1
|
Hintermayer MA, Juźwik CA, Morquette B, Hua E, Zhang J, Drake S, Shi SS, Rambaldi I, Vangoor V, Pasterkamp J, Moore C, Fournier AE. A miR-383-5p Signaling Hub Coordinates the Axon Regeneration Response to Inflammation. J Neurosci 2024; 44:e1822232024. [PMID: 39266301 PMCID: PMC11529811 DOI: 10.1523/jneurosci.1822-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 08/07/2024] [Accepted: 09/05/2024] [Indexed: 09/14/2024] Open
Abstract
Neuroinflammation can positively influence axon regeneration following injury in the central nervous system. Inflammation promotes the release of neurotrophic molecules and stimulates intrinsic proregenerative molecular machinery in neurons, but the detailed mechanisms driving this effect are not fully understood. We evaluated how microRNAs are regulated in retinal neurons in response to intraocular inflammation to identify their potential role in axon regeneration. We found that miR-383-5p is downregulated in retinal ganglion cells in response to zymosan-induced intraocular inflammation. MiR-383-5p downregulation in neurons is sufficient to promote axon growth in vitro, and the intravitreal injection of a miR-383-5p inhibitor into the eye promotes axon regeneration following optic nerve crush. MiR-383-5p directly targets ciliary neurotrophic factor (CNTF) receptor components, and miR-383-5p inhibition sensitizes adult retinal neurons to the outgrowth-promoting effects of CNTF. Interestingly, we also demonstrate that CNTF treatment is sufficient to reduce miR-383-5p levels in neurons, constituting a positive-feedback module, whereby initial CNTF treatment reduces miR-383-5p levels, which then disinhibits CNTF receptor components to sensitize neurons to the ligand. Additionally, miR-383-5p inhibition derepresses the mitochondrial antioxidant protein peroxiredoxin-3 (PRDX3) which was required for the proregenerative effects associated with miR-383-5p loss-of-function in vitro. We have thus identified a positive-feedback mechanism that facilitates neuronal CNTF sensitivity in neurons and a new molecular signaling module that promotes inflammation-induced axon regeneration.
Collapse
Affiliation(s)
- Matthew A Hintermayer
- Montréal Neurological Institute, McGill University, Montréal, Quebec H3A 2B4, Canada
| | - Camille A Juźwik
- Montréal Neurological Institute, McGill University, Montréal, Quebec H3A 2B4, Canada
| | - Barbara Morquette
- Montréal Neurological Institute, McGill University, Montréal, Quebec H3A 2B4, Canada
| | - Elizabeth Hua
- Montréal Neurological Institute, McGill University, Montréal, Quebec H3A 2B4, Canada
| | - Julia Zhang
- Montréal Neurological Institute, McGill University, Montréal, Quebec H3A 2B4, Canada
| | - Sienna Drake
- Montréal Neurological Institute, McGill University, Montréal, Quebec H3A 2B4, Canada
| | - Shan Shan Shi
- Montréal Neurological Institute, McGill University, Montréal, Quebec H3A 2B4, Canada
| | - Isabel Rambaldi
- Montréal Neurological Institute, McGill University, Montréal, Quebec H3A 2B4, Canada
| | - Vamshi Vangoor
- Department of Translation Neuroscience, University Medical Center Brain Center, Utrecht University, Utrecht 3584 CG, Netherlands
| | - Jeroen Pasterkamp
- Department of Translation Neuroscience, University Medical Center Brain Center, Utrecht University, Utrecht 3584 CG, Netherlands
| | - Craig Moore
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL A1B 3V6, Canada
| | - Alyson E Fournier
- Montréal Neurological Institute, McGill University, Montréal, Quebec H3A 2B4, Canada
| |
Collapse
|
2
|
Yilmaz SN, Steiner K, Marksteiner J, Faserl K, Villunger M, Sarg B, Humpel C. From Organotypic Mouse Brain Slices to Human Alzheimer's Plasma Biomarkers: A Focus on Nerve Fiber Outgrowth. Biomolecules 2024; 14:1326. [PMID: 39456259 PMCID: PMC11506054 DOI: 10.3390/biom14101326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 10/08/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease characterized by memory loss and progressive deterioration of cognitive functions. Being able to identify reliable biomarkers in easily available body fluids such as blood plasma is vital for the disease. To achieve this, we used a technique that applied human plasma to organotypic brain slice culture via microcontact printing. After a 2-week culture period, we performed immunolabeling for neurofilament and myelin oligodendrocyte glycoprotein (MOG) to visualize newly formed nerve fibers and oligodendrocytes. There was no significant change in the number of new nerve fibers in the AD plasma group compared to the healthy control group, while the length of the produced fibers significantly decreased. A significant increase in the number of MOG+ dots around these new fibers was detected in the patient group. According to our hypothesis, there are factors in the plasma of AD patients that affect the growth of new nerve fibers, which also affect the oligodendrocytes. Based on these findings, we selected the most promising plasma samples and conducted mass spectrometry using a differential approach and we identified three putative biomarkers: aldehyde-dehydrogenase 1A1, alpha-synuclein and protein S100-A4. Our method represents a novel and innovative approach for translating research findings from mouse models to human applications.
Collapse
Affiliation(s)
- Sakir Necat Yilmaz
- Laboratory of Psychiatry and Experimental Alzheimer’s Research, Medical University of Innsbruck, 6020 Innsbruck, Austria; (S.N.Y.); (K.S.)
- Department of Histology and Embryology, Faculty of Medicine, Mersin University, Mersin 33130, Turkey
| | - Katharina Steiner
- Laboratory of Psychiatry and Experimental Alzheimer’s Research, Medical University of Innsbruck, 6020 Innsbruck, Austria; (S.N.Y.); (K.S.)
| | - Josef Marksteiner
- Department of Psychiatry and Psychotherapy A, Hall State Hospital, 6060 Hall in Tirol, Austria;
| | - Klaus Faserl
- Protein Core Facility, Institute of Medical Biochemistry, CCB-Biocenter, Medical University of Innsbruck, 6020 Innsbruck, Austria; (K.F.); (M.V.); (B.S.)
| | - Mathias Villunger
- Protein Core Facility, Institute of Medical Biochemistry, CCB-Biocenter, Medical University of Innsbruck, 6020 Innsbruck, Austria; (K.F.); (M.V.); (B.S.)
| | - Bettina Sarg
- Protein Core Facility, Institute of Medical Biochemistry, CCB-Biocenter, Medical University of Innsbruck, 6020 Innsbruck, Austria; (K.F.); (M.V.); (B.S.)
| | - Christian Humpel
- Laboratory of Psychiatry and Experimental Alzheimer’s Research, Medical University of Innsbruck, 6020 Innsbruck, Austria; (S.N.Y.); (K.S.)
| |
Collapse
|
3
|
Hazer Rosberg DB, Stenberg L, Mahlapuu M, Dahlin LB. PXL01 alters macrophage response with no effect on axonal outgrowth or Schwann cell response after nerve repair in rats. Regen Med 2024; 19:327-343. [PMID: 38957920 PMCID: PMC11346556 DOI: 10.1080/17460751.2024.2361515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 05/23/2024] [Indexed: 07/04/2024] Open
Abstract
Background: Adjunctive pharmacological treatment may improve nerve regeneration. We investigated nerve regeneration processes of PXL01 - a lactoferrin-derived peptide - after repair of the sciatic nerve in healthy Wistar rats.Materials & methods: PXL01, sodium hyaluronate (carrier) or sodium chloride was administered around the repair. After 6 days axonal outgrowth, Schwann cell response, pan- (CD68) and pro-healing (CD206) macrophages in sciatic nerve, sensory neuronal response in dorsal root ganglia (DRG) and expression of heat shock protein 27 (HSP27) in sciatic nerves and DRGs were analyzed.Results: Despite a lower number of pan-macrophages, other investigated variables in sciatic nerves or DRGs did not differ between the treatment groups.Conclusion: PLX01 applied locally inhibits inflammation through pan-macrophages in repaired sciatic nerves without any impact on nerve regeneration or pro-healing macrophages.
Collapse
Affiliation(s)
- Derya Burcu Hazer Rosberg
- Department of Translational Medicine – Hand Surgery, Lund University, Skåne University Hospital, SE-20502, Malmö, Sweden
- Department of Hand Surgery, Skåne University Hospital, MalmöSE-20502, Sweden
| | - Lena Stenberg
- Department of Translational Medicine – Hand Surgery, Lund University, Skåne University Hospital, SE-20502, Malmö, Sweden
| | - Margit Mahlapuu
- Department of Chemistry & Molecular Biology, Göteborg University , SE-40530, Göteborg, Sweden
| | - Lars B Dahlin
- Department of Translational Medicine – Hand Surgery, Lund University, Skåne University Hospital, SE-20502, Malmö, Sweden
- Department of Hand Surgery, Skåne University Hospital, MalmöSE-20502, Sweden
- Department of Biomedical & Clinical Sciences, Linköping University, LinköpingSE-58183, Sweden
| |
Collapse
|
4
|
Xue R, Xie M, Wu Z, Wang S, Zhang Y, Han Z, Li C, Tang Q, Wang L, Li D, Wang S, Yang H, Zhao RC. Mesenchymal Stem Cell-Derived Exosomes Promote Recovery of The Facial Nerve Injury through Regulating Macrophage M1 and M2 Polarization by Targeting the P38 MAPK/NF-Κb Pathway. Aging Dis 2024; 15:851-868. [PMID: 37548941 PMCID: PMC10917525 DOI: 10.14336/ad.2023.0719-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 07/19/2023] [Indexed: 08/08/2023] Open
Abstract
Facial nerve (FN) injury seriously affects human social viability and causes a heavy economic and social burden. Although mesenchymal stem cell-derived exosomes (MSC-Exos) promise therapeutic benefits for injury repair, there has been no evaluation of the impact of MSC-Exos administration on FN repair. Herein, we explore the function of MSC-Exos in the immunomodulation of macrophages and their effects in repairing FN injury. An ultracentrifugation technique was used to separate exosomes from the MSC supernatant. Administrating MSC-Exos to SD rats via local injection after FN injury promoted axon regeneration and myelination and alleviated local and systemic inflammation. MSC-Exos facilitated M2 polarization and reduced the M1-M2 polarization ratio. miRNA sequencing of MSC-Exos and previous literature showed that the MAPK/NF-κb pathway was a downstream target of macrophage polarization. We confirmed this hypothesis both in vivo and in vitro. Our findings show that MSC-Exos are a potential candidate for treating FN injury because they may have superior benefits for FN injury recovery and can decrease inflammation by controlling the heterogeneity of macrophages, which is regulated by the p38 MAPK/NF-κb pathway.
Collapse
Affiliation(s)
- Ruoyan Xue
- Department of Otolaryngology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Mengyao Xie
- Department of Otolaryngology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Zhiyuan Wu
- Department of Otolaryngology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Shu Wang
- Department of Otolaryngology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Yongli Zhang
- Department of Otolaryngology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Zhijin Han
- Department of Otolaryngology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Chen Li
- Department of Otolaryngology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Qi Tang
- Department of Otolaryngology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Liping Wang
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Peking Union Medical College Hospital, Center of Excellence in Tissue Engineering Chinese Academy of Medical Sciences, Beijing Key Laboratory, Beijing, China.
| | - Di Li
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Peking Union Medical College Hospital, Center of Excellence in Tissue Engineering Chinese Academy of Medical Sciences, Beijing Key Laboratory, Beijing, China.
| | - Shihua Wang
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Peking Union Medical College Hospital, Center of Excellence in Tissue Engineering Chinese Academy of Medical Sciences, Beijing Key Laboratory, Beijing, China.
| | - Hua Yang
- Department of Otolaryngology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Robert Chunhua Zhao
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Peking Union Medical College Hospital, Center of Excellence in Tissue Engineering Chinese Academy of Medical Sciences, Beijing Key Laboratory, Beijing, China.
- School of Life Sciences, Shanghai University, Shanghai, China.
| |
Collapse
|
5
|
Sun Y, Chen D, Dai T, Yu Z, Xie H, Wang X, Zhang W. Cell-free fat extract promotes axon regeneration and retinal ganglion cells survival in traumatic optic neuropathy. Front Cell Neurosci 2024; 18:1344853. [PMID: 38515790 PMCID: PMC10954833 DOI: 10.3389/fncel.2024.1344853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 02/27/2024] [Indexed: 03/23/2024] Open
Abstract
Injuries to axons within the central nervous system (CNS) pose a substantial clinical challenge due to their limited regenerative capacity. This study investigates the therapeutic potential of Cell-free fat extract (CEFFE) in CNS injury. CEFFE was injected intravitreally after the optic nerve was crushed. Two weeks post-injury, quantification of regenerated axons and survival rates of retinal ganglion cells (RGCs) were performed. Subsequently, comprehensive gene ontology (GO) an-notation elucidated the cellular origins and functional attributes of CEFFE components. Molecular mechanisms underlying CEFFE's therapeutic effects were explored through Western blotting (WB). Additionally, levels of inflammatory factors within CEFFE were determined using enzyme-linked immunosorbent assay (ELISA), and histological staining of microglia was conducted to assess its impact on neuroinflammation. CEFFE demonstrated a significant capacity to promote axon re-generation and enhance RGCs survival. GO annotation revealed the involvement of 146 proteins within CEFFE in axonogenesis and neurogenesis. WB analysis unveiled the multifaceted pathways through which CEFFE exerts its therapeutic effects. Elevated levels of inflammatory factors were detected through ELISA, and CEFFE exhibited a modulatory effect on microglial activation in the retinal tissue following optic nerve crush (ONC). The present study highlights the therapeutic promise of CEFFE in the management of CNS injuries, exemplified by its ability to foster axon regeneration and improve RGCs survival.
Collapse
Affiliation(s)
- Yiyu Sun
- Department of Wound Reconstructive Surgery, Tongji Hospital Affiliated to Tongji University, School of Medicine, Tongji University, Shanghai, China
| | - Di Chen
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Tao Dai
- Department of Wound Reconstructive Surgery, Tongji Hospital Affiliated to Tongji University, School of Medicine, Tongji University, Shanghai, China
| | - Ziyou Yu
- Shanghai Key Laboratory of Tissue Engineering, Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Hui Xie
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiangsheng Wang
- Department of Plastic and Reconstructive Surgery, Affiliated Hangzhou First People’s Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, China
| | - Wenjie Zhang
- Shanghai Key Laboratory of Tissue Engineering, Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
6
|
Lisek M, Tomczak J, Boczek T, Zylinska L. Calcium-Associated Proteins in Neuroregeneration. Biomolecules 2024; 14:183. [PMID: 38397420 PMCID: PMC10887043 DOI: 10.3390/biom14020183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 01/27/2024] [Accepted: 02/01/2024] [Indexed: 02/25/2024] Open
Abstract
The dysregulation of intracellular calcium levels is a critical factor in neurodegeneration, leading to the aberrant activation of calcium-dependent processes and, ultimately, cell death. Ca2+ signals vary in magnitude, duration, and the type of neuron affected. A moderate Ca2+ concentration can initiate certain cellular repair pathways and promote neuroregeneration. While the peripheral nervous system exhibits an intrinsic regenerative capability, the central nervous system has limited self-repair potential. There is evidence that significant variations exist in evoked calcium responses and axonal regeneration among neurons, and individual differences in regenerative capacity are apparent even within the same type of neurons. Furthermore, some studies have shown that neuronal activity could serve as a potent regulator of this process. The spatio-temporal patterns of calcium dynamics are intricately controlled by a variety of proteins, including channels, ion pumps, enzymes, and various calcium-binding proteins, each of which can exert either positive or negative effects on neural repair, depending on the cellular context. In this concise review, we focus on several calcium-associated proteins such as CaM kinase II, GAP-43, oncomodulin, caldendrin, calneuron, and NCS-1 in order to elaborate on their roles in the intrinsic mechanisms governing neuronal regeneration following traumatic damage processes.
Collapse
Affiliation(s)
| | | | | | - Ludmila Zylinska
- Department of Molecular Neurochemistry, Medical University of Lodz, 92-215 Lodz, Poland; (M.L.); (J.T.); (T.B.)
| |
Collapse
|
7
|
Andries L, Kancheva D, Masin L, Scheyltjens I, Van Hove H, De Vlaminck K, Bergmans S, Claes M, De Groef L, Moons L, Movahedi K. Immune stimulation recruits a subset of pro-regenerative macrophages to the retina that promotes axonal regrowth of injured neurons. Acta Neuropathol Commun 2023; 11:85. [PMID: 37226256 DOI: 10.1186/s40478-023-01580-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 05/08/2023] [Indexed: 05/26/2023] Open
Abstract
The multifaceted nature of neuroinflammation is highlighted by its ability to both aggravate and promote neuronal health. While in mammals retinal ganglion cells (RGCs) are unable to regenerate following injury, acute inflammation can induce axonal regrowth. However, the nature of the cells, cellular states and signalling pathways that drive this inflammation-induced regeneration have remained elusive. Here, we investigated the functional significance of macrophages during RGC de- and regeneration, by characterizing the inflammatory cascade evoked by optic nerve crush (ONC) injury, with or without local inflammatory stimulation in the vitreous. By combining single-cell RNA sequencing and fate mapping approaches, we elucidated the response of retinal microglia and recruited monocyte-derived macrophages (MDMs) to RGC injury. Importantly, inflammatory stimulation recruited large numbers of MDMs to the retina, which exhibited long-term engraftment and promoted axonal regrowth. Ligand-receptor analysis highlighted a subset of recruited macrophages that exhibited expression of pro-regenerative secreted factors, which were able to promote axon regrowth via paracrine signalling. Our work reveals how inflammation may promote CNS regeneration by modulating innate immune responses, providing a rationale for macrophage-centred strategies for driving neuronal repair following injury and disease.
Collapse
Affiliation(s)
- Lien Andries
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Division, Department of Biology, Leuven Brain Institute, KU Leuven, Naamsestraat 61, Box 2464, 3000, Louvain, Belgium
| | - Daliya Kancheva
- Laboratory for Molecular and Cellular Therapy, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium
| | - Luca Masin
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Division, Department of Biology, Leuven Brain Institute, KU Leuven, Naamsestraat 61, Box 2464, 3000, Louvain, Belgium
| | - Isabelle Scheyltjens
- Laboratory for Molecular and Cellular Therapy, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium
| | - Hannah Van Hove
- Laboratory for Molecular and Cellular Therapy, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium
| | - Karen De Vlaminck
- Laboratory for Molecular and Cellular Therapy, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium
| | - Steven Bergmans
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Division, Department of Biology, Leuven Brain Institute, KU Leuven, Naamsestraat 61, Box 2464, 3000, Louvain, Belgium
| | - Marie Claes
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Division, Department of Biology, Leuven Brain Institute, KU Leuven, Naamsestraat 61, Box 2464, 3000, Louvain, Belgium
| | - Lies De Groef
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Division, Department of Biology, Leuven Brain Institute, KU Leuven, Naamsestraat 61, Box 2464, 3000, Louvain, Belgium
- Cellular Communication and Neurodegeneration Research Group, Animal Physiology and Neurobiology Division, Department of Biology, Leuven Brain Institute, KU Leuven, 3000, Louvain, Belgium
| | - Lieve Moons
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Division, Department of Biology, Leuven Brain Institute, KU Leuven, Naamsestraat 61, Box 2464, 3000, Louvain, Belgium.
| | - Kiavash Movahedi
- Laboratory for Molecular and Cellular Therapy, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium.
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium.
| |
Collapse
|
8
|
Shin YK, Jo YR, Lee SH, Park HT, Shin JE. Regulation of the V-ATPase subunit ATP6V0D2 and its role in demyelination after peripheral nerve injury. Biochem Biophys Res Commun 2023; 646:1-7. [PMID: 36689911 DOI: 10.1016/j.bbrc.2023.01.039] [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: 01/03/2023] [Accepted: 01/13/2023] [Indexed: 01/15/2023]
Abstract
After peripheral nerve injury, demyelinating Schwann cells discharge myelin debris and macrophages execute myelin degradation, leading to demyelination of degenerating axons, which is essential for efficient nerve regeneration. In this study, we show that vacuolar-type proton ATPase subunit d2 (Atp6v0d2) is among the most highly upregulated genes in degenerating mouse sciatic nerves after nerve injury using microarray analysis. ATP6V0D2 is mostly expressed in macrophages of injured nerves. Atp6v0d2 knockout mice display delayed peripheral nerve demyelination and significantly attenuated myelin lipid digestion after nerve injury. However, macrophage recruitment and Schwann cell dedifferentiation are unaffected by loss of Atp6v0d2 expression. Taken together, these data demonstrate that ATP6V0D2 in macrophages is specifically required for demyelination during Wallerian degeneration.
Collapse
Affiliation(s)
- Yoon Kyung Shin
- Peripheral Neuropathy Research Center (PNRC), Department of Molecular Neuroscience, College of Medicine, Dong-A University, Busan, 49201, Republic of Korea.
| | - Young Rae Jo
- Peripheral Neuropathy Research Center (PNRC), Department of Molecular Neuroscience, College of Medicine, Dong-A University, Busan, 49201, Republic of Korea
| | - Seoung Hoon Lee
- Department of Oral Microbiology and Immunology, College of Dentistry, Wonkwang University, Iksan, 54538, Republic of Korea
| | - Hwan Tae Park
- Peripheral Neuropathy Research Center (PNRC), Department of Molecular Neuroscience, College of Medicine, Dong-A University, Busan, 49201, Republic of Korea; Department of Translational Biomedical Sciences, Graduate School of Dong-A University, Busan, 49201, Republic of Korea
| | - Jung Eun Shin
- Peripheral Neuropathy Research Center (PNRC), Department of Molecular Neuroscience, College of Medicine, Dong-A University, Busan, 49201, Republic of Korea; Department of Translational Biomedical Sciences, Graduate School of Dong-A University, Busan, 49201, Republic of Korea.
| |
Collapse
|
9
|
Wang X, Yang C, Wang X, Miao J, Chen W, Zhou Y, Xu Y, An Y, Cheng A, Ye W, Chen M, Song D, Yuan X, Wang J, Qian P, Ruohao Wu A, Zhang ZY, Liu K. Driving axon regeneration by orchestrating neuronal and non-neuronal innate immune responses via the IFNγ-cGAS-STING axis. Neuron 2023; 111:236-255.e7. [PMID: 36370710 PMCID: PMC9851977 DOI: 10.1016/j.neuron.2022.10.028] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 09/20/2022] [Accepted: 10/18/2022] [Indexed: 11/13/2022]
Abstract
The coordination mechanism of neural innate immune responses for axon regeneration is not well understood. Here, we showed that neuronal deletion of protein tyrosine phosphatase non-receptor type 2 sustains the IFNγ-STAT1 activity in retinal ganglion cells (RGCs) to promote axon regeneration after injury, independent of mTOR or STAT3. DNA-damage-induced cGAMP synthase (cGAS)-stimulator of interferon genes (STINGs) activation is the functional downstream signaling. Directly activating neuronal STING by cGAMP promotes axon regeneration. In contrast to the central axons, IFNγ is locally translated in the injured peripheral axons and upregulates cGAS expression in Schwann cells and infiltrating blood cells to produce cGAMP, which promotes spontaneous axon regeneration as an immunotransmitter. Our study demonstrates that injured peripheral nervous system (PNS) axons can direct the environmental innate immune response for self-repair and that the neural antiviral mechanism can be harnessed to promote axon regeneration in the central nervous system (CNS).
Collapse
Affiliation(s)
- Xu Wang
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China,Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China,Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, China,Biomedical Research Institute, Shenzhen Peking University–The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, China,Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen, Guangdong 518057, China,Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangzhou 510515, China,These authors contributed equally
| | - Chao Yang
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China,Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China,Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, China,Biomedical Research Institute, Shenzhen Peking University–The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, China,Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen, Guangdong 518057, China,Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangzhou 510515, China,These authors contributed equally
| | - Xuejie Wang
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China,Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Jinmin Miao
- Department of Medicinal Chemistry and Molecular Pharmacology, Department of Chemistry, Center for Cancer Research and Institute for Drug Discovery, Purdue University, West Lafayette, IN, USA
| | - Weitao Chen
- Biomedical Research Institute, Shenzhen Peking University–The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, China
| | - Yiren Zhou
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Ying Xu
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China,Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Yongyan An
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Aifang Cheng
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, China,Department of Ocean Science, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Wenkang Ye
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, China,Department of Ocean Science, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Mengxian Chen
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Dong Song
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China,Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Xue Yuan
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Jiguang Wang
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China,Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Peiyuan Qian
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, China,Department of Ocean Science, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Angela Ruohao Wu
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China,Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, China,Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Hong Kong, China,Center for Aging Science, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Zhong-Yin Zhang
- Department of Medicinal Chemistry and Molecular Pharmacology, Department of Chemistry, Center for Cancer Research and Institute for Drug Discovery, Purdue University, West Lafayette, IN, USA
| | - Kai Liu
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China; Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China; Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, China; Biomedical Research Institute, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, China; Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen, Guangdong 518057, China; Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangzhou 510515, China.
| |
Collapse
|
10
|
Zhao J, Wang X, He Y, Xu P, Lai L, Chung Y, Pan X. The Role of T Cells in Alzheimer's Disease Pathogenesis. Crit Rev Immunol 2023; 43:15-23. [PMID: 37943150 DOI: 10.1615/critrevimmunol.2023050145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2023]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder associated with memory decline and cognitive impairment, which is related to hallmark protein aggregates, amyloid-β (Аβ) plaques and neurofibrillary tangles; the latter are accumulated with hyperphosphorylated Tau protein. Immune cells play an important role in AD pathogenesis. Although the role of T cells in AD remains controversial, studies have shown that T cell deficiency is associated with increased AD pathology. In contrast, transplantation of T cells reduces AD pathology. T cells can help B cells generate anti-Аβ antibody to neutralize the toxin of Аβ and hyperphosphorylated Tau. T cells also activate macrophages to phagocytose misfolded proteins including Аβ and Tau. Recent data have also shown that AD animals have a damaged thymic microenvironment, especially thymic epithelial cells (TECs), resulting in decreased T cell numbers, which contribute to AD pathology. Therefore, regulation of T cell regeneration, for example by rejuvenating the thymic microenvironment, has the potential to be used in the treatment of AD.
Collapse
Affiliation(s)
- Jin Zhao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, and Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Guangzhou, Guangdong, 510515, China; ZhuHai Hengqin ImStem Biotechnology Co. Ltd., Hengqin New District Huandao Donglu 1889 Building 3, Zhuhai, Guangdong, 519000, China; Key Laboratory of Mental Health of the Ministry of Education, Guangdong Province Key Laboratory of Psychiatric Disorders, School of Basic Medical Sciences, Southern Medical University, Guangdong, 510515, China
| | - Xiaofang Wang
- ZhuHai Hengqin ImStem Biotechnology Co. Ltd., Hengqin New District Huandao Donglu 1889 Building 3, Zhuhai, Guangdong, 519000, China; ImStem Biotechnology, Inc., 400 Farmington Avenue R1808, Farmington, CT 06030, USA
| | - Yusheng He
- ZhuHai Hengqin ImStem Biotechnology Co. Ltd., Hengqin New District Huandao Donglu 1889 Building 3, Zhuhai, Guangdong, 519000, China
| | - Pingyi Xu
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Laijun Lai
- Department of Allied Health Sciences, University of Connecticut, Storrs, CT, USA; University of Connecticut Stem Cell Institute, University of Connecticut, Storrs, CT, USA
| | - Younggie Chung
- ZhuHai Hengqin ImStem Biotechnology Co. Ltd., Hengqin New District Huandao Donglu 1889 Building 3, Zhuhai, Guangdong, 519000, China; ImStem Biotechnology, Inc., 400 Farmington Avenue R1808, Farmington, CT 06030, USA
| | - Xinghua Pan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, and Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Guangzhou, Guangdong, 510515, China; Key Laboratory of Mental Health of the Ministry of Education, Guangdong Province Key Laboratory of Psychiatric Disorders, School of Basic Medical Sciences, Southern Medical University, Guangdong, 510515, China
| |
Collapse
|
11
|
Molecular mechanisms of exercise contributing to tissue regeneration. Signal Transduct Target Ther 2022; 7:383. [PMID: 36446784 PMCID: PMC9709153 DOI: 10.1038/s41392-022-01233-2] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 10/03/2022] [Accepted: 10/17/2022] [Indexed: 12/03/2022] Open
Abstract
Physical activity has been known as an essential element to promote human health for centuries. Thus, exercise intervention is encouraged to battle against sedentary lifestyle. Recent rapid advances in molecular biotechnology have demonstrated that both endurance and resistance exercise training, two traditional types of exercise, trigger a series of physiological responses, unraveling the mechanisms of exercise regulating on the human body. Therefore, exercise has been expected as a candidate approach of alleviating a wide range of diseases, such as metabolic diseases, neurodegenerative disorders, tumors, and cardiovascular diseases. In particular, the capacity of exercise to promote tissue regeneration has attracted the attention of many researchers in recent decades. Since most adult human organs have a weak regenerative capacity, it is currently a key challenge in regenerative medicine to improve the efficiency of tissue regeneration. As research progresses, exercise-induced tissue regeneration seems to provide a novel approach for fighting against injury or senescence, establishing strong theoretical basis for more and more "exercise mimetics." These drugs are acting as the pharmaceutical alternatives of those individuals who cannot experience the benefits of exercise. Here, we comprehensively provide a description of the benefits of exercise on tissue regeneration in diverse organs, mainly focusing on musculoskeletal system, cardiovascular system, and nervous system. We also discuss the underlying molecular mechanisms associated with the regenerative effects of exercise and emerging therapeutic exercise mimetics for regeneration, as well as the associated opportunities and challenges. We aim to describe an integrated perspective on the current advances of distinct physiological mechanisms associated with exercise-induced tissue regeneration on various organs and facilitate the development of drugs that mimics the benefits of exercise.
Collapse
|
12
|
Sex Differences in Neuropathy: The Paradigmatic Case of MetFormin. Int J Mol Sci 2022; 23:ijms232314503. [PMID: 36498830 PMCID: PMC9738696 DOI: 10.3390/ijms232314503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/16/2022] [Accepted: 11/17/2022] [Indexed: 11/24/2022] Open
Abstract
As a widely prescribed anti-diabetic drug, metformin has been receiving novel attention for its analgesic potential. In the study of the complex etiology of neuropathic pain (NeP), male and female individuals exhibit quite different responses characterized by higher pain sensitivity and greater NeP incidence in women. This "gender gap" in our knowledge of sex differences in pain processing strongly limits the sex-oriented treatment of patients suffering from NeP. Besides, the current investigation of the analgesic potential of metformin has not addressed the "gender gap" problem. Hence, this study focuses on metformin and sex-dependent analgesia in a murine model of NeP induced by chronic constriction injury of the sciatic nerve. We investigated sexual dimorphism in signaling pathways involved by 7 days of metformin administration, such as changes in AMP-activated protein kinase and the positive regulation of autophagy machinery, discovering that metformin affected in a sexually dimorphic manner the immunological and inflammatory response to nerve lesion. These effects were complemented by morphological and adaptive changes occurring after peripheral nerve injury. Altogether these data can contribute to explaining a number of potential mechanisms responsible for the complete recovery from NeP found in male mice, as opposed to the failure of long-lasting recovery in female animals.
Collapse
|
13
|
Qin HJ, Li H, Chen JZ, Zhang KR, Zhao XQ, Qin JQ, Yu B, Yang J. Artificial nerve graft constructed by coculture of activated Schwann cells and human hair keratin for repair of peripheral nerve defects. Neural Regen Res 2022; 18:1118-1123. [PMID: 36255001 PMCID: PMC9827759 DOI: 10.4103/1673-5374.355817] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Studies have shown that human hair keratin (HHK) has no antigenicity and excellent mechanical properties. Schwann cells, as unique glial cells in the peripheral nervous system, can be induced by interleukin-1β to secrete nerve growth factor, which promotes neural regeneration. Therefore, HHK with Schwann cells may be a more effective approach to repair nerve defects than HHK without Schwann cells. In this study, we established an artificial nerve graft by loading an HHK skeleton with activated Schwann cells. We found that the longitudinal HHK microfilament structure provided adhesion medium, space and direction for Schwann cells, and promoted Schwann cell growth and nerve fiber regeneration. In addition, interleukin-1β not only activates Schwann cells, but also strengthens their activity and increases the expression of nerve growth factors. Activated Schwann cells activate macrophages, and activated macrophages secrete interleukin-1β, which maintains the activity of Schwann cells. Thus, a beneficial cycle forms and promotes nerve repair. Furthermore, our studies have found that the newly constructed artificial nerve graft promotes the improvements in nerve conduction function and motor function in rats with sciatic nerve injury, and increases the expression of nerve injury repair factors fibroblast growth factor 2 and human transforming growth factor B receptor 2. These findings suggest that this artificial nerve graft effectively repairs peripheral nerve injury.
Collapse
Affiliation(s)
- Han-Jun Qin
- Department of Orthopedics, Nanfang Hospital, Southern Medical University; Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Hang Li
- Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Jun-Ze Chen
- Department of Orthopedics, Baiyun Branch of Southern Hospital, Guangzhou, Guangdong Province, China
| | - Kai-Rui Zhang
- Department of Orthopedics, Nanfang Hospital, Southern Medical University; Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Xing-Qi Zhao
- Department of Orthopedics, Nanfang Hospital, Southern Medical University; Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Jian-Qiang Qin
- Department of Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Bin Yu
- Department of Orthopedics, Nanfang Hospital, Southern Medical University; Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China,Correspondence to: Jun Yang, ; Bin Yu, .
| | - Jun Yang
- Department of Orthopedics, Nanfang Hospital, Southern Medical University; Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China,Department of Orthopedics, The 74th Group Military Hospital of PLA, Guangzhou, Guangdong Province, China,Correspondence to: Jun Yang, ; Bin Yu, .
| |
Collapse
|
14
|
Asiedu K. Role of ocular surface neurobiology in neuronal-mediated inflammation in dry eye disease. Neuropeptides 2022; 95:102266. [PMID: 35728484 DOI: 10.1016/j.npep.2022.102266] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 06/07/2022] [Accepted: 06/08/2022] [Indexed: 01/18/2023]
Abstract
Inflammation is the consequence of dry eye disease regardless of its etiology. Several injurious or harmless processes to the ocular surface neurons promote ocular surface neurogenic inflammation, leading to the vicious cycle of dry eye disease. These processes include the regular release of neuromediators during the conduction of ocular surface sensations, hyperosmolarity-induced ocular surface neuronal damage, neuro-regenerative activities, and neuronal-mediated dendritic cell activities. Neurogenic inflammation appears to be the main culprit, instigating the self-perpetuating inflammation observed in patients with dry eye disease.
Collapse
Affiliation(s)
- Kofi Asiedu
- School of Optometry & Vision Science, Faculty of Medicine and Health, University of New South Wales, Sydney, Australia.
| |
Collapse
|
15
|
Eid SA, Savelieff MG, Eid AA, Feldman EL. Nox, Nox, Are You There? The Role of NADPH Oxidases in the Peripheral Nervous System. Antioxid Redox Signal 2022; 37:613-630. [PMID: 34861780 PMCID: PMC9634986 DOI: 10.1089/ars.2021.0135] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 11/23/2021] [Accepted: 11/26/2021] [Indexed: 02/06/2023]
Abstract
Significance: Reactive oxygen species (ROS) contribute to multiple aspects of peripheral nervous system (PNS) biology ranging from physiological processes (e.g., axonal outgrowth and regeneration) to pathophysiology (e.g., nerve degeneration). Although ROS are derived from multiple sources, NADPH oxidase (Nox) family members are dedicated to ROS generation. Noxs are expressed in the PNS, and their overexpression is associated with detrimental effects on nerve function and contributes, at least in part, to peripheral neuropathies. Recent Advances: Of the seven members, studies mostly focused on Nox1, Nox2, and Nox4, which are expressed in the PNS in a cell-specific manner. We have also recently identified human Nox5 in sural nerve biopsies. When maintained at homeostatic levels, Noxs regulate several aspects of peripheral nerve health, most notably neurite outgrowth and axonal regeneration following nerve lesion. While Nox2 and Nox4 dysregulation is a major source of oxidative stress in PNS disorders, including neuropathic pain and diabetic peripheral neuropathy, recent evidence also implicates Nox1 and Nox5. Critical Issues: Although there is compelling evidence for a direct role of Noxs on nerve function, little is known about their subcellular localization, intercellular regulation, and interaction. These, together with redox signaling, are considered crucial components of nerve redox status. In addition, the lack of isoform-specific inhibitors limits conclusions about the physiological role of Noxs in the PNS and their therapeutic potential in peripheral neuropathies. Future Directions: Future research using isoform-specific genetic and pharmacological approaches are therefore needed to better understand the significance of Nox enzymes in PNS (patho) physiology. Antioxid. Redox Signal. 37, 613-630.
Collapse
Affiliation(s)
- Stéphanie A. Eid
- Department of Neurology, School of Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Department of Neurology, NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, Michigan, USA
| | - Masha G. Savelieff
- Department of Neurology, NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, Michigan, USA
| | - Assaad A. Eid
- Department of Anatomy, Cell Biology, and Physiological Sciences, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon
| | - Eva L. Feldman
- Department of Neurology, School of Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Department of Neurology, NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
16
|
Jacobi A, Tran NM, Yan W, Benhar I, Tian F, Schaffer R, He Z, Sanes JR. Overlapping transcriptional programs promote survival and axonal regeneration of injured retinal ganglion cells. Neuron 2022; 110:2625-2645.e7. [PMID: 35767994 PMCID: PMC9391321 DOI: 10.1016/j.neuron.2022.06.002] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 04/08/2022] [Accepted: 06/03/2022] [Indexed: 12/12/2022]
Abstract
Injured neurons in the adult mammalian central nervous system often die and seldom regenerate axons. To uncover transcriptional pathways that could ameliorate these disappointing responses, we analyzed three interventions that increase survival and regeneration of mouse retinal ganglion cells (RGCs) following optic nerve crush (ONC) injury, albeit not to a clinically useful extent. We assessed gene expression in each of 46 RGC types by single-cell transcriptomics following ONC and treatment. We also compared RGCs that regenerated with those that survived but did not regenerate. Each intervention enhanced survival of most RGC types, but type-independent axon regeneration required manipulation of multiple pathways. Distinct computational methods converged on separate sets of genes selectively expressed by RGCs likely to be dying, surviving, or regenerating. Overexpression of genes associated with the regeneration program enhanced both survival and axon regeneration in vivo, indicating that mechanistic analysis can be used to identify novel therapeutic strategies.
Collapse
Affiliation(s)
- Anne Jacobi
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, and Department of Neurology, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA; Department of Molecular and Cellular Biology, Center for Brain Science, Harvard University, 52 Oxford Street, Cambridge, MA 02138, USA
| | - Nicholas M Tran
- Department of Molecular and Cellular Biology, Center for Brain Science, Harvard University, 52 Oxford Street, Cambridge, MA 02138, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA
| | - Wenjun Yan
- Department of Molecular and Cellular Biology, Center for Brain Science, Harvard University, 52 Oxford Street, Cambridge, MA 02138, USA
| | - Inbal Benhar
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Feng Tian
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, and Department of Neurology, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Rebecca Schaffer
- Department of Molecular and Cellular Biology, Center for Brain Science, Harvard University, 52 Oxford Street, Cambridge, MA 02138, USA
| | - Zhigang He
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, and Department of Neurology, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA.
| | - Joshua R Sanes
- Department of Molecular and Cellular Biology, Center for Brain Science, Harvard University, 52 Oxford Street, Cambridge, MA 02138, USA.
| |
Collapse
|
17
|
The Anti-inflammation Property of Olfactory Ensheathing Cells in Neural Regeneration After Spinal Cord Injury. Mol Neurobiol 2022; 59:6447-6459. [PMID: 35962300 DOI: 10.1007/s12035-022-02983-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 07/29/2022] [Indexed: 10/15/2022]
Abstract
Neural regeneration has troubled investigators worldwide in the past decades. Currently, cell transplantation emerged as a breakthrough targeted therapy for spinal cord injury (SCI) in the neurotrauma field, which provides a promising strategy in neural regeneration. Olfactory ensheathing cells (OECs), a specialized type of glial cells, is considered as the excellent candidate due to its unique variable and intrinsic regeneration-supportive properties. In fact, OECs could support olfactory receptor neuron turnover and axonal extension, which is essential to maintain the function of olfactory nervous system. Hitherto, an increasing number of literatures demonstrate that transplantation of OECs exerts vital roles in neural regeneration and functional recovery after neural injury, including central and peripheral nervous system. It is common knowledge that the deteriorating microenvironment (ischemia, hypoxia, scar, acute and chronic inflammation, etc.) resulting from injured nervous system is adverse for neural regeneration. Interestingly, recent studies indicated that OECs could promote neural repair through improvement of the disastrous microenvironments, especially to the overwhelmed inflammatory responses. Although OECs possess unusual advantages over other cells for neural repair, particularly in SCI, the mechanisms of OEC-mediated neural repair are still controversial with regard to anti-inflammation. Therefore, it is significant to summarize the anti-inflammation property of OECs, which is helpful to understand the biological characteristics of OECs and drive future studies. Here, we mainly focus on the anti-inflammatory role of OECs to make systematic review and discuss OEC-based therapy for CNS injury.
Collapse
|
18
|
Pinelli F, Pizzetti F, Veneruso V, Petillo E, Raghunath M, Perale G, Veglianese P, Rossi F. Biomaterial-Mediated Factor Delivery for Spinal Cord Injury Treatment. Biomedicines 2022; 10:biomedicines10071673. [PMID: 35884981 PMCID: PMC9313204 DOI: 10.3390/biomedicines10071673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 05/24/2022] [Accepted: 07/05/2022] [Indexed: 11/19/2022] Open
Abstract
Spinal cord injury (SCI) is an injurious process that begins with immediate physical damage to the spinal cord and associated tissues during an acute traumatic event. However, the tissue damage expands in both intensity and volume in the subsequent subacute phase. At this stage, numerous events exacerbate the pathological condition, and therein lies the main cause of post-traumatic neural degeneration, which then ends with the chronic phase. In recent years, therapeutic interventions addressing different neurodegenerative mechanisms have been proposed, but have met with limited success when translated into clinical settings. The underlying reasons for this are that the pathogenesis of SCI is a continued multifactorial disease, and the treatment of only one factor is not sufficient to curb neural degeneration and resulting paralysis. Recent advances have led to the development of biomaterials aiming to promote in situ combinatorial strategies using drugs/biomolecules to achieve a maximized multitarget approach. This review provides an overview of single and combinatorial regenerative-factor-based treatments as well as potential delivery options to treat SCIs.
Collapse
Affiliation(s)
- Filippo Pinelli
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Via Mancinelli 7, 20131 Milan, Italy; (F.P.); (F.P.); (E.P.)
| | - Fabio Pizzetti
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Via Mancinelli 7, 20131 Milan, Italy; (F.P.); (F.P.); (E.P.)
| | - Valeria Veneruso
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy;
| | - Emilia Petillo
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Via Mancinelli 7, 20131 Milan, Italy; (F.P.); (F.P.); (E.P.)
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy;
| | - Michael Raghunath
- Center for Cell Biology and Tissue Engineering, Institute for Chemistry and Biotechnology (ICBT), Zurich University of Applied Sciences (ZHAW), 8820 Wädenswil, Switzerland;
| | - Giuseppe Perale
- Faculty of Biomedical Sciences, University of Southern Switzerland (USI), Via Buffi 13, 6900 Lugano, Switzerland;
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Donaueschingenstrasse 13, 1200 Vienna, Austria
| | - Pietro Veglianese
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy;
- Correspondence: (P.V.); (F.R.); Tel.: +39-02-3901-4205 (P.V.); +39-02-2399-3145 (F.R.)
| | - Filippo Rossi
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Via Mancinelli 7, 20131 Milan, Italy; (F.P.); (F.P.); (E.P.)
- Correspondence: (P.V.); (F.R.); Tel.: +39-02-3901-4205 (P.V.); +39-02-2399-3145 (F.R.)
| |
Collapse
|
19
|
Shen YY, Zhang RR, Liu QY, Li SY, Yi S. Robust temporal changes of cellular senescence and proliferation after sciatic nerve injury. Neural Regen Res 2022; 17:1588-1595. [PMID: 34916445 PMCID: PMC8771116 DOI: 10.4103/1673-5374.330619] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 07/28/2021] [Accepted: 09/03/2021] [Indexed: 01/10/2023] Open
Abstract
Cellular senescence and proliferation are essential for wound healing and tissue remodeling. However, senescence-proliferation cell fate after peripheral nerve injury has not been clearly revealed. Here, post-injury gene expression patterns in rat sciatic nerve stumps (SRP113121) and L4-5 dorsal root ganglia (SRP200823) obtained from the National Center for Biotechnology Information were analyzed to decipher cellular senescence and proliferation-associated genetic changes. We first constructed a rat sciatic nerve crush model. Then, β-galactosidase activities were determined to indicate the existence of cellular senescence in the injured sciatic nerve. Ki67 and EdU immunostaining was performed to indicate cellular proliferation in the injured sciatic nerve. Both cellular senescence and proliferation were less vigorous in the dorsal root ganglia than in sciatic nerve stumps. These results reveal the dynamic changes of injury-induced cellular senescence and proliferation from both genetic and morphological aspects, and thus extend our understanding of the biological processes following peripheral nerve injury. The study was approved by the Animal Ethics Committee of Nantong University, China (approval No. 20190226-001) on February 26, 2019.
Collapse
Affiliation(s)
- Yin-Ying Shen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu Province, China
| | - Rui-Rui Zhang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu Province, China
| | - Qian-Yan Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu Province, China
| | - Shi-Ying Li
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu Province, China
| | - 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 Province, China
| |
Collapse
|
20
|
Haidar MA, Shakkour Z, Reslan MA, Al-Haj N, Chamoun P, Habashy K, Kaafarani H, Shahjouei S, Farran SH, Shaito A, Saba ES, Badran B, Sabra M, Kobeissy F, Bizri M. SARS-CoV-2 involvement in central nervous system tissue damage. Neural Regen Res 2022; 17:1228-1239. [PMID: 34782556 PMCID: PMC8643043 DOI: 10.4103/1673-5374.327323] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 06/30/2021] [Accepted: 07/28/2021] [Indexed: 12/18/2022] Open
Abstract
As the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continues to spread globally, it became evident that the SARS-CoV-2 virus infects multiple organs including the brain. Several clinical studies revealed that patients with COVID-19 infection experience an array of neurological signs ranging in severity from headaches to life-threatening strokes. Although the exact mechanism by which the SARS-CoV-2 virus directly impacts the brain is not fully understood, several theories have been suggested including direct and indirect pathways induced by the virus. One possible theory is the invasion of SARS-CoV-2 to the brain occurs either through the bloodstream or via the nerve endings which is considered to be the direct route. Such findings are based on studies reporting the presence of viral material in the cerebrospinal fluid and brain cells. Nevertheless, the indirect mechanisms, including blood-clotting abnormalities and prolonged activation of the immune system, can result in further tissue and organ damages seen during the course of the disease. This overview attempts to give a thorough insight into SARS-CoV-2 coronavirus neurological infection and highlights the possible mechanisms leading to the neurological manifestations observed in infected patients.
Collapse
Affiliation(s)
- Muhammad Ali Haidar
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Zaynab Shakkour
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Mohammad Amine Reslan
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Nadine Al-Haj
- Faculty of Health Sciences, University of Balamand, Beirut, Lebanon
| | - Perla Chamoun
- Faculty of Medicine, University of Balamand, Koura, Lebanon
| | - Karl Habashy
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | | | - Shima Shahjouei
- Program for Neurotrauma, Neuroproteomics & Biomarkers Research, Departments of Emergency Medicine, University of Florida, Gainesville, FL, USA
| | - Sarah H. Farran
- Department of Pathology and Laboratory Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | | | - Esber S. Saba
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Bassam Badran
- Laboratory of Cancer Biology and Molecular Immunology, Faculty of Sciences I, Lebanese University, Hadath, Beirut, Lebanon
| | - Mirna Sabra
- Faculty of Medicine, Lebanese University, Neuroscience Research Center (NRC), Beirut, Lebanon
| | - Firas Kobeissy
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Neuroscience Institute, Neurology Department, Geisinger Health System, PA, USA
| | - Maya Bizri
- Department of Psychiatry, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
21
|
Cooke P, Janowitz H, Dougherty SE. Neuronal Redevelopment and the Regeneration of Neuromodulatory Axons in the Adult Mammalian Central Nervous System. Front Cell Neurosci 2022; 16:872501. [PMID: 35530177 PMCID: PMC9074815 DOI: 10.3389/fncel.2022.872501] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 03/24/2022] [Indexed: 01/09/2023] Open
Abstract
One reason that many central nervous system injuries, including those arising from traumatic brain injury, spinal cord injury, and stroke, have limited recovery of function is that neurons within the adult mammalian CNS lack the ability to regenerate their axons following trauma. This stands in contrast to neurons of the adult mammalian peripheral nervous system (PNS). New evidence, provided by single-cell expression profiling, suggests that, following injury, both mammalian central and peripheral neurons can revert to an embryonic-like growth state which is permissive for axon regeneration. This “redevelopment” strategy could both facilitate a damage response necessary to isolate and repair the acute damage from injury and provide the intracellular machinery necessary for axon regrowth. Interestingly, serotonin neurons of the rostral group of raphe nuclei, which project their axons into the forebrain, display a robust ability to regenerate their axons unaided, counter to the widely held view that CNS axons cannot regenerate without experimental intervention after injury. Furthermore, initial evidence suggests that norepinephrine neurons within the locus coeruleus possess similar regenerative abilities. Several morphological characteristics of serotonin axon regeneration in adult mammals, observable using longitudinal in vivo imaging, are distinct from the known characteristics of unaided peripheral nerve regeneration, or of the regeneration seen in the spinal cord and optic nerve that occurs with experimental intervention. These results suggest that there is an alternative CNS program for axon regeneration that likely differs from that displayed by the PNS.
Collapse
Affiliation(s)
- Patrick Cooke
- Linden Lab, Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Haley Janowitz
- Linden Lab, Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Sarah E Dougherty
- Linden Lab, Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
22
|
What Is Parvalbumin for? Biomolecules 2022; 12:biom12050656. [PMID: 35625584 PMCID: PMC9138604 DOI: 10.3390/biom12050656] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 04/25/2022] [Accepted: 04/28/2022] [Indexed: 12/28/2022] Open
Abstract
Parvalbumin (PA) is a small, acidic, mostly cytosolic Ca2+-binding protein of the EF-hand superfamily. Structural and physical properties of PA are well studied but recently two highly conserved structural motifs consisting of three amino acids each (clusters I and II), which contribute to the hydrophobic core of the EF-hand domains, have been revealed. Despite several decades of studies, physiological functions of PA are still poorly known. Since no target proteins have been revealed for PA so far, it is believed that PA acts as a slow calcium buffer. Numerous experiments on various muscle systems have shown that PA accelerates the relaxation of fast skeletal muscles. It has been found that oxidation of PA by reactive oxygen species (ROS) is conformation-dependent and one more physiological function of PA in fast muscles could be a protection of these cells from ROS. PA is thought to regulate calcium-dependent metabolic and electric processes within the population of gamma-aminobutyric acid (GABA) neurons. Genetic elimination of PA results in changes in GABAergic synaptic transmission. Mammalian oncomodulin (OM), the β isoform of PA, is expressed mostly in cochlear outer hair cells and in vestibular hair cells. OM knockout mice lose their hearing after 3–4 months. It was suggested that, in sensory cells, OM maintains auditory function, most likely affecting outer hair cells’ motility mechanisms.
Collapse
|
23
|
Yeoh S, Warner WS, Merchant SS, Hsu EW, Agoston DV, Mahan MA. Incorporating Blood Flow in Nerve Injury and Regeneration Assessment. Front Surg 2022; 9:862478. [PMID: 35529911 PMCID: PMC9069240 DOI: 10.3389/fsurg.2022.862478] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 04/04/2022] [Indexed: 01/22/2023] Open
Abstract
Peripheral nerve injury is a significant public health challenge, with limited treatment options and potential lifelong impact on function. More than just an intrinsic part of nerve anatomy, the vascular network of nerves impact regeneration, including perfusion for metabolic demands, appropriate signaling and growth factors, and structural scaffolding for Schwann cell and axonal migration. However, the established nerve injury classification paradigm proposed by Sydney Sunderland in 1951 is based solely on hierarchical disruption to gross anatomical nerve structures and lacks further information regarding the state of cellular, metabolic, or inflammatory processes that are critical in determining regenerative outcomes. This review covers the anatomical structure of nerve-associated vasculature, and describes the biological processes that makes these vessels critical to successful end-organ reinnervation after severe nerve injuries. We then propose a theoretical framework that incorporates measurements of blood vessel perfusion and inflammation to unify perspectives on all mechanisms of nerve injury.
Collapse
Affiliation(s)
- Stewart Yeoh
- Department of Neurosurgery, University of Utah, Salt Lake City, Utah, United States
| | - Wesley S. Warner
- Department of Neurosurgery, University of Utah, Salt Lake City, Utah, United States
| | - Samer S. Merchant
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah, United States
| | - Edward W. Hsu
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah, United States
| | - Denes v. Agoston
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States
| | - Mark A. Mahan
- Department of Neurosurgery, University of Utah, Salt Lake City, Utah, United States
| |
Collapse
|
24
|
Xie L, Cen LP, Li Y, Gilbert HY, Strelko O, Berlinicke C, Stavarache MA, Ma M, Wang Y, Cui Q, Kaplitt MG, Zack DJ, Benowitz LI, Yin Y. Monocyte-derived SDF1 supports optic nerve regeneration and alters retinal ganglion cells' response to Pten deletion. Proc Natl Acad Sci U S A 2022; 119:e2113751119. [PMID: 35394873 PMCID: PMC9169637 DOI: 10.1073/pnas.2113751119] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 02/23/2022] [Indexed: 12/22/2022] Open
Abstract
Although mammalian retinal ganglion cells (RGCs) normally cannot regenerate axons nor survive after optic nerve injury, this failure is partially reversed by inducing sterile inflammation in the eye. Infiltrative myeloid cells express the axogenic protein oncomodulin (Ocm) but additional, as-yet-unidentified, factors are also required. We show here that infiltrative macrophages express stromal cell–derived factor 1 (SDF1, CXCL12), which plays a central role in this regard. Among many growth factors tested in culture, only SDF1 enhances Ocm activity, an effect mediated through intracellular cyclic AMP (cAMP) elevation and phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K) activation. SDF1 deficiency in myeloid cells (CXCL12flx/flxLysM-Cre−/+ mice) or deletion of the SDF1 receptor CXCR4 in RGCs (intraocular AAV2-Cre in CXCR4flx/flx mice) or SDF1 antagonist AMD3100 greatly suppresses inflammation-induced regeneration and decreases RGC survival to baseline levels. Conversely, SDF1 induces optic nerve regeneration and RGC survival, and, when combined with Ocm/cAMP, SDF1 increases axon regeneration to levels similar to those induced by intraocular inflammation. In contrast to deletion of phosphatase and tensin homolog (Pten), which promotes regeneration selectively from αRGCs, SDF1 promotes regeneration from non-αRGCs and enables the latter cells to respond robustly to Pten deletion; however, SDF1 surprisingly diminishes the response of αRGCs to Pten deletion. When combined with inflammation and Pten deletion, SDF1 enables many RGCs to regenerate axons the entire length of the optic nerve. Thus, SDF1 complements the effects of Ocm in mediating inflammation-induced regeneration and enables different RGC subtypes to respond to Pten deletion.
Collapse
Affiliation(s)
- Lili Xie
- Department of Neurosurgery, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115
- F. M. Kirby Neurobiology Center, Boston Children’s Hospital, Boston, MA 02115
| | - Ling-Ping Cen
- Department of Neurosurgery, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115
- F. M. Kirby Neurobiology Center, Boston Children’s Hospital, Boston, MA 02115
- Joint Shantou International Eye Center, Shantou University and The Chinese University of Hong Kong, Shantou 515000, China
| | - Yiqing Li
- Department of Neurosurgery, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115
- F. M. Kirby Neurobiology Center, Boston Children’s Hospital, Boston, MA 02115
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510085, China
| | - Hui-Ya Gilbert
- Department of Neurosurgery, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115
- F. M. Kirby Neurobiology Center, Boston Children’s Hospital, Boston, MA 02115
| | - Oleksandr Strelko
- Department of Neurosurgery, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115
- F. M. Kirby Neurobiology Center, Boston Children’s Hospital, Boston, MA 02115
| | - Cynthia Berlinicke
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21287
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21287
| | - Mihaela A. Stavarache
- Laboratory of Molecular Neurosurgery, Department of Neurological Surgery, Weill Cornell Medical College, New York, NY 10065
| | - Madeline Ma
- Department of Neurosurgery, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115
- F. M. Kirby Neurobiology Center, Boston Children’s Hospital, Boston, MA 02115
| | - Yongting Wang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Qi Cui
- Department of Neurosurgery, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115
- F. M. Kirby Neurobiology Center, Boston Children’s Hospital, Boston, MA 02115
- Joint Shantou International Eye Center, Shantou University and The Chinese University of Hong Kong, Shantou 515000, China
| | - Michael G. Kaplitt
- Laboratory of Molecular Neurosurgery, Department of Neurological Surgery, Weill Cornell Medical College, New York, NY 10065
| | - Donald J. Zack
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21287
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21287
| | - Larry I. Benowitz
- Department of Neurosurgery, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115
- F. M. Kirby Neurobiology Center, Boston Children’s Hospital, Boston, MA 02115
- Department of Ophthalmology, Harvard Medical School, Boston, MA 02115
- Program in Neuroscience, Harvard Medical School, Boston, MA 02115
| | - Yuqin Yin
- Department of Neurosurgery, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115
- F. M. Kirby Neurobiology Center, Boston Children’s Hospital, Boston, MA 02115
| |
Collapse
|
25
|
Jiao L, Zhang Y, Wang H, Fan D. Corneal confocal microscopy in the evaluation of immune-related motor neuron disease syndrome. BMC Neurol 2022; 22:138. [PMID: 35410142 PMCID: PMC8996532 DOI: 10.1186/s12883-022-02667-5] [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: 12/28/2021] [Accepted: 04/07/2022] [Indexed: 11/21/2022] Open
Abstract
Background To investigate the sensitivity and specificity of corneal confocal microscopy (CCM) in the diagnosis of immune-related motor neuron disease syndrome and evaluation of the response to immunosuppressive therapy. Methods Seventy-two patients with clinical manifestations of motor neuron disease (MND) were analysed. According to whether they had concomitant rheumatic immune disease or rheumatic immune antibody abnormalities, they were divided into an MND group (33 patients) and an immune-related MND syndrome group (39 patients). Another 10 healthy adults were selected as the control group. All individuals were examined by CCM. Results For Langerhans cell(LC) density, the area under the receiver operating characteristic(ROC)curve was 0.8, the best cut-off was 67.7 cells/mm2, the sensitivity was 79.5%, and the specificity was 72.7%. For inferior whorl length (IWL), the area under the ROC curve was 0.674, the best cut-off was 17.41 mm/mm2, the sensitivity was 69.2%, and the specificity was 66.7%. After immunosuppressive therapy in 5 patients with immune-related MND syndrome, the LCD was significantly reduced (P < 0.05), and there was no statistically significant change in the IWL (P > 0.05). Conclusion The LC density and IWL are ideal for distinguishing MND from immune-related MND syndrome. The LC density reflects the immunotherapy response sensitively.
Collapse
Affiliation(s)
- Lin Jiao
- Department of Neurology, Peking University Third Hospital, Beijing, China.,Beijing Municipal Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases, Beijing, China
| | - Yuanjin Zhang
- Department of Neurology, Peking University Third Hospital, Beijing, China.,Beijing Municipal Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases, Beijing, China
| | - Haikun Wang
- Department of Ophthalmology, Peking University Third Hospital, Beijing, China
| | - Dongsheng Fan
- Department of Neurology, Peking University Third Hospital, Beijing, China. .,Beijing Municipal Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases, Beijing, China.
| |
Collapse
|
26
|
Impact of Yiqi Huoxue Decoction on the Relationship between Remodeling of Cardiac Nerves and Macrophages after Myocardial Infarction in Rats. JOURNAL OF HEALTHCARE ENGINEERING 2022; 2022:4441603. [PMID: 35432831 PMCID: PMC9010163 DOI: 10.1155/2022/4441603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 03/18/2022] [Indexed: 11/17/2022]
Abstract
Sympathetic nerve remodeling after myocardial infarction (MI) has an indispensable role in cardiac remodeling. Numerous works have shown that sympathetic nerve remodeling can be delayed by inhibition of inflammatory response. Earlier studies have shown improvement in ventricular remodeling and inhibited chronic stage neural remodeling by Yiqi Huoxue decoction (YQHX). Therefore, the current study looked at the inhibitory effect of YQHX prescription on proinflammatory mediators and macrophages and the effect on neural remodeling at 3 and 7 days after MI. YQHX inhibited the expression of Toll-like receptor 4 (TLR4) and nuclear factor kappa B (NF-κB) proteins and macrophage infiltration within 7 days after myocardial infarction. YQHX could decrease Th-positive nerve fiber density in the area around infarction and reduce the expression of growth-associated protein 43 (GAP43), nerve growth factor (NGF), and tyrosine hydroxylase (TH) proteins, which was associated with the remodeling of sympathetic nerves. Thus, the nerve remodeling inhibition after MI due to YQHX may be through its anti-inflammatory action. These data provide direct evidence for the potential application of traditional Chinese medicine (TCM) in the remodeling of sympathetic nerves after MI.
Collapse
|
27
|
Molnár K, Nógrádi B, Kristóf R, Mészáros Á, Pajer K, Siklós L, Nógrádi A, Wilhelm I, Krizbai IA. Motoneuronal inflammasome activation triggers excessive neuroinflammation and impedes regeneration after sciatic nerve injury. J Neuroinflammation 2022; 19:68. [PMID: 35305649 PMCID: PMC8934511 DOI: 10.1186/s12974-022-02427-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 03/01/2022] [Indexed: 12/12/2022] Open
Abstract
Background Peripheral nerve injuries are accompanied by inflammatory reactions, over-activation of which may hinder recovery. Among pro-inflammatory pathways, inflammasomes are one of the most potent, leading to release of active IL-1β. Our aim was to understand how inflammasomes participate in central inflammatory reactions accompanying peripheral nerve injury. Methods After axotomy of the sciatic nerve, priming and activation of the NLRP3 inflammasome was examined in cells of the spinal cord. Regeneration of the nerve was evaluated after coaptation using sciatic functional index measurements and retrograde tracing. Results In the first 3 days after the injury, elements of the NLRP3 inflammasome were markedly upregulated in the L4–L5 segments of the spinal cord, followed by assembly of the inflammasome and secretion of active IL-1β. Although glial cells are traditionally viewed as initiators of neuroinflammation, in this acute phase of inflammation, inflammasome activation was found exclusively in affected motoneurons of the ventral horn in our model. This process was significantly inhibited by 5-BDBD, a P2X4 receptor inhibitor and MCC950, a potent NLRP3 inhibitor. Although at later time points the NLRP3 protein was upregulated in microglia too, no signs of inflammasome activation were detected in these cells. Inhibition of inflammasome activation in motoneurons in the first days after nerve injury hindered development of microgliosis in the spinal cord. Moreover, P2X4 or inflammasome inhibition in the acute phase significantly enhanced nerve regeneration on both the morphological and the functional levels. Conclusions Our results indicate that the central reaction initiated by sciatic nerve injury starts with inflammasome activation in motoneurons of the ventral horn, which triggers a complex inflammatory reaction and activation of microglia. Inhibition of neuronal inflammasome activation not only leads to a significant reduction of microgliosis, but has a beneficial effect on the recovery as well. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02427-9.
Collapse
Affiliation(s)
- Kinga Molnár
- Institute of Biophysics, Biological Research Centre, Eötvös Loránd Research Network (ELKH), Temesvári krt. 62, 6726, Szeged, Hungary
| | - Bernát Nógrádi
- Institute of Biophysics, Biological Research Centre, Eötvös Loránd Research Network (ELKH), Temesvári krt. 62, 6726, Szeged, Hungary.,Theoretical Medicine Doctoral School, University of Szeged, Szeged, Hungary.,Department of Neurology, University of Szeged, Szeged, Hungary
| | - Rebeka Kristóf
- Institute of Biophysics, Biological Research Centre, Eötvös Loránd Research Network (ELKH), Temesvári krt. 62, 6726, Szeged, Hungary.,Theoretical Medicine Doctoral School, University of Szeged, Szeged, Hungary
| | - Ádám Mészáros
- Institute of Biophysics, Biological Research Centre, Eötvös Loránd Research Network (ELKH), Temesvári krt. 62, 6726, Szeged, Hungary.,Doctoral School of Biology, University of Szeged, Szeged, Hungary
| | - Krisztián Pajer
- Department of Anatomy, Histology and Embryology, University of Szeged, Szeged, Hungary
| | - László Siklós
- Institute of Biophysics, Biological Research Centre, Eötvös Loránd Research Network (ELKH), Temesvári krt. 62, 6726, Szeged, Hungary
| | - Antal Nógrádi
- Department of Anatomy, Histology and Embryology, University of Szeged, Szeged, Hungary
| | - Imola Wilhelm
- Institute of Biophysics, Biological Research Centre, Eötvös Loránd Research Network (ELKH), Temesvári krt. 62, 6726, Szeged, Hungary. .,Institute of Life Sciences, Vasile Goldiş Western University of Arad, Arad, Romania.
| | - István A Krizbai
- Institute of Biophysics, Biological Research Centre, Eötvös Loránd Research Network (ELKH), Temesvári krt. 62, 6726, Szeged, Hungary. .,Institute of Life Sciences, Vasile Goldiş Western University of Arad, Arad, Romania.
| |
Collapse
|
28
|
Winter CC, He Z, Jacobi A. Axon Regeneration: A Subcellular Extension in Multiple Dimensions. Cold Spring Harb Perspect Biol 2022; 14:a040923. [PMID: 34518340 PMCID: PMC8886981 DOI: 10.1101/cshperspect.a040923] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Axons are a unique cellular structure that allows for the communication between neurons. Axon damage compromises neuronal communications and often leads to functional deficits. Thus, developing strategies that promote effective axon regeneration for functional restoration is highly desirable. One fruitful approach is to dissect the regenerative mechanisms used by some types of neurons in both mammalian and nonmammalian systems that exhibit spontaneous regenerative capacity. Additionally, numerous efforts have been devoted to deciphering the barriers that prevent successful axon regeneration in the most regeneration-refractory system-the adult mammalian central nervous system. As a result, several regeneration-promoting strategies have been developed, but significant limitations remain. This review is aimed to summarize historic progression and current understanding of this exciting yet incomplete endeavor.
Collapse
Affiliation(s)
- Carla C Winter
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, Massachusetts 02115, USA
- Department of Neurology and Ophthalmology, Harvard Medical School, Boston, Massachusetts 02115, USA
- PhD Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Zhigang He
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, Massachusetts 02115, USA
- Department of Neurology and Ophthalmology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Anne Jacobi
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, Massachusetts 02115, USA
- Department of Neurology and Ophthalmology, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
29
|
Traumatic optic neuropathy: a review of current studies. Neurosurg Rev 2022; 45:1895-1913. [PMID: 35034261 DOI: 10.1007/s10143-021-01717-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 10/26/2021] [Accepted: 12/09/2021] [Indexed: 10/24/2022]
Abstract
Traumatic optic neuropathy (TON) is a serious complication of craniofacial trauma that directly or indirectly damages the optic nerve and can cause severe vision loss. The incidence of TON has been gradually increasing in recent years. Research on the protection and regeneration of the optic nerve after the onset of TON is still at the level of laboratory studies and which is insufficient to support clinical treatment of TON. And, due to without clear guidelines, there is much ambiguity regarding its diagnosis and management. Clinical interventions for TON include observation only, treatment with corticosteroids alone, or optic canal (OC) decompression (with or without steroids). There is controversy in clinical practice concerning which treatment is the best. A review of available studies shows that the visual acuity of patients with TON can be significantly improved after OC decompression surgery (especially endoscopic transnasal/transseptal optic canal decompression (ETOCD)) with or without the use of corticosteroids. And new findings of laboratory studies such as mitochondrial therapy, lipid change studies, and other studies in favor of TON therapy have also been identified. In this review, we discuss the evolving perspective of surgical treatment and experimental study.
Collapse
|
30
|
Blaszkiewicz M, Gunsch G, Willows JW, Gardner ML, Sepeda JA, Sas AR, Townsend KL. Adipose Tissue Myeloid-Lineage Neuroimmune Cells Express Genes Important for Neural Plasticity and Regulate Adipose Innervation. Front Endocrinol (Lausanne) 2022; 13:864925. [PMID: 35795142 PMCID: PMC9251313 DOI: 10.3389/fendo.2022.864925] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 05/05/2022] [Indexed: 12/19/2022] Open
Abstract
Peripheral nerves allow a bidirectional communication between brain and adipose tissues, and many studies have clearly demonstrated that a loss of the adipose nerve supply results in tissue dysfunction and metabolic dysregulation. Neuroimmune cells closely associate with nerves in many tissues, including subcutaneous white adipose tissue (scWAT). However, in scWAT, their functions beyond degrading norepinephrine in an obese state remain largely unexplored. We previously reported that a myeloid-lineage knockout (KO) of brain-derived neurotrophic factor (BDNF) resulted in decreased innervation of scWAT, accompanied by an inability to brown scWAT after cold stimulation, and increased adiposity after a high-fat diet. These data underscored that adipose tissue neuroimmune cells support the peripheral nerve supply to adipose and impact the tissue's metabolic functions. We also reported that a subset of myeloid-lineage monocyte/macrophages (Ly6c+CCR2+Cx3cr1+) is recruited to scWAT in response to cold, a process known to increase neurite density in adipose and promote metabolically healthy processes. These cold-induced neuroimmune cells (CINCs) also expressed BDNF. Here we performed RNAseq on CINCs from cold-exposed and room temperature-housed mice, which revealed a striking and coordinated differential expression of numerous genes involved in neuronal function, including neurotrophin signaling and axonal guidance, further supporting that CINCs fulfill a nerve-supporting role in adipose. The increased expression of leukocyte transendothelial migration genes in cold-stimulated CINCs also confirms prior evidence that they are recruited to scWAT and are not tissue resident. We now provide whole-depot imaging of scWAT from LysM-BDNF KO mice, revealing a striking reduction of innervation across the depot fitting with their reduced energy expenditure phenotype. By contrast, Cx3cr1-BDNF KO mice (a macrophage subset of LysM+ cells) exhibited increased thermogenesis and energy expenditure, with compensatory increased food intake and no change in adiposity or body weight. While these KO mice also exhibit a significantly reduced innervation of scWAT, especially around the subiliac lymph node, they displayed an increase in small fiber sympathetic neurite branching, which may underlie their increased thermogenesis. We propose a homeostatic role of scWAT myeloid-lineage neuroimmune cells together in nerve maintenance and neuro-adipose regulation of energy expenditure.
Collapse
Affiliation(s)
- Magdalena Blaszkiewicz
- Neurobiology and Energy Balance Laboratory, Department of Neurological Surgery, The Ohio State University, Wexner Medical Center, Columbus, OH, United States
| | - Gilian Gunsch
- Neurobiology and Energy Balance Laboratory, Department of Neurological Surgery, The Ohio State University, Wexner Medical Center, Columbus, OH, United States
| | - Jake W. Willows
- Neurobiology and Energy Balance Laboratory, Department of Neurological Surgery, The Ohio State University, Wexner Medical Center, Columbus, OH, United States
| | - Miranda L. Gardner
- Department of Cancer Biology and Genetics, Comprehensive Cancer Center, The Ohio State University, Wexner Medical Center, Columbus, OH, United States
- Campus Chemical Instrument Center, Mass Spectrometry and Proteomics Facility, The Ohio State University, Columbus, OH, United States
| | - Jesse A. Sepeda
- Department of Neurology, The Ohio State University, Wexner Medical Center, Columbus, OH, United States
| | - Andrew R. Sas
- Department of Neurology, The Ohio State University, Wexner Medical Center, Columbus, OH, United States
| | - Kristy L. Townsend
- Neurobiology and Energy Balance Laboratory, Department of Neurological Surgery, The Ohio State University, Wexner Medical Center, Columbus, OH, United States
- *Correspondence: Kristy L. Townsend,
| |
Collapse
|
31
|
The Conditioning Lesion Response in Dorsal Root Ganglion Neurons Is Inhibited in Oncomodulin Knock-Out Mice. eNeuro 2022; 9:ENEURO.0477-21.2022. [PMID: 35131866 PMCID: PMC8874952 DOI: 10.1523/eneuro.0477-21.2022] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 01/20/2022] [Accepted: 01/30/2022] [Indexed: 11/21/2022] Open
Abstract
Regeneration can occur in peripheral neurons after injury, but the mechanisms involved are not fully delineated. Macrophages in dorsal root ganglia (DRGs) are involved in the enhanced regeneration that occurs after a conditioning lesion (CL), but how macrophages stimulate this response is not known. Oncomodulin (Ocm) has been proposed as a proregenerative molecule secreted by macrophages and neutrophils, is expressed in the DRG after axotomy, and stimulates neurite outgrowth by DRG neurons in culture. Wild-type (WT) and Ocm knock-out (KO) mice were used to investigate whether Ocm plays a role in the CL response in DRG neurons after sciatic nerve transection. Neurite outgrowth was measured after 24 and 48 h in explant culture 7 d after a CL. Sciatic nerve regeneration was also measured in vivo 7 d after a CL and 2 d after a subsequent sciatic nerve crush. The magnitude of the increased neurite outgrowth following a CL was significantly smaller in explants from Ocm KO mice than in explants from WT mice. In vivo after a CL, increased regeneration was found in WT animals but not in KO animals. Macrophage accumulation and levels of interleukin-6 (IL-6) mRNA were measured in axotomized DRG from WT and Ocm KO animals, and both were significantly higher than in sham-operated ganglia. At 6 h after axotomy, Il-6 mRNA was higher in WT than in Ocm KO mice. Our data support the hypothesis that Ocm plays a necessary role in producing a normal CL response and that its effects possibly result in part from stimulation of the expression of proregenerative macrophage cytokines such as IL-6.
Collapse
|
32
|
Neural Stem Cells: Promoting Axonal Regeneration and Spinal Cord Connectivity. Cells 2021; 10:cells10123296. [PMID: 34943804 PMCID: PMC8699545 DOI: 10.3390/cells10123296] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 11/16/2021] [Accepted: 11/16/2021] [Indexed: 11/17/2022] Open
Abstract
Spinal cord injury (SCI) leads to irreversible functional impairment caused by neuronal loss and the disruption of neuronal connections across the injury site. While several experimental strategies have been used to minimize tissue damage and to enhance axonal growth and regeneration, the corticospinal projection, which is the most important voluntary motor system in humans, remains largely refractory to regenerative therapeutic interventions. To date, one of the most promising pre-clinical therapeutic strategies has been neural stem cell (NSC) therapy for SCI. Over the last decade we have found that host axons regenerate into spinal NSC grafts placed into sites of SCI. These regenerating axons form synapses with the graft, and the graft in turn extends very large numbers of new axons from the injury site over long distances into the distal spinal cord. Here we discuss the pathophysiology of SCI that makes the spinal cord refractory to spontaneous regeneration, the most recent findings of neural stem cell therapy for SCI, how it has impacted motor systems including the corticospinal tract and the implications for sensory feedback.
Collapse
|
33
|
Du YL, Sergeeva EG, Stein DG. Visual recovery following optic nerve crush in male and female wild-type and TRIF-deficient mice. Restor Neurol Neurosci 2021; 38:355-368. [PMID: 32986632 DOI: 10.3233/rnn-201019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND There is growing evidence that the TIR-domain-containing adapter-inducing interferon-β (TRIF) pathway is implicated in the modulation of neuroinflammation following injuries to the brain and retina. After exposure to injury or to excitotoxic pathogens, toll-like receptors (TLR) activate the innate immune system signaling cascade and stimulate the release of inflammatory cytokines. Inhibition of the TLR4 receptor has been shown to enhance retinal ganglion cell (RGC) survival in optic nerve crush (ONC) and in ischemic injury to other parts of the brain. OBJECTIVE Based on this evidence, we tested the hypothesis that mice with the TRIF gene knocked out (TKO) will demonstrate decreased inflammatory responses and greater functional recovery after ONC. METHODS Four experimental groups -TKO ONC (12 males and 8 females), WT ONC (10 males and 8 females), TKO sham (9 males and 5 females), and WT sham (7 males and 5 females) -were used as subjects. Visual evoked potentials (VEP) were recorded in the left and right primary visual cortices and optomotor response were assessed in all mice at 14, 30, and 80 days after ONC. GFAP and Iba-1 were used as markers for astrocytes and microglial cells respectively at 7 days after ONC, along with NF-kB to measure inflammatory effects downstream of TRIF activation; RMPBS marker was used to visualize RGC survival and GAP-43 was used as a marker of regenerating optic nerve axons at 30 days after ONC. RESULTS We found reduced inflammatory response in the retina at 7 days post-ONC, less RGC loss and greater axonal regeneration 30 days post-ONC, and better recovery of visual function 80 days post-ONC in TKO mice compared to WT mice. CONCLUSIONS Our study showed that the TRIF pathway is involved in post-ONC inflammatory response and gliosis and that deletion of TRIF induces better RGC survival and regeneration and better functional recovery in mice. Our results suggest the TRIF pathway as a potential therapeutic target for reducing the inflammatory damage caused by nervous system injury.
Collapse
Affiliation(s)
- Yimeng Lina Du
- Emory University College of Arts and Sciences, Neuroscience and Behavioral Biology Program, GA, USA
| | - Elena G Sergeeva
- Department of Emergency Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Donald G Stein
- Emory University College of Arts and Sciences, Neuroscience and Behavioral Biology Program, GA, USA.,Department of Emergency Medicine, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
34
|
Alesci A, Pergolizzi S, Lo Cascio P, Fumia A, Lauriano ER. Neuronal regeneration: Vertebrates comparative overview and new perspectives for neurodegenerative diseases. ACTA ZOOL-STOCKHOLM 2021. [DOI: 10.1111/azo.12397] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Alessio Alesci
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences University of Messina Messina Italy
| | - Simona Pergolizzi
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences University of Messina Messina Italy
| | - Patrizia Lo Cascio
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences University of Messina Messina Italy
| | - Angelo Fumia
- Department of Clinical and Experimental Medicine University of Messina Messina Italy
| | - Eugenia Rita Lauriano
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences University of Messina Messina Italy
| |
Collapse
|
35
|
Basu A, Behera S, Bhardwaj S, Dey S, Ghosh-Roy A. Regulation of UNC-40/DCC and UNC-6/Netrin by DAF-16 promotes functional rewiring of the injured axon. Development 2021; 148:268990. [PMID: 34109380 DOI: 10.1242/dev.198044] [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: 10/26/2020] [Accepted: 05/04/2021] [Indexed: 01/02/2023]
Abstract
The adult nervous system has a limited capacity to regenerate after accidental damage. Post-injury functional restoration requires proper targeting of the injured axon to its postsynaptic cell. Although the initial response to axonal injury has been studied in great detail, it is rather unclear what controls the re-establishment of a functional connection. Using the posterior lateral microtubule neuron in Caenorhabditis elegans, we found that after axotomy, the regrowth from the proximal stump towards the ventral side and accumulation of presynaptic machinery along the ventral nerve cord correlated to the functional recovery. We found that the loss of insulin receptor DAF-2 promoted 'ventral targeting' in a DAF-16-dependent manner. We further showed that coordinated activities of DAF-16 in neuron and muscle promoted 'ventral targeting'. In response to axotomy, expression of the Netrin receptor UNC-40 was upregulated in the injured neuron in a DAF-16-dependent manner. In contrast, the DAF-2-DAF-16 axis contributed to the age-related decline in Netrin expression in muscle. Therefore, our study revealed an important role for insulin signaling in regulating the axon guidance molecules during the functional rewiring process.
Collapse
Affiliation(s)
- Atrayee Basu
- Department of Cellular and Molecular Neuroscience, National Brain Research Centre, Manesar, Nainwal Mode, Gurgaon, Haryana 122051, India
| | - Sibaram Behera
- Department of Cellular and Molecular Neuroscience, National Brain Research Centre, Manesar, Nainwal Mode, Gurgaon, Haryana 122051, India
| | - Smriti Bhardwaj
- Department of Cellular and Molecular Neuroscience, National Brain Research Centre, Manesar, Nainwal Mode, Gurgaon, Haryana 122051, India
| | - Shirshendu Dey
- Fluorescence Microscopy Division, Bruker India Scientific PvT Ltd, International Trade Tower, Nehru Place, New Delhi 110019, India
| | - Anindya Ghosh-Roy
- Department of Cellular and Molecular Neuroscience, National Brain Research Centre, Manesar, Nainwal Mode, Gurgaon, Haryana 122051, India
| |
Collapse
|
36
|
Echternacht SR, Chacon MA, Leckenby JI. Central versus peripheral nervous system regeneration: is there an exception for cranial nerves? Regen Med 2021; 16:567-579. [PMID: 34075805 DOI: 10.2217/rme-2020-0096] [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] [Indexed: 01/08/2023] Open
Abstract
There exists a dichotomy in regenerative capacity between the PNS and CNS, which poses the question - where do cranial nerves fall? Through the discussion of the various cells and processes involved in axonal regeneration, we will evaluate whether the assumption that cranial nerve regeneration is analogous to peripheral nerve regeneration is valid. It is evident from this review that much remains to be clarified regarding both PNS and CNS regeneration. Furthermore, it is not clear if cranial nerves follow the PNS model, CNS model or possess an alternative novel regenerative process altogether. Future research should continue to focus on elucidating how cranial nerves regenerate; and the various cellular interactions, molecules and pathways involved.
Collapse
Affiliation(s)
- Scott R Echternacht
- University of Rochester School of Medicine & Dentistry, 601 Elmwood Avenue, Rochester, NY 14642, USA.,Division of Plastic Surgery, University of Rochester Medical Center, 601 Elmwood Avenue, Box 661, Rochester, NY 14642, USA
| | - Miranda A Chacon
- Division of Plastic Surgery, University of Rochester Medical Center, 601 Elmwood Avenue, Box 661, Rochester, NY 14642, USA.,Department of Surgery, University of Rochester Medical Center, 601 Elmwood Avenue, Box 661, Rochester, NY 14642, USA
| | - Jonathan I Leckenby
- Division of Plastic Surgery, University of Rochester Medical Center, 601 Elmwood Avenue, Box 661, Rochester, NY 14642, USA
| |
Collapse
|
37
|
Capsazepine decreases corneal pain syndrome in severe dry eye disease. J Neuroinflammation 2021; 18:111. [PMID: 33975636 PMCID: PMC8114509 DOI: 10.1186/s12974-021-02162-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 04/29/2021] [Indexed: 11/26/2022] Open
Abstract
Background Dry eye disease (DED) is a multifactorial disease of the ocular surface accompanied by neurosensory abnormalities. Here, we evaluated the effectiveness of transient receptor potential vanilloid-1 (TRPV1) blockade to alleviate ocular pain, neuroinflammation, and anxiety-like behavior associated with severe DED. Methods Chronic DED was induced by unilateral excision of the Harderian and extraorbital lacrimal glands of adult male mice. Investigations were conducted at 21 days after surgery. The mRNA levels of TRPV1, transient receptor potential ankyrin-1 (TRPA1), and acid-sensing ion channels 1 and 3 (ASIC1 and ASIC3) in the trigeminal ganglion (TG) were evaluated by RNAscope in situ hybridization. Multi-unit extracellular recording of ciliary nerve fiber activity was used to monitor spontaneous and stimulated (cold, heat, and acid) corneal nerve responsiveness in ex vivo eye preparations. DED mice received topical instillations of the TRPV1 antagonist (capsazepine) twice a day for 2 weeks from d7 to d21 after surgery. The expression of genes involved in neuropathic and inflammatory pain was evaluated in the TG using a global genomic approach. Chemical and mechanical corneal nociception and spontaneous ocular pain were monitored. Finally, anxiety-like behaviors were assessed by elevated plus maze and black and white box tests. Results First, in situ hybridization showed DED to trigger upregulation of TRPV1, TRPA1, ASIC1, and ASIC3 mRNA in the ophthalmic branch of the TG. DED also induced overexpression of genes involved in neuropathic and inflammatory pain in the TG. Repeated instillations of capsazepine reduced corneal polymodal responsiveness to heat, cold, and acidic stimulation in ex vivo eye preparations. Consistent with these findings, chronic capsazepine instillation inhibited the upregulation of genes involved in neuropathic and inflammatory pain in the TG of DED animals and reduced the sensation of ocular pain, as well as anxiety-like behaviors associated with severe DED. Conclusion These data provide novel insights on the effectiveness of TRPV1 antagonist instillation in alleviating abnormal corneal neurosensory symptoms induced by severe DED, opening an avenue for the repositioning of this molecule as a potential analgesic treatment for patients suffering from chronic DED.
Collapse
|
38
|
Singh RK, Binette F, Seiler M, Petersen-Jones SM, Nasonkin IO. Pluripotent Stem Cell-Based Organoid Technologies for Developing Next-Generation Vision Restoration Therapies of Blindness. J Ocul Pharmacol Ther 2021; 37:147-156. [PMID: 33052761 PMCID: PMC8060716 DOI: 10.1089/jop.2020.0016] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 07/28/2020] [Indexed: 12/25/2022] Open
Abstract
Blindness, associated with death of retinal cells at the back of the eye, is caused by a number of conditions with high prevalence such as glaucoma, age-related macular degeneration, and diabetic retinopathy. In addition, a large number of orphan inherited (mostly monogenic) conditions, such as retinitis pigmentosa and Leber Congenital Amaurosis, add to the overall number of patients with blinding retinal degenerative diseases. Blindness caused by deterioration and loss of retina is so far incurable. Modern biomedical research leveraging molecular and regenerative medicine approaches had a number of groundbreaking discoveries and proof-of-principle treatments of blindness in animals. However, these methods are slow to be standardized and commercialized as therapies to benefit people losing their eyesight due to retinal degenerative conditions. In this review, we will outline major regenerative medicine approaches, which are emerging as promising for preserving or/and restoring vision. We discuss the potential of each of these approaches to reach commercialization step and be converted to treatments, which could at least ameliorate blindness caused by retinal cell death.
Collapse
Affiliation(s)
| | | | - Magdalene Seiler
- Stem Cell Research Center, University of California, Irvine, Irvine, California, USA
- Department of Physical Medicine & Rehabilitation, University of California, Irvine, Irvine, California, USA
- Department of Ophthalmology, University of California, Irvine, Irvine, California, USA
- Department of Anatomy & Neurobiology, University of California, Irvine, Irvine, California, USA
| | - Simon M. Petersen-Jones
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, Michigan, USA
| | | |
Collapse
|
39
|
Van Dyck A, Bollaerts I, Beckers A, Vanhunsel S, Glorian N, van Houcke J, van Ham TJ, De Groef L, Andries L, Moons L. Müller glia-myeloid cell crosstalk accelerates optic nerve regeneration in the adult zebrafish. Glia 2021; 69:1444-1463. [PMID: 33502042 DOI: 10.1002/glia.23972] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 01/12/2021] [Accepted: 01/15/2021] [Indexed: 12/18/2022]
Abstract
Neurodegenerative disorders, characterized by progressive neuronal loss, eventually lead to functional impairment in the adult mammalian central nervous system (CNS). Importantly, these deteriorations are irreversible, due to the very limited regenerative potential of these CNS neurons. Stimulating and redirecting neuroinflammation was recently put forward as an important approach to induce axonal regeneration, but it remains elusive how inflammatory processes and CNS repair are intertwined. To gain more insight into these interactions, we investigated how immunomodulation affects the regenerative outcome after optic nerve crush (ONC) in the spontaneously regenerating zebrafish. First, inducing intraocular inflammation using zymosan resulted in an acute inflammatory response, characterized by an increased infiltration and proliferation of innate blood-borne immune cells, reactivation of Müller glia, and altered retinal cytokine expression. Strikingly, inflammatory stimulation also accelerated axonal regrowth after optic nerve injury. Second, we demonstrated that acute depletion of both microglia and macrophages in the retina, using pharmacological treatments with both the CSF1R inhibitor PLX3397 and clodronate liposomes, compromised optic nerve regeneration. Moreover, we observed that csf1ra/b double mutant fish, lacking microglia in both retina and brain, displayed accelerated RGC axonal regrowth after ONC, which was accompanied with unusual Müller glia proliferative gliosis. Altogether, our results highlight the importance of altered glial cell interactions in the axonal regeneration process after ONC in adult zebrafish. Unraveling the relative contribution of the different cell types, as well as the signaling pathways involved, may pinpoint new targets to stimulate repair in the vertebrate CNS.
Collapse
Affiliation(s)
- Annelies Van Dyck
- Neural Circuit Development and Regeneration Research Group, Department of Biology, KU Leuven, Leuven, Belgium
| | - Ilse Bollaerts
- Neural Circuit Development and Regeneration Research Group, Department of Biology, KU Leuven, Leuven, Belgium
| | - An Beckers
- Neural Circuit Development and Regeneration Research Group, Department of Biology, KU Leuven, Leuven, Belgium
| | - Sophie Vanhunsel
- Neural Circuit Development and Regeneration Research Group, Department of Biology, KU Leuven, Leuven, Belgium
| | - Nynke Glorian
- Neural Circuit Development and Regeneration Research Group, Department of Biology, KU Leuven, Leuven, Belgium
| | - Jessie van Houcke
- Neural Circuit Development and Regeneration Research Group, Department of Biology, KU Leuven, Leuven, Belgium
| | - Tjakko J van Ham
- Department of Clinical Genetics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Lies De Groef
- Neural Circuit Development and Regeneration Research Group, Department of Biology, KU Leuven, Leuven, Belgium.,Leuven Brain Institute (LBI), KU Leuven, Leuven, Belgium
| | - Lien Andries
- Neural Circuit Development and Regeneration Research Group, Department of Biology, KU Leuven, Leuven, Belgium
| | - Lieve Moons
- Neural Circuit Development and Regeneration Research Group, Department of Biology, KU Leuven, Leuven, Belgium.,Leuven Brain Institute (LBI), KU Leuven, Leuven, Belgium
| |
Collapse
|
40
|
da Silva-Junior AJ, Mesentier-Louro LA, Nascimento-Dos-Santos G, Teixeira-Pinheiro LC, Vasques JF, Chimeli-Ormonde L, Bodart-Santos V, de Carvalho LRP, Santiago MF, Mendez-Otero R. Human mesenchymal stem cell therapy promotes retinal ganglion cell survival and target reconnection after optic nerve crush in adult rats. Stem Cell Res Ther 2021; 12:69. [PMID: 33468246 PMCID: PMC7814601 DOI: 10.1186/s13287-020-02130-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Accepted: 12/28/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Optic-nerve injury results in impaired transmission of visual signals to central targets and leads to the death of retinal ganglion cells (RGCs) and irreversible vision loss. Therapies with mesenchymal stem cells (MSCs) from different sources have been used experimentally to increase survival and regeneration of RGCs. METHODS We investigated the efficacy of human umbilical Wharton's jelly-derived MSCs (hWJ-MSCs) and their extracellular vesicles (EVs) in a rat model of optic nerve crush. RESULTS hWJ-MSCs had a sustained neuroprotective effect on RGCs for 14, 60, and 120 days after optic nerve crush. The same effect was obtained using serum-deprived hWJ-MSCs, whereas transplantation of EVs obtained from those cells was ineffective. Treatment with hWJ-MSCs also promoted axonal regeneration along the optic nerve and reinnervation of visual targets 120 days after crush. CONCLUSIONS The observations showed that this treatment with human-derived MSCs promoted sustained neuroprotection and regeneration of RGCs after optic nerve injury. These findings highlight the possibility to use cell therapy to preserve neurons and to promote axon regeneration, using a reliable source of human MSCs.
Collapse
Affiliation(s)
- Almir Jordão da Silva-Junior
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil. .,Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa-REGENERE, Rio de Janeiro, RJ, Brazil. .,Rede NanoSaúde, Rio de Janeiro, RJ, Brazil.
| | - Louise Alessandra Mesentier-Louro
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil.,Department of Ophthalmology, Stanford University, Palo Alto, CA, USA
| | - Gabriel Nascimento-Dos-Santos
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil.,Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa-REGENERE, Rio de Janeiro, RJ, Brazil
| | - Leandro Coelho Teixeira-Pinheiro
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil.,Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa-REGENERE, Rio de Janeiro, RJ, Brazil
| | - Juliana F Vasques
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil.,Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa-REGENERE, Rio de Janeiro, RJ, Brazil
| | - Luiza Chimeli-Ormonde
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil.,Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa-REGENERE, Rio de Janeiro, RJ, Brazil
| | - Victor Bodart-Santos
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil.,Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa-REGENERE, Rio de Janeiro, RJ, Brazil
| | - Luiza Rachel Pinheiro de Carvalho
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil.,Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa-REGENERE, Rio de Janeiro, RJ, Brazil
| | - Marcelo Felippe Santiago
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil.,Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa-REGENERE, Rio de Janeiro, RJ, Brazil
| | - Rosalia Mendez-Otero
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil.,Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa-REGENERE, Rio de Janeiro, RJ, Brazil.,Rede NanoSaúde, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
41
|
Makker PGS, Keating BA, Lees JG, Burke D, Howells J, Moalem-Taylor G. Electrophysiological investigation of motor axonal excitability in a mouse model of nerve constriction injury. J Peripher Nerv Syst 2021; 26:99-112. [PMID: 33432642 DOI: 10.1111/jns.12430] [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: 11/09/2020] [Revised: 12/19/2020] [Accepted: 12/30/2020] [Indexed: 11/29/2022]
Abstract
Peripheral nerve injuries caused by focal constriction are characterised by local nerve ischaemia, axonal degeneration, demyelination, and neuroinflammation. The aim of this study was to understand temporal changes in the excitability properties of injured motor axons in a mouse model of nerve constriction injury (NCI). The excitability of motor axons following unilateral sciatic NCI was studied in male C57BL/6J mice distal to the site of injury at the acute (6 hours-1 week) and chronic (up to 20 weeks) phases of injury, using threshold tracking. Multiple measures of nerve excitability, including strength-duration properties, threshold electrotonus, current-threshold relationship, and recovery cycle were examined using the automated nerve excitability protocol (TRONDNF). Acutely, injured motor axons developed a pattern of excitability characteristic of ischemic depolarisation. In most cases, the sciatic nerve became transiently inexcitable. When a liminal compound muscle action potential could again be recorded, it had an increase in threshold and latency, compared to both pre-injury baseline and sham-injured groups. These axons showed a greater threshold change in response to hyperpolarising threshold electrotonus and a significant upward shift in the recovery cycle. Mathematical modelling suggested that the changes seen in chronically injured axons involve shortened internodes, reduced myelination, and exposed juxtaparanodal fast K+ conductances. The findings of this study demonstrate long-term changes in motor excitability following NCI (involving alterations in axonal properties and ion channel activity) and are important for understanding the mechanisms of neurapraxic injuries and traumatic mononeuropathies.
Collapse
Affiliation(s)
- Preet G S Makker
- Translational Neuroscience Facility, School of Medical Sciences, The University of New South Wales (UNSW), Sydney, New South Wales, Australia
| | - Brooke A Keating
- Translational Neuroscience Facility, School of Medical Sciences, The University of New South Wales (UNSW), Sydney, New South Wales, Australia
| | - Justin G Lees
- Translational Neuroscience Facility, School of Medical Sciences, The University of New South Wales (UNSW), Sydney, New South Wales, Australia
| | - David Burke
- Central Clinical School, Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - James Howells
- Central Clinical School, Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Gila Moalem-Taylor
- Translational Neuroscience Facility, School of Medical Sciences, The University of New South Wales (UNSW), Sydney, New South Wales, Australia
| |
Collapse
|
42
|
Lopez-Silva TL, Cristobal CD, Edwin Lai CS, Leyva-Aranda V, Lee HK, Hartgerink JD. Self-assembling multidomain peptide hydrogels accelerate peripheral nerve regeneration after crush injury. Biomaterials 2021; 265:120401. [PMID: 33002786 PMCID: PMC7669633 DOI: 10.1016/j.biomaterials.2020.120401] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 09/12/2020] [Accepted: 09/17/2020] [Indexed: 12/24/2022]
Abstract
Multidomain peptide (MDP) hydrogels are a class of self-assembling materials that have been shown to elicit beneficial responses for soft tissue regeneration. However, their capacity to promote nervous system regeneration remains unknown. The peripheral nervous system (PNS) substantially recovers after injury, partly due to the abundance of extracellular matrix (ECM) components in its basal lamina. However, severe peripheral nerve injuries that significantly damage the ECM continue to be a major clinical challenge as they occur at a high rate and can be extremely detrimental to patients' quality of life. In this study, a panel of eight MDPs were designed to contain various motifs mimicking extracellular matrix components and growth factors and successfully self-assembled into injectable, nanofibrous hydrogels. Using an in vitro screening system, various lysine based MDPs were found to enhance neurite outgrowth. To test their capacity to promote nerve regeneration in vivo, rat sciatic nerve crush injury was performed with MDP hydrogels injected directly into the injury sites. MDP hydrogels were found to enhance macrophage recruitment to the injury site and degrade efficiently over time. Rats that were injected with the MDP hydrogel K2 and laminin motif-containing MDPs K2-IIKDI and K2-IKVAV were found to have significantly accelerated functional recovery and remyelination compared to those injected with HBSS or other MDPs. These results demonstrate that MDPs enhance neurite outgrowth and promote a multicellular pro-regenerative response in peripheral nerve injury. This study provides important insights into the potential of MDPs as biomaterials for nerve regeneration and other clinical applications.
Collapse
Affiliation(s)
- Tania L Lopez-Silva
- Department of Chemistry and Bioengineering, Rice University, Houston, TX, 77005, USA
| | - Carlo D Cristobal
- Integrative Program in Molecular and Biomedical Sciences, Baylor College of Medicine, Houston, TX, 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, 77030, USA
| | - Cheuk Sun Edwin Lai
- Department of Chemistry and Bioengineering, Rice University, Houston, TX, 77005, USA
| | | | - Hyun Kyoung Lee
- Integrative Program in Molecular and Biomedical Sciences, Baylor College of Medicine, Houston, TX, 77030, USA; Department of Pediatrics-Neurology, Baylor College of Medicine, Houston, TX, 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, 77030, USA.
| | - Jeffrey D Hartgerink
- Department of Chemistry and Bioengineering, Rice University, Houston, TX, 77005, USA.
| |
Collapse
|
43
|
da Costa A, Metais T, Mouthon F, Kerkovich D, Charvériat M. Evaluating and modulating TFEB in the control of autophagy: toward new treatments in CNS disorders. Fundam Clin Pharmacol 2020; 35:539-551. [PMID: 33259088 DOI: 10.1111/fcp.12634] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 10/15/2020] [Accepted: 11/27/2020] [Indexed: 12/11/2022]
Abstract
TFEB is a mammalian transcription factor that binds directly to the CLEAR consensus sequence (5'-GTCACGTGAC-3') present in the regulatory regions of genes inducing autophagosome formation, autophagosome-lysosome fusion, hydrolase enzyme expression, and lysosomal exocytosis. By modulating these activities, TFEB coordinates on-demand control over each cell's degradation pathway. Thus, a nuclear signaling pathway regulates cellular energy metabolism through TFEB. Our growing understanding of the role of TFEB and CLEAR in the promotion of healthy clearance together with in vitro and in vivo preclinical findings in various animal models of disease supports the conclusion that the pharmacological activation of TFEB could clear toxic proteins to treat both rare and common forms of neurodegenerative disease.
Collapse
|
44
|
Zhang F, Miao Y, Liu Q, Li S, He J. Changes of pro-inflammatory and anti-inflammatory macrophages after peripheral nerve injury. RSC Adv 2020; 10:38767-38773. [PMID: 35518415 PMCID: PMC9057386 DOI: 10.1039/d0ra06607a] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 10/01/2020] [Indexed: 12/12/2022] Open
Abstract
Macrophages are notable immune cells that are recruited to the injury sites after peripheral nerve injury. Following peripheral nerve injury, increasing numbers of macrophages engulf debris and promote nerve regeneration. However, changes of pro-inflammatory (M1) and anti-inflammatory (M2) macrophages, two types of macrophages with dissimilar biological functions, have not been discovered. In the current study, the expression profiles of M1 and M2 macrophage marker genes in the sciatic nerve stumps and dorsal root ganglions (DRGs) after rat sciatic nerve injury were determined using RNA sequencing. Robust up-regulation of macrophage marker genes was observed in the injured sciatic nerve stumps as compared with in the DRGs. Measurement of the dynamic expression levels of M1 macrophage specific marker genes CD38 and Gpr18 as well as M2 macrophage specific marker genes Egr2 and Myc suggested that M1 macrophages were highly involved at all tested time points after peripheral nerve injury while M2 macrophage might be more involved in the later phase after nerve injury. Dynamic changes of M1 macrophage-inducing miRNAs showed that miR-18a, miR-19b, miR-21, miR-29a, and miR-29b were elevated in the injured nerve stump. These up-regulated miRNAs might mediate macrophage polarization by targeting multiple genes, such as Pten. Collectively, our study explored the unique temporal patterns of pro-inflammatory and anti-inflammatory macrophages after peripheral nerve injury for genetic aspects and provided a deeper understanding of the cellular and molecular basis of microenvironment reconstruction after peripheral nerve injury. The temporal patterns of pro-inflammatory and anti-inflammatory macrophages after peripheral nerve injury.![]()
Collapse
Affiliation(s)
- Fuchao Zhang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University Nantong Jiangsu 226001 China
| | - Yang Miao
- Department of Pharmacy, Yancheng City No. 1 Peoples' Hospital Yancheng China
| | - Qianyan Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University Nantong Jiangsu 226001 China
| | - Shiying Li
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University Nantong Jiangsu 226001 China
| | - Jianghong He
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University Nantong Jiangsu 226001 China
| |
Collapse
|
45
|
TGFβ1 Induces Axonal Outgrowth via ALK5/PKA/SMURF1-Mediated Degradation of RhoA and Stabilization of PAR6. eNeuro 2020; 7:ENEURO.0104-20.2020. [PMID: 32887692 PMCID: PMC7540929 DOI: 10.1523/eneuro.0104-20.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 07/21/2020] [Accepted: 08/06/2020] [Indexed: 12/11/2022] Open
Abstract
Transforming growth factor (TGF)β1 has repeatedly been associated with axonal regeneration and recovery after injury to the CNS. We found TGFβ1 upregulated in the stroke-denervated mouse spinal cord after ischemic injury to the motor cortex as early as 4 d postinjury (dpi) and persisting up to 28 dpi. Given the potential role of TGFβ1 in structural plasticity and functional recovery after stroke highlighted in several published studies, we investigated its downstream signaling in an in vitro model of neurite outgrowth. We found that in this model, TGFβ1 rescues neurite outgrowth under growth inhibitory conditions via the canonical TGFβR2/ALK5 signaling axis. Thereby, protein kinase A (PKA)-mediated phosphorylation of the E3 ubiquitin ligase SMURF1 induces a switch of its substrate preference from PAR6 to the Ras homolog A (RhoA), in this way enhancing outgrowth on the level of the cytoskeleton. This proposed mechanism of TGFβ1 signaling could underly the observed increase in structural plasticity after stroke in vivo as suggested by the temporal and spatial expression of TGFβ1. In accordance with previous publications, this study corroborates the potential of TGFβ1 and associated signaling cascades as a target for future therapeutic interventions to enhance structural plasticity and functional recovery for stroke patients.
Collapse
|
46
|
The Effects of a Ketogenic Diet on Sensorimotor Function in a Thoracolumbar Mouse Spinal Cord Injury Model. eNeuro 2020; 7:ENEURO.0178-20.2020. [PMID: 32680835 PMCID: PMC7433893 DOI: 10.1523/eneuro.0178-20.2020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 06/14/2020] [Accepted: 07/10/2020] [Indexed: 01/26/2023] Open
Abstract
Spinal cord injury and peripheral nerve injuries are traumatic events that greatly impact quality of life. One factor that is being explored throughout patient care is the idea of diet and the role it has on patient outcomes. But the effects of diet following neurotrauma need to be carefully explored in animal models to ensure that they have beneficial effects. The ketogenic diet provides sufficient daily caloric requirements while being potentially neuroprotective and analgesic. In this study, animals were fed a high-fat, low-carbohydrate diet that led to a high concentration of blood ketone that was sustained for as long as the animals were on the diet. Mice fed a ketogenic diet had significantly lower levels of tyrosine and tryptophan, but the levels of other monoamines within the spinal cord remained similar to those of control mice. Mice were fed a standard or ketogenic diet for 7 d before and 28 d following the injury. Our results show that mice hemisected over the T10–T11 vertebrae showed no beneficial effects of being on a ketogenic diet over a 28 d recovery period. Similarly, ligation of the common peroneal and tibial nerve showed no differences between mice fed normal or ketogenic diets. Tests included von Frey, open field, and ladder-rung crossing. We add to existing literature showing protective effects of the ketogenic diet in forelimb injuries by focusing on neurotrauma in the hindlimbs. The results suggest that ketogenic diets need to be assessed based on the type and location of neurotrauma.
Collapse
|
47
|
Zhao J, Su M, Lin Y, Liu H, He Z, Lai L. Administration of Amyloid Precursor Protein Gene Deleted Mouse ESC-Derived Thymic Epithelial Progenitors Attenuates Alzheimer's Pathology. Front Immunol 2020; 11:1781. [PMID: 32849642 PMCID: PMC7431620 DOI: 10.3389/fimmu.2020.01781] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 07/03/2020] [Indexed: 01/07/2023] Open
Abstract
Alzheimer's disease (AD) is a devastating neurodegenerative disorder and the most common cause of dementia in older adults. Although amyloid-beta (Aβ) plaque deposition and chronic neuroinflammation in the central nervous system (CNS) contribute to AD pathology, neither Aβ plaque removal nor anti-inflammatory therapy has shown much clinical success, suggesting that the combinational therapies for the disease-causative factors may be needed for amelioration. Recent data also suggest that systemic immunity in AD should be boosted, rather than suppressed, to drive an immune-dependent cascade needed for Aβ clearance and brain repair. Thymic epithelial cells (TECs) not only play a critical role in supporting T cell development but also mediate the deletion of autoreactive T cells by expressing autoantigens. We have reported that embryonic stem cells (ESCs) can be selectively induced to differentiate into thymic epithelial progenitors (TEPs) in vitro that further develop into TECs in vivo to support T cell development. We show here that transplantation of mouse ESC (mESC)-TEPs into AD mice reduced cerebral Aβ plaque load and improved cognitive performance, in correlation with an increased number of T cells, enhanced choroid plexus (CP) gateway activity, and increased number of macrophages in the brain. Furthermore, transplantation of the amyloid precursor protein (APP) gene deleted mESC-TEPs (APP-/-) results in more effective reduction of AD pathology as compared to wild-type (APP+/+) mESC-TEPs. This is associated with the generation of Aβ-specific T cells, which leads to an increase of anti-Aβ antibody (Ab)-producing B cells in the spleen and enhanced levels of anti-Aβ antibodies in the serum, as well as an increase of Aβ phagocytosing macrophages in the CNS. Our results suggest that transplantation of APP-/- human ESC- or induced pluripotent stem cell (iPSC)-derived TEPs may provide a new tool to mitigate AD in patients.
Collapse
Affiliation(s)
- Jin Zhao
- Guizhou Provincial Key Laboratory for Regenerative Medicine, Tissue Engineering and Stem Cell Research Center, Department of Immunology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China.,Key Laboratory of Adult Stem Cell Translational Research, Chinese Academy of Medical Sciences, Guiyang, China
| | - Min Su
- Department of Allied Health Sciences, University of Connecticut, Storrs, CT, United States
| | - Yujun Lin
- Department of Allied Health Sciences, University of Connecticut, Storrs, CT, United States
| | - Haiyan Liu
- Department of Allied Health Sciences, University of Connecticut, Storrs, CT, United States
| | - Zhixu He
- Key Laboratory of Adult Stem Cell Translational Research, Chinese Academy of Medical Sciences, Guiyang, China.,Department of Pediatrics, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Laijun Lai
- Department of Allied Health Sciences, University of Connecticut, Storrs, CT, United States.,University of Connecticut Stem Cell Institute, University of Connecticut, Storrs, CT, United States
| |
Collapse
|
48
|
Belrose JL, Prasad A, Sammons MA, Gibbs KM, Szaro BG. Comparative gene expression profiling between optic nerve and spinal cord injury in Xenopus laevis reveals a core set of genes inherent in successful regeneration of vertebrate central nervous system axons. BMC Genomics 2020; 21:540. [PMID: 32758133 PMCID: PMC7430912 DOI: 10.1186/s12864-020-06954-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 07/27/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND The South African claw-toed frog, Xenopus laevis, is uniquely suited for studying differences between regenerative and non-regenerative responses to CNS injury within the same organism, because some CNS neurons (e.g., retinal ganglion cells after optic nerve crush (ONC)) regenerate axons throughout life, whereas others (e.g., hindbrain neurons after spinal cord injury (SCI)) lose this capacity as tadpoles metamorphose into frogs. Tissues from these CNS regions (frog ONC eye, tadpole SCI hindbrain, frog SCI hindbrain) were used in a three-way RNA-seq study of axotomized CNS axons to identify potential core gene expression programs for successful CNS axon regeneration. RESULTS Despite tissue-specific changes in expression dominating the injury responses of each tissue, injury-induced changes in gene expression were nonetheless shared between the two axon-regenerative CNS regions that were not shared with the non-regenerative region. These included similar temporal patterns of gene expression and over 300 injury-responsive genes. Many of these genes and their associated cellular functions had previously been associated with injury responses of multiple tissues, both neural and non-neural, from different species, thereby demonstrating deep phylogenetically conserved commonalities between successful CNS axon regeneration and tissue regeneration in general. Further analyses implicated the KEGG adipocytokine signaling pathway, which links leptin with metabolic and gene regulatory pathways, and a novel gene regulatory network with genes regulating chromatin accessibility at its core, as important hubs in the larger network of injury response genes involved in successful CNS axon regeneration. CONCLUSIONS This study identifies deep, phylogenetically conserved commonalities between CNS axon regeneration and other examples of successful tissue regeneration and provides new targets for studying the molecular underpinnings of successful CNS axon regeneration, as well as a guide for distinguishing pro-regenerative injury-induced changes in gene expression from detrimental ones in mammals.
Collapse
Affiliation(s)
- Jamie L Belrose
- Department of Biological Sciences, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY, 12222, USA
- Center for Neuroscience Research, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY, 12222, USA
| | - Aparna Prasad
- Department of Biological Sciences, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY, 12222, USA
- Center for Neuroscience Research, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY, 12222, USA
| | - Morgan A Sammons
- Department of Biological Sciences, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY, 12222, USA
| | - Kurt M Gibbs
- Department of Biology and Chemistry, Morehead State University, Morehead, KY, 40351, USA
| | - Ben G Szaro
- Department of Biological Sciences, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY, 12222, USA.
- Center for Neuroscience Research, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY, 12222, USA.
| |
Collapse
|
49
|
Eroglu U, Yakar F, Bozkurt M, Kahilogullari G, Shukriyev B, Ugur HC, Unlu A. Surgical Results of the Use of Expanded Polytetrafluor Ethylene as an Adhesion Inhibitory Membrane in Anastomosis Surgery for Total Peripheral Nerve Cut. Indian J Surg 2020. [DOI: 10.1007/s12262-020-02068-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
|
50
|
Chen T, Li Y, Ni W, Tang B, Wei Y, Li J, Yu J, Zhang L, Gao J, Zhou J, Zhang W, Xu H, Hu J. Human Neural Stem Cell-Conditioned Medium Inhibits Inflammation in Macrophages Via Sirt-1 Signaling Pathway In Vitro and Promotes Sciatic Nerve Injury Recovery in Rats. Stem Cells Dev 2020; 29:1084-1095. [PMID: 32560594 DOI: 10.1089/scd.2020.0020] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Chronic persistent inflammation is thought to impede axon regeneration and cause demyelinating disease also with neuropathic pain, leading to more severe dysfunction after peripheral nerve injury. Increasing evidence indicates that neural stem cells (NSCs) have immunomodulatory effects, and previous studies have shown that many of the beneficial effects attributed to stem cell therapy may exert their therapeutic effects through paracrine mechanisms. In this research, the repairing effect of NSC-conditioned medium (NSC-CM) on sciatic nerve injury and its mechanism of repair were further explored. The present research showed that NSC-CM promoted histopathological and functional recovery after crush injury in rats, and what counts is that NSC-CM inhibited the inflammation of sciatic nerve in the late stage of injury. NSC-CM significantly downregulated the infiltration of proinflammatory factors [tumor necrosis factor alpha (TNF-α), interleukin 6 (IL-6), and IL-1β] as well as decreased the CD68 inflammatory macrophages infiltrating in the sciatic nerve. In addition, to study the effect of NSC-CM on the inflammatory state of macrophages in vitro, lipopolysaccharide (LPS) was used to induce the proinflammation of macrophages. The results showed that NSC-CM decreased the expression of macrophage proinflammatory-related proteins (IL-6, IL-1β, TNF-α, inducible nitric oxide synthase) induced by LPS. The activation of Sirt-1 signaling in macrophages effectively countered the proinflammation induced by LPS in the presence of NSC-CM. Using Sirt-1-specific inhibitor EX527 partially weakened the anti-inflammatory effect of NSC-CM. Altogether, this study demonstrated for the first time that NSC-CM promotes functional recovery after sciatic nerve crush injury in vivo and also inhibits the inflammation in activated macrophages by activating Sirt-1 signaling pathway in vitro.
Collapse
Affiliation(s)
- Tianyan Chen
- Jinagsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Yilei Li
- Jinagsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Wei Ni
- Jinagsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Bin Tang
- Jinagsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Yusheng Wei
- Jinagsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Jing Li
- Jinagsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Jiahong Yu
- Jinagsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Lei Zhang
- Jinagsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Jianyi Gao
- Jinagsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Jiqin Zhou
- Jinagsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Weining Zhang
- Jinagsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Hong Xu
- Department of Clinical Laboratory, Zhenjiang Centre for Disease Prevention and Control, Zhenjiang, China
| | - Jiabo Hu
- Jinagsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| |
Collapse
|