Skip to main content

REVIEW article

Front. Pharmacol., 22 October 2020
Sec. Ethnopharmacology

Pharmacological Activities of Psoralidin: A Comprehensive Review of the Molecular Mechanisms of Action

  • 1Phytochemistry Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
  • 2Science and Technology Application and Research Center (BITAUM), Bursa Uludag University, Bursa, Turkey
  • 3Faculty of Chemistry and Chemical Technology, Al-Farabi Kazakh National University, Almaty, Kazakhstan
  • 4Department of Medicinal and Aromatic Plants, University of Health Sciences, Istanbul, Turkey
  • 5Department of Chemistry, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, India
  • 6Medical Ethics and Law Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
  • 7Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, Craiova, Romania
  • 8Department of Toxicology, University of Medicine and Pharmacy of Craiova, Craiova, Romania
  • 9Faculty of Allied Health Sciences, University Institute of Diet and Nutritional Sciences, The University of Lahore, Lahore, Pakistan
  • 10Laboratorio de Análisis Químico, Departamento de Producción Vegetal, Facultad de Agronomía, Universidad de Concepción, Concepción, Chile
  • 11Department of Botany, University of Fort Hare, Alice, South Africa
  • 12Department of Nutrition and Dietetics, Faculty of Pharmacy, Centre for Healthy Living, University of Concepción, Concepción, Chile
  • 13Unidad de Desarrollo Tecnológico, UDT, Universidad de Concepción, Concepción, Chile

Analysis of the most relevant studies on the pharmacological properties and molecular mechanisms of psoralidin, a bioactive compound from the seeds of Cullen corylifolium (L.) Medik. confirmed its complex therapeutic potential. In the last years, the interest of the scientific community regarding psoralidin increased, especially after the discovery of its benefits in estrogen-related diseases and as a chemopreventive agent. Growing preclinical pieces of evidence indicate that psoralidin has anticancer, antiosteoporotic, anti-inflammatory, anti-vitiligo, antibacterial, antiviral, and antidepressant-like effects. Here, we provide a comprehensive and critical review of psoralidin on its bioavailability, pharmacological activities with focus on molecular mechanisms and cell signaling pathways. In this review, we conducted literature research on the PubMed database using the following keywords: “Psoralidin” or “therapeutic effects” or “biological activity” or “Cullen corylifolium” in order to identify relevant studies regarding PSO bioavailability and mechanisms of therapeutic effects in different diseases based on preclinical, experimental studies. In the light of psoralidin beneficial actions for human health, this paper gathers complete information on its pharmacotherapeutic effects and opens new natural therapeutic perspectives in chronic diseases.

Introduction

Used since ancient times in traditional medicine, Cullen corylifolium (L.) Medik. (syn. Psoralea corylifolia L.) has proven its therapeutic properties over the centuries. One of the main bioactive compounds is psoralidin (PSO), a coumestan derivative with a lot of pharmacological properties (Chopra et al., 2013). A diversity of pharmacological activities of PSO compound has been identified and described. PSO exerts therapeutic effects on cardiovascular and inflammatory diseases (Chopra et al., 2013; Yan et al., 2015).

PSO has a lot of beneficial health effects as an antibacterial against gram-negative and gram-positive germs (Borate et al., 2014), antioxidant (Xiao et al., 2010), anti-inflammatory modulating the inhibition of proinflammatory cytokines (Lee et al., 2012), antimycobacterial (Khushboo et al., 2010), antipsoriatic (Dwarampudi et al., 2012), and antidepressant (Yi et al., 2008). Besides, PSO possesses osteoblast proliferation-stimulating effects (Larsson and Fazzalari, 2014), estrogenic-like effects (Xin et al., 2019), and antitumor properties (Szliszka et al., 2011; Luo et al., 2013; Pal et al., 2017a; Badarau et al., 2019; Rusu et al., 2019). PSO is a natural phenolic coumarin in the same class with isopsoralen, psoralen, bakuchiol, bakuchalcone, flavones, and bavachinin (Zhang et al., 2016) isolated and identified from the seeds of the medicinal plant Cullen corylifolium. The plant is widely distributed in different regions of Asia, India and Europe (Zhang et al., 2019). PSO is a coumestan derivative with an isopentenyl group at the second carbon position of coumestrol (Liu et al., 2014) (Figure 1).

FIGURE 1
www.frontiersin.org

FIGURE 1. Chemical structure of psoralidin (3,9-dihydroxy-2-prenyl-coumestan).

The insolubility of PSO in water is one of the main reason why in vivo studies are difficult to be conducted (Hao et al., 2014). However, dozens of studies have been performed (Figure 2) since 1961, when the first report of the structure of PSO was published by Khastgir (Xin et al., 2019). Initially, the studies focused on the evaluation of PSO cytotoxicity, but further in vivo showed its efficacy in preventing or reducing the progression of many pathologies.

FIGURE 2
www.frontiersin.org

FIGURE 2. Psoralidin citations that appear along the time from 1961 till 2019. The Pubmed database was searched by the use of “psoralidin” as a keyword. The plot shows the cumulative number of hits identified for each year after the first report of psoralidin (PSO).

The literature available in the PubMed database was researched for ethnomedicine uses, phytochemistry, pharmacological molecular mechanisms and PSO bioavailability by applying the following keywords: “Psoralea corylifolium,” “Cullen corylifolium,” “phytochemistry,” “bioavailability,” “anticancer,” “Anti-osteoporosis,” “Anti-inflammatory,” “antibacterial,” “SARS-CoV antiviral,” “neuro-protective,” “anti-vitiligo,” “antiprotozoal,” “vasodilatory,” “anti-apoptotic,” “antidepressant-like effects,” and “mechanism of action” their titles corresponding to the medical subject (MeSH) using OR/AND conjunctions. The research focused on studies in ethnomedicine, phytochemical, preclinical and clinical reports available to understand the mechanisms of action. Searches were limited to works in extenso in English and did not include studies with homeopathic preparations.

Very few clinical studies have been found in the literature on the efficacy of PSO, so more clinical research is needed in the future. Highlighting the molecular pharmacological mechanisms of PSO is the strength of this study because it can lead to the development of useful pharmaceutical formulations in the treatment of many chronic diseases.

Bioavailability of Psoralidin

The bioavailability reflects the real available concentration at the site of drug action after absorption of these compounds from the gastrointestinal tract following oral administration of a dosage form (solution, suspension, tablet, capsule, powder, or elixir) (Chow, 2014; Salehi et al., 2019b). The percentage of a medicinal product absorbed unmodified in the systemic circulation represent the bioavailability of that product (Pandareesh et al., 2015; Salehi et al., 2020a).

PSO is an orally bioavailable natural compound. New pharmaceutical forms such as nanoencapsulation have been developed to increase the reduced bioavailability of PSO (Yi et al., 2008; Chen et al., 2017; Zhang et al., 2019). The results of Yin et al. (2016) demonstrate that nanoencapsulation of PSO (via chitosan and Eudragit S100) increased the oral delivery efficacy. By analyzing the pharmacokinetic profiles and some non-pharmacokinetic facings by the non-compartmental model of some suspensions of PSO and PSO-nanoencapsulated, it was found that the blood concentrations of the nanoencapsulated pharmaceutical forms are much higher at all moments of determination. The maximum concentration of nanoencapsulated PSO was 943 ng/ml compared to the simple PSO suspension whose concentration was 357 ng/ml, proving that nanoencapsulation favors the increased oral absorption of PSO more than conventional suspensions. Moreover, the peak time and the half-time remained the same, not being modified. The bioavailability of PSO in nanoencapsulated pharmaceutical forms was 339.02% higher compared to the control, a simple suspension with PSO. As a result, PSO pharmaceutical nanoformulations with chitosan and Eudragit S100 compounds have shown great potential for improving PSO bioavailability. It is still a need for further studies to improve the bioavailability of this promising compound.

Pharmacological Importance of Psoralidin: In Vitro and In Vivo Studies

The studies have proven that PSO has the next pharmacological properties: antidepressant (Farahani et al., 2015), modulator of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) signaling pathway (Rana et al., 2014) and it is an potential estrogen receptor modulator (Yang et al., 2019). The main therapeutic properties of PSO and its mechanisms of action are summarized in Figure 3.

FIGURE 3
www.frontiersin.org

FIGURE 3. Summarized scheme of the main pharmacological activities of psoralidin. ↑, increase; ↓, decrease; NF-κB, nuclear factor kappa beta; COX, cyclooxygenase; PGE2, prostaglandin E2.

In Vitro Studies

Cancer is one of the most feared diseases, and the fight against it has concentrated the efforts of thousands of medical researchers (Mishra et al., 2018; Salehi et al., 2019a). For hundreds of years, plants with anticancer effects have succeeded in improving the condition of the patients and, in some cases, to stimulate defense mechanisms of the body against certain types of cancer (Sharifi-Rad et al., 2020). It is not surprising that in recent years researchers have focused on the thesaurus of medicinal plant substances and have conducted research to identify compounds that are effective in anticancer therapies (Sani et al., 2017; Salehi et al., 2019a). PSO has been used in many in vitro models to evaluate its anticancerous effects and the underlying mechanisms (Xin et al., 2019). Determination of cytotoxicity after PSO contact with cells is done by calculating the percentage of dead cells and IC50 or the concentration of PSO that inhibited by 50% cell death (He et al., 2016).

PSO promotes cellular apoptosis and inhibits the cancer cells proliferation in prostate cancer (Szliszka et al., 2011; Das et al., 2014); breast cancer (Dwarampudi et al., 2012); liver cancer (Luo et al., 2013); colon cancer (Jin et al., 2016a); esophagal carcinoma (Jin et al., 2016b), or modulate the autophagy in the lung (Hao et al., 2014; Xin et al., 2019).

According to various studies, PSO has been recognized to inhibit forskolin-induced corticotrophin-releasing factor gene transcription (Chen et al., 2008). In a recent survey, it has shown potent in vitro activity against colon, gastric, breast and prostate cancer lines. It has been shown to exhibit a substantial effect on inhibiting protein tyrosine phosphatase 1B, a crucial metabolite involved in insulin signaling (Ren et al., 2019).

Different studies were conducted on the cytotoxicity of PSO suggested that it has shown the potential to induce cytotoxicity against breast (MCF-7) cancer, colon (HT-29), and gastric (SNU-1, SNU-16) cells. In prostate cancer cells, androgen-independent (DU-145, PC-3) androgen-dependent (LNCaP, C4-2B), the treatment with PSO also caused apoptosis. The growth of PC-3 xenograft tumor was also inhibited in nude mice by PSO (Srinivasan et al., 2010; Chiou et al., 2011; Hao et al., 2014).

Studies have been shown that PSO has a benefic role against diabetes complications, oxidative stress, obesity, osteoporosis, as well as on cell proliferation, autophagy, and apoptosis (Lim et al., 2011). PSO is implicated in the modulation of the autophagy process and inhibiting cadmium-transformed prostate epithelial cell xenograft growth thought placenta-specific 8 inhibition expression, reducing the levels nuclear factor kappa B (NF-κB) and B-cell lymphoma-2 (Bcl-2) expression and enhancing apoptotic gene expression (Pal et al., 2017a). Moreover, PSO has been found to decrease blood glucose, improve glucose tolerance and increase serum insulin levels (Seo et al., 2014).

PSO also has been found to promote the mineralization, suppress the adipocytes formation, increase the alkaline phosphatase activity, calcium nodule formation, and osteocalcin levels in bone marrow mesenchymal stem cells (BMSCs) whereas also down-regulated adipogenesis related factors. Moreover, PSO regulates the protein kinase B/glycogen synthase kinase 3β/β-catenin signaling pathway during the suppression of adipocyte differentiation (Cao et al., 2019). PSO significantly decreases cell proliferation and promotes apoptosis of Eca9706 cells in a dose-dependent manner. Moreover, it suppresses caspase-3 and NF-κB activities and also decreases the phosphatidylinositol-3-kinase (PI3K) and protein kinase B (Akt) protein expression dose-dependently (Jin et al., 2016b). Other in vitro studies on cell lines reported that PSO binds to both types of alpha and beta estrogen receptors, activating the classic estrogen receptor signaling pathway, thus explaining the effect of reducing bone loss as well as the antioxidant action (Liu et al., 2014). Furthermore, a very recent study has shown the dual beneficial antiosteoporotic effect of PSO by inhibiting adipogenesis and stimulating osteogenesis (Cao et al., 2019).

Researchers have used a mouse model of depression, and PSO has shown positive results in the forced swim test. 5-Hydroxyindole acetic acid and 5-hydroxytryptamine levels were significantly increased in the brain after PSO treatment, along with the changes in dopamine levels. Serum corticosterone, adrenal corticotropin-releasing hormone and corticotropin-releasing factor, hormones involved in stress control in mice, were reduced by PSO (Yi et al., 2008).

In Vivo Studies

According to the animal studies carried out in till now, PSO has shown a variety of biological activities including antidepressant (Yi et al., 2008), anti-inflammatory (Yang et al., 2011), anti-osteoclastogenic (Kong et al., 2017), and anti-osteoporotic (Zhai et al., 2018) (Table 1).

TABLE 1
www.frontiersin.org

TABLE 1. The most relevant information, including the preclinical pharmacological activities and mechanism of action of PSO.

Yi et al. (2008) investigated the effect of PSO against depression in male mice. In this study, the forced swimming test was used to screen the antidepressant effect of PSO. PSO in doses 20, 40, and 60 mg/kg was administered to mice using as vehicle water 15 ml/kg by gastric gavage. The results revealed that administration of PSO significantly decreased immobility time and increased swimming behavior without altering climbing behavior in the mouse forced swimming test. Locomotor activity studied by the open-field test was not affected by PSO.

Moreover, the levels of monoamine neurotransmitters involved in the pathophysiology of depression as 5-hydroxytryptamine and 5-hydroxyindoleacetic acid (5-HIAA, the main metabolite of serotonin) increased significantly in various brain regions after PSO treatment. In mice exposed to forced swimming test, striatum dopamine levels were also changed. In mice, PSO also suppressed the increased level of serum corticotropin-releasing factor, adrenal corticotropin-releasing hormone and corticosterone concentrations induced by swimming stress. Overall, the results obtained in this study suggested that PSO antidepressant properties are mediated via the hypothalamic-pituitary-adrenal axis and monoamine neurotransmitter systems (Yi et al., 2008).

Inflammation acts as a significant limiting factor in the radiotherapy (Buga et al., 2019). In a study carried out a few years ago (Yang et al., 2011), the PSO (5 mg/kg) anti-inflammatory effect was investigated in IR-irradiated mouse lung (20 Gy at a dose rate of 0.81 Gy/min, for 12 h or 1 week). Twelve hours after treatment, PSO suppressed the radiation-increased mRNA levels of TNF-α, TGF-β, and ICAM-1 by half, and inhibited IL-6 and IL-1 α/β by one quarter. The PSO inhibitory effects were found to be more effective 1 week after irradiation. In conclusion, data obtained in this study suggested that PSO may regulate the inflammation process in the ionizing radiation-irradiated lung (Yang et al., 2011).

Regarding the osteoporosis, it occurs primarily in women with premature menopause, due to estrogen hormone deficiency (Salehi et al., 2020b). In the absence of this hormone, disorders occur in the assimilation and fixation of calcium which leads to weakening of the bones over time (Larsson and Fazzalari, 2014). Hormone replacement therapy has minimal indications due to severe side effects (thromboembolism, stroke, endometrial cancer, breast cancer, and cardiovascular disease) (Tsatsakis et al., 2019). Therefore, research has focused on finding natural estrogen analogues with more excellent therapeutic safety (He et al., 2017).

The similar structure of PSO with coumestrol, a phytoestrogen, allows PSO to bind efficiently to estrogen receptors. In vivo studies performed in ovariectomized rats showed that PSO could inhibit bone resorption of osteoclasts and stimulate the bone formation (Zhai et al., 2017).

In another study performed by Kong et al. (2017), the impact of PSO on osteoclastogenesis and bone loss was investigated in mice. A low (5 mg/kg) and high dose (50 mg/kg) PSO was administered orally before LPS injection (5 mg/kg) and then every other seven days. The femurs of mice were collected on day 10. The results showed that PSO treatment inhibited LPS-induced bone loss and osteoclastogenesis formation and markedly down-regulated inflammatory cytokines (TNF-α, IL-1β, and IL-6) expression.

In a similar study performed by Zhai et al. (2018), osteotropic activities of PSO from Cullen corylifolium and coumestrol from Medicago sativa L. were studied. PSO and coumestrol (at doses of 10 mg/kg body weight/day each) were administered to ovariectomized rats, and after 12 weeks of intervention, samples were collected. Both PSO and coumestrol suppressed ovariectomized bone loss in vivo. It was demonstrated that PSO suppresses bone loss and have better osteoprotective effects than coumestrol.

Clinical Studies

Regarding human clinical trials with PSO, there is a scarcity. There are only a few that have included a reduced number of patients with dermatological conditions such as acne and vitiligo.

A small clinical study included 76 adolescent and young patients diagnosed with grade II and III acne vulgaris, who received double medication for 1 month: oral tablets that had as active principle PSO from Cullen corylifolium and topical cream Clarine with antimicrobial, anti-inflammatory, healing, antioxidant, astringent, and emollient properties. The results showed that patients with grade II acne had an excellent response (56.3% of cases) and good response (43.8% of cases). Patients with grade III acne had a good response (only 38.3% of cases) and a good response in (56.7% of cases). Thus, the combination of the oral and topical preparations had a synergistic, beneficial anti-acne effect (Gopal et al., 2001; Khushboo et al., 2009).

In another study, the combination of PSO with trioxalene (a chemical derivative) and the exposure to solar ultraviolet is an effective psoriasis treatment. Another clinical study included 30 patients with lip vitiligo who topically applied an ointment with Cullen corylifolium as the main ingredient. Patients with mild forms of vitiligo showed maximum improvement in 1–10 months, while chronic cases with lip vitiligo showed an inadequate response. Exposure of the patient to sunlight (5–30 min daily) with oral administration of this for 1–7 weeks gave improved the beneficial effects (Sharma et al., 2005). Possible mechanism of action: PSO interacts with ultraviolet in the epidermis forming photosensitive fragments of DNA. The maximum activity is at a wavelength of 325 nm having as effects: 1) slowing down the proliferation of keratinocytes; 2) suppression of the immune skin reaction; and 3) affects melanocytes, fibroblasts, and endothelial cells (Chopra et al., 2013).

Although there are many treatments for vitiligo, none work unanimously for all patients. Patients with vitiligo have areas with completely pigment-free skin. PUVA phototherapy (PSO + UVA) has proven to be effective over time, but narrow-band UVB therapy is gaining ground as a safe and effective therapeutic option. A recent study aimed to statistically compare the effectiveness of PUVA and nb-UVB treatments (type B ultraviolet, narrow-band). A group of 69 vitiligo patients who were treated with PUVA or nb-UVB were retrospectively analyzed. The two groups of patients were compared regarding the stage of repigmentation, the number of treatments until complete repigmentation, the appearance of newly affected areas or the increase in the size of existing ones, adverse effects of therapy, stability of repigmentation, and matching of pigment color. It has been shown that in the group treated with PUVA (31 patients), nine showed complete repigmentation (23.6%) and 14 a moderate improvement (36.8%), while in the group of those treated with nb-UVB (31 patients), 13 showed complete repigmentation (41.9%), and a moderate improvement (32.2%). Increased stability of repigmentation and better matching of pigment color to the nb-UVB-treated group was observed. The conclusions of the study were that nb-UVB is a more effective therapeutic method and the repigmentation induced by nb-UVB is statistically more stable (Parsad et al., 2006).

Discussion

The strong point of this study was that many systematic reviews were included that highlighted the mechanisms of PSO pharmacological activities in many chronic diseases. Thus, the article provides an up-to-date overview of the potential therapeutic applications of this natural compound.

Although, this study presented the beneficial pharmacological activities of PSO for human health, but the main clinical therapeutic limitation derives from low absorption. Several methods have been tried to improve PSO absorption, of which prenylation has been shown to be the most effective, as prenylated metabolite it has to be more bioavailable, increasing estrogenic activity and thus improving the anti-osteoporotic effects (Kretzschmar et al., 2010; Mukai et al., 2013) and antioxidant (Li et al., 2014).

Psoralidin is a prenylated coumestans derivative, with multiple therapeutic effects proven in preclinical studies, of which the most important and with the greatest clinical applicability as adjuvant therapies are: antiosteoporotic and anticancer.

Osteoporosis is a condition that manifests itself by decreased bone mass and damage to bone tissue. Thus, even bone fractures can occur as a result of minor traumas, the most affected being the spine, hip, forearm, ribs and hand joint (Salehi et al., 2020b). PSO has osteoprotective effects, promotes osteogenic differentiation, mineralization of osteoblasts, inhibits bone resorption by reducing osteoclast differentiation.

Cancer is a malignant tumor (Zlatian et al., 2015). Tumors appear when cells start to proliferate uncontrollably and can be benign or malignant (Elsebai et al., 2016). Unlike benign tumors, malignant tumors can differentiate (and lose their normal function), having the ability to invade neighboring tissues and to metastasize, that means to form tumors secondary to other parts of the body than the site of origin (Buga et al., 2019).

Carcinogenesis is a multi-stage and multi-factorial process during which profound changes occur, allowing cells to proliferate uncontrollably and ultimately invading other tissues. This process can be described in three major stages: initiation, promotion, and tumor progression. The results of our study showed that PSO could reduce the proliferation of malignant cells and stimulate apoptosis, thus being a potential adjunct in anticancer treatment.

Viral infections play an important role in human disease, and the recent COVID-19 pandemic in the emergence of globalization and ease of travel have highlighted their prevention as a critical issue in protecting public health (Alam Riaz et al., 2020; Docea et al., 2020; Goumenou et al., 2020; Tsatsakis et al., 2020). Despite advances in immunization and drug development, many viruses lack preventive vaccines and effective antiviral therapies, which are often caused by the generation of viral escape mutants (Calina et al., 2020a; Calina et al., 2020b). Thus, the identification of new antiviral alternative treatments is of critical importance, and bioactive natural compounds are an excellent source for such discoveries (Sarkar et al., 2020). In a recent study, it was shown that ethanolic extract from P. corylifolia seeds has good activity against papain-like protease (PLpro), an essential enzyme involved in SARS-CoV replication. The bioassay-guided fractionation of this extract identified six polyphenolic compounds as the bioactive compounds responsible for action against SARS-CoV PLpro. Of these, the most potent compounds that inhibited SARS-CoV PLpro were PSO and isobavachalcone (with IC50 ranging from 4.2 to 38.4 μM). The anti-SARS-CoV-2 mechanism is a type I, whereby PSO binds selectively to the free enzyme and not to the enzyme substrate. Based on these preliminary results it can be considered PSO as being one of the natural compound as potential agents against coronaviruses (Kim et al., 2014; Mani et al., 2020).

Although promising preclinical pharmacological studies have highlighted the mechanisms underlying PSO use as adjunctive therapy, long-term toxicity studies and data on possible interactions with other drugs are required. Besides, additional clinical trials are needed to confirm the clinical utility in medical practice.

Conclusion

The numerous bioactivities of PSO are summarized in this review outlining research approaches focusing on pharmacological activities and clinical trials. PSO has been proved to be a therapeutic option for cancer treatment. Based on this extensive research, we also suggest that PSO has potential as an antidepressant, anti-inflammatory, anti-osteoclastogenic, anti-osteoporotic and also bring beneficial effects in estrogen-related diseases. Regarding the bioavailability, we describe PSO as a non-toxic but low oral bioavailability compound; thus, new and further researches regarding the bioavailability of PSO as well as studies on pharmacokinetic are still required to improve the efficacy and delivery of PSO. The various therapeutic effects of PSO support its use as a potential drug in a wide range of medical conditions. Further research and studies are needed to confirm its benefits and efficacy in pharmaceutical formulations.

Author Contributions

JS-R, MM, and DC: conceptualization. SK, BY, AB, BS, and MAA: validation investigation. DC, AD, NA, and MM: resources. MI, NA, MERR, and JS-R: data curation. MM, DC, BS, AM, and JS-R: review and editing. All authors: writing. All authors read and approved the final manuscript and contributed equally to the manuscript.

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.

Acknowledgments

This work was supported by CONICYT PIA/APOYO CCTE AFB170007.

References

Alam Riaz, T., Ali, E., Rahman, M., Mubarak, M., and Martorell, M. (2020). A perspective on emerging therapeutic interventions for COVID-19. Front. Public Health 8, 281. doi: 10.3389/fpubh.2020.00281

PubMed Abstract | CrossRef Full Text | Google Scholar

Badarau, D., Wang, A. S., Swamy, M. K., Shaw, S., Maggi, F., Da Silva, L. E., et al. (2019). Arctium species secondary metabolites chemodiversity and bioactivities. Front. Plant Sci. 10, 834. doi: 10.3389/fpls.2019.00834

PubMed Abstract | CrossRef Full Text | Google Scholar

Borate, A., Khambhapati, A., Udgire, M., Paul, D., and Mathur, S. (2014). Preliminary phytochemical studies and evaluation of antibacterial activity of Psoralea corylifolia seed extract. Am. J. Phytomed. Clin. Ther. 2, 095–101

Google Scholar

Bronikowska, J., Szliszka, E., Jaworska, D., Czuba, Z. P., and Krol, W. (2012). The coumarin psoralidin enhances anticancer effect of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). Molecules 17, 6449–6464. doi: 10.3390/molecules17066449

PubMed Abstract | CrossRef Full Text | Google Scholar

Buga, A. M., Docea, A. O., Albu, C., Malin, R. D., Branisteanu, D. E., Ianosi, G., et al. (2019). Molecular and cellular stratagem of brain metastases associated with melanoma. Oncol. Lett. 17, 4170–4175. doi: 10.3892/ol.2019.9933

PubMed Abstract | CrossRef Full Text | Google Scholar

Calina, D., Docea, A., Petrakis, D., Egorov, A., Ishmukhametov, A., Gabibov, A., et al. (2020a). Towards effective COVID-19 vaccines: updates, perspectives and challenges. Int. J. Mol. Med. 46, 3–16. doi: 10.3892/ijmm.2020.4596

PubMed Abstract | CrossRef Full Text | Google Scholar

Calina, D., Hartung, T., Docea, A. O., Spandidos, D. A., Egorov, A. M., Shtilman, M. I., et al. (2020b). COVID-19 vaccines: ethical framework concerning human challenge studies. Daru J. Pharm. Sci., 1–6. doi: 10.1007/s40199-020-00371-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Cao, H.-J., Li, C.-R., Wang, L.-Y., Ziadlou, R., Grad, S., Zhang, Y., et al. (2019). Effect and mechanism of psoralidin on promoting osteogenesis and inhibiting adipogenesis. Phytomedicine 61, 152860. doi: 10.1016/j.phymed.2019.152860

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, C.-H., Hwang, T.-L., Chen, L.-C., Chang, T.-H., Wei, C.-S., and Chen, J.-J. (2017). Isoflavones and anti-inflammatory constituents from the fruits of Psoralea corylifolia. Phytochemistry 143, 186–193. doi: 10.1016/j.phytochem.2017.08.004

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, Y., Cheung, Y.-T., Kong, L.-D., Ng, T. B., Qiao, C., Mo, S.-F., et al. (2008). Transcriptional regulation of corticotrophin releasing factor gene by furocoumarins isolated from seeds of Psoralea corylifolia. Life Sci. 82, 1117. doi: 10.1016/j.lfs.2008.03.014

PubMed Abstract | CrossRef Full Text | Google Scholar

Chiou, W.-F., Don, M.-J., Liao, J.-F., and Wei, B.-L. (2011). Psoralidin inhibits LPS-induced iNOS expression via repressing Syk-mediated activation of PI3K-IKK-IκB signaling pathways. Eur. J. Pharmacol. 650, 102–109. doi: 10.1016/j.ejphar.2010.10.004

PubMed Abstract | CrossRef Full Text | Google Scholar

Chopra, B., Dhingra, A. K., and Dhar, K. L. (2013). Psoralea corylifolia L. (Buguchi)—folklore to modern evidence: review. Fitoterapia 90, 44–56. doi: 10.1016/j.fitote.2013.06.016

PubMed Abstract | CrossRef Full Text | Google Scholar

Chow, S.-C. (2014). Bioavailability and bioequivalence in drug development. Wiley Interdiscip. Rev. Comput. Stat. 6, 304–312. doi: 10.1002/wics.1310

PubMed Abstract | CrossRef Full Text | Google Scholar

Das, T. P., Suman, S., and Damodaran, C. (2014). Induction of reactive oxygen species generation inhibits epithelial-mesenchymal transition and promotes growth arrest in prostate cancer cells. Mol. Carcinog. 53, 537–547. doi: 10.1002/mc.22014

PubMed Abstract | CrossRef Full Text | Google Scholar

Docea, A. O., Tsatsakis, A., Albulescu, D., Cristea, O., Zlatian, O., Vinceti, M., et al. (2020). A new threat from an old enemy: re-emergence of coronavirus. Int. J. Mol. Med. 45, 1631–1643. doi: 10.3892/ijmm.2020.4555

PubMed Abstract | CrossRef Full Text | Google Scholar

Dwarampudi, L. P., Dhanabal, S., Shanmugam, R., and Muruganantham, N. (2012). Antipsoriatic activity and cytotoxicity of ethanolic extract of Psoralia corylifolia seeds. Hygeia J. Drug Med. 4, 41–48.

Google Scholar

Elsebai, M. F., Mocan, A., and Atanasov, A. G. (2016). Cynaropicrin: a comprehensive research review and therapeutic potential as an anti-hepatitis C virus agent. Front. Pharmacol. 7, 472. doi: 10.3389/fphar.2016.00472

PubMed Abstract | CrossRef Full Text | Google Scholar

Farahani, M. S., Bahramsoltani, R., Farzaei, M. H., Abdollahi, M., and Rahimi, R. (2015). Plant-derived natural medicines for the management of depression: an overview of mechanisms of action. Rev. Neurosci. 26, 305–21. doi: 10.1515/revneuro-2014-0058

PubMed Abstract | CrossRef Full Text | Google Scholar

Gebremeskel, A., Wijerathne, T. D., Kim, J. H., Kim, M. J., Seo, C. S., Shin, H. K., et al. (2017). Psoralea corylifolia extract induces vasodilation in rat arteries through both endothelium-dependent and -independent mechanisms involving inhibition of TRPC3 channel activity and elaboration of prostaglandin. Pharmaceut. Biol. 55, 2136–2144. doi: 10.1080/13880209.2017.1383484

PubMed Abstract | CrossRef Full Text | Google Scholar

Gopal, M., Farahana, B., and Pramesh, R. (2001). Effectiveness of herbal medications in the treatment of acne vulgaris—a pilot study. Indian Pract. 54, 723.

Google Scholar

Goumenou, M., Sarigiannis, D., Tsatsakis, A., Anesti, O., Docea, A. O., Petrakis, D., et al. (2020). COVID-19 in Northern Italy: an integrative overview of factors possibly influencing the sharp increase of the outbreak. Mol. Med. Rep. 22, 20–32. doi: 10.3892/mmr.2020.11079

PubMed Abstract | CrossRef Full Text | Google Scholar

Hao, W., Zhang, X., Zhao, W., and Chen, X. (2014). Psoralidin induces autophagy through ROS generation which inhibits the proliferation of human lung cancer A549 cells. PeerJ 2, e555. doi: 10.7717/peerj.555

PubMed Abstract | CrossRef Full Text | Google Scholar

He, J.-B., Chen, M.-H., and Lin, D.-K. (2017). New insights into the tonifying kidney-yin herbs and formulas for the treatment of osteoporosis. Arch. Osteoporos. 12, 14. doi: 10.1007/s11657-016-0301-4

PubMed Abstract | CrossRef Full Text | Google Scholar

He, Y., Zhu, Q., Chen, M., Huang, Q., Wang, W., Li, Q., et al. (2016). The changing 50% inhibitory concentration (IC50) of cisplatin: a pilot study on the artifacts of the MTT assay and the precise measurement of density-dependent chemoresistance in ovarian cancer. Oncotarget 7, 70803–70821. doi: 10.18632/oncotarget.12223

PubMed Abstract | CrossRef Full Text | Google Scholar

Jin, Z., Yan, W., Jin, H., Ge, C., and Xu, Y. (2016a). Differential effect of psoralidin in enhancing apoptosis of colon cancer cells via nuclear factor-κB and B-cell lymphoma-2/B-cell lymphoma-2-associated X protein signaling pathways. Oncol. Lett. 11, 267–272. doi: 10.3892/ol.2015.3861

PubMed Abstract | CrossRef Full Text | Google Scholar

Jin, Z., Yan, W., Jin, H., Ge, C., and Xu, Y. (2016b). Psoralidin inhibits proliferation and enhances apoptosis of human esophageal carcinoma cells via NF-κB and PI3K/Akt signaling pathways. Oncol. Lett. 12, 971–976. doi: 10.3892/ol.2016.4716

PubMed Abstract | CrossRef Full Text | Google Scholar

Khushboo, P., Jadhav, V., and Kadam, V. (2009). Development and validation of a HPTLC method for determination of psoralen in Psoralea corylifolia (Bavachi). Int. J. PharmTech Res. 1, 1122–1128.

Google Scholar

Khushboo, P., Jadhav, V., Kadam, V., and Sathe, N. (2010). Psoralea corylifolia Linn.—"Kushtanashini". Pharmacogn. Rev. 4, 69. doi: 10.4103/0973-7847.65331

PubMed Abstract | CrossRef Full Text | Google Scholar

Kim, D. W., Seo, K. H., Curtis-Long, M. J., Oh, K. Y., Oh, J.-W., Cho, J. K., et al. (2014). Phenolic phytochemical displaying SARS-CoV papain-like protease inhibition from the seeds of Psoralea corylifolia. J. Enzyme Inhib. Med. Chem. 29, 59–63. doi: 10.3109/14756366.2012.753591

PubMed Abstract | CrossRef Full Text | Google Scholar

Kim, Y., Lim, H.-S., Lee, J., and Jeong, S.-J. (2016). Quantitative analysis of Psoralea corylifolia Linne and its neuroprotective and anti-neuroinflammatory effects in HT22 hippocampal cells and BV-2 microglia. Molecules 21, 1076. doi: 10.3390/molecules21081076

PubMed Abstract | CrossRef Full Text | Google Scholar

Kong, L., Ma, R., Yang, X., Zhu, Z., Guo, H., He, B., et al. (2017). Psoralidin suppresses osteoclastogenesis in BMMs and attenuates LPS-mediated osteolysis by inhibiting inflammatory cytokines. Int. Immunopharm. 51, 31–39. doi: 10.1016/j.intimp.2017.07.003

PubMed Abstract | CrossRef Full Text | Google Scholar

Kretzschmar, G., Zierau, O., Wober, J., Tischer, S., Metz, P., and Vollmer, G. (2010). Prenylation has a compound specific effect on the estrogenicity of naringenin and genistein. J. Steroid Biochem. Mol. Biol. 118, 1–6. doi: 10.1016/j.jsbmb.2009.08.005

PubMed Abstract | CrossRef Full Text | Google Scholar

Kumar, R., Srinivasan, S., Koduru, S., Pahari, P., Rohr, J., Kyprianou, N., et al. (2009). Psoralidin, an herbal molecule, inhibits phosphatidylinositol 3-kinase-mediated akt signaling in androgen-independent prostate cancer cells. Cancer Prev. Res. 2, 234–243. doi: 10.1158/1940-6207.capr-08-0129

PubMed Abstract | CrossRef Full Text | Google Scholar

Kumar, R., Srinivasan, S., Pahari, P., Rohr, J., and Damodaran, C. (2010). Activating stress-activated protein kinase-mediated cell death and inhibiting epidermal growth factor receptor signaling: a promising therapeutic strategy for prostate cancer. Mol. Cancer Ther. 9, 2488–2496. doi: 10.1158/1535-7163.mct-10-0180

PubMed Abstract | CrossRef Full Text | Google Scholar

Larsson, S., and Fazzalari, N. L. (2014). Anti-osteoporosis therapy and fracture healing. Arch. Orthop. Trauma Surg. 134, 291–297. doi: 10.1007/s00402-012-1558-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Lee, S.-J., Nam, K.-W., and Mar, W. (2009). Induction of quinone reductase activity by psoralidin isolated from Psoralea corylifolia in mouse hepa 1c1c7 cells. Arch. Pharm. Res. (Seoul) 32, 1061–1065. doi: 10.1007/s12272-009-1712-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Lee, S., Yun, B., Kim, M., Park, C., Lee, W., Oh, H.-M., et al. (2012). Phenolic compounds isolated from Psoralea corylifolia inhibit IL-6-induced STAT3 activation. Planta Med. 78, 903–906. doi: 10.1055/s-0031-1298482

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, M., Dong, H., Chen, L., and Zhang, Y. (2014). Study on quercetin isopentenyl modification and free radical scavenging activity. Sci. Technol. Food Ind. 35, 84–87.

Google Scholar

Lim, S.-H., Ha, T.-Y., Ahn, J., and Kim, S. (2011). Estrogenic activities of Psoralea corylifolia L. seed extracts and main constituents. Phytomedicine 18, 425–430. doi: 10.1016/j.phymed.2011.02.002

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, X., Nam, J.-W., Song, Y. S., Viswanath, A. N. I., Pae, A. N., Kil, Y.-S., et al. (2014). Psoralidin, a coumestan analogue, as a novel potent estrogen receptor signaling molecule isolated from Psoralea corylifolia. Bioorg. Med. Chem. Lett. 24, 1403–1406. doi: 10.1016/j.bmcl.2014.01.029

PubMed Abstract | CrossRef Full Text | Google Scholar

Luo, W., Liu, Y., Zhang, J., Luo, X., Lin, C., and Guo, J. (2013). Andrographolide inhibits the activation of NF-κB and MMP-9 activity in H3255 lung cancer cells. Exp. Ther. Med. 6, 743–746. doi: 10.3892/etm.2013.1196

PubMed Abstract | CrossRef Full Text | Google Scholar

Mani, J. S., Johnson, J. B., Steel, J. C., Broszczak, D. A., Neilsen, P. M., Walsh, K. B., et al. (2020). Natural product-derived phytochemicals as potential agents against coronaviruses: a review. Virus Res., 284, 197989. doi: 10.1016/j.virusres.2020.197989

PubMed Abstract | CrossRef Full Text | Google Scholar

Mar, W., Je, K.-H., and Seo, E.-K. (2001). Cytotoxic constituents of Psoralea corylifolia. Arch. Pharm. Res. (Seoul) 24, 211. doi: 10.1007/bf02978259

PubMed Abstract | CrossRef Full Text | Google Scholar

Mishra, A. P., Salehi, B., Sharifi-Rad, M., Pezzani, R., Kobarfard, F., Sharifi-Rad, J., et al. (2018). Programmed cell death, from a cancer perspective: an overview. Mol. Diagn. Ther. 22, 281–295. doi: 10.1007/s40291-018-0329-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Mukai, R., Fujikura, Y., Murota, K., Uehara, M., Minekawa, S., Matsui, N., et al. (2013). Prenylation enhances quercetin uptake and reduces efflux in Caco-2 cells and enhances tissue accumulation in mice fed long-term. J. Nutr. 143, 1558–1564. doi: 10.3945/jn.113.176818

PubMed Abstract | CrossRef Full Text | Google Scholar

Pal, D., Kolluru, V., Chandrasekaran, B., Baby, B. V., Aman, M., Suman, S., et al. (2017a). Targeting aberrant expression of Notch-1 in ALDH+ cancer stem cells in breast cancer. Mol. Carcinog. 56, 1127–1136. doi: 10.1002/mc.22579

PubMed Abstract | CrossRef Full Text | Google Scholar

Pal, D., Suman, S., Kolluru, V., Sears, S., Das, T. P., Alatassi, H., et al. (2017b). Inhibition of autophagy prevents cadmium-induced prostate carcinogenesis. Br. J. Cancer 117, 56–64. doi: 10.1038/bjc.2017.143

PubMed Abstract | CrossRef Full Text | Google Scholar

Pandareesh, M. D., Mythri, R. B., and Srinivas Bharath, M. M. (2015). Bioavailability of dietary polyphenols: factors contributing to their clinical application in CNS diseases. Neurochem. Int. 89, 198–208. doi: 10.1016/j.neuint.2015.07.003

PubMed Abstract | CrossRef Full Text | Google Scholar

Parsad, D., Kanwar, A., and Kumar, B. (2006). Psoralen-ultraviolet A vs. narrow-band ultraviolet B phototherapy for the treatment of vitiligo. J. Eur. Acad. Dermatol. Venerol. 20, 175–177. doi: 10.1111/j.1468-3083.2006.01413.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Rao, Z., Wang, S., and Wang, J. (2018). Protective effects of psoralidin on IL-1β-induced chondrocyte apoptosis. Mol. Med. Rep. 17, 3418–3424. doi: 10.3892/mmr.2017.8248

PubMed Abstract | CrossRef Full Text | Google Scholar

Rana, A., Attar, R., Qureshi, M. Z., Gasparri, M. L., Donato, V. D., Ali, G. M., et al. (2014). Dealing naturally with stumbling blocks on highways and byways of TRAIL induced signaling. Asian Pac. J. Cancer Prev. 15, 8041–8046. doi: 10.7314/apjcp.2014.15.19.8041

PubMed Abstract | CrossRef Full Text | Google Scholar

Ren, G., Luo, W., Sun, W., Niu, Y., Ma, D.-L., Leung, C.-H., et al. (2016). Psoralidin induced reactive oxygen species (ROS)-dependent DNA damage and protective autophagy mediated by NOX4 in breast cancer cells. Phytomedicine 23, 939–947. doi: 10.1016/j.phymed.2016.05.008

PubMed Abstract | CrossRef Full Text | Google Scholar

Ren, L., Li, L.-Z., Huang, J., Huang, L.-Z., Li, J.-H., Li, Y.-M., et al. (2019). New compounds from the seeds of Psoralea corylifolia with their protein tyrosine phosphatase 1B inhibitory activity. J. Asian Nat. Prod. Res., 22 (8), 732–737. doi: 10.1080/10286020.2019.1621852

PubMed Abstract | CrossRef Full Text | Google Scholar

Rusu, M. E., Mocan, A., Ferreira, I. C., and Popa, D.-S. (2019). Health benefits of nut consumption in middle-aged and elderly population. Antioxidants 8, 302. doi: 10.3390/antiox8080302

PubMed Abstract | CrossRef Full Text | Google Scholar

Salehi, B., Calina, D., Docea, A., Koirala, N., Aryal, S., Lombardo, D., et al. (2020a). Curcumin’s nanomedicine formulations for therapeutic application in neurological diseases. J. Clin. Med. 9, 430. doi: 10.3390/jcm9020430

PubMed Abstract | CrossRef Full Text | Google Scholar

Salehi, B., Lopez-Jornet, P., Pons-Fuster López, E., Calina, D., Sharifi-Rad, M., Ramírez-Alarcón, K., et al. (2019a). Plant-derived bioactives in oral mucosal lesions: a key emphasis to curcumin, lycopene, chamomile, aloe vera, green tea and coffee properties. Biomolecules 9, 106. doi: 10.3390/biom9030106

PubMed Abstract | CrossRef Full Text | Google Scholar

Salehi, B., Rescigno, A., Dettori, T., Calina, D., Docea, A. O., Singh, L., et al. (2020b). Avocado-soybean unsaponifiables: a panoply of potentialities to Be exploited. Biomolecules 10, 130. doi: 10.3390/biom10010130

PubMed Abstract | CrossRef Full Text | Google Scholar

Salehi, B., Sestito, S., Rapposelli, S., Peron, G., Calina, D., Sharifi-Rad, M., et al. (2019b). Epibatidine: a promising natural alkaloid in health. Biomolecules 9, 6. doi: 10.3390/biom9030106

PubMed Abstract | CrossRef Full Text | Google Scholar

Sani, T. A., Mohammadpour, E., Mohammadi, A., Memariani, T., Yazdi, M. V., Rezaee, R., et al. (2017). Cytotoxic and apoptogenic properties of Dracocephalum kotschyi aerial part different fractions on calu-6 and mehr-80 lung cancer cell lines. Farmacia 65, 189–199.

Google Scholar

Sarkar, C., Mondal, M., Torequl Islam, M., Martorell, M., Oana Docea, A., Maroyi, A., et al. (2020). Potential therapeutic options for COVID-19: current status, challenges and future perspectives. Front. Pharmacol. 11, 1428. doi: 10.3389/fphar.2020.572870

PubMed Abstract | CrossRef Full Text | Google Scholar

Seo, E., Lee, E.-K., Lee, C. S., Chun, K.-H., Lee, M.-Y., and Jun, H.-S. (2014). Psoralea corylifolia L. seed extract ameliorates streptozotocin-induced diabetes in mice by inhibition of oxidative stress. Oxid. Med. Cell. Longev. 2014, 897296. doi: 10.1155/2014/897296

PubMed Abstract | CrossRef Full Text | Google Scholar

Sharifi-Rad, J., Rodrigues, C. F., Sharopov, F., Docea, A. O., Can Karaca, A., Sharifi-Rad, M., et al. (2020). Diet, lifestyle and cardiovascular diseases: linking pathophysiology to cardioprotective effects of natural bioactive compounds. Int. J. Environ. Res. Public Health 17, 2326. doi: 10.3390/ijerph17072326

PubMed Abstract | CrossRef Full Text | Google Scholar

Sharma, P., Yelne, M., and Dennis, T. (2005). Database on medicinal plants used in Ayurveda. India: Central Council for Research in Ayurveda and Siddha

Google Scholar

Shi, M., Zhang, Y., Song, M., Sun, Y., Li, C., and Kang, W. (2018). Screening the marker components in Psoralea corylifolia L. with the aids of spectrum-effect relationship and component knock-out by UPLC-MS2. Int. J. Mol. Sci. 19, 3439. doi: 10.3390/ijms19113439

PubMed Abstract | CrossRef Full Text | Google Scholar

Song, K., Ling, F., Huang, A., Dong, W., Liu, G., Jiang, C., et al. (2015). In vitro and in vivo assessment of the effect of antiprotozoal compounds isolated from Psoralea corylifolia against Ichthyophthirius multifiliis in fish. Int. J. Parasitol. Drugs Drug Resist. 5, 58–64. doi: 10.1016/j.ijpddr.2015.04.001

PubMed Abstract | CrossRef Full Text | Google Scholar

Srinivasan, S., Kumar, R., Koduru, S., Chandramouli, A., and Damodaran, C. (2010). Inhibiting TNF-mediated signaling: a novel therapeutic paradigm for androgen independent prostate cancer. Apoptosis 15, 153–161. doi: 10.1007/s10495-009-0416-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Sun, D.-X., Ge, G.-B., Dong, P.-P., Cao, Y.-F., Fu, Z.-W., Ran, R.-X., et al. (2016). Inhibition behavior of fructus psoraleae’s ingredients towards human carboxylesterase 1 (hCES1). Xenobiotica 46, 503–510. doi: 10.3109/00498254.2015.1091521

PubMed Abstract | CrossRef Full Text | Google Scholar

Sun, H., Ma, Z., Lu, D., and Wu, B. (2015). Regio- and isoform-specific glucuronidation of psoralidin: evaluation of 3-O-glucuronidation as a functional marker for UGT1A9. J. Pharmaceut. Sci. 104, 2369–2377. doi: 10.1002/jps.24464

PubMed Abstract | CrossRef Full Text | Google Scholar

Szliszka, E., Czuba, Z. P., Sędek, Ł., Paradysz, A., and Król, W. (2011). Enhanced TRAIL-mediated apoptosis in prostate cancer cells by the bioactive compounds neobavaisoflavone and psoralidin isolated from Psoralea corylifolia. Pharmacol. Rep. 63, 139–148. doi: 10.1016/s1734-1140(11)70408-x

PubMed Abstract | CrossRef Full Text | Google Scholar

Tsatsakis, A., Docea, A. O., Calina, D., Tsarouhas, K., Zamfira, L.-M., Mitrut, R., et al. (2019). A mechanistic and pathophysiological approach for stroke associated with drugs of abuse. J. Clin. Med. 8, 1295. doi: 10.3390/jcm8091295

PubMed Abstract | CrossRef Full Text | Google Scholar

Tsatsakis, A., Petrakis, D., Nikolouzakis, T. K., Docea, A. O., Calina, D., Vinceti, M., et al. (2020). COVID-19, an opportunity to reevaluate the correlation between long-term effects of anthropogenic pollutants on viral epidemic/pandemic events and prevalence. Food Chem. Toxicol., 141, 111418. doi: 10.1016/j.fct.2020.111418

PubMed Abstract | CrossRef Full Text | Google Scholar

Xiao, G., Li, G., Chen, L., Zhang, Z., Yin, J.-J., Wu, T., et al. (2010). Isolation of antioxidants from Psoralea corylifolia fruits using high-speed counter-current chromatography guided by thin layer chromatography-antioxidant autographic assay. J. Chromatogr. A 1217, 5470–5476. doi: 10.1016/j.chroma.2010.06.041

PubMed Abstract | CrossRef Full Text | Google Scholar

Xin, Z., Wu, X., Yu, Z., Shang, J., Xu, B., Jiang, S., et al. (2019). Mechanisms explaining the efficacy of psoralidin in cancer and osteoporosis, a review. Pharmacol. Res., 147, 104334. doi: 10.1016/j.phrs.2019.104334

PubMed Abstract | CrossRef Full Text | Google Scholar

Xu, Q., Pan, Y., Yi, L.-T., Li, Y.-C., Mo, S.-F., Jiang, F.-X., et al. (2008). Antidepressant-like effects of psoralen isolated from the seeds of Psoralea corylifolia in the mouse forced swimming test. Biol. Pharm. Bull. 31, 1109–1114. doi: 10.1248/bpb.31.1109

PubMed Abstract | CrossRef Full Text | Google Scholar

Yan, C., Wu, Y., Weng, Z., Gao, Q., Yang, G., Chen, Z., et al. (2015). Development of an HPLC method for absolute quantification and QAMS of flavonoids components in Psoralea corylifolia L. J. Anal. Methods Chem. 2015. doi: 10.1155/2015/792637

PubMed Abstract | CrossRef Full Text | Google Scholar

Yang, H. J., Youn, H., Seong, K. M., Yun, Y. J., Kim, W., Kim, Y. H., et al. (2011). Psoralidin, a dual inhibitor of COX-2 and 5-LOX, regulates ionizing radiation (IR)-induced pulmonary inflammation. Biochem. Pharmacol. 82, 524–534. doi: 10.1016/j.bcp.2011.05.027

PubMed Abstract | CrossRef Full Text | Google Scholar

Yang, J., Wen, L., Jiang, Y., and Yang, B. (2019). Natural Estrogen Receptor Modulators and Their Heterologous Biosynthesis. Trends Endocrinol. Metab. 30, 66–76. doi: 10.1016/j.tem.2018.11.002

PubMed Abstract | CrossRef Full Text | Google Scholar

Yi, L.-T., Li, Y.-C., Pan, Y., Li, J.-M., Xu, Q., Mo, S.-F., et al. (2008). Antidepressant-like effects of psoralidin isolated from the seeds of Psoralea Corylifolia in the forced swimming test in mice. Prog. Neuropsychopharmacol. Biol. Psychiatry 32, 510–519. doi: 10.1016/j.pnpbp.2007.10.005

PubMed Abstract | CrossRef Full Text | Google Scholar

Yin, J., Xiang, C., and Song, X. (2016). Nanoencapsulation of psoralidin via chitosan and Eudragit S100 for enhancement of oral bioavailability. Int. J. Pharm. 510, 203–209. doi: 10.1016/j.ijpharm.2016.05.007

PubMed Abstract | CrossRef Full Text | Google Scholar

Yu, B., Wang, A.-H., Zhou, K., Chai, L.-J., and Liu, L. (2019). Molecular pathway of psoralidin-induced apoptosis in HepG2 cell line. Chin. J. Integr. Med. 25, 757–762. doi: 10.1007/s11655-016-2251-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhai, Y., Li, Y., Wang, Y., Cui, J., Feng, K., Kong, X., et al. (2017). Psoralidin, a prenylated coumestan, as a novel anti-osteoporosis candidate to enhance bone formation of osteoblasts and decrease bone resorption of osteoclasts. Eur. J. Pharmacol. 801, 62–71. doi: 10.1016/j.ejphar.2017.03.001

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhai, Y., Wang, Q., Li, Y., Cui, J., Feng, K., Kong, X., et al. (2018). The higher osteoprotective activity of psoralidin in vivo than coumestrol is attributed by its presence of an isopentenyl group and through activated PI3K/Akt axis. Biomed. Pharmacother. 102, 1015–1024. doi: 10.1016/j.biopha.2018.03.166

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, R., Shi, W., Li, L., Huang, X., Xu, D., and Wu, L. (2019). Biological activity and health promoting effects of psoralidin. Pharmazie 74, 67–72. doi: 10.1691/ph.2019.8619

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, X., Zhao, W., Wang, Y., Lu, J., and Chen, X. (2016). The chemical constituents and bioactivities of Psoralea corylifolia Linn.: a review. Am. J. Chin. Med. 44, 35–60. doi: 10.1142/s0192415x16500038

PubMed Abstract | CrossRef Full Text | Google Scholar

Zlatian, O. M., Comănescu, M. V., Roşu, A. F., Roşu, L., Cruce, M., Găman, A. E., et al. (2015). Histochemical and immunohistochemical evidence of tumor heterogeneity in colorectal cancer. Rom. J. Morphol. Embryol. 56, 175–181.

PubMed Abstract | Google Scholar

Keywords: Psoralea corylifolia, herbal medicine, psoralidin, bioavailability, biological properties, molecular mechanisms, cell signalling pathways

Citation: Sharifi-Rad J, Kamiloglu S, Yeskaliyeva B, Beyatli A, Alfred MA, Salehi B, Calina D, Docea AO, Imran M, Anil Kumar NV, Romero-Román ME, Maroyi A and Martorell M (2020) Pharmacological Activities of Psoralidin: A Comprehensive Review of the Molecular Mechanisms of Action. Front. Pharmacol. 11:571459. doi: 10.3389/fphar.2020.571459

Received: 10 June 2020; Accepted: 28 September 2020;
Published: 22 October 2020.

Edited by:

Andrei Mocan, Iuliu Haţieganu University of Medicine and Pharmacy, Romania

Reviewed by:

Claudio Ferrante, University “G. d’Annunzio” of Chieti-Pescara, Italy
Monica Paoliello, Albert Einstein College of Medicine, United States
Luigi Menghini, University “G. d’Annunzio”of Chieti-Pescara, Italy

Copyright © 2020 Sharifi-Rad, Kamiloglu, Yeskaliyeva, Beyatli, Alfred, Salehi, Calina, Docea, Imran, Anil Kumar, Eugenia Romero-Román, Maroyi and Martorell. 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: Javad Sharifi-Rad, javad.sharifirad@gmail.com, Bahare Salehi, bahar.saheli007@gmail.com, Daniela Calina calinadaniela@gmail.com, Alfred Maroyi, amaroyi@ufh.ac.za, Miquel Martorell, martorellpons@gmail.com

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.