Int J Pharm Pharm Sci, Vol 15, Issue 12, 1-12Review Article

NANOENCAPSULATION OF LUTEOLIN: ENHANCING BIOAVAILABILITY AND MEDICINAL BENEFITS

RAKSHA B., VAISHNAVI M., DURGA M.*, BRINDHA BANU B., DEEPIKAA R.

Department of Biochemistry and Bioinformatics, Dr. MGR Janaki College of Arts and Science for Women, Adyar, Chennai-600028, Tamil Nadu, India
*Corresponding author: Durga M.; *Email: durgam2k7@gmail.com

Received: 20 Sep 2023, Revised and Accepted: 18 Oct 2023


ABSTRAC

Luteolin is a naturally occurring chemical widely found in plants ranging from Bryophyta to Magnoliophyta. It can be obtained from several dietary sources such as carrots, olive oil, celery, spinach, oregano, and, fossils of some organisms such as Celtis and Ulmus dating back 36 to 25 million years. It is synthesized by the Shikimate pathway. The major qualities and therapeutic benefits of luteolin include cytoprotective abilities, Antioxidant, Anti-inflammatory, Anticancer, Antidepressant, Antidiabetic, Antiallergic, Reactive Oxygen Species Scavenging and High radical scavenging. The antioxidant and Reactive Oxygen Species scavenging activity of luteolin aids in treating and curing inflammatory skin processes. It has been proven to act as a therapeutic drug with a wide spectrum of scope in the prevention and treatment of a vast range of malignant and benign cancers, extending from bladder cancer to breast cancer and from oral cancer to glioblastoma, which is achieved by its anticancer, antioxidant properties and cytoprotective abilities. Apart from its anticancer properties, it has a great scope in the restoration from neuropsychiatric disease and high-level fatigue due to Long COVID syndrome-associated brain fog and Chemo fog. The poor solubility and low bioavailability of luteolin limit its use in food and medicine. Synthetic and Natural polymer-based delivery systems have been developed to improve its stability and bioavailability. This review will highlight recent research on its nanoencapsulation and provide more information on luteolin to help readers have a better grasp of the compound's medicinal benefits.

Keywords: Luteolin, Antioxidant, Anticancer, Anti-inflammatory, Flavonoid, Nanoencapsulation, COVID, Antidepressant, Neuroprotector, Anti-diabetic


INTRODUCTION

For this comprehensive review, we conducted a thorough search of specialized databases covering the years 1977-2023, including renowned sources such as Elsevier and Pubmed. We utilized a range of targeted keywords, such as "Luteolin," "Luteolin anti-cancer activity," "Luteolin antioxidant," "Luteolin nanoencapsulation," and "Luteolin updated review," to ensure a comprehensive and thorough analysis. In addition, we also included articles from top-tier journals like Springer, reputable internet sources, and online publications from well-known journals such as International Journal of Agriculture and Life Sciences, Planta Medica, International Journal of Current Pharmaceutical Research, The Asian Journal of Pharmaceutical and Clinical Research, The International Journal of Pharma and Biosciences, and International Journal of Applied Pharmaceutics.

The great majority of chemical substances utilized daily are found in plants. As we are aware of the medicinal advantages that plants offer, the bioactive substances in them are being widely investigated in the past few decades to treat and prevent a variety of human ailments [1]. Flavonoids are polyphenols that have a significant part in protecting plant cells from ultraviolet (UV) irradiation, insects, and microorganisms [2]. Cell culture, animal, and human population research gives an idea that flavonoids are advantageous to both animal and human health [3]. Luteolin, also known as 3′,4′,5,7-tetrahydroxyflavone, is a naturally occurring chemical that belongs to flavonoids, which are extensively distributed in the plant kingdom [2]. Luteolin belongs to the flavone group of flavonoids and has a C6-C3-C6 structure that contains two benzene rings, a third ring that contains oxygen, and a double bond between two and three carbon atoms fig. 1. It also has hydroxyl groups at carbons 3', 4', 5 and 7 [4]. The vast majority of luteolin's bioactivity is caused by the presence of a hydroxyl moiety at carbon positions of 3', 4', 5, and 7. Being mostly derived from fruits, vegetables, and other edible plant parts, luteolin is a flavonoid chemical that is widely distributed.

The biological importance of Luteolin compounds has been studied in recent decades, and these researches have revealed their anti-cancer, antioxidant, anti-inflammatory, and neuroprotective properties [5, 6]. The luteolin's pharmacological effects may be connected on a functional level. For example, luteolin's ability to reduce inflammation may be related to its ability to fight cancer. Luteolin's anticancer function is connected with initiating apoptosis, which involves DNA damage, redox regulation, and protein kinases in limiting cancer cell proliferation, reducing angiogenesis and metastasis. Furthermore, luteolin makes cancer cells more susceptible to medically induced cell toxicity by decreasing cell survival pathways and energizing the apoptotic pathways. Notably, luteolin may pass through the blood-brain barrier, making it useful for treating illnesses of the central nervous system, such as brain tumors [7].

Fig. 1: Diagrammatic representation of luteolin compound

Sources of luteolin

Luteolin is a flavone that can be found in a variety of vegetables and medicinal plants [8]. Plants are the primary source of luteolin and its derivatives [9]. However, concentrations are often low when compared to other flavonols, such as quercetin or kaempferol [8]. It makes up a minimal proportion of our daily diet (less than 1 mg/d) when compared to other secondary plant substances [10]. Luteolin and its glycosides are found in many different plant families, including the Bryophyta, Pteridophyta, Pinophyta, and Magnoliophyta, and are widely distributed across the plant world [9]. Some spices, such as thyme, sage, and parsley as well as wild carrots and, artichokes, contain significant amounts [8]. Carrots, peppers, celery, spinach, lettuce, olive oil, peppermint, thyme, rosemary, and oregano are all good sources of luteolin [8, 9]. Even while luteolin only makes up a small amount of the flavonoids found in food, it can be obtained in large quantities from peanut hulls and the plant Reseda luteola L., which is being used as a dyeing agent because of its high luteolin concentration for thousands of years [11]. Even the fossils of the Celtis and Ulmus species, which date back 36 and 25 million years, respectively, attest to its existence [12].

Fig. 2: Sources of luteolin [8, 9, 11]

Biosynthesis of luteolin

Plants contain a diverse set of metabolic substances that let them perform basic processes and respond to different stimuli [13]. Phenyl propanoids are compounds derived from the phenylalanine amino acid that are involved in plant development, cellular metabolism, and biotic and abiotic stimuli [13]. The phenylpropanoid pathway follows the shikimate pathway, which has been investigated for decades [14]. Via the phenylpropanoid and flavonoid pathways, which branch off from the principal secondary metabolite pathway, the Luteolin molecule is synthesized.

Fig. 3: Diagrammatic representation of biosynthesis of luteolin. PAL-phenylalanine ammonia lyase; C4H-trans-cinnamate 4-hydroxylase; 4CL-coumarate 4-ligase; CHS-chalcone synthase; CHI-chalcone isomerase; F3’H-flavonoid 3’-hydroxylase; FNS-flavone synthase [13, 15-20]

Luteolin biosynthesis starts with the conversion of amino acid phenylalanine to trans-cinnamic acid by the enzyme phenylalanine ammonia-lyase, followed by the formation of trans-coumaric acid from trans-cinnamic acid with the help of the enzyme trans-cinnamate 4-hydroxylase (C4H) [fig. 3]. Then, p-coumaroyl CoA is formed from trans-coumaric acid by the action of enzyme coumarate 4-ligase (4CL) [13, 15]. It is followed by the conversion of p-coumaroyl CoA into naringenin chalcone (NC) by chalcone synthase (CHS) [16]. The next step is the key to the biosynthesis of Luteolin and it happens through the action of the enzyme chalcone isomerase (CHI), which converts naringenin chalcone into naringenin [17]. The introduction of a hydroxyl group at the 3’ position in the beta ring of naringenin occurs through the action of the enzyme flavonoid 3’-hydroxylase (F3’H), leading to the formation of eriodictyol [18]. Eventually, Luteolin is produced from the substrates naringenin and eriodictyol by the enzyme flavone synthase (FNS) [19, 20].

Qualities of luteolin

Plants produce luteolin molecule in two different forms: as an aglycone without sugar moiety and as a glycoside with sugar moiety attached. It has a molecular weight of 286.236 g/mol and with the molecular formula of C15H10O6 [21]. It is predominantly found in plants as glycosides that are cleaved following nutritional absorption. The aglycones are subsequently conjugated and processed [22]. Luteolin, like other flavonoids, is a pleiotropic compound, which means that its pharmacological properties may not be explained by a single biochemical activity [22].

Luteolin as an antioxidant

The most important effect of luteolin includes its effective anti-oxidative activity, which includes high radical scavenging and cytoprotective abilities [23, 24]. It acts as a reactive oxygen species (ROS) scavenger by oxidizing itself [25]. As a result, the anti-inflammatory properties of luteolin may be linked in part with its anti-oxidative properties. This is especially essential, considering oxidative stress plays a significant role in many inflammatory skin processes [26, 27]. Other anti-oxidants include vitamins and cellular redox mechanisms as well as luteolin interact with one another. Luteolin can enhance its anti-oxidative strength in this way [28]. Because of its glycosidic group, it has anti-scavenging activity, which aids in the eradication of reactive nitrogen and oxygen species [29-33].

In Wister rats, luteolin (50 mg/kg orally) pre-treatment protects from renal failure via a detoxifying mechanism mediated by antioxidation activity, as well as anti-inflammatory and anti-apoptotic mechanisms [34]. It aids in minimizing the impact of intestinal mucositis-related mucosal damage brought on by cancer treatment [35]. Furthermore, Luteolin antioxidant activity has been shown to cause apoptosis through increasing antioxidant activity [36]. By enhancing the activity of several antioxidant enzymes, the rat model's hepatotoxicity caused by carbon tetrachloride (CCl4) was minimized [37]. Due to its antioxidant properties, it also functions as a chemoprotective molecule while treating patients with doxorubicin, a medicine that damages the hepatorenal system and increases the effectiveness of treatment by removing the drug's adverse effects [38]. Thus, flavonoids, which function as primary antioxidants or free radical scavengers, aid in numerous health ailments [39].

Luteolin as an anti-cancer agent

Each year, over 18 million new cases of cancer are recorded worldwide. Cancer has a greater impact on vulnerable groups and strains health and the economy [40]. Epidemiological research shows that flavonoids provide a variety of health advantages. Dietary flavonoids' anticancer abilities have been demonstrated by several studies [41]. According to research by Sabzichi et al., luteolin packed in phytosomes increases the passive targeting of breast cancer cells in MDA-MB 231 cells. On the other hand, the treatment of cells with doxorubicin and luteolin-containing nanoparticles resulted in the highest percentage of cells dying. To a larger extent than luteolin alone, nanoparticles loaded with luteolin reduced the expression of downstream Nrf2 gene genes at the messenger Ribonucleic acid (mRNA) level in cells. Likewise, these nanoparticles loaded with luteolin strongly decreased the expression of Nrf2 downstream genes, including heme oxygenase 1 (Ho1) inhibition and multi-drug resistance gene (MDR1), and significantly increased cancer cell mortality [42].

Luteolin’s efficient inhibition of cancer cell progression when examined in vivo at doses of 3 to 50 µM and in vitro at doses of 5 to 10 mg/kg demonstrated its effectiveness [43]. In another study, luteolin was administered to MCF-7 cells at a dose of 60 mol/l for 48 h, which inhibited the growth of cancer in a dose and time-dependent manner by lowering the expression of Bcl-2 protein, lowering the migration rate by 71.07% and lowering the expression of AEG-1 and MMP-2 by 82.34% and 85.70% respectively [44].

It has the benefit of treating skin cancer due to its capacity to penetrate the skin. Its action against stomach cancer was demonstrated in studies using human carcinoma cells at an IC50 value of 7.1 g/ml. Its effective action against lung cancer was seen at an IC50 value of 11.7 g/ml and it was effective against bladder cancer at IC50 value of 19.5 g/ml [45].

Leukaemia, a type of blood cancer that generates abnormal white blood cells and frequently results in fatalities, is another serious illness that affects people. This Luteolin substance inhibited the growth of the human leukemic cell lines CEM-C7 and CEM-C1 [46, 47].

Epidemiological research suggests that human lung, prostate, stomach, and breast cancer risk is inversely correlated with dietary intake of flavonoids [48-50]. In human breast cancer treatment, Luteolin and paclitaxel, when combined with MDA-MB-231 cells, reduced tumor size and weight, activated caspases-8 and-3, and improved Fas ligand expression. In an orthotropic tumor model, the rise in Fas expression was also ascribed to the inhibition of STAT3 [51].

Fig. 4: Luteolin-mediated extrinsic and intrinsic apoptosis in breast cancer [52]

Fig. 5: Potential therapeutic targets for luteolin in psoriasis and dermatitis [53]

Table 1: Luteolin affections in different types of cancer [54]

Cancer type Cell proliferation Cell survival signaling Apoptosis Angiogenesis Metastasis Dose of luteolin Reference
Breast cancer Inhibit MAPKs, PI3K-Akt, CDK2 Inhibit PI3K-Akt, EGFR, NF-κB, MAPKs Activate DR5, caspases-8 and-9, Fas, Bax Inhibit VEGF, MMP-9, PI3K/Akt Inhibit PI3K/Akt 10 mg/kg 60μmol/l for 48 h, suppressed the proliferation of cancer [54]
Colon cancer - - - Inhibit MMP-9 - 1.2 mg/kg b. w [54]
Pancreatic cancer - Inhibit EGFR, NF-κB Activate Bax Inhibit NF-κB Inhibit NF-κB - [54]
Prostate cancer - - Inhibit FASN Inhibit VEGF, MMP-9 Inhibit IL-6 - [54]
Glioblastoma

Inhibit

P13K-Akt

Inhibit

P13K-Akt, PKC

Activate P53,

Inhibit XIAP

Inhibit NF-κB, PI3K/Akt Inhibit NF-κB, PI3K/Akt - [54]
Oral cancer - - Activate Fas, P53 Inhibit IL-6 - [54]
Lung cancer Inhibit MAPKs

Inhibit

NF-κB, MAPKs

Activate caspases-3 and-9, Bax, JNK

Inhibit Bcl-XL

Inhibit VEGF, MMP-9, NF-κB, HIF-1α Inhibit IL-6, FAK, NF-κB 50μM [54]
Kidney cancer - - Activate DR5, Capsases, Bax, p53, JNK - - - [54]
Cervical and placental cancer

Inhibit

P13K-Akt

Inhibit

P13K-Akt

Activate DR5

Inhibit Bcl-XL

Inhibit

P13K-Akt

- [54]
Ovarian cancer - - - - Inhibit FAK - [54]
Skin cancer - - - Inhibit MMP-9 - [54]
Liver cancer

Inhibit

PI3K-Akt

Inhibit

PI3k-Akt, NF-κB

Activate Bax, P53

Inhibit Bcl-XL

Inhibit NF-κB Inhibit NF-κB - [54]
Gastric cancer - - Activate Bax, P53 Inhibit VEGF, MMP-9 - 40 mg/kg [54]
Oesophageal and bladder cancer - - Activate p53, JNK - - - [54]

Fig. 6: Luteolin can alter macrophage polarization from M1 to M2 phenotype [55]

Anti-diabetic activity of luteolin

Diabetes is a serious health issue that exists worldwide. Every developed nation as well as a developing one is affected by its prevalence. According to the International Diabetic Federation (IDF) estimate of 2017, about 451 million people are affected by it, and by 2045, that number is expected to rise to 693 million. Additionally, it has negative socioeconomic effects. Type 2 diabetes among the growing younger population alarmed society. Diabetes is one of the most common illnesses that influence the health of the global population and can result in a number of life-threatening conditions [56]. As a result of oxidative stress, diabetes damages heart muscles and results in myocardial ischemia/reperfusion (I/R). The redirection of the oxidation reaction caused by activating the sestrin 2-Nrf2-based feedback loop during Luteolin therapy lowers oxidative stress and cardiac damage [57].

Long-term diabetes damages the neurons in the cerebral cortex; the treatment of luteolin greatly reduces diabetic symptoms such as peroxidation of lipids, which rises in diabetic rat brains. In addition, it lowers GS4, superoxide dismutase, and catalase activity which sharply declines in the hippocampus and cerebral cortex of rats after luteolin administration. It is believed that luteolin's antioxidant effect enhances CA1 neurons by minimizing neuronal apoptosis since ChE activity is a result of diabetes and leads to progressive cognitive decline and neurological dysfunction. Luteolin inhibits the ChE activity, which improves the situation in diabetic rats [58].

Anti-inflammatory and anti-allergic properties of luteolin

One of the body's defense mechanisms, inflammation, aids in the healing of wounds and protects against infection. However, persistent inflammation can lead to dangerous conditions like cancer, chronic obstructive pulmonary disease, and arthritis [59-61]. The inflammation action is necessary to lessen the influence of the stimuli, which would otherwise disrupt the normal cells, but it must be minimized because chronic inflammation interferes with proper functioning. Anti-inflammatory molecules are introduced to treat it in order to safeguard cells from negative effects [62]. During inflammation, macrophages are triggered by a variety of chemicals, including cytokines from the host and pathogen toxins. Lipopolysaccharide (LPS), a part of Gram-negative bacteria's outer membrane, is frequently used as an endotoxin and inflammatory trigger. Tumor necrosis factor (TNF), free radicals-ROS and reactive nitrogen species (RNS), and interleukins (ILs) are vigorously produced by the activated macrophages, which attract inflammatory cells like neutrophils and lymphocytes to the site of infection and clear the pathogens [61, 63, 64]. Luteolin, which is a flavonoid, is said to possess an anti-allergic effect [65]. Persistent synthesis of these chemicals during the time of chronic inflammation can lead to illnesses such as cancer. Luteolin exhibits an anti-inflammatory effect as it blocks the synthesis of such cytokines and their signal transduction pathways [66-68]. Luteolin reduces oxLDL-activated inflammation in vitro by blocking STAT3, a signal transducer as well as an activator of transcription. Its interaction with STAT3 was primarily demonstrated in one study by hydrogen bonding [69].

Luteolin as a neuroprotector

Important disorders with a high global occurrence are anxiety and depression [70]. The most ubiquitous neurodegenerative diseases are Parkinson's disease (PD) and Alzheimer's disease (AD). Although oxidative stress is thought to play a significant part in the development of both illnesses, other variables such as the buildup of misfolded proteins, also play a role [71]. Some of their symptoms can be alleviated by antidepressant medications, but they are accompanied by many negative effects. Luteolin was given to male 129 Sv/Ev mice along with palmitoylethanolamide in a trial to determine its possible antidepressant impact. The results demonstrated a strong antidepressant effect at low doses, suggesting that this combination could be considered a new approach to treating depressive symptoms [70].

Luteolin as protection against alzheimer’s disease

The most prevalent cause of memory loss in the world's population is Alzheimer's disease. The disease's primary symptom is the buildup of amyloid peptides within the brain's extracellular matrix [72]. There is currently no cure known for the illness. The search for an Alzheimer's disease cure is still ongoing worldwide. According to this theory, the secondary metabolite Luteolin may be able to decrease the impact of the disease. Due to the (direct) interaction between the gene expression of an antioxidant enzyme involved in free radical scavenging and ROS, Luteolin efficiently lowers the signs and symptoms of Alzheimer's disease as well as the formation of A42 aggregation in transgenic drosophila. This is shown by the concentration-mediated reduction of AchE activity, which delays the emergence of symptoms like those of Alzheimer's disease [73]. Luteolin prevents ER (Endoplasmic Reticulum) stress, which impairs learning and memory in mice, from causing neuro-inflammatory aggravation. As a result, it enhances 3XTg's brain histomorphology and minimizes protein plaques in mice with Alzheimer's illness [74].

Fig. 7: Luteolin’s action in ER stress. Unfolded proteins are created as a result of ER stress in the cells. The UPR leads to neuroinflammatory aggravation, which leads to memory and learning impairment in mice. Luteolin suppresses neuroinflammation [74]

Additionally, it boosts the expression of Bcl2 and significantly decreases the expression of Bax and caspase-3. High concentrations of Luteolin may be hazardous, blocking A25-35 and causing cell death. Additionally, it causes apoptosis by selectively acting on ER to protect Bcl2 cells from A-25-35 and stimulates the ER/ERK/MAPK transmission pathway [75]. Insulin resistance in the brain may be reduced by luteolin. The current research discovered that the Luteolin therapy enhanced hepatic insulin sensitivity and tightly controlled cell function, which boosted glucose metabolism and potentiated insulin signaling in the hippocampus [76].

Luteolin in Parkinson's disease treatment

Coherently, drugs that can trigger autophagy, the process by which intracellular trash is degraded, may aid in the removal of harmful chemicals from neurons, having a neuroprotective impact. According to this theory, injection of luteolin into male C57/BL6 mice with palmitoylethanolamide as an endogenous autophagic promoter improved tissue structure stimulated autophagy, and improved neurobehavioral functioning [71]. The luteolin generated during defense in the in vitro effect on oxidation is connected to the erratic amplification of endogenous free radical repression of the mitochondrial viability of membrane potential of mitochondria and a decrease in glutathione content. The catalyzing activity suggests that the multilayer modulatory route contributes to the neuroprotective effects of luteolin. The possible maintaining of the antioxidation or pro-oxidation ratio leads to protection.

Additionally, the neuroprotective pathway aids in reviving the ROS scavenging activity, a depleted endogenous enzymatic and non-enzymatic antioxidative defense system [77]. Luteolin improves mouse behavior in the traction and pole trait test, suggesting its potential in applied Parkinson’s disease therapy by boosting the Bcl2/Bax ratio by lowering caspase-3 and also preventing the loss of TH+ve neurons in the substantia nigra (SN) and neural fibers in the striatum [78].

Luteolin in obesity treatment

Obesity acts as a major public health risk and contributes significantly to the burden of non-communicable diseases in the world, such as type 2 diabetes, hypertension, cardiovascular disease, and some malignancies. It is believed to cause premature mortality [79]. It is characterized by an abnormal buildup of body fat and associated with a significant risk of metabolic comorbidities, such as non-alcoholic fatty liver disease, type 2 diabetes, and cardiovascular disease. Adipose tissue is an essential immunological and endocrine organ as well as a key regulator of energy storage and metabolism in lean individuals. A persistent energy imbalance causes Adipose tissue remodeling, adipocyte hypotrophy and hyperplasia, chronic low-grade inflammation, and adipocyte malfunction in Adipose tissue. These changes eventually result in ectopic lipid accumulation and systemic insulin resistance [80].

Luteolin has been shown to help manage obesity when taken as a dietary supplement. By altering the Toll-like receptor signaling pathway, luteolin supplementation reduced macrophage infiltration and adipokine/cytokine dysregulation in rat models [81]. It has been shown to help combat obesity and related metabolic illnesses by increasing Adipose tissue thermogenesis and systemic energy expenditure. It has also been shown to reduce Adipose tissue lipogenesis, inflammation, and ectopic lipid deposition [80].

In a study, luteolin was found to be involved in the regulation of efflux genes of cholesterol, such as liver ATP-binding cassette transporter G1 (ABCG1), X receptor (LXR-), and scavenger receptor class B member 1 (SRB1). It demonstrated that luteolin lowers cholesterol by controlling the different genes associated with the cholesterol export process [82]. By lowering proinflammatory mediators in macrophages like tumor necrosis factor (TNF), monocyte chemoattractant protein (MCP-1), and NO while co-cultivating with 3T3-L1 adipocytes and RAW264 macrophages, luteolin reduces the obesity-related adipocyte inflammation that is observed after administration. The ability of luteolin to lessen inflammation in adipose tissue serves as proof of this [83].

Luteolin in cardiac health

Any condition, abnormality, or poor function linked to the heart, blood vessels, or circulation is referred to as cardiovascular disease (CVDs) [84]. The most effective approach for preventing the start of this illness is to improve dietary and lifestyle uses and make them affordable and accessible to the general public. Diet is a significant external factor in the development of CVDs [85, 86]. The luteolin molecule mitigates the likelihood of myocardial infarction as integrating it into food may help lower the risk of CVD. In a study using rats with myocardial ischemia/reperfusion (I/R) (MIRM) damage, treatment with luteolin decreased the damage to the heart valves by downregulating the Src homology 2 domain-containing protein tyrosine phosphatase 1 (SHP-1) regulation and upregulating the STAT3 pathway, which reduced the inflammatory response [87].

The anti-apoptosis property proves essential in avoiding harm to cardiac tissue. In one study, giving luteolin prevented apoptosis by enhancing AKT signaling in the simulated ischemia/reperfusion (sI/R) paradigm [88]. Luteolin serves to avoid cardiac abnormalities such as Ca+2 transport and contractile dysfunction, which worsen in failing cardiomyocytes and are stopped by controlling the SERCA2a gene. As a result, it improves cardiac health [89]. The SERCA proteins are important for maintaining heart health. By triggering the p38 MAPK pathway in the cardiomyocytes and simulated ischemia/reperfusion rat models, luteolin aids in upregulating its expression [90].

Fig. 8: Schematic diagram for the cardioprotection of luteolin against I/R injury in the diabetic heart [91]

Fig. 9: By up-regulating AKT, up-regulating BCL-2, and down-regulating BAX, lut reduces I/R injury by suppressing apoptosis. The main protein involved in Ca2+ absorption from the cytosol into the SR is called SERCA2a. Following PI3K/AKT signaling pathway activation, SERCA2a activity is increased. At the same time, luteolin functions as a p38 mapk pathway inhibitor to prevent the phosphorylation of PLN, increasing SERCA2a activity and decreasing Ca2+ overload. Through HO-1, lut prevents oxidative damage and improves nrf2's ability to bind to the ARE. Lut inhibits JNK and raises p-ERK1/2 to promote cardiomyocyte contraction. to shield the heart from I/R injury, lut activates the myocardial eNOS pathway and suppresses the mitochondrial permeability transition pore [92]

Luteolin for Long-COVID syndrome-associated brain fog and chemo fog

SARS-CoV-2 infection causes COVID-19, whose severity is a result of the host's inflammatory response and the release of a cascade of pro-inflammatory cytokines [93]. As a result of COVID-19, auto-immune and inflammation-related diseases are in particular, becoming increasingly prevalent [94]. Additionally, multiple "mystery" illnesses have been attributed to cytokine storms [95]. One such illness called "brain fog", affects survivors of COVID-19 and is linked to extremely high levels of fatigue and neuropsychiatric symptoms [89]. The terms "chronic COVID syndrome," "post-COVID syndrome," and "long haulers COVID syndrome" are also used to describe this condition [96]. Patients with long-COVID syndrome report symptoms that are strikingly similar [97] to those of those with myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) [98, 99], mast cell activation syndrome (MCAS) [100, 101], or systemic mastocytosis (SM) [102], conditions in which stress, pathogens, and environmental stimuli activate the body's special tissue immune cells called mast cells.

Fig. 10: Symptoms of long-COVID syndrome [93]

Patients receiving chemotherapy are more prone to getting COVID-19 infection [103]. Additionally, cognitive dysfunction, often considered a sign of long-COVID syndrome, is experienced by more than 50% of patients undergoing or finishing chemotherapy [93]. Cognitive dysfunction, also known as "chemofog" [104, 105] or "chemobrain," [106-110] has been linked to specific neuroimaging findings [89]. Several medications, including doxorubicin [111-113], methotrexate [114, 115], lenalidomide [116], rituximab [116], and trastuzumab [117], have been associated with "chemobrain". The hypothalamic-pituitary-adrenal (HPA) axis, normally stimulated by stress and has the ability to further impair the emotional stability of those affected by COVID-19 [118, 119], is also susceptible to being impacted by COVID-19 [120].

It seems that long-COVID syndrome does not have any clinically viable treatments [121, 122]. Furthermore, since T cells and antibody production seem to be protective but proinflammatory cytokines appear to be harmful, it is difficult to determine whether it would be preferable to stimulate or repress the immune system [123, 124]. Inhibition of mast cell-related neuroinflammation would be a reasonable strategy, particularly for brain fog linked with long-COVID syndrome, MCAS, ME/CFS, and chemotherapy-induced "chemobrain" [93]. The COVID-19 or long-COVID syndrome might be helped by mast cell inhibition; however, there are currently no known potent inhibitors of mast cells [125, 126]. Alternatively, the commonly accessible and generally well-regarded safe [127-131] natural flavonoids quercetin and luteolin [132-136], which have structurally similar properties, could suppress mast cells [132-136]. Both flavonoids have wide anti-viral characteristics, inhibiting virus entry into host cells, inhibiting neuroinflammation [137], and reducing cognitive loss [138]. In addition, luteolin has been shown to have improved brain penetration, inhibit microglia and mast cells, and lessen neuroinflammation as well as cognitive impairment, including Alzheimer's disease, in both people and animal models [93].

Nanoencapsulation of luteolin

Flavonoids have a variety of biological effects and may be utilized to treat or prevent disease. Because they showcase a remarkable range of biochemical and pharmacological actions, including anti-inflammatory, anti-oxidant, cytostatic, apoptotic, and estrogenic activities, flavonoids have attracted considerable interest for research and application in functional foods, nutraceutical products, and pharmaceuticals [139, 140]. Among the flavonoids, luteolin (3',4',5,7-tetrahydroxyflavone) is capable of improving insulin sensitivity and is present in a variety of plants, including celery, green peppers, perilla leaves, chamomile tea, broccoli, and carrots. Additionally, because luteolin can pass through the brain-blood barrier, it can be used to treat ailments of the central nervous system [141-144]. Due to luteolin's low oral bioavailability and the need for high bioavailability for it to demonstrate pharmacological activity in vivo, the development of innovative formulations may be useful in maximizing luteolin's pharmacological activity [145]. The stability, bioactivity, and bioavailability of these substances must, therefore, be preserved by product formulators in order to ensure that they are delivered to consumers in their active molecular form. The main objective of nanoparticle systems is to have these characteristics [146, 147]. By enhancing bioavailability, solubility, and retention duration, biodegradable nanoparticles are widely employed to enhance the beneficial value of diverse water-soluble/insoluble medical medicines and bioactive compounds. These drug-nanoparticle compositions improve the therapeutic index, specificity, tolerability, and efficacy of the related medications. They also lower costs for the patient toxicity risks and have a number of benefits, such as preventing premature degradation and interaction with biological systems and enhancing intracellular penetration [148].

The use of luteolin in food and medicine is severely constrained by its poor solubility and low bioavailability. Some delivery systems based on synthetic polymers, such as hyaluronic acid/poly (N-isopropyl acrylamide) polymer network hydrogels and monomethoxy poly (ethylene glycol)-poly (-caprolactone) (MPEG-PCL) micelles, have been advanced to increase the stability and bioavailability of the compound luteolin. However, there are few reports of the nano-delivery system made from plant-based polymers for encapsulating luteolin [149].

Due to their tiny size, high surface-to-volume ratio, and potent dispersibility, many nanoscale delivery methods, including emulsifiers and liposomes have been extensively exploited in recent years to increase the bioavailability and stability of bioactive chemicals. When designing delivery systems for nanoparticles with great encapsulating capacity, high penetration of biological barrier, and well-controlled release property, starch is frequently used because of its accessibility, availability at a lesser cost, renewability, and biodegradable quality. According to reports, molecular modification may be able to give starch the physiochemical characteristics it needs for use in encapsulating systems [149].

The usage of oxidized lotus root starch nanoparticles, which are utilized to encapsulate luteolin, has amylopectin (70%–80%) and amylose (20%–30%). Currently, traditional meals are frequently prepared using lotus root starch as a food additive. However, due to its limited solubility in water at ambient temperature (25 °C), the potential applications of this starch have undergone little research. According to earlier research, oxidation may increase starch's solubility and have an impact on its capacity to encapsulate substances. Sodium hypochlorite (NaClO)-mediated 2,2,6,6-tetramethyl-1-piperidinyloxy (TEMPO) can oxidize lotus root starch. Because of its low cost, easy operating conditions, and low possibility of sample contamination, the resulting oxidized lotus root starch (OLRS) was subsequently utilized to create luteolin-OLRS nanoparticles [149]. This nanoencapsulation of luteolin may enhance its various properties, including its free radical scavenging property [150].

CONCLUSION

This article covers almost every aspect of luteolin's biological, physical, and chemical properties, including its anti-oxidant, anti-cancer, anti-diabetic, anti-inflammatory, neuroprotective, cardioprotective, anti-depressant, and aid in long-covid syndrome effects. The nano-encapsulated luteolin in synthetic and bio-polymers, with increased bioavailability and activity, was also reviewed. However, there has to be further investigation done on how to integrate luteolin alongside additional therapeutic compounds and treatments.

ABBREVIATIONS

Reactive oxygen species (ROS), Ischemia/reperfusion (I/R), cardiovascular disease (CVDs)

FUNDING

Nil

AUTHORS CONTRIBUTIONS

All the authors have contributed equally.

CONFLICT OF INTERESTS

Declared none

REFERENCES

  1. Imran M, Rauf A, Abu-Izneid T, Nadeem M, Shariati MA, Khan IA. Corrigendum to “Luteolin, a flavonoid, as an anticancer agent: a review”. Biomed Pharmacother Biomed Pharmacother. 2019;116:109084. doi: 10.1016/j.biopha.2019.109084, PMID 31178263.

  2. Harborne JB, Williams CA. Advances in flavonoid research since 1992. Phytochemistry. 2000 Nov;55(6):481-504. doi: 10.1016/s0031-9422(00)00235-1, PMID 11130659.

  3. Birt DF, Hendrich S, Wang W. Dietary agents in cancer prevention: flavonoids and isoflavonoids. Pharmacol Ther. 2001 May-Jun;90(2-3):157-77. doi: 10.1016/s0163-7258(01)00137-1, PMID 11578656.

  4. Ross JA, Kasum CM. Dietary flavonoids: bioavailability, metabolic effects, and safety. Annu Rev Nutr. 2002;22:19-34. doi: 10.1146/annurev.nutr.22.111401.144957, PMID 12055336.

  5. Ou HC, Pandey S, Hung MY, Huang SH, Hsu PT, Day CH. Luteolin: a natural flavonoid enhances the survival of HUVECs against oxidative stress by modulating AMPK/PKC pathway. Am J Chin Med. 2019;47(3):541-57. doi: 10.1142/S0192415X19500289, PMID 30966772.

  6. Cook MT. Mechanism of metastasis suppression by luteolin in breast cancer. Breast Cancer Dove Med. 2018 Jun 12;10:89-100:S144202. doi: 10.2147/BCTT, PMID 29928143, PMCID PMC6003288.

  7. Wruck CJ, Claussen M, Fuhrmann G, Romer L, Schulz A, Pufe T. Luteolin protects rat PC12 and C6 cells against MPP+ induced toxicity via an ERK dependent Keap1-Nrf2-ARE pathway. J Neural Transm Suppl. 2007;72(72):57-67. doi: 10.1007/978-3-211-73574-9_9, PMID 17982879.

  8. Manzoor MF, Ahmad N, Manzoor A, Kalsoom A. Food based phytochemical luteolin their derivatives, sources and medicinal benefits. Int J Agric Life Sci. 2017;3:11:s12200084. doi: 10.22573/spg.ijals.017.

  9. Muruganathan N, Dhanapal AR, Baskar V, Muthuramalingam P, Selvaraj D, Aara H. Recent updates on source, biosynthesis, and therapeutic potential of natural flavonoid luteolin: a review. Metabolites. 2022 Nov 20;12(11):1145. doi: 10.3390/metabo12111145, PMID 36422285, PMCID PMC9696498.

  10. Seelinger G, Merfort I, Schempp CM. Anti-oxidant, anti-inflammatory and anti-allergic activities of luteolin. Planta Med. 2008 Nov;74(14):1667-77. doi: 10.1055/s-0028-1088314, PMID 18937165.

  11. Ge L, Xia F, Song Y, Yang K, Qin Z, Li L. Solubility of luteolin in several imidazole-based ionic liquids and extraction from peanut shells using selected ionic liquid as solvent. Sep Purif Technol. 2014 Oct 15;135:223-8. doi: 10.1016/j.seppur.2014.08.022.

  12. Flavonoid and other chemical constituents of fossil Miocene celtis and Ulmus (succor creek flora). Science. 1977 Aug 19;197(4305):765-7. doi: 10.1126/science.197.4305.765, PMID 17790771.

  13. Vogt T. Phenylpropanoid biosynthesis. Mol Plant. 2010 Jan;3(1):2-20. doi: 10.1093/mp/ssp106, PMID 20035037.

  14. Herrmann KM, Weaver LM. The shikimate pathway. Annu Rev Plant Physiol Plant Mol Biol. 1999 Jun;50:473-503. doi: 10.1146/annurev.arplant.50.1.473, PMID 15012217.

  15. Ferrer JL, Austin MB, Stewart C Jr, Noel JP. Structure and function of enzymes involved in the biosynthesis of phenylpropanoids. Plant Physiol Biochem. 2008 Mar;46(3):356-70. doi: 10.1016/j.plaphy.2007.12.009, PMID 18272377, PMCID PMC2860624.

  16. Ferrer JL, Jez JM, Bowman ME, Dixon RA, Noel JP. Structure of chalcone synthase and the molecular basis of plant polyketide biosynthesis. Nat Struct Biol. 1999 Aug;6(8):775-84. doi: 10.1038/11553, PMID 10426957.

  17. Jez JM, Bowman ME, Dixon RA, Noel JP. Structure and mechanism of the evolutionarily unique plant enzyme chalcone isomerase. Nat Struct Biol. 2000 Sep;7(9):786-91. doi: 10.1038/79025, PMID 10966651.

  18. Croft KD. The chemistry and biological effects of flavonoids and phenolic acids. Ann N Y Acad Sci. 1998 Nov 20;854:435-42. doi: 10.1111/j.1749-6632.1998.tb09922.x, PMID 9928450.

  19. Martens S, Mithofer A. Corrigendum to “Flavones and flavone synthases”. Phytochemistry Phytochemistry. 2006;67(5). doi: 10.1016/j.phytochem.2006.01.004.

  20. Nabavi SM, Samec D, Tomczyk M, Milella L, Russo D, Habtemariam S. Flavonoid biosynthetic pathways in plants: versatile targets for metabolic engineering. Biotechnol Adv. 2020 Jan-Feb;38:107316. doi: 10.1016/j.biotechadv.2018.11.005, PMID 30458225.

  21. Yang K, Song Y, Ge L, Su J, Wen Y, Long Y. Measurement and correlation of the solubilities of luteolin and rutin in five imidazole-based ionic liquids. Fluid Ph. 2013 Apr 25;344:27-31. doi: 10.1016/j.fluid.2013.01.026.

  22. Aziz N, Kim MY, Cho JY. Anti-inflammatory effects of luteolin: a review of in vitro, in vivo, and in silico studies. J Ethnopharmacol. 2018 Oct 28;225:342-58. doi: 10.1016/j.jep.2018.05.019, PMID 29801717.

  23. Havsteen BH. The biochemistry and medical significance of the flavonoids. Pharmacol Ther. 2002 Nov-Dec;96(2-3):67-202. doi: 10.1016/s0163-7258(02)00298-x, PMID 12453566.

  24. Heim KE, Tagliaferro AR, Bobilya DJ. Flavonoid antioxidants: chemistry, metabolism and structure-activity relationships. J Nutr Biochem. 2002 Oct;13(10):572-84. doi: 10.1016/s0955-2863(02)00208-5, PMID 12550068.

  25. Lien EJ, Ren S, Bui HH, Wang R. Quantitative structure-activity relationship analysis of phenolic antioxidants. Free Radic Biol Med. 1999 Feb;26(3-4):285-94. doi: 10.1016/s0891-5849(98)00190-7, PMID 9895218.

  26. Sander CS, Chang H, Hamm F, Elsner P, Thiele JJ. Role of oxidative stress and the antioxidant network in cutaneous carcinogenesis. Int J Dermatol. 2004 May;43(5):326-35. doi: 10.1111/j.1365-4632.2004.02222.x, PMID 15117361.

  27. Seelinger G, Merfort I, Wolfle U, Schempp CM. Anti-carcinogenic effects of the flavonoid luteolin. Molecules. 2008 Oct 22;13(10):2628-51. doi: 10.3390/molecules13102628, PMID 18946424, PMCID PMC6245397.

  28. Wolfle U, Haarhaus B, Schempp CM. The photoprotective and antioxidative properties of luteolin are synergistically augmented by tocopherol and ubiquinone. Planta Med. 2013 Jul;79(11):963-5. doi: 10.1055/s-0032-1328716. PMID 23839819.

  29. Choi CW, Jung HA, Kang SS, Choi JS. Antioxidant constituents and a new triterpenoid glycoside from flos lonicerae. Arch Pharm Res. 2007 Jan;30(1):1-7. doi: 10.1007/BF02977770, PMID 17328234.

  30. Wu MJ, Huang CL, Lian TW, Kou MC, Wang L. Antioxidant activity of glossogyne tenuifolia. J Agric Food Chem. 2005 Aug 10;53(16):6305-12. doi: 10.1021/jf050511a, PMID 16076111.

  31. Cai Q, Rahn RO, Zhang R. Dietary flavonoids, quercetin, luteolin and genistein, reduce oxidative DNA damage and lipid peroxidation and quench free radicals. Cancer Lett. 1997 Oct 28;119(1):99-107. doi: 10.1016/s0304-3835(97)00261-9, PMID 18372528.

  32. Horvathova K, Chalupa I, Sebova L, Tothova D, Vachalkova A. Protective effect of quercetin and luteolin in human melanoma HMB-2 cells. Mutat Res. 2005 Jan 3;565(2):105-12. doi: 10.1016/j.mrgentox.2004.08.013, PMID 15661608.

  33. Cheng IF, Breen K. On the ability of four flavonoids, baicilein, luteolin, naringenin, and quercetin, to suppress the Fenton reaction of the iron-ATP complex. Biometals. 2000 Mar;13(1):77-83. doi: 10.1023/a:1009229429250, PMID 10831228.

  34. Albarakati AJA, Baty RS, Aljoudi AM, Habotta OA, Elmahallawy EK, Kassab RB. Luteolin protects against lead acetate-induced nephrotoxicity through antioxidant, anti-inflammatory, anti-apoptotic, and Nrf2/HO-1 signaling pathways. Mol Biol Rep. 2020 Apr;47(4):2591-603. doi: 10.1007/s11033-020-05346-1, PMID 32144527.

  35. Boeing T, de Souza P, Speca S, Somensi LB, Mariano LNB, Cury BJ. Luteolin prevents irinotecan-induced intestinal mucositis in mice through antioxidant and anti-inflammatory properties. Br J Pharmacol. 2020 May;177(10):2393-408. doi: 10.1111/bph.14987, PMID 31976547, PMCID PMC7174882.

  36. Kang KA, Piao MJ, Hyun YJ, Zhen AX, Cho SJ, Ahn MJ. Luteolin promotes apoptotic cell death via upregulation of Nrf2 expression by DNA demethylase and the interaction of Nrf2 with p53 in human colon cancer cells. Exp Mol Med. 2019 Apr 15;51(4):1-14. doi: 10.1038/s12276-019-0238-y, PMID 30988303, PMCID PMC6465248.

  37. Yan Y, Jun C, Lu Y, Jiangmei S. Combination of metformin and luteolin synergistically protects carbon tetrachloride-induced hepatotoxicity: mechanism involves antioxidant, anti-inflammatory, antiapoptotic, and Nrf2/HO-1 signaling pathway. BioFactors. 2019 Jul;45(4):598-606. doi: 10.1002/biof.1521, PMID 31336028.

  38. Owumi SE, Lewu DO, Arunsi UO, Oyelere AK. Luteolin attenuates doxorubicin-induced derangements of liver and kidney by reducing oxidative and inflammatory stress to suppress apoptosis. Hum Exp Toxicol. 2021 Oct;40(10):1656-72. doi: 10.1177/09603271211006171, PMID 33827303.

  39. Johney J, Johney J, Ragunathan R. Evaluation of antioxidant, antimicrobial, anticancer, and wound healing properties of leaf extracts of acanthus ilicifolius L. Int J Curr Pharm Sci. 2023 Jan 15;15(1):22-9. doi: 10.22159/ijcpr.2023v15i1.2066.

  40. Tiwary S, Hussain MS. Functional foods for prevention and treatment of cancer. Asian J Pharm Clin Res. 2021;14(3):4-10. doi: 10.22159/ajpcr.2021.v14i3.40426.

  41. Lotha RO, Sivasubramanian AR. Flavonoids nutraceuticals in prevention and treatment of cancer: a review. Asian J Pharm Clin Res. 2018;11(1):42-7. doi: 10.22159/ajpcr.2017.v11i1.23410.

  42. Sun DW, Zhang HD, Mao L, Mao CF, Chen W, Cui M. Luteolin inhibits breast cancer development and progression in vitro and in vivo by suppressing Notch signaling and regulating MiRNAs. Cell Physiol Biochem. 2015;37(5):1693-711. doi: 10.1159/000438535, PMID 26545287.

  43. Kawaii S, Tomono Y, Katase E, Ogawa K, Yano M. Antiproliferative activity of flavonoids on several cancer cell lines. Biosci Biotechnol Biochem. 1999 May;63(5):896-9. doi: 10.1271/bbb.63.896, PMID 10380632.

  44. Sato Y, Sasaki N, Saito M, Endo N, Kugawa F, Ueno A. Luteolin attenuates doxorubicin-induced cytotoxicity to MCF-7 human breast cancer cells. Biol Pharm Bull. 2015;38(5):703-9. doi: 10.1248/bpb.b14-00780, PMID 25947916.

  45. Cherng JM, Shieh DE, Chiang W, Chang MY, Chiang LC. Chemopreventive effects of minor dietary constituents in common foods on human cancer cells. Biosci Biotechnol Biochem. 2007 Jun;71(6):1500-4. doi: 10.1271/bbb.70008, PMID 17587681.

  46. Post JF, Varma RS. Growth inhibitory effects of bioflavonoids and related compounds on human leukemic CEM-C1 and CEM-C7 cells. Cancer Lett. 1992 Dec 24;67(2-3):207-13. doi: 10.1016/0304-3835(92)90145-l, PMID 1483269.

  47. Seelinger G, Merfort I, Wölfle U, Schempp CM. Anti-carcinogenic effects of the flavonoid luteolin. Molecules. 2008 Oct 22;13(10):2628-51. doi: 10.3390/molecules13102628, PMID 18946424, PMCID PMC6245397.

  48. Neuhouser ML. Dietary flavonoids and cancer risk: evidence from human population studies. Nutr Cancer. 2004;50(1):1-7. doi: 10.1207/s15327914nc5001_1, PMID 15572291.

  49. Knekt P, Jarvinen R, Seppanen R, Hellovaara M, Teppo L, Pukkala E. Dietary flavonoids and the risk of lung cancer and other malignant neoplasms. Am J Epidemiol. 1997 Aug 1;146(3):223-30. doi: 10.1093/oxfordjournals.aje.a009257, PMID 9247006.

  50. Wright ME, Mayne ST, Stolzenberg-Solomon RZ, Li Z, Pietinen P, Taylor PR. Development of a comprehensive dietary antioxidant index and application to lung cancer risk in a cohort of male smokers. Am J Epidemiol. 2004 Jul 1;160(1):68-76. doi: 10.1093/aje/kwh173, PMID 15229119.

  51. Cook MT, Liang Y, Besch Williford C, Goyette S, Mafuvadze B, Hyder SM. Luteolin inhibits progestin-dependent angiogenesis, stem cell-like characteristics, and growth of human breast cancer xenografts. Springerplus. 2015 Aug 22;4:444. doi: 10.1186/s40064-015-1242-x, PMID 26312209, PMCID PMC4546074.

  52. Cook MT. Mechanism of metastasis suppression by luteolin in breast cancer. Breast cancer (dove Med press). 2018 Jun 12;10:89-100:S144202. doi: 10.2147/BCTT, PMID 29928143, PMCID PMC6003288.

  53. Sharma A, Chabloz S, Lapides RA, Roider E, Ewald CY. Potential synergistic supplementation of NAD+promoting compounds as a strategy for increasing healthspan. Nutrients. 2023 Jan 14;15(2):445. doi: 10.3390/nu15020445, PMID 36678315, PMCID PMC9861325.

  54. Imran M, Rauf A, Abu-Izneid T, Nadeem M, Shariati MA, Khan IA. Corrigendum to “Luteolin, a flavonoid, as an anticancer agent: a review”. Biomed Pharmacother Biomed Pharmacother. 2019;116:109084. doi: 10.1016/j.biopha.2019.109084, PMID 31178263.

  55. Wang S, Cao M, Xu S, Shi J, Mao X, Yao X. Luteolin alters macrophage polarization to inhibit inflammation. Inflammation. 2020 Feb;43(1):95-108. doi: 10.1007/s10753-019-01099-7, PMID 31673976.

  56. Lin X, Xu Y, Pan X, Xu J, Ding Y, Sun X. Global, regional, and national burden and trend of diabetes in 195 countries and territories: an analysis from 1990 to 2025. Sci Rep. 2020 Sep 8;10(1):14790. doi: 10.1038/s41598-020-71908-9, PMID 32901098, PMCID PMC7478957.

  57. Zhou XR, Ru XC, Xiao C, Pan J, Lou YY, Tang LH. Sestrin2 is involved in the Nrf2-regulated antioxidative signaling pathway in luteolin-induced prevention of the diabetic rat heart from ischemia/reperfusion injury. Food Funct. 2021 Apr 21;12(8):3562-71. doi: 10.1039/d0fo02942d, PMID 33900303.

  58. Liu Y, Tian X, Gou L, Sun L, Ling X, Yin X. Luteolin attenuates diabetes-associated cognitive decline in rats. Brain Res Bull. 2013 May;94:23-9. doi: 10.1016/j.brainresbull.2013.02.001, PMID 23415807.

  59. Brody JS, Spira A. State of the art. Chronic obstructive pulmonary disease, inflammation, and lung cancer. Proc Am Thorac Soc. 2006 Aug;3(6):535-7. doi: 10.1513/pats.200603-089MS, PMID 16921139.

  60. Hussain SP, Harris CC. Inflammation and cancer: an ancient link with novel potentials. Int J Cancer. 2007 Dec 1;121(11):2373-80. doi: 10.1002/ijc.23173, PMID 17893866.

  61. Karin M, Lawrence T, Nizet V. Innate immunity gone awry: linking microbial infections to chronic inflammation and cancer. Cell. 2006 Feb 24;124(4):823-35. doi: 10.1016/j.cell.2006.02.016, PMID 16497591.

  62. Aziz N, Kim MY, Cho JY. Anti-inflammatory effects of luteolin: a review of in vitro, in vivo, and in silico studies. J Ethnopharmacol. 2018 Oct 28;225:342-58. doi: 10.1016/j.jep.2018.05.019, PMID 29801717.

  63. Brody JS, Spira A. State of the art. Chronic obstructive pulmonary disease, inflammation, and lung cancer. Proc Am Thorac Soc. 2006 Aug;3(6):535-7. doi: 10.1513/pats.200603-089MS, PMID 16921139.

  64. Hussain SP, Harris CC. Inflammation and cancer: an ancient link with novel potentials. Int J Cancer. 2007 Dec 1;121(11):2373-80. doi: 10.1002/ijc.23173, PMID 17893866.

  65. Durga M, Nathiya S, Devasena T. Multifarious actions of dietary flavonoids-implications in cancer and cataract. Int J Pharm Biol Sci. 2014;5(2):404-6.

  66. Xagorari A, Papapetropoulos A, Mauromatis A, Economou M, Fotsis T, Roussos C. Luteolin inhibits an endotoxin-stimulated phosphorylation cascade and proinflammatory cytokine production in macrophages. J Pharmacol Exp Ther. 2001 Jan;296(1):181-7, PMID 11123379.

  67. Chen CC, Chow MP, Huang WC, Lin YC, Chang YJ. Flavonoids inhibit tumor necrosis factor-alpha-induced up-regulation of intercellular adhesion molecule-1 (ICAM-1) in respiratory epithelial cells through activator protein-1 and nuclear factor-kappaB: structure-activity relationships. Mol Pharmacol. 2004 Sep;66(3):683-93. doi: 10.1124/mol.66.3, PMID 15322261.

  68. Kumazawa Y, Kawaguchi K, Takimoto H. Immunomodulating effects of flavonoids on acute and chronic inflammatory responses caused by tumor necrosis factor alpha. Curr Pharm Des. 2006;12(32):4271-9. doi: 10.2174/138161206778743565, PMID 17100629.

  69. Ding X, Zheng L, Yang B, Wang X, Ying Y. Luteolin attenuates atherosclerosis via modulating signal transducer and activator of transcription 3-mediated inflammatory response. Drug Des Devel Ther. 2019;13:3899-911. doi: 10.2147/DDDT.S207185, PMID 31819365.

  70. Crupi R, Paterniti I, Ahmad A, Campolo M, Esposito E, Cuzzocrea S. Effects of palmitoylethanolamide and luteolin in an animal model of anxiety/depression. CNS Neurol Disord Drug Targets. 2013 Nov;12(7):989-1001. doi: 10.2174/18715273113129990084, PMID 23844686.

  71. Siracusa R, Paterniti I, Impellizzeri D, Cordaro M, Crupi R, Navarra M. The association of palmitoylethanolamide with luteolin decreases neuroinflammation and stimulates autophagy in Parkinson’s disease model. CNS Neurol Disord Drug Targets. 2015;14(10):1350-65. doi: 10.2174/1871527314666150821102823, PMID 26295827.

  72. Uwishema O, Mahmoud A, Sun J, Correia IFS, Bejjani N, Alwan M. Is Alzheimer’s disease an infectious neurological disease? A review of the literature. Brain Behav. 2022 Aug;12(8):e2728. doi: 10.1002/brb3.2728, PMID 35879909, PMCID PMC9392514.

  73. Ali F, Rahul, Jyoti S, Naz F, Ashafaq M, Shahid M. Therapeutic potential of luteolin in transgenic Drosophila model of Alzheimer’s disease. Neurosci Lett. 2019;692:90-9. doi: 10.1016/j.neulet.2018.10.053, PMID 30420334.

  74. Kou JJ, Shi JZ, He YY, Hao JJ, Zhang HY, Luo DM. Luteolin alleviates cognitive impairment in Alzheimer’s disease mouse model via inhibiting endoplasmic reticulum stress-dependent neuroinflammation. Acta Pharmacol Sin. 2022 Apr;43(4):840-9. doi: 10.1038/s41401-021-00702-8, PMID 34267346, PMCID PMC8975883.

  75. Wang HR, Pei SY, Fan DX, Liu YH, Pan XF, Song FX. Luteolin protects pheochromocytoma (PC-12) cells against Aβ25-35-induced cell apoptosis through the ER/ERK/MAPK signalling pathway. Evid Based Complement Alternat Med. 2020 Nov 30;2020:2861978. doi: 10.1155/2020/2861978, PMID 33335556, PMCID PMC7723489.

  76. Park S, Kim DS, Kang S, Kim HJ. The combination of luteolin and l-theanine improved Alzheimer disease-like symptoms by potentiating hippocampal insulin signaling and decreasing neuroinflammation and norepinephrine degradation in amyloid-β-infused rats. Nutr Res. 2018 Dec;60:116-31. doi: 10.1016/j.nutres.2018.09.010, PMID 30527255.

  77. Zhao G, Yao Yue C, Qin GW, Guo LH. Luteolin from purple perilla mitigates ROS insult particularly in primary neurons. Neurobiol Aging. 2012 Jan;33(1):176-86. doi: 10.1016/j.neurobiolaging.2010.02.013, PMID 20382451.

  78. Qin L, Chen Z, Yang L, Shi H, Wu H, Zhang B. Luteolin-7-O-glucoside protects dopaminergic neurons by activating estrogen-receptor-mediated signaling pathway in MPTP-induced mice. Toxicology. 2019 Oct 1;426:152256. doi: 10.1016/j.tox.2019.152256, PMID 31381935.

  79. Loos RJF, Yeo GSH. The genetics of obesity: from discovery to biology. Nat Rev Genet. 2022 Feb;23(2):120-33. doi: 10.1038/s41576-021-00414-z. PMID 34556834, PMCID PMC8459824.

  80. Zhang Z, Wang J, Lin Y, Chen J, Liu J, Zhang X. Nutritional activities of luteolin in obesity and associated metabolic diseases: an eye on adipose tissues. Crit Rev Food Sci Nutr. 2022 Oct 27:1-15. doi: 10.1080/10408398.2022.2138257, PMID 36300856.

  81. Kwon EY, Choi MS. Luteolin targets the toll-like receptor signaling pathway in prevention of hepatic and adipocyte fibrosis and insulin resistance in diet-induced obese mice. Nutrients. 2018 Oct 3;10(10):1415. doi: 10.3390/nu10101415, PMID 30282902, PMCID PMC6213163.

  82. Park HS, Lee K, Kim SH, Hong MJ, Jeong NJ, Kim MS. Luteolin improves hypercholesterolemia and glucose intolerance through LXRα-dependent pathway in diet-induced obese mice. J Food Biochem. 2020 Sep;44(9):e13358. doi: 10.1111/jfbc.13358, PMID 32598492.

  83. Ando C, Takahashi N, Hirai S, Nishimura K, Lin S, Uemura T. Luteolin, a food-derived flavonoid, suppresses adipocyte-dependent activation of macrophages by inhibiting JNK activation. FEBS Lett. 2009 Nov 19;583(22):3649-54. doi: 10.1016/j.febslet.2009.10.045, PMID 19854181.

  84. Ike S, Onyema C. Cardiovascular diseases in Nigeria: what has happened in the past 20 years? Nig J Cardiol. 2020;17(1). doi: 10.4103/njc.njc_33_19.

  85. Mendis S, Puska P, Norrving B, World Health Organization. Global atlas on cardiovascular disease prevention and control. World Health Organization; 2011.

  86. Alissa EM, Ferns GA. Dietary fruits and vegetables and cardiovascular diseases risk. Crit Rev Food Sci Nutr. 2017 Jun 13;57(9):1950-62. doi: 10.1080/10408398.2015.1040487, PMID 26192884.

  87. Liu D, Luo H, Qiao C. SHP-1/STAT3 interaction is related to luteolin-induced myocardial ischemia protection. Inflammation. 2022 Feb;45(1):88-99. doi: 10.1007/s10753-021-01530-y, PMID 34460026, PMCID PMC8403691.

  88. Hu W, Xu T, Wu P, Pan D, Chen J, Chen J. Luteolin improves cardiac dysfunction in heart failure rats by regulating sarcoplasmic reticulum Ca2+-ATPase 2a. Sci Rep. 2017 Jan 23;7:41017. doi: 10.1038/srep41017, PMID 28112209, PMCID PMC5253630.

  89. Fang F, Li D, Pan H, Chen D, Qi L, Zhang R. Luteolin inhibits apoptosis and improves cardiomyocyte contractile function through the PI3K/Akt pathway in simulated ischemia/reperfusion. Pharmacology. 2011;88(3-4):149-58. doi: 10.1159/000330068, PMID 21934351.

  90. Zhu S, Xu T, Luo Y, Zhang Y, Xuan H, Ma Y. Luteolin enhances sarcoplasmic reticulum Ca2+-ATPase activity through p38 MAPK signaling thus improving rat cardiac function after ischemia/reperfusion. Cell Physiol Biochem. 2017;41(3):999-1010. doi: 10.1159/000460837, PMID 28222421.

  91. Xiao C, Xia ML, Wang J, Zhou XR, Lou YY, Tang LH. Luteolin attenuates cardiac ischemia/reperfusion injury in diabetic rats by modulating Nrf2 antioxidative function. Oxid Med Cell Longev. 2019 Apr 8;2019:2719252. doi: 10.1155/2019/2719252, PMID 31089405, PMCID PMC6476158.

  92. Luo Y, Shang P, Li D. Luteolin: a flavonoid that has multiple cardio-protective effects and its molecular mechanisms. Front Pharmacol. 2017 Oct 6;8:692. doi: 10.3389/fphar.2017.00692, PMID 29056912, PMCID PMC5635727.

  93. Theoharides TC, Cholevas C, Polyzoidis K, Politis A. Long-COVID syndrome-associated brain fog and chemofog: luteolin to the rescue. BioFactors. 2021 Mar;47(2):232-41. doi: 10.1002/biof.1726, PMID 33847020, PMCID PMC8250989.

  94. Galeotti C, Bayry J. Autoimmune and inflammatory diseases following COVID-19. Nat Rev Rheumatol. 2020 Aug;16(8):413-4. doi: 10.1038/s41584-020-0448-7, PMID 32499548, PMCID PMC7271827.

  95. Canna SW, Cron RQ. Highways to hell: mechanism-based management of cytokine storm syndromes. J Allergy Clin Immunol. 2020 Nov;146(5):949-59. doi: 10.1016/j.jaci.2020.09.016, PMID 33007328, PMCID PMC7522622.

  96. Baig AM. Chronic COVID syndrome: need for an appropriate medical terminology for long-COVID and COVID long-haulers. J Med Virol. 2021 May;93(5):2555-6. doi: 10.1002/jmv.26624, PMID 33095459.

  97. Theoharides TC, Conti P. COVID-19 and multisystem inflammatory syndrome, or is it mast cell activation syndrome? J Biol Regul Homeost Agents. 2020 Sep-Oct;34(5):1633-6. doi: 10.23812/20-EDIT3, PMID 33023287.

  98. Hatziagelaki E, Adamaki M, Tsilioni I, Dimitriadis G, Theoharides TC. Myalgic encephalomyelitis/chronic fatigue syndrome-metabolic disease or disturbed homeostasis due to focal inflammation in the hypothalamus? J Pharmacol Exp Ther. 2018 Oct;367(1):155-67. doi: 10.1124/jpet.118.250845, PMID 30076265.

  99. Natelson BH. Myalgic encephalomyelitis/chronic fatigue syndrome and fibromyalgia: definitions, similarities, and differences. Clin Ther. 2019 Apr;41(4):612-8. doi: 10.1016/j.clinthera.2018.12.016, PMID 30795933, PMCID PMC6589349.

  100. Akin C, Valent P, Metcalfe DD. Mast cell activation syndrome: proposed diagnostic criteria. J Allergy Clin Immunol. 2010 Dec;126(6):1099-104.e4. doi: 10.1016/j.jaci.2010.08.035, PMID 21035176, PMCID PMC3753019.

  101. Theoharides TC, Tsilioni I, Ren H. Recent advances in our understanding of mast cell activation-or should it be mast cell mediator disorders? Expert Rev Clin Immunol. 2019 Jun;15(6):639-56. doi: 10.1080/1744666X.2019.1596800, PMID 30884251, PMCID PMC7003574.

  102. Theoharides TC, Valent P, Akin C. Mast Cells, Mastocytosis, and related disorders. N Engl J Med. 2015 Jul 9;373(2):163-72. doi: 10.1056/NEJMra1409760, PMID 26154789.

  103. Liang W, Guan W, Chen R, Wang W, Li J, Xu K. Cancer patients in SARS-CoV-2 infection: a nationwide analysis in China. Lancet Oncol. 2020 Mar;21(3):335-7. doi: 10.1016/S1470-2045(20)30096-6, PMID 32066541, PMCID PMC7159000.

  104. Raffa RB, Duong PV, Finney J, Garber DA, Lam LM, Mathew SS. Is ’chemo-fog’/’chemo-brain’ caused by cancer chemotherapy? J Clin Pharm Ther. 2006 Apr;31(2):129-38. doi: 10.1111/j.1365-2710.2006.00726.x, PMID 16635046.

  105. Raffa RB. A proposed mechanism for chemotherapy-related cognitive impairment (”chemo-fog”). J Clin Pharm Ther. 2011 Jun;36(3):257-9. doi: 10.1111/j.1365-2710.2010.01188.x, PMID 21545608, PMCID PMC3249621.

  106. Mitchell T, Turton P. 'Chemobrain’: concentration and memory effects in people receiving chemotherapy-a descriptive phenomenological study. Eur J Cancer Care (Engl). 2011 Jul;20(4):539-48. doi: 10.1111/j.1365-2354.2011.01244.x, PMID 21443746.

  107. Plos One Staff. Correction: chemobrain experienced by breast cancer survivors: a meta-ethnography study investigating research and care implications. Plos One. 2015;10(2):e0117740. doi: 10.1371/journal.pone.0117740, PMID 25647507.

  108. Baer W. Chemobrain: an opportunity in cancer survivorship to enhance patient wellness. J Oncol Pract. 2017 Dec;13(12):794-6. doi: 10.1200/JOP.2017.027987, PMID 29232541.

  109. Gutmann DH. Clearing the fog surrounding chemobrain. Cell. 2019 Jan 10;176(1-2):2-4. doi: 10.1016/j.cell.2018.12.027, PMID 30633904.

  110. Henderson FM, Cross AJ, Baraniak AR. ”A new normal with chemobrain”: experiences of the impact of chemotherapy-related cognitive deficits in long-term breast cancer survivors. Health Psychol Open. 2019 Mar 5;6(1):2055102919832234. doi: 10.1177/2055102919832234, PMID 30873289, PMCID PMC6405778.

  111. Eide S, Feng ZP. Doxorubicin chemotherapy-induced ’chemo-brain’: meta-analysis. Eur J Pharmacol. 2020 Aug 15;881:173078. doi: 10.1016/j.ejphar.2020.173078, PMID 32505665.

  112. El-Agamy SE, Abdel Aziz AK, Esmat A, Azab SS. Chemotherapy and cognition: comprehensive review on doxorubicin-induced chemobrain. Cancer Chemother Pharmacol. 2019 Jul;84(1):1-14. doi: 10.1007/s00280-019-03827-0, PMID 30955080.

  113. Ongnok B, Chattipakorn N, Chattipakorn SC. Doxorubicin and cisplatin induced cognitive impairment: the possible mechanisms and interventions. Exp Neurol. 2020 Feb;324:113118. doi: 10.1016/j.expneurol.2019.113118, PMID 31756316.

  114. Gibson EM, Nagaraja S, Ocampo A, Tam LT, Wood LS, Pallegar PN. Methotrexate chemotherapy induces persistent tri-glial dysregulation that underlies chemotherapy-related cognitive impairment. Cell. 2019 Jan 10;176(1-2):43-55.e13. doi: 10.1016/j.cell.2018.10.049, PMID 30528430, PMCID PMC6329664.

  115. Levy D, Burstein R, Kainz V, Jakubowski M, Strassman AM. Mast cell degranulation activates a pain pathway underlying migraine headache. Pain. 2007 Jul;130(1-2):166-76. doi: 10.1016/j.pain.2007.03.012, PMID 17459586, PMCID PMC2045157.

  116. Calvi E, Marchetti M, Santagata F, Luppi C, Coppo E, Massaia M. Similar neurocognitive patterns in patients treated with lenalidomide: chemobrain effect? Neurocase. 2019 Dec;25(6):259-62. doi: 10.1080/13554794.2019.1666876, PMID 31522586.

  117. Lee S, Lee HJ, Kang H, Kim EH, Lim YC, Park H. Trastuzumab induced chemobrain, atorvastatin rescued chemobrain with enhanced anticancer effect and without hair loss-side effect. J Clin Med. 2019 Feb 11;8(2):234. doi: 10.3390/jcm8020234, PMID 30754707, PMCID PMC6406319.

  118. Theoharides TC. Stress, inflammation, and autoimmunity: the 3 modern Erinyes. Clin Ther. 2020 May;42(5):742-4. doi: 10.1016/j.clinthera.2020.04.002. PMID 32354496, PMCID PMC7165270.

  119. Theoharides TC. The impact of psychological stress on mast cells. Ann Allergy Asthma Immunol. 2020 Oct;125(4):388-92. doi: 10.1016/j.anai.2020.07.007, PMID 32687989.

  120. Steenblock C, Todorov V, Kanczkowski W, Eisenhofer G, Schedl A, Wong ML. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and the neuroendocrine stress axis. Mol Psychiatry. 2020 Aug;25(8):1611-7. doi: 10.1038/s41380-020-0758-9, PMID 32382135, PMCID PMC7204611.

  121. Ye Q, Wang B, Mao J. The pathogenesis and treatment of the `Cytokine Storm’ in COVID-19. J Infect. 2020;80(6):607-13. doi: 10.1016/j.jinf.2020.03.037, PMID 32283152.

  122. Iannaccone G, Scacciavillani R, Del Buono MG, Camilli M, Ronco C, Lavie CJ. Weathering the cytokine storm in COVID-19: therapeutic implications. Cardiorenal Med. 2020;10(5):277-87. doi: 10.1159/000509483, PMID 32599589, PMCID PMC7360507.

  123. Jamilloux Y, Henry T, Belot A, Viel S, Fauter M, El Jammal T. Should we stimulate or suppress immune responses in COVID-19? Cytokine and anti-cytokine interventions. Autoimmun Rev. 2020 Jul;19(7):102567. doi: 10.1016/j.autrev.2020.102567, PMID 32376392, PMCID PMC7196557.

  124. Ronconi G, Tete G, Kritas SK, Gallenga CE, Al Caraffa RR, Ross R. SARS-CoV-2, which induces COVID-19, causes Kawasaki-like disease in children: role of pro-inflammatory and anti-inflammatory cytokines. J Biol Regul Homeost Agents. 2020 May-Jun;34(3):767-73. doi: 10.23812/editorial-ronconi-E-59, PMID 32476380.

  125. Kazama I. Stabilizing mast cells by commonly used drugs: a novel therapeutic target to relieve post-COVID syndrome? Drug Discov Ther. 2020 Nov 4;14(5):259-61. doi: 10.5582/ddt.2020.03095, PMID 33116043.

  126. Theoharides TC. COVID-19, pulmonary mast cells, cytokine storms, and beneficial actions of luteolin. BioFactors. 2020 May;46(3):306-8. doi: 10.1002/biof.1633, PMID 32339387, PMCID PMC7267424.

  127. Harwood M, Danielewska Nikiel B, Borzelleca JF, Flamm GW, Williams GM, Lines TC. A critical review of the data related to the safety of quercetin and lack of evidence of in vivo toxicity, including lack of genotoxic/carcinogenic properties. Food Chem Toxicol. 2007 Nov;45(11):2179-205. doi: 10.1016/j.fct.2007.05.015. PMID 17698276.

  128. Okamoto T. Safety of quercetin for clinical application (Review). Int J Mol Med. 2005 Aug;16(2):275-8. PMID 16012761.

  129. Taliou A, Zintzaras E, Lykouras L, Francis K. An open-label pilot study of a formulation containing the anti-inflammatory flavonoid luteolin and its effects on behavior in children with autism spectrum disorders. Clin Ther. 2013 May;35(5):592-602. doi: 10.1016/j.clinthera.2013.04.006, PMID 23688534.

  130. Theoharides TC, Conti P, Economu M. Brain inflammation, neuropsychiatric disorders, and immunoendocrine effects of luteolin. J Clin Psychopharmacol. 2014 Apr;34(2):187-9. doi: 10.1097/JCP.0000000000000084, PMID 24525647.

  131. Andres S, Pevny S, Ziegenhagen R, Bakhiya N, Schafer B, Hirsch Ernst KI. Safety aspects of the use of quercetin as a dietary supplement. Mol Nutr Food Res. 2018 Jan;62(1). doi: 10.1002/mnfr.201700447, PMID 29127724.

  132. Kempuraj D, Madhappan B, Christodoulou S, Boucher W, Cao J, Papadopoulou N. Flavonols inhibit proinflammatory mediator release, intracellular calcium ion levels and protein kinase C theta phosphorylation in human mast cells. Br J Pharmacol. 2005 Aug;145(7):934-44. doi: 10.1038/sj.bjp.0706246, PMID 15912140, PMCID PMC1576204.

  133. Seelinger G, Merfort I, Schempp CM. Anti-oxidant, anti-inflammatory and anti-allergic activities of luteolin. Planta Med. 2008 Nov;74(14):1667-77. doi: 10.1055/s-0028-1088314, PMID 18937165.

  134. Calis Z, Mogulkoc R, Baltaci AK. The roles of flavonols/flavonoids in neurodegeneration and neuroinflammation. Mini Rev Med Chem. 2020;20(15):1475-88. doi: 10.2174/1389557519666190617150051, PMID 31288717.

  135. Jager AK, Saaby L. Flavonoids and the CNS. Molecules. 2011 Feb 10;16(2):1471-85. doi: 10.3390/molecules16021471, PMID 21311414, PMCID PMC6259921.

  136. Leyva Lopez N, Gutierrez Grijalva EP, Ambriz Perez DL, Heredia JB. Flavonoids as cytokine modulators: a possible therapy for inflammation-related diseases. Int J Mol Sci. 2016 Jun 9;17(6):921. doi: 10.3390/ijms17060921, PMID 27294919, PMCID PMC4926454.

  137. Calis Z, Mogulkoc R, Baltaci AK. The roles of flavonols/flavonoids in neurodegeneration and neuroinflammation. Mini Rev Med Chem. 2020;20(15):1475-88. doi: 10.2174/1389557519666190617150051, PMID 31288717.

  138. Devi SA, Chamoli A. Polyphenols as an effective therapeutic intervention against cognitive decline during normal and pathological brain aging. Adv Exp Med Biol. 2020;1260:159-74. doi: 10.1007/978-3-030-42667-5_7, PMID 32304034.

  139. Del Rio JA, Fuster MD, Gomez P, Porras I, Garcia Lidon A, Ortuno A. Citrus limon: a source of flavonoids of pharmaceutical interest. Food Chem. 2004 Feb;84(3):457-61. doi: 10.1016/S0308-8146(03)00272-3.

  140. Havsteen BH. The biochemistry and medical significance of the flavonoids. Pharmacol Ther. 2002 Nov-Dec;96(2-3):67-202. doi: 10.1016/s0163-7258(02)00298-x, PMID 12453566.

  141. Lopez Lazaro M. Distribution and biological activities of the flavonoid luteolin. Mini Rev Med Chem. 2009 Jan;9(1):31-59. doi: 10.2174/138955709787001712, PMID 19149659.

  142. Choi CW, Jung HA, Kang SS, Choi JS. Antioxidant constituents and a new triterpenoid glycoside from Flos Lonicerae. Arch Pharm Res. 2007 Jan;30(1):1-7. doi: 10.1007/BF02977770, PMID 17328234.

  143. Miyazawa M, Hisama M. Antimutagenic activity of flavonoids from Chrysanthemum morifolium. Biosci Biotechnol Biochem. 2003 Oct;67(10):2091-9. doi: 10.1271/bbb.67.2091, PMID 14586095.

  144. Lin Y, Shi R, Wang X, Shen HM. Luteolin, a flavonoid with potential for cancer prevention and therapy. Curr Cancer Drug Targets. 2008 Nov;8(7):634-46. doi: 10.2174/156800908786241050, PMID 18991571, PMCID PMC2615542.

  145. Kumar SS, Shanmugasundaram P, Komala M, Bhargavi B, Padmavathy J. Nanoparticle formulation of bioflavonoids for enhanced anti-cancer activity. Int J App Pharm. 2020;12(5):29-35. doi: 10.22159/ijap.2020v12i5.38425.

  146. Fessi HP, Puisieux F, Devissaguet JP, Ammoury N, Benita S. Nanocapsule formation by interfacial polymer deposition following solvent displacement. International Journal of Pharmaceutics. 1989 Oct 1;55(1):R1-4. doi: 10.1016/0378-5173(89)90281-0.

  147. Reis CP, Neufeld RJ, Ribeiro AJ, Veiga F. Nanoencapsulation I. Methods for preparation of drug-loaded polymeric nanoparticles. Nanomedicine. 2006;2(1):8-21. doi: 10.1016/j.nano.2005.12.003, PMID 17292111.

  148. Kumari A, Yadav SK, Yadav SC. Biodegradable polymeric nanoparticles based drug delivery systems. Colloids Surf B Biointerfaces. 2010 Jan 1;75(1):1-18. doi: 10.1016/j.colsurfb.2009.09.001, PMID 19782542.

  149. Zanwar AA, Badole SL, Shende PS, Hegde MV, Bodhankar SL. Antioxidant role of catechin in health and disease. Inpolyphenols Hum Health Dis. 2014 Jan 1:267-71.

  150. Balakrishnan BB, Krishnasamy K. Evaluation of free radical screening and antioxidant potential of moringa concanensis nimmo-a medicinal plant used in Indian traditional medication system. Int J Pharm Pharm Sci. 2018;10(7):91-7. doi: 10.22159/ijpps.2018v10i7.26403.