IMR Press / FBL / Volume 26 / Issue 12 / DOI: 10.52586/5058
Open Access Review
Puerarin—an isoflavone with beneficial effects on bone health
Show Less
1 Department of Gastronomy Sciences and Functional Foods, Faculty of Food Science and Nutrition, Poznań University of Life Sciences, 60-624Poznań, Poland
2 Department of Human Nutrition and Dietetics, Faculty of Food Science and Nutrition, Poznań University of Life Sciences, 60-624 Poznań, Poland
*Correspondence: anna.gramza@up.poznan.pl (Anna Gramza-Michałowska)
Academic Editor: Graham Pawelec
Front. Biosci. (Landmark Ed) 2021, 26(12), 1653–1667; https://doi.org/10.52586/5058
Submitted: 11 August 2021 | Revised: 1 November 2021 | Accepted: 17 November 2021 | Published: 30 December 2021
Copyright: © 2021 The Author(s). Published by BRI.
This is an open access article under the CC BY 4.0 license (https://creativecommons.org/licenses/by/4.0/).
Abstract

Puerarin is a compound from the group of isoflavones, naturally occurring in plants of the genus Pueraria, whose representatives include, among others, Pueraria lobata and Pueraria mirifica. Relatively many scientific studies on the biological activity of puerarin have been conducted so far. It seems that most attention was paid to the effect of puerarin on bone health. However, until now, no published studies have been collected and discussed in that regard. Based on the available data obtained from in vitro studies and on the animal model, it can be clearly shown that puerarin is an effective compound in inhibiting bone resorption and improving bone structure. Consumption of puerarin may be associated with the prevention of bone mass loss and thus can reduce the risk of developing osteoporosis. However, it is necessary to conduct human intervention studies to confirm the effectiveness of such action.

Keywords
Isoflavones
Puerarin
Bone
Osteoporosis
Pueraria lobata
2. Introduction

Osteoporosis is a metabolic bone disease associated with reduced bone mass as well as increased degradation of bone microarchitecture. As a result, it is characterized by reduced bone strength and increased risk of fractures [1]. Osteoporosis is a major public health challenge worldwide [2]. It is estimated that in 2010 osteoporotic fractures caused 43,000 deaths in the European Union alone [3]. As for the USA, nearly 54 million Americans have too low bone mass and show an increased risk of fractures [4]. In addition, more than 8.9 million fractures per year are reported worldwide. The majority of these fractures concern the European population (34.8%) [1]. Therefore, it seems necessary to take all measures to prevent this disease. The most important, modifiable risk factors for osteoporosis include lack of physical activity, smoking and improper nutrition, mainly related to the deficiency of nutrients essential for bone structure and homeostasis, including vitamin D and calcium [5]. However, a number of other compounds present in the diet are also significant for bone health. These include vitamin C [6], potassium, magnesium, omega-3 fatty acids [7], carotenoids (e.g., lycopene, beta-carotene), polyphenols [8]. A group of phytochemicals particularly well-studied in terms of their effect on bone health includes soy isoflavones, including, above all, genistein and daidzein [9]. However, there are many other compounds among isoflavones which are derived not only from soya. One of isoflavones, puerarin (daidzein-8-C-glucoside) (Fig. 1), has become an object of interest in recent years. It is a major bioactive component found in the root of plants of the genus Pueraria (common name kudzu). Puerarin was isolated from such species as Pueraria lobata [10] and Pueraria mirifica [11]. Puerarin was extracted for the first time in 1950 [12]. Since then, its biological activity has been broadly analyzed. A number of health benefits are attributed to puerarin, i.e., antioxidative [13], anti-inflammatory [14], neuroprotective [15], hepatoprotective [16], anticancer [17], antidiabetic [18], cardioprotective [19], and anti-atherosclerotic effects [20]. This study reviews the current state of knowledge regarding the importance of puerarin for bone health. Based on the results of in vitro experiments and animal studies, it was discussed how it affects bone mineral density, bone markers and structural parameters.

Fig. 1.

Chemical structure of puerarin (daidzein-8-C-glucoside).

3. Effect of puerarin on bone health
3.1 In vitro studies

Many in vitro experiments in which the effect of puerarin on bone health is analyzed via various mechanisms, have been conducted so far (Fig. 2). In their research, scientists focused primarily on determining the effects of puerarin on osteoblasts and osteoclasts. The results obtained are discussed below.

Fig. 2.

The effect of puerarin on bone health.

3.1.1 Effect on osteoblasts

Li et al. [21] analyzed the effect of puerarin (5–20 μM) on the proliferation of osteoblasts from female mice using the MTT assay. They noted that the proliferation of osteoblasts increased after 24 hours of culture. It should be noted that after 48 h and 72 h even higher proliferation was observed but this effect differed depending on the dose (5 μM, p < 0.05; 10 and 20 μM, p < 0.01). Additionally, the authors of the experiment found a statistically significant decrease in RANKL (receptor activator of nuclear factor kappa-B ligand) protein expression in osteoblasts after treatment with puerarin. At the same time, they noted an increase in OPG (osteoprotegerin) protein expression. These results showed that puerarin may inhibit osteoclastogenesis-related processes and thus prevent bone resorption [21]. Similar results were obtained by Yuan et al. [22] who showed that puerarin increased mRNA expression of OPG/GAPD in M3T3-E1 cells and simultaneously decreased RANKL mRNA expression. Wang et al. [23] confirmed that puerarin stimulates osteogenesis in osteoblast-like MC3T3-E1 cells. The observed increase in the viability of MC3T3-E1 cells indicates a significant effect of puerarin on proliferation, while the increase in alkaline phosphatase (ALP) activity (which is a marker of osteoblast activity) shows a stimulating effect of osteoblastic differentiation. However, the most beneficial effect was observed in the case of puerarin administration at 20 μM. In addition, it has been noted that puerarin may stimulate osteocalcin (a marker of bone formation) secretion in MC3T3-E1 cells. The authors of the experiment suggest that puerarin may stimulate osteogenesis by up-regulating the expression of OPN (osteopontin) and OPG, which constitute bone turnover markers. The results of an experiment by Shan et al. [24] confirmed that puerarin (10–40 μM) stimulates cell differentiation in MC3TC-E1 cells. As a result of treatment with puerarin, the researchers observed an increase in ALP activity, collagen type 1 secretion as well as osteocalcin secretion. Furthermore, the experiment showed an increase in bone mineralization of MC3T3-E1 cells. Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) showed an increase in expression of miR-106b after treatment of MC3T3-E1 cells with puerarin. This may indicate the anti-osteoporotic properties of puerarin, as miR-106b can have an inhibitory effect on osteoclastogenesis and osteolysis. A similar effect was observed by Feng et al. [25] who confirmed that puerarin stimulates the viability and differentiation of MC3T3-E1 cells by downregulation of miR-204. An in vitro experiment published in 2012 showed that puerarin does not affect rat osteoblast proliferation and cell viability. On the other hand, it has been found that puerarin (0.01–0.1 μM) may stimulate osteoblast differentiation. It was observed that cells treated with puerarin had higher ALP activity and Col I (type I collagen) secretion. In addition, this study showed that treatment with puerarin caused a significant increase in phospho-p38 MAPK (mitogen-activated protein kinase) and β-catenin proteins, indicating that puerarin could increase osteoblast differentiation via p38 MAPK and Wnt/b-catenin pathways [26]. Another experiment proved that puerarin promoted the proliferation and differentiation of human osteoblastic MG-63 cells. The highest puerarin activity was found at a dose of 1 μM. Additionally, it was observed that puerarin increased ALP activity as well as stimulated collagen synthesis in osteoblastic cells. It was also noted that puerarin inhibited cisplatin-induced apoptosis in MG-63 cells via ER-dependent MEK/ERK and PI3K (phosphatidylinositol 3-kinase-protein kinase B)/Akt activation [27]. Similarly, Zhang et al. [28] concluded and proved that puerarin (2.5–100 μM) positively affects rat calvaria osteoblastic bone formation through activation of the PI3K/AKt signaling pathway. An experiment by Liu et al. [29] has provided evidence that puerarin may prevent bone loss. The authors of the research noted that puerarin protected human osteoblasts from serum-free-induced apoptosis. The highest effectiveness was found for puerarin concentration of 10-8 M. Additionally, treatment with puerarin caused an increase in Bcl-2 (B-cell lymphoma 2) proteins while decreasing the expression of Bax in osteoblasts. It was also found that the anti-apoptotic effect was mediated by the ERK signaling pathway. Wang et al. [30] proved that puerarin (0.01–1 μM) stimulated the production of osteoprotegerin by human osteoblastic MG-63 cells. It should be noted that the increase in OPG expression caused by puerarin was comparable to the effect obtained with the use of 17β-estradiol. Simultaneously, puerarin inhibits the production of RANKL and IL-6 (interleukin 6), which may indicate the inhibition of bone resorption. The researchers also showed that puerarin stimulates ER-alpha, ER-beta, and steroid hormone receptor coactivator expression, which proves that puerarin acts via the ERE (estrogen response elements) pathway. Tiyasatkulkovit et al. [31] further proved that both puerarin (1000 nM) and Pueraria mirifica (PM) extract (100 μg/mL may increase the expression of ALP over type I collagen in primary baboon osteoblasts. At the same time, they did not observe the effect of puerarin and PM on other markers of bone formation: Runx2 (run-related transcription factor 2), osteocalcin and osterix. Additionally, the authors of the experiment noted that both puerarin and PM extract lowered the levels of RANKL mRNA expression (bone resorption marker). However, they did not cause any changes in OPG expression. An increase in ALP activity after treatment with puerarin was also observed in a study performed by Wang et al. [32] The highest ALP activity was found after the application of puerarin at 10-6 mol/L. At the indicated dose of puerarin, the most significant positive effect on the number of mineralized modules (5.64) was also noted. It should also be noted that too high doses of puerarin (10-3 mol/L) may inhibit ALP activity, osteoblast differentiation, reduce the formation of mineralized osteoblast nodules and, as a result, adversely affect bone formation. In turn, Wang et al. [32] proved that puerarin can stimulate proliferation and differentiation of osteoblasts in a high-glucose environment. This is an important fact because high glucose levels can disturb the balance between osteoclastic/osteogenic activity. Consistent with the obtained results, scientists suggest that puerarin may be a promising agent in the treatment of diabetic osteoporosis (DOP) [33].

3.1.2 Effect on osteoclasts

An experiment conducted by Yuan et al. [22] has proven that puerarin (10-7–10-6 mol/L) may inhibit osteoclast formation induced by RANKL. The researchers observed that RAW264.7 cells treated with puerarin formed a lower number of TRAP-positive cells (tartrate resistant acid phosphatase). One study showed that puerarin (10–50 μM) inhibited LPS-induced (lipopolysaccharide) differentiation from osteoclast precursor RAW264.7 cells. According to the researchers, puerarin was significantly capable of inhibiting the production of pro-inflammatory cytokines: PGE2 (prostaglandin E2), IL-1β (interleukin 1β) and TNF-α (tumor necrosis factor alpha), which may stimulate osteoclast differentiation. In consequence, a statistically significant decrease in mRNA expression of TRAP, cathepsin K and MMP-9 (metalloproteinase 9) occurred. At the same time, it was observed that puerarin inhibited the activation of Akt in RAW264.7 cells, thus preventing osteoclastogenesis [34]. The studies conducted by Yang et al. [35] provided conclusions about the beneficial effect of puerarin (1–25 μM) on the inhibition of RANKL-induced osteoclast activation in bone marrow-derived macrophages. The researchers observed that the cells treated with puerarin formed a lower number of TRAP-positive cells. Additionally, it was observed that puerarin decreased the expression of osteoclast-related genes including cathepsin K, a nuclear factor of activated T cell cytoplasmic 1 and calcitonin receptor. The observed effects differed depending on the dose, and the most beneficial results were obtained after treatment with puerarin at a dose of 25 μM. Based on these data, puerarin was found to be able to inhibit osteoclast formation [35]. The effect of puerarin on the inhibition of osteoclastogenesis was also confirmed by Lin et al. [36]. They observed that puerarin caused the inhibition of migration of osteoclast precursors (OCP) by blocking the production of monocyte chemotactic protein-1 (MCP-1). It should be mentioned here that MCP-1 is a key mediator of osteoclastogenesis and plays a role in bone resorption.

3.1.3 Effect on other cells

Li and Peng proved that puerarin (10–100 μM) stimulated the proliferation of human periodontal ligament stem cells. This effect differed depending on the dose and the increase in puerarin concentration (100 μM) was correlated with greater proliferation. Additionally, they observed that puerarin contributed to ALP activity. This research also showed that puerarin increased the osteogenic differentiation of periodontal ligament stem cells [37]. Wang et al. [38] noted that the osteocalcin level and ALP activity in bone marrow cells treated with ethanol + puerarin (0.01 mg/mL) were higher compared to cells treated with ethanol only. The results obtained show that puerarin may protect bone marrow stromal cells from alcohol-induced osteonecrosis [38]. The essence of the presented results of in vitro studies on puerarin is presented in Table 1 (Ref. [21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38]).

Table 1.Effect of puerarin on biomarkers of bone health (in vitro).
Cell cultures Study design Source of puerarin Effects References
Osteoblasts from female, 3-months old mice Puerarin 6’O-xyloside, 5, 10, 20 mM Shanghai Tauto Biotech Ltd. Co., Shanghai, China Proliferation of osteoblasts [21]
RANKL expression in osteoblasts
OPG expression in osteoblats
RAW264.7 cells Puerarin, 10-7, 10-6, 10-5μM, E2, 10-1μM PUMC Pharmaceutical Co., Ltd., Hebei, China The numbers of osteoclasts [22]
Mouse osteoblastic MC3T3-E1 cells Puerarin, 1 μM, E2, 0.1 μM PUMC Pharmaceutical Co., Ltd., Hebei, China mRNA expression of OPG/GAPDH [22]
mRNA expression of RANKL/GAPDH
Human osteoblastic MG-63 cells ICI Puerarin, 0.1 µM, Puerarin + ICI The National Institutes for Food and Drug Control, Beijing, China [purity >98% by HPLC analysis] ALP activity [23]
Type 1 collagen content
Cells in G1 phase
Cells in G2 + S phase
Cyclin B1 expression
Cyclin D1 expression
Cell death
Osteoblastic MC3T3-E1 cells Puerarin, 5, 10, 20, 40 μM National Institutes for Food and Drug Control, Beijing, China Optical density [24]
ALP activity
Type 1 collagen content
Content of OCN
Mineralized nodule
Relative expression of miR-106b
Osteoblastic MC3T3-E1 cells Puerarin, 0.1, 1, 10 μM No data Mineralized nodule [25]
ALP activity
Cell viability
Relative miR-204 expression level
Osteoblasts from Sprague-Dawley rats Puerarin, 0.01, 0.03, 0.1 mM National Institutes for Food and Drug Control, Beijing, China ALP activity [26]
Col I secretion
Osteoblastic MC3T3-E1 cells Puerarin, 1, 10, 20 μM Nanjing TCM institute of Chinese Materia Medica (TCM054-110528, China) Cell viability of MC3T3-E1 cells [27]
ALP activity
Secreted osteocalcin
OPN expression
OPG expression
Rat calvaria osteoblasts 17-beta-estriadol group, Puerarin, 2.5, 5, 10, 25, 50, 100 µmol/L Beijing Institutes for Food and Drug Control, Beijing China [purity 98.5%] ALP activity [28]
Cell viability
Mineral nodules formation
p-Akt/Total Akt
Human osteoblasts from femur Puerarin, 10-10, 10-9, 10-8, 10-7, 10-6 µM.E2, 10 µM Sigma, Inc., USA Cell apoptosis [29]
Bax expression
Bcl-2 expression
Human osteoblastic MG-63 cells ICI, 100 µM, Puerarin, 0.1 µM, Puerarin + ICI, Puerarin, 0.01, 0.1, 1 μM National Institutes for Food and Drug Control, Beijing, China [purity >98%] IL-6 production [30]
OPG expression
RANKL expression
ERα expression
ERβ expression
Primary baboon osteoblasts P. mirifica extract, 100 μg/mL, Genistein, 1000 nM, Puerarin, Puerarin, 1000 nM P. mirifica (Chiang Mai Province, Northern Thailand) ALP activity [31]
Puerarin (LKT Laboratories Inc., St. Paul, MN, USA) Type I collagen
RANKL mRNA expression
Osteoblasts from calvarial bone of Wistar rats Puerarin, 10-10, 10-9, 10-8, 10-7, 10-6, 10-5, 10-4, 10-3μM National Institutes for Food and Drug Control, Beijing, China Osteoblast proliferation [32]
ALP activity
Number of mineralized nodules
Osteoblasts from newly born Sprague Dawley rats Puerarin, 0.01, 0.1, 1 μM National Institutes for Food and Drug Control (NIFDC), China ALP activity [33]
RAW264.7 cells LPS + Puerarin, 10, 25, 50 μM National Institutes for Food and Drug Control, Beijing, China [purity 99.8% by HPLC] TNF-α [34]
IL-1β
PGE2
TRAP mRNA expression
Cathepsin K mRNA expression
MMP-9 mRNA expression
Activation of Akt
Bone marrow-derived macrophages from femur of male C57BL/6J mice Puerarin, 1, 5, 25 μM Sigma-Aldrich (St. Louis, USA) [purity 98.0%] Number of osteoclasts per wall [35]
Percentage of osteoclast area
Ring number per wall
Area of bone resorption
CTR mRNA expression
CTR mRNA expression
NFATc1 mRNA expression
c-fos mRNA expression
RAW264.7 cells Puerarin, 10, 25, 50, 100 μM No data MCP-1 secretion [36]
Human periodontal ligament stem cells (PDLSC) Puerarin, 10, 50, 100 μM Inchem Corp., Rock Hill, SC, USA [purity 95%] MTT optical density [37]
ALP activity
mRNA expression of RUNX2
mRNA expression of collagen I
mRNA expression of OPN
mRNA expression of OCN
Bone marrow cells Puerarin, 0.01 mg/mL - ALP activity [38]
from the midshafts
of 6- to 8-week-old
male and female
mouse femurs Osteocalcin level
ALP, Alkaline phosphatase; BAP, Bone alkaline phosphatase; BMC, Bone mineral content; BMD, Bone mineral density; BV/TV, Bone surface/total volume; BS, Bone surface; Ct, Cortical bone tissue; CTx, C-terminal telopeptide; Ca, calcium; DPD, Deoxypyridinoline; EE, 17 alpha-ethinylestradiol; FM, Femoral metaphysis; FD, Femoral diaphysis; L4, Fourth lumbar vertebra; LPS, Lipopolysaccharides; MCF-7 cell, epithelial luminal cell line; MMP, Matrix metalloproteinase; NFATc1, Nuclear factor of activated T-cell, cytoplasmic 1; Ob.S, Osteoblast surface; Oc.S, Osteoclast surface; OVX, ovariectomized group; ORX, orchidectomized; OPG, Osteoprotegerin; OPN, Osteopontin; PPARγ, Peroxisome proliferator-activated receptor γ; PTIF, total isoflavones from Pueraria lobata; PVEE, kudzu vine ethanol extracts; RANKL, receptor activator of nuclear factor-κB ligand; Tb, Trabecula; Tb.N/Tb.Tn, Trabecular number; Tb.Sp, Trabecular separation; TD, Tibial diaphysis; TM, Tibial metaphysis; TRAP, Tartrate resistant acid phosphatase.
3.2 Animal model studies

The results obtained from in vitro studies are widely confirmed in tests carried out on animal models. It has been proven many times that the administration of puerarin had a beneficial effect on bone markers, bone mineral density and bone structural parameters.

3.2.1 Effect on bone markers

Li et al. [21] proved the anti-osteoporotic activity of puerarin 6-O-xyloside. They showed that ovariectomized rats (OVX) treated with puerarin (intraperitoneal injection) (20, 40, 60 mg/kg/d) had significantly higher blood calcium levels compared to the control OVX group. Additionally, higher blood phosphorus levels were observed in animals treated with puerarin. However, this effect was differed depending on the dose. The highest level of the indicated components was found with the administration of puerarin at 60 mg/kg/d. This research also assessed ALP activity. It was observed that the use of puerarin at doses of 40 or 60 mg/kg/d increased serum levels of ALP. At the same time, an increase in osteoprotegerin levels was observed [21]. Another study that proved the anti-osteoporotic effect of puerarin was conducted by Michihara et al. [39]. They showed that in ovariectomized mice during 8-week oral (per os) administration of puerarin (5 mg/kg/d) urinary DPD (doxypyridinoline) levels were lower compared to mice that did not consume this isoflavonoid. In the conducted experiment it was observed that the control group had a significantly lower number of femoral trabeculae than the OVX-puerarin group, which indicates that puerarin may protect the trabecular structure from destruction. In the group fed with puerarin a significantly lower activity of TRAP, which is a bone absorption marker, was also found. It should be noted, however, that the administration of puerarin did not affect the level of osteocalcin. Based on the observed lack of effect of puerarin on morphology and weight of oviducts and uterus, the researchers indicated that the observed properties are independent of the estrogen receptor-mediated pathway. They also proved this effect by observing that puerarin did not stimulate the growth of MCF-7 cells [39]. Similarly, Tanaka et al. [40] noted that puerarin may inhibit ovariectomy-induced bone loss. In the experiment, the researchers administered kudzu vine ethanol extract (PVEE) per os to female mice. Of isoflavones present in it, puerarin was found in the highest amounts (52%). This extract was administered at 20 mg/kg/d for 8 weeks. The authors of this research showed that animals which consumed PVEE had lower levels of bone resorption markers such as DPD and TRAP compared to OVX mice. At the same time, it was noted that the use of PVEE inhibited the reduction in femoral bone mineral density. A lower number of matured osteoclasts in the distal femur was also observed in the group treated with PVEE. On the other hand, administration of PVEE did not affect the serum bone-specific alkaline phosphatase activity. At the same time, Liang et al. [41] noted that puerarin may have a beneficial effect on bones in rats with diabetes. It was observed that daily intraperitoneal administration of puerarin (60 mg/kg/d) caused a decrease in caspase-3 expression in osteoblasts. The authors of the experiment suggest that high blood glucose levels are associated with an increase in the expression of caspase-3, which in turn may contribute to osteoblast apoptosis and lead to the development of osteoporosis. Moreover, Luo et al. [42] proved that oral administration of kudzu root extract (0.45, 0.9 and 1.8 g/kg/d) to ovariectomized rats may have a beneficial effect on bone health. In the conducted studies it was observed that the use of kudzu root extract resulted in lowering the serum levels of C-terminal telopeptide of collagen type I (CTx). This indicates that this extract may inhibit bone resorption. Additionally, it has been shown that the administration of P. lobata extract (100 mg/kg/d) results in a statistically significant reduction in the levels of osteocalcin as well as ALP and CTx, which are markers of bone resorption and remodeling [43]. It is also worth mentioning that studies conducted by Wang et al. [38] on an animal model showed that puerarin may inhibit alcohol-induced osteonecrosis. Marrow and bone necrosis were found in mice belonging to the model group treated with ethanol (intragastric). An increase in the number of empty osteocyte lacunae in the femur was also observed. Such adverse lesions were not found in mice treated with intramuscular injection of puerarin (0.5 g/kg/d). Additionally, higher expression of osteocalcin mRNA was found in animals treated with puerarin [38]. Research results published in 2020 also indicated that puerarin (15.4 and 30.8 mg/kg/d) administered i.p. inhibits titanium particle-induced osteolysis [44]. Zhang et al. [34], on the other hand, proved that calvarial injection of puerarin (1 mg/kg/d) is capable of inhibiting LPS-induced bone loss. The authors found that after the injection of LPS there was a significant increase in the number of osteoclasts and an increase in the area of osteolysis. At the same time, significant reductions in calvaria weight were observed. Mice additionally treated with puerarin had a significantly lower number of osteoclasts as well as decreased osteolysis area. Lower bone mass loss, compared to the puerarin-free group (68.5 vs. 47.9 mg of calvaria weight), was observed as well [45]. The beneficial effect of Pueraria lobata extract (PE) was also confirmed by Lee et al. [46]. They observed that the administration of PE (25–1600 mg/kg) to ovariectomized (OVX) rats resulted in a reduction in the level of bone turnover markers, such as osteocalcin, C-terminal telopeptide fragment of type I collagen, deoxypyridinoline, and pyridinoline, the concentration of which was elevated in OVX rats. The highest effect was seen in animals receiving 1600 mg PE/kg. Moreover, researchers confirmed that puerarin caused an increase in the plasma level of estradiol, which indicates that PE has estrogenic activity and its activity is similar to that of phytoestrogens. Therefore, it seems that puerarin may slow down the osteoporotic changes associated with estrogen deficiency, which is characteristic in postmenopausal women [46].

3.2.2 Effect on bone mass/bone mineral density

An experiment by Tanaka et al. [40] showed that ovariectomized mice fed with kudzu vine ethanol extracts (PVEE) had a lower degree of trabecular destruction than mice belonging to the control OVX-group. The beneficial effect of puerarin on bones was also noted in a research conducted by Yuan et al. [22]. Ovariectomized mice received a diet containing puerarin at different doses: 2, 4 or 8 mg/d. The authors observed that mice receiving food enriched with puerarin (2 mg/d) had a higher mineral density of various regions of the femur – proximal femur, middle femur, distal femur compared to animals fed without this additive [22]. In an experiment conducted by Liang et al. [41], femur X-rays showed that animals treated with puerarin had a lower loss of bone mass compared to those not treated with this additive. It was shown that femoral BMD (bone mineral density) in rats treated with puerarin was 9.7% higher than in the control group. Moreover, rats with diabetes had a higher number of osteoclasts clumped together as well as higher cortical bone reduction. In rats treated with puerarin, these lesions were milder [41]. Similar results were also obtained by Cho et al. [47] who administered isoflavones from Pueraria lobata per os to ovariectomized mice. Of isoflavones found in P. lobata extract, puerarin was present at the highest concentration (7.5%). Daidzein (4.2%) and genistein (1.9%) were present in smaller amounts. The researchers found that mice fed with P. lobata extract (200 and 500 mg/kg/d) had significantly higher femoral BMD than those fed without this additive [47]. Similarly, in another study, researchers showed that rats consuming the kudzu root had higher BMD of the middle femur [42]. In turn, Wang et al. [26] proved that puerarin stimulates bone formation. They showed that ovariectomized rats treated with puerarin (i.g.; 20 mg/kg/d) had a higher femur BMD and BMC (bone mineral content). The positive effect of Pueraria mirifica administration on bone health was also noted by Suthon et al. [48]. The researchers showed that rats treated with P. mirifica (50 mg/kg/d) by gavage had a higher total BMD of L4 (fourth lumbar vertebra), as well as trabecular BMD of L4 and tibial metaphysis, compared to rats not treated with isoflavones. The effect of dietary isoflavones from Puerariae Radix on bone metabolism in ovariectomized rats was also studied by Lim et al. [43]. The researchers administered orally P. lobata extract, containing high amounts of isoflavones, to rats. Puerarin constituted 57.6% of all isoflavones. The remaining isoflavones were daidzein (30.4%) and genistein (12.0%). They found that rats treated with the extract at 100 mg/kg/d had a significantly higher femoral bone mineral density compared to the control OVX-group. Such an effect was not observed in the group treated with P. lobata extract at 30 mg/kg/d. Yang et al. [49] showed that rats with ligature-induced periodontitis and treated with puerarin at 200 mg/kg/d by gavage had a lower volume of bone loss compared to those not treated with isoflavonoids. In this research, it was noted that the consumption of puerarin inhibited alveolar bone loss by inhibiting RANKL production and reducing the number of active osteoclasts [49]. The experiment performed on healthy male C57BL/6J mice showed that the intraperitoneal administration of puerarin (10 and 50 mg/kg/d) caused inhibition of osteolysis induced by titanium particles. The authors of the research noted that mice treated with puerarin had higher BMD and BV/TV (bone surface/total volume). However, this effect was dose-dependent, and a more beneficial effect was observed when a higher dose of puerarin was used. In addition, micro-CT scanning showed a lower number of pores in mice treated with puerarin. The researchers also noted a statistically significant reduction in eroded surface area, in the number of TRAP-positive cells, as well as in the osteoclast surface to bone surface ratio [49]. Interesting research results were observed by Liu et al. [50], who noted that administration of puerarin combined with zinc (50 mg/kg/d + 0.25 mg/kg/d) by gavage could prevent mandibular bone loss in OVX rats more effectively than administration of these compounds separately. The experiment showed that rats treated with puerarin + zinc had statistically significantly higher bone mineral density compared to rats fed without these additives, as well as compared to those consuming puerarin + zinc separately. In contrast, studies performed on adult ovariectomized rats showed that rats treated with puerarin (administered i.p.) (50 mg/kg/d) from Pueraria lobata had a lower BMD of the proximal tibia compared to animals not treated with this isoflavonoid. This means that puerarin was not capable of reducing loss of BMD induced by ovariectomy [47]. In 2020, the results of a meta-analysis on the effect of puerarin administration on bone mass for OVX-induced postmenopausal osteoporosis in a murine model were published. Based on eight randomized studies, the authors showed that the use of puerarin can have a beneficial effect on the increase in bone mineral density [51]. The beneficial effect of puerarin on bones was also noted by Li et al. [52]. They showed that administration of puerarin at 100 mg/kg/d resulted in a statistically significant increase in cortex and trabeculae BMD. Simultaneously, the authors of the experiment noticed that in the tested rats there was an improvement in the integrity of the intestinal mucosa. Moreover, positive changes in gut microbiota were noted. For example, Lactobacillaceae and Bifidobacteriaceae bacteria were found to grow. Additionally, puerarin increased the content of SCFAs. The authors suggested that puerarin may prevent osteoporotic changes by modulating the community of gut microbiota and repairing intestinal mucosal integrity [52]. The essence of the presented results of puerarin studies in animal models is presented in Table 2 (Ref. [21, 22, 26, 34, 35, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 52, 53, 54, 55, 56]).

Table 2.Effect of puerarin on biomarkers of bone health in animal models.
Study size Study design Source of puerarin Effects References
50 female rats, 3 months old Puerarin 6’O-xyloside, 40, 60 mg/kg/d (i.p.) Shanghai Tauto Biotech Ltd. Co., Shanghai, China Blood calcium level [21]
Blood phosphorus level
Serum ALP level
Duration of treatment: 12 weeks Serum OPG level
48 Kunming female mice 8 weeks old OVX + Puerarin, 2, 4, 8 mg PUMC Pharmaceutical Co., Ltd., Hebei, China Proximal femur BMD [22]
Middle femur BMD
Distal femur BMD
Duration of treatment: 4 weeks Total femur BMD
12 Sprague-Dawley rats OVX group treated with puerarin (20 mg/kg/d, intragastric administration) National Institutes for Food and Drug Control, Beijing, China BMD [26]
BMC
Tb.N
Tb.Tn
Duration of treatment: 12 weeks Tb.Sp
36 ICR male mice, 6–8 weeks old Puerarin, 1 mg/kg/d (calvarial injection) National Institutes for Food and Drug Control, Beijing, China [purity 99.8% by HPLC] Number of osteoclasts [34]
Osteolysis area
Duration of treatment: 2 weeks Calvaria weight
Pathogen-free and healthy male C57BL/6J mice, 6–8 weeks old Puerarin, 10, 50 mg/kg/d (i.p.) Sigma-Aldrich (St. Louis, USA) [purity 98.0%] BMD [35]
BV/TV
Total porosity
Number of pores
Eroded surface
TRAP positive cells
Duration of treatment: 14 days Oc.S/BS
216 female Kunming mice, 4 weeks old experimental group received spirits (20 mL/kg) [46% ethanol; intragastrically] + puerarin (0.5 g/kg) [intramuscular injection] - ALP activity [38]
Empty osteocyte lacuna
Largest fat cell diameter in femoral heads
Expression of PPARγ mRNA
Duration of treatment: 4–10 months Expression of osteocalcin mRNA
Slc: ddY female mice, 9 weeks old Sham-puerarin group (5 mg/kg/d) - Urine DPD concentration [39]
OVX-puerarin group (5 mg/kg/d)
Duration of treatment: 8 weeks Serum TRAP activity
Slc: ddY female mice, 10 weeks old OVX-PVEE group (20 mg/kg/d) Kudzu vine ethanol extract (PVEE) Urine DPD [40]
TRAP activity
Femoral BMD
Duration of treatment: 8 weeks Number of matured osteoclasts
30 Sprague-Dawley male rats Diabetes treated with puerarin (intraperitoneally injected, 60 mg/kg/d) control group Limin Pharmaceutical Corporation, Jinan, Shandong, China Caspase-3 expression [41]
Femoral BMD)
Number of osteoclasts clumped together
Cortical bone reduction
Duration of treatment: 6 weeks Deteriorated bone micro-architecture
60 female Sprague-Dawley rats, 6 months old OVX + kudzu, 0.45, 0.9, 1.8 g/kg/d Kudu root extract Serum CTX-I [42]
Duration of treatment: 6 weeks Femoral BMD
Female Sprague-Dawley rats, 8 weeks old OVX + 17β-estradiol (10 μg/kg/d, i.p.) P. lobata extract (Kapdang Co., Seoul, Korea) Femur BMD [43]
ALP
Osteocalcin
OVX + PTIF (30 or 100 mg/kg/d) CTx
Duration of treatment: 8 weeks Estradiol
20 Male Sprague-Dawley rats Puerarin, 15.4, 30.8 mg/kg/d (i.p.) Sigma-Aldrich, Saint Louis, MO, USA BMD [44]
BV/TV
Tb.Th
NFAT-c1 positive cells
MMP9-positive cells
60 adult Sprague-Dawley rats Estradiol, 10 µg/kg (i.p.) Beijing Four Rings Biopharmaceutical Co., Ltd. Tb.N [45]
BV/TV
Puerarin, 50 mg/kg (i.p.) Osteocalcin
Duration of treatment: 12 weeks BMD
48 nine-week-old female Sprague Dawley rats Puerarin, 25, 100, 400, 1600 mg/kg/d Sejun F & B Co., Ltd., Gangwon, Korea Osteocalcin [46]
C-terminal telopeptide fragment of type I collagen
Deoxypyridinoline
Duration of treatment: 8 weeks Pyridinoline
20 female mice, 30 weeks old OVX treated with 200 mg or 500 IPL/kg/d Pueraria lobata (Willd.) extract Femoral BMD [47]
Duration of treatment: 4 weeks
Sprague-Dawley female rats, 6 months old OVX + P. mirifica, 5, 25, 50 mg/g/d (by gavage) OVX + Puerarin, 7 mg/kg/d (subcutaneously injected) P. mirifica powder (Smith Natural Co., Ltd) Trabecular BMD of tibia metaphysis [48]
Total BMD of L4
Trabecular BMD of L4
BV/TV
Tb.Sp
Tb.N
Duration of treatment: 12 weeks Ob.S/BS
40 male Sprague-Dawley rats, 7 weeks old Puerarin, 200 mg/kg/d (by gavage) Shanghai Winherb Medical S&T Development Co. Ltd., Shanghai, China [98% purity] BV/TV [49]
Bone resorption area
Number of RANKL-positive cells
RANKL/OPG expression
Number of OPG-positive cells
Number of active osteoclasts
Female Sprague-Dawley rats, 8 weeks old OVX + 17 beta-estradiol group (10 ug/kg/d) Sigma-Aldrich, Saint Louis, MO, USA [Analytically pure] BMD in bone trabecula of mandible [50]
BMD in inferior margin of mandible
BV/TV
Tb/Tb.N
Tb.Sp
OVX + puerarin group (50 mg/kg/d) Ca
OVX + zinc group (0.25 mg/kg/d) TRAP
OVX + puerarin + zinc group RANKL
Duration of treatment: 12 weeks vs. OVX+puearin or OVX+zinc group
40 10-week-old Sprague-Dawley female rats Puerarin, 50, 100 mg/kg/d J&K Scientific Ltd, Beijing, China [purity 98.0%] Cortex BMD [52]
Duration of treatment: 14 weeks Trabeculae BMD
Male Sprague-Dawley rats, 7 months old ORX + P. mirifica, 0, 10, 100, 1000 mg/kg/d Pueraria mirifica (Chiang Mai Province, Thailand) TbBMD of TM/FM/L4 [53]
CtBMD of TM/FM/L4/TD
TbBMC of TM/FM/L4
Duration of treatment: 3 months CtBMC of TM/FM/L4/TD/FD
18 eleven-week-old C57BL/6J mice Puerarin, 100 mg/kg intraperitoneal injections every two days Sigma-Aldrich, Sydney, Australia BV/TV [54]
BS/BV
BS/TV
Duration of treatment: 6 weeks Tb.N.
30 7–8 weeks old Male Sprague-Dawley rats Puerarin, 50 mg/kg/d Sigma-Aldrich, USA [99% purity] BV/TV [55]
Tb.N
Tb.Sp
OPG
Duration of treatment: 14 weeks CTX
30 female 6-week-old outbred ICR mice Fermented puerarin, 100 mg/kg/d Daegu University, Daegu, Korea BMD [56]
BV/TV
Duration of treatment: 12 weeks Tb.Th
Tb.N
Tb.Sp
ALP, Alkaline phosphatase; BAP, Bone alkaline phosphatase; BMC, Bone mineral content; BMD, Bone mineral density; BV/TV, Bone surface/total volume; BS, Bone surface; Ct, Cortical bone tissue; CTx, C-terminal telopeptide; Ca, calcium; DPD, Deoxypyridinoline; EE, 17 alpha-ethinylestradiol; FM, Femoral metaphysis; FD, Femoral diaphysis; L4, Fourth lumbar vertebra; LPS, Lipopolysaccharides; MCF-7 cell, epithelial luminal cell line; MMP, Matrix metalloproteinase; NFATc1, Nuclear factor of activated T-cell, cytoplasmic 1; Ob.S, Osteoblast surface; Oc.S, Osteoclast surface; OVX, ovariectomized group; ORX, orchidectomized; OPG, Osteoprotegerin; OPN, Osteopontin; PPARγ, Peroxisome proliferator-activated receptor γ; PTIF, total isoflavones from Pueraria lobata; PVEE, kudzu vine ethanol extracts; RANKL, receptor activator of nuclear factor-κB ligand; Tb, Trabecula; Tb.N/Tb.Tn, Trabecular number; Tb.Sp, Trabecular separation; TD, Tibial diaphysis; TM, Tibial metaphysis; TRAP, Tartrate resistant acid phosphatase.
3.2.3 Effect on structural parameters (histomorphometric data)

Li et al. [21] observed that the administration of puerarin 6-O-xyloside to rats had a beneficial effect on bone microarchitecture. Compared to the control OVX group, in animals treated with puerarin, the cortical and trabecular bone were thickened. It should be noted that the level of pathological lesions in femoral bone tissue was dependent on the dose of puerarin administered. The least adverse lesions occurred after administration of 60 mg puerarin/kg/d [21]. Urasopon et al. [53] conducted research in which they administered Pueraria mirifica to rats after orchidectomy. They found that the enrichment of food with P. mirifica inhibited bone loss in trabecular and cortical bones. It should be stressed that this effect was dependent on the dose of P. mirifica. The most positive results were achieved in the group of rats treated with the highest amounts of P. mirifica (1000 mg/kg/d) [53]. Wang et al. [26] showed that the administration of puerarin to rats resulted in the improvement of trabecular bone structure. They noted that both trabecular number and trabecular thickness were significantly higher in the OVX + puerarin group compared to the OVX group [26]. The beneficial effect on bone histomorphometry was also confirmed by Suthon et al. [48]. They observed that rats treated with P. mirificashowed an improvement in such parameters as bone volume (BV), trabecular separation (Tb.Sp), trabecular number (Tb.N), and osteoblast surface (Ob.S) [48]. Similarly, Lim et al. [43] observed that puerarin can effectively improve the histomorphometry parameters. It was noted that the proximal tibia had significantly higher trabecular number (Tb.N) as well as BV/TV. However, no difference was found for such parameters as bone surface/bone volume (BS/BV), trabecular thickness (Tb.Th) and Tb.Sp. This research also showed an increase in plasma osteocalcin level in the group treated with puerarin, with a simultaneous slight decrease in alkaline phosphatase activity [43]. Additionally, the bone morphometry of the trabeculae of mandibles proved that animals treated with puerarin + zinc showed higher values for such parameters as BV/TV and Tb/Tb.N. At the same time, a significant reduction in Tb.Sp compared to OVX + puerarin and OVX + zinc group was noted. The authors also compared the levels of serum bone biochemical markers. They showed significantly higher calcium levels and simultaneously lower levels of TRAP and RANKL. The results of this study showed that the combined use of puerarin and zinc may be an effective way to inhibit osteoclastogenesis [50]. Xiao et al. [54] confirmed that puerarin protects against bone mass loss in ovariectomy-induced osteoporosis model mice. They reported that the animals that received puerarin had improved the bone structural features. The following parameters were improved: BV/TV, BS/BV, BS/TV, Tb.N. The authors of the experiment proved that puerarin has a bone protective effect by suppressing osteoclastogenesis via inhibition of the TRAF6/ROS-dependent MAPK/NF-κB signaling pathway [54]. Interesting observations were made by Guo et al. [55], who proved that puerarin inhibits the osteoporotic changes associated with streptozotocin (STZ)-induced diabetes. The authors observed that puerarin improves bone microstructure. Higher BV/TV and Tb.N values were noted in rats receiving puerarin. At the same time, the reduction in Tb.Sp was noted. Moreover, lower levels of OPG and CTX were found. The scientists indicated that this effect may be related to suppressed inflammation [55]. Anti-osteoporotic activity was also demonstrated by Pueraria lobata fermented (FPE) with Lactobacillus paracasei JS1. Researchers observed that ovariectomized mice treated with FPE were characterized by improved bone architecture. BV/TV, Tb.Th, and Tb.N were higher compared to OVX group. At the same time, a lower value of Tb.Sp was noted. Additionally, an increase in bone mineral density was noted in mice receiving FBE [56].

4. Summary and perspective

The current state of knowledge clearly shows that puerarin has a positive effect on bone health. It has been repeatedly proven that this compound stimulates osteoblast differentiation and simultaneously inhibits osteoclastogenesis. The studies carried out on an animal model proved that puerarin has anti-osteoporotic properties and can inhibit bone loss in ovariectomized and orchidectomized animals. Many experiments showed that animals treated with puerarin had higher bone mineral density and bone mineral content. It should be noted, however, that despite promising indications concerning the beneficial effect of puerarin on bones, there is a lack of research that would prove such an effect among people. For this reason, the initiation of intervention studies with the participation of people, especially because there are good grounds for this, is worth considering.

5. Author contributions

Conceptualization and methodology—BK, AGM; investigation and data curation—BK, AS; writing—original draft preparation—BK; writing—review and editing—BK, JS, AGM; supervision—AGM.

6. Ethics approval and consent to participate

Not applicable.

7. Acknowledgment

Thanks to all the peer reviewers for their opinions and suggestions.

8. Funding

This research received no external funding.

9. Conflict of interest

The authors declare no conflict of interest.

Abbreviations

ALP, Alkaline phosphatase; BAP, Bone alkaline phosphatase; BMC, Bone mineral content; BMD, Bone mineral density; BV/TV, Bone surface/bone volume; BS, Bone surface; Ct, Cortical bone tissue; CTx, C-terminal telopeptide; Ca, calcium; DPD, Deoxypyridinoline; EE, 17 alpha-ethinylestradiol; FM, Femoral metaphysis; FD, Femoral diaphysis; L4, Fourth lumbar vertebra; LPS, Lipopolysaccharides; MCF-7 cell, epithelial luminal cell line; MMP, Matrix metalloproteinase; NFATc1, Nuclear factor of activated T-cell, cytoplasmic 1; Ob.S, Osteoblast surface; Oc.S, Osteoclast surface; OVX, ovariectomized group; ORX, orchidectomized; OPG, Osteoprotegerin; OPN, Osteopontin; PPARγ, Peroxisome proliferator-activated receptor γ; PTIF, total isoflavones from Pueraria lobata; PVEE, kudzu vine ethanol extracts; RANKL, receptor activator of nuclear factor-κB ligand; Tb, Trabeculars; Tb.N/Tb.Tn, Trabecular number; Tb.Sp, Trabecular separation; TD, Tibial diaphysis; TM, Tibial metaphysis; TRAP, Tartrate resistant acid phosphatase.

References
[1]
Pouresmaeili F, Kamalidehghan B, Kamarehei M, Goh MG. A comprehensive overview on osteoporosis and its risk factors. Therapeutics and Clinical Risk Management. 2018; 14: 2029–2049.
[2]
Chen L, Ko N, Chen K. Medical Treatment for Osteoporosis: from Molecular to Clinical Opinions. International Journal of Molecular Sciences. 2019; 20: 2213.
[3]
Brzezińska O, Łukasik Z, Makowska J, Walczak K. Role of vitamin C in osteoporosis development and treatment — a literature review. Nutrients. 2020; 12: 2394.
[4]
des Bordes J, Prasad S, Pratt G, Suarez-Almazor ME, Lopez-Olivo MA. Knowledge, beliefs, and concerns about bone health from a systematic review and metasynthesis of qualitative studies. Public Library of Science One. 2020; 15: e0227765.
[5]
Yao P, Bennett D, Mafham M, Lin X, Chen Z, Armitage J, et al. Vitamin D and Calcium for the Prevention of Fracture. JAMA Network Open. 2019; 2: e1917789.
[6]
Aghajanian P, Hall S, Wongworawat MD, Mohan S. The Roles and Mechanisms of Actions of Vitamin C in Bone: New Developments. Journal of Bone and Mineral Research. 2015; 30: 1945–1955.
[7]
Sahni S, Mangano KM, McLean RR, Hannan MT, Kiel DP. Dietary Approaches for Bone Health: Lessons from the Framingham Osteoporosis Study. Current Osteoporosis Reports. 2015; 13: 245–255.
[8]
Sacco SM, Horcajada M, Offord E. Phytonutrients for bone health during ageing. British Journal of Clinical Pharmacology. 2013; 75: 697–707.
[9]
Zheng X, Lee S, Chun OK. Soy Isoflavones and Osteoporotic Bone Loss: a Review with an Emphasis on Modulation of Bone Remodeling. Journal of Medicinal Food. 2016; 19: 1–14.
[10]
Zhou Y, Zhang H, Peng C. Puerarin: a review of pharmacological effects. Phytotherapy Research. 2014; 28: 961–975.
[11]
Kakehashi A, Yoshida M, Tago Y, Ishii N, Okuno T, Gi M, et al. Pueraria mirifica Exerts Estrogenic Effects in the Mammary Gland and Uterus and Promotes Mammary Carcinogenesis in Donryu Rats. Toxins. 2016; 8: 275.
[12]
Wei S, Chen Y, Xu X. Progress on the pharmacological research of puerarin: a review. Chinese Journal of Natural Medicines. 2014; 12: 407–414.
[13]
Xiao C, Li J, Dong X, He X, Niu X, Liu C, et al. Anti-oxidative and TNF-α suppressive activities of puerarin derivative (4AC) in RAW264.7 cells and collagen-induced arthritic rats. European Journal of Pharmacology. 2011; 666: 242–250.
[14]
Jeon Y, Lee J, Lee Y, Kim D. Puerarin inhibits inflammation and oxidative stress in dextran sulfate sodium-induced colitis mice model. Biomedicine & Pharmacotherapy. 2020; 124: 109847.
[15]
Zhu G, Wang X, Wu S, Li X, Li Q. Neuroprotective effects of puerarin on 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine induced Parkinson’s disease model in mice. Phytotherapy Research. 2014; 28: 179–186.
[16]
Zhao M, Du Y, Yuan L, Wang N. Protective effect of puerarin on acute alcoholic liver injury. The American Journal of Chinese Medicine. 2010; 38: 241–249.
[17]
Ahmad B, Khan S, Liu Y, Xue M, Nabi G. Molecular mechanisms of anticancer activities of puerarin. Cancer Management and Research. 2020; 12: 79–90.
[18]
Chen X, Yu J, Shi J. Management of Diabetes Mellitus with Puerarin, a Natural Isoflavone from Pueraria lobata. The American Journal of Chinese Medicine. 2018; 46: 1771–1789.
[19]
Li X, Yuan T, Chen D, Chen Y, Sun S, Wang D, et al. Cardioprotective effects of puerarin-V on isoproterenol-induced myocardial infarction mice is associated with regulation of PPAR-Υ/NF-κB pathway. Molecules. 2018; 23: 3322.
[20]
Bao L, Zhang Y, Wei G, Wang Y, Ma R, Cheng R, et al. The anti-atherosclerotic effects of puerarin on induced-atherosclerosis in rabbits. Biomedical Papers of the Medical Faculty of the University Palacky Olomouc Czechoslovakia. 2015; 159: 53–59.
[21]
Li H, Chen B, Pang G, Chen J, Xie J, Huang H. Anti-osteoporotic activity of puerarin 6”-O-xyloside on ovariectomized mice and its potential mechanism. Pharmaceutical Biology. 2016; 54: 111–117.
[22]
Yuan S, Sheng T, Liu L, Zhang Y, Liu X, Ma T, et al. Puerarin prevents bone loss in ovariectomized mice and inhibits osteoclast formation in vitro. Chinese Journal of Natural Medicines. 2016; 14: 265–269.
[23]
Wang N, Wang X, Cheng W, Cao H, Zhang P, Qin L. Puerarin promotes osteogenesis and inhibits adipogenesis in vitro. Chinese Medicine. 2013; 8: 17.
[24]
Shan Z, Cheng N, Huang R, Zhao B, Zhou Y. Puerarin promotes the proliferation and differentiation of MC3T3-E1 cells via microRNA-106b by targeting receptor activator of nuclear factor-κB ligand. Experimental and Therapeutic Medicine. 2018; 15: 55–60.
[25]
Feng Q, Cheng SY, Yang R, Zeng XW, Zhao FM, Zhan XQ. Puerarin promotes the viability and differentiation of MC3T3-E1 cells by enhancing LC3B-mediated autophagy through downregulation of miR-204. Experimental and therapeutic Medicine. 2020; 19: 883–890.
[26]
Wang P, Zhu X, Yang L, Liang H, Feng S, Zhang R. Puerarin stimulates osteoblasts differentiation and bone formation through estrogen receptor, p38 MAPK, and Wnt/β-catenin pathways. Journal of Asian Natural Products Research. 2012; 14: 897–905.
[27]
Wang Y, Wang WL, Xie WL, Li LZ, Sun J, Sun WJ, et al. Puerarin stimulates proliferation and differentiation and protects against cell death in human osteoblastic MG-63 cells via ER-dependent MEK/ERK and PI3K/Akt activation. Phytomedicine. 2013; 20: 787–796.
[28]
Zhang Y, Zeng X, Zhang L, Zheng X. Stimulatory effect of puerarin on bone formation through activation of PI3K/Akt pathway in rat calvaria osteoblasts. Planta Medica. 2007; 73: 341–347.
[29]
Liu L, Liu L, Bo T, Li S, Zhu Z, Cui R, et al. Puerarin Suppress Apoptosis of Human Osteoblasts via ERK Signaling Pathway. International Journal of Endocrinology. 2013; 2013: 1–6.
[30]
Wang Y, Yang C, Xie WL, Zhao YW, Li ZM, Sun WJ, et al. Puerarin concurrently stimulates osteoprotegerin and inhibits receptor activator of NF-κB ligand (RANKL) and interleukin-6 production in human osteoblastic MG-63 cells. Phytomedicine. 2014; 21: 1032–1036.
[31]
Tiyasatkulkovit W, Malaivijitnond S, Charoenphandhu N, Havill LM, Ford AL, VandeBerg JL. Pueraria mirifica extract and puerarin enhance proliferation and expression of alkaline phosphatase and type i collagen in primary baboon osteoblasts. Phytomedicine. 2014; 21: 1498–1503.
[32]
Wang C, Meng M, Tang X, Chen K, Zhang L, Liu W, et al. The proliferation, differentiation, and mineralization effects of puerarin on osteoblasts in vitro. Chinese Journal of Natural Medicines. 2014; 12: 436–442.
[33]
Wang XH, Shi XW, Luo XX, Zhang DH. Effect of puerarin on the proliferation and differentiation of osteoblasts and expression of type I collagen mRNA in a high-glucose environment. Acta Endocronologica. 2020; 16: 288–294.
[34]
Zhang Y, Yan M, Yu Q, Yang P, Zhang H, Sun Y, et al. Puerarin Prevents LPS-Induced Osteoclast Formation and Bone Loss via Inhibition of Akt Activation. Biological & Pharmaceutical Bulletin. 2016; 39: 2028–2035.
[35]
Yang C, Li J, Zhu K, Yuan X, Cheng T, Qian Y, et al. Puerarin Exerts Protective Effects on Wear Particle-Induced Inflammatory Osteolysis. Frontiers in Pharmacology. 2019; 10: 1113.
[36]
Lin S, Ke D, Lin Y, Fu X, Yu Y. Puerarin inhibits the migration of osteoclast precursors and osteoclastogenesis by inhibiting MCP-1 production. Bioscience, Biotechnology, and Biochemistry. 2020; 84: 1455–1459.
[37]
Li J, Peng Y. Effect of puerarin on osteogenic differentiation of human periodontal ligament stem cells. Journal of International Medical Research. 2020; 48: 030006051985164.
[38]
Wang Y, Yin L, Li Y, Liu P, Cui Q. Preventive effects of puerarin on alcohol-induced osteonecrosis. Clinical Orthopaedics and Related Research. 2008; 466: 1059–1067.
[39]
Michihara S, Tanaka T, Uzawa Y, Moriyama T, Kawamura Y. Puerarin exerted anti-osteoporotic action independent of estrogen receptor-mediated pathway. Journal of Nutritional Science and Vitaminology. 2012; 58: 202–209.
[40]
Tanaka T, Tang H, Yu F, Michihara S, Uzawa Y, Zaima N, et al. Kudzu (Pueraria lobata) vine ethanol extracts improve ovariectomy-induced bone loss in female mice. Journal of Agricultural and Food Chemistry. 2011; 59: 13230–13237.
[41]
Liang J, Chen H, Pan W, Xu C. Puerarin inhibits caspase-3 expression in osteoblasts of diabetic rats. Molecular Medicine Reports. 2012; 5: 1419–1422.
[42]
Luo Y, Zheng S, Ding Y, Dai Y, Zhou Y, Xiang R, et al. Preventive effects of kudzu root on bone loss and cartilage degradation in ovariectomized rats. American Journal of Translational Research. 2017; 9: 5180.
[43]
Lim DW, Kim JG, Kim YT. Effects of dietary isoflavones from Puerariae radix on lipid and bone metabolism in ovariectomized rats. Nutrients. 2013; 5: 2734–2746.
[44]
Tang W, Xiao L, Ge G, Zhong M, Zhu J, Qin J, et al. Puerarin inhibits titanium particle‐induced osteolysis and RANKL‐induced osteoclastogenesis via suppression of the NF‐κB signaling pathway. Journal of Cellular and Molecular Medicine. 2020; 24: 11972–11983.
[45]
Li B, Liu H, Jia S. Puerarin enhances bone mass by promoting osteoblastogenesis and slightly lowering bone marrow adiposity in ovariectomized rats. Biological & Pharmaceutical Bulletin. 2014; 37: 1919–1925.
[46]
Lee M, Kim B, Lee Y, Park S, Shim J, Chung B, et al. Ameliorative Effects of Pueraria lobata Extract on Postmenopausal Symptoms through Promoting Estrogenic Activity and Bone Markers in Ovariectomized Rats. Evidence-Based Complementary and Alternative Medicine. 2021; 2021: 1–8.
[47]
Cho HJ, Jun H, Lee JH, Jia Y, Hoang MH, Shim J, et al. Acute effect of high-dose isoflavones from Pueraria lobata (Willd.) Ohwi on lipid and bone metabolism in ovariectomized mice. Phytotherapy Research. 2012; 26: 1864–1871.
[48]
Suthon S, Jaroenporn S, Charoenphandhu N, Suntornsaratoon P, Malaivijitnond S. Anti-osteoporotic effects of Pueraria candollei var. mirifica on bone mineral density and histomorphometry in estrogen-deficient rats. Journal of Natural Medicines. 2016; 70: 225–233.
[49]
Yang X, Zhang H, Wang J, Zhang Z, Li C. Puerarin decreases bone loss and collagen destruction in rats with ligature-induced periodontitis. Journal of Periodontal Research. 2015; 50: 748–757.
[50]
Liu H, Li W, Jia S, Li B. Puerarin and zinc additively prevent mandibular bone loss through inhibiting osteoclastogenesis in ovariectomized rats. Histology and Histopathology. 2017; 32: 851–860.
[51]
Yang X, Zheng H, Liu Y, Hao D, He B, Kong L. Puerarin for OVX-Induced Postmenopausal Osteoporosis in Murine Model: Systematic Review and Meta-Analysis. Current Stem Cell Research & Therapy. 2020; 15: 37–42.
[52]
Li B, Liu M, Wang Y, Gong S, Yao W, Li W, et al. Puerarin improves the bone micro-environment to inhibit OVX-induced osteoporosis via modulating SCFAs released by the gut microbiota and repairing intestinal mucosal integrity. Biomedicine & Pharmacotherapy. 2020; 132: 110923.
[53]
Urasopon N, Hamada Y, Asaoka K, Cherdshewasart W, Malaivijitnond S. Pueraria mirifica, a phytoestrogen-rich herb, prevents bone loss in orchidectomized rats. Maturitas. 2007; 56: 322–331.
[54]
Xiao L, Zhong M, Huang Y, Zhu J, Tang W, Li D, et al. Puerarin alleviates osteoporosis in the ovariectomy-induced mice by suppressing osteoclastogenesis via inhibition of TRAF6/ROS-dependent MAPK/NF-κB signaling pathways. Aging. 2020; 12: 21706–21729.
[55]
Guo C, Xie J, Hong R, Pan H, Zhang F, Liang Y. Puerarin alleviates streptozotocin (STZ)-induced osteoporosis in rats through suppressing inflammation and apoptosis via HDAC1/HDAC3 signaling. Biomedicine & Pharmacotherapy. 2019; 115: 108570.
[56]
Kim SY, Lee HJ, Kim T, Lee YG, Kwon JE, Kang SC. Anti-Osteoporotic Activity of Pueraria lobata Fermented with Lactobacillus paracasei JS1 by Regulation of Osteoblast Differentiation and Protection against Bone Loss in Ovariectomized Mice. Fermentation 2021; 7: 186.
Share
Back to top