Abstract
Spinal cord injury (SCI) is an incurable trauma that frequently results in partial or complete loss of motor and sensory function. Massive neurons are damaged after the initial mechanical insult. Secondary injuries, which are triggered by immunological and inflammatory responses, also result in neuronal loss and axon retraction. This results in defects in the neural circuit and a deficiency in the processing of information. Although inflammatory responses are necessary for spinal cord recovery, conflicting evidence of their contributions to specific biological processes have made it difficult to define the specific role of inflammation in SCI. This review summarizes our understanding of the complex role of inflammation in neural circuit events following SCI, such as cell death, axon regeneration and neural remodeling. We also review the drugs that regulate immune responses and inflammation in the treatment of SCI and discuss the roles of these drugs in the modulation of neural circuits. Finally, we provide evidence about the critical role of inflammation in facilitating spinal cord neural circuit regeneration in zebrafish, an animal model with robust regenerative capacity, to provide insights into the regeneration of the mammalian central nervous system.
Keywords: Inflammation, Immune response, Spinal cord injury, Axon regeneration, Cell death, Zebrafish, Inflammatory drugs, Trauma, Neurological damage
Highlights.
The complex roles of inflammation in neural circuit events following SCI are discussed.
The drugs regulating immune responses and inflammation in the treatment of SCI are reviewed, as well as their roles in the pathophysiology of SCI.
The positive roles of inflammation in facilitating spinal cord neural circuit regeneration in zebrafish are reviewed.
Background
The inflammatory responses that occur after spinal cord injury (SCI) are complicated and can have both beneficial and detrimental effects. For example, drugs that suppress immune responses or inflammation, such as methylprednisolone and minocycline, have been employed to treat SCI [1,2]. On the other hand, drugs that activate inflammatory signaling can also mitigate secondary damage [3]. The use of these medications is constrained by their poor therapeutic efficacy and the possibility of negative side effects. To date, no effective anti-inflammation drugs have been developed for SCI, primarily due to limited understanding of the SCI-associated inflammatory responses. For instance, researchers are still looking into the periods in which inflammation plays a role as well as the biological processes that inflammation affects. Poor functional recovery in SCI is a result of the degeneration of neural circuits brought on by neuronal loss, axonal retraction or synaptic damage, which limits the processing of ascending or descending signals. Therefore, it is crucial to comprehend how inflammation affects modifications to neural circuits following SCI.
While mammalian SCI is irreversible, some teleosts, including zebrafish, have a high capacity for neural circuit regeneration [4]. Because inflammation develops differently in fish and mammals, this divergence could lead to a significant advance in the research of regeneration.
Review
Immune and inflammation responses in SCI
SCI consists of primary injury and secondary injury. Primary injury is related to the initial traumatic injury and is caused by the destructive impact, resulting in immediate and irreversible mechanical injury. Primary injury triggers a pathophysiological cascade, including the secondary injury stage. Secondary injury begins as early as a few minutes after the initial trauma and is divided into three stages: acute, subacute and chronic [5–8]. Although the pathological mechanism of SCI is similar across different species, the rate of illness evolution varies, such as between mice and humans. In rodents, secondary injury can be divided into an acute stage (<24 h), a subacute stage (from 24 h to 7 days) and a chronic stage (>7 days) [8]. In humans, the transition between the acute phase and subacute phase usually occurs within a few hours to 48 h after injury, and the change from acute to chronic phase is thought to happen at 6 months [5]. The acute secondary injury stage is characterized by vascular injury, excitotoxicity, ionic imbalance, oxidative damage, the inflammatory response and immune cell activation. Acute secondary injury leads to the subacute secondary injury stage, which is characterized by neuronal apoptosis, Wallerian degeneration, reactive astrocyte activation, axonal remodeling and glial scar formation. Subacute secondary injury leads to chronic secondary injury, which is characterized by the development of cysts, axonal dieback and glial scar maturation (Figure 1) [6,7,9,10].
Figure 1.

Cellular events following SCI over time. Damage and cellular events, as well as immune cell activation, are revealed. Primary injury triggers a pathophysiological cascade, including the secondary injury stage. Secondary injury begins as early as a few minutes after the initial trauma and is divided into three stages: acute, subacute and chronic injury. The changes of various cells (neutrophils, microglia, monocytes/macrophages, lymphocytes and NK cells) in the process of SCI are marked. This Figure is modified from [11,99] with permission. SCI spinal cord injury, NK cells natural killer cells
Immediately after SCI, neutrophils and microglia become activated and secrete a number of cytokines, such as interleukin-1β (IL-1β), tumor necrosis factor alpha (TNF-α) and interleukin 6 (IL-6). This causes the subsequent infiltration of monocyte-derived macrophages, which play an important role in scavenging tissue debris at the lesion site. During the subacute phase, monocytes and macrophages infiltrate the spinal cord and produce proinflammatory cytokines, chemokines, autoantibodies, reactive oxygen and nitrogen species and other inflammatory mediators. The neuroinflammatory response is important for secondary injury, which results in cell death and tissue degeneration in the subacute and chronic stages. The inflammatory process and secreted cytokines have been extensively examined [11]. In this review, we will concentrate on the role of inflammation in different damaged neural circuits after SCI.
Cell death and inflammation
There are two main types of cell death, non-programmed cell death and programmed cell death. Non-programmed cell death, also known as necrosis, is an unregulated passive process. It is generally accepted that this type of non-programmed cell death can be triggered by proinflammatory cytokines, glutamate excitotoxicity, ionic imbalance and toxic components. Apoptosis, necroptosis, autophagy, ferroptosis and pyroptosis are examples of programmed cell death, which is a type of cell death controlled by regulatory mechanisms. Pyroptosis is a form of cell death that is triggered by proinflammatory signaling and requires the activation of caspase-1 and/or caspase11 (caspase 4/5 in humans). Pyroptosis is most commonly observed in phagocytes of the myeloid lineage, such as macrophages, dendritic cells and neutrophils [12]. The function of inflammasomes in pyroptosis has been thoroughly covered in other articles [13]. Here we will focus on apoptosis and necroptosis.
Apoptosis and inflammation
Apoptosis has been considered to be an immunologically silent form of cell death, and dying cells are cleared by phagocytes during this process. Apoptosis does not result in a proinflammatory reaction because the membrane of dying cells that are engulfed by phagocytes is still intact and the cell contents do not come into contact other surrounding cells [14]. Furthermore, apoptotic cells maintain this anti-inflammatory state even after entering secondary necrosis and leaking their cellular contents [14,15]. Apoptosis can be regulated by inflammation even when it does not trigger proinflammatory responses. TNF-α is a proinflammatory cytokine that can induce apoptosis in non-immune cells via its receptor TNF receptor 1, whose intracellular death domain activates caspase-8 and caspase-10, resulting in cell death. This process can be regulated by other inflammatory signaling pathways. For example, inducible NO synthase is a key inflammatory mediator that can activate TNF-α-mediated signaling [16,17]. Fas (also known as CD95) is another extrinsic immune-related signaling protein that is expressed by multiple cell types after SCI, including astrocytes, oligodendrocytes and microglia. In both the acute and subacute stages of SCI, activation of this cell-death receptor causes apoptosis and an inflammatory response [18]. Following SCI, Fas-deficient mice have a significant decrease in apoptosis, as well as inflammation, as shown by a decrease in cytokine expression [18]. Although cell death is unavoidable during primary SCI, studies have shown that inhibiting apoptosis can promote functional recovery. Interestingly, inhibiting Fas signaling in transgenic fas-deficient mice or administering soluble Fas receptor to block receptor activity can significantly improve functional recovery after SCI [18,19].
Other evidence suggests a correlation between inflammation and apoptosis. For example, an increase in p38 expression was associated with inflammation and apoptosis after SCI, and the p38 inhibitor SB203580 alleviated secondary SCI by suppressing inflammation and apoptosis [20]. Deficiency in progranulin, a 593 amino acid-long secreted glycoprotein, is detrimental to SCI recovery by promoting apoptosis and neuroinflammation [21]. Metformin inhibited neuronal apoptosis and the inflammatory response by increasing the expression of catenin and brain-derived neurotrophic factor and promoted motor functional recovery in rats following SCI in a recent study [22]. The above research results show that studying the relationship between apoptosis and inflammation and looking for ways to inhibit apoptosis after SCI may have important clinical significance for further treatment of SCI.
Necroptosis and inflammation
Necroptosis, which is also known as programmed necrosis, results in similar morphological changes to necrosis, such as ruptured cell membranes, enlarged cell volumes and swollen organelles. In necroptosis, activated TNF-α stimulates receptor-interacting protein kinases 1/3 (RIPK1/3) and mixed lineage kinase domain-like protein (MLKL) signaling. In contrast to apoptosis, necroptosis requires RIPK activity, which is regulated by a caspase-independent pathway [23]. After SCI, the expression levels of RIPK1, RIPK3 and MLKL are upregulated, although their peak expression varies temporally; RIPK3 and MLKL peak 1 day post-injury (dpi) [24] and RIPK1 peaks 3 dpi [25]. Inhibiting MLKL activity in SCI can facilitate neurological function recovery [26].
It has been discovered that multiple inflammatory signaling pathways are actively involved in the process of necroptosis. Reactive astrocytes undergo necroptosis in response to the inflammatory response-related gene toll-like receptor 4 (TLR4) and myeloid differentiation primary response gene 88 [27,28]. Lipopolysaccharide-induced neuroinflammation promotes necroptosis in neurons, which is facilitated by smad ubiquitination regulatory factor 1 [29]. According to a study showing that the necroptosis inhibitor necrostatin-1 can lessen tissue loss and speed up functional recovery following SCI in mice, suppressing necroptosis appears to be advantageous [30]. Notably, in contrast to apoptosis, necroptosis results in the release of cytokines or other inflammatory mediators into the extracellular environment. Therefore, there might be complex crosstalk between inflammation and necroptosis.
Future investigations should look into the relationship between inflammation and cell death, specifically programmed cell death, in secondary injury in SCI. It would also be intriguing to investigate whether cell death is a potential target for inflammation-modulated SCI therapy.
Role of inflammation in neural remodeling and axon regeneration
Both neural remodeling and axon regeneration are essential biological processes for the reorganization of neural circuitry, and inflammation signaling appears to play multiple roles in both of these processes. Microglia can interact with damaged axons and phagocytose dendrites [31], potentially exacerbating synaptic damage. This process may involve matrix metallopeptidase 9 and chemokine [32–34]. Axon sprouting, synaptic remodeling and functional recovery are all enhanced by deletion of CX3CR1, a chemokine receptor that binds to CX3CL1 in microglia/macrophages [35]. On the other hand, inflammatory stimulation can also promote axon regeneration after SCI. Intraspinal injection of the TLR2 agonist Pam2CSK4 prevents axonal dieback [3]. This outcome is consistent with research that indicates that the potent inflammatory stimulus zymosan can encourage axon regeneration by activating macrophages [36].
It is worth noting that macrophages can either promote axon regrowth or exacerbate tissue remodeling. The different roles of macrophages may come from their heterogeneity. Many of the negative effects of macrophage interactions have been attributed to the M1-like phenotype, which orchestrates glutamate- and nitric oxide-induced neuronal death [37]. M2-like macrophages, which are normally repressed, are beneficial, and their activation may reduce axonal dieback by reducing gliosis and the expression of proinflammatory cytokines such as IL-1 [38,39]. Therefore, when targeting macrophages, the diverse roles of macrophages must be considered [36].
Other immune-related cells associated with axonal recovery have been identified. Neutrophils are a diverse immune cell population that is essential for immune defense. A subset of neutrophils with axon regenerative properties has been discovered [40], which is defined as CD14 + Ly6Glo granulocytes, and these cells resemble immature neutrophils. This subpopulation promotes axonal regrowth in SCI [40]. In SCI, activated B cells secrete the antibody complement component 1q, which exacerbates axon pathology and demyelination, and this effect is ameliorated in mice with B-cell deficiency [41]. Leukocytes can be induced by IL-6 or leukemia inhibitory factor and inhibit axonal growth and impair locomotor recovery [42]. T cells can be activated by vaccination with myelin basic protein and exacerbate demyelination and axonal pathology in both mice and rats, leading to increased tissue destruction [43,44]. Inflammatory signals generated or stimulated by these cells may be critical for functional recovery after SCI. Therefore, it is necessary to investigate how various cell types orchestrate inflammatory signaling and contribute to neural remodeling.
Zebrafish inflammation in SCI
Both larval and adult zebrafish can regenerate axons and functionally recover from SCI [45]. Because of the high molecular and genetic conservation between teleosts and mammals, zebrafish research may be able to overcome the limitations of non-regenerative mammalian models.
Zebrafish with central nervous system injury may benefit from acute inflammation, which promotes healing [46]. For instance, injection of the inflammatory mediator IL-6 into the ventricles of embryonic zebrafish promotes spinal cord axon regrowth [47]. Microglia/macrophages in zebrafish express typical vertebrate macrophage genes, including a variety of transcriptional regulators, immune pathogen receptors and pruning-associated genes, suggesting functional conservation between mammals and fish [48]. It is interesting to note that in early SCI, many M2-type macrophage genes are upregulated, whereas changes in the expression of M1-type macrophage genes are very limited [49]. This rapid expansion of M2 anti-inflammatory macrophages may contribute to the advantageous benefits of acute inflammation in zebrafish.
Rapid remission of inflammation is observed in zebrafish following SCI. Temporal activation of microglia and macrophages differs between zebrafish and mammals, although early activation of microglia and the infiltration of blood-borne macrophages have been observed at the wound site in the two species [50]. In mammals, macrophages and microglia persist at the injury site (for at least 42 days in rodents and 12 months in humans) [51–53]. After damage, M1-polarized macrophages persist for an extended period of time and exert neurotoxic effects that result in chronic inflammation and poor functional recovery [54–56]. However, macrophage depletion was observed 10 dpi in adult zebrafish spinal cords [50], while in larval zebrafish, microglia and macrophages peaked at 2 dpi before significantly declining by 5 dpi [47]. Additionally, neutrophil counts peaked at 2 hour post injury in larval zebrafish and then rapidly declined thereafter [47]. The rapid resolution of inflammation in zebrafish SCI may provide a favorable extracellular environment for axon growth.
Regulatory T cells, a distinct subtype of T cells that plays a central role in preserving self-antigen tolerance and reducing inflammatory tissue injury, are another element in the treatment of SCI. Zebrafish regulatory T cells quickly migrate to the injured region in damaged organs and produce tissue-specific regenerative factors through a procedure distinct from the classic anti-inflammatory pathway to promote the proliferation of regenerative precursor cells [57].
These findings provide compelling evidence that inflammation plays a role in the pathogenesis of SCI. The timing and level of immune cell activation may determine whether inflammation is beneficial or detrimental. The results in zebrafish show that acute inflammation is mostly advantageous. However, prolonged inflammation may deteriorate the reorganization of neural circuits. Understanding how to maximize the role of acute inflammation in neural regeneration and efficiently resolve inflammation is important for the development of regenerative medicines.
Immune-targeting strategies for spinal cord regeneration
To date, methylprednisolone is the only pharmacological agent that has received clinical approval for the treatment of SCI, but the risks associated with corticosteroid treatment (e.g. gastrointestinal bleeding and wound infection) and limitations in functional recovery restrict its use [1]. Despite these drawbacks, it has been demonstrated that methylprednisolone is beneficial in suppressing inflammation, particularly microglial and macrophage activation, which aids in preventing tissue loss. Minocycline is a clinically available antibiotic with anti-inflammatory properties. Numerous animal models have revealed promising neuroprotective effects of minocycline. Minocycline pretreatment and treatment can significantly prevent production of the proinflammatory and neurotoxic cytokines TNF-α and IL-1β, thereby significantly reducing spinal cord tissue damage and functional and sensory complications after injury [2]. In clinical trials, minocycline can effectively suppress inflammation [58], and its administration tends to promote sensory and function outcomes, though no statistical significance has been observed [59]. The combination of minocycline and methylprednisolone is more effective than either drug alone. After SCI, combination therapy can effectively lower levels of lipid peroxidation as well as TNF-α and IL-6 levels, preventing the death of neuronal and glial cells [60].
A variety of targeting strategies for microglia and macrophages have been suggested to be effective. Pro-inflammatory M1 macrophages and anti-inflammatory M2 macrophages have been identified as a result of research into macrophage heterogeneity [54]. Overall, a strategy that boosts M2 activation or prevents the activation of M1 cells is advantageous for SCI recovery. For example, the administration of cytokines associated with M2 activation, including IL-4, IL-10 and IL-13, improves functional outcomes in SCI. Blocking M1 activation by blocking IL-6 or TNF-α facilitates tissue repair and functional recovery [61]. Minocycline can reduce macrophage activation (Table 1). Granulocyte-colony stimulating factor (G-CSF) and substance P can induce M2 macrophages (Table 1). These strategies have significantly enhanced neural circuit reorganization in SCI. Clinical trials revealed that G-CSF can slightly improve sensory and motor function in SCI patients, as shown by anti-inflammatory, myelin-protective and axon-regenerative effects [62], though its long-term effects require further investigation [63]. Table 1 also summarizes additional anti-inflammatory drugs used in the treatment of SCI and their potential roles in neural circuits.
Table 1.
Popular drugs targeting inflammation in the treatment of SCI
| Drugs | Inflammation processes regulated | Biological processes affected | Beneficial or detrimental for function recovery | Reference | |
|---|---|---|---|---|---|
| PPAR-γ agonists | 15d-PGJ2 | 15d-PGJ2 prevents neutrophil infiltration | Reduces cell death in neurons and oligodendrocytes | Improves motor, sensory function and neuroprotection | [64–67] |
| Rosiglitazone | Rosiglitazone exerts anti-inflammatory effects by inhibiting NLRP3 inflammasome in neurons | ||||
| Pioglitazone | Pioglitazone reduces inflammatory genes | ||||
| Methylprednisolone | Binds with glucocorticosteroid receptors to prevent nuclear translocation of proinflammatory transcription factors | Reduces astrocyte cell death and microglial activation, inhibits A1 astrocytes activation and free oxygen radical induced lipid peroxidation | Improves motor and sensory function but is still controversial in clinical practice Increases the extravasation of plasma components and enhances tissue swelling and edema High dose administration can lead to a variety of physical complications, including wound infection, sepsis, gastrointestinal bleeding, pulmonary embolism and even mortality |
[1,68–71] | |
| Minocycline | Inhibits the proinflammatory and neurotoxic cytokines TNF-α and IL-1β and reduces microglial/macrophage activation | Reduces the lesion area, increases the number of descending sympathetic axons passing through the injury site, inhibits inflammation, axonal dieback and microglial/macrophage activation and improves axonal regeneration | Improves motor function and has neuroprotective effect | [2,71–75] | |
| Erythropoietin | Inhibits leukocytes infiltration and reduces the level of pro-inflammatory cytokines such as TNF-α, IL-6 and MCP-1 | Prevents neuronal apoptosis through crosstalk between JAK-2 and NF-κB signal cascade Promotes neurogenesis and oligodendrocyte survival, enhances axon regeneration and reduces myelin loss through activating AMPK and inactivating mTOR signals |
Improves motor function and has neuroprotective effect Long-term administration can lead to excessive erythropoiesis and increased blood viscosity. |
[76–79] | |
| Estrogen | Inhibits the activation of microglia and astrocytes | Reduces edema and the inflammatory response, improves axonal and myelin loss, and decreases cell death | Improves motor and sensory function and relieves neuropathic pain, but is still controversial in clinical practice High-dose administration has serious safety problems, such as the risk of deep venous thrombosis and coronary heart disease |
[70,77,80,81] | |
| Etanercept (anti-TNF-α agents) | Inhibites TNF-α and IL-1β expression | Modulates the post-traumatic inflammatory response, attenuates neuronal injury, reduces tissue damage and cell apoptosis | Improves motor function and reduces mechanical allodynia | [70,82,83] | |
| Rolipram (PDE4 inhibitor) | Inhibits TNF-α and IL-1β production, prevents IL-10 reduction and increases white matter sparing | Protects neurons and oligodendrocytes, promotes axon regeneration and attenuates the formation of glial scars | Improves motor function, promotes neuroprotection and enhances myelinated tissue sparing Side effects such as nausea, vomiting and sedation may occur during use |
[70,84–89] | |
| Anti-α4β1 integrin | Decreases the intraspinal influx of neutrophils and monocyte/macrophages and reduces leukocyte activation and migration | Inhibits inflammation and secondary injury | Improves motor function, decreases neuropathic pain and has neuroprotective effect | [70,90,91] | |
| Substance P | Stimulates IL-10 expression and induces M2 macrophages | Activates neural stem cells, increases neuronal cells and reduces apoptotic cells | Improves motor function and repairs damaged tissue | [92–95] | |
| G-CSF | Alleviates inflammation signaling and promotes M2 macrophage activation | Inhibits the apoptosis of nerve cells and oligodendrocytes, reduces glial scars, enhances axonal myelination and regeneration | Improves motor and sensory function Side effects such as urinary tract infection, mild hepatopathy and gastric ulcer in clinical trials |
[62,69,71,96–98] | |
SCI spinal cord injury, PPAR peroxisome proliferator-activated receptor, 15d-PGJ2 15-deoxy-Δ12,14-PGJ2, NLRP3 NLR family pyrin domain containing 3, TNF-α tumor necrosis factor α, IL-1β interleukin-1β, IL-6 interleukin-6, MCP-1 macrophage chemotactic protein-1, JAK janus kinase, NF-κB nuclear factor kappa-light-chain-enhancer of activated B cells, AMPK 5′adenosine monophosphate-activated protein kinase, mTOR mammalian target of rapamycin, PDE4 phosphodiesterase 4, IL-10 interleukin-10, G-CSF granulocyte-colony stimulating factor
Conclusions
Inflammation plays a complex role in SCI injury and regeneration. Generally, early inflammatory events are critical for removing pathogens and cellular debris, as well as limiting the severity of acute injury. Excessive inflammatory responses after acute stages may impair axonal regeneration, neuronal regrowth and remyelination, resulting in severe neurological dysfunction. Notably, some inflammatory events, such as the activation of M2 macrophages, may be beneficial for regeneration. Inflammation management has emerged as one of the most important therapeutic strategies preventing apoptosis and oxidative damage, as well as promoting angiogenesis and neuronal regeneration. Future studies may focus on the precise function of distinct immune cells and the downstream signaling networks triggered by inflammation, as well as the ideal way to develop regenerative medicine strategies by effectively targeting inflammation. Due to the complexity of inflammation, it is feasible that concurrently targeting multiple immune cells or signaling may have synergistic effects that separate signaling cannot foresee.
Abbreviations
- dpi:
Days post-injury; G-CSF: Granulocyte-colony stimulating factor; IL-1β: Interleukin-1β; IL-4: Interleukin-4; IL-6: interleukin-6; IL-10: interleukin-10; IL-13: interleukin-13; MLKL: Mixed lineage kinase domain-like protein; RIPK1/3: Receptor-interacting protein kinases 1/3; SCI: Spinal cord injury; TLR2: Toll-like receptor 2; TLR4: Toll-like receptor 4; TNF-α: Tumor necrosis factor α.
Funding
This work was supported by the National Key R&D Program of China (2022YFA1105900), the National Natural Science Foundation of China (81701127), the Nantong Science and Technology Foundation of China (JC2021058) and the Large Instruments Open Foundation of Nantong University (KFJN2231, KFJN2275). TL is supported by the National Health and Medical Research Council (NHMRC) Early Career Fellowship (Grant No. 1112258) and WSU Vice-Chancellor’s Senior Research Fellowship.
Authors’ contributions
Conceptualization: LX and GC. Writing—original draft: YS, YJ, JL and LX. Writing—review and editing: YJ, TL, LX and GC. Analysis and collation of literature: GL, BL and YG. Supervision: LX and GC. Project administration: LX. All authors have read and approved the final manuscript.
Conflicts of interest
The authors declare that they have no conflicts of interest.
Contributor Information
Yan Jin, Key Laboratory of Neuroregeneration of Jiangsu and the Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products,Nantong University, Nantong 226006, China; School of Life Sciences, Nantong University, Nantong 226019, China.
Yixing Song, Key Laboratory of Neuroregeneration of Jiangsu and the Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products,Nantong University, Nantong 226006, China.
Jiaqi Lin, School of Medicine, Nantong University, Nantong 226006, China.
Tianqing Liu, NICM Health Research Institute, Western Sydney University, Westmead, NSW 2145, Australia.
Guicai Li, Key Laboratory of Neuroregeneration of Jiangsu and the Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products,Nantong University, Nantong 226006, China.
Biqin Lai, Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat-sen University), Ministry of Education, Guangzhou 510275, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226006, China.
Yun Gu, Key Laboratory of Neuroregeneration of Jiangsu and the Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products,Nantong University, Nantong 226006, China.
Gang Chen, School of Medicine, Nantong University, Nantong 226006, China.
Lingyan Xing, Key Laboratory of Neuroregeneration of Jiangsu and the Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products,Nantong University, Nantong 226006, China.
References
- 1. Bracken MB, Shepard MJ, Collins WF, Holford TR, Young W, Baskin DS, et al. A randomized, controlled trial of methylprednisolone or naloxone in the treatment of acute spinal-cord injury. Results of the second national acute spinal cord injury study. N Engl J Med. 1990;322:1405–11. [DOI] [PubMed] [Google Scholar]
- 2. Afshary K, Chamanara M, Talari B, Rezaei P, Nassireslami E. Therapeutic effects of minocycline pretreatment in the locomotor and sensory complications of spinal cord injury in an animal model. J Mol Neurosci. 2020;70:1064–72. [DOI] [PubMed] [Google Scholar]
- 3. Stirling DP, Cummins K, Mishra M, Teo W, Yong VW, Stys P. Toll-like receptor 2-mediated alternative activation of microglia is protective after spinal cord injury. Brain. 2014;137:707–23. [DOI] [PubMed] [Google Scholar]
- 4. El-Daher F, Becker CG. Neural circuit reorganisation after spinal cord injury in zebrafish. Curr Opin Genet Dev. 2020;64:44–51. [DOI] [PubMed] [Google Scholar]
- 5. Ahuja CS, Wilson JR, Nori S, Kotter MRN, Druschel C, Curt A, et al. Traumatic spinal cord injury. Nat Rev Dis Primers. 2017;3:17018. [DOI] [PubMed] [Google Scholar]
- 6. Alizadeh A, Dyck SM, Karimi-Abdolrezaee S. Traumatic spinal cord injury: an overview of pathophysiology, models and acute injury mechanisms. Front Neurol. 2019;10:282. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Anjum A, Yazid MD, Fauzi Daud M, Idris J, Ng AMH, Selvi Naicker A, et al. Spinal cord injury: pathophysiology, multimolecular interactions, and underlying recovery mechanisms. Int J Mol Sci. 2020;21:7533. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Li JJ, Liu H, Zhu Y, Yan L, Liu R, Wang G, et al. Animal models for treating spinal cord injury using biomaterials-based tissue engineering strategies. Tissue Eng Part B Rev. 2022;28:79–100. [DOI] [PubMed] [Google Scholar]
- 9. Pinchi E, Frati A, Cantatore S, D'errico S, Russa R, Maiese A, et al. Acute spinal cord injury: a systematic review investigating mirna families involved. Int J Mol Sci. 2019;20:1841. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Zhang Y, Yang S, Liu C, Han X, Gu X, Zhou S. Deciphering glial scar after spinal cord injury. Burns. Trauma. 2021;9:tkab035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Hellenbrand DJ, Quinn CM, Piper ZJ, Morehouse CN, Fixel JA, Hanna AS. Inflammation after spinal cord injury: a review of the critical timeline of signaling cues and cellular infiltration. J Neuroinflammation. 2021;18:284. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Vande Walle L, Lamkanfi M. Pyroptosis. Curr Biol. 2016;26:R568–r72. [DOI] [PubMed] [Google Scholar]
- 13. Al Mamun A, Wu Y, Monalisa I, Jia C, Zhou K, Munir F, et al. Role of pyroptosis in spinal cord injury and its therapeutic implications. J Adv Res. 2021;28:97–109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Wallach D, Kovalenko A. Keeping inflammation at bay. elife. 2014;3:e02583. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Szondy Z, Sarang Z, Kiss B, Garabuczi É, Köröskényi K. Anti-inflammatory mechanisms triggered by apoptotic cells during their clearance. Front Immunol. 2017;8:909. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Xu J, Kim GM, Chen S, Yan P, Ahmed SH, Ku G, et al. Inos and nitrotyrosine expression after spinal cord injury. J Neurotrauma. 2001;18:523–32. [DOI] [PubMed] [Google Scholar]
- 17. Hausmann ON. Post-traumatic inflammation following spinal cord injury. Spinal Cord. 2003;41:369–78. [DOI] [PubMed] [Google Scholar]
- 18. Yu WR, Fehlings MG. Fas/FasL-mediated apoptosis and inflammation are key features of acute human spinal cord injury: implications for translational, clinical application. Acta Neuropathol. 2011;122:747–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Ackery A, Robins S, Fehlings MG. Inhibition of Fas-mediated apoptosis through administration of soluble Fas receptor improves functional outcome and reduces posttraumatic axonal degeneration after acute spinal cord injury. J Neurotrauma. 2006;23:604–16. [DOI] [PubMed] [Google Scholar]
- 20. Zhang HW, Ding JD, Zhang ZS, Zhao SS, Duan KY, Zhu BQ, et al. Critical role of p38 in spinal cord injury by regulating inflammation and apoptosis in a rat model. Spine (Phila Pa 1976. 2020;45:E355–e63. [DOI] [PubMed] [Google Scholar]
- 21. Wang C, Zhang L, Ndong JC, Hettinghouse A, Sun G, Chen C, et al. Progranulin deficiency exacerbates spinal cord injury by promoting neuroinflammation and cell apoptosis in mice. J Neuroinflammation. 2019;16:238. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Zhang T, Wang F, Li K, Lv C, Gao K, Lv C. Therapeutic effect of metformin on inflammation and apoptosis after spinal cord injury in rats through the Wnt/β-catenin signaling pathway. Neurosci Lett. 2020;739:135440. [DOI] [PubMed] [Google Scholar]
- 23. Holler N, Zaru R, Micheau O, Thome M, Attinger A, Valitutti S, et al. Fas triggers an alternative, caspase-8-independent cell death pathway using the kinase RIP as effector molecule. Nat Immunol. 2000;1:489–95. [DOI] [PubMed] [Google Scholar]
- 24. Liu S, Li Y, Choi HMC, Sarkar C, Koh EY, Wu J, et al. Lysosomal damage after spinal cord injury causes accumulation of RIPK1 and RIPK3 proteins and potentiation of necroptosis. Cell Death Dis. 2018;9:476. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Kanno H, Ozawa H, Handa K, Murakami T, Itoi E. Changes in expression of receptor-interacting protein kinase 1 in secondary neural tissue damage following spinal cord injury. Neurosci Insights. 2020;15:2633105520906402. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Jiao J, Wang Y, Ren P, Sun S, Wu M. Necrosulfonamide ameliorates neurological impairment in spinal cord injury by improving antioxidative capacity. Front Pharmacol. 2019;10:1538. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Fan H, Tang HB, Kang J, Shan L, Song H, Zhu K, et al. Involvement of endoplasmic reticulum stress in the necroptosis of microglia/macrophages after spinal cord injury. Neuroscience. 2015;311:362–73. [DOI] [PubMed] [Google Scholar]
- 28. Fan H, Zhang K, Shan L, Kuang F, Chen K, Zhu K, et al. Reactive astrocytes undergo m1 microglia/macrohpages-induced necroptosis in spinal cord injury. Mol Neurodegener. 2016;11:14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Shao L, Liu X, Zhu S, Liu C, Gao Y, Xu X. The role of Smurf1 in neuronal necroptosis after lipopolysaccharide-induced neuroinflammation. Cell Mol Neurobiol. 2018;38:809–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Liu M, Wu W, Li H, Li S, Huang LT, Yang YQ, et al. Necroptosis, a novel type of programmed cell death, contributes to early neural cells damage after spinal cord injury in adult mice. J Spinal Cord Med. 2015;38:745–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Greenhalgh AD, 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;34:6316–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Horn KP, Busch SA, Hawthorne AL, Van Rooijen N, Silver J. Another barrier to regeneration in the cns: activated macrophages induce extensive retraction of dystrophic axons through direct physical interactions. J Neurosci. 2008;28:9330–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Evans TA, Barkauskas DS, Myers JT, Hare EG, You JQ, Ransohoff RM, et al. High-resolution intravital imaging reveals that blood-derived macrophages but not resident microglia facilitate secondary axonal dieback in traumatic spinal cord injury. Exp Neurol. 2014;254:109–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Tran AP, Warren PM, Silver J. The biology of regeneration failure and success after spinal cord injury. Physiol Rev. 2018;98:881–917. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Freria CM, Hall JC, Wei P, Guan Z, Mctigue DM, Popovich PG. Deletion of the fractalkine receptor, CX3CR1, improves endogenous repair, axon sprouting, and synaptogenesis after spinal cord injury in mice. J Neurosci. 2017;37:3568–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Gensel JC, Nakamura S, Guan Z, Van Rooijen N, Ankeny DP, Popovich PG. Macrophages promote axon regeneration with concurrent neurotoxicity. J Neurosci. 2009;29:3956–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Yawata I, Takeuchi H, Doi Y, Liang J, Mizuno T, Suzumura A. Macrophage-induced neurotoxicity is mediated by glutamate and attenuated by glutaminase inhibitors and gap junction inhibitors. Life Sci. 2008;82:1111–6. [DOI] [PubMed] [Google Scholar]
- 38. Busch SA, Hamilton JA, Horn KP, Cuascut FX, Cutrone R, Lehman N, et al. Multipotent adult progenitor cells prevent macrophage-mediated axonal dieback and promote regrowth after spinal cord injury. J Neurosci. 2011;31:944–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Depaul MA, Palmer M, Lang BT, Cutrone R, Tran AP, Madalena KM, et al. Intravenous multipotent adult progenitor cell treatment decreases inflammation leading to functional recovery following spinal cord injury. Sci Rep. 2015;5:16795. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Sas AR, Carbajal KS, Jerome AD, Menon R, Yoon C, Kalinski AL, et al. A new neutrophil subset promotes cns neuron survival and axon regeneration. Nat Immunol. 2020;21:1496–505. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Ankeny DP, Guan Z, Popovich PG. B cells produce pathogenic antibodies and impair recovery after spinal cord injury in mice. J Clin Invest. 2009;119:2990–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Lacroix S, Chang L, Rose-John S, Tuszynski MH. Delivery of hyper-interleukin-6 to the injured spinal cord increases neutrophil and macrophage infiltration and inhibits axonal growth. J Comp Neurol. 2002;454:213–28. [DOI] [PubMed] [Google Scholar]
- 43. Jones TB, Basso DM, Sodhi A, Pan JZ, Hart RP, Maccallum RC, et al. Pathological CNS autoimmune disease triggered by traumatic spinal cord injury: implications for autoimmune vaccine therapy. J Neurosci. 2002;22:2690–700. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Jones TB, Ankeny DP, Guan Z, Mcgaughy V, Fisher LC, Basso DM, et al. Passive or active immunization with myelin basic protein impairs neurological function and exacerbates neuropathology after spinal cord injury in rats. J Neurosci. 2004;24:3752–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Ghosh S, Hui SP. Axonal regeneration in zebrafish spinal cord. Regeneration (Oxf). 2018;5:43–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Iribarne M. Inflammation induces zebrafish regeneration. Neural Regen Res. 2021;16:1693–701. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Tsarouchas TM, Wehner D, Cavone L, Munir T, Keatinge M, Lambertus M, et al. Dynamic control of proinflammatory cytokines IL-1β and TNF-α by macrophages in zebrafish spinal cord regeneration. Nat Commun. 2018;9:4670. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Oosterhof N, Holtman IR, Kuil LE, Van Der Linde HC, Boddeke EW, Eggen BJ, et al. Identification of a conserved and acute neurodegeneration-specific microglial transcriptome in the zebrafish. Glia. 2017;65:138–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Hui SP, Sengupta D, Lee SG, Sen T, Kundu S, Mathavan S, et al. Genome wide expression profiling during spinal cord regeneration identifies comprehensive cellular responses in zebrafish. PLoS One. 2014;9:e84212. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Hui SP, Dutta A, Ghosh S. Cellular response after crush injury in adult zebrafish spinal cord. Dev Dyn. 2010;239:2962–79. [DOI] [PubMed] [Google Scholar]
- 51. Fleming JC, Norenberg MD, Ramsay DA, Dekaban GA, Marcillo AE, Saenz AD, et al. The cellular inflammatory response in human spinal cords after injury. Brain. 2006;129:3249–69. [DOI] [PubMed] [Google Scholar]
- 52. Kigerl KA, Mcgaughy VM, Popovich PG. Comparative analysis of lesion development and intraspinal inflammation in four strains of mice following spinal contusion injury. J Comp Neurol. 2006;494:578–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Donnelly DJ, Popovich PG. Inflammation and its role in neuroprotection, axonal regeneration and functional recovery after spinal cord injury. Exp Neurol. 2008;209:378–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Kigerl KA, Gensel JC, Ankeny DP, Alexander JK, Donnelly DJ, Popovich PG. Identification of two distinct macrophage subsets with divergent effects causing either neurotoxicity or regeneration in the injured mouse spinal cord. J Neurosci. 2009;29:13435–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Beck KD, Nguyen HX, Galvan MD, Salazar DL, Woodruff TM, Anderson AJ. Quantitative analysis of cellular inflammation after traumatic spinal cord injury: evidence for a multiphasic inflammatory response in the acute to chronic environment. Brain. 2010;133:433–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Wang X, Cao K, Sun X, Chen Y, Duan Z, Sun L, et al. Macrophages in spinal cord injury: phenotypic and functional change from exposure to myelin debris. Glia. 2015;63:635–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Gupta S, Adhikary S, Hui SP. Decoding the proregenerative competence of regulatory t cells through complex tissue regeneration in zebrafish. Clin Exp Immunol. 2021;206:346–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Casha S, Rice T, Stirling DP, Silva C, Gnanapavan S, Giovannoni G, et al. Cerebrospinal fluid biomarkers in human spinal cord injury from a phase II minocycline trial. J Neurotrauma. 2018;35:1918–28. [DOI] [PubMed] [Google Scholar]
- 59. Casha S, Zygun D, Mcgowan MD, Bains I, Yong VW, Hurlbert RJ. Results of a phase II placebo-controlled randomized trial of minocycline in acute spinal cord injury. Brain. 2012;135:1224–36. [DOI] [PubMed] [Google Scholar]
- 60. Sencar L, Yilmaz DM, Tuli A, Polat S. Effects of combined treatment of minocycline and methylprednisolone on the expression of tumor necrosis factor alpha and interleukine-6 in experimental spinal cord injury: a light and electron microscopic study. Ultrastruct Pathol. 2020;44:283–99. [DOI] [PubMed] [Google Scholar]
- 61. Ren Y, Young W. Managing inflammation after spinal cord injury through manipulation of macrophage function. Neural Plast. 2013;2013:945034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Aschauer-Wallner S, Leis S, Bogdahn U, Johannesen S, Couillard-Despres S, Aigner L. Granulocyte colony-stimulating factor in traumatic spinal cord injury. Drug Discov Today. 2021;26:1642–55. [DOI] [PubMed] [Google Scholar]
- 63. Koda M, Hanaoka H, Fujii Y, Hanawa M, Kawasaki Y, Ozawa Y, et al. Randomized trial of granulocyte colony-stimulating factor for spinal cord injury. Brain. 2021;144:789–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Genovese T, Esposito E, Mazzon E, Di Paola R, Muià C, Meli R, et al. Effect of cyclopentanone prostaglandin 15-deoxy-Δ12,14-PGJ2 on early functional recovery from experimental spinal cord injury. Shock. 2008;30:142–52. [DOI] [PubMed] [Google Scholar]
- 65. Meng QQ, Feng ZC, Zhang XL, Hu LQ, Wang M, Zhang HF, et al. PPAR-γ activation exerts an anti-inflammatory effect by suppressing the NLRP3 inflammasome in spinal cord-derived neurons. Mediat Inflamm. 2019;2019:6386729. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Mctigue DM, Tripathi R, Wei P, Lash AT. The ppar gamma agonist pioglitazone improves anatomical and locomotor recovery after rodent spinal cord injury. Exp Neurol. 2007;205:396–406. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Park SW, Yi JH, Miranpuri G, Satriotomo I, Bowen K, Resnick DK, et al. Thiazolidinedione class of peroxisome proliferator-activated receptor gamma agonists prevents neuronal damage, motor dysfunction, myelin loss, neuropathic pain, and inflammation after spinal cord injury in adult rats. J Pharmacol Exp Ther. 2007;320:1002–12. [DOI] [PubMed] [Google Scholar]
- 68. Zou HJ, Guo SW, Zhu L, Xu X, Liu JB. Methylprednisolone induces neuro-protective effects via the inhibition of A1 astrocyte activation in traumatic spinal cord injury mouse models. Front Neurosci. 2021;15:628917. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Chio JCT, Xu KJ, Popovich P, David S, Fehlings MG. Neuroimmunological therapies for treating spinal cord injury: evidence and future perspectives. Exp Neurol. 2021;341:113704. [DOI] [PubMed] [Google Scholar]
- 70. Singh PL, Agarwal N, Barrese JC, Heary RF. Current therapeutic strategies for inflammation following traumatic spinal cord injury. Neural Regen Res. 2012;7:1812–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Zhang Y, Al Mamun A, Yuan Y, Lu Q, Xiong J, Yang S, et al. Acute spinal cord injury: pathophysiology and pharmacological intervention (review). Mol Med Rep. 2021;23:1–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Squair JW, Ruiz I, Phillips AA, Zheng MMZ, Sarafis ZK, Sachdeva R, et al. Minocycline reduces the severity of autonomic dysreflexia after experimental spinal cord injury. J Neurotrauma. 2018;35:2861–71. [DOI] [PubMed] [Google Scholar]
- 73. Stirling DP, Khodarahmi K, Liu J, Mcphail LT, Mcbride CB, Steeves JD, et al. Minocycline treatment reduces delayed oligodendrocyte death, attenuates axonal dieback, and improves functional outcome after spinal cord injury. J Neurosci. 2004;24:2182–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Badhiwala JH, Ahuja CS, Fehlings MG. Time is spine: a review of translational advances in spinal cord injury. J Neurosurg Spine. 2018;30:1–18. [DOI] [PubMed] [Google Scholar]
- 75. Afshari K, Momeni Roudsari N, Lashgari NA, Haddadi NS, Haj-Mirzaian A, Hassan Nejad M, et al. Antibiotics with therapeutic effects on spinal cord injury: a review. Fundam Clin Pharmacol. 2021;35:277–304. [DOI] [PubMed] [Google Scholar]
- 76. Wang P, Xie ZD, Xie CN, Lin CW, Wang JL, Xuan LN, et al. Amp-activated protein kinase-dependent induction of autophagy by erythropoietin protects against spinal cord injury in rats. CNS Neurosci Ther. 2018;24:1185–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Wang H, Zhou WX, Huang JF, Zheng XQ, Tian HJ, Wang B, et al. Endocrine therapy for the functional recovery of spinal cord injury. Front Neurosci. 2020;14:590570. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Mofidi A, Bader A, Pavlica S. The use of erythropoietin and its derivatives to treat spinal cord injury. Mini Rev Med Chem. 2011;11:763–70. [DOI] [PubMed] [Google Scholar]
- 79. Matis GK, Birbilis TA. Erythropoietin in spinal cord injury. Eur Spine J. 2009;18:314–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Lee JY, Choi HY, Ju BG, Yune TY. Estrogen alleviates neuropathic pain induced after spinal cord injury by inhibiting microglia and astrocyte activation. Biochim Biophys Acta Mol basis Dis. 2018;1864:2472–80. [DOI] [PubMed] [Google Scholar]
- 81. Sribnick EA, Wingrave JM, Matzelle DD, Wilford GG, Ray SK, Banik NL. Estrogen attenuated markers of inflammation and decreased lesion volume in acute spinal cord injury in rats. J Neurosci Res. 2005;82:283–93. [DOI] [PubMed] [Google Scholar]
- 82. Hasturk AE, Baran C, Yilmaz ER, Arikan M, Togral G, Hayirli N, et al. Etanercept prevents histopathological damage after spinal cord injury in rats. Asian J Neurosurg. 2018;13:37–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Marchand F, Tsantoulas C, Singh D, Grist J, Clark AK, Bradbury EJ, et al. Effects of etanercept and minocycline in a rat model of spinal cord injury. Eur J Pain. 2009;13:673–81. [DOI] [PubMed] [Google Scholar]
- 84. Schaal SM, Garg MS, Ghosh M, Lovera L, Lopez M, Patel M, et al. The therapeutic profile of rolipram, PDE target and mechanism of action as a neuroprotectant following spinal cord injury. PLoS One. 2012;7:e43634. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Whitaker CM, Beaumont E, Wells MJ, Magnuson DS, Hetman M, Onifer SM. Rolipram attenuates acute oligodendrocyte death in the adult rat ventrolateral funiculus following contusive cervical spinal cord injury. Neurosci Lett. 2008;438:200–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Costa LM, Pereira JE, Filipe VM, Magalhães LG, Couto PA, Gonzalo-Orden JM, et al. Rolipram promotes functional recovery after contusive thoracic spinal cord injury in rats. Behav Brain Res. 2013;243:66–73. [DOI] [PubMed] [Google Scholar]
- 87. Beaumont E, Whitaker CM, Burke DA, Hetman M, Onifer SM. Effects of rolipram on adult rat oligodendrocytes and functional recovery after contusive cervical spinal cord injury. Neuroscience. 2009;163:985–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Iannotti CA, Clark M, Horn KP, Van Rooijen N, Silver J, Steinmetz MP. A combination immunomodulatory treatment promotes neuroprotection and locomotor recovery after contusion sci. Exp Neurol. 2011;230:3–15. [DOI] [PubMed] [Google Scholar]
- 89. Nikulina E, Tidwell JL, Dai HN, Bregman BS, Filbin MT. The phosphodiesterase inhibitor rolipram delivered after a spinal cord lesion promotes axonal regeneration and functional recovery. Proc Natl Acad Sci U S A. 2004;101:8786–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Bao F, Omana V, Brown A, Weaver LC. The systemic inflammatory response after spinal cord injury in the rat is decreased by α4β1 integrin blockade. J Neurotrauma. 2012;29:1626–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Fleming JC, Bao F, Chen Y, Hamilton EF, Relton JK, Weaver LC. Alpha4beta1 integrin blockade after spinal cord injury decreases damage and improves neurological function. Exp Neurol. 2008;214:147–59. [DOI] [PubMed] [Google Scholar]
- 92. Mashaghi A, Marmalidou A, Tehrani M, Grace PM, Pothoulakis C, Dana R. Neuropeptide substance P and the immune response. Cell Mol Life Sci. 2016;73:4249–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Jiang MH, Chung E, Chi GF, Ahn W, Lim JE, Hong HS, et al. Substance P induces M2-type macrophages after spinal cord injury. Neuroreport. 2012;23:786–92. [DOI] [PubMed] [Google Scholar]
- 94. Yang L, Li G, Ye J, Lu D, Chen Z, Xiang AP, et al. Substance P enhances endogenous neurogenesis to improve functional recovery after spinal cord injury. Int J Biochem Cell Biol. 2017;89:110–9. [DOI] [PubMed] [Google Scholar]
- 95. Jiang MH, Lim JE, Chi GF, Ahn W, Zhang M, Chung E, et al. Substance P reduces apoptotic cell death possibly by modulating the immune response at the early stage after spinal cord injury. Neuroreport. 2013;24:846–51. [DOI] [PubMed] [Google Scholar]
- 96. Khorasanizadeh M, Eskian M, Vaccaro AR, Rahimi-Movaghar V. Granulocyte colony-stimulating factor (G-CSF) for the treatment of spinal cord injury. CNS Drugs. 2017;31:911–37. [DOI] [PubMed] [Google Scholar]
- 97. Torregrossa F, Sallì M, Grasso G. Emerging therapeutic strategies for traumatic spinal cord injury. World Neurosurg. 2020;140:591–601. [DOI] [PubMed] [Google Scholar]
- 98. Kawabe J, Koda M, Hashimoto M, Fujiyoshi T, Furuya T, Endo T, et al. Neuroprotective effects of granulocyte colony-stimulating factor and relationship to promotion of angiogenesis after spinal cord injury in rats: laboratory investigation. J Neurosurg Spine. 2011;15:414–21. [DOI] [PubMed] [Google Scholar]
- 99. Putatunda R, Bethea JR, Hu WH. Potential immunotherapies for traumatic brain and spinal cord injury. Chin J Traumatol. 2018;21:125–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
