Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Mar 1.
Published in final edited form as: J Immunol Regen Med. 2019 Nov 5;7:100023. doi: 10.1016/j.regen.2019.100023

The role of the immune system during regeneration of the central nervous system

KZ Sabin 1, K Echeverri 1
PMCID: PMC7451963  NIHMSID: NIHMS1545897  PMID: 32864529

Abstract

Central nervous system damage in mammals leads to neuronal cell death, axonal degeneration, and formation of a glial scar resulting in functional and behavioral defects. Other vertebrates, like fish and salamanders, have retained the ability to functionally regenerate after central nervous system injury. To date research from many research organisms has led to a more concise understanding of the response of local neural cells to injury. However, it has become clear that non-neural cells of the immune system play an important role in determining the tissue response to injury. In this review we briefly consider the mammalian response to injury compared to organisms with the natural ability to regenerate. We then discuss similarities and differences in how cells of the innate and adaptive immune system respond and contribute to tissue repair in various species.

1. Introduction

Humans are unable to functionally regenerate after central nervous system (CNS) traumas. Various animal models (i.e. mice and rats) of mechanical or contusive injury to the brain and spinal cord have provided a wealth of information regarding our inability to regenerate lost or damaged CNS tissue. However, we still do not know how to therapeutically stimulate robust functional recovery. This is due, in part, to the complexity of CNS traumas, which involves multiple cell types, both neural and non-neural, axonal degeneration, and formation of a growth inhibitory glial scar. Remarkably, other vertebrates, such as fish, frogs, and salamanders, are capable of regenerating a fully functional CNS after injury. Functional CNS regeneration in these species is characterized by the lack of reactive gliosis or a glial scar, robust axon regeneration, and a pro-regenerative glial cell response. Better understanding of the mechanisms utilized by naturally regenerating species could identify particular processes or pathways for therapeutic intervention.

Apart from the endogenous neural response to CNS trauma, both the innate and adaptive immune system play an important role in regulating a tissues response to injury. Immediately following injury, dead and necrotic cells release damage associated molecular patterns (DAMPs), reactive oxygen species (ROS), purine metabolites (ATP), mitochondrial components, and diverse polysaccharide components 1, 2 These factors bind to pattern recognition receptors (PRRs) (i.e. Toll-like receptors (TLRs) or P2X7) on the surface of neighboring cells, including resident immune cells, which stimulates the release of cytokines that are responsible for mobilizing an immune response 3 While this immune response is critical to protect the host from wound-related infections, functional studies have shown that this cascade of events also directly regulates the regenerative outcome.

In this review, we will briefly discuss the different response to CNS trauma in several species, including mice, zebrafish, and salamanders. We will then focus on the role of the immune system following injury. Specifically, on the role of cells of the innate immune system, the molecules they secrete, and how that influences the response of local neural cells. Finally, we will review the poorly understood and emerging role of the adaptive immune system during CNS regeneration.

2. Vertebrates Response to Central Nervous System Damage

2.1. Spinal Cord Regeneration in Naturally Regenerating Organisms

Some vertebrates, like fish, frogs, and salamanders, have the remarkable ability to regenerate the CNS after injury 4. Specifically, after spinal cord injury there is robust axon regeneration, which results in function restoration. Early retrograde tracing approaches in zebrafish and salamanders identified specific brain regions that project axons to the spinal cord. Following SCI, every spinal cord projecting brain region regenerates axons through the lesion. However, in zebrafish the organization of the regenerated axon tracts is slightly different from the pre-injury organization 5-7. Instead of regenerating through the white matter, axons preferentially migrate through the spinal cord grey matter before making connections with cells caudal to the lesion 8. Further investigations into the regenerative potential of neuronal subpopulations using transgenic reporter animals confirmed that the organization of regenerated TH+ (dopaminergic) and 5-HT+ (serotonergic) axons is drastically different compared to uninjured spinal cords 9. Interestingly, dopamine released from regenerating TH+ axons directly regulated the neural stem cell response to injury and promoted motor neuron regeneration in the zebrafish spinal cord 10.

In addition to extensive axon regeneration, naturally regenerating organisms exhibit a pro-regenerative glial cell response to injury. After injury glial cells adjacent to the lesion proliferate and migrate to repair the damaged spinal cord and create a permissive environment for axon regeneration 11-14. After spinal cord injury in zebrafish, GFAP+ glial cells adjacent to the injury adopt a bipolar, elongated morphology and form a glial bridge connecting the rostral and caudal stumps 13, 15. Formation of the glial bridge is dependent on FGF and connective tissue growth factor (CTGF) signaling, and overexpression of CTGF is sufficient to speed up bridge formation and functional recovery 13, 15 Regenerating axons use the glial bridge as a substrate to migrate across the lesion and restore motor function. Whether these same signaling mechanisms regulate glial bridge formation in lamprey is not clear.

In contrast to zebrafish, GFAP+ glial cells in the salamander spinal cord do not form a glial bridge. Early reports show that after spinal cord transection, glial cells seal over the rostral and caudal stumps and migrate towards each other 11, 12, 14, 16. A detailed characterization of spinal cord regeneration in newts suggests that glial cells and regenerating axons are intimately associated with each other throughout the regeneration process 14. This is slightly different than in zebrafish where the glial bridge forms first and then axons regenerate over the bridge and through the lesion. Regardless of the mechanism, the overall conclusion remains similar: glial cells adjacent to the lesion undergo a pro-regenerative response to injury, function to reconnect the spinal cord, and create a permissive/instructive environment for axon regeneration.

More recent molecular analyses have identified several signaling pathways that are important for the pro-regenerative glial cell response. Following spinal cord transection or tail amputation in zebrafish and salamanders classical developmental signaling molecules like Wnt/PCP, FGF, Notch, Sonic Hedgehog, and Sox2 play critical roles in regulating the pro-regenerative glial cell response, spinal cord patterning, asymmetric neural stem cell division, and neuronal differentiation 17-23. Additionally, microRNAs play a critical role in promoting spinal cord regeneration after spinal cord transection and tail amputation. Inhibition of a subset of these microRNAs results in defects in glial cell proliferation, spinal cord patterning, ependymal bulb formation, and expression of various axon guidance molecules or transcription factors 24-27. Recently, it was shown that axolotl glial cells up-regulate the non-canonical AP-1cFos/JunB after spinal cord transection, which functions to promote regeneration and inhibit glial scar formation 26. This is in contrast to damaged mammalian astrocytes, which express the canonical AP-1cFos/cJun, which functions to promote reactive gliosis and glial scar formation 28-30. How changes in the composition of the AP-1 transcriptional complex results in drastically different cellular responses to injury is still not clear but remains an intriguing question.

2.2. Mammalian Response to CNS Damage

Damage to the mammalian CNS results in neuronal cell death, axon degeneration, and formation of a glial and fibrotic scar that collectively lead to behavioral defects (Figure 1). There are many factors that contribute to regenerative failure in mammals, including the inability of neurons to initiate new axon growth 31-34, as well as a lesion microenvironment that inhibits axon regeneration 35-38. Following axon damage there is unrestricted influx of calcium into the axoplasm and breakdown of the cytoskeletal components 34, 39-41. These events result in massive cytoskeletal rearrangement, activation of various signaling pathways, and proteases, such as calpains 39, 41, 42. Activation of these pathways in response to axon damage are important for sealing the damaged axon and supporting growth cone formation in organisms with the innate ability to regenerate, like Aplysia and the sea lamprey 34, 40, 43. However, in mammals these early signaling events fail to induce formation of new growth cones and instead result in formation of dystrophic end bulbs 39. It was historically thought that dystrophic end bulbs were stagnant structures but actually there is persistent and highly dynamic remodeling of the actin and microtubule cytoskeleton, even into the chronic stages of injury 32, 44-46. Remarkably, when exogenously supplied with neurotrophins up to a year after injury some dystrophic axons formed new growth cones and migrated through the lesion 32, 45. Collectively, these studies suggest that damaged mammalian axons can be induced to a regenerative response if given the correct signals.

Figure 1: Inflammatory signaling after spinal cord injury.

Figure 1:

Following spinal cord injury damaged axons degenerate, form dystrophic end bulbs, but largely fail to migrate across the lesion. Astrocytes adjacent to the injury undergo reactive gliosis and contribute to the formation of the glial scar. Dead and necrotic cells release DAMPs, which signal to local neural cells and recruit leukocytes into the lesion. Collectively, the interaction between local cells and leukocytes influences the CNS response to injury by regulating reactive gliosis, glial scar formation, and neuronal survival.

Apart from environmental cues, there are neuron intrinsic roadblocks to axon regeneration. A handful of molecules and pathways have been identified as modulators of axon regeneration after injury. Experimental activation of mTOR signaling or overexpression of various KLF transcription factors in corticospinal tract neurons promotes axon regeneration and sprouting after spinal cord injury 47-51. Additionally, overexpression of STAT3 in dorsal root ganglion neurons enhances regeneration of the centrally projecting axons 52. Finally, conditional knockout of suppressor of cytokine signaling 3 (SOCS3) in retinal ganglion cells promotes axon regeneration after optic nerve crush 53. However, the precise mechanisms by which these molecules stimulate axon regeneration remain unclear. An additional caveat to these experiments is that gene manipulation occurred prior to injury. An outstanding question in the field is whether and to what extent manipulation of these pathways after injury can active a regenerative response. Remarkably, forced activation of mTOR signaling in corticospinal tract neurons one year after injury is sufficient to stimulate axon regeneration through the lesion 54. While the regenerated axons formed synaptic structures caudal to the lesion whether there was a functional improvement was not addressed 54. How to ensure that regenerated axons form the appropriate connections with the right post-synaptic partner to restore lost function represents an additional level of complexity to the problem.

In addition to neuron intrinsic pathways, non-neuronal cells, including astrocytes, oligodendrocytes, microglia, and peripheral inflammatory cells exquisitely regulate the mammalian response to CNS damage 36, 37, 55-57. Injury in the CNS result in the formation of two distinct compartments: 1) a fibrotic lesion at the core of the injury, which is comprised of fibroblasts, inflammatory cells, endothelial cells, and growth inhibitory extracellular matrix35-38, 58 and 2) a glial scar comprised of reactive astrocytes 38, 58-60. Reactive astrocytes are the major glial cell type contributing to the glial scar. Following injury, astrocytes adjacent to the injury site proliferate, undergo reactive gliosis, up-regulate expression of the intermediate filament proteins glial fibrillary acidic protein (GFAP) 61-63 and vimentin 64, and migrate towards the lesion. The reactive astrocytes originate from pre-existing astrocytes that re-enter the cell cycle, proliferate, and migrate towards the lesion 29, 65, 66. Lineage tracing experiments suggest ependymal cells, a putative spinal cord neural stem cell population, could give rise to astrocytes after injury, which then undergo reactive gliosis and contribute to the glial scar 67, 68. Reactive astrocytes within the glial scar express axon growth inhibitory molecules such as chondroitin sulfate proteoglycans (CSPGs), semaphorins, and ephrins 36, 37.

While reactive astrocytes do secrete growth inhibitory molecules, recent reports using sophisticated cell ablation and conditional gene knockout approaches suggest a protective role for the glial scar. Ablation of proliferative, scar forming reactive astrocytes led to increased axonal degeneration, a failure to repair the blood brain barrier, and decreased functional recovery compared to wild type mice 66. The increased axonal degeneration was attributed to a massive influx of inflammatory cells that are normally restricted to the fibrotic scar at the lesion center 66. However, ablation of reactive astrocytes led to a breakdown of the glial barrier separating the fibrotic lesion core from surrounding spinal cord tissue 66, allowing the aberrant infiltration of inflammatory cells into the spinal cord parenchyma. Further analysis identified the transcription factor STAT3 as being necessary for induction of reactive gliosis in spinal cord astrocytes after injury 69. Specific knockout of STAT3 from astrocytes using a GFAP-Cre phenocopied the cell ablation experiments, further highlighting a potentially protective role of reactive astrocytes early after injury 69. While early work largely focused on the effect of reactive astrocytes on axon regeneration, it is now clear that reactive astrocytes also regulate and respond to the inflammatory response to CNS trauma. The interaction between astrocytes and inflammatory cells can have major consequences on the tissue response to injury. We will now briefly consider what is known about these interactions.

3. Reactive Astrocytes and Inflammatory Signaling

Traumatic brain and spinal cord injury results in the death of local cells and disruption of the blood brain barrier (BBB) 55, 56. These events result in dead and necrotic cells releasing various DAMPs, including ATP and the DNA scaffolding protein high mobility group B1 (HMGB1), into the surrounding environment. These DAMPs will diffuse away from the lesion and bind to Toll-like receptors and other PRRs on neighboring cells and enter into circulation to stimulate a systemic inflammatory response. While the subsequent inflammatory response is essential to protect against infection, it also plays a major role in regulating the tissue response to injury (Figure 1).

While immune cells primarily express PRRs they are also expressed by astrocytes 70, 71. Activation of PRR signaling in astrocytes by ATP or HMGB1 directly or indirectly activates pro-inflammatory transcriptional cascades, like NF-κB or STAT3. Subsequent NF-κB activation contributes to cytotoxic edema associated with traumatic brain injury 72. Paradoxically, NF-κB signaling also stimulates astrocytes to express key pro-survival, neurotrophic molecules and could also mediate a neuroprotective function 73.

In addition to regulating the astrocytic response to injury, PRRs can signal in local, non-neural cells as well. HMGB1 signaling in endothelial cells stimulates vascular remodeling and BBB repair after brain injury 74, 75. Additionally, HMGB1 induces IL-6 expression in microglia 76. Increased IL-6 results in STAT3 activation in astrocytes leading to the up-regulation of aquaporin 4, which further exacerbates trauma-associated edema 76. Interestingly, STAT3 is a key regulator of reactive gliosis and glial scar formation in astrocytes, suggesting STAT3 likely regulates multiple processes in astrocytes after injury 69. Other signaling molecules that function in astrocytes to promote reactive gliosis and glial scar formation are SOCS3 and NF-κB 77, 78. Intriguingly, these pathways play important roles in regulating and responding to inflammatory signals, suggesting that the inflammatory response and reactive gliosis are linked at the molecular level. Consistent with this hypothesis, recent reports have identified microglia as important regulators of the reactive phenotype.

Transcriptional profiling of reactive astrocytes after stroke or systemic lipopolysaccharide injections revealed distinct transcriptional differences between the resulting populations 79. Recently, 2 subtypes of reactive astrocytes were genetically and functionally described, where A1 reactive astrocytes were induced by classically activated microglia, were pro-inflammatory, and exhibited cytotoxic effects on neighboring neurons 80. The A1 phenotype could be induced in vivo and in vitro by exposing astrocytes to TNF-α, IL-1α, and the complement component C1q 80. Conversely, A2 reactive astrocytes were induced after ischemic injury and secreted neurotrophic and angiogenic factors that conferred a more neuroprotective effect 80. These studies suggest that different kinds of stimuli (i.e. ischemia, inflammation, mechanical injury, degeneration) could induce a unique reactive state in astrocytes. Whether reactive astrocytes have a predetermined state or could transition along a spectrum of phenotypes remains an intriguing question 57.

4. An Immunological Blueprint for Nervous System Regeneration

4.1. Neutrophils and mammalian nervous system response to injury

Neurological damage in mammals results in the loss of sensory, motor, and cognitive function. Historically, research has focused on elucidating the cellular and molecular mechanisms that lead to regenerative failure in neural tissue, such as why neurons are not replaced, why axons fail to regenerate, and how reactive astrocytes and oligodendrocytes contribute to the glial scar. However, in the last few decades it is clear that the non-neural immune system plays a key role in regulating the response of neural tissue to injury 81-84. After spinal cord injury or traumatic injury to the brain, neutrophils are the first inflammatory cell type to traffic to the lesion, appearing within hours, peaking around 24 hours, and largely cleared from the lesion 3-5 days post injury (Figure 2) 85-90.

Figure 2: Dynamics of Leukocyte Infiltration in various species after Spinal Cord or Brain Injury.

Figure 2:

Schematic representation of leukocyte infiltration after mammalian spinal cord injury91, 200, adult zebrafish spinal cord injury,8, 13 larval zebrafish spinal cord injury, 150 Axolotl spinal cord injury,151, 152 mammalian brain injury,84 adult zebrafish brain injury,135, 136, 201 and larval zebrafish brain injury 141.

In general, neutrophil function seems to inhibit nervous system regeneration and exacerbate tissue damage 91. After injury, local neural cells respond to DAMPs released from damaged cells and additionally release various signals that recruit neutrophils to the injury site. Experimental perturbation of some of these damage responsive pathways like, leukotriene B4, cAMP, NF-κB signaling, CXCR2, and myeloperoxidase signaling results in decreased neutrophil trafficking to the lesion and an overall better histological score following injury 92-95. In vitro experiments suggest that neutrophils exert neurotoxic effects on cultured neurons via cell-cell interactions and through the secretion of factors including MMP9, ROS, and TNF-α 96, 97. Knockout models of neutrophil elastase exhibited decreased edema and fewer apoptotic cells after traumatic brain injury 98. However there was no effect on neutrophil migration or MMP9 expression, and no long-term functional benefits.

One caveat of these experiments is that many of these genetic perturbations are not neutrophil specific, but instead target the entire myeloid lineage. Approaches utilizing antibody-based depletion of Ly6G/Gr-1+ neutrophils have reported a potentially beneficial role for neutrophils after spinal cord injury. Neutrophil depleted mice exhibit a much larger ROS burst after injury as well as decreased vasculogenesis and a heightened inflammatory response 99-101. These results suggest that after spinal cord injury, neutrophils could play a neuroprotective role by mitigating ROS and the inflammatory response while promoting new blood vessel formation. Whether neutrophil depletion resulted in long-term functional benefits after spinal cord injury remains unclear. In contrast, after brain injury, neutrophil depletion appears to be beneficial. Neutrophil depletion resulted in less edema, fewer apoptotic cells, and reduced macrophage/microglia activation in response to brain injury 102. However, as with elastase knockout, there were no long-term functional improvements.

4.2. Macrophages/Microglia and mammalian nervous system regeneration

Unlike neutrophil infiltration, the kinetics of macrophage/microglia migration is slightly different after spinal cord and brain injury. In the spinal cord, macrophages/microglia appear during the sub-acute phase 2-7 days post injury and can persist within the lesion months after the initial injury (Figure 2) 91, 103. Whether these chronically persistent macrophages are the result of continued monocyte trafficking or proliferation of lesion-associated macrophages is not clear 103. However, in the brain, macrophages/microglia appear 3-7 days after injury and are largely cleared by chronic stages of injury 82, 84, 104. When chronically activated macrophages/microglia persist around the lesion, this is often correlated with a poor cognitive prognosis 105, 106.

In mouse models of spinal cord injury, ablation of macrophages/microglia with Clodronate loaded liposomes resulted in improved motor recovery 107-109. However, macrophage ablation using thymidine kinase or diphtheria toxin receptor expression in CD11b+ myeloid cells did not result in improved repair after brain injury 110, 111. However, ablation of CD11b+ cells even in uninjured animals resulted in an inflammatory response making it difficult to draw definitive conclusions. Inhibition of macrophage/microglia migration to the lesion by blocking CCR2 does help to combat cognitive decline, suggesting an overall detrimental role during functional recovery 112-114. It is experimentally difficult to separate macrophages from microglia, leading many researchers to investigate the role that collective cell populations have on nervous tissue repair. However, attempts to differentiate between the two populations using various classification schemes suggest that peripheral macrophages and resident microglia do play distinct roles after nervous system injury 83, 115-117.

Utilizing bone marrow chimeric mice, peripheral macrophages were GFP labeled based on expression of the chemokine receptor CX3CR1, which allowed for differentiation between peripheral macrophages (GFP+) and microglia (GFP) 117-121. These approaches revealed that macrophages are enriched in the fibrotic center of the lesion while microglia are largely associated with the glial scar, which has a high concentration of chondroitin sulfate proteoglycans. Interestingly, microglia express CD44, which binds to CSPGs expressed by cells in the glial scar leading to the expression of neurotrophins and an overall neuroprotective effect 122. Activated microglia also secrete various cytokines that stimulate local astrocytes to undergo reactive gliosis and thus facilitates formation of the glial scar 123. Yet, this too could prove beneficial as the glial scar mitigates the acute inflammatory response to injury and prevents excessive secondary damage 66, 69, 124. However, during chronic phases of spinal cord injury the glial scar does represent a major barrier to axon regeneration and functional restoration. It is important to mention that Ly6hi/CX3CR1lo pro-inflammatory peripheral macrophages are also found in the glial scar, although the functional significance of this finding is not clear 103, 118, 121, 125.

Traumatic brain injury models in CX3CR1 knockout mice have fewer macrophages/microglia that infiltrate the lesion. This results in an early neuroprotective effect after injury but leads to the persistence of a chronic pro-inflammatory lesion, which is normally resolved in wild type animals 126. The chronic pro-inflammatory lesion results in increased neuronal cell death and decreased cognitive performance. This suggests that early inhibition of CX3CR1+ cell migration to the lesion could be beneficial following injury. However, these cells are necessary at later time points to resolve the inflammatory response and facilitate functional recovery. It is not clear whether the same or two functionally distinct cell populations mediate these opposing processes. Following spinal cord injury, there does appear to be two temporally distinct waves of peripheral macrophage infiltration. Inhibition of the later wave results in decreased functional recovery, suggesting this later population of macrophages is beneficial to repair 85. These differing results could be due to the heterogeneity of macrophage phenotypes 127.

Pro-inflammatory M1 macrophages are generally thought to be detrimental to regeneration while the anti-inflammatory M2 macrophage is thought to promote regeneration 128-130. While this system implies a binary cell state, accumulating evidence supports the notion that macrophage phenotypes reside on a spectrum between the M1 and M2 classification 131. M2 macrophages are present after spinal cord injury but only within the first 7 days and are quickly replaced by M1 macrophages, which persist into chronic stages 132. These results suggest that the chronic persistence of M1 macrophages could be responsible for a pro-inflammatory microenvironment that impedes functional recovery. Similarly, after traumatic brain injury there is an influx of M1 macrophages at 3 and 7 days post injury with a transient increase in M2 macrophages at 5 days post injury 133. Whether these dynamics are necessary to avoid catastrophic effects due to prolonged M1 or M2 signaling is not clear. Furthermore, it would be interesting to determine if activation of a more M2-like signature during chronic stages of nervous system repair would be sufficient to confer a more regenerative response.

4.3. Inflammation and functional nervous system regeneration

Recent studies in zebrafish are beginning to shed light on the role of various immune cell types during nervous system regeneration (Figure 2). An acute inflammatory response is necessary for zebrafish brain regeneration after stab injury or neuronal ablation 134-139. Inhibition of the inflammatory response leads to decreased neural stem cell proliferation and neuronal differentiation 136. Circulating macrophages are essential for the re-establishment of the blood brain barrier in response to damage 140. After stab injury in the larval zebrafish brain there is rapid recruitment of microglia to the lesion however tissue repair largely occurs in the absence of neutrophils 141. Intriguingly, microglia in the newt brain seem to play an inhibitory role during dopaminergic neuron regeneration following chemical ablation 142. Following neuronal ablation newts normally regenerate dopaminergic neurons within 30 days and this is dependent on neural stem cell differentiation 143, 144. Interestingly, inhibition of the microglial response to neuronal ablation results in more TH+ neurons 142. While inhibition of microglia does not affect neural stem cell proliferation, it does lead to fewer apoptotic neurons suggesting that microglia normally impede neuronal survival 142, 145.

Adult and larval zebrafish are able to regenerate spinal cord motor neurons after mechanical injury or chemical ablation 17, 146. Specific ablation of motor neurons using the nitroreductase system 147 stimulates a robust inflammatory response, which is necessary for motor neuron regeneration 148. The precise mechanism by which inflammatory cells stimulate the endogenous neural stem cells to differentiate into new motor neurons is still not clear. However, stab injury in the zebrafish brain leads to expression of the chemokine receptor CXCR5 in neural stem cells and is necessary for regenerative neurogenesis 149. This suggests that injury leads to differential expression of immune modulatory molecules, potentially sensitizing local cells to subsequent inflammatory signals to promote a regenerative response. While a similar reaction occurs after injury in mammals, this more often than not leads to a scarring or fibrotic response, however an important difference is the duration of the immune response in pro-regenerative species versus non-regenerative mammals.

The inflammatory response to injury also plays an important role in regulating axon regeneration after spinal cord injury. In larval zebrafish, there is a rapid and dynamic influx of macrophages/microglia and neutrophils to the lesion, which secrete TNF-α or IL-1β, respectively. Macrophages and macrophage-derived TNF-α is necessary for axon regeneration and neutrophil clearance. Genetic ablation of macrophages or CRISPR-mediated TNF-α knockout resulted in sustained neutrophil presence and chronic IL-1β signaling, which blocked axon regeneration 150. Leukocyte infiltration is slightly different in adult zebrafish where macrophages/microglia persist caudal to the lesion up to 2 weeks after injury and neutrophils are only rarely observed 8, 13.

Early work in the axolotl identified recruitment of macrophages or microglia to the lesion within 24 hours and persisted until 2 weeks post crush injury (Figure 2) 151. Whether these cells are necessary to promote regeneration is not known. A recent microarray analysis of axolotl spinal cord regeneration revealed that IL-1β is up-regulated transiently at 24 hours post injury, which appears to coincide with blood cell infiltration within the lesion 152. Whether these cells are macrophages, neutrophils, or some other cell type is not clear. While transient IL-1β signaling is crucial for zebrafish spinal cord regeneration, it is also important for fin regeneration suggesting an overall pro-regenerative effect 150, 153, 154. It would be interesting to determine if early and transient up-regulation of IL-1β in axolotl plays a similar pro-regenerative role. Furthermore, the zebrafish JunB homologue junbb is necessary for injury-induced up-regulation of IL-1β 153, 154. In axolotl, the non-canonical AP-1cFos/JunB is up-regulated in glial cells after spinal cord injury and functions to promote a pro-regenerative response 26. Whether this JunB-containing AP-1 complex similarly regulates IL-1β expression in axolotl glial cells is an interesting question.

5. The role of T cells during Regeneration

5.1. Effector lymphocytes during tissue repair

While a lot is known about how the innate immune system effects regeneration relatively little is known about the role of the adaptive immune system after injury 155, 156. In general, it seems that the contribution of T cells to the regenerative response is highly context dependent. For example, a contusive model of brain injury in RAG−/− mice, which lack T and B cells, showed that there is largely no effect on functional outcome 157. However, it is worth mentioning that there appeared to be fewer neutrophils and macrophages around the lesion area. Additionally, inhibition of T cell trafficking to the lesion by injection of the sphingosine-1-phosphate receptor agonist FTY720 similarly had no effect on functional restoration 158. However, after ischemia/reperfusion injury, RAG−/− and severe combined immunodeficiency (SCID) mice exhibit better neurological recovery 159-161. It has been proposed that after ischemia/reperfusion, canonical T cell cytokines like interferon-γ, IL-17, IL-21, and IL-23 could promote neuronal degeneration 156. However, it is not clear why lymphocytes would be beneficial after ischemia/reperfusion but have no effect after contusive brain injuries. These potential discrepancies could be addressed using experimental approaches that specifically ablate B cells or subsets of effector and regulatory T cells.

A specific role for CD4 T cells in providing neuroprotective signals has been reported after injury and motor neuron disease. The ability of CD4 T cells to confer a protective effect is largely due to secretion of IL-4, which stimulates expression of various neurotrophic factors in local neurons 162, 163. In this way, CD4 T cells delay axonal degeneration and mitigate neuronal apoptosis. Interestingly, many of the CD4 T cells that traffic to central nervous system lesions are autoreactive for particular antigens, like myelin-associated proteins 164-167. Injection of mice with autologous dendritic cells pulsed with myelin components stimulated a strong adaptive immune response and improved functional recovery after spinal cord injury 168, 169. While it is still not clear what the precise role of the adaptive immune system is after injury, it seems that therapeutic stimulation could elicit a pro-reparative response. Indeed, various approaches utilizing active and passive vaccination approaches are beginning to elucidate the molecular mechanisms that stimulate functional recovery after central nervous system injuries 170.

It is also becoming clear that T cells play an important role during mammalian wound healing. The skin has a complement of tissue resident T cells, which play integral roles during immune surveillance for malignant cells, viral, and bacterial infections 171. Resident skin T cells are a major source of insulin-like growth factor-1 (IGF-1), which promotes wound healing after injury 172. Apart from typical effector T cells, a subset of specialized T cells that express γ and δ T cell receptor chains are important for wound healing. The use of genetic mouse models deficient for γδ T cells revealed that this cell population is a critical source of keratinocyte growth factor, which is necessary for normal wound healing 173. Furthermore, wound beds deficient for γδ T cells have a higher proportion of myeloid-derived suppressor cells, fewer pro-regenerative M2 macrophages, and an altered cytokine profile 174. However the functional outcome of these altered cellular dynamics to injury has not been reported. Additionally, whether γδ T cells play a similar role during injury response in the CNS is not clear but remains an interesting area of research.

The potential contribution of the adaptive immune system to tissue regeneration in non-mammalian models is almost completely unexplored. It is known that zebrafish and salamanders possess similar compliments of adaptive immune cells relative to humans. This suggests that these cells are present and could potentiate a pro-regenerative response to injury. As discussed above, complete and functional tissue regeneration in salamanders and fish, occurs despite a robust and well-orchestrated inflammatory response. These observations highlight that differences likely exist in which leukocytes arrive when, how long they stay, where in the injury they reside, and the specific cytokine profiles (pro- versus anti-inflammatory) they present.

5.2. Regulatory T cells during tissue repair

Regulatory T (Tregs) cells have recently been implicated as major regulators of mammalian tissue regeneration that generally seem to play a pro-regenerative or protective role. Tregs are defined by the expression of the transcription factor FoxP3 and the high affinity IL-2 receptor subunit CD25 175. Tregs play important roles in regulating effector cell functions during inflammatory responses, help to prevent autoimmunity, and have an emerging role in promoting tissue regeneration 176-179. Similar to their role during immune responses, Tregs seem to promote tissue repair by directly regulating the inflammatory response to injury. For example, Tregs secrete TGF-β and IL-10, which mitigate the pro-inflammatory response to injury 180, 181. However, Tregs also release molecules that elicit a pro-reparative response from resident cells in the lesion. Specifically, Tregs secrete the epidermal growth factor-like molecule Amphiregulin, which promotes lung, brain, and muscle repair 182-184. Depletion of Tregs or conditional knockout of Amphiregulin in Tregs leads to a heightened pro-inflammatory response and decreased functional recovery 182-188. Additionally, in mammals Tregs recruited to sites of demyelination directly regulate oligodendrocyte progenitor cell differentiation and re-myelination by secretion of cellular communication network factor 3 (CCN3)189. Remarkably, Tregs directly regulate epithelial stem cell differentiation to stimulate hair follicle replacement after depilation 190. Whether Tregs similarly influence epithelial stem cell differentiation after full thickness wounding remains to be seen. Adoptive transfer of Tregs into RAG−/− mice or expansion of the Treg pool results in better functional recovery in a variety of injury models 184-188. Interestingly, epidermal growth factor receptor signaling in Tregs is necessary for normal cutaneous healing after full thickness wounds 191. However, it is not clear if Amphiregulin secreted by Tregs then signals in an autocrine manner though EGFR to potentiate the pro-regenerative functions of Tregs. Whether Tregs in mammals exert a similar regulatory role over neural stem cells or reactive astrocytes after CNS injuries is poorly understood but remains an interesting and exciting question.

Similar to mammals, zebrafish Tregs (zTregs) promote functional regeneration after a variety of injury models. zTregs traffic to injury sites in the spinal cord, heart, and retina and function to elicit pro-regenerative response from local cells. Interestingly, the expression of pro-regenerative molecules was tissue dependent, where zTregs up-regulated neurotrophin-3 in the damaged spinal cord, insulin-like growth factor-1 in the retina, and neuregulin-1 in the heart192. Xenopus tadpoles are able to regenerate their tails after amputation, however there is a developmental stage (st45-47) where they temporarily lose this ability193. One study suggests this refractory period coincides with the development of the immune system 194. Pharmacologic and genetic perturbation in immune system development is sufficient to improve regenerative out comes 194. Additionally, FoxP3 expression is highest during the post-refractory period suggesting that development of Tregs coincides with restoration of regenerative potential 194. It would be interesting to use cell transplantation and cell ablation approaches to determine if a causal relationship exists between Treg development and the restoration of regeneration.

6. Perspective

The precise function of the immune system during tissue repair and regeneration remains enigmatic. It is clear that the immune system exerts a strong regulatory role after injury and it is not simply the presence or absence of an inflammatory response that dictates the reparative outcome (i.e. functional regeneration versus scarring). Instead, the cytokine profile and spatiotemporal dynamics of leukocyte recruitment to a lesion likely play integral roles in determining functional outcome.

For example, after spinal cord injury in larval zebrafish neutrophils arrive first and then macrophages infiltrate the lesion, a process which is similar to mammals 91, 103, 150. However in zebrafish, both cell types are present in the lesion for a much shorter period of time than in mammals and express either IL-1β or TNF-α, respectively. In contrast, mammalian neutrophils and macrophages have both been reported to secrete IL-1β and TNF-α 91, 103. Admittedly, leukocyte recruitment may be different after spinal cord injury in adult versus larval zebrafish, as neutrophils were not commonly found in the injured adult spinal cord 13. This is reminiscent of larval zebrafish brain regeneration where microglia but not neutrophils infiltrate the lesion, which is in contrast to mammalian brain injury where neutrophils arrive first 84, 141.

In the future it will be important to conduct comparative studies across multiple species with varied regenerative abilities using standardized injury paradigms. The use of such approaches will provide valuable insight into the precise role that the immune system plays after injury. For example, the zebrafish is able to regenerate the heart after amputation but the medaka, a closely related species, is not 195-197. Compared to zebrafish, the medaka exhibits a blunted acute inflammatory response to injury 196. The inflammatory response was necessary for heart regeneration in the zebrafish and sufficient to stimulate a regenerative response in medaka 196. Additionally, the African spiny mouse (Acomys spp.) is able to regenerate ear hole punches that the standard Mus musculus cannot 198. Initial characterization of the inflammatory response to ear hole punch in Acomys and Mus revealed that both organisms undergo an inflammatory response to injury. However, the timing of leukocyte infiltration, the macrophage polarization state, and specific leukocyte localization in the lesion is different between the two species and likely contributes to the differences in regenerative outcome 199. Collectively, these studies highlight the importance of comparative analysis across closely related species with varied regenerative capabilities. As genome editing and conditional transgenesis techniques continue to improve, such comparative studies will become more feasible and shed light on the complex role of the immune system during tissue repair and regeneration.

Acknowledgements

The authors would like to thank Jessica Dewey helpful comments regarding the manuscript.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Conflicts of Interest

The authors have no conflicts of interest to report.

References:

  • 1.Alvarez K and Vasquez G, Damage-associated molecular patterns and their role as initiators of inflammatory and auto-immune signals in systemic lupus erythematosus. Int Rev Immunol, 2017. 36(5): p. 259–270. [DOI] [PubMed] [Google Scholar]
  • 2.Kubelkova K and Macela A, Innate Immune Recognition: An Issue More Complex Than Expected. Front Cell Infect Microbiol, 2019. 9: p. 241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Takeuchi O and Akira S, Pattern recognition receptors and inflammation. Cell, 2010. 140(6): p. 805–20. [DOI] [PubMed] [Google Scholar]
  • 4.Tanaka EM and Ferretti P, Considering the evolution of regeneration in the central nervous system. Nat Rev Neurosci, 2009. 10(10): p. 713–23. [DOI] [PubMed] [Google Scholar]
  • 5.Becker T, Bernhardt RR, Reinhard E, et al. , Readiness of zebrafish brain neurons to regenerate a spinal axon correlates with differential expression of specific cell recognition molecules. J Neurosci, 1998. 18(15): p. 5789–803. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Clarke JD, Alexander R, and Holder N, Regeneration of descending axons in the spinal cord of the axolotl. Neurosci Lett, 1988. 89(1): p. 1–6. [DOI] [PubMed] [Google Scholar]
  • 7.Becker T, Wullimann MF, Becker CG, et al. , Axonal regrowth after spinal cord transection in adult zebrafish. J Comp Neurol, 1997. 377(4): p. 577–95. [DOI] [PubMed] [Google Scholar]
  • 8.Becker T and Becker CG, Regenerating descending axons preferentially reroute to the gray matter in the presence of a general macrophage/microglial reaction caudal to a spinal transection in adult zebrafish. J Comp Neurol, 2001. 433(1): p. 131–47. [DOI] [PubMed] [Google Scholar]
  • 9.Kuscha V, Barreiro-Iglesias A, Becker CG, et al. , Plasticity of tyrosine hydroxylase and serotonergic systems in the regenerating spinal cord of adult zebrafish. J Comp Neurol, 2012. 520(5): p. 933–51. [DOI] [PubMed] [Google Scholar]
  • 10.Reimer MM, Norris A, Ohnmacht J, et al. , Dopamine from the brain promotes spinal motor neuron generation during development and adult regeneration. Dev Cell, 2013. 25(5): p. 478–91. [DOI] [PubMed] [Google Scholar]
  • 11.Butler EG and Ward MB, Reconstitution of the spinal cord following ablation in urodele larvae. J Exp Zool, 1965. 160(1): p. 47–65. [DOI] [PubMed] [Google Scholar]
  • 12.Butler EG and Ward MB, Reconstitution of the spinal cord after ablation in adult Triturus. Dev Biol, 1967. 15(5): p. 464–86. [DOI] [PubMed] [Google Scholar]
  • 13.Goldshmit Y, Sztal TE, Jusuf PR, et al. , Fgf-dependent glial cell bridges facilitate spinal cord regeneration in zebrafish. J Neurosci, 2012. 32(22): p. 7477–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Zukor KA, Kent DT, and Odelberg SJ, Meningeal cells and glia establish a permissive environment for axon regeneration after spinal cord injury in newts. Neural Dev, 2011. 6: p. 1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Mokalled MH, Patra C, Dickson AL, et al. , Injury-induced ctgfa directs glial bridging and spinal cord regeneration in zebrafish. Science, 2016. 354(6312): p. 630–634. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Sabin K, Santos-Ferreira T, Essig J, et al. , Dynamic membrane depolarization is an early regulator of ependymoglial cell response to spinal cord injury in axolotl. Dev Biol, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Becker CG and Becker T, Neuronal regeneration from ependymo-radial glial cells: cook, little pot, cook! Dev Cell, 2015. 32(4): p. 516–27. [DOI] [PubMed] [Google Scholar]
  • 18.Dias TB, Yang YJ, Ogai K, et al. , Notch signaling controls generation of motor neurons in the lesioned spinal cord of adult zebrafish. J Neurosci, 2012. 32(9): p. 3245–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Reimer MM, Kuscha V, Wyatt C, et al. , Sonic hedgehog is a polarized signal for motor neuron regeneration in adult zebrafish. J Neurosci, 2009. 29(48): p. 15073–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Rodrigo Albors A, Tazaki A, Rost F, et al. , Planar cell polarity-mediated induction of neural stem cell expansion during axolotl spinal cord regeneration. Elife, 2015. 4: p. e10230. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Schnapp E, Kragl M, Rubin L, et al. , Hedgehog signaling controls dorsoventral patterning, blastema cell proliferation and cartilage induction during axolotl tail regeneration. Development, 2005. 132(14): p. 3243–53. [DOI] [PubMed] [Google Scholar]
  • 22.Tazaki A, Tanaka EM, and Fei JF, Salamander spinal cord regeneration: The ultimate positive control in vertebrate spinal cord regeneration. Dev Biol, 2017. 432(1): p. 63–71. [DOI] [PubMed] [Google Scholar]
  • 23.Zhang F, Clarke JD, and Ferretti P, FGF-2 Up-regulation and proliferation of neural progenitors in the regenerating amphibian spinal cord in vivo. Dev Biol, 2000. 225(2): p. 381–91. [DOI] [PubMed] [Google Scholar]
  • 24.Diaz Quiroz JF, Tsai E, Coyle M, et al. , Precise control of miR-125b levels is required to create a regeneration-permissive environment after spinal cord injury: a cross-species comparison between salamander and rat. Dis Model Mech, 2014. 7(6): p. 601–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Gearhart MD, Erickson JR, Walsh A, et al. , Identification of Conserved and Novel MicroRNAs during Tail Regeneration in the Mexican Axolotl. Int J Mol Sci, 2015. 16(9): p. 22046–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Sabin KZ, Jiang P, Gearhart MD, et al. , AP-1(cFos/JunB)/miR-200a regulate the pro-regenerative glial cell response during axolotl spinal cord regeneration. Commun Biol, 2019. 2: p. 91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Sehm T, Sachse C, Frenzel C, et al. , miR-196 is an essential early-stage regulator of tail regeneration, upstream of key spinal cord patterning events. Dev Biol, 2009. 334(2): p. 468–80. [DOI] [PubMed] [Google Scholar]
  • 28.Cao Z, Wu XF, Peng Y, et al. , Scorpion Venom Heat-Resistant Peptide Attenuates Glial Fibrillary Acidic Protein Expression via c-Jun/AP-1. Cell Mol Neurobiol, 2015. 35(8): p. 1073–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Gadea A, Schinelli S, and Gallo V, Endothelin-1 regulates astrocyte proliferation and reactive gliosis via a JNK/c-Jun signaling pathway. J Neurosci, 2008. 28(10): p. 2394–408. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Gao K, Wang CR, Jiang F, et al. , Traumatic scratch injury in astrocytes triggers calcium influx to activate the JNK/c-Jun/AP-1 pathway and switch on GFAP expression. Glia, 2013. 61(12): p. 2063–77. [DOI] [PubMed] [Google Scholar]
  • 31.Ferguson TA and Son YJ, Extrinsic and intrinsic determinants of nerve regeneration. J Tissue Eng, 2011. 2(1): p. 2041731411418392. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Kadoya K, Tsukada S, Lu P, et al. , Combined intrinsic and extrinsic neuronal mechanisms facilitate bridging axonal regeneration one year after spinal cord injury. Neuron, 2009. 64(2): p. 165–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Liu K, Tedeschi A, Park KK, et al. , Neuronal intrinsic mechanisms of axon regeneration. Annu Rev Neurosci, 2011. 34: p. 131–52. [DOI] [PubMed] [Google Scholar]
  • 34.Mar FM, Bonni A, and Sousa MM, Cell intrinsic control of axon regeneration. EMBO Rep, 2014. 15(3): p. 254–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Diaz Quiroz JF and Echeverri K, Spinal cord regeneration: where fish, frogs and salamanders lead the way, can we follow? Biochem J, 2013. 451(3): p. 353–64. [DOI] [PubMed] [Google Scholar]
  • 36.Rolls A, Shechter R, and Schwartz M, The bright side of the glial scar in CNS repair. Nat Rev Neurosci, 2009. 10(3): p. 235–41. [DOI] [PubMed] [Google Scholar]
  • 37.Silver J and Miller JH, Regeneration beyond the glial scar. Nat Rev Neurosci, 2004. 5(2): p. 146–56. [DOI] [PubMed] [Google Scholar]
  • 38.Sofroniew MV, Molecular dissection of reactive astrogliosis and glial scar formation. Trends Neurosci, 2009. 32(12): p. 638–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Bradke F, Fawcett JW, and Spira ME, Assembly of a new growth cone after axotomy: the precursor to axon regeneration. Nat Rev Neurosci, 2012. 13(3): p. 183–93. [DOI] [PubMed] [Google Scholar]
  • 40.Ziv NE and Spira ME, Localized and transient elevations of intracellular Ca2+ induce the dedifferentiation of axonal segments into growth cones. J Neurosci, 1997. 17(10): p. 3568–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.He Z and Jin Y, Intrinsic Control of Axon Regeneration. Neuron, 2016. 90(3): p. 437–51. [DOI] [PubMed] [Google Scholar]
  • 42.Kamber D, Erez H, and Spira ME, Local calcium-dependent mechanisms determine whether a cut axonal end assembles a retarded endbulb or competent growth cone. Exp Neurol, 2009. 219(1): p. 112–25. [DOI] [PubMed] [Google Scholar]
  • 43.Jin LQ, Zhang G, Jamison C Jr., et al. , Axon regeneration in the absence of growth cones: acceleration by cyclic AMP. J Comp Neurol, 2009. 515(3): p. 295–312. [DOI] [PubMed] [Google Scholar]
  • 44.Houle JD, Demonstration of the potential for chronically injured neurons to regenerate axons into intraspinal peripheral nerve grafts. Exp Neurol, 1991. 113(1): p. 1–9. [DOI] [PubMed] [Google Scholar]
  • 45.Kobayashi NR, Fan DP, Giehl KM, et al. , BDNF and NT-4/5 prevent atrophy of rat rubrospinal neurons after cervical axotomy, stimulate GAP-43 and Talpha1-tubulin mRNA expression, and promote axonal regeneration. J Neurosci, 1997. 17(24): p. 9583–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Li Y and Raisman G, Sprouts from cut corticospinal axons persist in the presence of astrocytic scarring in long-term lesions of the adult rat spinal cord. Exp Neurol, 1995. 134(1): p. 102–11. [DOI] [PubMed] [Google Scholar]
  • 47.Blackmore MG, Wang Z, Lerch JK, et al. , Kruppel-like Factor 7 engineered for transcriptional activation promotes axon regeneration in the adult corticospinal tract. Proc Natl Acad Sci U S A, 2012. 109(19): p. 7517–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Liu K, Lu Y, Lee JK, et al. , PTEN deletion enhances the regenerative ability of adult corticospinal neurons. Nat Neurosci, 2010. 13(9): p. 1075–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Moore DL, Blackmore MG, Hu Y, et al. , KLF family members regulate intrinsic axon regeneration ability. Science, 2009. 326(5950): p. 298–301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Park KK, Liu K, Hu Y, et al. , Promoting axon regeneration in the adult CNS by modulation of the PTEN/mTOR pathway. Science, 2008. 322(5903): p. 963–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Zukor K, Belin S, Wang C, et al. , Short hairpin RNA against PTEN enhances regenerative growth of corticospinal tract axons after spinal cord injury. J Neurosci, 2013. 33(39): p. 15350–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Bareyre FM, Garzorz N, Lang C, et al. , In vivo imaging reveals a phase-specific role of STAT3 during central and peripheral nervous system axon regeneration. Proc Natl Acad Sci U S A, 2011. 108(15): p. 6282–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Smith PD, Sun F, Park KK, et al. , SOCS3 deletion promotes optic nerve regeneration in vivo. Neuron, 2009. 64(5): p. 617–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Du K, Zheng S, Zhang Q, et al. , Pten Deletion Promotes Regrowth of Corticospinal Tract Axons 1 Year after Spinal Cord Injury. J Neurosci, 2015. 35(26): p. 9754–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Burda JE, Bernstein AM, and Sofroniew MV, Astrocyte roles in traumatic brain injury. Exp Neurol, 2016. 275 Pt 3: p. 305–315. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Burda JE and Sofroniew MV, Reactive gliosis and the multicellular response to CNS damage and disease. Neuron, 2014. 81(2): p. 229–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Liddelow SA and Barres BA, Reactive Astrocytes: Production, Function, and Therapeutic Potential. Immunity, 2017. 46(6): p. 957–967. [DOI] [PubMed] [Google Scholar]
  • 58.Adams KL and Gallo V, The diversity and disparity of the glial scar. Nat Neurosci, 2018. 21(1): p. 9–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Faulkner JR, Herrmann JE, Woo MJ, et al. , Reactive astrocytes protect tissue and preserve function after spinal cord injury. J Neurosci, 2004. 24(9): p. 2143–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Voskuhl RR, Peterson RS, Song B, et al. , Reactive astrocytes form scar-like perivascular barriers to leukocytes during adaptive immune inflammation of the CNS. J Neurosci, 2009. 29(37): p. 11511–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Bignami A and Dahl D, Astrocyte-specific protein and neuroglial differentiation. An immunofluorescence study with antibodies to the glial fibrillary acidic protein. J Comp Neurol, 1974. 153(1): p. 27–38. [DOI] [PubMed] [Google Scholar]
  • 62.Eng LF, Glial fibrillary acidic protein (GFAP): the major protein of glial intermediate filaments in differentiated astrocytes. J Neuroimmunol, 1985. 8(4-6): p. 203–14. [DOI] [PubMed] [Google Scholar]
  • 63.Yang Z and Wang KK, Glial fibrillary acidic protein: from intermediate filament assembly and gliosis to neurobiomarker. Trends Neurosci, 2015. 38(6): p. 364–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Yang HY, Lieska N, Shao D, et al. , Proteins of the intermediate filament cytoskeleton as markers for astrocytes and human astrocytomas. Mol Chem Neuropathol, 1994. 21(2-3): p. 155–76. [DOI] [PubMed] [Google Scholar]
  • 65.Buffo A, Rite I, Tripathi P, et al. , Origin and progeny of reactive gliosis: A source of multipotent cells in the injured brain. Proc Natl Acad Sci U S A, 2008. 105(9): p. 3581–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Bush TG, Puvanachandra N, Horner CH, et al. , Leukocyte infiltration, neuronal degeneration, and neurite outgrowth after ablation of scar-forming, reactive astrocytes in adult transgenic mice. Neuron, 1999. 23(2): p. 297–308. [DOI] [PubMed] [Google Scholar]
  • 67.Barnabe-Heider F, Goritz C, Sabelstrom H, et al. , Origin of new glial cells in intact and injured adult spinal cord. Cell Stem Cell, 2010. 7(4): p. 470–82. [DOI] [PubMed] [Google Scholar]
  • 68.Meletis K, Barnabe-Heider F, Carlen M, et al. , Spinal cord injury reveals multilineage differentiation of ependymal cells. PLoS Biol, 2008. 6(7): p. e182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Herrmann JE, Imura T, Song B, et al. , STAT3 is a critical regulator of astrogliosis and scar formation after spinal cord injury. J Neurosci, 2008. 28(28): p. 7231–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Gorina R, Font-Nieves M, Marquez-Kisinousky L, et al. , Astrocyte TLR4 activation induces a proinflammatory environment through the interplay between MyD88-dependent NFkappaB signaling, MAPK, and Jak1/Stat1 pathways. Glia, 2011. 59(2): p. 242–55. [DOI] [PubMed] [Google Scholar]
  • 71.Ponath G, Schettler C, Kaestner F, et al. , Autocrine S100B effects on astrocytes are mediated via RAGE. J Neuroimmunol, 2007. 184(1-2): p. 214–22. [DOI] [PubMed] [Google Scholar]
  • 72.Jayakumar AR, Tong XY, Ruiz-Cordero R, et al. , Activation of NF-kappaB mediates astrocyte swelling and brain edema in traumatic brain injury. J Neurotrauma, 2014. 31(14): p. 1249–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Zaheer A, Yorek MA, and Lim R, Effects of glia maturation factor overexpression in primary astrocytes on MAP kinase activation, transcription factor activation, and neurotrophin secretion. Neurochem Res, 2001. 26(12): p. 1293–9. [DOI] [PubMed] [Google Scholar]
  • 74.Hayakawa K, Miyamoto N, Seo JH, et al. , High-mobility group box 1 from reactive astrocytes enhances the accumulation of endothelial progenitor cells in damaged white matter. J Neurochem, 2013. 125(2): p. 273–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Hayakawa K, Pham LD, Katusic ZS, et al. , Astrocytic high-mobility group box 1 promotes endothelial progenitor cell-mediated neurovascular remodeling during stroke recovery. Proc Natl Acad Sci U S A, 2012. 109(19): p. 7505–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Laird MD, Shields JS, Sukumari-Ramesh S, et al. , High mobility group box protein-1 promotes cerebral edema after traumatic brain injury via activation of toll-like receptor 4. Glia, 2014. 62(1): p. 26–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Brambilla R, Bracchi-Ricard V, Hu WH, et al. , Inhibition of astroglial nuclear factor kappaB reduces inflammation and improves functional recovery after spinal cord injury. J Exp Med, 2005. 202(1): p. 145–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Okada S, Nakamura M, Katoh H, et al. , Conditional ablation of Stat3 or Socs3 discloses a dual role for reactive astrocytes after spinal cord injury. Nat Med, 2006. 12(7): p. 829–34. [DOI] [PubMed] [Google Scholar]
  • 79.Zamanian JL, Xu L, Foo LC, et al. , Genomic analysis of reactive astrogliosis. J Neurosci, 2012. 32(18): p. 6391–410. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Liddelow SA, Guttenplan KA, Clarke LE, et al. , Neurotoxic reactive astrocytes are induced by activated microglia. Nature, 2017. 541(7638): p. 481–487. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Bastien D and Lacroix S, Cytokine pathways regulating glial and leukocyte function after spinal cord and peripheral nerve injury. Exp Neurol, 2014. 258: p. 62–77. [DOI] [PubMed] [Google Scholar]
  • 82.Jassam YN, Izzy S, Whalen M, et al. , Neuroimmunology of Traumatic Brain Injury: Time for a Paradigm Shift. Neuron, 2017. 95(6): p. 1246–1265. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Karve IP, Taylor JM, and Crack PJ, The contribution of astrocytes and microglia to traumatic brain injury. Br J Pharmacol, 2016. 173(4): p. 692–702. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.McKee CA and Lukens JR, Emerging Roles for the Immune System in Traumatic Brain Injury. Front Immunol, 2016. 7: p. 556. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Beck KD, Nguyen HX, Galvan MD, et al. , Quantitative analysis of cellular inflammation after traumatic spinal cord injury: evidence for a multiphasic inflammatory response in the acute to chronic environment. Brain, 2010. 133(Pt 2): p. 433–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Clark RS, Schiding JK, Kaczorowski SL, et al. , Neutrophil accumulation after traumatic brain injury in rats: comparison of weight drop and controlled cortical impact models. J Neurotrauma, 1994. 11(5): p. 499–506. [DOI] [PubMed] [Google Scholar]
  • 87.Fleming JC, Norenberg MD, Ramsay DA, et al. , The cellular inflammatory response in human spinal cords after injury. Brain, 2006. 129(Pt 12): p. 3249–69. [DOI] [PubMed] [Google Scholar]
  • 88.Soares HD, Hicks RR, Smith D, et al. , Inflammatory leukocytic recruitment and diffuse neuronal degeneration are separate pathological processes resulting from traumatic brain injury. J Neurosci, 1995. 15(12): p. 8223–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Stirling DP and Yong VW, Dynamics of the inflammatory response after murine spinal cord injury revealed by flow cytometry. J Neurosci Res, 2008. 86(9): p. 1944–58. [DOI] [PubMed] [Google Scholar]
  • 90.Whalen MJ, Carlos TM, Kochanek PM, et al. , Neutrophils do not mediate blood-brain barrier permeability early after controlled cortical impact in rats. J Neurotrauma, 1999. 16(7): p. 583–94. [DOI] [PubMed] [Google Scholar]
  • 91.Neirinckx V, Coste C, Franzen R, et al. , Neutrophil contribution to spinal cord injury and repair. J Neuroinflammation, 2014. 11: p. 150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Bao F, Fleming JC, Golshani R, et al. , A selective phosphodiesterase-4 inhibitor reduces leukocyte infiltration, oxidative processes, and tissue damage after spinal cord injury. J Neurotrauma, 2011. 28(6): p. 1035–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Kubota K, Saiwai H, Kumamaru H, et al. , Myeloperoxidase exacerbates secondary injury by generating highly reactive oxygen species and mediating neutrophil recruitment in experimental spinal cord injury. Spine (Phila Pa 1976), 2012. 37(16): p. 1363–9. [DOI] [PubMed] [Google Scholar]
  • 94.Saiwai H, Ohkawa Y, Yamada H, et al. , The LTB4-BLT1 axis mediates neutrophil infiltration and secondary injury in experimental spinal cord injury. Am J Pathol, 2010. 176(5): p. 2352–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Semple BD, Bye N, Ziebell JM, et al. , Deficiency of the chemokine receptor CXCR2 attenuates neutrophil infiltration and cortical damage following closed head injury. Neurobiol Dis, 2010. 40(2): p. 394–403. [DOI] [PubMed] [Google Scholar]
  • 96.Dinkel K, Dhabhar FS, and Sapolsky RM, Neurotoxic effects of polymorphonuclear granulocytes on hippocampal primary cultures. Proc Natl Acad Sci U S A, 2004. 101(1): p. 331–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Nguyen HX, O'Barr TJ, and Anderson AJ, Polymorphonuclear leukocytes promote neurotoxicity through release of matrix metalloproteinases, reactive oxygen species, and TNF-alpha. J Neurochem, 2007. 102(3): p. 900–12. [DOI] [PubMed] [Google Scholar]
  • 98.Semple BD, Trivedi A, Gimlin K, et al. , Neutrophil elastase mediates acute pathogenesis and is a determinant of long-term behavioral recovery after traumatic injury to the immature brain. Neurobiol Dis, 2015. 74: p. 263–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Conlan JW and North RJ, Neutrophils are essential for early anti-Listeria defense in the liver, but not in the spleen or peritoneal cavity, as revealed by a granulocyte-depleting monoclonal antibody. J Exp Med, 1994. 179(1): p. 259–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Daley JM, Thomay AA, Connolly MD, et al. , Use of Ly6G-specific monoclonal antibody to deplete neutrophils in mice. J Leukoc Biol, 2008. 83(1): p. 64–70. [DOI] [PubMed] [Google Scholar]
  • 101.de Castro R Jr., Hughes MG, Xu GY, et al. , Evidence that infiltrating neutrophils do not release reactive oxygen species in the site of spinal cord injury. Exp Neurol, 2004. 190(2): p. 414–24. [DOI] [PubMed] [Google Scholar]
  • 102.Kenne E, Erlandsson A, Lindbom L, et al. , Neutrophil depletion reduces edema formation and tissue loss following traumatic brain injury in mice. J Neuroinflammation, 2012. 9: p. 17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Milich LM, Ryan CB, and Lee JK, The origin, fate, and contribution of macrophages to spinal cord injury pathology. Acta Neuropathol, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Loane DJ and Kumar A, Microglia in the TBI brain: The good, the bad, and the dysregulated. Exp Neurol, 2016. 275 Pt 3: p. 316–327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Block ML and Hong JS, Chronic microglial activation and progressive dopaminergic neurotoxicity. Biochem Soc Trans, 2007. 35(Pt 5): p. 1127–32. [DOI] [PubMed] [Google Scholar]
  • 106.Gao HM and Hong JS, Why neurodegenerative diseases are progressive: uncontrolled inflammation drives disease progression. Trends Immunol, 2008. 29(8): p. 357–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.lannotti CA, Clark M, Horn KP, et al. , A combination immunomodulatory treatment promotes neuroprotection and locomotor recovery after contusion SCI. Exp Neurol, 2011. 230(1): p. 3–15. [DOI] [PubMed] [Google Scholar]
  • 108.Lee SM, Rosen S, Weinstein P, et al. , Prevention of both neutrophil and monocyte recruitment promotes recovery after spinal cord injury. J Neurotrauma, 2011. 28(9): p. 1893–907. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Popovich PG, Guan Z, Wei P, et al. , Depletion of hematogenous macrophages promotes partial hindlimb recovery and neuroanatomical repair after experimental spinal cord injury. Exp Neurol, 1999. 158(2): p. 351–65. [DOI] [PubMed] [Google Scholar]
  • 110.Bennett RE and Brody DL, Acute reduction of microglia does not alter axonal injury in a mouse model of repetitive concussive traumatic brain injury. J Neurotrauma, 2014. 31(19): p. 1647–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Frieler RA, Nadimpalli S, Boland LK, et al. , Depletion of macrophages in CD11b diphtheria toxin receptor mice induces brain inflammation and enhances inflammatory signaling during traumatic brain injury. Brain Res, 2015. 1624: p. 103–112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Gyoneva S, Kim D, Katsumoto A, et al. , Ccr2 deletion dissociates cavity size and tau pathology after mild traumatic brain injury. J Neuroinflammation, 2015. 12: p. 228. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Hsieh CL, Niemi EC, Wang SH, et al. , CCR2 deficiency impairs macrophage infiltration and improves cognitive function after traumatic brain injury. J Neurotrauma, 2014. 31(20): p. 1677–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Morganti JM, Jopson TD, Liu S, et al. , CCR2 antagonism alters brain macrophage polarization and ameliorates cognitive dysfunction induced by traumatic brain injury. J Neurosci, 2015. 35(2): p. 748–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.David S and Kroner A, Repertoire of microglial and macrophage responses after spinal cord injury. Nat Rev Neurosci, 2011. 12(7): p. 388–99. [DOI] [PubMed] [Google Scholar]
  • 116.Greenhalgh AD and David S, Differences in the phagocytic response of microglia and peripheral macrophages after spinal cord injury and its effects on cell death. J Neurosci, 2014. 34(18): p. 6316–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Shechter R, London A, Varol C, et al. , Infiltrating blood-derived macrophages are vital cells playing an anti-inflammatory role in recovery from spinal cord injury in mice. PLoS Med, 2009. 6(7): p. e1000113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Donnelly DJ, Longbrake EE, Shawler TM, et al. , Deficient CX3CR1 signaling promotes recovery after mouse spinal cord injury by limiting the recruitment and activation of Ly6Clo/iNOS+ macrophages. J Neurosci, 2011. 31(27): p. 9910–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Rolls A, Shechter R, London A, et al. , Two faces of chondroitin sulfate proteoglycan in spinal cord repair: a role in microglia/macrophage activation. PLoS Med, 2008. 5(8): p. e171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Wang IE, Lapan SW, Scimone ML, et al. , Hedgehog signaling regulates gene expression in planarian glia. Elife, 2016. 5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Zhu Y, Soderblom C, Krishnan V, et al. , Hematogenous macrophage depletion reduces the fibrotic scar and increases axonal growth after spinal cord injury. Neurobiol Dis, 2015. 74: p. 114–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Raposo C and Schwartz M, Glial scar and immune cell involvement in tissue remodeling and repair following acute CNS injuries. Glia, 2014. 62(11): p. 1895–904. [DOI] [PubMed] [Google Scholar]
  • 123.Rohl C, Lucius R, and Sievers J, The effect of activated microglia on astrogliosis parameters in astrocyte cultures. Brain Res, 2007. 1129(1): p. 43–52. [DOI] [PubMed] [Google Scholar]
  • 124.Anderson MA, Burda JE, Ren Y, et al. , Astrocyte scar formation aids central nervous system axon regeneration. Nature, 2016. 532(7598): p. 195–200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Wang X, Cao K, Sun X, et al. , Macrophages in spinal cord injury: phenotypic and functional change from exposure to myelin debris. Glia, 2015. 63(4): p. 635–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Zanier ER, Marchesi F, Ortolano F, et al. , Fractalkine Receptor Deficiency Is Associated with Early Protection but Late Worsening of Outcome following Brain Trauma in Mice. J Neurotrauma, 2016. 33(11): p. 1060–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Das A, Sinha M, Datta S, et al. , Monocyte and macrophage plasticity in tissue repair and regeneration. Am J Pathol, 2015. 185(10): p. 2596–606. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Mills CD, Kincaid K, Alt JM, et al. , M-1/M-2 macrophages and the Th1/Th2 paradigm. J Immunol, 2000. 164(12): p. 6166–73. [DOI] [PubMed] [Google Scholar]
  • 129.Mills CD, Shearer J, Evans R, et al. , Macrophage arginine metabolism and the inhibition or stimulation of cancer. J Immunol, 1992. 149(8): p. 2709–14. [PubMed] [Google Scholar]
  • 130.Miron VE, Boyd A, Zhao JW, et al. , M2 microglia and macrophages drive oligodendrocyte differentiation during CNS remyelination. Nat Neurosci, 2013. 16(9): p. 1211–1218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Martinez FO and Gordon S, The M1 and M2 paradigm of macrophage activation: time for reassessment. F1000Prime Rep, 2014. 6: p. 13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Kigerl KA, Gensel JC, Ankeny DP, et al. , Identification of two distinct macrophage subsets with divergent effects causing either neurotoxicity or regeneration in the injured mouse spinal cord. J Neurosci, 2009. 29(43): p. 13435–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Wang G, Zhang J, Hu X, et al. , Microglia/macrophage polarization dynamics in white matter after traumatic brain injury. J Cereb Blood Flow Metab, 2013. 33(12): p. 1864–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Kroehne V, Freudenreich D, Hans S, et al. , Regeneration of the adult zebrafish brain from neurogenic radial glia-type progenitors. Development, 2011. 138(22): p. 4831–41. [DOI] [PubMed] [Google Scholar]
  • 135.Baumgart EV, Barbosa JS, Bally-Cuif L, et al. , Stab wound injury of the zebrafish telencephalon: a model for comparative analysis of reactive gliosis. Glia, 2012. 60(3): p. 343–57. [DOI] [PubMed] [Google Scholar]
  • 136.Kyritsis N, Kizil C, Zocher S, et al. , Acute inflammation initiates the regenerative response in the adult zebrafish brain. Science, 2012. 338(6112): p. 1353–6. [DOI] [PubMed] [Google Scholar]
  • 137.Martin M, Leffler J, and Blom AM, Annexin A2 and A5 serve as new ligands for C1q on apoptotic cells. J Biol Chem, 2012. 287(40): p. 33733–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Oosterhof N, Holtman IR, Kuil LE, et al. , Identification of a conserved and acute neurodegeneration-specific microglial transcriptome in the zebrafish. Glia, 2017. 65(1): p. 138–149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Skaggs K, Goldman D, and Parent JM, Excitotoxic brain injury in adult zebrafish stimulates neurogenesis and long-distance neuronal integration. Glia, 2014. 62(12): p. 2061–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Liu C, Wu C, Yang Q, et al. , Macrophages Mediate the Repair of Brain Vascular Rupture through Direct Physical Adhesion and Mechanical Traction. Immunity, 2016. 44(5): p. 1162–76. [DOI] [PubMed] [Google Scholar]
  • 141.Herzog C, Pons Garcia L, Keatinge M, et al. , Rapid clearance of cellular debris by microglia limits secondary neuronal cell death after brain injury in vivo. Development, 2019. 146(9). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Kirkham M, Berg DA, and Simon A, Microglia activation during neuroregeneration in the adult vertebrate brain. Neurosci Lett, 2011. 497(1): p. 11–6. [DOI] [PubMed] [Google Scholar]
  • 143.Berg DA, Kirkham M, Beljajeva A, et al. , Efficient regeneration by activation of neurogenesis in homeostatically quiescent regions of the adult vertebrate brain. Development, 2010. 137(24): p. 4127–34. [DOI] [PubMed] [Google Scholar]
  • 144.Parish CL, Beljajeva A, Arenas E, et al. , Midbrain dopaminergic neurogenesis and behavioural recovery in a salamander lesion-induced regeneration model. Development, 2007. 134(15): p. 2881–7. [DOI] [PubMed] [Google Scholar]
  • 145.Hameed LS, Berg DA, Belnoue L, et al. , Environmental changes in oxygen tension reveal ROS-dependent neurogenesis and regeneration in the adult newt brain. Elife, 2015. 4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Cardozo MJ, Mysiak KS, Becker T, et al. , Reduce, reuse, recycle - Developmental signals in spinal cord regeneration. Dev Biol, 2017. 432(1): p. 53–62. [DOI] [PubMed] [Google Scholar]
  • 147.Curado S, Anderson RM, Jungblut B, et al. , Conditional targeted cell ablation in zebrafish: a new tool for regeneration studies. Dev Dyn, 2007. 236(4): p. 1025–35. [DOI] [PubMed] [Google Scholar]
  • 148.Ohnmacht J, Yang Y, Maurer GW, et al. , Spinal motor neurons are regenerated after mechanical lesion and genetic ablation in larval zebrafish. Development, 2016. 143(9): p. 1464–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Kizil C, Dudczig S, Kyritsis N, et al. , The chemokine receptor cxcr5 regulates the regenerative neurogenesis response in the adult zebrafish brain. Neural Dev, 2012. 7: p. 27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Tsarouchas TM, Wehner D, Cavone L, et al. , Dynamic control of proinflammatory cytokines Il-1beta and Tnf-alpha by macrophages in zebrafish spinal cord regeneration. Nat Commun, 2018. 9(1): p. 4670. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Zammit PS, Clarke JD, Golding JP, et al. , Macrophage response during axonal regeneration in the axolotl central and peripheral nervous system. Neuroscience, 1993. 54(3): p. 781–9. [DOI] [PubMed] [Google Scholar]
  • 152.Sabin K, Santos-Ferreira T, Essig J, et al. , Dynamic membrane depolarization is an early regulator of ependymoglial cell response to spinal cord injury in axolotl. Dev Biol, 2015. 408(1): p. 14–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Hasegawa T, Hall CJ, Crosier PS, et al. , Transient inflammatory response mediated by interleukin-1beta is required for proper regeneration in zebrafish fin fold. Elife, 2017. 6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Ishida T, Nakajima T, Kudo A, et al. , Phosphorylation of Junb family proteins by the Jun N-terminal kinase supports tissue regeneration in zebrafish. Dev Biol, 2010. 340(2): p. 468–79. [DOI] [PubMed] [Google Scholar]
  • 155.Ankeny DP and Popovich PG, Mechanisms and implications of adaptive immune responses after traumatic spinal cord injury. Neuroscience, 2009. 158(3): p. 1112–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Filiano AJ, Gadani SP, and Kipnis J, How and why do T cells and their derived cytokines affect the injured and healthy brain? Nat Rev Neurosci, 2017. 18(6): p. 375–384. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Weckbach S, Neher M, Losacco JT, et al. , Challenging the role of adaptive immunity in neurotrauma: Rag1(−/−) mice lacking mature B and T cells do not show neuroprotection after closed head injury. J Neurotrauma, 2012. 29(6): p. 1233–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Mencl S, Hennig N, Hopp S, et al. , FTY720 does not protect from traumatic brain injury in mice despite reducing posttraumatic inflammation. J Neuroimmunol, 2014. 274(1-2): p. 125–31. [DOI] [PubMed] [Google Scholar]
  • 159.Hurn PD, Subramanian S, Parker SM, et al. , T- and B-cell-deficient mice with experimental stroke have reduced lesion size and inflammation. J Cereb Blood Flow Metab, 2007. 27(11): p. 1798–805. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Subramanian S, Zhang B, Kosaka Y, et al. , Recombinant T cell receptor ligand treats experimental stroke. Stroke, 2009. 40(7): p. 2539–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Yilmaz G, Arumugam TV, Stokes KY, et al. , Role of T lymphocytes and interferon-gamma in ischemic stroke. Circulation, 2006. 113(17): p. 2105–12. [DOI] [PubMed] [Google Scholar]
  • 162.Gadani SP, Cronk JC, Norris GT, et al. , IL-4 in the brain: a cytokine to remember. J Immunol, 2012. 189(9): p. 4213–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Walsh JT, Hendrix S, Boato F, et al. , MHCII-independent CD4+ T cells protect injured CNS neurons via IL-4. J Clin Invest, 2015. 125(6): p. 2547. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Barouch R and Schwartz M, Autoreactive T cells induce neurotrophin production by immune and neural cells in injured rat optic nerve: implications for protective autoimmunity. FASEB J, 2002. 16(10): p. 1304–6. [DOI] [PubMed] [Google Scholar]
  • 165.Kipnis J, Mizrahi T, Hauben E, et al. , Neuroprotective autoimmunity: naturally occurring CD4+CD25+ regulatory T cells suppress the ability to withstand injury to the central nervous system. Proc Natl Acad Sci U S A, 2002. 99(24): p. 15620–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Kipnis J, Yoles E, Schori H, et al. , Neuronal survival after CNS insult is determined by a genetically encoded autoimmune response. J Neurosci, 2001. 21(13): p. 4564–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Moalem G, Leibowitz-Amit R, Yoles E, et al. , Autoimmune T cells protect neurons from secondary degeneration after central nervous system axotomy. Nat Med, 1999. 5(1): p. 49–55. [DOI] [PubMed] [Google Scholar]
  • 168.Hauben E, Butovsky O, Nevo U, et al. , Passive or active immunization with myelin basic protein promotes recovery from spinal cord contusion. J Neurosci, 2000. 20(17): p. 6421–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Hauben E, Gothilf A, Cohen A, et al. , Vaccination with dendritic cells pulsed with peptides of myelin basic protein promotes functional recovery from spinal cord injury. J Neurosci, 2003. 23(25): p. 8808–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Jones TB, Lymphocytes and autoimmunity after spinal cord injury. Exp Neurol, 2014. 258: p. 78–90. [DOI] [PubMed] [Google Scholar]
  • 171.Cruz MS, Diamond A, Russell A, et al. , Human alphabeta and gammadelta T Cells in Skin Immunity and Disease. Front Immunol, 2018. 9: p. 1304. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Toulon A, Breton L, Taylor KR, et al. , A role for human skin-resident T cells in wound healing. J Exp Med, 2009. 206(4): p. 743–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Jameson J, Ugarte K, Chen N, et al. , A role for skin gammadelta T cells in wound repair. Science, 2002. 296(5568): p. 747–9. [DOI] [PubMed] [Google Scholar]
  • 174.Rani M, Zhang Q, and Schwacha MG, Gamma delta T cells regulate wound myeloid cell activity after burn. Shock, 2014. 42(2): p. 133–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Josefowicz SZ, Lu LF, and Rudensky AY, Regulatory T cells: mechanisms of differentiation and function. Annu Rev Immunol, 2012. 30: p. 531–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Ali N and Rosenblum MD, Regulatory T cells in skin. Immunology, 2017. 152(3): p. 372–381. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Li J, Tan J, Martino MM, et al. , Regulatory T-Cells: Potential Regulator of Tissue Repair and Regeneration. Front Immunol, 2018. 9: p. 585. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Panduro M, Benoist C, and Mathis D, Tissue Tregs. Annu Rev Immunol, 2016. 34: p. 609–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Zaiss DM, Minutti CM, and Knipper JA, Immune- and non-immune-mediated roles of regulatory T-cells during wound healing. Immunology, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Rubtsov YP, Rasmussen JP, Chi EY, et al. , Regulatory T cell-derived interleukin-10 limits inflammation at environmental interfaces. Immunity, 2008. 28(4): p. 546–58. [DOI] [PubMed] [Google Scholar]
  • 181.Worthington JJ, Kelly A, Smedley C, et al. , Integrin alphavbeta8-Mediated TGF-beta Activation by Effector Regulatory T Cells Is Essential for Suppression of T-Cell-Mediated Inflammation. Immunity, 2015. 42(5): p. 903–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Arpaia N, Green JA, Moltedo B, et al. , A Distinct Function of Regulatory T Cells in Tissue Protection. Cell, 2015. 162(5): p. 1078–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Burzyn D, Kuswanto W, Kolodin D, et al. , A special population of regulatory T cells potentiates muscle repair. Cell, 2013. 155(6): p. 1282–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Ito M, Komai K, Mise-Omata S, et al. , Brain regulatory T cells suppress astrogliosis and potentiate neurological recovery. Nature, 2019. 565(7738): p. 246–250. [DOI] [PubMed] [Google Scholar]
  • 185.Liesz A, Suri-Payer E, Veltkamp C, et al. , Regulatory T cells are key cerebroprotective immunomodulators in acute experimental stroke. Nat Med, 2009. 15(2): p. 192–9. [DOI] [PubMed] [Google Scholar]
  • 186.Sharir R, Semo J, Shimoni S, et al. , Experimental myocardial infarction induces altered regulatory T cell hemostasis, and adoptive transfer attenuates subsequent remodeling. PLoS One, 2014. 9(12): p. e113653. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Villalta SA, Rosenthal W, Martinez L, et al. , Regulatory T cells suppress muscle inflammation and injury in muscular dystrophy. Sci Transl Med, 2014. 6(258): p. 258ra142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Weirather J, Hofmann UD, Beyersdorf N, et al. , Foxp3+ CD4+ T cells improve healing after myocardial infarction by modulating monocyte/macrophage differentiation. Circ Res, 2014. 115(1): p. 55–67. [DOI] [PubMed] [Google Scholar]
  • 189.Dombrowski Y, O'Hagan T, Dittmer M, et al. , Regulatory T cells promote myelin regeneration in the central nervous system. Nat Neurosci, 2017. 20(5): p. 674–680. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Ali N, Zirak B, Rodriguez RS, et al. , Regulatory T Cells in Skin Facilitate Epithelial Stem Cell Differentiation. Cell, 2017. 169(6): p. 1119–1129 e11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Nosbaum A, Prevel N, Truong HA, et al. , Cutting Edge: Regulatory T Cells Facilitate Cutaneous Wound Healing. J Immunol, 2016. 196(5): p. 2010–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Hui SP, Sheng DZ, Sugimoto K, et al. , Zebrafish Regulatory T Cells Mediate Organ-Specific Regenerative Programs. Dev Cell, 2017. 43(6): p. 659–672 e5. [DOI] [PubMed] [Google Scholar]
  • 193.Beck CW, Christen B, and Slack JM, Molecular pathways needed for regeneration of spinal cord and muscle in a vertebrate. Dev Cell, 2003. 5(3): p. 429–39. [DOI] [PubMed] [Google Scholar]
  • 194.Fukazawa T, Naora Y, Kunieda T, et al. , Suppression of the immune response potentiates tadpole tail regeneration during the refractory period. Development, 2009. 136(14): p. 2323–7. [DOI] [PubMed] [Google Scholar]
  • 195.Ito K, Morioka M, Kimura S, et al. , Differential reparative phenotypes between zebrafish and medaka after cardiac injury. Dev Dyn, 2014. 243(9): p. 1106–15. [DOI] [PubMed] [Google Scholar]
  • 196.Lai SL, Marin-Juez R, Moura PL, et al. , Reciprocal analyses in zebrafish and medaka reveal that harnessing the immune response promotes cardiac regeneration. Elife, 2017. 6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Poss KD, Wilson LG, and Keating MT, Heart regeneration in zebrafish. Science, 2002. 298(5601): p. 2188–90. [DOI] [PubMed] [Google Scholar]
  • 198.Gawriluk TR, Simkin J, Thompson KL, et al. , Comparative analysis of ear-hole closure identifies epimorphic regeneration as a discrete trait in mammals. Nat Commun, 2016. 7: p. 11164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Simkin J, Gawriluk TR, Gensel JC, et al. , Macrophages are necessary for epimorphic regeneration in African spiny mice. Elife, 2017. 6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Donnelly DJ and Popovich PG, Inflammation and its role in neuroprotection, axonal regeneration and functional recovery after spinal cord injury. Exp Neurol, 2008. 209(2): p. 378–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Marz M, Schmidt R, Rastegar S, et al. , Regenerative response following stab injury in the adult zebrafish telencephalon. Dev Dyn, 2011. 240(9): p. 2221–31. [DOI] [PubMed] [Google Scholar]

RESOURCES