Subscribe to RSS
DOI: 10.1055/a-2441-6488
Pharmacological Effects of Paeonia lactiflora Focusing on Painful Diabetic Neuropathy
This work is funded in-house.
- Abstract
- Introduction
- Search Strategy
- Phytochemical Characteristic of Paeonia lactiflora
- In Silico Data
- Preclinical Data
- Clinical Data
- Summary and Conclusion
- Contributorsʼ Statement
- References
Abstract
Painful diabetic neuropathy (PDN) is a highly prevalent complication in patients suffering from diabetes mellitus. Given the inadequate pain-relieving effect of current therapies for PDN, there is a high unmet medical need for specialized therapeutic options. In traditional Chinese medicine (TCM), various herbal formulations have been implemented for centuries to relieve pain, and one commonly used plant in this context is Paeonia lactiflora (P. lactiflora). Here, we summarize the chemical constituents of P. lactiflora including their pharmacological mechanisms-of-action and discuss potential benefits for the treatment of PDN. For this, in silico data, as well as preclinical and clinical studies, were critically reviewed and comprehensively compiled. Our findings reveal that P. lactiflora and its individual constituents exhibit a variety of pharmacological properties relevant for PDN, including antinociceptive, anti-inflammatory, antioxidant, and antiapoptotic activities. Through this multifaceted and complex combination of various pharmacological effects, relevant hallmarks of PDN are specifically addressed, suggesting that P. lactiflora may represent a promising source for novel therapeutic approaches for PDN.
#
Keywords
Paeonia lactiflora - Paeoniaceae - painful diabetic neuropathy - nociception - neuroprotection - neuroinflammation - oxidative stressAbbreviations
Introduction
Diabetes mellitus is one of the most serious pandemics of the 21st century. The global prevalence was estimated to be 11% in 2021, which corresponds to more than 500 million people. By 2045, the global number of people affected is predicted to rise to up to 12%, with around 90% of the patients suffering from diabetes mellitus type 2 (DMT2) [1], [2]. DMT2 leads to numerous complications, including cardiovascular effects, renal disease, and retinopathy [2]. Notably, one of the most common chronic complications is peripheral neuropathy, with a highly variable lifetime prevalence of up to 50% of diabetic patients, depending on the country, as well as on age and the duration of the diabetes [3], [4], [5]. Pain is a common clinical manifestation associated with neuropathy, affecting approximately 20% of the patient population [6], [7]. However, these statistics are most likely underestimated. For example, the PROTECT study, carried out in Germany between 2013 and 2016, analyzed the presence and prior diagnosis of distal sensory neuropathy in 1850 patients with or without a history of diabetes. Apparently, 43% of DMT2 patients had been diagnosed with painful sensory neuropathy, while 62% actually demonstrated neuropathic pain during the medical examination, a discrepancy suggesting a noticeably higher number of undiagnosed patients with PDN [8]. Overall, as PDN significantly reduces the quality of life, there is a high unmet medical need to alleviate or even prevent the symptoms [9], [10], [11].
Diabetic neuropathy leads to various structural and functional alterations of the peripheral nervous system (PNS), including reduced nerve fiber density in the epidermis, axonal atrophy, and demyelination, while sensory neurons are predominantly affected. The cell bodies of the sensory neurons are localized in the dorsal root ganglia (DRG), with long axons extending far into distal areas of the body ([Fig. 1]) [12], [13]. Highly specialized sensory neurons, the so-called nociceptors that can be divided into thinly myelinated Aδ fibers and unmyelinated C fibers, are relevant for the development of pain [14]. Nociceptors express ion channels such as voltage-gated sodium channels, particularly NaV 1.7 and NaV 1.8, transient receptor potential vanilloid 1 (TRPV1), and transient receptor potential ankyrin 1 (TRPA1), as well as voltage-gated calcium channels like CaV 3.2 and many others ([Fig. 1]) [15]. The activation of these channels triggers an action potential, thereby generating further nociceptive signaling to the central nervous system (CNS) [14]. Notably, impaired sensory neurons can become hyperexcitable, resulting in spontaneous activity in the absence of a triggering signal, which is associated with subsequent central sensitization and nociception [16]. Schwann cells, major players for the structural and functional integrity of neurons in the PNS, also appear to have an impaired metabolism in diabetic patients, causing further destabilization of the axons ([Fig. 1]) [17], [18]. Furthermore, dysfunction of the vascular endothelia cells can lead to deficiencies in blood supply and hypoxia, subsequently causing damage to sensory neurons and Schwann cells [13]. Ultimately, cell injury in the PNS leads to the activation of macrophages [19]. [Fig. 1] presents the anatomy of a peripheral nerve and the cell types relevant for the pathophysiology of PDN.


To date, the complex pathogenesis of diabetic neuropathy is not yet completely elucidated, but various key signaling pathways have been reported that can be assigned to the three main cell damaging pillars of DMT2: hyperglycemia, dyslipidemia, and insulin resistance [20], [21]. [Fig. 2] illustrates the complex signaling interplay of these three pillars.


In sensory neurons and Schwann cells, glucose as well as fatty acids are used as substrates in the energy metabolism to generate acetyl-CoA by glycolysis or by β-oxidation. Acetyl-CoA is then incorporated into the tricarboxylic acid (TCA) cycle in the mitochondria, producing the redox-active cofactors NADH and dihydroflavin-adenine dinucleotide (FADH2), which are used to produce ATP by oxidative phosphorylation. Oxidative phosphorylation, however, is associated with the production of small amounts of reactive oxygen species (ROS), which are easily eliminated under physiological conditions by cellular antioxidant mechanisms [13].
Since glucose uptake in the PNS involves insulin-independent glucose transporters (GLUT) 1 and GLUT3, impaired insulin signaling in DMT2 is not associated with reduced glucose uptake in sensory neurons or Schwann cells, resulting in an increased activation of various glucose-dependent metabolic pathways in these cells in the presence of increased blood glucose levels ([Fig. 2]) [13], [22]. As a consequence, elevated glycolysis can lead to a disruption of the respiratory chain due to a negative feedback loop. However, in the onset of DMT2, elevated glycolysis contributes to an increased formation of ROS that exceeds the capacity of the endogenous redox systems [12], [21].
During glycolysis, fructose-6-phosphate (F6P) is converted into glyceraldehyde-3-phosphate (G3P), which can be further transformed into the reactive glucose metabolite methylglyoxal ([Fig. 2]) [23]. Both glucose and methylglyoxal lead to unspecific glycation of lipids, nucleotides, and proteins, forming so-called advanced glycation end products (AGEs) with altered functionality [21]. Extracellular AGE can bind to receptors for AGE (RAGE), which are able to trigger downstream signaling pathways via the activation of the transcription factor nuclear factor κB (NF-κB) [24]. Similarly, ion channels at the terminals of nociceptors can undergo glycation, forming AGEs contributing to the hyperexcitability of sensory neurons as described above [15].
Furthermore, G3P is in equilibrium with dihydroxyacetone-phosphate (DHAP), which can be converted to diacylglycerol (DAG). DAG, on the other hand, is able to activate protein kinase C (PKC), which is associated with the disruption of the Na/K ATPase activity, a key protein for neuronal activity, and with the induction of endoplasmic reticulum (ER) stress [25], [26].
Moreover, the glycolysis intermediate F6P increases the synthesis of uridine diphosphate N-acetylglucosamine (UDP-GlcNAC) via the hexosamine pathway ([Fig. 2]). UDP-GlcNAC attaches either to transcription factors or to cytosolic proteins, thereby modifying gene expressions and protein functions linked to ER stress [27].
Glucose can also be metabolized to sorbitol via the polyol pathway. Sorbitol damages the cells due to its osmotic effect and inhibits the expression of Na/K ATPase by decreasing intracellular myoinositol [23]. Additionally, a depletion of NADPH leads to a reduction in glutathione (GSH), a major detoxification system of our cells, resulting in an increased susceptibility to oxidative stress [25].
Additionally, dyslipidemia plays a role in the development of PDN. Plasma lipoproteins, low-density lipoprotein (LDL) in particular, can undergo glycation or oxidation ([Fig. 2]) [21]. These modified molecules, like AGE-LDL and oxidized LDL, can bind to extracellular receptors such as oxidized LDL receptor 1 (LOX1), toll-like receptor 4 (TLR4), or RAGE, triggering a variety of signaling cascades, including the activation of caspase 3 and the DNA degradation, leading to apoptosis. It also leads to an increased NADPH oxidase activity, resulting in oxidative stress [28], [29].
Finally, insulin resistance interferes with the phosphoinositide 3-kinase/protein kinase B (PI3K-Akt) signaling pathway ([Fig. 2]). Insulin resistance leads to reduced phosphorylation of the insulin receptor substrate (IRS), thereby inhibiting downstream signaling cascades, including the activation of PI3K and ultimately Akt, which is associated with neurotrophic effects such as regeneration, survival, and axonal growth [12], [30].
Due to the complex pathophysiology and the limited understanding of the exact mechanistic interplay leading to PDN, current treatments remain inadequate [31]. In the absence of a disease-modifying treatment strategy, current guideline therapy is mainly restricted to the alleviation of pain symptoms [32]. First-line therapy options include anticonvulsants, tricyclic antidepressants, and serotonin-norepinephrine reuptake inhibitors (SNRI), but the exact mechanism-of-action in neuropathic pain is not yet fully elucidated [24], [33]. The most common therapeutic approach is the anticonvulsant pregabalin, which inhibits voltage-dependent calcium channels of presynaptic neurons in the CNS. This prevents the release of neurotransmitters like glutamate, noradrenaline, and substance P and the subsequent transmission of nociceptive stimuli [20], [33]. Moreover, a study hypothesized an effect on the PNS by inhibiting NF-κB activation in rat DRG pre-treated with substance P [34]. Tricyclic antidepressants and SNRI, on the other hand, influence the reuptake of neurotransmitters in the CNS [20]. However, these treatments are only effective to a limited extent, with only one-third of patients achieving significant pain relief [35]. Additionally, systemically applied therapies carry a high risk of adverse effects, especially in patients with polypharmacy like diabetic patients with increasing age [12]. In conclusion, there is a high unmet medical need for specialized therapeutic options [36].
Paeonia lactiflora (P. lactiflora) belongs to the family of Paeoniaceae and has been applied in traditional Chinese medicine (TCM) for centuries for the treatment of various disorders including pain symptoms [37]. In TCM, the dried root is used, which can be separated according to its processing method into Paeoniae radix alba ‘white peony root’ (baishao) and Paeoniae radix rubra ‘red peony root’ (chishao) [38]. Monographs of these drugs are not only listed in the Chinese but also the European Pharmacopoeia [39], [40].
Numerous pharmacological properties are attributed to the constituents of P. lactiflora that have an influence on the key factors affected in PDN: the nervous system, the vascular system, and the immune system. These characteristics include antinociceptive, anti-inflammatory, antioxidant, and antiapoptotic effects [41], [42]. Paeoniflorin, for example, the main constituent of P. lactiflora, is reported to show analgesic effects on different types of pain, including PDN. These effects are mediated via different pathways, amongst them the modulation of nociceptor excitability and the inhibition of inflammatory response [43]. Studies on Schwann cells also demonstrated that apoptosis induced by oxidative stress in the form of hydrogen peroxide is inhibited by paeoniflorin through inhibition of the phosphorylation of p38 mitogen-activated protein kinases (p38MAPK) and a reduction in the levels of caspase3, cleaved-caspase3, and cleaved-caspase7 [44]. On the other hand, paeonol presented pain-relieving activity in diabetic mice by enhancing antioxidant enzymes, thereby reducing oxidative stress [45]. Both paeonol and kaempferol additionally reduced neuroinflammation due to the modulation of microglia activation by switching M1 macrophages with a pro-inflammatory phenotype to M2 macrophages with an anti-inflammatory phenotype [46], [47].
The clinical relevance of P. lactiflora was demonstrated in a recently published meta-analysis that evaluated the efficacy and safety of herbal formulations from TCM with respect to the treatment of PDN [35]. Noteworthy, P. lactiflora turned out to be one of the most frequently used plants in prescriptions for PDN. For the herbal formulations in general, higher clinical efficacy and a lower rate of adverse effects compared to corresponding control groups with “classical” pharmaceuticals are reported [35].
Despite the given evidence for the use of P. lactiflora in PDN, to date, there is no review highlighting the potential of P. lactiflora as a substantial treatment strategy. Thus, this article provides a systematic insight into the existing research on the constituents of P. lactiflora and their pharmacological effects presented in various pain models, which also play a potential role in the treatment of PDN.
#
Search Strategy
We considered relevant articles from the following electronic databases until July 2024: PubMed, Springer, Web of Science, Google Scholar, Science Direct, and China National Knowledge Infrastructure. The search terms included “painful diabetic neuropathy”, “analgesic”, “antinociceptive”, “neuroprotective”, “clinical trial”, and “network pharmacology”, combined with the plant name or the names of main constituents. In silico data, as well as preclinical and clinical studies, were reviewed and analyzed.
#
Phytochemical Characteristic of Paeonia lactiflora
To date, approximately 300 compounds have been isolated and structurally identified from P. lactiflora, including monoterpenes and their glycosides, sesquiterpenes, triterpenes and steroid compounds, tannins, flavonoids, lignans, stilbenes, volatile oils, and other compounds [48], [49], [50]. Among them, monoterpenes and their glycosides are considered to be the predominant active components in P. lactiflora that have been shown to possess significant therapeutic effects in various nervous system diseases, including neuropathic pain, neuroinflammation, and neurotoxicity. As illustrated in the following chapters, the reported components from P. lactiflora exert pharmacological activities through multilateral mechanisms, such as antinociceptive, anti-inflammatory, antioxidant, and antiapoptotic activities [51], [52].
Monoterpenes and their glycosides
Symbol structures for the genus Paeonia and the basis for the plantʼs pharmacological effects are the pinane type and p-menthane type monoterpene glycosides. In particular, pinane-type monoterpene glycosides, such as the primary compounds paeoniflorin (1) and albiflorin (2), have been extensively studied and demonstrated to have neuroprotective, anti-inflammatory, antioxidant, and analgesic effects that play an important role in the treatment of PDN [51], [52]. Pinane-type monoterpenes are characterized by their “cage-like” pinane skeleton and exist as mono-cargo dibenzoate of monoterpene glycosides, which differ in the aromatic ring substitutions or sugar moieties and various stereocenters in the pinane skeleton, forming abundant varieties ([Fig. 3]) [49], [53]. For example, in contrast to paeoniflorin, the isomer albiflorin (2) exhibits a lactone ring in the pinane skeleton. Furthermore, oxypaeoniflorin, benzoylpaeoniflorin, benzoyloxypaeoniflorin, and galloylpaeoniflorin have similar structures to paeoniflorin but with different substituents. The other important type of monoterpene glycosides in P. lactiflora, the p-menthane type, includes paeonilactone A – C ([Fig. 3]) [54]. In addition, numerous novel types of monoterpene glycosides were isolated in recent years, such as nor-monoterpenes, labile monoterpenes, or dimeric monoterpenes [49].


#
Terpenes and steroids
In addition to monoterpenes, sesquiterpenes and triterpenes can also be isolated from P. lactiflora. Most triterpenes are pentacyclic triterpenoids, such as oleanolic acid, hederagenin, and betulinic acid ([Fig. 4]) [55]. Recently, three new 30-noroleanane triterpenoids paeonenoides L – N were isolated from the root section of P. lactiflora that show anti-inflammatory, antioxidant, and antidiabetic activities [56], [57]. Furthermore, the steroids palbinone, daucosterol, and β-sitosterol (3) were isolated from P. lactiflora ([Fig. 4]) [58], [59]. Many clinical roles of steroids are related to their potent anti-inflammatory and immune-modulating properties [60]. For example, palbinone suppresses glucose-induced retinal inflammation and oxidative stress in a diabetic rat model [61], while β-Sitosterol improves glycemic control in type 2 diabetic rats and protects against oxidative damages in diabetic mice [62].


#
Tannins
Recently, a study using high-resolution accurate-mass LC-MS instruments (UHPLC-Q-Exactive Orbitrap MS) identified 106 tannin constituents in the spectrum of the dried root of P. lactiflora [50]. Based on their structure, tannins can be divided into two types ([Fig. 5]), hydrolyzed tannins and condensed tannins, and most of them present anti-inflammatory and antioxidant activities [63]. Gallic acid and its derivatives, gallotannins and ellagitannins, such as ellagic acid and strictinin, comprise the main group of tannins in P. lactiflora [38]. Among them, a series of high molecular weight hydrolyzed tannins, including tetra-, penta-, hexa-, hepta-, octa-, nona-, and deca-galloylglucoses displaying a 1,2,3,4,6-penta-O-galloyl-β-D-glucose core show potent glucose-lowering activity [64], [65]. Moreover, many condensed tannins, such as (−)-epicatechin gallate, theaflavin-3-gallate, and theaflavin-3′-gallate were isolated from P. lactiflora [50]. Condensed tannins, also referred to as proanthocyanidins, are oligomers or polymers of flavan-3-ols, which are characterized by immunomodulatory, antidiabetic, and neuroprotective properties [66].


#
Flavonoids
Flavonoid extracts of P. lactiflora are known for their antioxidant, anti-inflammatory, anticancer, and antibacterial activities [67], [68]. They can be divided into several structural classes ([Fig. 6]), including the following: flavonols, such as kaempferol (4) and isoquercetin [69]; anthocyanidins, such as, for example, pelargonidin-3-glucoside and cyanidin-3-glucoside [70]; flavones, such as luteolin [71] and scutellarin methylester [69]; flavanone, including liquiritin apioside [72]; chalcone, such as chalcone-2′-O-glucoside [67]; and flavan-3-ol, such as, for example, catechin [73].


#
Lignans
Most of the lignans isolated from P. lactiflora display a benzofuran or tetrahydrofuran substructure ([Fig. 7]), such as machilusol C, rel-(2α,3β)-7-O-methylcedrusin, and rel-7S,8R,8′R-forsythialan C, while some representatives have a 1-phenyltetralin skeleton, such as, for example, isolariciresinol and (+)-lyoniresinol [48], and some belong to neolignans, such as (7S, 8S)-3′,4,7,9′-tetrahydroxy-3-methoxy-8-O-4′-neolignan [74]. Lignans exhibit diverse biological effects, including anti-inflammatory and antioxidant activities.


#
Stilbenes
Stilbenes are a group of specialized compounds with a C6-C2-C6 structure usually composed of two isomers. To date, less than 500 naturally occurring stilbenes have been isolated from plants and only a few from P. lactiflora. Stilbenes discovered in seeds of P. lactiflora represent resveratrol oligomers, including trans-resveratrol and its glycosides, trans-ε-viniferin, cis-ε-viniferin, gnetin H, suffruticosol A, and suffruticosol B ([Fig. 8]). For stilbenes, a variety of biological activities have been reported including neuroprotective, antidiabetic, antioxidant, and anti-inflammatory effects [75]. For example, suffruticosol A demonstrates anti-inflammatory activity by inhibiting the production of nitric oxide, as well as the expression of inducible nitric oxide synthase and pro-inflammatory cytokines in lipopolysaccharide (LPS)-stimulated macrophages [76].


#
Volatile oils and other compounds
More than 70 different types of volatile oils and other compounds are described and distributed in all parts of P. lactiflora including phenols, benzenoids, phenylpropanoids, alkyl hydrocarbons, fatty acid derivatives, coumarin, anthraquinone, and others [48], [77]. Some of them are the main constituents of the plantʼs fragrance [78], [79]. Paeonol (5) ([Fig. 9]) is an important bioactive phenolic compound of P. lactiflora. Despite its simple structure, it has great pharmacological potential with regard to the treatment of PDN including anti-inflammatory, antidiabetic, and neuroprotective effects [80], [81], [82].


#
#
In Silico Data
The network pharmacology approach reflects a systematic data analysis where networks of drug-target interactions and disease-target phenotypes are correlated, offering the opportunity of mapping active substances with pathophysiological pathways to uncover possible modes-of-action [83], [84].
Noteworthy, a recent study revealed potential mechanisms of the antinociceptive effect of P. lactiflora using in silico methods. In this study, 11 active constituents were identified that may exert analgesic effects, mainly via an inflammation-regulated transient receptor potential (TRP) channel pathway, including TRPV1, which is sensitive to heat pain, or the calcium signaling pathway, as well as the serotonin (5-HT) receptor [85]. The effect of the monoterpene glycoside albiflorin ([Fig. 3]) on 5-HT receptors has already been investigated in more detail. A high affinity of albiflorin to 5-HT and to norepinephrine receptors has been reported, hinting at a mechanism-of-action similar to that of antidepressants [86]. Furthermore, Hu et al. demonstrated that the steroid compound β-sitosterol ([Fig. 4]) could be one of the key active constituents acting on neuroinflammation and immune regulation by binding to the peroxisome proliferator-activated receptor gamma (PPARγ) and to TNF-α [87]. Another study assessed the mechanisms-of-action of the herb pair P. lactiflora and Ramulus cinnamomi (cassiae) in chronic pain with comorbid anxiety and depression. The main pathways involved were the AGE-RAGE axis and the TNF signaling pathway, which contribute to neuroinflammation [88]. In addition to peaoniflorin, albiflorin, palbinone, β-sitosterol, and kaemferol were identified as active compounds inhibiting neuropathic and inflammatory pain [85], [87], [88].
#
Preclinical Data
Neuropathic pain caused by diabetes mellitus
Diabetes and diabetic peripheral neuropathy can be induced in rats using streptozotocin, leading amongst other things to a decreased threshold for mechanical and thermal pain [89]. Adki et al. used this model to demonstrate that treatment with the phenolic constituent paeonol ([Fig. 9]) resulted in the suppression of mechanical allodynia and hyperalgesia, as well as thermal hyperalgesia and improved sensory nerve conduction velocity. Furthermore, they showed the antioxidant effect of paeonol by increasing the content of the antioxidant enzymes GSH, superoxide dismutase (SOD), and catalase (CAT) and the anti-inflammatory effect by decreasing NF-κB activity in the sciatic nerve. All these effects of paeonol had a comparable significance to the control treatment with pregabalin [45]. Similar effects have been shown for treatment with the flavonoid kaempferol ([Fig. 6]). Mechanical hyperalgesia and allodynia were alleviated in diabetic rats by modulating oxidative stress, especially by increasing GSH levels, and by reducing the formation of AGE, leading to decreased concentrations of the proinflammatory cytokines TNF-α and IL-1β [90].
#
Neuropathic pain caused by nerve injury
One of the most researched constituents of P. lactiflora is paeoniflorin, which also represents the major active monoterpene of the total glycoside of paeony (TGP) ([Fig. 3]). To verify the inflammatory mediator regulation of TRP channels found in the pharmacological network analysis [85], a study analyzed mRNA levels of different TRP channels, as well as the phosphorylation of p38MAPK (p-p38MAPK) in a rat model of chronic construction injury (CCI) in comparison to the positive control pregabalin. Expression levels of TRPA1, TRPV1, transient receptor potential vanilloid 4 (TRPV4), transient receptor potential melastatin 8 (TRPM8), and p-p38MAPK in rat DRG in combination with serum levels of pro-inflammatory cytokines, such as IL-6 and TNF-α, decreased in the presence of paeoniflorin, demonstrating the antinociceptive effect via the reduction in inflammatory factors by inhibiting the p38MAPK pathway. The antinociceptive effect was confirmed in behavioral tests, in which paeoniflorin attenuated mechanical pain and thermal pain [91].
Moreover, Zhou et al. also showed that paeoniflorin, as well as albiflorin ([Fig. 3]), could relieve neuropathic pain in a model of mechanical hyperalgesia induced by CCI in rats by inhibiting microglia activation in the CNS by reducing the activated p38MAPK signaling pathway. This led to reduced IL-1β and TNF-α levels, suggesting that the reported antinociceptive effect of paeoniflorin and albiflorin is mediated via the inhibition of neuroinflammation [92]. In this context, the role of the NOD-like receptor protein 3 (NLRP3) inflammasome and the influence of paeoniflorin were elucidated. Paeoniflorin was found to reduce the activation of the NLRP3 inflammasome in the spinal cord, which mediates the development of neuropathic pain. Paeoniflorin also suppressed NF-κB activity in the spinal cord, thereby inhibiting neuroinflammation [93]. Likewise, the isolated constituent albiflorin reduced the expression of NLRP3, the levels of IL-1β and ROS, and the activity of NF-κB, thereby alleviating pain [94].
The CCI model of neuropathic pain was additionally used to assess the analgesic effect of β-sitosterol ([Fig. 4]) with the non-steroidal anti-inflammatory drug ibuprofen as a positive control. In this study, β-sitosterol was able to relieve mechanical pain, presumably by decreasing TLR4 expression and NF-κB activity, resulting in reduced levels of the proinflammatory cytokines IL-1β and IL-8. [95]. Moreover, paeonol ([Fig. 9]) alleviated mechanical and thermal pain in the CCI model by reducing levels of TNF-α, IL-1β, and IL-6, associated with the inhibition of neuroinflammation [96].
In a mouse model for neuropathic pain and associated insomnia, where the sciatic nerve is partially ligated, paeoniflorin ([Fig. 3]) has relieved both mechanical and thermal pain and improved sleep. Since administration of an adenosine A1 receptor (A1R) antagonist abolished the antinociceptive and hypnotic effects, it was hypothesized that the efficacy of paeoniflorin may be mediated by A1Rs [97].
To investigate postoperative pain, mice underwent plantar incision surgery. The subsequent administration of paeoniflorin was able to alleviate the mechanical pain. In addition, paeoniflorin was able to inhibit microglial activation by reducing p-p38MAPK and by preventing the upregulation of IL-1β in the spinal cord. The reduction in neuronal cFOS expression, a marker of the activation of nociceptive neurons in the spinal cord, was also reduced by paeoniflorin [98].
#
Neuropathic pain caused by chemotherapy
Noteworthy, a study focusing on the paclitaxel-induced model of peripheral neuropathic pain in mice demonstrated that not only the oral but also the repeated topical application of paeoniflorin ([Fig. 3]) could attenuate mechanical pain [99]. Andoh et al. additionally reported that paeoniflorin downregulated the expression of the transcription factor C/EBP homologous protein (CHOP) in Schwann cells, which is a marker of endoplasmic reticulum stress. As this effect could be counteracted by an A1R antagonist, they similarly suggested A1R to be a potential mediator of the analgesic effects of paeoniflorin [100].
#
Inflammatory pain
Another study focused on the effect of paeoniflorin on complete Freundʼs adjuvant (CFA)-induced inflammatory pain in mice, where paeoniflorin could inhibit the spinal microglial activation and reduce NF-κB expression, leading to reduced production of the proinflammatory cytokines TNF-α, IL-6, and IL-1β in the CNS, both in vivo and in vitro. Behavioral tests also showed a relief of mechanical pain and thermal pain with paeoniflorin [101]. This is consistent with a study in which the DRG neurons of mice were examined in the CFA model. The authors hypothesized that the attenuation of inflammatory pain by paeoniflorin is partially due to the modulation of pyruvate and succinate dehydrogenase activity in the TCA cycle, resulting in a downstream inhibition of NLRP3 inflammasome expression. In addition, they found a reduction in serum IL-6, TNF-α, and IL-1β levels. Most importantly, they demonstrated that paeoniflorin could directly suppress the response of DRG neurons to capsaicin and reduce the release of substance P, a neurotransmitter relevant to pain perception [102].
Furthermore, paeoniflorin alleviated inflammatory pain in mice with LPS-induced pain by inhibiting the production of the proinflammatory cytokines IL-1β, TNF-α, and IL-6, as well as intracellular Ca2+ levels and PKC activity, in addition to NF-κB activation and TRPV1 expression [103].
A detailed summary of the pharmacological effects of the key constituents of P. lactiflora acting on PDN and other pain-related disorders of different pathological genesis is summarized in [Table 1].
Compound |
Pain Model |
Mechanisms |
References |
---|---|---|---|
Abbreviations: CCI: chronic constriction injury; A1R: adenosine A1 receptor; AGEs: advanced glycation end products; CAT: catalase; CFA: complete Freundʼs adjuvant; CHOP: C/EBP homologous protein; GSH: glutathione; IL-1β: interleukin-1 beta; IL-6: interleukin-6; IL-8: interleukin-8; LPS: lipopolysaccharide; MDA: malondialdehyde; NF-κB: nuclear factor kappa B; NLRP3: nucleotide-binding oligomerization domain (NOD)-like receptor (NLR) pyrin domain containing 3; p38 MAPK: p38 mitogen-activated protein kinase; PKC: protein kinase C; ROS: reactive oxygen species; SOD: superoxide dismutase; TLR4: toll-like receptor 4; TNF-α: tumor necrosis factor-α; TRPA1: transient receptor potenzial ankyrin 1; TRPM8: transient receptor potenzial melastatin 8; TRPV1: transient receptor potenzial vanilloid 1; TRPV4: transient receptor potenzial vanilloid 4 |
|||
Paeoniflorin (1) |
CCI neuropathic pain |
↓TRPM8, TRPA1, TRPV1, TRPV4, IL-6, TNF-α, p38 MAPK, IL-1β, NLRP3, NF-κB, cFOS |
|
LPS-induced or CFA-induced inflammatory pain |
↓IL-1β, TNF-α, IL-6, intracellular Ca2+, PKC, NF-κB, TRPV1, NLRP3, substance P |
||
Paclitaxel-induced neuropathic pain |
↓CHOP |
[100] |
|
Albiflorin (2) |
CCI neuropathic pain |
↓p38 MAPK, IL-1β, TNF-α, NLRP3, ROS, NF-κB |
|
β-Sitosterol (3) |
CCI neuropathic pain |
↓TLR4, NF-κB, IL-1β, IL-8 |
[95] |
Kaempferol (4) |
Diabetic neuropathic pain |
↓AGEs, TNF-α, IL-1β
|
[90] |
Paeonol (5) |
Diabetic neuropathic pain |
↓NF-κB, MDA |
[45] |
CCI neuropathic pain |
↓TNF-α, IL-1β, IL-6 |
[96] |
#
#
Clinical Data
To date, for the highlighted monomers isolated from P. lactiflora, mostly preclinical data are available. However, P. lactiflora extracts have been used for centuries as an important ingredient in TCM formulations for the treatment of PDN. Many classical TCM formulations involved P. lactiflora as one of the predominant compositions, such as the Shaoyao Gancao decoction [104], Buyang Huanwu decoction [105], Danggui Sini decoction [106], Shentong Zhuyu decoction [107], Huangqi Guizhi Wuwu decoction [108], Yiqi Huoxue Tongmai decoction [109], or Mudan granules [110]. Among them, the Shaoyao Gancao decoction, which originated in the Dong Han Dynasty (25 – 220 A. D.), is one of the most influential classical TCM formulations. It consists of only two ingredients, Paeoniae Radix Alba (poeny) and Glycyrrhizae Radix et Rhizoma Praeparata Cum Melle (licorice), with a ratio of 1 : 1. This formulation is widely used in Asian countries to treat various types of pain, including diabetic neuropathic pain [104]. Notably, in a recent literature review and meta-analysis, the effectiveness and safety of Chinese herbal medicine in the therapy of PDN were assessed in a total of 21 randomized controlled trials with 1737 PDN patients. Noteworthy, after analyzing all TCM formulations, P. lactiflora belonged to the top 10 (Paeoniae Radix Rubra ranked No. 4 and Paeoniae Radix Alba ranked No. 9) most frequently used herbal medicines in all formulations proven to enhance nerve conduction velocity, reduce pain, and promote clinical efficacy during the therapy of PDN [35].
Mudan granules are a patented TCM drug approved for diabetic peripheral neuropathy by the National Medical Products Administration in 2008 and listed in the National Health Insurance Catalogue as an urgently needed medicine in China [111]. The 2024 Expert Consensus of the China Association of Chinese Medicine summarizes the recent research and clinical effects of Mudan granules in treating diabetic peripheral neuropathy. This drug can significantly improve the syndrome of diabetic peripheral neuropathy, enhance peripheral nerve conduction velocity, and alleviate symptoms of peripheral sensory abnormalities in patients, including pain and numbness. Moreover, it can be used alone or in combination with other medicines to treat diabetic peripheral neuropathy. Therefore, Mudan granules were considered a highly recommended drug for the treatment of PDN [112].
The main components of Mudan granules are the roots of P. lactiflora (Paeoniae Radix Rubra), Astragalus membranacea, Panax notoginseng, Corydalis yanhusuo and five other medicinal herbs. In an ongoing clinical trial for Mudan granules that had been registered in the open science framework (OSF) in 2022, 93 PDN patients were recruited and randomly divided into a treatment group (Mudan granules combined with pregabalin) and a control group (placebo combined with pregabalin) to evaluate the efficacy and safety of Mudan granules in treating PDN [110]. Meanwhile, a post-marketing evaluation of Mudan granules as an intervention for type 2 diabetic peripheral neuropathy was initiated in 2021. This is a 14-center, double-blind, randomized, placebo-controlled, parallel-arm trial involving 402 people [113]. It is designed to evaluate the efficacy of Mudan granules in combination with methylcobalamin, an active form of vitamin B12 that has been proven in numerous clinical trials to alleviate the symptoms of peripheral diabetic neuropathy [114].
#
Summary and Conclusion
In general, the molecular interplay of oxidative stress and inflammation represents a major part in the pathogenesis of PDN, increasing the sensitivity of sensory neurons to nociceptive signals. Furthermore, the increased susceptibility to apoptosis is associated with a higher prevalence of the degeneration of sensory neurons. Overall, in silico and preclinical data, as well as clinical studies, strengthen the analgesic, anti-inflammatory, antioxidant, and antiapoptotic evidence of P. lactiflora extracts and its secondary metabolites as a great potential for the treatment of PDN.
Especially, the major monoterpene glycoside paeoniflorin exhibits antinociceptive effects in various pain models. This effect is mainly mediated by the suppression of inflammatory cytokines, by downregulating the expression of ion channels TRPM8, TRPA1, TRPV1, and TRPV4, relevant for the generation of pain signals, and by reducing the neurotransmitter substance P. Similarly, the monoterpene glycoside albiflorin exhibits analgesic properties by decreasing ROS and inflammatory signaling through the inhibition of the NLRP3 inflammasome and of the transcription factor NF-κB. Both, the phenolic compound paeonol and the flavonoid kaempferol alleviate painful diabetic neuropathy analogously by reducing neuroinflammation via the inhibition of NF-κB and the subsequent suppression of inflammatory cytokines. In addition, they increase a protective function against reactive oxygen species by enhancing antioxidant enzymes. Furthermore, the steroid β-sitosterol demonstrates anti-inflammatory properties by inhibiting the TLR4 signaling pathway and thereby relieving neuropathic pain.
Altogether, these synergistic effects of P. lactiflora metabolites allow a simultaneous influence on the key features of the complex pathophysiology of PDN including inflammation, oxidative stress, and hyperexcitability of neuronal cells, thereby offering the advantage of a holistic therapy. In line, the multi-target therapeutic approach used in TCM for centuries confirms a high efficacy and low toxicity in clinical studies, suggesting that P. lactiflora and its constituents might be a specific treatment option with a low rate of adverse effects. Furthermore, since the side effects of local medications are generally less than those of systemic medications, the topical antinociceptive effect of paeoniflorin might serve as a promising basis for the development of topical formulations of isolated, biologically active secondary metabolites in the future.
However, given the use of herbal formulations in TCM, one limitation in the assessment of the effects of P. lactiflora for the treatment of PDN is the small number of clinical studies focusing exclusively on P. lactiflora. Considering this lack of sufficient clinical evaluations, the need for randomized controlled clinical trials to confirm the promising effects has been identified.
In addition to the summarized secondary metabolites directly isolated from the plant, the next logical step is the investigation of endophytic fungi from P. lactiflora that may significantly impact its therapeutic potential. Endophytic fungi symbiotically inhabit plants without harming the host, and some fungi have developed pathways analogous to their host, synthesizing bioactive compounds that were originally associated with the plant [115]. This has already been demonstrated for taxol, a chemotherapeutic agent first isolated from the Pacific yew and later from the endophyte Taxomyces andreanae [116], suggesting that not only the individual plant P. lactiflora but also associated endophytic fungi might be valuable resources for the isolation of pharmacologically active compounds targeting PDN in the future.
#
Contributorsʼ Statement
Y.G. and V. W. carried out the literature search, data collection and interpretation, produced the first draft. N.T. was responsible for conceiving the topic, provided funding and supervision, as well as reviewed and edited the final draft. All authors approved the manuscript in its final form.
#
#
Conflict of Interest
The authors declare that they have no conflict of interest.
-
References
- 1 Sun H, Saeedi P, Karuranga S, Pinkepank M, Ogurtsova K, Duncan BB, Stein C, Basit A, Chan JC, Mbanya JC. IDF diabetes atlas: Global, regional and country-level diabetes prevalence estimates for 2021 and projections for 2045. Diabetes Res Clin Pract 2022; 183: 109119
- 2 Zheng Y, Ley SH, Hu FB. Global aetiology and epidemiology of type 2 diabetes mellitus and its complications. Nat Rev Endocrinol 2018; 14: 88-98
- 3 Zhu J, Hu Z, Luo Y, Liu Y, Luo W, Du X, Luo Z, Hu J, Peng S. Diabetic peripheral neuropathy: Pathogenetic mechanisms and treatment. Front Endocrinol (Lausanne) 2024; 14: 1265372
- 4 Lu Y, Xing P, Cai X, Luo D, Li R, Lloyd C, Sartorius N, Li M. Prevalence and risk factors for diabetic peripheral neuropathy in type 2 diabetic patients from 14 countries: Estimates of the INTERPRET-DD study. Front Public Health 2020; 8: 534372
- 5 Hicks CW, Selvin E. Epidemiology of peripheral neuropathy and lower extremity disease in diabetes. Curr Diab Rep 2019; 19: 1-8
- 6 Sloan G, Shillo P, Selvarajah D, Wu J, Wilkinson ID, Tracey I, Anand P, Tesfaye S. A new look at painful diabetic neuropathy. Diabetes Res Clin Pract 2018; 144: 177-191
- 7 Abdissa D. Prevalence and associated factors of painful diabetic peripheral neuropathy among diabetic patients on follow up at Jimma University Medical Center. J Diabetes Metab Disord 2020; 19: 1407-1413
- 8 Ziegler D, Landgraf R, Lobmann R, Reiners K, Rett K, Schnell O, Strom A. Painful and painless neuropathies are distinct and largely undiagnosed entities in subjects participating in an educational initiative (PROTECT study). Diabetes Res Clin Pract 2018; 139: 147-154
- 9 Alleman CJ, Westerhout KY, Hensen M, Chambers C, Stoker M, Long S, van Nooten FE. Humanistic and economic burden of painful diabetic peripheral neuropathy in Europe: A review of the literature. Diabetes Res Clin Pract 2015; 109: 215-225
- 10 Girach A, Julian TH, Varrassi G, Paladini A, Vadalouka A, Zis P. Quality of life in painful peripheral neuropathies: A systematic review. Pain Res Manag 2019; 2019: 2091960
- 11 Gylfadottir SS, Christensen DH, Nicolaisen SK, Andersen H, Callaghan BC, Itani M, Khan KS, Kristensen AG, Nielsen JS, Sindrup SH. Diabetic polyneuropathy and pain, prevalence, and patient characteristics: A cross-sectional questionnaire study of 5, 514 patients with recently diagnosed type 2 diabetes. Pain 2020; 161: 574-583
- 12 Sloan G, Selvarajah D, Tesfaye S. Pathogenesis, diagnosis and clinical management of diabetic sensorimotor peripheral neuropathy. Nat Rev Endocrinol 2021; 17: 400-420
- 13 Eid SA, Rumora AE, Beirowski B, Bennett DL, Hur J, Savelieff MG, Feldman EL. New perspectives in diabetic neuropathy. Neuron 2023; 111: 2623-2641
- 14 Lee Y, Lee CH, Oh U. Painful channels in sensory neurons. Mol Cells 2005; 20: 315-324
- 15 Wang Q, Ye Y, Yang L, Xiao L, Liu J, Zhang W, Du G. Painful diabetic neuropathy: The role of ion channels. Biomed Pharmacother 2024; 173: 116417
- 16 Joksimovic SL, Jevtovic-Todorovic V, Todorovic SM. The mechanisms of plasticity of nociceptive ion channels in painful diabetic neuropathy. Front Pain Res (Lausanne) 2022; 3: 869735
- 17 Niimi N, Yako H, Takaku S, Chung SK, Sango K. Aldose reductase and the polyol pathway in schwann cells: Old and new problems. Int J Mol Sci 2021; 22: 1031
- 18 Gonçalves NP, Vægter CB, Andersen H, Østergaard L, Calcutt NA, Jensen TS. Schwann cell interactions with axons and microvessels in diabetic neuropathy. Nat Rev Neurol 2017; 13: 135-147
- 19 Msheik Z, El Massry M, Rovini A, Billet F, Desmoulière A. The macrophage: A key player in the pathophysiology of peripheral neuropathies. J Neuroinflammation 2022; 19: 97
- 20 Yang K, Wang Y, Li YW, Chen YG, Xing N, Lin HB, Zhou P, Yu XP. Progress in the treatment of diabetic peripheral neuropathy. Biomed Pharmacother 2022; 148: 112717
- 21 Feldman EL, Callaghan BC, Pop-Busui R, Zochodne DW, Wright DE, Bennett DL, Bril V, Russell JW, Viswanathan V. Diabetic neuropathy. Nat Rev Dis Primers 2019; 5: 1-18
- 22 Rawat A, Morrison BM. Metabolic transporters in the peripheral Nerve–What, where, and why?. Neurotherapeutics 2021; 18: 2185-2199
- 23 Dewanjee S, Das S, Das AK, Bhattacharjee N, Dihingia A, Dua TK, Kalita J, Manna P. Molecular mechanism of diabetic neuropathy and its pharmacotherapeutic targets. Eur J Pharmacol 2018; 833: 472-523
- 24 Qureshi Z, Ali MN, Khalid M. An insight into potential pharmacotherapeutic agents for painful diabetic neuropathy. J Diabetes Res 2022; 2022: 9989272
- 25 Feldman EL, Nave KA, Jensen TS, Bennett DL. New horizons in diabetic neuropathy: mechanisms, bioenergetics, and pain. Neuron 2017; 93: 1296-1313
- 26 Kan YY, Chang YS, Liao WC, Chao TN, Hsieh YL. Roles of neuronal protein kinase Cε on endoplasmic reticulum stress and autophagic formation in diabetic neuropathy. Mol Neurobiol 2024; 61: 2481-2495
- 27 Mizukami H, Osonoi S. Collateral glucose-utlizing pathwaya in diabetic polyneuropathy. Int J Mol Sci 2020; 22: 94
- 28 Vincent AM, Hayes JM, McLean LL, Vivekanandan-Giri A, Pennathur S, Feldman EL. Dyslipidemia-induced neuropathy in mice: The role of oxLDL/LOX-1. Diabetes 2009; 58: 2376-2385
- 29 Vincent AM, Callaghan BC, Smith AL, Feldman EL. Diabetic neuropathy: Cellular mechanisms as therapeutic targets. Nat Rev Neurol 2011; 7: 573-583
- 30 Kim B, Feldman EL. Insulin resistance in the nervous system. Trends Endocrinol Metab 2012; 23: 133-141
- 31 Rastogi A, Jude E. Novel treatment modalities for painful diabetic neuropathy. Diabetes Metab Syndr 2021; 15: 287-293
- 32 Callaghan BC, Gallagher G, Fridman V, Feldman EL. Diabetic neuropathy: What does the future hold?. Diabetologia 2020; 63: 891-897
- 33 Frampton JE, Scott LJ. Pregabalin: In the treatment of painful diabetic peripheral neuropathy. Drugs 2004; 64: 2813-2820
- 34 Park S, Ahn ES, Han DW, Lee JH, Min KT, Kim H, Hong YW. Pregabalin and gabapentin inhibit substance P-induced NF-κB activation in neuroblastoma and glioma cells. J Cell Biochem 2008; 105: 414-423
- 35 Song M, Huai B, Shi Z, Li W, Xi Y, Liu Z, Zhang J, Zhou J, Qiao Y, Liu D. The efficacy and safety of Chinese herbal medicine in the treatment of painful diabetic neuropathy: A systematic review and meta-analysis. Front Pharmacol 2023; 14: 1072991
- 36 Papanas N, Ziegler D. Emerging drugs for diabetic peripheral neuropathy and neuropathic pain. Expert Opin Emerg Drugs 2016; 21: 393-407
- 37 He DY, Dai SM. Anti-inflammatory and immunomodulatory effects of Paeonia lactiflora Pall., a traditional Chinese herbal medicine. Front Pharmacol 2011; 2: 10
- 38 Parker S, May B, Zhang C, Zhang AL, Lu C, Xue CC. A pharmacological review of bioactive constituents of Paeonia lactiflora Pallas and Paeonia veitchii Lynch. Phytother Res 2016; 30: 1445-1473
- 39 European Pharmacopoeia Commission. European Pharmacopoeia, 11th Ed. Stuttgart: Deutscher Apotheker Verlag; 2022
- 40 Chinese Pharmacopoeia Commission. Chinese Pharmacopeia. Beijing: China Medical Science Press; 2020
- 41 Hong H, Lu X, Wu C, Chen J, Chen C, Zhang J, Huang C, Cui Z. A review for the pharmacological effects of paeoniflorin in the nervous system. Front Pharmacol 2022; 13: 898955
- 42 Tan YQ, Chen HW, Li J, Wu QJ. Efficacy, chemical constituents, and pharmacological actions of Radix Paeoniae Rubra and Radix Paeoniae Alba . Front Pharmacol 2020; 11: 1054
- 43 Li M, Zhu X, Zhang M, Yu J, Jin S, Hu X, Piao H. The analgesic effect of paeoniflorin: A focused review. Open Life Sci 2024; 19: 20220905
- 44 Zhang D, Bing Y, Chang SQ, Sheng-Suo M, Jian-Xin S, Lin Y, Xing L, Hui-Mei S, Bei J, Zheng YC. Protective effect of paeoniflorin on H2O2 induced Schwann cells injury based on network pharmacology and experimental validation. Chin J Nat Med 2021; 19: 90-99
- 45 Adki KM, Kulkarni YA. Neuroprotective effect of paeonol in streptozotocin-induced diabetes in rats. Life Sci 2021; 271: 119202
- 46 Chang S, Li X, Zheng Y, Shi H, Zhang D, Jing B, Chen Z, Qian G, Zhao G. Kaempferol exerts a neuroprotective effect to reduce neuropathic pain through TLR4/NF-ĸB signaling pathway. Phytother Res 2022; 36: 1678-1691
- 47 Li X, Shi H, Zhang D, Jing B, Chen Z, Zheng Y, Chang S, Gao L, Zhao G. Paeonol alleviates neuropathic pain by modulating microglial M1 and M2 polarization via the RhoA/p 38MAPK signaling pathway. CNS Neurosci Ther 2023; 29: 2666-2679
- 48 Fan Z, Liu J, Wang X, Yang S, Wang Q, Yan L, Zhang Y, Wu X. Paeoniae radix rubra: A review of ethnopharmacology, phytochemistry, pharmacological activities, therapeutic mechanism for blood stasis syndrome, and quality control. Chem Biodivers 2024; 21: e202401119
- 49 Li P, Shen J, Wang Z, Liu S, Liu Q, Li Y, He C, Xiao P. Genus Paeonia: A comprehensive review on traditional uses, phytochemistry, pharmacological activities, clinical application, and toxicology. J Ethnopharmacol 2021; 269: 113708
- 50 Xiong P, Qin SH, Li KL, Liu MJ, Zhu L, Peng J, Shi SL, Tang SN, Tian AP, Cai W. Identification of the tannins in traditional Chinese medicine Paeoniae Radix Alba by UHPLC-Q-Exactive Orbitrap MS. Arab J Chem 2021; 14: 103398
- 51 Xu SY, Cao HY, Yang RH, Xu RX, Zhu XY, Ma W, Liu XB, Yan XY, Fu P. Genus Paeonia monoterpene glycosides: A systematic review on their pharmacological activities and molecular mechanisms. Phytomedicine 2024; 127: 155483
- 52 Jiang H, Li J, Wang L, Wang S, Nie X, Chen Y, Fu Q, Jiang M, Fu C, He Y. Total glucosides of paeony: A review of its phytochemistry, role in autoimmune diseases, and mechanisms of action. J Ethnopharmacol 2020; 258: 112913
- 53 Aimi N, Inaba M, Watanabe M, Shibata S. Chemical studies on the oriental plant drugs–XXIII: Paeoniflorin, a glucoside of Chinese paeony root. Tetrahedron 1969; 25: 1825-1838
- 54 Hayashi T, Shinbo T, Shimizu M, Arisawa M, Morita N, Kimura M, Matsuda S, Kikuchi T. Paeonilactone-A, -B, and -C, new monoterpenoids from paeony root. Tetrahedron Lett 1985; 26: 3699-3702
- 55 Ikuta A, Kamiya K, Satake T, Saiki Y. Triterpenoids from callus tissue cultures of Paeonia species. Phytochemistry 1995; 38: 1203-1207
- 56 Xia XF, Wang LY, Xia GY, Xia H, Zhou LN, Li WT, Lin PC, Lin S. Oleanane and 30-noroleanane triterpenoids from the roots of Paeonia lactiflora . Fitoterapia 2024; 176: 105981
- 57 Del Prado-Audelo ML, Cortés H, Caballero-Florán IH, González-Torres M, Escutia-Guadarrama L, Bernal-Chávez SA, Giraldo-Gomez DM, Magaña JJ, Leyva-Gómez G. Therapeutic applications of terpenes on inflammatory diseases. Front Pharmacol 2021; 12: 704197
- 58 Kadota S, Terashima S, Kikuchi T, Namba T. Palbinone, a potent inhibitor of 3α-hydroxy dehydrogenase from Paeonia albiflora . Tetrahedron Lett 1992; 33: 255-256
- 59 Kadota S, Basnet P, Terashima S, Li JX, Namba T, Kageyu A. Palbinone, a novel terpenoid from Paeonia albiflora: A potent inhibitory activity on human monocyte interleukin-1β . Phytother Res 1995; 9: 379-381
- 60 Ericson-Neilsen W, Kaye AD. Steroids: pharmacology, complications, and practice delivery issues. Ochsner J 2014; 14: 203-207
- 61 Shi Q, Wang J, Cheng Y, Dong X, Zhang M, Pei C. Palbinone alleviates diabetic retinopathy in STZ-induced rats by inhibiting NLRP3 inflammatory activity. J Biochem Mol Toxicol 2020; 34: e22489
- 62 Khan Z, Nath N, Rauf A, Emran TB, Mitra S, Islam F, Chandran D, Barua J, Khandaker MU, Idris AM, Wilairatana P, Thiruvengadam M. Multifunctional roles and pharmacological potential of β-sitosterol: Emerging evidence toward clinical applications. Chem Biol Interact 2022; 365: 110117
- 63 Pizzi A. Tannins medical/pharmacological and related applications: A critical review. Sustain Chem Pharm 2021; 22: 100481
- 64 Nishizawa M, Yamagishi T, Nonaka G, Nishioka I, Nagasawa T, Oura H. Tannins and related compounds. XII. Isolation and characterization of galloylglucoses from Paeoniae Radix and their effect on urea-nitrogen concentration in rat serum. Chem Pharm Bull 1983; 31: 2593-2600
- 65 Juan YC, Chang CC, Tsai WJ, Lin YL, Hsu YS, Liu HK. Pharmacological evaluation of insulin mimetic novel suppressors of PEPCK gene transcription from Paeoniae Rubra Radix. J Ethnopharmacol 2011; 137: 592-600
- 66 Smeriglio A, Barreca D, Bellocco E, Trombetta D. Proanthocyanidins and hydrolysable tannins: Occurrence, dietary intake and pharmacological effects. Br J Pharmacol 2017; 174: 1244-1262
- 67 Lv M, Yang Y, Choisy P, Xu T, Pays K, Zhang L, Zhu J, Wang Q, Li S, Wang L. Flavonoid components and anti-photoaging activity of flower extracts from six Paeonia cultivars . Ind Crops Prod 2023; 200: 116707
- 68 Belwal T, Singh G, Jeandet P, Pandey A, Giri L, Ramola S, Bhatt ID, Venskutonis PR, Georgiev MI, Clément C, Luo Z. Anthocyanins, multi-functional natural products of industrial relevance: Recent biotechnological advances. Biotechnol Adv 2020; 43: 107600
- 69 Liu L, Yuan Y, Zuo J, Tao J. Composition and antioxidant activity of Paeonia lactiflora petal flavonoid extract and underlying mechanisms of the protective effect on H2O2-induced oxidative damage in BRL3A cells. Hortic Plant J 2023; 9: 335-344
- 70 Hosoki T, Seo M. Flower anthocyanins of herbaceous peony. Bull Fac Agr Shimane Univ 1991; 25: 11-14
- 71 Kim HJ, Chung SK, Park SW. Lipoxygenase inhibitors from Paeonia lactiflora seeds. Prev Nutr Food Sci 1999; 4: 163-166
- 72 Tanaka T, Zhang H, Jiang ZH, Kouno I. Relationship between hydrophobicity and structure of hydrolyzable tannins, and association of tannins with crude drug constituents in aqueous solution. Chem Pharm Bull 1997; 45: 1891-1897
- 73 Ulubelen A, Cetin ET, Isildatici S, Öztürk S. Phytochemical investigation of Paeonia decora . Lloydia 1968; 31: 249-251
- 74 Liu X, Yang MH, Wang XB, Xie SS, Li ZR, Kim DH, Park JS, Kong LY. Lignans from the root of Paeonia lactiflora and their anti-β-amyloid aggregation activities. Fitoterapia 2015; 103: 136-142
- 75 Teka T, Zhang L, Ge X, Li Y, Han L, Yan X. Stilbenes: Source plants, chemistry, biosynthesis, pharmacology, application and problems related to their clinical Application-A comprehensive review. Phytochemistry 2022; 197: 113128
- 76 Ryu HW, Song HH, Shin IS, Cho BO, Jeong SH, Kim DY, Ahn KS, Oh SR. Suffruticosol A isolated from Paeonia lactiflora seedcases attenuates airway inflammation in mice induced by cigarette smoke and LPS exposure. J Funct Foods 2015; 17: 774-784
- 77 Nie R, Zhang Y, Jin Q, Zhang S, Wu G, Chen L, Zhang H, Wang X. Identification and characterisation of bioactive compounds from the seed kernels and hulls of Paeonia lactiflora Pall by UPLC-QTOF-MS. Food Res Int 2021; 139: 109916
- 78 Zhao Q, Gu L, Li Y, Zhi H, Luo J, Zhang Y. Volatile composition and classification of Paeonia lactiflora flower aroma types and identification of the fragrance-related genes. Int J Mol Sci 2023; 24: 9410
- 79 Wang T, Xie A, Zhang D, Zemiao L, Li X, Li Y, Sun X. Analysis of the volatile components in flowers of Paeonia lactiflora Pall. and Paeonia lactiflora Pall. var. Trichocarpa . Am J Plant Sci 2021; 12: 146-162
- 80 Zhang L, Li DC, Liu LF. Paeonol: Pharmacological effects and mechanisms of action. Int Immunopharmacol 2019; 72: 413-421
- 81 Adki KM, Kulkarni YA. Chemistry, pharmacokinetics, pharmacology and recent novel drug delivery systems of paeonol. Life Sci 2020; 250: 117544
- 82 Wu R, Liu Y, Zhang F, Dai S, Xue X, Peng C, Li Y, Li Y. Protective mechanism of Paeonol on central nervous system. Phytother Res 2024; 38: 470-488
- 83 Hopkins AL. Network pharmacology: The next paradigm in drug discovery. Nat Chem Biol 2008; 4: 682-690
- 84 Guney E, Menche J, Vidal M, Barábasi AL. Network-based in silico drug efficacy screening. Nat Commun 2016; 7: 10331
- 85 Di Z, Ma S, Sun J. Mechanisms involved in antineuralgic effects of Paeonia Lactiflora: Prediction based on network pharmacology. TMR Clinical Research 2019; 2: 43-56
- 86 Jin ZL, Gao N, Xu W, Xu P, Li S, Zheng YY, Xue M. Receptor and transporter binding and activity profiles of albiflorin extracted from Radix paeoniae Alba. Sci Rep 2016; 6: 33793
- 87 Hu F, Lin J, Xiong L, Li Z, Liu WK, Zheng YJ. Exploring the molecular mechanism of Xuebifang in the treatment of diabetic peripheral neuropathy based on bioinformatics and network pharmacology. Front Endocrinol (Lausanne) 2024; 15: 1275816
- 88 Pan HT, Xi ZQ, Wei XQ, Wang K. A network pharmacology approach to predict potential targets and mechanisms of “Ramulus Cinnamomi (cassiae)–Paeonia lactiflora” herb pair in the treatment of chronic pain with comorbid anxiety and depression. Ann Med 2022; 54: 413-425
- 89 Bishnoi M, Bosgraaf CA, Abooj M, Zhong L, Premkumar LS. Streptozotocin-induced early thermal hyperalgesia is independent of glycemic state of rats: Role of transient receptor potential vanilloid 1 (TRPV1) and inflammatory mediators. Mol Pain 2011; 7: 52
- 90 Kishore L, Kaur N, Singh R. Effect of Kaempferol isolated from seeds of Eruca sativa on changes of pain sensitivity in Streptozotocin-induced diabetic neuropathy. Inflammopharmacology 2018; 26: 993-1003
- 91 Zhang D, Jing B, Li X, Shi H, Chen Z, Chang S, Zheng Y, Lin Y, Pan Y, Sun J. Antihyperalgesic effect of Paeniflorin based on chronic constriction injury in rats. Rev Bras Farmacogn 2022; 32: 375-385
- 92 Zhou J, Wang L, Wang J, Wang C, Yang Z, Wang C, Zhu Y, Zhang J. Paeoniflorin and albiflorin attenuate neuropathic pain via MAPK pathway in chronic constriction injury rats. Evid Based Complement Alternat Med 2016; 2016: 8082753
- 93 Liu P, Cheng J, Ma S, Zhou J. Paeoniflorin attenuates chronic constriction injury-induced neuropathic pain by suppressing spinal NLRP3 inflammasome activation. Inflammopharmacology 2020; 28: 1495-1508
- 94 Liu P, Chen J, Ma S, Zhang J, Zhou J. Albiflorin attenuates mood disorders under neuropathic pain state by suppressing the hippocampal NLRP3 inflammasome activation during chronic constriction injury. Int J Neuropsychopharmacol 2021; 24: 64-76
- 95 Zheng Y, Zhao J, Chang S, Zhuang Z, Waimei S, Li X, Chen Z, Jing B, Zhang D, Zhao G. β-sitosterol alleviates neuropathic pain by affect microglia polarization through inhibiting TLR4/NF-κB signaling pathway. J Neuroimmune Pharmacol 2023; 18: 690-703
- 96 Cai L, Zeng R, Huang Q, Liu X, Cao Z, Guo Q. Paeonol inhibits chronic constriction injury-induced astrocytic activation and neuroinflammation in rats via the HDAC/miR-15a pathway. Drug Dev Res 2022; 83: 1758-1765
- 97 Yin D, Liu YY, Wang TX, Hu ZZ, Qu WM, Chen JF, Cheng NN, Huang ZL. Paeoniflorin exerts analgesic and hypnotic effects via adenosine A 1 receptors in a mouse neuropathic pain model. Psychopharmacology (Berl) 2016; 233: 281-293
- 98 Fan YX, Hu L, Zhu SH, Han Y, Liu WT, Yang YJ, Li QP. Paeoniflorin attenuates postoperative pain by suppressing matrix Metalloproteinase-9/2 in mice. Eur J Pain 2018; 22: 272-281
- 99 Andoh T, Kurokawa Y, Kato M, Juntado ME. Local preventive effects of shakuyakukanzoto and paeoniflorin external gel on paclitaxel-induced peripheral neuropathic pain in mice. Tradit Kampo Med 2022; 9: 186-191
- 100 Andoh T, Kobayashi N, Uta D, Kuraishi Y. Prophylactic topical paeoniflorin prevents mechanical allodynia caused by paclitaxel in mice through adenosine A1 receptors. Phytomedicine 2017; 25: 1-7
- 101 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
- 102 Ruan Y, Ling J, Ye F, Cheng N, Wu F, Tang Z, Cheng X, Liu H. Paeoniflorin alleviates CFA-induced inflammatory pain by inhibiting TRPV1 and succinate/SUCNR1-HIF-1α/NLPR3 pathway. Int Immunopharmacol 2021; 101: 108364
- 103 Yin N, Gao Q, Tao W, Chen J, Bi J, Ding F, Wang Z. Paeoniflorin relieves LPS-induced inflammatory pain in mice by inhibiting NLRP3 inflammasome activation via transient receptor potential vanilloid 1. J Leukoc Biol 2020; 108: 229-241
- 104 Feng LM, Chen YY, Xu DQ, Fu RJ, Yue SJ, Zhao Q, Huang YX, Bai X, Wang M, Xing LM, Tang YP, Duan JA. An integrated strategy for discovering effective components of Shaoyao Gancao decoction for treating neuropathic pain by the combination of partial least-squares regression and multi-index comprehensive method. J Ethnopharmacol 2020; 260: 113050
- 105 Meizhen Z, Xiaohui H, Yiting T, Yupeng C, Puyu HE, Liming Z, Bing P, Qing NI. Efficacy and safety of Buyang Huanwu decoction for diabetic peripheral neuropathy: A systematic review and metaanalysis. J Tradit Chin Med 2023; 43: 841-850
- 106 Wang Y, Sheng C. Clinical research of modified Danggui-Sini decoction combined with mecobalamin in treatment of diabetic peripheral neuropathy. Int J Trad Chin Med 2017; 11: 981-984
- 107 Wang C, Lin J, Xie H, Chen L, Chen P, Wu L, Gong Q, Xia D, Wang X. Study on analgesic effect of Shentong Zhuyu Decoction in neuropathic pain rats by network pharmacology and RNA-Seq. J Ethnopharmacol 2024; 330: 118189
- 108 Pang B, Zhao TY, Zhao LH, Wan F, Ye R, Zhou Q, Tian F, Tong XL. Huangqi Guizhi Wuwu Decoction for treating diabetic peripheral neuropathy: A meta-analysis of 16 randomized controlled trials. Neural Regen Res 2016; 11: 1347-1358
- 109 Zhan G, Zheng Y, Li D, Zhang H. Clinical effect of Yiqi Huoxue Tongmai decoction in the treatment of diabetic peripheral neuropathy. Pak J Zool 2024; 56: 1-6
- 110 Zhang A, Wang Q, Liu M, Tan M, Zhang X, Wu R. Efficacy and safety of Mudan granules for painful diabetic peripheral neuropathy: A protocol for a double-blind randomized controlled trial. Medicine (Baltimore) 2022; 101: e28896
- 111 Zhang Y, Wu X, Yao W, Ni Y, Ding X. Advances of traditional Chinese medicine preclinical mechanisms and clinical studies on diabetic peripheral neuropathy. Pharm Biol 2024; 62: 544-561
- 112 China Association of Chinese Medicine. Expert consensus on the clinical application of Mudan granules in the treatment of diabetic peripheral neuropathy. Accessed July 04, 2024 at: http://www.cacm.org.cn
- 113 Zhang Y, Jin D, Duan Y, Hao R, Chen K, Yu T, Lian F, Tong X. Efficacy of Mudan Granule (combined with methylcobalamin) on type 2 diabetic peripheral neuropathy: study protocol for a double-blind, randomized, placebo-controlled, parallel-arm, multi-center trial. Front Pharmacol 2021; 12: 676503
- 114 Sawangjit R, Thongphui S, Chaichompu W, Phumart P. Efficacy and safety of mecobalamin on peripheral neuropathy: A systematic review and meta-analysis of randomized controlled trials. J Altern Complement Med 2020; 26: 1117-1129
- 115 Cheng X, Wei Z, Pu S, Xiang M, Yan A, Zhang Y, Wang X. Diversity of endophytic fungi of Paeonia Lactiflora Pallas and screening for fungal paeoniflorin producers. FEMS Microbiol Lett 2018; 365: fny263
- 116 Stierle A, Strobel G, Stierle D. Taxol and taxane production by Taxomyces andreanae, an endophytic fungus of Pacific yew. Science 1993; 260: 214-216
Correspondence
Publication History
Received: 25 July 2024
Accepted after revision: 03 October 2024
Article published online:
29 October 2024
© 2024. Thieme. All rights reserved.
Georg Thieme Verlag KG
Rüdigerstraße 14, 70469 Stuttgart, Germany
-
References
- 1 Sun H, Saeedi P, Karuranga S, Pinkepank M, Ogurtsova K, Duncan BB, Stein C, Basit A, Chan JC, Mbanya JC. IDF diabetes atlas: Global, regional and country-level diabetes prevalence estimates for 2021 and projections for 2045. Diabetes Res Clin Pract 2022; 183: 109119
- 2 Zheng Y, Ley SH, Hu FB. Global aetiology and epidemiology of type 2 diabetes mellitus and its complications. Nat Rev Endocrinol 2018; 14: 88-98
- 3 Zhu J, Hu Z, Luo Y, Liu Y, Luo W, Du X, Luo Z, Hu J, Peng S. Diabetic peripheral neuropathy: Pathogenetic mechanisms and treatment. Front Endocrinol (Lausanne) 2024; 14: 1265372
- 4 Lu Y, Xing P, Cai X, Luo D, Li R, Lloyd C, Sartorius N, Li M. Prevalence and risk factors for diabetic peripheral neuropathy in type 2 diabetic patients from 14 countries: Estimates of the INTERPRET-DD study. Front Public Health 2020; 8: 534372
- 5 Hicks CW, Selvin E. Epidemiology of peripheral neuropathy and lower extremity disease in diabetes. Curr Diab Rep 2019; 19: 1-8
- 6 Sloan G, Shillo P, Selvarajah D, Wu J, Wilkinson ID, Tracey I, Anand P, Tesfaye S. A new look at painful diabetic neuropathy. Diabetes Res Clin Pract 2018; 144: 177-191
- 7 Abdissa D. Prevalence and associated factors of painful diabetic peripheral neuropathy among diabetic patients on follow up at Jimma University Medical Center. J Diabetes Metab Disord 2020; 19: 1407-1413
- 8 Ziegler D, Landgraf R, Lobmann R, Reiners K, Rett K, Schnell O, Strom A. Painful and painless neuropathies are distinct and largely undiagnosed entities in subjects participating in an educational initiative (PROTECT study). Diabetes Res Clin Pract 2018; 139: 147-154
- 9 Alleman CJ, Westerhout KY, Hensen M, Chambers C, Stoker M, Long S, van Nooten FE. Humanistic and economic burden of painful diabetic peripheral neuropathy in Europe: A review of the literature. Diabetes Res Clin Pract 2015; 109: 215-225
- 10 Girach A, Julian TH, Varrassi G, Paladini A, Vadalouka A, Zis P. Quality of life in painful peripheral neuropathies: A systematic review. Pain Res Manag 2019; 2019: 2091960
- 11 Gylfadottir SS, Christensen DH, Nicolaisen SK, Andersen H, Callaghan BC, Itani M, Khan KS, Kristensen AG, Nielsen JS, Sindrup SH. Diabetic polyneuropathy and pain, prevalence, and patient characteristics: A cross-sectional questionnaire study of 5, 514 patients with recently diagnosed type 2 diabetes. Pain 2020; 161: 574-583
- 12 Sloan G, Selvarajah D, Tesfaye S. Pathogenesis, diagnosis and clinical management of diabetic sensorimotor peripheral neuropathy. Nat Rev Endocrinol 2021; 17: 400-420
- 13 Eid SA, Rumora AE, Beirowski B, Bennett DL, Hur J, Savelieff MG, Feldman EL. New perspectives in diabetic neuropathy. Neuron 2023; 111: 2623-2641
- 14 Lee Y, Lee CH, Oh U. Painful channels in sensory neurons. Mol Cells 2005; 20: 315-324
- 15 Wang Q, Ye Y, Yang L, Xiao L, Liu J, Zhang W, Du G. Painful diabetic neuropathy: The role of ion channels. Biomed Pharmacother 2024; 173: 116417
- 16 Joksimovic SL, Jevtovic-Todorovic V, Todorovic SM. The mechanisms of plasticity of nociceptive ion channels in painful diabetic neuropathy. Front Pain Res (Lausanne) 2022; 3: 869735
- 17 Niimi N, Yako H, Takaku S, Chung SK, Sango K. Aldose reductase and the polyol pathway in schwann cells: Old and new problems. Int J Mol Sci 2021; 22: 1031
- 18 Gonçalves NP, Vægter CB, Andersen H, Østergaard L, Calcutt NA, Jensen TS. Schwann cell interactions with axons and microvessels in diabetic neuropathy. Nat Rev Neurol 2017; 13: 135-147
- 19 Msheik Z, El Massry M, Rovini A, Billet F, Desmoulière A. The macrophage: A key player in the pathophysiology of peripheral neuropathies. J Neuroinflammation 2022; 19: 97
- 20 Yang K, Wang Y, Li YW, Chen YG, Xing N, Lin HB, Zhou P, Yu XP. Progress in the treatment of diabetic peripheral neuropathy. Biomed Pharmacother 2022; 148: 112717
- 21 Feldman EL, Callaghan BC, Pop-Busui R, Zochodne DW, Wright DE, Bennett DL, Bril V, Russell JW, Viswanathan V. Diabetic neuropathy. Nat Rev Dis Primers 2019; 5: 1-18
- 22 Rawat A, Morrison BM. Metabolic transporters in the peripheral Nerve–What, where, and why?. Neurotherapeutics 2021; 18: 2185-2199
- 23 Dewanjee S, Das S, Das AK, Bhattacharjee N, Dihingia A, Dua TK, Kalita J, Manna P. Molecular mechanism of diabetic neuropathy and its pharmacotherapeutic targets. Eur J Pharmacol 2018; 833: 472-523
- 24 Qureshi Z, Ali MN, Khalid M. An insight into potential pharmacotherapeutic agents for painful diabetic neuropathy. J Diabetes Res 2022; 2022: 9989272
- 25 Feldman EL, Nave KA, Jensen TS, Bennett DL. New horizons in diabetic neuropathy: mechanisms, bioenergetics, and pain. Neuron 2017; 93: 1296-1313
- 26 Kan YY, Chang YS, Liao WC, Chao TN, Hsieh YL. Roles of neuronal protein kinase Cε on endoplasmic reticulum stress and autophagic formation in diabetic neuropathy. Mol Neurobiol 2024; 61: 2481-2495
- 27 Mizukami H, Osonoi S. Collateral glucose-utlizing pathwaya in diabetic polyneuropathy. Int J Mol Sci 2020; 22: 94
- 28 Vincent AM, Hayes JM, McLean LL, Vivekanandan-Giri A, Pennathur S, Feldman EL. Dyslipidemia-induced neuropathy in mice: The role of oxLDL/LOX-1. Diabetes 2009; 58: 2376-2385
- 29 Vincent AM, Callaghan BC, Smith AL, Feldman EL. Diabetic neuropathy: Cellular mechanisms as therapeutic targets. Nat Rev Neurol 2011; 7: 573-583
- 30 Kim B, Feldman EL. Insulin resistance in the nervous system. Trends Endocrinol Metab 2012; 23: 133-141
- 31 Rastogi A, Jude E. Novel treatment modalities for painful diabetic neuropathy. Diabetes Metab Syndr 2021; 15: 287-293
- 32 Callaghan BC, Gallagher G, Fridman V, Feldman EL. Diabetic neuropathy: What does the future hold?. Diabetologia 2020; 63: 891-897
- 33 Frampton JE, Scott LJ. Pregabalin: In the treatment of painful diabetic peripheral neuropathy. Drugs 2004; 64: 2813-2820
- 34 Park S, Ahn ES, Han DW, Lee JH, Min KT, Kim H, Hong YW. Pregabalin and gabapentin inhibit substance P-induced NF-κB activation in neuroblastoma and glioma cells. J Cell Biochem 2008; 105: 414-423
- 35 Song M, Huai B, Shi Z, Li W, Xi Y, Liu Z, Zhang J, Zhou J, Qiao Y, Liu D. The efficacy and safety of Chinese herbal medicine in the treatment of painful diabetic neuropathy: A systematic review and meta-analysis. Front Pharmacol 2023; 14: 1072991
- 36 Papanas N, Ziegler D. Emerging drugs for diabetic peripheral neuropathy and neuropathic pain. Expert Opin Emerg Drugs 2016; 21: 393-407
- 37 He DY, Dai SM. Anti-inflammatory and immunomodulatory effects of Paeonia lactiflora Pall., a traditional Chinese herbal medicine. Front Pharmacol 2011; 2: 10
- 38 Parker S, May B, Zhang C, Zhang AL, Lu C, Xue CC. A pharmacological review of bioactive constituents of Paeonia lactiflora Pallas and Paeonia veitchii Lynch. Phytother Res 2016; 30: 1445-1473
- 39 European Pharmacopoeia Commission. European Pharmacopoeia, 11th Ed. Stuttgart: Deutscher Apotheker Verlag; 2022
- 40 Chinese Pharmacopoeia Commission. Chinese Pharmacopeia. Beijing: China Medical Science Press; 2020
- 41 Hong H, Lu X, Wu C, Chen J, Chen C, Zhang J, Huang C, Cui Z. A review for the pharmacological effects of paeoniflorin in the nervous system. Front Pharmacol 2022; 13: 898955
- 42 Tan YQ, Chen HW, Li J, Wu QJ. Efficacy, chemical constituents, and pharmacological actions of Radix Paeoniae Rubra and Radix Paeoniae Alba . Front Pharmacol 2020; 11: 1054
- 43 Li M, Zhu X, Zhang M, Yu J, Jin S, Hu X, Piao H. The analgesic effect of paeoniflorin: A focused review. Open Life Sci 2024; 19: 20220905
- 44 Zhang D, Bing Y, Chang SQ, Sheng-Suo M, Jian-Xin S, Lin Y, Xing L, Hui-Mei S, Bei J, Zheng YC. Protective effect of paeoniflorin on H2O2 induced Schwann cells injury based on network pharmacology and experimental validation. Chin J Nat Med 2021; 19: 90-99
- 45 Adki KM, Kulkarni YA. Neuroprotective effect of paeonol in streptozotocin-induced diabetes in rats. Life Sci 2021; 271: 119202
- 46 Chang S, Li X, Zheng Y, Shi H, Zhang D, Jing B, Chen Z, Qian G, Zhao G. Kaempferol exerts a neuroprotective effect to reduce neuropathic pain through TLR4/NF-ĸB signaling pathway. Phytother Res 2022; 36: 1678-1691
- 47 Li X, Shi H, Zhang D, Jing B, Chen Z, Zheng Y, Chang S, Gao L, Zhao G. Paeonol alleviates neuropathic pain by modulating microglial M1 and M2 polarization via the RhoA/p 38MAPK signaling pathway. CNS Neurosci Ther 2023; 29: 2666-2679
- 48 Fan Z, Liu J, Wang X, Yang S, Wang Q, Yan L, Zhang Y, Wu X. Paeoniae radix rubra: A review of ethnopharmacology, phytochemistry, pharmacological activities, therapeutic mechanism for blood stasis syndrome, and quality control. Chem Biodivers 2024; 21: e202401119
- 49 Li P, Shen J, Wang Z, Liu S, Liu Q, Li Y, He C, Xiao P. Genus Paeonia: A comprehensive review on traditional uses, phytochemistry, pharmacological activities, clinical application, and toxicology. J Ethnopharmacol 2021; 269: 113708
- 50 Xiong P, Qin SH, Li KL, Liu MJ, Zhu L, Peng J, Shi SL, Tang SN, Tian AP, Cai W. Identification of the tannins in traditional Chinese medicine Paeoniae Radix Alba by UHPLC-Q-Exactive Orbitrap MS. Arab J Chem 2021; 14: 103398
- 51 Xu SY, Cao HY, Yang RH, Xu RX, Zhu XY, Ma W, Liu XB, Yan XY, Fu P. Genus Paeonia monoterpene glycosides: A systematic review on their pharmacological activities and molecular mechanisms. Phytomedicine 2024; 127: 155483
- 52 Jiang H, Li J, Wang L, Wang S, Nie X, Chen Y, Fu Q, Jiang M, Fu C, He Y. Total glucosides of paeony: A review of its phytochemistry, role in autoimmune diseases, and mechanisms of action. J Ethnopharmacol 2020; 258: 112913
- 53 Aimi N, Inaba M, Watanabe M, Shibata S. Chemical studies on the oriental plant drugs–XXIII: Paeoniflorin, a glucoside of Chinese paeony root. Tetrahedron 1969; 25: 1825-1838
- 54 Hayashi T, Shinbo T, Shimizu M, Arisawa M, Morita N, Kimura M, Matsuda S, Kikuchi T. Paeonilactone-A, -B, and -C, new monoterpenoids from paeony root. Tetrahedron Lett 1985; 26: 3699-3702
- 55 Ikuta A, Kamiya K, Satake T, Saiki Y. Triterpenoids from callus tissue cultures of Paeonia species. Phytochemistry 1995; 38: 1203-1207
- 56 Xia XF, Wang LY, Xia GY, Xia H, Zhou LN, Li WT, Lin PC, Lin S. Oleanane and 30-noroleanane triterpenoids from the roots of Paeonia lactiflora . Fitoterapia 2024; 176: 105981
- 57 Del Prado-Audelo ML, Cortés H, Caballero-Florán IH, González-Torres M, Escutia-Guadarrama L, Bernal-Chávez SA, Giraldo-Gomez DM, Magaña JJ, Leyva-Gómez G. Therapeutic applications of terpenes on inflammatory diseases. Front Pharmacol 2021; 12: 704197
- 58 Kadota S, Terashima S, Kikuchi T, Namba T. Palbinone, a potent inhibitor of 3α-hydroxy dehydrogenase from Paeonia albiflora . Tetrahedron Lett 1992; 33: 255-256
- 59 Kadota S, Basnet P, Terashima S, Li JX, Namba T, Kageyu A. Palbinone, a novel terpenoid from Paeonia albiflora: A potent inhibitory activity on human monocyte interleukin-1β . Phytother Res 1995; 9: 379-381
- 60 Ericson-Neilsen W, Kaye AD. Steroids: pharmacology, complications, and practice delivery issues. Ochsner J 2014; 14: 203-207
- 61 Shi Q, Wang J, Cheng Y, Dong X, Zhang M, Pei C. Palbinone alleviates diabetic retinopathy in STZ-induced rats by inhibiting NLRP3 inflammatory activity. J Biochem Mol Toxicol 2020; 34: e22489
- 62 Khan Z, Nath N, Rauf A, Emran TB, Mitra S, Islam F, Chandran D, Barua J, Khandaker MU, Idris AM, Wilairatana P, Thiruvengadam M. Multifunctional roles and pharmacological potential of β-sitosterol: Emerging evidence toward clinical applications. Chem Biol Interact 2022; 365: 110117
- 63 Pizzi A. Tannins medical/pharmacological and related applications: A critical review. Sustain Chem Pharm 2021; 22: 100481
- 64 Nishizawa M, Yamagishi T, Nonaka G, Nishioka I, Nagasawa T, Oura H. Tannins and related compounds. XII. Isolation and characterization of galloylglucoses from Paeoniae Radix and their effect on urea-nitrogen concentration in rat serum. Chem Pharm Bull 1983; 31: 2593-2600
- 65 Juan YC, Chang CC, Tsai WJ, Lin YL, Hsu YS, Liu HK. Pharmacological evaluation of insulin mimetic novel suppressors of PEPCK gene transcription from Paeoniae Rubra Radix. J Ethnopharmacol 2011; 137: 592-600
- 66 Smeriglio A, Barreca D, Bellocco E, Trombetta D. Proanthocyanidins and hydrolysable tannins: Occurrence, dietary intake and pharmacological effects. Br J Pharmacol 2017; 174: 1244-1262
- 67 Lv M, Yang Y, Choisy P, Xu T, Pays K, Zhang L, Zhu J, Wang Q, Li S, Wang L. Flavonoid components and anti-photoaging activity of flower extracts from six Paeonia cultivars . Ind Crops Prod 2023; 200: 116707
- 68 Belwal T, Singh G, Jeandet P, Pandey A, Giri L, Ramola S, Bhatt ID, Venskutonis PR, Georgiev MI, Clément C, Luo Z. Anthocyanins, multi-functional natural products of industrial relevance: Recent biotechnological advances. Biotechnol Adv 2020; 43: 107600
- 69 Liu L, Yuan Y, Zuo J, Tao J. Composition and antioxidant activity of Paeonia lactiflora petal flavonoid extract and underlying mechanisms of the protective effect on H2O2-induced oxidative damage in BRL3A cells. Hortic Plant J 2023; 9: 335-344
- 70 Hosoki T, Seo M. Flower anthocyanins of herbaceous peony. Bull Fac Agr Shimane Univ 1991; 25: 11-14
- 71 Kim HJ, Chung SK, Park SW. Lipoxygenase inhibitors from Paeonia lactiflora seeds. Prev Nutr Food Sci 1999; 4: 163-166
- 72 Tanaka T, Zhang H, Jiang ZH, Kouno I. Relationship between hydrophobicity and structure of hydrolyzable tannins, and association of tannins with crude drug constituents in aqueous solution. Chem Pharm Bull 1997; 45: 1891-1897
- 73 Ulubelen A, Cetin ET, Isildatici S, Öztürk S. Phytochemical investigation of Paeonia decora . Lloydia 1968; 31: 249-251
- 74 Liu X, Yang MH, Wang XB, Xie SS, Li ZR, Kim DH, Park JS, Kong LY. Lignans from the root of Paeonia lactiflora and their anti-β-amyloid aggregation activities. Fitoterapia 2015; 103: 136-142
- 75 Teka T, Zhang L, Ge X, Li Y, Han L, Yan X. Stilbenes: Source plants, chemistry, biosynthesis, pharmacology, application and problems related to their clinical Application-A comprehensive review. Phytochemistry 2022; 197: 113128
- 76 Ryu HW, Song HH, Shin IS, Cho BO, Jeong SH, Kim DY, Ahn KS, Oh SR. Suffruticosol A isolated from Paeonia lactiflora seedcases attenuates airway inflammation in mice induced by cigarette smoke and LPS exposure. J Funct Foods 2015; 17: 774-784
- 77 Nie R, Zhang Y, Jin Q, Zhang S, Wu G, Chen L, Zhang H, Wang X. Identification and characterisation of bioactive compounds from the seed kernels and hulls of Paeonia lactiflora Pall by UPLC-QTOF-MS. Food Res Int 2021; 139: 109916
- 78 Zhao Q, Gu L, Li Y, Zhi H, Luo J, Zhang Y. Volatile composition and classification of Paeonia lactiflora flower aroma types and identification of the fragrance-related genes. Int J Mol Sci 2023; 24: 9410
- 79 Wang T, Xie A, Zhang D, Zemiao L, Li X, Li Y, Sun X. Analysis of the volatile components in flowers of Paeonia lactiflora Pall. and Paeonia lactiflora Pall. var. Trichocarpa . Am J Plant Sci 2021; 12: 146-162
- 80 Zhang L, Li DC, Liu LF. Paeonol: Pharmacological effects and mechanisms of action. Int Immunopharmacol 2019; 72: 413-421
- 81 Adki KM, Kulkarni YA. Chemistry, pharmacokinetics, pharmacology and recent novel drug delivery systems of paeonol. Life Sci 2020; 250: 117544
- 82 Wu R, Liu Y, Zhang F, Dai S, Xue X, Peng C, Li Y, Li Y. Protective mechanism of Paeonol on central nervous system. Phytother Res 2024; 38: 470-488
- 83 Hopkins AL. Network pharmacology: The next paradigm in drug discovery. Nat Chem Biol 2008; 4: 682-690
- 84 Guney E, Menche J, Vidal M, Barábasi AL. Network-based in silico drug efficacy screening. Nat Commun 2016; 7: 10331
- 85 Di Z, Ma S, Sun J. Mechanisms involved in antineuralgic effects of Paeonia Lactiflora: Prediction based on network pharmacology. TMR Clinical Research 2019; 2: 43-56
- 86 Jin ZL, Gao N, Xu W, Xu P, Li S, Zheng YY, Xue M. Receptor and transporter binding and activity profiles of albiflorin extracted from Radix paeoniae Alba. Sci Rep 2016; 6: 33793
- 87 Hu F, Lin J, Xiong L, Li Z, Liu WK, Zheng YJ. Exploring the molecular mechanism of Xuebifang in the treatment of diabetic peripheral neuropathy based on bioinformatics and network pharmacology. Front Endocrinol (Lausanne) 2024; 15: 1275816
- 88 Pan HT, Xi ZQ, Wei XQ, Wang K. A network pharmacology approach to predict potential targets and mechanisms of “Ramulus Cinnamomi (cassiae)–Paeonia lactiflora” herb pair in the treatment of chronic pain with comorbid anxiety and depression. Ann Med 2022; 54: 413-425
- 89 Bishnoi M, Bosgraaf CA, Abooj M, Zhong L, Premkumar LS. Streptozotocin-induced early thermal hyperalgesia is independent of glycemic state of rats: Role of transient receptor potential vanilloid 1 (TRPV1) and inflammatory mediators. Mol Pain 2011; 7: 52
- 90 Kishore L, Kaur N, Singh R. Effect of Kaempferol isolated from seeds of Eruca sativa on changes of pain sensitivity in Streptozotocin-induced diabetic neuropathy. Inflammopharmacology 2018; 26: 993-1003
- 91 Zhang D, Jing B, Li X, Shi H, Chen Z, Chang S, Zheng Y, Lin Y, Pan Y, Sun J. Antihyperalgesic effect of Paeniflorin based on chronic constriction injury in rats. Rev Bras Farmacogn 2022; 32: 375-385
- 92 Zhou J, Wang L, Wang J, Wang C, Yang Z, Wang C, Zhu Y, Zhang J. Paeoniflorin and albiflorin attenuate neuropathic pain via MAPK pathway in chronic constriction injury rats. Evid Based Complement Alternat Med 2016; 2016: 8082753
- 93 Liu P, Cheng J, Ma S, Zhou J. Paeoniflorin attenuates chronic constriction injury-induced neuropathic pain by suppressing spinal NLRP3 inflammasome activation. Inflammopharmacology 2020; 28: 1495-1508
- 94 Liu P, Chen J, Ma S, Zhang J, Zhou J. Albiflorin attenuates mood disorders under neuropathic pain state by suppressing the hippocampal NLRP3 inflammasome activation during chronic constriction injury. Int J Neuropsychopharmacol 2021; 24: 64-76
- 95 Zheng Y, Zhao J, Chang S, Zhuang Z, Waimei S, Li X, Chen Z, Jing B, Zhang D, Zhao G. β-sitosterol alleviates neuropathic pain by affect microglia polarization through inhibiting TLR4/NF-κB signaling pathway. J Neuroimmune Pharmacol 2023; 18: 690-703
- 96 Cai L, Zeng R, Huang Q, Liu X, Cao Z, Guo Q. Paeonol inhibits chronic constriction injury-induced astrocytic activation and neuroinflammation in rats via the HDAC/miR-15a pathway. Drug Dev Res 2022; 83: 1758-1765
- 97 Yin D, Liu YY, Wang TX, Hu ZZ, Qu WM, Chen JF, Cheng NN, Huang ZL. Paeoniflorin exerts analgesic and hypnotic effects via adenosine A 1 receptors in a mouse neuropathic pain model. Psychopharmacology (Berl) 2016; 233: 281-293
- 98 Fan YX, Hu L, Zhu SH, Han Y, Liu WT, Yang YJ, Li QP. Paeoniflorin attenuates postoperative pain by suppressing matrix Metalloproteinase-9/2 in mice. Eur J Pain 2018; 22: 272-281
- 99 Andoh T, Kurokawa Y, Kato M, Juntado ME. Local preventive effects of shakuyakukanzoto and paeoniflorin external gel on paclitaxel-induced peripheral neuropathic pain in mice. Tradit Kampo Med 2022; 9: 186-191
- 100 Andoh T, Kobayashi N, Uta D, Kuraishi Y. Prophylactic topical paeoniflorin prevents mechanical allodynia caused by paclitaxel in mice through adenosine A1 receptors. Phytomedicine 2017; 25: 1-7
- 101 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
- 102 Ruan Y, Ling J, Ye F, Cheng N, Wu F, Tang Z, Cheng X, Liu H. Paeoniflorin alleviates CFA-induced inflammatory pain by inhibiting TRPV1 and succinate/SUCNR1-HIF-1α/NLPR3 pathway. Int Immunopharmacol 2021; 101: 108364
- 103 Yin N, Gao Q, Tao W, Chen J, Bi J, Ding F, Wang Z. Paeoniflorin relieves LPS-induced inflammatory pain in mice by inhibiting NLRP3 inflammasome activation via transient receptor potential vanilloid 1. J Leukoc Biol 2020; 108: 229-241
- 104 Feng LM, Chen YY, Xu DQ, Fu RJ, Yue SJ, Zhao Q, Huang YX, Bai X, Wang M, Xing LM, Tang YP, Duan JA. An integrated strategy for discovering effective components of Shaoyao Gancao decoction for treating neuropathic pain by the combination of partial least-squares regression and multi-index comprehensive method. J Ethnopharmacol 2020; 260: 113050
- 105 Meizhen Z, Xiaohui H, Yiting T, Yupeng C, Puyu HE, Liming Z, Bing P, Qing NI. Efficacy and safety of Buyang Huanwu decoction for diabetic peripheral neuropathy: A systematic review and metaanalysis. J Tradit Chin Med 2023; 43: 841-850
- 106 Wang Y, Sheng C. Clinical research of modified Danggui-Sini decoction combined with mecobalamin in treatment of diabetic peripheral neuropathy. Int J Trad Chin Med 2017; 11: 981-984
- 107 Wang C, Lin J, Xie H, Chen L, Chen P, Wu L, Gong Q, Xia D, Wang X. Study on analgesic effect of Shentong Zhuyu Decoction in neuropathic pain rats by network pharmacology and RNA-Seq. J Ethnopharmacol 2024; 330: 118189
- 108 Pang B, Zhao TY, Zhao LH, Wan F, Ye R, Zhou Q, Tian F, Tong XL. Huangqi Guizhi Wuwu Decoction for treating diabetic peripheral neuropathy: A meta-analysis of 16 randomized controlled trials. Neural Regen Res 2016; 11: 1347-1358
- 109 Zhan G, Zheng Y, Li D, Zhang H. Clinical effect of Yiqi Huoxue Tongmai decoction in the treatment of diabetic peripheral neuropathy. Pak J Zool 2024; 56: 1-6
- 110 Zhang A, Wang Q, Liu M, Tan M, Zhang X, Wu R. Efficacy and safety of Mudan granules for painful diabetic peripheral neuropathy: A protocol for a double-blind randomized controlled trial. Medicine (Baltimore) 2022; 101: e28896
- 111 Zhang Y, Wu X, Yao W, Ni Y, Ding X. Advances of traditional Chinese medicine preclinical mechanisms and clinical studies on diabetic peripheral neuropathy. Pharm Biol 2024; 62: 544-561
- 112 China Association of Chinese Medicine. Expert consensus on the clinical application of Mudan granules in the treatment of diabetic peripheral neuropathy. Accessed July 04, 2024 at: http://www.cacm.org.cn
- 113 Zhang Y, Jin D, Duan Y, Hao R, Chen K, Yu T, Lian F, Tong X. Efficacy of Mudan Granule (combined with methylcobalamin) on type 2 diabetic peripheral neuropathy: study protocol for a double-blind, randomized, placebo-controlled, parallel-arm, multi-center trial. Front Pharmacol 2021; 12: 676503
- 114 Sawangjit R, Thongphui S, Chaichompu W, Phumart P. Efficacy and safety of mecobalamin on peripheral neuropathy: A systematic review and meta-analysis of randomized controlled trials. J Altern Complement Med 2020; 26: 1117-1129
- 115 Cheng X, Wei Z, Pu S, Xiang M, Yan A, Zhang Y, Wang X. Diversity of endophytic fungi of Paeonia Lactiflora Pallas and screening for fungal paeoniflorin producers. FEMS Microbiol Lett 2018; 365: fny263
- 116 Stierle A, Strobel G, Stierle D. Taxol and taxane production by Taxomyces andreanae, an endophytic fungus of Pacific yew. Science 1993; 260: 214-216

















