1
|
Zhou M, Xu Z, Zhong H, Ning G, Feng S. Spinal cord injury and inflammatory mediators: Role in "fire barrier" formation and potential for neural regeneration. Neural Regen Res 2026; 21:923-937. [PMID: 39995083 DOI: 10.4103/nrr.nrr-d-24-00792] [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: 07/18/2024] [Accepted: 01/07/2025] [Indexed: 02/26/2025] Open
Abstract
Traumatic spinal cord injury result in considerable and lasting functional impairments, triggering complex inflammatory and pathological events. Spinal cord scars, often metaphorically referred to as "fire barriers," aim to control the spread of neuroinflammation during the acute phase but later hinder axon regeneration in later stages. Recent studies have enhanced our understanding of immunomodulation, revealing that injury-associated inflammation involves various cell types and molecules with positive and negative effects. This review employs bibliometric analysis to examine the literature on inflammatory mediators in spinal cord injury, highlighting recent research and providing a comprehensive overview of the current state of research and the latest advances in studies on neuroinflammation related to spinal cord injury. We summarize the immune and inflammatory responses at different stages of spinal cord injury, offering crucial insights for future research. Additionally, we review repair strategies based on inflammatory mediators for the injured spinal cord. Finally, this review discusses the current status and future directions of translational research focused on immune-targeting strategies, including pharmaceuticals, biomedical engineering, and gene therapy. The development of a combined, precise, and multitemporal strategy for the repair of injured spinal cords represents a promising direction for future research.
Collapse
Affiliation(s)
- Mi Zhou
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
- International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin, China
- Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin, China
| | - Zhengyu Xu
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
- International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin, China
- Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin, China
| | - Hao Zhong
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
- International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin, China
- Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin, China
| | - Guangzhi Ning
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
- International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin, China
- Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin, China
| | - Shiqing Feng
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
- International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin, China
- Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin, China
- Department of Orthopedics, Qilu Hospital of Shandong University, Shandong University Center for Orthopedics, Advanced Medical Research Institute, Shandong University, Jinan, Shandong Province, China
- Orthopedic Research Center of Shandong University & Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| |
Collapse
|
2
|
Yanai R, Yasunaga G, Tsuji S, Honda T, Iwata A, Miyagawa E, Yoshida K, Kishimoto M, Sakai H, Fujise Y, Asagiri M, Mitamura Y. Dietary intake of whale oil-containing ω-3 long-chain polyunsaturated fatty acids attenuates choroidal neovascularization in mice. FASEB J 2025; 39:e70378. [PMID: 39937567 PMCID: PMC11818653 DOI: 10.1096/fj.202402041r] [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: 09/01/2024] [Revised: 01/16/2025] [Accepted: 01/28/2025] [Indexed: 02/13/2025]
Abstract
Age-related macular degeneration (AMD) is a leading cause of blindness in Western and developing countries. Since antivascular endothelial growth factor (VEGF) therapy is available for the regression of choroidal neovascularization (CNV), it does not work for the pathophysiology of AMD so a cure is increasingly demanded. Whale oil promotes various bodily functions, such as anti-inflammatory effects for cardiovascular disease, but its physiological mechanisms are still unclarified. Here, we examined the effects of whale oil on a mouse model of AMD. The area of CNV measured in choroidal flat-mount preparations at 7 days after laser photocoagulation was significantly smaller in mice fed whale oil compared with control mice free of ω-3 long-chain polyunsaturated fatty acids (LCPUFAs). The plasma concentrations of ω-3 LCPUFAs were higher, whereas those of ω-6 LCPUFAs were lower in mice fed the diet containing whale oil than in those fed the control diet. The concentrations of various inflammatory cytokines and chemokines in the retina or choroid at 3 or 7 days after CNV induction differed between the two groups of mice. Furthermore, the concentration of VEGF was decreased in the retina but increased in the choroid at 7 or 3 days after photocoagulation, respectively. Our results thus show that dietary intake of whale oil-containing ω-3 LCPUFAs attenuated CNV in association with changes in inflammatory mediator levels and VEGF expression in the retina and choroid of mice, and it, therefore, warrants further study as a means to protect against AMD in humans.
Collapse
Affiliation(s)
- Ryoji Yanai
- Department of Ophthalmology, Institute of Biomedical SciencesTokushima University Graduate SchoolTokushimaJapan
| | | | - Shunya Tsuji
- Department of PharmacologyYamaguchi University Graduate School of MedicineUbeYamaguchiJapan
| | - Takeshi Honda
- Department of PharmacologyYamaguchi University Graduate School of MedicineUbeYamaguchiJapan
| | - Arihiro Iwata
- Yamaguchi Prefectural Industrial Technology InstituteUbeYamaguchiJapan
| | - Eiji Miyagawa
- Yamaguchi Prefectural Industrial Technology InstituteUbeYamaguchiJapan
| | - Koji Yoshida
- Yoshida General Techno Co., Ltd.ShimonosekiYamaguchiJapan
| | | | | | | | - Masataka Asagiri
- Department of PharmacologyYamaguchi University Graduate School of MedicineUbeYamaguchiJapan
| | - Yoshinori Mitamura
- Department of Ophthalmology, Institute of Biomedical SciencesTokushima University Graduate SchoolTokushimaJapan
| |
Collapse
|
3
|
John RK, Vogel SP, Zia S, Lee KV, Nguyen AT, Torres-Espin A, Fenrich KK, Ng C, Schmidt EKA, Vavrek R, Raposo PJF, Smith K, Fouad K, Plemel JR. Reawakening inflammation in the chronically injured spinal cord using lipopolysaccharide induces diverse microglial states. J Neuroinflammation 2025; 22:56. [PMID: 40022205 PMCID: PMC11871772 DOI: 10.1186/s12974-025-03379-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 02/14/2025] [Indexed: 03/03/2025] Open
Abstract
BACKGROUND Rehabilitative training is an effective method to promote recovery following spinal cord injury (SCI), with lower training efficacy observed in the chronic stage. The increased training efficacy during the subacute period is associated with a shift towards a more adaptive or proreparative state induced by the SCI. A potential link is SCI-induced inflammation, which is elevated in the subacute period, and, as injection of lipopolysaccharide (LPS) alongside training improves recovery in chronic SCI, suggesting LPS could reopen a window of plasticity late after injury. Microglia may play a role in LPS-mediated plasticity as they react to LPS and are implicated in facilitating recovery following SCI. However, it is unknown how microglia change in response to LPS following SCI to promote neuroplasticity. MAIN BODY Here we used single-cell RNA sequencing to examine microglial responses in subacute and chronic SCI with and without an LPS injection. We show that subacute SCI is characterized by a disease-associated microglial (DAM) signature, while chronic SCI is highly heterogeneous, with both injury-induced and homeostatic states. DAM states exhibit predicted metabolic pathway activity and neuronal interactions that are associated with potential mediators of plasticity. With LPS injection, microglia shifted away from the homeostatic signature to a primed, translation-associated state and increased DAM in degenerated tracts caudal to the injury. CONCLUSION Microglial states following an inflammatory stimulus in chronic injury incompletely recapitulate the subacute injury environment, showing both overlapping and distinct microglial signatures across time and with LPS injection. Our results contribute to an understanding of how microglia and LPS-induced neuroinflammation contribute to plasticity following SCI.
Collapse
Affiliation(s)
- Rebecca K John
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| | - Sadie P Vogel
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| | - Sameera Zia
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| | - Kelly V Lee
- Department of Medicine, Division of Neurology, University of Alberta, Edmonton, Canada
| | - Antoinette T Nguyen
- Department of Physical Therapy, Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, Canada
| | - Abel Torres-Espin
- Department of Physical Therapy, Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, Canada
- School of Public Health Sciences, Faculty of Health, University of Waterloo, Waterloo, Canada
| | - Keith K Fenrich
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
- Department of Physical Therapy, Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, Canada
| | - Carmen Ng
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| | - Emma K A Schmidt
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| | - Romana Vavrek
- Department of Physical Therapy, Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, Canada
| | - Pamela J F Raposo
- Department of Physical Therapy, Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, Canada
| | - Keira Smith
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| | - Karim Fouad
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada.
- Department of Physical Therapy, Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, Canada.
| | - Jason R Plemel
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada.
- Department of Medicine, Division of Neurology, University of Alberta, Edmonton, Canada.
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Canada.
- Li Ka Shing Institute of Virology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada.
| |
Collapse
|
4
|
Liu ZG, Zhou LY, Sun YQ, Ma YH, Liu CM, Zhang BY. Unlocking the potential for optic nerve regeneration over long distances: a multi-therapeutic intervention. Front Neurol 2025; 15:1526973. [PMID: 39850731 PMCID: PMC11754882 DOI: 10.3389/fneur.2024.1526973] [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: 11/15/2024] [Accepted: 12/24/2024] [Indexed: 01/25/2025] Open
Abstract
Retinal ganglion cells (RGCs) generally fail to regenerate axons, resulting in irreversible vision loss after optic nerve injury. While many studies have shown that modulating specific genes can enhance RGCs survival and promote optic nerve regeneration, inducing long-distance axon regeneration in vivo through single-gene manipulation remains challenging. Nevertheless, combined multi-gene therapies have proven effective in significantly enhancing axonal regeneration. At present, research on promoting optic nerve regeneration remains slow, with most studies unable to achieve axonal growth beyond the optic chiasm or reestablish connections with the brain. Future research priorities include directing axonal growth along correct pathways, facilitating synapse formation and myelination, and modifying the inhibitory microenvironment. These strategies are crucial not only for optic nerve regeneration but also for broader applications in central nervous system repair. In this review, we discuss multifactors therapeutic strategies for optic nerve regeneration, offering insights into advancing nerve regeneration research.
Collapse
Affiliation(s)
- Zhen-Gang Liu
- Department of Orthopaedics, China-Japan Union Hospital of Jilin University, Changchun, China
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Lai-Yang Zhou
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Yong-Quan Sun
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Yi-Hang Ma
- Department of Orthopaedics, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Chang-Mei Liu
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Bo-Yin Zhang
- Department of Orthopaedics, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
5
|
Masin L, Bergmans S, Van Dyck A, Farrow K, De Groef L, Moons L. Local glycolysis supports injury-induced axonal regeneration. J Cell Biol 2024; 223:e202402133. [PMID: 39352499 PMCID: PMC11451009 DOI: 10.1083/jcb.202402133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 04/09/2024] [Accepted: 09/09/2024] [Indexed: 10/06/2024] Open
Abstract
Successful axonal regeneration following injury requires the effective allocation of energy. How axons withstand the initial disruption in mitochondrial energy production caused by the injury and subsequently initiate regrowth is poorly understood. Transcriptomic data showed increased expression of glycolytic genes after optic nerve crush in retinal ganglion cells with the co-deletion of Pten and Socs3. Using retinal cultures in a multicompartment microfluidic device, we observed increased regrowth and enhanced mitochondrial trafficking in the axons of Pten and Socs3 co-deleted neurons. While wild-type axons relied on mitochondrial metabolism, after injury, in the absence of Pten and Socs3, energy production was supported by local glycolysis. Specific inhibition of lactate production hindered injury survival and the initiation of regrowth while slowing down glycolysis upstream impaired regrowth initiation, axonal elongation, and energy production. Together, these observations reveal that glycolytic ATP, combined with sustained mitochondrial transport, is essential for injury-induced axonal regrowth, providing new insights into the metabolic underpinnings of axonal regeneration.
Collapse
Affiliation(s)
- Luca Masin
- Department of Biology, Animal Physiology and Neurobiology Section, KU Leuven, Leuven Brain Institute, Leuven, Belgium
| | - Steven Bergmans
- Department of Biology, Animal Physiology and Neurobiology Section, KU Leuven, Leuven Brain Institute, Leuven, Belgium
| | - Annelies Van Dyck
- Department of Biology, Animal Physiology and Neurobiology Section, KU Leuven, Leuven Brain Institute, Leuven, Belgium
| | - Karl Farrow
- Department of Biology, Animal Physiology and Neurobiology Section, KU Leuven, Leuven Brain Institute, Leuven, Belgium
- Neuro-Electronics Research Flanders, Vlaams Instituut voor Biotechnologie, Leuven, Belgium
- imec, Leuven, Belgium
| | - Lies De Groef
- Department of Biology, Animal Physiology and Neurobiology Section, KU Leuven, Leuven Brain Institute, Leuven, Belgium
| | - Lieve Moons
- Department of Biology, Animal Physiology and Neurobiology Section, KU Leuven, Leuven Brain Institute, Leuven, Belgium
| |
Collapse
|
6
|
Cucarian J, Raposo P, Vavrek R, Nguyen A, Nelson B, Monnier P, Torres-Espin A, Fenrich K, Fouad K. No impact of anti-inflammatory medication on inflammation-driven recovery following cervical spinal cord injury in rats. Exp Neurol 2024; 383:115039. [PMID: 39481514 DOI: 10.1016/j.expneurol.2024.115039] [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: 06/13/2024] [Revised: 10/24/2024] [Accepted: 10/27/2024] [Indexed: 11/02/2024]
Abstract
Following spinal cord injury (SCI), inflammation is associated with the exacerbation of damage to spinal tissue. Consequently, managing inflammation during the acute and subacute phases is a common target in SCI treatment. However, inflammation may also induce potential benefits, including the stimulation of neuroplasticity and repair. This positive role of inflammation in spinal cord healing and functional recovery is not fully understood. To address this knowledge gap, we examined the effects of two common anti-inflammatory medications, Diphenhydramine and Methylprednisolone, on the efficacy of rehabilitative motor training on recovery from subacute cervical SCI in adult rats. Training depends critically on neuroplasticity thus if inflammation is a key regulator, we propose that anti-inflammatory drugs will reduce subsequent recovery. Both drugs were administered orally over one month, alongside task-specific reaching and grasping training. After treatment, no substantial changes in motor recovery or lesion size between the treated and control groups were observed. Treated animals also did not show any discernible changes in sensory function or anxiety-like behavior. Taken together, our data indicate that the prolonged use of these anti-inflammatory agents at commonly used doses did not profoundly impact recovery following an SCI. Therefore, considering earlier reports of the benefits of pro-inflammatory stimuli on plasticity, further studies in this area are imperative to elucidate the true impact of treating inflammation and its implications for recovery after spinal cord injuries.
Collapse
Affiliation(s)
- Jaison Cucarian
- Neuroscience and Mental Health Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada.
| | - Pamela Raposo
- Department of Physical Therapy, Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, Canada
| | - Romana Vavrek
- Department of Physical Therapy, Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, Canada
| | - Antoinette Nguyen
- Department of Physical Therapy, Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, Canada
| | - Brooklynn Nelson
- Neuroscience and Mental Health Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Philippe Monnier
- Department of Ophthalmology and Vision Science, University of Toronto, Toronto, ON, Canada; Donald K. Johnson Eye Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Abel Torres-Espin
- Department of Physical Therapy, Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, Canada; School of Public Health Sciences, Faculty of Health, University of Waterloo, Waterloo, ON, Canada; Department of Neurological Surgery and Brain and Spinal Injury Center (BASIC), Faculty of Medicine, University of California San Francisco, San Francisco, USA
| | - Keith Fenrich
- Neuroscience and Mental Health Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada; Department of Physical Therapy, Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, Canada
| | - Karim Fouad
- Neuroscience and Mental Health Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada; Department of Physical Therapy, Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, Canada
| |
Collapse
|
7
|
Gobrecht P, Gebel J, Hilla A, Gisselmann G, Fischer D. Targeting Vasohibins to Promote Axon Regeneration. J Neurosci 2024; 44:e2031232024. [PMID: 38429108 PMCID: PMC10993095 DOI: 10.1523/jneurosci.2031-23.2024] [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: 10/27/2023] [Revised: 12/13/2023] [Accepted: 02/07/2024] [Indexed: 03/03/2024] Open
Abstract
Treatments accelerating axon regeneration in the nervous system are still clinically unavailable. However, parthenolide promotes adult sensory neurons' axon growth in culture by inhibiting microtubule detyrosination. Here, we show that overexpression of vasohibins increases microtubule detyrosination in growth cones and compromises growth in culture and in vivo. Moreover, overexpression of these proteins increases the required parthenolide concentrations to promote axon regeneration. At the same time, the partial knockdown of endogenous vasohibins or their enhancer SVBP in neurons facilitates axon growth, verifying them as pharmacological targets for promoting axon growth. In vivo, repeated intravenous application of parthenolide or its prodrug di-methyl-amino-parthenolide (DMAPT) markedly facilitates the regeneration of sensory, motor, and sympathetic axons in injured murine and rat nerves, leading to acceleration of functional recovery. Moreover, orally applied DMAPT was similarly effective in promoting nerve regeneration. Thus, pharmacological inhibition of vasohibins facilitates axon regeneration in different species and nerves, making parthenolide and DMAPT the first promising drugs for curing nerve injury.
Collapse
Affiliation(s)
- Philipp Gobrecht
- Center of Pharmacology, Institute II, Medical Faculty, University of Cologne, Cologne D-50931, Germany
- Department of Cell Physiology, Ruhr University of Bochum, Bochum 44780, Germany
| | - Jeannette Gebel
- Center of Pharmacology, Institute II, Medical Faculty, University of Cologne, Cologne D-50931, Germany
- Department of Cell Physiology, Ruhr University of Bochum, Bochum 44780, Germany
| | - Alexander Hilla
- Department of Cell Physiology, Ruhr University of Bochum, Bochum 44780, Germany
| | - Günter Gisselmann
- Department of Cell Physiology, Ruhr University of Bochum, Bochum 44780, Germany
| | - Dietmar Fischer
- Center of Pharmacology, Institute II, Medical Faculty, University of Cologne, Cologne D-50931, Germany
- Department of Cell Physiology, Ruhr University of Bochum, Bochum 44780, Germany
| |
Collapse
|
8
|
Namestnikova DD, Kovalenko DB, Pokusaeva IA, Chudakova DA, Gubskiy IL, Yarygin KN, Baklaushev VP. Mesenchymal stem cells in the treatment of ischemic stroke. КЛИНИЧЕСКАЯ ПРАКТИКА 2024; 14:49-64. [DOI: 10.17816/clinpract624157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2025] Open
Abstract
Over the past two decades, multiple preclinical studies have shown that transplantation of mesenchymal stem cells leads to a pronounced positive effect in animals with experimental stroke. Based on the promising results of preclinical studies, several clinical trials on the transplantation of mesenchymal stem cells to stroke patients have also been conducted. In this review, we present and analyze the results of completed clinical trials dedicated to the mesenchymal stem cells transplantation in patients with ischemic stroke. According to the obtained results, it can be concluded that transplantation of mesenchymal stem cells is safe and feasible from the economic and biomedical point of view. For the further implementa-tion of this promising approach into the clinical practice, randomized, placebo-controlled, multicenter clinical trials are needed with a large sample of patients and optimized cell transplantation protocols and patient inclusion criteria. In this review we also discuss possi-ble strategies to enhance the effectiveness of cell therapy with the use of mesenchymal stem cells.
Collapse
Affiliation(s)
- Daria D. Namestnikova
- Federal Center of Brain Research and Neurotechnologies
- Pirogov Russian National Research Medical University
| | | | | | | | - Ilya L. Gubskiy
- Federal Center of Brain Research and Neurotechnologies
- Pirogov Russian National Research Medical University
| | | | - Vladimir P. Baklaushev
- Federal Center of Brain Research and Neurotechnologies
- Pirogov Russian National Research Medical University
- Federal Scientific and Clinical Center for Specialized Medical Assistance and Medical Technologies of the Federal Medical Biological Agency
| |
Collapse
|
9
|
Glass J, Robinson RL, Greenway G, Jones G, Sharma S. Diabetic Müller-Glial-Cell-Specific Il6ra Knockout Mice Exhibit Accelerated Retinal Functional Decline and Thinning of the Inner Nuclear Layer. Invest Ophthalmol Vis Sci 2023; 64:1. [PMID: 38038619 PMCID: PMC10697173 DOI: 10.1167/iovs.64.15.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 11/08/2023] [Indexed: 12/02/2023] Open
Abstract
Purpose Interleukin-6 (IL-6) is implicated in the pathology of diabetic retinopathy (DR). IL-6 trans-signaling via soluble IL-6 receptor (IL-6R) is primarily responsible for its pro-inflammatory functions, whereas cis-signaling via membrane-bound IL-6R is anti-inflammatory. Using a Müller-glial-cell-specific Il6ra-/- mouse, we examined how loss of IL-6 cis-signaling in Müller glial cells (MGCs) affected retinal thinning and electroretinography (ERG) response over 9 months of diabetes. Methods Diabetes was induced in wildtype and knockout mice with streptozotocin (40 mg/kg, daily for 5 days). Spectral domain optical coherence tomography (SD-OCT), ERG, and fundoscopy/fluorescein angiography (FA) were assessed at 2, 6, and 9 months of diabetes. MGCs and bipolar neurons were examined in retinal tissue sections by immunofluorescence. Results Diabetic MGC Il6ra-/- mice had significantly thinner retinas than diabetic wildtype mice at 2 (-7.6 µm), 6 (-12.0 µm), and 9 months (-5.0 µm) of diabetes, as well as significant thinning of the inner nuclear layer (INL). Diabetic MGC Il6ra-/- mice also showed a reduction in scotopic B-wave amplitude and B-wave/A-wave ratio earlier than wildtype diabetic mice. In retinal sections, we found a decrease in bipolar neuronal marker PKCα only in diabetic MGC Il6ra-/- mice, which was significantly lower than both controls and diabetic wildtype mice. Glutamine synthetase, a Müller cell marker, was reduced in both wildtype and MGC Il6ra-/- diabetic mice compared to their respective controls. Conclusions IL-6 cis-signaling in MGCs contributes to maintenance of the INL in diabetes, and loss of the IL-6 receptor reduces MGC-mediated neuroprotection of bipolar neurons in the diabetic retina.
Collapse
Affiliation(s)
- Joshua Glass
- Center for Biotechnology and Genomic Medicine, Augusta University, Augusta, Georgia, United States
| | - Rebekah L. Robinson
- Center for Biotechnology and Genomic Medicine, Augusta University, Augusta, Georgia, United States
| | - Grace Greenway
- Center for Biotechnology and Genomic Medicine, Augusta University, Augusta, Georgia, United States
| | - Garrett Jones
- Center for Biotechnology and Genomic Medicine, Augusta University, Augusta, Georgia, United States
| | - Shruti Sharma
- Center for Biotechnology and Genomic Medicine, Augusta University, Augusta, Georgia, United States
- Culver Vision Discovery Institute, Augusta University, Augusta, Georgia, United States
- Department of Ophthalmology, Augusta University, Augusta, Georgia, United States
| |
Collapse
|
10
|
Estera LA, Walsh SP, Headen JA, Williamson RE, Kalinski AL. Neuroinflammation: Breaking barriers and bridging gaps. Neurosci Res 2023; 197:9-17. [PMID: 34748905 DOI: 10.1016/j.neures.2021.11.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 11/01/2021] [Indexed: 01/04/2023]
Abstract
Neurons are the cells of the nervous system and are responsible for every thought, movement and perception. Immune cells are the cells of the immune system, constantly protecting from foreign pathogens. Understanding the interaction between the two systems is especially important in disease states such as autoimmune or neurodegenerative disease. Unfortunately, this interaction is typically detrimental to the host. However, recent efforts have focused on how neurons and immune cells interact, either directly or indirectly, following traumatic injury to the nervous system. The outcome of this interaction can be beneficial - leading to successful neural repair, or detrimental - leading to functional deficits, depending on where the injury occurs. This review will discuss our understanding of neuron-immune cell interactions after traumatic injury to both the peripheral and central nervous system.
Collapse
Affiliation(s)
- Lora A Estera
- Department of Biology, Ball State University, Muncie, IN 47306, USA
| | - Sam P Walsh
- Department of Biology, Ball State University, Muncie, IN 47306, USA
| | - Jordan A Headen
- Department of Biology, Ball State University, Muncie, IN 47306, USA
| | | | - Ashley L Kalinski
- Department of Biology, Ball State University, Muncie, IN 47306, USA.
| |
Collapse
|
11
|
Leibinger M, Zeitler C, Paulat M, Gobrecht P, Hilla A, Andreadaki A, Guthoff R, Fischer D. Inhibition of microtubule detyrosination by parthenolide facilitates functional CNS axon regeneration. eLife 2023; 12:RP88279. [PMID: 37846146 PMCID: PMC10581688 DOI: 10.7554/elife.88279] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2023] Open
Abstract
Injured axons in the central nervous system (CNS) usually fail to regenerate, causing permanent disabilities. However, the knockdown of Pten knockout or treatment of neurons with hyper-IL-6 (hIL-6) transforms neurons into a regenerative state, allowing them to regenerate axons in the injured optic nerve and spinal cord. Transneuronal delivery of hIL-6 to the injured brain stem neurons enables functional recovery after severe spinal cord injury. Here we demonstrate that the beneficial hIL-6 and Pten knockout effects on axon growth are limited by the induction of tubulin detyrosination in axonal growth cones. Hence, cotreatment with parthenolide, a compound blocking microtubule detyrosination, synergistically accelerates neurite growth of cultured murine CNS neurons and primary RGCs isolated from adult human eyes. Systemic application of the prodrug dimethylamino-parthenolide (DMAPT) facilitates axon regeneration in the injured optic nerve and spinal cord. Moreover, combinatorial treatment further improves hIL-6-induced axon regeneration and locomotor recovery after severe SCI. Thus, DMAPT facilitates functional CNS regeneration and reduces the limiting effects of pro-regenerative treatments, making it a promising drug candidate for treating CNS injuries.
Collapse
Affiliation(s)
- Marco Leibinger
- Center for Pharmacology, Institute II, Medical Faculty and University of CologneCologneGermany
- Department of Cell Physiology, Ruhr University of BochumBochumGermany
| | - Charlotte Zeitler
- Center for Pharmacology, Institute II, Medical Faculty and University of CologneCologneGermany
- Department of Cell Physiology, Ruhr University of BochumBochumGermany
| | - Miriam Paulat
- Department of Cell Physiology, Ruhr University of BochumBochumGermany
| | - Philipp Gobrecht
- Center for Pharmacology, Institute II, Medical Faculty and University of CologneCologneGermany
- Department of Cell Physiology, Ruhr University of BochumBochumGermany
| | - Alexander Hilla
- Department of Cell Physiology, Ruhr University of BochumBochumGermany
| | - Anastasia Andreadaki
- Center for Pharmacology, Institute II, Medical Faculty and University of CologneCologneGermany
- Department of Cell Physiology, Ruhr University of BochumBochumGermany
| | - Rainer Guthoff
- Eye Hospital, Heinrich Heine University DüsseldorfDüsseldorfGermany
| | - Dietmar Fischer
- Center for Pharmacology, Institute II, Medical Faculty and University of CologneCologneGermany
- Department of Cell Physiology, Ruhr University of BochumBochumGermany
| |
Collapse
|
12
|
Xiao R, Lei C, Zhang Y, Zhang M. Interleukin-6 in retinal diseases: From pathogenesis to therapy. Exp Eye Res 2023:109556. [PMID: 37385535 DOI: 10.1016/j.exer.2023.109556] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 06/03/2023] [Accepted: 06/25/2023] [Indexed: 07/01/2023]
Abstract
Interleukin-6 (IL-6) is a pleiotropic cytokine that participates in immunomodulation, inflammation, increases vascular permeability, hematopoiesis, and stimulates cell proliferation, among other biological processes. It exerts effects primarily through the classic and trans-signaling pathways. Many studies have demonstrated that IL-6 plays a critical role in the development of retinal diseases including diabetic retinopathy, uveitis, age-related macular degeneration, glaucoma, retinal vein occlusion, central serous chorioretinopathy and proliferative vitreoretinopathy. Thus, the progressive development of drugs targeting IL-6 and IL-6 receptor may play a role in the treatment of multiple retinal diseases. In this article, we comprehensively review the IL-6's biological functions of and its mechanisms in the pathogenesis of various retinal diseases. Furthermore, we summarize the drugs targeting IL-6 and its receptor and prospect their potential application in retinal diseases, hoping to provide new ideas for the treatment of retinal diseases.
Collapse
Affiliation(s)
- Ruihan Xiao
- Department of Ophthalmology and Research Laboratory of Macular Disease, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Chunyan Lei
- Department of Ophthalmology and Research Laboratory of Macular Disease, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yi Zhang
- Department of Ophthalmology and Research Laboratory of Macular Disease, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Meixia Zhang
- Department of Ophthalmology and Research Laboratory of Macular Disease, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
13
|
Zhang ZY, Zuo ZY, Liang Y, Zhang SM, Zhang CX, Chi J, Fan B, Li GY. Promotion of axon regeneration and protection on injured retinal ganglion cells by rCXCL2. Inflamm Regen 2023; 43:31. [PMID: 37340465 DOI: 10.1186/s41232-023-00283-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 05/31/2023] [Indexed: 06/22/2023] Open
Abstract
BACKGROUND In addition to rescuing injured retinal ganglion cells (RGCs) by stimulating the intrinsic growth ability of damaged RGCs in various retinal/optic neuropathies, increasing evidence has shown that the external microenvironmental factors also play a crucial role in restoring the survival of RGCs by promoting the regrowth of RGC axons, especially inflammatory factors. In this study, we aimed to screen out the underlying inflammatory factor involved in the signaling of staurosporine (STS)-induced axon regeneration and verify its role in the protection of RGCs and the promotion of axon regrowth. METHODS We performed transcriptome RNA sequencing for STS induction models in vitro and analyzed the differentially expressed genes. After targeting the key gene, we verified the role of the candidate factor in RGC protection and promotion of axon regeneration in vivo with two RGC-injured animal models (optic nerve crush, ONC; retinal N-methyl-D-aspartate, NMDA damage) by using cholera toxin subunit B anterograde axon tracing and specific immunostaining of RGCs. RESULTS We found that a series of inflammatory genes expressed upregulated in the signaling of STS-induced axon regrowth and we targeted the candidate CXCL2 gene since the level of the chemokine CXCL2 gene elevated significantly among the top upregulated genes. We further demonstrated that intravitreal injection of rCXCL2 robustly promoted axon regeneration and significantly improved RGC survival in ONC-injured mice in vivo. However, different from its role in ONC model, the intravitreal injection of rCXCL2 was able to simply protect RGCs against NMDA-induced excitotoxicity in mouse retina and maintain the long-distance projection of RGC axons, yet failed to promote significant axon regeneration. CONCLUSIONS We provide the first in vivo evidence that CXCL2, as an inflammatory factor, is a key regulator in the axon regeneration and neuroprotection of RGCs. Our comparative study may facilitate deciphering the exact molecular mechanisms of RGC axon regeneration and developing high-potency targeted drugs.
Collapse
Affiliation(s)
- Zi-Yuan Zhang
- Department of Ophthalmology, Second Hospital of Jilin University, Changchun, 130041, China
| | - Zhao-Yang Zuo
- Department of Ophthalmology, Second Hospital of Jilin University, Changchun, 130041, China
| | - Yang Liang
- Department of Ophthalmology, Second Hospital of Jilin University, Changchun, 130041, China
| | - Si-Ming Zhang
- Department of Ophthalmology, Second Hospital of Jilin University, Changchun, 130041, China
| | - Chun-Xia Zhang
- Department of Ophthalmology, Second Hospital of Jilin University, Changchun, 130041, China
| | - Jing Chi
- Department of Ophthalmology, Second Hospital of Jilin University, Changchun, 130041, China
| | - Bin Fan
- Department of Ophthalmology, Second Hospital of Jilin University, Changchun, 130041, China.
| | - Guang-Yu Li
- Department of Ophthalmology, Second Hospital of Jilin University, Changchun, 130041, China.
| |
Collapse
|
14
|
Mays EA, Ellis EB, Hussain Z, Parajuli P, Sundararaghavan HG. Enzyme-Mediated Nerve Growth Factor Release from Nanofibers Using Gelatin Microspheres. Tissue Eng Part A 2023; 29:333-343. [PMID: 37016821 DOI: 10.1089/ten.tea.2022.0205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/06/2023] Open
Abstract
Spinal cord injury is a complex environment, with many conflicting growth factors present at different times throughout the injury timeline. Delivery of multiple growth factors has received mixed results, highlighting a need to consider the timing of delivery for possibly antagonistic growth factors. Cell-mediated degradation of delivery vehicles for delayed release of growth factors offers an attractive way to exploit the highly active immune response in the spinal cord injury environment. In this study, growth factor-loaded gelatin microspheres (GMS) combined with methacrylated hyaluronic acid (MeHA) were electrospun to create GMS fibers (GMSF) for delayed release of growth factors (GFs). GMS were successfully combined with MeHA while electrospinning, with an average fiber diameter of 365 ± 10 nm and 44% ± 8% fiber alignment. GMSF with nerve growth factor (NGF) was tested on dissociated chick dorsal root ganglia cells. We further tested the effect of M1 macrophage-conditioned media (M1CM) to simulate macrophage invasion after spinal cord injury for cell-mediated degradation. We hypothesized that neurons grown on GMSF with loaded NGF would exhibit longer neurites in M1CM, showing a release of functional NGF, as compared with controls. GMSF in M1CM was significantly different from MeHA in serum-free media (SFM) and M0-conditioned media (M0CM), as well as GMSF in M0CM (p < 0.05). Moreover, GMSF + NGF in all media conditions were significantly different from MeHA in SFM and M0CM (p < 0.05). The goal of this study was to develop a biomaterial system where drug delivery is triggered by immune response, allowing for more control and longer exposure to encapsulated drugs. The spinal cord injury microenvironment is known to have a robust immune response, making this immune-medicated drug release system particularly significant for directed repair.
Collapse
Affiliation(s)
- Elizabeth A Mays
- Department of Biomedical Engineering, Wayne State University, Detroit, Michigan, USA
| | - Eric B Ellis
- Department of Chemical Engineering and Materials Science, Wayne State University, Detroit, Michigan, USA
| | - Zahin Hussain
- School of Medicine, Michigan State University, East Lansing, Michigan, USA
| | - Prahlad Parajuli
- Department of Pharmaceutical Sciences, Wayne State University, Detroit, Michigan, USA
| | | |
Collapse
|
15
|
Barron A, Manna S, McElwain CJ, Musumeci A, McCarthy FP, O’Keeffe GW, McCarthy CM. Maternal pre-eclampsia serum increases neurite growth and mitochondrial function through a potential IL-6-dependent mechanism in differentiated SH-SY5Y cells. Front Physiol 2023; 13:1043481. [PMID: 36714304 PMCID: PMC9877349 DOI: 10.3389/fphys.2022.1043481] [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: 09/13/2022] [Accepted: 12/28/2022] [Indexed: 01/15/2023] Open
Abstract
Introduction: Pre-eclampsia (PE) is a common and serious hypertensive disorder of pregnancy, which affects 3%-5% of first-time pregnancies and is a leading cause of maternal and neonatal morbidity and mortality. Prenatal exposure to PE is associated with an increased risk of neurodevelopmental disorders in affected offspring, although the cellular and molecular basis of this increased risk is largely unknown. Methods: Here, we examined the effects of exposure to maternal serum from women with PE or a healthy uncomplicated pregnancy on the survival, neurite growth and mitochondrial function of neuronally differentiated human SH-SY5Y neuroblastoma cells, which are commonly used to study neurite growth. Neurite growth and mitochondrial function are two strongly linked neurodevelopmental parameters in which alterations have been implicated in neurodevelopmental disorders. Following this, we investigated the pleiotropic cytokine interleukin-6 (IL-6) levels as a potential mechanism. Results: Cells exposed to 3% (v/v) PE serum for 72 h exhibited increased neurite growth (p < 0.05), which was validated in the human neural progenitor cell line, ReNcell® VM (p < 0.01), and mitochondrial respiration (elevated oxygen consumption rate (p < 0.05), basal mitochondrial respiration, proton leak, ATP synthesis, and non-mitochondrial respiration) compared to control serum-treated cells. ELISA analysis showed elevations in maternal IL-6 in PE sera (p < 0.05) and placental explants (p < 0.05). In support of this, SH-SY5Y cells exposed to 3% (v/v) PE serum for 24 h had increased phospho-STAT3 levels, which is a key intracellular mediator of IL-6 signalling (p < 0.05). Furthermore, treatment with anti-IL-6 neutralizing antibody blocked the effects of PE serum on neurite growth (p < 0.05), and exposure to IL-6 promoted neurite growth in SH-SY5Y cells (p < 0.01). Discussion: Collectively these data show elevated serum levels of maternal IL-6 in PE, which increases neurite growth and mitochondrial function in SH-SY5Y cells. This rationalizes the further study of IL-6 as a potential mediator between PE exposure and neurodevelopmental outcome in the offspring.
Collapse
Affiliation(s)
- Aaron Barron
- Department of Anatomy and Neuroscience, University College, Cork, Ireland,Department of Pharmacology and Therapeutics, University College Cork, Cork, Ireland
| | - Samprikta Manna
- Department of Pharmacology and Therapeutics, University College Cork, Cork, Ireland,Department of Obstetrics and Gynaecology, Cork University Maternity Hospital, Cork, Ireland
| | - Colm J. McElwain
- Department of Pharmacology and Therapeutics, University College Cork, Cork, Ireland
| | - Andrea Musumeci
- Department of Pharmacology and Therapeutics, University College Cork, Cork, Ireland
| | - Fergus P. McCarthy
- Department of Obstetrics and Gynaecology, Cork University Maternity Hospital, Cork, Ireland
| | - Gerard W. O’Keeffe
- Department of Anatomy and Neuroscience, University College, Cork, Ireland,Cork Neuroscience Centre, University College Cork, Cork, Ireland,*Correspondence: Gerard W. O’Keeffe, ; Cathal M. McCarthy,
| | - Cathal M. McCarthy
- Department of Pharmacology and Therapeutics, University College Cork, Cork, Ireland,*Correspondence: Gerard W. O’Keeffe, ; Cathal M. McCarthy,
| |
Collapse
|
16
|
Tang S, Wang K, Qi X. Neuro-protective effects of n-butylphthalide on carbon monoxide poisoning rats by modulating IL-2, AKT and BCL-2. J Toxicol Sci 2023; 48:495-505. [PMID: 37661366 DOI: 10.2131/jts.48.495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Acute carbon monoxide poisoning (CO-poisoning) causes neurotoxicity by inducing necrosis, apoptosis, lipid peroxidation, and oxidative stress. DL-3-n-butylphthalide (NBP) is a synthetic compound originally extracted from the seeds of Chinese celery and based on pure l-3-n-butylphthalide. In ischemia/reperfusion, it exerts neuroprotective effects through its anti-apoptotic, anti-necrotic and antioxidant properties, and activation of pro-survival pathways. Our study performed bioinformatic analysis to identify the differential expression genes. CO-poisoning patients' blood was collected to confirm the findings. Male rats were exposed to CO 3000 ppm for 40 min, and NBP (100 mg/kg/day) was continuously injected intraperitoneally immediately after poisoning and for the next 15 days. After NBP treatment, the rats were evaluated by Morris water maze test. At the end of experiments, blood and brain tissues of the rats were collected to evaluate the expression levels of IL-2, AKT and BCL-2. We found that IL-2 was elevated in CO-poisoning patients and animal models. Brain tissue damage in CO-poisoning rats was significantly alleviated after NBP treatment. Furthermore, NBP increased the expression of IL-2, AKT and BCL-2 in rat CO-poisoning model. NBP showed neuroprotective action by increasing IL-2, AKT, and BCL-2 expressions.
Collapse
Affiliation(s)
- Shengtao Tang
- The Second School of Clinical Medicine, Southern Medical University, China
- Department of Neurology, The First People's Hospital of Chenzhou, China
| | - Kunyu Wang
- Department of Neurology, The First Teaching Hospital of Jilin University, China
| | - Xiaokun Qi
- The Second School of Clinical Medicine, Southern Medical University, China
- Department of Neurology, The Sixth Medical Center of the General Hospital of the Chinese People's Liberation Army, China
| |
Collapse
|
17
|
Gaire BP. Microglia as the Critical Regulators of Neuroprotection and Functional Recovery in Cerebral Ischemia. Cell Mol Neurobiol 2022; 42:2505-2525. [PMID: 34460037 PMCID: PMC11421653 DOI: 10.1007/s10571-021-01145-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 08/25/2021] [Indexed: 12/13/2022]
Abstract
Microglial activation is considered as the critical pathogenic event in diverse central nervous system disorders including cerebral ischemia. Proinflammatory responses of activated microglia have been well reported in the ischemic brain and neuroinflammatory responses of activated microglia have been believed to be the potential therapeutic strategy. However, despite having proinflammatory roles, microglia can have significant anti-inflammatory roles and they are associated with the production of growth factors which are responsible for neuroprotection and recovery after ischemic injury. Microglia can directly promote neuroprotection by preventing ischemic infarct expansion and promoting functional outcomes. Indirectly, microglia are involved in promoting anti-inflammatory responses, neurogenesis, and angiogenesis in the ischemic brain which are crucial pathophysiological events for ischemic recovery. In fact, anti-inflammatory cytokines and growth factors produced by microglia can promote neuroprotection and attenuate neurobehavioral deficits. In addition, microglia regulate phagocytosis, axonal regeneration, blood-brain barrier protection, white matter integrity, and synaptic remodeling, which are essential for ischemic recovery. Microglia can also regulate crosstalk with neurons and other cell types to promote neuroprotection and ischemic recovery. This review mainly focuses on the roles of microglia in neuroprotection and recovery following ischemic injury. Furthermore, this review also sheds the light on the therapeutic potential of microglia in stroke patients.
Collapse
Affiliation(s)
- Bhakta Prasad Gaire
- Department of Neurology and Anesthesiology, Shock Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
| |
Collapse
|
18
|
Kiaie N, Gorabi AM, Loveless R, Teng Y, Jamialahmadi T, Sahebkar A. The regenerative potential of glial progenitor cells and reactive astrocytes in CNS injuries. Neurosci Biobehav Rev 2022; 140:104794. [PMID: 35902044 DOI: 10.1016/j.neubiorev.2022.104794] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 07/20/2022] [Accepted: 07/21/2022] [Indexed: 10/16/2022]
Abstract
Cell therapeutic approaches focusing on the regeneration of damaged tissue have been a popular topic among researchers in recent years. In particular, self-repair scarring from the central nervous system (CNS) can significantly complicate the treatment of an injured patient. In CNS regeneration schemes, either glial progenitor cells or reactive glial cells have key roles to play. In this review, the contribution and underlying mechanisms of these progenitor/reactive glial cells during CNS regeneration are discussed, as well as their role in CNS-related diseases.
Collapse
Affiliation(s)
- Nasim Kiaie
- Research Center for Advanced Technologies in Cardiovascular Medicine, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Armita Mahdavi Gorabi
- Department of Tissue Engineering and Applied Cell Science, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Reid Loveless
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Yong Teng
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Tannaz Jamialahmadi
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
19
|
Liu H, Bell K, Herrmann A, Arnhold S, Mercieca K, Anders F, Nagel-Wolfrum K, Thanos S, Prokosch V. Crystallins Play a Crucial Role in Glaucoma and Promote Neuronal Cell Survival in an In Vitro Model Through Modulating Müller Cell Secretion. Invest Ophthalmol Vis Sci 2022; 63:3. [PMID: 35816047 PMCID: PMC9284462 DOI: 10.1167/iovs.63.8.3] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose The aim of this study was to explore the roles of crystallins in the context of aging in glaucoma and potential mechanisms of neuroprotection in an experimental animal model of glaucoma. Methods Intraocular pressure (IOP) was significantly elevated for 8 weeks in animals at different ages (10 days, 12 weeks, and 44 weeks) by episcleral vein cauterization. Retinal ganglion cells (RGCs) were quantified by anti-Brn3a immunohistochemical staining (IHC). Proteomics using ESI-LTQ Orbitrap XL-MS was used to analyze the presence and abundance of crystallin isoforms the retinal samples, respectively. Neuroprotective property and localization of three selected crystallins CRYAB, CRYBB2, and CRYGB as most significantly changed in retina and retinal layers were determined by IHC. Their expressions and endocytic uptakes into Müller cells were analyzed by IHC and Western blotting. Müller cell secretion of neurotrophic factors into the supernatant following CRYAB, CRYBB2, and CRYGB supplementation in vitro was measured via microarray. Results IOP elevation resulted in significant RGC loss in all age groups (P < 0.001). The loss increased with aging. Proteomics analysis revealed in parallel a significant decrease of crystallin abundance – especially CRYAB, CRYBB2, and CRYGB. Significant neuroprotective effects of CRYAB, CRYBB2, and CRYGB after addition to retinal cultures were demonstrated (P < 0.001). Endocytic uptake of CRYAB, CRYBB2, and CRYGB was seen in Müller cells with subsequent increased secretion of various neurotrophic factors into the supernatant, including nerve growth factor, clusterin, and matrix metallopeptidase 9. Conclusions An age-dependent decrease in CRYAB, CRYBB2, and CRYGB abundance is found going along with increased RGC loss. Addition of CRYAB, CRYBB2, and CRYGB to culture protected RGCs in vitro. CRYAB, CRYBB2, and CRYGB were uptaken into Müller cells. Secretion of neurotrophic factors was increased as a potential mode of action.
Collapse
Affiliation(s)
- Hanhan Liu
- Department of Ophthalmology, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Katharina Bell
- Singapore Eye Research Institute and Singapore National Eye Center, Singapore; Duke-NUS Medical School, Singapore
| | - Anja Herrmann
- Institute of Veterinary Anatomy, Histology and Embryology, Justus-Liebig-University Gießen, Gießen, Germany
| | - Stefan Arnhold
- Institute of Veterinary Anatomy, Histology and Embryology, Justus-Liebig-University Gießen, Gießen, Germany
| | - Karl Mercieca
- Department of Ophthalmology, University Medical Center Bonn, Bonn, Germany
| | - Fabian Anders
- Department of Ophthalmology, University Medical Centre of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Kerstin Nagel-Wolfrum
- Institute for Molecular Physiology, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Solon Thanos
- Department of Ophthalmology, Experimental Ophthalmology, University Medical Center Münster, Münster, Germany
| | - Verena Prokosch
- Department of Ophthalmology, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
20
|
Casagrande BP, Bueno AA, Pisani LP, Estadella D. Hepatic glycogen participates in the regulation of hypothalamic pAkt/Akt ratio in high-sugar/high-fat diet-induced obesity. Metab Brain Dis 2022; 37:1423-1434. [PMID: 35316448 DOI: 10.1007/s11011-022-00944-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 02/22/2022] [Indexed: 01/07/2023]
Abstract
The hypothalamus is a major integrating centre that controls energy homeostasis and plays a major role in hepatic glycogen (HGlyc) turnover. Not only do hypothalamic and hepatic Akt levels influence glucose homeostasis and glycogen synthesis, but exposure to high-sugar/high-fat diets (HSHF) can also lead to hypothalamic inflammation and HGlyc accumulation. HSHF withdrawal overall restores energy and glucose homeostasis, but the actual relationship between hypothalamic inflammation and HGlyc after short-term HSHF withdrawal has not yet been fully elucidated. Here we investigated the short-term effects of HSHF withdrawal preceded by a 30-day HSHF intake on the liver-hypothalamus crosstalk and glucose homeostasis. Sixty-day old male Wistar rats were fed for 30 days a control chow (n = 10) (Ct), or an HSHF diet (n = 20). On the 30th day of dietary intervention, a random HSHF subset (n = 10) had their diets switched to control chow for 48 h (Hw) whilst the remaining HSHF rats remained in the HSHF diet (n = 10) (Hd). All rats were anaesthetized and euthanized at the end of the protocol. We quantified HGlyc, Akt phosphorylation, inflammation and glucose homeostasis biomarkers. We also assessed the effect of propensity to obesity on those biomarkers, as detailed previously. Hd rats showed impaired glucose homeostasis, higher HGlyc and hypothalamic inflammation, and lower pAkt/Akt. Increased HGlyc was significantly associated with HSHF intake on pAkt/Akt lowered levels. We also found that HGlyc breakdown may have prevented a further pAkt/Akt drop after HSHF withdrawal. Propensity to obesity showed no apparent effect on hypothalamic inflammation or glucose homeostasis. Our findings suggest a comprehensive role of HGlyc as a structural and functional modulator of energy metabolism, and such roles may come into play relatively rapidly.
Collapse
Affiliation(s)
- Breno P Casagrande
- Biosciences Department, Institute of Health and Society, Federal University of São Paulo-UNIFESP/BS, 1015-020, Santos, São Paulo, Brazil
| | - Allain A Bueno
- College of Health, Life and Environmental Sciences, University of Worcester, Henwick Grove, WR2 6AJ, Worcester, United Kingdom
| | - Luciana P Pisani
- Biosciences Department, Institute of Health and Society, Federal University of São Paulo-UNIFESP/BS, 1015-020, Santos, São Paulo, Brazil
| | - Debora Estadella
- Biosciences Department, Institute of Health and Society, Federal University of São Paulo-UNIFESP/BS, 1015-020, Santos, São Paulo, Brazil.
| |
Collapse
|
21
|
Claes M, Geeraerts E, Plaisance S, Mentens S, Van den Haute C, De Groef L, Arckens L, Moons L. Chronic Chemogenetic Activation of the Superior Colliculus in Glaucomatous Mice: Local and Retrograde Molecular Signature. Cells 2022; 11:1784. [PMID: 35681479 PMCID: PMC9179903 DOI: 10.3390/cells11111784] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 05/20/2022] [Accepted: 05/22/2022] [Indexed: 12/13/2022] Open
Abstract
One important facet of glaucoma pathophysiology is axonal damage, which ultimately disrupts the connection between the retina and its postsynaptic brain targets. The concurrent loss of retrograde support interferes with the functionality and survival of the retinal ganglion cells (RGCs). Previous research has shown that stimulation of neuronal activity in a primary retinal target area-i.e., the superior colliculus-promotes RGC survival in an acute mouse model of glaucoma. To build further on this observation, we applied repeated chemogenetics in the superior colliculus of a more chronic murine glaucoma model-i.e., the microbead occlusion model-and performed bulk RNA sequencing on collicular lysates and isolated RGCs. Our study revealed that chronic target stimulation upon glaucomatous injury phenocopies the a priori expected molecular response: growth factors were pinpointed as essential transcriptional regulators both in the locally stimulated tissue and in distant, unstimulated RGCs. Strikingly, and although the RGC transcriptome revealed a partial reversal of the glaucomatous signature and an enrichment of pro-survival signaling pathways, functional rescue of injured RGCs was not achieved. By postulating various explanations for the lack of RGC neuroprotection, we aim to warrant researchers and drug developers for the complexity of chronic neuromodulation and growth factor signaling.
Collapse
Affiliation(s)
- Marie Claes
- Neural Circuit Development and Regeneration Research Group, Department of Biology, KU Leuven, Leuven Brain Institute, 3000 Leuven, Belgium; (M.C.); (E.G.); (S.M.)
| | - Emiel Geeraerts
- Neural Circuit Development and Regeneration Research Group, Department of Biology, KU Leuven, Leuven Brain Institute, 3000 Leuven, Belgium; (M.C.); (E.G.); (S.M.)
| | | | - Stephanie Mentens
- Neural Circuit Development and Regeneration Research Group, Department of Biology, KU Leuven, Leuven Brain Institute, 3000 Leuven, Belgium; (M.C.); (E.G.); (S.M.)
- Cellular Communication and Neurodegeneration Research Group, Department of Biology, KU Leuven, Leuven Brain Institute, 3000 Leuven, Belgium;
- Neuroplasticity and Neuroproteomics Research Group, Department of Biology, KU Leuven, Leuven Brain Institute, 3000 Leuven, Belgium;
| | - Chris Van den Haute
- Neurobiology and Gene Therapy Research Group, Department of Neurosciences, KU Leuven, Leuven Brain Institute, 3000 Leuven, Belgium;
- KU Leuven Viral Vector Core, 3000 Leuven, Belgium
| | - Lies De Groef
- Cellular Communication and Neurodegeneration Research Group, Department of Biology, KU Leuven, Leuven Brain Institute, 3000 Leuven, Belgium;
| | - Lut Arckens
- Neuroplasticity and Neuroproteomics Research Group, Department of Biology, KU Leuven, Leuven Brain Institute, 3000 Leuven, Belgium;
| | - Lieve Moons
- Neural Circuit Development and Regeneration Research Group, Department of Biology, KU Leuven, Leuven Brain Institute, 3000 Leuven, Belgium; (M.C.); (E.G.); (S.M.)
| |
Collapse
|
22
|
Lin MW, Fang SY, Hsu JYC, Huang CY, Lee PH, Huang CC, Chen HF, Lam CF, Lee JS. Mitochondrial Transplantation Attenuates Neural Damage and Improves Locomotor Function After Traumatic Spinal Cord Injury in Rats. Front Neurosci 2022; 16:800883. [PMID: 35495036 PMCID: PMC9039257 DOI: 10.3389/fnins.2022.800883] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Accepted: 03/18/2022] [Indexed: 01/11/2023] Open
Abstract
Mitochondrial dysfunction is a hallmark of secondary neuroinflammatory responses and neuronal death in spinal cord injury (SCI). Even though mitochondria-based therapy is an attractive therapeutic option for SCI, the efficacy of transplantation of allogeneic mitochondria in the treatment of SCI remains unclear. Herein, we determined the therapeutic effects of mitochondrial transplantation in the traumatic SCI rats. Compressive SCI was induced by applying an aneurysm clip on the T10 spinal cord of rats. A 100-μg bolus of soleus-derived allogeneic mitochondria labeled with fluorescent tracker was transplanted into the injured spinal cords. The results showed that the transplanted mitochondria were detectable in the injured spinal cord up to 28 days after treatment. The rats which received mitochondrial transplantation exhibited better recovery of locomotor and sensory functions than those who did not. Both the expression of dynamin-related protein 1 and severity of demyelination in the injured cord were reduced in the mitochondrial transplanted groups. Mitochondrial transplantation also alleviated SCI-induced cellular apoptosis and inflammation responses. These findings suggest that transplantation of allogeneic mitochondria at the early stage of SCI reduces mitochondrial fragmentation, neuroapoptosis, neuroinflammation, and generation of oxidative stress, thus leading to improved functional recovery following traumatic SCI.
Collapse
Affiliation(s)
- Ming-Wei Lin
- Department of Medical Research, E-Da Hospital, E-Da Cancer Hospital, Kaohsiung City, Taiwan
- Department of Nursing, College of Medicine, I-Shou University, Kaohsiung City, Taiwan
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung City, Taiwan
| | - Shih-Yuan Fang
- Department of Anesthesiology, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan City, Taiwan
| | - Jung-Yu C. Hsu
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan City, Taiwan
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan City, Taiwan
| | - Chih-Yuan Huang
- Section of Neurosurgery, Department of Surgery, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan City, Taiwan
| | - Po-Hsuan Lee
- Section of Neurosurgery, Department of Surgery, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan City, Taiwan
| | - Chi-Chen Huang
- Section of Neurosurgery, Department of Surgery, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan City, Taiwan
| | - Hui-Fang Chen
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan City, Taiwan
| | - Chen-Fuh Lam
- Department of Medical Research, E-Da Hospital, E-Da Cancer Hospital, Kaohsiung City, Taiwan
- Department of Anesthesiology, E-Da Hospital, E-Da Cancer Hospital, Kaohsiung City, Taiwan
- College of Medicine, I-Shou University, Kaohsiung City, Taiwan
| | - Jung-Shun Lee
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan City, Taiwan
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan City, Taiwan
- Section of Neurosurgery, Department of Surgery, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan City, Taiwan
- *Correspondence: Jung-Shun Lee,
| |
Collapse
|
23
|
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: 32] [Impact Index Per Article: 10.7] [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
|
24
|
García-Juárez M, Camacho-Morales A. Defining the role of anti- and pro-inflammatory outcomes of Interleukin-6 in mental health. Neuroscience 2022; 492:32-46. [DOI: 10.1016/j.neuroscience.2022.03.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 03/03/2022] [Accepted: 03/16/2022] [Indexed: 01/03/2023]
|
25
|
Zhang Q, Li Y, Zhuo Y. Synaptic or Non-synaptic? Different Intercellular Interactions with Retinal Ganglion Cells in Optic Nerve Regeneration. Mol Neurobiol 2022; 59:3052-3072. [PMID: 35266115 PMCID: PMC9016027 DOI: 10.1007/s12035-022-02781-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 02/24/2022] [Indexed: 12/31/2022]
Abstract
Axons of adult neurons in the mammalian central nervous system generally fail to regenerate by themselves, and few if any therapeutic options exist to reverse this situation. Due to a weak intrinsic potential for axon growth and the presence of strong extrinsic inhibitors, retinal ganglion cells (RGCs) cannot regenerate their axons spontaneously after optic nerve injury and eventually undergo apoptosis, resulting in permanent visual dysfunction. Regarding the extracellular environment, research to date has generally focused on glial cells and inflammatory cells, while few studies have discussed the potentially significant role of interneurons that make direct connections with RGCs as part of the complex retinal circuitry. In this study, we provide a novel angle to summarize these extracellular influences following optic nerve injury as "intercellular interactions" with RGCs and classify these interactions as synaptic and non-synaptic. By discussing current knowledge of non-synaptic (glial cells and inflammatory cells) and synaptic (mostly amacrine cells and bipolar cells) interactions, we hope to accentuate the previously neglected but significant effects of pre-synaptic interneurons and bring unique insights into future pursuit of optic nerve regeneration and visual function recovery.
Collapse
Affiliation(s)
- Qi Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Yiqing Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, Guangzhou, 510060, China.
| | - Yehong Zhuo
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, Guangzhou, 510060, China.
| |
Collapse
|
26
|
Khan A, Parray A, Akhtar N, Agouni A, Kamran S, Pananchikkal SV, Priyanka R, Gad H, Ponirakis G, Petropoulos IN, Chen KH, Tayyab K, Saqqur M, Shuaib A, Malik RA. Corneal nerve loss in patients with TIA and acute ischemic stroke in relation to circulating markers of inflammation and vascular integrity. Sci Rep 2022; 12:3332. [PMID: 35228650 PMCID: PMC8885663 DOI: 10.1038/s41598-022-07353-7] [Citation(s) in RCA: 4] [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: 10/09/2021] [Accepted: 02/14/2022] [Indexed: 12/01/2022] Open
Abstract
Vascular and inflammatory mechanisms are implicated in the development of cerebrovascular disease and corneal nerve loss occurs in patients with transient ischemic attack (TIA) and acute ischemic stroke (AIS). We have assessed whether serum markers of inflammation and vascular integrity are associated with the severity of corneal nerve loss in patients with TIA and AIS. Corneal confocal microscopy (CCM) was performed to quantify corneal nerve fiber density (CNFD), corneal nerve branch density (CNBD) and corneal nerve fiber length (CNFL) in 105 patients with TIA (n = 24) or AIS (n = 81) and age matched control subjects (n = 56). Circulating levels of IL-6, MMP-2, MMP-9, E-Selectin, P-Selectin and VEGF were quantified in patients within 48 h of presentation with a TIA or AIS. CNFL (P = 0.000, P = 0.000), CNFD (P = 0.122, P = 0.000) and CNBD (P = 0.002, P = 0.000) were reduced in patients with TIA and AIS compared to controls, respectively with no difference between patients with AIS and TIA. The NIHSS Score (P = 0.000), IL-6 (P = 0.011) and E-Selectin (P = 0.032) were higher in patients with AIS compared to TIA with no difference in MMP-2 (P = 0.636), MMP-9 (P = 0.098), P-Selectin (P = 0.395) and VEGF (P = 0.831). CNFL (r = 0.218, P = 0.026) and CNFD (r = 0.230, P = 0.019) correlated with IL-6 and multiple regression analysis showed a positive association of CNFL and CNFD with IL-6 (P = 0.041, P = 0.043). Patients with TIA and AIS have evidence of corneal nerve loss and elevated IL6 and E-selectin levels. Larger longitudinal studies are required to determine the association between inflammatory and vascular markers and corneal nerve fiber loss in patients with cerebrovascular disease.
Collapse
Affiliation(s)
- Adnan Khan
- Department of Medicine, Research Division, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Aijaz Parray
- Department of Neurology and Institute of Neurosciences, Hamad Medical Corporation, Doha, Qatar
| | - Naveed Akhtar
- Department of Neurology and Institute of Neurosciences, Hamad Medical Corporation, Doha, Qatar
| | - Abdelali Agouni
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha, Qatar
| | - Saadat Kamran
- Department of Neurology and Institute of Neurosciences, Hamad Medical Corporation, Doha, Qatar
| | - Sajitha V Pananchikkal
- Department of Neurology and Institute of Neurosciences, Hamad Medical Corporation, Doha, Qatar
| | - Ruth Priyanka
- Department of Neurology and Institute of Neurosciences, Hamad Medical Corporation, Doha, Qatar
| | - Hoda Gad
- Department of Medicine, Research Division, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Georgios Ponirakis
- Department of Medicine, Research Division, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Ioannis N Petropoulos
- Department of Medicine, Research Division, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Kuan-Han Chen
- Department of Medicine, Research Division, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Kausar Tayyab
- Department of Medicine, Research Division, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Maher Saqqur
- Department of Neurology, University of Toronto Mississauga, Mississauga, ON, Canada
| | - Ashfaq Shuaib
- Stroke Program, Department of Neurology, University of Alberta, Alberta, Canada
| | - Rayaz A Malik
- Department of Medicine, Research Division, Weill Cornell Medicine-Qatar, Doha, Qatar.
| |
Collapse
|
27
|
Guo S, Moore RM, Charlesworth MC, Johnson KL, Spinner RJ, Windebank AJ, Wang H. The proteome of distal nerves: implication in delayed repair and poor functional recovery. Neural Regen Res 2022; 17:1998-2006. [PMID: 35142689 PMCID: PMC8848594 DOI: 10.4103/1673-5374.335159] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Chronic denervation is one of the key factors that affect nerve regeneration. Chronic axotomy deteriorates the distal nerve stump, causes protein changes, and renders the microenvironment less permissive for regeneration. Some of these factors/proteins have been individually studied. To better delineate the comprehensive protein expression profiles and identify proteins that contribute to or are associated with this detrimental effect, we carried out a proteomic analysis of the distal nerve using an established delayed rat sciatic nerve repair model. Four rats that received immediate repair after sciatic nerve transection served as control, whereas four rats in the experimental group (chronic denervation) had their sciatic nerve repaired after a 12-week delay. All the rats were sacrificed after 16 weeks to harvest the distal nerves for extracting proteins. Twenty-five micrograms of protein from each sample were fractionated in SDS-PAGE gels. NanoLC-MS/MS analysis was applied to the gels. Protein expression levels of nerves on the surgery side were compared to those on the contralateral side. Any protein with a P value of less than 0.05 and a fold change of 4 or higher was deemed differentially expressed. All the differentially expressed proteins in both groups were further stratified according to the biological processes. A PubMed search was also conducted to identify the differentially expressed proteins that have been reported to be either beneficial or detrimental to nerve regeneration. Ingenuity Pathway Analysis (IPA) software was used for pathway analysis. The results showed that 709 differentially expressed proteins were identified in the delayed repair group, with a bigger proportion of immune and inflammatory process-related proteins and a smaller proportion of proteins related to axon regeneration and lipid metabolism in comparison to the control group where 478 differentially expressed proteins were identified. The experimental group also had more beneficial proteins that were downregulated and more detrimental proteins that were upregulated. IPA revealed that protective pathways such as LXR/RXR, acute phase response, RAC, ERK/MAPK, CNTF, IL-6, and FGF signaling were inhibited in the delayed repair group, whereas three detrimental pathways, including the complement system, PTEN, and apoptosis signaling, were activated. An available database of the adult rodent sciatic nerve was used to assign protein changes to specific cell types. The poor regeneration seen in the delayed repair group could be associated with the down-regulation of beneficial proteins and up-regulation of detrimental proteins. The proteins and pathways identified in this study may offer clues for future studies to identify therapeutic targets.
Collapse
Affiliation(s)
- Song Guo
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, USA
| | - Raymond M Moore
- Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
| | | | | | - Robert J Spinner
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, USA
| | | | - Huan Wang
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
28
|
Terheyden-Keighley D, Leibinger M, Fischer D. Transneuronal delivery of designer cytokines: perspectives for spinal cord injury. Neural Regen Res 2022; 17:338-340. [PMID: 34269207 PMCID: PMC8464008 DOI: 10.4103/1673-5374.317974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Affiliation(s)
| | - Marco Leibinger
- Department of Cell Physiology, Ruhr University of Bochum, Bochum, Germany
| | - Dietmar Fischer
- Department of Cell Physiology, Ruhr University of Bochum, Bochum, Germany
| |
Collapse
|
29
|
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: 2.3] [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
|
30
|
Stem Cell Secretome for Spinal Cord Repair: Is It More than Just a Random Baseline Set of Factors? Cells 2021; 10:cells10113214. [PMID: 34831436 PMCID: PMC8625005 DOI: 10.3390/cells10113214] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 11/02/2021] [Accepted: 11/16/2021] [Indexed: 11/29/2022] Open
Abstract
Hundreds of thousands of people suffer spinal cord injuries each year. The experimental application of stem cells following spinal cord injury has opened a new era to promote neuroprotection and neuroregeneration of damaged tissue. Currently, there is great interest in the intravenous administration of the secretome produced by mesenchymal stem cells in acute or subacute spinal cord injuries. However, it is important to highlight that undifferentiated neural stem cells and induced pluripotent stem cells are able to adapt to the damaged environment and produce the so-called lesion-induced secretome. This review article focuses on current research related to the secretome and the lesion-induced secretome and their roles in modulating spinal cord injury symptoms and functional recovery, emphasizing different compositions of the lesion-induced secretome in various models of spinal cord injury.
Collapse
|
31
|
Mirabella F, Desiato G, Mancinelli S, Fossati G, Rasile M, Morini R, Markicevic M, Grimm C, Amegandjin C, Termanini A, Peano C, Kunderfranco P, di Cristo G, Zerbi V, Menna E, Lodato S, Matteoli M, Pozzi D. Prenatal interleukin 6 elevation increases glutamatergic synapse density and disrupts hippocampal connectivity in offspring. Immunity 2021; 54:2611-2631.e8. [PMID: 34758338 PMCID: PMC8585508 DOI: 10.1016/j.immuni.2021.10.006] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 04/24/2021] [Accepted: 10/07/2021] [Indexed: 02/07/2023]
Abstract
Early prenatal inflammatory conditions are thought to be a risk factor for different neurodevelopmental disorders. Maternal interleukin-6 (IL-6) elevation during pregnancy causes abnormal behavior in offspring, but whether these defects result from altered synaptic developmental trajectories remains unclear. Here we showed that transient IL-6 elevation via injection into pregnant mice or developing embryos enhanced glutamatergic synapses and led to overall brain hyperconnectivity in offspring into adulthood. IL-6 activated synaptogenesis gene programs in glutamatergic neurons and required the transcription factor STAT3 and expression of the RGS4 gene. The STAT3-RGS4 pathway was also activated in neonatal brains during poly(I:C)-induced maternal immune activation, which mimics viral infection during pregnancy. These findings indicate that IL-6 elevation at early developmental stages is sufficient to exert a long-lasting effect on glutamatergic synaptogenesis and brain connectivity, providing a mechanistic framework for the association between prenatal inflammatory events and brain neurodevelopmental disorders.
Collapse
Affiliation(s)
- Filippo Mirabella
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, Milan, Italy; IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Genni Desiato
- IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy; Institute of Neuroscience - National Research Council, 20139 Milan, Italy
| | - Sara Mancinelli
- IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Giuliana Fossati
- IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Marco Rasile
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, Milan, Italy
| | - Raffaella Morini
- IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Marija Markicevic
- Neuroscience Center Zürich, ETH Zürich and University of Zürich, Zürich 8057, Switzerland
| | - Christina Grimm
- Neuroscience Center Zürich, ETH Zürich and University of Zürich, Zürich 8057, Switzerland
| | - Clara Amegandjin
- Department of Neurosciences, Université de Montréal, Montréal, QC, Canada; CHU Sainte-Justine Research Center, Montréal, QC, Canada
| | - Alberto Termanini
- Bioinformatic Unit, Humanitas Clinical and Research Center, 20089 Rozzano, Milan, Italy
| | - Clelia Peano
- Institute of Genetic and Biomedical Research, UoS Milan, National Research Council, 20089 Rozzano, Milan, Italy; Genomic Unit, Humanitas Clinical and Research Center, 20089 Rozzano, Milan, Italy
| | - Paolo Kunderfranco
- Bioinformatic Unit, Humanitas Clinical and Research Center, 20089 Rozzano, Milan, Italy
| | - Graziella di Cristo
- Department of Neurosciences, Université de Montréal, Montréal, QC, Canada; CHU Sainte-Justine Research Center, Montréal, QC, Canada
| | - Valerio Zerbi
- Neuroscience Center Zürich, ETH Zürich and University of Zürich, Zürich 8057, Switzerland; Neural Control of Movement Lab, Department of Health Sciences and Technology, ETH Zürich, Zürich 8057, Switzerland
| | - Elisabetta Menna
- IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy; Institute of Neuroscience - National Research Council, 20139 Milan, Italy
| | - Simona Lodato
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, Milan, Italy; IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Michela Matteoli
- IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy; Institute of Neuroscience - National Research Council, 20139 Milan, Italy.
| | - Davide Pozzi
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, Milan, Italy; IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy.
| |
Collapse
|
32
|
Wareham LK, Echevarria FD, Sousa JL, Konlian DO, Dallas G, Formichella CR, Sankaran P, Goralski PJ, Gustafson JR, Sappington RM. Interleukin-6 promotes microtubule stability in axons via Stat3 protein-protein interactions. iScience 2021; 24:103141. [PMID: 34646984 PMCID: PMC8496173 DOI: 10.1016/j.isci.2021.103141] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 06/02/2021] [Accepted: 09/14/2021] [Indexed: 10/25/2022] Open
Abstract
The interleukin-6 (IL-6) family of cytokines and its downstream effector, STAT3, are important mediators of neuronal health, repair, and disease throughout the CNS, including the visual system. Here, we elucidate a transcription-independent mechanism for the neuropoietic activities of IL-6 related to axon development, regeneration, and repair. We examined the outcome of IL-6 deficiency on structure and function of retinal ganglion cell (RGC) axons, which form the optic projection. We found that IL-6 deficiency substantially delays anterograde axon transport in vivo. The reduced rate of axon transport is accompanied by changes in morphology, structure, and post-translational modification of microtubules. In vivo and in vitro studies in mice and swine revealed that IL-6-dependent microtubule phenotypes arise from protein-protein interactions between STAT3 and stathmin. As in tumor cells and T cells, this STAT3-stathmin interaction stabilizes microtubules in RGCs. Thus, this IL-6-STAT3-dependent mechanism for axon architecture is likely a fundamental mechanism for microtubule stability systemically.
Collapse
Affiliation(s)
- Lauren K Wareham
- Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | | | - Jennifer L Sousa
- Department of Neurobiology and Anatomy, Wake Forest School of Medicine, Atrium Health Wake Forest Baptist Medical Center, 1 Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | - Danielle O Konlian
- Department of Neurobiology and Anatomy, Wake Forest School of Medicine, Atrium Health Wake Forest Baptist Medical Center, 1 Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | - Gabrielle Dallas
- Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Cathryn R Formichella
- Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Priya Sankaran
- Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Peter J Goralski
- Department of Neurobiology and Anatomy, Wake Forest School of Medicine, Atrium Health Wake Forest Baptist Medical Center, 1 Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | - Jenna R Gustafson
- Department of Neurobiology and Anatomy, Wake Forest School of Medicine, Atrium Health Wake Forest Baptist Medical Center, 1 Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | - Rebecca M Sappington
- Department of Neurobiology and Anatomy, Wake Forest School of Medicine, Atrium Health Wake Forest Baptist Medical Center, 1 Medical Center Boulevard, Winston-Salem, NC 27157, USA.,Department of Ophthalmology, Wake Forest School of Medicine, Winston-Salem, NC 27106, USA
| |
Collapse
|
33
|
Yow YY, Goh TK, Nyiew KY, Lim LW, Phang SM, Lim SH, Ratnayeke S, Wong KH. Therapeutic Potential of Complementary and Alternative Medicines in Peripheral Nerve Regeneration: A Systematic Review. Cells 2021; 10:cells10092194. [PMID: 34571842 PMCID: PMC8472132 DOI: 10.3390/cells10092194] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 08/20/2021] [Accepted: 08/20/2021] [Indexed: 12/12/2022] Open
Abstract
Despite the progressive advances, current standards of treatments for peripheral nerve injury do not guarantee complete recovery. Thus, alternative therapeutic interventions should be considered. Complementary and alternative medicines (CAMs) are widely explored for their therapeutic value, but their potential use in peripheral nerve regeneration is underappreciated. The present systematic review, designed according to guidelines of Preferred Reporting Items for Systematic Review and Meta-Analysis Protocols, aims to present and discuss the current literature on the neuroregenerative potential of CAMs, focusing on plants or herbs, mushrooms, decoctions, and their respective natural products. The available literature on CAMs associated with peripheral nerve regeneration published up to 2020 were retrieved from PubMed, Scopus, and Web of Science. According to current literature, the neuroregenerative potential of Achyranthes bidentata, Astragalus membranaceus, Curcuma longa, Panax ginseng, and Hericium erinaceus are the most widely studied. Various CAMs enhanced proliferation and migration of Schwann cells in vitro, primarily through activation of MAPK pathway and FGF-2 signaling, respectively. Animal studies demonstrated the ability of CAMs to promote peripheral nerve regeneration and functional recovery, which are partially associated with modulations of neurotrophic factors, pro-inflammatory cytokines, and anti-apoptotic signaling. This systematic review provides evidence for the potential use of CAMs in the management of peripheral nerve injury.
Collapse
Affiliation(s)
- Yoon-Yen Yow
- Department of Biological Sciences, School of Medicine and Life Sciences, Sunway University, Petaling Jaya 47500, Malaysia; (T.-K.G.); (K.-Y.N.); (S.R.)
- Correspondence: (Y.-Y.Y.); (L.-W.L.); (K.-H.W.); Tel.: +603-7491-8622 (Y.-Y.Y.); +852-3917-6830 (L.-W.L.); +603-7967-4729 (K.-H.W.)
| | - Tiong-Keat Goh
- Department of Biological Sciences, School of Medicine and Life Sciences, Sunway University, Petaling Jaya 47500, Malaysia; (T.-K.G.); (K.-Y.N.); (S.R.)
| | - Ke-Ying Nyiew
- Department of Biological Sciences, School of Medicine and Life Sciences, Sunway University, Petaling Jaya 47500, Malaysia; (T.-K.G.); (K.-Y.N.); (S.R.)
| | - Lee-Wei Lim
- Neuromodulation Laboratory, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, L4 Laboratory Block, Hong Kong
- Correspondence: (Y.-Y.Y.); (L.-W.L.); (K.-H.W.); Tel.: +603-7491-8622 (Y.-Y.Y.); +852-3917-6830 (L.-W.L.); +603-7967-4729 (K.-H.W.)
| | - Siew-Moi Phang
- Institute of Ocean and Earth Sciences, Universiti Malaya, Kuala Lumpur 50603, Malaysia;
- Faculty of Applied Sciences, UCSI University, Cheras, Kuala Lumpur 56000, Malaysia
| | - Siew-Huah Lim
- Department of Chemistry, Faculty of Science, Universiti Malaya, Kuala Lumpur 50603, Malaysia;
| | - Shyamala Ratnayeke
- Department of Biological Sciences, School of Medicine and Life Sciences, Sunway University, Petaling Jaya 47500, Malaysia; (T.-K.G.); (K.-Y.N.); (S.R.)
| | - Kah-Hui Wong
- Department of Anatomy, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia
- Correspondence: (Y.-Y.Y.); (L.-W.L.); (K.-H.W.); Tel.: +603-7491-8622 (Y.-Y.Y.); +852-3917-6830 (L.-W.L.); +603-7967-4729 (K.-H.W.)
| |
Collapse
|
34
|
Signal transducer and activator of transcription-3 mediated neuroprotective effect of interleukin-6 on cobalt chloride mimetic hypoxic cell death in R28 cells. Mol Biol Rep 2021; 48:6197-6203. [PMID: 34318437 DOI: 10.1007/s11033-021-06586-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 07/20/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Hypoxic injury to retinal ganglionic cells and adjoining glia is implicated in glaucomatous optic neuropathy. The present study evaluates the effect of IL-6 on R28 retinal precursor cell line exposed to hypoxic injury. METHODS AND RESULTS Apoptotic cell death induced by hypoxia mimetic CoCl2 in R28 cells with or without IL-6 treatment was measured using cell viability assays and apoptotic markers. Oxidative stress was also measured. Hypoxia induced by mimetic CoCl2 led to a time and concentration dependent apoptosis of cells mediated by disruption of mitochondrial membrane potential and activation of caspase 3. Cells pre-treated with IL-6 demonstrated significantly higher viability and mitochondrial integrity under hypoxic conditions. A critical role of STAT3 was observed in mediating the cytoprotective effects of IL-6. Treatment of cells with IL-6 led to STAT3-mediated expression of the Bcl-2 family proteins and MnSOD. CONCLUSIONS The data from the present study indicate cytoprotective role of IL-6 and suggest a previously unreported mechanism of neuroprotection via STAT3 mediated signaling.
Collapse
|
35
|
Suggestive evidence of genetic association of -572G > C polymorphism with primary open angle glaucoma in a North Indian Punjabi population. Hum Immunol 2021; 82:791-797. [PMID: 34301420 DOI: 10.1016/j.humimm.2021.06.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 05/09/2021] [Accepted: 06/23/2021] [Indexed: 11/21/2022]
Abstract
BACKGROUND IL6 is an important candidate gene implicated in the pathogenesis of glaucoma. The present study assessed the genetic association of -174G > C and -572G > C polymorphisms in the IL6 promoter region with primary open angle glaucoma (POAG) and primary angle closure glaucoma (PACG) in a north Indian Punjabi cohort. METHODS 910 subjects (313 POAG, 148 PACG cases and 449 controls) were recruited. Genotyping was done by TaqMan assays. Genetic association was tested under different genetic models using Plink. Diplotype and linkage disequilibrium (LD) analysis was done through Haploview. Association of clinical parameters with the genotypes was assessed by one-way ANOVA. Adjustment for potential confounding variables was done by binary logistic regression. IL6 levels were measured in POAG patients and controls. RESULTS 572G > C variant showed marginal difference in genotype frequency between pooled cases and POAG subgroup with respect to controls (p = 0.042; OR = 1.33; and p = 0.041; OR = 1.37). The GC genotype conferred 1.37-fold protection under codominant model in POAG cases (p = 0.034, OR = 1.37, 95% CI = 1.02-1.85; pcorr = 0.025, OR = 1.45, 95% CI = 1.04-2.02). The mean value for IOP was elevated among cases having 'CC' genotype at the -572G > C locus (p = 0.037). Lower levels of IL6 were detected in POAG patients in plasma samples (p = 0.0001). CONCLUSION The study reports suggestive evidence for -572G > C variant in IL6 in affecting genetic susceptibility to POAG in the targeted North Indian Punjabi cohort. A correlation of IL6 levels in aqueous humor (AH) and systemic circulation in POAG was observed, the functional and diagnostic relevance of which may be further investigated.
Collapse
|
36
|
Pikatza-Menoio O, Elicegui A, Bengoetxea X, Naldaiz-Gastesi N, López de Munain A, Gerenu G, Gil-Bea FJ, Alonso-Martín S. The Skeletal Muscle Emerges as a New Disease Target in Amyotrophic Lateral Sclerosis. J Pers Med 2021; 11:671. [PMID: 34357138 PMCID: PMC8307751 DOI: 10.3390/jpm11070671] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 07/09/2021] [Accepted: 07/14/2021] [Indexed: 01/02/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disorder that leads to progressive degeneration of motor neurons (MNs) and severe muscle atrophy without effective treatment. Most research on ALS has been focused on the study of MNs and supporting cells of the central nervous system. Strikingly, the recent observations of pathological changes in muscle occurring before disease onset and independent from MN degeneration have bolstered the interest for the study of muscle tissue as a potential target for delivery of therapies for ALS. Skeletal muscle has just been described as a tissue with an important secretory function that is toxic to MNs in the context of ALS. Moreover, a fine-tuning balance between biosynthetic and atrophic pathways is necessary to induce myogenesis for muscle tissue repair. Compromising this response due to primary metabolic abnormalities in the muscle could trigger defective muscle regeneration and neuromuscular junction restoration, with deleterious consequences for MNs and thereby hastening the development of ALS. However, it remains puzzling how backward signaling from the muscle could impinge on MN death. This review provides a comprehensive analysis on the current state-of-the-art of the role of the skeletal muscle in ALS, highlighting its contribution to the neurodegeneration in ALS through backward-signaling processes as a newly uncovered mechanism for a peripheral etiopathogenesis of the disease.
Collapse
Affiliation(s)
- Oihane Pikatza-Menoio
- Neuromuscular Diseases Group, Neurosciences Area, Biodonostia Health Research Institute, 20014 Donostia/San Sebastián, Spain; (O.P.-M.); (A.E.); (X.B.); (N.N.-G.); (A.L.d.M.); (G.G.); (F.J.G.-B.)
- CIBERNED, Carlos III Institute, Spanish Ministry of Economy & Competitiveness, 28031 Madrid, Spain
| | - Amaia Elicegui
- Neuromuscular Diseases Group, Neurosciences Area, Biodonostia Health Research Institute, 20014 Donostia/San Sebastián, Spain; (O.P.-M.); (A.E.); (X.B.); (N.N.-G.); (A.L.d.M.); (G.G.); (F.J.G.-B.)
- CIBERNED, Carlos III Institute, Spanish Ministry of Economy & Competitiveness, 28031 Madrid, Spain
| | - Xabier Bengoetxea
- Neuromuscular Diseases Group, Neurosciences Area, Biodonostia Health Research Institute, 20014 Donostia/San Sebastián, Spain; (O.P.-M.); (A.E.); (X.B.); (N.N.-G.); (A.L.d.M.); (G.G.); (F.J.G.-B.)
| | - Neia Naldaiz-Gastesi
- Neuromuscular Diseases Group, Neurosciences Area, Biodonostia Health Research Institute, 20014 Donostia/San Sebastián, Spain; (O.P.-M.); (A.E.); (X.B.); (N.N.-G.); (A.L.d.M.); (G.G.); (F.J.G.-B.)
- CIBERNED, Carlos III Institute, Spanish Ministry of Economy & Competitiveness, 28031 Madrid, Spain
| | - Adolfo López de Munain
- Neuromuscular Diseases Group, Neurosciences Area, Biodonostia Health Research Institute, 20014 Donostia/San Sebastián, Spain; (O.P.-M.); (A.E.); (X.B.); (N.N.-G.); (A.L.d.M.); (G.G.); (F.J.G.-B.)
- CIBERNED, Carlos III Institute, Spanish Ministry of Economy & Competitiveness, 28031 Madrid, Spain
- Department of Neurology, Donostialdea Integrated Health Organization, Osakidetza Basque Health Service, 20014 Donostia/San Sebastián, Spain
- Department of Neurosciences, Faculty of Medicine and Nursery, University of the Basque Country UPV-EHU, 20014 Donostia/San Sebastián, Spain
| | - Gorka Gerenu
- Neuromuscular Diseases Group, Neurosciences Area, Biodonostia Health Research Institute, 20014 Donostia/San Sebastián, Spain; (O.P.-M.); (A.E.); (X.B.); (N.N.-G.); (A.L.d.M.); (G.G.); (F.J.G.-B.)
- CIBERNED, Carlos III Institute, Spanish Ministry of Economy & Competitiveness, 28031 Madrid, Spain
- Department of Physiology, University of the Basque Country UPV-EHU, 48940 Leioa, Spain
| | - Francisco Javier Gil-Bea
- Neuromuscular Diseases Group, Neurosciences Area, Biodonostia Health Research Institute, 20014 Donostia/San Sebastián, Spain; (O.P.-M.); (A.E.); (X.B.); (N.N.-G.); (A.L.d.M.); (G.G.); (F.J.G.-B.)
- CIBERNED, Carlos III Institute, Spanish Ministry of Economy & Competitiveness, 28031 Madrid, Spain
| | - Sonia Alonso-Martín
- Neuromuscular Diseases Group, Neurosciences Area, Biodonostia Health Research Institute, 20014 Donostia/San Sebastián, Spain; (O.P.-M.); (A.E.); (X.B.); (N.N.-G.); (A.L.d.M.); (G.G.); (F.J.G.-B.)
- CIBERNED, Carlos III Institute, Spanish Ministry of Economy & Competitiveness, 28031 Madrid, Spain
| |
Collapse
|
37
|
MMP2 Modulates Inflammatory Response during Axonal Regeneration in the Murine Visual System. Cells 2021; 10:cells10071672. [PMID: 34359839 PMCID: PMC8307586 DOI: 10.3390/cells10071672] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/21/2021] [Accepted: 06/26/2021] [Indexed: 12/17/2022] Open
Abstract
Neuroinflammation has been put forward as a mechanism triggering axonal regrowth in the mammalian central nervous system (CNS), yet little is known about the underlying cellular and molecular players connecting these two processes. In this study, we provide evidence that MMP2 is an essential factor linking inflammation to axonal regeneration by using an in vivo mouse model of inflammation-induced axonal regeneration in the optic nerve. We show that infiltrating myeloid cells abundantly express MMP2 and that MMP2 deficiency results in reduced long-distance axonal regeneration. However, this phenotype can be rescued by restoring MMP2 expression in myeloid cells via a heterologous bone marrow transplantation. Furthermore, while MMP2 deficiency does not affect the number of infiltrating myeloid cells, it does determine the coordinated expression of pro- and anti-inflammatory molecules. Altogether, in addition to its role in axonal regeneration via resolution of the glial scar, here, we reveal a new mechanism via which MMP2 facilitates axonal regeneration, namely orchestrating the expression of pro- and anti-inflammatory molecules by infiltrating innate immune cells.
Collapse
|
38
|
Zwanzig A, Meng J, Müller H, Bürger S, Schmidt M, Pankonin M, Wiedemann P, Unterlauft JD, Eichler W. Neuroprotective effects of glial mediators in interactions between retinal neurons and Müller cells. Exp Eye Res 2021; 209:108689. [PMID: 34216615 DOI: 10.1016/j.exer.2021.108689] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 05/31/2021] [Accepted: 06/29/2021] [Indexed: 12/18/2022]
Abstract
Progressive retinal ganglion cell (RGC) loss underlies a number of retinal neurodegenerative disorders, which may lead to permanent vision loss. However, secreted neuroprotective factors, such as PEDF, VEGF and IL-6, which are produced by Müller cells, have been shown to promote RGC survival. Assuming that the communication of RGCs with Müller cells involves a release of glioactive substances we sought to determine whether retinal neurons are able to modulate expression levels of Müller cell-derived PEDF, VEGF and IL-6. We demonstrate elevated mRNA levels of these factors in Müller cells in co-cultures with RGCs or R28 cells when compared to homotypic Müller cell cultures. Furthermore, R28 cells were more protected from apoptosis when co-cultured with Müller cells. IL-6 and VEGF were upregulated in Müller cells under hypoxia. Both cytokines, as well as PEDF, induced an altered neuronal expression of members of the Bcl-2 family, which are central molecules in the regulation of apoptosis. These results suggest that in retinal ischemia, via own secreted mediators, RGCs can resist a potential demise by stimulating Müller cells to increase production of neuroprotective factors, which counteract RGC apoptosis.
Collapse
Affiliation(s)
- Annette Zwanzig
- Leipzig University, Department of Ophthalmology and Eye Hospital, Liebigstrasse 10-14, D-04103 Leipzig, Germany
| | - Jie Meng
- Leipzig University, Department of Ophthalmology and Eye Hospital, Liebigstrasse 10-14, D-04103 Leipzig, Germany
| | - Heidi Müller
- Leipzig University, Department of Ophthalmology and Eye Hospital, Liebigstrasse 10-14, D-04103 Leipzig, Germany
| | - Susanne Bürger
- Leipzig University, Department of Ophthalmology and Eye Hospital, Liebigstrasse 10-14, D-04103 Leipzig, Germany
| | - Manuela Schmidt
- Leipzig University, Department of Ophthalmology and Eye Hospital, Liebigstrasse 10-14, D-04103 Leipzig, Germany
| | - Maik Pankonin
- Leipzig University, Department of Ophthalmology and Eye Hospital, Liebigstrasse 10-14, D-04103 Leipzig, Germany
| | - Peter Wiedemann
- Leipzig University, Department of Ophthalmology and Eye Hospital, Liebigstrasse 10-14, D-04103 Leipzig, Germany
| | - Jan Darius Unterlauft
- Leipzig University, Department of Ophthalmology and Eye Hospital, Liebigstrasse 10-14, D-04103 Leipzig, Germany
| | - Wolfram Eichler
- Leipzig University, Department of Ophthalmology and Eye Hospital, Liebigstrasse 10-14, D-04103 Leipzig, Germany.
| |
Collapse
|
39
|
Wu CL, Yang YT, Wang YH, Lin TY, Lin IC, Sung CW. Association of interleukin-6 gene polymorphisms and glaucoma: Systematic review and meta-analysis. Eur J Ophthalmol 2021; 31:1862-1869. [PMID: 32628041 DOI: 10.1177/1120672120940198] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
AIM To conduct a systematic review and meta-analysis to explore the association between IL-6 gene polymorphisms (rs1800795 and rs1524107) and glaucoma. METHODS A comprehensive search was performed to select eligible studies regarding IL-6 polymorphisms and glaucoma. The effect sizes in the fixed-effects model were calculated using odds ratios (ORs) with 95% confidence intervals (CIs). RESULTS Four eligible studies comprising 762 cases and 799 controls were selected for meta-analysis. Regarding the association between the IL-6 rs1800795 polymorphism and glaucoma, those who carried the G/G+G/C genotypes had a non-significant higher risk of glaucoma (OR = 1.29, 95% CI = 0.76-2.19) in the dominant model. However, no obvious association (OR = 0.97, 95% CI = 0.68-1.37) was found for the recessive model (G/G vs G/C+C/C). In the subgroup analysis stratified by ethnicity, no significant associations were observed in populations of Asian or European heritage. Significantly higher glaucoma risks of 15.9 and 99.0 were observed for the dominant (C/C+C/T vs T/T) and recessive (C/C vs C/T+T/T) models, respectively. CONCLUSION No statistically significant glaucoma risks were observed for the rs1800795 except rs1524107 polymorphism of IL-6. Further studies with a larger sample size are required to validate the effects of IL-6 polymorphisms on glaucoma risk.
Collapse
Affiliation(s)
- Chien-Liang Wu
- Department of Ophthalmology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Ophthalmology, Taipei Municipal Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Ya-Ting Yang
- Department of Dentistry, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Yuan-Hung Wang
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Medical Research, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Tzu-Yu Lin
- Department of Ophthalmology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - I-Chan Lin
- Department of Ophthalmology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Ophthalmology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Chih-Wei Sung
- Department of Emergency Medicine, National Taiwan University Hospital Hsin-Chu Branch, Hsinchu, Taiwan
| |
Collapse
|
40
|
Feizi S, Alemzadeh-Ansari M, Karimian F, Esfandiari H. Use of erythropoietin in ophthalmology: a review. Surv Ophthalmol 2021; 67:427-439. [PMID: 34157346 DOI: 10.1016/j.survophthal.2021.06.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 06/08/2021] [Accepted: 06/14/2021] [Indexed: 12/17/2022]
Abstract
Erythropoietin (EPO) is a glycoprotein hormone that regulates hematopoiesis in the human body. The presence of EPO and its receptors in different tissues indicates that this hormone has extramedullary effects in other tissues, including the eye. We focus on the biological roles of this hormone in the development and normal physiologic functions of the eye. Furthermore, we explore the role of EPO in the management of different ocular diseases - including diabetic retinopathy, retinopathy of prematurity, inherited retinal degeneration, branch and central retinal vein occlusion, retinal detachment, traumatic optic neuropathy, optic neuritis, methanol optic neuropathy, nonarteritic anterior ischemic optic neuropathy, glaucoma, and scleral necrosis.
Collapse
Affiliation(s)
- Sepehr Feizi
- Ophthalmic Research Center, Department of Ophthalmology, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | | | - Farid Karimian
- Ophthalmic Research Center, Department of Ophthalmology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamed Esfandiari
- Department of Ophthalmology, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
41
|
Hilla AM, Baehr A, Leibinger M, Andreadaki A, Fischer D. CXCR4/CXCL12-mediated entrapment of axons at the injury site compromises optic nerve regeneration. Proc Natl Acad Sci U S A 2021; 118:e2016409118. [PMID: 34011605 PMCID: PMC8166183 DOI: 10.1073/pnas.2016409118] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Regenerative failure in the mammalian optic nerve is generally attributed to axotomy-induced retinal ganglion cell (RGC) death, an insufficient intrinsic regenerative capacity, and an extrinsic inhibitory environment. Here, we show that a chemoattractive CXCL12/CXCR4-dependent mechanism prevents the extension of growth-stimulated axons into the distal nerve. The chemokine CXCL12 is chemoattractive toward axonal growth cones in an inhibitory environment, and these effects are entirely abolished by the specific knockout of its receptor, CXCR4 (CXCR4-/-), in cultured regenerating RGCs. Notably, 8% of naïve RGCs express CXCL12 and transport the chemokine along their axons in the nerve. Thus, axotomy causes its release at the injury site. However, most osteopontin-positive α-RGCs, the main neuronal population that survives optic nerve injury, express CXCR4 instead. Thus, CXCL12-mediated attraction prevents growth-stimulated axons from regenerating distally in the nerve, indicated by axons returning to the lesion site. Accordingly, specific depletion of CXCR4 in RGC reduces aberrant axonal growth and enables long-distance regeneration. Likewise, CXCL12 knockout in RGCs fully mimics these CXCR4-/- effects. Thus, active CXCL12/CXCR4-mediated entrapment of regenerating axons to the injury site contributes to regenerative failure in the optic nerve.
Collapse
Affiliation(s)
- Alexander M Hilla
- Department of Cell Physiology, Faculty of Biology and Biotechnology, Ruhr University, 44780 Bochum, Germany
| | - Annemarie Baehr
- Department of Cell Physiology, Faculty of Biology and Biotechnology, Ruhr University, 44780 Bochum, Germany
| | - Marco Leibinger
- Department of Cell Physiology, Faculty of Biology and Biotechnology, Ruhr University, 44780 Bochum, Germany
| | - Anastasia Andreadaki
- Department of Cell Physiology, Faculty of Biology and Biotechnology, Ruhr University, 44780 Bochum, Germany
| | - Dietmar Fischer
- Department of Cell Physiology, Faculty of Biology and Biotechnology, Ruhr University, 44780 Bochum, Germany
| |
Collapse
|
42
|
Extracellular CIRP Activates the IL-6Rα/STAT3/Cdk5 Pathway in Neurons. Mol Neurobiol 2021; 58:3628-3640. [PMID: 33783711 PMCID: PMC10404139 DOI: 10.1007/s12035-021-02368-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 03/22/2021] [Indexed: 10/21/2022]
Abstract
Extracellular cold-inducible RNA-binding protein (eCIRP) stimulates microglial inflammation causing neuronal damage during ischemic stroke and is a critical mediator of alcohol-induced cognitive impairment. However, the precise role of eCIRP in mediating neuroinflammation remains unknown. In this study, we report that eCIRP activates neurotoxic cyclin-dependent kinase-5 (Cdk5)/p25 through the induction of IL-6Rα/STAT3 pathway in neurons. Amyloid β (Aβ)-mediated neuronal stress, which is associated with Alzheimer's disease, increased the levels of eCIRP released from BV2 microglial cells. The released eCIRP levels from BV2 cells increased 3.2-fold upon stimulation with conditioned medium from Neuro-2a (N2a) cells containing Aβ compared to control N2a supernatant in a time-dependent manner. Stimulation of N2a cells and primary neurons with eCIRP upregulated the neuronal Cdk5 activator p25 expression in a dose- and time-dependent manner. eCIRP directly induced neuronal STAT3 phosphorylation and p25 increase via its novel receptor IL-6Rα. Next, we showed using surface plasmon resonance that eCIRP-derived peptide C23 inhibited the binding of eCIRP to IL-6Rα at 25 μM, with a 40-fold increase in equilibrium dissociation constant (Kd) value (from 8.08 × 10-8 M to 3.43 × 10-6 M), and completely abrogated the binding at 50 μM. Finally, C23 reversed the eCIRP-induced increase in neuronal STAT3 phosphorylation and p25 levels. In conclusion, the current study demonstrates that the upregulation of neuronal IL-6Rα/STAT3/Cdk5 pathway is a key mechanism of eCIRP's role in neuroinflammation and that C23 as a potent inhibitor of this pathway has translational potential in neurodegenerative pathologies controlled by eCIRP.
Collapse
|
43
|
Lindborg JA, Tran NM, Chenette DM, DeLuca K, Foli Y, Kannan R, Sekine Y, Wang X, Wollan M, Kim IJ, Sanes JR, Strittmatter SM. Optic nerve regeneration screen identifies multiple genes restricting adult neural repair. Cell Rep 2021; 34:108777. [PMID: 33657370 PMCID: PMC8009559 DOI: 10.1016/j.celrep.2021.108777] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 12/21/2020] [Accepted: 01/29/2021] [Indexed: 12/22/2022] Open
Abstract
Adult mammalian central nervous system (CNS) trauma interrupts neural networks and, because axonal regeneration is minimal, neurological deficits persist. Repair via axonal growth is limited by extracellular inhibitors and cell-autonomous factors. Based on results from a screen in vitro, we evaluate nearly 400 genes through a large-scale in vivo regeneration screen. Suppression of 40 genes using viral-driven short hairpin RNAs (shRNAs) promotes retinal ganglion cell (RGC) axon regeneration after optic nerve crush (ONC), and most are validated by separate CRISPR-Cas9 editing experiments. Expression of these axon-regeneration-suppressing genes is not significantly altered by axotomy. Among regeneration-limiting genes, loss of the interleukin 22 (IL-22) cytokine allows an early, yet transient, inflammatory response in the retina after injury. Reduced IL-22 drives concurrent activation of signal transducer and activator of transcription 3 (Stat3) and dual leucine zipper kinase (DLK) pathways and upregulation of multiple neuron-intrinsic regeneration-associated genes (RAGs). Including IL-22, our screen identifies dozens of genes that limit CNS regeneration. Suppression of these genes in the context of axonal damage could support improved neural repair.
Collapse
Affiliation(s)
- Jane A Lindborg
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Nicholas M Tran
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA
| | - Devon M Chenette
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Kristin DeLuca
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Yram Foli
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Ramakrishnan Kannan
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Yuichi Sekine
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Xingxing Wang
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Marius Wollan
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT 06536, USA
| | - In-Jung Kim
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Joshua R Sanes
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA
| | - Stephen M Strittmatter
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT 06536, USA.
| |
Collapse
|
44
|
Kaarijärvi R, Kaljunen H, Ketola K. Molecular and Functional Links between Neurodevelopmental Processes and Treatment-Induced Neuroendocrine Plasticity in Prostate Cancer Progression. Cancers (Basel) 2021; 13:cancers13040692. [PMID: 33572108 PMCID: PMC7915380 DOI: 10.3390/cancers13040692] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 02/04/2021] [Accepted: 02/05/2021] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Treatment-induced neuroendocrine prostate cancer (t-NEPC) is a subtype of castration-resistant prostate cancer (CRPC) which develops under prolonged androgen deprivation therapy. The mechanisms and pathways underlying the t-NEPC are still poorly understood and there are no effective treatments available. Here, we summarize the literature on the molecules and pathways contributing to neuroendocrine phenotype in prostate cancer in the context of their known cellular neurodevelopmental processes. We also discuss the role of tumor microenvironment in neuroendocrine plasticity, future directions, and therapeutic options under clinical investigation for neuroendocrine prostate cancer. Abstract Neuroendocrine plasticity and treatment-induced neuroendocrine phenotypes have recently been proposed as important resistance mechanisms underlying prostate cancer progression. Treatment-induced neuroendocrine prostate cancer (t-NEPC) is highly aggressive subtype of castration-resistant prostate cancer which develops for one fifth of patients under prolonged androgen deprivation. In recent years, understanding of molecular features and phenotypic changes in neuroendocrine plasticity has been grown. However, there are still fundamental questions to be answered in this emerging research field, for example, why and how do the prostate cancer treatment-resistant cells acquire neuron-like phenotype. The advantages of the phenotypic change and the role of tumor microenvironment in controlling cellular plasticity and in the emergence of treatment-resistant aggressive forms of prostate cancer is mostly unknown. Here, we discuss the molecular and functional links between neurodevelopmental processes and treatment-induced neuroendocrine plasticity in prostate cancer progression and treatment resistance. We provide an overview of the emergence of neurite-like cells in neuroendocrine prostate cancer cells and whether the reported t-NEPC pathways and proteins relate to neurodevelopmental processes like neurogenesis and axonogenesis during the development of treatment resistance. We also discuss emerging novel therapeutic targets modulating neuroendocrine plasticity.
Collapse
|
45
|
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: 3.3] [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
|
46
|
Transneuronal delivery of hyper-interleukin-6 enables functional recovery after severe spinal cord injury in mice. Nat Commun 2021; 12:391. [PMID: 33452250 PMCID: PMC7810685 DOI: 10.1038/s41467-020-20112-4] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Accepted: 11/13/2020] [Indexed: 12/27/2022] Open
Abstract
Spinal cord injury (SCI) often causes severe and permanent disabilities due to the regenerative failure of severed axons. Here we report significant locomotor recovery of both hindlimbs after a complete spinal cord crush. This is achieved by the unilateral transduction of cortical motoneurons with an AAV expressing hyper-IL-6 (hIL-6), a potent designer cytokine stimulating JAK/STAT3 signaling and axon regeneration. We find collaterals of these AAV-transduced motoneurons projecting to serotonergic neurons in both sides of the raphe nuclei. Hence, the transduction of cortical neurons facilitates the axonal transport and release of hIL-6 at innervated neurons in the brain stem. Therefore, this transneuronal delivery of hIL-6 promotes the regeneration of corticospinal and raphespinal fibers after injury, with the latter being essential for hIL-6-induced functional recovery. Thus, transneuronal delivery enables regenerative stimulation of neurons in the deep brain stem that are otherwise challenging to access, yet highly relevant for functional recovery after SCI.
Collapse
|
47
|
Terheyden L, Roider J, Klettner A. Basolateral activation with TLR agonists induces polarized cytokine release and reduces barrier function in RPE in vitro. Graefes Arch Clin Exp Ophthalmol 2021; 259:413-424. [PMID: 32949301 PMCID: PMC7843481 DOI: 10.1007/s00417-020-04930-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 08/24/2020] [Accepted: 09/10/2020] [Indexed: 01/18/2023] Open
Abstract
PURPOSE Systemic inflammation may be of importance in the development of AMD. RPE cells can recognize danger signals with toll-like receptors (TLR) and may react in a pro-inflammatory manner. In this study, we evaluated the basal and apical secretions of TNFα, IL-6, and IL-1β in primary RPE cells and RPE/choroid explant cells under basolateral stimulation of TLR2, 3, and 4; the effects on barrier function; and their influence on neuronal cell viability. METHODS RPE/choroid tissue explants were prepared from porcine eyes and cultivated in modified Ussing chambers; primary porcine RPE cells on transwell plates. Cells were basally stimulated with agonists Pam2CSK4 (Pam; TLR2), polyinosinic/polycytidylic acid (Poly I:C; TLR3), and lipopolysaccharide (LPS; TLR4) for 24 h. Supernatants were evaluated with ELISA for cytokines TNFα, IL-6, and IL-1β. Apical supernatants were applied to SHSY-5Y cells, and cell viability was evaluated in MTT assay. Barrier function was tested by measuring transepithelial electrical resistance (TER) and occludin immunostaining. RESULTS None of the tested TLR agonists was toxic on RPE cells after 24 h of exposure. Unstimulated RPE cells secreted hardly any cytokines. Pam induced IL-6, IL-1ß, and TNFα on the basal and apical sides at all concentrations tested. Poly I:C induced IL-6 and TNFα primarily at the basal side at lower but on both sides at higher concentrations. LPS induced IL-6, IL-1ß, and TNFα apically and basally at all concentrations tested. In the RPE/choroid, a strong difference between apical and basal secretions could be found. IL-6 was constitutively secreted basally, but not apically, but was induced by all agonists on both sides. IL-1ß and TNFα alpha were strongly induced on the basal side by all agonists. TER was reduced by all agonists, with Pam and LPS being effective in all concentrations tested. Occludin expression was unaltered, but the distribution was influenced by the agonists, with a less distinct localization at the cell borders after treatment. None of the agonists or supernatants of treated RPE and RPE/choroid organ cultures exerted any effect on viability of SHSY-5Y cells. CONCLUSIONS Danger signals activating TLRs can induce polarized cytokine expression and contribute to the loss of barrier function in the RPE.
Collapse
Affiliation(s)
- Laura Terheyden
- grid.9764.c0000 0001 2153 9986University Medical Center, Department of Ophthalmology, University of Kiel, Arnold-Heller-Str. 3, Haus B2, 24105 Kiel, Germany
| | - Johann Roider
- grid.9764.c0000 0001 2153 9986University Medical Center, Department of Ophthalmology, University of Kiel, Arnold-Heller-Str. 3, Haus B2, 24105 Kiel, Germany
| | - Alexa Klettner
- grid.9764.c0000 0001 2153 9986University Medical Center, Department of Ophthalmology, University of Kiel, Arnold-Heller-Str. 3, Haus B2, 24105 Kiel, Germany
| |
Collapse
|
48
|
Punyte V, Vilkeviciute A, Gedvilaite G, Kriauciuniene L, Liutkeviciene R. Association of VEGFA, TIMP-3, and IL-6 gene polymorphisms with predisposition to optic neuritis and optic neuritis with multiple sclerosis. Ophthalmic Genet 2020; 42:35-44. [PMID: 33121296 DOI: 10.1080/13816810.2020.1839916] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
BACKGROUND The etiology of the inflammatory ON is multifactorial. Much attention is paid to the inflammatory and immune processes that are likely to contribute to the demyelination and MS development. IL-6, VEGFA, and TIMP-3 genes are thought to be involved in the inflammatory processes and manifestation of CNS demyelination, so we aimed to determine the relationship between VEGFA rs1413711, TIMP-3 rs9621532, IL-6 rs1800796 gene polymorphisms and ON, and ON with MS. MATERIALS AND METHODS Patients with ON, ON with MS, and a random sample of healthy population were enrolled. The genotyping of VEGFA rs1413711, TIMP-3 rs9621532, and IL-6 rs1800796 polymorphisms was carried out using the real-time polymerase chain reaction method. RESULTS T/C and C/C genotypes of VEGFA rs1413711 were associated with about threefold increased odds of developing ON in the dominant and codominant models. Each allele C at VEGFA rs1413711 was associated with 1.7-fold increased odds of ON development. IL-6 rs1800796 allele C was more frequent in the ON with MS group compared to the control: 17.6% vs. 7.5%, respectively (p = .040). No statistically significant associations were found between TIMP-3 rs9621532 and the ON development. CONCLUSION: VEGFA rs1413711 is associated with the ON development.
Collapse
Affiliation(s)
- Vaida Punyte
- Lithuanian University of Health Sciences, Medical Academy , Kaunas, Lithuania
| | - Alvita Vilkeviciute
- Lithuanian University of Health Sciences, Medical Academy , Kaunas, Lithuania.,Neuroscience Institute, Lithuanian University of Health Sciences, Medical Academy , Kaunas, Lithuania
| | - Greta Gedvilaite
- Lithuanian University of Health Sciences, Medical Academy , Kaunas, Lithuania.,Neuroscience Institute, Lithuanian University of Health Sciences, Medical Academy , Kaunas, Lithuania
| | - Loresa Kriauciuniene
- Lithuanian University of Health Sciences, Medical Academy , Kaunas, Lithuania.,Neuroscience Institute, Lithuanian University of Health Sciences, Medical Academy , Kaunas, Lithuania
| | - Rasa Liutkeviciene
- Lithuanian University of Health Sciences, Medical Academy , Kaunas, Lithuania.,Neuroscience Institute, Lithuanian University of Health Sciences, Medical Academy , Kaunas, Lithuania
| |
Collapse
|
49
|
Teixeira-Pinheiro LC, Toledo MF, Nascimento-Dos-Santos G, Mendez-Otero R, Mesentier-Louro LA, Santiago MF. Paracrine signaling of human mesenchymal stem cell modulates retinal microglia population number and phenotype in vitro. Exp Eye Res 2020; 200:108212. [PMID: 32910940 DOI: 10.1016/j.exer.2020.108212] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 08/13/2020] [Accepted: 08/31/2020] [Indexed: 01/13/2023]
Abstract
PURPOSE Cellular therapy with mesenchymal stem cells (MSC) is emerging as an effective option to treat optic neuropathies. In models of retinal degeneration, MSC injected in the vitreous body protects injured retinal ganglion cells and stimulate their regeneration, however the mechanism is still unknown. Considering the immunomodulating proprieties of MSC and the controversial role of microglial contribution on retinal regeneration, we developed an in vitro co-culture model to analyze the effect of MSC on retinal microglia population. METHODS We used whole adult rat retinal explants in co-culture with human Wharton's jelly mesenchymal stem cells (hMSC) separated by a transwell membrane and analyzed hMSC effect on both retinal ganglion cells (RGCs) and retinal microglia. RESULTS hMSC in co-culture protected RGCs after 3 days in vitro by paracrine signaling. In addition, hMSC reduced microglia population and inhibited the pro-inflammatory phenotype of the remaining microglia. CONCLUSIONS Using a co-culture model, we demonstrated the paracrine effect of hMSC on RGC survival after injury concomitant with a reduction of microglial population. Paracrine signaling of hMSC also changed microglia phenotype and the expression of antiinflammatory factors in the retina. Our results are consistent with a detrimental effect of microglia on RGC survival and regeneration after injury.
Collapse
Affiliation(s)
- Leandro C Teixeira-Pinheiro
- Instituto de Biofisica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, RJ, 21941-902, Brazil; Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa, Brazil.
| | - Maria F Toledo
- Instituto de Biofisica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, RJ, 21941-902, Brazil; Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa, Brazil
| | | | - Rosalia Mendez-Otero
- Instituto de Biofisica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, RJ, 21941-902, Brazil; Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa, Brazil
| | - Louise A Mesentier-Louro
- Instituto de Biofisica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, RJ, 21941-902, Brazil; Department of Ophthalmology, Stanford University, Palo Alto, USA
| | - Marcelo F Santiago
- Instituto de Biofisica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, RJ, 21941-902, Brazil; Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa, Brazil
| |
Collapse
|
50
|
Klettner A, Brinkmann A, Winkelmann K, Käckenmeister T, Hildebrandt J, Roider J. Effect of long-term inflammation on viability and function of RPE cells. Exp Eye Res 2020; 200:108214. [PMID: 32898511 DOI: 10.1016/j.exer.2020.108214] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 08/12/2020] [Accepted: 09/01/2020] [Indexed: 12/20/2022]
Abstract
PURPOSE Degenerative ocular disorders like age-related macular degeneration (AMD) are associated with long-term pro-inflammatory signals on retinal pigment epithelial (RPE) cells. In this study, we investigated the effect of long term treatment of RPE cells with agonists of toll-like receptor (TLR) -3 (Polyinosinic:polycytidylic acid, Poly I:C), TLR-4 (lipopolysaccharide, LPS) and the pro-inflammatory cytokine TNFα. METHODS All tests were conducted with primary porcine RPE. Cells were stimulated with Poly I:C (1, 10, 100 μg/ml), LPS (0.1, 1, 10 μg/ml) or TNFα (12.5, 25 or 50 ng/ml) for 1 day, 7 days or 4 weeks. Cell viability tests (MTT) were additionally tested in ARPE-19 cells. Cytokine secretion (IL-6, IL-1β, IL-8, TNFα, TGF-β) was tested in ELISA, phagocytosis in a microscopic assay, and expression of RPE65 in Western blot. Barrier function was tested in transwell-cultured cells by measuring transepithelial resistance for up to 3 days. RESULTS LPS and TNFα significantly reduce cell viability after 1 day and 7 days, Poly I:C after 7 days and 4 weeks. LPS, Poly I:C and TNFα significantly induce the secretion of IL-6 and IL-8 at all tested time points. IL-1β is increased by LPS and Poly I:C after 1 day, but not by TNFα. TNFα secretion is increased by Poly I:C and LPS after 1 day but not at later time points. TGF-β secretion is not influenced by any stimulus. Concerning RPE function, LPS decreased phagocytosis after 7 days, while Poly I:C and TNFα showed no effect. RPE65 expression was strongly reduced by TNFα and LPS after 4 weeks. Wound healing capacity was reduced by Poly I:C but induced by LPS after 7 d and 4 w. Barrier function was not affected by Poly I:C or LPS, while TNFα reduced barrier function after 1 h, 4 h and 3 days. CONCLUSION Long term pro-inflammatory stimuli reduce RPE viability, barrier properties and cellular function and induce pro-inflammatory cytokines and therefore may contribute directly to atrophic changes in AMD.
Collapse
Affiliation(s)
- Alexa Klettner
- University of Kiel, University Medical Center, Department of Ophthalmology, Arnold-Heller-Str. 3, Haus B2, 24105, Kiel, Germany.
| | - Anna Brinkmann
- University of Kiel, University Medical Center, Department of Ophthalmology, Arnold-Heller-Str. 3, Haus B2, 24105, Kiel, Germany
| | - Katrin Winkelmann
- University of Kiel, University Medical Center, Department of Ophthalmology, Arnold-Heller-Str. 3, Haus B2, 24105, Kiel, Germany
| | - Tom Käckenmeister
- University of Kiel, University Medical Center, Department of Ophthalmology, Arnold-Heller-Str. 3, Haus B2, 24105, Kiel, Germany
| | - Julia Hildebrandt
- University of Kiel, University Medical Center, Department of Ophthalmology, Arnold-Heller-Str. 3, Haus B2, 24105, Kiel, Germany
| | - Johann Roider
- University of Kiel, University Medical Center, Department of Ophthalmology, Arnold-Heller-Str. 3, Haus B2, 24105, Kiel, Germany
| |
Collapse
|