Skip to main content

MINI REVIEW article

Front. Integr. Neurosci., 13 July 2022
This article is part of the Research Topic Horizons in Integrative Neuroscience 2022 View all 9 articles

Formononetin: A Pathway to Protect Neurons

  • 1The Second Clinical Medical School, Nanjing Medical University, Nanjing, China
  • 2Department of Physiology, Nanjing Medical University, Nanjing, China

Formononetin (FMN) is a phytoestrogen member of the flavonoid family, which has the pharmacological effects of antioxidative, antihypertensive, antitumor, and anti-infective. FMN demonstrates potential in the prevention and treatment of diseases, specifically neurological diseases, such as traumatic brain injury (TBI), spinal cord injury (SCI), ischemic stroke, cerebral ischemia-reperfusion, Alzheimer’s disease, and nerve tumor. Herein, a literature search is conducted to provide information on the signaling pathways of neuroprotection of formononetin based on the neuroprotective study. The significant neuroprotective function of FMN makes it a novel candidate for the development of drugs targeting the central nervous system.

Introduction

Formononetin (FMN) is a phytoestrogen member of the flavonoid family, which is the main active component of the legume red clover and one of the active components of commonly used Chinese herbal medicines such as Angelica sinensis, Astragalus membranaceus, Kudzu root, and Caulis spatholobi. The chemical molecular formula of FMN (C16H12O4) is shown in Figure 1. FMN can be obtained from natural plants by drug extraction and purification methods, such as extraction, ultrasound, microwave, supercritical water application, and pressurization, and can also be synthesized and modified by artificial methods. FMN has antioxidative, antihypertensive, antitumor, anti-infective, estrogen-like, and other pharmacological effects (Machado Dutra et al., 2021).

FIGURE 1
www.frontiersin.org

Figure 1. Structure of formononetin.

In the past few decades, FMN has been intensively investigated in the repair of tissues, following lung injury (Chen et al., 2021), kidney injury (Huang et al., 2017; Oza and Kulkarni, 2019; Lv et al., 2020), myocardial injury (Wang et al., 2020), atherosclerosis (Sun et al., 2013; Ma et al., 2020), peripheral nerve injury (Fang et al., 2020; Oza and Kulkarni, 2020), wound healing (Li et al., 2015), articular cartilage metabolism (Cho et al., 2019), and hair regeneration (Kim et al., 2016; El-Arabey and Abdalla, 2020). It is actively involved in antibacterial and antifungal activities against candida strains (das Neves et al., 2016), and also provides inhibitory effects on various malignant tumors, such as gastric cancer (Yao et al., 2019; Wang and Zhao, 2021), bladder cancer (Wu et al., 2017), and breast cancer (Zhou et al., 2019). In addition, FMN demonstrates potential in the prevention and treatment of diseases, specifically neurological diseases, such as Alzheimer’s disease (AD) and cerebral ischemia (Sun et al., 2011). Thus, we aim to introduce the central neuroprotective role and mechanism of FMN by a mini review.

Traumatic Brain Injury

Traumatic brain injury (TBI) is a brain injury caused by external force, and oxidative stress may be one of the main causes of TBI. Interestingly, growing evidence suggests that FMN has antioxidant properties. Therefore, FMN may have potential applications in the treatment of oxidative stress injury in TBI. Specifically, FMN may protect TBI rats from neurological damage, and the mechanism underlying this protection is related to the inhibition of intracranial inflammatory responses and oxidative stress (Li et al., 2014). Recently, FMN has been found to upregulate the expression of nuclear factor E2-related factor 2 (Nrf2) in TBI rats (Berry et al., 2009; Soltani et al., 2015; Engler-Chiurazzi et al., 2017). Li et al. (2017) found that FMN, a typical isoflavone phytoestrogen, improves brain dysfunction, reduces brain edema, and inhibits neuronal apoptosis. Moreover, the administration of FMN attenuated TBI-induced oxidative stress by upregulating heme oxygenase-1 (HO-1) expression and downregulating the BTB domain and CNC homolog 1 expression. This process increases the expression of miR-155, which suggests that FMN exerts neuroprotective effects by regulating key molecules related to the Nrf2/antioxidant response element [ARE]/HO-1 antioxidant pathway and regulating redox homeostasis in TBI rats. Accumulating evidence presents that the Nrf2/ARE/HO-1 pathway is involved in oxidative stress in secondary brain injury, following TBI (Figure 2; Cheng et al., 2016; Liu Z. et al., 2017). As a key regulator of ARE, Nrf2 activates transcription in response to oxidative stress; however, this process is significantly different between animal models and clinical treatments. To verify the relationship between the Nrf2/ARE/HO-1 pathway activation and FMN efficacy in the treatment of TBI, further studies should evaluate the mechanism of injury response and functional deficits (Figure 2).

FIGURE 2
www.frontiersin.org

Figure 2. The underlying mechanisms of FMN involved in TBI.

In addition, Li et al. (2014) suggested that TBI increases the levels of tumor necrosis factor-α (TNF-α) and interleukin (IL)-6 and upregulates cyclooxygenase-2 (COX-2) mRNA levels in damaged brain areas. The results are consistent with significant hydrocephalus in rats model. Therefore, the inflammatory response is believed to accelerate TBI deterioration. Following FMN treatment, a significant decrease was noted in the concentration of inflammatory cytokines in the damaged brain, accompanied by a corresponding decrease in COX-2 mRNA expression. The inhibition of COX-2 activity in astrocytes, neurons, and microglia by FMN effectively reduces intracranial inflammatory mediators, such as TNF-α and IL-6. The modulation of the inflammatory cascade in the central nervous system may be a therapeutic target for TBI. TBI results in significant cell necrosis or death in the cortex, which induces nerve injury (Li et al., 2018). In biological mechanisms, FMN-mediated activation of IL-10 expression promotes cell growth by binding to IL-10-specific receptors in cortical neurons, which subsequently inhibits cortical injury in TBI. Increased serum IL-10 levels with FMN treatment may be related to IL-10-based biological pathways in peripheral cortical neurons to counteract TBI conditions. These studies have demonstrated that FMN may have neuroprotective effects against TBI by inducing neuronal IL-10 expression. Collectively, FMN is implicated in suppressing inflammatory responses and oxidative stress in the brain to enhance neuroprotection, and, thus, FMN may be promising agent for the treatment of TBI.

Spinal Cord Injury

Spinal cord injury (SCI) has become a challenging chronic problem because of the interaction of multiple factors such as ischemia, oxidative stress, excitotoxicity, and immune-inflammatory response in the damaged local tissues. SCI results in neuronal and oligodendrocyte apoptosis and necrosis, peripheral syrinx, and glial scar formation in the damaged area, and limited regenerative capacity of the central nervous system. However, few effective strategies for promoting nerve regeneration in SCI have been established. Thus, the use of growth-permissive substrates is necessary at the injury site (Roman et al., 2011; Yamane et al., 2018).

Given that gelatin methacryloyl (GelMA) has excellent biocompatibility and mechanical properties, favoring cell adhesion and proliferation, it can be used for nerve tissue repair at diseased sites (Soucy et al., 2018). de Vasconcelos et al. (2020) used FMN to assess the functionalization process of carbon nanotubes (CNs) and developed a GelMA formulation, containing CN-FMN that was applied to spinal cord lesions by in situ photopolymerization. CN has been evaluated for the treatment of SCI because of its electrical properties and nanometer size. A photopolymerized preparation containing FMN-functionalized CNs was developed for in situ SCI treatment; thus, functionalized CNs can be incorporated into GelMA formulations. Constant hydrogel formation in situ after exposure to UV radiation makes this product a treatment option for SCI.

In addition, Novais et al. (2021) further developed a GelMA-hydroalcoholic extract of red propolis (HERP) formulation (the main bioactive maker of the formulation is FMN), and verified the function of the formulation in the experimental model of rat SCI. The results showed that the formulations containing FMN had greater post-injury recovery rates compared to formulations without FMN. Histomorphometric techniques demonstrated that the GelMA-HERP formulations treated tissue with less inflammation and less cavitation at the injury site.

Ischemic Stroke

In animal models of local and global cerebral ischemia, the dietary intake of phytoestrogens may reduce stroke injury (Lovekamp-Swan et al., 2007; Liang et al., 2008; Donzelli et al., 2010). However, the potential protective effects of phytoestrogens on ischemic injury are still unclear (Schreihofer and Redmond, 2009). Recent studies have shown that FMN has neuroprotective effects on ischemic stroke through its anti-apoptotic effects (Liang et al., 2014). FMN may protect against ischemic stroke by downregulating the Bax/Bcl-2 ratio, activating PI3K/AKT and anti-apoptosis, and reducing the TNF-α level (Li et al., 2014; Liang et al., 2014). However, whether FMN can restore neurological functions after ischemic stroke is still unknown.

Axons are components of neurons, and axonal growth promotes the differentiation of damaged neurons, establishes new connections with other neurons, forms neural networks, and restores impaired neurological function (Rao and Pearse, 2016). βIII-tubulin is a protein that makes up the neuronal skeleton, and the upregulation of βIII-tubulin expression can modify the axonal cytoskeleton, which is important for the regrowth of axons (Moskowitz and Oblinger, 1995). Growth-associated protein 43 (GAP-43) is a neuron-specific axonal membrane protein involved in the regulation of the growth and synaptic development of neurons (Donnelly et al., 2013). FMN upregulated the expressions of βIII-tubulin and GAP-43, indicating that it may induce neuronal differentiation. The nerve growth factor (NGF) is involved in the development, differentiation, growth, regeneration, and functional properties of neurons (Aloe et al., 2015). Synaptic connections between nerve cells are called synaptic plasticity and are the basis for neuronal regeneration and repair (Castrén and Antila, 2017). Brain-derived neurotrophic factor (BDNF) is typically involved in synaptic plasticity and promotes neuronal development, differentiation, growth, and regeneration (Begni et al., 2017). Wu et al. (2020) found FMN significantly increases expressions of NGF and BDNF, which may contribute to the beneficial effects of FMN on synaptic plasticity to restore neurological function.

It is worth noting that, although there are gender differences in stroke-induced injury and stroke repair, most of the current animal models are based on male rats, so future studies should further explore the gender differences in FMN treatment of stroke.

Cerebral Ischemia Reperfusion

Cerebral ischemia reperfusion (I/R) injury leads to a series of injuries, including cell death (necrosis and apoptosis), brain edema, and other cellular reactions, such as angiogenesis and reconstruction of functional microvessels to promote stroke recovery (Folkman, 1995; Beck and Plate, 2009; Chen et al., 2012). Vascular endothelial growth factor (VEGF)-signaling proteins are stimulated by ischemia and are essential for the angiogenesis and prevention of ischemic injury (Carmeliet, 2003). The upregulation of VEGF not only promotes angiogenesis but also increases microvascular permeability (Dvorak et al., 1995). Platelet endothelial cell adhesion molecule 1 (PECAM-1) is a member of the immunoglobulin gene superfamily of cell adhesion molecules and is composed of all contiguous human endothelial cells (Muller et al., 1992). PECAM-1 mediates cell-cell adhesion through homophilic and heterophilic interactions and transduces intracellular signals that upregulate integrin function on leukocytes (Delisser et al., 1997). At present, sodiumformononetin-3′-sulfonate (SUL-F) has been synthesized. SUL-F (3, 7.5, 15, and 30 mg/kg administered intravenously) was found to have protective effects against cerebral I/R injury by improving neurological function, inhibiting apoptosis, and increasing the expression levels of VEGF and PECAM-1 caused by middle cerebral artery occlusion (Figure 3). Treatment with SUL-F (10 and 20 μg/ml) significantly increased cell migration, angiogenesis, and VEGF and PECAM levels in human umbilical vein endothelial cells (Zhu et al., 2014). SUL-F was found to have significant neuroprotective effects on cerebral I/R injury in rats and could improve cerebral angiogenesis in human umbilical vein endothelial cells. The protective mechanism of SUL-F is attributed to the promotion of the expressions of VEGF and PECAM, inhibition of apoptosis, and improvement of cerebral angiogenesis.

FIGURE 3
www.frontiersin.org

Figure 3. Structure of SUL-F.

Estrogen has neuroprotective properties, and dietary intake of phytoestrogens can attenuate stroke injury in animal I/R models. To investigate the molecular mechanism underlying the neuroprotective effects of FMN on I/R rats, some studies have found that FMN significantly reduces the infarct volume and brain water content, improves neurological deficits, upregulates ER-α and p-Akt, and downregulates the Bax/Bcl-2 ratio, while FMN had little effects on p-ERK1/2 protein expressions (Figure 4; Liang et al., 2014). FMN demonstrated neuroprotective effects on cerebral I/R injury rats, and the molecular mechanism may be associated with the downregulation of the Bax/Bcl-2 ratio and the activation of the PI3K/Akt signaling pathway. FMN has some protective effects on I/R injury rats, and the mechanism may be related to anti-oxidative stress and anti-apoptosis. These results suggest the potential of FMN as an effective and promising agent for the treatment of cerebral I/R injury.

FIGURE 4
www.frontiersin.org

Figure 4. The underlying mechanisms of FMN involved in cerebral ischemia/reperfusion.

Alzheimer’s Disease

Alzheimer’s disease (AD) is the most common cause of dementia in the elderly population. Its main pathological features include the accumulation of extracellular amyloid-β (Aβ) plaques and intracellular neurofibrillary tangles (Bloom, 2014). Studies have established a strong correlation between AD symptoms and the accumulation of these plaques and tangles because they induce neurodegeneration that mediates memory loss and cognitive loss. Disruptions in the Aβ transport across the blood-brain barrier are key elements in the pathogenesis of AD. In the vascular endothelial cells of the hippocampus, Aβ transport is mainly mediated by low-density lipoprotein-associated protein 1 (LRP1) and advanced glycation end receptor (RAGE) products. Fei et al. (2018) investigated the effects of FMN on improving learning and memory in amyloid precursor protein (APP)/presenilin-1 mice and related mechanisms. They found that FMN significantly improved learning and memory by inhibiting APP processing to produce Aβ, RAGE-dependent inflammatory signaling, and promoting LRP1-dependent brain Aβ clearance pathways (Fei et al., 2018). Moreover, FMN treatment alleviated ultrastructural changes in hippocampal vascular endothelial cells. Chen et al. (2017) reported the protective effects of FMN on Aβ25-35-induced neurotoxicity in HT22 cells. FMN significantly increased the viability of HT22 cells and decreased the apoptosis rate when challenged with Aβ25-35 (Chen et al., 2017). The inhibitory effects of FMN were associated with the PI3K/Akt signaling pathway, as the PI3K inhibitor (LY294002) or the ERα-specific inhibitor prevented this effect (Figure 5). The form also accelerates the non-amyloidogenic process of amyloid protein (APP) by enhancing α-secretase activity and sAPPα release (Figure 5). Taken together, FMN may be an effective and promising treatment of AD.

FIGURE 5
www.frontiersin.org

Figure 5. The underlying mechanisms of FMN involved in AD.

Emerging evidence continues to demonstrate that neuroinflammation is also a major pathological component of AD (Webers et al., 2020). Several animal experimental and clinical studies have also provided a close link between neuroinflammation and AD pathogenesis. Accumulating evidence suggests that the inflammatory response in the brain is a major factor in AD pathogenesis (Heppner et al., 2015; Fu et al., 2019). High levels of proinflammatory mediators were detected in the brains of patients with AD (Hesse et al., 2016). Therefore, the anti-inflammatory effects of FMN have the potential to delay the disease onset or slow disease progression in AD. Diet-based FMN treatment can be applied for neuroprotection against oxidative stress and may reduce the incidence of neurodegenerative diseases. Future studies should investigate the activity of FMN in vivo and elucidate more details of the underlying mechanisms.

Nerve Tumor

For more than a decade, FMN has been intensively investigated because of its apoptosis-promoting and antiproliferative effects and potential as an anticancer agent. These anticancer properties have been observed in various cancer models, such as gastric cancer (Yao et al., 2019; Wang and Zhao, 2021), bladder cancer (Wu et al., 2017), and breast cancer (Zhou et al., 2019). In addition, FMN may also attenuate metastasis and tumor growth. Specifically, FMN may contribute to antiproliferative characteristics and cell cycle arrest-inducing properties of cells (Ong et al., 2019).

In recent years, increasing evidence has shown that most anticancer drugs are derived from natural products, such as including paclitaxel, vinblastine, and camptothecin (Sethi et al., 2012; Cragg and Pezzuto, 2016; Liu D. et al., 2017). For instance, polysaccharides obtained from Astragalus plants can counteract the adverse effects of chemotherapeutic drugs by significantly reducing myelosuppression in the patients with cancer (Duan and Wang, 2002). In addition to its antiproliferative and pro-apoptotic properties, FMN enhances the transcriptional activity of p53 by increasing its phosphorylation at Ser15 and Ser20, thereby upregulating the expression level of p53 in a concentration-dependent manner (Yang et al., 2014). For example, FMN exhibits synergy when combined with other chemotherapeutic agents. Temozolomide (TMZ) is an oral chemotherapeutic agent usually used in the treatment of certain brain cancers, such as glioblastoma multiforme. However, TMZ has undesirable side effects, including hematological complications and intrinsic and acquired resistance (van den Bent et al., 2003; Zhang et al., 2018). While FMN and TMZ alone can sufficiently inhibit the growth of C6 glioma cells in a concentration-dependent manner, they demonstrate a synergistic effect on C6 cells when they are used in combination. This drug combination increased Bax protein expressions, cleaved caspase-3 and caspase-9, attenuated Bcl-2 expression, and promoted tumor cell apoptosis (Zhang et al., 2018). As FMN has protective effects against certain malignancies, FMN has been clinically used as one of the basic herbs for the treatment of cancer in traditional medicine (Wang et al., 2018).

In conclusion, the beneficial effects of FMN can be attributed to its antiproliferative and cell cycle arrest-inducing properties. FMN regulates various transcription factors and growth factor-mediated oncogenic pathways, thereby mitigating possible causes of chronic inflammation associated with the survival of tumor cells and their resistance to chemotherapy.

Outlook

The evidence presented herein provides a comprehensive summary of the potential neuroprotective effects of FMN in in vitro and in vivo studies and the current progress in clinical research. Many molecular targets and mechanisms of action are involved in the protective properties of central nervous system diseases. Moreover, numerous in vitro studies have demonstrated that the safety and the efficacy of FMN and its metabolites in biological systems are further confirmed in in vivo studies. The potential role of FMN in prospective drug development is supported by these findings. However, further in vivo and clinical studies should assess the efficacy and safety of FMN in the prevention and treatment of various central nerve conditions. FMN derivatives and metabolites should have different kinetic properties and activities and be fully elucidated; thus, further studies should assess if this bioactive phytochemical is safe for clinical usage. The significant neuroprotective function of FMN makes it a novel candidate for the development of drugs targeting the central nervous system.

Author Contributions

JW: study concept and design. XM and JW: drafting of the manuscript. Both authors contributed to the article and approved the submitted version.

Conflict of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s Note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

Aloe, L., Rocco, M. L., Balzamino, B. O., and Micera, A. (2015). Nerve growth factor: a focus on neuroscience and therapy. Curr. Neuropharmacol. 13, 294–303. doi: 10.2174/1570159x13666150403231920

PubMed Abstract | CrossRef Full Text | Google Scholar

Beck, H., and Plate, K. H. (2009). Angiogenesis after cerebral ischemia. Acta Neuropathol. 117, 481–496. doi: 10.1007/s00401-009-0483-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Begni, V., Riva, M. A., and Cattaneo, A. (2017). Cellular and molecular mechanisms of the brain-derived neurotrophic factor in physiological and pathological conditions. Clin. Sci. (Lond). 131, 123–138. doi: 10.1042/CS20160009

PubMed Abstract | CrossRef Full Text | Google Scholar

Berry, C., Ley, E. J., Tillou, A., Cryer, G., Margulies, D. R., and Salim, A. (2009). The effect of gender on patients with moderate to severe head injuries. J. Trauma. 67, 950–953. doi: 10.1097/TA.0b013e3181ba3354

PubMed Abstract | CrossRef Full Text | Google Scholar

Bloom, G. S. (2014). Amyloid-β and tau: the trigger and bullet in Alzheimer disease pathogenesis. JAMA Neurol. 71, 505–508. doi: 10.1001/jamaneurol.2013.5847

PubMed Abstract | CrossRef Full Text | Google Scholar

Carmeliet, P. (2003). Angiogenesis in health and disease. Nat. Med. 9, 653–660. doi: 10.1038/nm0603-653

PubMed Abstract | CrossRef Full Text | Google Scholar

Castrén, E., and Antila, H. (2017). Neuronal plasticity and neurotrophic factors in drug responses. Mol. Psychiatry 22, 1085–1095. doi: 10.1038/mp.2017.61

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, L., Ou, S., Zhou, L., Tang, H., Xu, J., and Guo, K. (2017). Formononetin attenuates Aβ25-35-induced cytotoxicity in HT22 cells via PI3K/Akt signaling and non-amyloidogenic cleavage of APP. Neurosci. Lett. 639, 36–42. doi: 10.1016/j.neulet.2016.12.064

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, Y. C., Wu, J. S., Yang, S. T., Huang, C. Y., Chang, C., Sun, G. Y., et al. (2012). Stroke, angiogenesis and phytochemicals. Front. Biosci. (Schol Ed). 4:599–610. doi: 10.2741/s287

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, Y., Wei, D., Zhao, J., Xu, X., and Chen, J. (2021). Reduction of hyperoxic acute lung injury in mice by formononetin. PLoS One 16:e0245050. doi: 10.1371/journal.pone.0245050

PubMed Abstract | CrossRef Full Text | Google Scholar

Cheng, T., Wang, W., Li, Q., Han, X., Xing, J., Qi, C., et al. (2016). Cerebroprotection of flavanol (–)-epicatechin after traumatic brain injury via Nrf2-dependent and –independent pathways. Free Radic Biol. Med. 92, 15–28. doi: 10.1016/j.freeradbiomed.2015.12.027

PubMed Abstract | CrossRef Full Text | Google Scholar

Cho, I. A., Kim, T. H., Lim, H., Park, J. H., Kang, K. R., Lee, S. Y., et al. (2019). Formononetin antagonizes the interleukin-1β-induced catabolic effects through suppressing inflammation in primary rat chondrocytes. Inflammation 42, 1426–1440. doi: 10.1007/s10753-019-01005-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Cragg, G. M., and Pezzuto, J. M. (2016). Natural products as a vital source for the discovery of cancer chemotherapeutic and chemopreventive agents. Med. PrincPract. 25, 41–59. doi: 10.1159/000443404

PubMed Abstract | CrossRef Full Text | Google Scholar

das Neves, M. V., da Silva, T. M., Lima Ede, O., da Cunha, E. V., and Oliveira Ede, J. (2016). Isoflavone formononetin from red propolis acts as a fungicide against candida sp. Braz. J. Microbiol. 47, 159–166. doi: 10.1016/j.bjm.2015.11.009

PubMed Abstract | CrossRef Full Text | Google Scholar

de Vasconcelos, A. C. P., Morais, R. P., Novais, G. B., da Barroso, S., Menezes, L. R. O., Dos Santos, S., et al. (2020). In situ photocrosslinkable formulation of nanocomposites based on multi-walled carbon nanotubes and formononetin for potential application in spinal cord injury treatment. Nanomedicine 29:102272. doi: 10.1016/j.nano.2020.102272

PubMed Abstract | CrossRef Full Text | Google Scholar

Delisser, H. M., Baldwin, H. S., and Albelda, S. M. (1997). Platelet endothelial cell adhesion molecule 1 (PECAM-1/CD31): a multifunctional vascular cell adhesion molecule. Trends Cardiovasc Med. 7, 203–210. doi: 10.1016/S1050-1738(97)00049-2

CrossRef Full Text | Google Scholar

Donnelly, C. J., Park, M., Spillane, M., Yoo, S., Pacheco, A., Gomes, C., et al. (2013). Axonally synthesized β-actin and GAP-43 proteins support distinct modes of axonal growth. J. Neurosci. 33, 3311–3322. doi: 10.1523/JNEUROSCI.1722-12.2013

PubMed Abstract | CrossRef Full Text | Google Scholar

Donzelli, A., Braida, D., Finardi, A., Capurro, V., Valsecchi, A. E., Colleoni, M., et al. (2010). Neuroprotective effects of genistein in mongolian gerbils: estrogen receptor-β involvement. J. Pharmacol. Sci. 114, 158–167. doi: 10.1254/jphs.10164fp

PubMed Abstract | CrossRef Full Text | Google Scholar

Duan, P., and Wang, Z. M. (2002). [Clinical study on effect of astragalus in efficacy enhancing and toxicity reducing of chemotherapy in patients of malignant tumor]. Zhongguo Zhong Xi Yi Jie He Za Zhi. 22, 515–517.

PubMed Abstract | Google Scholar

Dvorak, H. F., Brown, L. F., Detmar, M., and Dvorak, A. M. (1995). Vascular permeability factor/vascular endothelial growth factor, microvascular hyperpermeability, and angiogenesis. Am. J. Pathol. 146, 1029–1039.

Google Scholar

El-Arabey, A. A., and Abdalla, M. (2020). Formononetin: novel deal for androgenetic alopecia. Arch. Med. Res. 51, 30–31. doi: 10.1016/j.arcmed.2019.12.001

PubMed Abstract | CrossRef Full Text | Google Scholar

Engler-Chiurazzi, E. B., Brown, C. M., Povroznik, J. M., and Simpkins, J. W. (2017). Estrogens as neuroprotectants: estrogenic actions in the context of cognitive aging and brain injury. Prog. Neurobiol. 157, 188–211. doi: 10.1016/j.pneurobio.2015.12.008

PubMed Abstract | CrossRef Full Text | Google Scholar

Fang, Y., Ye, J., Zhao, B., Sun, J., Gu, N., Chen, X., et al. (2020). Formononetin ameliorates oxaliplatin-induced peripheral neuropathy via the KEAP1-NRF2-GSTP1 axis. Redox Biol. 36:101677. doi: 10.1016/j.redox.2020.101677

PubMed Abstract | CrossRef Full Text | Google Scholar

Fei, H. X., Zhang, Y. B., Liu, T., Zhang, X. J., and Wu, S. L. (2018). Neuroprotective effect of formononetin in ameliorating learning and memory impairment in mouse model of Alzheimer’s disease. Biosci. Biotechnol. Biochem. 82, 57–64. doi: 10.1080/09168451.2017.1399788

PubMed Abstract | CrossRef Full Text | Google Scholar

Folkman, J. (1995). Angiogenesis in cancer, vascular, rheumatoid and other disease. Nat. Med. 1, 27–31. doi: 10.1038/nm0195-27

PubMed Abstract | CrossRef Full Text | Google Scholar

Fu, W. Y., Wang, X., and Ip, N. Y. (2019). Targeting neuroinflammation as a therapeutic strategy for Alzheimer’s disease: mechanisms. Drug Can. New Opport. ACS Chem. Neurosci. 10, 872–879. doi: 10.1021/acschemneuro.8b00402

PubMed Abstract | CrossRef Full Text | Google Scholar

Heppner, F. L., Ransohoff, R. M., and Becher, B. (2015). Immune attack: the role of inflammation in Alzheimer disease. Nat. Rev. Neurosci. 16, 358–372. doi: 10.1038/nrn3880

PubMed Abstract | CrossRef Full Text | Google Scholar

Hesse, R., Wahler, A., Gummert, P., Kirschmer, S., Otto, M., Tumani, H., et al. (2016). Decreased IL-8 levels in CSF and serum of AD patients and negative correlation of MMSE and IL-1β. BMC Neurol. 16:185. doi: 10.1186/s12883-016-0707-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Huang, D., Wang, C., Duan, Y., Meng, Q., Liu, Z., Huo, X., et al. (2017). Targeting Oct2 and P53: formononetin prevents cisplatin-induced acute kidney injury. Toxicol Appl. Pharmacol. 326, 15–24. doi: 10.1016/j.taap.2017.04.013

PubMed Abstract | CrossRef Full Text | Google Scholar

Kim, M. H., Choi, Y. Y., Lee, J. E., Kim, K., and Yang, W. M. (2016). Topical treatment of hair loss with formononetin by modulating apoptosis. Planta Med. 82, 65–69. doi: 10.1055/s-0035-1557897

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, S., Dang, Y., Zhou, X., Huang, B., Huang, X., Zhang, Z., et al. (2015). Formononetin promotes angiogenesis through the estrogen receptor alpha-enhanced ROCK pathway. Sci. Rep. 5:16815. doi: 10.1038/srep16815

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, Z., Dong, X., Zhang, J., Zeng, G., Zhao, H., Liu, Y., et al. (2014). Formononetin protects TBI rats against neurological lesions and the underlying mechanism. J. Neurol. Sci. 338, 112–117. doi: 10.1016/j.jns.2013.12.027

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, Z., Wang, Y., Zeng, G., Zheng, X., Wang, W., Ling, Y., et al. (2017). Increased miR-155 and heme oxygenase-1 expression is involved in the protective effects of formononetin in traumatic brain injury in rats. Am. J. Transl Res. 9, 5653–5661.

PubMed Abstract | Google Scholar

Li, Z., Zeng, G., Zheng, X., Wang, W., Ling, Y., Tang, H., et al. (2018). Neuroprotective effect of formononetin against TBI in rats via suppressing inflammatory reaction in cortical neurons. Biomed. Pharmacother. 106, 349–354. doi: 10.1016/j.biopha.2018.06.041

PubMed Abstract | CrossRef Full Text | Google Scholar

Liang, H. W., Qiu, S. F., Shen, J., Sun, L. N., Wang, J. Y., Bruce, I. C., et al. (2008). Genistein attenuates oxidative stress and neuronal damage following transient global cerebral ischemia in rat hippocampus. Neurosci. Lett. 438, 116–120. doi: 10.1016/j.neulet.2008.04.058

PubMed Abstract | CrossRef Full Text | Google Scholar

Liang, K., Ye, Y., Wang, Y., Zhang, J., and Li, C. (2014). Formononetin mediates neuroprotection against cerebral ischemia/reperfusion in rats via downregulation of the Bax/Bcl-2 ratio and upregulation PI3K/Akt signaling pathway. J. Neurol. Sci. 344, 100–104. doi: 10.1016/j.jns.2014.06.033

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, D., Miao, H., Zhao, Y., Kang, X., Shang, S., Xiang, W., et al. (2017). NF-κB potentiates tumor growth by suppressing a novel target LPTS. Cell Commun. Signal. 15:39. doi: 10.1186/s12964-017-0196-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, Z., Wang, H., Shi, X., Li, L., Zhou, M., Ding, H., et al. (2017). DL-3-n-butylphthalide (NBP) provides neuroprotection in the mice models after traumatic brain injury via Nrf2-ARE signaling pathway. Neurochem. Res. 42, 1375–1386. doi: 10.1007/s11064-017-2186-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Lovekamp-Swan, T., Glendenning, M., and Schreihofer, D. A. (2007). A high soy diet reduces programmed cell death and enhances bcl-xL expression in experimental stroke. Neuroscience 148, 644–652. doi: 10.1016/j.neuroscience.2007.06.046

PubMed Abstract | CrossRef Full Text | Google Scholar

Lv, J., Zhuang, K., Jiang, X., Huang, H., and Quan, S. (2020). Renoprotective effect of formononetin by suppressing smad3 expression in Db/Db mice. Diabetes MetabSyndrObes 13, 3313–3324. doi: 10.2147/DMSO.S272147

PubMed Abstract | CrossRef Full Text | Google Scholar

Ma, C., Xia, R., Yang, S., Liu, L., Zhang, J., Feng, K., et al. (2020). Formononetin attenuates atherosclerosis via regulating interaction between KLF4 and SRA in apoE-/- mice. Theranostics 10, 1090–1106. doi: 10.7150/thno.38115

PubMed Abstract | CrossRef Full Text | Google Scholar

Machado Dutra, J., Espitia, P. J. P., and Andrade Batista, R. (2021). Formononetin: biological effects and uses – a review. Food Chem. 359:129975. doi: 10.1016/j.foodchem.2021.129975

PubMed Abstract | CrossRef Full Text | Google Scholar

Moskowitz, P. F., and Oblinger, M. M. (1995). Sensory neurons selectively upregulate synthesis and transport of the beta III-tubulin protein during axonal regeneration. J. Neurosci. 15, 1545–1555. doi: 10.1523/JNEUROSCI.15-02-01545.1995

PubMed Abstract | CrossRef Full Text | Google Scholar

Muller, W. A., Berman, M. E., Newman, P. J., DeLisser, H. M., and Albelda, S. M. (1992). A heterophilic adhesion mechanism for platelet/endothelial cell adhesion molecule 1 (CD31). J. Exp. Med. 175, 1401–1404. doi: 10.1084/jem.175.5.1401

PubMed Abstract | CrossRef Full Text | Google Scholar

Novais, G. B., Dos Santos, S., Santana, R. J. R., Filho, R. N. P., Cunha, J. L. S., Lima, B. S., et al. (2021). Development of a new formulation based on in situ photopolymerized polymer for the treatment of spinal cord injury. Polymers (Basel). 13:4274. doi: 10.3390/polym13244274

PubMed Abstract | CrossRef Full Text | Google Scholar

Ong, S. K. L., Shanmugam, M. K., Fan, L., Fraser, S. E., Arfuso, F., Ahn, K. S., et al. (2019). Focus on formononetin: anticancer potential and molecular targets. Cancers (Basel). 11:611. doi: 10.3390/cancers11050611

PubMed Abstract | CrossRef Full Text | Google Scholar

Oza, M. J., and Kulkarni, Y. A. (2019). Formononetin attenuates kidney damage in type 2 diabetic rats. Life Sci. 219, 109–121. doi: 10.1016/j.lfs.2019.01.013

PubMed Abstract | CrossRef Full Text | Google Scholar

Oza, M. J., and Kulkarni, Y. A. (2020). Formononetin ameliorates diabetic neuropathy by increasing expression of SIRT1 and NGF. Chem. Biodivers. 17:e2000162. doi: 10.1002/cbdv.202000162

PubMed Abstract | CrossRef Full Text | Google Scholar

Rao, S. N., and Pearse, D. D. (2016). Regulating axonal responses to injury: the intersection between signaling pathways involved in axon myelination and the inhibition of axon regeneration. Front. Mol. Neurosci. 9:33. doi: 10.3389/fnmol.2016.00033

PubMed Abstract | CrossRef Full Text | Google Scholar

Roman, J. A., Niedzielko, T. L., Haddon, R. C., Parpura, V., and Floyd, C. L. (2011). Single-walled carbon nanotubes chemically functionalized with polyethylene glycol promote tissue repair in a rat model of spinal cord injury. J. Neurotrauma. 28, 2349–2362. doi: 10.1089/neu.2010.1409

PubMed Abstract | CrossRef Full Text | Google Scholar

Schreihofer, D. A., and Redmond, L. (2009). Soy phytoestrogens are neuroprotective against stroke-like injury in vitro. Neuroscience 158, 602–609. doi: 10.1016/j.neuroscience.2008.10.003

PubMed Abstract | CrossRef Full Text | Google Scholar

Sethi, G., Shanmugam, M. K., Ramachandran, L., Kumar, A. P., and Tergaonkar, V. (2012). Multifaceted link between cancer and inflammation. Biosci. Rep. 32, 1–15. doi: 10.1042/BSR20100136

PubMed Abstract | CrossRef Full Text | Google Scholar

Soltani, Z., Khaksari, M., Jafari, E., Iranpour, M., and Shahrokhi, N. (2015). Is genistein neuroprotective in traumatic brain injury? Physiol. Behav. 152, 26–31. doi: 10.1016/j.physbeh.2015.08.037

PubMed Abstract | CrossRef Full Text | Google Scholar

Soucy, J. R., Shirzaei, Sani E, Portillo, Lara R, Diaz, D., Dias, F., Weiss, A. S., et al. (2018). Photocrosslinkable gelatin/tropoelastin hydrogel adhesives for peripheral nerve repair. Tissue Eng. Part A. 24, 1393–1405. doi: 10.1089/ten.TEA.2017.0502

PubMed Abstract | CrossRef Full Text | Google Scholar

Sun, T., Liu, R., and Cao, Y. X. (2011). Vasorelaxant and antihypertensive effects of formononetin through endothelium-dependent and –independent mechanisms. Acta Pharmacol. Sin. 32, 1009–1018. doi: 10.1038/aps.2011.51

PubMed Abstract | CrossRef Full Text | Google Scholar

Sun, T., Wang, J., Huang, L. H., and Cao, Y. X. (2013). Antihypertensive effect of formononetin through regulating the expressions of eNOS, 5-HT2A/1B receptors and α1-adrenoceptors in spontaneously rat arteries. Eur. J. Pharmacol. 699, 241–249. doi: 10.1016/j.ejphar.2012.10.031

PubMed Abstract | CrossRef Full Text | Google Scholar

van den Bent, M. J., Taphoorn, M. J., Brandes, A. A., Menten, J., Stupp, R., Frenay, M., et al. (2003). European organization for research and treatment of cancer brain tumor group. phase II study of first-line chemotherapy with temozolomide in recurrent oligodendroglial tumors: the european organization for research and treatment of cancer brain tumor group study 26971. J. Clin. Oncol. 21, 2525–2528. doi: 10.1200/JCO.2003.12.015

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, A. L., Li, Y., Zhao, Q., and Fan, L. Q. (2018). Formononetin inhibits colon carcinoma cell growth and invasion by microRNA 149 mediated EphB3 downregulation and inhibition of PI3K/AKT and STAT3 signaling pathways. Mol. Med. Rep. 17, 7721–7729. doi: 10.3892/mmr.2018.8857

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, D. S., Yan, L. Y., Yang, D. Z., Lyu, Y., Fang, L. H., Wang, S. B., et al. (2020). Formononetin ameliorates myocardial ischemia/reperfusion injury in rats by suppressing the ROS-TXNIP-NLRP3 pathway. Biochem. Biophys. Res. Commun. 525, 759–766. doi: 10.1016/j.bbrc.2020.02.147

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, W. S., and Zhao, C. S. (2021). Formononetin exhibits anticancer activity in gastric carcinoma cell and regulating miR-542-5p. Kaohsiung J. Med. Sci. 37, 215–225. doi: 10.1002/kjm2.12322

PubMed Abstract | CrossRef Full Text | Google Scholar

Webers, A., Heneka, M. T., and Gleeson, P. A. (2020). The role of innate immune responses and neuroinflammation in amyloid accumulation and progression of Alzheimer’s disease. Immunol. Cell Biol. 98, 28–41. doi: 10.1111/imcb.12301

PubMed Abstract | CrossRef Full Text | Google Scholar

Wu, Q. L., Cheng, Y. Q., Liu, A. J., and Zhang, W. D. (2020). Formononetin recovered injured nerve functions by enhancing synaptic plasticity in ischemic stroke rats. Biochem. Biophys. Res. Commun. 17:15. doi: 10.1016/j.bbrc.2020.02.015

PubMed Abstract | CrossRef Full Text | Google Scholar

Wu, Y., Zhang, X., Li, Z., Yan, H., Qin, J., and Li, T. (2017). Formononetin inhibits human bladder cancer cell proliferation and invasiveness via regulation of miR-21 and PTEN. Food Funct. 8, 1061–1066. doi: 10.1039/c6fo01535b

PubMed Abstract | CrossRef Full Text | Google Scholar

Yamane, K., Mazaki, T., Shiozaki, Y., Yoshida, A., Shinohara, K., Nakamura, M., et al. (2018). Collagen-binding hepatocyte growth factor (HGF) alone or with a gelatin- furfurylamine hydrogel enhances functional recovery in mice after spinal cord injury. Sci. Rep. 8:917. doi: 10.1038/s41598-018-19316-y

PubMed Abstract | CrossRef Full Text | Google Scholar

Yang, Y., Zhao, Y., Ai, X., Cheng, B., and Lu, S. (2014). Formononetin suppresses the proliferation of human non-small cell lung cancer through induction of cell cycle arrest and apoptosis. Int. J. Clin. Exp. Pathol. 7, 8453–8461.

PubMed Abstract | Google Scholar

Yao, J. N., Zhang, X. X., Zhang, Y. Z., Li, J. H., Zhao, D. Y., Gao, B., et al. (2019). Discovery and anticancer evaluation of a formononetin derivative against gastric cancer SGC7901 cells. Invest. New Drugs. 37, 1300–1308. doi: 10.1007/s10637-019-00767-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, X., Ni, Q., Wang, Y., Fan, H., and Li, Y. (2018). Synergistic anticancer effects of formononetin and temozolomide on glioma C6 cells. Biol. Pharm. Bull. 41, 1194–1202. doi: 10.1248/bpb.b18-00002

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhou, Q., Zhang, W., Li, T., Tang, R., Li, C., Yuan, S., et al. (2019). Formononetin enhances the tumoricidal effect of everolimus in breast cancer MDA-MB-468 cells by suppressing the mTOR pathway. Evid. Based Complement Alternat Med. 2019:9610629. doi: 10.1155/2019/9610629

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhu, H., Zou, L., Tian, J., Lin, F., He, J., and Hou, J. (2014). Protective effects of sulphonated formononetin in a rat model of cerebral ischemia and reperfusion injury. Planta Med. 80, 262–268. doi: 10.1055/s-0033-1360340

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: CNS, formononetin, neuroprotecion, signaling pathway, molecular target

Citation: Ma X and Wang J (2022) Formononetin: A Pathway to Protect Neurons. Front. Integr. Neurosci. 16:908378. doi: 10.3389/fnint.2022.908378

Received: 30 March 2022; Accepted: 14 June 2022;
Published: 13 July 2022.

Edited by:

Joseph F. X. DeSouza, York University, Canada

Reviewed by:

Manisha Jignesh Oza, SVKM’s Dr. Bhanuben Nanavati College of Pharmacy, India
Sumana Chakravarty, Indian Institute of Chemical Technology (CSIR), India

Copyright © 2022 Ma and Wang. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Juejin Wang, juejinwang@njmu.edu.cn

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.