1
|
Zhu H, Mu L, Xu X, Huang T, Wang Y, Xu S, Wang Y, Wang W, Wang Z, Wang H, Xue C. EZH2-dependent myelination following sciatic nerve injury. Neural Regen Res 2025; 20:2382-2394. [PMID: 39359095 PMCID: PMC11759024 DOI: 10.4103/nrr.nrr-d-23-02040] [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: 12/17/2023] [Revised: 02/26/2024] [Accepted: 03/29/2024] [Indexed: 10/04/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202508000-00028/figure1/v/2024-09-30T120553Z/r/image-tiff Demyelination and remyelination have been major focal points in the study of peripheral nerve regeneration following peripheral nerve injury. Notably, the gene regulatory network of regenerated myelin differs from that of native myelin. Silencing of enhancer of zeste homolog 2 (EZH2) hinders the differentiation, maturation, and myelination of Schwann cells in vitro. To further determine the role of EZH2 in myelination and recovery post-peripheral nerve injury, conditional knockout mice lacking Ezh2 in Schwann cells (Ezh2fl/fl;Dhh-Cre and Ezh2fl/fl;Mpz-Cre) were generated. Our results show that a significant proportion of axons in the sciatic nerve of Ezh2-depleted mice remain unmyelinated. This highlights the crucial role of Ezh2 in initiating Schwann cell myelination. Furthermore, we observed that 21 days after inducing a sciatic nerve crush injury in these mice, most axons had remyelinated at the injury site in the control nerve, while Ezh2fl/fl;Mpz-Cre mice had significantly fewer remyelinated axons compared with their wild-type littermates. This suggests that the absence of Ezh2 in Schwann cells impairs myelin formation and remyelination. In conclusion, EZH2 has emerged as a pivotal regulatory factor in the process of demyelination and myelin regeneration following peripheral nerve injury. Modulating EZH2 activity during these processes may offer a promising therapeutic target for the treatment of peripheral nerve injuries.
Collapse
Affiliation(s)
- Hui Zhu
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu Province, China
| | - Li Mu
- Medical School of Nantong University, Nantong, Jiangsu Province, China
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Xi Xu
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu Province, China
- Department of Rehabilitation Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Tianyi Huang
- Medical School of Nantong University, Nantong, Jiangsu Province, China
| | - Ying Wang
- Medical School of Nantong University, Nantong, Jiangsu Province, China
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Siyuan Xu
- Medical School of Nantong University, Nantong, Jiangsu Province, China
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Yiting Wang
- Medical School of Nantong University, Nantong, Jiangsu Province, China
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Wencong Wang
- Medical School of Nantong University, Nantong, Jiangsu Province, China
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Zhiping Wang
- Medical School of Nantong University, Nantong, Jiangsu Province, China
- Department of Critical Care Medicine, Nantong Fourth People’s Hospital, Nantong, Jiangsu Province, China
| | - Hongkui Wang
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu Province, China
| | - Chengbin Xue
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu Province, China
| |
Collapse
|
2
|
Marymonchyk A, Rodriguez-Aller R, Willis A, Beaupré F, Warsi S, Snapyan M, Clavet-Fournier V, Lavoie-Cardinal F, Kaplan DR, Miller FD, Saghatelyan A. Neural stem cell quiescence and activation dynamics are regulated by feedback input from their progeny under homeostatic and regenerative conditions. Cell Stem Cell 2025; 32:445-462.e9. [PMID: 39919748 DOI: 10.1016/j.stem.2025.01.001] [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: 03/28/2024] [Revised: 11/03/2024] [Accepted: 01/02/2025] [Indexed: 02/09/2025]
Abstract
Life-long maintenance of stem cells implies that feedback mechanisms from the niche regulate their quiescence/activation dynamics. Here, in the mouse adult subventricular neural stem cell (NSC) niche, we charted a precise spatiotemporal map of functional responses in NSCs induced by multiple niche cells and used machine learning to predict NSC interactions with specific niche cell types. We revealed a feedback mechanism whereby the NSC proliferative state is directly repressed by transient amplifying cells (TAPs), their rapidly dividing progeny. NSC processes wrap around TAPs and display hotspots of Ca2+ activity at their points of contact, mediated by ephrin (Efn) signaling. The modulation of Efn signaling or TAP ablation altered the Ca2+ signature of NSCs, leading to their activation. In vivo optogenetic modulation of Ca2+ dynamics abrogated NSC activation and prevented niche replenishment. Thus, TAP-to-NSC feedback signaling controls stem cell quiescence and activation, providing a mechanism to maintain stem cell pools throughout life.
Collapse
Affiliation(s)
- Alina Marymonchyk
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada; Université Laval, Quebec City, QC, Canada
| | - Raquel Rodriguez-Aller
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada; Université Laval, Quebec City, QC, Canada
| | - Ashleigh Willis
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
| | - Frédéric Beaupré
- Université Laval, Quebec City, QC, Canada; CERVO Brain Research Center, Quebec City, QC, Canada
| | - Sareen Warsi
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Marina Snapyan
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Valérie Clavet-Fournier
- Université Laval, Quebec City, QC, Canada; CERVO Brain Research Center, Quebec City, QC, Canada
| | - Flavie Lavoie-Cardinal
- Université Laval, Quebec City, QC, Canada; CERVO Brain Research Center, Quebec City, QC, Canada
| | - David R Kaplan
- Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada; Program for Neuroscience and Mental Health, Hospital for Sick Children, Toronto, ON, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Freda D Miller
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada; Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada; Program for Neuroscience and Mental Health, Hospital for Sick Children, Toronto, ON, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada; Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Armen Saghatelyan
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada.
| |
Collapse
|
3
|
Londono R, Pan Z, Hudnall ML, Lozito TP. Simulation of adult limb regeneration with lizard tail spinal cord implants reveals distinct roles of radial glia and microglia populations. RESEARCH SQUARE 2025:rs.3.rs-6010337. [PMID: 40092433 PMCID: PMC11908338 DOI: 10.21203/rs.3.rs-6010337/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Lizards are the closest relatives of humans able to suppress fibrosis and regrow multiple tissue lineages following appendage regeneration. As amniotes capable of tail, but not limb regrowth, lizards are also distinguished as the only vertebrate group that include both regenerative and non-regenerative appendages in the same animal. Lizard tail stumps naturally form blastemas - heterogenous collections of fibroblasts, adult stem cells, and immune cells that suppress scar formation and potentiate new tissue growth. Conversely, amputated lizard limbs form scars similar to those observed in human patients. Lizard blastema formation is dependent upon tail spinal cord tissue, which contains distinct populations of radial glia and microglia. Using the parthenogenetic lizard Lepidodactylus lugubris as a platform for tail-to-limb spinal cord implantations, we developed an ectopic blastema model toward defining the roles of radial glial and microglia in appendage regeneration. Removal of either population inhibits fibroblast proliferation and blastema formation, but only microglia depletion leads to enhanced fibrosis. Similarly, effects of radial glia, but not microglia, depletion on fibroblast proliferation are reversed via Hedgehog agonism. Taken together, these results indicate that lizard limbs contain all the necessary cell types and biological responses necessary for blastema formation but lack the proliferative and anti-fibrotic signals provided by tail spinal cord radial glia and microglia, respectively. Radial glia contribute Hedgehog signals that cause fibroblast proliferation but do not affect fibrosis. Conversely, microglia enhance fibroblast sensitivity to Hedgehog signaling and inhibit differentiation into fibrocytes. In summary, this study demonstrates blastema stimulation in amputated limbs of adult amniotes with application of lizard spinal cord cells and holds promise as a blueprint for limiting painful scarring and supporting new tissue growth following amputation injuries in human patients.
Collapse
Affiliation(s)
- Ricardo Londono
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Zheyu Pan
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, 1540 Alcazar St, Los Angeles, CA 90089, USA
| | - Megan L Hudnall
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, 1540 Alcazar St, Los Angeles, CA 90089, USA
| | - Thomas P Lozito
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, 1540 Alcazar St, Los Angeles, CA 90089, USA
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, 1425 San Pablo St, Los Angeles, CA 90033, US
| |
Collapse
|
4
|
Willis A, Jeong D, Liu Y, Lithopoulos MA, Yuzwa SA, Frankland PW, Kaplan DR, Miller FD. Single cell approaches define neural stem cell niches and identify microglial ligands that can enhance precursor-mediated oligodendrogenesis. Cell Rep 2025; 44:115194. [PMID: 39823226 DOI: 10.1016/j.celrep.2024.115194] [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: 03/22/2024] [Revised: 05/24/2024] [Accepted: 12/19/2024] [Indexed: 01/19/2025] Open
Abstract
Here, we used single cell RNA sequencing and single cell spatial transcriptomics to characterize the forebrain neural stem cell (NSC) niche under homeostatic and injury conditions. We defined the dorsal and lateral ventricular-subventricular zones (V-SVZs) as two distinct neighborhoods and showed that, after white matter injury, NSCs are activated to make oligodendrocytes dorsally for remyelination. This activation is coincident with an increase in transcriptionally distinct microglia in the dorsal V-SVZ niche. We modeled ligand-receptor interactions within this changing niche and identified two remyelination-associated microglial ligands, insulin growth factor 1 and oncostatin M, that promote precursor proliferation and oligodendrogenesis in culture. Infusion of either ligand into the lateral ventricles also enhanced oligodendrogenesis, even in the lateral V-SVZ, where NSCs normally make neuroblasts. These data support a model where gliogenesis versus neurogenesis is determined by the local NSC neighborhood and where injury-induced niche alterations promote NSC activation, local oligodendrogenesis, and likely contribute to myelin repair.
Collapse
Affiliation(s)
- Ashleigh Willis
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Danielle Jeong
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Yunlong Liu
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Marissa A Lithopoulos
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Scott A Yuzwa
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Paul W Frankland
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Psychology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - David R Kaplan
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Freda D Miller
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
5
|
Schneider A, Won S, Armstrong EA, Cooper AJ, Suresh A, Rivera R, Barrett‐Wilt G, Denu JM, Simcox JA, Svaren J. The role of ATP citrate lyase in myelin formation and maintenance. Glia 2025; 73:105-121. [PMID: 39318247 PMCID: PMC11660526 DOI: 10.1002/glia.24620] [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: 03/22/2024] [Revised: 09/05/2024] [Accepted: 09/14/2024] [Indexed: 09/26/2024]
Abstract
Formation of myelin by Schwann cells is tightly coupled to peripheral nervous system development and is important for neuronal function and long-term maintenance. Perturbation of myelin causes a number of specific disorders that are among the most prevalent diseases affecting the nervous system. Schwann cells synthesize myelin lipids de novo rather than relying on uptake of circulating lipids, yet one unresolved matter is how acetyl CoA, a central metabolite in lipid formation is generated during myelin formation and maintenance. Recent studies have shown that glucose-derived acetyl CoA itself is not required for myelination. However, the importance of mitochondrially-derived acetyl CoA has never been tested for myelination in vivo. Therefore, we have developed a Schwann cell-specific knockout of the ATP citrate lyase (Acly) gene to determine the importance of mitochondrial metabolism to supply acetyl CoA in nerve development. Intriguingly, the ACLY pathway is important for myelin maintenance rather than myelin formation. In addition, ACLY is required to maintain expression of a myelin-associated gene program and to inhibit activation of the latent Schwann cell injury program.
Collapse
Affiliation(s)
- Andrew Schneider
- Waisman CenterUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Seongsik Won
- Waisman CenterUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Eric A. Armstrong
- Wisconsin Institute of DiscoveryUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Aaron J. Cooper
- Waisman CenterUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
- Department of Comparative Biosciences, School of Veterinary MedicineUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Amulya Suresh
- Waisman CenterUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Rachell Rivera
- Waisman CenterUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | | | - John M. Denu
- Wisconsin Institute of DiscoveryUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Judith A. Simcox
- Howard Hughes Medical Institute, Department of BiochemistryUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - John Svaren
- Waisman CenterUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
- Department of Comparative Biosciences, School of Veterinary MedicineUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| |
Collapse
|
6
|
Yuan X, Li W, Liu Q, Ou Y, Li J, Yan Q, Zhang P. Single-Cell RNA-Seq Reveals the Pseudo-temporal Dynamic Evolution Characteristics of ADSCs to Neuronal Differentiation. Cell Mol Neurobiol 2024; 45:5. [PMID: 39661257 PMCID: PMC11634962 DOI: 10.1007/s10571-024-01524-y] [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: 08/06/2024] [Accepted: 12/05/2024] [Indexed: 12/12/2024]
Abstract
Adipose-derived stromal cells (ADSCs) are commonly used in regenerative medicine, but the genetic features of their development into neuronal cells are unknown. This study used single-cell RNA sequencing (scRNA-seq) to reveal gene expression changes during ADSCs to neuronal differentiation. Sequencing of the ADSCs group, the prei-1d group, and the induction 1 h, 3 h, 5 h, 6 h, and 8 h groups was performed using the BD Rhapsody platform. Sequence data were analyzed using t-SNE, Monocle2, GO, and KEGG algorithms. Results showed that a total of 38,453 cells were collected, which were divided into 0-13 clusters. Monocle2 structured analysis revealed that ADSCs were located at the beginning of the trajectory, and the cells after 5 h of induction were mainly distributed at the end of the trajectory in branches 1 and 2. Up-regulated differentially expressed genes (DEGs) at 5 h after induction enriched GO items including cellular protein metabolism, cell adhesion, endocytosis, and cell migration. KEGG analysis showed that induced 6 h and 8 h groups mainly enriched pathways were oxidative phosphorylation, glutathione metabolism, and expression of Parkinson's disease-related genes. In conclusion, two distinct cell state mechanisms stimulate ADSCs to develop into mature neurons. ADSCs induced for 5 h had developed into mature neurons. Later, the differentiated cells undergo degenerative changes associated with senescence.
Collapse
Affiliation(s)
- Xiaodong Yuan
- Department of Neurology, Kailuan General Hospital Affiliated North China University of Science and Technology, 57 Xinhua East Road, Lubei District, Tangshan, 063000, Hebei, China.
- Department of Neurology, Hebei Provincial Key Laboratory of Neurobiological Function, Tangshan, 063000, Hebei, China.
| | - Wen Li
- Department of Neurology, Kailuan General Hospital Affiliated North China University of Science and Technology, 57 Xinhua East Road, Lubei District, Tangshan, 063000, Hebei, China
- Department of Neurology, Hebei Provincial Key Laboratory of Neurobiological Function, Tangshan, 063000, Hebei, China
| | - Qing Liu
- Department of Neurology, Kailuan General Hospital Affiliated North China University of Science and Technology, 57 Xinhua East Road, Lubei District, Tangshan, 063000, Hebei, China
- Department of Neurology, Hebei Provincial Key Laboratory of Neurobiological Function, Tangshan, 063000, Hebei, China
| | - Ya Ou
- Department of Neurology, Kailuan General Hospital Affiliated North China University of Science and Technology, 57 Xinhua East Road, Lubei District, Tangshan, 063000, Hebei, China
| | - Jing Li
- Department of Radiology, Tangshan Maternal and Child Health Hospital, Tangshan, 063000, Hebei, China
| | - Qi Yan
- Department of Neurology, Kailuan General Hospital Affiliated North China University of Science and Technology, 57 Xinhua East Road, Lubei District, Tangshan, 063000, Hebei, China
- Department of Neurology, Hebei Provincial Key Laboratory of Neurobiological Function, Tangshan, 063000, Hebei, China
| | - Pingshu Zhang
- Department of Neurology, Kailuan General Hospital Affiliated North China University of Science and Technology, 57 Xinhua East Road, Lubei District, Tangshan, 063000, Hebei, China.
- Department of Neurology, Hebei Provincial Key Laboratory of Neurobiological Function, Tangshan, 063000, Hebei, China.
| |
Collapse
|
7
|
Thomas MJ, Xu H, Wang A, Beg MA, Sorci-Thomas MG. PCPE2: Expression of multifunctional extracellular glycoprotein associated with diverse cellular functions. J Lipid Res 2024; 65:100664. [PMID: 39374805 PMCID: PMC11567036 DOI: 10.1016/j.jlr.2024.100664] [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: 03/31/2024] [Revised: 09/21/2024] [Accepted: 10/01/2024] [Indexed: 10/09/2024] Open
Abstract
Procollagen C-endopeptidase enhancer 2, known as PCPE2 or PCOC2 (gene name, PCOLCE2) is a glycoprotein that resides in the extracellular matrix, and is similar in domain organization to PCPE1/PCPE, PCOC1 (PCOLCE1/PCOLCE). Due to the many similarities between the two related proteins, PCPE2 has been assumed to have biological functions similar to PCPE. PCPE is a well-established enhancer of procollagen processing activating the enzyme, BMP-1. However, reports show that PCPE2 has a strikingly different tissue expression profile compared to PCPE. With that in mind and given the paucity of published studies on PCPE2, this review examines the current literature citing PCPE2 and its association with specific cell types and signaling pathways. Additionally, this review will present a brief history of PCPE2's discovery, highlighting structural and functional similarities and differences compared to PCPE. Considering the widespread use of RNA sequencing techniques to examine associations between cell-specific gene expression and disease states, we will show that PCPE2 is repeatedly found as a differentially regulated gene (DEG) significantly associated with a number of cellular processes, well beyond the scope of procollagen fibril processing.
Collapse
Affiliation(s)
- Michael J Thomas
- Division of Endocrinology and Molecular Medicine, Department of Pharmacology & Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA; Cardiovascular Research Center, Division of Endocrinology and Molecular Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Hao Xu
- Division of Endocrinology and Molecular Medicine, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Angela Wang
- Division of Endocrinology and Molecular Medicine, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Mirza Ahmar Beg
- Division of Endocrinology and Molecular Medicine, Department of Pharmacology & Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA; Cardiovascular Research Center, Division of Endocrinology and Molecular Medicine, Medical College of Wisconsin, Milwaukee, WI, USA; Division of Endocrinology and Molecular Medicine, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Mary G Sorci-Thomas
- Division of Endocrinology and Molecular Medicine, Department of Pharmacology & Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA; Cardiovascular Research Center, Division of Endocrinology and Molecular Medicine, Medical College of Wisconsin, Milwaukee, WI, USA; Division of Endocrinology and Molecular Medicine, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA.
| |
Collapse
|
8
|
Zhao L, Jiang C, Yu B, Zhu J, Sun Y, Yi S. Single-cell profiling of cellular changes in the somatic peripheral nerves following nerve injury. Front Pharmacol 2024; 15:1448253. [PMID: 39415832 PMCID: PMC11479879 DOI: 10.3389/fphar.2024.1448253] [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: 06/13/2024] [Accepted: 09/20/2024] [Indexed: 10/19/2024] Open
Abstract
Injury to the peripheral nervous system disconnects targets to the central nervous system, disrupts signal transmission, and results in functional disability. Although surgical and therapeutic treatments improve nerve regeneration, it is generally hard to achieve fully functional recovery after severe peripheral nerve injury. A better understanding of pathological changes after peripheral nerve injury helps the development of promising treatments for nerve regeneration. Single-cell analyses of the peripheral nervous system under physiological and injury conditions define the diversity of cells in peripheral nerves and reveal cell-specific injury responses. Herein, we review recent findings on the single-cell transcriptome status in the dorsal root ganglia and peripheral nerves following peripheral nerve injury, identify the cell heterogeneity of peripheral nerves, and delineate changes in injured peripheral nerves, especially molecular changes in neurons, glial cells, and immune cells. Cell-cell interactions in peripheral nerves are also characterized based on ligand-receptor pairs from coordinated gene expressions. The understanding of cellular changes following peripheral nerve injury at a single-cell resolution offers a comprehensive and insightful view for the peripheral nerve repair process, provides an important basis for the exploration of the key regulators of neuronal growth and microenvironment reconstruction, and benefits the development of novel therapeutic drugs for the treatment of peripheral nerve injury.
Collapse
Affiliation(s)
- Li Zhao
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Chunyi Jiang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Bin Yu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Jianwei Zhu
- Department of Orthopedic, Affiliated Hospital of Nantong University, Nantong, China
| | - Yuyu Sun
- Department of Orthopedic, Nantong Third People’s Hospital, Nantong University, Nantong, China
| | - Sheng Yi
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| |
Collapse
|
9
|
Yoo K, Jo YW, Yoo T, Hann SH, Park I, Kim YE, Kim YL, Rhee J, Song IW, Kim JH, Baek D, Kong YY. Muscle-resident mesenchymal progenitors sense and repair peripheral nerve injury via the GDNF-BDNF axis. eLife 2024; 13:RP97662. [PMID: 39324575 PMCID: PMC11426970 DOI: 10.7554/elife.97662] [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] [Indexed: 09/27/2024] Open
Abstract
Fibro-adipogenic progenitors (FAPs) are muscle-resident mesenchymal progenitors that can contribute to muscle tissue homeostasis and regeneration, as well as postnatal maturation and lifelong maintenance of the neuromuscular system. Recently, traumatic injury to the peripheral nerve was shown to activate FAPs, suggesting that FAPs can respond to nerve injury. However, questions of how FAPs can sense the anatomically distant peripheral nerve injury and whether FAPs can directly contribute to nerve regeneration remained unanswered. Here, utilizing single-cell transcriptomics and mouse models, we discovered that a subset of FAPs expressing GDNF receptors Ret and Gfra1 can respond to peripheral nerve injury by sensing GDNF secreted by Schwann cells. Upon GDNF sensing, this subset becomes activated and expresses Bdnf. FAP-specific inactivation of Bdnf (Prrx1Cre; Bdnffl/fl) resulted in delayed nerve regeneration owing to defective remyelination, indicating that GDNF-sensing FAPs play an important role in the remyelination process during peripheral nerve regeneration. In aged mice, significantly reduced Bdnf expression in FAPs was observed upon nerve injury, suggesting the clinical relevance of FAP-derived BDNF in the age-related delays in nerve regeneration. Collectively, our study revealed the previously unidentified role of FAPs in peripheral nerve regeneration, and the molecular mechanism behind FAPs' response to peripheral nerve injury.
Collapse
Affiliation(s)
- Kyusang Yoo
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Young-Woo Jo
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Takwon Yoo
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Sang-Hyeon Hann
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Inkuk Park
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Yea-Eun Kim
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Ye Lynne Kim
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Joonwoo Rhee
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - In-Wook Song
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Ji-Hoon Kim
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
- Molecular Recognition Research Center, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Daehyun Baek
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Young-Yun Kong
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
10
|
Salzer J, Feltri ML, Jacob C. Schwann Cell Development and Myelination. Cold Spring Harb Perspect Biol 2024; 16:a041360. [PMID: 38503507 PMCID: PMC11368196 DOI: 10.1101/cshperspect.a041360] [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] [Indexed: 03/21/2024]
Abstract
Glial cells in the peripheral nervous system (PNS), which arise from the neural crest, include axon-associated Schwann cells (SCs) in nerves, synapse-associated SCs at the neuromuscular junction, enteric glia, perikaryon-associated satellite cells in ganglia, and boundary cap cells at the border between the central nervous system (CNS) and the PNS. Here, we focus on axon-associated SCs. These SCs progress through a series of formative stages, which culminate in the generation of myelinating SCs that wrap large-caliber axons and of nonmyelinating (Remak) SCs that enclose multiple, small-caliber axons. In this work, we describe SC development, extrinsic signals from the axon and extracellular matrix (ECM) and the intracellular signaling pathways they activate that regulate SC development, and the morphogenesis and organization of myelinating SCs and the myelin sheath. We review the impact of SCs on the biology and integrity of axons and their emerging role in regulating peripheral nerve architecture. Finally, we explain how transcription and epigenetic factors control and fine-tune SC development and myelination.
Collapse
Affiliation(s)
- James Salzer
- Neuroscience Institute, New York University Grossman School of Medicine, New York, New York 10016, USA
| | - M Laura Feltri
- Institute for Myelin and Glia Exploration, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York 14203, USA
- IRCCS Neurological Institute Carlo Besta, Milano 20133, Italy
- Department of Biotechnology and Translational Sciences, Universita' Degli Studi di Milano, Milano 20133, Italy
| | - Claire Jacob
- Faculty of Biology, Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University Mainz, Mainz 55128, Germany
| |
Collapse
|
11
|
Xu Y, Yan Y, Zipfel GJ, MacEwan M, Ray WZ, Athiraman U. Isoflurane conditioning improves functional outcomes after peripheral nerve injury in a sciatic cut repair murine model. Front Neurol 2024; 15:1406463. [PMID: 39211813 PMCID: PMC11357975 DOI: 10.3389/fneur.2024.1406463] [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: 03/27/2024] [Accepted: 08/05/2024] [Indexed: 09/04/2024] Open
Abstract
Introduction Anesthetic conditioning has been shown to provide neuroprotection in several neurological disorders. Whether anesthetic conditioning provides protection against peripheral nerve injuries remains unknown. The aim of our current study is to investigate the impact of isoflurane conditioning on the functional outcomes after peripheral nerve injury (PNI) in a rodent sciatic nerve injury model. Methods Adult male Lewis rats underwent sciatic nerve cut and repair and exposed to none (Group 1, sham), single isoflurane exposure (Group 2), three-time isoflurane exposure (Group 3), and six-time isoflurane exposure (Group 4). Isoflurane conditioning was established by administration of 2% isoflurane for 1 hour, beginning 1-hour post sciatic nerve cut and repair. Groups 3 and 4 were exposed to isoflurane for 1 hour, 3 and 6 consecutive days respectively. Functional outcomes assessed included compound muscle action potential (CMAP), evoked muscle force (tetanic and specific tetanic force), wet muscle mass, and axonal counting. Results We observed an increase in axons, myelin width and a decrease in G-ratio in the isoflurane conditioning groups (3- and 6-days). This correlated with a significant improvement in tetanic and specific tetanic forces, observed in both groups 3 and 4. Discussion Isoflurane conditioning (3- and 6-day groups) resulted in improvement in functional outcomes at 12 weeks post peripheral nerve injury and repair in a murine model. Future experiments should be focused on identifying the therapeutic window of isoflurane conditioning and exploring the underlying molecular mechanisms responsible for isoflurane conditioning induced neuroprotection in PNI.
Collapse
Affiliation(s)
- Yameng Xu
- The Institute of Materials Science & Engineering, Washington University, St. Louis, MO, United States
| | - Ying Yan
- Department of Neurological Surgery, Washington University, St. Louis, MO, United States
| | - Gregory J. Zipfel
- Department of Neurological Surgery, Washington University, St. Louis, MO, United States
- Department of Neurology, Washington University, St. Louis, MO, United States
| | - Matthew MacEwan
- Department of Neurological Surgery, Washington University, St. Louis, MO, United States
| | - Wilson Z. Ray
- Department of Neurological Surgery, Washington University, St. Louis, MO, United States
- Department of Orthopedic Surgery, Washington University, St. Louis, MO, United States
- Department of Biomedical Engineering, Washington University, St. Louis, MO, United States
| | - Umeshkumar Athiraman
- Department of Neurological Surgery, Washington University, St. Louis, MO, United States
- Department of Anesthesiology, Washington University, St. Louis, MO, United States
| |
Collapse
|
12
|
Hu R, Dun X, Singh L, Banton MC. Runx2 regulates peripheral nerve regeneration to promote Schwann cell migration and re-myelination. Neural Regen Res 2024; 19:1575-1583. [PMID: 38051902 PMCID: PMC10883509 DOI: 10.4103/1673-5374.387977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 09/16/2023] [Indexed: 12/07/2023] Open
Abstract
Abstract
JOURNAL/nrgr/04.03/01300535-202407000-00038/figure1/v/2023-11-20T171125Z/r/image-tiff
Runx2 is a major regulator of osteoblast differentiation and function; however, the role of Runx2 in peripheral nerve repair is unclear. Here, we analyzed Runx2 expression following injury and found that it was specifically up-regulated in Schwann cells. Furthermore, using Schwann cell-specific Runx2 knockout mice, we studied peripheral nerve development and regeneration and found that multiple steps in the regeneration process following sciatic nerve injury were Runx2-dependent. Changes observed in Runx2 knockout mice include increased proliferation of Schwann cells, impaired Schwann cell migration and axonal regrowth, reduced re-myelination of axons, and a block in macrophage clearance in the late stage of regeneration. Taken together, our findings indicate that Runx2 is a key regulator of Schwann cell plasticity, and therefore peripheral nerve repair. Thus, our study shows that Runx2 plays a major role in Schwann cell migration, re-myelination, and peripheral nerve functional recovery following injury.
Collapse
Affiliation(s)
- Rong Hu
- School of Traditional Chinese Medicine, Department of Traditional Chinese Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Xinpeng Dun
- The Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Lolita Singh
- Faculty of Health, University of Plymouth, Plymouth, UK
| | | |
Collapse
|
13
|
Yuan X, Li W, Liu Q, Long Q, Yan Q, Zhang P. Genomic characteristics of adipose-derived stromal cells induced into neurons based on single-cell RNA sequencing. Heliyon 2024; 10:e33079. [PMID: 38984299 PMCID: PMC11231542 DOI: 10.1016/j.heliyon.2024.e33079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 06/13/2024] [Accepted: 06/13/2024] [Indexed: 07/11/2024] Open
Abstract
Adipose-derived stromal cells (ADSCs) can be induced to differentiate into neurons, representing the most promising avenue for cell therapy. However, the molecular mechanism and genomic characteristics of the differentiation of ADSCs into neurons remain poorly understood. In this study, cells from the adult ADSCs group, induction 1h, 3h, 5h, 6h, and 8h groups were selected for single-cell RNA sequencing (scRNA-Seq). Samples from these seven-time points were sequenced and analyzed. The expression of neuron marker genes, including NES, MAP2, TMEM59L, PTK2B, CHN1, DNM1, NRSN2, FBLN2, SCAMP1, SLC1A1, DLG4, CDK5, and ENO2, was found to be low in the ADSCs group, but highly expressed in differentiated cell clusters. The expression of stem cell marker genes, including CCND1, IL1B, MMP1, MMP3, MYO10, and BMP2, was the highest in the ADSCs cluster. This expression decreased significantly with the extension of induction time. Gene ontology (GO) enrichment analysis of upregulated genes in the induced samples showed that the biological processes related to neuronal differentiation and development, such as neuronal differentiation, projection, and apoptosis, were significantly upregulated with a longer induction time during cell cluster differentiation. The results of the cell communication analysis demonstrated the gradual formation of complex neural network connections between ADSC-derived neurons through receptor and ligand pairs at 5h after the induction of differentiation.
Collapse
Affiliation(s)
- Xiaodong Yuan
- Department of Neurology of Kailuan General Hospital Affiliated North China University of Science and Technology, China
- Hebei Provincial Key Laboratory of Neurobiological Function, China
| | - Wen Li
- Department of Neurology of Kailuan General Hospital Affiliated North China University of Science and Technology, China
- Hebei Provincial Key Laboratory of Neurobiological Function, China
| | - Qing Liu
- Department of Neurology of Kailuan General Hospital Affiliated North China University of Science and Technology, China
- Hebei Provincial Key Laboratory of Neurobiological Function, China
| | - Qingxi Long
- Department of Neurology of Kailuan General Hospital Affiliated North China University of Science and Technology, China
- Hebei Provincial Key Laboratory of Neurobiological Function, China
| | - Qi Yan
- Department of Neurology of Kailuan General Hospital Affiliated North China University of Science and Technology, China
- Hebei Provincial Key Laboratory of Neurobiological Function, China
| | - Pingshu Zhang
- Department of Neurology of Kailuan General Hospital Affiliated North China University of Science and Technology, China
- Hebei Provincial Key Laboratory of Neurobiological Function, China
| |
Collapse
|
14
|
He L, Sato JE, Sundar P, Azimi T, Beachy PA, Bekale LA, Pepper JP. Localized application of SAG21k-loaded fibrin hydrogels for targeted modulation of the hedgehog pathway in facial nerve injury. Int J Biol Macromol 2024; 269:131747. [PMID: 38670196 PMCID: PMC11774140 DOI: 10.1016/j.ijbiomac.2024.131747] [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: 04/02/2024] [Accepted: 04/20/2024] [Indexed: 04/28/2024]
Abstract
Given the broad biological effects of the Hedgehog (Hh) pathway, there is potential clinical value in local application of Hh pathway modulators to restrict pathway activation of target tissues and avoid systemic pathway activation. One option to limit Hh pathway activation is using fibrin hydrogels to deliver pathway modulators directly to tissues of interest, bypassing systemic distribution of the drug. In this study, we loaded the potent Hh pathway agonist, SAG21k, into fibrin hydrogels. We describe the binding between fibrin and SAG21k and achieve sustained release of the drug in vitro. SAG21k-loaded fibrin hydrogels exhibit strong biological activity in vitro, using a pathway-specific reporter cell line. To test in vivo activity, we used a mouse model of facial nerve injury. Application of fibrin hydrogels is a common adjunct to surgical nerve repair, and the Hh pathway is known to play an important role in facial nerve injury and regeneration. Local application of the Hh pathway agonist SAG21k using a fibrin hydrogel applied to the site of facial nerve injury successfully activates the Hh pathway in treated nerve tissue. Importantly, this method appears to avoid systemic pathway activation when Hh-responsive organs are analyzed for transcriptional pathway activation. This method of local tissue Hh pathway agonist administration allows for effective pathway targeting surgically accessible tissues and may have translational value in situations where supranormal pathway activation is therapeutic.
Collapse
Affiliation(s)
- Lili He
- Department of Otolaryngology - Head & Neck Surgery, Stanford University School of Medicine; 300 Pasteur Drive, Stanford, CA 94305, United States
| | - Justine Esther Sato
- Department of Otolaryngology - Head & Neck Surgery, Stanford University School of Medicine; 300 Pasteur Drive, Stanford, CA 94305, United States
| | - Praveen Sundar
- Department of Otolaryngology - Head & Neck Surgery, Stanford University School of Medicine; 300 Pasteur Drive, Stanford, CA 94305, United States
| | - Tannaz Azimi
- Department of Otolaryngology - Head & Neck Surgery, Stanford University School of Medicine; 300 Pasteur Drive, Stanford, CA 94305, United States
| | - Philip Arden Beachy
- Departments of Urology, and Developmental Biology, Stanford University School of Medicine, Stanford, CA, United States; Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, United States
| | - Laurent Adonis Bekale
- Department of Otolaryngology - Head & Neck Surgery, Stanford University School of Medicine; 300 Pasteur Drive, Stanford, CA 94305, United States.
| | - Jon-Paul Pepper
- Department of Otolaryngology - Head & Neck Surgery, Stanford University School of Medicine; 300 Pasteur Drive, Stanford, CA 94305, United States.
| |
Collapse
|
15
|
Wu HF, Saito-Diaz K, Huang CW, McAlpine JL, Seo DE, Magruder DS, Ishan M, Bergeron HC, Delaney WH, Santori FR, Krishnaswamy S, Hart GW, Chen YW, Hogan RJ, Liu HX, Ivanova NB, Zeltner N. Parasympathetic neurons derived from human pluripotent stem cells model human diseases and development. Cell Stem Cell 2024; 31:734-753.e8. [PMID: 38608707 PMCID: PMC11069445 DOI: 10.1016/j.stem.2024.03.011] [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: 01/02/2023] [Revised: 01/16/2024] [Accepted: 03/13/2024] [Indexed: 04/14/2024]
Abstract
Autonomic parasympathetic neurons (parasymNs) control unconscious body responses, including "rest-and-digest." ParasymN innervation is important for organ development, and parasymN dysfunction is a hallmark of autonomic neuropathy. However, parasymN function and dysfunction in humans are vastly understudied due to the lack of a model system. Human pluripotent stem cell (hPSC)-derived neurons can fill this void as a versatile platform. Here, we developed a differentiation paradigm detailing the derivation of functional human parasymNs from Schwann cell progenitors. We employ these neurons (1) to assess human autonomic nervous system (ANS) development, (2) to model neuropathy in the genetic disorder familial dysautonomia (FD), (3) to show parasymN dysfunction during SARS-CoV-2 infection, (4) to model the autoimmune disease Sjögren's syndrome (SS), and (5) to show that parasymNs innervate white adipocytes (WATs) during development and promote WAT maturation. Our model system could become instrumental for future disease modeling and drug discovery studies, as well as for human developmental studies.
Collapse
Affiliation(s)
- Hsueh-Fu Wu
- Center for Molecular Medicine, University of Georgia, Athens, GA 30602, USA; Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA 30602, USA
| | - Kenyi Saito-Diaz
- Center for Molecular Medicine, University of Georgia, Athens, GA 30602, USA
| | - Chia-Wei Huang
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA 30602, USA; Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA
| | - Jessica L McAlpine
- Center for Molecular Medicine, University of Georgia, Athens, GA 30602, USA; Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA 30602, USA
| | - Dong Eun Seo
- Center for Molecular Medicine, University of Georgia, Athens, GA 30602, USA; Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA 30602, USA
| | - D Sumner Magruder
- Department of Genetics, Department of Computer Science, Wu Tsai Institute, Program for Computational Biology and Bioinformatics, Yale University, New Haven, CT 06520, USA
| | - Mohamed Ishan
- Regenerative Bioscience Center, Department of Animal and Dairy Science College of Agricultural and Environmental Sciences, University of Georgia, Athens, GA 30602, USA
| | - Harrison C Bergeron
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
| | - William H Delaney
- Center for Molecular Medicine, University of Georgia, Athens, GA 30602, USA
| | - Fabio R Santori
- Center for Molecular Medicine, University of Georgia, Athens, GA 30602, USA
| | - Smita Krishnaswamy
- Department of Genetics, Department of Computer Science, Wu Tsai Institute, Program for Computational Biology and Bioinformatics, Yale University, New Haven, CT 06520, USA
| | - Gerald W Hart
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA 30602, USA; Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA
| | - Ya-Wen Chen
- Department of Otolaryngology, Department of Cell, Developmental, and Regenerative Biology, Institute for Airway Sciences, Institute for Regenerative Medicine, Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Robert J Hogan
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA; Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
| | - Hong-Xiang Liu
- Regenerative Bioscience Center, Department of Animal and Dairy Science College of Agricultural and Environmental Sciences, University of Georgia, Athens, GA 30602, USA
| | - Natalia B Ivanova
- Center for Molecular Medicine, University of Georgia, Athens, GA 30602, USA; Department of Genetics, University of Georgia, Athens, GA 30602, USA
| | - Nadja Zeltner
- Center for Molecular Medicine, University of Georgia, Athens, GA 30602, USA; Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA 30602, USA; Department of Cellular Biology, University of Georgia, Athens, GA 30602, USA.
| |
Collapse
|
16
|
Yeo E, Shim J, Oh SJ, Choi Y, Noh H, Kim H, Park JH, Lee KT, Kim SH, Lee D, Lee JH. Revisiting roles of mast cells and neural cells in keloid: exploring their connection to disease activity. Front Immunol 2024; 15:1339336. [PMID: 38524141 PMCID: PMC10957560 DOI: 10.3389/fimmu.2024.1339336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 02/19/2024] [Indexed: 03/26/2024] Open
Abstract
Background Mast cells (MCs) and neural cells (NCs) are important in a keloid microenvironment. They might contribute to fibrosis and pain sensation within the keloid. However, their involvement in pathological excessive scarring has not been adequately explored. Objectives To elucidate roles of MCs and NCs in keloid pathogenesis and their correlation with disease activity. Methods Keloid samples from chest and back regions were analyzed. Single-cell RNA sequencing (scRNA-seq) was conducted for six active keloids (AK) samples, four inactive keloids (IK) samples, and three mature scar (MS) samples from patients with keloids. Results The scRNA-seq analysis demonstrated notable enrichment of MCs, lymphocytes, and macrophages in AKs, which exhibited continuous growth at the excision site when compared to IK and MS samples (P = 0.042). Expression levels of marker genes associated with activated and degranulated MCs, including FCER1G, BTK, and GATA2, were specifically elevated in keloid lesions. Notably, MCs within AK lesions exhibited elevated expression of genes such as NTRK1, S1PR1, and S1PR2 associated with neuropeptide receptors. Neural progenitor cell and non-myelinating Schwann cell (nmSC) genes were highly expressed in keloids, whereas myelinating Schwann cell (mSC) genes were specific to MS samples. Conclusions scRNA-seq analyses of AK, IK, and MS samples unveiled substantial microenvironmental heterogeneity. Such heterogeneity might be linked to disease activity. These findings suggest the potential contribution of MCs and NCs to keloid pathogenesis. Histopathological and molecular features observed in AK and IK samples provide valuable insights into the mechanisms underlying pain and pruritus in keloid lesions.
Collapse
Affiliation(s)
- Eunhye Yeo
- Department of Dermatology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Department of Medical Device Management and Research, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, Republic of Korea
| | - Joonho Shim
- Department of Dermatology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Se Jin Oh
- Department of Dermatology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - YoungHwan Choi
- Department of Dermatology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Department of Medical Device Management and Research, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, Republic of Korea
| | - Hyungrye Noh
- Department of Dermatology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Heeyeon Kim
- Department of Dermatology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Ji-Hye Park
- Department of Dermatology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Kyeong-Tae Lee
- Department of Plastic Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Seok-Hyung Kim
- Department of Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Dongyoun Lee
- Department of Dermatology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jong Hee Lee
- Department of Dermatology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Department of Medical Device Management and Research, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, Republic of Korea
| |
Collapse
|
17
|
Zou X, Dong Y, Alhaskawi A, Zhou H, Ezzi SHA, Kota VG, Abdulla MHAH, Abdalbary SA, Lu H, Wang C. Techniques and graft materials for repairing peripheral nerve defects. Front Neurol 2024; 14:1307883. [PMID: 38318237 PMCID: PMC10839026 DOI: 10.3389/fneur.2023.1307883] [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: 10/05/2023] [Accepted: 12/15/2023] [Indexed: 02/07/2024] Open
Abstract
Peripheral nerve defects refer to damage or destruction occurring in the peripheral nervous system, typically affecting the limbs and face. The current primary approaches to address peripheral nerve defects involve the utilization of autologous nerve transplants or the transplantation of artificial material. Nevertheless, these methods possess certain limitations, such as inadequate availability of donor nerve or unsatisfactory regenerative outcomes post-transplantation. Biomaterials have been extensively studied as an alternative approach to promote the repair of peripheral neve defects. These biomaterials include both natural and synthetic materials. Natural materials consist of collagen, chitosan, and silk, while synthetic materials consist of polyurethane, polylactic acid, and polycaprolactone. Recently, several new neural repair technologies have also been developed, such as nerve regeneration bridging technology, electrical stimulation technology, and stem cell therapy technology. Overall, biomaterials and new neural repair technologies provide new methods and opportunities for repairing peripheral nerve defects. However, these methods still require further research and development to enhance their effectiveness and feasibility.
Collapse
Affiliation(s)
- Xiaodi Zou
- Department of Orthopedics, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Yanzhao Dong
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Ahmad Alhaskawi
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Haiying Zhou
- Faculty of Medicine, The Chinese University of Hong Kong School of Biomedical Science, Shatin, China
| | | | | | | | - Sahar Ahmed Abdalbary
- Department of Orthopedic Physical Therapy, Faculty of Physical Therapy, Nahda University in Beni Suef, Beni Suef, Egypt
| | - Hui Lu
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University, Hangzhou, China
- Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Zhejiang University, Hangzhou, China
| | - Changxin Wang
- Department of Orthopedics, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
18
|
Bhat GP, Maurizio A, Motta A, Podini P, Diprima S, Malpighi C, Brambilla I, Martins L, Badaloni A, Boselli D, Bianchi F, Pellegatta M, Genua M, Ostuni R, Del Carro U, Taveggia C, de Pretis S, Quattrini A, Bonanomi D. Structured wound angiogenesis instructs mesenchymal barrier compartments in the regenerating nerve. Neuron 2024; 112:209-229.e11. [PMID: 37972594 DOI: 10.1016/j.neuron.2023.10.025] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 09/19/2023] [Accepted: 10/18/2023] [Indexed: 11/19/2023]
Abstract
Organ injury stimulates the formation of new capillaries to restore blood supply raising questions about the potential contribution of neoangiogenic vessel architecture to the healing process. Using single-cell mapping, we resolved the properties of endothelial cells that organize a polarized scaffold at the repair site of lesioned peripheral nerves. Transient reactivation of an embryonic guidance program is required to orient neovessels across the wound. Manipulation of this structured angiogenic response through genetic and pharmacological targeting of Plexin-D1/VEGF pathways within an early window of repair has long-term impact on configuration of the nerve stroma. Neovessels direct nerve-resident mesenchymal cells to mold a provisionary fibrotic scar by assembling an orderly system of stable barrier compartments that channel regenerating nerve fibers and shield them from the persistently leaky vasculature. Thus, guided and balanced repair angiogenesis enables the construction of a "bridge" microenvironment conducive for axon regrowth and homeostasis of the regenerated tissue.
Collapse
Affiliation(s)
- Ganesh Parameshwar Bhat
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, via Olgettina 60, 20132 Milan, Italy
| | - Aurora Maurizio
- Center for Omics Sciences, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Alessia Motta
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, via Olgettina 60, 20132 Milan, Italy
| | - Paola Podini
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, via Olgettina 60, 20132 Milan, Italy; Institute of Experimental Neurology, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Santo Diprima
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, via Olgettina 60, 20132 Milan, Italy; Center for Omics Sciences, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Chiara Malpighi
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, via Olgettina 60, 20132 Milan, Italy
| | - Ilaria Brambilla
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, via Olgettina 60, 20132 Milan, Italy
| | - Luis Martins
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, via Olgettina 60, 20132 Milan, Italy
| | - Aurora Badaloni
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, via Olgettina 60, 20132 Milan, Italy
| | - Daniela Boselli
- FRACTAL-Flow cytometry Resource Advanced Cytometry Technical Applications Laboratory, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Francesca Bianchi
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, via Olgettina 60, 20132 Milan, Italy
| | - Marta Pellegatta
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, via Olgettina 60, 20132 Milan, Italy
| | - Marco Genua
- San Raffaele Telethon Institute for Gene therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Renato Ostuni
- San Raffaele Telethon Institute for Gene therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Ubaldo Del Carro
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, via Olgettina 60, 20132 Milan, Italy
| | - Carla Taveggia
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, via Olgettina 60, 20132 Milan, Italy
| | - Stefano de Pretis
- Center for Omics Sciences, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Angelo Quattrini
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, via Olgettina 60, 20132 Milan, Italy; Institute of Experimental Neurology, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Dario Bonanomi
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, via Olgettina 60, 20132 Milan, Italy.
| |
Collapse
|
19
|
Jaafar AK, Techer R, Chemello K, Lambert G, Bourane S. PCSK9 and the nervous system: a no-brainer? J Lipid Res 2023; 64:100426. [PMID: 37586604 PMCID: PMC10491654 DOI: 10.1016/j.jlr.2023.100426] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/08/2023] [Accepted: 08/10/2023] [Indexed: 08/18/2023] Open
Abstract
In the past 20 years, PCSK9 has been shown to play a pivotal role in LDL cholesterol metabolism and cardiovascular health by inducing the lysosomal degradation of the LDL receptor. PCSK9 was discovered by the cloning of genes up-regulated after apoptosis induced by serum deprivation in primary cerebellar neurons, but despite its initial identification in the brain, the precise role of PCSK9 in the nervous system remains to be clearly established. The present article is a comprehensive review of studies published or in print before July 2023 that have investigated the expression pattern of PCSK9, its effects on lipid metabolism as well as its putative roles specifically in the central and peripheral nervous systems, with a special focus on cerebrovascular and neurodegenerative diseases.
Collapse
Affiliation(s)
- Ali K Jaafar
- Laboratoire Inserm UMR 1188 DéTROI, Saint-Pierre, La Réunion, France
| | - Romuald Techer
- Laboratoire Inserm UMR 1188 DéTROI, Saint-Pierre, La Réunion, France
| | - Kévin Chemello
- Laboratoire Inserm UMR 1188 DéTROI, Saint-Pierre, La Réunion, France
| | - Gilles Lambert
- Laboratoire Inserm UMR 1188 DéTROI, Saint-Pierre, La Réunion, France; Faculté de Médecine, Université de La Réunion, Saint-Pierre, La Réunion, France.
| | - Steeve Bourane
- Laboratoire Inserm UMR 1188 DéTROI, Saint-Pierre, La Réunion, France
| |
Collapse
|
20
|
Zhang X, Lu P, Shen X. Morphologies and potential roles of telocytes in nervous tissue. CLINICAL AND TRANSLATIONAL DISCOVERY 2023; 3. [DOI: 10.1002/ctd2.186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 03/16/2023] [Indexed: 01/03/2025]
Abstract
AbstractStructurally similar cells have been found and termed telocytes (TCs) since the first characterisation of interstitial Cajal‐like cells in 1911. TCs are a novel and peculiar interstitial cell type with a small cellular body, markedly long cell processes named telopodes and a wide distribution in numerous tissues throughout the body. Besides specific morphological characteristics and immunohistochemical profiles, TCs build three‐dimensional mixed networks through homocellular (connection to each other) and/or heterocellular contacts (connection with other cell types), interaction with extracellular matrix and their vicinity to nerve endings, and thus might play, as part of an integrated system, roles in maintaining organ/tissue function. In this mini‐review, we summarise physical properties, general characteristics and distribution of TCs in diverse organs and tissues, focusing on their potential functions in nervous tissue and current challenges in investigating TCs as a distinct cell type.
Collapse
Affiliation(s)
- Xu Zhang
- Institute for Reproductive Health Shanghai Institute for Biomedical and Pharmaceutical Technologies Shanghai China
| | - Ping Lu
- Institute for Reproductive Health Shanghai Institute for Biomedical and Pharmaceutical Technologies Shanghai China
| | - Xiaorong Shen
- Institute for Reproductive Health Shanghai Institute for Biomedical and Pharmaceutical Technologies Shanghai China
| |
Collapse
|
21
|
Zhao L, Huang W, Yi S. Cellular complexity of the peripheral nervous system: Insights from single-cell resolution. Front Neurosci 2023; 17:1098612. [PMID: 36998728 PMCID: PMC10043217 DOI: 10.3389/fnins.2023.1098612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 02/21/2023] [Indexed: 03/15/2023] Open
Abstract
Single-cell RNA sequencing allows the division of cell populations, offers precise transcriptional profiling of individual cells, and fundamentally advances the comprehension of cellular diversity. In the peripheral nervous system (PNS), the application of single-cell RNA sequencing identifies multiple types of cells, including neurons, glial cells, ependymal cells, immune cells, and vascular cells. Sub-types of neurons and glial cells have further been recognized in nerve tissues, especially tissues in different physiological and pathological states. In the current article, we compile the heterogeneities of cells that have been reported in the PNS and describe cellular variability during development and regeneration. The discovery of the architecture of peripheral nerves benefits the understanding of the cellular complexity of the PNS and provides a considerable cellular basis for future genetic manipulation.
Collapse
Affiliation(s)
- Lili Zhao
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, China
- NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu, China
| | - Weixiao Huang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, China
- NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu, China
- School of Medicine and Life Sciences, Nanjing University of Chinese Medicine, Nanjing, China
| | - Sheng Yi
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, China
- NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu, China
- *Correspondence: Sheng Yi,
| |
Collapse
|
22
|
Iron Metabolism and Ferroptosis in Peripheral Nerve Injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:5918218. [PMID: 36506935 PMCID: PMC9733998 DOI: 10.1155/2022/5918218] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 11/07/2022] [Accepted: 11/21/2022] [Indexed: 12/04/2022]
Abstract
Peripheral nerve injury (PNI) is a major clinical problem that may lead to different levels of sensory and motor dysfunction including paralysis. Due to the high disability rate and unsatisfactory prognosis, the exploration and revealment of the mechanisms involved in the PNI are urgently required. Ferroptosis, a recently identified novel form of cell death, is an iron-dependent process. It is a unique modality of cell death, closely associated with iron concentrations, generation of reactive oxygen species, and accumulation of the lipid reactive oxygen species. These processes are regulated by multiple cellular metabolic pathways, including iron overloading, lipid peroxidation, and the glutathione/glutathione peroxidase 4 pathway. Furthermore, ferroptosis is accompanied by morphological changes in the mitochondria, such as increased membrane density and shrunken mitochondria; this association between ferroptosis and mitochondrial damage has been detected in various diseases, including spinal cord injury and PNI. The inhibition of ferroptosis can promote the repair of damaged peripheral nerves, reduce mitochondrial damage, and promote the recovery of neurological function. In this review, we intend to discuss the detailed mechanisms of ferroptosis and summarize the current researches on ferroptosis with respect to nerve injury. This review also aims at providing new insights on targeting ferroptosis for PNI treatment.
Collapse
|
23
|
Chau MJ, Quintero JE, Blalock E, Byrum S, Mackintosh SG, Samaan C, Gerhardt GA, van Horne CG. Transection injury differentially alters the proteome of the human sural nerve. PLoS One 2022; 17:e0260998. [PMID: 36417411 PMCID: PMC9683555 DOI: 10.1371/journal.pone.0260998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 09/09/2022] [Indexed: 11/25/2022] Open
Abstract
Regeneration after severe peripheral nerve injury is often poor. Knowledge of human nerve regeneration and the growth microenvironment is greatly lacking. We aimed to identify the regenerative proteins in human peripheral nerve by comparing the proteome before and after a transection injury. In a unique study design, we collected closely matched samples of naïve and injured sural nerve. Naïve and injured (two weeks after injury) samples were analyzed using mass spectrometry and immunoassays. We found significantly altered levels following the nerve injury. Mass spectrometry revealed that injury samples had 568 proteins significantly upregulated and 471 significantly downregulated compared to naïve samples (q-value ≤ 0.05 and Z ≥ |2| (log2)). We used Gene Ontology (GO) pathway overrepresentation analysis to highlight groups of proteins that were significantly upregulated or downregulated with injury-induced degeneration and regeneration. Significant protein changes in key pathways were identified including growth factor levels, Schwann cell de-differentiation, myelination downregulation, epithelial-mesenchymal transition (EMT), and axonal regeneration pathways. The proteomes of the uninjured nerve compared to the degenerating/regenerating nerve may reveal biomarkers to aid in the development of repair strategies such as infusing supplemental trophic factors and in monitoring neural tissue regeneration.
Collapse
Affiliation(s)
- Monica J. Chau
- Brain Restoration Center, College of Medicine, University of Kentucky, Lexington, KY, United States of America
- Department of Neurosurgery, College of Medicine, University of Kentucky, Lexington, KY, United States of America
| | - Jorge E. Quintero
- Brain Restoration Center, College of Medicine, University of Kentucky, Lexington, KY, United States of America
- Department of Neurosurgery, College of Medicine, University of Kentucky, Lexington, KY, United States of America
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, United States of America
| | - Eric Blalock
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY, United States of America
| | - Stephanie Byrum
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR, United States of America
| | - Samuel G. Mackintosh
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR, United States of America
| | - Christopher Samaan
- Brain Restoration Center, College of Medicine, University of Kentucky, Lexington, KY, United States of America
- Department of Neurosurgery, College of Medicine, University of Kentucky, Lexington, KY, United States of America
| | - Greg A. Gerhardt
- Brain Restoration Center, College of Medicine, University of Kentucky, Lexington, KY, United States of America
- Department of Neurosurgery, College of Medicine, University of Kentucky, Lexington, KY, United States of America
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, United States of America
- Department of Neurology, College of Medicine, University of Kentucky, Lexington, KY, United States of America
| | - Craig G. van Horne
- Brain Restoration Center, College of Medicine, University of Kentucky, Lexington, KY, United States of America
- Department of Neurosurgery, College of Medicine, University of Kentucky, Lexington, KY, United States of America
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, United States of America
- * E-mail:
| |
Collapse
|
24
|
Hermans F, Bueds C, Hemeryck L, Lambrichts I, Bronckaers A, Vankelecom H. Establishment of inclusive single-cell transcriptome atlases from mouse and human tooth as powerful resource for dental research. Front Cell Dev Biol 2022; 10:1021459. [PMID: 36299483 PMCID: PMC9590651 DOI: 10.3389/fcell.2022.1021459] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 09/12/2022] [Indexed: 11/13/2022] Open
Abstract
Single-cell (sc) omics has become a powerful tool to unravel a tissue's cell landscape across health and disease. In recent years, sc transcriptomic interrogation has been applied to a variety of tooth tissues of both human and mouse, which has considerably advanced our fundamental understanding of tooth biology. Now, an overarching and integrated bird's-view of the human and mouse tooth sc transcriptomic landscape would be a powerful multi-faceted tool for dental research, enabling further decipherment of tooth biology and development through constantly progressing state-of-the-art bioinformatic methods as well as the exploration of novel hypothesis-driven research. To this aim, we re-assessed and integrated recently published scRNA-sequencing datasets of different dental tissue types (healthy and diseased) from human and mouse to establish inclusive tooth sc atlases, and applied the consolidated data map to explore its power. For mouse tooth, we identified novel candidate transcriptional regulators of the ameloblast lineage. Regarding human tooth, we provide support for a developmental connection, not advanced before, between specific epithelial compartments. Taken together, we established inclusive mouse and human tooth sc atlases as powerful tools to potentiate innovative research into tooth biology, development and disease. The maps are provided online in an accessible format for interactive exploration.
Collapse
Affiliation(s)
- Florian Hermans
- Laboratory of Tissue Plasticity in Health and Disease, Cluster of Stem Cell and Developmental Biology, Department of Development and Regeneration, Leuven Stem Cell Institute, KU Leuven (University of Leuven), Leuven, Belgium
- UHasselt-Hasselt University, Biomedical Research Institute (BIOMED), Department of Cardio and Organ Systems, Diepenbeek, Belgium
| | - Celine Bueds
- Laboratory of Tissue Plasticity in Health and Disease, Cluster of Stem Cell and Developmental Biology, Department of Development and Regeneration, Leuven Stem Cell Institute, KU Leuven (University of Leuven), Leuven, Belgium
| | - Lara Hemeryck
- Laboratory of Tissue Plasticity in Health and Disease, Cluster of Stem Cell and Developmental Biology, Department of Development and Regeneration, Leuven Stem Cell Institute, KU Leuven (University of Leuven), Leuven, Belgium
| | - Ivo Lambrichts
- UHasselt-Hasselt University, Biomedical Research Institute (BIOMED), Department of Cardio and Organ Systems, Diepenbeek, Belgium
| | - Annelies Bronckaers
- UHasselt-Hasselt University, Biomedical Research Institute (BIOMED), Department of Cardio and Organ Systems, Diepenbeek, Belgium
| | - Hugo Vankelecom
- Laboratory of Tissue Plasticity in Health and Disease, Cluster of Stem Cell and Developmental Biology, Department of Development and Regeneration, Leuven Stem Cell Institute, KU Leuven (University of Leuven), Leuven, Belgium
| |
Collapse
|
25
|
Ramesh R, Manurung Y, Ma KH, Blakely T, Won S, Moreno-Ramos OA, Wyatt E, Awatramani R, Svaren J. JUN Regulation of Injury-Induced Enhancers in Schwann Cells. J Neurosci 2022; 42:6506-6517. [PMID: 35906072 PMCID: PMC9410756 DOI: 10.1523/jneurosci.2533-21.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 06/22/2022] [Accepted: 07/14/2022] [Indexed: 11/21/2022] Open
Abstract
Schwann cells play a critical role after peripheral nerve injury by clearing myelin debris, forming axon-guiding bands of Büngner, and remyelinating regenerating axons. Schwann cells undergo epigenomic remodeling to differentiate into a repair state that expresses unique genes, some of which are not expressed at other stages of Schwann cell development. We previously identified a set of enhancers that are activated in Schwann cells after nerve injury, and we determined whether these enhancers are preprogrammed into the Schwann cell epigenome as poised enhancers before injury. Poised enhancers share many attributes of active enhancers, such as open chromatin, but are marked by repressive histone H3 lysine 27 (H3K27) trimethylation rather than H3K27 acetylation. We find that most injury-induced enhancers are not marked as poised enhancers before injury indicating that injury-induced enhancers are not preprogrammed in the Schwann cell epigenome. Injury-induced enhancers are enriched with AP-1 binding motifs, and the c-JUN subunit of AP-1 had been shown to be critical to drive the transcriptional response of Schwann cells after injury. Using in vivo chromatin immunoprecipitation sequencing analysis in rat, we find that c-JUN binds to a subset of injury-induced enhancers. To test the role of specific injury-induced enhancers, we focused on c-JUN-binding enhancers upstream of the Sonic hedgehog (Shh) gene, which is only upregulated in repair Schwann cells compared with other stages of Schwann cell development. We used targeted deletions in male/female mice to show that the enhancers are required for robust induction of the Shh gene after injury.SIGNIFICANCE STATEMENT The proregenerative actions of Schwann cells after nerve injury depends on profound reprogramming of the epigenome. The repair state is directed by injury-induced transcription factors, like JUN, which is uniquely required after nerve injury. In this study, we test whether the injury program is preprogrammed into the epigenome as poised enhancers and define which enhancers bind JUN. Finally, we test the roles of these enhancers by performing clustered regularly interspaced short palindromic repeat (CRISPR)-mediated deletion of JUN-bound injury enhancers in the Sonic hedgehog gene. Although many long-range enhancers drive expression of Sonic hedgehog at different developmental stages of specific tissues, these studies identify an entirely new set of enhancers that are required for Sonic hedgehog induction in Schwann cells after injury.
Collapse
Affiliation(s)
- Raghu Ramesh
- Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin 53705
- Comparative Biomedical Sciences Graduate Program, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin 53706
| | - Yanti Manurung
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611
| | - Ki H Ma
- Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin 53705
| | - Todd Blakely
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611
| | - Seongsik Won
- Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin 53705
| | - Oscar Andrés Moreno-Ramos
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611
| | - Eugene Wyatt
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611
| | - Rajeshwar Awatramani
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611
| | - John Svaren
- Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin 53705
- Comparative Biomedical Sciences Graduate Program, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin 53706
- Department of Comparative Biosciences, School of Veterinary Medicine University of Wisconsin-Madison, Madison, Wisconsin 53705
| |
Collapse
|
26
|
Johnston APW, Miller FD. The Contribution of Innervation to Tissue Repair and Regeneration. Cold Spring Harb Perspect Biol 2022; 14:a041233. [PMID: 35667791 PMCID: PMC9438784 DOI: 10.1101/cshperspect.a041233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Animals such as amphibians have an incredible capacity for regeneration with some being able to regrow their tail or appendages. Although some mammalian tissues like the skin and bones can repair following injury, there are only a few examples of true multilineage regeneration, including the distal portion of the digit tip. In both amphibians and mammals, however, to achieve successful repair or regeneration, it is now appreciated that intact nerve innervation is a necessity. Here, we review the current state of literature and discuss recent advances that identify axon-derived signals, Schwann cells, and nerve-derived mesenchymal cells as direct and indirect supporters of adult tissue homeostasis and repair. We posit that understanding how nerves positively influence repair and regeneration could lead to targeted regenerative medicine strategies to enhance tissue repair in humans.
Collapse
Affiliation(s)
- Adam P W Johnston
- Department of Applied Human Sciences; Department of Biomedical Sciences, University of Prince Edward Island, Charlottetown, Prince Edward Island C1A 4P3, Canada
| | - Freda D Miller
- Michael Smith Laboratories; Department of Medical Genetics; School of Biomedical Engineering, University of British Columbia, Vancouver V6T 1Z3, Canada
| |
Collapse
|
27
|
Li Y, Xu S, Ma S, Wu M. Network-based cancer heterogeneity analysis incorporating multi-view of prior information. Bioinformatics 2022; 38:2855-2862. [PMID: 35561185 PMCID: PMC9113254 DOI: 10.1093/bioinformatics/btac183] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 02/22/2022] [Accepted: 03/22/2022] [Indexed: 02/03/2023] Open
Abstract
MOTIVATION Cancer genetic heterogeneity analysis has critical implications for tumour classification, response to therapy and choice of biomarkers to guide personalized cancer medicine. However, existing heterogeneity analysis based solely on molecular profiling data usually suffers from a lack of information and has limited effectiveness. Many biomedical and life sciences databases have accumulated a substantial volume of meaningful biological information. They can provide additional information beyond molecular profiling data, yet pose challenges arising from potential noise and uncertainty. RESULTS In this study, we aim to develop a more effective heterogeneity analysis method with the help of prior information. A network-based penalization technique is proposed to innovatively incorporate a multi-view of prior information from multiple databases, which accommodates heterogeneity attributed to both differential genes and gene relationships. To account for the fact that the prior information might not be fully credible, we propose a weighted strategy, where the weight is determined dependent on the data and can ensure that the present model is not excessively disturbed by incorrect information. Simulation and analysis of The Cancer Genome Atlas glioblastoma multiforme data demonstrate the practical applicability of the proposed method. AVAILABILITY AND IMPLEMENTATION R code implementing the proposed method is available at https://github.com/mengyunwu2020/PECM. The data that support the findings in this paper are openly available in TCGA (The Cancer Genome Atlas) at https://portal.gdc.cancer.gov/. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Yang Li
- Center for Applied Statistics, School of Statistics, Statistical Consulting Center, and RSS and China-Re Life Joint Lab on Public Health and Risk Management, Renmin University of China, Beijing 100872, China
| | - Shaodong Xu
- Center for Applied Statistics, School of Statistics, Statistical Consulting Center, and RSS and China-Re Life Joint Lab on Public Health and Risk Management, Renmin University of China, Beijing 100872, China
| | - Shuangge Ma
- Department of Biostatistics, Yale School of Public Health, New Haven, CT 06520, USA
| | - Mengyun Wu
- School of Statistics and Management, Shanghai University of Finance and Economics, Shanghai 200433, China
| |
Collapse
|
28
|
Brosius Lutz A, Lucas TA, Carson GA, Caneda C, Zhou L, Barres BA, Buckwalter MS, Sloan SA. An RNA-sequencing transcriptome of the rodent Schwann cell response to peripheral nerve injury. J Neuroinflammation 2022; 19:105. [PMID: 35501870 PMCID: PMC9063194 DOI: 10.1186/s12974-022-02462-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 04/13/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The important contribution of glia to mechanisms of injury and repair of the nervous system is increasingly recognized. In stark contrast to the central nervous system (CNS), the peripheral nervous system (PNS) has a remarkable capacity for regeneration after injury. Schwann cells are recognized as key contributors to PNS regeneration, but the molecular underpinnings of the Schwann cell response to injury and how they interact with the inflammatory response remain incompletely understood. METHODS We completed bulk RNA-sequencing of Schwann cells purified acutely using immunopanning from the naïve and injured rodent sciatic nerve at 3, 5, and 7 days post-injury. We used qRT-PCR and in situ hybridization to assess cell purity and probe dataset integrity. Finally, we used bioinformatic analysis to probe Schwann cell-specific injury-induced modulation of cellular pathways. RESULTS Our data confirm Schwann cell purity and validate RNAseq dataset integrity. Bioinformatic analysis identifies discrete modules of genes that follow distinct patterns of regulation in the 1st days after injury and their corresponding molecular pathways. These findings enable improved differentiation of myeloid and glial components of neuroinflammation after peripheral nerve injury and highlight novel molecular aspects of the Schwann cell injury response such as acute downregulation of the AGE/RAGE pathway and of secreted molecules Sparcl1 and Sema5a. CONCLUSIONS We provide a helpful resource for further deciphering the Schwann cell injury response and a depth of transcriptional data that can complement the findings of recent single cell sequencing approaches. As more data become available on the response of CNS glia to injury, we anticipate that this dataset will provide a valuable platform for understanding key differences in the PNS and CNS glial responses to injury and for designing approaches to ameliorate CNS regeneration.
Collapse
Affiliation(s)
- Amanda Brosius Lutz
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA, 94305-5125, USA.
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA, 94305-5125, USA.
- Department of Obstetrics and Gynecology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland.
| | - Tawaun A Lucas
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, 94305-5125, USA
| | - Glenn A Carson
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA, 94305-5125, USA
| | - Christine Caneda
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA, 94305-5125, USA
| | - Lu Zhou
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA, 94305-5125, USA
| | - Ben A Barres
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA, 94305-5125, USA
| | - Marion S Buckwalter
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, 94305-5125, USA
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, 94305-5125, USA
| | - Steven A Sloan
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA, 94305-5125, USA
- Department of Human Genetics, Emory University, 30322, Atlanta, Georgia
| |
Collapse
|
29
|
Formation of the Mouse Internal Capsule and Cerebral Peduncle: A Pioneering Role for Striatonigral Axons as Revealed in Isl1 Conditional Mutants. J Neurosci 2022; 42:3344-3364. [PMID: 35273083 PMCID: PMC9034787 DOI: 10.1523/jneurosci.2291-21.2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 02/09/2022] [Accepted: 02/13/2022] [Indexed: 01/05/2023] Open
Abstract
The projection neurons of the striatum, the principal nucleus of the basal ganglia, belong to one of the following two major pathways: the striatopallidal (indirect) pathway or the striatonigral (direct) pathway. Striatonigral axons project long distances and encounter ascending tracts (thalamocortical) while coursing alongside descending tracts (corticofugal) as they extend through the internal capsule and cerebral peduncle. These observations suggest that striatal circuitry may help to guide their trajectories. To investigate the developmental contributions of striatonigral axons to internal capsule formation, we have made use of Sox8-EGFP (striatal direct pathway) and Fezf2-TdTomato (corticofugal pathway) BAC transgenic reporter mice in combination with immunohistochemical markers to trace these axonal pathways throughout development. We show that striatonigral axons pioneer the internal capsule and cerebral peduncle and are temporally and spatially well positioned to provide guidance for corticofugal and thalamocortical axons. Using Isl1 conditional knock-out (cKO) mice, which exhibit disrupted striatonigral axon outgrowth, we observe both corticofugal and thalamocortical axon defects with either ventral forebrain- or telencephalon-specific Isl1 inactivation, despite Isl1 not being expressed in either cortical or thalamic projection neurons. Striatonigral axon defects can thus disrupt internal capsule formation. Our genome-wide transcriptomic analysis in Isl1 cKOs reveals changes in gene expression relevant to cell adhesion, growth cone dynamics, and extracellular matrix composition, suggesting potential mechanisms by which the striatonigral pathway exerts this guidance role. Together, our data support a novel pioneering role for the striatal direct pathway in the correct assembly of the ascending and descending axon tracts within the internal capsule and cerebral peduncle.SIGNIFICANCE STATEMENT The basal ganglia are a group of subcortical nuclei with established roles in the coordination of voluntary motor programs, aspects of cognition, and the selection of appropriate social behaviors. Hence, disruptions in basal ganglia connectivity have been implicated in the motor, cognitive, and social dysfunction characterizing common neurodevelopmental disorders such as attention-deficit/hyperactivity disorder, autism spectrum disorder, obsessive-compulsive disorder, and tic disorder. Here, we identified a novel role for the striatonigral (direct) pathway in pioneering the internal capsule and cerebral peduncle, and in guiding axons extending to and from the cortex. Our findings suggest that the abnormal development of basal ganglia circuits can drive secondary internal capsule defects and thereby may contribute to the pathology of these disorders.
Collapse
|
30
|
Yim AKY, Wang PL, Bermingham JR, Hackett A, Strickland A, Miller TM, Ly C, Mitra RD, Milbrandt J. Disentangling glial diversity in peripheral nerves at single-nuclei resolution. Nat Neurosci 2022; 25:238-251. [PMID: 35115729 PMCID: PMC9060899 DOI: 10.1038/s41593-021-01005-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 12/14/2021] [Indexed: 11/09/2022]
Abstract
The peripheral nerve contains diverse cell types that support its proper function and maintenance. In this study, we analyzed multiple peripheral nerves using single-nuclei RNA sequencing, which allowed us to circumvent difficulties encountered in analyzing cells with complex morphologies via conventional single-cell methods. The resultant mouse peripheral nerve cell atlas highlights a diversity of cell types, including multiple subtypes of Schwann cells (SCs), immune cells and stromal cells. We identified a distinct myelinating SC subtype that expresses Cldn14, Adamtsl1 and Pmp2 and preferentially ensheathes motor axons. The number of these motor-associated Pmp2+ SCs is reduced in both an amyotrophic lateral sclerosis (ALS) SOD1G93A mouse model and human ALS nerve samples. Our findings reveal the diversity of SCs and other cell types in peripheral nerve and serve as a reference for future studies of nerve biology and disease.
Collapse
Affiliation(s)
- Aldrin K Y Yim
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Peter L Wang
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - John R Bermingham
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Amber Hackett
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Amy Strickland
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Timothy M Miller
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Cindy Ly
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Robi D Mitra
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Jeffrey Milbrandt
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
31
|
Zotter B, Dagan O, Brady J, Baloui H, Samanta J, Salzer JL. Gli1 Regulates the Postnatal Acquisition of Peripheral Nerve Architecture. J Neurosci 2022; 42:183-201. [PMID: 34772739 PMCID: PMC8802940 DOI: 10.1523/jneurosci.3096-20.2021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 10/17/2021] [Accepted: 10/19/2021] [Indexed: 11/21/2022] Open
Abstract
Peripheral nerves are organized into discrete compartments. Axons, Schwann cells (SCs), and endoneurial fibroblasts (EFs) reside within the endoneurium and are surrounded by the perineurium, a cellular sheath comprised of layers of perineurial glia (PNG). SC secretion of Desert Hedgehog (Dhh) regulates this organization. In Dhh nulls, the perineurium is deficient and the endoneurium is subdivided into small compartments termed minifascicles. Human Dhh mutations cause a neuropathy with similar defects. Here we examine the role of Gli1, a canonical transcriptional effector of hedgehog signaling, in regulating peripheral nerve organization in mice of both genders. We identify PNG, EFs, and pericytes as Gli1-expressing cells by genetic fate mapping. Although expression of Dhh by SCs and Gli1 in target cells is coordinately regulated with myelination, Gli1 expression unexpectedly persists in Dhh null EFs. Thus, Gli1 is expressed in EFs noncanonically (i.e., independent of hedgehog signaling). Gli1 and Dhh also have nonredundant activities. Unlike Dhh nulls, Gli1 nulls have a normal perineurium. Like Dhh nulls, Gli1 nulls form minifascicles, which we show likely arise from EFs. Thus, Dhh and Gli1 are independent signals: Gli1 is dispensable for perineurial development but functions cooperatively with Dhh to drive normal endoneurial development. During development, Gli1 also regulates endoneurial extracellular matrix production, nerve vascular organization, and has modest, nonautonomous effects on SC sorting and myelination of axons. Finally, in adult nerves, induced deletion of Gli1 is sufficient to drive minifascicle formation. Thus, Gli1 regulates the development and is required to maintain the endoneurial architecture of peripheral nerves.SIGNIFICANCE STATEMENT Peripheral nerves are organized into distinct cellular/ECM compartments: the epineurium, perineurium, and endoneurium. This organization, with its associated cellular constituents, is critical for the structural and metabolic support of nerves and their response to injury. Here, we show that Gli1, a transcription factor normally expressed downstream of hedgehog signaling, is required for the proper organization of the endoneurium but not the perineurium. Unexpectedly, Gli1 expression by endoneurial cells is independent of, and functions nonredundantly with, Schwann Cell-derived Desert Hedgehog in regulating peripheral nerve architecture. These results further delineate how peripheral nerves acquire their distinctive organization during normal development, and highlight mechanisms that may regulate their reorganization in pathologic settings, including peripheral neuropathies and nerve injury.
Collapse
Affiliation(s)
- Brendan Zotter
- Department of Neuroscience and Physiology, Neuroscience Institute, NYU Langone Medical Center, New York, New York 10016
| | - Or Dagan
- Department of Neuroscience and Physiology, Neuroscience Institute, NYU Langone Medical Center, New York, New York 10016
| | - Jacob Brady
- Department of Neuroscience and Physiology, Neuroscience Institute, NYU Langone Medical Center, New York, New York 10016
| | - Hasna Baloui
- Departments of Neuroscience and Clinical Neuroscience, Karolinska Institutet, Stockholm, 17177, Sweden
| | - Jayshree Samanta
- Department of Comparative Biosciences, School of Veterinary Medicine, Stem Cell and Regenerative Medicine Center, University of Wisconsin-Madison, Madison, Wisconsin 53706
| | - James L Salzer
- Department of Neuroscience and Physiology, Neuroscience Institute, NYU Langone Medical Center, New York, New York 10016
| |
Collapse
|
32
|
Chau MJ, Quintero JE, Monje PV, Voss SR, Welleford AS, Gerhardt GA, van Horne CG. Using a Transection Paradigm to Enhance the Repair Mechanisms of an Investigational Human Cell Therapy. Cell Transplant 2022; 31:9636897221123515. [PMID: 36169034 PMCID: PMC9523845 DOI: 10.1177/09636897221123515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 08/16/2022] [Indexed: 12/02/2022] Open
Abstract
One promising strategy in cell therapies for Parkinson's disease (PD) is to harness a patient's own cells to provide neuroprotection in areas of the brain affected by neurodegeneration. No treatment exists to replace cells in the brain. Thus, our goal has been to support sick neurons and slow neurodegeneration by transplanting living repair tissue from the peripheral nervous system into the substantia nigra of those with PD. Our group has pioneered the transplantation of transection-activated sural nerve fascicles into the brain of human subjects with PD. Our experience in sural nerve transplantation has supported the safety and feasibility of this approach. As part of a paradigm to assess the reparative properties of human sural nerve following a transection injury, we collected nerve tissue approximately 2 weeks after sural nerve transection for immunoassays from 15 participants, and collected samples from two additional participants for single nuclei RNA sequencing. We quantified the expression of key neuroprotective and select anti-apoptotic genes along with their corresponding protein levels using immunoassays. The single nuclei data clustered into 10 distinctive groups defined on the basis of previously published cell type-specific genes. Transection-induced reparative peripheral nerve tissue showed RNA expression of neuroprotective factors and anti-apoptotic factors across multiple cell types after nerve injury induction. Key proteins of interest (BDNF, GDNF, beta-NGF, PDGFB, and VEGF) were upregulated in reparative tissue. These results provide insight on this repair tissue's utility as a neuroprotective cell therapy.
Collapse
Affiliation(s)
- Monica J. Chau
- Brain Restoration Center, College of Medicine, University of Kentucky, Lexington, KY, USA
- Department of Neurosurgery, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Jorge E. Quintero
- Brain Restoration Center, College of Medicine, University of Kentucky, Lexington, KY, USA
- Department of Neurosurgery, College of Medicine, University of Kentucky, Lexington, KY, USA
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Paula V. Monje
- Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Stephen Randal Voss
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Andrew S. Welleford
- Department of Neurology, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Greg A. Gerhardt
- Brain Restoration Center, College of Medicine, University of Kentucky, Lexington, KY, USA
- Department of Neurosurgery, College of Medicine, University of Kentucky, Lexington, KY, USA
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, USA
- Department of Neurology, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Craig G. van Horne
- Brain Restoration Center, College of Medicine, University of Kentucky, Lexington, KY, USA
- Department of Neurosurgery, College of Medicine, University of Kentucky, Lexington, KY, USA
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
33
|
A Shared Transcriptional Identity for Forebrain and Dentate Gyrus Neural Stem Cells from Embryogenesis to Adulthood. eNeuro 2022; 9:ENEURO.0271-21.2021. [PMID: 35027446 PMCID: PMC8856713 DOI: 10.1523/eneuro.0271-21.2021] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 11/10/2021] [Accepted: 11/15/2021] [Indexed: 12/12/2022] Open
Abstract
Adult neural stem cells (NSCs) reside in two distinct niches in the mammalian brain, the ventricular-subventricular zone (V-SVZ) of the forebrain lateral ventricles and the subgranular zone (SGZ) of the hippocampal dentate gyrus. They are thought to be molecularly distinct since V-SVZ NSCs produce inhibitory olfactory bulb (OB) interneurons and SGZ NSCs excitatory dentate granule neurons. Here, we have asked whether this is so by directly comparing V-SVZ and SGZ NSCs from embryogenesis to adulthood using single-cell transcriptional data. We show that the embryonic radial glial precursor (RP) parents of these two NSC populations are very similar, but differentially express a small cohort of genes involved in glutamatergic versus GABAergic neurogenesis. These different RPs then undergo a similar gradual transition to a dormant adult NSC state over the first three postnatal weeks. This dormancy state involves transcriptional shutdown of genes that maintain an active, proliferative, prodifferentiation state and induction of genes involved in sensing and regulating their niche environment. Moreover, when reactivated to generate adult-born progeny, both populations reacquire a development-like state and re-express proneurogenic genes. Thus, V-SVZ and SGZ NSCs share a common transcriptional state throughout their lifespans and transition into and out of dormancy via similar trajectories.
Collapse
|
34
|
Savelieff MG, Noureldein MH, Feldman EL. Systems Biology to Address Unmet Medical Needs in Neurological Disorders. Methods Mol Biol 2022; 2486:247-276. [PMID: 35437727 PMCID: PMC9446424 DOI: 10.1007/978-1-0716-2265-0_13] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Neurological diseases are highly prevalent and constitute a significant cause of mortality and disability. Neurological disorders encompass a heterogeneous group of neurodegenerative conditions, broadly characterized by injury to the peripheral and/or central nervous system. Although the etiology of neurological diseases varies greatly, they share several characteristics, such as heterogeneity of clinical presentation, non-cell autonomous nature, and diversity of cellular, subcellular, and molecular pathways. Systems biology has emerged as a valuable platform for addressing the challenges of studying heterogeneous neurological diseases. Systems biology has manifold applications to address unmet medical needs for neurological illness, including integrating and correlating different large datasets covering the transcriptome, epigenome, proteome, and metabolome associated with a specific condition. This is particularly useful for disentangling the heterogeneity and complexity of neurological conditions. Hence, systems biology can help in uncovering pathophysiology to develop novel therapeutic targets and assessing the impact of known treatments on disease progression. Additionally, systems biology can identify early diagnostic biomarkers, to help diagnose neurological disease preceded by a long subclinical phase, as well as define the exposome, the collection of environmental toxicants that increase risk of certain neurological diseases. In addition to these current applications, there are numerous potential emergent uses, such as precision medicine.
Collapse
Affiliation(s)
- Masha G Savelieff
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI, USA
| | - Mohamed H Noureldein
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI, USA
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Eva L Feldman
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI, USA.
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
35
|
Li M, Min Q, Banton MC, Dun X. Single-Cell Regulatory Network Inference and Clustering Identifies Cell-Type Specific Expression Pattern of Transcription Factors in Mouse Sciatic Nerve. Front Cell Neurosci 2021; 15:676515. [PMID: 34955748 PMCID: PMC8693779 DOI: 10.3389/fncel.2021.676515] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 10/28/2021] [Indexed: 02/05/2023] Open
Abstract
Advances in single-cell RNA sequencing technologies and bioinformatics methods allow for both the identification of cell types in a complex tissue and the large-scale gene expression profiling of various cell types in a mixture. In this report, we analyzed a single-cell RNA sequencing (scRNA-seq) dataset for the intact adult mouse sciatic nerve and examined cell-type specific transcription factor expression and activity during peripheral nerve homeostasis. In total, we identified 238 transcription factors expressed in nine different cell types of intact mouse sciatic nerve. Vascular smooth muscle cells have the lowest number of transcription factors expressed with 17 transcription factors identified. Myelinating Schwann cells (mSCs) have the highest number of transcription factors expressed, with 61 transcription factors identified. We created a cell-type specific expression map for the identified 238 transcription factors. Our results not only provide valuable information about the expression pattern of transcription factors in different cell types of adult peripheral nerves but also facilitate future studies to understand the function of key transcription factors in the peripheral nerve homeostasis and disease.
Collapse
Affiliation(s)
- Mingchao Li
- Department of Neurology, The Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Qing Min
- School of Pharmacy, Hubei University of Science and Technology, Xianning, China
| | - Matthew C Banton
- School of Biomedical Science, Faculty of Health, University of Plymouth, Plymouth, United Kingdom
| | - Xinpeng Dun
- School of Pharmacy, Hubei University of Science and Technology, Xianning, China.,The Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| |
Collapse
|
36
|
Zhang F, Wu LB, Yu Q, Wang MJ, Zeng XL, Wei XT, Wu ZJ, Cai RL, Hu L. Neurotropic Viruses as a Tool for Neural Circuit-Tracing. NEUROCHEM J+ 2021. [DOI: 10.1134/s1819712421040176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
37
|
Li M, Banton MC, Min Q, Parkinson DB, Dun X. Meta-Analysis Reveals Transcription Factor Upregulation in Cells of Injured Mouse Sciatic Nerve. Front Cell Neurosci 2021; 15:688243. [PMID: 34744629 PMCID: PMC8567084 DOI: 10.3389/fncel.2021.688243] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 09/30/2021] [Indexed: 11/13/2022] Open
Abstract
Following peripheral nerve injury, transcription factors upregulated in the distal nerve play essential roles in Schwann cell reprogramming, fibroblast activation and immune cell function to create a permissive distal nerve environment for axonal regrowth. In this report, we first analysed four microarray data sets to identify transcription factors that have at least twofold upregulation in the mouse distal nerve stump at day 3 and day 7 post-injury. Next, we compared their relative mRNA levels through the analysis of an available bulk mRNA sequencing data set at day 5 post-injury. We then investigated the expression of identified TFs in analysed single-cell RNA sequencing data sets for the distal nerve at day 3 and day 9 post-injury. These analyses identified 55 transcription factors that have at least twofold upregulation in the distal nerve following mouse sciatic nerve injury. Expression profile for the identified 55 transcription factors in cells of the distal nerve stump was further analysed on the scRNA-seq data. Transcription factor network and functional analysis were performed in Schwann cells. We also validated the expression pattern of Jun, Junb, Runx1, Runx2, and Sox2 in the mouse distal nerve stump by immunostaining. The findings from our study not only could be used to understand the function of key transcription factors in peripheral nerve regeneration but also could be used to facilitate experimental design for future studies to investigate the function of individual TFs in peripheral nerve regeneration.
Collapse
Affiliation(s)
- Mingchao Li
- Department of Neurology, The Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Matthew C Banton
- School of Biomedical Science, Faculty of Health, University of Plymouth, Plymouth, United Kingdom
| | - Qing Min
- School of Pharmacy, Hubei University of Science and Technology, Xianning, China
| | - David B Parkinson
- Peninsula Medical School, Faculty of Health, University of Plymouth, Plymouth, United Kingdom
| | - Xinpeng Dun
- School of Pharmacy, Hubei University of Science and Technology, Xianning, China.,The Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| |
Collapse
|
38
|
Jha MK, Passero JV, Rawat A, Ament XH, Yang F, Vidensky S, Collins SL, Horton MR, Hoke A, Rutter GA, Latremoliere A, Rothstein JD, Morrison BM. Macrophage monocarboxylate transporter 1 promotes peripheral nerve regeneration after injury in mice. J Clin Invest 2021; 131:e141964. [PMID: 34491913 DOI: 10.1172/jci141964] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 09/02/2021] [Indexed: 12/28/2022] Open
Abstract
Peripheral nerves have the capacity for regeneration, but the rate of regeneration is so slow that many nerve injuries lead to incomplete recovery and permanent disability for patients. Macrophages play a critical role in the peripheral nerve response to injury, contributing to both Wallerian degeneration and nerve regeneration, and their function has recently been shown to be dependent on intracellular metabolism. To date, the impact of their intracellular metabolism on peripheral nerve regeneration has not been studied. We examined conditional transgenic mice with selective ablation in macrophages of solute carrier family 16, member 1 (Slc16a1), which encodes monocarboxylate transporter 1 (MCT1), and found that MCT1 contributed to macrophage metabolism, phenotype, and function, specifically in regard to phagocytosis and peripheral nerve regeneration. Adoptive cell transfer of wild-type macrophages ameliorated the impaired nerve regeneration in macrophage-selective MCT1-null mice. We also developed a mouse model that overexpressed MCT1 in macrophages and found that peripheral nerves in these mice regenerated more rapidly than in control mice. Our study provides further evidence that MCT1 has an important biological role in macrophages and that manipulations of macrophage metabolism can enhance recovery from peripheral nerve injuries, for which there are currently no approved medical therapies.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Samuel L Collins
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Maureen R Horton
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | - Guy A Rutter
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Alban Latremoliere
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | | |
Collapse
|
39
|
Abstract
The sympathetic nervous system prepares the body for 'fight or flight' responses and maintains homeostasis during daily activities such as exercise, eating a meal or regulation of body temperature. Sympathetic regulation of bodily functions requires the establishment and refinement of anatomically and functionally precise connections between postganglionic sympathetic neurons and peripheral organs distributed widely throughout the body. Mechanistic studies of key events in the formation of postganglionic sympathetic neurons during embryonic and early postnatal life, including axon growth, target innervation, neuron survival, and dendrite growth and synapse formation, have advanced the understanding of how neuronal development is shaped by interactions with peripheral tissues and organs. Recent progress has also been made in identifying how the cellular and molecular diversity of sympathetic neurons is established to meet the functional demands of peripheral organs. In this Review, we summarize current knowledge of signalling pathways underlying the development of the sympathetic nervous system. These findings have implications for unravelling the contribution of sympathetic dysfunction stemming, in part, from developmental perturbations to the pathophysiology of peripheral neuropathies and cardiovascular and metabolic disorders.
Collapse
|
40
|
Saio S, Konishi K, Hohjoh H, Tamura Y, Masutani T, Iddamalgoda A, Ichihashi M, Hasegawa H, Mizutani KI. Extracellular Environment-Controlled Angiogenesis, and Potential Application for Peripheral Nerve Regeneration. Int J Mol Sci 2021; 22:ijms222011169. [PMID: 34681829 PMCID: PMC8541280 DOI: 10.3390/ijms222011169] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/12/2021] [Accepted: 10/13/2021] [Indexed: 12/18/2022] Open
Abstract
Endothelial cells acquire different phenotypes to establish functional vascular networks. Vascular endothelial growth factor (VEGF) signaling induces endothelial proliferation, migration, and survival to regulate vascular development, which leads to the construction of a vascular plexuses with a regular morphology. The spatiotemporal localization of angiogenic factors and the extracellular matrix play fundamental roles in ensuring the proper regulation of angiogenesis. This review article highlights how and what kinds of extracellular environmental molecules regulate angiogenesis. Close interactions between the vascular and neural systems involve shared molecular mechanisms to coordinate developmental and regenerative processes. This review article focuses on current knowledge about the roles of angiogenesis in peripheral nerve regeneration and the latest therapeutic strategies for the treatment of peripheral nerve injury.
Collapse
Affiliation(s)
- Shingo Saio
- Laboratory of Stem Cell Biology, Graduate School of Pharmaceutical Sciences, Kobe Gakuin University, 1-1-3 Minatojima, Chuo-ku, Kobe 650-8586, Japan; (S.S.); (K.K.); (Y.T.); (M.I.)
| | - Kanna Konishi
- Laboratory of Stem Cell Biology, Graduate School of Pharmaceutical Sciences, Kobe Gakuin University, 1-1-3 Minatojima, Chuo-ku, Kobe 650-8586, Japan; (S.S.); (K.K.); (Y.T.); (M.I.)
| | - Hirofumi Hohjoh
- Laboratory of Hygienic Sciences, Kobe Pharmaceutical University, 4-19-1, Motoyamakitamachi, Higashinada-ku, Kobe 658-8558, Japan;
| | - Yuki Tamura
- Laboratory of Stem Cell Biology, Graduate School of Pharmaceutical Sciences, Kobe Gakuin University, 1-1-3 Minatojima, Chuo-ku, Kobe 650-8586, Japan; (S.S.); (K.K.); (Y.T.); (M.I.)
| | - Teruaki Masutani
- Research & Development Dept., Ichimaru Pharcos Co., Ltd., 318-1 Asagi, Motosu 501-0475, Japan; (T.M.); (A.I.)
- Medical Education Development Center, Gifu University School of Medicine, 1-1 Yanagido, Gifu 501-1194, Japan
| | - Arunasiri Iddamalgoda
- Research & Development Dept., Ichimaru Pharcos Co., Ltd., 318-1 Asagi, Motosu 501-0475, Japan; (T.M.); (A.I.)
| | - Masamitsu Ichihashi
- Laboratory of Stem Cell Biology, Graduate School of Pharmaceutical Sciences, Kobe Gakuin University, 1-1-3 Minatojima, Chuo-ku, Kobe 650-8586, Japan; (S.S.); (K.K.); (Y.T.); (M.I.)
| | - Hiroshi Hasegawa
- Laboratory of Hygienic Sciences, Kobe Pharmaceutical University, 4-19-1, Motoyamakitamachi, Higashinada-ku, Kobe 658-8558, Japan;
- Correspondence: (H.H.); (K.-i.M.)
| | - Ken-ichi Mizutani
- Laboratory of Stem Cell Biology, Graduate School of Pharmaceutical Sciences, Kobe Gakuin University, 1-1-3 Minatojima, Chuo-ku, Kobe 650-8586, Japan; (S.S.); (K.K.); (Y.T.); (M.I.)
- Correspondence: (H.H.); (K.-i.M.)
| |
Collapse
|
41
|
Shen Y, Zhu J, Liu Q, Ding S, Dun X, He J. Up-Regulation of CD146 in Schwann Cells Following Peripheral Nerve Injury Modulates Schwann Cell Function in Regeneration. Front Cell Neurosci 2021; 15:743532. [PMID: 34720881 PMCID: PMC8552958 DOI: 10.3389/fncel.2021.743532] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 09/23/2021] [Indexed: 11/13/2022] Open
Abstract
CD146 is cell adhesion molecule and is implicated in a variety of physiological and pathological processes. However, the involvement of CD146 in peripheral nerve regeneration has not been studied yet. Here, we examine the spatial and temporal expression pattern of CD146 in injured mouse sciatic nerve via high-throughput data analysis, RT-PCR and immunostaining. By microarray data analysis and RT-PCR validation, we show that CD146 mRNA is significantly up-regulated in the nerve bridge and in the distal nerve stump following mouse sciatic nerve transection injury. By single cell sequencing data analysis and immunostaining, we demonstrate that CD146 is up-regulated in Schwann cells and cells associated with blood vessels following mouse peripheral nerve injury. Bioinformatic analysis revealed that CD146 not only has a key role in promoting of blood vessel regeneration but also regulates cell migration. The biological function of CD146 in Schwann cells was further investigated by knockdown of CD146 in rat primary Schwann cells. Functional assessments showed that knockdown of CD146 decreases viability and proliferation of Schwann cells but increases Schwann cell migration. Collectively, our findings imply that CD146 could be a key cell adhesion molecule that is up-regulated in injured peripheral nerves to regulate peripheral nerve regeneration.
Collapse
Affiliation(s)
- Yinying Shen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Jun Zhu
- Department of Thoracic Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Qianyan Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Shiyan Ding
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xinpeng Dun
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Jianghong He
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| |
Collapse
|
42
|
Gerber D, Pereira JA, Gerber J, Tan G, Dimitrieva S, Yángüez E, Suter U. Transcriptional profiling of mouse peripheral nerves to the single-cell level to build a sciatic nerve ATlas (SNAT). eLife 2021; 10:e58591. [PMID: 33890853 PMCID: PMC8064760 DOI: 10.7554/elife.58591] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 04/13/2021] [Indexed: 12/11/2022] Open
Abstract
Peripheral nerves are organ-like structures containing diverse cell types to optimize function. This interactive assembly includes mostly axon-associated Schwann cells, but also endothelial cells of supporting blood vessels, immune system-associated cells, barrier-forming cells of the perineurium surrounding and protecting nerve fascicles, and connective tissue-resident cells within the intra-fascicular endoneurium and inter-fascicular epineurium. We have established transcriptional profiles of mouse sciatic nerve-inhabitant cells to foster the fundamental understanding of peripheral nerves. To achieve this goal, we have combined bulk RNA sequencing of developing sciatic nerves up to the adult with focused bulk and single-cell RNA sequencing of Schwann cells throughout postnatal development, extended by single-cell transcriptome analysis of the full sciatic nerve both perinatally and in the adult. The results were merged in the transcriptome resource Sciatic Nerve ATlas (SNAT: https://www.snat.ethz.ch). We anticipate that insights gained from our multi-layered analysis will serve as valuable interactive reference point to guide future studies.
Collapse
Affiliation(s)
- Daniel Gerber
- Department of Biology, Institute of Molecular Health Sciences, Swiss Federal Institute of Technology, ETH ZurichZurichSwitzerland
| | - Jorge A Pereira
- Department of Biology, Institute of Molecular Health Sciences, Swiss Federal Institute of Technology, ETH ZurichZurichSwitzerland
| | - Joanne Gerber
- Department of Biology, Institute of Molecular Health Sciences, Swiss Federal Institute of Technology, ETH ZurichZurichSwitzerland
| | - Ge Tan
- Functional Genomics Center Zurich, ETH Zurich/University of ZurichZurichSwitzerland
| | - Slavica Dimitrieva
- Functional Genomics Center Zurich, ETH Zurich/University of ZurichZurichSwitzerland
| | - Emilio Yángüez
- Functional Genomics Center Zurich, ETH Zurich/University of ZurichZurichSwitzerland
| | - Ueli Suter
- Department of Biology, Institute of Molecular Health Sciences, Swiss Federal Institute of Technology, ETH ZurichZurichSwitzerland
| |
Collapse
|
43
|
Chen B, Banton MC, Singh L, Parkinson DB, Dun XP. Single Cell Transcriptome Data Analysis Defines the Heterogeneity of Peripheral Nerve Cells in Homeostasis and Regeneration. Front Cell Neurosci 2021; 15:624826. [PMID: 33828460 PMCID: PMC8019921 DOI: 10.3389/fncel.2021.624826] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 02/26/2021] [Indexed: 12/17/2022] Open
Abstract
The advances in single-cell RNA sequencing technologies and the development of bioinformatics pipelines enable us to more accurately define the heterogeneity of cell types in a selected tissue. In this report, we re-analyzed recently published single-cell RNA sequencing data sets and provide a rationale to redefine the heterogeneity of cells in both intact and injured mouse peripheral nerves. Our analysis showed that, in both intact and injured peripheral nerves, cells could be functionally classified into four categories: Schwann cells, nerve fibroblasts, immune cells, and cells associated with blood vessels. Nerve fibroblasts could be sub-clustered into epineurial, perineurial, and endoneurial fibroblasts. Identified immune cell clusters include macrophages, mast cells, natural killer cells, T and B lymphocytes as well as an unreported cluster of neutrophils. Cells associated with blood vessels include endothelial cells, vascular smooth muscle cells, and pericytes. We show that endothelial cells in the intact mouse sciatic nerve have three sub-types: epineurial, endoneurial, and lymphatic endothelial cells. Analysis of cell type-specific gene changes revealed that Schwann cells and endoneurial fibroblasts are the two most important cell types promoting peripheral nerve regeneration. Analysis of communication between these cells identified potential signals for early blood vessel regeneration, neutrophil recruitment of macrophages, and macrophages activating Schwann cells. Through this analysis, we also report appropriate marker genes for future single cell transcriptome data analysis to identify cell types in intact and injured peripheral nerves. The findings from our analysis could facilitate a better understanding of cell biology of peripheral nerves in homeostasis, regeneration, and disease.
Collapse
Affiliation(s)
- Bing Chen
- Department of Neurology, The Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Matthew C Banton
- Faculty of Health, School of Biomedical Science, University of Plymouth, Plymouth, United Kingdom
| | - Lolita Singh
- Faculty of Health, Peninsula Medical School, University of Plymouth, Plymouth, United Kingdom
| | - David B Parkinson
- Faculty of Health, Peninsula Medical School, University of Plymouth, Plymouth, United Kingdom
| | - Xin-Peng Dun
- Faculty of Health, Peninsula Medical School, University of Plymouth, Plymouth, United Kingdom.,School of Pharmacy, Hubei University of Science and Technology, Xianning, China
| |
Collapse
|
44
|
Schwann cell plasticity regulates neuroblastic tumor cell differentiation via epidermal growth factor-like protein 8. Nat Commun 2021; 12:1624. [PMID: 33712610 PMCID: PMC7954855 DOI: 10.1038/s41467-021-21859-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 02/17/2021] [Indexed: 01/31/2023] Open
Abstract
Adult Schwann cells (SCs) possess an inherent plastic potential. This plasticity allows SCs to acquire repair-specific functions essential for peripheral nerve regeneration. Here, we investigate whether stromal SCs in benign-behaving peripheral neuroblastic tumors adopt a similar cellular state. We profile ganglioneuromas and neuroblastomas, rich and poor in SC stroma, respectively, and peripheral nerves after injury, rich in repair SCs. Indeed, stromal SCs in ganglioneuromas and repair SCs share the expression of nerve repair-associated genes. Neuroblastoma cells, derived from aggressive tumors, respond to primary repair-related SCs and their secretome with increased neuronal differentiation and reduced proliferation. Within the pool of secreted stromal and repair SC factors, we identify EGFL8, a matricellular protein with so far undescribed function, to act as neuritogen and to rewire cellular signaling by activating kinases involved in neurogenesis. In summary, we report that human SCs undergo a similar adaptive response in two patho-physiologically distinct situations, peripheral nerve injury and tumor development.
Collapse
|
45
|
Balakrishnan A, Belfiore L, Chu TH, Fleming T, Midha R, Biernaskie J, Schuurmans C. Insights Into the Role and Potential of Schwann Cells for Peripheral Nerve Repair From Studies of Development and Injury. Front Mol Neurosci 2021; 13:608442. [PMID: 33568974 PMCID: PMC7868393 DOI: 10.3389/fnmol.2020.608442] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 12/31/2020] [Indexed: 12/13/2022] Open
Abstract
Peripheral nerve injuries arising from trauma or disease can lead to sensory and motor deficits and neuropathic pain. Despite the purported ability of the peripheral nerve to self-repair, lifelong disability is common. New molecular and cellular insights have begun to reveal why the peripheral nerve has limited repair capacity. The peripheral nerve is primarily comprised of axons and Schwann cells, the supporting glial cells that produce myelin to facilitate the rapid conduction of electrical impulses. Schwann cells are required for successful nerve regeneration; they partially “de-differentiate” in response to injury, re-initiating the expression of developmental genes that support nerve repair. However, Schwann cell dysfunction, which occurs in chronic nerve injury, disease, and aging, limits their capacity to support endogenous repair, worsening patient outcomes. Cell replacement-based therapeutic approaches using exogenous Schwann cells could be curative, but not all Schwann cells have a “repair” phenotype, defined as the ability to promote axonal growth, maintain a proliferative phenotype, and remyelinate axons. Two cell replacement strategies are being championed for peripheral nerve repair: prospective isolation of “repair” Schwann cells for autologous cell transplants, which is hampered by supply challenges, and directed differentiation of pluripotent stem cells or lineage conversion of accessible somatic cells to induced Schwann cells, with the potential of “unlimited” supply. All approaches require a solid understanding of the molecular mechanisms guiding Schwann cell development and the repair phenotype, which we review herein. Together these studies provide essential context for current efforts to design glial cell-based therapies for peripheral nerve regeneration.
Collapse
Affiliation(s)
- Anjali Balakrishnan
- Biological Sciences Platform, Sunnybrook Research Institute (SRI), Toronto, ON, Canada.,Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Lauren Belfiore
- Biological Sciences Platform, Sunnybrook Research Institute (SRI), Toronto, ON, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Tak-Ho Chu
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Taylor Fleming
- Biological Sciences Platform, Sunnybrook Research Institute (SRI), Toronto, ON, Canada
| | - Rajiv Midha
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Jeff Biernaskie
- Department of Comparative Biology and Experimental Medicine, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Carol Schuurmans
- Biological Sciences Platform, Sunnybrook Research Institute (SRI), Toronto, ON, Canada.,Department of Biochemistry, University of Toronto, Toronto, ON, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
46
|
Henzi A, Aguzzi A. The prion protein is not required for peripheral nerve de- and remyelination after crush injury. PLoS One 2021; 16:e0245944. [PMID: 33481951 PMCID: PMC7822300 DOI: 10.1371/journal.pone.0245944] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 01/11/2021] [Indexed: 12/15/2022] Open
Abstract
The cellular prion protein (PrP) is essential to the long-term maintenance of myelin sheaths in peripheral nerves. PrP activates the adhesion G-protein coupled receptor Adgrg6 on Schwann cells and initiates a pro-myelination cascade of molecular signals. Because Adgrg6 is crucial for peripheral myelin development and regeneration after nerve injury, we investigated the role of PrP in peripheral nerve repair. We performed experimental sciatic nerve crush injuries in co-isogenic wild-type and PrP-deficient mice, and examined peripheral nerve repair processes. Generation of repair Schwann cells, macrophage recruitment and remyelination were similar in PrP-deficient and wild-type mice. We conclude that PrP is dispensable for sciatic nerve de- and remyelination after crush injury. Adgrg6 may sustain its function in peripheral nerve repair independently of its activation by PrP.
Collapse
Affiliation(s)
- Anna Henzi
- Institute of Neuropathology, University of Zurich, Zurich, Switzerland
| | - Adriano Aguzzi
- Institute of Neuropathology, University of Zurich, Zurich, Switzerland
- * E-mail:
| |
Collapse
|