1
|
Martinez P, Sabatier JM. Malignant tumors in vagal-innervated organs: Exploring its homeostatic role. Cancer Lett 2025; 617:217539. [PMID: 39954934 DOI: 10.1016/j.canlet.2025.217539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 01/28/2025] [Accepted: 02/06/2025] [Indexed: 02/17/2025]
Abstract
Cancer remains a significant global health challenge, with its progression shaped by complex and multifactorial mechanisms. Recent research suggests that the vagus nerve could play a critical role in mediating communication between the tumor microenvironment and the central nervous system (CNS). This review highlights the diversity of vagal afferent receptors, which could position the vagus nerve as a unique pathway for transmitting immune, metabolic, mechanical, and chemical signals from tumors to the CNS. Such signaling could influence systemic disease progression and tumor-related responses. Additionally, the vagus nerve's interactions with the microbiome and the renin-angiotensin system (RAS)-both implicated in cancer biology-further underscore its potential central role in modulating tumor-related processes. Contradictions in the literature, particularly concerning vagal fibers, illustrate the complexity of its involvement in tumor progression, with both tumor-promoting and tumor-suppressive effects reported depending on cancer type and context. These contradictions often overlook certain experimental biases, such as the failure to distinguish between vagal afferent and efferent fibers during vagotomies or the localized parasympathetic effects that cannot always be extrapolated to the systemic level. By focusing on the homeostatic role of the vagus nerve, understanding these mechanisms could open the door to new perspectives in cancer research related to the vagus nerve and lead to potential therapeutic innovations.
Collapse
Affiliation(s)
| | - Jean-Marc Sabatier
- Institut de NeuroPhysiopathologie (INP), CNRS UMR 7051, 27 Bd Jean Moulin, 13005, Marseille, France
| |
Collapse
|
2
|
Kallogjerovic S, Velázquez‐Quesada I, Hadap R, Gligorijevic B. Retrograde tracing of breast cancer-associated sensory neurons. J Microsc 2025; 298:232-244. [PMID: 38881512 PMCID: PMC11847563 DOI: 10.1111/jmi.13340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 05/24/2024] [Accepted: 06/05/2024] [Indexed: 06/18/2024]
Abstract
Breast cancer is one of the leading causes of mortality among women. The tumour microenvironment, consisting of host cells and extracellular matrix, has been increasingly studied for its interplay with cancer cells, and the resulting effect on tumour progression. While the breast is one of the most innervated organs in the body, the role of neurons, and specifically sensory neurons, has been understudied, mostly for technical reasons. One of the reasons is the anatomy of sensory neurons: sensory neuron somas are located in the spine, and their axons can extend longer than a meter across the body to provide innervation in the breast. Next, neurons are challenging to culture, and there are no cell lines adequately representing the diversity of sensory neurons. Finally, sensory neurons are responsible for transporting several different types of signals to the brain, and there are many different subtypes of sensory neurons. The subtypes of sensory neurons, which innervate and interact with breast tumours, are unknown. To establish the tools for labelling and subtyping neurons that interact with breast cancer cells, we utilised two retrograde tracer's standards in neuroscience, wheat-germ agglutinin (WGA) and cholera toxin subunit B (CTB). In vitro, we employed primary sensory neurons isolated from mouse dorsal root ganglia, cultured in a custom-built microfluidic device DACIT, that mimics the anatomical compartmentalisation of the sensory neuron's soma and axons. In vivo, we utilised both syngeneic and transgenic mouse models of mammary carcinoma. We show that CTB and WGA trace different but overlapping sensory neuronal subpopulations: while WGA is more efficient in labelling CGRP+ neurons, CTB is superior in labelling the NF200+ neurons. Surprisingly, both tracers are also taken up by a significant population of breast cancer cells, both in vitro and in vivo. In summary, we have established methodologies for retrograde tracing of sensory neurons interacting with breast cancer cells. Our tools will be useful for future studies of breast tumour innervation, and development of therapies targeting breast cancer-associated neuron subpopulations of sensory neurons. Lay description: Breast cancer is an aggressive disease that affects both women and men throughout the world. While it has been reported that the increasing size of nerves in breast cancer correlates to bad prognosis in patients, the role of nerves, especially sensory nerves, in breast cancer progression, has remained largely understudied. Sensory nerves are responsible for delivering signals such as pain, mechanical forces (pressure, tension, stretch, touch) and temperature to the brain. The human body is densely innervated, and nerves extending into peripheral organs can be as long as a few meters. Nerve classification and function can be very complex, as they contain bundles of extensions (axons) originating in different neuronal bodies (soma). Maintaining neurons and growing axons in cell culture conditions in order to mimic innervation is technically challenging, as it involves multiple organs of the human body. Here, we focus on tracing sensory axons from the breast tumours back to the neuronal soma, located in the dorsal root ganglia, inside the spine. To do so, we are using two different 'retrograde' tracers, WGA and CTB, which are proteins with a natural ability to enter axons and travel in a retrograde fashion, arriving at the soma, even if it means to travel distances longer than a meter. Both tracers are fluorescently labelled, making them visible using high-resolution fluorescent microscopy. We show that both WGA and CTB can label sensory neurons in tumours, or in cell culture conditions. The two tracers differ in efficiency of tracing different sensory neurons subpopulations: while WGA is more efficient in tracing small C-fibres (CGRP-positive), CTB is more efficient in tracing A-fibres (NF200+) of sensory neurons. In summary, we have successfully established retrograde tracing techniques for sensory neurons towards studying and targeting breast cancer innervation.
Collapse
Affiliation(s)
| | | | - Rutva Hadap
- Bioengineering DepartmentTemple UniversityPhiladelphiaPennsylvaniaUSA
| | - Bojana Gligorijevic
- Bioengineering DepartmentTemple UniversityPhiladelphiaPennsylvaniaUSA
- Cancer Signaling and Microenvironment ProgramFox Chase Cancer CenterPhiladelphiaPennsylvaniaUSA
- Fels Cancer Institute for Personalized MedicineLewis Katz School of MedicineTemple UniversityPhiladelphiaPennsylvaniaUSA
| |
Collapse
|
3
|
Joshi O, Cooper A, Powell R, Martin MK, Rodriguez R, Kuechle JB, Bhattacharjee A. Localization of AP2α2, TRPV1 and PIEZO2 to the Large Dense Core Vesicles of Human Dorsal Root Ganglion Neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.31.646357. [PMID: 40236095 PMCID: PMC11996434 DOI: 10.1101/2025.03.31.646357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Dorsal Root Ganglia (DRG) consist of both peptidergic and non-peptidergic nociceptive neurons. CGRP, an inflammatory neuropeptide, is a classical marker of peptidergic nociceptors and CGRP is stored within the large dense core vesicles (LDCVs) of these neurons. In addition to storing large peptide neurotransmitters, LDCVs might also serve to transport key membrane proteins to the peripheral terminals. This immunohistochemical study investigated the localization of different membrane proteins to the LDCVs of human DRG neurons. Previously validated antibodies against the endocytotic subunit AP2α2, the heat-activated channel TRPV1 and the mechanosensitive channel PIEZO2 were used in conjunction with an antibody against CGRP on sections of intact human DRG isolated from de-identified human subjects. Immunohistochemical studies were also performed on human synovial tissue to examine peripheral terminals. High magnification confocal microscopy was used to determine the co-localization signal of these membrane proteins with CGRP. We observed a strong co-localization of AP2α2 with the CGRP containing LDCVs signifying its role in membrane recycling. Moreover, we also observed a strong colocalization of TRPV1 and PIEZO2 with CGRP suggesting that LDCV release controls the trafficking of these channels to the membrane. It is likely that during injury, bulk exocytosis of CGRP will concomitantly increase the surface expression of TRPV1 and PIEZO2 channels enhancing the responsiveness of these neurons to painful stimuli. This model suggests that neurons that co-localize TRPV1 and PIEZO2 to CGRP containing LDCVs are likely silent nociceptors.
Collapse
|
4
|
Mwirigi JM, Sankaranarayanan I, Tavares-Ferreira D, Gabriel KA, Palomino S, Li Y, Uhelski ML, Shiers S, Franco-Enzástiga Ú, Wangzhou A, Lesnak JB, Bandaru S, Shrivastava A, Inturi N, Albrecht PJ, Dockum M, Cervantes AM, Horton P, Funk G, North RY, Tatsui CE, Corrales G, Yousuf MS, Curatolo M, Gereau RW, Patwardhan A, Dussor G, Dougherty PM, Rice FL, Price TJ. Expansion of OSMR expression and signaling in the human dorsal root ganglion links OSM to neuropathic pain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.26.645611. [PMID: 40236060 PMCID: PMC11996445 DOI: 10.1101/2025.03.26.645611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
RNA sequencing studies on human dorsal root ganglion (hDRG) from patients suffering from neuropathic pain show upregulation of OSM, linking this IL-6 family cytokine to pain disorders. In mice, however, OSM signaling causes itch behaviors through a direct effect on its cognate receptor expressed uniquely by pruriceptive sensory neurons. We hypothesized that an expansion in function of OSM-OSM receptor (OSMR) in sensory disorders in humans could be explained by species differences in receptor expression and signaling. Our in situ hybridization and immunohistochemical findings demonstrate broad expression of OSMR in DRG nociceptors and afferent fibers innervating the superficial and deep skin of humans. In patch-clamp electrophysiology, OSM directly activates human sensory neurons engaging MAPK signaling to promote action potential firing. Using CRISPR editing we show that OSM activation of MAPK signaling is dependent on OSMR and not LIFR in hDRG. Bulk, single-nuclei, and single-cell RNA-seq of OSM-treated hDRG cultures reveal expansive similarities in the transcriptomic signature observed in pain DRGs from neuropathic patients, indicating that OSM alone can orchestrate transcriptomic signatures associated with pain. We conclude that OSM-OSMR signaling via MAPKs is a critical signaling factor for DRG plasticity that may underlie neuropathic pain in patients.
Collapse
|
5
|
Palomino SM, Gabriel KA, Mwirigi JM, Cervantes A, Horton P, Funk G, Moutal A, Martin LF, Khanna R, Price TJ, Patwardhan A. Genetic editing of primary human dorsal root ganglion neurons using CRISPR-Cas9. Sci Rep 2025; 15:11116. [PMID: 40169710 PMCID: PMC11961745 DOI: 10.1038/s41598-025-91153-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 02/18/2025] [Indexed: 04/03/2025] Open
Abstract
CRISPR-Cas9 is now the leading method for genome editing and is advancing for the treatment of human disease. CRIPSR has promise in treating neurological diseases, but traditional viral-vector-delivery approaches have neurotoxicity limiting their use. Here we describe a simple method for non-viral transfection of primary human DRG (hDRG) neurons for CRISPR-Cas9 editing. We edited TRPV1, NTSR2, and CACNA1E using a lipofection method with CRISPR-Cas9 plasmids containing reporter tags (GFP or mCherry). Transfection was successfully demonstrated by the expression of the reporters two days post-administration. CRISPR-Cas9 editing was confirmed at the genome level with a T7-endonuclease-I assay; protein level with immunocytochemistry and Western blot; and functional level through capsaicin-induced Ca2+ accumulation in a high-throughput compatible fluorescent imaging plate reader (FLIPR) system. This work establishes a reliable, target specific, non-viral CRISPR-Cas9-mediated genetic editing in primary human neurons with potential for future clinical application for sensory diseases.
Collapse
Affiliation(s)
- Seph M Palomino
- Department of Anesthesiology and Pain Management, University of Texas Southwestern Medical Center, 6202 Harry Hines Blvd., 9th Floor, Dallas, 75235, TX, USA
| | - Katherin A Gabriel
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, 800 W Campbell Rd, Richardson, TX, 75080, USA
| | - Juliet M Mwirigi
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, 800 W Campbell Rd, Richardson, TX, 75080, USA
| | - Anna Cervantes
- Southwest Transplant Alliance, Manderville Ln, Dallas, TX, 8190, 75231, USA
| | - Peter Horton
- Southwest Transplant Alliance, Manderville Ln, Dallas, TX, 8190, 75231, USA
| | - Geoffrey Funk
- Southwest Transplant Alliance, Manderville Ln, Dallas, TX, 8190, 75231, USA
| | - Aubin Moutal
- Department of Pharmacology and Physiology, Saint Louis University, 1402 S. Grand Blvd, St. Louis, Mo, 63104, USA
| | - Laurent F Martin
- Department of Pharmacology, University of Arizona, 1501 N Campbell Ave, Tucson, AZ, 85721, USA
| | - Rajesh Khanna
- Department of Pharmacology and Therapeutics, University of Florida, 1200 Newell Drive, Gainesville, FL, ARB R5-234, 32610-0267, USA
| | - Theodore J Price
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, 800 W Campbell Rd, Richardson, TX, 75080, USA.
| | - Amol Patwardhan
- Department of Anesthesiology and Pain Management, University of Texas Southwestern Medical Center, 6202 Harry Hines Blvd., 9th Floor, Dallas, 75235, TX, USA.
| |
Collapse
|
6
|
Haberberger RV, Matusica D, Shiers S, Sankaranarayanan I, Price TJ. Transcriptomic and Histological Characterization of Telocytes in the Human Dorsal Root Ganglion. J Comp Neurol 2025; 533:e70044. [PMID: 40097369 PMCID: PMC11913768 DOI: 10.1002/cne.70044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 01/23/2025] [Accepted: 03/05/2025] [Indexed: 03/19/2025]
Abstract
Telocytes are interstitial cells characterized by long processes that span considerable distances within tissues, likely facilitating coordination and interaction with various cell types. Although present in central and peripheral neuronal tissues, their role remains elusive. Dorsal root ganglia (DRG) house pseudounipolar afferent neurons responsible for transmitting signals related to temperature, proprioception, and nociception. This study aimed to investigate the presence and function of telocytes in human DRG by examining their transcriptional profile, anatomical location, and ultrastructure. Combined expression of CD34 and PDGFRA is a marker gene set for telocytes, and our sequencing data revealed CD34 and PDGFRA expressing cells comprise roughly 1.5%-3% of DRG cells. Combined expression of CD34 and PDGFRA is a putative marker gene set for telocytes. Further analysis identified nine subclusters with enriched cluster-specific genes. Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) pathway analysis suggested vascular, immune, and connective tissue-associated putative telocyte subtypes, mapping over 3000 potential receptor-ligand interactions between sensory neurons and these CD34 and PDGFRA expressing putative telocytes were identified using a ligand-receptors interactome platform. Immunohistochemistry identified CD34+ve telocytes in the endoneural space of DRGs, next to neuron-satellite complexes, in perivascular spaces and in the endoneural space between nerve fiber bundles, consistent with pathway analysis. Transmission electron microscopy (TEM) confirmed their location identifying characteristic elongated nucleus, long and thin telopodes containing vesicles, often surrounded by a basal lamina. This study provides the first gene expression analysis of telocytes in complex human tissue, specifically the DRG, highlighting functional differences based on tissue location while revealing no significant ultrastructural variations.
Collapse
Affiliation(s)
- Rainer V. Haberberger
- Department of Anatomy and Pathology, School of BiomedicineThe University of AdelaideAdelaideAustralia
- Anatomy, Histology & Pathology, College of Medicine and Public HealthFlinders UniversityAdelaideAustralia
| | - Dusan Matusica
- Anatomy, Histology & Pathology, College of Medicine and Public HealthFlinders UniversityAdelaideAustralia
| | - Stephanie Shiers
- Department of Neuroscience, Center for Advanced Pain StudiesThe University of Texas at DallasRichardsonTexasUSA
| | - Ishwarya Sankaranarayanan
- Department of Neuroscience, Center for Advanced Pain StudiesThe University of Texas at DallasRichardsonTexasUSA
| | - Theodore J. Price
- Department of Neuroscience, Center for Advanced Pain StudiesThe University of Texas at DallasRichardsonTexasUSA
| |
Collapse
|
7
|
Weiner S, Perleth S, Kampf T, Lau K, Hessenauer F, Homola G, Nordbeck P, Üçeyler N, Sommer C, Pham M, Schindehütte M. MRI T2 Mapping of Dorsal Root Ganglia Reveals Increased T2 Relaxation Time in Classical Fabry Disease. Biomedicines 2025; 13:592. [PMID: 40149569 PMCID: PMC11940213 DOI: 10.3390/biomedicines13030592] [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: 01/31/2025] [Revised: 02/18/2025] [Accepted: 02/25/2025] [Indexed: 03/29/2025] Open
Abstract
Background/Objectives: Fabry disease (FD) is a rare X-linked lysosomal storage disorder characterised by progressive glycolipid accumulation affecting multiple organs, including the peripheral nervous system. The dorsal root ganglia (DRG) play a key role in Fabry-related neuropathy, but non-invasive biomarkers of DRG involvement and their association with overall disease severity remain limited. This study evaluated lumbosacral DRG T2 relaxation time (DRG-T2) in FD patients as a potential imaging biomarker of FD severity. Methods: In a prospective, single-centre study, 80 genetically confirmed FD patients underwent 3T MRI with quantitative T2 mapping of the lumbosacral DRG. DRG-T2 was analysed in relation to sex, genetic subtype and Fabry-specific biomarkers. Results: Results showed that DRG-T2 was higher in patients with classical FD mutations than in those with nonclassical mutations (p = 0.03). Furthermore, DRG-T2 showed a negative correlation with body weight (ρ = -0.31, p = 0.005) and BMI (ρ = -0.32, p = 0.004), while no associations were found with lyso-Gb3 levels or alpha-galactosidase A activity. The inter-rater and test-retest reliability of DRG-T2 were good to excellent (ICC = 0.76 and 0.89, respectively). Conclusions: These results demonstrate DRG-T2 as a marker of neuronal involvement, making it a strong and reliable imaging biomarker of disease severity in FD. However, future studies need to correlate its changes with clinical and histological studies.
Collapse
Affiliation(s)
- Simon Weiner
- Department of Neuroradiology, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Sarah Perleth
- Department of Neuroradiology, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Thomas Kampf
- Department of Neuroradiology, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Kolja Lau
- Department of Internal Medicine, University Hospital Würzburg, 97080 Würzburg, Germany
- Fabry Center for Interdisciplinary Therapy (FAZiT), University Hospital Würzburg, 97080 Würzburg, Germany
| | - Florian Hessenauer
- Department of Neuroradiology, University Hospital Würzburg, 97080 Würzburg, Germany
| | - György Homola
- Department of Neuroradiology, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Peter Nordbeck
- Department of Internal Medicine, University Hospital Würzburg, 97080 Würzburg, Germany
- Fabry Center for Interdisciplinary Therapy (FAZiT), University Hospital Würzburg, 97080 Würzburg, Germany
| | - Nurcan Üçeyler
- Fabry Center for Interdisciplinary Therapy (FAZiT), University Hospital Würzburg, 97080 Würzburg, Germany
- Department of Neurology, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Claudia Sommer
- Fabry Center for Interdisciplinary Therapy (FAZiT), University Hospital Würzburg, 97080 Würzburg, Germany
- Department of Neurology, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Mirko Pham
- Department of Neuroradiology, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Magnus Schindehütte
- Department of Neuroradiology, University Hospital Würzburg, 97080 Würzburg, Germany
- Fabry Center for Interdisciplinary Therapy (FAZiT), University Hospital Würzburg, 97080 Würzburg, Germany
| |
Collapse
|
8
|
Marth AA, Feuerriegel GC, Wanivenhaus F, Nanz D, Sutter R. 7 T Lumbosacral Plexus Neurography: Feasibility and Comparison of Spinal Nerve Visualization With 3 T MRI. Invest Radiol 2025; 60:145-150. [PMID: 39212499 DOI: 10.1097/rli.0000000000001113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
OBJECTIVES 7 T magnetic resonance (MR) imaging can offer superior spatial resolution compared with lower field strengths. However, its use for imaging of the lumbosacral plexus has been constrained by technical challenges and therefore remained relatively unexplored. Therefore, this study investigated the feasibility of 7 T MR neurography by means of comparing the visibility of the spinal nerves and image quality to 3 T MR neurography. MATERIALS AND METHODS In this monocentric, institutional review board-approved, prospective study, 30 healthy subjects underwent acquisition time-matched 7 T MR neurography and 3 T MR neurography of the lumbar spine using a 3-dimensional dual-echo steady-state sequence. Visibility of the nerve root, dorsal root ganglia, and spinal nerve fascicles of L1-S1, along with image artifacts and overall image quality, were compared between the different field strengths by 2 radiologists using 4-point Likert scales (1 = poor, 4 = excellent). Comparisons between field strengths were made using the Wilcoxon signed rank test, and interobserver agreement was assessed. RESULTS 7 T MR neurography enabled significantly improved visualization of the lumbar nerve roots, dorsal root ganglia, and spinal nerve fascicles ( P ≤ 0.002). Compared with 3 T MR neurography, no difference in overall image quality was observed ( P = 0.211), although 7 T MR imaging exhibited significantly increased image artifacts ( P < 0.001). Interobserver agreement (κ) for qualitative measures ranged from 0.71 to 0.88 for 7 T, and from 0.75 to 0.91 for 3 T. CONCLUSIONS 7 T MR neurography allowed for improved visualization of lumbar spinal nerves, whereas overall image quality was comparable to 3 T MR neurography. This supports the feasibility of 7 T MR neurography of the lumbosacral plexus, even though image artifacts at 7 T were significantly increased.
Collapse
Affiliation(s)
- Adrian A Marth
- From the Department of Radiology, Balgrist University Hospital, Zurich, Switzerland (A.A.M., G.C.F., R.S.); Swiss Center for Musculoskeletal Imaging, Balgrist Campus AG, Zurich, Switzerland (A.A.M., D.N.); Department of Orthopedic Surgery, Balgrist University Hospital, Zurich, Switzerland (F.W.); and Medical Faculty, University of Zurich, Zurich, Switzerland (D.N., R.S.)
| | | | | | | | | |
Collapse
|
9
|
Hayashi K, Hsieh TH, Huang YL, Chuang DCC. Using Transcranial Magnetic Nerve Stimulation to Differentiate Motor and Sensory Fascicles in a Mixed Nerve: Experimental Rat Study. J Reconstr Microsurg 2025. [PMID: 39775443 DOI: 10.1055/a-2483-5556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
BACKGROUND Accurately matching the correct fascicles in a ruptured mixed nerve is critical for functional recovery. This study investigates the use of transcranial magnetic stimulation (TMS) to differentiate motor and sensory fascicles in a mixed nerve. METHODS In all 40 rats, the median nerve in the left upper arm was evenly split into three segments. The rats were separated into two groups. In Group A (20 rats), the segment with the highest amplitude during TMS was selected as the motor neurotizer and transferred to the musculocutaneous nerve. In Group B (20 rats), only the medial one-third segment was selected and transferred without using TMS. The results were compared using grooming tests, nerve electrophysiological studies, muscle tetanus contraction force measurements, muscle weight, and axon counts at 16 weeks. RESULTS The grooming test showed that Group A performed significantly better than Group B at 12 and 16 weeks postoperatively. Tetanic muscle contraction force measurements also revealed that Group A had significantly better outcomes than Group B. However, electrophysiological testing, muscle weight, and axon counts showed no significant differences between the two groups. CONCLUSION This study suggests that TMS can be used to distinguish motor fascicles from sensory fascicles in a mixed nerve. It is desirable to apply this technique intraoperatively to differentiate motor and sensory fascicles for appropriate nerve matching and to select the motor fascicles as a motor neurotizer for functioning free muscle innervation in human mixed nerve injury.
Collapse
Affiliation(s)
- Kota Hayashi
- Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital, Taipei, Linkou, Chang Gung University, Taoyuan, Taiwan
| | - Tsung-Hsun Hsieh
- School of Physical Therapy and Graduate Institute of Rehabilitation Science, Chang Gung University, Taoyuan, Taiwan
- Neuroscience Research Center, Chang Gung Memorial Hospital, Linkou, Taoyuan, Taiwan
| | - Yen-Lin Huang
- School of Medicine, National Tsing-Hua University, Hsinchu, Taiwan
- Department of Anatomic Pathology, Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital, Linkou, Taoyuan, Taiwan
| | - David Chwei-Chin Chuang
- Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital, Taipei, Linkou, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
10
|
McIlvried LA, Del Rosario JS, Pullen MY, Wangzhou A, Sheahan TD, Shepherd AJ, Slivicki RA, Lemen JA, Price TJ, Copits BA, Gereau RW. Intrinsic adaptive plasticity in mouse and human sensory neurons. J Gen Physiol 2025; 157:e202313488. [PMID: 39688836 DOI: 10.1085/jgp.202313488] [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: 09/23/2023] [Revised: 06/07/2024] [Accepted: 11/27/2024] [Indexed: 12/18/2024] Open
Abstract
In response to changes in activity induced by environmental cues, neurons in the central nervous system undergo homeostatic plasticity to sustain overall network function during abrupt changes in synaptic strengths. Homeostatic plasticity involves changes in synaptic scaling and regulation of intrinsic excitability. Increases in spontaneous firing and excitability of sensory neurons are evident in some forms of chronic pain in animal models and human patients. However, whether mechanisms of homeostatic plasticity are engaged in sensory neurons of the peripheral nervous system (PNS) is unknown. Here, we show that sustained depolarization (induced by 24-h incubation in 30 mM KCl) induces compensatory changes that decrease the excitability of mouse and human sensory neurons without directly opposing membrane depolarization. Voltage-clamp recordings show that sustained depolarization produces no significant alteration in voltage-gated potassium currents, but a robust reduction in voltage-gated sodium currents, likely contributing to the overall decrease in neuronal excitability. The compensatory decrease in neuronal excitability and reduction in voltage-gated sodium currents reversed completely following a 24-h recovery period in a normal medium. Similar adaptive changes were not observed in response to 24 h of sustained action potential firing induced by optogenetic stimulation at 1 Hz, indicating the need for prolonged depolarization to drive engagement of this adaptive mechanism in sensory neurons. Our findings show that mouse and human sensory neurons are capable of engaging adaptive mechanisms to regulate intrinsic excitability in response to sustained depolarization in a manner similar to that described in neurons in the central nervous system.
Collapse
Affiliation(s)
- Lisa A McIlvried
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA
| | - John Smith Del Rosario
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Melanie Y Pullen
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Andi Wangzhou
- Department of Neuroscience and Center for Advanced Pain Studies, The University of Texas at Dallas, Dallas, TX, USA
| | - Tayler D Sheahan
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Andrew J Shepherd
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Richard A Slivicki
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Theodore J Price
- Department of Neuroscience and Center for Advanced Pain Studies, The University of Texas at Dallas, Dallas, TX, USA
| | - Bryan A Copits
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Robert W Gereau
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neuroscience and Department of Biomedical Engineering, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
11
|
Sierra S, Herz SM, On D, Dozmorov MG, Damaj MI, Gonzalez-Maeso J. Upregulation of the neuropeptide receptor calcitonin receptor-like in the spinal cord via MLL2 in a mouse model of paclitaxel-induced peripheral neuropathy. Mol Pain 2025; 21:17448069251314857. [PMID: 39905828 DOI: 10.1177/17448069251314857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2025] Open
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a prevalent and severe side effect affecting cancer patients undergoing paclitaxel treatment. Growing evidence underscores the pivotal role of calcitonin-related peptide (CGRP) in the development of CIPN. Repeated administration of paclitaxel induces alterations in CGRP release from sensory neurons within the dorsal root ganglia (DRG). The density of the CGRP receptor is most prominent in the dorsal horn of the spinal cord, where it overlaps with the distribution of CGRP. However, the impact of chemotherapy treatment on expression of the CGRP receptor in the spinal cord remains unclear, as well as the potential therapeutic benefits of a CGRP receptor antagonist in an animal model of CIPN. Using a mouse model of paclitaxel-induced mechanical hypersensitivity, we show upregulation of Calcitonin receptor-like receptor (Calcrl) mRNA expression in the spinal cord, an event that occurred in association with upregulation of the H3K4 methyltransferase MLL2. This effect of repeated paclitaxel administration was also linked to an increase in the recruitment of MLL2, thereby enhancing levels of the active mark H3K4me2 at the Calcrl promoter. Furthermore, administration of the CGRP receptor antagonist BIBN4096 mitigated mechanical and cold hypersensitivity in paclitaxel-treated mice. Together, these observations suggest the CGRP receptor in the spinal cord as a potential target for reducing paclitaxel-induced neuropathic pain in animal models.
Collapse
Affiliation(s)
- Salvador Sierra
- Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | - Sara M Herz
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Doan On
- Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Mikhail G Dozmorov
- Department of Biostatistics, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - M Imad Damaj
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Javier Gonzalez-Maeso
- Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| |
Collapse
|
12
|
Jang K, Garraway SM. TrkB Agonist (7,8-DHF)-Induced Responses in Dorsal Root Ganglia Neurons Are Decreased after Spinal Cord Injury: Implication for Peripheral Pain Mechanisms. eNeuro 2025; 12:ENEURO.0219-24.2024. [PMID: 39753357 PMCID: PMC11728855 DOI: 10.1523/eneuro.0219-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 11/08/2024] [Accepted: 12/04/2024] [Indexed: 01/15/2025] Open
Abstract
Brain-derived neurotrophic factor (BDNF) and tropomyosin receptor kinase B (TrkB) are known to contribute to both protective and pronociceptive processes. However, their contribution to neuropathic pain after spinal cord injury (SCI) needs further investigation. In a recent study utilizing TrkBF616A mice, it was shown that systemic pharmacogenetic inhibition of TrkB signaling with 1NM-PP1 (1NMP) immediately after SCI delayed the onset of pain hypersensitivity, implicating maladaptive TrkB signaling in pain after SCI. To examine potential neural mechanisms underlying the behavioral outcome, patch-clamp recording was performed in small-diameter dissociated thoracic (T) dorsal root ganglia (DRG) neurons to evaluate TrkB signaling in uninjured mice and after T10 contusion SCI. Bath-applied 7,8-dihydroxyflavone (7,8-DHF), a selective TrkB agonist, induced a robust inward current in neurons from uninjured mice, which was attenuated by 1NMP treatment. SCI also decreased 7,8-DHF-induced current while increasing the latency to its peak amplitude. Western blot revealed a concomitant decrease in TrkB expression in DRGs adjacent to the spinal lesion. Analyses of cellular and membrane properties showed that SCI increased neuronal excitability, evident by an increase in resting membrane potential and the number of spiking neurons. However, SCI did not increase spontaneous firing in DRG neurons. These results suggest that SCI induced changes in TrkB activation in DRG neurons even though these alterations are likely not contributing to pain hypersensitivity by nociceptor hyperexcitability. Overall, this reveals complex interactions involving TrkB signaling and provides an opportunity to investigate other, presumably peripheral, mechanisms by which TrkB contributes to pain hypersensitivity after SCI.
Collapse
Affiliation(s)
- Kyeongran Jang
- Department of Cell Biology, School of Medicine, Emory University, Atlanta, Georgia 30322
| | - Sandra M Garraway
- Department of Cell Biology, School of Medicine, Emory University, Atlanta, Georgia 30322
| |
Collapse
|
13
|
Tay SH, Pang JKS, Ng W, Ng CY, Khong ZJ, Chong ZS, Soh BS, Ng SY. iPSC-derived human sensory neurons reveal a subset of TRPV1 antagonists as anti-pruritic compounds. Sci Rep 2024; 14:31182. [PMID: 39732807 DOI: 10.1038/s41598-024-82549-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 12/05/2024] [Indexed: 12/30/2024] Open
Abstract
Signaling interplay between the histamine 1 receptor (H1R) and transient receptor potential cation channel subfamily V member 1 (TRPV1) in mediating histaminergic itch has been well-established in mammalian models, but whether this is conserved in humans remains to be confirmed due to the difficulties in obtaining human sensory neurons (SNs) for experimentation. Additionally, previously reported species-specific differences in TRPV1 function indicate that use of human SNs is vital for drug candidate screening to have a higher chance of identifying clinically effective TRPV1 antagonists. In this study, we built a histamine-dependent itch model using peripheral SNs derived from human induced pluripotent stem cells (hiPSC-SNs), which provides an accessible source of human SNs for pre-clinical drug screening. We validated channel functionality using immunostaining, calcium imaging, and multielectrode array (MEA) recordings, and confirmed the interdependence of H1R and TRPV1 signalling in human SNs. We further tested the amenability of our model for pre-clinical studies by screening multiple TRPV1 antagonists in parallel, identifying SB366791 as a potent inhibitor of H1R activation and potential candidate for alleviating histaminergic itch. Notably, some of the results using our model corroborated with efficacy and side effect findings from human clinical trials, underscoring the importance of this species-specific platform. Taken together, our results present a robust in vitro human model for histaminergic itch, which can be used to further interrogate the molecular basis of human SN function as well as screen for TRPV1 activity-modifying compounds for a number of clinical indications.
Collapse
Affiliation(s)
- Shermaine Huiping Tay
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Republic of Singapore
| | - Jeremy Kah Sheng Pang
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Republic of Singapore
| | - Winanto Ng
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Republic of Singapore
| | - Chong Yi Ng
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Republic of Singapore
| | - Zi Jian Khong
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Republic of Singapore
| | - Zheng-Shan Chong
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Republic of Singapore
| | - Boon Seng Soh
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Republic of Singapore
| | - Shi-Yan Ng
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Republic of Singapore.
- Yong Loo Lin School of Medicine (Physiology), National University of Singapore, Singapore, 117456, Republic of Singapore.
- National Neuroscience Institute, Singapore, 308433, Republic of Singapore.
| |
Collapse
|
14
|
Deep RNA sequencing of human dorsal root ganglion neurons reveals somatosensory mechanisms. Nat Neurosci 2024; 27:2276-2277. [PMID: 39506062 DOI: 10.1038/s41593-024-01795-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2024]
|
15
|
Sodmann A, Degenbeck J, Aue A, Schindehütte M, Schlott F, Arampatzi P, Bischler T, Schneider M, Brack A, Monoranu CM, Gräfenhan T, Bohnert M, Pham M, Antoniadis G, Blum R, Rittner HL. Human dorsal root ganglia are either preserved or completely lost after deafferentation by brachial plexus injury. Br J Anaesth 2024; 133:1250-1262. [PMID: 39393999 PMCID: PMC11589459 DOI: 10.1016/j.bja.2024.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 08/05/2024] [Accepted: 09/13/2024] [Indexed: 10/13/2024] Open
Abstract
BACKGROUND Plexus injury results in lifelong suffering from flaccid paralysis, sensory loss, and intractable pain. For this clinical problem, regenerative medicine concepts set high expectations. However, it is largely unknown how dorsal root ganglia (DRG) are affected by accidental deafferentation. METHODS Here, we phenotyped DRG of a clinically and MRI-characterised cohort of 13 patients with plexus injury. Avulsed DRG were collected during reconstructive nerve surgery. For control, we used DRG from forensic autopsy. The cellular composition of the DRG was analysed in histopathological slices with multicolour high-resolution immunohistochemistry, tile microscopy, and deep-learning-based bioimage analysis. We then sequenced the bulk RNA of corresponding DRG slices. RESULTS In about half of the patients we found loss of the typical DRG units consisting of neurones and satellite glial cells. The DRG cells were replaced by mesodermal/connective tissue. In the remaining patients, the cellular units were well preserved. Preoperative plexus MRI neurography was not able to distinguish the two types. Patients with 'neuronal preservation' had less maximum pain than patients with 'neuronal loss'. Arm function improved after nerve reconstruction, but severe pain persisted. Transcriptome analysis of preserved DRGs revealed expression of subtype-specific sensory neurone marker genes, but downregulation of neuronal attributes. Furthermore, they showed signs of ongoing inflammation and connective tissue remodelling. CONCLUSIONS Patients with plexus injury separate into two groups with either neuronal preservation or neuronal loss. The former could benefit from anti-inflammatory therapy. For the latter, studies should explore mechanisms of neuronal loss especially for regenerative approaches. CLINICAL TRIAL REGISTRATION DRKS00017266.
Collapse
Affiliation(s)
- Annemarie Sodmann
- Department of Anesthesiology, Intensive Care, Emergency Medicine and Pain Therapy, Centre for Interdisciplinary Pain Medicine, University Hospital of Würzburg, Würzburg, Germany; Department of Neurology, University Hospital of Würzburg, Würzburg, Germany
| | - Johannes Degenbeck
- Department of Anesthesiology, Intensive Care, Emergency Medicine and Pain Therapy, Centre for Interdisciplinary Pain Medicine, University Hospital of Würzburg, Würzburg, Germany
| | - Annemarie Aue
- Department of Anesthesiology, Intensive Care, Emergency Medicine and Pain Therapy, Centre for Interdisciplinary Pain Medicine, University Hospital of Würzburg, Würzburg, Germany
| | - Magnus Schindehütte
- Institute of Neuroradiology, University Hospital of Würzburg, Würzburg, Germany
| | - Felicitas Schlott
- Department of Anesthesiology, Intensive Care, Emergency Medicine and Pain Therapy, Centre for Interdisciplinary Pain Medicine, University Hospital of Würzburg, Würzburg, Germany; Department of Neurology, University Hospital of Würzburg, Würzburg, Germany
| | - Panagiota Arampatzi
- Core Unit Systems Medicine, University Hospital of Würzburg, Würzburg, Germany
| | - Thorsten Bischler
- Core Unit Systems Medicine, University Hospital of Würzburg, Würzburg, Germany
| | - Max Schneider
- Department of Neurosurgery, Peripheral Nerve Surgery Unit, University of Ulm, Günzburg, Germany
| | - Alexander Brack
- Department of Anesthesiology, Intensive Care, Emergency Medicine and Pain Therapy, Centre for Interdisciplinary Pain Medicine, University Hospital of Würzburg, Würzburg, Germany
| | - Camelia M Monoranu
- Institute of Pathology, Section Neuropathology, University of Würzburg, Würzburg, Germany
| | - Tom Gräfenhan
- Core Unit Systems Medicine, University Hospital of Würzburg, Würzburg, Germany
| | - Michael Bohnert
- Institute of Forensic Medicine, University of Würzburg, Würzburg, Germany
| | - Mirko Pham
- Institute of Neuroradiology, University Hospital of Würzburg, Würzburg, Germany
| | - Gregor Antoniadis
- Department of Neurosurgery, Peripheral Nerve Surgery Unit, University of Ulm, Günzburg, Germany
| | - Robert Blum
- Department of Neurology, University Hospital of Würzburg, Würzburg, Germany.
| | - Heike L Rittner
- Department of Anesthesiology, Intensive Care, Emergency Medicine and Pain Therapy, Centre for Interdisciplinary Pain Medicine, University Hospital of Würzburg, Würzburg, Germany.
| |
Collapse
|
16
|
Yu H, Nagi SS, Usoskin D, Hu Y, Kupari J, Bouchatta O, Yan H, Cranfill SL, Gautam M, Su Y, Lu Y, Wymer J, Glanz M, Albrecht P, Song H, Ming GL, Prouty S, Seykora J, Wu H, Ma M, Marshall A, Rice FL, Li M, Olausson H, Ernfors P, Luo W. Leveraging deep single-soma RNA sequencing to explore the neural basis of human somatosensation. Nat Neurosci 2024; 27:2326-2340. [PMID: 39496796 PMCID: PMC11614738 DOI: 10.1038/s41593-024-01794-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 09/23/2024] [Indexed: 11/06/2024]
Abstract
The versatility of somatosensation arises from heterogeneous dorsal root ganglion (DRG) neurons. However, soma transcriptomes of individual human (h)DRG neurons-critical information to decipher their functions-are lacking due to technical difficulties. In this study, we isolated somata from individual hDRG neurons and conducted deep RNA sequencing (RNA-seq) to detect, on average, over 9,000 unique genes per neuron, and we identified 16 neuronal types. These results were corroborated and validated by spatial transcriptomics and RNAscope in situ hybridization. Cross-species analyses revealed divergence among potential pain-sensing neurons and the likely existence of human-specific neuronal types. Molecular-profile-informed microneurography recordings revealed temperature-sensing properties across human sensory afferent types. In summary, by employing single-soma deep RNA-seq and spatial transcriptomics, we generated an hDRG neuron atlas, which provides insights into human somatosensory physiology and serves as a foundation for translational work.
Collapse
Affiliation(s)
- Huasheng Yu
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Saad S Nagi
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Dmitry Usoskin
- Department of Medical Biochemistry and Biophysics, Division of Molecular Neurobiology, Karolinska Institute, Stockholm, Sweden
| | - Yizhou Hu
- Department of Medical Biochemistry and Biophysics, Division of Molecular Neurobiology, Karolinska Institute, Stockholm, Sweden
| | - Jussi Kupari
- Department of Medical Biochemistry and Biophysics, Division of Molecular Neurobiology, Karolinska Institute, Stockholm, Sweden
| | - Otmane Bouchatta
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Hanying Yan
- Department of Biostatistics in Biostatistics and Epidemiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Suna Li Cranfill
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mayank Gautam
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yijing Su
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - You Lu
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - James Wymer
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Max Glanz
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Phillip Albrecht
- Neuroscience & Pain Research Group, Integrated Tissue Dynamics, LLC, Rensselaer, NY, USA
| | - Hongjun Song
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Guo-Li Ming
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Stephen Prouty
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - John Seykora
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Hao Wu
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Minghong Ma
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Andrew Marshall
- Pain Research Institute, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Frank L Rice
- Neuroscience & Pain Research Group, Integrated Tissue Dynamics, LLC, Rensselaer, NY, USA
| | - Mingyao Li
- Department of Biostatistics in Biostatistics and Epidemiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Håkan Olausson
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden.
| | - Patrik Ernfors
- Department of Medical Biochemistry and Biophysics, Division of Molecular Neurobiology, Karolinska Institute, Stockholm, Sweden.
| | - Wenqin Luo
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
17
|
Antoine JC. Inflammatory sensory neuronopathies. Rev Neurol (Paris) 2024; 180:1037-1046. [PMID: 38472032 DOI: 10.1016/j.neurol.2023.12.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 12/15/2023] [Accepted: 12/22/2023] [Indexed: 03/14/2024]
Abstract
Inflammatory sensory neuronopathies are rare disorders mediated by dysimmune mechanisms targeting sensory neurons in the dorsal root ganglia. They constitute a heterogeneous group of disorders with acute, subacute, or chronic courses, and occur with cancer, systemic autoimmune diseases, notably Sjögren syndrome, and viral infections but a noticeable proportion of them remains isolated. Identifying inflammatory sensory neuronopathies is crucial because they have the potential to be stabilized or even to improve with immunomodulatory or immunosuppressant treatments provided that the treatment is applied at an early stage of the disease, before a definitive degeneration of neurons. Biomarkers, and notably antibodies, are crucial for this early identification, which is the first step to develop therapeutic trials.
Collapse
Affiliation(s)
- J-C Antoine
- Department of Neurology, University Hospital of Saint-Etienne, 42055 Saint-Étienne cedex, France.
| |
Collapse
|
18
|
Kuete CF, Granja-Vazquez R, Truong V, Walsh P, Price T, Biswas S, Dussor G, Pancrazio J, Kolber B. Profiling Human iPSC-Derived Sensory Neurons for Analgesic Drug Screening Using a Multi-Electrode Array. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.18.623405. [PMID: 39605708 PMCID: PMC11601878 DOI: 10.1101/2024.11.18.623405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Chronic pain is a major global health issue, yet effective treatments are limited by poor translation from preclinical studies to humans. To address this, we developed a high-content screening (HCS) platform for analgesic discovery using hiPSC-derived nociceptors. These cells were cultured on multi-well micro-electrode arrays to monitor activity, achieving nearly 100% active electrodes by week two, maintaining stable activity for at least two weeks. After maturation (28 days), we exposed the nociceptors to various drugs, assessing their effects on neuronal activity, with excellent assay performance (Z' values >0.5). Pharmacological tests showed responses to analgesic targets, including ion channels (Nav, Cav, Kv, TRPV1), neurotransmitter receptors (AMPAR, GABA-R), and kinase inhibitors (tyrosine, JAK1/2). Transcriptomic analysis confirmed the presence of these drug targets, although expression levels varied compared to primary human dorsal root ganglion cells. This HCS platform facilitates the rapid discovery of novel analgesics, reducing the risk of preclinical-to-human translation failure. Motivation Chronic pain affects approximately 1.5 billion people worldwide, yet effective treatments remain elusive. A significant barrier to progress in analgesic drug discovery is the limited translation of preclinical findings to human clinical outcomes. Traditional rodent models, although widely used, often fail to accurately predict human responses, while human primary tissues are limited by scarcity, technical difficulties, and ethical concerns. Recent advancements have identified human induced pluripotent stem cell (hiPSC)-derived nociceptors as promising alternatives; however, current differentiation protocols produce cells with inconsistent and physiologically questionable phenotypes.To address these challenges, our study introduces a novel high-content screening (HCS) platform using hiPSC-derived nociceptors cultured on multi-well micro-electrode arrays (MEAs). The "Anatomic" protocol, used to generate these nociceptors, ensures cells with transcriptomic profiles closely matching human primary sensory neurons. Our platform achieves nearly 100% active electrode yield within two weeks and demonstrates sustained, stable activity over time. Additionally, robust Z' factor analysis (exceeding 0.5) confirms the platform's reliability, while pharmacological validation establishes the functional expression of critical analgesic targets. This innovative approach improves both the efficiency and clinical relevance of analgesic drug screening, potentially bridging the translational gap between preclinical studies and human clinical trials, and offering new hope for effective pain management.
Collapse
|
19
|
Handlin LJ, Macchi NL, Dumaire NLA, Salih L, Lessie EN, McCommis KS, Moutal A, Dai G. Membrane lipid nanodomains modulate HCN pacemaker channels in nociceptor DRG neurons. Nat Commun 2024; 15:9898. [PMID: 39548079 PMCID: PMC11568329 DOI: 10.1038/s41467-024-54053-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 10/29/2024] [Indexed: 11/17/2024] Open
Abstract
Cell membranes consist of heterogeneous lipid nanodomains that influence key cellular processes. Using FRET-based fluorescent assays and fluorescence lifetime imaging microscopy (FLIM), we find that the dimension of cholesterol-enriched ordered membrane domains (OMD) varies considerably, depending on specific cell types. Particularly, nociceptor dorsal root ganglion (DRG) neurons exhibit large OMDs. Disruption of OMDs potentiated action potential firing in nociceptor DRG neurons and facilitated the opening of native hyperpolarization-activated cyclic nucleotide-gated (HCN) pacemaker channels. This increased neuronal firing is partially due to an increased open probability and altered gating kinetics of HCN channels. The gating effect on HCN channels is likely due to a direct modulation of their voltage sensors by OMDs. In animal models of neuropathic pain, we observe reduced OMD size and a loss of HCN channel localization within OMDs. Additionally, cholesterol supplementation inhibited HCN channels and reduced neuronal hyperexcitability in pain models. These findings suggest that disturbances in lipid nanodomains play a critical role in regulating HCN channels within nociceptor DRG neurons, influencing pain modulation.
Collapse
Affiliation(s)
- Lucas J Handlin
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, Saint Louis, USA
| | - Natalie L Macchi
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, Saint Louis, USA
| | - Nicolas L A Dumaire
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, USA
| | - Lyuba Salih
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, USA
| | - Erin N Lessie
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, Saint Louis, USA
| | - Kyle S McCommis
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, Saint Louis, USA
| | - Aubin Moutal
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, USA
| | - Gucan Dai
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, Saint Louis, USA.
| |
Collapse
|
20
|
Tarasiuk O, Invernizzi C, Alberti P. In vitro neurotoxicity testing: lessons from chemotherapy-induced peripheral neurotoxicity. Expert Opin Drug Metab Toxicol 2024; 20:1037-1052. [PMID: 39246127 DOI: 10.1080/17425255.2024.2401584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 08/30/2024] [Accepted: 09/03/2024] [Indexed: 09/10/2024]
Abstract
INTRODUCTION Chemotherapy induced peripheral neurotoxicity (CIPN) is a long-lasting, or even permanent, late toxicity caused by largely used anticancer drugs. CIPN affects a growing population of cancer survivors and diminishes their quality of life since there is no curative/preventive treatment. Among several reasons for this unmet clinical need, there is an incomplete knowledge on mechanisms leading to CIPN. Therefore, bench side research is still greatly needed: in vitro studies are pivotal to both evaluate neurotoxicity mechanisms and potential neuroprotection strategies. AREAS COVERED Advantages and disadvantages of in vitro approaches are addressed with respect to their applicability to the CIPN field. Different cell cultures and techniques to assess neurotoxicity/neuroprotection are described. PubMed search-string: (chemotherapy-induced) AND (((neuropathy) OR neurotoxicity) OR neuropathic pain) AND (in vitro) AND (((((model) OR SH-SY5Y) OR PC12) OR iPSC) OR DRG neurons); (chemotherapy-induced) AND (((neuropathy) OR neurotoxicity) OR neuropathic pain) AND (model) AND (((neurite elongation) OR cell viability) OR morphology). No articles published before 1990 were selected. EXPERT OPINION CIPN is an ideal experimental setting to test axonal damage and, in general, peripheral nervous system mechanisms of disease and neuroprotection. Therefore, starting from robust preclinical data in this field, potentially, relevant biological rationale can be transferred to other human spontaneous diseases of the peripheral nervous system.
Collapse
Affiliation(s)
- Olga Tarasiuk
- Experimental Neurology Unit, School of Medicine and Surgery, Monza, Italy
- NeuroMI (Milan Center for Neuroscience), Milan, Italy
| | - Chiara Invernizzi
- Experimental Neurology Unit, School of Medicine and Surgery, Monza, Italy
- Neuroscience, School of Medicine and Surgery, Monza, Italy
| | - Paola Alberti
- Experimental Neurology Unit, School of Medicine and Surgery, Monza, Italy
- NeuroMI (Milan Center for Neuroscience), Milan, Italy
- Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| |
Collapse
|
21
|
Chen J, Li Y, Wang F, Gu Y, Zhou X, Liu W, Liu X, Wang Y, Ye Q. Fentanyl induces analgesic effect through miR-381-3p/TRPM7 when combined with bupivacaine in subarachnoid injection. Eur J Pharm Sci 2024; 202:106888. [PMID: 39191357 DOI: 10.1016/j.ejps.2024.106888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 07/31/2024] [Accepted: 08/23/2024] [Indexed: 08/29/2024]
Abstract
Fentanyl combined with bupivacaine in subarachnoid anesthesia exerts a strong synergistic analgesic effect, extending the duration of analgesia. However, the mechanism of enhanced analgesic effect of fentanyl remains elusive. The present study investigated the potential mechanism of the analgesic effect of fentanyl when combined with bupivacaine. The subarachnoid injection (SI) rat model was employed, and SI of fentanyl or/and bupivacaine was used to investigate their analgesic effect. Dorsal root ganglion (DRG)' RNA sequencing (RNA-Seq) and bioinformatics analysis were performed to evaluate the downstream mechanisms of MicroRNAs (miRNAs). Further validation tests included RT-PCR, Western blot, and immunofluorescence. A single SI of fentanyl or bupivacaine decreased the positive responses to stimulation when used alone or in combination. RNA-seq results revealed that miR-381-3p played a role in the fentanyl-driven promotion of analgesia. Bioinformatics analysis and dual-luciferase reporter identified TRPM7 as a direct downstream target gene of miR-381-3p. In vitro, overexpression of miR-381-3p could further block fentanyl-induced expression of TRPM7, p-ERK1/2, CGRP, and SP. In addition, antagomir-381-3p reversed the inhibitory effect of fentanyl on the expression of TRPM7, p-ERK1/2, CGRP, and SP, in vivo; however, TRPM7 siRNA rescued the effect of antagomir-381-3p. In conclusion, fentanyl inhibits p-ERK by targeting TRPM7 via miR-381-3p, lowering the production of CGRP and SP, and ultimately inducing analgesic effects.
Collapse
Affiliation(s)
- Jiaxin Chen
- Ningxia Medical University, Yinchuan 750004, Ningxia, China
| | - Yan Li
- Ningxia Medical University, Yinchuan 750004, Ningxia, China
| | - Fa Wang
- Ningxia Medical University, Yinchuan 750004, Ningxia, China
| | - Yinghua Gu
- Ningxia Medical University, Yinchuan 750004, Ningxia, China
| | - Xiaohong Zhou
- Ningxia Medical University, Yinchuan 750004, Ningxia, China
| | - Wenxun Liu
- Department of Anesthesiology, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan 750002, Ningxia, China
| | - Xin Liu
- Ningxia Medical University, Yinchuan 750004, Ningxia, China
| | - Yun Wang
- Department of Anesthesiology, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan 750002, Ningxia, China
| | - Qingshan Ye
- Department of Anesthesiology, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan 750002, Ningxia, China.
| |
Collapse
|
22
|
Hall BE, Mazhar K, Macdonald E, Cassidy M, Doty M, Judkins C, Terse A, Shiers S, Tadros S, Yun S, Burton MD, Price TJ, Kulkarni AB. Transcriptome analysis of rheumatoid arthritis uncovers genes linked to inflammation-induced pain. Sci Rep 2024; 14:25893. [PMID: 39472517 PMCID: PMC11522505 DOI: 10.1038/s41598-024-77212-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 10/21/2024] [Indexed: 11/02/2024] Open
Abstract
Autoimmune diseases such as rheumatoid arthritis (RA) can promote states of chronic inflammation with accompanying tissue destruction and pain. RA can cause inflammatory synovitis in peripheral joints, particularly within the hands and feet, but can also sometimes trigger temporomandibular joint (TMJ) arthralgia. To better understand the effects of ongoing inflammation-induced pain signaling, dorsal root ganglia (DRGs) were acquired from individuals with RA for transcriptomic study. We conducted RNA sequencing from the L5 DRGs because it contains the soma of the sensory neurons that innervate the affected joints in the foot. DRGs from 5 RA patients were compared with 9 non-arthritic controls. RNA-seq of L5 DRGs identified 128 differentially expressed genes (DEGs) that were dysregulated in the RA subjects as compared to the non-arthritic controls. The DRG resides outside the blood brain barrier and, as such, our initial transcriptome analysis detected signs of an autoimmune disorder including the upregulated expression of immunoglobulins and other immunologically related genes within the DRGs of the RA donors. Additionally, we saw the upregulation in genes implicated in neurogenesis that could promote pain hypersensitivity. Overall, our DRG analysis suggests that there are upregulated inflammatory and pain signaling pathways that can contribute to chronic pain in RA.
Collapse
Affiliation(s)
- Bradford E Hall
- Functional Genomics Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Room 130, Bethesda, MD, 20892, USA
| | - Khadijah Mazhar
- Department of Neuroscience and Center for Advanced Pain Studies, The University of Texas at Dallas, Dallas, TX, 75080, USA
| | - Emma Macdonald
- Functional Genomics Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Room 130, Bethesda, MD, 20892, USA
- NIH Graduate Partnerships Program, Brown University, Providence, RI, 02912, USA
| | - Margaret Cassidy
- Functional Genomics Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Room 130, Bethesda, MD, 20892, USA
- U. Penn, Philadelphia, PA, 19104, USA
| | - Megan Doty
- Functional Genomics Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Room 130, Bethesda, MD, 20892, USA
- , Dartmouth, Hanover, NH, 03755, USA
| | - Christian Judkins
- Functional Genomics Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Room 130, Bethesda, MD, 20892, USA
- Millipore Sigma, Rockville, MD, 20850, USA
| | - Anita Terse
- Functional Genomics Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Room 130, Bethesda, MD, 20892, USA
| | - Stephanie Shiers
- Department of Neuroscience and Center for Advanced Pain Studies, The University of Texas at Dallas, Dallas, TX, 75080, USA
| | - Saber Tadros
- Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Sijung Yun
- Predictiv Care, Inc, Mountain View, CA, 94040, USA
| | - Michael D Burton
- Neuroimmunology and Behavior Laboratory, Department of Neuroscience, Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX, USA
| | - Theodore J Price
- Department of Neuroscience and Center for Advanced Pain Studies, The University of Texas at Dallas, Dallas, TX, 75080, USA
| | - Ashok B Kulkarni
- Functional Genomics Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Room 130, Bethesda, MD, 20892, USA.
| |
Collapse
|
23
|
Brown RI, Barber HM, Kucenas S. Satellite glial cell manipulation prior to axotomy enhances developing dorsal root ganglion central branch regrowth into the spinal cord. Glia 2024; 72:1766-1784. [PMID: 39141572 PMCID: PMC11325082 DOI: 10.1002/glia.24581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 05/30/2024] [Accepted: 06/02/2024] [Indexed: 08/16/2024]
Abstract
The central and peripheral nervous systems (CNS and PNS, respectively) exhibit remarkable diversity in the capacity to regenerate following neuronal injury with PNS injuries being much more likely to regenerate than those that occur in the CNS. Glial responses to damage greatly influence the likelihood of regeneration by either promoting or inhibiting axonal regrowth over time. However, despite our understanding of how some glial lineages participate in nerve degeneration and regeneration, less is known about the contributions of peripheral satellite glial cells (SGC) to regeneration failure following central axon branch injury of dorsal root ganglia (DRG) sensory neurons. Here, using in vivo, time-lapse imaging in larval zebrafish coupled with laser axotomy, we investigate the role of SGCs in axonal regeneration. In our studies we show that SGCs respond to injury by relocating their nuclei to the injury site during the same period that DRG neurons produce new central branch neurites. Laser ablation of SGCs prior to axon injury results in more neurite growth attempts and ultimately a higher rate of successful central axon regrowth, implicating SGCs as inhibitors of regeneration. We also demonstrate that this SGC response is mediated in part by ErbB signaling, as chemical inhibition of this receptor results in reduced SGC motility and enhanced central axon regrowth. These findings provide new insights into SGC-neuron interactions under injury conditions and how these interactions influence nervous system repair.
Collapse
Affiliation(s)
- Robin I Brown
- Department of Biology, University of Virginia, Charlottesville, Virginia, USA
- Program in Fundamental Neuroscience, University of Virginia, Charlottesville, Virginia, USA
| | - Heather M Barber
- Program in Fundamental Neuroscience, University of Virginia, Charlottesville, Virginia, USA
- Cell & Developmental Biology Graduate Program, University of Virginia, Charlottesville, Virginia, USA
| | - Sarah Kucenas
- Department of Biology, University of Virginia, Charlottesville, Virginia, USA
- Program in Fundamental Neuroscience, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
24
|
Haberberger RV, Matusica D, Shiers S, Sankaranarayanan I, Price TJ. Transcriptomic and histological characterization of telocytes in the human dorsal root ganglion. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.24.614693. [PMID: 39386553 PMCID: PMC11463542 DOI: 10.1101/2024.09.24.614693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Telocytes are interstitial cells with long processes that cover distances in tissues and likely coordinate interacts with other cell types. Though present in central and peripheral neuronal tissues, their role remains unclear. Dorsal root ganglia (DRG) house pseudounipolar afferent neurons responsible for signals such as temperature, proprioception and nociception. This study aimed to investigate the presence and function of telocytes in human DRG by investigating their transcriptional profile, location and ultrastructure. Sequencing data revealed CD34 and PDGFRA expressing cells comprise roughly 1.5-3% of DRG cells. Combined expression of CD34 and PDGFRA is a putative marker gene set for telocytes. Further analysis identified nine subclusters with enriched cluster-specific genes. KEGG and GO pathway analysis suggested vascular, immune and connective tissue associated putative telocyte subtypes. Over 3000 potential receptor-ligand interactions between sensory neurons and these CD34 and PDGFRA expressing putative telocytes were identified using a ligand-receptors interactome platform. Immunohisto-chemistry showed CD34+ telocytes in the endoneural space of DRGs, next to neuron-satellite complexes, in perivascular spaces and in the endoneural space between nerve fibre bundles, consistent with pathway analysis. Transmission electron microscopy (TEM) confirmed their location identifying characteristic elongated nucleus, long and thin telopods containing vesicles, surrounded by a basal lamina. This is the first study that provides gene expression analysis of telocytes in complex human tissue such as the DRG, highlighting functional differences based on tissue location with no significant ultrastructural variation.
Collapse
Affiliation(s)
- Rainer V Haberberger
- Department of Anatomy and Pathology, School of Biomedicine, The University of Adelaide, Adelaide, Australia
- 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
| | - Stephanie Shiers
- Department of Neuroscience, Center for Advanced Pain Studies, The University of Texas at Dallas. Richardson, TX
| | - Ishwarya Sankaranarayanan
- Department of Neuroscience, Center for Advanced Pain Studies, The University of Texas at Dallas. Richardson, TX
| | - Theodore J Price
- Department of Neuroscience, Center for Advanced Pain Studies, The University of Texas at Dallas. Richardson, TX
| |
Collapse
|
25
|
Ege E, Briggi D, Vu P, Cheng J, Lin F, Xu J. Targeting dorsal root ganglia for chemotherapy-induced peripheral neuropathy: from bench to bedside. Ther Adv Neurol Disord 2024; 17:17562864241252718. [PMID: 39318973 PMCID: PMC11421407 DOI: 10.1177/17562864241252718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/26/2024] Open
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a debilitating condition affecting an increasing number of cancer survivors worldwide. However, insights into its pathophysiology and availability of effective therapies remain lacking. Dorsal root ganglia (DRG) have been studied as a key component of chemotherapeutic drug toxicity and a potential therapeutic target for CIPN treatment. This comprehensive review aims to synthesize, summarize, and correlate the results of both preclinical and clinical studies relevant to the pathophysiology and management of CIPN in relation to the DRG. Design: Review. A thorough literature search was conducted using the terms 'dorsal root ganglion' and 'chemotherapy-induced peripheral neuropathy', along with appropriate variations. Searched databases included PubMed, EMBASE, Medline, Cochrane Library, Wiley Library, and Web of Science. Inclusion criteria targeted all English language, peer-reviewed original research from the inception of these databases to the present year. Review articles, book chapters, and other nonoriginal publications were excluded. Of 134 relevant studies identified, the majority were preclinical studies elucidating how various chemotherapeutic agents, especially taxanes, disrupt neurotransmission, inflammatory processes, and apoptotic pathways within sensory neurons of DRG. Not only do these effects correlate with the presentation of CIPN, but their disruption has also been shown to reduce CIPN symptoms in preclinical models. However, clinical studies addressing DRG interventions are very limited in number and scope at this time. These results reveal various pathways within DRG that may be effective targets for CIPN treatment. While limited, clinical studies do offer promise in the utility of DRG neuromodulation in managing painful CIPN. In the future, clinical trials are needed to assess interventions aimed at these neuronal and nonneuronal pathological targets to better treat this complex condition.
Collapse
Affiliation(s)
- Eliana Ege
- Department of Physical Medicine and Rehabilitation, Baylor College of Medicine, Houston, TX, USA
| | - Daniel Briggi
- Department of Physical Medicine and Rehabilitation, Baylor College of Medicine, Houston, TX, USA
| | - Peter Vu
- Department of Physical Medicine and Rehabilitation, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Jianguo Cheng
- Department of Pain Management, Cleveland Clinic, Cleveland, OH, USA
- Department of Neuroscience, Cleveland Clinic, Cleveland, OH, USA
| | - Feng Lin
- Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, OH, USA
| | - Jijun Xu
- Department of Pain Management and Inflammation and Immunity, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| |
Collapse
|
26
|
Jiang Z, Luo W, Long Z, Chen J. The role of TRPV1 in chronic prostatitis: a review. Front Pharmacol 2024; 15:1459683. [PMID: 39364048 PMCID: PMC11446813 DOI: 10.3389/fphar.2024.1459683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 09/10/2024] [Indexed: 10/05/2024] Open
Abstract
Chronic prostatitis is a prevalent male urinary system disorder characterized by pelvic discomfort or pain, bladder dysfunction, sexual dysfunction, and infertility. Pain and lower urinary tract symptoms (LUTS) are the most common symptoms, significantly impacting patients' quality of life and driving them to seek medical attention. Transient receptor potential vanilloid subtype 1 (TRPV1) is a non-selective calcium ion-dependent cation channel in the TRPV channel family that is widely distributed in neural tissue and plays a role in signal transmission. In this review, we provide a comprehensive overview of the current understanding of the role of TRPV1 in chronic prostatitis. The discussion focuses on the connection between TRPV1 and prostatitis pain and LUTS, and highlights the potential for targeting this channel in the development of novel treatment strategies.
Collapse
Affiliation(s)
- Zhipeng Jiang
- Third Affiliated Hospital of Zunyi Medical University (First People's Hospital of Zunyi), Zunyi, China
| | - Wen Luo
- Third Affiliated Hospital of Zunyi Medical University (First People's Hospital of Zunyi), Zunyi, China
| | - Zongmin Long
- Third Affiliated Hospital of Zunyi Medical University (First People's Hospital of Zunyi), Zunyi, China
| | - Jie Chen
- Kweichow Moutai Hospital, Zunyi, China
| |
Collapse
|
27
|
Handlin LJ, Macchi NL, Dumaire NLA, Salih L, Lessie EN, McCommis KS, Moutal A, Dai G. Membrane Lipid Nanodomains Modulate HCN Pacemaker Channels in Nociceptor DRG Neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.02.556056. [PMID: 37732182 PMCID: PMC10508734 DOI: 10.1101/2023.09.02.556056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
Cell membranes consist of heterogeneous lipid nanodomains that influence key cellular processes. Using FRET-based fluorescent assays and fluorescence lifetime imaging microscopy (FLIM), we found that the dimension of cholesterol-enriched ordered membrane domains (OMD) varies considerably, depending on specific cell types. Particularly, nociceptor dorsal root ganglion (DRG) neurons exhibit large OMDs. Disruption of OMDs potentiated action potential firing in nociceptor DRG neurons and facilitated the opening of native hyperpolarization-activated cyclic nucleotide-gated (HCN) pacemaker channels. This increased neuronal firing is partially due to an increased open probability and altered gating kinetics of HCN channels. The gating effect on HCN channels was likely due to a direct modulation of their voltage sensors by OMDs. In animal models of neuropathic pain, we observed reduced OMD size and a loss of HCN channel localization within OMDs. Additionally, cholesterol supplementation inhibited HCN channels and reduced neuronal hyperexcitability in pain models. These findings suggest that disturbances in lipid nanodomains play a critical role in regulating HCN channels within nociceptor DRG neurons, influencing pain modulation.
Collapse
|
28
|
St Clair-Glover M, Finol-Urdaneta RK, Maddock M, Wallace E, Miellet S, Wallace G, Yue Z, Dottori M. Efficient fabrication of 3D bioprinted functional sensory neurons using an inducible Neurogenin-2 human pluripotent stem cell line. Biofabrication 2024; 16:045022. [PMID: 39084624 DOI: 10.1088/1758-5090/ad69c4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 07/31/2024] [Indexed: 08/02/2024]
Abstract
Three-dimensional (3D) tissue models have gained recognition for their improved ability to mimic the native cell microenvironment compared to traditional two-dimensional models. This progress has been driven by advances in tissue-engineering technologies such as 3D bioprinting, a promising method for fabricating biomimetic living tissues. While bioprinting has succeeded in generating various tissues to date, creating neural tissue models remains challenging. In this context, we present an accelerated approach to fabricate 3D sensory neuron (SN) structures using a transgenic human pluripotent stem cell (hPSC)-line that contains an inducible Neurogenin-2 (NGN2) expression cassette. The NGN2 hPSC line was first differentiated to neural crest cell (NCC) progenitors, then incorporated into a cytocompatible gelatin methacryloyl-based bioink for 3D bioprinting. Upregulated NGN2 expression in the bioprinted NCCs resulted in induced SN (iSN) populations that exhibited specific cell markers, with 3D analysis revealing widespread neurite outgrowth through the scaffold volume. Calcium imaging demonstrated functional activity of iSNs, including membrane excitability properties and voltage-gated sodium channel (NaV) activity. This efficient approach to generate 3D bioprinted iSN structures streamlines the development of neural tissue models, useful for the study of neurodevelopment and disease states and offering translational potential.
Collapse
Affiliation(s)
- Mitchell St Clair-Glover
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW 2522, Australia
- School of Medical, Indigenous, and Health Sciences, Molecular Horizons, University of Wollongong, Wollongong, NSW 2522, Australia
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, University of Wollongong, NSW 2522, Australia
| | - Rocio K Finol-Urdaneta
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW 2522, Australia
- School of Medical, Indigenous, and Health Sciences, Molecular Horizons, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Marnie Maddock
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW 2522, Australia
- School of Medical, Indigenous, and Health Sciences, Molecular Horizons, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Eileen Wallace
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW 2522, Australia
- School of Medical, Indigenous, and Health Sciences, Molecular Horizons, University of Wollongong, Wollongong, NSW 2522, Australia
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, University of Wollongong, NSW 2522, Australia
| | - Sara Miellet
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW 2522, Australia
- School of Medical, Indigenous, and Health Sciences, Molecular Horizons, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Gordon Wallace
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, University of Wollongong, NSW 2522, Australia
| | - Zhilian Yue
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, University of Wollongong, NSW 2522, Australia
| | - Mirella Dottori
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW 2522, Australia
- School of Medical, Indigenous, and Health Sciences, Molecular Horizons, University of Wollongong, Wollongong, NSW 2522, Australia
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, University of Wollongong, NSW 2522, Australia
| |
Collapse
|
29
|
Zedde M, Romani I, Scaravilli A, Cocozza S, Trojano L, Ragno M, Rifino N, Bersano A, Gerevini S, Pantoni L, Valzania F, Pascarella R. Expanding the Neurological Phenotype of Anderson-Fabry Disease: Proof of Concept for an Extrapyramidal Neurodegenerative Pattern and Comparison with Monogenic Vascular Parkinsonism. Cells 2024; 13:1131. [PMID: 38994983 PMCID: PMC11240674 DOI: 10.3390/cells13131131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 06/18/2024] [Accepted: 06/25/2024] [Indexed: 07/13/2024] Open
Abstract
Anderson-Fabry disease (AFD) is a genetic sphingolipidosis involving virtually the entire body. Among its manifestation, the involvement of the central and peripheral nervous system is frequent. In recent decades, it has become evident that, besides cerebrovascular damage, a pure neuronal phenotype of AFD exists in the central nervous system, which is supported by clinical, pathological, and neuroimaging data. This neurodegenerative phenotype is often clinically characterized by an extrapyramidal component similar to the one seen in prodromal Parkinson's disease (PD). We analyzed the biological, clinical pathological, and neuroimaging data supporting this phenotype recently proposed in the literature. Moreover, we compared the neurodegenerative PD phenotype of AFD with a classical monogenic vascular disease responsible for vascular parkinsonism and cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL). A substantial difference in the clinical and neuroimaging features of neurodegenerative and vascular parkinsonism phenotypes emerged, with AFD being potentially responsible for both forms of the extrapyramidal involvement, and CADASIL mainly associated with the vascular subtype. The available studies share some limitations regarding both patients' information and neurological and genetic investigations. Further studies are needed to clarify the potential association between AFD and extrapyramidal manifestations.
Collapse
Affiliation(s)
- Marialuisa Zedde
- Neurology Unit, Stroke Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Viale Risorgimento 80, 42123 Reggio Emilia, Italy;
| | - Ilaria Romani
- Department of Neurosciences, Psychology, Pharmacology and Child Health, University of Florence, 50139 Firenze, Italy;
| | - Alessandra Scaravilli
- Department of Advanced Biomedical Sciences, University of Naples “Federico II”, 80133 Napoli, Italy; (A.S.); (S.C.)
| | - Sirio Cocozza
- Department of Advanced Biomedical Sciences, University of Naples “Federico II”, 80133 Napoli, Italy; (A.S.); (S.C.)
| | - Luigi Trojano
- Dipartimento di Psicologia, Università della Campania ‘Luigi Vanvitelli’, viale Ellittico 31, 81100 Caserta, Italy;
| | - Michele Ragno
- Centro Medico Salute 23, Via O. Licini 5, 63066 Grottammare (AP), Italy;
| | - Nicola Rifino
- Cerebrovascular Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milano, Italy; (N.R.); (A.B.)
| | - Anna Bersano
- Cerebrovascular Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milano, Italy; (N.R.); (A.B.)
| | - Simonetta Gerevini
- Head Diagnostic Dept and Neuroradiology Unit, ASST Papa Giovanni XXIII, 24127 Bergamo, Italy;
| | - Leonardo Pantoni
- Neuroscience Research Center, Department of Biomedical and Clinical Science, University of Milan, 20122 Milano, Italy;
| | - Franco Valzania
- Neurology Unit, Stroke Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Viale Risorgimento 80, 42123 Reggio Emilia, Italy;
| | - Rosario Pascarella
- Neuroradiology Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Viale Risorgimento 80, 42123 Reggio Emilia, Italy;
| |
Collapse
|
30
|
Cuendias P, Vega JA, García-Suárez O, Suazo I, Cobo R, García-Piqueras J, García-Mesa Y. Axonal and Glial PIEZO1 and PIEZO2 Immunoreactivity in Human Clitoral Krause's Corpuscles. Int J Mol Sci 2024; 25:6722. [PMID: 38928429 PMCID: PMC11203881 DOI: 10.3390/ijms25126722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 06/13/2024] [Accepted: 06/17/2024] [Indexed: 06/28/2024] Open
Abstract
Krause's corpuscles are typical of cutaneous mucous epithelia, like the lip vermillion or the glans clitoridis, and are associated with rapidly adapting low-threshold mechanoreceptors involved in gentle touch or vibration. PIEZO1 and PIEZO2 are transmembrane mechano-gated proteins that form a part of the cationic ion channels required for mechanosensitivity in mammalian cells. They are involved in somatosensitivity, especially in the different qualities of touch, but also in pain and proprioception. In the present study, immunohistochemistry and immunofluorescence were used to analyze the occurrence and cellular location of PIEZO1 and PIEZO2 in human clitoral Krause's corpuscles. Both PIEZO1 and PIEZO2 were detected in Krause's corpuscles in both the axon and the terminal glial cells. The presence of PIEZOs in the terminal glial cells of Kraus's corpuscles is reported here for the first time. Based on the distribution of PIEZO1 and PIEZO2, it may be assumed they could be involved in mechanical stimuli, sexual behavior, and sexual pleasure.
Collapse
Affiliation(s)
- Patricia Cuendias
- Grupo de Investigación SINPOS, Departamento de Morfología y Biología Celular, Universidad de Oviedo, 33006 Oviedo, Spain; (P.C.); (J.A.V.); (O.G.-S.); (J.G.-P.)
| | - José A. Vega
- Grupo de Investigación SINPOS, Departamento de Morfología y Biología Celular, Universidad de Oviedo, 33006 Oviedo, Spain; (P.C.); (J.A.V.); (O.G.-S.); (J.G.-P.)
- Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Providencia, Santiago de Chile 4810010, Chile;
| | - Olivia García-Suárez
- Grupo de Investigación SINPOS, Departamento de Morfología y Biología Celular, Universidad de Oviedo, 33006 Oviedo, Spain; (P.C.); (J.A.V.); (O.G.-S.); (J.G.-P.)
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Iván Suazo
- Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Providencia, Santiago de Chile 4810010, Chile;
| | - Ramón Cobo
- Servicio de Otorrinolaringología, Hospital Universitario “Marqués de Valdecilla”, 39008 Santander, Spain;
| | - Jorge García-Piqueras
- Grupo de Investigación SINPOS, Departamento de Morfología y Biología Celular, Universidad de Oviedo, 33006 Oviedo, Spain; (P.C.); (J.A.V.); (O.G.-S.); (J.G.-P.)
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Departamento de Anatomía, Histología y Neurociencia, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Yolanda García-Mesa
- Grupo de Investigación SINPOS, Departamento de Morfología y Biología Celular, Universidad de Oviedo, 33006 Oviedo, Spain; (P.C.); (J.A.V.); (O.G.-S.); (J.G.-P.)
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| |
Collapse
|
31
|
Schindehütte M, Weiner S, Klug K, Hölzli L, Nauroth-Kreß C, Hessenauer F, Kampf T, Homola GA, Nordbeck P, Wanner C, Sommer C, Üçeyler N, Pham M. Dorsal root ganglion magnetic resonance imaging biomarker correlations with pain in Fabry disease. Brain Commun 2024; 6:fcae155. [PMID: 38751382 PMCID: PMC11095551 DOI: 10.1093/braincomms/fcae155] [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: 08/25/2023] [Revised: 01/20/2024] [Accepted: 04/29/2024] [Indexed: 05/18/2024] Open
Abstract
Fabry disease is a rare monogenetic, X-linked lysosomal storage disorder with neuropathic pain as one characteristic symptom. Impairment of the enzyme alpha-galactosidase A leads to an accumulation of globotriaosylceramide in the dorsal root ganglia. Here, we investigate novel dorsal root ganglia MR imaging biomarkers and their association with Fabry genotype and pain phenotype. In this prospective study, 89 Fabry patients were examined using a standardized 3 T MRI protocol of the dorsal root ganglia. Fabry pain was assessed through a validated Fabry pain questionnaire. The genotype was determined by diagnostic sequencing of the alpha-galactosidase A gene. MR imaging end-points were dorsal root ganglia volume by voxel-wise morphometric analysis and dorsal root ganglia T2 signal. Reference groups included 55 healthy subjects and Fabry patients of different genotype categories without Fabry pain. In patients with Fabry pain, T2 signal of the dorsal root ganglia was increased by +39.2% compared to healthy controls (P = 0.001) and by +29.4% compared to painless Fabry disease (P = 0.017). This effect was pronounced in hemizygous males (+40.7% compared to healthy; P = 0.008 and +29.1% compared to painless; P = 0.032) and was consistently observed across the genotype spectrum of nonsense (+38.1% compared to healthy, P < 0.001) and missense mutations (+39.2% compared to healthy; P = 0.009). T2 signal of dorsal root ganglia and globotriaosylsphingosine levels were the only independent predictors of Fabry pain (P = 0.047; P = 0.002). Volume of dorsal root ganglia was enlarged by +46.0% in Fabry males in the nonsense compared to missense genotype category (P = 0.005) and by +34.5% compared to healthy controls (P = 0.034). In painful Fabry disease, MRI T2 signal of dorsal root ganglia is increased across different genotypes. Dorsal root ganglion MRI T2 signal as a novel in vivo imaging biomarker may help to better understand whether Fabry pain is modulated or even caused by dorsal root ganglion pathology.
Collapse
Affiliation(s)
- Magnus Schindehütte
- Department of Neuroradiology, University Hospital Würzburg, Würzburg 97080, Germany
| | - Simon Weiner
- Department of Neuroradiology, University Hospital Würzburg, Würzburg 97080, Germany
| | - Katharina Klug
- Department of Neurology, University Hospital Würzburg, Würzburg 97080, Germany
| | - Lea Hölzli
- Department of Neuroradiology, University Hospital Würzburg, Würzburg 97080, Germany
| | | | - Florian Hessenauer
- Department of Neuroradiology, University Hospital Würzburg, Würzburg 97080, Germany
| | - Thomas Kampf
- Department of Neuroradiology, University Hospital Würzburg, Würzburg 97080, Germany
| | - György A Homola
- Department of Neuroradiology, University Hospital Würzburg, Würzburg 97080, Germany
| | - Peter Nordbeck
- Department of Internal Medicine, University Hospital Würzburg, Würzburg 97080, Germany
| | - Christoph Wanner
- Department of Internal Medicine, University Hospital Würzburg, Würzburg 97080, Germany
| | - Claudia Sommer
- Department of Neurology, University Hospital Würzburg, Würzburg 97080, Germany
| | - Nurcan Üçeyler
- Department of Neurology, University Hospital Würzburg, Würzburg 97080, Germany
| | - Mirko Pham
- Department of Neuroradiology, University Hospital Würzburg, Würzburg 97080, Germany
| |
Collapse
|
32
|
Hall BE, Mazhar K, Macdonald E, Cassidy M, Doty M, Judkins C, Terse A, Shiers S, Tadros S, Yun S, Burton MD, Price TJ, Kulkarni A. Transcriptome Analysis of Rheumatoid Arthritis Uncovers Genes Linked to Inflammation-Induced Pain. RESEARCH SQUARE 2024:rs.3.rs-4218885. [PMID: 38712195 PMCID: PMC11071542 DOI: 10.21203/rs.3.rs-4218885/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Autoimmune diseases such as rheumatoid arthritis (RA) can promote states of chronic Inflammation with accompanying tissue destruction and pain. RA can cause inflammatory synovitis in peripheral joints, particularly within the hands and feet, but can also sometimes trigger temporomandibular joint (TMJ) arthralgia. To better understand the effects of ongoing Inflammation-induced pain signaling, dorsal root ganglia (DRGs) were acquired from individuals with RA for transcriptomic study. We conducted RNA sequencing from the L5 DRGs because it contains the soma of the sensory neurons that innervate the affected joints in the foot. DRGs from 5 RA patients were compared with 9 non-arthritic controls. RNA-seq of L5 DRGs identified 128 differentially expressed genes (DEGs) that were dysregulated in the RA subjects as compared to the non-arthritic controls. The DRG resides outside the blood brain barrier and, as such, our initial transcriptome analysis detected signs of an autoimmune disorder including the upregulated expression of immunoglobulins and other immunologically related genes within the DRGs of the RA donors. Additionally, we saw the upregulation in genes implicated in neurogenesis that could promote pain hypersensitivity. overall, our DRG analysis suggests that there are upregulated inflammatory and pain signaling pathways that can contribute to chronic pain in RA.
Collapse
Affiliation(s)
- Bradford E Hall
- National Institute of Dental and Craniofacial Research, National Institutes of Health
| | | | - Emma Macdonald
- National Institute of Dental and Craniofacial Research, National Institutes of Health
| | - Margaret Cassidy
- National Institute of Dental and Craniofacial Research, National Institutes of Health
| | - Megan Doty
- National Institute of Dental and Craniofacial Research, National Institutes of Health
| | - Christian Judkins
- National Institute of Dental and Craniofacial Research, National Institutes of Health
| | - Anita Terse
- National Institute of Dental and Craniofacial Research, National Institutes of Health
| | | | - Saber Tadros
- National Cancer Institute, National Institutes of Health
| | | | | | | | - Ashok Kulkarni
- National Institute of Dental and Craniofacial Research, National Institutes of Health
| |
Collapse
|
33
|
Bhowmick S, Graham RD, Verma N, Trevathan JK, Franke M, Nieuwoudt S, Fisher LE, Shoffstall AJ, Weber DJ, Ludwig KA, Lempka SF. Computational modeling of dorsal root ganglion stimulation using an Injectrode. J Neural Eng 2024; 21:026039. [PMID: 38502956 PMCID: PMC11007586 DOI: 10.1088/1741-2552/ad357f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 02/23/2024] [Accepted: 03/19/2024] [Indexed: 03/21/2024]
Abstract
Objective.Minimally invasive neuromodulation therapies like the Injectrode, which is composed of a tightly wound polymer-coated Platinum/Iridium microcoil, offer a low-risk approach for administering electrical stimulation to the dorsal root ganglion (DRG). This flexible electrode is aimed to conform to the DRG. The stimulation occurs through a transcutaneous electrical stimulation (TES) patch, which subsequently transmits the stimulation to the Injectrode via a subcutaneous metal collector. However, it is important to note that the effectiveness of stimulation through TES relies on the specific geometrical configurations of the Injectrode-collector-patch system. Hence, there is a need to investigate which design parameters influence the activation of targeted neural structures.Approach.We employed a hybrid computational modeling approach to analyze the impact of Injectrode system design parameters on charge delivery and neural response to stimulation. We constructed multiple finite element method models of DRG stimulation, followed by the implementation of multi-compartment models of DRG neurons. By calculating potential distribution during monopolar stimulation, we simulated neural responses using various parameters based on prior acute experiments. Additionally, we developed a canonical monopolar stimulation and full-scale model of bipolar bilateral L5 DRG stimulation, allowing us to investigate how design parameters like Injectrode size and orientation influenced neural activation thresholds.Main results.Our findings were in accordance with acute experimental measurements and indicate that the minimally invasive Injectrode system predominantly engages large-diameter afferents (Aβ-fibers). These activation thresholds were contingent upon the surface area of the Injectrode. As the charge density decreased due to increasing surface area, there was a corresponding expansion in the stimulation amplitude range before triggering any pain-related mechanoreceptor (Aδ-fibers) activity.Significance.The Injectrode demonstrates potential as a viable technology for minimally invasive stimulation of the DRG. Our findings indicate that utilizing a larger surface area Injectrode enhances the therapeutic margin, effectively distinguishing the desired Aβactivation from the undesired Aδ-fiber activation.
Collapse
Affiliation(s)
- Sauradeep Bhowmick
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States of America
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, United States of America
| | - Robert D Graham
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States of America
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, United States of America
| | - Nishant Verma
- Department of Biomedical Engineering, University of Wisconsin–Madison, Madison, WI, United States of America
- Wisconsin Institute for Translational Neuroengineering (WITNe)–Madison, Madison, WI, United States of America
| | - James K Trevathan
- Department of Biomedical Engineering, University of Wisconsin–Madison, Madison, WI, United States of America
- Wisconsin Institute for Translational Neuroengineering (WITNe)–Madison, Madison, WI, United States of America
| | | | | | - Lee E Fisher
- Rehab Neural Engineering Labs (RNEL), Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Andrew J Shoffstall
- Neuronoff Inc., Cleveland, OH, United States of America
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States of America
| | - Douglas J Weber
- Department of Mechanical Engineering, Carnegie Mellon University, Pittsburgh, PA, United States of America
- Neuroscience Institute, Carnegie Mellon University, Pittsburgh, PA, United States of America
| | - Kip A Ludwig
- Department of Biomedical Engineering, University of Wisconsin–Madison, Madison, WI, United States of America
- Wisconsin Institute for Translational Neuroengineering (WITNe)–Madison, Madison, WI, United States of America
- Department of Neurosurgery, University of Wisconsin–Madison, Madison, WI, United States of America
| | - Scott F Lempka
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States of America
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, United States of America
- Department of Anesthesiology, University of Michigan, Ann Arbor, MI, United States of America
| |
Collapse
|
34
|
Falconieri A, Folino P, Da Palmata L, Raffa V. Nano-pulling stimulates axon regeneration in dorsal root ganglia by inducing stabilization of axonal microtubules and activation of local translation. Front Mol Neurosci 2024; 17:1340958. [PMID: 38633213 PMCID: PMC11022966 DOI: 10.3389/fnmol.2024.1340958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Accepted: 03/11/2024] [Indexed: 04/19/2024] Open
Abstract
Introduction Axonal plasticity is strongly related to neuronal development as well as regeneration. It was recently demonstrated that active mechanical tension, intended as an extrinsic factor, is a valid contribution to the modulation of axonal plasticity. Methods In previous publications, our team validated a the "nano-pulling" method used to apply mechanical forces to developing axons of isolated primary neurons using magnetic nanoparticles (MNP) actuated by static magnetic fields. This method was found to promote axon growth and synaptic maturation. Here, we explore the use of nano-pulling as an extrinsic factor to promote axon regeneration in a neuronal tissue explant. Results Whole dorsal root ganglia (DRG) were thus dissected from a mouse spinal cord, incubated with MNPs, and then stretched. We found that particles were able to penetrate the ganglion and thus become localised both in the somas and in sprouting axons. Our results highlight that nano-pulling doubles the regeneration rate, and this is accompanied by an increase in the arborizing capacity of axons, an accumulation of cellular organelles related to mass addition (endoplasmic reticulum and mitochondria) and pre-synaptic proteins with respect to spontaneous regeneration. In line with the previous results on isolated hippocampal neurons, we observed that this process is coupled to an increase in the density of stable microtubules and activation of local translation. Discussion Our data demonstrate that nano-pulling enhances axon regeneration in whole spinal ganglia exposed to MNPs and external magnetic fields. These preliminary data represent an encouraging starting point for proposing nano-pulling as a biophysical tool for the design of novel therapies based on the use of force as an extrinsic factor for promoting nerve regeneration.
Collapse
|
35
|
Hanani M. Satellite Glial Cells in Human Disease. Cells 2024; 13:566. [PMID: 38607005 PMCID: PMC11011452 DOI: 10.3390/cells13070566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 03/19/2024] [Accepted: 03/21/2024] [Indexed: 04/13/2024] Open
Abstract
Satellite glial cells (SGCs) are the main type of glial cells in sensory ganglia. Animal studies have shown that these cells play essential roles in both normal and disease states. In a large number of pain models, SGCs were activated and contributed to the pain behavior. Much less is known about SGCs in humans, but there is emerging recognition that SGCs in humans are altered in a variety of clinical states. The available data show that human SGCs share some essential features with SGCs in rodents, but many differences do exist. SGCs in DRG from patients suffering from common painful diseases, such as rheumatoid arthritis and fibromyalgia, may contribute to the pain phenotype. It was found that immunoglobulins G (IgG) from fibromyalgia patients can induce pain-like behavior in mice. Moreover, these IgGs bind preferentially to SGCs and activate them, which can sensitize the sensory neurons, causing nociception. In other human diseases, the evidence is not as direct as in fibromyalgia, but it has been found that an antibody from a patient with rheumatoid arthritis binds to mouse SGCs, which leads to the release of pronociceptive factors from them. Herpes zoster is another painful disease, and it appears that the zoster virus resides in SGCs, which acquire an abnormal morphology and may participate in the infection and pain generation. More work needs to be undertaken on SGCs in humans, and this review points to several promising avenues for better understanding disease mechanisms and developing effective pain therapies.
Collapse
Affiliation(s)
- Menachem Hanani
- Laboratory of Experimental Surgery, Hadassah-Hebrew University Medical Center, Mount Scopus, Jerusalem 91240, Israel; ; Tel.: +972-2-5844721
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 91120, Israel
| |
Collapse
|
36
|
Ma J, Eglauf J, Grad S, Alini M, Serra T. Engineering Sensory Ganglion Multicellular System to Model Tissue Nerve Ingrowth. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308478. [PMID: 38113315 PMCID: PMC10953573 DOI: 10.1002/advs.202308478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 12/04/2023] [Indexed: 12/21/2023]
Abstract
Discogenic pain is associated with deep nerve ingrowth in annulus fibrosus tissue (AF) of intervertebral disc (IVD). To model AF nerve ingrowth, primary bovine dorsal root ganglion (DRG) micro-scale tissue units are spatially organised around an AF explant by mild hydrodynamic forces within a collagen matrix. This results in a densely packed multicellular system mimicking the native DRG tissue morphology and a controlled AF-neuron distance. Such a multicellular organisation is essential to evolve populational-level cellular functions and in vivo-like morphologies. Pro-inflammatory cytokine-primed AF demonstrates its neurotrophic and neurotropic effects on nociceptor axons. Both effects are dependent on the AF-neuron distance underpinning the role of recapitulating inter-tissue/organ anatomical proximity when investigating their crosstalk. This is the first in vitro model studying AF nerve ingrowth by engineering mature and large animal tissues in a morphologically and physiologically relevant environment. The new approach can be used to biofabricate multi-tissue/organ models for untangling pathophysiological conditions and develop novel therapies.
Collapse
Affiliation(s)
- Junxuan Ma
- AO Research InstituteClavadelerstrasse 8Davos7270Switzerland
| | - Janick Eglauf
- AO Research InstituteClavadelerstrasse 8Davos7270Switzerland
- ETH ZürichRämistrasse 101Zürich8092Switzerland
| | - Sibylle Grad
- AO Research InstituteClavadelerstrasse 8Davos7270Switzerland
| | - Mauro Alini
- AO Research InstituteClavadelerstrasse 8Davos7270Switzerland
| | - Tiziano Serra
- AO Research InstituteClavadelerstrasse 8Davos7270Switzerland
- Complex Tissue Regeneration DepartmentMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229ETNetherlands
| |
Collapse
|
37
|
Kallogjerovic S, Velázquez-Quesada I, Hadap R, Gligorijevic B. Retrograde tracing of breast cancer-associated sensory neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.26.582088. [PMID: 38463981 PMCID: PMC10925213 DOI: 10.1101/2024.02.26.582088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Breast cancer is one of the leading causes of mortality among women. The tumor microenvironment, consisting of host cells and extracellular matrix, has been increasingly studied for its interplay with cancer cells, and the resulting effect on tumor progression. While the breast is one of the most innervated organs in the body, the role of neurons, and specifically sensory neurons, has been understudied, mostly for technical reasons. One of the reasons is the anatomy of sensory neurons: sensory neuron somas are located in the spine, and their axons can extend longer than a meter across the body to provide innervation in the breast. Next, neurons are challenging to culture, and there are no cell lines adequately representing the diversity of sensory neurons. Finally, sensory neurons are responsible for transporting several different types of signals to the brain, and there are many different subtypes of sensory neurons. The subtypes of sensory neurons which innervate and interact with breast tumors are unknown. To establish the tools for labeling and subtyping neurons that interact with breast cancer cells, we utilized two retrograde tracer's standards in neuroscience, wheat-germ agglutinin (WGA) and cholera toxin subunit B (CTB). In vitro , we employed primary sensory neurons isolated from mouse dorsal root ganglia, cultured in a custom-built microfluidic device DACIT, that mimics the anatomical compartmentalization of the sensory neuron's soma and axons. In vivo , we utilized both syngeneic and transgenic mouse models of mammary carcinoma. We show that CTB and WGA trace different but overlapping sensory neuronal subpopulations: while WGA is more efficient in labeling CGRP+ neurons, CTB is superior in labeling the NF200+ neurons. Surprisingly, both tracers are also taken up by a significant population of breast cancer cells, both in vitro and in vivo . In summary, we have established methodologies for retrograde tracing of sensory neurons interacting with breast cancer cells. Our tools will be useful for future studies of breast tumor innervation, and development of therapies targeting breast cancer-associated neuron subpopulations of sensory neurons.
Collapse
|
38
|
He H, Luo H, Qian B, Xu H, Zhang G, Zou X, Zou J. Autonomic Nervous System Dysfunction Is Related to Chronic Prostatitis/Chronic Pelvic Pain Syndrome. World J Mens Health 2024; 42:1-28. [PMID: 37118962 PMCID: PMC10782122 DOI: 10.5534/wjmh.220248] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 02/09/2023] [Accepted: 02/14/2023] [Indexed: 04/30/2023] Open
Abstract
Chronic prostatitis/chronic pelvic pain syndrome (CP/CPPS) is a common and non-lethal urological condition with painful symptoms. The complexity of CP/CPPS's pathogenesis and lack of efficient etiological diagnosis results in incomplete treatment and recurrent episodes, causing long-term mental and psychological suffering in patients. Recent findings indicate that the autonomic nervous system involves in CP/CPPS, including sensory, sympathetic, parasympathetic, and central nervous systems. Neuro-inflammation and sensitization of sensory nerves lead to persistent inflammation and pain. Sympathetic and parasympathetic alterations affect the cardiovascular and reproductive systems and the development of prostatitis. Central sensitization lowers pain thresholds and increases pelvic pain perception in chronic prostatitis. Therefore, this review summarized the detailed processes and mechanisms of the critical role of the autonomic nervous system in developing CP/CPPS. Furthermore, it describes the neurologically relevant substances and channels or receptors involved in this process, which provides new perspectives for new therapeutic approaches to CP/CPPS.
Collapse
Affiliation(s)
- Hailan He
- Department of Graduate, First Clinical Colledge, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Urology, the First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Hui Luo
- Department of Graduate, First Clinical Colledge, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Urology, the First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Biao Qian
- Department of Urology, the First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Institute of Urology, the First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Jiangxi Engineering Technology Research Center of Calculi Prevention, Ganzhou, Jiangxi, China
| | - Hui Xu
- Department of Urology, the First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Institute of Urology, the First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Jiangxi Engineering Technology Research Center of Calculi Prevention, Ganzhou, Jiangxi, China
| | - Guoxi Zhang
- Department of Urology, the First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Institute of Urology, the First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Jiangxi Engineering Technology Research Center of Calculi Prevention, Ganzhou, Jiangxi, China
| | - Xiaofeng Zou
- Department of Urology, the First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Institute of Urology, the First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Jiangxi Engineering Technology Research Center of Calculi Prevention, Ganzhou, Jiangxi, China
| | - Junrong Zou
- Department of Urology, the First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Institute of Urology, the First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Jiangxi Engineering Technology Research Center of Calculi Prevention, Ganzhou, Jiangxi, China.
| |
Collapse
|
39
|
Jang K, Garraway SM. A review of dorsal root ganglia and primary sensory neuron plasticity mediating inflammatory and chronic neuropathic pain. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2024; 15:100151. [PMID: 38314104 PMCID: PMC10837099 DOI: 10.1016/j.ynpai.2024.100151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 01/04/2024] [Accepted: 01/19/2024] [Indexed: 02/06/2024]
Abstract
Pain is a sensory state resulting from complex integration of peripheral nociceptive inputs and central processing. Pain consists of adaptive pain that is acute and beneficial for healing and maladaptive pain that is often persistent and pathological. Pain is indeed heterogeneous, and can be expressed as nociceptive, inflammatory, or neuropathic in nature. Neuropathic pain is an example of maladaptive pain that occurs after spinal cord injury (SCI), which triggers a wide range of neural plasticity. The nociceptive processing that underlies pain hypersensitivity is well-studied in the spinal cord. However, recent investigations show maladaptive plasticity that leads to pain, including neuropathic pain after SCI, also exists at peripheral sites, such as the dorsal root ganglia (DRG), which contains the cell bodies of sensory neurons. This review discusses the important role DRGs play in nociceptive processing that underlies inflammatory and neuropathic pain. Specifically, it highlights nociceptor hyperexcitability as critical to increased pain states. Furthermore, it reviews prior literature on glutamate and glutamate receptors, voltage-gated sodium channels (VGSC), and brain-derived neurotrophic factor (BDNF) signaling in the DRG as important contributors to inflammatory and neuropathic pain. We previously reviewed BDNF's role as a bidirectional neuromodulator of spinal plasticity. Here, we shift focus to the periphery and discuss BDNF-TrkB expression on nociceptors, non-nociceptor sensory neurons, and non-neuronal cells in the periphery as a potential contributor to induction and persistence of pain after SCI. Overall, this review presents a comprehensive evaluation of large bodies of work that individually focus on pain, DRG, BDNF, and SCI, to understand their interaction in nociceptive processing.
Collapse
Affiliation(s)
- Kyeongran Jang
- Department of Cell Biology, Emory University, School of Medicine, Atlanta, GA, 30322, USA
| | - Sandra M. Garraway
- Department of Cell Biology, Emory University, School of Medicine, Atlanta, GA, 30322, USA
| |
Collapse
|
40
|
Xian H, Guo H, Liu YY, Zhang JL, Hu WC, Yu MJ, Zhao R, Xie RG, Zhang H, Cong R. Peripheral BDNF Regulates Somatosensory-Sympathetic Coupling in Brachial Plexus Avulsion-Induced Neuropathic Pain. Neurosci Bull 2023; 39:1789-1806. [PMID: 37335428 PMCID: PMC10661543 DOI: 10.1007/s12264-023-01075-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Accepted: 03/19/2023] [Indexed: 06/21/2023] Open
Abstract
Brachial plexus avulsion (BPA) is a combined injury involving the central and peripheral nervous systems. Patients with BPA often experience severe neuropathic pain (NP) in the affected limb. NP is insensitive to the existing treatments, which makes it a challenge to researchers and clinicians. Accumulated evidence shows that a BPA-induced pain state is often accompanied by sympathetic nervous dysfunction, which suggests that the excitation state of the sympathetic nervous system is correlated with the existence of NP. However, the mechanism of how somatosensory neural crosstalk with the sympathetic nerve at the peripheral level remains unclear. In this study, through using a novel BPA C7 root avulsion mouse model, we found that the expression of BDNF and its receptor TrκB in the DRGs of the BPA mice increased, and the markers of sympathetic nervous system activity including α1 and α2 adrenergic receptors (α1-AR and α2-AR) also increased after BPA. The phenomenon of superexcitation of the sympathetic nervous system, including hypothermia and edema of the affected extremity, was also observed in BPA mice by using CatWalk gait analysis, an infrared thermometer, and an edema evaluation. Genetic knockdown of BDNF in DRGs not only reversed the mechanical allodynia but also alleviated the hypothermia and edema of the affected extremity in BPA mice. Further, intraperitoneal injection of adrenergic receptor inhibitors decreased neuronal excitability in patch clamp recording and reversed the mechanical allodynia of BPA mice. In another branch experiment, we also found the elevated expression of BDNF, TrκB, TH, α1-AR, and α2-AR in DRG tissues from BPA patients compared with normal human DRGs through western blot and immunohistochemistry. Our results revealed that peripheral BDNF is a key molecule in the regulation of somatosensory-sympathetic coupling in BPA-induced NP. This study also opens a novel analgesic target (BDNF) in the treatment of this pain with fewer complications, which has great potential for clinical transformation.
Collapse
Affiliation(s)
- Hang Xian
- Department of Orthopedics, Xijing Hospital, The Air Force Medical University, Xi'an, 710032, China
| | - Huan Guo
- Pain and Related Diseases Research Laboratory, Medical College of Shantou University, Shantou, 515041, China
- Department of Neurobiology, School of Basic Medicine, The Air Force Medical University, Xi'an, 710032, China
| | - Yuan-Ying Liu
- School of Life Science and Research Center for Resource Peptide Drugs, Shaanxi Engineering and Technological Research Center for Conversation and Utilization of Regional Biological Resources, Yanan University, Yanan, 716000, China
- Department of Neurobiology, School of Basic Medicine, The Air Force Medical University, Xi'an, 710032, China
| | - Jian-Lei Zhang
- Department of Orthopedics, Xijing Hospital, The Air Force Medical University, Xi'an, 710032, China
| | - Wen-Chao Hu
- Department of Neurobiology, School of Basic Medicine, The Air Force Medical University, Xi'an, 710032, China
- The Sixth Regiment, School of Basic Medicine, The Air Force Medical University, Xi'an, 710032, China
| | - Ming-Jun Yu
- The Tenth Squadron of the Third Regiment, School of Basic Medicine, The Air Force Medical University, Xi'an, 710032, China
| | - Rui Zhao
- Department of Orthopedics, Xijing Hospital, The Air Force Medical University, Xi'an, 710032, China
| | - Rou-Gang Xie
- Department of Neurobiology, School of Basic Medicine, The Air Force Medical University, Xi'an, 710032, China.
| | - Hang Zhang
- Department of Orthopedics, Xijing Hospital, The Air Force Medical University, Xi'an, 710032, China.
| | - Rui Cong
- Department of Orthopedics, Xijing Hospital, The Air Force Medical University, Xi'an, 710032, China.
| |
Collapse
|
41
|
Krawczyk A, Mozel S, Rycerz K, Jaworska-Adamu J, Arciszewski MB. Immunoreactivity of glutamine synthetase in satellite glia around various subpopulations of lumbar dorsal root ganglia neurons in adult rats treated with monosodium glutamate. J Chem Neuroanat 2023; 134:102347. [PMID: 37838216 DOI: 10.1016/j.jchemneu.2023.102347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 09/30/2023] [Accepted: 10/09/2023] [Indexed: 10/16/2023]
Abstract
Satellite glial cells (SGCs), involved inter alia in glutamate (Glu) metabolism, form a glial sheath around sensory neurons of dorsal root ganglia (DRGs). SGCs show a presence of glutamine synthetase (GS) which transform uptaken Glu into glutamine (Gln). In DRGs, this aminoacid is used mainly by small neurons which are able to synthetize substance P (SP) that play a crucial role in nociception. The aim of the study was to define the influence of monosodium glutamate (MSG) on GS immunoreactivity in satellite glia around various subpopulations of neurons including SP immunopositive cells in DRGs of adult rats. The studies were carried out on lumbar DRGs slides in rats which received subcutaneous injection of saline solution (control group) or 4 g/kg b. w. of MSG (MSG group). Immunofluorescence reactions were conducted with use of anti-GS and anti-SP antibodies. Administration of MSG to adult rats increased the GS immunoexpression in SGCs. In rats receiving MSG, a number of small neurons with GS-immunopositive glial sheath was not altered when compared to control individuals, whereas there was a statistically significant increase of GS immunoexpression in SGCs around large and medium neurons. Moreover, in these animals, a statistically significant increase in the number of small SP-positive neurons with GS-positive glial sheath was observed. SP is responsible for transmission of pain, thus the obtained results may be useful for further research concerning the roles of glia in nociceptive pathway regulation.
Collapse
Affiliation(s)
- Aleksandra Krawczyk
- Department of Animal Anatomy and Histology, Faculty of Veterinary Medicine, University of Life Sciences, Lublin, Poland
| | - Sylwia Mozel
- Department of Animal Anatomy and Histology, Faculty of Veterinary Medicine, University of Life Sciences, Lublin, Poland.
| | - Karol Rycerz
- Department of Animal Anatomy and Histology, Faculty of Veterinary Medicine, University of Life Sciences, Lublin, Poland
| | - Jadwiga Jaworska-Adamu
- Department of Animal Anatomy and Histology, Faculty of Veterinary Medicine, University of Life Sciences, Lublin, Poland
| | - Marcin Bartłomiej Arciszewski
- Department of Animal Anatomy and Histology, Faculty of Veterinary Medicine, University of Life Sciences, Lublin, Poland
| |
Collapse
|
42
|
Veshchitskii A, Merkulyeva N. Calcium-binding protein parvalbumin in the spinal cord and dorsal root ganglia. Neurochem Int 2023; 171:105634. [PMID: 37967669 DOI: 10.1016/j.neuint.2023.105634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 09/20/2023] [Accepted: 10/26/2023] [Indexed: 11/17/2023]
Abstract
Parvalbumin is one of the calcium-binding proteins. In the spinal cord, it is mainly expressed in inhibitory neurons; in the dorsal root ganglia, it is expressed in proprioceptive neurons. In contrast to in the brain, weak systematization of parvalbumin-expressing neurons occurs in the spinal cord. The aim of this paper is to provide a systematic review of parvalbumin-expressing neuronal populations throughout the spinal cord and the dorsal root ganglia of mammals, regarding their mapping, co-expression with some functional markers. The data reviewed are mostly concerning rodentia species because they are predominantly presented in literature.
Collapse
Affiliation(s)
- Aleksandr Veshchitskii
- Neuromorphology Lab, Pavlov Institute of Physiology Russian Academy of Sciences, Saint Petersburg, Russia
| | - Natalia Merkulyeva
- Neuromorphology Lab, Pavlov Institute of Physiology Russian Academy of Sciences, Saint Petersburg, Russia.
| |
Collapse
|
43
|
Willemen HLDM, Santos Ribeiro PS, Broeks M, Meijer N, Versteeg S, Tiggeler A, de Boer TP, Małecki JM, Falnes PØ, Jans J, Eijkelkamp N. Inflammation-induced mitochondrial and metabolic disturbances in sensory neurons control the switch from acute to chronic pain. Cell Rep Med 2023; 4:101265. [PMID: 37944527 PMCID: PMC10694662 DOI: 10.1016/j.xcrm.2023.101265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 07/24/2023] [Accepted: 10/10/2023] [Indexed: 11/12/2023]
Abstract
Pain often persists in patients with an inflammatory disease, even when inflammation has subsided. The molecular mechanisms leading to this failure in pain resolution and the transition to chronic pain are poorly understood. Mitochondrial dysfunction in sensory neurons links to chronic pain, but its role in resolution of inflammatory pain is unclear. Transient inflammation causes neuronal plasticity, called hyperalgesic priming, which impairs resolution of pain induced by a subsequent inflammatory stimulus. We identify that hyperalgesic priming in mice increases the expression of a mitochondrial protein (ATPSc-KMT) and causes mitochondrial and metabolic disturbances in sensory neurons. Inhibition of mitochondrial respiration, knockdown of ATPSCKMT expression, or supplementation of the affected metabolite is sufficient to restore resolution of inflammatory pain and prevents chronic pain development. Thus, inflammation-induced mitochondrial-dependent disturbances in sensory neurons predispose to a failure in resolution of inflammatory pain and development of chronic pain.
Collapse
Affiliation(s)
- Hanneke L D M Willemen
- Center for Translational Immunology, Department of Immunology, University Medical Center Utrecht, Utrecht University, 3508 Utrecht, the Netherlands
| | - Patrícia Silva Santos Ribeiro
- Center for Translational Immunology, Department of Immunology, University Medical Center Utrecht, Utrecht University, 3508 Utrecht, the Netherlands
| | - Melissa Broeks
- Section Metabolic Diagnostics, Department of Genetics, University Medical Center Utrecht, Utrecht University, 3508 Utrecht, the Netherlands
| | - Nils Meijer
- Section Metabolic Diagnostics, Department of Genetics, University Medical Center Utrecht, Utrecht University, 3508 Utrecht, the Netherlands
| | - Sabine Versteeg
- Center for Translational Immunology, Department of Immunology, University Medical Center Utrecht, Utrecht University, 3508 Utrecht, the Netherlands
| | - Annefien Tiggeler
- Center for Translational Immunology, Department of Immunology, University Medical Center Utrecht, Utrecht University, 3508 Utrecht, the Netherlands
| | - Teun P de Boer
- Department of Medical Physiology, Division of Heart & Lungs, University Medical Center Utrecht, Yalelaan 50, 3584 Utrecht, the Netherlands
| | - Jędrzej M Małecki
- Department of Biosciences, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway; CRES-O - Centre for Embryology and Healthy Development, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Pål Ø Falnes
- Department of Biosciences, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway; CRES-O - Centre for Embryology and Healthy Development, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Judith Jans
- Section Metabolic Diagnostics, Department of Genetics, University Medical Center Utrecht, Utrecht University, 3508 Utrecht, the Netherlands
| | - Niels Eijkelkamp
- Center for Translational Immunology, Department of Immunology, University Medical Center Utrecht, Utrecht University, 3508 Utrecht, the Netherlands.
| |
Collapse
|
44
|
Yazar U, Guvercin AR, Rouhikia M, Aktoklu M, Demirci MA, Erbay I, Ayar A. Cerebrolysin provides effective protection on high glucose-induced neuropathy in cultured rat dorsal root ganglion neurons. J Recept Signal Transduct Res 2023; 43:109-114. [PMID: 38079610 DOI: 10.1080/10799893.2023.2291566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 11/23/2023] [Indexed: 01/25/2024]
Abstract
Cerebrolysin, an endogenous peptide with neuroprotective and neurotrophic properties, indicated to be beneficial on diabetic neuropathy by preliminary clinical and experimental studies but without evidence on central or peripheral action. Dorsal root ganglion (DRG) neurons, based on involvement of pain sensation in both health and disease as first relay centers for transmission and processing of peripheral nociceptive sensory signals, was used to investigate possible effects of Cerebrolysin on high glucose-induced neuropathy, as model. DRG's were obtained from adult rats and the isolated neurons were seeded on E-Plate®'s equipped with gold microelectrodes, and incubated in culture media in a CO2 incubator at 37 C. DRGs were exposed to high glucose (50 mM) in the absence and presence of different concentrations of Cerebrolysin ® (2-40 mg/ml). Cell index (derived from cell viability and neurite outgrowth) was recorded with Real-Time Cell Analyzer and was used as primary outcome measure. High glucose-induced cellular neuropathy and neuroprotective effects of Cerebrolysin was evaluated from area under the curve (AUC) of cell index-time graphs. Exposure of DRG neurons to high glucose caused a rapid and persistent decrease in the mean AUC values compared to normoglycemic controls. Co-treatment with Cerebrolysin (40 mg/ml) attenuated this high glucose-induced effect in a concentration-dependent manner. In normoglycemic conditions, treatment with Cerebrolysin caused a dose-dependent increase in the mean AUC values. Cerebrolysin treatment resulted in maintenance of the functional integrity, survival, and promotion of neurite outgrowth of the cultured DRG neurons exposed to high glucose, indicating involvement of peripheral sensory neurons.
Collapse
Affiliation(s)
- Ugur Yazar
- Departments of Neurosurgery, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey
| | - Ali Rıza Guvercin
- Departments of Neurosurgery, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey
| | - Mahindokht Rouhikia
- Departments of Physiology, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey
| | - Mehmet Aktoklu
- Departments of Neurosurgery, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey
| | - Mehmet Ali Demirci
- Departments of Neurosurgery, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey
| | - Ibrahim Erbay
- Departments of Physiology, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey
| | - Ahmet Ayar
- Departments of Physiology, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey
| |
Collapse
|
45
|
Stewart ZE. Safety of local anesthetics in cervical nerve root injections: a narrative review. Skeletal Radiol 2023; 52:1893-1900. [PMID: 36326881 DOI: 10.1007/s00256-022-04220-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 10/23/2022] [Accepted: 10/24/2022] [Indexed: 11/06/2022]
Abstract
Severe neurological adverse events have been reported after fluoroscopically guided cervical nerve root injections. Particulate corticosteroids inadvertently injected intraarterially and iatrogenic vertebral artery trauma have been implicated in these outcomes. This has raised concern for the potential consequences of including local anesthetic with these injections. As a result, some providers have now discontinued the routine administration of local anesthetic with corticosteroid when performing cervical nerve root injections. At present, there is no consensus regarding whether the use of local anesthetic in this context is safe. Here, the current literature is synthesized into a narrative review aiming to clarify current perspectives of the safety of local anesthetics in cervical nerve root injections.
Collapse
Affiliation(s)
- Zachary E Stewart
- Massachusetts General Hospital, Harvard Medical School Boston MA, Boston, USA.
| |
Collapse
|
46
|
Yu H, Usoskin D, Nagi SS, Hu Y, Kupari J, Bouchatta O, Cranfill SL, Gautam M, Su Y, Lu Y, Wymer J, Glanz M, Albrecht P, Song H, Ming GL, Prouty S, Seykora J, Wu H, Ma M, Rice FL, Olausson H, Ernfors P, Luo W. Single-Soma Deep RNA sequencing of Human DRG Neurons Reveals Novel Molecular and Cellular Mechanisms Underlying Somatosensation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.17.533207. [PMID: 36993480 PMCID: PMC10055202 DOI: 10.1101/2023.03.17.533207] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
The versatility of somatosensation arises from heterogeneous dorsal root ganglion (DRG) neurons. However, soma transcriptomes of individual human DRG (hDRG) neurons-critical in-formation to decipher their functions-are lacking due to technical difficulties. Here, we developed a novel approach to isolate individual hDRG neuron somas for deep RNA sequencing (RNA-seq). On average, >9,000 unique genes per neuron were detected, and 16 neuronal types were identified. Cross-species analyses revealed remarkable divergence among pain-sensing neurons and the existence of human-specific nociceptor types. Our deep RNA-seq dataset was especially powerful for providing insight into the molecular mechanisms underlying human somatosensation and identifying high potential novel drug targets. Our dataset also guided the selection of molecular markers to visualize different types of human afferents and the discovery of novel functional properties using single-cell in vivo electrophysiological recordings. In summary, by employing a novel soma sequencing method, we generated an unprecedented hDRG neuron atlas, providing new insights into human somatosensation, establishing a critical foundation for translational work, and clarifying human species-species properties.
Collapse
|
47
|
Brant BJA, Yu Y, Omar AA, Jaramillo Polanco JO, Lopez Lopez CD, Jiménez Vargas NN, Tsang Q, McDonell A, Takami K, Reed DE, Lomax AE, Vanner SJ, Tuck CJ. Dietary monosodium glutamate increases visceral hypersensitivity in a mouse model of visceral pain. Neurogastroenterol Motil 2023; 35:e14596. [PMID: 37248774 DOI: 10.1111/nmo.14596] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 01/24/2023] [Accepted: 04/03/2023] [Indexed: 05/31/2023]
Abstract
BACKGROUND Monosodium glutamate (MSG) has been identified as a trigger of abdominal pain in irritable bowel syndrome (IBS), but the mechanism is unknown. This study examined whether MSG causes visceral hypersensitivity using a water-avoidance stress (WAS) mouse model of visceral pain. METHODS Mice were divided into four groups receiving treatment for 6 days: WAS + MSG gavage, WAS + saline gavage, sham-WAS + MSG gavage, and sham-WAS + saline gavage. The acute effects of intraluminal administration of 10 μM MSG on jejunal extrinsic afferent nerve sensitivity to distension (0-60 mmHg) were examined using ex vivo extracellular recordings. MSG was also applied directly to jejunal afferents from untreated mice. Glutamate concentration was measured in serum, and in the serosal compartment of Ussing chambers following apical administration. KEY RESULTS Acute intraluminal MSG application increased distension responses of jejunal afferent nerves from mice exposed to WAS + MSG. This effect was mediated by wide dynamic range and high-threshold units at both physiologic and noxious pressures (10-60 mmHg, p < 0.05). No effect of MSG was observed in the other groups, or when applied directly to the jejunal afferent nerves. Serum glutamate was increased in mice exposed to WAS + MSG compared to sham-WAS + saline, and serosal glutamate increased using WAS tissue (p = 0.0433). CONCLUSIONS AND INFERENCES These findings demonstrate that repeated exposure to MSG in mice leads to sensitization of jejunal afferent nerves to acute ex vivo exposure to MSG. This may contribute to visceral hypersensitivity reported in response to MSG in patients with IBS.
Collapse
Affiliation(s)
- Bailey J A Brant
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| | - Yang Yu
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| | - Amal Abu Omar
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| | | | - Cintya D Lopez Lopez
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| | | | - Quentin Tsang
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| | - Abby McDonell
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| | - Kaede Takami
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| | - David E Reed
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| | - Alan E Lomax
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| | - Stephen J Vanner
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| | - Caroline J Tuck
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
- Department of Dietetics, Nutrition and Sport, La Trobe University, Melbourne, Victoria, Australia
| |
Collapse
|
48
|
Navickaitė I, Ališauskienė M, Petrauskienė S, Žemgulytė G. Sarcoidosis-Associated Sensory Ganglionopathy and Harlequin Syndrome: A Case Report. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1495. [PMID: 37629785 PMCID: PMC10456357 DOI: 10.3390/medicina59081495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 08/11/2023] [Accepted: 08/18/2023] [Indexed: 08/27/2023]
Abstract
Background and Objectives: Sensory ganglionopathy is a rare neurological disorder caused by degeneration of the neurons composing the dorsal root ganglia. It manifests as various sensory disturbances in the trunk, proximal limbs, face, or mouth in a patchy and asymmetrical pattern. Harlequin syndrome is characterized by unilateral flushing and sweating of the face, neck, and upper chest, concurrent with contralateral anhidrosis. Here, we present and discuss a clinical case of sarcoidosis-associated ganglionopathy and Harlequin syndrome. Case presentation: A 31-year-old woman complained of burning pain in the right side of the upper chest and the feet. She also experienced episodes of intense flushing and sweating on the right side of her face, neck, and upper chest. Three years before these symptoms began, the patient was diagnosed with pulmonary sarcoidosis. On neurological examination, sensory disturbances were present. In the trunk, the patient reported pronounced hyperalgesia and allodynia in the upper part of the right chest and some patches on the right side of the upper back. In the extremities, hypoalgesia in the tips of the fingers and hyperalgesia in the feet were noted. An extensive diagnostic workup was performed to eliminate other possible causes of these disorders. A broad range of possible metabolic, immunological, and structural causes were ruled out. Thus, the final clinical diagnosis of sarcoidosis-induced sensory ganglionopathy, small-fiber neuropathy, and Harlequin syndrome was made. Initially, the patient was treated with pregabalin and amitriptyline, but the effect was inadequate for the ganglionopathy-induced pain. Therefore, therapeutic plasma exchange as an immune-modulating treatment was selected, leading to partial pain relief. Conclusions: This case report demonstrates the possible autoimmune origin of both sensory ganglionopathy and Harlequin syndrome. It suggests that an autoimmune etiology for these disorders should be considered and the diagnostic workup should include screening for the most common autoimmune conditions.
Collapse
Affiliation(s)
- Ieva Navickaitė
- Department of Neurology, Medical Academy, Lithuanian University of Health Sciences, A. Mickeviciaus Str. 9, LT-44307 Kaunas, Lithuania; (M.A.); (G.Ž.)
| | - Miglė Ališauskienė
- Department of Neurology, Medical Academy, Lithuanian University of Health Sciences, A. Mickeviciaus Str. 9, LT-44307 Kaunas, Lithuania; (M.A.); (G.Ž.)
| | - Sandra Petrauskienė
- Department of Preventive and Paediatric Dentistry, Lithuanian University of Health Sciences, Luksos-Daumanto Str. 6, LT-50106 Kaunas, Lithuania;
| | - Gintarė Žemgulytė
- Department of Neurology, Medical Academy, Lithuanian University of Health Sciences, A. Mickeviciaus Str. 9, LT-44307 Kaunas, Lithuania; (M.A.); (G.Ž.)
| |
Collapse
|
49
|
Vroman R, Hunter RS, Wood MJ, Davis OC, Malfait Z, George DS, Ren D, Tavares-Ferreira D, Price TJ, Miller RJ, Malfait AM, Malfait F, Miller RE, Syx D. Analysis of matrisome expression patterns in murine and human dorsal root ganglia. Front Mol Neurosci 2023; 16:1232447. [PMID: 37664243 PMCID: PMC10471487 DOI: 10.3389/fnmol.2023.1232447] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 07/27/2023] [Indexed: 09/05/2023] Open
Abstract
The extracellular matrix (ECM) is a dynamic structure of molecules that can be divided into six different categories and are collectively called the matrisome. The ECM plays pivotal roles in physiological processes in many tissues, including the nervous system. Intriguingly, alterations in ECM molecules/pathways are associated with painful human conditions and murine pain models. Nevertheless, mechanistic insight into the interplay of normal or defective ECM and pain is largely lacking. The goal of this study was to integrate bulk, single-cell, and spatial RNA sequencing (RNAseq) datasets to investigate the expression and cellular origin of matrisome genes in male and female murine and human dorsal root ganglia (DRG). Bulk RNAseq showed that about 65% of all matrisome genes were expressed in both murine and human DRG, with proportionally more core matrisome genes (glycoproteins, collagens, and proteoglycans) expressed compared to matrisome-associated genes (ECM-affiliated genes, ECM regulators, and secreted factors). Single cell RNAseq on male murine DRG revealed the cellular origin of matrisome expression. Core matrisome genes, especially collagens, were expressed by fibroblasts whereas matrisome-associated genes were primarily expressed by neurons. Cell-cell communication network analysis with CellChat software predicted an important role for collagen signaling pathways in connecting vascular cell types and nociceptors in murine tissue, which we confirmed by analysis of spatial transcriptomic data from human DRG. RNAscope in situ hybridization and immunohistochemistry demonstrated expression of collagens in fibroblasts surrounding nociceptors in male and female human DRG. Finally, comparing human neuropathic pain samples with non-pain samples also showed differential expression of matrisome genes produced by both fibroblasts and by nociceptors. This study supports the idea that the DRG matrisome may contribute to neuronal signaling in both mouse and human, and that dysregulation of matrisome genes is associated with neuropathic pain.
Collapse
Affiliation(s)
- Robin Vroman
- Department of Biomolecular Medicine, Center for Medical Genetics, Ghent University, Ghent, Belgium
| | - Rahel S. Hunter
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, IL, United States
| | - Matthew J. Wood
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, IL, United States
| | - Olivia C. Davis
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, United States
| | - Zoë Malfait
- Department of Biomolecular Medicine, Center for Medical Genetics, Ghent University, Ghent, Belgium
| | - Dale S. George
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Dongjun Ren
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Diana Tavares-Ferreira
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, United States
| | - Theodore J. Price
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, United States
| | - Richard J. Miller
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Anne-Marie Malfait
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, IL, United States
| | - Fransiska Malfait
- Department of Biomolecular Medicine, Center for Medical Genetics, Ghent University, Ghent, Belgium
| | - Rachel E. Miller
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, IL, United States
| | - Delfien Syx
- Department of Biomolecular Medicine, Center for Medical Genetics, Ghent University, Ghent, Belgium
| |
Collapse
|
50
|
Isaifan D, Crovella S, Soubra L, Al-Nesf M, Steinhoff M. Fc Epsilon RI-Neuroimmune Interplay in Pruritus Triggered by Particulate Matter in Atopic Dermatitis Patients. Int J Mol Sci 2023; 24:11851. [PMID: 37511610 PMCID: PMC10380572 DOI: 10.3390/ijms241411851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/25/2023] [Accepted: 03/29/2023] [Indexed: 07/30/2023] Open
Abstract
Atopic dermatitis (AD) is the most common chronic relapsing neuroinflammatory skin disease that is characterized by a complex and multifactorial pathophysiology. It reflects a profound interplay between genetic and environmental factors, and a recently disclosed neuroimmune dysregulation that drives skin barrier disruption, pruritus, and microbial imbalance. In terms of the key external environmental players that impact AD, air quality and itch severity linkage have been thoroughly researched. The impact of ambient air pollutants including particulate matter (PM) and AD pruritic exacerbation has been recorded despite reductions in air pollution levels in in developed countries. The developing countries have, on the contrary, experienced significant urbanization and industrialization with limited environmental protection standards in the past decades. This unprecedented construction, petrochemical industry utilization, and increment in population counts has been paired with consistent exposure to outdoor PM. This may present a key cause of AD pruritic exacerbation supported by the fact that AD prevalence has intensified globally in the past 50 years, indicating that environmental exposure may act as a trigger that could flare up itch in vulnerable persons. At the molecular level, the impact of PM on severe pruritus in AD could be interpreted by the toxic effects on the complex neuroimmune pathways that govern this disease. AD has been recently viewed as a manifestation of the disruption of both the immune and neurological systems. In light of these facts, this current review aims to introduce the basic concepts of itch sensory circuits in the neuroimmune system. In addition, it describes the impact of PM on the potential neuroimmune pathways in AD pathogenesis with a special focus on the Fc Epsilon RI pathway. Finally, the review proposes potential treatment lines that could be targeted to alleviate pruritus based on immune mediators involved in the Fc Epsilon signaling map.
Collapse
Affiliation(s)
- Dina Isaifan
- Department of Biological and Environmental Sciences, College of Arts and Sciences, Qatar University, Doha P.O. Box 2713, Qatar
| | - Sergio Crovella
- Laboratory of Animal Research Center (LARC), Qatar University, Doha P.O. Box 2713, Qatar
| | - Lama Soubra
- Department of Biological and Environmental Sciences, College of Arts and Sciences, Qatar University, Doha P.O. Box 2713, Qatar
| | - Maryam Al-Nesf
- Allergy and Immunology Division, Hamad Medical Corporation, Doha P.O. Box 3050, Qatar
| | - Martin Steinhoff
- Department of Dermatology & Venereology, Weill Cornell Medicine, New York, NY 10065, USA
| |
Collapse
|