1
|
Liu Y, Xu T, Yu Z, Xu B. Neurophysiological Basis of Electroacupuncture Stimulation in the Treatment of Cardiovascular-Related Diseases: Vagal Interoceptive Loops. Brain Behav 2024; 14:e70076. [PMID: 39344397 PMCID: PMC11440030 DOI: 10.1002/brb3.70076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 08/28/2024] [Accepted: 09/07/2024] [Indexed: 10/01/2024] Open
Abstract
PURPOSE The vagal sensory nerve (VSN) is an essential interoceptive pathway that is connected to every level of the body. Its intricate genetic coding provides sustenance for physiological processes, including controlling blood pressure and respiration. Electroacupuncture (EA) is a proven surface stimulation therapy that can regulate vagal nerve activity, which can effectively prevent cardiovascular diseases. A growing number of studies have concentrated on the mapping of VSN codes, but little is known, and the physiological background of how EA influences interoceptive has not been fully explored. METHOD Here, we incorporate the hypothesized interaction among EA targets, VSNs, and the heart. This offers suggestions for using a versatile and focused EA strategy to modify vagal interoceptive awareness to enhance cardiovascular conditions. We first clarified the major role of vagal nerve in the control of cardiac activity. Additionally, we clarified the multidimensional coding pattern in the VSNs, revealing that the targeted control of multimodal interoceptive is the functional basis of the synchronization of cardiovascular system. FINDING We propose a strategy in which EA of the VSNs is employed to activate the interoceptive loop and reduce the risk of cardiovascular disease.
Collapse
Affiliation(s)
- Yun Liu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of EducationNanjing University of Chinese MedicineNanjingChina
| | - Tiancheng Xu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of EducationNanjing University of Chinese MedicineNanjingChina
| | - Zhi Yu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of EducationNanjing University of Chinese MedicineNanjingChina
| | - Bin Xu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of EducationNanjing University of Chinese MedicineNanjingChina
| |
Collapse
|
2
|
Qiao LY. Satellite Glial Cells Bridge Sensory Neuron Crosstalk in Visceral Pain and Cross-Organ Sensitization. J Pharmacol Exp Ther 2024; 390:213-221. [PMID: 38777604 PMCID: PMC11264254 DOI: 10.1124/jpet.123.002061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 05/07/2024] [Accepted: 05/08/2024] [Indexed: 05/25/2024] Open
Abstract
Following colonic inflammation, the uninjured bladder afferent neurons are also activated. The mechanisms and pathways underlying this sensory neuron cross-activation (from injured neurons to uninjured neurons) are not fully understood. Colonic and bladder afferent neurons reside in the same spinal segments and are separated by satellite glial cells (SGCs) and extracellular matrix in dorsal root ganglia (DRG). SGCs communicate with sensory neurons in a bidirectional fashion. This review summarizes the differentially regulated genes/proteins in the injured and uninjured DRG neurons and explores the role of SGCs in regulation of sensory neuron crosstalk in visceral cross-organ sensitization. The review also highlights the paracrine pathways in mediating neuron-SGC and SGC-neuron coupling with an emphasis on the neurotrophins and purinergic systems. Finally, I discuss the results from recent RNAseq profiling of SGCs to reveal useful molecular markers for characterization, functional study, and therapeutic targets of SGCs. SIGNIFICANCE STATEMENT: Satellite glial cells (SGCs) are the largest glial subtypes in sensory ganglia and play a critical role in mediating sensory neuron crosstalk, an underlying mechanism in colon-bladder cross-sensitization. Identification of novel and unique molecular markers of SGCs can advance the discovery of therapeutic targets in treatment of chronic pain including visceral pain comorbidity.
Collapse
Affiliation(s)
- Liya Y Qiao
- Department of Physiology and Biophysics, Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| |
Collapse
|
3
|
Areti A, Komirishetty P, Zochodne DW. Collaborative Roles for RAC1, ERM Proteins and PTEN During Adult Sensory Axon Regeneration. Mol Neurobiol 2024:10.1007/s12035-024-04273-7. [PMID: 38904854 DOI: 10.1007/s12035-024-04273-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 05/29/2024] [Indexed: 06/22/2024]
Abstract
The role of local of growth cone (GC) manipulation in adult regenerative systems is largely unexplored despite substantial translational importance. Here we investigated collaboration among Rac1 GTPase, its partnering ERM proteins and PTEN in adult sensory neurons and adult nerve regeneration. We confirmed expression of both Rac1 and ERM in adults and noted substantial impacts on neurite outgrowth in naïve and pre-injured adult sensory neurons. PTEN inhibition added to this outgrowth. Rac1 activation acted directly on adult GCs facilitating both attractive turning and advancement. In vivo regeneration indices including electrophysiological recovery, return of sensation, walking, repopulation of myelinated axons and reinnervation of the target epidermis indicated benefits of local Rac1 activation. These indices suggested maximal GC activation whereas local PTEN inhibition offered only limited added improvement. Our findings provide support for the concept of manipulating adult GCs, by emphasizing local Rac1 activation in directing therapy for nerve repair.
Collapse
Affiliation(s)
- Aparna Areti
- Division of Neurology, Department of Medicine and the Neuroscience and Mental Health Institute, University of Alberta, 7-132 Clinical Sciences Building 11350-83 Ave, T6G 2G3, Edmonton, AB, Canada
| | - Prashanth Komirishetty
- Division of Neurology, Department of Medicine and the Neuroscience and Mental Health Institute, University of Alberta, 7-132 Clinical Sciences Building 11350-83 Ave, T6G 2G3, Edmonton, AB, Canada
| | - Douglas W Zochodne
- Division of Neurology, Department of Medicine and the Neuroscience and Mental Health Institute, University of Alberta, 7-132 Clinical Sciences Building 11350-83 Ave, T6G 2G3, Edmonton, AB, Canada.
| |
Collapse
|
4
|
Lian Y, Wu C, Liu L, Li X. Prediction of cell-cell communication patterns of dorsal root ganglion cells: single-cell RNA sequencing data analysis. Neural Regen Res 2024; 19:1367-1374. [PMID: 37905887 PMCID: PMC11467928 DOI: 10.4103/1673-5374.384067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 07/05/2023] [Accepted: 07/28/2023] [Indexed: 11/02/2023] Open
Abstract
Dorsal root ganglion neurons transmit peripheral somatic information to the central nervous system, and dorsal root ganglion neuron excitability affects pain perception. Dorsal root ganglion stimulation is a new approach for managing pain sensation. Knowledge of the cell-cell communication among dorsal root ganglion cells may help in the development of new pain and itch management strategies. Here, we used the single-cell RNA-sequencing (scRNA-seq) database to investigate intercellular communication networks among dorsal root ganglion cells. We collected scRNA-seq data from six samples from three studies, yielding data on a total of 17,766 cells. Based on genetic profiles, we identified satellite glial cells, Schwann cells, neurons, vascular endothelial cells, immune cells, fibroblasts, and vascular smooth muscle cells. Further analysis revealed that eight types of dorsal root ganglion neurons mediated proprioceptive, itch, touch, mechanical, heat, and cold sensations. Moreover, we predicted several distinct forms of intercellular communication among dorsal root ganglion cells, including cell-cell contact, secreted signals, extracellular matrix, and neurotransmitter-mediated signals. The data mining predicted that Mrgpra3 -positive neurons robustly express the genes encoding the adenosine Adora2b (A2B) receptor and glial cell line-derived neurotrophic factor family receptor alpha 1 (GFRα-1). Our immunohistochemistry results confirmed the coexpression of the A2B receptor and GFRα-1. Intrathecal injection of the A2B receptor antagonist PSB-603 effectively prevented histamine-induced scratching behaviour in a dose-dependent manner. Our results demonstrate the involvement of the A2B receptor in the modulation of itch sensation. Furthermore, our findings provide insight into dorsal root ganglion cell-cell communication patterns and mechanisms. Our results should contribute to the development of new strategies for the regulation of dorsal root ganglion excitability.
Collapse
Affiliation(s)
- Yanna Lian
- National Health Commission and Chinese Academy of Medical Sciences Key Laboratory of Medical Neurobiology, Ministry of Education Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
- International Institutes of Medicine, the Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, Zhejiang Province, China
| | - Cheng Wu
- National Health Commission and Chinese Academy of Medical Sciences Key Laboratory of Medical Neurobiology, Ministry of Education Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Haining, Zhejiang Province, China
- Biomedical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK
| | - Li Liu
- Core Facilities of the School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Xiangyao Li
- National Health Commission and Chinese Academy of Medical Sciences Key Laboratory of Medical Neurobiology, Ministry of Education Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
- International Institutes of Medicine, the Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, Zhejiang Province, China
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Haining, Zhejiang Province, China
- Biomedical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK
| |
Collapse
|
5
|
Du J, Sudlow LC, Luzhansky ID, Berezin MY. DRG Explant Model: Elucidating Mechanisms of Oxaliplatin-Induced Peripheral Neuropathy and Identifying Potential Therapeutic Targets. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.05.560580. [PMID: 37873159 PMCID: PMC10592953 DOI: 10.1101/2023.10.05.560580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Oxaliplatin triggered chemotherapy induced peripheral neuropathy (CIPN) is a common and debilitating side effect of cancer treatment which limits the efficacy of chemotherapy and negatively impacts patients quality of life dramatically. For better understanding the mechanisms of CIPN and screen for potential therapeutic targets, it is critical to have reliable in vitro assays that effectively mirror the neuropathy in vivo . In this study, we established a dorsal root ganglia (DRG) explant model. This model displayed dose-dependent inhibition of neurite outgrowth in response to oxaliplatin, while oxalic acid exhibited no significant impact on the regrowth of DRG. The robustness of this assay was further demonstrated by the inhibition of OCT2 transporter, which facilitates oxaliplatin accumulation in neurons, fully restoring the neurite regrowth capacity. Using this model, we revealed that oxaliplatin triggered a substantial increase of oxidative stress in DRG. Notably, inhibition of TXNIP with verapamil significantly reduced oxidative stress level. Our results demonstrated the use of DRG explants as an efficient model to study the mechanisms of CIPN and screen for potential treatments.
Collapse
|
6
|
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
|
7
|
Starinets A, Tyrtyshnaia A, Manzhulo I. Anti-Inflammatory Activity of Synaptamide in the Peripheral Nervous System in a Model of Sciatic Nerve Injury. Int J Mol Sci 2023; 24:6273. [PMID: 37047247 PMCID: PMC10093792 DOI: 10.3390/ijms24076273] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/22/2023] [Accepted: 03/25/2023] [Indexed: 03/29/2023] Open
Abstract
N-docosahexaenoylethanolamine (DHEA), or synaptamide, is an endogenous metabolite of docosahexaenoic acid (DHA) that exhibits synaptogenic and neurogenic effects. In our previous studies, synaptamide administration inhibited the neuropathic pain-like behavior and reduced inflammation in the central nervous system following sciatic nerve injury. In the present study, we examine the effect of synaptamide on the peripheral nervous system in a neuropathic pain condition. The dynamics of ionized calcium-binding adapter molecule 1 (iba-1), CD68, CD163, myelin basic protein, and the production of interleukin 1β and 6 within the sciatic nerve, as well as the neuro-glial index and the activity of iba-1, CD163, glial fibrillary acidic protein (GFAP), neuronal NO synthase (nNOS), substance P (SP), activating transcription factor 3 (ATF3) in the dorsal root ganglia (DRG), are studied. According to our results, synaptamide treatment (4 mg/kg/day) (1) decreases the weight-bearing deficit after nerve trauma; (2) enhances the remyelination process in the sciatic nerve; (3) shows anti-inflammatory properties in the peripheral nervous system; (4) decreases the neuro-glial index and GFAP immunoreactivity in the DRG; (5) inhibits nNOS- and SP-ergic activity in the DRG, which might contribute to neuropathic pain attenuation. In general, the current study demonstrates the complex effect of synaptamide on nerve injury, which indicates its high potential for neuropathic pain management.
Collapse
Affiliation(s)
| | | | - Igor Manzhulo
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, 690041 Vladivostok, Russia; (A.S.); (A.T.)
| |
Collapse
|
8
|
Alcayaga J, Vera J, Reyna-Jeldes M, Covarrubias AA, Coddou C, Díaz-Jara E, Del Rio R, Retamal MA. Activation of Intra-nodose Ganglion P2X7 Receptors Elicit Increases in Neuronal Activity. Cell Mol Neurobiol 2023:10.1007/s10571-023-01318-8. [PMID: 36680690 DOI: 10.1007/s10571-023-01318-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 01/05/2023] [Indexed: 01/22/2023]
Abstract
Vagus nerve innervates several organs including the heart, stomach, and pancreas among others. Somas of sensory neurons that project through the vagal nerve are located in the nodose ganglion. The presence of purinergic receptors has been reported in neurons and satellite glial cells in several sensory ganglia. In the nodose ganglion, calcium depletion-induced increases in neuron activity can be partly reversed by P2X7 blockers applied directly into the ganglion. The later suggest a possible role of P2X7 receptors in the modulation of neuronal activity within this sensory ganglion. We aimed to characterize the response to P2X7 activation in nodose ganglion neurons under physiological conditions. Using an ex vivo preparation for electrophysiological recordings of the neural discharges of nodose ganglion neurons, we found that treatments with ATP induce transient neuronal activity increases. Also, we found a concentration-dependent increase in neural activity in response to Bz-ATP (ED50 = 0.62 mM, a selective P2X7 receptor agonist), with a clear desensitization pattern when applied every ~ 30 s. Electrophysiological recordings from isolated nodose ganglion neurons reveal no differences in the responses to Bz-ATP and ATP. Finally, we showed that the P2X7 receptor was expressed in the rat nodose ganglion, both in neurons and satellite glial cells. Additionally, a P2X7 receptor negative allosteric modulator decreased the duration of Bz-ATP-induced maximal responses without affecting their amplitude. Our results show the presence of functional P2X7 receptors under physiological conditions within the nodose ganglion of the rat, and suggest that ATP modulation of nodose ganglion activity may be in part mediated by the activation of P2X7 receptors.
Collapse
Affiliation(s)
- Julio Alcayaga
- Laboratorio de Fisiología Celular, Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Casilla 653, Santiago, Chile. .,Millennium Nucleus for the Study of Pain (MiNuSPain), Santiago, Chile.
| | - Jorge Vera
- Laboratorio de Fisiología Celular, Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Casilla 653, Santiago, Chile
| | - Mauricio Reyna-Jeldes
- Laboratorio de Señalización Purinérgica, Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica del Norte, Coquimbo, Chile.,Millennium Nucleus for the Study of Pain (MiNuSPain), Santiago, Chile
| | - Alejandra A Covarrubias
- Laboratorio de Señalización Purinérgica, Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica del Norte, Coquimbo, Chile.,Millennium Nucleus for the Study of Pain (MiNuSPain), Santiago, Chile
| | - Claudio Coddou
- Laboratorio de Señalización Purinérgica, Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica del Norte, Coquimbo, Chile.,Millennium Nucleus for the Study of Pain (MiNuSPain), Santiago, Chile
| | - Esteban Díaz-Jara
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Rodrigo Del Rio
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile.,Centro de Envejecimiento y Regeneración (CARE-UC), Pontificia Universidad Católica de Chile, Santiago, Chile.,Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile
| | - Mauricio A Retamal
- Universidad de Desarrollo, Programa de Comunicación Celular en Cáncer. Facultad de Medicina Clínica Alemana., Santiago, Chile. .,Universidad del Desarrollo. , Centro de Fisiología Celular e Integrativa, Clínica Alemana Facultad de Medicina., Santiago, Chile.
| |
Collapse
|
9
|
Avraham O, Chamessian A, Feng R, Yang L, Halevi AE, Moore AM, Gereau RW, Cavalli V. Profiling the molecular signature of satellite glial cells at the single cell level reveals high similarities between rodents and humans. Pain 2022; 163:2348-2364. [PMID: 35503034 PMCID: PMC9522926 DOI: 10.1097/j.pain.0000000000002628] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 02/23/2022] [Indexed: 11/25/2022]
Abstract
ABSTRACT Peripheral sensory neurons located in dorsal root ganglia relay sensory information from the peripheral tissue to the brain. Satellite glial cells (SGCs) are unique glial cells that form an envelope completely surrounding each sensory neuron soma. This organization allows for close bidirectional communication between the neuron and its surrounding glial coat. Morphological and molecular changes in SGC have been observed in multiple pathological conditions such as inflammation, chemotherapy-induced neuropathy, viral infection, and nerve injuries. There is evidence that changes in SGC contribute to chronic pain by augmenting the neuronal activity in various rodent pain models. Satellite glial cells also play a critical role in axon regeneration. Whether findings made in rodent model systems are relevant to human physiology have not been investigated. Here, we present a detailed characterization of the transcriptional profile of SGC in mice, rats, and humans at the single cell level. Our findings suggest that key features of SGC in rodent models are conserved in humans. Our study provides the potential to leverage rodent SGC properties and identify potential targets in humans for the treatment of nerve injuries and alleviation of painful conditions.
Collapse
Affiliation(s)
- Oshri Avraham
- Department of Neuroscience, Washington University School of Medicine, St Louis 63110, Missouri, USA
| | - Alexander Chamessian
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St Louis 63110, Missouri, USA
- Department of Neurology, Washington University School of Medicine, St Louis 63110, Missouri, USA
| | - Rui Feng
- Department of Neuroscience, Washington University School of Medicine, St Louis 63110, Missouri, USA
| | - Lite Yang
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St Louis 63110, Missouri, USA
- Neuroscience Program, Washington University School of Medicine, St Louis 63110, Missouri, USA
| | - Alexandra E. Halevi
- Department of Plastic and Reconstructive Surgery, Washington University School of Medicine, St Louis 63110, Missouri, USA
| | - Amy M. Moore
- Department of Plastic and Reconstructive Surgery, The Ohio State University, Columbus Ohio, USA
| | - Robert W. Gereau
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St Louis 63110, Missouri, USA
| | - Valeria Cavalli
- Department of Neuroscience, Washington University School of Medicine, St Louis 63110, Missouri, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, Missouri 63110, USA
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| |
Collapse
|
10
|
Survival of compromised adult sensory neurons involves macrovesicular formation. Cell Death Dis 2022; 8:462. [PMID: 36424403 PMCID: PMC9691713 DOI: 10.1038/s41420-022-01247-3] [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: 07/08/2021] [Revised: 10/26/2022] [Accepted: 11/02/2022] [Indexed: 11/25/2022]
Abstract
Adult neurons are recognized as post-mitotically arrested cells with limited regenerative potential. Given these restraints, it is perplexing how neurons sustain routine physiological and occasional reparative stress without compromising their density and integrity. We observed that specific insults or physiological alterations drive adult sensory neurons to attempt cell cycle entry. In this context, we demonstrate that at least a small population of sensory neurons modify their cytoskeleton as a survival mechanism in settings of growth arrest and associated stress. Most notably, among their apparent survival modifications is included a unique, and uncharacterized form of macrovesicle shedding and a subsequent neuron size adjustment. Using time-lapse imaging, we demonstrate macrovesicle shedding in some neurons subjected to growth restraint, but not associated with apoptosis. In axotomized neurons in vivo, cell cycle entry was rare to absent and macrovesicles were not observed, but we nonetheless identified changes in mRNA associated with autophagy. In vivo, neighbouring macrophages may have a role in modifying the neuron cytoskeleton after axotomy. Overall, the findings identify previously unrecognized structural adaptations in adult sensory neurons that may provide resilience to diverse insults.
Collapse
|
11
|
Gong Y, Laheji F, Berenson A, Qian A, Park SO, Kok R, Selig M, Hahn R, Sadjadi R, Kemp S, Eichler F. Peroxisome Metabolism Contributes to PIEZO2-Mediated Mechanical Allodynia. Cells 2022; 11:1842. [PMID: 35681537 PMCID: PMC9180358 DOI: 10.3390/cells11111842] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Revised: 05/21/2022] [Accepted: 06/01/2022] [Indexed: 11/17/2022] Open
Abstract
Mutations in the peroxisomal half-transporter ABCD1 cause X-linked adrenoleukodystrophy, resulting in elevated very long-chain fatty acids (VLCFA), progressive neurodegeneration and an associated pain syndrome that is poorly understood. In the nervous system of mice, we found ABCD1 expression to be highest in dorsal root ganglia (DRG), with satellite glial cells (SGCs) displaying higher expression than neurons. We subsequently examined sensory behavior and DRG pathophysiology in mice deficient in ABCD1 compared to wild-type mice. Beginning at 8 months of age, Abcd1-/y mice developed persistent mechanical allodynia. DRG had a greater number of IB4-positive nociceptive neurons expressing PIEZO2, the mechanosensitive ion channel. Blocking PIEZO2 partially rescued the mechanical allodynia. Beyond affecting neurons, ABCD1 deficiency impacted SGCs, as demonstrated by high levels of VLCFA, increased glial fibrillary acidic protein (GFAP), as well as genes disrupting neuron-SGC connectivity. These findings suggest that lack of the peroxisomal half-transporter ABCD1 leads to PIEZO2-mediated mechanical allodynia as well as SGC dysfunction. Given the known supportive role of SGCs to neurons, this elucidates a novel mechanism underlying pain in X-linked adrenoleukodystrophy.
Collapse
Affiliation(s)
- Yi Gong
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (Y.G.); (F.L.); (A.B.); (A.Q.); (S.-O.P.); (M.S.); (R.H.); (R.S.)
| | - Fiza Laheji
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (Y.G.); (F.L.); (A.B.); (A.Q.); (S.-O.P.); (M.S.); (R.H.); (R.S.)
| | - Anna Berenson
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (Y.G.); (F.L.); (A.B.); (A.Q.); (S.-O.P.); (M.S.); (R.H.); (R.S.)
| | - April Qian
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (Y.G.); (F.L.); (A.B.); (A.Q.); (S.-O.P.); (M.S.); (R.H.); (R.S.)
| | - Sang-O Park
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (Y.G.); (F.L.); (A.B.); (A.Q.); (S.-O.P.); (M.S.); (R.H.); (R.S.)
| | - Rene Kok
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam University Medical Center, Amsterdam Gastroenterology Endocrinology Metabolism, University of Amsterdam, 1105 Amsterdam, The Netherlands; (R.K.); (S.K.)
| | - Martin Selig
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (Y.G.); (F.L.); (A.B.); (A.Q.); (S.-O.P.); (M.S.); (R.H.); (R.S.)
| | - Ryan Hahn
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (Y.G.); (F.L.); (A.B.); (A.Q.); (S.-O.P.); (M.S.); (R.H.); (R.S.)
| | - Reza Sadjadi
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (Y.G.); (F.L.); (A.B.); (A.Q.); (S.-O.P.); (M.S.); (R.H.); (R.S.)
| | - Stephan Kemp
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam University Medical Center, Amsterdam Gastroenterology Endocrinology Metabolism, University of Amsterdam, 1105 Amsterdam, The Netherlands; (R.K.); (S.K.)
- Department of Pediatric Neurology, Emma Children’s Hospital, Amsterdam University Medical Center, Amsterdam Neuroscience, University of Amsterdam, 1105 Amsterdam, The Netherlands
| | - Florian Eichler
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (Y.G.); (F.L.); (A.B.); (A.Q.); (S.-O.P.); (M.S.); (R.H.); (R.S.)
| |
Collapse
|
12
|
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
|
13
|
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
|
14
|
Avraham O, Feng R, Ewan EE, Rustenhoven J, Zhao G, Cavalli V. Profiling sensory neuron microenvironment after peripheral and central axon injury reveals key pathways for neural repair. eLife 2021; 10:e68457. [PMID: 34586065 PMCID: PMC8480984 DOI: 10.7554/elife.68457] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 09/12/2021] [Indexed: 12/19/2022] Open
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.
Collapse
Affiliation(s)
- Oshri Avraham
- Department of Neuroscience, Washington University School of MedicineSaint LouisUnited States
| | - Rui Feng
- Department of Neuroscience, Washington University School of MedicineSaint LouisUnited States
| | - Eric Edward Ewan
- Department of Neuroscience, Washington University School of MedicineSaint LouisUnited States
| | - Justin Rustenhoven
- Department of Pathology and Immunology, Washington University School of MedicineSt LouisUnited States
- Center for Brain Immunology and Glia (BIG), Washington University School of MedicineSt LouisUnited States
| | - Guoyan Zhao
- Department of Neuroscience, Washington University School of MedicineSaint LouisUnited States
- Department of Pathology and Immunology, Washington University School of MedicineSt LouisUnited States
| | - Valeria Cavalli
- Department of Neuroscience, Washington University School of MedicineSaint LouisUnited States
- Center of Regenerative Medicine, Washington University School of MedicineSt. LouisUnited States
- Hope Center for Neurological Disorders, Washington University School of MedicineSt. LouisUnited States
| |
Collapse
|
15
|
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
|
16
|
Inactivation of vimentin in satellite glial cells affects dorsal root ganglion intermediate filament expression and neuronal axon growth in vitro. Mol Cell Neurosci 2021; 115:103659. [PMID: 34400333 DOI: 10.1016/j.mcn.2021.103659] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 08/05/2021] [Accepted: 08/10/2021] [Indexed: 11/20/2022] Open
Abstract
Peripheral nerve trauma and regeneration are complex events, and little is known concerning how occurrences in the distal stump affect the cell body's response to injury. Intermediate filament (IF) proteins underpin cellular architecture and take part in nerve cell proliferation, differentiation and axon regeneration, but their role in these processes is not yet fully understood. The present study aimed to investigate the regulation and interrelationship of major neural IFs in adult dorsal root ganglion (DRG) neurons and satellite glial cells (SGCs) following sciatic nerve injury. We demonstrated that the expression of neural IFs in DRG neurons and SGCs after axotomy depends on vimentin activity. In intact DRGs, synemin M and peripherin proteins are detected in small neurons while neurofilament L (NFL) and synemin L characterize large neurons. Both neuronal populations are surrounded by vimentin positive- and glial fibrillary acidic protein (GFAP)-negative SGCs. In response to axotomy, synemin M and peripherin were upregulated in large wild-type DRG neurons and, to a lesser extent, in vim-/- and synm-/- DRG neurons, suggesting the role for these IFs in axon regeneration. However, an increase in the number of NFL-positive small neurons was observed in vim-/- mice, accompanied by a decrease of peripherin-positive small neurons. These findings suggest that vimentin is required for injury-induced neuronal IF remodeling. We further show that vimentin is also indispensable for nerve injury-induced GFAP upregulation in perineuronal SGCs and that inactivation of vimentin and synemin appears to accelerate the rate of DRG neurite regeneration at early stages in vitro.
Collapse
|
17
|
Lee N, Lee MY, Lee J, Kwon SH, Seung H, Lim J, Kim S. Hepatocyte growth factor induces pErk and pSTAT3 (Ser 727) to promote mitochondrial activity and neurite outgrowth in primary dorsal root ganglion cultures. Neuroreport 2021; 32:525-530. [PMID: 33788814 DOI: 10.1097/wnr.0000000000001622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Hepatocyte growth factor (HGF) promotes the neurite outgrowth of sensory neurons in developmental stages, but its role in injured peripheral nerves in adult mice remains largely been unexplored. In this study, we investigated the role of HGF in the regeneration of injured peripheral nerves using cultured dorsal root ganglions (DRGs). When cells were treated with HGF protein, the length of the neurite was increased 1.4-fold compared to the untreated control group. HGF greatly increased the level of phosphorylated STAT3 at serine 727 [pSTAT3 (Ser 727)], thereby translocating the protein to the mitochondria. HGF treatment increased the activity of mitochondrial complex I. When DRGs were cultured in the presence of U0126, a pharmacological inhibitor of Erk, the HGF-mediated increase in neurite outgrowth and the level of pSTAT3 (Ser 727) were both suppressed. Taken together, these results suggest that the HGF/c-met pathway might promote neurite outgrowth by controlling mitochondrial activity through the HGF/Erk/STAT3 axis.
Collapse
Affiliation(s)
- Nayeon Lee
- Department of Biological Sciences, Seoul National University.,Division of Gene Therapy, Helixmith Co Ltd., Seoul, Korea
| | - Mi-Young Lee
- Division of Gene Therapy, Helixmith Co Ltd., Seoul, Korea
| | - Junghun Lee
- Division of Gene Therapy, Helixmith Co Ltd., Seoul, Korea
| | - Sang Ho Kwon
- Division of Gene Therapy, Helixmith Co Ltd., Seoul, Korea
| | - Hana Seung
- Division of Gene Therapy, Helixmith Co Ltd., Seoul, Korea
| | - Jaegook Lim
- Division of Gene Therapy, Helixmith Co Ltd., Seoul, Korea
| | - Sunyoung Kim
- Division of Gene Therapy, Helixmith Co Ltd., Seoul, Korea
| |
Collapse
|
18
|
Regulation of Mitochondrial Function by Epac2 Contributes to Acute Inflammatory Hyperalgesia. J Neurosci 2021; 41:2883-2898. [PMID: 33593853 DOI: 10.1523/jneurosci.2368-20.2021] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 01/14/2021] [Accepted: 02/09/2021] [Indexed: 12/12/2022] Open
Abstract
Gαs-coupled receptors signaling through cAMP provide a key mechanism for the sensitization of nociceptive sensory neurons, and the cAMP effector Epac has been implicated in the transition from acute to chronic pain. Epac exerts its effects through Rap1 and protein kinase C (PKC). To identify targets of Epac-PKC signaling in sensory neurons of the mouse dorsal root ganglion (DRG), we profiled PKC substrate proteins phosphorylated in response to the activation of Epac with the proinflammatory prostaglandin E2 (PGE2). A prominent Epac-dependent phospho-protein band induced by PGE2 was identified by mass spectrometry as the mitochondrial enzyme pyruvate dehydrogenase (Pdha1). In dissociated DRG from both males and females, the recruitment of Pdha1 to phospho-protein fractions was rapidly induced by PGE2 and prevented by selective inhibition of Epac2. Epac activation increased mitochondrial respiration, consistent with an increase in Pdha1 function mediated by Epac2. Hindpaw injection of PGE2 induced heat hyperalgesia in males and females, but Pdha1 phosphorylation occurred only in males. Hyperalgesia was attenuated in males but not in females by systemic inhibition of Epac2, and also by a mitochondrial membrane potential uncoupler, dinitrophenol, supporting a role for mitochondrial regulation in acute hyperalgesia. These findings identify a mechanism for the regulation of mitochondrial function by Epac2 that contributes to acute inflammatory hyperalgesia in male mice. Systemic administration of the cyclooxygenase 2 inhibitor celecoxib suppressed both PGE2-induced heat hyperalgesia and Pdha1 phosphorylation in DRG of males but not females, suggesting that prostaglandin synthesis within the DRG mediates the phosphorylation of Pdha1 in response to hindpaw insult.SIGNIFICANCE STATEMENT There has been extensive investigation of mitochondrial dysfunction as a causative factor in neuropathic pain disorders. In contrast, results reported here implicate enhanced mitochondrial function as a contributing factor in the development of acute inflammatory hyperalgesia. We describe a mechanism in which Epac2 activation by prostaglandin receptors leads to phosphorylation of pyruvate dehydrogenase and an increase in mitochondrial respiration in peripheral sensory neurons. Although Epac2 activation leads to Pdha1 (pyruvate dehydrogenase) phosphorylation in dissociated neurons from mice of both sexes, induction of this pathway in vivo by hindpaw insult is restricted to males and appears to require intraganglionic prostaglandin synthesis. These findings support a model in which Gs-coupled receptor modulation of mitochondrial function promotes acute nociceptive signaling and inflammatory hyperalgesia.
Collapse
|
19
|
Krishnan A, Dwivedi S, Chandrasekhar A, Areti A, Zochodne DW. In vitro priming response in dorsal root ganglia partially mimics injury-driven pre-conditioning response and reprograms neurons for enhanced outgrowth. Mol Cell Neurosci 2020; 110:103573. [PMID: 33248236 DOI: 10.1016/j.mcn.2020.103573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 11/02/2020] [Accepted: 11/23/2020] [Indexed: 11/15/2022] Open
Abstract
Peripheral nerve injuries have the potential to bring about long-term disabilities in individuals. The major issue in repairing nerve injuries is the poor growth rate of axons. Although several molecules have been identified as potential candidates for improving axon growth, their potential translation into clinical practice is preliminary and largely unexplored. This necessitates identifying additional molecular candidates with superior potential to improve axon growth. Lack of a simple non-surgical screening model also poses a hurdle in rapidly screening potential candidate molecules. In this work, we developed a novel, rapid screening model for nerve regeneration therapeutics that retains a focus on adult neurons. The model involves simple incubation of sensory ganglia over a period of 24 h prior to dissociation. Surprisingly, this model features unique events that reprogram both sensory neurons and supporting glia favoring axon growth. Moreover, several associated cellular and molecular changes involved in this model partially mimic classic axotomy-induced changes in sensory ganglia. Overall, this model presents with a platform that not only allows rapid screening of drug candidates but offers opportunities in studying novel intrinsic molecular changes in both neurons and glial cells directed towards improving the pace of axon growth.
Collapse
Affiliation(s)
- Anand Krishnan
- Division of Neurology, Department of Medicine, and the Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada; Dept. of Anatomy, Physiology, and Pharmacology, College of Medicine, University of Saskatchewan, Canada; Cameco MS Neuroscience Research Centre, Saskatoon, Saskatchewan, Canada
| | - Shubham Dwivedi
- Dept. of Anatomy, Physiology, and Pharmacology, College of Medicine, University of Saskatchewan, Canada; Cameco MS Neuroscience Research Centre, Saskatoon, Saskatchewan, Canada
| | - Ambika Chandrasekhar
- Division of Neurology, Department of Medicine, and the Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Aparna Areti
- Division of Neurology, Department of Medicine, and the Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Douglas W Zochodne
- Division of Neurology, Department of Medicine, and the Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
20
|
Avraham O, Deng PY, Jones S, Kuruvilla R, Semenkovich CF, Klyachko VA, Cavalli V. Satellite glial cells promote regenerative growth in sensory neurons. Nat Commun 2020; 11:4891. [PMID: 32994417 PMCID: PMC7524726 DOI: 10.1038/s41467-020-18642-y] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 09/04/2020] [Indexed: 01/11/2023] Open
Abstract
Peripheral sensory neurons regenerate their axon after nerve injury to enable functional recovery. Intrinsic mechanisms operating in sensory neurons are known to regulate nerve repair, but whether satellite glial cells (SGC), which completely envelop the neuronal soma, contribute to nerve regeneration remains unexplored. Using a single cell RNAseq approach, we reveal that SGC are distinct from Schwann cells and share similarities with astrocytes. Nerve injury elicits changes in the expression of genes related to fatty acid synthesis and peroxisome proliferator-activated receptor (PPARα) signaling. Conditional deletion of fatty acid synthase (Fasn) in SGC impairs axon regeneration. The PPARα agonist fenofibrate rescues the impaired axon regeneration in mice lacking Fasn in SGC. These results indicate that PPARα activity downstream of FASN in SGC contributes to promote axon regeneration in adult peripheral nerves and highlight that the sensory neuron and its surrounding glial coat form a functional unit that orchestrates nerve repair. The contribution of satellite glia to peripheral nerve regeneration is unclear. Here, the authors show that satellite glia are transcriptionally distinct from Schwann cells, share similarities with astrocytes, and, upon injury, they contribute to axon regeneration via Fasn-PPARα signalling pathway.
Collapse
Affiliation(s)
- Oshri Avraham
- Department of Neuroscience, Washington University School of Medicine, St Louis, MO, 63110, USA
| | - Pan-Yue Deng
- Department of Cell Biology and Physiology, Washington University School of Medicine, St Louis, MO, 63110, USA
| | - Sara Jones
- Department of Neuroscience, Washington University School of Medicine, St Louis, MO, 63110, USA
| | - Rejji Kuruvilla
- Department of Biology, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Clay F Semenkovich
- Department of Cell Biology and Physiology, Washington University School of Medicine, St Louis, MO, 63110, USA.,Division of Endocrinology, Metabolism & Lipid Research, Washington University School of Medicine, St Louis, MO, 63110, USA
| | - Vitaly A Klyachko
- Department of Cell Biology and Physiology, Washington University School of Medicine, St Louis, MO, 63110, USA
| | - Valeria Cavalli
- Department of Neuroscience, Washington University School of Medicine, St Louis, MO, 63110, USA. .,Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA. .,Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| |
Collapse
|
21
|
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
|
22
|
Krishnan A, Bhavanam S, Zochodne D. An Intimate Role for Adult Dorsal Root Ganglia Resident Cycling Cells in the Generation of Local Macrophages and Satellite Glial Cells. J Neuropathol Exp Neurol 2019; 77:929-941. [PMID: 30169768 DOI: 10.1093/jnen/nly072] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The intricate interactions between neurons, glial, and inflammatory cells within peripheral ganglia are physiologically important, but not well explored. Here, we show that adult dorsal root ganglia (DRG) contain populations of self-renewing cells, collectively referred as DRG resident cycling cells (DRCCs), that are active not only in "quiescent" ganglia but also accelerate their turnover in response to distal axotomy. An unexpected proportion of DRCCs were resident macrophages. These cells closely accompanied, and aligned with recycling satellite glial cells (SGCs) that were juxtaposed to sensory neurons and possessed stem cell-like properties. Selective inhibition of colony stimulating factor 1 receptor prevented the local proliferation of macrophages. Interestingly, DRCC turnover was accompanied by apoptosis at later intervals indicating a balanced cellular milieu in the DRGs. These findings identify a complex interactive multicellular DRG microenvironment supporting self-renewal of both macrophages and SGCs and its potential implications in the overall response of adult DRGs to injury.
Collapse
Affiliation(s)
- Anand Krishnan
- Neuroscience and Mental Health Institute.,Division of Neurology, Department of Medicine.,Alberta Diabetes Institute
| | - Sudha Bhavanam
- Division of Laboratory Medicine and Pathology, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Douglas Zochodne
- Neuroscience and Mental Health Institute.,Division of Neurology, Department of Medicine.,Alberta Diabetes Institute
| |
Collapse
|
23
|
Zhang Q, Zhao J, Shen J, Zhang X, Ren R, Ma Z, He Y, Kang Q, Wang Y, Dong X, Sun J, Liu Z, Yi X. The ATP-P2X7 Signaling Pathway Participates in the Regulation of Slit1 Expression in Satellite Glial Cells. Front Cell Neurosci 2019; 13:420. [PMID: 31607866 PMCID: PMC6761959 DOI: 10.3389/fncel.2019.00420] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 09/02/2019] [Indexed: 11/17/2022] Open
Abstract
Slit1 is one of the known signaling factors of the slit family and can promote neurite growth by binding to its receptor, Robo2. Upregulation of Slit1 expression in dorsal root ganglia (DRG) after peripheral nerve injury plays an important role in nerve regeneration. Each sensory neuronal soma in the DRG is encapsulated by several surrounding satellite glial cells (SGCs) to form a neural structural unit. However, the temporal and spatial patterns of Slit1 upregulation in SGCs in DRG and its molecular mechanisms are not well understood. This study examined the spatial and temporal patterns of Slit1 expression in DRG after sciatic nerve crush by immunohistochemistry and western blotting. The effect of neuronal damage signaling on the expression of Slit1 in SGCs was studied in vivo by fluorescent gold retrograde tracing and double immunofluorescence staining. The relationship between the expression of Slit1 in SGCs and neuronal somas was also observed by culturing DRG cells and double immunofluorescence labeling. The molecular mechanism of Slit1 was further explored by immunohistochemistry and western blotting after intraperitoneal injection of Bright Blue G (BBG, P2X7R inhibitor). The results showed that after peripheral nerve injury, the expression of Slit1 in the neurons and SGCs of DRG increased. The expression of Slit1 was presented with a time lag in SGCs than in neurons. The expression of Slit1 in SGCs was induced by contact with surrounding neuronal somas. Through injured cell localization, it was found that the expression of Slit1 was stronger in SGCs surrounding injured neurons than in SGCs surrounding non-injured neurons. The expression of vesicular nucleotide transporter (VNUT) in DRG neurons was increased by injury signaling. After the inhibition of P2X7R, the expression of Slit1 in SGCs was downregulated, and the expression of VNUT in DRG neurons was upregulated. These results indicate that the ATP-P2X7R pathway is involved in signal transduction from peripheral nerve injury to SGCs, leading to the upregulation of Slit1 expression.
Collapse
Affiliation(s)
- Quanpeng Zhang
- Department of Anatomy, Hainan Medical University, Haikou, China.,Joint Laboratory for Neuroscience, Hainan Medical University, Fourth Military Medical University, Haikou, China
| | - Jiuhong Zhao
- Department of Anatomy, Hainan Medical University, Haikou, China.,Joint Laboratory for Neuroscience, Hainan Medical University, Fourth Military Medical University, Haikou, China
| | - Jing Shen
- Department of Ophthalmology, First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Xianfang Zhang
- Department of Anatomy, Hainan Medical University, Haikou, China.,Joint Laboratory for Neuroscience, Hainan Medical University, Fourth Military Medical University, Haikou, China
| | - Rui Ren
- Department of Anatomy, Hainan Medical University, Haikou, China.,Joint Laboratory for Neuroscience, Hainan Medical University, Fourth Military Medical University, Haikou, China
| | - Zhijian Ma
- Department of Anatomy, Hainan Medical University, Haikou, China.,Joint Laboratory for Neuroscience, Hainan Medical University, Fourth Military Medical University, Haikou, China
| | - Yuebin He
- Joint Laboratory for Neuroscience, Hainan Medical University, Fourth Military Medical University, Haikou, China
| | - Qian Kang
- Infection Control Department, People's Hospital of Xing'an County, Guilin, China
| | - Yanshan Wang
- Quality Inspection Department, Minghui Industry (Shenzhen) Co., Ltd., Shenzhen, China
| | - Xu Dong
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical University, Haikou, China
| | - Jin Sun
- Department of Clinical Medicine, Hainan Medical University, Haikou, China
| | - Zhuozhou Liu
- Department of Clinical Medicine, Hainan Medical University, Haikou, China
| | - Xinan Yi
- Department of Anatomy, Hainan Medical University, Haikou, China.,Joint Laboratory for Neuroscience, Hainan Medical University, Fourth Military Medical University, Haikou, China
| |
Collapse
|
24
|
Abstract
Permanent disabilities following CNS injuries result from the failure of injured axons to regenerate and rebuild functional connections with their original targets. By contrast, injury to peripheral nerves is followed by robust regeneration, which can lead to recovery of sensory and motor functions. This regenerative response requires the induction of widespread transcriptional and epigenetic changes in injured neurons. Considerable progress has been made in recent years in understanding how peripheral axon injury elicits these widespread changes through the coordinated actions of transcription factors, epigenetic modifiers and, to a lesser extent, microRNAs. Although many questions remain about the interplay between these mechanisms, these new findings provide important insights into the pivotal role of coordinated gene expression and chromatin remodelling in the neuronal response to injury.
Collapse
Affiliation(s)
- Marcus Mahar
- Department of Neuroscience, Hope Center for Neurological Disorders and Center of Regenerative Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Valeria Cavalli
- Department of Neuroscience, Hope Center for Neurological Disorders and Center of Regenerative Medicine, Washington University School of Medicine, St Louis, MO, USA.
| |
Collapse
|
25
|
Carlin D, Halevi AE, Ewan EE, Moore AM, Cavalli V. Nociceptor Deletion of Tsc2 Enhances Axon Regeneration by Inducing a Conditioning Injury Response in Dorsal Root Ganglia. eNeuro 2019; 6:ENEURO.0168-19.2019. [PMID: 31182472 PMCID: PMC6595439 DOI: 10.1523/eneuro.0168-19.2019] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 05/15/2019] [Accepted: 05/18/2019] [Indexed: 12/30/2022] Open
Abstract
Neurons of the PNS are able to regenerate injured axons, a process requiring significant cellular resources to establish and maintain long-distance growth. Genetic activation of mTORC1, a potent regulator of cellular metabolism and protein translation, improves axon regeneration of peripheral neurons by an unresolved mechanism. To gain insight into this process, we activated mTORC1 signaling in mouse nociceptors via genetic deletion of its negative regulator Tsc2. Perinatal deletion of Tsc2 in nociceptors enhanced initial axon growth after sciatic nerve crush, however by 3 d post-injury axon elongation rate became similar to controls. mTORC1 inhibition prior to nerve injury was required to suppress the enhanced axon growth. Gene expression analysis in purified nociceptors revealed that Tsc2-deficient nociceptors had increased activity of regeneration-associated transcription factors (RATFs), including cJun and Atf3, in the absence of injury. Additionally, nociceptor deletion of Tsc2 activated satellite glial cells and macrophages in the dorsal root ganglia (DRG) in a similar manner to nerve injury. Surprisingly, these changes improved axon length but not percentage of initiating axons in dissociated cultures. The pro-regenerative environment in naïve DRG was recapitulated by AAV8-mediated deletion of Tsc2 in adult mice, suggesting that this phenotype does not result from a developmental effect. Consistently, AAV8-mediated Tsc2 deletion did not improve behavioral recovery after a sciatic nerve crush injury despite initially enhanced axon growth. Together, these data show that neuronal mTORC1 activation induces an incomplete pro-regenerative environment in the DRG that improves initial but not later axon growth after nerve injury.
Collapse
Affiliation(s)
- Dan Carlin
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO 63110
| | - Alexandra E Halevi
- Division of Plastic and Reconstructive Surgery, Washington University School of Medicine, St. Louis, MO 63110
| | - Eric E Ewan
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO 63110
| | - Amy M Moore
- Division of Plastic and Reconstructive Surgery, Washington University School of Medicine, St. Louis, MO 63110
| | - Valeria Cavalli
- Department of Neuroscience, Center of Regenerative Medicine, Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110
| |
Collapse
|
26
|
Pontes RB, Lisboa MRP, Pereira AF, Lino JA, de Oliveira FFB, de Mesquita AKV, de Freitas Alves BW, Lima-Júnior RCP, Vale ML. Involvement of Endothelin Receptors in Peripheral Sensory Neuropathy Induced by Oxaliplatin in Mice. Neurotox Res 2019; 36:688-699. [PMID: 31228092 DOI: 10.1007/s12640-019-00074-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 05/24/2019] [Accepted: 06/11/2019] [Indexed: 01/10/2023]
Abstract
The aim of this study was to evaluate the participation of the endothelin ETA and ETB receptors and the effects of bosentan in oxaliplatin-induced peripheral sensory neuropathy (OIN) in mice. Adult male Swiss mice received 1 mg/kg of oxaliplatin intravenously, twice a week for 5 weeks. Dorsal root ganglia (DRG) and spinal cords were removed for evaluation of the endothelin ETA and ETB receptor expression. Afterwards, selective (BQ-123 and BQ-788; 10 nmol in 30 μL, intraplantarly) and non-selective (bosentan, 100 mg/kg, orally) antagonists were administered in order to evaluate the involvement of the endothelin receptors in OIN. Mechanical and thermal nociception tests were performed once a week for 56 days. Oxaliplatin induced mechanical and thermal hypersensitivity and increased the endothelin ETA receptor expression in both the DRG and spinal cord (P < 0.05). Endothelin ETB receptor expression was increased in the DRG (P < 0.05) but not in the spinal cord. Both endothelin ETA and ETB receptor selective antagonists partially prevented mechanical hyperalgesia in mice with OIN (P < 0.05). Moreover, bosentan prevented mechanical and thermal hypersensitivity in oxaliplatin-treated mice (P < 0.05). In conclusion, both endothelin ETA and ETB receptors seem to be involved in the OIN in mice and they should be considered possible targets for the management of this clinical feature.
Collapse
Affiliation(s)
- Renata Bessa Pontes
- Department of Physical Therapy, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, 60430-160, Brazil
| | - Mario Roberto Pontes Lisboa
- Department of Morphology, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, 60430-170, Brazil
| | - Anamaria Falcão Pereira
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará (UFC), R. Cel. Nunes de Melo, 1127, Rodolfo Teófilo, Fortaleza, CE, 60430-270, Brazil
| | - Juliana Arcanjo Lino
- Department of Clinical Medicine, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, 60430-140, Brazil
| | - Francisco Fábio Bezerra de Oliveira
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará (UFC), R. Cel. Nunes de Melo, 1127, Rodolfo Teófilo, Fortaleza, CE, 60430-270, Brazil
| | | | | | - Roberto César Pereira Lima-Júnior
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará (UFC), R. Cel. Nunes de Melo, 1127, Rodolfo Teófilo, Fortaleza, CE, 60430-270, Brazil
| | - Mariana Lima Vale
- Department of Morphology, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, 60430-170, Brazil.
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará (UFC), R. Cel. Nunes de Melo, 1127, Rodolfo Teófilo, Fortaleza, CE, 60430-270, Brazil.
| |
Collapse
|
27
|
Pannese E. Biology and Pathology of Perineuronal Satellite Cells in Sensory Ganglia. BIOLOGY AND PATHOLOGY OF PERINEURONAL SATELLITE CELLS IN SENSORY GANGLIA 2018. [DOI: 10.1007/978-3-319-60140-3_1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
28
|
Coutinho-Budd JC, Sheehan AE, Freeman MR. The secreted neurotrophin Spätzle 3 promotes glial morphogenesis and supports neuronal survival and function. Genes Dev 2017; 31:2023-2038. [PMID: 29138279 PMCID: PMC5733495 DOI: 10.1101/gad.305888.117] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 10/20/2017] [Indexed: 10/27/2022]
Abstract
Most glial functions depend on establishing intimate morphological relationships with neurons. Significant progress has been made in understanding neuron-glia signaling at synaptic and axonal contacts, but how glia support neuronal cell bodies is unclear. Here we explored the growth and functions of Drosophila cortex glia (which associate almost exclusively with neuronal cell bodies) to understand glia-soma interactions. We show that cortex glia tile with one another and with astrocytes to establish unique central nervous system (CNS) spatial domains that actively restrict glial growth, and selective ablation of cortex glia causes animal lethality. In an RNAi-based screen, we identified αSNAP (soluble NSF [N-ethylmalemeide-sensitive factor] attachment protein α) and several components of vesicle fusion and recycling machinery as essential for the maintenance of cortex glial morphology and continued contact with neurons. Interestingly, loss of the secreted neurotrophin Spätzle 3 (Spz3) phenocopied αSNAP phenotypes, which included loss of glial ensheathment of neuron cell bodies, increased neuronal cell death, and defects in animal behavior. Rescue experiments suggest that Spz3 can exert these effects only over very short distances. This work identifies essential roles for glial ensheathment of neuronal cell bodies in CNS homeostasis as well as Spz3 as a novel signaling factor required for maintenance of cortex glial morphology and neuron-glia contact.
Collapse
Affiliation(s)
- Jaeda C Coutinho-Budd
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, Massachusetts 01605, USA
| | - Amy E Sheehan
- Vollum Institute, Oregon Health and Sciences University, Portland, Oregon 97239, USA
| | - Marc R Freeman
- Vollum Institute, Oregon Health and Sciences University, Portland, Oregon 97239, USA
| |
Collapse
|
29
|
Fadda A, Bärtschi M, Hemphill A, Widmer HR, Zurbriggen A, Perona P, Vidondo B, Oevermann A. Primary Postnatal Dorsal Root Ganglion Culture from Conventionally Slaughtered Calves. PLoS One 2016; 11:e0168228. [PMID: 27936156 PMCID: PMC5148591 DOI: 10.1371/journal.pone.0168228] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 11/28/2016] [Indexed: 12/13/2022] Open
Abstract
Neurological disorders in ruminants have an important impact on veterinary health, but very few host-specific in vitro models have been established to study diseases affecting the nervous system. Here we describe a primary neuronal dorsal root ganglia (DRG) culture derived from calves after being conventionally slaughtered for food consumption. The study focuses on the in vitro characterization of bovine DRG cell populations by immunofluorescence analysis. The effects of various growth factors on neuron viability, neurite outgrowth and arborisation were evaluated by morphological analysis. Bovine DRG neurons are able to survive for more than 4 weeks in culture. GF supplementation is not required for neuronal survival and neurite outgrowth. However, exogenously added growth factors promote neurite outgrowth. DRG cultures from regularly slaughtered calves represent a promising and sustainable host specific model for the investigation of pain and neurological diseases in bovines.
Collapse
Affiliation(s)
- A. Fadda
- Division of Neurological Sciences, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, Theodor Kocher Institute, University of Bern, Switzerland
| | - M. Bärtschi
- Division of Neurological Sciences, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - A. Hemphill
- Institute for Parasitology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - H. R. Widmer
- Neurocenter and Regenerative Neuroscience Cluster, University Hospital and University of Bern, Bern, Switzerland
| | - A. Zurbriggen
- Division of Neurological Sciences, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - P. Perona
- School of Engineering, The University of Edinburgh, Edinburgh, United Kingdom
| | - B. Vidondo
- Veterinary Public Health Institute (VPHI), Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - A. Oevermann
- Division of Neurological Sciences, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- * E-mail:
| |
Collapse
|
30
|
Ciglieri E, Ferrini F, Boggio E, Salio C. An improved method for in vitro morphofunctional analysis of mouse dorsal root ganglia. Ann Anat 2016; 207:62-7. [PMID: 27224901 DOI: 10.1016/j.aanat.2016.04.032] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 03/17/2016] [Accepted: 04/21/2016] [Indexed: 12/11/2022]
Abstract
Sensory neurons in dorsal root ganglia (DRGs) are the first-order neurons along the pathway conveying sensory information from the periphery to the central nervous system. The analysis of the morphological and physiological features of these neurons and their alterations in pathology is the necessary prerequisite to understand pain encoding mechanisms. Here, we describe an in vitro procedure for combined morphofunctional analysis of mouse DRGs. Freshly excised DRGs obtained from adult mice were incubated in collagenase to dissolve the ensheathing connective capsule. The degradation of the connective tissue facilitates both access to the neurons by classical recording glass pipettes and the penetration of primary antibodies for immunohistochemical procedures. The entire DRGs were then imaged using a confocal microscope obtaining a fine 3D representation of their cytoarchitecture without requiring tissue sectioning. Thus, our proposed whole-mount preparation represents a flexible in vitro approach for both functional and phenotypic analysis of DRG neurons by at the same time preserving their neuroanatomical relationships.
Collapse
Affiliation(s)
- E Ciglieri
- Department of Veterinary Sciences, University of Turin, Largo Braccini 2, 10095 Grugliasco, TO, Italy
| | - F Ferrini
- Department of Veterinary Sciences, University of Turin, Largo Braccini 2, 10095 Grugliasco, TO, Italy
| | - E Boggio
- Department of Veterinary Sciences, University of Turin, Largo Braccini 2, 10095 Grugliasco, TO, Italy
| | - C Salio
- Department of Veterinary Sciences, University of Turin, Largo Braccini 2, 10095 Grugliasco, TO, Italy.
| |
Collapse
|