Next Article in Journal
A Bacteriophage Microgel Effectively Treats the Multidrug-Resistant Acinetobacter baumannii Bacterial Infections in Burn Wounds
Next Article in Special Issue
Multidirectional Effects of Terpenoids from Sorbus intermedia (EHRH.) PERS Fruits in Cellular Model of Benign Prostate Hyperplasia
Previous Article in Journal
Cutaneous Polymeric-Micelles-Based Hydrogel Containing Origanum vulgare L. Essential Oil: In Vitro Release and Permeation, Angiogenesis, and Safety Profile In Ovo
Previous Article in Special Issue
Margaritaria nobilis L.f. (Phyllanthaceae) Ethanolic Extract: Low Acute Oral Toxicity and Antinociceptive Activity
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Effects of Natural Product-Derived Compounds on Inflammatory Pain via Regulation of Microglial Activation

1
Division of Functional Food Research, Korea Food Research Institute, Wanju 55365, Republic of Korea
2
Department of Food Biotechnology, Korea University of Science and Technology, Daejeon 34113, Republic of Korea
3
Department of Anesthesiology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
*
Author to whom correspondence should be addressed.
Pharmaceuticals 2023, 16(7), 941; https://0-doi-org.brum.beds.ac.uk/10.3390/ph16070941
Submission received: 8 June 2023 / Revised: 22 June 2023 / Accepted: 26 June 2023 / Published: 29 June 2023

Abstract

:
Inflammatory pain is a type of pain caused by tissue damage associated with inflammation and is characterized by hypersensitivity to pain and neuroinflammation in the spinal cord. Neuroinflammation is significantly increased by various neurotransmitters and cytokines that are expressed in activated primary afferent neurons, and it plays a pivotal role in the development of inflammatory pain. The activation of microglia and elevated levels of pro-inflammatory cytokines are the hallmark features of neuroinflammation. During the development of neuroinflammation, various intracellular signaling pathways are activated or inhibited in microglia, leading to the regulation of inflammatory proteins and cytokines. Numerous attempts have been conducted to alleviate inflammatory pain by inhibiting microglial activation. Natural products and their compounds have gained attention as potential candidates for suppressing inflammatory pain due to verified safety through centuries of use. Many studies have also shown that natural product-derived compounds have the potential to suppress microglial activation and alleviate inflammatory pain. Herein, we review the literature on inflammatory mediators and intracellular signaling involved in microglial activation in inflammatory pain, as well as natural product-derived compounds that have been found to suppress microglial activation. This review suggests that natural product-derived compounds have the potential to alleviate inflammatory pain through the suppression of microglial activation.

1. Introduction

The International Association for the Study of Pain (IASP) describes pain as an unpleasant sensory and emotional experience associated with, or resembling that associated with, actual or potential tissue damage. If pain persists for more than 3 months, which is considered the tissue healing period, it is diagnosed as chronic pain. In 2019, approximately 20% of adults in the US were diagnosed with chronic pain, and the number of patients with chronic pain is increasing with the increase in the aging population [1]. According to a National Institute of Health (NIH) report, the cost of treating chronic pain exceeds that of a few major diseases related to the highest morbidity and mortality, such as cardiovascular diseases (USD 309 billion), cancers (USD 243 billion), and injuries (USD 205 billion) [2]. Patients with chronic pain have a poor quality of life for reasons such as difficulty engaging in daily activities and prolonged treatment. Patients with chronic pain also suffer from mental disorders such as depression, anxiety disorders, and sleep disturbances [3]. As the average lifespan continues to increase, the importance of pain management is also increasing.
Pain perception is a complex and highly orchestrated process involving a series of sequential events [4]. Following inflammation-related tissue damage, nociceptors, which are sensory neurons responsible for detecting harmful stimuli, initiate a response. Nociceptors convert stimuli into electrical signals, which are then transmitted to the central nervous system (CNS). Subsequently, these electrical signals are transmitted to secondary afferent neurons located in the dorsal horn of the spinal cord. With the repetitive transmission of pain signals, neuroinflammation is strongly induced within the spinal cord. Neuroinflammation profoundly influences synaptic transmission, thereby contributing to the persistence of pain.
The causes of chronic pain include nerve injury, cancer, muscle injury, and inflammation [5]. Among the various causes, pain caused by inflammation is called inflammatory pain. Currently, non-steroidal anti-inflammatory drugs (NSAIDs) are the most commonly used drugs to treat inflammatory pain [6]. The main mechanism underlying the analgesic and anti-inflammatory effects of NSAIDs is inhibition of the cyclooxygenase (COX2) enzyme, which produces prostaglandins. Prostaglandin is a representative inflammatory mediator that induces fever, inflammation, and pain. Aspirin, naproxen, and ibuprofen are some of the commonly used NSAIDs. However, since NSAIDs have adverse effects, such as indigestion, stomach ulcers, headaches, drowsiness, and dizziness, interest in natural products as agents for alleviating inflammatory pain has recently increased. Natural products have been used for centuries to treat various diseases related with inflammation, without causing side effects [7]. Therefore, natural products have the potential to be developed into new drugs against inflammatory pain.
In this review, we aim to summarize the current understanding of inflammatory factors and intracellular signaling involved in the development of inflammatory pain as well as to highlight the potential of natural products in treating inflammatory pain.

2. Mechanism Underlying the Development of Inflammatory Pain

Inflammatory pain is characterized by a heightened sensitivity to pain due to tissue damage resulting from an inflammatory or immune response. The two typical symptoms of pain are hyperalgesia and allodynia. Hyperalgesia is characterized by an abnormally increased sensitivity to pain and an extreme response to pain. Allodynia is recognized as pain for common stimuli that do not normally cause pain. Chronic inflammatory diseases and infections, such as arthritis, shingles, and tissue injury are representative causes of inflammatory pain [8]. Inflammatory mediators are released locally by immune cells at the site of inflammation and can directly activate sensory neurons in peripheral tissues. Activated sensory neurons then release neuropeptides, such as substance P, calcitonin gene-related peptide, and prostanoids, into the dorsal horn of the spinal cord. Repetitive and persistent stimulation of sensory neurons can lead to the over-release of neuropeptides, resulting in neuroinflammation of the spinal cord.
Spinal neuroinflammation, caused by peripheral inflammation, is characterized by the activation of microglia and increased expression of inflammatory mediators in the spinal cord [9]. Activated microglia are major sources of pro-inflammatory cytokines and inflammation-related proteins that are regulated by various intracellular signaling (Figure 1). Along with spinal neuroinflammation, microglial activation is significantly induced, resulting in pain hypersensitivity through central sensitization. Central sensitization, a leading cause of chronic pain, represents the reinforcement of the function between pre- and post-synaptic neurons in the nociceptive pathway caused by increasing excitatory transmission and strengthening of synapses in response to inflammation and nerve injury. The increased levels of pro-inflammatory cytokines can enhance synaptic transmission by increasing excitatory synaptic transmission and decreasing inhibitory synaptic transmission in the dorsal horn of spinal cord [10,11]. Pro-inflammatory cytokines may induce gene expression by activating cAMP response element-binding protein (CREB) transcription factors, leading to long-term potentiation [12]. These results comprehensively contribute to the persistence and hypersensitivity of pain through central sensitization.
Microglia, the resident macrophage-like cells located in the CNS, play an important role in the development of chronic pain associated with neuroinflammation [13,14]. As the first immune cell in the CNS, microglia are essential for brain maintenance and homeostasis as they are involved in removing cell debris, infectious agents, and other unnecessary elements. Although the primary function of microglia is to protect the CNS, they can have destructive effects on neurons. Various signaling molecules are released from sensory neurons damaged by inflammation and eventually activate microglia [13]. Microglial activation is characterized by the increased production of inflammatory mediators, such as iNOS, COX-2, MMP-9, TNF-α, IL-1β, IL-6, MCP1, and MCP3. The expression of these inflammatory mediators is regulated by intracellular signaling pathways, including NF-B, MAPK, JAK2-STAT3, Nrf2, and autophagy. The expression and secretion of inflammatory mediators are induced by intracellular signaling in activated microglia, resulting in an increase in neuroinflammation [15]. Inflammatory mediators contribute to increased neuroinflammation to damage to cells. Furthermore, these mediators and cytokines are involved in the induction and maintenance of central sensitization by upregulating the NMDA receptor in excitatory synaptic neurons [16]. In agreement with these findings, pro-inflammatory cytokines, such as TNF-α, IL-1β, or IL-6, modulate the function of receptors associated with central sensitization in the spinal cord [12]. Experimental studies using rodent models have shown that intrathecal injection of pro-inflammatory cytokines induces pain hypersensitivity [10,17,18]. A previous study showed that the specific deletion of microglia in the spinal cord had an inhibitory effect on formalin-induced inflammatory pain via the modulation of central sensitization [19]. Inhibition of microglial activation using chemogenetic approaches, specifically DREADD, alleviate neuroinflammation and chronic pain following nerve injury [20]. Inhibition of microglial activation attenuated nerve injury-induced pain hypersensitivity in the early phase but not the late phase. Therefore, understanding and targeting the processes and factors involved in microglial activation-induced neuroinflammation may offer an effective approach to prevent the early phase of inflammatory pain, which has the potential to become chronic pain.

3. Expression of Inflammatory Mediators in Activated Microglia

3.1. Inducible Nitric Oxide Synthase

Nitric oxide synthase (NOS) is an enzyme that catalyzes the production of nitric oxide (NO) from L-arginine. Among the different isoforms of NOS, inducible nitric oxide synthase (iNOS) plays a significant role in the development of inflammatory pain. iNOS is induced in various cells and tissues by cytokines and other molecules. Although previous studies have reported the role of iNOS in various inflammatory diseases except for the CNS, it has been recently confirmed that iNOS contributes to the development of chronic pain [21,22]. iNOS continuously produces large amounts of NO until it is degraded. High amounts of NO result in the production of high levels of reactive nitrogen oxide species (RNOS), causing damage to the surrounding tissue and cells. Among various cells in the CNS, microglia are the major cellular sources of iNOS [23]. Many previous studies have shown that activated microglia remarkably increase the expression of iNOS, leading to the excessive production of NO [24,25,26]. Additionally, iNOS expression and neuroinflammation in the dorsal horn of the spinal cord were found to have significantly increased in CFA-injected mice [27]. Osborne et al. reported that carrageenan-induced thermal hyperalgesia was significantly alleviated by the intrathecal injection of the nonselective NOS inhibitor L-NAME in rats. Furthermore, a selective iNOS inhibitor suppressed thermal hypersensitivity during carrageenan-induced inflammatory pain [28]. Formalin-induced pain behavior was attenuated in iNOS knockout mice. Moreover, nerve injury-induced pain hypersensitivity and microglial activation in the spinal cord were suppressed in the iNOS knockout mice compared with in wild-type mice [26]. These results demonstrated that iNOS expression in activated microglia can exacerbate neuroinflammation, resulting in increased pain sensitivity.

3.2. Cyclooxygenase-2

Cyclooxygenase-2 (COX-2) is a primary target for reducing inflammation and pain. COX-2 converts arachidonic acid to prostaglandin E2 (PGE2), which is associated with inflammation and pain. COX-2 is induced in response to inflammatory stimulation and is primarily expressed by monocytes, macrophages, fibroblasts, neurons, and microglia [29,30]. COX-2 inhibition is expected to reduce inflammation and pain without causing side effects [31]. Many studies have shown that lipopolysaccharide (LPS) treatment increased COX-2 expression in microglia [32,33,34,35]. These studies suggest that neuroinflammation can be suppressed by inhibition of COX-2 expression in microglia. In addition, various COX-2 inhibitors suppressed neuroinflammation by inhibiting the release of inflammatory mediators by microglia [36]. Naproxen is a representative oral NSAID; it attenuated CFA-induced pain hypersensitivity by inhibiting COX-2 expression in the spinal cord [37]. Therefore, microglia-specific inhibition of COX-2 expression has the potential to alleviate inflammatory pain.

3.3. Matrix Metalloproteinases-9

Matrix metalloproteinases-9 (MMP-9) is a member of the zinc metalloproteinase family involved in the degradation of the extracellular matrix and is strongly implicated in the development of various neuroinflammation-related diseases [38]. Microglia are a major source of MMP-9. Nerve injury has been found to increase the expression of MMP-9 rapidly and temporarily in the dorsal root ganglion (DRG), leading to the induction of neuropathic pain [39,40]. Microglial activation is also increased by nerve injury-induced MMP-9 expression in the spinal cord. Intrathecal injection of siMMP-9 significantly attenuated nerve injury-induced pain hypersensitivity and inhibited microglial activation in the spinal cord. A previous study found that activated microglia significantly increased the expression of MMP-9 in LPS-treated microglial cells [41]. Additionally, in one study, MMP-9 expression was notably upregulated in the DRG and spinal cord in a CFA-induced inflammatory pain model [42]. This study showed that inhibition of MMP-9 had inhibitory effects on CFA-induced pain hypersensitivity in rats. These studies indicated that MMP-9 is involved in the development of microglial activation-mediated inflammatory pain.

3.4. Pro-Inflammatory Cytokines

Cytokines are secreted mainly by the immune and glial cells of the CNS. These cytokines act as intercellular mediators that control the function and differentiation of other cells [43]. In response to peripheral inflammation and tissue injury, microglia can be activated to secrete pro-inflammatory cytokines, such as TNF-α, interleukin-1β (IL-1β), IL-6, monocyte chemotactic protein-1 (MCP1), and MCP3 [24,33,34,37]. These pro-inflammatory cytokines are strongly involved in neuroinflammation and mainly contribute to the exacerbation of chronic pain.

3.4.1. TNF-α

Tumor necrosis factor-α (TNF-α) is a cytokine that causes inflammation-related diseases and might be a potential therapeutic target. Moreover, TNF-α is proposed to be a pro-inflammatory cytokine that plays a critical role in the development of chronic pain [43]. Microglia express and secrete TNF-α in response to stimuli and can also be activated by TNF-α via TNF receptors (TNFRs) [44]. In one study, TNF-α induced microglial activation, as evidenced by the increased expression of iNOS, IL-1β, and IL-6 in primary cells [45]. This study showed that LPS-induced microglial activation was partially blocked by treatment with TNFR type 1 (TNFR1) antibody. Moreover, intrathecal injection of TNF-α significantly induced pain hypersensitivity in mice. In addition, TNFR1 knockout mice exhibited better inhibition of pain hypersensitivity than TNFR2 knockout mice in CFA and formalin-induced inflammatory pain models [10]. Thus, TNF-α can activate microglia via binding to TNFR1 to increase pain sensitivity.

3.4.2. Interleukin-1β

Interleukin-1β (IL-1β) is one of the important mediators of the inflammatory response and is implicated in microglial activation-mediated inflammatory pain. The IL-1β precursor is cleaved by cytosolic caspase 1 and activated to mediate the inflammatory response. Previous studies showed that IL-1β could activate microglia, as revealed by the increased expression of IL-6, MCP1, and CXCL10 in human microglia cells [46,47]. IL-1β is a major mediator that increases the expression of COX-2 in the spinal cord, resulting in the development of CFA-induced inflammatory pain [48]. In one study of a mouse model, inhibition of IL-1β in the spinal cord decreased sensitivity to pain by decreasing the expression of COX2. Another study reported that intravenous injection of IL-1β significantly increased pain hypersensitivity and microglial activation in dorsal horn of the spinal cord [49].

3.4.3. Interelukin-6

Interelukin-6 (IL-6) is also a well-known inflammatory cytokine along with TNF-α and IL-1β. IL-6 is an important mediator of fever and pathogenesis of chronic pain. In one study, exposure to LPS remarkably induced the expression of IL-6 in microglia [50]. Furthermore, formalin- and CFA-injected mice showed notably increased microglial activation and IL-6 expression in the spinal cord [51,52]. In addition, nerve injury-induced IL-6 expression was decreased by microglia inhibitor in serum and spinal cord [17,53]. This study showed that intrathecal injection of IL-6 significantly increased microglial activation in the spinal cord. These data indicate the presence of a positive feedback loop between IL-6 and microglial activation, resulting in the pathogenesis of microglial activation-mediated inflammatory pain.

3.4.4. Monocyte Chemoattractant Protein-1

MCP1 is one of the key chemokines that control the migration and infiltration of monocyte. MCP1 plays an important role in the development of chronic pain. Although MCP1 is known to interact with several receptors, C-C chemokine receptor type 2 (CCR2) is its preferred receptor [54]. Direct injection of MCP1 into the spinal cord induced pain hypersensitivity, whereas co-treatment with an MCP1 inhibitor reduces sensitivity to pain by blocking central sensitization [11]. This study showed that CFA-induced pain hypersensitivity in mice was significantly attenuated by intrathecal injection of a CCR2 inhibitor. Additionally, the intrathecal injection of MCP1 induced microglial activation in the spinal cord [55]; spinal microglial activation was also markedly decreased by an antibody against MCP1. A previous study reported that MCP1 was strongly increased in DRG neurons due to peripheral inflammation and was transported into the spinal cord [56]. In addition, neuron-derived MCP1 notably induced microglial activation, as evidenced by the upregulated expression of iNOS, COX2, IL-1β, and IL-6 [57]. Several in vitro experiments showed that activated microglia could secrete MCP1 [24,58]. These results suggest that peripheral inflammation induces MCP1 expression in the neurons. MCP1 is transported into the spinal cord, followed by autocrine activation of microglia by MCP1. Therefore, peripheral inflammation-induced MCP1 expression may increase pain sensitivity by activation of microglia through a positive feedback loop.

3.4.5. Monocyte Chemoattractant Protein-3

MCP3 is a small cytokine that is closely related to MCP1. MCP3 has been found to play a role in the development of chronic pain. In one study, nerve injury remarkably induced MCP3 expression in the spinal cords of mice [18]. Moreover, nerve injury-induced microglial activation was decreased in the spinal cord of CCR2 knockout mice compared to in normal mice. Moreover, intrathecal injection of MCP3 induced pain hypersensitivity in a dose-dependent manner, while intrathecal injection of a CCR2 inhibitor or antibody against MCP3 reduced sensitivity to pain in mice. This study suggests that MCP3 is primarily expressed in astrocytes. Astrocyte-derived MCP3 plays a key role in the development of neuropathic pain. However, many studies have reported that MCP3 is also expressed in activated microglia [24,59]. Furthermore, CFA-induced pain hypersensitivity was attenuated by inhibition of microglial activation and MCP3 expression in the spinal cord. Microglial activation was inhibited by MCP3 knockdown. Therefore, inhibition of MCP3 expression may alleviate inflammation-induced pain hypersensitivity by regulating microglial activation in the spinal cord.

4. Intracellular Signaling in Activated Microglia

Activated microglia release inflammatory mediators that may contribute to hypersensitivity to inflammatory pain. Many studies have demonstrated the importance of intracellular signaling pathways that are strongly involved in microglial activation-mediated inflammatory pain. Activation of microglia leads to the induction of cascades of numerous intracellular signaling pathways. These signaling pathways may contribute to changes in the function of microglia, and gene expression resulting from these signaling pathways may influence the functions and structures of nearby cells and tissues, resulting in exacerbated inflammatory pain. Understanding how intracellular signaling pathways work in activated microglia may help identify new therapeutic targets for inflammatory pain (Figure 2).

4.1. Nuclear Factor-κB

Nuclear factor-κB (NF-κB) is a representative of the family of transcript factors associated with the inflammatory response. The activation of NF-κB signaling involves two major pathways: canonical and noncanonical. Although both pathways are important for regulating the inflammatory response, the noncanonical pathway is particularly involved in regulating specific functions of the adaptive immune system [60]. This chapter focuses on canonical NF-κB signaling, which is associated with microglial activation. Canonical NF-κB signaling is triggered by various stimuli [61]. The first step in the canonical NF-κB signaling pathway is the activation of the IκB kinase (IKK) complex, which comprises IKKα, IKKβ, and IKKγ subunits [62]. The IKK complex phosphorylates IκBα, leading to ubiquitylation and proteasomal degradation. This results in the phosphorylation and nuclear translocation of the NF-κB dimer (p65 and p50). The translocated NF-κB dimer binds to a specific DNA sequence and promotes the transcription of target genes. NF-κB signaling is strongly associated with the development of microglial activation, as evidenced by increased expression of inflammatory mediators and cytokines [63]. Previous studies have shown that microglial activation was significantly reduced by treatment with NF-κB inhibitors [64,65]. Moreover, LPS-induced expression of inflammatory cytokines and mediators was reduced by suppression of NF-κB activity through IKK-specific deletion in microglia [66]. Inhibition of NF-κB in the spinal cord showed an alleviative effect on CFA-induced pain hypersensitivity and microglial activation [67]. Furthermore, nerve injury-induced upregulation of pain sensitivity was alleviated by the suppression of NF-κB signaling in microglial activation [68]. These data demonstrate that activation of NF-κB signaling in microglia can lead to increased pain sensitivity in inflammatory pain.

4.2. Mitogen-Activated Protein Kinase

The mitogen-activated protein kinase (MAPK) signaling pathway, which comprises the c-Jun N-terminal kinase (JNK), extracellular signal-regulated kinase (ERK), and p38 mitogen-activated protein (p38) kinase, plays a crucial role in regulating various cellular functions such as proliferation, differentiation, development, and migration [69]. MAPK is involved in protein kinase cascades, in which they are activated in a sequential manner by the upstream signals such as MAPKK and MAPKKK.
In the nervous system, JNK is implicated in the pathogenesis of various neuroinflammation-related diseases [70,71]. JNK is activated by its upstream signals, MKK4 and MKK7, leading to the phosphorylation of the downstream signal, c-Jun. The activated JNK could induce the production of inflammatory mediators and cytokines in the CNS. Thus, the inhibition of JNK has been considered a therapeutic target for the treatment of neurodegenerative diseases. In addition, the inhibition of JNK in microglia has been suggested to attenuate inflammatory pain. Previously, LPS-induced microglial activation was remarkably suppressed by treatment with a JNK inhibitor, as revealed by the reduced expression of inflammatory mediators and cytokines [72]. Additionally, CFA-induced pain hypersensitivity was attenuated by intrathecal injection of a JNK inhibitor in rats [73].
Among the MAPK family members, ERK1/2 activation by MEK1/2 is generally considered to regulate cell survival, proliferation, and differentiation. The activation of ERK in microglia leads to neuroinflammation by increasing the expression of pro-inflammatory cytokines and inflammatory mediators [74]. These effects have become the cornerstone in the development of neurodegenerative diseases. A previous study identified that ERK activation in microglia was significantly increased in the early stages of nerve injury-induced chronic pain [75]. In in vitro experiments, LPS-induced expression of iNOS, COX-2, and pro-inflammatory cytokines was notably reduced by treatment with ERK inhibitor in microglia [76,77]. In addition, direct injection of ERK inhibitor into the spinal cord showed inhibitory effects on CFA-induced pain hypersensitivity in mice [78,79]; these effects were accompanied by a reduction in COX-2 expression in the spinal cord.
p38 is more strongly involved in the development of chronic pain related to microglial activation than other MAPK family members. p38 is activated by the upstream kinases MKK3 and MKK6 and plays an important role in the inflammatory response. p38 inhibitors have been found to alleviate inflammatory diseases [80]. During the development of chronic pain, p38 activation is notably increased in microglia compared to that in other cells [81]. In addition, the activation of p38 in microglia was upregulated in CFA-induced inflammatory pain. Moreover, intrathecal injection of p38 inhibitor attenuated CFA- or formalin-induced pain-like behaviors and significantly suppressed expression of IL-1β and IL-6 in the spinal cord [82,83,84]. In in vitro studies, LPS-induced NO overproduction and expression of iNOS and COX2 were reduced by treatment with a p38 inhibitor in a dose-dependent manner [37]. Taken together, these results demonstrate that MAPK signaling is critical for microglial activation and leads to the pathogenesis of inflammatory pain.

4.3. Janus Kinase 2 (JAK2)/Signal Transducer and Activator of Transcription 3

The Janus kinase 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3) pathway is an intracellular signaling pathway activated by cytokines. The JAK2/STAT3 pathway is involved in immune cell division, development, recruitment, and activation. Many studies have indicated that microglial activation is dependent on the phosphorylation of the JAK2/STA3 signaling pathway caused by various stimuli [85,86,87]. These data revealed that inhibition of JAK2/STAT3 signaling suppressed microglial activation, as shown by the decreased expression of inflammatory cytokines and mediators. Additionally, CFA-induced pain hypersensitivity and spinal microglial activation were significantly reduced by inhibition of the JAK2/STAT3 signaling pathway in rodents [24,88,89]. These results indicate that JAK2/STAT3 signaling plays an important role in microglial activation and development of inflammatory pain.

4.4. Nuclear Factor-Erythroid 2-Related Factor 2

Nuclear factor-erythroid 2-related factor 2 (Nrf2) is a transcription factor that regulates antioxidant enzymes to protect against damage caused by oxidative stress. Oxidative stress is a hallmark of neuroinflammation and neurodegeneration that leads to disease progression [90]. Accumulating evidence has shown that microglial activation was significantly increased in the CNS of Nrf2-deficient mice, contributing to the exacerbation of neurodegenerative diseases (Figure 3) [91,92]. Previous studies have shown that microglial activation was increased in Nrf2 knockout mice, as revealed by the upregulated expression of pro-inflammatory cytokines and inflammatory mediators [93,94]. Nrf2 induces gene expression via interaction with an antioxidant response element (ARE) that is known to encode antioxidant enzymes. Nrf2-dependent gene expression exerts a protective effect against oxidative stress in microglia, resulting in the suppression of neuroinflammation. Moreover, neuroinflammation-mediated chronic pain is regulated by the activation of Nrf2 in microglia. Among Nrf2-dependent genes, the heme oxygenase-1 (HO-1) gene is a representative gene with strong antioxidant effects. Previously, Nrf2-dependent HO-1 expression in microglia showed inhibitory effects against CFA-induced pain hypersensitivity and microglial activation in mice [75]. Additionally, the administration of an HO-1 inducer significantly attenuated formalin-induced pain-like behavior in mice. However, the attenuative effect of the HO-1 inducer was reversed in Nrf2-knockout mice [95]. These results indicated that Nrf2 inhibits inflammation-induced microglial activation and pain hypersensitivity via HO-1 induction.

4.5. Autophagy

Autophagy is a lysosomal degradation pathway responsible for the removal and recycling of unnecessary or dysfunctional molecules maintain cellular homeostasis [96]. The process begins by the marking of unwanted or damaged molecules for removal, followed by the formation of an autophagosome that envelopes the unwanted molecules. This autophagosome then combines with a lysosome to degrade the cargo, after which unwanted molecules are removed and recycled. Autophagy has a protective effect, as demonstrated by its ability to remove amyloid-β, a hallmark of Alzheimer’s disease, and prevent neurodegeneration in mice [97]. Additionally, microglial autophagy has been found to play a role in regulating neuroinflammation. LPS-treated microglia showed inhibited autophagic activity, leading to increased neuroinflammation. However, treatment with the autophagy inducer, rapamycin, significantly reduced LPS-induced neuroinflammation in microglia [98]. CFA-induced pain hypersensitivity was attenuated by the induction of autophagy in the spinal cord [24,99,100]. In addition, CFA-induced expression of pro-inflammatory cytokines and microglial activation were decreased by enhanced autophagy in the spinal cord. These results suggested that autophagy activation may have a protective effect against microglial activation and inflammatory pain.

5. Natural Product-Derived Compounds against Microglial Activation-Mediated Inflammatory Pain

Multiple studies have demonstrated that microglial activation contributes significantly to the development of inflammatory pain. Therefore, targeting microglial activation through the regulation of inflammatory mediators has been proposed as a therapeutic strategy for the treatment of inflammatory pain. Many natural products and their compounds have been found to exert protective effects against inflammation [101]. These studies suggest that natural product-derived compounds with anti-inflammatory effects inhibit inflammatory pain by suppressing microglial activation. Table 1 and Figure 4 present a summary of natural product-derived compounds that have been found to have the potential to alleviate microglial activation-mediated inflammatory pain.

5.1. 3,5-Dicaffeoylquinic Acid

3,5-Dicaffeoylquinic acid (3,5-DCQA) is a phenolic nutraceutical present in Arctium lappa and Aster yomena; it has shown inhibitory effects on the LPS-induced expression of iNOS and COX-2 and secretion of TNF-α, IL-1β, IL-6, MCP1, and MCP3 in BV2 microglial cells [24]. Suppression of MCP3 expression by 3,5-DCQA enhanced autophagy by suppressing LPS-induced activation of JAK2-STAT3, resulting in the reduction of microglial activation. Furthermore, CFA-induced pain hypersensitivity was attenuated by the administration of 3,5-DCQA. Additionally, the administration of 3,5-DCQA suppressed microglial activation in the spinal cord of CFA-injected mice.

5.2. Chlorogenic Acid

Chlorogenic acid, an ester of caffeic and quinic acids, is a natural phenolic compound found in plants. One study showed that chlorogenic acid significantly inhibited LPS-induced NO production and expression of iNOS and TNF-α in primary microglia [102]. In addition, LPS-induced phosphorylation of NF-κB signaling was suppressed by treatment with chlorogenic acid. Carrageenan-induced foot swelling and formalin-induced pain-like behavior were significantly reduced by oral administration of chlorogenic acid in mice [103]. In a clinical study, plasma antioxidant capacity was significantly increased in the group that consumed chlorogenic acid-rich coffee/day. Participants had consumed a maximum of 480 mg/day chlorogenic acid for 8 weeks and did not experience any adverse effects [104]. Furthermore, a clinical study identified that the consumption of chlorogenic acid resulted in improved neuronal function [105].

5.3. Ferulic Acid

Ferulic acid, a well-known phenolic compound, is a bioactive compound found in medicinal herbs, including Ferula asafoetida. One study showed that LPS-induced expression of iNOS and TNF-α was reduced after treatment with ferulic acid in BV2 microglial cells in a dose-dependent manner [106]. Additionally, LPS-induced phosphorylation of JNK and NF-κB was reduced by ferulic acid. Moreover, formalin-induced pain-like behavior in mice was alleviated by intraperitoneal injection of ferulic acid [107]. In a clinical study, participants were administered 1000 mg/day ferulic acid for 6 weeks. No toxicity associated with this dose of ferulic acid was observed. The oxidative stress marker was significantly reduced in the group supplemented with ferulic acid. Moreover, TNF-α was remarkably reduced in blood samples [108]. Another clinical study also showed clinical positive effective of ferulic acid on neuronal functioning [109].

5.4. 6-Gingerol

6-gingerol, present in Zingiber officinale, is a bioactive phenolic compound, which is known to have a neuroprotective effect. Previously, LPS-induced NO production and expression of iNOS, IL-1β, and IL-6 were dose-dependently suppressed by treatment with 6-gingerol, leading to inhibition of microglial activation [110]. LPS-induced phosphorylation of STAT3 in microglia was significantly reduced by 6-gingerol treatment. Furthermore, intraperitoneal injection of 6-gingerol was found to attenuate acetic acid- and formalin-induced pain-like behaviors, such as writhing and licking, in mice [111]. Carrageenan-induced paw swelling was also suppressed by the administration of 6-gingerol. No clinical trials have specifically investigated analgesic effects of 6-gingerol alone. However, numerous studies have reported alleviative effects of ginger on inflammation-related pain in humans [112]. Given that 6-gingerol had no adverse effects at a concentration of 10 mg twice a day for 12 weeks, it is necessary to evaluate the analgesic effects of 6-gingerol using this dosage [113].

5.5. Curcumin

Curcumin is a bright yellow bioactive component found in Curcuma longa. LPS-induced expression of iNOS, TNF-α, and IL-1β was decreased after treating BV2 microglial cells with curcumin [114]. Additionally, lipoteichoic acid (LTA) treatment increased the production of NO and PGE3, and expression of iNOS, COX-2, and TNF-α, reversed by treatment with curcumin in BV2 microglial cells. In one study, curcumin showed a suppressive effect on microglial activation via inhibition of NF-κB and MAPK signaling and induction of Nrf2 in BV2 microglial cells [115]. Moreover, in another study CFA-induced hyperalgesia was attenuated via suppression of TNF-α, IL-1β, and IL-6 in the spinal cord by administration of curcumin [116]. In a clinical study identifying anti-inflammatory effects of curcumin, the reduction in pain sensitivity and inflammation at the surgical site was evaluated in a group of patients who receive 400 mg of curcumin three times a day for 6 days [117].

5.6. Kaempferol

Kaempferol, one of the most common flavonoids found in numerous medicinal herbs, is known to have antioxidant and anti-inflammatory effects. In one study, LPS-induced microglial activation was suppressed by treatment with kaempferol, as revealed by decreased production of NO and PGE2 and decreased expression of iNOS, COX-2, MMP9, TNF-α, and IL-1β in microglia [118]. The underlying inhibitory mechanism of kaempferol is the inhibition of NF-κB and MAPK signaling pathways in microglia. Moreover, formalin-induced pain hypersensitivity was alleviated by intrathecal injection of kaempferol in mice [119]. Administration of kaempferol showed an inhibitory effect on the formalin-induced expression of TNF-α, IL-1β, and IL-6 in the spinal cord. A variety of clinical studies had provided evidence for the preventive effects of kaempferol on diseases associated with inflammation [120,121]. The consumption of 50 mg a day of kaempferol for 4 weeks is safe in adults [122].

5.7. Quercetin

Quercetin is considered an antioxidant, anti-inflammatory, and anti-nociceptive compound. Studies have shown that quercetin inhibits LPS-induced NO production and iNOS expression in BV2 microglial cells. Furthermore, LPS-induced NF-κB activation was reduced by quercetin treatment [123]. Additionally, in one study, quercetin led to the activation of Nrf/HO-1 signaling, resulting in the inhibition of NO production in microglia. Moreover, CFA-induced chronic inflammatory hyperalgesia was attenuated by the inhibition of ERK1/2 and NF-κB in the spinal cord after the administration of quercetin [124]. CFA-induced TNF-α expression was also decreased in the spinal cord by quercetin administration. A previous study investigated the effect of quercetin supplementation on inflammation and pain in women diagnosed with rheumatoid arthritis [125]. The patients were given 500 mg of quercetin a day for 8 weeks. The results revealed significant reductions in plasma levels of TNF-α and improvements in symptoms related to swelling and pain in patients following quercetin supplementation. Notably, no side effects were observed in the patients.

5.8. Formononetin

Formononetin is a bioactive isoflavone found in various plants including Trifolium pratense L. In one study, LPS-induced microglial activation was reduced by treatment of formononetin, as revealed by a decrease in the expression of TNF-α, IL-1β, and IL-6 [126]. Additionally, LPS-induced expression of iNOS and COX-2 was suppressed in BV2 microglial cells. Moreover, formononetin showed an inhibitory effect on LPS-induced activation of NF-κB signaling. In the CFA-induced inflammatory pain model, the administration of formononetin alleviated mechanical allodynia and thermal hyperalgesia in mice [127]. Formononetin has been studied in preclinical tests for other diseases, but clinical studies for the use of formononetin alone have yet to be performed. A previous study showed that extracts containing rich-formononetin (50 mg/day for at least 1 year) exhibited beneficial effects on the bone, with no significantly adverse effects [128]. However, further clinical research is required to determine the safety and effects of formononetin specifically on inflammatory pain.

5.9. Naringenin

Naringenin is a flavonoid with antioxidant, anti-inflammatory, and anti-cancer properties. One study revealed that naringenin blocked transformation into LPS-induced activation, as evidenced by expression of iNOS, TNF-α, and IL-1β in BV2 microglia cells [129]. In addition, the LPS-induced phosphorylation of MAPK members, including JNK, ERK, and p38, was notably inhibited by naringenin treatment. In mice with inflammatory pain, carrageenan-, capsaicin-, CFA-, and PGE2-induced mechanical hyperalgesia was significantly alleviated by the oral administration of naringenin without gastric or hepatic toxicity [130]. A study conducted on healthy adults to evaluate the safety and pharmacokinetics of naringenin reported that the half-life was 3 h and almost disappeared from the serum after 24 h of ingestion [131]. No adverse events were reported up to 900 mg of naringenin. Clinical trials using orange juice, which is known to contain naringenin, showed anti-inflammatory effects as evidenced by increasing pro-inflammatory cytokines [132]. These findings suggest that naringenin has the potential to attenuate pain through the regulation of inflammation.

5.10. Resveratrol

Resveratrol is a bioactive component produced in grapes and is a representative inducer of autophagy. One study reported that LPS/interferon γ (IFNγ)-induced expression of iNOS, TNF-α, and IL-1β was suppressed by resveratrol treatment in N9 microglial cells [133]. In the case of LPS/IFNγ-induced microglial activation, activation of NF-κB was inhibited by resveratrol treatment. Moreover, in one study, CFA-induced temporomandibular disorders, resveratrol dose-dependently attenuated pain-like behavior in mice [134]. TNF-α in activated microglia of spinal trigeminal nucleus caudalis is also inhibited by resveratrol treatment. In a clinical study, resveratrol was evaluated for its effects on inflammation and pain in patients with knee osteoarthritis [135]. A total of 110 patients were treated with 500 mg/day resveratrol for 90 days. In the group that received oral administration of resveratrol, pain sensitivity and pro-inflammatory cytokines in serum were significantly decreased compared to the control group.

5.11. Honokiol

Honokiol, a natural polyphenolic compound, is extracted from the bark and seeds of Magnolia officinalis. Honokiol is an autophagy inducer that suppresses skin cancer [136]. LPS-induced NO production and expression of iNOS, IL-1β, and IL-6 in primary microglia were suppressed after treatment with honokiol [137]. In addition, in one study, carrageenan- and CFA-induced mechanical hyperalgesia, allodynia, and thermal hyperalgesia were alleviated by intraperitoneal injection of honokiol in mice [138]. In a clinical study aimed at evaluating safety, 50 mg per kg of honokiol was intravenously injected into cancer patients [139]. There were no serious adverse effects and a positive clinical response was achieved in patients. Therefore, an evaluation is needed of the anti-inflammatory and analgesic effects of honokiol at the same concentration in humans.

5.12. Ligustilide

Ligustilide, a major compound found in the roots of Angelica sinensis, has a protective effect against inflammation in microglia. Studies have shown that LPS-induced NO production and expression of iNOS and COX2 were remarkably reduced in microglia after ligustilide treatment. LPS-induced production of TNF-α, IL-1β, IL-6, and MCP1 was also suppressed by ligustilide treatment in previous studies [140,141]. Furthermore, in one study CFA-induced pain hypersensitivity and microglial activation in the spinal cord were significantly reduced after ligustilide treatment; ligustilide treatment alleviated acetic acid- and formalin-induced pain in mice [142]. Following a safety evaluation of ligustilide in rats, oral administration of 90 mg/kg ligustilide had good health status, without any histopathological change [143]. Moreover, tissue analysis indicated that ligustilide could penetrate the blood–brain barrier. Based on these results, it is suggested that ligustilide needs to be evaluated for safety and effects on inflammatory pain in clinical settings.

5.13. Glycyrrhizin

Glycyrrhizin, a triterpene saponin present in Glycyrrhiza glabra, has shown inhibitory effects on inflammatory pain by suppressing microglial activation [144]. In one study, LPS-induced microglial activation was significantly reduced by treatment with glycyrrhizin, as evidenced by decreased NO production and expression of pro-inflammatory cytokines. Glycyrrhizin inhibited LPS-induced HMGB1/TLR4/NF-κB signaling in microglia, leading to reducing microglial activation. Additionally, CFA-induced pain hypersensitivity was attenuated by the administration of glycyrrhizin in mice. Glycyrrhizin suppresses CFA-induced expression of pro-inflammatory cytokines and activation of NF-κB in the spinal cord. In a clinical study, glycyrrhizin was administered to patients with the selective serotonin reuptake inhibitor (SSRI) in order to evaluate the effects on depression and inflammation [145]. The results showed serum levels of TNF-α, and IL-1β were significantly reduced and depressive symptoms were improved. No patients experienced severe adverse events with 150 mg/3 times a day glycyrrhizin in combination with a 10 mg/day SSRI for 4 weeks.

5.14. Docosahexaenoic Acid

Docosahexaenoic acid (DHA) is the major bioactive omega-3 polyunsaturated fatty acid. DHA is known to regulate the inflammatory responses in neurodegenerative diseases. In one study, carrageenan-induced inflammatory pain and microglial activation were inhibited by the administration of DHA [146]. Carrageenan-induced mechanical allodynia was inhibited by intrathecal injection of DHA in mice. After DHA treatment, carrageenan-induced microglial activation was suppressed by p38 inhibition in spinal microglia. Further, LPS-induced expression of TNF-α, IL-1β, IL-6, MCP1, CCL3, and CXCL10 was significantly suppressed by DHA treatment of BV2 microglial cells. The consumption of omega-3 fatty acid showed improvements in pain and function in patients with osteoarthritis [147]. The patients who consumed 0.45 omega-3 fatty acids/day for 24 months experienced beneficial effects on osteoarthritis without any adverse events. Thus, it is necessary to evaluate the analgesic effects and safety specifically using DHA alone in clinical settings.

5.15. Paeoniflorin

Paeoniflorin, the main active ingredient of Paeonia lactiflora, is known to reduce CFA-induced pain hypersensitivity and mRNA expression of TNF-α, IL-1β, and IL-6 in the spinal cord [148]. Furthermore, in one study, CFA-induced microglial activation in the dorsal horn was inhibited by paeoniflorin. In in vitro experiments, LPS-induced pro-inflammatory cytokines were reduced by treatment with paeoniflorin via inhibiting AKT- NF-κB in microglia. In a clinical study, intravenous injection of powders containing 35.8 mg/day paeoniflorin for 7 days showed no adverse events in healthy adults [149].

5.16. Sinomenine

Sinomenine is found in Sinomenium acutum and is known to have various pharmacological effects such as anti-cancer, anti-inflammation, and antioxidant effects. One study found that treatment with sinomenine suppressed amyloid-β-induced microglial activation, as evidenced by the reduction in NO production and expression of TNF-α, IL-1β, and MCP1 in BV2 cells [150]. Moreover, CFA-induced pain hypersensitivity was inhibited by intraperitoneal injection of sinomenine in mice [151]. In addition, CFA-induced expression of TNF-α, IL-1β, IL-6, and COX-2 and PGE2 production was inhibited by the activation of p38 and NF-κB in the spinal cord. In a clinical study, patients with osteoarthritis were orally administered 20 mg/2 times a day sinomenine for 3 months [152]. The results indicated that disease symptoms were attenuated and plasma levels of pro-inflammatory cytokines were significantly reduced by sinomenine in patients. No adverse events were observed during the study. Based on these findings, further evaluation is necessary to assess the attenuative effects of sinomenine on inflammatory pain.

5.17. Muscone

Muscone is found in musk, which is a glandular secretion of musk deer; it is a pharmacologically bioactive compound that has been used in medicine for centuries. One study revealed that muscone had an inhibitory effect on LPS-induced NO production and expression of iNOS, IL-1β, and IL-6 in BV2 microglial cells [89]. In addition, LPS-induced activation of JAK2/STAT3 signaling was significantly suppressed by muscone treatment of BV2 cells. In an inflammatory pain model, CFA-induced pain hypersensitivity was attenuated by the intraperitoneal injection of muscone in mice. Moreover, muscone administration suppressed CFA-induced expression of pro-inflammatory cytokines and phosphorylation of JAK2/STAT3 signaling in the spinal cord of mice. Muscone exhibited liver toxicity in Kunming mice at doses exceeding 50 mg/kg [153]. As a result, additional studies in pro-clinical and clinical trials are required to further investigate the safety and effects of muscone on inflammatory pain.

5.18. Urolithins

Urolithins are secondary metabolites formed by gut microbiome from ellagic acid and ellagitannins found in foods like pomegranate. Urolithins suppressed LPS-induced production of NO and mRNA expression of TNF-α, IL-1β, IL-6, iNOS, and COX-2 in BV2 microglial cells. Additionally, LPS-induced activation of ERK, p38, and NF-κB signaling were significantly reduced by urolithins in BV2 cells [154]. Another group induced experimental osteoarthritis to study the effects of urolithins on inflammatory pain. The meniscotibial and medial collateral ligaments were transected in the knees of mice to induce osteoarthritis as inflammatory pain model. Mice were given a diet containing urolithins to identify the effects on pain. Results showed urolithins reduced pain hypersensitivity and slowed down disease progression in mice [155]. In clinical trials of older adults, supplementation with 1000 mg/day urolithin for 4 months showed no adverse events [156]. Additionally, plasma levels of inflammatory biomarkers were significantly reduced in a group that consumed urolithin. However, further experiments are needed to evaluate the effects of urolithins on pain relief.
Table 1. Natural product-derived compounds attenuating microglial activation-mediated inflammatory pain.
Table 1. Natural product-derived compounds attenuating microglial activation-mediated inflammatory pain.
Class of
Phytochemicals
SubclassMajor
Compound
SourceTargeting
Inflammatory
Mediators
Targeting
Intracellular
Signaling
Inducer in
Animal
Model
Safety Dosage in Clinical StudyEffects in
Clinical Study
Reference
PhenolicsPhenolic acid3,5-Dicaffeoylquinic acidArctium
lappa, aster yomena
TNF-α, IL-1β, IL-6,
MCP1, MCP3, iNOS, COX2
JAK2/STAT3,
Autophagy
CFA [24]
PhenolicsPhenolic acidChlorogenic acid NO, iNOS, TNF-αNF-κBCarrageenan, Formalin480 mg/day for 8 weeksImprovements in neuronal function[102,103,104,105]
PhenolicsPhenolic acidFerulic acidferula
asafetida
TNF-α, iNOSNF-κB, JNKFormalin1000 mg/day for 6 weeksAnti-oxidant, anti-inflammation[106,107,108,109].
PhenolicsPhenolic acid6-gingerolzingiber
officinale
NO, iNOS, IL-1β, IL-6STAT3Acetic acid,
Formalin,
Carrageenan
20 mg/day for 12 weeks [110,111,113]
PhenolicsflavonoidsCurcuminCurcuma
longa
NO, PGE2, iNOS, COX2, TNF-α, IL-1β, IL-6NF-κB, MAPK, Nrf2CFA1200 mg/day for 6 daysAnalgesic effects, anti-inflammation[114,115,116,117]
PhenolicsflavonoidsKaempferolTea, broccoliNO, PGE2, iNOS,
COX2, MMP-9,
TNF-α, IL-1β, IL-6
NF-κB, JNK,
ERK, p38
Formalin50 mg/day for 4 weeksAnti-inflammation[118,119,120,121,122]
PhenolicsflavonoidsQuercetin NO, iNOS, TNF-αNF-κB, Nrf2, ERKCFA500 mg/day for 8 weeksAnalgesic effects, anti-inflammation[123,124,125]
PhenolicsflavonoidsFormononetinTrifolium
pretense L.
TNF-α, IL-1β, IL-6,
iNOS, COX2
NF-κBCFA [126,127]
PhenolicsflavonoidsNaringenin TNF-α, IL-1β, iNOSMAPKCarrageenan, Capsaicin,
CFA, PGE2
900 mg for a day [129,130,131]
PhenolicsstilbenesResveratrolgrapeTNF-α, IL-1β, iNOSNF-κB,
Autophagy
CFA500 mg/day for 90 daysAnalgesic effects, anti-inflammation[133,134,135]
PhenolicslignanHonokiolMagnolia
officinlis
NO, iNOS, TNF-α,
IL-1β, IL-6
AutophagyCarrageenan, CFA50 mg/kg for a week [136,137,138,139]
Non-phenolicsphthalideLigustilidethe roof of Angelica
sinensis
NO, iNOS, COX-2,
TNF-α, IL-1β, IL-6,
MCP1
NF-κBCFA,
Acetic acid,
Formalin
[140,141,142]
Non-phenolicssaponinGlycyrrhizinGlycyrrhiza
glabra
NO, TNF-α,
IL-1β, IL-6
NF-κBCFA450 mg/day for 4 weeksAnti-inflammation[144,145]
Non-phenolicsomega-3 fatty acidDocosahexaenoic acidOmega-3 polyunsaturated fatty acidTNF-α, IL-1β, IL-6,
MCP1, CCL3,
CXCL10
p38Carrageenan [146]
Non-phenolicsmonoterpenePaeoniflorinPaeonia
lactiflora
TNF-α, IL-1β, IL-6NF-κBCFA35.8 mg/day for 7 days [148,149]
Non-phenolicsalkaloidSinomenineSinomenium
acutum
NO, TNF-α, IL-1β,
IL-6, MCP1
NF-κB, p38CFA40 mg/day for 3 monthsAnti-inflammation[150,151,152]
MusconeMuskNO, TNF-α,
IL-1β, IL-6
JAK2/STAT3CFA [89]
UrolithinsSecondary metaboliteTNF-α, IL-1β, IL-6, iNOS, and COX-2ERK, p38, and NF-κBSurgery1000 mg/day for 4 monthsAnti-inflammation[154,155,156]
MusconeMuskNO, TNF-α,
IL-1β, IL-6
JAK2/STAT3CFA [89]
UrolithinsSecondary metaboliteTNF-α, IL-1β, IL-6, iNOS, and COX-2ERK, p38, and NF-κBSurgery1000 mg/day for 4 monthsAnti-inflammation[154,155,156]

6. Methods

Reference lists were searched for articles published until 19–20 June 2023, using the keywords “Microglial activation”, “Neuroinflammation”, “Inflammatory biomarkers”, “Inflammatory pain”, “Chronic pain”, “Intracellular signaling”, “Natural products”, “Pharmaceuticals”.
In our search for this review, we applied no limits for country of origin or study design. Articles published in a language other than English were excluded.

7. Conclusions

Inflammation in peripheral tissues can lead to the activation of microglia in the dorsal horn of the spinal cord, which is a significant contributor to neuroinflammation and inflammatory pain. Several studies have shown that natural products and their compounds have the ability to regulate microglial activation. Additionally, many studies have proposed that microglial activation-mediated inflammatory pain can be modulated using natural product-derived compounds. However, it is important to consider the possibility of drug–drug interactions (DDIs) when multiple drugs are consumed in combination, as these interactions can potentially affect the pharmacological effects of each drug [157]. In the case of muscone, a previous study reported that it reduced the hypnotic and analgesic effects of ketamine, which is a widely used anesthetic [158]. Taken together, although toxicological and pharmacological studies are required to determine their safety in humans, natural product-derived compounds are potential therapeutic candidates for the treatment of inflammatory pain.

Author Contributions

J.P.: Writing—Original Draft, Conceptualization, C.L.: Funding acquisition, Y.T.K.: Writing—Review and Editing, Supervision. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Main Research Program of the Korea Food Research Institute (KFRI), funded by the Korean Ministry of Science and ICT, grant number E0210201-03.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Zelaya, C.E.; Dahlhamer, J.M.; Lucas, J.W.; Connor, E.M. Chronic Pain and High-Impact Chronic Pain among US Adults, 2019. 2020. Available online: https://stacks.cdc.gov/view/cdc/97308 (accessed on 7 June 2023).
  2. Lee, J.; Jotwani, R.; Robert, S.W. The economic cost of racial disparities in chronic pain. J. Comp. Eff. Res. 2020, 9, 903–906. [Google Scholar] [CrossRef] [PubMed]
  3. McWilliams, L.A.; Cox, B.J.; Enns, M.W. Mood and anxiety disorders associated with chronic pain: An examination in a nationally representative sample. Pain 2003, 106, 127–133. [Google Scholar] [CrossRef]
  4. Reddi, D.; Curran, N.; Stephens, R. An introduction to pain pathways and mechanisms. Br. J. Hosp. Med. 2013, 74 (Suppl. S12), C188–C191. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Treede, R.D.; Rief, W.; Barke, A.; Aziz, Q.; Bennett, M.I.; Benoliel, R.; Cohen, M.; Evers, S.; Finnerup, N.B.; First, M.B.; et al. Chronic pain as a symptom or a disease: The IASP Classification of Chronic Pain for the International Classification of Diseases (ICD-11). Pain 2019, 160, 19–27. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Ghlichloo, I.; Gerriets, V. Nonsteroidal Anti-Inflammatory Drugs (NSAIDs); StatPearls: Treasure Island, FL, USA, 2022. [Google Scholar]
  7. Harvey, A.L.; Edrada-Ebel, R.; Quinn, R.J. The re-emergence of natural products for drug discovery in the genomics era. Nat. Rev. Drug Discov. 2015, 14, 111–129. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Ji, R.R.; Xu, Z.Z.; Gao, Y.J. Emerging targets in neuroinflammation-driven chronic pain. Nat. Rev. Drug Discov. 2014, 13, 533–548. [Google Scholar] [CrossRef] [Green Version]
  9. Matsuda, M.; Huh, Y.; Ji, R.R. Roles of inflammation, neurogenic inflammation, and neuroinflammation in pain. J. Anesth. 2019, 33, 131–139. [Google Scholar] [CrossRef]
  10. Zhang, L.; Berta, T.; Xu, Z.Z.; Liu, T.; Park, J.Y.; Ji, R.R. TNF-α contributes to spinal cord synaptic plasticity and inflammatory pain: Distinct role of TNF receptor subtypes 1 and 2. Pain 2011, 152, 419–427. [Google Scholar] [CrossRef] [Green Version]
  11. Xie, R.G.; Gao, Y.J.; Park, C.K.; Lu, N.; Luo, C.; Wang, W.T.; Wu, S.X.; Ji, R.R. Spinal CCL2 Promotes Central Sensitization, Long-Term Potentiation, and Inflammatory Pain via CCR2: Further Insights into Molecular, Synaptic, and Cellular Mechanisms. Neurosci. Bull. 2018, 34, 13–21. [Google Scholar] [CrossRef]
  12. Kawasaki, Y.; Zhang, L.; Cheng, J.K.; Ji, R.R. Cytokine mechanisms of central sensitization: Distinct and overlapping role of interleukin-1β, interleukin-6, and tumor necrosis factor-α in regulating synaptic and neuronal activity in the superficial spinal cord. J. Neurosci. 2008, 28, 5189–5194. [Google Scholar] [CrossRef] [Green Version]
  13. Chen, G.; Zhang, Y.Q.; Qadri, Y.J.; Serhan, C.N.; Ji, R.R. Microglia in Pain: Detrimental and Protective Roles in Pathogenesis and Resolution of Pain. Neuron 2018, 100, 1292–1311. [Google Scholar] [CrossRef] [Green Version]
  14. Vergne-Salle, P.; Bertin, P. Chronic pain and neuroinflammation. Jt. Bone Spine 2021, 88, 105222. [Google Scholar] [CrossRef]
  15. Lim, E.Y.; Kim, Y.T. Food-Derived Natural Compounds for Pain Relief in Neuropathic Pain. BioMed Res. Int. 2016, 2016, 7917528. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Ji, R.R.; Nackley, A.; Huh, Y.; Terrando, N.; Maixner, W. Neuroinflammation and Central Sensitization in Chronic and Widespread Pain. Anesthesiology 2018, 129, 343–366. [Google Scholar] [CrossRef] [PubMed]
  17. Latremoliere, A.; Mauborgne, A.; Masson, J.; Bourgoin, S.; Kayser, V.; Hamon, M.; Pohl, M. Differential implication of proinflammatory cytokine interleukin-6 in the development of cephalic versus extracephalic neuropathic pain in rats. J. Neurosci. 2008, 28, 8489–8501. [Google Scholar] [CrossRef] [Green Version]
  18. Imai, S.; Ikegami, D.; Yamashita, A.; Shimizu, T.; Narita, M.; Niikura, K.; Furuya, M.; Kobayashi, Y.; Miyashita, K.; Okutsu, D.; et al. Epigenetic transcriptional activation of monocyte chemotactic protein 3 contributes to long-lasting neuropathic pain. Brain 2013, 136, 828–843. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Gu, N.; Yi, M.H.; Murugan, M.; Xie, M.; Parusel, S.; Peng, J.; Eyo, U.B.; Hunt, C.L.; Dong, H.; Wu, L.J. Spinal microglia contribute to sustained inflammatory pain via amplifying neuronal activity. Mol. Brain 2022, 15, 86. [Google Scholar] [CrossRef]
  20. Yi, M.H.; Liu, Y.U.; Liu, K.; Chen, T.; Bosco, D.B.; Zheng, J.; Xie, M.; Zhou, L.; Qu, W.; Wu, L.J. Chemogenetic manipulation of microglia inhibits neuroinflammation and neuropathic pain in mice. Brain Behav. Immun. 2021, 92, 78–89. [Google Scholar] [CrossRef]
  21. Zamora, R.; Vodovotz, Y.; Billiar, T.R. Inducible nitric oxide synthase and inflammatory diseases. Mol. Med. 2000, 6, 347–373. [Google Scholar] [CrossRef] [Green Version]
  22. Rocha, P.A.; Ferreira, A.F.B.; Da Silva, J.T.; Alves, A.S.; Martins, D.O.; Britto, L.R.G.; Chacur, M. Effects of selective inhibition of nNOS and iNOS on neuropathic pain in rats. Mol. Cell Neurosci. 2020, 105, 103497. [Google Scholar] [CrossRef]
  23. Lull, M.E.; Block, M.L. Microglial activation and chronic neurodegeneration. Neurotherapeutics 2010, 7, 354–365. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Park, J.; Kim, Y.; Lee, C.; Kim, Y.T. 3,5-Dicaffeoylquinic acid attenuates microglial activation-mediated inflammatory pain by enhancing autophagy through the suppression of MCP3/JAK2/STAT3 signaling. Biomed. Pharmacother. 2022, 153, 113549. [Google Scholar] [CrossRef] [PubMed]
  25. Zhang, L.Q.; Gao, S.J.; Sun, J.; Li, D.Y.; Wu, J.Y.; Song, F.H.; Liu, D.Q.; Zhou, Y.Q.; Mei, W. DKK3 ameliorates neuropathic pain via inhibiting ASK-1/JNK/p-38-mediated microglia polarization and neuroinflammation. J. Neuroinflamm. 2022, 19, 129. [Google Scholar] [CrossRef] [PubMed]
  26. Kuboyama, K.; Tsuda, M.; Tsutsui, M.; Toyohara, Y.; Tozaki-Saitoh, H.; Shimokawa, H.; Yanagihara, N.; Inoue, K. Reduced spinal microglial activation and neuropathic pain after nerve injury in mice lacking all three nitric oxide synthases. Mol. Pain 2011, 7, 50. [Google Scholar] [CrossRef] [Green Version]
  27. Chen, Y.; Zhou, Y.; Li, X.C.; Ma, X.; Mi, W.L.; Chu, Y.X.; Wang, Y.Q.; Mao-Ying, Q.L. Neuronal GRK2 regulates microglial activation and contributes to electroacupuncture analgesia on inflammatory pain in mice. Biol. Res. 2022, 55, 5. [Google Scholar] [CrossRef]
  28. Osborne, M.G.; Coderre, T.J. Effects of intrathecal administration of nitric oxide synthase inhibitors on carrageenan-induced thermal hyperalgesia. Br. J. Pharmacol. 1999, 126, 1840–1846. [Google Scholar] [CrossRef] [Green Version]
  29. Jiang, M.; Deng, H.; Chen, X.; Lin, Y.; Xie, X.; Bo, Z. The efficacy and safety of selective COX-2 inhibitors for postoperative pain management in patients after total knee/hip arthroplasty: A meta-analysis. J. Orthop. Surg. Res. 2020, 15, 39. [Google Scholar] [CrossRef] [Green Version]
  30. Hoozemans, J.J.; Rozemuller, A.J.; Janssen, I.; De Groot, C.J.; Veerhuis, R.; Eikelenboom, P. Cyclooxygenase expression in microglia and neurons in Alzheimer’s disease and control brain. Acta Neuropathol. 2001, 101, 2–8. [Google Scholar] [CrossRef]
  31. Sinatra, R. Role of COX-2 inhibitors in the evolution of acute pain management. J. Pain Symptom Manag. 2002, 24, S18–S27. [Google Scholar] [CrossRef]
  32. Cho, N.; Moon, E.H.; Kim, H.W.; Hong, J.; Beutler, J.A.; Sung, S.H. Inhibition of Nitric Oxide Production in BV2 Microglial Cells by Triterpenes from Tetrapanax papyriferus. Molecules 2016, 21, 459. [Google Scholar] [CrossRef] [Green Version]
  33. Sun, Z.; Li, G.; Tong, T.; Chen, J. Micheliolide suppresses LPS-induced neuroinflammatory responses. PLoS ONE 2017, 12, e0186592. [Google Scholar] [CrossRef] [Green Version]
  34. Kim, S.Y.; Jin, C.Y.; Kim, C.H.; Yoo, Y.H.; Choi, S.H.; Kim, G.Y.; Yoon, H.M.; Park, H.T.; Choi, Y.H. Isorhamnetin alleviates lipopolysaccharide-induced inflammatory responses in BV2 microglia by inactivating NF-κB, blocking the TLR4 pathway and reducing ROS generation. Int. J. Mol. Med. 2019, 43, 682–692. [Google Scholar] [CrossRef] [Green Version]
  35. Park, J.; Lim, E.Y.; Kim, Y.T. The inhibitory effects of Aster yomena extract on microglial activation-mediated inflammatory response and pain by modulation of the NF-κB and MAPK signaling pathways. J. Funct. Foods 2021, 85, 104659. [Google Scholar] [CrossRef]
  36. Dhapola, R.; Hota, S.S.; Sarma, P.; Bhattacharyya, A.; Medhi, B.; Reddy, D.H. Recent advances in molecular pathways and therapeutic implications targeting neuroinflammation for Alzheimer’s disease. Inflammopharmacology 2021, 29, 1669–1681. [Google Scholar] [CrossRef]
  37. Park, J.; Kim, Y.T. Erythronium japonicum Alleviates Inflammatory Pain by Inhibiting MAPK Activation and by Suppressing NF-κB Activation via ERK/Nrf2/HO-1 Signaling Pathway. Antioxidants 2020, 9, 626. [Google Scholar] [CrossRef] [PubMed]
  38. Konnecke, H.; Bechmann, I. The role of microglia and matrix metalloproteinases involvement in neuroinflammation and gliomas. Clin. Dev. Immunol. 2013, 2013, 914104. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Ji, R.R.; Xu, Z.Z.; Wang, X.; Lo, E.H. Matrix metalloprotease regulation of neuropathic pain. Trends Pharmacol. Sci. 2009, 30, 336–340. [Google Scholar] [CrossRef] [Green Version]
  40. Kawasaki, Y.; Xu, Z.Z.; Wang, X.; Park, J.Y.; Zhuang, Z.Y.; Tan, P.H.; Gao, Y.J.; Roy, K.; Corfas, G.; Lo, E.H.; et al. Distinct roles of matrix metalloproteases in the early- and late-phase development of neuropathic pain. Nat. Med. 2008, 14, 331–336. [Google Scholar] [CrossRef] [Green Version]
  41. Lee, E.J.; Kim, H.S. Inhibitory mechanism of MMP-9 gene expression by ethyl pyruvate in lipopolysaccharide-stimulated BV2 microglial cells. Neurosci. Lett. 2011, 493, 38–43. [Google Scholar] [CrossRef]
  42. Kular, L.; Rivat, C.; Lelongt, B.; Calmel, C.; Laurent, M.; Pohl, M.; Kitabgi, P.; Melik-Parsadaniantz, S.; Martinerie, C. NOV/CCN3 attenuates inflammatory pain through regulation of matrix metalloproteinases-2 and -9. J. Neuroinflamm. 2012, 9, 36. [Google Scholar] [CrossRef]
  43. Leung, L.; Cahill, C.M. TNF-α and neuropathic pain—A review. J. Neuroinflamm. 2010, 7, 27. [Google Scholar] [CrossRef] [Green Version]
  44. Duan, Y.W.; Chen, S.X.; Li, Q.Y.; Zang, Y. Neuroimmune Mechanisms Underlying Neuropathic Pain: The Potential Role of TNF-α-Necroptosis Pathway. Int. J. Mol. Sci. 2022, 23, 7191. [Google Scholar] [CrossRef] [PubMed]
  45. Kuno, R.; Wang, J.; Kawanokuchi, J.; Takeuchi, H.; Mizuno, T.; Suzumura, A. Autocrine activation of microglia by tumor necrosis factor-α. J. Neuroimmunol. 2005, 162, 89–96. [Google Scholar] [CrossRef] [PubMed]
  46. Hankittichai, P.; Lou, H.J.; Wikan, N.; Smith, D.R.; Potikanond, S.; Nimlamool, W. Oxyresveratrol Inhibits IL-1β-Induced Inflammation via Suppressing AKT and ERK1/2 Activation in Human Microglia, HMC3. Int. J. Mol. Sci. 2020, 21, 6054. [Google Scholar] [CrossRef]
  47. Davis, R.L.; Buck, D.J.; McCracken, K.; Cox, G.W.; Das, S. Interleukin-1β-induced inflammatory signaling in C20 human microglial cells. Neuroimmunol. Neuroinflamm. 2018, 5, 50. [Google Scholar] [CrossRef]
  48. Samad, T.A.; Moore, K.A.; Sapirstein, A.; Billet, S.; Allchorne, A.; Poole, S.; Bonventre, J.V.; Woolf, C.J. Interleukin-1β-mediated induction of Cox-2 in the CNS contributes to inflammatory pain hypersensitivity. Nature 2001, 410, 471–475. [Google Scholar] [CrossRef] [PubMed]
  49. Gui, W.S.; Wei, X.; Mai, C.L.; Murugan, M.; Wu, L.J.; Xin, W.J.; Zhou, L.J.; Liu, X.G. Interleukin-1β overproduction is a common cause for neuropathic pain, memory deficit, and depression following peripheral nerve injury in rodents. Mol. Pain 2016, 12, 1744806916646784. [Google Scholar] [CrossRef] [Green Version]
  50. Jang, S.; Kelley, K.W.; Johnson, R.W. Luteolin reduces IL-6 production in microglia by inhibiting JNK phosphorylation and activation of AP-1. Proc. Natl. Acad. Sci. USA 2008, 105, 7534–7539. [Google Scholar] [CrossRef] [Green Version]
  51. Lin, T.; Li, K.; Zhang, F.Y.; Zhang, Z.K.; Light, A.R.; Fu, K.Y. Dissociation of spinal microglia morphological activation and peripheral inflammation in inflammatory pain models. J. Neuroimmunol. 2007, 192, 40–48. [Google Scholar] [CrossRef] [Green Version]
  52. Yang, Y.; Zhou, W.; Xu, X.; Ge, X.; Wang, F.; Zhang, G.Q.; Miao, L.; Deng, X. Aprepitant Inhibits JNK and p38/MAPK to Attenuate Inflammation and Suppresses Inflammatory Pain. Front. Pharmacol. 2021, 12, 811584. [Google Scholar] [CrossRef]
  53. Zanjani, T.M.; Sabetkasaei, M.; Mosaffa, N.; Manaheji, H.; Labibi, F.; Farokhi, B. Suppression of interleukin-6 by minocycline in a rat model of neuropathic pain. Eur. J. Pharmacol. 2006, 538, 66–72. [Google Scholar] [CrossRef]
  54. Bose, S.; Cho, J. Role of chemokine CCL2 and its receptor CCR2 in neurodegenerative diseases. Arch. Pharm. Res. 2013, 36, 1039–1050. [Google Scholar] [CrossRef]
  55. Thacker, M.A.; Clark, A.K.; Bishop, T.; Grist, J.; Yip, P.K.; Moon, L.D.; Thompson, S.W.; Marchand, F.; McMahon, S.B. CCL2 is a key mediator of microglia activation in neuropathic pain states. Eur. J. Pain 2009, 13, 263–272. [Google Scholar] [CrossRef] [PubMed]
  56. Dansereau, M.A.; Midavaine, E.; Begin-Lavallee, V.; Belkouch, M.; Beaudet, N.; Longpre, J.M.; Melik-Parsadaniantz, S.; Sarret, P. Mechanistic insights into the role of the chemokine CCL2/CCR2 axis in dorsal root ganglia to peripheral inflammation and pain hypersensitivity. J. Neuroinflamm. 2021, 18, 79. [Google Scholar] [CrossRef]
  57. Zhang, L.; Tan, J.; Jiang, X.; Qian, W.; Yang, T.; Sun, X.; Chen, Z.; Zhu, Q. Neuron-derived CCL2 contributes to microglia activation and neurological decline in hepatic encephalopathy. Biol. Res. 2017, 50, 26. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Cai, Q.; Li, Y.; Pei, G. Polysaccharides from Ganoderma lucidum attenuate microglia-mediated neuroinflammation and modulate microglial phagocytosis and behavioural response. J. Neuroinflamm. 2017, 14, 63. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Kwiatkowski, K.; Popiolek-Barczyk, K.; Piotrowska, A.; Rojewska, E.; Ciapala, K.; Makuch, W.; Mika, J. Chemokines CCL2 and CCL7, but not CCL12, play a significant role in the development of pain-related behavior and opioid-induced analgesia. Cytokine 2019, 119, 202–213. [Google Scholar] [CrossRef] [PubMed]
  60. Sun, S.C.; Liu, Z.G. A special issue on NF-κB signaling and function. Cell Res. 2011, 21, 1–2. [Google Scholar] [CrossRef] [Green Version]
  61. Zhang, H.; Sun, S.C. NF-κB in inflammation and renal diseases. Cell Biosci. 2015, 5, 63. [Google Scholar] [CrossRef] [Green Version]
  62. Hayden, M.S.; Ghosh, S. Shared principles in NF-κB signaling. Cell 2008, 132, 344–362. [Google Scholar] [CrossRef] [Green Version]
  63. Brás, J.P.; Bravo, J.; Freitas, J.; Barbosa, M.A.; Santos, S.G.; Summavielle, T.; Almeida, M.I. TNF-α-induced microglia activation requires miR-342: Impact on NF-kB signaling and neurotoxicity. Cell Death Dis. 2020, 11, 415. [Google Scholar] [CrossRef] [PubMed]
  64. Wen, X.; Xiao, L.; Zhong, Z.; Wang, L.; Li, Z.; Pan, X.; Liu, Z. Astaxanthin acts via LRP-1 to inhibit inflammation and reverse lipopolysaccharide-induced M1/M2 polarization of microglial cells. Oncotarget 2017, 8, 69370–69385. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Gibson, C.J.; Hossain, M.M.; Richardson, J.R.; Aleksunes, L.M. Inflammatory regulation of ATP binding cassette efflux transporter expression and function in microglia. J. Pharmacol. Exp. Ther. 2012, 343, 650–660. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Cho, I.H.; Hong, J.; Suh, E.C.; Kim, J.H.; Lee, H.; Lee, J.E.; Lee, S.; Kim, C.H.; Kim, D.W.; Jo, E.K.; et al. Role of microglial IKKbeta in kainic acid-induced hippocampal neuronal cell death. Brain 2008, 131, 3019–3033. [Google Scholar] [CrossRef]
  67. Zhang, J.; Deng, X. Bupivacaine effectively relieves inflammation-induced pain by suppressing activation of the NF-κB signalling pathway and inhibiting the activation of spinal microglia and astrocytes. Exp. Ther. Med. 2017, 13, 1074–1080. [Google Scholar] [CrossRef] [Green Version]
  68. Li, X.; Wu, G.; Li, M.; Zhang, Z. Oleanolic acid administration alleviates neuropathic pain after a peripheral nerve injury by regulating microglia polarization-mediated neuroinflammation. RSC Adv. 2020, 10, 12920–12928. [Google Scholar] [CrossRef]
  69. Zhang, W.; Liu, H.T. MAPK signal pathways in the regulation of cell proliferation in mammalian cells. Cell Res. 2002, 12, 9–18. [Google Scholar] [CrossRef]
  70. Yarza, R.; Vela, S.; Solas, M.; Ramirez, M.J. c-Jun N-terminal Kinase (JNK) Signaling as a Therapeutic Target for Alzheimer’s Disease. Front. Pharmacol. 2015, 6, 321. [Google Scholar] [CrossRef] [Green Version]
  71. Peng, J.; Andersen, J.K. The role of c-Jun N-terminal kinase (JNK) in Parkinson’s disease. IUBMB Life 2003, 55, 267–271. [Google Scholar] [CrossRef]
  72. Waetzig, V.; Czeloth, K.; Hidding, U.; Mielke, K.; Kanzow, M.; Brecht, S.; Goetz, M.; Lucius, R.; Herdegen, T.; Hanisch, U.K. c-Jun N-terminal kinases (JNKs) mediate pro-inflammatory actions of microglia. Glia 2005, 50, 235–246. [Google Scholar] [CrossRef]
  73. Wang, Y.R.; Xu, H.; Tao, M.; Xu, L.H.; Fu, X.C. Ligustilide Relieves Complete Freund’s Adjuvant-induced Mechanical Hyperalgesia through Inhibiting the Activation of Spinal c-Jun N-terminal Kinase/c-Jun Pathway in Rats. Pharmacogn. Mag. 2017, 13, 634–638. [Google Scholar] [CrossRef]
  74. Gasco, H.A.; Ros-Bernal, F.; Castillo-Gómez, E.; Olucha-Bordonau, F. MAPK/ERK Dysfunction in Neurodegenerative Diseases. Available online: https://pdfs.semanticscholar.org/25e1/3caa0ba99099e466e319cf166cfe22d4a2a9.pdf (accessed on 7 June 2023).
  75. Zhuang, Z.Y.; Gerner, P.; Woolf, C.J.; Ji, R.R. ERK is sequentially activated in neurons, microglia, and astrocytes by spinal nerve ligation and contributes to mechanical allodynia in this neuropathic pain model. Pain 2005, 114, 149–159. [Google Scholar] [CrossRef] [PubMed]
  76. Kang, C.-H.; Jayasooriya, R.G.P.T.; Dilshara, M.G.; Choi, Y.H.; Jeong, Y.-K.; Kim, N.D.; Kim, G.-Y. Caffeine suppresses lipopolysaccharide-stimulated BV2 microglial cells by suppressing Akt-mediated NF-κB activation and ERK phosphorylation. Food Chem. Toxicol. 2012, 50, 4270–4276. [Google Scholar] [CrossRef]
  77. Ryu, K.-Y.; Lee, H.-J.; Woo, H.; Kang, R.-J.; Han, K.-M.; Park, H.; Lee, S.M.; Lee, J.-Y.; Jeong, Y.J.; Nam, H.-W. Dasatinib regulates LPS-induced microglial and astrocytic neuroinflammatory responses by inhibiting AKT/STAT3 signaling. J. Neuroinflamm. 2019, 16, 190. [Google Scholar] [CrossRef]
  78. Zhang, H.; Ma, S.B.; Gao, Y.J.; Xing, J.L.; Xian, H.; Li, Z.Z.; Shen, S.N.; Wu, S.X.; Luo, C.; Xie, R.G. Spinal CCL2 Promotes Pain Sensitization by Rapid Enhancement of NMDA-Induced Currents Through the ERK-GluN2B Pathway in Mouse Lamina II Neurons. Neurosci. Bull. 2020, 36, 1344–1354. [Google Scholar] [CrossRef] [PubMed]
  79. Cao, D.L.; Zhang, Z.J.; Xie, R.G.; Jiang, B.C.; Ji, R.R.; Gao, Y.J. Chemokine CXCL1 enhances inflammatory pain and increases NMDA receptor activity and COX-2 expression in spinal cord neurons via activation of CXCR2. Exp. Neurol. 2014, 261, 328–336. [Google Scholar] [CrossRef] [Green Version]
  80. Kumar, S.; Boehm, J.; Lee, J.C. p38 MAP kinases: Key signalling molecules as therapeutic targets for inflammatory diseases. Nat. Rev. Drug Discov. 2003, 2, 717–726. [Google Scholar] [CrossRef] [PubMed]
  81. Ji, R.R.; Suter, M.R. p38 MAPK, microglial signaling, and neuropathic pain. Mol. Pain 2007, 3, 33. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Ji, R.R.; Samad, T.A.; Jin, S.X.; Schmoll, R.; Woolf, C.J. p38 MAPK activation by NGF in primary sensory neurons after inflammation increases TRPV1 levels and maintains heat hyperalgesia. Neuron 2002, 36, 57–68. [Google Scholar] [CrossRef] [Green Version]
  83. Qi, J.; Chen, C.; Meng, Q.X.; Wu, Y.; Wu, H.; Zhao, T.B. Crosstalk between Activated Microglia and Neurons in the Spinal Dorsal Horn Contributes to Stress-induced Hyperalgesia. Sci. Rep. 2016, 6, 39442. [Google Scholar] [CrossRef] [Green Version]
  84. Taves, S.; Berta, T.; Liu, D.L.; Gan, S.; Chen, G.; Kim, Y.H.; Van de Ven, T.; Laufer, S.; Ji, R.R. Spinal inhibition of p38 MAP kinase reduces inflammatory and neuropathic pain in male but not female mice: Sex-dependent microglial signaling in the spinal cord. Brain Behav. Immun. 2016, 55, 70–81. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Fan, Z.; Zhang, W.; Cao, Q.; Zou, L.; Fan, X.; Qi, C.; Yan, Y.; Song, B.; Wu, B. JAK2/STAT3 pathway regulates microglia polarization involved in hippocampal inflammatory damage due to acute paraquat exposure. Ecotoxicol. Environ. Saf. 2022, 234, 113372. [Google Scholar] [CrossRef] [PubMed]
  86. Xiong, J.; Wang, C.; Chen, H.; Hu, Y.; Tian, L.; Pan, J.; Geng, M. Aβ-induced microglial cell activation is inhibited by baicalin through the JAK2/STAT3 signaling pathway. Int. J. Neurosci. 2014, 124, 609–620. [Google Scholar] [CrossRef] [PubMed]
  87. Porro, C.; Cianciulli, A.; Trotta, T.; Lofrumento, D.D.; Panaro, M.A. Curcumin Regulates Anti-Inflammatory Responses by JAK/STAT/SOCS Signaling Pathway in BV-2 Microglial Cells. Biology 2019, 8, 51. [Google Scholar] [CrossRef] [Green Version]
  88. Liu, J.; Xie, X.; Qin, K.; Xu, L.; Peng, J.; Li, X.; Li, X.; Liu, Z. Dexamethasone and potassium canrenoate alleviate hyperalgesia by competitively regulating IL-6/JAK2/STAT3 signaling pathway during inflammatory pain in vivo and in vitro. Immun. Inflamm. Dis. 2022, 10, e721. [Google Scholar] [CrossRef]
  89. Yu, S.; Zhao, G.; Han, F.; Liang, W.; Jiao, Y.; Li, Z.; Li, L. Muscone relieves inflammatory pain by inhibiting microglial activation-mediated inflammatory response via abrogation of the NOX4/JAK2-STAT3 pathway and NLRP3 inflammasome. Int. Immunopharmacol. 2020, 82, 106355. [Google Scholar] [CrossRef]
  90. Simpson, D.S.A.; Oliver, P.L. ROS Generation in Microglia: Understanding Oxidative Stress and Inflammation in Neurodegenerative Disease. Antioxidants 2020, 9, 743. [Google Scholar] [CrossRef]
  91. Ren, P.; Chen, J.; Li, B.; Zhang, M.; Yang, B.; Guo, X.; Chen, Z.; Cheng, H.; Wang, P.; Wang, S.; et al. Nrf2 Ablation Promotes Alzheimer’s Disease-Like Pathology in APP/PS1 Transgenic Mice: The Role of Neuroinflammation and Oxidative Stress. Oxid. Med. Cell Longev. 2020, 2020, 3050971. [Google Scholar] [CrossRef]
  92. Rojo, A.I.; Pajares, M.; Garcia-Yague, A.J.; Buendia, I.; Van Leuven, F.; Yamamoto, M.; Lopez, M.G.; Cuadrado, A. Deficiency in the transcription factor NRF2 worsens inflammatory parameters in a mouse model with combined tauopathy and amyloidopathy. Redox Biol. 2018, 18, 173–180. [Google Scholar] [CrossRef]
  93. Velagapudi, R.; El-Bakoush, A.; Olajide, O.A. Activation of Nrf2 Pathway Contributes to Neuroprotection by the Dietary Flavonoid Tiliroside. Mol. Neurobiol. 2018, 55, 8103–8123. [Google Scholar] [CrossRef] [Green Version]
  94. Li, Y.; Lv, O.; Zhou, F.; Li, Q.; Wu, Z.; Zheng, Y. Linalool Inhibits LPS-Induced Inflammation in BV2 Microglia Cells by Activating Nrf2. Neurochem. Res. 2015, 40, 1520–1525. [Google Scholar] [CrossRef] [PubMed]
  95. Rosa, A.O.; Egea, J.; Lorrio, S.; Rojo, A.I.; Cuadrado, A.; Lopez, M.G. Nrf2-mediated haeme oxygenase-1 up-regulation induced by cobalt protoporphyrin has antinociceptive effects against inflammatory pain in the formalin test in mice. Pain 2008, 137, 332–339. [Google Scholar] [CrossRef] [PubMed]
  96. Aman, Y.; Schmauck-Medina, T.; Hansen, M.; Morimoto, R.I.; Simon, A.K.; Bjedov, I.; Palikaras, K.; Simonsen, A.; Johansen, T.; Tavernarakis, N.; et al. Autophagy in healthy aging and disease. Nat. Aging 2021, 1, 634–650. [Google Scholar] [CrossRef] [PubMed]
  97. Fang, E.F.; Hou, Y.; Palikaras, K.; Adriaanse, B.A.; Kerr, J.S.; Yang, B.; Lautrup, S.; Hasan-Olive, M.M.; Caponio, D.; Dan, X.; et al. Mitophagy inhibits amyloid-β and tau pathology and reverses cognitive deficits in models of Alzheimer’s disease. Nat. Neurosci. 2019, 22, 401–412. [Google Scholar] [CrossRef] [PubMed]
  98. Ye, X.; Zhu, M.; Che, X.; Wang, H.; Liang, X.J.; Wu, C.; Xue, X.; Yang, J. Lipopolysaccharide induces neuroinflammation in microglia by activating the MTOR pathway and downregulating Vps34 to inhibit autophagosome formation. J. Neuroinflamm. 2020, 17, 18. [Google Scholar] [CrossRef] [Green Version]
  99. Liu, C.; Zheng, X.; Liu, L.; Hu, Y.; Zhu, Q.; Zhang, J.; Wang, H.; Gu, E.W.; Yang, Z.; Xu, G. Caloric Restriction Alleviates CFA-Induced Inflammatory Pain via Elevating β-Hydroxybutyric Acid Expression and Restoring Autophagic Flux in the Spinal Cord. Front. Neurosci. 2022, 16, 828278. [Google Scholar] [CrossRef] [PubMed]
  100. Liang, L.; Tao, B.; Fan, L.; Yaster, M.; Zhang, Y.; Tao, Y.X. mTOR and its downstream pathway are activated in the dorsal root ganglion and spinal cord after peripheral inflammation, but not after nerve injury. Brain Res. 2013, 1513, 17–25. [Google Scholar] [CrossRef] [Green Version]
  101. Azab, A.; Nassar, A.; Azab, A.N. Anti-Inflammatory Activity of Natural Products. Molecules 2016, 21, 1321. [Google Scholar] [CrossRef] [Green Version]
  102. Shen, W.; Qi, R.; Zhang, J.; Wang, Z.; Wang, H.; Hu, C.; Zhao, Y.; Bie, M.; Wang, Y.; Fu, Y.; et al. Chlorogenic acid inhibits LPS-induced microglial activation and improves survival of dopaminergic neurons. Brain Res. Bull. 2012, 88, 487–494. [Google Scholar] [CrossRef]
  103. dos Santos, M.D.; Almeida, M.C.; Lopes, N.P.; de Souza, G.E. Evaluation of the anti-inflammatory, analgesic and antipyretic activities of the natural polyphenol chlorogenic acid. Biol. Pharm. Bull. 2006, 29, 2236–2240. [Google Scholar] [CrossRef] [Green Version]
  104. Agudelo-Ochoa, G.M.; Pulgarin-Zapata, I.C.; Velasquez-Rodriguez, C.M.; Duque-Ramirez, M.; Naranjo-Cano, M.; Quintero-Ortiz, M.M.; Lara-Guzman, O.J.; Munoz-Durango, K. Coffee Consumption Increases the Antioxidant Capacity of Plasma and Has No Effect on the Lipid Profile or Vascular Function in Healthy Adults in a Randomized Controlled Trial. J. Nutr. 2016, 146, 524–531. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Tajik, N.; Tajik, M.; Mack, I.; Enck, P. The potential effects of chlorogenic acid, the main phenolic components in coffee, on health: A comprehensive review of the literature. Eur. J. Nutr. 2017, 56, 2215–2244. [Google Scholar] [CrossRef] [PubMed]
  106. Rehman, S.U.; Ali, T.; Alam, S.I.; Ullah, R.; Zeb, A.; Lee, K.W.; Rutten, B.P.F.; Kim, M.O. Ferulic Acid Rescues LPS-Induced Neurotoxicity via Modulation of the TLR4 Receptor in the Mouse Hippocampus. Mol. Neurobiol. 2019, 56, 2774–2790. [Google Scholar] [CrossRef]
  107. Priebe, A.; Hunke, M.; Tonello, R.; Sonawane, Y.; Berta, T.; Natarajan, A.; Bhuvanesh, N.; Pattabiraman, M.; Chandra, S. Ferulic acid dimer as a non-opioid therapeutic for acute pain. J. Pain Res. 2018, 11, 1075. [Google Scholar] [CrossRef] [Green Version]
  108. Bumrungpert, A.; Lilitchan, S.; Tuntipopipat, S.; Tirawanchai, N.; Komindr, S. Ferulic Acid Supplementation Improves Lipid Profiles, Oxidative Stress, and Inflammatory Status in Hyperlipidemic Subjects: A Randomized, Double-Blind, Placebo-Controlled Clinical Trial. Nutrients 2018, 10, 713. [Google Scholar] [CrossRef] [Green Version]
  109. Di Giacomo, S.; Percaccio, E.; Gulli, M.; Romano, A.; Vitalone, A.; Mazzanti, G.; Gaetani, S.; Di Sotto, A. Recent Advances in the Neuroprotective Properties of Ferulic Acid in Alzheimer’s Disease: A Narrative Review. Nutrients 2022, 14, 3709. [Google Scholar] [CrossRef] [PubMed]
  110. Liu, Y.; Deng, S.; Zhang, Z.; Gu, Y.; Xia, S.; Bao, X.; Cao, X.; Xu, Y. 6-Gingerol attenuates microglia-mediated neuroinflammation and ischemic brain injuries through Akt-mTOR-STAT3 signaling pathway. Eur. J. Pharmacol. 2020, 883, 173294. [Google Scholar] [CrossRef]
  111. Young, H.Y.; Luo, Y.L.; Cheng, H.Y.; Hsieh, W.C.; Liao, J.C.; Peng, W.H. Analgesic and anti-inflammatory activities of [6]-gingerol. J. Ethnopharmacol. 2005, 96, 207–210. [Google Scholar] [CrossRef]
  112. Sharma, S.; Shukla, M.K.; Sharma, K.C.; Tirath; Kumar, L.; Anal, J.M.H.; Upadhyay, S.K.; Bhattacharyya, S.; Kumar, D. Revisiting the therapeutic potential of gingerols against different pharmacological activities. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2023, 396, 633–647. [Google Scholar] [CrossRef]
  113. Konmun, J.; Danwilai, K.; Ngamphaiboon, N.; Sripanidkulchai, B.; Sookprasert, A.; Subongkot, S. A phase II randomized double-blind placebo-controlled study of 6-gingerol as an anti-emetic in solid tumor patients receiving moderately to highly emetogenic chemotherapy. Med. Oncol. 2017, 34, 69. [Google Scholar] [CrossRef]
  114. Gao, F.; Lei, J.; Zhang, Z.; Yang, Y.; You, H. Curcumin alleviates LPS-induced inflammation and oxidative stress in mouse microglial BV2 cells by targeting miR-137-3p/NeuroD1. RSC Adv. 2019, 9, 38397–38406. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Yu, Y.; Shen, Q.; Lai, Y.; Park, S.Y.; Ou, X.; Lin, D.; Jin, M.; Zhang, W. Anti-inflammatory Effects of Curcumin in Microglial Cells. Front. Pharmacol. 2018, 9, 386. [Google Scholar] [CrossRef] [Green Version]
  116. Singh, A.K.; Vinayak, M. Curcumin attenuates CFA induced thermal hyperalgesia by modulation of antioxidant enzymes and down regulation of TNF-α, IL-1β and IL-6. Neurochem. Res. 2015, 40, 463–472. [Google Scholar] [CrossRef]
  117. Satoskar, R.R.; Shah, S.J.; Shenoy, S.G. Evaluation of anti-inflammatory property of curcumin (diferuloyl methane) in patients with postoperative inflammation. Int. J. Clin. Pharmacol. Ther. Toxicol. 1986, 24, 651–654. [Google Scholar] [PubMed]
  118. Park, S.E.; Sapkota, K.; Kim, S.; Kim, H.; Kim, S.J. Kaempferol acts through mitogen-activated protein kinases and protein kinase B/AKT to elicit protection in a model of neuroinflammation in BV2 microglial cells. Br. J. Pharmacol. 2011, 164, 1008–1025. [Google Scholar] [CrossRef] [Green Version]
  119. Jabbari, S.; Bananej, M.; Zarei, M.; Komaki, A.; Hajikhani, R. Effects of intrathecal and intracerebroventricular microinjection of kaempferol on pain: Possible mechanisms of action. Res. Pharm. Sci. 2021, 16, 203–216. [Google Scholar] [CrossRef]
  120. Alam, W.; Khan, H.; Shah, M.A.; Cauli, O.; Saso, L. Kaempferol as a Dietary Anti-Inflammatory Agent: Current Therapeutic Standing. Molecules 2020, 25, 4073. [Google Scholar] [CrossRef]
  121. Ren, J.; Lu, Y.; Qian, Y.; Chen, B.; Wu, T.; Ji, G. Recent progress regarding kaempferol for the treatment of various diseases. Exp. Ther. Med. 2019, 18, 2759–2776. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Akiyama, M.; Mizokami, T.; Ito, H.; Ikeda, Y. A randomized, placebo-controlled trial evaluating the safety of excessive administration of kaempferol aglycone. Food Sci. Nutr. 2023. [Google Scholar] [CrossRef]
  123. Kang, C.H.; Choi, Y.H.; Moon, S.K.; Kim, W.J.; Kim, G.Y. Quercetin inhibits lipopolysaccharide-induced nitric oxide production in BV2 microglial cells by suppressing the NF-κB pathway and activating the Nrf2-dependent HO-1 pathway. Int. Immunopharmacol. 2013, 17, 808–813. [Google Scholar] [CrossRef]
  124. Kumar, S.; Vinayak, M. Quercetin Ameliorates CFA-Induced Chronic Inflammatory Hyperalgesia via Modulation of ROS-Mediated ERK1/2 Signaling and Inhibition of Spinal Glial Activation In Vivo. Neuromol. Med. 2020, 22, 517–533. [Google Scholar] [CrossRef] [PubMed]
  125. Javadi, F.; Ahmadzadeh, A.; Eghtesadi, S.; Aryaeian, N.; Zabihiyeganeh, M.; Rahimi Foroushani, A.; Jazayeri, S. The Effect of Quercetin on Inflammatory Factors and Clinical Symptoms in Women with Rheumatoid Arthritis: A Double-Blind, Randomized Controlled Trial. J. Am. Coll. Nutr. 2017, 36, 9–15. [Google Scholar] [CrossRef] [PubMed]
  126. El-Bakoush, A.; Olajide, O.A. Formononetin inhibits neuroinflammation and increases estrogen receptor beta (ERβ) protein expression in BV2 microglia. Int. Immunopharmacol. 2018, 61, 325–337. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Wang, X.S.; Guan, S.Y.; Liu, A.; Yue, J.; Hu, L.N.; Zhang, K.; Yang, L.K.; Lu, L.; Tian, Z.; Zhao, M.G.; et al. Anxiolytic effects of Formononetin in an inflammatory pain mouse model. Mol. Brain 2019, 12, 36. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Ong, S.K.L.; Shanmugam, M.K.; Fan, L.; Fraser, S.E.; Arfuso, F.; Ahn, K.S.; Sethi, G.; Bishayee, A. Focus on Formononetin: Anticancer Potential and Molecular Targets. Cancers 2019, 11, 611. [Google Scholar] [CrossRef] [Green Version]
  129. Zhang, B.; Wei, Y.Z.; Wang, G.Q.; Li, D.D.; Shi, J.S.; Zhang, F. Targeting MAPK Pathways by Naringenin Modulates Microglia M1/M2 Polarization in Lipopolysaccharide-Stimulated Cultures. Front. Cell Neurosci. 2018, 12, 531. [Google Scholar] [CrossRef] [Green Version]
  130. Pinho-Ribeiro, F.A.; Zarpelon, A.C.; Fattori, V.; Manchope, M.F.; Mizokami, S.S.; Casagrande, R.; Verri, W.A., Jr. Naringenin reduces inflammatory pain in mice. Neuropharmacology 2016, 105, 508–519. [Google Scholar] [CrossRef]
  131. Rebello, C.J.; Beyl, R.A.; Lertora, J.J.L.; Greenway, F.L.; Ravussin, E.; Ribnicky, D.M.; Poulev, A.; Kennedy, B.J.; Castro, H.F.; Campagna, S.R.; et al. Safety and pharmacokinetics of naringenin: A randomized, controlled, single-ascending-dose clinical trial. Diabetes Obes. Metab. 2020, 22, 91–98. [Google Scholar] [CrossRef]
  132. Cara, K.C.; Beauchesne, A.R.; Wallace, T.C.; Chung, M. Effects of 100% Orange Juice on Markers of Inflammation and Oxidation in Healthy and At-Risk Adult Populations: A Scoping Review, Systematic Review, and Meta-analysis. Adv. Nutr. 2022, 13, 116–137. [Google Scholar] [CrossRef]
  133. Zhang, S.; Gao, L.; Liu, X.; Lu, T.; Xie, C.; Jia, J. Resveratrol Attenuates Microglial Activation via SIRT1-SOCS1 Pathway. Evid. Based Complement. Alternat. Med. 2017, 2017, 8791832. [Google Scholar] [CrossRef] [Green Version]
  134. Ma, Y.; Liu, S.; Shu, H.; Crawford, J.; Xing, Y.; Tao, F. Resveratrol alleviates temporomandibular joint inflammatory pain by recovering disturbed gut microbiota. Brain Behav. Immun. 2020, 87, 455–464. [Google Scholar] [CrossRef] [PubMed]
  135. Marouf, B.H.; Hussain, S.A.; Ali, Z.S.; Ahmmad, R.S. Resveratrol Supplementation Reduces Pain and Inflammation in Knee Osteoarthritis Patients Treated with Meloxicam: A Randomized Placebo-Controlled Study. J. Med. Food 2018, 21, 1253–1259. [Google Scholar] [CrossRef] [PubMed]
  136. Lin, S.R.; Fu, Y.S.; Tsai, M.J.; Cheng, H.; Weng, C.F. Natural Compounds from Herbs that can Potentially Execute as Autophagy Inducers for Cancer Therapy. Int. J. Mol. Sci. 2017, 18, 1412. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Rickert, U.; Cossais, F.; Heimke, M.; Arnold, P.; Preusse-Prange, A.; Wilms, H.; Lucius, R. Anti-inflammatory properties of Honokiol in activated primary microglia and astrocytes. J. Neuroimmunol. 2018, 323, 78–86. [Google Scholar] [CrossRef] [PubMed]
  138. Khalid, S.; Ullah, M.Z.; Khan, A.U.; Afridi, R.; Rasheed, H.; Khan, A.; Ali, H.; Kim, Y.S.; Khan, S. Antihyperalgesic Properties of Honokiol in Inflammatory Pain Models by Targeting of NF-κB and Nrf2 Signaling. Front. Pharmacol. 2018, 9, 140. [Google Scholar] [CrossRef] [Green Version]
  139. Eliaz, I.; Weil, E. Intravenous Honokiol in Drug-Resistant Cancer: Two Case Reports. Integr. Cancer Ther. 2020, 19, 1534735420922615. [Google Scholar] [CrossRef] [PubMed]
  140. Zhu, M.D.; Zhao, L.X.; Wang, X.T.; Gao, Y.J.; Zhang, Z.J. Ligustilide inhibits microglia-mediated proinflammatory cytokines production and inflammatory pain. Brain Res. Bull. 2014, 109, 54–60. [Google Scholar] [CrossRef]
  141. Wang, J.; Du, J.R.; Wang, Y.; Kuang, X.; Wang, C.Y. Z-ligustilide attenuates lipopolysaccharide-induced proinflammatory response via inhibiting NF-κB pathway in primary rat microglia. Acta Pharmacol. Sin. 2010, 31, 791–797. [Google Scholar] [CrossRef] [Green Version]
  142. Du, J.; Yu, Y.; Ke, Y.; Wang, C.; Zhu, L.; Qian, Z.M. Ligustilide attenuates pain behavior induced by acetic acid or formalin. J. Ethnopharmacol. 2007, 112, 211–214. [Google Scholar] [CrossRef]
  143. Zhang, Y.; Zhang, Y.; Han, Y.; Tian, Y.; Wu, P.; Xin, A.; Wei, X.; Shi, Y.; Zhang, Z.; Su, G.; et al. Pharmacokinetics, tissue distribution, and safety evaluation of a ligustilide derivative (LIGc). J. Pharm. Biomed. Anal. 2020, 182, 113140. [Google Scholar] [CrossRef] [PubMed]
  144. Sun, X.; Zeng, H.; Wang, Q.; Yu, Q.; Wu, J.; Feng, Y.; Deng, P.; Zhang, H. Glycyrrhizin ameliorates inflammatory pain by inhibiting microglial activation-mediated inflammatory response via blockage of the HMGB1-TLR4-NF-kB pathway. Exp. Cell Res. 2018, 369, 112–119. [Google Scholar] [CrossRef]
  145. Cao, Z.Y.; Liu, Y.Z.; Li, J.M.; Ruan, Y.M.; Yan, W.J.; Zhong, S.Y.; Zhang, T.; Liu, L.L.; Wu, R.; Wang, B.; et al. Glycyrrhizic acid as an adjunctive treatment for depression through anti-inflammation: A randomized placebo-controlled clinical trial. J. Affect. Disord. 2020, 265, 247–254. [Google Scholar] [CrossRef]
  146. Lu, Y.; Zhao, L.X.; Cao, D.L.; Gao, Y.J. Spinal injection of docosahexaenoic acid attenuates carrageenan-induced inflammatory pain through inhibition of microglia-mediated neuroinflammation in the spinal cord. Neuroscience 2013, 241, 22–31. [Google Scholar] [CrossRef] [PubMed]
  147. Hill, C.L.; March, L.M.; Aitken, D.; Lester, S.E.; Battersby, R.; Hynes, K.; Fedorova, T.; Proudman, S.M.; James, M.; Cleland, L.G.; et al. Fish oil in knee osteoarthritis: A randomised clinical trial of low dose versus high dose. Ann. Rheum. Dis. 2016, 75, 23–29. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Hu, B.; Xu, G.; Zhang, X.; Xu, L.; Zhou, H.; Ma, Z.; Shen, X.; Zhu, J.; Shen, R. Paeoniflorin Attenuates Inflammatory Pain by Inhibiting Microglial Activation and Akt-NF-κB Signaling in the Central Nervous System. Cell Physiol. Biochem. 2018, 47, 842–850. [Google Scholar] [CrossRef]
  149. Li, X.; Shi, F.; Zhang, R.; Sun, C.; Gong, C.; Jian, L.; Ding, L. Pharmacokinetics, Safety, and Tolerability of Amygdalin and Paeoniflorin after Single and Multiple Intravenous Infusions of Huoxue-Tongluo Lyophilized Powder for Injection in Healthy Chinese Volunteers. Clin. Ther. 2016, 38, 327–337. [Google Scholar] [CrossRef]
  150. Shukla, S.M.; Sharma, S.K. Sinomenine inhibits microglial activation by Aβ and confers neuroprotection. J. Neuroinflamm. 2011, 8, 117. [Google Scholar] [CrossRef] [Green Version]
  151. Yuan, Y.; Zhang, Y.; He, X.; Fan, S. Protective effects of sinomenine on CFA-induced inflammatory pain in rats. Med. Sci. Monit. Int. Med. J. Exp. Clin. Res. 2018, 24, 2018–2024. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Liu, W.; Zhang, Y.; Zhu, W.; Ma, C.; Ruan, J.; Long, H.; Wang, Y. Sinomenine Inhibits the Progression of Rheumatoid Arthritis by Regulating the Secretion of Inflammatory Cytokines and Monocyte/Macrophage Subsets. Front. Immunol. 2018, 9, 2228. [Google Scholar] [CrossRef] [PubMed]
  153. Liu, S.; Cheng, Y.; Rao, M.; Tang, M.; Dong, Z. Muscone Induces CYP1A2 and CYP3A4 Enzyme Expression in L02 Human Liver Cells and CYP1A2 and CYP3A11 Enzyme Expression in Kunming Mice. Pharmacology 2017, 99, 205–215. [Google Scholar] [CrossRef]
  154. Xu, J.; Yuan, C.; Wang, G.; Luo, J.; Ma, H.; Xu, L.; Mu, Y.; Li, Y.; Seeram, N.P.; Huang, X.; et al. Urolithins Attenuate LPS-Induced Neuroinflammation in BV2Microglia via MAPK, Akt, and NF-κB Signaling Pathways. J. Agric. Food Chem. 2018, 66, 571–580. [Google Scholar] [CrossRef]
  155. D’Amico, D.; Olmer, M.; Fouassier, A.M.; Valdes, P.; Andreux, P.A.; Rinsch, C.; Lotz, M. Urolithin A improves mitochondrial health, reduces cartilage degeneration, and alleviates pain in osteoarthritis. Aging Cell 2022, 21, e13662. [Google Scholar] [CrossRef] [PubMed]
  156. Liu, S.; D’Amico, D.; Shankland, E.; Bhayana, S.; Garcia, J.M.; Aebischer, P.; Rinsch, C.; Singh, A.; Marcinek, D.J. Effect of Urolithin A Supplementation on Muscle Endurance and Mitochondrial Health in Older Adults: A Randomized Clinical Trial. JAMA Netw. Open. 2022, 5, e2144279. [Google Scholar] [CrossRef]
  157. Niu, J.; Straubinger, R.M.; Mager, D.E. Pharmacodynamic Drug-Drug Interactions. Clin. Pharmacol. Ther. 2019, 105, 1395–1406. [Google Scholar] [CrossRef] [PubMed]
  158. Liu, C.; Huang, Z.; Li, Z.; Li, J.; Li, Y. Muscone reduced the hypnotic and analgesic effect of ketamine in mice. J. Chin. Med. Assoc. 2020, 83, 148–155. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Inflammatory mediators associated with the development of microglial activation-mediated inflammatory pain. Tissue damage associated with inflammation lead to microglial activation by releasing diverse signaling molecules from sensory neurons. Activated microglia induce neuroinflammation through increasing the expression of inflammatory mediators, such as excessive NO, iNOS, COX-2, TNF-α, IL-1β, IL-6, MCP1, and MCP3. These mediators lead to central sensitization, resulting in increased sensitivity to pain. Activated microglia can trigger inflammatory pain via inflammatory mediator-related signaling.
Figure 1. Inflammatory mediators associated with the development of microglial activation-mediated inflammatory pain. Tissue damage associated with inflammation lead to microglial activation by releasing diverse signaling molecules from sensory neurons. Activated microglia induce neuroinflammation through increasing the expression of inflammatory mediators, such as excessive NO, iNOS, COX-2, TNF-α, IL-1β, IL-6, MCP1, and MCP3. These mediators lead to central sensitization, resulting in increased sensitivity to pain. Activated microglia can trigger inflammatory pain via inflammatory mediator-related signaling.
Pharmaceuticals 16 00941 g001
Figure 2. Intracellular signaling that can induce inflammatory mediators in activated microglia. NF-κB, MAPK, and JAK2/STAT3 signaling can be activated in microglia. And this signaling can induce expression of inflammatory mediators in activated microglia, leading to exacerbation of inflammatory pain.
Figure 2. Intracellular signaling that can induce inflammatory mediators in activated microglia. NF-κB, MAPK, and JAK2/STAT3 signaling can be activated in microglia. And this signaling can induce expression of inflammatory mediators in activated microglia, leading to exacerbation of inflammatory pain.
Pharmaceuticals 16 00941 g002
Figure 3. Intracellular signaling that can regulate inflammatory mediators in microglia. Nrf2 signaling and autophagy have a protective effect on microglial activation. Inflammatory mediators expressed in activated microglia are suppressed by activation of Nrf2 signaling and autophagy.
Figure 3. Intracellular signaling that can regulate inflammatory mediators in microglia. Nrf2 signaling and autophagy have a protective effect on microglial activation. Inflammatory mediators expressed in activated microglia are suppressed by activation of Nrf2 signaling and autophagy.
Pharmaceuticals 16 00941 g003
Figure 4. Effect of natural product-derived compounds on inflammatory pain via the suppression of microglial activation. Natural products and phytochemicals may have suppressive effects on microglial activation. Each compound may inhibit microglial activation by regulating the intracellular signaling pathways. The suppression of microglial activation by the modulation of intracellular signaling via natural product-derived compounds shows potential for attenuating inflammatory pain.
Figure 4. Effect of natural product-derived compounds on inflammatory pain via the suppression of microglial activation. Natural products and phytochemicals may have suppressive effects on microglial activation. Each compound may inhibit microglial activation by regulating the intracellular signaling pathways. The suppression of microglial activation by the modulation of intracellular signaling via natural product-derived compounds shows potential for attenuating inflammatory pain.
Pharmaceuticals 16 00941 g004
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Park, J.; Lee, C.; Kim, Y.T. Effects of Natural Product-Derived Compounds on Inflammatory Pain via Regulation of Microglial Activation. Pharmaceuticals 2023, 16, 941. https://0-doi-org.brum.beds.ac.uk/10.3390/ph16070941

AMA Style

Park J, Lee C, Kim YT. Effects of Natural Product-Derived Compounds on Inflammatory Pain via Regulation of Microglial Activation. Pharmaceuticals. 2023; 16(7):941. https://0-doi-org.brum.beds.ac.uk/10.3390/ph16070941

Chicago/Turabian Style

Park, Joon, Changho Lee, and Yun Tai Kim. 2023. "Effects of Natural Product-Derived Compounds on Inflammatory Pain via Regulation of Microglial Activation" Pharmaceuticals 16, no. 7: 941. https://0-doi-org.brum.beds.ac.uk/10.3390/ph16070941

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