the largest to the sacral spinal ganglia. The density of neuronal arrangement per 0.1 mm2 of the area of the spinal ganglia correlated with their sizes: the highest parameter was identified for the thoracic spinal ganglia, the lowest – for the sacral. The conducted studies revealed that histo- and cyto-structure of the spinal ganglia is characteristic of notable differentiation of the nervous cells of small sizes. Therefore, we differentiated neurons of the spinal ganglia into large, medium and small. The highest quantity of large neurons was found in the sacral ganglia, and largest amount of medium-sized neurons – in the ganglia of the lumbar enlargement. In other ganglia, small neurons dominated. Correspondingly, different nuclear-cytoplasmic ratio in these neurons was determined, indicating different extent of morphofunctional condition of nervous cells. We determined content of localization and separation of nucleic acids in histostructure of the spinal cord at the tissue and cellular levels.
Collapse
|
17
|
Hanani M, Spray DC. Emerging importance of satellite glia in nervous system function and dysfunction. Nat Rev Neurosci 2020; 21:485-498. [PMID: 32699292 PMCID: PMC7374656 DOI: 10.1038/s41583-020-0333-z] [Citation(s) in RCA: 180] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/09/2020] [Indexed: 02/08/2023]
Abstract
Satellite glial cells (SGCs) closely envelop cell bodies of neurons in sensory, sympathetic and parasympathetic ganglia. This unique organization is not found elsewhere in the nervous system. SGCs in sensory ganglia are activated by numerous types of nerve injury and inflammation. The activation includes upregulation of glial fibrillary acidic protein, stronger gap junction-mediated SGC-SGC and neuron-SGC coupling, increased sensitivity to ATP, downregulation of Kir4.1 potassium channels and increased cytokine synthesis and release. There is evidence that these changes in SGCs contribute to chronic pain by augmenting neuronal activity and that these changes are consistent in various rodent pain models and likely also in human pain. Therefore, understanding these changes and the resulting abnormal interactions of SGCs with sensory neurons could provide a mechanistic approach that might be exploited therapeutically in alleviation and prevention of pain. We describe how SGCs are altered in rodent models of four common types of pain: systemic inflammation (sickness behaviour), post-surgical pain, diabetic neuropathic pain and post-herpetic pain.
Collapse
Affiliation(s)
- Menachem Hanani
- Laboratory of Experimental Surgery, Hadassah-Hebrew University Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel.
| | - David C Spray
- Dominick Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY, USA
| |
Collapse
|
18
|
Wang Y, Wang S, Wu H, Liu X, Ma J, Khan MA, Riaz A, Wang L, Qiu HJ, Sun Y. Compartmentalized Neuronal Culture for Viral Transport Research. Front Microbiol 2020; 11:1470. [PMID: 32760359 PMCID: PMC7373733 DOI: 10.3389/fmicb.2020.01470] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 06/05/2020] [Indexed: 01/12/2023] Open
Abstract
Neuron-invading viruses usually enter via the peripheral organs/tissues of their mammalian hosts and are transported to the neurons. Virus trafficking is critical for transport or spread within the nervous system. Primary culture of neurons is a valuable and indispensable method for neurobiological research, allowing researchers to investigate basic mechanisms of diverse neuronal functions as well as retrograde and anterograde virus transport in neuronal axons. Primary ganglion sensory neurons from mice can be cultured in a compartmentalized culture device, which allows spatial fluidic separation of cell bodies and distal axons. These neurons serve as an important model for investigating the transport of viruses between the neuronal soma and distal axons. Alphaherpesviruses are fascinating and important human and animal pathogens, they replicate and establish lifelong latent infection in the peripheral nervous system, the mechanism of the viral transport along the axon is the key to understand the virus spread in the nervous system. In this review, we briefly introduce and evaluate the most frequently used compartmentalization tools in viral transport research, with particular emphasis on alphaherpesviruses.
Collapse
Affiliation(s)
- Yimin Wang
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China
| | - Shan Wang
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China
| | - Hongxia Wu
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Xinxin Liu
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China
| | - Jinyou Ma
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China
| | - Muhammad Akram Khan
- Department of Veterinary Pathology, Faculty of Veterinary and Animal Science, PMAS-Arid Agriculture University, Rawalpindi, Pakistan
| | - Aayesha Riaz
- Department of Parasitology and Microbiology, Faculty of Veterinary and Animal Science, PMAS-Arid Agriculture University, Rawalpindi, Pakistan
| | - Lei Wang
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China
| | - Hua-Ji Qiu
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Yuan Sun
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| |
Collapse
|
19
|
Jansen AJG, Spaan T, Low HZ, Di Iorio D, van den Brand J, Tieke M, Barendrecht A, Rohn K, van Amerongen G, Stittelaar K, Baumgärtner W, Osterhaus A, Kuiken T, Boons GJ, Huskens J, Boes M, Maas C, van der Vries E. Influenza-induced thrombocytopenia is dependent on the subtype and sialoglycan receptor and increases with virus pathogenicity. Blood Adv 2020; 4:2967-2978. [PMID: 32609845 PMCID: PMC7362372 DOI: 10.1182/bloodadvances.2020001640] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 05/18/2020] [Indexed: 12/18/2022] Open
Abstract
Thrombocytopenia is a common complication of influenza virus infection, and its severity predicts the clinical outcome of critically ill patients. The underlying cause(s) remain incompletely understood. In this study, in patients with an influenza A/H1N1 virus infection, viral load and platelet count correlated inversely during the acute infection phase. We confirmed this finding in a ferret model of influenza virus infection. In these animals, platelet count decreased with the degree of virus pathogenicity varying from 0% in animals infected with the influenza A/H3N2 virus, to 22% in those with the pandemic influenza A/H1N1 virus, up to 62% in animals with a highly pathogenic A/H5N1 virus infection. This thrombocytopenia is associated with virus-containing platelets that circulate in the blood. Uptake of influenza virus particles by platelets requires binding to sialoglycans and results in the removal of sialic acids by the virus neuraminidase, a trigger for hepatic clearance of platelets. We propose the clearance of influenza virus by platelets as a paradigm. These insights clarify the pathophysiology of influenza virus infection and show how severe respiratory infections, including COVID-19, may propagate thrombocytopenia and/or thromboembolic complications.
Collapse
MESH Headings
- Animals
- Blood Platelets/metabolism
- Blood Platelets/pathology
- Blood Platelets/virology
- Disease Models, Animal
- Ferrets
- Host-Pathogen Interactions
- Humans
- Influenza A Virus, H1N1 Subtype/pathogenicity
- Influenza A Virus, H1N1 Subtype/physiology
- Influenza A Virus, H3N2 Subtype/pathogenicity
- Influenza A Virus, H3N2 Subtype/physiology
- Influenza A Virus, H5N1 Subtype/pathogenicity
- Influenza A Virus, H5N1 Subtype/physiology
- Influenza A virus/pathogenicity
- Influenza A virus/physiology
- Influenza, Human/complications
- Influenza, Human/metabolism
- Influenza, Human/pathology
- Influenza, Human/virology
- N-Acetylneuraminic Acid/metabolism
- Orthomyxoviridae Infections/complications
- Orthomyxoviridae Infections/metabolism
- Orthomyxoviridae Infections/pathology
- Orthomyxoviridae Infections/virology
- Polysaccharides/metabolism
- Thrombocytopenia/etiology
- Thrombocytopenia/metabolism
- Thrombocytopenia/pathology
- Thrombocytopenia/virology
- Virus Internalization
Collapse
Affiliation(s)
- A J Gerard Jansen
- Department of Plasma Proteins, Sanquin, Amsterdam, The Netherlands
- Department of Hematology, Erasmus MC, Cancer Institute, Rotterdam, The Netherlands
| | - Thom Spaan
- Department of Clinical Chemistry and Hematology, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Infectious Diseases and Immunology, University of Utrecht, Utrecht, The Netherlands
| | - Hui Zhi Low
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine, Hannover, Germany
| | - Daniele Di Iorio
- Molecular Nanofabrication Group, MESA+ Institute for Nanotechnology, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands
| | | | - Malte Tieke
- Department of Infectious Diseases and Immunology, University of Utrecht, Utrecht, The Netherlands
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine, Hannover, Germany
| | - Arjan Barendrecht
- Department of Clinical Chemistry and Hematology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Kerstin Rohn
- Department of Pathology, University of Veterinary Medicine, Hannover, Germany
| | | | | | | | - Albert Osterhaus
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine, Hannover, Germany
| | - Thijs Kuiken
- Department of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Geert-Jan Boons
- Department of Pharmacy, University of Utrecht, Utrecht, The Netherlands; and
| | - Jurriaan Huskens
- Molecular Nanofabrication Group, MESA+ Institute for Nanotechnology, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands
| | - Marianne Boes
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Coen Maas
- Department of Clinical Chemistry and Hematology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Erhard van der Vries
- Department of Clinical Chemistry and Hematology, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Infectious Diseases and Immunology, University of Utrecht, Utrecht, The Netherlands
- Department of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
20
|
Eikelberg D, Lehmbecker A, Brogden G, Tongtako W, Hahn K, Habierski A, Hennermann JB, Naim HY, Felmy F, Baumgärtner W, Gerhauser I. Axonopathy and Reduction of Membrane Resistance: Key Features in a New Murine Model of Human G M1-Gangliosidosis. J Clin Med 2020; 9:jcm9041004. [PMID: 32252429 PMCID: PMC7230899 DOI: 10.3390/jcm9041004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 03/29/2020] [Accepted: 03/30/2020] [Indexed: 12/13/2022] Open
Abstract
GM1-gangliosidosis is caused by a reduced activity of β-galactosidase (Glb1), resulting in intralysosomal accumulations of GM1. The aim of this study was to reveal the pathogenic mechanisms of GM1-gangliosidosis in a new Glb1 knockout mouse model. Glb1−/− mice were analyzed clinically, histologically, immunohistochemically, electrophysiologically and biochemically. Morphological lesions in the central nervous system were already observed in two-month-old mice, whereas functional deficits, including ataxia and tremor, did not start before 3.5-months of age. This was most likely due to a reduced membrane resistance as a compensatory mechanism. Swollen neurons exhibited intralysosomal storage of lipids extending into axons and amyloid precursor protein positive spheroids. Additionally, axons showed a higher kinesin and lower dynein immunoreactivity compared to wildtype controls. Glb1−/− mice also demonstrated loss of phosphorylated neurofilament positive axons and a mild increase in non-phosphorylated neurofilament positive axons. Moreover, marked astrogliosis and microgliosis were found, but no demyelination. In addition to the main storage material GM1, GA1, sphingomyelin, phosphatidylcholine and phosphatidylserine were elevated in the brain. In summary, the current Glb1−/− mice exhibit a so far undescribed axonopathy and a reduced membrane resistance to compensate the functional effects of structural changes. They can be used for detailed examinations of axon–glial interactions and therapy trials of lysosomal storage diseases.
Collapse
Affiliation(s)
- Deborah Eikelberg
- Department of Pathology, University of Veterinary Medicine Hannover, D-30559 Hannover, Germany; (D.E.); (A.L.); (W.T.); (K.H.); (A.H.); (I.G.)
| | - Annika Lehmbecker
- Department of Pathology, University of Veterinary Medicine Hannover, D-30559 Hannover, Germany; (D.E.); (A.L.); (W.T.); (K.H.); (A.H.); (I.G.)
| | - Graham Brogden
- Department of Physiological Chemistry, University of Veterinary Medicine Hannover, D-30559 Hannover, Germany; (G.B.); (H.Y.N.)
| | - Witchaya Tongtako
- Department of Pathology, University of Veterinary Medicine Hannover, D-30559 Hannover, Germany; (D.E.); (A.L.); (W.T.); (K.H.); (A.H.); (I.G.)
- c/o Faculty of Veterinary Science, Prince of Sonkla University, 5 Karnjanavanich Rd., Hat Yai, Songkhla 90110, Thailand
| | - Kerstin Hahn
- Department of Pathology, University of Veterinary Medicine Hannover, D-30559 Hannover, Germany; (D.E.); (A.L.); (W.T.); (K.H.); (A.H.); (I.G.)
| | - Andre Habierski
- Department of Pathology, University of Veterinary Medicine Hannover, D-30559 Hannover, Germany; (D.E.); (A.L.); (W.T.); (K.H.); (A.H.); (I.G.)
| | - Julia B. Hennermann
- Villa Metabolica, University of Mainz, Langenbeckstraße 2, D-55131 Mainz, Germany;
| | - Hassan Y. Naim
- Department of Physiological Chemistry, University of Veterinary Medicine Hannover, D-30559 Hannover, Germany; (G.B.); (H.Y.N.)
| | - Felix Felmy
- Department for Physiology and Cell Biology, University of Veterinary Medicine Hannover, 30559 Hannover, Germany;
| | - Wolfgang Baumgärtner
- Department of Pathology, University of Veterinary Medicine Hannover, D-30559 Hannover, Germany; (D.E.); (A.L.); (W.T.); (K.H.); (A.H.); (I.G.)
- Correspondence:
| | - Ingo Gerhauser
- Department of Pathology, University of Veterinary Medicine Hannover, D-30559 Hannover, Germany; (D.E.); (A.L.); (W.T.); (K.H.); (A.H.); (I.G.)
| |
Collapse
|
21
|
Neurotrophic effects of G M1 ganglioside, NGF, and FGF2 on canine dorsal root ganglia neurons in vitro. Sci Rep 2020; 10:5380. [PMID: 32214122 PMCID: PMC7096396 DOI: 10.1038/s41598-020-61852-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 03/04/2020] [Indexed: 01/26/2023] Open
Abstract
Dogs share many chronic morbidities with humans and thus represent a powerful model for translational research. In comparison to rodents, the canine ganglioside metabolism more closely resembles the human one. Gangliosides are components of the cell plasma membrane playing a role in neuronal development, intercellular communication and cellular differentiation. The present in vitro study aimed to characterize structural and functional changes induced by GM1 ganglioside (GM1) in canine dorsal root ganglia (DRG) neurons and interactions of GM1 with nerve growth factor (NGF) and fibroblast growth factor (FGF2) using immunofluorescence for several cellular proteins including neurofilaments, synaptophysin, and cleaved caspase 3, transmission electron microscopy, and electrophysiology. GM1 supplementation resulted in increased neurite outgrowth and neuronal survival. This was also observed in DRG neurons challenged with hypoxia mimicking neurodegenerative conditions due to disruptions of energy homeostasis. Immunofluorescence indicated an impact of GM1 on neurofilament phosphorylation, axonal transport, and synaptogenesis. An increased number of multivesicular bodies in GM1 treated neurons suggested metabolic changes. Electrophysiological changes induced by GM1 indicated an increased neuronal excitability. Summarized, GM1 has neurotrophic and neuroprotective effects on canine DRG neurons and induces functional changes. However, further studies are needed to clarify the therapeutic value of gangliosides in neurodegenerative diseases.
Collapse
|
22
|
Jager SE, Pallesen LT, Richner M, Harley P, Hore Z, McMahon S, Denk F, Vaegter CB. Changes in the transcriptional fingerprint of satellite glial cells following peripheral nerve injury. Glia 2020; 68:1375-1395. [PMID: 32045043 DOI: 10.1002/glia.23785] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 01/09/2020] [Accepted: 01/10/2020] [Indexed: 01/13/2023]
Abstract
Satellite glial cells (SGCs) are homeostatic cells enveloping the somata of peripheral sensory and autonomic neurons. A wide variety of neuronal stressors trigger activation of SGCs, contributing to, for example, neuropathic pain through modulation of neuronal activity. However, compared to neurons and other glial cells of the nervous system, SGCs have received modest scientific attention and very little is known about SGC biology, possibly due to the experimental challenges associated with studying them in vivo and in vitro. Utilizing a recently developed method to obtain SGC RNA from dorsal root ganglia (DRG), we took a systematic approach to characterize the SGC transcriptional fingerprint by using next-generation sequencing and, for the first time, obtain an overview of the SGC injury response. Our RNA sequencing data are easily accessible in supporting information in Excel format. They reveal that SGCs are enriched in genes related to the immune system and cell-to-cell communication. Analysis of SGC transcriptional changes in a nerve injury-paradigm reveal a differential response at 3 days versus 14 days postinjury, suggesting dynamic modulation of SGC function over time. Significant downregulation of several genes linked to cholesterol synthesis was observed at both time points. In contrast, regulation of gene clusters linked to the immune system (MHC protein complex and leukocyte migration) was mainly observed after 14 days. Finally, we demonstrate that, after nerve injury, macrophages are in closer physical proximity to both small and large DRG neurons, and that previously reported injury-induced proliferation of SGCs may, in fact, be proliferating macrophages.
Collapse
Affiliation(s)
- Sara E Jager
- Danish Research Institute of Translational Neuroscience (DANDRITE), Nordic-EMBL Partnership for Molecular Medicine, Department of Biomedicine, Aarhus University, Aarhus C, Denmark.,Wolfson Centre for Age-Related Diseases, King's College London, Guy's Campus, London, UK
| | - Lone T Pallesen
- Danish Research Institute of Translational Neuroscience (DANDRITE), Nordic-EMBL Partnership for Molecular Medicine, Department of Biomedicine, Aarhus University, Aarhus C, Denmark
| | - Mette Richner
- Danish Research Institute of Translational Neuroscience (DANDRITE), Nordic-EMBL Partnership for Molecular Medicine, Department of Biomedicine, Aarhus University, Aarhus C, Denmark
| | - Peter Harley
- Wolfson Centre for Age-Related Diseases, King's College London, Guy's Campus, London, UK
| | - Zoe Hore
- Wolfson Centre for Age-Related Diseases, King's College London, Guy's Campus, London, UK
| | - Stephen McMahon
- Wolfson Centre for Age-Related Diseases, King's College London, Guy's Campus, London, UK
| | - Franziska Denk
- Wolfson Centre for Age-Related Diseases, King's College London, Guy's Campus, London, UK
| | - Christian B Vaegter
- Danish Research Institute of Translational Neuroscience (DANDRITE), Nordic-EMBL Partnership for Molecular Medicine, Department of Biomedicine, Aarhus University, Aarhus C, Denmark
| |
Collapse
|
23
|
Ganchingco JRC, Fukuyama T, Yoder JA, Bäumer W. Calcium imaging of primary canine sensory neurons: Small-diameter neurons responsive to pruritogens and algogens. Brain Behav 2019; 9:e01428. [PMID: 31571393 PMCID: PMC6908857 DOI: 10.1002/brb3.1428] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 08/28/2019] [Accepted: 08/31/2019] [Indexed: 01/19/2023] Open
Abstract
INTRODUCTION Rodent primary sensory neurons are commonly used for studying itch and pain neurophysiology, but translation from rodents to larger mammals and humans is not direct and requires further validation to make correlations. METHODS This study developed a primary canine sensory neuron culture from dorsal root ganglia (DRG) excised from cadaver dogs. Additionally, the canine DRG cell cultures developed were used for single-cell ratiometric calcium imaging, with the activation of neurons to the following pruritogenic and algogenic substances: histamine, chloroquine, canine protease-activated receptor 2 (PAR2) activating peptide (SLIGKT), compound 48/80, 5-hydroxytryptamine receptor agonist (5-HT), bovine adrenal medulla peptide (BAM8-22), substance P, allyl isothiocyanate (AITC), and capsaicin. RESULTS This study demonstrates a simple dissection and rapid processing of DRG collected from canine cadavers used to create viable primary sensory neuron cultures to measure responses to pruritogens and algogens. CONCLUSION Ratiometric calcium imaging demonstrated that small-diameter canine sensory neurons can be activated by multiple stimuli, and a single neuron can react to both a pruritogenic stimulation and an algogenic stimulation.
Collapse
Affiliation(s)
- Joy Rachel C Ganchingco
- Department of Molecular Biomedical Sciences, North Carolina State University College of Veterinary Medicine, Raleigh, NC, USA
| | - Tomoki Fukuyama
- Department of Molecular Biomedical Sciences, North Carolina State University College of Veterinary Medicine, Raleigh, NC, USA.,Laboratory of Veterinary Pharmacology, Azabu University, Kanagawa, Japan
| | - Jeffrey A Yoder
- Department of Molecular Biomedical Sciences, North Carolina State University College of Veterinary Medicine, Raleigh, NC, USA
| | - Wolfgang Bäumer
- Department of Molecular Biomedical Sciences, North Carolina State University College of Veterinary Medicine, Raleigh, NC, USA.,Institute of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Freie Universität Berlin, Berlin, Germany
| |
Collapse
|
24
|
Pertl K, Borchers M, Baumgärtner W, Wohlsein P. Expression of Neuroectodermal Markers in Atypical Fibromas in Two Dwarf Hamsters (Phodopus spp.). J Comp Pathol 2019; 172:53-57. [PMID: 31690416 DOI: 10.1016/j.jcpa.2019.09.001] [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: 07/19/2019] [Revised: 08/14/2019] [Accepted: 09/04/2019] [Indexed: 10/25/2022]
Abstract
A 2-year-old male Djungarian hamster (Phodopus sungorus) and a 1-year-old male Roborovski hamster (Phodopus roborovskii) were presented with expansile subcutaneous masses. The well-demarcated, firm, grey, multilobulated nodules displayed an homogeneous, white-grey cut surface. Histological examination revealed a neoplasm of variable cellularity consisting of spindle-shaped to polygonal 'ganglion cell-like' cells with abundant, amphophilic, vacuolated cytoplasm. Immunohistochemically, the tumour cells were labelled intensely for vimentin and nestin, moderately for neuron specific enolase and weakly for melan-A. The histological and immunohistochemical findings were suggestive of an atypical fibroma with evidence of a neuroectodermal phenotype.
Collapse
Affiliation(s)
- K Pertl
- Department of Pathology, University of Veterinary Medicine, Hannover, Germany
| | - M Borchers
- Clinic of Small Animals Herrenhausen, Hannover, Germany
| | - W Baumgärtner
- Department of Pathology, University of Veterinary Medicine, Hannover, Germany
| | - P Wohlsein
- Department of Pathology, University of Veterinary Medicine, Hannover, Germany.
| |
Collapse
|
25
|
Chiocchetti R, Galiazzo G, Tagliavia C, Stanzani A, Giancola F, Menchetti M, Militerno G, Bernardini C, Forni M, Mandrioli L. Cellular Distribution of Canonical and Putative Cannabinoid Receptors in Canine Cervical Dorsal Root Ganglia. Front Vet Sci 2019; 6:313. [PMID: 31608295 PMCID: PMC6761858 DOI: 10.3389/fvets.2019.00313] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 09/02/2019] [Indexed: 12/11/2022] Open
Abstract
Growing evidence indicates cannabinoid receptors as potential therapeutic targets for chronic pain. Consequently, there is an increasing interest in developing cannabinoid receptor agonists for treating human and veterinary pain. To better understand the actions of a drug, it is of paramount importance to know the cellular distribution of its specific receptor(s). The distribution of canonical and putative cannabinoid receptors in the peripheral and central nervous system of dogs is still in its infancy. In order to help fill this anatomical gap, the present ex vivo study has been designed to identify the cellular sites of cannabinoid and cannabinoid-related receptors in canine spinal ganglia. In particular, the cellular distribution of the cannabinoid receptors type 1 and 2 (CB1 and CB2) and putative cannabinoid receptors G protein-coupled receptor 55 (GPR55), nuclear peroxisome proliferator-activated receptor alpha (PPARα), and transient receptor potential vanilloid type 1 (TRPV1) have been immunohistochemically investigated in the C6–C8 cervical ganglia of dogs. About 50% of the neuronal population displayed weak to moderate CB1 receptor and TRPV1 immunoreactivity, while all of them were CB2-positive and nearly 40% also expressed GPR55 immunolabeling. Schwann cells, blood vessel smooth muscle cells, and pericyte-like cells all expressed CB2 receptor immunoreactivity, endothelial cell being also PPARα-positive. All the satellite glial cells (SGCs) displayed bright GPR55 receptor immunoreactivity. In half of the study dogs, SGCs were also PPARα-positive, and limited to older dogs displayed TRPV1 immunoreactivity. The present study may represent a morphological substrate to consider in order to develop therapeutic strategies against chronic pain.
Collapse
Affiliation(s)
- Roberto Chiocchetti
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | - Giorgia Galiazzo
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | - Claudio Tagliavia
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | - Agnese Stanzani
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | - Fiorella Giancola
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | - Marika Menchetti
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | - Gianfranco Militerno
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | - Chiara Bernardini
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | - Monica Forni
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | - Luciana Mandrioli
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
26
|
Schwarz S, Spitzbarth I, Baumgärtner W, Lehmbecker A. Cryopreservation of Canine Primary Dorsal Root Ganglion Neurons and Its Impact upon Susceptibility to Paramyxovirus Infection. Int J Mol Sci 2019; 20:ijms20051058. [PMID: 30823498 PMCID: PMC6429404 DOI: 10.3390/ijms20051058] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 02/22/2019] [Accepted: 02/25/2019] [Indexed: 12/26/2022] Open
Abstract
Canine dorsal root ganglion (DRG) neurons, isolated post mortem from adult dogs, could provide a promising tool to study neuropathogenesis of neurotropic virus infections with a non-rodent host spectrum. However, access to canine DRG is limited due to lack of donor tissue and the cryopreservation of DRG neurons would greatly facilitate experiments. The present study aimed (i) to establish canine DRG neurons as an in vitro model for canine distemper virus (CDV) infection; and (ii) to determine whether DRG neurons are cryopreservable and remain infectable with CDV. Neurons were characterized morphologically and phenotypically by light microscopy, immunofluorescence, and functionally, by studying their neurite outgrowth and infectability with CDV. Cryopreserved canine DRG neurons remained in culture for at least 12 days. Furthermore, both non-cryopreserved and cryopreserved DRG neurons were susceptible to infection with two different strains of CDV, albeit only one of the two strains (CDV R252) provided sufficient absolute numbers of infected neurons. However, cryopreserved DRG neurons showed reduced cell yield, neurite outgrowth, neurite branching, and soma size and reduced susceptibility to CDV infection. In conclusion, canine primary DRG neurons represent a suitable tool for investigations upon the pathogenesis of neuronal CDV infection. Moreover, despite certain limitations, cryopreserved canine DRG neurons generally provide a useful and practicable alternative to address questions regarding virus tropism and neuropathogenesis.
Collapse
Affiliation(s)
- Sarah Schwarz
- Department of Pathology, University of Veterinary Medicine, 30559 Hannover, Germany.
- Center for Systems Neuroscience, 30559 Hannover, Germany.
| | - Ingo Spitzbarth
- Department of Pathology, University of Veterinary Medicine, 30559 Hannover, Germany.
- Center for Systems Neuroscience, 30559 Hannover, Germany.
| | - Wolfgang Baumgärtner
- Department of Pathology, University of Veterinary Medicine, 30559 Hannover, Germany.
- Center for Systems Neuroscience, 30559 Hannover, Germany.
| | - Annika Lehmbecker
- Department of Pathology, University of Veterinary Medicine, 30559 Hannover, Germany.
- Center for Systems Neuroscience, 30559 Hannover, Germany.
| |
Collapse
|
27
|
Wang XB, Ma W, Luo T, Yang JW, Wang XP, Dai YF, Guo JH, Li LY. A novel primary culture method for high-purity satellite glial cells derived from rat dorsal root ganglion. Neural Regen Res 2019; 14:339-345. [PMID: 30531018 PMCID: PMC6301172 DOI: 10.4103/1673-5374.244797] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Satellite glial cells surround neurons within dorsal root ganglia. Previous studies have focused on single-cell suspensions of cultured neurons derived from rat dorsal root ganglia. At present, the primary culture method for satellite glial cells derived from rat dorsal root ganglia requires no digestion skill. Hence, the aim of the present study was to establish a novel primary culture method for satellite glial cells derived from dorsal root ganglia. Neonatal rat spine was collected and an incision made to expose the transverse protrusion and remove dorsal root ganglia. Dorsal root ganglia were freed from nerve fibers, connective tissue, and capsule membranes, then rinsed and transferred to 6-well plates, and cultured in a humidified 5% CO2 incubator at 37°C. After 3 days in culture, some cells had migrated from dorsal root ganglia. After subculture, cells were identified by immunofluorescence labeling for three satellite glial cell-specific markers: glutamine synthetase, glial fibrillary acidic protein, and S100β. Cultured cells expressed glutamine synthetase, glial fibrillary acidic protein, and S100β, suggesting they are satellite glial cells with a purity of > 95%. Thus, we have successfully established a novel primary culture method for obtaining high-purity satellite glial cells from rat dorsal root ganglia without digestion.
Collapse
Affiliation(s)
- Xian-Bin Wang
- Institute of Neuroscience, Kunming Medical University, Kunming, Yunnan Province, China
| | - Wei Ma
- Institute of Neuroscience, Kunming Medical University, Kunming, Yunnan Province, China
| | - Tao Luo
- Institute of Neuroscience, Kunming Medical University, Kunming, Yunnan Province; Medical Faculty, Panzhihua University, Panzhihua, Sichuan Province, China
| | - Jin-Wei Yang
- Institute of Neuroscience, Kunming Medical University; Second Department of General Surgery, First People's Hospital of Yunnan Province, Kunming, Yunnan Province, China
| | - Xiang-Peng Wang
- Institute of Neuroscience, Kunming Medical University, Kunming, Yunnan Province, China
| | - Yun-Fei Dai
- Institute of Neuroscience, Kunming Medical University, Kunming, Yunnan Province, China
| | - Jian-Hui Guo
- Second Department of General Surgery, First People's Hospital of Yunnan Province, Kunming, Yunnan Province, China
| | - Li-Yan Li
- Institute of Neuroscience, Kunming Medical University, Kunming, Yunnan Province, China
| |
Collapse
|
28
|
Sun W, Ma M, Yu H, Yu H. Inhibition of lncRNA X inactivate-specific transcript ameliorates inflammatory pain by suppressing satellite glial cell activation and inflammation by acting as a sponge of miR-146a to inhibit Na v 1.7. J Cell Biochem 2018; 119:9888-9898. [PMID: 30129228 DOI: 10.1002/jcb.27310] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 06/25/2018] [Indexed: 12/28/2022]
Abstract
Long noncoding RNAs (lncRNA) has been validated to participate in nociception in inflammatory pain, presenting as a potential target against anesthesia. Previous research work confirmed the correlation between lncRNA X inactivate-specific transcript (XIST) and inflammation. However, its role in inflammatory pain is undefined. In animal pain models, voltage-gated sodium channels (VGSCs) reportedly participate in neural excitation. In this study, we observed the high expression of XIST and VGSC 1.7 (Nav 1.7) in the dorsal root ganglion (DRG) of the complete Freund's adjuvant (CFA)-induced rat inflammatory pain model. Furthermore, XIST inhibition alleviated pain behavior and the activation of DRG satellite glial cells by suppressing glial fibrillary acidic protein (GFAP) expression, as well as inflammatory cytokine levels of interleukin-6 and tumor necrosis factor-α. XIST downregulation increased the mechanical pain threshold in an inflammatory pain model. Moreover, the expression of miR-146a was decreased in CFA rats. In vitro, XIST acted as a sponge of miR-146a, which targeted Nav 1.7 via bioinformatic prediction, luciferase reporter, and pull-down assay. More importantly, activation of the Nav 1.7 pathway or miR-146 depression both reversed XIST knockdown-inhibited satellite glial cell activation and inflammatory pain in CFA rats. These results suggest that cessation of XIST may ameliorate inflammatory pain by acting as a sponge of miR-146a to inhibit Nav1.7, implying a promising strategy against inflammatory pain.
Collapse
Affiliation(s)
- Wenbo Sun
- Department of Anesthesia, Cangzhou Central Hospital, CangZhou, China
| | - Meina Ma
- Department of Anesthesia, Cangzhou Central Hospital, CangZhou, China
| | - Hongmei Yu
- Department of Anesthesia, Cangzhou Central Hospital, CangZhou, China
| | - Hong Yu
- Department of Anesthesia, Cangzhou Central Hospital, CangZhou, China
| |
Collapse
|