1
|
Wang F, Zhao C, Jing Z, Wang Q, Li M, Lu B, Huo A, Liang W, Hu W, Fu X. The dual roles of chemokines in peripheral nerve injury and repair. Inflamm Regen 2025; 45:11. [PMID: 40217284 PMCID: PMC11987372 DOI: 10.1186/s41232-025-00375-4] [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: 01/08/2025] [Accepted: 04/03/2025] [Indexed: 04/14/2025] Open
Abstract
Peripheral nerve injuries (PNI) occur in approximately 13-23 per 100,000 individuals, predominantly affecting young and middle-aged adults. These injuries often require a lengthy recovery period, placing substantial burdens on healthcare systems and national economies. Current treatment strategies have not significantly shortened this lengthy regenerative process, highlighting the urgent need for innovative therapeutic interventions. Chemokines were originally noted for their powerful ability to recruit immune cells; however, as research has advanced, it has become increasingly evident that their role in peripheral nerve repair has been underestimated. In this review, we provide the first comprehensive overview of chemokine expression and activity during peripheral nerve injury and regeneration. We summarize the existing literature on chemokine family members, detailing their expression patterns and localization in injured nerves to facilitate further mechanistic investigations. For chemokines that remain controversial, such as CXCL1 and CCL2, we critically examine experimental methodologies and discuss factors underlying conflicting results, ultimately affirming their contributions to promoting nerve repair. Importantly, we highlight the dual nature of chemokines: in the early stages of injury, they initiate reparative responses, activate Schwann cells, regulate Wallerian degeneration, and support nerve recovery; but when the axons are connected and the repair enters the later stages, their persistent proinflammatory effects during later stages may impede the healing process. Additionally, we emphasize that certain chemokines, including CXCL5, CXCL12, and CCL2, can act directly on neurons/axons, thereby accelerating axonal regeneration. Future research should focus on precisely mapping the localization and temporal expression profiles of these chemokines and exploring therapeutic approaches.
Collapse
Affiliation(s)
- Fangyuan Wang
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Henan Provincial Key Laboratory of Cranial Nerve Diseases, ZhengZhou, China
| | - Chenglin Zhao
- Department of Neurosurgery, People's Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Henan Provincial Key Laboratory of Cranial Nerve Diseases, ZhengZhou, China
| | - Zhou Jing
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Henan Provincial Key Laboratory of Cranial Nerve Diseases, ZhengZhou, China
| | - Qingyi Wang
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Henan Provincial Key Laboratory of Cranial Nerve Diseases, ZhengZhou, China
| | - Minghe Li
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Henan Provincial Key Laboratory of Cranial Nerve Diseases, ZhengZhou, China
| | - Bingqi Lu
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Henan Provincial Key Laboratory of Cranial Nerve Diseases, ZhengZhou, China
| | - Ao Huo
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Henan Provincial Key Laboratory of Cranial Nerve Diseases, ZhengZhou, China
| | - Wulong Liang
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Henan Provincial Key Laboratory of Cranial Nerve Diseases, ZhengZhou, China
| | - Weihua Hu
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Henan Provincial Key Laboratory of Cranial Nerve Diseases, ZhengZhou, China
| | - Xudong Fu
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China.
- Henan Provincial Key Laboratory of Cranial Nerve Diseases, ZhengZhou, China.
| |
Collapse
|
2
|
Haider S, Sassu E, Stefanovska D, Stoyek MR, Preissl S, Hortells L. News from the old: Aging features in the intracardiac, musculoskeletal, and enteric nervous systems. Ageing Res Rev 2025; 105:102690. [PMID: 39947485 DOI: 10.1016/j.arr.2025.102690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 01/08/2025] [Accepted: 02/10/2025] [Indexed: 02/18/2025]
Abstract
Aging strongly affects the peripheral nervous system (PNS), triggering alterations that vary depending on the innervated tissue. The most frequent alteration in peripheral nerve aging is reduced nerve fiber and glial density which can lead to abnormal nerve functionality. Interestingly, the activation of a destructive phenotype takes place in macrophages across the PNS while a reduced number of neuronal bodies is a unique feature of some enteric ganglia. Single cell/nucleus RNA-sequencing has unveiled a striking complexity of cell populations in the peripheral nerves, and these refined cell type annotations could facilitate a better understanding of PNS aging. While the effects of senescence on individual PNS cell types requires further characterization, the use of senolytics appears to improve general PNS function in models of aging. Here, we review the current understanding of age-related changes of the intracardiac, musculoskeletal, and enteric nervous system sub-sections of the PNS, highlighting their commonalities and differences.
Collapse
Affiliation(s)
- Severin Haider
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg 79110, Germany
| | - Eliza Sassu
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg 79110, Germany
| | - Dragana Stefanovska
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg 79110, Germany
| | - Mathew R Stoyek
- Department of Physiology & Biophysics, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Sebastian Preissl
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg 79110, Germany; Institute of Pharmaceutical Sciences, Pharmacology & Toxicology, University of Graz, Graz 8010, Austria; Field of Excellence BioHealth - University of Graz, Graz, Austria
| | - Luis Hortells
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg 79110, Germany; Cardiovascular Research Group, Department of Medical Biology, Faculty of Health Science, UiT-The Arctic University of Norway, Tromsø 9019, Norway.
| |
Collapse
|
3
|
Guest JD, Santamaria AJ, Solano JP, de Rivero Vaccari JP, Dietrich WD, Pearse DD, Khan A, Levi AD. Challenges in advancing Schwann cell transplantation for spinal cord injury repair. Cytotherapy 2025; 27:36-50. [PMID: 39387736 DOI: 10.1016/j.jcyt.2024.08.005] [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: 03/17/2024] [Revised: 08/05/2024] [Accepted: 08/09/2024] [Indexed: 10/15/2024]
Abstract
BACKGROUND AIMS In this article we aimed to provide an expert synthesis of the current status of Schwann cell (SC)therapeutics and potential steps to increase their clinical utility. METHODS We provide an expert synthesis based on preclinical, clinical and manufacturing experience. RESULTS Schwann cells (SCs) are essential for peripheral nerve regeneration and are of interest in supporting axonal repair after spinal cord injury (SCI). SCs can be isolated and cultivated in tissue culture from adult nerve biopsies or generated from precursors and neural progenitors using specific differentiation protocols leading to expanded quantities. In culture, they undergo dedifferentiation to a state similar to "repair" SCs. The known repertoire of SC functions is increasing beyond axon maintenance, myelination, and axonal regeneration to include immunologic regulation and the release of potentially therapeutic extracellular vesicles. Recently, autologous human SC cultures purified under cGMP conditions have been tested in both nerve repair and subacute and chronic SCI clinical trials. Although the effects of SCs to support nerve regeneration are indisputable, their efficacy for clinical SCI has been limited according to the outcomes examined. CONCLUSIONS This review discusses the current limitations of transplanted SCs within the damaged spinal cord environment. Limitations include limited post-transplant cell survival, the inability of SCs to migrate within astrocytic parenchyma, and restricted axonal regeneration out of SC-rich graft regions. We describe steps to amplify the survival and integration of transplanted SCs and to expand the repertoire of uses of SCs, including SC-derived extracellular vesicles. The relative merits of transplanting autologous versus allogeneic SCs and the role that endogenous SCs play in spinal cord repair are described. Finally, we briefly describe the issues requiring solutions to scale up SC manufacturing for commercial use.
Collapse
Affiliation(s)
- James D Guest
- The Miami Project to Cure Paralysis and Neurological Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA.
| | - Andrea J Santamaria
- The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Juan P Solano
- Pediatric Critical Care, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Juan P de Rivero Vaccari
- The Miami Project to Cure Paralysis and Neurological Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - William D Dietrich
- The Miami Project to Cure Paralysis and Neurological Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Damien D Pearse
- The Miami Project to Cure Paralysis and Neurological Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Aisha Khan
- The Stem Cell Institute, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Allan D Levi
- The Miami Project to Cure Paralysis and Neurological Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| |
Collapse
|
4
|
Olsen TC, LaGuardia JS, Chen DR, Lebens RS, Huang KX, Milek D, Noble M, Leckenby JI. Influencing factors and repair advancements in rodent models of peripheral nerve regeneration. Regen Med 2024; 19:561-577. [PMID: 39469920 PMCID: PMC11633413 DOI: 10.1080/17460751.2024.2405318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 09/12/2024] [Indexed: 10/30/2024] Open
Abstract
Peripheral nerve injuries lead to severe functional impairments, with rodent models essential for studying regeneration. This review examines key factors affecting outcomes. Age-related declines, like reduced nerve fiber density and impaired axonal transport of vesicles, hinder recovery. Hormonal differences influence regeneration, with BDNF/trkB critical for testosterone and nerve growth factor for estrogen signaling pathways. Species and strain selection impact outcomes, with C57BL/6 mice and Sprague-Dawley rats exhibiting varying regenerative capacities. Injury models - crush for early regeneration, chronic constriction for neuropathic pain, stretch for traumatic elongation and transection for severe lacerations - provide insights into clinically relevant scenarios. Repair techniques, such as nerve grafts and conduits, show that autografts are the gold standard for gaps over 3 cm, with success influenced by graft type and diameter. Time course analysis highlights crucial early degeneration and regeneration phases within the first month, with functional recovery stabilizing by three to six months. Early intervention optimizes regeneration by reducing scar tissue formation, while later interventions focus on remyelination. Understanding these factors is vital for designing robust preclinical studies and translating research into effective clinical treatments for peripheral nerve injuries.
Collapse
Affiliation(s)
- Timothy C Olsen
- Division of Plastic & Reconstructive Surgery, University of Rochester Medical Center, 601 Elmwood Avenue Box 661Rochester, NY14642, USA
| | - Jonnby S LaGuardia
- Division of Plastic & Reconstructive Surgery, University of Rochester Medical Center, 601 Elmwood Avenue Box 661Rochester, NY14642, USA
| | - David R Chen
- University of California, 410 Charles E. Young Drive, East Los Angeles, CA90095, USA
| | - Ryan S Lebens
- University of California, 410 Charles E. Young Drive, East Los Angeles, CA90095, USA
| | - Kelly X Huang
- University of California, 410 Charles E. Young Drive, East Los Angeles, CA90095, USA
| | - David Milek
- Division of Plastic & Reconstructive Surgery, University of Rochester Medical Center, 601 Elmwood Avenue Box 661Rochester, NY14642, USA
| | - Mark Noble
- Department of Biomedical Genetics, University of Rochester Medical Center, 601 Elmwood Avenue Box 661Rochester, NY14642, USA
| | - Jonathan I Leckenby
- Division of Plastic & Reconstructive Surgery, University of Rochester Medical Center, 601 Elmwood Avenue Box 661Rochester, NY14642, USA
| |
Collapse
|
5
|
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
|
6
|
Xin GD, Liu XY, Fan XD, Zhao GJ. Exosomes repairment for sciatic nerve injury: a cell-free therapy. Stem Cell Res Ther 2024; 15:214. [PMID: 39020385 PMCID: PMC11256477 DOI: 10.1186/s13287-024-03837-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 07/05/2024] [Indexed: 07/19/2024] Open
Abstract
Sciatic nerve injury (SNI) is a common type of peripheral nerve injury typically resulting from trauma, such as contusion, sharp force injuries, drug injections, pelvic fractures, or hip dislocations. It leads to both sensory and motor dysfunctions, characterized by pain, numbness, loss of sensation, muscle atrophy, reduced muscle tone, and limb paralysis. These symptoms can significantly diminish a patient's quality of life. Following SNI, Wallerian degeneration occurs, which activates various signaling pathways, inflammatory factors, and epigenetic regulators. Despite the availability of several surgical and nonsurgical treatments, their effectiveness remains suboptimal. Exosomes are extracellular vesicles with diameters ranging from 30 to 150 nm, originating from the endoplasmic reticulum. They play a crucial role in facilitating intercellular communication and have emerged as highly promising vehicles for drug delivery. Increasing evidence supports the significant potential of exosomes in repairing SNI. This review delves into the pathological progression of SNI, techniques for generating exosomes, the molecular mechanisms behind SNI recovery with exosomes, the effectiveness of combining exosomes with other approaches for SNI repair, and the changes and future outlook for utilizing exosomes in SNI recovery.
Collapse
Affiliation(s)
- Guang-Da Xin
- Nephrology Department, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, 130000, China
| | - Xue-Yan Liu
- Cardiology Department, China-Japan Union Hospital of Jilin Universit, Changchun, Jilin Province, 130000, China
| | - Xiao-Di Fan
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, 130000, China
| | - Guan-Jie Zhao
- Nephrology Department, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, 130000, China.
| |
Collapse
|
7
|
Ratan Y, Rajput A, Pareek A, Pareek A, Kaur R, Sonia S, Kumar R, Singh G. Recent Advances in Biomolecular Patho-Mechanistic Pathways behind the Development and Progression of Diabetic Neuropathy. Biomedicines 2024; 12:1390. [PMID: 39061964 PMCID: PMC11273858 DOI: 10.3390/biomedicines12071390] [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: 05/07/2024] [Revised: 06/12/2024] [Accepted: 06/19/2024] [Indexed: 07/28/2024] Open
Abstract
Diabetic neuropathy (DN) is a neurodegenerative disorder that is primarily characterized by distal sensory loss, reduced mobility, and foot ulcers that may potentially lead to amputation. The multifaceted etiology of DN is linked to a range of inflammatory, vascular, metabolic, and other neurodegenerative factors. Chronic inflammation, endothelial dysfunction, and oxidative stress are the three basic biological changes that contribute to the development of DN. Although our understanding of the intricacies of DN has advanced significantly over the past decade, the distinctive mechanisms underlying the condition are still poorly understood, which may be the reason behind the lack of an effective treatment and cure for DN. The present study delivers a comprehensive understanding and highlights the potential role of the several pathways and molecular mechanisms underlying the etiopathogenesis of DN. Moreover, Schwann cells and satellite glial cells, as integral factors in the pathogenesis of DN, have been enlightened. This work will motivate allied research disciplines to gain a better understanding and analysis of the current state of the biomolecular mechanisms behind the pathogenesis of DN, which will be essential to effectively address every facet of DN, from prevention to treatment.
Collapse
Affiliation(s)
- Yashumati Ratan
- Department of Pharmacy, Banasthali Vidyapith, Banasthali 304022, Rajasthan, India; (A.R.); (A.P.); (A.P.)
| | - Aishwarya Rajput
- Department of Pharmacy, Banasthali Vidyapith, Banasthali 304022, Rajasthan, India; (A.R.); (A.P.); (A.P.)
| | - Ashutosh Pareek
- Department of Pharmacy, Banasthali Vidyapith, Banasthali 304022, Rajasthan, India; (A.R.); (A.P.); (A.P.)
| | - Aaushi Pareek
- Department of Pharmacy, Banasthali Vidyapith, Banasthali 304022, Rajasthan, India; (A.R.); (A.P.); (A.P.)
| | - Ranjeet Kaur
- Adesh Institute of Dental Sciences and Research, Bathinda 151101, Punjab, India;
| | - Sonia Sonia
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar 143005, Punjab, India;
| | - Rahul Kumar
- Baba Ragav Das Government Medical College, Gorakhpur 273013, Uttar Pradesh, India;
| | - Gurjit Singh
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, IL 60607, USA
| |
Collapse
|
8
|
Su Y, Huang M, Thomas AG, Maragakis J, Huizar KDJ, Zheng Y, Wu Y, Farah MH, Slusher BS. GCPII Inhibition Promotes Remyelination after Peripheral Nerve Injury in Aged Mice. Int J Mol Sci 2024; 25:6893. [PMID: 39000003 PMCID: PMC11241013 DOI: 10.3390/ijms25136893] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 06/18/2024] [Accepted: 06/21/2024] [Indexed: 07/14/2024] Open
Abstract
Peripheral nerve injuries (PNIs) represent a significant clinical challenge, particularly in elderly populations where axonal remyelination and regeneration are impaired. Developing therapies to enhance these processes is crucial for improving PNI repair outcomes. Glutamate carboxypeptidase II (GCPII) is a neuropeptidase that plays a pivotal role in modulating glutamate signaling through its enzymatic cleavage of the abundant neuropeptide N-acetyl aspartyl glutamate (NAAG) to liberate glutamate. Within the PNS, GCPII is expressed in Schwann cells and activated macrophages, and its expression is amplified with aging. In this study, we explored the therapeutic potential of inhibiting GCPII activity following PNI. We report significant GCPII protein and activity upregulation following PNI, which was normalized by the potent and selective GCPII inhibitor 2-(phosphonomethyl)-pentanedioic acid (2-PMPA). In vitro, 2-PMPA robustly enhanced myelination in dorsal root ganglion (DRG) explants. In vivo, using a sciatic nerve crush injury model in aged mice, 2-PMPA accelerated remyelination, as evidenced by increased myelin sheath thickness and higher numbers of remyelinated axons. These findings suggest that GCPII inhibition may be a promising therapeutic strategy to enhance remyelination and potentially improve functional recovery after PNI, which is especially relevant in elderly PNI patients where this process is compromised.
Collapse
Affiliation(s)
- Yu Su
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (Y.S.); (M.H.); (A.G.T.); (J.M.); (K.D.J.H.); (Y.Z.); (Y.W.)
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Meixiang Huang
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (Y.S.); (M.H.); (A.G.T.); (J.M.); (K.D.J.H.); (Y.Z.); (Y.W.)
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ajit G. Thomas
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (Y.S.); (M.H.); (A.G.T.); (J.M.); (K.D.J.H.); (Y.Z.); (Y.W.)
| | - John Maragakis
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (Y.S.); (M.H.); (A.G.T.); (J.M.); (K.D.J.H.); (Y.Z.); (Y.W.)
| | - Kaitlyn D. J. Huizar
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (Y.S.); (M.H.); (A.G.T.); (J.M.); (K.D.J.H.); (Y.Z.); (Y.W.)
| | - Yuxin Zheng
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (Y.S.); (M.H.); (A.G.T.); (J.M.); (K.D.J.H.); (Y.Z.); (Y.W.)
| | - Ying Wu
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (Y.S.); (M.H.); (A.G.T.); (J.M.); (K.D.J.H.); (Y.Z.); (Y.W.)
| | - Mohamed H. Farah
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Barbara S. Slusher
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (Y.S.); (M.H.); (A.G.T.); (J.M.); (K.D.J.H.); (Y.Z.); (Y.W.)
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Psychiatry and Behavioral Science, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
9
|
Moss KR, Mi R, Kawaguchi R, Ehmsen JT, Shi Q, Vargas PI, Mukherjee-Clavin B, Lee G, Höke A. hESC- and hiPSC-derived Schwann cells are molecularly comparable and functionally equivalent. iScience 2024; 27:109855. [PMID: 38770143 PMCID: PMC11103364 DOI: 10.1016/j.isci.2024.109855] [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: 11/07/2022] [Revised: 02/11/2024] [Accepted: 04/26/2024] [Indexed: 05/22/2024] Open
Abstract
Establishing robust models of human myelinating Schwann cells is critical for studying peripheral nerve injury and disease. Stem cell differentiation has emerged as a key human cell model and disease motivating development of Schwann cell differentiation protocols. Human embryonic stem cells (hESCs) are considered the ideal pluripotent cell but ethical concerns regarding their use have propelled the popularity of human induced pluripotent stem cells (hiPSCs). Given that the equivalence of hESCs and hiPSCs remains controversial, we sought to compare the molecular and functional equivalence of hESC- and hiPSC-derived Schwann cells generated with our previously reported protocol. We identified only modest transcriptome differences by RNA sequencing and insignificant proteome differences by antibody array. Additionally, both cell types comparably improved nerve regeneration and function in a chronic denervation and regeneration animal model. Our findings demonstrate that Schwann cells derived from hESCs and hiPSCs with our protocol are molecularly comparable and functionally equivalent.
Collapse
Affiliation(s)
- Kathryn R. Moss
- Department of Neurology, Neuromuscular Division, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ruifa Mi
- Department of Neurology, Neuromuscular Division, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Riki Kawaguchi
- Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles David Geffen School of Medicine, Los Angeles, CA 90095, USA
| | - Jeffrey T. Ehmsen
- Department of Neurology, Neuromuscular Division, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Qiang Shi
- Department of Neurology, Neuromuscular Division, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Paula I. Vargas
- Department of Neurology, Neuromuscular Division, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Bipasha Mukherjee-Clavin
- Department of Neurology, Neuromuscular Division, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Gabsang Lee
- Department of Neurology, Neuromuscular Division, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ahmet Höke
- Department of Neurology, Neuromuscular Division, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
10
|
Hagen KM, Gordon P, Frederick A, Palmer AL, Edalat P, Zonta YR, Scott L, Flancia M, Reid JK, Joel M, Ousman SS. CRYAB plays a role in terminating the presence of pro-inflammatory macrophages in the older, injured mouse peripheral nervous system. Neurobiol Aging 2024; 133:1-15. [PMID: 38381471 DOI: 10.1016/j.neurobiolaging.2023.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 10/10/2023] [Accepted: 10/11/2023] [Indexed: 02/22/2024]
Abstract
Evidence indicates that dysfunction of older Schwann cells and macrophages contributes to poor regeneration of more mature peripheral nervous system (PNS) neurons after damage. Since the underlying molecular factors are largely unknown, we investigated if CRYAB, a small heat shock protein that is expressed by Schwann cells and axons and whose expression declines with age, impacts prominent deficits in the injured, older PNS including down-regulation of cholesterol biosynthesis enzyme genes, Schwann cell dysfunction, and macrophage persistence. Following sciatic nerve transection injury in 3- and 12-month-old wildtype and CRYAB knockout mice, we found by bulk RNA sequencing and RT-PCR, that while gene expression of cholesterol biosynthesis enzymes is markedly dysregulated in the aging, injured PNS, CRYAB is not involved. However, immunohistochemical staining of crushed sciatic nerves revealed that more macrophages of the pro-inflammatory but not immunosuppressive phenotype persisted in damaged 12-month-old knockout nerves. These pro-inflammatory macrophages were more efficient at engulfing myelin debris. CRYAB thus appears to play a role in resolving pro-inflammatory macrophage responses after damage to the older PNS.
Collapse
Affiliation(s)
- Kathleen Margaret Hagen
- Department of Neuroscience, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Paul Gordon
- Cumming School of Medicine Centre for Health Genomics and Informatics, University of Calgary, Calgary, Alberta, Canada
| | - Ariana Frederick
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Alexandra Louise Palmer
- Department of Neuroscience, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Pariya Edalat
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Yohan Ricci Zonta
- Department of Neuroscience, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Lucas Scott
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Melissa Flancia
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Jacqueline Kelsey Reid
- Department of Neuroscience, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Matthew Joel
- Department of Neuroscience, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Shalina Sheryl Ousman
- Departments of Clinical Neurosciences and Cell Biology and Anatomy, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
11
|
Kuang R, Zhang Y, Wu G, Zhu Z, Xu S, Liu X, Xu Y, Luo Y. Long Non-coding RNAs Influence Aging Process of Sciatic Nerves in SD Rats. Comb Chem High Throughput Screen 2024; 27:2140-2150. [PMID: 37691192 PMCID: PMC11348477 DOI: 10.2174/1386207326666230907115800] [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: 06/23/2023] [Revised: 08/19/2023] [Accepted: 08/23/2023] [Indexed: 09/12/2023]
Abstract
OBJECTIVES To investigate the long non-coding RNAs (lncRNAs) changes in the sciatic nerve (SN) in Sprague Dawley (SD) rats during aging. METHODS Eighteen healthy SD rats were selected at the age of 1 month (1M) and 24 months (24M) and SNs were collected. High-throughput transcriptome sequencing and bioinformatics analysis were performed. Protein-protein interaction (PPI) networks and competing endogenous RNA (ceRNA) networks were established according to differentially expressed genes (DEGs). RESULT As the length of lncRNAs increased, its proportion to the total number of lncRNAs decreased. A total of 4079 DElncRNAs were identified in Con vs. 24M. GO analysis was primarily clustered in nerve and lipid metabolism, extracellular matrix, and vascularization-related fields. There were 17 nodes in the PPI network of the target genes of up-regulating genes including Itgb2, Lox, Col11a1, Wnt5a, Kras, etc. Using quantitative RT-PCR, microarray sequencing accuracy was validated. There were 169 nodes constructing the PPI network of down-regulated target genes, mainly including Col1a1, Hmgcs1, Hmgcr. CeRNA interaction networks were constructed. CONCLUSION Lipid metabolism, angiogenesis, and ECM fields might play an important role in the senescence process in SNs. Col3a1, Serpinh1, Hmgcr, and Fdps could be candidates for nerve aging research.
Collapse
Affiliation(s)
- Rui Kuang
- Department of Plastic Surgery, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan Road 2, Guangzhou 510080, China
| | - Yi Zhang
- Department of Plastic Surgery, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan Road 2, Guangzhou 510080, China
| | - Guanggeng Wu
- Department of Plastic Surgery, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan Road 2, Guangzhou 510080, China
| | - Zhaowei Zhu
- Department of Plastic Surgery, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan Road 2, Guangzhou 510080, China
| | - Shuqia Xu
- Department of Plastic Surgery, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan Road 2, Guangzhou 510080, China
| | - Xiangxia Liu
- Department of Plastic Surgery, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Yangbin Xu
- Department of Plastic Surgery, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan Road 2, Guangzhou 510080, China
| | - Yunxiang Luo
- Department of Plastic Surgery, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan Road 2, Guangzhou 510080, China
| |
Collapse
|
12
|
Fuentes-Flores A, Geronimo-Olvera C, Girardi K, Necuñir-Ibarra D, Patel SK, Bons J, Wright MC, Geschwind D, Hoke A, Gomez-Sanchez JA, Schilling B, Rebolledo DL, Campisi J, Court FA. Senescent Schwann cells induced by aging and chronic denervation impair axonal regeneration following peripheral nerve injury. EMBO Mol Med 2023; 15:e17907. [PMID: 37860842 DOI: 10.15252/emmm.202317907] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 09/14/2023] [Accepted: 09/18/2023] [Indexed: 10/21/2023] Open
Abstract
Following peripheral nerve injury, successful axonal growth and functional recovery require Schwann cell (SC) reprogramming into a reparative phenotype, a process dependent upon c-Jun transcription factor activation. Unfortunately, axonal regeneration is greatly impaired in aged organisms and following chronic denervation, which can lead to poor clinical outcomes. While diminished c-Jun expression in SCs has been associated with regenerative failure, it is unclear whether the inability to maintain a repair state is associated with the transition into an axonal growth inhibition phenotype. We here find that reparative SCs transition into a senescent phenotype, characterized by diminished c-Jun expression and secretion of inhibitory factors for axonal regeneration in aging and chronic denervation. In both conditions, the elimination of senescent SCs by systemic senolytic drug treatment or genetic targeting improved nerve regeneration and functional recovery, increased c-Jun expression and decreased nerve inflammation. This work provides the first characterization of senescent SCs and their influence on axonal regeneration in aging and chronic denervation, opening new avenues for enhancing regeneration and functional recovery after peripheral nerve injuries.
Collapse
Affiliation(s)
- Andrés Fuentes-Flores
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism (GERO), Santiago, Chile
| | - Cristian Geronimo-Olvera
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism (GERO), Santiago, Chile
| | - Karina Girardi
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism (GERO), Santiago, Chile
| | - David Necuñir-Ibarra
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism (GERO), Santiago, Chile
| | | | - Joanna Bons
- Buck Institute for Research on Aging, Novato, CA, USA
| | - Megan C Wright
- Departments of Neurology and Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Daniel Geschwind
- Departments of Neurology, Psychiatry, and Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Ahmet Hoke
- Departments of Neurology and Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Jose A Gomez-Sanchez
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Alicante, Spain
- Instituto de Neurociencias de Alicante, UMH-CSIC, San Juan de Alicante, Spain
| | | | - Daniela L Rebolledo
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism (GERO), Santiago, Chile
| | | | - Felipe A Court
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism (GERO), Santiago, Chile
- Buck Institute for Research on Aging, Novato, CA, USA
| |
Collapse
|
13
|
Huang W, Yi S, Zhao L. Genetic Features of Young and Aged Animals After Peripheral Nerve Injury: Implications for Diminished Regeneration Capacity. Cell Mol Neurobiol 2023; 43:4363-4375. [PMID: 37922116 PMCID: PMC10661822 DOI: 10.1007/s10571-023-01431-8] [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: 07/04/2023] [Accepted: 10/27/2023] [Indexed: 11/05/2023]
Abstract
The spontaneous regeneration capacity of peripheral nerves is fundamentally reduced with advancing age, leading to severe and long-term functional loss. The cellular and molecular basis underlying incomplete and delayed recovery of aging peripheral nerves is still murky. Here, we collected sciatic nerves of aged rats at 1d, 4d, and 7d after nerve injury, systematically analyzed the transcriptional changes of injured sciatic nerves, and examined the differences of injury responses between aged rats and young rats. RNA sequencing revealed that sciatic nerves of aged and young rats exhibit distinctive expression patterns after nerve injury. Acute and vigorous immune responses, including motivated B cell receptor signaling pathway, occurred in injured sciatic nerves of both aged and young rats. Different from young rats, aged rats have more CD8+ T cells and B cells in normal state and the elevation of M2 macrophages seemed to be more robust in sciatic nerves, especially at later time points after nerve injury. Young rats, on the other hand, showed strong and early up-regulation of cell cycle-related genes. These identified unique transcriptional signatures of aged and young rats help the understanding of aged-associated injury responses in the wound microenvironments and provide essential basis for the treatment of regeneration deficits in aged population.
Collapse
Affiliation(s)
- Weixiao Huang
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Sheng Yi
- 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, China
| | - Lili Zhao
- 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, China.
| |
Collapse
|
14
|
Zhang WJ, Liu SC, Ming LG, Yu JW, Zuo C, Hu DX, Luo HL, Zhang Q. Potential role of Schwann cells in neuropathic pain. Eur J Pharmacol 2023; 956:175955. [PMID: 37541365 DOI: 10.1016/j.ejphar.2023.175955] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 07/26/2023] [Accepted: 08/01/2023] [Indexed: 08/06/2023]
Abstract
Neuropathic pain (NPP) is a common syndrome associated with most forms of disease, which poses a serious threat to human health. NPP may persist even after the nociceptive stimulation is eliminated, and treatment is extremely challenging in such cases. Schwann cells (SCs) form the myelin sheaths around neuronal axons and play a crucial role in neural information transmission. SCs can secrete trophic factors to nourish and protect axons, and can further secrete pain-related factors to induce pain. SCs may be activated by peripheral nerve injury, triggering the transformation of myelinated and non-myelinated SCs into cell phenotypes that specifically promote repair. These differentiated SCs provide necessary signals and spatial clues for survival, axonal regeneration, and nerve regeneration of damaged neurons. They can further change the microenvironment around the regions of nerve injury, and relieve the pain by repairing the injured nerve. Herein, we provide a comprehensive overview of the biological characteristics of SCs, discuss the relationship between SCs and nerve injury, and explore the potential mechanism of SCs and the occurrence of NPP. Moreover, we summarize the feasible strategies of SCs in the treatment of NPP, and attempt to elucidate the deficiencies and defects of SCs in the treatment of NPP.
Collapse
Affiliation(s)
- Wen-Jun Zhang
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Nanchang University, Nanchang City, Jiangxi province, 343000, China
| | - Si-Cheng Liu
- Department of Gastrointestinal surgery, The Second Affiliated Hospital, Nanchang University, Nanchang City, Jiangxi province, 343000, China
| | - Li-Guo Ming
- Department of Gastrointestinal surgery, The Second Affiliated Hospital, Nanchang University, Nanchang City, Jiangxi province, 343000, China
| | - Jian-Wen Yu
- Department of Gastrointestinal surgery, The Second Affiliated Hospital, Nanchang University, Nanchang City, Jiangxi province, 343000, China
| | - Cheng Zuo
- Department of Gastrointestinal surgery, The Second Affiliated Hospital, Nanchang University, Nanchang City, Jiangxi province, 343000, China
| | - Dong-Xia Hu
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Nanchang University, Nanchang City, Jiangxi province, 343000, China
| | - Hong-Liang Luo
- Department of Gastrointestinal surgery, The Second Affiliated Hospital, Nanchang University, Nanchang City, Jiangxi province, 343000, China.
| | - Qiao Zhang
- Orthopedics Department, The Second Affiliated Hospital, Nanchang University, Nanchang City, Jiangxi province, 343000, China.
| |
Collapse
|
15
|
Shashi KK, Shahin MM, Johnston P, Shaikh R. Cryoablation for Bone and Soft Tissue Lesions in Pediatric Patients: Complications and Preventive Measures. Cardiovasc Intervent Radiol 2023; 46:1249-1256. [PMID: 37580423 DOI: 10.1007/s00270-023-03523-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 07/21/2023] [Indexed: 08/16/2023]
Abstract
PURPOSE To evaluate complications associated with cryoablation in a pediatric population and review preventive measures to mitigate these complications. MATERIAL AND METHODS Retrospective study including all the image guided cryoablations performed on pediatric population. Immediate and delayed complications were analyzed, and we identified the different protective measures used and the clinical outcomes from follow-up. Point estimates for the percentage of complications were calculated by maximum likelihood, and 95% confidence intervals for the true percentages were calculated using the Clopper-Pearson exact method. RESULTS Eighty-seven ablations were performed on 68 patients (age range of 2-18 years, mean 12.4 years) for non-neoplastic (70%) and neoplastic (30%) lesions. The percentage of ablations resulting in complications was 18% (95% confidence interval (CI) 11% to 28%). Of these, 5% (95% CI 1% to 11%) were grade 3 complications, and 14% (95% CI 7% to 23%) were grade 2 complications. Thermal protection was performed in 27.6% of ablations (n = 24). The mean clinical follow-up duration was 348 days. CONCLUSION Cryoablation in pediatric patients is relatively safe, with a major complication rate per ablation of 5%. Thermal protective measures can be considered to mitigate these complications.
Collapse
Affiliation(s)
- Kumar Kempegowda Shashi
- Department of Radiology, Arkansas Children's Hospital, 1 Children's Way, Little Rock, AR, 72202, USA
| | - Mohamed M Shahin
- Department of Radiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
| | - Patrick Johnston
- Department of Radiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
| | - Raja Shaikh
- Department of Radiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA.
| |
Collapse
|
16
|
Zhang H, Zhang Z, Lin H. Research progress on the reduced neural repair ability of aging Schwann cells. Front Cell Neurosci 2023; 17:1228282. [PMID: 37545880 PMCID: PMC10398339 DOI: 10.3389/fncel.2023.1228282] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 07/06/2023] [Indexed: 08/08/2023] Open
Abstract
Peripheral nerve injury (PNI) is associated with delayed repair of the injured nerves in elderly patients, resulting in loss of nerve function, chronic pain, muscle atrophy, and permanent disability. Therefore, the mechanism underlying the delayed repair of peripheral nerves in aging patients should be investigated. Schwann cells (SCs) play a crucial role in repairing PNI and regulating various nerve-repair genes after injury. SCs also promote peripheral nerve repair through various modalities, including mediating nerve demyelination, secreting neurotrophic factors, establishing Büngner bands, clearing axon and myelin debris, and promoting axon remyelination. However, aged SCs undergo structural and functional changes, leading to demyelination and dedifferentiation disorders, decreased secretion of neurotrophic factors, impaired clearance of axonal and myelin debris, and reduced capacity for axon remyelination. As a result, aged SCs may result in delayed repair of nerves after injury. This review article aimed to examine the mechanism underlying the diminished neural repair ability of aging SCs.
Collapse
|
17
|
Maita KC, Garcia JP, Avila FR, Ricardo A TG, Ho OA, Claudia C S C, Eduardo N C, Forte AJ. Evaluation of the Aging Effect on Peripheral Nerve Regeneration: A Systematic Review. J Surg Res 2023; 288:329-340. [PMID: 37060859 DOI: 10.1016/j.jss.2023.03.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 01/18/2023] [Accepted: 03/16/2023] [Indexed: 04/17/2023]
Abstract
INTRODUCTION Peripheral nerve injuries have been associated with increased healthcare costs and decreased patients' quality of life. Aging represents one factor that slows the speed of peripheral nervous system (PNS) regeneration. Since cellular homeostasis imbalance associated with aging lead to an increased failure in nerve regeneration in mammals of advanced age, this systematic review aims to determine the main molecular and cellular mechanisms involved in peripheral nerve regeneration in aged murine models after a peripheral nerve injuries. METHODS Following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines, a literature search of 4 databases was conducted in July 2022 for studies comparing the peripheral nerve regeneration capability between young and aged murine models. RESULTS After the initial search yielded 744 publications, ten articles fulfilled the inclusion criteria. These studies show that age-related changes such as chronic inflammatory state, delayed macrophages' response to injury, dysfunctional Schwann Cells (SCs), and microenvironment alterations cause a reduction in the regenerative capability of the PNS in murine models. Furthermore, identifying altered gene expression patterns of SC after nerve damage can contribute to the understanding of physiological modifications produced by aging. CONCLUSIONS The interaction between macrophages and SC plays a crucial role in the nerve regeneration of aged models. Therefore, studies aimed at developing new and promising therapies for nerve regeneration should focus on these cellular groups to enhance the regenerative capabilities of the PNS in elderly populations.
Collapse
Affiliation(s)
- Karla C Maita
- Division of Plastic Surgery, Mayo Clinic, Jacksonville, Florida
| | - John P Garcia
- Division of Plastic Surgery, Mayo Clinic, Jacksonville, Florida
| | | | | | - Olivia A Ho
- Division of Plastic Surgery, Mayo Clinic, Jacksonville, Florida
| | - Chini Claudia C S
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Jacksonville, Florida
| | - Chini Eduardo N
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Jacksonville, Florida
| | - Antonio J Forte
- Division of Plastic Surgery, Mayo Clinic, Jacksonville, Florida.
| |
Collapse
|
18
|
Abd Razak NH, Zainey AS, Idris J, Daud MF. The Fundamentals of Schwann Cell Biology. INDUSTRIAL REVOLUTION IN KNOWLEDGE MANAGEMENT AND TECHNOLOGY 2023:105-113. [DOI: 10.1007/978-3-031-29265-1_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
|
19
|
Li C, Li X, Shi Z, Wu P, Fu J, Tang J, Qing L. Exosomes from LPS-preconditioned bone marrow MSCs accelerated peripheral nerve regeneration via M2 macrophage polarization: Involvement of TSG-6/NF-κB/NLRP3 signaling pathway. Exp Neurol 2022; 356:114139. [PMID: 35690131 DOI: 10.1016/j.expneurol.2022.114139] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 06/02/2022] [Accepted: 06/03/2022] [Indexed: 11/30/2022]
Abstract
Lipopolysaccharide (LPS)-preconditioned mesenchymal stem cells (MSCs) possessed strong immunomodulatory and anti-inflammatory functions by secreting exosomes as major paracrine effectors. However, the specific effect of exosomes from LPS pre-MSCs (LPS pre-Exos) on peripheral nerve regeneration has yet to be documented. Here, we established a sciatic nerve injury model in rats and an inflammatory model in RAW264.7 cells to explore the potential mechanism between LPS pre-Exos and peripheral nerve repair. The local injection of LPS pre-Exos into the nerve injury site resulted in an accelerated functional recovery, axon regeneration and remyelination, and an enhanced M2 Macrophage polarization. Consistent with the data in vivo, LPS pre-Exos were able to shift the pro-inflammation macrophage into a pro-regeneration macrophage. Notably, TNF stimulated gene-6 (TSG-6) was found to be highly enriched in LPS pre-Exos. We obtained si TSG-6 Exo by the knockdown of TSG-6 in LPS pre-Exos to demonstrate the role of TSG-6 in macrophage polarization, and found that TSG-6 served as a critical mediator in LPS pre-Exos-induced regulatory effects through the inhibition of NF-ΚΒ and NOD-like receptor protein 3 (NLRP3). In conclusion, our findings suggested that LPS pre-Exos promoted macrophage polarization toward an M2 phenotype by shuttling TSG-6 to inactivate the NF-ΚΒ/NLRP3 signaling axis, and could provide a potential therapeutic avenue for peripheral nerve repair.
Collapse
Affiliation(s)
- Cheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoxiao Li
- Department of Pathology, Changsha Medical University, Changsha, China; Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
| | - Zhen Shi
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China; Department of Plastic Surgery, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, China
| | - Panfeng Wu
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
| | - Jinfei Fu
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
| | - Juyu Tang
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
| | - Liming Qing
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
20
|
Identification and quantification of nociceptive Schwann cells in mice with and without Streptozotocin-induced diabetes. J Chem Neuroanat 2022; 123:102118. [DOI: 10.1016/j.jchemneu.2022.102118] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 06/03/2022] [Accepted: 06/04/2022] [Indexed: 12/31/2022]
|
21
|
Li J, Yao Y, Wang Y, Xu J, Zhao D, Liu M, Shi S, Lin Y. Modulation of the Crosstalk between Schwann Cells and Macrophages for Nerve Regeneration: A Therapeutic Strategy Based on a Multifunctional Tetrahedral Framework Nucleic Acids System. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2202513. [PMID: 35483031 DOI: 10.1002/adma.202202513] [Citation(s) in RCA: 84] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/17/2022] [Indexed: 02/05/2023]
Abstract
Peripheral nerve injury (PNI) is currently recognized as one of the most significant public health issues and affects the general well-being of millions of individuals worldwide. Despite advances in nerve tissue engineering, nerve repair still cannot guarantee complete functional recovery. In the present study, an innovative approach is adopted to establish a multifunctional tetrahedral framework nucleic acids (tFNAs) system, denoted as MiDs, which can integrate the powerful programmability, permeability, and structural stability of tFNAs, with the nerve regeneration potential of microRNA-22 to enhance the communication between Schwann cells (SCs) and macrophages for more effective functional rehabilitation of peripheral nerves. Relevant results demonstrate that MiDs can amplify the ability of SCs to recruit macrophages and facilitate their polarization into the pro-healing M2 phenotype to reconstruct the post-injury microenvironment. Furthermore, MiDs can initiate the adaptive intracellular reprogramming of SCs within a short period to further promote axon regeneration and remyelination. MiDs represent a new possibility for enhancing nerve repair and may have critical clinical applications in the future.
Collapse
Affiliation(s)
- Jiajie Li
- State Key Laboratory of Oral Diseases National Clinical Research Center for Oral Diseases West China Hospital of Stomatology Sichuan University Chengdu 610041 P. R. China
| | - Yangxue Yao
- State Key Laboratory of Oral Diseases National Clinical Research Center for Oral Diseases West China Hospital of Stomatology Sichuan University Chengdu 610041 P. R. China
| | - Yun Wang
- State Key Laboratory of Oral Diseases National Clinical Research Center for Oral Diseases West China Hospital of Stomatology Sichuan University Chengdu 610041 P. R. China
| | - Jiangshan Xu
- College of Biomedical Engineering Sichuan University Chengdu 610041 P. R. China
| | - Dan Zhao
- State Key Laboratory of Oral Diseases National Clinical Research Center for Oral Diseases West China Hospital of Stomatology Sichuan University Chengdu 610041 P. R. China
| | - Mengting Liu
- State Key Laboratory of Oral Diseases National Clinical Research Center for Oral Diseases West China Hospital of Stomatology Sichuan University Chengdu 610041 P. R. China
| | - Sirong Shi
- State Key Laboratory of Oral Diseases National Clinical Research Center for Oral Diseases West China Hospital of Stomatology Sichuan University Chengdu 610041 P. R. China
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases National Clinical Research Center for Oral Diseases West China Hospital of Stomatology Sichuan University Chengdu 610041 P. R. China
- College of Biomedical Engineering Sichuan University Chengdu 610041 P. R. China
| |
Collapse
|
22
|
Moss KR, Johnson AE, Bopp TS, Yu ATY, Perry K, Chung T, Höke A. SARM1 knockout does not rescue neuromuscular phenotypes in a Charcot-Marie-Tooth disease Type 1A mouse model. J Peripher Nerv Syst 2022; 27:58-66. [PMID: 35137510 PMCID: PMC8940700 DOI: 10.1111/jns.12483] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 01/25/2022] [Accepted: 02/02/2022] [Indexed: 11/28/2022]
Abstract
Charcot-Marie-Tooth disease Type 1A (CMT1A) is caused by duplication of the PMP22 gene and is the most common inherited peripheral neuropathy. Although CMT1A is a dysmyelinating peripheral neuropathy, secondary axon degeneration has been suggested to drive functional deficits in patients. Given that SARM1 knockout is a potent inhibitor of the programmed axon degeneration pathway, we asked whether SARM1 knockout rescues neuromuscular phenotypes in CMT1A model (C3-PMP) mice. CMT1A mice were bred with SARM1 knockout mice to generate CMT1A/SARM1-/- mice. A series of behavioral assays were employed to evaluate motor and sensorimotor function. Electrophysiological and histological studies of the tibial branch of the sciatic nerve were performed. Additionally, gastrocnemius and soleus muscle morphology were evaluated histologically. Although clear behavioral and electrophysiological deficits were observed in CMT1A model mice, genetic deletion of SARM1 conferred no significant improvement. Nerve morphometry revealed predominantly myelin deficits in CMT1A model mice and SARM1 knockout yielded no improvement in all nerve morphometry measures. Similarly, muscle morphometry deficits in CMT1A model mice were not improved by SARM1 knockout. Our findings demonstrate that programmed axon degeneration pathway inhibition does not provide therapeutic benefit in C3-PMP CMT1A model mice. Our results indicate that the clinical phenotypes observed in CMT1A mice are likely caused primarily by prolonged dysmyelination, motivate further investigation into mechanisms of dysmyelination in these mice and necessitate the development of improved CMT1A rodent models that recapitulate the secondary axon degeneration observed in patients.
Collapse
Affiliation(s)
- Kathryn R. Moss
- Department of Neurology, Neuromuscular Division, Johns Hopkins School of Medicine, Baltimore, MD
| | - Anna E. Johnson
- Department of Neurology, Neuromuscular Division, Johns Hopkins School of Medicine, Baltimore, MD
| | - Taylor S. Bopp
- Department of Physical Medicine and Rehabilitation, Johns Hopkins School of Medicine, Baltimore, MD
| | - Andrew T-Y. Yu
- Department of Neurology, Neuromuscular Division, Johns Hopkins School of Medicine, Baltimore, MD
| | - Ken Perry
- Department of Neurology, Neuromuscular Division, Johns Hopkins School of Medicine, Baltimore, MD
| | - Tae Chung
- Department of Physical Medicine and Rehabilitation, Johns Hopkins School of Medicine, Baltimore, MD
| | - Ahmet Höke
- Department of Neurology, Neuromuscular Division, Johns Hopkins School of Medicine, Baltimore, MD,Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD,Corresponding Author: Ahmet Höke MD, PhD, Johns Hopkins School of Medicine, 855 N. Wolfe St., Baltimore, MD 21205, Tel: 410-955-2227, Fax: 410-502-5459,
| |
Collapse
|
23
|
Sardella-Silva G, Mietto BS, Ribeiro-Resende VT. Four Seasons for Schwann Cell Biology, Revisiting Key Periods: Development, Homeostasis, Repair, and Aging. Biomolecules 2021; 11:1887. [PMID: 34944531 PMCID: PMC8699407 DOI: 10.3390/biom11121887] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 12/08/2021] [Accepted: 12/10/2021] [Indexed: 01/28/2023] Open
Abstract
Like the seasons of the year, all natural things happen in stages, going through adaptations when challenged, and Schwann cells are a great example of that. During maturation, these cells regulate several steps in peripheral nervous system development. The Spring of the cell means the rise and bloom through organized stages defined by time-dependent regulation of factors and microenvironmental influences. Once matured, the Summer of the cell begins: a high energy stage focused on maintaining adult homeostasis. The Schwann cell provides many neuron-glia communications resulting in the maintenance of synapses. In the peripheral nervous system, Schwann cells are pivotal after injuries, balancing degeneration and regeneration, similarly to when Autumn comes. Their ability to acquire a repair phenotype brings the potential to reconnect axons to targets and regain function. Finally, Schwann cells age, not only by growing old, but also by imposed environmental cues, like loss of function induced by pathologies. The Winter of the cell presents as reduced activity, especially regarding their role in repair; this reflects on the regenerative potential of older/less healthy individuals. This review gathers essential information about Schwann cells in different stages, summarizing important participation of this intriguing cell in many functions throughout its lifetime.
Collapse
Affiliation(s)
- Gabriela Sardella-Silva
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil;
- Núcleo Multidisciplinar de Pesquisa em Biologia (Numpex-Bio), Campus de Duque de Caxias Geraldo Guerra Cidade, Universidade Federal do Rio de Janeiro, Duque de Caxias 25255-030, RJ, Brazil
| | - Bruno Siqueira Mietto
- Instituto de Ciências Biológicas, Universidade Federal de Juiz de Fora, Juiz de Fora 36036-900, MG, Brazil;
| | - Victor Túlio Ribeiro-Resende
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil;
- Núcleo Multidisciplinar de Pesquisa em Biologia (Numpex-Bio), Campus de Duque de Caxias Geraldo Guerra Cidade, Universidade Federal do Rio de Janeiro, Duque de Caxias 25255-030, RJ, Brazil
| |
Collapse
|
24
|
Shi G, Hao D, Zhang L, Qin J, Tian G, Ma B, Zhou X. Endocytosis-associated patterns in nerve regeneration after peripheral nerve injury. J Orthop Translat 2021; 31:10-19. [PMID: 34760620 PMCID: PMC8551787 DOI: 10.1016/j.jot.2021.09.004] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 08/29/2021] [Accepted: 09/13/2021] [Indexed: 11/25/2022] Open
Abstract
Background Clearance of myelin debris and remyelination of myelin are necessary steps for peripheral nerve remodeling and regeneration. It has yet to be clarified which genes or proteins are involved in endocytosis or exocytosis in the removal of myelin debris during peripheral nerve repair. Methods For this project, a rat model of subacute stage of sciatic nerve injury was established first. Subsequently, normal Schwann cells (NSCs) and activated Schwann cells (ASCs) were harvest before and after peripheral nerve injury (PNI). Following methylated DNA immunoprecipitation sequencing (MeDIP-seq) and tandem mass tags (TMT) labeling analysis of NSCs and ASCs, what common biomarkers changes in peripheral nervous systems remain to be elucidated. Results A total of 14,770 different expression genes (DEGs) and 3249 different expression proteins (DEPs) were screened between ASCs and NSCs. For the exosomes, the diameter and particles concentration of exosomes were 141.7 nm and 2.97 × 107 particles/mL, respectively. The size distribution of exosomes was 50–200 nm. ASCs showed higher cellular uptake ability than the NSCs by cellular uptake test. Moreover, RAB7A, ARF6, ARF1, VPS45, RAB11A, DNM3, and NEDD4 were the core markers and may control the molecular mechanism of the Endocytosis pathway. Conclusion These biomarkers may play significant roles in the initiation phase of demyelination and axon regeneration. The translational potential of this article This study explores that the endocytosis-associated patterns of Schwann cells may be new therapeutic strategy for nerve tissue engineering and nerve regeneration.
Collapse
Affiliation(s)
- Guidong Shi
- Department of Orthopaedics, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.,Shandong University Centre for Orthopaedics, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Dingyu Hao
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Lei Zhang
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Jia Qin
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Guangyuan Tian
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Boyuan Ma
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Xianhu Zhou
- The Affiliated Hospital of Medical School, Ningbo University, 247 People Road, Jiangbei District, Ningbo, Zhejiang, China
| |
Collapse
|
25
|
Slavin BR, Sarhane KA, von Guionneau N, Hanwright PJ, Qiu C, Mao HQ, Höke A, Tuffaha SH. Insulin-Like Growth Factor-1: A Promising Therapeutic Target for Peripheral Nerve Injury. Front Bioeng Biotechnol 2021; 9:695850. [PMID: 34249891 PMCID: PMC8264584 DOI: 10.3389/fbioe.2021.695850] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 06/02/2021] [Indexed: 01/27/2023] Open
Abstract
Patients who sustain peripheral nerve injuries (PNIs) are often left with debilitating sensory and motor loss. Presently, there is a lack of clinically available therapeutics that can be given as an adjunct to surgical repair to enhance the regenerative process. Insulin-like growth factor-1 (IGF-1) represents a promising therapeutic target to meet this need, given its well-described trophic and anti-apoptotic effects on neurons, Schwann cells (SCs), and myocytes. Here, we review the literature regarding the therapeutic potential of IGF-1 in PNI. We appraised the literature for the various approaches of IGF-1 administration with the aim of identifying which are the most promising in offering a pathway toward clinical application. We also sought to determine the optimal reported dosage ranges for the various delivery approaches that have been investigated.
Collapse
Affiliation(s)
- Benjamin R Slavin
- Department of Plastic and Reconstructive Surgery, School of Medicine, Johns Hopkins University, Baltimore, MD, United States.,Division of Plastic and Reconstructive Surgery, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Karim A Sarhane
- Department of Plastic and Reconstructive Surgery, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Nicholas von Guionneau
- Department of Plastic and Reconstructive Surgery, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Phillip J Hanwright
- Department of Plastic and Reconstructive Surgery, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Chenhu Qiu
- Department of Materials Science and Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, United States.,Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, United States
| | - Hai-Quan Mao
- Department of Materials Science and Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, United States.,Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, United States.,Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD, United States.,Translational Tissue Engineering Center, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Ahmet Höke
- Department of Neurology, School of Medicine, Johns Hopkins University, Baltimore, MD, United States.,Department of Neuroscience, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Sami H Tuffaha
- Department of Plastic and Reconstructive Surgery, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
26
|
Ward PJ, Davey RA, Zajac JD, English AW. Neuronal androgen receptor is required for activity dependent enhancement of peripheral nerve regeneration. Dev Neurobiol 2021; 81:411-423. [PMID: 33864349 DOI: 10.1002/dneu.22826] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 03/29/2021] [Accepted: 03/30/2021] [Indexed: 11/11/2022]
Abstract
Neuronal activity after nerve injury can enhance axon regeneration and the restoration of function. The mechanism for this enhancement relies in part on hormone receptors, and we previously demonstrated that systemic androgen receptor antagonism blocked the effect of exercise or electrical stimulation on enhancing axon regeneration after nerve injury in both sexes. Here, we tested the hypothesis that the site of this androgen receptor signaling is both neuronal and involves the classical, genomic signaling pathway. In vivo, dorsal root ganglion neurons successfully regenerate in response to activity-dependent neuronal activation, and conditional deletion of the DNA-binding domain of the androgen receptor in adults blocks this effect in males and females. Motoneurons in males and females also respond in this manner, but we also observed a sex difference. In vitro, cultured sensory dorsal root ganglion neurons respond to androgens via traditional androgen receptor signaling mechanisms leading to enhanced neurite growth and did not respond to a testosterone conjugate that is unable to cross the cell membrane. Given our previous observation of a requirement for activity-dependent androgen receptor signaling to promote regeneration in both sexes, we interpret our results to indicate that genomic neuronal androgen receptor signaling is required for activity-dependent axon regeneration in both sexes.
Collapse
Affiliation(s)
- Patricia J Ward
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, USA
| | - Rachel A Davey
- Department of Medicine, Austin Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Jeffrey D Zajac
- Department of Medicine, Austin Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Arthur W English
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
27
|
van Erp S, van Berkel AA, Feenstra EM, Sahoo PK, Wagstaff LJ, Twiss JL, Fawcett JW, Eva R, Ffrench-Constant C. Age-related loss of axonal regeneration is reflected by the level of local translation. Exp Neurol 2021; 339:113594. [PMID: 33450233 PMCID: PMC8024785 DOI: 10.1016/j.expneurol.2020.113594] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 12/07/2020] [Accepted: 12/17/2020] [Indexed: 01/08/2023]
Abstract
Regeneration capacity is reduced as CNS axons mature. Using laser-mediated axotomy, proteomics and puromycin-based tagging of newly-synthesized proteins in a human embryonic stem cell-derived neuron culture system that allows isolation of axons from cell bodies, we show here that efficient regeneration in younger axons (d45 in culture) is associated with local axonal protein synthesis (local translation). Enhanced regeneration, promoted by co-culture with human glial precursor cells, is associated with increased axonal synthesis of proteins, including those constituting the translation machinery itself. Reduced regeneration, as occurs with the maturation of these axons by d65 in culture, correlates with reduced levels of axonal proteins involved in translation and an inability to respond by increased translation of regeneration promoting axonal mRNAs released from stress granules. Together, our results provide evidence that, as in development and in the PNS, local translation contributes to CNS axon regeneration.
Collapse
Affiliation(s)
- Susan van Erp
- MRC Centre for Regenerative Medicine and MS Society Edinburgh Centre, Edinburgh bioQuarter, University of Edinburgh, Edinburgh, UK.
| | - Annemiek A van Berkel
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research (CNCR), VU University Amsterdam, De Boelelaan 1085, 1081, HV, Amsterdam, the Netherlands
| | - Eline M Feenstra
- MRC Centre for Regenerative Medicine and MS Society Edinburgh Centre, Edinburgh bioQuarter, University of Edinburgh, Edinburgh, UK
| | - Pabitra K Sahoo
- Department of Biological Sciences, University of South Carolina, Columbia 29208, SC, USA
| | - Laura J Wagstaff
- MRC Centre for Regenerative Medicine and MS Society Edinburgh Centre, Edinburgh bioQuarter, University of Edinburgh, Edinburgh, UK
| | - Jeffery L Twiss
- Department of Biological Sciences, University of South Carolina, Columbia 29208, SC, USA
| | - James W Fawcett
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK; Centre for Reconstructive Neuroscience, Institute of Experimental Medicine, Czech Academy of Sciences, Prague, Czech Republic
| | - Richard Eva
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Charles Ffrench-Constant
- MRC Centre for Regenerative Medicine and MS Society Edinburgh Centre, Edinburgh bioQuarter, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
28
|
Nazareth L, St John J, Murtaza M, Ekberg J. Phagocytosis by Peripheral Glia: Importance for Nervous System Functions and Implications in Injury and Disease. Front Cell Dev Biol 2021; 9:660259. [PMID: 33898462 PMCID: PMC8060502 DOI: 10.3389/fcell.2021.660259] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 03/17/2021] [Indexed: 12/30/2022] Open
Abstract
The central nervous system (CNS) has very limited capacity to regenerate after traumatic injury or disease. In contrast, the peripheral nervous system (PNS) has far greater capacity for regeneration. This difference can be partly attributed to variances in glial-mediated functions, such as axon guidance, structural support, secretion of growth factors and phagocytic activity. Due to their growth-promoting characteristic, transplantation of PNS glia has been trialed for neural repair. After peripheral nerve injuries, Schwann cells (SCs, the main PNS glia) phagocytose myelin debris and attract macrophages to the injury site to aid in debris clearance. One peripheral nerve, the olfactory nerve, is unique in that it continuously regenerates throughout life. The olfactory nerve glia, olfactory ensheathing cells (OECs), are the primary phagocytes within this nerve, continuously clearing axonal debris arising from the normal regeneration of the nerve and after injury. In contrast to SCs, OECs do not appear to attract macrophages. SCs and OECs also respond to and phagocytose bacteria, a function likely critical for tackling microbial invasion of the CNS via peripheral nerves. However, phagocytosis is not always effective; inflammation, aging and/or genetic factors may contribute to compromised phagocytic activity. Here, we highlight the diverse roles of SCs and OECs with the focus on their phagocytic activity under physiological and pathological conditions. We also explore why understanding the contribution of peripheral glia phagocytosis may provide us with translational strategies for achieving axonal regeneration of the injured nervous system and potentially for the treatment of certain neurological diseases.
Collapse
Affiliation(s)
- Lynn Nazareth
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia.,Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia
| | - James St John
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia.,Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia.,Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD, Australia
| | - Mariyam Murtaza
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia.,Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia.,Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD, Australia
| | - Jenny Ekberg
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia.,Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia.,Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD, Australia
| |
Collapse
|
29
|
Fuertes-Alvarez S, Izeta A. Terminal Schwann Cell Aging: Implications for Age-Associated Neuromuscular Dysfunction. Aging Dis 2021; 12:494-514. [PMID: 33815879 PMCID: PMC7990373 DOI: 10.14336/ad.2020.0708] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 07/08/2020] [Indexed: 12/11/2022] Open
Abstract
Action potential is transmitted to muscle fibers through specialized synaptic interfaces called neuromuscular junctions (NMJs). These structures are capped by terminal Schwann cells (tSCs), which play essential roles during formation and maintenance of the NMJ. tSCs are implicated in the correct communication between nerves and muscles, and in reinnervation upon injury. During aging, loss of muscle mass and strength (sarcopenia and dynapenia) are due, at least in part, to the progressive loss of contacts between muscle fibers and nerves. Despite the important role of tSCs in NMJ function, very little is known on their implication in the NMJ-aging process and in age-associated denervation. This review summarizes the current knowledge about the implication of tSCs in the age-associated degeneration of NMJs. We also speculate on the possible mechanisms underlying the observed phenotypes.
Collapse
Affiliation(s)
- Sandra Fuertes-Alvarez
- 1Biodonostia, Tissue Engineering Group, Paseo Dr. Begiristain, s/n, San Sebastian 20014, Spain
| | - Ander Izeta
- 1Biodonostia, Tissue Engineering Group, Paseo Dr. Begiristain, s/n, San Sebastian 20014, Spain.,2Tecnun-University of Navarra, School of Engineering, Department of Biomedical Engineering and Science, Paseo Mikeletegi, 48, San Sebastian 20009, Spain
| |
Collapse
|
30
|
Chakraborty A, Ciciriello AJ, Dumont CM, Pearson RM. Nanoparticle-Based Delivery to Treat Spinal Cord Injury-a Mini-review. AAPS PharmSciTech 2021; 22:101. [PMID: 33712968 PMCID: PMC8733957 DOI: 10.1208/s12249-021-01975-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 02/22/2021] [Indexed: 02/06/2023] Open
Abstract
There is an increasing need to develop improved and non-invasive strategies to treat spinal cord injury (SCI). Nanoparticles (NPs) are an enabling technology to improve drug delivery, modulate inflammatory responses, and restore functional responses following SCI. However, the complex pathophysiology associated with SCI presents several distinct challenges that must be overcome for sufficient NP drug delivery to the spinal cord. The objective of this mini-review is to highlight the physiological challenges and cell types available for modulation and discuss several promising advancements using NPs to improve SCI treatment. We will focus our discussion on recent innovative approaches in NP drug delivery and how the implementation of multifactorial approaches to address the proinflammatory and complex immune dysfunction in SCI offers significant potential to improve outcomes in SCI.
Collapse
Affiliation(s)
- Atanu Chakraborty
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 N. Pine Street, Baltimore, Maryland, 21201, USA
| | - Andrew J Ciciriello
- Department of Biomedical Engineering, University of Miami, 1251 Memorial Drive, Coral Gables, Florida, 33156, USA
- Biomedical Nanotechnology Institute at the University of Miami (BioNIUM), University of Miami, 1951 NW Seventh Avenue Suite 475, Miami, Florida, 33136, USA
| | - Courtney M Dumont
- Department of Biomedical Engineering, University of Miami, 1251 Memorial Drive, Coral Gables, Florida, 33156, USA.
- Biomedical Nanotechnology Institute at the University of Miami (BioNIUM), University of Miami, 1951 NW Seventh Avenue Suite 475, Miami, Florida, 33136, USA.
| | - Ryan M Pearson
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 N. Pine Street, Baltimore, Maryland, 21201, USA.
- Department of Molecular Microbiology and Immunology, University of Maryland School of Medicine, 685 W. Baltimore Street, Maryland, 21201, Baltimore, USA.
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, 22 S. Greene Street, Baltimore, Maryland, 21201, USA.
| |
Collapse
|
31
|
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: 76] [Impact Index Per Article: 19.0] [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
|
32
|
Wagstaff LJ, Gomez-Sanchez JA, Fazal SV, Otto GW, Kilpatrick AM, Michael K, Wong LYN, Ma KH, Turmaine M, Svaren J, Gordon T, Arthur-Farraj P, Velasco-Aviles S, Cabedo H, Benito C, Mirsky R, Jessen KR. Failures of nerve regeneration caused by aging or chronic denervation are rescued by restoring Schwann cell c-Jun. eLife 2021; 10:e62232. [PMID: 33475496 PMCID: PMC7819709 DOI: 10.7554/elife.62232] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 12/21/2020] [Indexed: 02/06/2023] Open
Abstract
After nerve injury, myelin and Remak Schwann cells reprogram to repair cells specialized for regeneration. Normally providing strong regenerative support, these cells fail in aging animals, and during chronic denervation that results from slow axon growth. This impairs axonal regeneration and causes significant clinical problems. In mice, we find that repair cells express reduced c-Jun protein as regenerative support provided by these cells declines during aging and chronic denervation. In both cases, genetically restoring Schwann cell c-Jun levels restores regeneration to control levels. We identify potential gene candidates mediating this effect and implicate Shh in the control of Schwann cell c-Jun levels. This establishes that a common mechanism, reduced c-Jun in Schwann cells, regulates success and failure of nerve repair both during aging and chronic denervation. This provides a molecular framework for addressing important clinical problems, suggesting molecular pathways that can be targeted to promote repair in the PNS.
Collapse
Affiliation(s)
- Laura J Wagstaff
- Department of Cell and Developmental Biology, University College LondonLondonUnited Kingdom
| | - Jose A Gomez-Sanchez
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández‐CSICSan Juan de AlicanteSpain
| | - Shaline V Fazal
- Department of Cell and Developmental Biology, University College LondonLondonUnited Kingdom
| | - Georg W Otto
- University College London Great Ormond Street Institute of Child HealthLondonUnited Kingdom
| | - Alastair M Kilpatrick
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of EdinburghEdinburghUnited Kingdom
| | - Kirolos Michael
- Department of Cell and Developmental Biology, University College LondonLondonUnited Kingdom
| | - Liam YN Wong
- Department of Cell and Developmental Biology, University College LondonLondonUnited Kingdom
| | - Ki H Ma
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin‐MadisonMadisonUnited States
| | - Mark Turmaine
- Department of Cell and Developmental Biology, University College LondonLondonUnited Kingdom
| | - John Svaren
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin‐MadisonMadisonUnited States
| | - Tessa Gordon
- Division of Plastic and Reconstructive Surgery, The Hospital for Sick ChildrenTorontoCanada
| | - Peter Arthur-Farraj
- John Van Geest Centre for Brain repair, Department of Clinical Neurosciences, University of CambridgeCambridgeUnited Kingdom
| | - Sergio Velasco-Aviles
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández‐CSICSan Juan de AlicanteSpain
- Hospital General Universitario de Alicante, ISABIALAlicanteSpain
| | - Hugo Cabedo
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández‐CSICSan Juan de AlicanteSpain
- Hospital General Universitario de Alicante, ISABIALAlicanteSpain
| | - Cristina Benito
- Department of Cell and Developmental Biology, University College LondonLondonUnited Kingdom
| | - Rhona Mirsky
- Department of Cell and Developmental Biology, University College LondonLondonUnited Kingdom
| | - Kristjan R Jessen
- Department of Cell and Developmental Biology, University College LondonLondonUnited Kingdom
| |
Collapse
|
33
|
de Jongh R, Spijkers XM, Pasteuning-Vuhman S, Vulto P, Pasterkamp RJ. Neuromuscular junction-on-a-chip: ALS disease modeling and read-out development in microfluidic devices. J Neurochem 2021; 157:393-412. [PMID: 33382092 DOI: 10.1111/jnc.15289] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 12/24/2020] [Accepted: 12/28/2020] [Indexed: 12/21/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal and progressive neurodegenerative disease affecting upper and lower motor neurons with no cure available. Clinical and animal studies reveal that the neuromuscular junction (NMJ), a synaptic connection between motor neurons and skeletal muscle fibers, is highly vulnerable in ALS and suggest that NMJ defects may occur at the early stages of the disease. However, mechanistic insight into how NMJ dysfunction relates to the onset and progression of ALS is incomplete, which hampers therapy development. This is, in part, caused by a lack of robust in vitro models. The ability to combine microfluidic and induced pluripotent stem cell (iPSC) technologies has opened up new avenues for studying molecular and cellular ALS phenotypes in vitro. Microfluidic devices offer several advantages over traditional culture approaches when modeling the NMJ, such as the spatial separation of different cell types and increased control over the cellular microenvironment. Moreover, they are compatible with 3D cell culture, which enhances NMJ functionality and maturity. Here, we review how microfluidic technology is currently being employed to develop more reliable in vitro NMJ models. To validate and phenotype such models, various morphological and functional read-outs have been developed. We describe and discuss the relevance of these read-outs and specifically illustrate how these read-outs have enhanced our understanding of NMJ pathology in ALS. Finally, we share our view on potential future directions and challenges.
Collapse
Affiliation(s)
- Rianne de Jongh
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Xandor M Spijkers
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands.,Mimetas B.V., Organ-on-a-chip Company, Leiden, The Netherlands
| | - Svetlana Pasteuning-Vuhman
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Paul Vulto
- Mimetas B.V., Organ-on-a-chip Company, Leiden, The Netherlands
| | - R Jeroen Pasterkamp
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
34
|
Hagen KM, Ousman SS. The Neuroimmunology of Guillain-Barré Syndrome and the Potential Role of an Aging Immune System. Front Aging Neurosci 2021; 12:613628. [PMID: 33584245 PMCID: PMC7873882 DOI: 10.3389/fnagi.2020.613628] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 12/11/2020] [Indexed: 12/15/2022] Open
Abstract
Guillain-Barré syndrome (GBS) is a paralyzing autoimmune condition affecting the peripheral nervous system (PNS). Within GBS there are several variants affecting different aspects of the peripheral nerve. In general, there appears to be a role for T cells, macrophages, B cells, and complement in initiating and perpetuating attacks on gangliosides of Schwann cells and axons. Of note, GBS has an increased prevalence and severity with increasing age. In addition, there are alterations in immune cell functioning that may play a role in differences in GBS with age alongside general age-related declines in reparative processes (e.g., delayed de-differentiation of Schwann cells and decline in phagocytic ability of macrophages). The present review will explore the immune response in GBS as well as in animal models of several variants of the disorder. In addition, the potential involvement of an aging immune system in contributing to the increased prevalence and severity of GBS with age will be theorized.
Collapse
Affiliation(s)
- Kathleen M. Hagen
- Department of Neuroscience, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Shalina S. Ousman
- Departments of Clinical Neurosciences and Cell Biology and Anatomy, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
35
|
Marshall KL, Farah MH. Axonal regeneration and sprouting as a potential therapeutic target for nervous system disorders. Neural Regen Res 2021; 16:1901-1910. [PMID: 33642358 PMCID: PMC8343323 DOI: 10.4103/1673-5374.308077] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Nervous system disorders are prevalent health issues that will only continue to increase in frequency as the population ages. Dying-back axonopathy is a hallmark of many neurologic diseases and leads to axonal disconnection from their targets, which in turn leads to functional impairment. During the course of many of neurologic diseases, axons can regenerate or sprout in an attempt to reconnect with the target and restore synapse function. In amyotrophic lateral sclerosis (ALS), distal motor axons retract from neuromuscular junctions early in the disease-course before significant motor neuron death. There is evidence of compensatory motor axon sprouting and reinnervation of neuromuscular junctions in ALS that is usually quickly overtaken by the disease course. Potential drugs that enhance compensatory sprouting and encourage reinnervation may slow symptom progression and retain muscle function for a longer period of time in ALS and in other diseases that exhibit dying-back axonopathy. There remain many outstanding questions as to the impact of distinct disease-causing mutations on axonal outgrowth and regeneration, especially in regards to motor neurons derived from patient induced pluripotent stem cells. Compartmentalized microfluidic chambers are powerful tools for studying the distal axons of human induced pluripotent stem cells-derived motor neurons, and have recently been used to demonstrate striking regeneration defects in human motor neurons harboring ALS disease-causing mutations. Modeling the human neuromuscular circuit with human induced pluripotent stem cells-derived motor neurons will be critical for developing drugs that enhance axonal regeneration, sprouting, and reinnervation of neuromuscular junctions. In this review we will discuss compensatory axonal sprouting as a potential therapeutic target for ALS, and the use of compartmentalized microfluidic devices to find drugs that enhance regeneration and axonal sprouting of motor axons.
Collapse
Affiliation(s)
| | - Mohamed H Farah
- Department of Neurology at Johns Hopkins School of Medicine, Baltimore, MD, USA
| |
Collapse
|
36
|
Qu WR, Zhu Z, Liu J, Song DB, Tian H, Chen BP, Li R, Deng LX. Interaction between Schwann cells and other cells during repair of peripheral nerve injury. Neural Regen Res 2021; 16:93-98. [PMID: 32788452 PMCID: PMC7818858 DOI: 10.4103/1673-5374.286956] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Peripheral nerve injury (PNI) is common and, unlike damage to the central nervous system injured nerves can effectively regenerate depending on the location and severity of injury. Peripheral myelinating glia, Schwann cells (SCs), interact with various cells in and around the injury site and are important for debris elimination, repair, and nerve regeneration. Following PNI, Wallerian degeneration of the distal stump is rapidly initiated by degeneration of damaged axons followed by morphologic changes in SCs and the recruitment of circulating macrophages. Interaction with fibroblasts from the injured nerve microenvironment also plays a role in nerve repair. The replication and migration of injury-induced dedifferentiated SCs are also important in repairing the nerve. In particular, SC migration stimulates axonal regeneration and subsequent myelination of regenerated nerve fibers. This mobility increases SC interactions with other cells in the nerve and the exogenous environment, which influence SC behavior post-injury. Following PNI, SCs directly and indirectly interact with other SCs, fibroblasts, and macrophages. In addition, the inter- and intracellular mechanisms that underlie morphological and functional changes in SCs following PNI still require further research to explain known phenomena and less understood cell-specific roles in the repair of the injured peripheral nerve. This review provides a basic assessment of SC function post-PNI, as well as a more comprehensive evaluation of the literature concerning the SC interactions with macrophages and fibroblasts that can influence SC behavior and, ultimately, repair of the injured nerve.
Collapse
Affiliation(s)
- Wen-Rui Qu
- Department of Hand Surgery, the Second Hospital of Jilin University, Changchun, Jilin Province, China
| | - Zhe Zhu
- Department of Hand Surgery, the Second Hospital of Jilin University, Changchun, Jilin Province, China
| | - Jun Liu
- Department of Hand Surgery, the Second Hospital of Jilin University, Changchun, Jilin Province, China
| | - De-Biao Song
- Department of Emergency and Critical Medicine, the Second Hospital of Jilin University, Changchun, Jilin Province, China
| | - Heng Tian
- Department of Hand Surgery, the Second Hospital of Jilin University, Changchun, Jilin Province, China
| | - Bing-Peng Chen
- Orthopedic Medical Center, the Second Hospital of Jilin University, Changchun, Jilin Province, China
| | - Rui Li
- Department of Hand Surgery, the Second Hospital of Jilin University, Changchun, Jilin Province, China
| | - Ling-Xiao Deng
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
37
|
Huang Z, Powell R, Phillips JB, Haastert-Talini K. Perspective on Schwann Cells Derived from Induced Pluripotent Stem Cells in Peripheral Nerve Tissue Engineering. Cells 2020; 9:E2497. [PMID: 33213068 PMCID: PMC7698557 DOI: 10.3390/cells9112497] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 11/13/2020] [Accepted: 11/16/2020] [Indexed: 02/06/2023] Open
Abstract
Schwann cells play a crucial role in successful peripheral nerve repair and regeneration by supporting both axonal growth and myelination. Schwann cells are therefore a feasible option for cell therapy treatment of peripheral nerve injury. However, sourcing human Schwann cells at quantities required for development beyond research is challenging. Due to their availability, rapid in vitro expansion, survival, and integration within the host tissue, stem cells have attracted considerable attention as candidate cell therapies. Among them, induced pluripotent stem cells (iPSCs) with the associated prospects for personalized treatment are a promising therapy to take the leap from bench to bedside. In this critical review, we firstly focus on the current knowledge of the Schwann cell phenotype in regard to peripheral nerve injury, including crosstalk with the immune system during peripheral nerve regeneration. Then, we review iPSC to Schwann cell derivation protocols and the results from recent in vitro and in vivo studies. We finally conclude with some prospects for the use of iPSCs in clinical settings.
Collapse
Affiliation(s)
- Zhong Huang
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, 30623 Hannover, Germany;
- Center for Systems Neuroscience (ZSN) Hannover, 30559 Hannover, Germany
| | - Rebecca Powell
- Department of Pharmacology, UCL School of Pharmacy, 29-39 Brunswick Square, London WC1N 1AX, UK;
- UCL Centre for Nerve Engineering, University College London, London WC1E 6BT, UK
| | - James B. Phillips
- Department of Pharmacology, UCL School of Pharmacy, 29-39 Brunswick Square, London WC1N 1AX, UK;
- UCL Centre for Nerve Engineering, University College London, London WC1E 6BT, UK
| | - Kirsten Haastert-Talini
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, 30623 Hannover, Germany;
- Center for Systems Neuroscience (ZSN) Hannover, 30559 Hannover, Germany
| |
Collapse
|
38
|
Rodriguez-Fontan F, Reeves B, Tuaño K, Colakoglu S, D' Agostino L, Banegas R. Tobacco use and neurogenesis: A theoretical review of pathophysiological mechanism affecting the outcome of peripheral nerve regeneration. J Orthop 2020; 22:59-63. [PMID: 32280170 PMCID: PMC7138932 DOI: 10.1016/j.jor.2020.03.026] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 03/22/2020] [Indexed: 02/07/2023] Open
Abstract
Peripheral nerve injury often requires medical intervention. Unfortunately, many patients never have a full recovery, despite a multi-disciplinary approach, including operative intervention and physical and/or occupational therapy. Outcomes are multifactorial, but are largely affected by the original injury severity, and patient comorbidities. A lcoholism, diabetes mellitus and ageing may detrimentally affect the outcomes of nerve injury; however little is known about tobacco's potential impact on nerve regeneration. Tobacco has known immunomodulatory effects, which suggests that it might affect peripheral nerve regeneration and functional recovery following injury. This review characterizes the effects of tobacco use on the complex cellular and chemokine interactions in peripheral nerve regeneration.
Collapse
Affiliation(s)
- Francisco Rodriguez-Fontan
- Department of Orthopedics, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
- Universidad de Buenos Aires, Facultad de Medicina, Buenos Aires, Argentina
| | - Bradley Reeves
- University of Colorado, School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Krystle Tuaño
- Division of Plastic and Reconstructive Surgery, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Salih Colakoglu
- Division of Plastic and Reconstructive Surgery, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | | | | |
Collapse
|
39
|
Stratton JA, Eaton S, Rosin NL, Jawad S, Holmes A, Yoon G, Midha R, Biernaskie J. Macrophages and Associated Ligands in the Aged Injured Nerve: A Defective Dynamic That Contributes to Reduced Axonal Regrowth. Front Aging Neurosci 2020; 12:174. [PMID: 32595489 PMCID: PMC7304384 DOI: 10.3389/fnagi.2020.00174] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 05/19/2020] [Indexed: 01/08/2023] Open
Abstract
The regenerative capacity of injured peripheral nerves is diminished with aging. To identify factors that contribute to this impairment, we compared the immune cell response in young vs. aged animals following nerve injury. First, we confirmed that macrophage accumulation is delayed in aged injured nerves which is due to defects in monocyte migration as a result of defects in site-specific recruitment signals in the aged nerve. Interestingly, impairment in both macrophage accumulation and functional recovery could be overcome by transplanting bone marrow from aged animals into young mice. That is, upon exposure to a youthful environment, monocytes/macrophages originating from the aged bone marrow behaved similarly to young cells. Transcriptional profiling of aged macrophages following nerve injury revealed that both pro- and anti-inflammatory genes were largely downregulated in aged compared to young macrophages. One ligand of particular interest was macrophage-associated secreted protein (MCP1), which exhibited a potent role in regulating aged axonal regrowth in vitro. Given that macrophage-derived MCP1 is significantly diminished in the aged injured nerve, our data suggest that age-associated defects in MCP1 signaling could contribute to the regenerative deficits that occur in the aged nervous system.
Collapse
Affiliation(s)
- Jo Anne Stratton
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada.,Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada.,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Shane Eaton
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Nicole L Rosin
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Sana Jawad
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Alexandra Holmes
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Grace Yoon
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Rajiv Midha
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada.,Department of Clinical Neuroscience, Faculty of Medicine, University of Calgary, Calgary, AB, Canada
| | - Jeff Biernaskie
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada.,Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada.,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
40
|
Sepsis Activates the TLR4/MyD88 Pathway in Schwann Cells to Promote Infiltration of Macrophages, Thereby Impeding Neuromuscular Function. Shock 2020; 55:90-99. [PMID: 32433207 DOI: 10.1097/shk.0000000000001557] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
INTRODUCTION Sepsis is a kind of maladjustment response to bacterial infection and activation of coagulation, which can induce neuromuscular dysfunction. However, there is scarce of experimental evidence about the relationship between Schwann cells (SCs) and sepsis in neuromuscular dysfunction. We therefore set out to identify the potential role of SCs in sepsis-induced neuromuscular dysfunction and to explore the underlying molecular mechanism. METHODS Primary SCs were isolated from the left hind limb sciatic nerve of sepsis mice, which was constructed by cecal ligation and puncture. Then, the SCs were infected with adenovirus encoding toll-like receptor 4 (TLR4), MyD88, or IL-1R (with lipopolysaccharide stimulation), and the Raw 264.7 macrophages were injected with adenovirus with CCR2 silencing (with mMCP-1 stimulation). Further investigation of the interleukin 1 beta (IL-1β) and macrophage cationic peptide 1 (MCP-1) expressions, we followed reverse transcription-quantitative polymerase chain reaction and enzyme-linked immunosorbent assay techniques, the F4/80 and Ki67 expressions was observed by immunofluorescence staining, while the expressions of CCR2, FAK/p-FAK, nuclear factor-κB (NFκB)/p-NFκB, and ERK1/2/p-ERK1/2 were determined by Western blot analysis. Last, but not the least, the cell migration ability and cell proliferation ability were detected by Transwell assay and Flow cytometry respectively. RESULTS Our results showed that in sepsis mice, the TLR4/MyD88/ERK pathway was activated in SCs, which triggered the cells to secrete IL-1β and MCP-1. The secreted IL-1β bound with IL-1β receptor on the surface of SCs, thereby activating the IL-1β/IL-1R/MyD88/ERK pathway and further promoting the secretion of MCP-1 by SCs. MCP-1 was found to bind to CCR2 on the surface of Raw264.7 macrophages to activate the TLR4/MyD88/ERK pathway which caused the inhibition of neuromuscular function. CONCLUSION Sepsis significantly promotes the infiltration of macrophages by activating the TLR4/MyD88 pathway in SCs, thereby impeding neuromuscular function. Consistently, our study provides a novel concept in the area of neuromuscular dysfunction therapeutics following sepsis.
Collapse
|
41
|
Pandya VA, Patani R. Decoding the relationship between ageing and amyotrophic lateral sclerosis: a cellular perspective. Brain 2020; 143:1057-1072. [PMID: 31851317 PMCID: PMC7174045 DOI: 10.1093/brain/awz360] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 09/13/2019] [Accepted: 09/21/2019] [Indexed: 12/13/2022] Open
Abstract
With an ageing population comes an inevitable increase in the prevalence of age-associated neurodegenerative diseases, such as amyotrophic lateral sclerosis (ALS), a relentlessly progressive and universally fatal disease characterized by the degeneration of upper and lower motor neurons within the brain and spinal cord. Indeed, the physiological process of ageing causes a variety of molecular and cellular phenotypes. With dysfunction at the neuromuscular junction implicated as a key pathological mechanism in ALS, and each lower motor unit cell type vulnerable to its own set of age-related phenotypes, the effects of ageing might in fact prove a prerequisite to ALS, rendering the cells susceptible to disease-specific mechanisms. Moreover, we discuss evidence for overlap between age and ALS-associated hallmarks, potentially implicating cell type-specific ageing as a key contributor to this multifactorial and complex disease. With a dearth of disease-modifying therapy currently available for ALS patients and a substantial failure in bench to bedside translation of other potential therapies, the unification of research in ageing and ALS requires high fidelity models to better recapitulate age-related human disease and will ultimately yield more reliable candidate therapeutics for patients, with the aim of enhancing healthspan and life expectancy.
Collapse
Affiliation(s)
- Virenkumar A Pandya
- Department of Neuromuscular Diseases, University College London Queen Square Institute of Neurology, Queen Square, London, UK
- The Francis Crick Institute, London, UK
| | - Rickie Patani
- Department of Neuromuscular Diseases, University College London Queen Square Institute of Neurology, Queen Square, London, UK
- The Francis Crick Institute, London, UK
| |
Collapse
|
42
|
Preconditioning of Rat Bone Marrow-Derived Mesenchymal Stromal Cells with Toll-Like Receptor Agonists. Stem Cells Int 2019; 2019:7692973. [PMID: 31531025 PMCID: PMC6721436 DOI: 10.1155/2019/7692973] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 07/02/2019] [Indexed: 12/29/2022] Open
Abstract
Bone marrow-derived mesenchymal stromal cells (BM-MSCs) are dynamic cells that can sense the environment, adapting their regulatory functions to different conditions. Accordingly, the therapeutic potential of BM-MSCs can be modulated by preconditioning strategies aimed at modifying their paracrine action. Although rat BM-MSCs (rBM-MSCs) have been widely tested in preclinical research, most preconditioning studies have employed human and mouse BM-MSCs. Herein, we investigated whether rBM-MSCs modify their phenotype and paracrine functions in response to Toll-like receptor (TLR) agonists. The data showed that rBM-MSCs expressed TLR3, TLR4, and MDA5 mRNA and were able to internalize polyinosinic-polycytidylic acid (Poly(I:C)), a TLR3/MDA5 agonist. rBM-MSCs were then stimulated with Poly(I:C) or with lipopolysaccharide (LPS, a TLR4 agonist) for 1 h and were grown under normal culture conditions. LPS or Poly(I:C) stimulation did not affect the viability or the morphology of rBM-MSCs and did not modify the expression pattern of key cell surface markers. Poly(I:C) did not induce statistically significant changes in the release of several inflammatory mediators and VEGF by rBM-MSCs, although it tended to increase IL-6 and MCP-1 secretion, whereas LPS increased the release of IL-6, MCP-1, and VEGF, three factors that were constitutively secreted by unstimulated cells. The neurotrophic activity of the conditioned medium from unstimulated and LPS-preconditioned rBM-MSCs was investigated using dorsal root ganglion explants, showing that soluble factors produced by unstimulated and LPS-preconditioned rBM-MSCs can stimulate neurite outgrowth similarly, in a VEGF-dependent manner. LPS-preconditioned cells, however, were slightly more efficient in increasing the number of regrowing axons in a model of sciatic nerve transection in rats. In conclusion, LPS preconditioning boosted the production of constitutively secreted factors by rBM-MSCs, without changing their mesenchymal identity, an effect that requires further investigation in exploratory preclinical studies.
Collapse
|
43
|
Jha MK, Lee Y, Russell KA, Yang F, Dastgheyb RM, Deme P, Ament XH, Chen W, Liu Y, Guan Y, Polydefkis MJ, Hoke A, Haughey NJ, Rothstein JD, Morrison BM. Monocarboxylate transporter 1 in Schwann cells contributes to maintenance of sensory nerve myelination during aging. Glia 2019; 68:161-177. [PMID: 31453649 DOI: 10.1002/glia.23710] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 07/18/2019] [Accepted: 08/01/2019] [Indexed: 12/15/2022]
Abstract
Schwann cell (SC)-specific monocarboxylate transporter 1 (MCT1) knockout mice were generated by mating MCT1 f/f mice with myelin protein zero (P0)-Cre mice. P0-Cre+/- , MCT1 f/f mice have no detectable early developmental defects, but develop hypomyelination and reduced conduction velocity in sensory, but not motor, peripheral nerves during maturation and aging. Furthermore, reduced mechanical sensitivity is evident in aged P0-Cre+/- , MCT1 f/f mice. MCT1 deletion in SCs impairs both their glycolytic and mitochondrial functions, leading to altered lipid metabolism of triacylglycerides, diacylglycerides, and sphingomyelin, decreased expression of myelin-associated glycoprotein, and increased expression of c-Jun and p75-neurotrophin receptor, suggesting a regression of SCs to a less mature developmental state. Taken together, our results define the contribution of SC MCT1 to both SC metabolism and peripheral nerve maturation and aging.
Collapse
Affiliation(s)
- Mithilesh Kumar Jha
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Youngjin Lee
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Biomedical Sciences, City University of Hong Kong, Hong Kong SAR, Hong Kong
| | - Katelyn A Russell
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Fang Yang
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Raha M Dastgheyb
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Pragney Deme
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Xanthe H Ament
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Weiran Chen
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ying Liu
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Yun Guan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Neurological Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Michael J Polydefkis
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ahmet Hoke
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Norman J Haughey
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jeffrey D Rothstein
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Brain Science Institute, Johns Hopkins University, Baltimore, Maryland
| | - Brett M Morrison
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
44
|
Mulroy E, Pelosi L. Carpal tunnel syndrome in advanced age: A sonographic and electrodiagnostic study. Muscle Nerve 2019; 60:236-241. [DOI: 10.1002/mus.26496] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/21/2019] [Indexed: 12/25/2022]
Affiliation(s)
- Eoin Mulroy
- Department of Neurology and Clinical NeurophysiologyAuckland District Health Board 2 Park Road, Grafton, Auckland 1023 New Zealand
| | - Luciana Pelosi
- Bay of Plenty District Health Board Tauranga New Zealand
| |
Collapse
|
45
|
Büttner R, Schulz A, Reuter M, Akula AK, Mindos T, Carlstedt A, Riecken LB, Baader SL, Bauer R, Morrison H. Inflammaging impairs peripheral nerve maintenance and regeneration. Aging Cell 2018; 17:e12833. [PMID: 30168637 PMCID: PMC6260910 DOI: 10.1111/acel.12833] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 06/14/2018] [Accepted: 07/21/2018] [Indexed: 12/13/2022] Open
Abstract
The regenerative capacity of peripheral nerves declines during aging, contributing to the development of neuropathies, limiting organism function. Changes in Schwann cells prompt failures in instructing maintenance and regeneration of aging nerves; molecular mechanisms of which have yet to be delineated. Here, we identified an altered inflammatory environment leading to a defective Schwann cell response, as an underlying mechanism of impaired nerve regeneration during aging. Chronic inflammation was detected in intact uninjured old nerves, characterized by increased macrophage infiltration and raised levels of monocyte chemoattractant protein 1 (MCP1) and CC chemokine ligand 11 (CCL11). Schwann cells in the old nerves appeared partially dedifferentiated, accompanied by an activated repair program independent of injury. Upon sciatic nerve injury, an initial delayed immune response was followed by a persistent hyperinflammatory state accompanied by a diminished repair process. As a contributing factor to nerve aging, we showed that CCL11 interfered with Schwann cell differentiation in vitro and in vivo. Our results indicate that increased infiltration of macrophages and inflammatory signals diminish regenerative capacity of aging nerves by altering Schwann cell behavior. The study identifies CCL11 as a promising target for anti‐inflammatory therapies aiming to improve nerve regeneration in old age.
Collapse
Affiliation(s)
- Robert Büttner
- Leibniz Institute on Aging; Fritz Lipmann Institute; Jena Germany
| | - Alexander Schulz
- Leibniz Institute on Aging; Fritz Lipmann Institute; Jena Germany
- Department of Genetics and Program in Cellular Neuroscience, Neurodegeneration and Repair; Yale University School of Medicine; New Haven Connecticut
| | - Michael Reuter
- Leibniz Institute on Aging; Fritz Lipmann Institute; Jena Germany
| | - Asha K. Akula
- Leibniz Institute on Aging; Fritz Lipmann Institute; Jena Germany
| | - Thomas Mindos
- Leibniz Institute on Aging; Fritz Lipmann Institute; Jena Germany
| | | | - Lars B. Riecken
- Leibniz Institute on Aging; Fritz Lipmann Institute; Jena Germany
| | | | - Reinhard Bauer
- Institute of Molecular Cell Biology; Jena University Hospital; Jena Germany
| | - Helen Morrison
- Leibniz Institute on Aging; Fritz Lipmann Institute; Jena Germany
| |
Collapse
|
46
|
Qing L, Chen H, Tang J, Jia X. Exosomes and Their MicroRNA Cargo: New Players in Peripheral Nerve Regeneration. Neurorehabil Neural Repair 2018; 32:765-776. [PMID: 30223738 PMCID: PMC6146407 DOI: 10.1177/1545968318798955] [Citation(s) in RCA: 120] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Peripheral nerve injury is a major clinical problem and often results in a poor functional recovery. Despite obvious clinical need, treatment strategies have been largely suboptimal. In the nervous system, exosomes, which are nanosized extracellular vesicles, play a critical role in mediating intercellular communication. More specifically, microRNA carried by exosomes are involved in various key processes such as nerve and vascular regeneration, and exosomes originating from Schwann cells, macrophages, and mesenchymal stem cells can promote peripheral nerve regeneration. In this review, the current knowledge of exosomes' and their miRNA cargo's role in peripheral nerve regeneration are summarized. The possible future roles of exosomes in therapy and the potential for microRNA-containing exosomes to treat peripheral nerve injuries are also discussed.
Collapse
Affiliation(s)
- Liming Qing
- Department of Hand & Microsurgery, Xiangya Hospital of
Central South University, Changsha, Hunan, 410008
- Department of Neurosurgery, University of Maryland School of
Medicine, Baltimore, MD 21201, USA
| | - Huanwen Chen
- Department of Neurosurgery, University of Maryland School of
Medicine, Baltimore, MD 21201, USA
| | - Juyu Tang
- Department of Hand & Microsurgery, Xiangya Hospital of
Central South University, Changsha, Hunan, 410008
| | - Xiaofeng Jia
- Department of Neurosurgery, University of Maryland School of
Medicine, Baltimore, MD 21201, USA
- Department of Orthopedics, University of Maryland School of
Medicine, Baltimore, MD 21201, USA
- Department of Anatomy and Neurobiology, University of Maryland
School of Medicine, Baltimore, MD 21201, USA
- Department of Biomedical Engineering, Johns Hopkins University
School of Medicine, Baltimore, MD 21205, USA
- Department of Anesthesiology and Critical Care Medicine, Johns
Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
47
|
Fledrich R, Abdelaal T, Rasch L, Bansal V, Schütza V, Brügger B, Lüchtenborg C, Prukop T, Stenzel J, Rahman RU, Hermes D, Ewers D, Möbius W, Ruhwedel T, Katona I, Weis J, Klein D, Martini R, Brück W, Müller WC, Bonn S, Bechmann I, Nave KA, Stassart RM, Sereda MW. Targeting myelin lipid metabolism as a potential therapeutic strategy in a model of CMT1A neuropathy. Nat Commun 2018; 9:3025. [PMID: 30072689 PMCID: PMC6072747 DOI: 10.1038/s41467-018-05420-0] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 06/28/2018] [Indexed: 01/17/2023] Open
Abstract
In patients with Charcot-Marie-Tooth disease 1A (CMT1A), peripheral nerves display aberrant myelination during postnatal development, followed by slowly progressive demyelination and axonal loss during adult life. Here, we show that myelinating Schwann cells in a rat model of CMT1A exhibit a developmental defect that includes reduced transcription of genes required for myelin lipid biosynthesis. Consequently, lipid incorporation into myelin is reduced, leading to an overall distorted stoichiometry of myelin proteins and lipids with ultrastructural changes of the myelin sheath. Substitution of phosphatidylcholine and phosphatidylethanolamine in the diet is sufficient to overcome the myelination deficit of affected Schwann cells in vivo. This treatment rescues the number of myelinated axons in the peripheral nerves of the CMT rats and leads to a marked amelioration of neuropathic symptoms. We propose that lipid supplementation is an easily translatable potential therapeutic approach in CMT1A and possibly other dysmyelinating neuropathies.
Collapse
Affiliation(s)
- R Fledrich
- Department of Neurogenetics, Max-Planck-Institute of Experimental Medicine, Göttingen, 37075, Germany.
- Institute of Anatomy, University of Leipzig, Leipzig, 04103, Germany.
- Department of Neuropathology, University Hospital Leipzig, Leipzig, 04103, Germany.
| | - T Abdelaal
- Department of Neurogenetics, Max-Planck-Institute of Experimental Medicine, Göttingen, 37075, Germany
- Department of Clinical Neurophysiology, University Medical Center Göttingen, Göttingen, 37075, Germany
- Chemistry of Natural and Microbial Products Department, Pharmaceutical and Drug Industries Division, National Research Centre, Giza, 12622, Egypt
| | - L Rasch
- Department of Neurogenetics, Max-Planck-Institute of Experimental Medicine, Göttingen, 37075, Germany
- Department of Clinical Neurophysiology, University Medical Center Göttingen, Göttingen, 37075, Germany
| | - V Bansal
- Center for Molecular Neurobiology, Institute of Medical Systems Biology, University Medical Center Hamburg-Eppendorf, Hamburg, 20251, Germany
| | - V Schütza
- Department of Neurogenetics, Max-Planck-Institute of Experimental Medicine, Göttingen, 37075, Germany
- Department of Neuropathology, University Hospital Leipzig, Leipzig, 04103, Germany
| | - B Brügger
- Heidelberg University Biochemistry Center (BZH), Heidelberg, 69120, Germany
| | - C Lüchtenborg
- Heidelberg University Biochemistry Center (BZH), Heidelberg, 69120, Germany
| | - T Prukop
- Department of Neurogenetics, Max-Planck-Institute of Experimental Medicine, Göttingen, 37075, Germany
- Department of Clinical Neurophysiology, University Medical Center Göttingen, Göttingen, 37075, Germany
- Institute of Clinical Pharmacology, University Medical Center Göttingen, Göttingen, 37075, Germany
| | - J Stenzel
- Department of Neurogenetics, Max-Planck-Institute of Experimental Medicine, Göttingen, 37075, Germany
- Department of Clinical Neurophysiology, University Medical Center Göttingen, Göttingen, 37075, Germany
| | - R U Rahman
- Center for Molecular Neurobiology, Institute of Medical Systems Biology, University Medical Center Hamburg-Eppendorf, Hamburg, 20251, Germany
| | - D Hermes
- Department of Neurogenetics, Max-Planck-Institute of Experimental Medicine, Göttingen, 37075, Germany
- Department of Clinical Neurophysiology, University Medical Center Göttingen, Göttingen, 37075, Germany
| | - D Ewers
- Department of Neurogenetics, Max-Planck-Institute of Experimental Medicine, Göttingen, 37075, Germany
- Department of Clinical Neurophysiology, University Medical Center Göttingen, Göttingen, 37075, Germany
| | - W Möbius
- Department of Neurogenetics, Max-Planck-Institute of Experimental Medicine, Göttingen, 37075, Germany
- Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, 37075, Germany
| | - T Ruhwedel
- Department of Neurogenetics, Max-Planck-Institute of Experimental Medicine, Göttingen, 37075, Germany
| | - I Katona
- Institute of Neuropathology, University Hospital Aachen, Aachen, 52074, Germany
| | - J Weis
- Institute of Neuropathology, University Hospital Aachen, Aachen, 52074, Germany
| | - D Klein
- Department of Neurology, Section of Developmental Neurobiology, University Hospital Wuerzburg, Wuerzburg, 97080, Germany
| | - R Martini
- Department of Neurology, Section of Developmental Neurobiology, University Hospital Wuerzburg, Wuerzburg, 97080, Germany
| | - W Brück
- Institute of Neuropathology, University Medical Center Göttingen, Göttingen, 37075, Germany
| | - W C Müller
- Department of Neuropathology, University Hospital Leipzig, Leipzig, 04103, Germany
| | - S Bonn
- Center for Molecular Neurobiology, Institute of Medical Systems Biology, University Medical Center Hamburg-Eppendorf, Hamburg, 20251, Germany
- German Center for Neurodegenerative Diseases, Tübingen, 72076, Germany
| | - I Bechmann
- Institute of Anatomy, University of Leipzig, Leipzig, 04103, Germany
| | - K A Nave
- Department of Neurogenetics, Max-Planck-Institute of Experimental Medicine, Göttingen, 37075, Germany.
| | - R M Stassart
- Department of Neurogenetics, Max-Planck-Institute of Experimental Medicine, Göttingen, 37075, Germany.
- Department of Neuropathology, University Hospital Leipzig, Leipzig, 04103, Germany.
- Institute of Neuropathology, University Medical Center Göttingen, Göttingen, 37075, Germany.
| | - M W Sereda
- Department of Neurogenetics, Max-Planck-Institute of Experimental Medicine, Göttingen, 37075, Germany.
- Department of Clinical Neurophysiology, University Medical Center Göttingen, Göttingen, 37075, Germany.
| |
Collapse
|
48
|
Xiao Y, Dong J, Yin Z, Wu Q, Zhou Y, Zhou X. Procyanidin B2 protects against d-galactose-induced mimetic aging in mice: Metabolites and microbiome analysis. Food Chem Toxicol 2018; 119:141-149. [PMID: 29751077 DOI: 10.1016/j.fct.2018.05.017] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 04/16/2018] [Accepted: 05/07/2018] [Indexed: 12/21/2022]
Abstract
To elucidate the possible mechanisms for the preventive effect of procyanidin B2 on aging, a combined analysis of metabolic profile and gut microbiome was carried out in the present study. The mimetic aged mice induced by d-galactose injection (500 mg/kg, sc daily), and the preventive group was fed with the diet plus 0.2% procyanidin B2. After 7 weeks of treatment, the spatial memory was assayed using the Morris water maze test. Procyanidin B2 significantly ameliorated the impaired memory and antioxidant abilities induced by d-galactose. Furthermore, metabolomics analysis of plasma based on LC/Q-TOF-MS demonstrated that phosphatidyl cholines, oleic acid, linoleic acid, carnitine, pantothenic acid, and taurocholic acid were significantly increased in the mice treated with procyanidin B2, and pyruvic acid, hydroxybutyric acid, hippuric acid, and cholic acid were decreased significantly. Together, gut microbiome analysis using Illumina sequencing showed that there were significant differences in the Firmicutes/Bacteroidetes ratio and abundance of Roseburia, Lachnospiraceae, and Bifidobacterium between the aging and supplemental procyanidin B2 groups. In summary, procyanidin B2 possessed potential prevention of the cognitive and oxidative impairment via the metabolic pathway regulation related to citrate cycle, fatty acid, and bile acid in the aged mice, accompanied by remodeling the gut flora.
Collapse
Affiliation(s)
- Ying Xiao
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, Shanghai, 201418, China.
| | - Jialin Dong
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, Shanghai, 201418, China
| | - Zhiting Yin
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, Shanghai, 201418, China
| | - Qiguo Wu
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, Shanghai, 201418, China
| | - Yiming Zhou
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, Shanghai, 201418, China
| | - Xiaoli Zhou
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, Shanghai, 201418, China.
| |
Collapse
|
49
|
Gonçalves NP, Vægter CB, Pallesen LT. Peripheral Glial Cells in the Development of Diabetic Neuropathy. Front Neurol 2018; 9:268. [PMID: 29770116 PMCID: PMC5940740 DOI: 10.3389/fneur.2018.00268] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Accepted: 04/06/2018] [Indexed: 12/15/2022] Open
Abstract
The global prevalence of diabetes is rapidly increasing, affecting more than half a billion individuals within the next few years. As diabetes negatively affects several physiological systems, this dramatic increase represents not only impaired quality of life on the individual level but also a huge socioeconomic challenge. One of the physiological consequences affecting up to half of diabetic patients is the progressive deterioration of the peripheral nervous system, resulting in spontaneous pain and eventually loss of sensory function, motor weakness, and organ dysfunctions. Despite intense research on the consequences of hyperglycemia on nerve functions, the biological mechanisms underlying diabetic neuropathy are still largely unknown, and treatment options lacking. Research has mainly focused directly on the neuronal component, presumably from the perspective that this is the functional signal-transmitting unit of the nerve. However, it is noteworthy that each single peripheral sensory neuron is intimately associated with numerous glial cells; the neuronal soma is completely enclosed by satellite glial cells and the length of the longest axons covered by at least 1,000 Schwann cells. The glial cells are vital for the neuron, but very little is still known about these cells in general and especially how they respond to diabetes in terms of altered neuronal support. We will discuss current knowledge of peripheral glial cells and argue that increased research in these cells is imperative for a better understanding of the mechanisms underlying diabetic neuropathy.
Collapse
Affiliation(s)
- Nádia Pereira Gonçalves
- Department of Biomedicine, Nordic-EMBL Partnership for Molecular Medicine, Danish Research Institute of Translational Neuroscience (DANDRITE), Aarhus University, Aarhus, Denmark.,The International Diabetic Neuropathy Consortium (IDNC), Aarhus University, Aarhus, Denmark
| | - Christian Bjerggaard Vægter
- Department of Biomedicine, Nordic-EMBL Partnership for Molecular Medicine, Danish Research Institute of Translational Neuroscience (DANDRITE), Aarhus University, Aarhus, Denmark.,The International Diabetic Neuropathy Consortium (IDNC), Aarhus University, Aarhus, Denmark
| | - Lone Tjener Pallesen
- Department of Biomedicine, Nordic-EMBL Partnership for Molecular Medicine, Danish Research Institute of Translational Neuroscience (DANDRITE), Aarhus University, Aarhus, Denmark
| |
Collapse
|
50
|
Macrophage Depletion Ameliorates Peripheral Neuropathy in Aging Mice. J Neurosci 2018; 38:4610-4620. [PMID: 29712789 DOI: 10.1523/jneurosci.3030-17.2018] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 02/06/2018] [Accepted: 03/09/2018] [Indexed: 01/02/2023] Open
Abstract
Aging is known as a major risk factor for the structure and function of the nervous system. There is urgent need to overcome such deleterious effects of age-related neurodegeneration. Here we show that peripheral nerves of 24-month-old aging C57BL/6 mice of either sex show similar pathological alterations as nerves from aging human individuals, whereas 12-month-old adult mice lack such alterations. Specifically, nerve fibers showed demyelination, remyelination and axonal lesion. Moreover, in the aging mice, neuromuscular junctions showed features typical for dying-back neuropathies, as revealed by a decline of presynaptic markers, associated with α-bungarotoxin-positive postsynapses. In line with these observations were reduced muscle strengths. These alterations were accompanied by elevated numbers of endoneurial macrophages, partially comprising the features of phagocytosing macrophages. Comparable profiles of macrophages could be identified in peripheral nerve biopsies of aging persons. To determine the pathological impact of macrophages in aging mice, we selectively targeted the cells by applying an orally administered CSF-1R specific kinase (c-FMS) inhibitor. The 6-month-lasting treatment started before development of degenerative changes at 18 months and reduced macrophage numbers in mice by ∼70%, without side effects. Strikingly, nerve structure was ameliorated and muscle strength preserved. We show, for the first time, that age-related degenerative changes in peripheral nerves are driven by macrophages. These findings may pave the way for treating degeneration in the aging peripheral nervous system by targeting macrophages, leading to reduced weakness, improved mobility, and eventually increased quality of life in the elderly.SIGNIFICANCE STATEMENT Aging is a major risk factor for the structure and function of the nervous system. Here we show that peripheral nerves of 24-month-old aging mice show similar degenerative alterations as nerves from aging human individuals. Both in mice and humans, these alterations were accompanied by endoneurial macrophages. To determine the pathological impact of macrophages in aging mice, we selectively targeted the cells by blocking a cytokine receptor, essential for macrophage survival. The treatment strongly reduced macrophage numbers and substantially improved nerve structure and muscle strength. We show, for the first time, that age-related degenerative changes in peripheral nerves are driven by macrophages. These findings may be helpful for treatment weakness and reduced mobility in the elderly.
Collapse
|