Next Article in Journal
Obesity: The Fat Tissue Disease Version of Cancer
Next Article in Special Issue
Flightless I Negatively Regulates Macrophage Surface TLR4, Delays Early Inflammation, and Impedes Wound Healing
Previous Article in Journal
Prostaglandin 2α Promotes Autophagy and Mitochondrial Energy Production in Fish Hepatocytes
Previous Article in Special Issue
Post-COVID Complications after Pressure Ulcer Surgery in Patients with Spinal Cord Injury Associate with Creatine Kinase Upregulation in Adipose Tissue
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Emerging Roles of Microglia Depletion in the Treatment of Spinal Cord Injury

1
Senior Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing 100037, China
2
National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing 100853, China
3
Department of Spine Surgery, Peking University People’s Hospital, Beijing 100044, China
4
Department of Pediatric Surgery, The Chinese PLA General Hospital, Beijing 100853, China
5
Beijing Key Lab of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Institute of Orthopaedics, The Chinese PLA General Hospital, Beijing 100853, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Cells 2022, 11(12), 1871; https://doi.org/10.3390/cells11121871
Submission received: 20 April 2022 / Revised: 31 May 2022 / Accepted: 6 June 2022 / Published: 9 June 2022
(This article belongs to the Special Issue The Cell Biology and Immunology of Wound Healing)

Abstract

:
Microglia, as the resident immune cells and first responder to neurological insults, play an extremely important role in the pathophysiological process of spinal cord injury. On the one hand, microglia respond rapidly and gather around the lesion in the early stage of injury to exert a protective role, but with the continuous stimulation of the injury, the excessive activated microglia secrete a large number of harmful substances, aggravate the injury of spinal cord tissue, and affect functional recovery. The effects of microglia depletion on the repair of spinal cord injury remain unclear, and there is no uniformly accepted paradigm for the removal methods and timing of microglia depletion, but different microglia depletion strategies greatly affect the outcomes after spinal cord injury. Therefore, this review summarizes the physiological and pathological roles of microglia, especially the effects of microglia depletion on spinal cord injury—sustained microglial depletion would aggravate injury and impair functional recovery, while the short-term depletion of microglial population in diseased conditions seems to improve tissue repair and promote functional improvement after spinal cord injury. Furthermore, we discuss the advantages and disadvantages of major strategies and timing of microglia depletion to provide potential strategy for the treatment of spinal cord injury.

1. Introduction

Spinal cord injury (SCI) is a severe traumatic condition of the central nervous system (CNS) that can cause motor, sensory, and autonomic dysfunction below the level of injury [1,2]. Currently, the estimated annual global incidence of SCI is 40 to 80 cases per million population with an approximate three million SCI patients worldwide [3]. After SCI, more than 45% of patients will have permanent dysfunction, and spontaneous functional recovery is extremely limited. Moreover, due to a series of complex pathophysiological processes, the current effective treatment for SCI is still unavailable [4,5].
The pathophysiological process of SCI includes the primary SCI and secondary SCI. The primary SCI is mainly caused by the initial traumatic impact, resulting in the direct damage to the spinal cord tissue. It usually occurs within minutes to hours following the injury, characterized by the cell necrosis, hemorrhage, edema, etc. On the basis of the primary injury, SCI further develops within a few days to several weeks, causing a series of secondary injuries including inflammatory response, apoptosis, lipid peroxidation, and the production of a large number of free radicals. The secondary injury leads to massive neuronal and glial cell death and glial scarring, and eventually forms necrotic cysts that hinder nerve regeneration. Among them, the inflammation is considered to play an extremely important role in the SCI [6,7].
Inflammation after SCI is complicated and regulated by various types of cells, including microglia, astrocytes, neutrophils, monocytes, macrophages, and lymphocytes [8,9,10]. In the initial stage of SCI, various types of major inflammatory cells aggregate to the lesion, clear the damaged tissue by actively phagocytosing myelin and dead cell debris, and gather around the lesion core to limit the further expansion of the injury. However, with the progression of the SCI, the aggregation of the above-mentioned cells induces a strong immune response and secretes a large number of cytotoxic substances, such as free radicals, nitric oxide, and various inflammatory factors, including interleukin and tumor necrosis factor, etc., which further aggravates the injury of spinal cord tissue. As resident immune cells of the CNS, microglia play a major and critical role in the inflammatory response following SCI. Despite the current mainstream view that microglia are the main detrimental factor in the inflammatory response after SCI, there is still controversy as to whether microglia need to be removed after SCI for tissue repair and functional recovery. To better evaluate the role of microglia in the repair of SCI, especially after their removal, this review systematically summarizes the research progress of microglia depletion on SCI repair, with focuses on the effect of microglia depletion on SCI repair, the methods of microglia depletion, and microglia removal timing.

2. Overview of Microglia

2.1. The Origin and Physiological Role of Microglia

Microglia are resident immune cells in the CNS, accounting for approximately 5–15% of the total number of cells in the CNS [11,12]. They originate from myeloid progenitor cells in the yolk sac during the embryonic period and are a special type of mononuclear macrophage [13,14,15]. Under physiological conditions, microglia are in a “resting state” and characterized by small cell diameters and various morphologies, similar to neuronal structures with many tiny protrusions extending hierarchically from the cell body. While the cell body does not move much, the branches of microglia are highly dynamic, constantly stretching, moving, and therefore surveying the CNS every now and then [16].
As the first line of defense, microglia in the “resting state” were reported to directly or indirectly contact neurons, astrocytes, oligodendrocytes, etc. through their multi-level branches, and then play a dynamic surveillance role in the CNS [17,18]. Specifically, they are able to make a quick response to the potential pathological damage, then quickly and effectively remove invading pathogens and cell debris through phagocytosis to maintain the stability of the neural network of the CNS [19,20,21]. On the other hand, microglia function as a special kind of secretory cells, which can secrete trophic factors in the “resting state” to regulate the activity of neurons and oligodendrocytes, thereby affecting important processes such as axon growth and myelination of neurons [22]. The absence or dysregulation of such cells may directly lead to abnormal immune regulation, inflammatory storm, and neuronal death in the CNS [23,24]. Notably, the microglia in the spinal cord and thalamus exclusively express the ST2 marker, which is the specific receptor for interleukin-33 and mediates the microglial engulfment of synapses in these two regions. This biological process is responsible for the important circuit formation in the spinal cord [25,26]. Therefore, microglia play an important regulatory role in the CNS under physiological conditions, and at the same time provide an important immune surveillance system to maintain the development and homeostasis of the CNS.

2.2. The Role of Microglia in SCI

Microglia, as resident immune cells of the central nervous system, play a major regulatory role in the inflammatory response after SCI, but its regulatory mechanism on SCI remains unclear [27,28]. Following SCI, microglia are rapidly activated by various pathological factors and lead to a range of alterations in morphology and function. For cell morphology, there is an “amoeba-like” change, which is manifested by the rapid enlargement of the cell body, and the shrinkage of the elongated branch structures (they become shorter and thicker). For cell function, the proliferation and migration activities of microglia are significantly increased, and the phagocytic activity and secretion of cytokines are significantly enhanced. Although moderately activated microglia have a certain positive effect after SCI, those continuously stimulated by the injury often exhibit abnormal activation and exert toxic effects on neuronal cells, which promotes inflammatory cell aggregation and infiltration and further activates the inflammatory response in turn, thereby mediating secondary injury in the spinal cord [29,30].
Previous studies [31,32] believed that microglia released a large amount of harmful substances after SCI, resulting in a large number of neural cells death, axonal degeneration, demyelination, etc. [4], and simultaneously provoked astrocytes through paracrine effects and interacted together to form glial scars, to hinder axonal regeneration. Liddelow et al. [33] found that activated microglia could secrete cytokines such as IL-1a, TNF, and C1q in the central nervous system, which induced neurotoxic reactive astrocytes and then caused the neuronal and oligodendrocytes deaths. If any of the IL-1a, TNF, and C1q-encoding genes were conditionally removed on microglia using gene editing technology, animals would have shown much better motor functional recovery after SCI [34,35,36]. Moreover, the persistent activated microglia were reported to induce chronic neuro-inflammation, leading to the further tissue injuries and cell deaths, as well as cognitive deficits [37].
As for the subsets of microglia, activated microglia were originally considered to be homogeneous cells. But recently, a growing number of studies have revealed that after activation, microglia mainly have two cell subtypes with different cell markers and functions, namely the classical activated type (M1 type) and selective activated type (M2 type) (Figure 1). Pro-inflammatory factors, such as lipopolysaccharide and γ-interferon, can induce the transformation of “resting” microglia into M1 cells [38]. When SCI happens, microglia quickly recognize harmful stimuli via a series of immune receptors such as toll-like receptors and nucleotide-binding oligomerization domain- like receptors. The activated microglia then shift their function and polarize into being pro-inflammatory due to exposure to interferon-r and cell debris [39,40]. M1-type microglia are associated with tissue damage and pro-inflammatory responses, and secrete pro-inflammatory cytokines such as tumor necrosis factor-a (TNF-a), interleukin 1b (IL-1b), chemokines, antigen-presenting molecules, etc., which recruit inflammatory cells to the injured lesions, increase the phagocytosis of necrotic cells, and improve the defense clearance of the CNS against invasive stimuli to some extent [41]. However, M1-type microglia are usually over-activated, and then aggravate the downstream inflammatory response storm, resulting in increased neuronal cells death and tissue injury. Therefore, M1-type microglia are also called pro-inflammatory microglia.
Unlike the M1-type microglia, M2-microglia are mainly involved in tissue repair, the regulation of inflammatory response, neuronal differentiation, etc. [42,43]. They can be activated by cytokines such as IL-4 and IL-10 to become M2-type microglia, which mainly express IL-4, IL-10, and growth factors such as insulin-like growth factor 1 and transforming growth factor TGF-β, etc. They also have an enhanced ability to remove myelin debris and other necrotic substances, and are able to promote repair and functional recovery after SCI. Therefore, M2 microglia are also known as anti-inflammatory microglia. Previous studies have shown that M2-type microglia can be further classified into three subtypes: M2a, M2b, and M2c, which are stimulated by different factors and have relatively different functional phenotypes [44]. M2a cells are the main subtype of M2 microglia induced by IL-4 or IL-13, and express those neuroprotective markers, including CD206, Ym1, and ARG. -They mainly secrete anti-inflammatory factors, neurotrophic factors and etc. to promote tissue repair and reconstruction after SCI; M2b type, which usually appears under the stimulation of virus infection and immune complexes, is considered to be a transitional microglia between M1 and M2a, and shows both pro-inflammatory and anti-inflammatory effects at the same time; the M2c type, also a phenotype of neuroprotective microglia, is often stimulated and produced by IL-10 and TGF-b. This type of microglia usually shows high expression of TGF-b, SOCS3, and IL4R-a markers after SCI, which are mainly associated with synaptic remodeling, phagocytosis, and wound healing [45]. M2a and M2c are regarded as the major neuroprotective cell types of microglia, which can effectively limit secondary inflammation-mediated tissue damage and promote spinal cord repair. Moreover, the expression of the aforementioned neuroprotective markers of M2a and M2c were positively correlated with neurological outcome after SCI, especially at the early-stage post-injury [45,46].
With the deepening of research on microglia, the simple classification of microglia into M1 and M2 type were gradually considered to be incorrect. Instead, the activation into M1/M2 microglia was regarded as a dynamic process. Within the first few hours after SCI, microglia are initially activated to M2-type cells temporarily and play an anti-inflammatory role. They are manifested as enhanced phagocytic activity, accelerated clearance of irritants and cellular debris, enhanced secretion of anti-inflammatory factors, and neurotrophic factors, which could inhibit the excessive immune-inflammatory response and promote the repair of damaged tissue. In a few hours to several days after SCI, due to the continuous stimulation of the spinal cord, microglia rapidly proliferate and gradually transform from M2 cells to M1 cells, exerting neurotoxic effects on neurons via the secretion of inflammatory factors, activation of the downstream immune response, and further aggravation of the secondary injury of the spinal cord. After entering the subacute and chronic phases of SCI, M1-type cells become the major cell type of microglia, and continuously exacerbate the SCI and hinder tissue repair [47,48]. Therefore, how to minimize activation of M1-type cells and maintain the activation of M2-type cells in microglia will be one of the important directions of the treatment for SCI.

3. The Effect of Microglia Depletion on SCI Repair

As mentioned above, although activated microglia after SCI play a certain role in neuroprotection and repair at the early stage, excessively activated microglia simultaneously release a large number of harmful substances, further aggravating SCI and impeding the damage repair and functional recovery. Thus, microglia are required to be regulated to minimize their deleterious effects. What if microglia depletion is performed during SCI? Is it possible to suppress inflammatory storms and promote nerve regeneration and functional recovery?

3.1. The Adverse Effects of Microglia Depletion on SCI

At present, most studies have found that the depletion of microglia before SCI significantly inhibits the repair and functional recovery in animals with SCI [49,50,51,52]. Li et al. [49] found that microglia could promote long-distance axon growth after spinal cord crush injury and removal of microglia will directly affect the process of nerve regeneration in neonatal mice with SCI. Furthermore, single-cell sequencing revealed that microglia can temporarily secrete fibronectin and its binding proteins to provide the extracellular-matrix bridge, thereby promoting spinal cord axon regeneration. Victor Bellver-Landete et al. [50] also found that microglia were an essential component of protective scars after spinal cord contusion injury, and the depletion of microglia directly led to decreased secretion of insulin-like growth factor-1, disordered scar structure, and ultimately hindered functional recovery in animals. Fan et al. [51] found that even M1-type microglia/macrophages were still able to promote spinal cord repair by inhibiting astrocyte activation after spinal cord contusion. Therefore, if the microglia had been completely removed, reactive astrocytes would then have been activated, which would further disrupt tissue repair and nerve regeneration. Consistently, Hakim et al. [52] showed that in mice with spinal cord contusion microglial activation exerted a strong positive effect on SCI repair as activated microglia were responsible for the removal of cell debris and immune-regulatory effects on inflammation and astrocytic response. Similarly, our previous study [53] also showed that 5-week-long microglia depletion would cause glial scar disorder, delay astrocyte aggregation, further axonal degeneration, and functional impairment on mice with spinal cord crush injury.
While the methods and strategies of microglia depletion in the aforementioned studies were different, all of them found that microglia depletion would further affect the repair after SCI. Thus, microglia depletion might not improve functional recovery after SCI, but even further aggravate injury on animals in these cases.

3.2. The Beneficial Effects of Microglia Depletion on SCI

Obviously, those above findings contradicted the scientists’ original assumption that the complete elimination of hyper-activated microglia should lead to beneficial outcomes. To this end, the researchers further explored whether microglia depletion could have beneficial effects? Victor Bellver-Landete et al. [50] changed their previous microglia depletion strategy by removing microglia three weeks before the injury and stopping microglia intervention after injury. The results showed that microglia depletion did not significantly affect SCI, but again no beneficial outcomes were found. Consistent with Victor’s study, Li et al. [54] found no locomotion functional recovery after spinal cord contusion via either pre- or post-injury microglial depletion for six weeks. However, their study demonstrated that microglial depletion significantly reduced the production of reactive oxygen species and improved neuronal survival and neurological recovery, including cognition and depressive-like behavior. Igor Jakovcevski et al. [55] pointed out that if microglia were continuously removed from 2 weeks before SCI to 2 weeks after spinal cord compression injury, the removal of microglia in the early stage of the injury (1 week after injury) could promote functional improvement after SCI, while in the late stage of the injury (5 weeks after injury), there was no significant improvement in functional outcomes after SCI even though there were some improvements in gliosis and cholinergic nerve regeneration at that time. Interestingly, Gaëtan Poulen et al. [56,57] reported that, regardless of rodents or non-human primates, a transient depletion of microglia could significantly enhance functional recovery with the alleviation of tissue damage, whereas prolonged microglia depletion was not beneficial for tissue repair after spinal cord hemisection. Ma et al. [58] found that two-week-long microglia depletion significantly improved the local microenvironment after SCI in rats, and enhanced the electrophysiological activity and functional recovery of animals. And if combined with their self-designed hydrogel-based scaffold, motor function recovery could be further improved after the complete transection spinal cord injury.
Taken together, the effects of microglia depletion on SCI are still controversial. Different microglia depleting approaches, microglia depleting periods, and SCI models directly determine the positive or negative outcomes after SCI. Notably, a growing body of evidence shows that the persistent microglia removal seemed to be not conducive to tissue repair after SCI, while selective and strategic removal of microglia might improve recovery after SCI.

4. The Strategy and Timing of Microglia Depletion

As previously mentioned, different microglia depletion strategies greatly affect the outcome of tissue repair after SCI. At present, there is no unified and recognized paradigm for microglia depletion. Each research team applied respective strategies of microglia depletion for special research purposes.

4.1. The Main Method of Microglia Depletion

The methods of microglia depletion can be mainly divided into two categories: gene manipulation and small-molecule intervention. The former mainly uses microglia-specific surface markers such as Cx3Cr1, TMEM119, Sall1, etc., to construct specific microglia transgenic animals and then achieve the conditional removal of microglia; the latter involves the design and construction of small-molecule compounds against specific microglia cell markers so as to induce apoptosis and necrosis of microglia and then deplete these cells.

4.1.1. Microglia Depletion via Gene Manipulation

Microglia depletion via gene manipulation usually requires the construction of specific transgenic mice combined with specific toxic substances, such as diphtheria toxin (DT) or herpes virus, to conditionally remove microglia. Parkhurst et al. [59] constructed Cx3cr1CreER: Rosa26iDTR transgenic mice and achieved conditional knockout of microglia through the CreERT system and DTR system. Specifically, they first constructed Cx3cr1CreER mice that specifically express Cre recombinase and estrogen receptor (ER) at the site of Cx3cr1+ cells and obtained Cx3Cr1CreER: Rosa26iDTR transgenic mice by crossing Cx3Cr1CreER mice with Rosa26iDTR mice. Then, they induced Cre recombinase expression by tamoxifen, and then injected DT intraperitoneally to achieve Cx3cr1+ cells knockout. Considering that peripheral monocyte-macrophages also express Cx3cr1 protein but with more frequent cell turnover (about 7 days), the intraperitoneal injection of DT would be performed 30 days after tamoxifen induction to achieve specific microglia depletion. Similarly, CX3CR1CreER: Rosa26iDTA [60], Cx3cr1Cre:Csf1rfl mice [49], and CD11bHSVTK transgenic mice [55] have been constructed successively to conditionally remove microglia in the CNS. They either used the CreERT system, the Cre-Flox system, or expressed suicide genes to achieve the depletion of microglia.
The advantage of microglia depletion via gene manipulation includes: (1) it can manipulate the time series to achieve microglia depletion at a specific time point or a specific period, allowing the experimenter to flexibly manipulate the observation window; (2) In addition, microglia-specific knockout can be achieved using the self-renewal characteristics of microglia. The disadvantage of microglia depletion at the genetic level is that: (1) it mainly relies on transgenic mice. The construction and feeding cycle are time-consuming, and special identification is also required; (2) At present, the construction of microglia transgenic animals is mainly carried out on mice, since it is difficult to expand to rats or other mammals; (3) Microglia depletion via gene manipulation is not of very high efficiency (about 70%) and cannot be maintained for a long time, since the renewed microglia do not express Cre recombinase. The application of tamoxifen and diphtheria toxin may affect the survival of mice or exacerbate function of mice with SCI.

4.1.2. Microglia Depletion via Small-Molecule Compounds

Unlike genetic manipulation to remove microglia, microglia depletion via small-molecule compounds is more convenient and diverse. They are either designed to impress those targets important for microglial survival, such as colony-stimulating factor 1 receptor (CSF1R), or they exert direct toxic effects leading to microglial cell death. For example, CSF1 is an extremely important regulator of microglia/macrophage proliferation, differentiation, and survival, so interventions on CSF1R can achieve targeted depletion of microglia. The small-molecule compound PLX3397 [14,33,61] and its second-generation product PLX5622 [50,62,63] are currently widely used specific inhibitors of CSF1R. They are highly specific to CSF1R and able to penetrate the blood–brain barrier and competitively occupy the site of CSF1, resulting in microglial cell death with up to 99% effectiveness. At the same time, studies have shown that PLX small-molecule compounds had little effect on other types of cells and animal behavior in mice, and thus are considered very good microglia-targeted regulation drugs.
In addition to these two star products, there are other CSF1R-specific inhibitors (GW2580 [64], BLZ945 [65], etc.), chlorophosphate liposomes [66], and M1-type microglia targeted depletion drugs such as gadolinium chloride [67], which could achieve microglia depletion to a varying degree. GW2580 is an orally selective inhibitor of the tyrosine kinase activity of CSF1R, which could selectively inhibit microglia/monocyte proliferation [64]. BLZ945, another brain-penetrant CSF1R inhibitor, could decrease the number of microglia by approx. 60% after its application [65]. Chlorophosphate liposomes can inhibit ADP/ATP transport in microglia and induce cell death by generating non-hydrolyzable ATP analogs, while gadolinium chloride competitively inhibits the active calcium activity of M1-type microglia and destroys the endoplasmic reticulum membrane to induce the death of microglia.
Different small-molecule compounds have distinct characteristics and modes of action, and the efficiency of microglial depletion is also different. Therefore, they should be selected according to their specific experimental purposes. The advantages of small-molecule compounds are very obvious: (1) It is not dependent on the species of animal. Most animals depend on CSF1 for microglial survival and proliferation. Taking PLX products as an example, PLX small-molecule compounds can theoretically work on all animals; (2) Small-molecule compounds can be taken orally with high selectivity, and some of them can pass through the blood–brain barrier, which is very convenient for application; (3) They are relatively safe, as they do not affect the stability of animal genomes. However, the shortcomings of small-molecule compounds are also obvious. For example: (1) It has been reported that [68] CSF1R inhibitors are not microglia-specific. Instead, they affect both hematopoietic and macrophage functions; (2) Manipulation at specific time points is unavailable to small-molecule compounds, and the starting point of the specific action on microglia is difficult to determine.
Taken together, when it comes to microglia depletion, there are two main strategies: gene manipulation and small-molecule intervention. Both strategies have their advantages and disadvantages and could be combined to make up for their respective shortcomings and achieve specific purposes (Table 1).

4.2. The Timing of Microglia Depletion

The optimal timing for microglia depletion remains controversial at present. The different purposes or approaches to microglia depletion also influence the timing selection of microglia depletion (Table 2).
Taking microglia depletion via gene manipulation as an example, some studies performed diphtheria toxin application 7 days after tamoxifen induction [72] and found that microglia were specifically reduced and lasted for a week. However, in theory, the peripheral monocytes and macrophages would also be specifically removed at this time. On the other hand, in order to obtain selective microglia depletion, many studies chose to use diphtheria toxin 3–4 weeks after tamoxifen induction [59,63], since the peripheral macrophages would have completed the cell turnover and no longer express the diphtheria toxin receptor, whereas microglia still expressed DT receptor. However, the time frame for microglia depletion via gene manipulation is relatively short and studies have shown [29] that the number of microglia return to normal one week after the application of diphtheria toxin.
As for the timing of microglia depletion via small-molecule compounds, it is more flexible and diverse. Taking the most widely used PLX3397 and PLX5622 as examples, most studies chose to apply CSF1R inhibitor three weeks or more before the injury [50,60] and maintain the drug to the end of the experiment. Additionally, some chose two weeks or one before the injury to remove microglia [33,49,53,73]. Furthermore, some studies on microglia depletion started on the day of SCI [58], or 3 days after SCI [50]. However, regardless of the timing, the results of the study showed that the number of microglia decreased significantly after the application of PLX CSF1R inhibitors and the number of microglia returned to normal levels after drug withdrawal. Taking into consideration the practical clinical application, it seems to be more reasonable if microglia depletion is obtained after injury, but the specific application period still needs further research to determine.
In conclusion, there is no unified timing of microglia depletion during SCI, and different strategies or purposes of microglia depletion determine the actual intervention timing. In terms of the length of the intervention time compared to the depletion of microglia for the entire experimental time window, the short-term removal of microglia seems more reasonable as microglia indeed play positive roles in the CNS [56].

5. Conclusions

Microglia play a very important role in normal physiological processes and after SCI. Following SCI, on the one hand, microglia respond quickly and play a certain role in neuroprotection and repair by removing invading pathogens and cell debris through phagocytosis. On the other hand, continuous activated microglia often exert adverse effects on neuronal cells, promote inflammatory cell aggregation and infiltration, and further activate the inflammatory response in turn, thereby mediating secondary injury in the spinal cord.
The effects of microglia depletion on SCI are still controversial, and different microglia depletion strategies greatly affect the outcome of tissue repair after SCI. Current studies have found that continuous microglia depletion is not conducive to tissue repair after SCI, and selective short-term depletion of microglia could effectively improve tissue repair and promote functional recovery after injury. Regarding the removal method and removal timing of microglia, there is still no uniform paradigm. Different strategies or purposes of microglia depletion determine the actual intervention timing and ultimately affect the outcome of animal SCI. Future research should focus more on: (1) How to achieve longer lasting microglia depletion via gene manipulation; (2) Exploring new and more microglia-specific small molecule compounds; (3) Identifying the optimal timing and duration of action of microglia depletion and elucidate its underlying mechanism. It is hoped that with the development of various new technologies and in-depth research on microglia, people can finally achieve the dynamic and selective regulation of microglia and make a breakthrough in the treatment of SCI in the future.

Author Contributions

J.D. wrote this initial manuscript. J.D., F.M., K.Z. and J.G. participated in drafting the manuscript. Z.L., M.L., X.L., J.L. and Y.W. were responsible for the draft reviewing and editing. L.Z. and P.T. instructed all the process of this study. All authors have read and agreed to the published version of the manuscript.

Funding

This research is supported by the Training Program of the Foundation for the Distinguished Young Scholars of Chinese PLA General Hospital projects (No. 2020-JQPY-003 to L.Z.) and the National Natural Science Foundation of China (No. 82101435 to K.Z.).

Acknowledgments

We appreciated the instructions and help of Jiang Peng from Orthopedics Institute of Chinese PLA.

Conflicts of Interest

The authors declare no conflict of interest in this study.

References

  1. Nakamura, M.; Okano, H. Cell transplantation therapies for spinal cord injury focusing on induced pluripotent stem cells. Cell Res. 2013, 23, 70–80. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Mothe, A.J.; Tator, C.H. Advances in stem cell therapy for spinal cord injury. J. Clin. Investig. 2012, 122, 3824–3834. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Assinck, P.; Duncan, G.J.; Hilton, B.J.; Plemel, J.R.; Tetzlaff, W. Cell transplantation therapy for spinal cord injury. Nat. Neurosci. 2017, 20, 637–647. [Google Scholar] [CrossRef] [PubMed]
  4. Courtine, G.; Sofroniew, M.V. Spinal cord repair: Advances in biology and technology. Nat. Med. 2019, 25, 898–908. [Google Scholar] [CrossRef] [PubMed]
  5. Hutson, T.H.; Di Giovanni, S. The translational landscape in spinal cord injury: Focus on neuroplasticity and regeneration. Nat. Rev. Neurol. 2019, 15, 732–745. [Google Scholar] [CrossRef]
  6. Silva, N.A.; Sousa, N.; Reis, R.L.; Salgado, A.J. From basics to clinical: A comprehensive review on spinal cord injury. Prog. Neurobiol. 2014, 114, 25–57. [Google Scholar] [CrossRef]
  7. Tator, C.H.; Fehlings, M.G. Review of the secondary injury theory of acute spinal cord trauma with emphasis on vascular mechanisms. J. Neurosurg. 1991, 75, 15–26. [Google Scholar] [CrossRef]
  8. Hilton, B.J.; Moulson, A.J.; Tetzlaff, W. Neuroprotection and secondary damage following spinal cord injury: Concepts and methods. Neurosci. Lett. 2017, 652, 3–10. [Google Scholar] [CrossRef]
  9. Kopper, T.J.; Gensel, J.C. Myelin as an inflammatory mediator: Myelin interactions with complement, macrophages, and microglia in spinal cord injury. J. Neurosci. Res. 2017, 96, 969–977. [Google Scholar] [CrossRef] [Green Version]
  10. Quadri, S.A.; Farooqui, M.; Ikram, A.; Zafar, A.; Khan, M.A.; Suriya, S.S.; Claus, C.F.; Fiani, B.; Rahman, M.; Ramachandran, A.; et al. Recent update on basic mechanisms of spinal cord injury. Neurosurg. Rev. 2018, 43, 425–441. [Google Scholar] [CrossRef]
  11. Thion, M.S.; Ginhoux, F.; Garel, S. Microglia and early brain development: An intimate journey. Science 2018, 362, 185–189. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Pasciuto, E.; Burton, O.T.; Roca, C.P.; Lagou, V.; Rajan, W.D.; Theys, T.; Mancuso, R.; Tito, R.Y.; Kouser, L.; Callaerts-Vegh, Z.; et al. Microglia Require CD4 T Cells to Complete the Fetal-to-Adult Transition. Cell 2020, 182, 625–640. [Google Scholar] [CrossRef] [PubMed]
  13. Hammond, T.R.; Dufort, C.; Dissing-Olesen, L.; Giera, S.; Young, A.; Wysoker, A.; Walker, A.J.; Gergits, F.; Segel, M.; Nemesh, J.; et al. Single-Cell RNA Sequencing of Microglia throughout the Mouse Lifespan and in the Injured Brain Reveals Complex Cell-State Changes. Immunity 2019, 50, 253–271. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Elmore, M.R.P.; Najafi, A.R.; Koike, M.A.; Dagher, N.N.; Spangenberg, E.E.; Rice, R.A.; Kitazawa, M.; Matusow, B.; Nguyen, H.; West, B.L.; et al. Colony-Stimulating Factor 1 Receptor Signaling Is Necessary for Microglia Viability, Unmasking a Microglia Progenitor Cell in the Adult Brain. Neuron 2014, 82, 380–397. [Google Scholar] [CrossRef] [Green Version]
  15. Ginhoux, F.; Greter, M.; Leboeuf, M.; Nandi, S.; See, P.; Gokhan, S.; Mehler, M.F.; Conway, S.J.; Ng, L.G.; Stanley, E.R.; et al. Fate mapping analysis reveals that adult microglia derive from primitive macrophages. Science 2010, 330, 841–845. [Google Scholar] [CrossRef] [Green Version]
  16. Morimoto, K.; Nakajima, K. Role of the Immune System in the Development of the Central Nervous System. Front. Neurosci. 2019, 13, 916. [Google Scholar] [CrossRef] [Green Version]
  17. Hughes, A.N.; Appel, B. Microglia phagocytose myelin sheaths to modify developmental myelination. Nat. Neurosci. 2020, 23, 1055–1066. [Google Scholar] [CrossRef]
  18. Nemes-Baran, A.D.; White, D.R.; DeSilva, T.M. Fractalkine-Dependent Microglial Pruning of Viable Oligodendrocyte Progenitor Cells Regulates Myelination. Cell Rep. 2020, 32, 108047. [Google Scholar] [CrossRef]
  19. Scott-Hewitt, N.; Perrucci, F.; Morini, R.; Erreni, M.; Mahoney, M.; Witkowska, A.; Carey, A.; Faggiani, E.; Schuetz, L.T.; Mason, S.; et al. Local externalization of phosphatidylserine mediates developmental synaptic pruning by microglia. EMBO J. 2020, 39, e105380. [Google Scholar] [CrossRef]
  20. Cheadle, L.; Rivera, S.A.; Phelps, J.S.; Ennis, K.A.; Stevens, B.; Burkly, L.C.; Lee, W.-C.A.; Greenberg, M.E. Sensory Experience Engages Microglia to Shape Neural Connectivity through a Non-Phagocytic Mechanism. Neuron 2020, 108, 451–468.e9. [Google Scholar] [CrossRef]
  21. Hanisch, U.-K.; Kettenmann, H. Microglia: Active sensor and versatile effector cells in the normal and pathologic brain. Nat. Neurosci. 2007, 10, 1387–1394. [Google Scholar] [CrossRef] [PubMed]
  22. Li, Q.; Barres, B.A. Microglia and macrophages in brain homeostasis and disease. Nat. Rev. Immunol. 2017, 18, 225–242. [Google Scholar] [CrossRef] [PubMed]
  23. Xu, L.; He, D.; Bai, Y. Microglia-Mediated Inflammation and Neurodegenerative Disease. Mol. Neurobiol. 2015, 53, 6709–6715. [Google Scholar] [CrossRef] [PubMed]
  24. Kierdorf, K.; Erny, D.; Goldmann, T.; Sander, V.; Schulz, C.; Perdiguero, E.G.; Wieghofer, P.; Heinrich, A.; Riemke, P.; Hölscher, C.; et al. Microglia emerge from erythromyeloid precursors via Pu.1- and Irf8-dependent pathways. Nat. Neurosci. 2013, 16, 273–280. [Google Scholar] [CrossRef]
  25. Vainchtein, I.D.; Chin, G.; Cho, F.S.; Kelley, K.W.; Miller, J.G.; Chien, E.C.; Liddelow, S.A.; Nguyen, P.T.; Nakao-Inoue, H.; Dorman, L.C.; et al. Astrocyte-derived interleukin-33 promotes microglial synapse engulfment and neural circuit development. Science 2018, 359, 1269–1273. [Google Scholar] [CrossRef] [Green Version]
  26. Zengeler, K.E.; Lukens, J.R. Innate immunity at the crossroads of healthy brain maturation and neurodevelopmental disorders. Nat. Rev. Immunol. 2021, 21, 454–468. [Google Scholar] [CrossRef]
  27. Kobashi, S.; Terashima, T.; Katagi, M.; Nakae, Y.; Okano, J.; Suzuki, Y.; Urushitani, M.; Kojima, H. Transplantation of M2-Deviated Microglia Promotes Recovery of Motor Function after Spinal Cord Injury in Mice. Mol. Ther. 2019, 28, 254–265. [Google Scholar] [CrossRef]
  28. Kroner, A.; Almanza, J.R. Role of microglia in spinal cord injury. Neurosci. Lett. 2019, 709, 134370. [Google Scholar] [CrossRef]
  29. Sierra, A.; de Castro, F.; Del, R.J.; Rafael, I.J.; Garrosa, M.; Kettenmann, H. The “Big-Bang” for modern glial biology: Translation and comments on Pio del Rio-Hortega 1919 series of papers on microglia. Glia 2016, 64, 1801–1840. [Google Scholar] [CrossRef]
  30. Venkatesh, K.; Ghosh, S.K.; Mullick, M.; Manivasagam, G.; Sen, D. Spinal cord injury: Pathophysiology, treatment strategies, associated challenges, and future implications. Cell Tissue Res. 2019, 377, 125–151. [Google Scholar] [CrossRef]
  31. Chen, J.; Wang, Z.; Zheng, Z.; Chen, Y.; Khor, S.; Shi, K.; He, Z.; Wang, Q.; Zhao, Y.; Zhang, H.; et al. Neuron and microglia/macrophage-derived FGF10 activate neuronal FGFR2/PI3K/Akt signaling and inhibit microglia/macrophages TLR4/NF-kappaB-dependent neuroinflammation to improve functional recovery after spinal cord injury. Cell Death Dis. 2017, 8, e3090. [Google Scholar] [CrossRef] [PubMed]
  32. Dietz, V.; Fouad, K. Restoration of sensorimotor functions after spinal cord injury. Brain 2013, 137, 654–667. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Liddelow, S.A.; Guttenplan, K.A.; Clarke, L.E.; Bennett, F.C.; Bohlen, C.J.; Schirmer, L.; Bennett, M.L.; Münch, A.E.; Chung, W.-S.; Peterson, T.C.; et al. Neurotoxic reactive astrocytes are induced by activated microglia. Nature 2017, 541, 481–487. [Google Scholar] [CrossRef] [PubMed]
  34. Bastien, D.; Bellver Landete, V.; Lessard, M.; Vallieres, N.; Champagne, M.; Takashima, A.; Tremblay, M.E.; Doyon, Y.; Lacroix, S. IL-1alpha Gene Deletion Protects Oligodendrocytes after Spinal Cord Injury through Upregulation of the Survival Factor. ToxJ Neurosci. 2015, 35, 10715–10730. [Google Scholar] [CrossRef] [Green Version]
  35. Kroner, A.; Greenhalgh, A.D.; Zarruk, J.G.; dos Santos, R.P.; Gaestel, M.; David, S. TNF and Increased Intracellular Iron Alter Macrophage Polarization to a Detrimental M1 Phenotype in the Injured Spinal Cord. Neuron 2014, 83, 1098–1116. [Google Scholar] [CrossRef] [Green Version]
  36. Galvan, M.D.; Luchetti, S.; Burgos, A.M.; Nguyen, H.X.; Hooshmand, M.J.; Hamers, F.P.T.; Anderson, A.J. Deficiency in Complement C1q Improves Histological and Functional Locomotor Outcome after Spinal Cord Injury. J. Neurosci. 2008, 28, 13876–13888. [Google Scholar] [CrossRef] [Green Version]
  37. Wang, A.-P.; Tian, Y.; Zhang, W.; Tian, T.; Gong, S.-X.; Huang, W.-Q.; Zhou, Q.-Y. Microglia-associated neuroinflammation is a potential therapeutic target for ischemic stroke. Neural Regen. Res. 2021, 16, 6–11. [Google Scholar] [CrossRef]
  38. Orihuela, R.; McPherson, C.A.; Harry, G.J. Microglial M1/M2 polarization and metabolic states. Br. J. Pharmacol. 2016, 173, 649–665. [Google Scholar] [CrossRef]
  39. Ren, Y.; Zhou, X.; He, X. Function of microglia and macrophages in secondary damage after spinal cord injury. Neural Regen. Res. 2014, 9, 1787–1795. [Google Scholar] [CrossRef]
  40. Liu, W.; Rong, Y.; Wang, J.; Zhou, Z.; Ge, X.; Ji, C.; Jiang, D.; Gong, F.; Li, L.; Chen, J.; et al. Exosome-shuttled miR-216a-5p from hypoxic preconditioned mesenchymal stem cells repair traumatic spinal cord injury by shifting microglial M1/M2 polarization. J. Neuroinflamm. 2020, 17, 47. [Google Scholar] [CrossRef] [Green Version]
  41. Kigerl, K.A.; Lai, W.; Wallace, L.M.; Yang, H.; Popovich, P.G. High mobility group box-1 (HMGB1) is increased in injured mouse spinal cord and can elicit neurotoxic inflammation. Brain Behav. Immun. 2017, 72, 22–33. [Google Scholar] [CrossRef] [PubMed]
  42. Kwon, H.S.; Koh, S.-H. Neuroinflammation in neurodegenerative disorders: The roles of microglia and astrocytes. Transl. Neurodegener. 2020, 9, 1–12. [Google Scholar] [CrossRef] [PubMed]
  43. Cherry, J.D.; Olschowka, J.A.; O’Banion, M.K. Neuroinflammation and M2 microglia: The good, the bad, and the inflamed. J. Neuroinflamm. 2014, 11, 98. [Google Scholar] [CrossRef] [Green Version]
  44. David, S.; Kroner, A. Repertoire of microglial and macrophage responses after spinal cord injury. Nat. Rev. Neurosci. 2011, 12, 388–399. [Google Scholar] [CrossRef] [PubMed]
  45. Kisucká, A.; Bimbová, K.; Bačová, M.; Gálik, J.; Lukáčová, N. Activation of Neuroprotective Microglia and Astrocytes at the Lesion Site and in the Adjacent Segments Is Crucial for Spontaneous Locomotor Recovery after Spinal Cord Injury. Cells 2021, 10, 1943. [Google Scholar] [CrossRef] [PubMed]
  46. Lukacova, N.; Kisucka, A.; Bimbova, K.K.; Bacova, M.; Ileninova, M.; Kuruc, T.; Galik, J. Glial-Neuronal Interactions in Pathogenesis and Treatment of Spinal Cord Injury. Int. J. Mol. Sci. 2021, 22, 13577. [Google Scholar] [CrossRef] [PubMed]
  47. Hu, X.; Leak, R.K.; Shi, Y.; Suenaga, J.; Gao, Y.; Zheng, P.; Chen, J. Microglial and macrophage polarization—New prospects for brain repair. Nat. Rev. Neurol. 2015, 11, 56–64. [Google Scholar] [CrossRef] [PubMed]
  48. Kigerl, K.A.; Gensel, J.C.; Ankeny, D.P.; Alexander, J.K.; Donnelly, D.J.; Popovich, P.G. Identification of Two Distinct Macrophage Subsets with Divergent Effects Causing either Neurotoxicity or Regeneration in the Injured Mouse Spinal Cord. J. Neurosci. 2009, 29, 13435–13444. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Li, Y.; He, X.; Kawaguchi, R.; Zhang, Y.; Wang, Q.; Monavarfeshani, A.; Yang, Z.; Chen, B.; Shi, Z.; Meng, H.; et al. Microglia-organized scar-free spinal cord repair in neonatal mice. Nature 2020, 587, 613–618. [Google Scholar] [CrossRef] [PubMed]
  50. Bellver-Landete, V.; Bretheau, F.; Mailhot, B.; Vallières, N.; Lessard, M.; Janelle, M.-E.; Vernoux, N.; Tremblay, M.; Fuehrmann, T.; Shoichet, M.S.; et al. Microglia are an essential component of the neuroprotective scar that forms after spinal cord injury. Nat. Commun. 2019, 10, 1–18. [Google Scholar] [CrossRef] [Green Version]
  51. Fan, H.; Zhang, K.; Shan, L.; Kuang, F.; Chen, K.; Zhu, K.; Ma, H.; Ju, G.; Wang, Y.-Z. Reactive astrocytes undergo M1 microglia/macrohpages-induced necroptosis in spinal cord injury. Mol. Neurodegener. 2016, 11, 1–16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Hakim, R.; Zachariadis, V.; Sankavaram, S.R.; Han, J.; Harris, R.A.; Brundin, L.; Enge, M.; Svensson, M. Spinal Cord Injury Induces Permanent Reprogramming of Microglia into a Disease-Associated State Which Contributes to Functional Recovery. J. Neurosci. 2021, 41, 8441–8459. [Google Scholar] [CrossRef] [PubMed]
  53. Fu, H.; Zhao, Y.; Hu, D.; Wang, S.; Yu, T.; Zhang, L. Depletion of microglia exacerbates injury and impairs function recovery after spinal cord injury in mice. Cell Death Dis. 2020, 11, 1–12. [Google Scholar] [CrossRef] [PubMed]
  54. Li, Y.; Ritzel, R.M.; Khan, N.; Cao, T.; He, J.; Lei, Z.; Matyas, J.J.; Sabirzhanov, B.; Liu, S.; Li, H.; et al. Delayed microglial depletion after spinal cord injury reduces chronic inflammation and neurodegeneration in the brain and improves neurological recovery in male mice. Theranostics 2020, 10, 11376–11403. [Google Scholar] [CrossRef] [PubMed]
  55. Jakovčevski, I.; Förster, E.; Reiss, G.; Schachner, M. Impact of Depletion of Microglia/Macrophages on Regeneration after Spinal Cord Injury. Neuroscience 2021, 459, 129–141. [Google Scholar] [CrossRef] [PubMed]
  56. Poulen, G.; Aloy, E.; Bringuier, C.M.; Mestre-Francés, N.; Artus, E.V.; Cardoso, M.; Perez, J.-C.; Goze-Bac, C.; Boukhaddaoui, H.; Lonjon, N.; et al. Inhibiting microglia proliferation after spinal cord injury improves recovery in mice and nonhuman primates. Theranostics 2021, 11, 8640–8659. [Google Scholar] [CrossRef]
  57. Poulen, G.; Bartolami, S.; Noristani, H.N.; Perrin, F.E.; Gerber, Y.N. Unlike Brief Inhibition of Microglia Proliferation after Spinal Cord Injury, Long-Term Treatment Does Not Improve Motor Recovery. Brain Sci. 2021, 11, 1643. [Google Scholar] [CrossRef]
  58. Ma, D.; Zhao, Y.; Huang, L.; Xiao, Z.; Chen, B.; Shi, Y.; Shen, H.; Dai, J. A novel hydrogel-based treatment for complete transection spinal cord injury repair is driven by microglia/macrophages repopulation. Biomaterials 2020, 237, 119830. [Google Scholar] [CrossRef]
  59. Parkhurst, C.N.; Yang, G.; Ninan, I.; Savas, J.N.; Yates, J.R., III.; Lafaille, J.J.; Hempstead, B.L.; Littman, D.R.; Gan, W.-B. Microglia promote learning-dependent synapse formation through brain-derived neurotrophic factor. Cell 2013, 155, 1596–1609. [Google Scholar] [CrossRef] [Green Version]
  60. Wu, W.; Li, Y.; Wei, Y.; Bosco, D.B.; Xie, M.; Zhao, M.-G.; Richardson, J.; Wu, L.-J. Microglial depletion aggravates the severity of acute and chronic seizures in mice. Brain Behav. Immun. 2020, 89, 245–255. [Google Scholar] [CrossRef]
  61. Wang, C.; Yue, H.; Hu, Z.; Shen, Y.; Ma, J.; Li, J.; Wang, X.-D.; Wang, L.; Sun, B.; Shi, P.; et al. Microglia mediate forgetting via complement-dependent synaptic elimination. Science 2020, 367, 688–694. [Google Scholar] [CrossRef] [PubMed]
  62. Willis, E.; MacDonald, K.P.; Nguyen, Q.; Garrido, A.L.; Gillespie, E.R.; Harley, S.B.; Bartlett, P.F.; Schroder, W.A.; Yates, A.G.; Anthony, D.; et al. Repopulating Microglia Promote Brain Repair in an IL-6-Dependent Manner. Cell 2020, 180, 833–846.e16. [Google Scholar] [CrossRef] [PubMed]
  63. Huang, Y.; Xu, Z.; Xiong, S.; Sun, F.; Qin, G.; Hu, G.; Wang, J.; Zhao, L.; Liang, Y.-X.; Wu, T.; et al. Repopulated microglia are solely derived from the proliferation of residual microglia after acute depletion. Nat. Neurosci. 2018, 21, 530–540. [Google Scholar] [CrossRef] [PubMed]
  64. Gerber, Y.N.; Saint-Martin, G.; Bringuier, C.M.; Bartolami, S.; Goze-Bac, C.; Noristani, H.; Perrin, F.E. CSF1R Inhibition Reduces Microglia Proliferation, Promotes Tissue Preservation and Improves Motor Recovery After Spinal Cord Injury. Front. Cell. Neurosci. 2018, 12, 368. [Google Scholar] [CrossRef] [Green Version]
  65. Beckmann, N.; Giorgetti, E.; Neuhaus, A.; Zurbruegg, S.; Accart, N.; Smith, P.; Perdoux, J.; Perrot, L.; Nash, M.; Desrayaud, S.; et al. Brain region-specific enhancement of remyelination and prevention of demyelination by the CSF1R kinase inhibitor BLZ. Acta Neuropathol. Commun. 2018, 6, 9. [Google Scholar] [CrossRef] [Green Version]
  66. Asai, H.; Ikezu, S.; Tsunoda, S.; Medalla, M.; Luebke, J.; Haydar, T.; Wolozin, B.; Butovsky, O.; Kügler, S.; Ikezu, T. Depletion of microglia and inhibition of exosome synthesis halt tau propagation. Nat. Neurosci. 2015, 18, 1584–1593. [Google Scholar] [CrossRef]
  67. Miron, V.E.; Boyd, A.; Zhao, J.-W.; Yuen, T.J.; Ruckh, J.M.; Shadrach, J.L.; van Wijngaarden, P.; Wagers, A.J.; Williams, A.; Franklin, R.J.M.; et al. M2 microglia and macrophages drive oligodendrocyte differentiation during CNS remyelination. Nat. Neurosci. 2013, 16, 1211–1218. [Google Scholar] [CrossRef] [Green Version]
  68. Lei, F.; Cui, N.; Zhou, C.; Chodosh, J.; Vavvas, D.G.; Paschalis, E.I. CSF1R inhibition by a small-molecule inhibitor is not microglia specific; affecting hematopoiesis and the function of macrophages. Proc. Natl. Acad. Sci. USA 2020, 117, 23336–23338. [Google Scholar] [CrossRef]
  69. Qin, Y.; Qiu, J.; Wang, P.; Liu, J.; Zhao, Y.; Jiang, F.; Lou, H. Impaired autophagy in microglia aggravates dopaminergic neurodegeneration by regulating NLRP3 inflammasome activation in experimental models of Parkinson’s disease. Brain Behav. Immun. 2020, 91, 324–338. [Google Scholar] [CrossRef]
  70. Heppner, F.; Greter, M.; Marino, D.; Falsig, J.; Raivich, G.; Hövelmeyer, N.; Waisman, A.; Rülicke, T.; Prinz, M.; Priller, J.; et al. Experimental autoimmune encephalomyelitis repressed by microglial paralysis. Nat. Med. 2005, 11, 146–152. [Google Scholar] [CrossRef]
  71. Varvel, N.H.; Grathwohl, S.A.; Baumann, F.; Liebig, C.; Bosch, A.; Brawek, B.; Thal, D.R.; Charo, I.F.; Heppner, F.L.; Aguzzi, A.; et al. Microglial repopulation model reveals a robust homeostatic process for replacing CNS myeloid cells. Proc. Natl. Acad. Sci. USA 2012, 109, 18150–18155. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Zhao, R.; Hu, W.; Tsai, J.; Li, W.; Gan, W.B. Microglia limit the expansion of beta-amyloid plaques in a mouse model of Alzheimer’s disease. Mol. Neurodegener. 2017, 12, 47. [Google Scholar] [CrossRef] [PubMed]
  73. Henry, R.J.; Ritzel, R.; Barrett, J.P.; Doran, S.J.; Jiao, Y.; Leach, J.B.; Szeto, G.L.; Wu, J.; Stoica, B.A.; Faden, A.I.; et al. Microglial Depletion with CSF1R Inhibitor During Chronic Phase of Experimental Traumatic Brain Injury Reduces Neurodegeneration and Neurological Deficits. J. Neurosci. 2020, 40, 2960–2974. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The polarization process of microglia after SCI.
Figure 1. The polarization process of microglia after SCI.
Cells 11 01871 g001
Table 1. The different strategy of microglia depletion and their respective characteristics.
Table 1. The different strategy of microglia depletion and their respective characteristics.
CategoryStrategyAdvantagesDisadvantagesReference
Gene manipulationCreERT systemPrecise timing control;
Microglia-specific removal;
Need for tamoxifen and diphtheria toxin induction;
Relatively time-consuming;
Species limitations;
Hakim et al. [52];
Parkhurst et al. [59].
Cre-Flox systemEasy to operate, and microglia are removed after birth;
No need for tamoxifen and diphtheria toxin induction;
Difficult to achieve precise timing control;
Possible adverse effects on peripheral mononuclear macrophages;
Relatively time-consuming;
Species limitations;
Li et al. [49];
Qin et al. [69].
HSVTK systemNo need for tamoxifen or diphtheria toxin induction;Difficult to achieve precise timing control;
Possible adverse effects on peripheral mononuclear macrophages;
Relatively time-consuming;
Species limitations;
Igor et al. [55];
Heppner et al. [70];
Varvel et al. [71].
Small-molecule compoundsCSF1R inhibitorsOral or injectable administration and some can pass through the blood–brain barrier;
Relatively time-saving;
No species limitation;
Not specific to microglia and affect hematopoietic and macrophage functions;
Difficult to achieve precise timing control;
Elmore et al. [14];
Lei et al. [68].
Chlorophosphate liposomesLocal microglia depletion;
Relatively time-saving;
No species limitation;
Need to inject into a specific site in the CNS so increase the risk of secondary damage;
Not specific for microglia.
Asai et al. [66].
Gadolinium chlorideSelectively remove M1-type microglia without affecting M2-type microglia;
Relatively time-saving;
No species limitation;
Need to inject into a specific site in the CNS so increase the risk of secondary damage.Miron et al. [67].
Table 2. The outcomes and therapeutic effects of the different timing of microglia depletion on SCI.
Table 2. The outcomes and therapeutic effects of the different timing of microglia depletion on SCI.
Depleting ApproachesDepleting TimingModelOutcomesTherapeutic EffectsReferences
CreERT systemMicroglia were depleted after Tamoxifen injectionContusionHarmfulAffect function recovery following injuryHakim et al. [52].
Cre-Flox systemMicroglia were depleted after birthCrushHarmfulImpair bridge formation and axon regenerationLi et al. [49].
HSVTK system2 weeks before SCI and 2 weeks post SCICompressionBeneficialImprove locomotor recovery in the early phase post SCI, but does not affect recovery in the following 4 weeksIgor et al. [55].
PLX56223 weeks before SCI to 5 weeks post SCIContusionHarmfulWorse functional recoveryVictor et al. [50].
PLX56221 day post SCI to 6 weeks post SCIContusionBeneficialEnhance cognitive behavior but failed to improve locomotion functionLi et al. [54].
PLX3397Pregnant mice received PLX from E14 and newborn pups continued to receive PLX 2 weeks post SCICrushHarmfulImpair bridge formation and axon regenerationLi et al. [49].
PLX33971 week before SCI to 4 weeks post SCICrushHarmfulImpair locomotor function and exacerbate tissue damageFu et al. [53].
PLX33970 day post SCI to 2 weeks post SCITransectionBeneficialEnhance the electrophysiological activity and functional recovery Ma et al. [58].
GW25804 weeks before SCI to 6 weeks post injuryHemisectionBeneficialImprove the fine motor function and decrease the gliosisGerber et al. [64].
GW25800 day post injury for 1 week (mice) or 2 weeks (non-human primates)HemisectionBeneficialPromote functional recovery and tissue repairPoulen et al. [56].
GW25800 day post injury for 6 weeks HemisectionNot beneficialNo impact on functional recoveryPoulen et al. [58].
GdCl31 day post SCI for onceContusionHarmfulActivate astrocytes and disrupt tissue repair and nerve regenerationFan et al. [51].
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Deng, J.; Meng, F.; Zhang, K.; Gao, J.; Liu, Z.; Li, M.; Liu, X.; Li, J.; Wang, Y.; Zhang, L.; et al. Emerging Roles of Microglia Depletion in the Treatment of Spinal Cord Injury. Cells 2022, 11, 1871. https://doi.org/10.3390/cells11121871

AMA Style

Deng J, Meng F, Zhang K, Gao J, Liu Z, Li M, Liu X, Li J, Wang Y, Zhang L, et al. Emerging Roles of Microglia Depletion in the Treatment of Spinal Cord Injury. Cells. 2022; 11(12):1871. https://doi.org/10.3390/cells11121871

Chicago/Turabian Style

Deng, Junhao, Fanqi Meng, Kexue Zhang, Jianpeng Gao, Zhongyang Liu, Ming Li, Xiao Liu, Jiantao Li, Yu Wang, Licheng Zhang, and et al. 2022. "Emerging Roles of Microglia Depletion in the Treatment of Spinal Cord Injury" Cells 11, no. 12: 1871. https://doi.org/10.3390/cells11121871

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop