Next Article in Journal
Serum-Based Biomarkers in Neurodegeneration and Multiple Sclerosis
Next Article in Special Issue
Meleagrin Isolated from the Red Sea Fungus Penicillium chrysogenum Protects against Bleomycin-Induced Pulmonary Fibrosis in Mice
Previous Article in Journal
Human iPSC Modeling of Genetic Febrile Seizure Reveals Aberrant Molecular and Physiological Features Underlying an Impaired Neuronal Activity
Previous Article in Special Issue
Post-Bariatric Hypoglycemia Is Associated with Endothelial Dysfunction and Increased Oxidative Stress
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Potential and Therapeutic Roles of Diosmin in Human Diseases

by
Etimad Huwait
1,2,* and
Mohammad Mobashir
3,4,*
1
Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah 22254, Saudi Arabia
2
Cell Culture Lab, Experimental Biochemistry Unit, King Fahad Medical Research Centre, King Abdulaziz University, Jeddah 22252, Saudi Arabia
3
SciLifeLab, Department of Oncology and Pathology, Karolinska Institutet, P.O. Box 1031, 17121 Stockholm, Sweden
4
Genome Biology Lab, Department of Biosciences, Faculty of Natural Science, Jamia Millia Islamia, New Delhi 110025, India
*
Authors to whom correspondence should be addressed.
Biomedicines 2022, 10(5), 1076; https://doi.org/10.3390/biomedicines10051076
Submission received: 20 March 2022 / Revised: 26 April 2022 / Accepted: 27 April 2022 / Published: 6 May 2022

Abstract

:
Because of their medicinal characteristics, effectiveness, and importance, plant-derived flavonoids have been a possible subject of research for many years, particularly in the last decade. Plants contain a huge number of flavonoids, and Diosmin, a flavone glycoside, is one of them. Numerous in-vitro and in-vivo studies have validated Diosmin’s extensive range of biological capabilities which present antioxidative, antihyperglycemic, anti-inflammatory, antimutagenic, and antiulcer properties. We have presented this review work because of the greater biological properties and influences of Diosmin. We have provided a brief overview of Diosmin, its pharmacology, major biological properties, such as anti-cancer, anti-diabetic, antibacterial, anticardiovascular, liver protection, and neuroprotection, therapeutic approach, potential Diosmin targets, and pathways that are known to be associated with it.

1. Introduction

Diosmin is a flavonoid found in citrus fruits. Flavonoids are anti-inflammatory plant compounds that protect your body from free radicals and other unstable molecules. The most prevalent uses for Diosmin include hemorrhoids and leg sores caused by poor blood flow. It is also claimed to heal a variety of diseases, albeit there is no hard evidence to back up these claims [1,2,3]. Hesperidin is frequently used with Diosmin which is another plant chemical. Diosmin may work by reducing swelling and restoring normal vein function. It also appears to have antioxidative effects [1,4,5,6]. Diosmin was first found in 1925 in the wort plant and has since been used as a natural treatment for hemorrhoids, varicose veins, venous insufficiency, leg ulcers, and other circulatory issues [7]. It may help people with venous insufficiency, a condition in which blood flow is restricted, reduce inflammation, and restore normal blood flow [8,9,10,11].
Due to its beneficial effects in a number of major human organ systems, such as the cardiovascular system, Diosmin is now one of the most sought-after natural compounds in the treatment of a variety of human diseases, including chronic venous insufficiency (CVI), a progressive disorder affecting an increasing number of people (Figure 1). Diosmin’s effects in combination with other flavonoids, as well as its delivery routes, have also been investigated in details [4,12,13,14]. Diosmin was shown to have anti-oxidant characteristics, allowing it to successfully modulate the activities of a range of variables (including enzymes and biomarkers) connected to oxidative imbalance in a variety of illnesses. Induced apoptosis has been shown to have anti-cancer characteristics and activity in a variety of cell types. In MCF-7, MDA-MB-231, and SKBR-3 breast cancer cells, as well as DU145 prostate cancer cells, A431 skin cancer cells, and colon, oral, urinary bladder, and esophageal carcinogenesis, Diosmin has been found to trigger apoptosis [15,16,17,18]. Diosmin has also been found to have anti-diabetic properties. Diosmin has therapeutic effects in the event of diabetes and associated complications, such as neuropathy and dyslipidemia. Some evidence of a weak antimicrobial action was also found. A combination of Diosmin and hesperidin has been shown to be particularly effective in the treatment of chronic venous insufficiency and hemorrhoids. Diosmin is a great medical medication that can be used alone or in conjunction with other flavonoids [1,14,17].
Intestinal microflora enzymes rapidly hydrolyze Diosmin into its aglycone form, diosmetin, which is easily absorbed and dispersed throughout the body, according to previous pharmacokinetic studies. In addition, Diosmin is broken down into phenolic acids or glycine-conjugated derivatives, which are excreted in the urine. The presence of break down products such as alkyl-phenolic acids demonstrated that the flavonoids followed a similar metabolic path to other flavonoids [12,18,19,20].
Diosmin’s anti-oxidant properties were discovered to have beneficial effects in a number of illnesses, where it successfully reduced the activities of several components associated to oxidative imbalance, such as enzymes/biomarkers. Induced apoptosis has been shown to have anti-cancer characteristics and activity in a variety of cell types [7,11,21]. In cancer cell lines, such as MCF-7, MDA-MB-231, SKBR-3; DU145; A431; colon, oral, urinary bladder, and esophageal carcinogenesis, Diosmin promotes apoptosis. Diosmin has also been found to have anti-diabetic properties. Diosmin has therapeutic effects in the event of diabetes and associated complications, such as neuropathy and dyslipidemia. Some evidence of a weak antimicrobial action was also found. A combination of Diosmin and hesperidin has been shown to be particularly effective in the treatment of chronic venous insufficiency and hemorrhoids. Diosmin is a great medical medication that can be used alone or in conjunction with other flavonoids [12,18,19,20].
Oxidative stress has been linked to the development of various diseases, including myocardial ischemia, cerebral ischemia–reperfusion injury, diabetes, neuronal cell injury, hypoxia, and cancer [16,22]. In rat models, Diosmin was found to exhibit anti-oxidant effects and to stimulate human neutrophils. Because of its anti-oxidant properties, Diosmin has been shown to offer a variety of therapeutic effects for illnesses characterized by oxidative stress. Many diseases, including arthritis, allergies, asthma, autoimmune diseases, atherosclerosis, diabetes, and cancer, are caused by inflammation. Inflammatory indicators, or biomarkers with elevated levels, can be used to detect inflammation [18,23]. The most common inflammatory markers include immune system cells, such as neutrophils, basophils, eosinophils, platelets, macrophages, and others, cell surface receptors and adhesion molecules such as selectins (L-selectin, P-selectin, and E-selectin), and soluble mediators such as cytokines (IL-1, IL-2, IL-6, TNF-α, TGF-ß, and IFN-γ) (complement factors, C-reactive protein and the coagulation factor fibrinogen) [8,10,15,18].
Recently published studies have shown that Diosmin has dose-dependent pro-apoptotic effects on a range of animal cancers, including breast, prostate, colon, oral, and urinary bladder tumors. Diosmin has been reported to trigger premature senescence in a variety of cancer cell types [23,24]. Other breast cancer cell lines, including MDA-MB-231 and SKBR-3, responded to Diosmin, but MCF-7 was found to be the most responsive. At lower doses, Diosmin produced G2/M cell cycle arrest, elevated p53, p21, and p27 levels, increased SA—gal activity, oxidative stress, and DNA damage in MCF-7 cells, all of which are associated with ageing. Apoptosis can be triggered by increased levels of nitric oxide, total ROS, total superoxide, mitochondrial production, and protein carbonylation. In the treatment of diabetes and its complications, Diosmin has been proven to have therapeutic effects. Diabetes mellitus is a chronic disease characterized by abnormal glucose, protein, and lipid metabolism due to a lack of or reduced insulin action. Diosmin possesses anti-hyperglycemic effects, according to numerous studies [13,14,17,22,25].
Plant-based bioactive components that are effective against fungus, yeasts, and bacteria, as well as insects, nematodes, and other plants, are known as phytochemicals. They can inhibit the synthesis of peptidoglycans, damage microbial membrane structures, alter the hydrophobicity of bacterial membrane surfaces, and interfere with quorum sensing (QS). The antibacterial activities of Diosmin were previously investigated by synthesizing silver nanoparticles from it (AgNPs). Flavonoids-rich diets aid in the improvement of cardiovascular parameters. Anti-platelet activity has been demonstrated for Diosmin. Overall binding in the heparin binding region, which includes helix A, D, and N-terminal residues, is expected to improve with Diosmin sulfation, potentially leading to the development of new anti-thrombin candidates. Diosmin’s anti-thrombotic properties are confirmed by changes in the protein composition of rats with venous thrombosis. Diosmin may stimulate endothelial cell growth by targeting the centrosome-associated protein 350 (CEP350) [5,13,26,27].
Hypoxia, angiogenesis, inflammation, and intrapulmonary vasodilation characterize hepatopulmonary syndrome (HPS), a severe consequence of hepatic cirrhosis. TNF/VEGF, IGF-1/PI3K/AKT, and FGF-1/ANG-2 signaling pathways may be involved in HPS development and can be restored by Diosmin therapy in a chronic bile duct ligation (CBDL)-induced rat model. In the central nervous system, many flavone derivatives have been found as potential GABAA receptor ligands, where they interact with the benzodiazepine binding site to generate depressive effects. Flavonoid glycosides cannot directly regulate the activity of the GABAA receptor. Diosmin contains sedative and sleep-inducing characteristics that have nothing to do with GABAA receptor regulation. 6-C-glycoside-diosmetin has been shown to have memory-enhancing and anxiolytic-like effects by activating the GABAA receptor, which is controversial [7,12,23].

2. Pharmacology of Diosmin

Flavonoids are phytochemicals that have been proven to have medicinal properties [28,29,30]. Diosmin, a flavone glycoside, is also found in hesperidin [9,12]. Daflon is an oral phlebotonic flavonoid combination based on Diosmin and Hesperidin that is widely used to treat and protect blood vessel disorders. After oral administration, Diosmin is converted to diosmetin, which is subsequently absorbed and esterified into glucuronide conjugates that are excreted in the urine [11,23,24,31]. Diosmin’s pharmacological properties have been investigated in a number of in vitro and in vivo studies, and it has been found to have anti-inflammatory, antioxidant, antidiabetic, antibacterial, antihyperlipidemic, and antifibrotic properties in a variety of disease models, as described in the previous section. In a number of clinical studies, Diosmin has also been proven to have a number of positive properties. According to toxicological investigations, Diosmin has a favorable safety profile. As a result, Diosmin has the potential to be an effective and safe treatment for a wide range of conditions. Diosmin, on the other hand, inhibits a variety of metabolic enzymes, encouraging clinical trials to look into its potential therapeutic efficacy and safety in a variety of illnesses while taking into consideration potential interactions. Diosmin is a venoactive drug that works on blood vessels in a variety of ways to improve circulation. It improves lymphatic drainage and microcirculation, as well as venous tone and suppleness. Diosmin is often used to improve vascular health in people with chronic venous disease for these reasons, and it has been demonstrated to improve quality of life. Diosmin also has antioxidant characteristics and scavenges oxygen free radicals, lowering oxidative stress levels that can be detected by biomarkers such as prostaglandin isoprostane precursors [2,11,12,31,32].

3. Critical Properties of Diosmin

3.1. Anti-Oxidant Property

Oxidative stress has been linked to the development of various diseases, including myocardial ischemia, cerebral ischemia–reperfusion injury, diabetes, neuronal cell injury, hypoxia, and cancer. In rat models, Diosmin was found to exhibit anti-oxidant effects and to stimulate human neutrophils. Because of its anti-oxidant properties, Diosmin has been shown to offer a variety of therapeutic effects for illnesses characterized by oxidative stress [22,33,34,35,36,37,38,39,40]. Diosmin was found to have significant anti-diabetic effects in diabetic rats fed streptozotocin nicotinamide (STZ-NA). When diabetic control rats were compared to normal control rats, anti-oxidant enzyme activities, such as glutathione-S-transferase (GST), glutathione peroxidase (GPx), superoxide dismutase (SOD), and catalase (CAT), as well as levels of low molecular weight antioxidants, such as vitamin C, vitamin E, and reduced glutathione (GSH), were found to be low, whereas lipid peroxidation Hyperglycemia with a significant drop in plasma insulin levels was also seen [7,11,27].
Oral administration of Diosmin enhanced the glycemic and anti-oxidant status of diabetic rats, according to previous studies. Diosmin treatment was also found to reduce lipid peroxidation [21,22,41]. Due to its anti-oxidant properties, Diosmin functions as an anti-hypertensive drug in deoxycorticosterone acetate (DOCA)-salt-induced hypertensive rats. Hypertension is characterized by an excessive production of reactive oxygen species. In DOCA-salt-treated rats, non-enzymatic and enzymatic antioxidants were found to be lower, while lipid peroxidation products (thiobarbituric acid reactive substances, lipid hydroperoxides, and conjugated dienes) were found to be significantly higher in blood plasma and tissues, such as the liver, kidney, heart, and aorta. Antioxidant levels were restored to near-normal levels, and lipid peroxidation products were reduced as a result of the Diosmin therapy. These findings were confirmed by histopathological examinations of the kidney and heart [1,11,31,42] Diosmin was found to have a hepatoprotective effect against ferrous sulfate-induced liver damage in adult male albino rats. Too much iron causes oxidative stress, reactive oxygen species (ROS), lipid peroxidation, inflammation, and tissue necrosis. Elevated ALT, AST, ALP, GGT, LDH activity, and bilirubin levels indicate hepatocyte membrane damage [21]. After Diosmin treatment, these values were considerably normalized. Because it maintains membrane integrity, decreases oxidative stress, and aids in the correction of dyslipidemia, Diosmin is a good hepatoprotective drug. The antioxidant and anti-inflammatory properties of Diosmin are primarily responsible for its hepatoprotective effects [11]. According to Senthamizhselvan et al., pre-treatment with Diosmin reduces oxidative stress in the rat heart after ischemia/reperfusion [43]. Ischemia–reperfusion injury occurs when ischemic tissue is reperfused, causing oxidative stress and cellular damage. The activities of enzyme antioxidants (SOD, CAT, and GPx) and GSH levels declined when hearts of control rats (no Diosmin pre-treatment) were subjected to an ischemia/reperfusion regimen, although the levels of lipid peroxidation products rose [44,45]. After 7 days of oral therapy with Diosmin (50 and 100 mg/kg), the activities of enzymatic antioxidants and GSH levels were found to be increased, while the levels of lipid peroxidation products were found to be reduced [6,43]. Ischemia–reperfusion injury produces edema and tissue destruction in the retina, resulting in vision loss. Diosmin was discovered to help male Wistar rats with retinal edema by maintaining the blood-retinal barrier and lowering vascular permeability. Diosmin’s ability to change the VEGF/PEDF ratio could explain its protective properties. The levels of malondialdehyde (MDA) and the activity of total-superoxide dismutase (T-SOD), glutathione peroxidase (GSH-Px), and catalase (CAT) in retinal tissues that had been altered by ischemia–reperfusion injury, recovered to normal after Diosmin administration (Tong et al., 2013). Diosmin protects male CD-1 mice from brain ischemia–reperfusion injury by stimulating the JAK2/STAT3 signaling pathway [3,13]. In human polymorphonuclear neutrophils stimulated in vitro by phorbol myristate acetate, Cypriani et al. discovered that S5682 (Daflon 500 mg), a purified flavonoid fraction including Diosmin and hesperidin in a 9:1 ratio, has anti-oxidative effects (Cypriani et al., 1993). Diosmin’s free-radical scavenging activity is one of the reasons for its protection against myocardial infarction [42,46].

3.2. Anti-Inflammatory Property

Many diseases, including arthritis, allergies, asthma, autoimmune diseases, atherosclerosis, diabetes, and cancer, are caused by inflammation. Inflammation can be detected by measuring the levels of certain biomarkers known as inflammatory markers. Immune cells, such as neutrophils, basophils, eosinophils, platelets, macrophages, and others, cell surface receptors and adhesion molecules such as selectins (L-selectin, P-selectin, and E-selectin), and soluble mediators, such as cytokines (IL-1, IL-2, IL-6, TNF-α, TGF-ß, and IFN-γ), chemokines, and NF-kB (complement factors, C-reactive protein and the coagulation factor fibrinogen) [8,9,10,15,18].
Diosmin has been shown in multiple studies to lower these markers due to its anti-inflammatory effects. In lung damage produced by lipopolysaccharide (LPS) therapy, pro-inflammatory cytokines (IL-2, IL-6, IL-17, and TNF-α) and NF-kB were shown to be increased. After a few days of Diosmin pre-treatment, the levels of these indicators were dramatically lowered. In this study, male adult Balb/c mice were employed (Imam et al., 2015). In rat colitis caused by trinitrobenzenesulfonic acid, Diosmin was found to decrease the production of LTB4 (eicosanoid) and colonic MDA (TNBS). In the TNBS-treated colon, LTB4 improves neutrophil chemotaxis, adhesion, and degranulation, whereas MDA is a lipid peroxidation product (Crespo et al., 1999). According to Tahir et al., Diosmin reduces COX-2 and iNOS levels in chemically induced hepatocarcinogenesis (inflammatory markers). In female Wistar rats, diethylnitrosamine (DEN) was utilized to induce hepatocarcinogenesis, while 2-acetylaminofluorene was employed to promote it (2-AAF). In a long-term experiment, Diosmin was given orally at doses of 10 and 20 mg/kg b.wt for 9 weeks (Tahir et al., 2013). Diosmin treatment was found to alter the levels of tumor necrosis factor (TNF-α) and cyclooxygenase-2 in acetic acid-induced ulcerative colitis (COX-II). TNF-α and COX-II levels were lowered in a dose-dependent manner in Diosmin-treated rats [10].

3.3. Anti-Cancer Property

Recent study has discovered that Diosmin has dose-dependent pro-apoptotic effects on a range of animal cancers, including breast, prostate, colon, oral, and urinary bladder tumors. Diosmin causes premature senescence and apoptosis in MCF-7 cells at various doses. Other breast cancer cell lines, including MDA-MB-231 and SKBR-3, responded to Diosmin, but MCF-7 was found to be the most responsive. At lower doses, Diosmin produced G2/M cell cycle arrest, elevated p53, p21, and p27 levels, increased SA—gal activity, oxidative stress, and DNA damage in MCF-7 cells, all of which are associated with ageing. Apoptosis can be triggered by increased levels of nitric oxide, total ROS, total superoxide, mitochondrial production, and protein carbonylation. In research on the androgen-independent prostate cancer cell line DU145, Diosmin’s pro-apoptotic activity was confirmed. The genome and cytotoxicity of three flavonoid glycosides (Diosmin, naringin, and hesperidin) were studied in the DU145 prostate cancer cell line. The most genotoxicity was caused by Diosmin. In DU145 cells, these flavonoids caused oxidative stress or intracellular redox disequilibrium, leading to changes in mitochondrial membrane potential and apoptotic cell death. Diosmin dramatically boosted overall ROS production. Diosmin therapy also increased the amount of double stranded breaks in DNA and the formation of micronuclei (genotoxicity) [16]. Diosmin has anti-proliferative effect in human colon cancer cell lines, according to Kuntz et al. [29]. Tanaka et al. discovered a chemopreventive effect of Diosmin on colon carcinogenesis produced by azoxymethane in male F344 rats [19,47]. The treatment of Diosmin orally reduced colon carcinogenesis, as evidenced by lower rates of colon cancers. They hypothesized that inhibiting ornithine decarboxylase (ODC), a rate-limiting enzyme in polyamine production, was responsible for the decrease in colonic cancers [47]. Cell apoptosis is produced by DNA damage when ODC is inhibited [48]. When exposed to carcinogens, ODC levels have been reported to rise in numerous tissues. ODC activity was likewise elevated in the colonic mucosa of azoxymethane-treated rats. Diosmin has a dose-dependent cytotoxic capability for A431 skin cancer cells, according to Buddhan et al. It inhibits the invasive capacity of A431 cells by inducing apoptosis through a ROS-mediated mechanism [21,27]. DNA fragmentation, overexpression of p53, caspase 3 and caspase 9 genes, and downregulation of Bcl-2, matrix metalloproteinases-2 and 9 genes were all observed in A431 cells after Diosmin treatment. Diosmin’s IC50 value was discovered to be 45 g/mL, at which point it produced significant ROS [49]. Diosmin was found to prevent oral carcinogenesis produced by 4-nitroquinoline 1-oxide in male F344 rats (4-NQO). They theorized that Diosmin had anti-cancer capabilities via a number of mechanisms. One of these mechanisms was the inhibition of ornithine decarboxylase (ODC). Agents that inhibit ODC activity have been shown to be effective in slowing tumor growth. Another hypothesized mechanism is the inhibition of DT-diaphorase activity, which is required for 4-NQO to have its carcinogenic effect [19,47]. Inhibiting oral carcinogenesis, Diosmin is more efficient than diosmentin (the aglycone version of Diosmin) [50]. Oral treatment of 1000 ppm Diosmin suppressed urinary-bladder carcinogenesis induced by N-butyl-N-(4-hydroxybutyl) nitrosamine in male ICR mice. These findings were supported by a count of silver-stained nucleolar-organizer-region-associated proteins (AgNORs) and a 5-bromodeoxyuridine (BUdR)-labeling index. Diosmin’s anti-carcinogenic effects may be due to a decrease in cell proliferation [13,51]. In male Wistar rats, Diosmin had a similar effect on esophageal carcinogenesis induced with N-methyl-N-amylnitrosamine (MNAN). Diosmin’s chemopreventive effects on several cancer kinds or cell lines are summarized in Figure 2.
Diosmin has also been shown to have anti-metastatic properties in metastatic lung melanoma (B16F10) [52]. Diosmin lowers the number of metastatic nodules, implant percentage, and invasion index in both micro- and macroscopic investigations. In the treatment of metastatic pulmonary melanoma, Diosmin acts in tandem with IFN-γ [53,54]. By disrupting the PI3K–Akt–MDM2 signaling pathway, Diosmin decreased HA22T cell growth (human hepatocellular cancer) in nude mice models and triggered G2/M cell cycle arrest through p53 activation [24,55].

3.4. Anti-Diabetic Property

In the treatment of diabetes and its complications, Diosmin has been proven to have therapeutic effects. Diabetes mellitus is a chronic disease characterized by abnormal glucose, protein, and lipid metabolism due to a lack of or decreased insulin activity. Diosmin possesses anti-hyperglycemic effects, according to numerous studies. According to Pari et al., Diosmin (in various doses) taken orally for 45 days can improve glycemic control. In the study, male albino Wistar strain rats were given streptozotocin-nicotinamide to develop diabetes (STZ-NA) [22,41]. The effect of Diosmin on plasma glucose levels was shown to be dose-dependent. In addition, oral Diosmin (100 mg/kg b.w.) treatment decreased glycosylated hemoglobin while raising hemoglobin and plasma insulin. Hexokinase and glucose-6-phosphate dehydrogenase, two essential liver enzymes, were also suppressed. In addition, there was a rise in body weight [22,41].
Diosmin has also been linked to improved lipid metabolism in people with diabetes. Hypercholesterolemia, lipid buildup in hepatic organs, and alterations in plasma lipid and lipoprotein profiles are all symptoms of metabolic dyslipidemia in people with type-2 diabetes [56,57]. Plasma lipids, tissue lipids (cholesterol, TGs, FFAs, and PLs), and plasma lipoproteins can all be efficiently normalized with Diosmin therapy (LDL, VLDL) [22,41]. Diosmin also corrects changes in glycoprotein profile caused by type-2 diabetes. When glucose is utilized by insulin-independent processes in diabetic people, glycoproteins build up. In STZ-NA-induced diabetic rats, the level of plasma glycoproteins increased dramatically. In diabetic rats’ liver and kidneys, hexose, hexosamine, and fucose levels were much greater, but sialic acid levels were significantly lower. Diosmin, when taken orally, has been shown to counteract these changes in glycoprotein profile [22,41].
Neuropathy is one of the most common diabetic consequences, affecting more than half of those who have the illness. Chronic untreated and uncontrolled hyperglycemia causes pain, tingling, and numbness in the periphery, as well as delayed nerve conduction. Hyperglycemia-induced oxidative stress results in the accumulation of polyols and advanced glycation end products, as well as impairment of (Na+/K+)-ATPase activity and endothelial function. Apoptosis is the death of neurons as a result of oxidative stress. Jain et al. employed streptozotocin and a high-fat diet to induce type-2 diabetes in male Sprague-Dawley rats to assess the effect of Diosmin on diabetic neuropathy. In rats, four weeks of treatment with Diosmin (50 and 100 mg/kg, p.o.) reduced the development of early diabetic neuropathy. It reduced oxidative stress by restoring GSH, NO, and SOD activity that had been changed [27,31,42,58]. Diosmin treatment resulted in considerable NF-kB normalization in alloxan-induced diabetic Wistar mice. NF-kB is important in the pathogenesis of diabetic neuropathy and other inflammatory illnesses [8]. In male Swiss mice, Diosmin was discovered to alleviate neuropathic pain produced by chronic constriction injury (CCI). Diosmin (1 or 10 mg/kg) was given intraperitoneally to reduce CCI-induced mechanical and thermal hyperalgesia. The role of the NO/cGMP/PKG/KATP channel signaling pathway was verified using inhibitors, such as L-NAME (an inhibitor of NOS), ODQ (an inhibitor of soluble guanylate cyclase), KT5823 (an inhibitor of PKG), or glibenclamide (an ATP-sensitive K+ channel blocker). Diosmin treatment also inhibited the production of cytokines (IL-1 and IL-33/St2) and reduced the activation of glial cells [5,31,59].

3.5. Anti-Bacterial Property

In bacteria, the majority of medications have developed resistance. Plants are being researched in the hope of discovering new and effective antibacterial agents. Plant-based bioactive components that are effective against fungus, yeasts, and bacteria, as well as insects, nematodes, and other plants, are known as phytochemicals. They can inhibit the synthesis of peptidoglycans, damage microbial membrane structures, alter the hydrophobicity of bacterial membrane surfaces, and interfere with quorum sensing (QS). By synthesizing silver nanoparticles of Diosmin, it has been shown the antibacterial properties (AgNPs) where they have tested its antimicrobial activity against Escherichia coli, Pseudomonas putida, and Staphylococcus aureus using the disc diffusion method. Diosmin produced hexagonal AgNPs that were slightly antimicrobial and had a size of roughly 5–40 nm. Diosmin inhibited E. coli, P. putida, and S. aureus with zones of inhibition of 6, 6, and 7 mm, respectively. Pits on the bacterial cell wall, changes in cell membrane permeability, obstruction of transduction, suppression of respiratory enzyme function due to free radical generation, and inactivation of various thiol-containing enzymes have all been proposed as antibacterial mechanisms [13,17,51].
Diosmin with amoxicillin-clavulanic acid (AMC) has been found to have mycobactericidal efficacy against Mycobacterium marinum. After treatment with a combination of AMC and Diosmin, the survival of M. marinum-infected Drosophila melanogaster fly models improved by 60%, providing in vitro proof. Its antibacterial activity was also proven against Mtb H37Ra and an MDR clinical isolate. The AMC-Diosmin combination was discovered to target L, D-transpeptidase (LDT) enzymes involved in Mtb cell wall synthesis, resulting in cellular leakage in M. marinum cells [14].

3.6. Cardiovascular Protection

Diets high in flavonoids help promote cardiovascular health. Diosmin has been shown to have anti-platelet action. Sulfation of Diosmin will very certainly improve overall binding at the heparin binding site, which contains helix A, D, and N-terminal residues, resulting in the development of novel anti-thrombin candidates. Diosmin’s anti-thrombotic properties are confirmed by changes in the protein composition of rats with venous thrombosis. Diosmin, which targets centrosome-associated protein 350, may increase endothelial cell proliferation (CEP350). In a rat model produced by the nitric oxide production inhibitor L-NAME, Diosmin’s antihypertensive effects are demonstrated [4,7,11,60,61,62,63,64]. Diosmin appears to protect rats from myocardial infarctions, hyaline arteriopathy, and fibrinoid necrosis caused by L-NAME. The elimination of superoxide anions by Diosmin could be the underlying mechanism for its anti-hypertensive actions. Diosmin has been shown to lower serum cardiac marker enzyme production, reduce plasma lipid peroxidation, and restrict lipid metabolism abnormalities in isoproterenol-induced myocardial-infarcted rats, resulting in anti-hyperlipidemia and cardio-protection. Isoproterenol has been reported to raise cardiac diagnostic markers, heart mitochondrial lipid peroxidation, and calcium ions while lowering anti-oxidant enzyme expression. Diosmin has been shown to be useful in preventing these traits. Left ventricular hypertrophy (LVH), ATPase dysfunction, and electrolyte imbalance all play a role in the etiology of isoproterenol-induced myocardial infarction. Pretreatment with Diosmin may attenuate the degenerative effects of isoproterenol in rats [4,5,7,11,31,65].
Reperfusion of ischemic tissues typically causes the generation of free radicals. Heart function recovery, anti-oxidant enzyme expression, and lipid peroxidation are all protected by Diosmin. In the presence of reperfusion stress, Diosmin successfully preserves TCA cycle enzyme activity. Diosmin reduces metabolic syndrome-related cardiovascular issues in rats, as demonstrated by improvements in systolic and diastolic blood pressure (BP) and ECG parameters. The anti-oxidant and anti-inflammatory effects of Diosmin in rats could explain these findings. Diosmetin protects mice from damage caused by ISO by upregulating AKT and NRF2 signaling while suppressing the NF-kB pathway. In venous smooth muscle action, Ca2+ is a key mediator. The contraction of the inferior vena cava (IVC) in normal Krebs and Ca2+-free Krebs has been shown to be unaffected by Diosmin. Diosmin, on the other hand, enhances the contractile response generated by KCl [23,24].
Sclerotherapy is a telangiectasia and varicose veins treatment that might cause irreversible endothelial damage. An exaggerated inflammatory response is usually induced during the sclerotherapy technique. MPFF has been shown to be useful in lowering inflammatory stress during sclerotherapy. By increasing venular diameter, preserving functional capillary density, reducing the number of leaky sites, and binding leukocytes, MPFF reduces post-sclerotherapy inflammation in a microvascular network. MPFF has been shown to lower the production of metalloproteinase-2 (MMP-2) and MMP-9 in rats while boosting MDA, which helps with varicose vein therapy. Linfadren, a combination of Diosmin, coumarin, and arbutin, has been shown in a randomized controlled trial to treat chronic hand edema in patients with post-trauma/surgery. Linfadren has been demonstrated to assist patients with breast cancer-related lymphedema when used in conjunction with extensive decongestive therapy [1,21,42,66].
Primary reflux from primary valve incompetence and venous thrombosis induces chronic venous illness, which includes pain, edema, skin damage, and ulceration. Two possible explanations are venous hypertension and microcirculation problems. Inflammation is both the beginning and the end of primary valve incompetence, as well as venous hypertension. Daflon 500 mg relieves clinical symptoms by reducing inflammatory reactions not only in the microcirculation but also in the vein wall and valve cusps. According to randomized trials, Daflon 500 mg for 60 days of therapy is also effective in elastic compression and speeding up the healing process in venous ulcers. A meta-analysis of Daflon 500 mg’s effects on venous leg ulcers found that it can be a valuable addition to standard therapy in big and long-standing ulcers. In randomized, double-blind, controlled trials, the therapeutic efficacy of Daflon 500 mg on chronic venous disease symptoms and edema was also studied. Diosmin may significantly reduce angiogenesis and inflammation, as evidenced by downregulation of TNF-α, IL-6, VEGF-C, VEGF-A, and FGF2 expression and overexpression of angiostatin expression in individuals with chronic venous issues. After six months of Daflon treatment, however, air plethysmography revealed no changes in venous hemodynamics. By magnifying adrenergic impact on microcirculation, Diosmin may cause adverse effects by raising creatine phosphokinase and serum lactic dehydrogenase levels [4,67,68,69].
Finally, Diosmin may protect against myocardial infarctions, hyaline arteriopathy, and fibrinoid necrosis caused by L-NAME by reducing serum cardiac marker enzyme production, plasma lipid peroxidation, and lipid metabolism alterations. By correcting isoproterenol-induced LVH, ATPase failure, and electrolyte imbalance, it improves metabolic syndrome-related cardiovascular issues. MPFF decreases MMP-2 and MMP-9 expression while increasing MDA, improving venular diameter, maintaining functional capillary density, reducing leaky sites, and sticking leukocytes. Inflammatory responses in the microcirculation, vein wall, and valve cusps are reduced with Daflon 500 mg. Daflon 500 mg has also been shown to be effective in the treatment of chronic venous disease symptoms and edema. TNF, IL-6, VEGF-C, VEGF-A, and FGF2 expression were all downregulated, whilst angiostatin expression was upregulated, showing that Diosmin reduces angiogenesis and inflammation in chronic venous disease patients. As evidenced by increases in SOD, CAT, GSH, and NO, as well as a decrease in MDA, Diosmin maintains redox equilibrium and downregulates the NF-B signaling pathway. Diosmin also lowers kidney weight, pH, total protein, calcium, and phosphorus in the urine, as well as potassium, sodium, magnesium, creatinine, and uric acid in the blood.

3.7. Liver Protection

Hypoxia, angiogenesis, inflammation, and intrapulmonary vasodilation characterize hepatopulmonary syndrome (HPS), a severe consequence of hepatic cirrhosis. TNF/VEGF, IGF-1/PI3K/AKT, and FGF-1/ANG-2 signaling pathways may be involved in HPS development and can be restored by Diosmin therapy in a chronic bile duct ligation (CBDL)-induced rat model. According to another study, Diosmin decreases BDL-induced liver abnormalities through modifying the Keap-1/NRF2 and p38-MAPK/NF-B/iNOS signaling pathways. Diosmin, pentoxifylline, and their combination have been shown to reduce BDL-induced liver cirrhosis via modulating the Keap-1/Nrf-2/GSH and NF-kB/p65/p38-MAPK signaling pathways. Diosmin also stimulates the production of cytoglobin, which contributes to the compound’s anti-oxidant, anti-inflammatory, and anti-fibrotic properties. Diosmin promotes the NRF2/Keap-1 pathway while suppressing the ROS-induced p38 MAPK pathways and activating the eNOS gene. By binding to the ARE sequence and upregulating the production of target genes, such as HO-1 and SOD, active NRF2 may enter the nucleus to promote transcriptional activity. The activity of the NF-kB, p53, and iNOS signaling pathways may be boosted when the p38 MAPK is triggered [6,7,13,27,41].

3.8. Neuroprotection

Diosmin performs a variety of actions that are linked to neuroprotection. Diosmin is a sedative that induces sleep and decreases the expression of IL-1, TNF, and IL-33/St2, as well as activating glial cells. It lowers mechanical and thermal hyperalgesia by activating D2, GABAA, and opioid receptors but not 5-HT1A receptors, reduces neuropathic pain by activating the NO/cGMP/PKG/KATP pathway, and suppresses IL-1, TNF-a, and other inflammatory mediators. Diosmin has been associated to thermal hyperalgesia, cold allodynia, and walking dysfunctions, as well as oxidative stress. It also helps with scopolamine-induced neural plasticity disruption and cognitive deficits by inhibiting TNF expression. Diosmin has an IC50 of 12.24 0.54 g mL and interacts with AChE enzyme via Tyr72, Tyr124, Trp286, Phe295, and Tyr341. Many flavone derivatives have been discovered as possible GABAA receptor ligands in the central nervous system, where they interact with the benzodiazepine binding site to produce depressive effects. The activity of the GABAA receptor cannot be controlled directly by flavonoid glycosides. Diosmin has sedative and sleep-inducing properties that are unrelated to GABAA receptor modulation. 6-C-glycoside-diosmetin activation of the GABAA receptor has been shown to have memory-enhancing and anxiolytic-like effects, which are controversial. As a result, more research into flavone derivatives’ interactions with the GABAA receptor is required. A range of harmful compounds in the peripheral and central nerve systems induces neuropathic pain. Diosmin coupled with hesperidin effectively reduces mechanical or thermal hyperalgesia in the chronic constriction injury (CCI) rat model. D2, GABAA, and opioid receptor antagonists may prevent these effects, while the 5-HT1A receptor inhibitor does not. In a CCI rat model, Diosmin alone has been shown to have anti-hyperalgesic effects via D2 and opioid receptors, as well as a reduction in pro-inflammatory cytokine (IL-1, TNF, and IL-6) expression [5,7].
Diosmin was found to reduce CCI-induced neuropathic pain by activating the NO/cGMP/PKG/KATP channel signaling pathway, lowering spinal cord cytokine (IL-1, TNF, and IL-33/St2), and activating glial cells in a separate investigation. Thermal hyperalgesia, cold allodynia, and walking dysfunctions, as well as oxidative stress, may be caused by STZ-induced diabetes in rats. The diabetes abnormalities in the brain system can be successfully corrected with Diosmin. Furthermore, diosmetin inhibits nociception in mice via antagonizing the transient receptor potential vanilloid 1 (TRPV1) receptor [12].
Misfolded proteins have been linked to neurodegenerative disorders. Diosmin was discovered to bind to hen egg white lysozyme (HEWL) in a sheet-shape, in an in vitro research, minimizing HEWL aggregation. These may play a role in treating amyloid-related illnesses. The synthesis and aggregation of amyloid-(A) peptide aids the degenerative course of Alzheimer’s disease (AD). Inhibiting GSK-3 using Diosmin and its aglycone derivative diosmetin has been demonstrated to reduce brain soluble A and A oligomer formation, as well as tau hyperphosphorylation, and improve cognitive impairment in rats. Diosmin may aid to ameliorate scopolamine-induced synaptic plasticity disruption and cognitive impairments by reducing the expression of the pro-inflammatory cytokine TNF in the rat hippocampus. Traumatic brain injury (TBI) can cause mental and cognitive disability. Diosmin inhibits the pro-inflammatory cytokine TNF-α, protecting against TBI-induced declines in neurological scores, memory, and long-term potentiation (Yizhou Zheng, 2020a).
Diosmin appears to protect PC12 cells from LPS-induced TNF production and apoptosis, as seen by decreased DNA fragmentation, decreased Bad and caspase-3 expression, and enhanced Bcl-2 expression. Diosmin interacts with critical residues Tyr72, Tyr124, Trp286, Phe295, and Tyr341 in the acetylcholinesterase (AChE) enzyme and has an IC50 value of 12.24 0.54 g mL−1. This has a similar binding orientation to Donepezil. Diosmin’s inhibitory impact on the AChE enzyme has been confirmed in silico and in vitro experiments. In an ELISA experiment using the Ellman method, Diosmin had no effect on the activity of AChE and butyrylcholinesterase (BChE).

3.9. Additional Roles

During ischemia–reperfusion injury, Diosmin protects against retinal edema by maintaining tight junction integrity and lowering capillary permeability [37,41,56]. With an IC50 value of 24 M, Diosmin suppresses AR activity. Diosmin reduces phosphorylation levels of JNK and p38-MAPK, reducing oxidative stress and apoptosis. Diosmin mediates fast current low-voltage-activated K+ channels, voltage-independent K+ channels, and the nitric oxide pathway, acting as a myorelaxant. Diosmin’s anti-oxidative, anti-inflammatory, and anti-apoptotic activities reduce MTX-induced histological changes and restore tissue architecture. By enhancing anti-oxidant enzyme expression and lowering inflammatory cytokine production, Diosmin protects against alcohol-induced abnormalities. Diosmin protects against changes in antioxidant expression, body and organ weight, and histopathology caused by cadmium.

4. Potential Diosmin Target Proteins and the Pathways

This article has gone over the proteins and pathways that have been identified as possible Diosmin targets. GSH and CAT are increased, while ROS, MDA, and lipid peroxidation are decreased. Diosmin is known to reduce the inflammatory proteins NF-kB, TNF-α, IL-1B, COX2, iNOS, and MAPKs. Diosmin controls the up (B-endorphin and glucose uptake) and down (gluconeogenesis and insulin resistance) of many diabetes-related pathways/pathway components [6,13,14,21]. Diosmin has been demonstrated to reduce proliferation, viability, and activate autophagy, all of which are known to play a role in various types of cancers. Platelet activity suppression, blood pressure, reperfusion stress, MMP-2/9, angiogenesis, and diagnostic markers are only a few of the cardiovascular parameters that Diosmin influences. Other pathways and their components linked to liver protection, neuroprotection, and antimicrobials include Keap-1, NRF2, p38-MAPK, iNOS, I/R-induced damage, EOF, RN4220, pUL5054, Ltd.Mt1, and Ltd.Mt2 [12,13,17,70,71]. Furthermore, we have mapped out the Diosmin target proteins and the respective pathways associated with them as shown in Figure 3. Furthermore, we have also presented a list of pathways which might be affected as a result of Diosmin intake (Table 1).
To prepare this pathways list, we have first used SwissTargetPrediction (http://www.swisstargetprediction.ch/predict.php) (accessed on 21 December 2021) to generate the proteins which could bind with Diosmin and then mapped out the KEGG pathways from the KEGG database (https://www.genome.jp/kegg/pathway.html) (accessed on 21 December 2021) for these Diosmin interactors (proteins). Table 1 contains the overall pathways and the respective proteins while similar information is presented in Figure 3 as a network of Diosmin, its interactors, and the pathways.

5. Combinational Therapy, Side Effects, and the Administrative Route

Diosmin is a venoactive drug administered orally for the treatment of chronic venous insufficiency (CVI). In CVI, veins have trouble sending blood from limbs back to the heart as a result of which blood gets pooled in the veins of legs. Reflux of the venous valves is the most common cause of CVI [80,81,82]. Edema and symptoms of chronic venous disease (CVD), particularly so-called venous pain, are treated with venoactive medicines (VAD). The usefulness of VAD is contested on a regular basis, despite the fact that it is well documented. Our goal was to gather all randomized controlled trials (RCTs) and meta-analyses devoted to VAD and symptoms in CVD, present them to a panel of international CVD experts, and have them vote by secret ballot on the level of efficacy of each drug, using EBM (Evidence-Based Medicine) rules and critical analysis [82,83,84]. Diosmin in combination with hesperidin (Daflon 500 mg) has been found more effective than Diosmin alone on venous symptoms [4,82]. The most common symptoms of CVI include leg ache, sensation of heaviness or tension, nocturnal cramps, sensation of swelling, restless legs, and itching. Burning, heaviness, weakness, and functional pain are reduced after two months of Daflon treatment. It also enhances the skin microcirculation’s blood velocity. Daflon is not considered a cure for CVI because it does not address the underlying cause of the condition; rather, it is used to alleviate the symptoms of the disease. Diosmin and Micronized Purified Flavonoid Fraction (MPFF) have been found to affect venous tone, lymphatic drainage, and microcirculation in CVI patients. They improve venous tone by blocking COMT (catechol-O-methyltransferase) from breaking down norepinephrine (noradrenaline) and so prolonging noradrenergic action. They enhance the number of functional lymphatics, lymphatic flow, capillary hematocrit, and red cell velocity while decreasing lymphatic channel width and intra-lymphatic pressure. They also prevent leukocyte adherence, intra-tissue movement, and the release of leukocyte (L-selectin) and endothelial (ICAM-1, VCAM-1) adhesion molecules, which protect microvascular permeability (Inflammatory mediators) [21,66].
Daflon treatment for four weeks (four tablets per day, in two divided doses) has also shown improvement in the symptoms associated with hemorrhoids (pain, heaviness, bleeding, pruritus and anal discharge) [85,86,87]. However, purified Diosmin has also been shown to reduce pain and bleeding. Daflon has also been reported to exhibit anti-oxidative property. Combination of Diosmin with amoxicillin-clavulanic acid (AMC) has been found to possess mycobactericidal activity against Mycobacterium Marinum [14,20,88].

6. Future Perspectives

Diosmin is an anti-oxidant, anti-cancer, anti-diabetic, and mild anti-bacterial flavone glycoside derived from citrus trees. Because of its characteristics, it is a good therapeutic treatment for a variety of disorders. It reduces oxidative stress by altering the activity of particular enzymes and promotes apoptosis in a variety of cancer cell lines via several ways. Its anti-inflammatory properties are due to its ability to lower the levels of numerous inflammation markers. It also helps alleviate the consequences of diabetes, such as neuropathy and dyslipidemia. It has been found to be quite useful in the treatment of chronic venous insufficiency and hemorrhoids when combined with other flavonoids, particularly hesperidin. Treatment with Diosmin appears to be promising in the treatment of many malignancies, diabetes, and disorders linked to oxidative stress and inflammation. Diosmin is an anti-hyperglycemic drug that also helps with the issues that come with it. Its ability to modulate the VEGF/PEDF ratio could be investigated further to determine if it is a pro- or anti-angiogenic factor. Its combination with hesperidin has been proven to be extremely helpful in the treatment of CVI and hemorrhoids, making it a good example of drug synergism. Its interaction with other flavonoids or phytochemicals could be investigated in the future using various methodologies, including system-level knowledge [77,89,90,91,92]. Furthermore, it could also be explored by generating, analyzing, exploring, and integrating large-scale datasets to understand the potentials of Diosmin in case of human diseases [72,73,74,76,77,78,79,90,93,94,95,96,97,98,99,100].

7. Conclusions

We have presented a review work that summarizes Diosmin’s pharmacology, significant biological features, such as anti-oxidant, anti-inflammatory, anti-cancer, anti-diabetic, antibacterial, anti-cardiovascular, liver protection, and neuroprotection, therapeutic strategy, possible Diosmin targets, and pathways connected with Diosmin (Figure 1) due to the diverse and potential biological and pharmacological properties and influences of Diosmin (Figure 1). Finally, we have summarized our review work where Diosmin actively plays critical roles in controlling well-known signaling components and the pathways. In this table, we clearly see that there are a number of proteins and these proteins are dominantly associated with inflammatory processes, cancer-associated pathways, antidiabetic, antioxidant, and antibacterial properties.

Author Contributions

Conceptualization, E.H. and M.M.; methodology, E.H. and M.M.; software, E.H. and M.M.; validation, E.H. and M.M.; formal analysis, E.H. and M.M.; investigation, E.H. and M.M.; resources, E.H. and M.M.; data curation, E.H. and M.M.; writing—original draft preparation, E.H. and M.M.; writing—review and editing, E.H. and M.M.; visualization, E.H. and M.M.; supervision, E.H. and M.M.; project administration, E.H. and M.M.; funding acquisition, E.H. and M.M.; funding acquisition, E.H. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We are thankful to K.A.U., K.I. and J.M.I. and for providing us the resources and the facility to carry out the work.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Bogucka-Kocka, A.; Woźniak, M.; Feldo, M.; Kockic, J.; Szewczyk, K. Diosmin—Isolation techniques, determination in plant material and pharmaceutical formulations, and clinical use. Nat. Prod. Commun. 2013, 8, 545–550. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Gerges, S.H.; Wahdan, S.A.; Elsherbiny, D.A.; El-Demerdash, E. Pharmacology of Diosmin, a Citrus Flavone Glycoside: An Updated Review. Eur. J. Drug Metab. Pharmacokinet. 2021, 47, 1–18. [Google Scholar] [CrossRef] [PubMed]
  3. Liu, W.Y.; Liou, S.-S.; Hong, T.-Y.; Liu, I.-M. The Benefits of the Citrus Flavonoid Diosmin on Human Retinal Pigment Epithelial Cells under High-Glucose Conditions. Molecules 2017, 22, 2251. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Feldo, M.; Wójciak-Kosior, M.; Sowa, I.; Kocki, J.; Bogucki, J.; Zubilewicz, T.; Kęsik, J.; Bogucka-Kocka, A. Effect of Diosmin Administration in Patients with Chronic Venous Disorders on Selected Factors Affecting Angiogenesis. Molecules 2019, 24, 3316. [Google Scholar] [CrossRef] [Green Version]
  5. Fattori, V.; Rasquel-Oliveira, F.S.; Artero, N.A.; Ferraz, C.R.; Borghi, S.M.; Casagrande, R.; Verri, W.A. Diosmin Treats Lipopolysaccharide-Induced Inflammatory Pain and Peritonitis by Blocking NF-κB Activation in Mice. J. Nat. Prod. 2020, 83, 1018–1026. [Google Scholar] [CrossRef]
  6. Imam, F.; Al-Harbi, N.O.; Al-Harbi, M.M.; Ansari, M.A.; Zoheir, K.M.A.; Iqbal, M.; Anwer, M.K.; Hoshani, A.A.R.; Attia, S.M.; Ahmad, S.F. Pharmacological Research. Pharmacol. Res. 2015, 102, 1–11. [Google Scholar] [CrossRef]
  7. Zhang, Z.; Liu, Q.; Yang, J.; Yao, H.; Fan, R.; Cao, C.; Liu, C.; Zhang, S.; Lei, X.; Xu, S. The proteomic profiling of multiple tissue damage in chickens for a selenium deficiency biomarker discovery. Food Funct. 2020, 11, 1312–1321. [Google Scholar] [CrossRef]
  8. Ahmed, S.; Mundhe, N.; Borgohain, M.; Chowdhury, L.; Kwatra, M.; Bolshette, N.; Ahmed, A.; Lahkar, M. Diosmin Modulates the NF-kB Signal Transduction Pathways and Downregulation of Various Oxidative Stress Markers in Alloxan-Induced Diabetic Nephropathy. Inflammation 2016, 39, 1783–1797. [Google Scholar] [CrossRef]
  9. Crespo, M.E.; Gálvez, J.; Cruz, T.; Ocete, M.A.; Zarzuelo, A. Anti-inflammatory activity of diosmin and hesperidin in rat colitis induced by TNBS. Planta Med. 1999, 65, 651–653. [Google Scholar] [CrossRef]
  10. Shalkami, A.S.; Hassan, M.; Bakr, A.G. Anti-inflammatory, antioxidant and anti-apoptotic activity of diosmin in acetic acid-induced ulcerative colitis. Hum. Exp. Toxicol. 2017, 37, 78–86. [Google Scholar] [CrossRef]
  11. Abdel-Reheim, M.A.; Messiha, B.A.S.; Abo-Saif, A.A. Hepatoprotective Effect of Diosmin on Iron-induced Liver Damage. Int. J. Pharmacol. 2017, 13, 529–540. [Google Scholar] [CrossRef]
  12. Carballo-Villalobos, A.I.; González-Trujano, M.-E.; Pellicer, F.; López-Muñoz, F.J. Antihyperalgesic Effect of Hesperidin Improves with Diosmin in Experimental Neuropathic Pain. BioMed Res. Int. 2016, 2016, 8263463. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Kilit, A.C.; Kose, E.O.; Imir, N.G.; Aydemir, E. Anticancer and antimicrobial activities of diosmin. Genet. Mol. Res. 2021, 20, GMR18752. [Google Scholar] [CrossRef]
  14. Pushkaran, A.C.; Vinod, V.; Vanuopadath, M.; Nair, S.S.; Nair, S.V.; Vasudevan, A.K.; Biswas, R.; Mohan, C.G. Combination of Repurposed Drug Diosmin with Amoxicillin-Clavulanic acid Causes Synergistic Inhibition of Mycobacterial Growth. Sci. Rep. 2019, 9, 6800. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Ali, N.; AlAsmari, A.F.; Imam, F.; Ahmed, M.Z.; Alqahtani, F.; Alharbi, M.; AlSwayyed, M.; AlAsmari, F.; Alasmari, M.; Alshammari, A.; et al. Protective effect of diosmin against doxorubicin-induced nephrotoxicity. Saudi J. Biol. Sci. 2021, 28, 4375–4383. [Google Scholar] [CrossRef]
  16. Lewinska, A.; Adamczyk-Grochala, J.; Kwasniewicz, E.; Deregowska, A.; Wnuk, M. Diosmin-induced senescence, apoptosis and autophagy in breast cancer cells of different p53 status and ERK activity. Toxicol. Lett. 2017, 265, 117–130. [Google Scholar] [CrossRef] [PubMed]
  17. Patel, K.; Gadewar, M.; Tahilyani, V.; Patel, D.K. A review on pharmacological and analytical aspects of diosmetin: A concise report. Chin. J. Integr. Med. 2013, 19, 792–800. [Google Scholar] [CrossRef]
  18. Yao, X.; Gu, X.; Jin, S.; Shi, K.; Gao, X.; Wang, Q.; Zhao, J.; Zhang, H.; Lai, X. Anticancer and Anti-inflammatory Effect of Diosmin against Dalton Ascitic Lymphoma Induced Leukemia. J. Oleo Sci. 2021, 70, 665–673. [Google Scholar] [CrossRef]
  19. Tanaka, T.; Makita, H.; Kawabata, K.; Mori, H.; Kakumoto, M.; Satoh, K.; Hara, A.; Sumida, T.; Fukutani, K.; Ogawa, H. Modulation of N-methyl-N-amylnitrosamine-induced rat oesophageal tumourigenesis by dietary feeding of diosmin and hesperidin, both alone and in combination. Carcinogenesis 1997, 18, 761–769. [Google Scholar] [CrossRef] [Green Version]
  20. Corsale, I.; Carrieri, P.; Martellucci, J.; Piccolomini, A.; Verre, L.; Rigutini, M.; Panicucci, S. Flavonoid mixture (diosmin, troxerutin, rutin, hesperidin, quercetin) in the treatment of I–III degree hemorroidal disease: A double-blind multicenter prospective comparative study. Int. J. Colorectal Dis. 2018, 33, 1595–1600. [Google Scholar] [CrossRef]
  21. Perumal, S. Effect of diosmin on apoptotic signaling molecules in N- nitrosodiethylamine-induced hepatocellular carcinoma in experimental rats. Mol. Cell. Biochem. 2018, 449, 27–37. [Google Scholar] [CrossRef] [PubMed]
  22. Srinivasan, S.; Pari, L. Ameliorative effect of diosmin, a citrus flavonoid against streptozotocin-nicotinamide generated oxidative stress induced diabetic rats. Chem.-Biol. Interact. 2012, 195, 43–51. [Google Scholar] [CrossRef] [PubMed]
  23. Soares, J.M.; Faria, B.M.D.; Ascari, L.M.; Souza, J.M.D.; Soares, A.G.; Cordeiro, Y.; Romão, L.F. Diosmin induces caspase-dependent apoptosis in human glioblastoma cells. An. Acad. Bras. Ciências 2019, 91, e20191031. [Google Scholar] [CrossRef] [PubMed]
  24. Dung, T.; Lin, C.-H.; Việt Bình, T.; Hsu, H.-H.; Su, C.-C.; Lin, Y.-M.; Tsai, C.-H.; Tsai, F.-J.; Kuo, W.-W.; Chen, L.-M.; et al. Diosmin induces cell apoptosis through protein phosphatase 2A activation in HA22T human hepatocellular carcinoma cells and blocks tumour growth in xenografted nude mice. Food Chem. 2012, 132, 2065–2073. [Google Scholar] [CrossRef]
  25. Dubey, K.; Dubey, R.; Gupta, R.; Gupta, A. Exploration of Diosmin to Control Diabetes and Its Complications-an In Vitro and In Silico Approach. Curr. Comput. Aided-Drug Des. 2021, 17, 307–313. [Google Scholar] [CrossRef]
  26. Li, T.; Zhu, W.; Liu, G.; Fang, C.; Quan, S. Diosmin for the prevention of ovarian hyperstimulation syndrome. Int. J. Gynecol. Obstet. 2020, 149, 166–170. [Google Scholar] [CrossRef]
  27. Eraslan, G.; Sarıca, Z.S.; Bayram, L.Ç.; Tekeli, M.Y.; Kanbur, M.; Karabacak, M. The effects of diosmin on aflatoxin-induced liver and kidney damage. Environ. Sci. Pollut. Res. Int. 2017, 24, 27931–27941. [Google Scholar] [CrossRef]
  28. Caltagirone, S.; Rossi, C.; Poggi, A.; Ranelletti, F.O.; Natali, P.G.; Brunetti, M.; Aiello, F.B.; Piantelli, M. Flavonoids apigenin and quercetin inhibit melanoma growth and metastatic potential. Int. J. Cancer 2000, 87, 595–600. [Google Scholar] [CrossRef]
  29. Kuntz, S.; Wenzel, U.; Daniel, H. Comparative analysis of the effects of flavonoids on proliferation, cytotoxicity, and apoptosis in human colon cancer cell lines. Eur. J. Nutr. 1999, 38, 133–142. [Google Scholar] [CrossRef]
  30. Kumar, S.; Pandey, A.K. Chemistry and Biological Activities of Flavonoids: An Overview. Sci. World J. 2013, 2013, 162750. [Google Scholar] [CrossRef] [Green Version]
  31. Bozdağ, M.; Eraslan, G. The effect of diosmin against lead exposure in rats. Naunyn Schmiedebergs Arch. Pharmacol. 2020, 393, 639–649. [Google Scholar] [CrossRef] [PubMed]
  32. Waring, M.J.; Arrowsmith, J.; Leach, A.R.; Leeson, P.D.; Mandrell, S.; Owen, R.M.; Pairaudeau, G.; Pennie, W.D.; Pickett, S.D.; Wang, J.; et al. An analysis of the attrition of drug candidates from four major pharmaceutical companies. Nat. Publ. Group 2015, 14, 475–486. [Google Scholar] [CrossRef] [PubMed]
  33. Chikara, S.; Nagaprashantha, L.D.; Singhal, J.; Horne, D.; Awasthi, S.; Singhal, S.S. Oxidative stress and dietary phytochemicals: Role in cancer chemoprevention and treatment. Cancer Lett. 2018, 413, 122–134. [Google Scholar] [CrossRef] [PubMed]
  34. Pfeffer, P.E.; Lu, H.; Mann, E.H.; Chen, Y.-H.; Ho, T.-R.; Cousins, D.J.; Corrigan, C.; Kelly, F.J.; Mudway, I.S.; Hawrylowicz, C.M. Effects of vitamin D on inflammatory and oxidative stress responses of human bronchial epithelial cells exposed to particulate matter. PLoS ONE 2018, 13, e0200040. [Google Scholar] [CrossRef] [Green Version]
  35. Zraika, S.; Hull, R.L.; Udayasankar, J.; Aston-Mourney, K.; Subramanian, S.L.; Kisilevsky, R.; Szarek, W.A.; Kahn, S.E. Oxidative stress is induced by islet amyloid formation and time-dependently mediates amyloid-induced beta cell apoptosis. Diabetologia 2009, 52, 626–635. [Google Scholar] [CrossRef] [Green Version]
  36. Khosravi, M.; Poursaleh, A.; Ghasempour, G.; Farhad, S.; Najafi, M. The effects of oxidative stress on the development of atherosclerosis. Biol. Chem. 2019, 400, 711–732. [Google Scholar] [CrossRef]
  37. Sepidarkish, M.; Farsi, F.; Akbari-Fakhrabadi, M.; Namazi, N.; Almasi-Hashiani, A.; Maleki Hagiagha, A.; Heshmati, J. The effect of vitamin D supplementation on oxidative stress parameters: A systematic review and meta-analysis of clinical trials. Pharmacol. Res. 2019, 139, 141–152. [Google Scholar] [CrossRef]
  38. Hallak, M.; Vazana, L.; Shpilberg, O.; Levy, I.; Mazar, J.; Nathan, I. A molecular mechanism for mimosine-induced apoptosis involving oxidative stress and mitochondrial activation. Apoptosis 2008, 13, 147–155. [Google Scholar] [CrossRef]
  39. De Las Heras, N.; Martín Giménez, V.M.; Ferder, L.; Manucha, W.; Lahera, V. Implications of Oxidative Stress and Potential Role of Mitochondrial Dysfunction in COVID-19: Therapeutic Effects of Vitamin D. Antioxidants 2020, 9, 897. [Google Scholar] [CrossRef]
  40. Poznyak, A.; Grechko, A.V.; Poggio, P.; Myasoedova, V.A.; Alfieri, V.; Orekhov, A.N. The Diabetes Mellitus-Atherosclerosis Connection: The Role of Lipid and Glucose Metabolism and Chronic Inflammation. Int. J. Mol. Sci. 2020, 21, 1835. [Google Scholar] [CrossRef] [Green Version]
  41. Pari, L.; Srinivasan, S. Antihyperglycemic effect of diosmin on hepatic key enzymes of carbohydrate metabolism in streptozotocin-nicotinamide-induced diabetic rats. Biomed. Pharmacother. 2010, 64, 477–481. [Google Scholar] [CrossRef] [PubMed]
  42. Adouani, I.; Qureshi, A.S.; Hang, T.-J. Preparation, evaluation and pharmacokinetics of diosmin herbosomein beagle dogs. Pak. J. Pharm. Sci. 2019, 33, 033–040. [Google Scholar]
  43. Senthamizhselvan, O.; Manivannan, J.; Silambarasan, T.; Raja, B. Diosmin pretreatment improves cardiac function and suppresses oxidative stress in rat heart after ischemia/reperfusion. Eur. J. Pharmacol. 2014, 736, 131–137. [Google Scholar] [CrossRef] [PubMed]
  44. Tong, N.; Zhang, Z.; Zhang, W.; Qiu, Y.; Gong, Y.; Yin, L.; Qiu, Q.; Wu, X. Diosmin Alleviates Retinal Edema by Protecting the Blood-Retinal Barrier and Reducing Retinal Vascular Permeability during Ischemia/Reperfusion Injury. PLoS ONE 2013, 8, e61794. [Google Scholar] [CrossRef] [Green Version]
  45. Calvert, J.W. Chapter 5—Ischemic Heart Disease and its Consequences. In Cellular and Molecular Pathobiology of Cardiovascular Disease; Willis, M.S., Homeister, J.W., Stone, J.R., Eds.; Cellular and Molecular Pathobiology of Cardiovascular Disease; Academic Press: San Diego, CA, USA, 2014; pp. 79–100. [Google Scholar]
  46. Queenthy, S.S.; John, B. Diosmin exhibits anti-hyperlipidemic effects in isoproterenol induced myocardial infarcted rats. Eur. J. Pharmacol. 2013, 718, 213–218. [Google Scholar] [CrossRef] [PubMed]
  47. Tanaka, T.; Makita, H.; Kawabata, K.; Mori, H.; Kakumoto, M.; Satoh, K.; Hara, A.; Sumida, T.; Tanaka, T.; Ogawa, H. Chemoprevention of azoxymethane-induced rat colon carcinogenesis by the naturally occurring flavonoids, diosmin and hesperidin. Carcinogenesis 1997, 18, 957–965. [Google Scholar] [CrossRef] [Green Version]
  48. Pendeville, H.; Carpino, N.; Marine, J.C.; Takahashi, Y.; Muller, M.; Martial, J.A.; Cleveland, J.L. The ornithine decarboxylase gene is essential for cell survival during early murine development. Mol. Cell. Biol. 2001, 21, 6549–6558. [Google Scholar] [CrossRef] [Green Version]
  49. Baskar, R.; Dai, J.; Wenlong, N.; Yeo, R.; Yeoh, K.-W. Biological response of cancer cells to radiation treatment. Front. Mol. Biosci. 2014, 1, 24. [Google Scholar] [CrossRef] [Green Version]
  50. Browning, A.M.; Walle, U.K.; Walle, T. Flavonoid glycosides inhibit oral cancer cell proliferation—role of cellular uptake and hydrolysis to the aglycones. J. Pharm. Pharmacol. 2005, 57, 1037–1041. [Google Scholar] [CrossRef]
  51. Sahu, N.; Soni, D.; Chandrashekhar, B.; Satpute, D.B.; Saravanadevi, S.; Sarangi, B.K.; Pandey, R.A. Synthesis of silver nanoparticles using flavonoids: Hesperidin, naringin and diosmin, and their antibacterial effects and cytotoxicity. Int. Nano Lett. 2016, 6, 173–181. [Google Scholar] [CrossRef] [Green Version]
  52. Martínez, C.; Vicente, V.; Yáñez, J.; Alcaraz, M.; Castells, M.T.; Canteras, M.; Benavente-García, O.; Castillo, J. The effect of the flavonoid diosmin, grape seed extract and red wine on the pulmonary metastatic B16F10 melanoma. Histol. Histopathol. 2005, 20, 1121–1129. [Google Scholar] [PubMed]
  53. Álvarez, N.; Vicente, V.; Martínez, C. Synergistic Effect of Diosmin and Interferon-α on Metastatic Pulmonary Melanoma. Cancer Biother. Radiopharm. 2009, 24, 347–352. [Google Scholar] [CrossRef] [PubMed]
  54. Martínez Conesa, C.; Vicente Ortega, V.; Yáñez Gascón, M.J.; Alcaraz Baños, M.; Canteras Jordana, M.; Benavente-García, O.; Castillo, J. Treatment of Metastatic Melanoma B16F10 by the Flavonoids Tangeretin, Rutin, and Diosmin. J. Agric. Food Chem. 2005, 53, 6791–6797. [Google Scholar] [CrossRef]
  55. Dung, T.D.; Day, C.H.; Binh, T.V.; Lin, C.-H.; Hsu, H.-H.; Su, C.-C.; Lin, Y.-M.; Tsai, F.-J.; Kuo, W.-W.; Chen, L.-M.; et al. PP2A mediates diosmin p53 activation to block HA22T cell proliferation and tumor growth in xenografted nude mice through PI3K–Akt–MDM2 signaling suppression. Food Chem. Toxicol. 2012, 50, 1802–1810. [Google Scholar] [CrossRef]
  56. Farmer, J.A. Diabetic dyslipidemia and atherosclerosis: Evidence from clinical trials. Curr. Diabetes Rep. 2008, 8, 71–77. [Google Scholar] [CrossRef] [PubMed]
  57. Shepherd, J. Does statin monotherapy address the multiple lipid abnormalities in type 2 diabetes? Atheroscler. Suppl. 2005, 6, 15–19. [Google Scholar] [CrossRef]
  58. Jain, D.; Bansal, M.K.; Dalvi, R.; Upganlawar, A.; Somani, R. Protective effect of diosmin against diabetic neuropathy in experimental rats. J. Integr. Med. 2014, 12, 35–41. [Google Scholar] [CrossRef]
  59. Zielinska, D.F.; Gnad, F.; Schropp, K.; Wiśniewski, J.R.; Mann, M. Short Article. MOLCEL 2012, 46, 542–548. [Google Scholar]
  60. Luepker, R.V. Cardiovascular disease: Rise, fall, and future prospects. Annu. Rev. Public Health 2011, 32, 1–3. [Google Scholar] [CrossRef] [Green Version]
  61. Nabel, E.G. Cardiovascular disease. N. Engl. J. Med. 2003, 349, 60–72. [Google Scholar] [CrossRef]
  62. Iafisco, M.; Alogna, A.; Miragoli, M.; Catalucci, D. Cardiovascular nanomedicine: The route ahead. Nanomedicine 2019, 14, 2391–2394. [Google Scholar] [CrossRef] [PubMed]
  63. Martín Giménez, V.M.; Kassuha, D.E.; Manucha, W. Nanomedicine applied to cardiovascular diseases: Latest developments. Ther. Adv. Cardiovasc. Dis. 2017, 11, 133–142. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Mashour, N.H.; Lin, G.I.; Frishman, W.H. Herbal medicine for the treatment of cardiovascular disease: Clinical considerations. Arch. Intern. Med. 1998, 158, 2225–2234. [Google Scholar] [CrossRef] [PubMed]
  65. Stansfield, W.E.; Ranek, M.; Pendse, A.; Schisler, J.C.; Wang, S.; Pulinilkunnil, T.; Willis, M.S. Chapter 4—The Pathophysiology of Cardiac Hypertrophy and Heart Failure. In Cellular and Molecular Pathobiology of Cardiovascular Disease; Willis, M.S., Homeister, J.W., Stone, J.R., Eds.; Cellular and Molecular Pathobiology of Cardiovascular Disease; Academic Press: San Diego, CA, USA, 2014; pp. 51–78. [Google Scholar]
  66. Katsenis, K. Micronized purified flavonoid fraction (MPFF): A review of its pharmacological effects, therapeutic efficacy and benefits in the management of chronic venous insufficiency. Curr. Vasc. Pharmacol. 2005, 3, 1–9. [Google Scholar] [CrossRef]
  67. Manning, B.D.; Cantley, L.C. AKT/PKB Signaling: Navigating Downstream. Cell 2007, 129, 1261–1274. [Google Scholar] [CrossRef] [Green Version]
  68. Munjal, A.; Khandia, R. Atherosclerosis: Orchestrating Cells and Biomolecules Involved in Its Activation and Inhibition; Elsevier Ltd.: Amsterdam, The Netherlands, 2019; pp. 1–38. [Google Scholar]
  69. Renehan, A.G.; Zwahlen, M.; Egger, M. Adiposity and cancer risk:new mechanistic insightsfrom epidemiology. Nat. Rev. Cancer 2015, 15, 484–498. [Google Scholar] [CrossRef]
  70. Lee, J.; Song, K.-M.; Jung, C.H. Diosmin restores the skin barrier by targeting the aryl hydrocarbon receptor in atopic dermatitis. Phytomedicine 2021, 81, 153418. [Google Scholar] [CrossRef]
  71. Singh, S.; Sharma, B.; Kanwar, S.S.; Kumar, A. Lead Phytochemicals for Anticancer Drug Development. Front. Plant Sci. 2016, 7, 1667. [Google Scholar] [CrossRef] [Green Version]
  72. Warsi, M.K.; Kamal, M.A.; Baeshen, M.N.; Izhari, M.A.; Mobashir, A.F.A.M. Comparative Study of Gene Expression Profiling Unravels Functions associated with Pathogenesis of Dengue Infection. Curr. Pharm. Des. 2020, 26, 5293–5299. [Google Scholar] [CrossRef]
  73. Kamal, M.A.; Warsi, M.K.; Alnajeebi, A.; Ali, H.A.; Helmi, N.; Izhari, M.A.; Mustafa, S.; Mobashir, M. Gene expression profiling and clinical relevance unravel the role hypoxia and immune signaling genes and pathways in breast cancer: Role of hypoxia and immune signaling genes in breast cancer. J. Intern. Med. Sci. Art 2020, 1, 2–10. [Google Scholar] [CrossRef]
  74. Bajrai, L.; Sohrab, S.S.; Alandijany, T.A.; Mobashir, M.; Parveen, S.; Kamal, M.A.; Azhar, E.I. Gene expression profiling of early acute febrile stage of dengue infection and its comparative analysis with Streptococcus pneumoniae infection. Front. Cell. Infect. Microbiol. 2021, 11, 1018. [Google Scholar] [CrossRef] [PubMed]
  75. Alexeyenko, A.; Sonnhammer, E.L.L. Global networks of functional coupling in eukaryotes from comprehensive data integration. Genome Res. 2009, 19, 1107–1116. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Mobashir, M.; Schraven, B.; Beyer, T. Simulated evolution of signal transduction networks. PLoS ONE 2012, 7, e50905. [Google Scholar] [CrossRef] [PubMed]
  77. Mobashir, M.; Madhusudhan, T.; Isermann, B.; Beyer, T.; Schraven, B. Negative Interactions and Feedback Regulations Are Required for Transient Cellular Response. Sci. Rep. 2014, 4, 3718. [Google Scholar] [CrossRef] [Green Version]
  78. Eldakhakhny, B.M.; Sadoun, A.H.; Choudhry, H.; Mobashir, M. In-Silico Study of Immune System Associated Genes in Case of Type-2 Diabetes with Insulin Action and Resistance, and/or Obesity. Front. Endocrinol. 2021, 12, 281. [Google Scholar] [CrossRef]
  79. Krishnamoorthy, P.K.P.; Kamal, M.A.; Warsi, M.K.; Alnajeebi, A.; Ali, H.A.; Helmi, N.; Izhari, M.A.; Mustafa, S.; Firoz, A.; Mobashir, M. In-silico study reveals immunological signaling pathways, their genes, and potential herbal drug targets in ovarian cancer. Inform. Med. Unlocked 2020, 20, 100422. [Google Scholar] [CrossRef]
  80. Christopoulos, D.; Nicolaides, A.N.; Szendro, G. Venous reflux: Quantification and correlation with the clinical severity of chronic venous disease. Br. J. Surg. 1988, 75, 352–356. [Google Scholar] [CrossRef]
  81. De Smet, F.; Christopoulos, A.; Carmeliet, P. Allosteric targeting of receptor tyrosine kinases. Nat. Biotechnol. 2014, 32, 1113–1120. [Google Scholar] [CrossRef]
  82. Ramelet, A.A.; Boisseau, M.R.; Allegra, C.; Nicolaides, A.; Jaeger, K.; Carpentier, P.; Cappelli, R.; Forconi, S. Veno-active drugs in the management of chronic venous disease. An international consensus statement: Current medical position, prospective views and final resolution. Clin. Hemorheol. Microcirc. 2005, 33, 309–319. [Google Scholar]
  83. Cazaubon, M.; Benigni, J.-P.; Steinbruch, M.; Jabbour, V.; Gouhier-Kodas, C. Is There a Difference in the Clinical Efficacy of Diosmin and Micronized Purified Flavonoid Fractio.on for the Treatment of Chronic Venous Disorders? Review of Available Evidence. Vasc. Health Risk Manag. 2021, 17, 591–600. [Google Scholar] [CrossRef]
  84. Serra, R.; Ielapi, N.; Bitonti, A.; Candido, S.; Fregola, S.; Gallo, A.; Loria, A.; Muraca, L.; Raimondo, L.; Velcean, L.; et al. Efficacy of a Low-Dose Diosmin Therapy on Improving Symptoms and Quality of Life in Patients with Chronic Venous Disease: Randomized, Double-Blind, Placebo-Controlled Trial. Nutrients 2021, 13, 999. [Google Scholar] [CrossRef]
  85. Meshikhes, A.W.N. Daflon for haemorrhoids: A prospective, multi-centre observational study. Surgeon 2004, 2, 335–338+361. [Google Scholar] [CrossRef]
  86. Cypriani, B.; Limasset, B.; Carrié, M.L.; Le Doucen, C.; Roussie, M.; de Paulet, A.C.; Damon, M. Antioxidant activity of micronized diosmin on oxygen species from stimulated human neutrophils. Biochem. Pharmacol. 1993, 45, 1531–1535. [Google Scholar] [CrossRef]
  87. Oh-hora, M.; Komatsu, N.; Pishyareh, M.; Feske, S.; Hori, S.; Taniguchi, M.; Rao, A.; Takayanagi, H. Agonist-Selected T Cell Development Requires Strong T Cell Receptor Signaling and Store-Operated Calcium Entry. Immunity 2013, 38, 881–895. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Castle, A.R.; Gill, A.C. Physiological Functions of the Cellular Prion Protein. Front. Mol. Biosci. 2017, 4, 19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Borisov, N.; Aksamitiene, E.; Kiyatkin, A.; Legewie, S.; Berkhout, J.; Maiwald, T.; Kaimachnikov, N.P.; Timmer, J.; Hoek, J.B.; Kholodenko, B.N. Systems-level interactions between insulin–EGF networks amplify mitogenic signaling. Mol. Syst. Biol. 2009, 5, 256. [Google Scholar] [CrossRef] [PubMed]
  90. Kumar, J.; Umar, T.; Kausar, T.; Mobashir, M.; Nayeem, S.M.; Hoda, N. Identification of lead BAY60-7550 analogues as potential inhibitors that utilize the hydrophobic groove in PDE2A: A molecular dynamics simulation study. J. Mol. Modeling 2017, 23, 7. [Google Scholar] [CrossRef]
  91. Pinu, F.R.; Beale, D.J.; Paten, A.M.; Kouremenos, K.; Swarup, S.; Schirra, H.J.; Wishart, D. Systems Biology and Multi-Omics Integration: Viewpoints from the Metabolomics Research Community. Metabolites 2019, 9, 76. [Google Scholar] [CrossRef] [Green Version]
  92. Rekhi, R.; Qutub, A.A. Systems approaches for synthetic biology: A pathway toward mammalian design. Front. Physiol. 2013, 4, 285. [Google Scholar] [CrossRef] [Green Version]
  93. Mustafa, S.; Mobashir, M. LC–MS and docking profiling reveals potential difference between the pure and crude fucoidan metabolites. Int. J. Biol. Macromol. 2020, 143, 11–29. [Google Scholar] [CrossRef]
  94. Mobashir, M. Mathematical Modeling and Evolution of Signal Transduction Pathways and Networks. Ph.D. Thesis, Magdeburg University, Magdeburg, Germany, 2013. [Google Scholar]
  95. Singh, A.; Maqbool, M.; Mobashir, M.; Hoda, N. Dihydroorotate dehydrogenase: A drug target for the development of antimalarials. Eur. J. Med. Chem. 2017, 125, 640–651. [Google Scholar] [CrossRef]
  96. Maqbool, M.; Mobashir, M.; Hoda, N. Pivotal role of glycogen synthase kinase-3: A therapeutic target for Alzheimer’s. Eur. J. Med. Chem. 2015, 107, 63–81. [Google Scholar] [CrossRef] [PubMed]
  97. Helmi, N.; Alammari, D.; Mobashir, M. Role of Potential COVID-19 Immune System Associated Genes and the Potential Pathwayslinkage with Type-2 Diabetes. Comb. Chem. High. Throughput Screen. 2021, 24, 1–11. [Google Scholar] [CrossRef] [PubMed]
  98. Kumar, J.; Meena, P.; Singh, A.; Jameel, E.; Maqbool, M.; Mobashir, M.; Shandilya, A.; Tiwari, M.; Hoda, N.; Jayaram, B. Synthesis and screening of triazolopyrimidine scaffold as multi-functional agents for Alzheimer’s disease therapies. Eur. J. Med. Chem. 2016, 119, 260–277. [Google Scholar] [CrossRef] [PubMed]
  99. Krishnamoorthy, P.K.P.; Subasree, S.; Arthi, U.; Mobashir, M.; Gowda, C.; Revanasiddappa, P.D. T-cell Epitope-based Vaccine Design for Nipah Virus by Reverse Vaccinology Approach. Comb. Chem. High. Throughput Screen. 2020, 23, 788–796. [Google Scholar] [CrossRef]
  100. Bajrai, L.H.; Sohrab, S.S.; Mobashir, M.; Kamal, M.A.; Rizvi, M.A.; Azhar, E.I. Understanding the role of potential pathways and its components including hypoxia and immune system in case of oral cancer. Sci. Rep. 2021, 11, 19576. [Google Scholar] [CrossRef]
Figure 1. A summary of Diosmin, the associated diseases and potential properties in terms of disease protection.
Figure 1. A summary of Diosmin, the associated diseases and potential properties in terms of disease protection.
Biomedicines 10 01076 g001
Figure 2. Effect of Diosmin on different types of cancer cell lines and their mode of actions.
Figure 2. Effect of Diosmin on different types of cancer cell lines and their mode of actions.
Biomedicines 10 01076 g002
Figure 3. Diosmin-associated proteins and the biological pathways. We have used FunCoup database for PPI (protein–protein interactions) and integrated it with the KEGG pathways list, and the same has been mentioned in the main text also and the same has been done for Table 1 [72,73,74,75,76,77,78,79].
Figure 3. Diosmin-associated proteins and the biological pathways. We have used FunCoup database for PPI (protein–protein interactions) and integrated it with the KEGG pathways list, and the same has been mentioned in the main text also and the same has been done for Table 1 [72,73,74,75,76,77,78,79].
Biomedicines 10 01076 g003
Table 1. Diosmin and its potential interactors followed by their pathways.
Table 1. Diosmin and its potential interactors followed by their pathways.
DiosminNOS2AArginine_and_proline_metabolism
DiosminNOS2ACalcium pathway
DiosminNOS2ALong-term_depression
DiosminNOS2ASmall_cell_lung_cancer
DiosminPTGS2Arachidonic_acid_metabolism
DiosminPTGS2VEGF pathway
DiosminPTGS2Leishmaniasis
DiosminPTGS2Pathways_in_cancer
DiosminPTGS2Small_cell_lung_cancer
DiosminMMP2Leukocyte_transendothelial_migration
DiosminMMP2GnRH pathway
DiosminMMP2Pathways_in_cancer
DiosminMMP2Bladder_cancer
DiosminICAM1Cell_adhesion_molecules_(CAMs)
DiosminICAM1Natural_killer_cell_mediated_cytotoxicity
DiosminICAM1Leukocyte_transendothelial_migration
DiosminICAM1Viral_myocarditis
DiosminCOMTSteroid_hormone_biosynthesis
DiosminCOMTTyrosine_metabolism
DiosminJAK2Chemokine pathway
DiosminJAK2Jak-STAT pathway
DiosminJAK2Adipocytokine pathway
DiosminJAK2Leishmaniasis
DiosminMMP9Leukocyte_transendothelial_migration
DiosminMMP9Pathways_in_cancer
DiosminMMP9Bladder_cancer
DiosminNFKB1MAPK pathway
DiosminNFKB1Chemokine pathway
DiosminNFKB1Apoptosis
DiosminNFKB1Toll-like_receptor pathway
DiosminNFKB1NOD-like_receptor pathway
DiosminNFKB1RIG-I-like_receptor pathway
DiosminNFKB1Cytosolic_DNA-sensing_pathway
DiosminNFKB1T_cell_receptorpathway
DiosminNFKB1B_cell_receptorpathway
DiosminNFKB1Neurotrophin pathway
DiosminNFKB1Adipocytokine pathway
DiosminNFKB1Epithelial_cell_signaling_in_Helicobacter_pylori_infection
DiosminNFKB1Leishmaniasis
DiosminNFKB1Chagas_disease
DiosminNFKB1Pathways_in_cancer
DiosminNFKB1Pancreatic_cancer
DiosminNFKB1Prostate_cancer
DiosminNFKB1Chronic_myeloid_leukemia
DiosminNFKB1Acute_myeloid_leukemia
DiosminNFKB1Small_cell_lung_cancer
DiosminIL2Cytokine-cytokine_receptor_interaction
DiosminIL2T_cell_receptor pathway
DiosminIL2Type_I_diabetes_mellitus
DiosminCDKN1BErbB pathway
DiosminCDKN1BCell_cycle
DiosminCDKN1BPathways_in_cancer
DiosminCDKN1BProstate_cancer
DiosminCDKN1BChronic_myeloid_leukemia
DiosminCDKN1BSmall_cell_lung_cancer
DiosminPIK3CAInositol_phosphate_metabolism
DiosminPIK3CAErbB pathway
DiosminPIK3CAChemokine pathway
DiosminPIK3CAPhosphatidylinositol_signaling_system
DiosminPIK3CAmTOR pathway
DiosminPIK3CAApoptosis
DiosminPIK3CAFocal_adhesion
DiosminPIK3CAToll-like_receptor pathway
DiosminPIK3CAJak-STAT pathway
DiosminPIK3CANatural_killer_cell_mediated_cytotoxicity
DiosminPIK3CAT_cell_receptor pathway
DiosminPIK3CAB_cell_receptor pathway
DiosminPIK3CAFc_epsilon_RI pathway
DiosminPIK3CAFc_gamma_R-mediated_phagocytosis
DiosminPIK3CALeukocyte_transendothelial_migration
DiosminPIK3CANeurotrophin pathway
DiosminPIK3CARegulation_of_actin_cytoskeleton
DiosminPIK3CAInsulinpathway
DiosminPIK3CAProgesterone-mediated_oocyte_maturation
DiosminPIK3CAType_II_diabetes_mellitus
DiosminPIK3CAAldosterone-regulated_sodium_reabsorption
DiosminPIK3CABacterial_invasion_of_epithelial_cells
DiosminPIK3CAChagas_disease
DiosminPIK3CAPathways_in_cancer
DiosminPIK3CAColorectal_cancer
DiosminPIK3CARenal_cell_carcinoma
DiosminPIK3CAPancreatic_cancer
DiosminPIK3CAEndometrial_cancer
DiosminPIK3CAGlioma
DiosminPIK3CAProstate_cancer
DiosminPIK3CAMelanoma
DiosminPIK3CAChronic_myeloid_leukemia
DiosminPIK3CAAcute_myeloid_leukemia
DiosminPIK3CASmall_cell_lung_cancer
DiosminPIK3CANon-small_cell_lung_cancer
DiosminCDKN1AErbB pathway
DiosminCDKN1ACell_cycle
DiosminCDKN1APathways_in_cancer
DiosminCDKN1AGlioma
DiosminCDKN1AProstate_cancer
DiosminCDKN1AMelanoma
DiosminCDKN1ABladder_cancer
DiosminCDKN1AChronic_myeloid_leukemia
DiosminIL1BMAPK pathway
DiosminIL1BCytokine-cytokine_receptor_interaction
DiosminIL1BApoptosis
DiosminIL1BToll-like_receptor pathway
DiosminIL1BHematopoietic_cell_lineage
DiosminIL1BType_I_diabetes_mellitus
DiosminIL1BAlzheimer’s_disease
DiosminBECN1Regulation_of_autophagy
DiosminCASP9Apoptosis
DiosminCASP9VEGF pathway
DiosminCASP9Alzheimer’s_disease
DiosminCASP9Parkinson’s_disease
DiosminCASP9Amyotrophic_lateral_sclerosis_(ALS)
DiosminCASP9Huntington’s_disease
DiosminCASP9Pathways_in_cancer
DiosminCASP9Colorectal_cancer
DiosminCASP9Pancreatic_cancer
DiosminCASP9Endometrial_cancer
DiosminCASP9Prostate_cancer
DiosminCASP9Small_cell_lung_cancer
DiosminCASP9Non-small_cell_lung_cancer
DiosminCASP9Viral_myocarditis
DiosminIL6Cytokine-cytokine_receptor_interaction
DiosminIL6Toll-like_receptor pathway
DiosminIL6Hematopoietic_cell_lineage
DiosminIL6Pathways_in_cancer
DiosminTP53MAPK pathway
DiosminTP53Cell_cycle
DiosminTP53Apoptosis
DiosminTP53Wnt pathway
DiosminTP53Neurotrophin pathway
DiosminTP53Pathways_in_cancer
DiosminTP53Colorectal_cancer
DiosminTP53Pancreatic_cancer
DiosminTP53Endometrial_cancer
DiosminTP53Glioma
DiosminTP53Prostate_cancer
DiosminTP5305216_Thyroid_cancer
DiosminTP5305217_Basal_cell_carcinoma
DiosminTP53Melanoma
DiosminTP53Bladder_cancer
DiosminTP53Chronic_myeloid_leukemia
DiosminTP53Small_cell_lung_cancer
DiosminTP53Non-small_cell_lung_cancer
DiosminPIK3R1ErbB pathway
DiosminPIK3R1Chemokine pathway
DiosminPIK3R1Phosphatidylinositol_signaling_system
DiosminPIK3R1mTOR pathway
DiosminPIK3R1Apoptosis
DiosminPIK3R1Focal_adhesion
DiosminPIK3R1Toll-like_receptor pathway
DiosminPIK3R1Jak-STATpathway
DiosminPIK3R1Natural_killer_cell_mediated_cytotoxicity
DiosminPIK3R1T_cell_receptor pathway
DiosminPIK3R1B_cell_receptor pathway
DiosminPIK3R1Fc_epsilon_RI pathway
DiosminPIK3R1Fc_gamma_R-mediated_phagocytosis
DiosminPIK3R1Leukocyte_transendothelial_migration
DiosminPIK3R1Neurotrophinpathway
DiosminPIK3R1Regulation_of_actin_cytoskeleton
DiosminPIK3R1Insulinpathway
DiosminPIK3R1Progesterone-mediated_oocyte_maturation
DiosminPIK3R1Type_II_diabetes_mellitus
DiosminPIK3R1Aldosterone-regulated_sodium_reabsorption
DiosminPIK3R1Bacterial_invasion_of_epithelial_cells
DiosminPIK3R1Chagas_disease
DiosminPIK3R1Pathways_in_cancer
DiosminPIK3R1Colorectal_cancer
DiosminPIK3R1Renal_cell_carcinoma
DiosminPIK3R1Pancreatic_cancer
DiosminPIK3R1Endometrial_cancer
DiosminPIK3R1Glioma
DiosminPIK3R1Prostate_cancer
DiosminPIK3R1Melanoma
DiosminPIK3R1Chronic_myeloid_leukemia
DiosminPIK3R1Acute_myeloid_leukemia
DiosminPIK3R1Small_cell_lung_cancer
DiosminPIK3R1Non-small_cell_lung_cancer
DiosminCDKN2ACell_cycle
DiosminCDKN2APathways_in_cancer
DiosminCDKN2APancreatic_cancer
DiosminCDKN2AGlioma
DiosminCDKN2AMelanoma
DiosminCDKN2ABladder_cancer
DiosminCDKN2AChronic_myeloid_leukemia
DiosminCDKN2ANon-small_cell_lung_cancer
DiosminVCAM1Cell_adhesion_molecules_(CAMs)
DiosminVCAM1Leukocyte_transendothelial_migration
DiosminCASP3MAPK pathway
DiosminCASP3Apoptosis
DiosminCASP3Natural_killer_cell_mediated_cytotoxicity
DiosminCASP3Alzheimer’s_disease
DiosminCASP3Parkinson’s_disease
DiosminCASP3Amyotrophic_lateral_sclerosis_(ALS)
DiosminCASP3Huntington’s_disease
DiosminCASP3Epithelial_cell_signaling_in_Helicobacter_pylori_infection
DiosminCASP3Pathways_in_cancer
DiosminCASP3Colorectal_cancer
DiosminCASP3Viral_myocarditis
DiosminSTAT3Chemokine pathway
DiosminSTAT3Jak-STAT pathway
DiosminSTAT3Adipocytokine pathway
DiosminSTAT3Pathways_in_cancer
DiosminSTAT3Pancreatic_cancer
DiosminSTAT3Acute_myeloid_leukemia
DiosminBCL2Apoptosis
DiosminBCL2Focal_adhesion
DiosminBCL2Amyotrophic_lateral_sclerosis_(ALS)
DiosminBCL2Pathways_in_cancer
DiosminBCL2Colorectal_cancer
DiosminBCL2Prostate_cancer
DiosminBCL2Small_cell_lung_cancer
DiosminBCL2Protein_processing_in_endoplasmic_reticulum
DiosminNFKB1Ras_signaling
DiosminPIK3CARas_signaling
DiosminPIK3R1Ras_signaling
DiosminPIK3CARap1_signaling
DiosminPIK3R1Rap1_signaling
DiosminBECN1Apelin_signaling
DiosminICAM1NF-kappa_B_signaling
DiosminIL1BNF-kappa_B_signaling
DiosminNFKB1NF-kappa_B_signaling
DiosminPTGS2NF-kappa_B_signaling
DiosminBCL2NF-kappa_B_signaling
DiosminVCAM1NF-kappa_B_signaling
DiosminICAM1TNF_signaling
DiosminIL1BTNF_signaling
DiosminIL6TNF_signaling
DiosminMMP9TNF_signaling
DiosminNFKB1TNF_signaling
DiosminPIK3CATNF_signaling
DiosminPIK3R1TNF_signaling
DiosminPTGS2TNF_signaling
DiosminVCAM1TNF_signaling
DiosminCASP3TNF_signaling
DiosminCDKN1AHIF-1_signaling
DiosminCDKN1BHIF-1_signaling
DiosminIL6HIF-1_signaling
DiosminNFKB1HIF-1_signaling
DiosminPIK3CAHIF-1_signaling
DiosminPIK3R1HIF-1_signaling
DiosminBCL2HIF-1_signaling
DiosminSTAT3HIF-1_signaling
DiosminCDKN1AFoxO_signaling
DiosminCDKN1BFoxO_signaling
DiosminIL6FoxO_signaling
DiosminPIK3CAFoxO_signaling
DiosminPIK3R1FoxO_signaling
DiosminSTAT3FoxO_signaling
DiosminPIK3CAPhospholipase_D_signaling
DiosminPIK3R1Phospholipase_D_signaling
DiosminNFKB1Sphingolipid_signaling
DiosminPIK3CASphingolipid_signaling
DiosminPIK3R1Sphingolipid_signaling
DiosminBCL2Sphingolipid_signaling
DiosminTP53Sphingolipid_signaling
DiosminNFKB1cAMP_signaling
DiosminPIK3CAcAMP_signaling
DiosminPIK3R1cAMP_signaling
DiosminCDKN1API3K-Akt_signaling
DiosminCDKN1BPI3K-Akt_signaling
DiosminIL2PI3K-Akt_signaling
DiosminIL6PI3K-Akt_signaling
DiosminJAK2PI3K-Akt_signaling
DiosminNFKB1PI3K-Akt_signaling
DiosminBCL2PI3K-Akt_signaling
DiosminTP53PI3K-Akt_signaling
DiosminCASP9PI3K-Akt_signaling
DiosminPIK3CAAMPK_signaling
DiosminPIK3R1AMPK_signaling
DiosminJAK2pathways_regulating_pluripotency_of_stem_cells
DiosminPIK3CApathways_regulating_pluripotency_of_stem_cells
DiosminPIK3R1pathways_regulating_pluripotency_of_stem_cells
DiosminSTAT3pathways_regulating_pluripotency_of_stem_cells
DiosminPTGS2Retrograde_endocannabinoid_signaling
DiosminIL1BInflammatory_mediator_regulation_of_TRP_channels
DiosminPIK3CAInflammatory_mediator_regulation_of_TRP_channels
DiosminPIK3R1Inflammatory_mediator_regulation_of_TRP_channels
DiosminIL1BOsteoclast_differentiation
DiosminNFKB1Osteoclast_differentiation
DiosminPIK3CAOsteoclast_differentiation
DiosminPIK3R1Osteoclast_differentiation
DiosminNFKB1Longevity_regulating_pathway
DiosminPIK3CALongevity_regulating_pathway
DiosminPIK3R1Longevity_regulating_pathway
DiosminTP53Longevity_regulating_pathway
DiosminIL1BHematopoietic_cell_lineage
DiosminIL6Hematopoietic_cell_lineage
DiosminPIK3CA04611_Platelet_activation
DiosminPIK3R104611_Platelet_activation
DiosminPTGS2Ovarian_steroidogenesis
DiosminMMP2Estrogen pathway
DiosminMMP9Estrogen pathway
DiosminPIK3CAEstrogen pathway
DiosminPIK3R1Estrogen pathway
DiosminJAK2Prolactin pathway
DiosminNFKB1Prolactin pathway
DiosminPIK3CAProlactin pathway
DiosminPIK3R1Prolactin pathway
DiosminSTAT3Prolactin pathway
DiosminCDKN1AOxytocin pathway
DiosminPTGS2Oxytocin pathway
DiosminPIK3CAThyroid_hormone pathway
DiosminPIK3R1Thyroid_hormone pathway
DiosminTP53Thyroid_hormone pathway
DiosminCASP9Thyroid_hormone pathway
DiosminBCL2Adrenergic_signaling_in_cardiomyocytes
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Huwait, E.; Mobashir, M. Potential and Therapeutic Roles of Diosmin in Human Diseases. Biomedicines 2022, 10, 1076. https://doi.org/10.3390/biomedicines10051076

AMA Style

Huwait E, Mobashir M. Potential and Therapeutic Roles of Diosmin in Human Diseases. Biomedicines. 2022; 10(5):1076. https://doi.org/10.3390/biomedicines10051076

Chicago/Turabian Style

Huwait, Etimad, and Mohammad Mobashir. 2022. "Potential and Therapeutic Roles of Diosmin in Human Diseases" Biomedicines 10, no. 5: 1076. https://doi.org/10.3390/biomedicines10051076

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop