1
|
Rosales TKO, da Silva FFA, Bernardes ES, Paulo Fabi J. Plant-derived polyphenolic compounds: nanodelivery through polysaccharide-based systems to improve the biological properties. Crit Rev Food Sci Nutr 2024; 64:11894-11918. [PMID: 37585699 DOI: 10.1080/10408398.2023.2245038] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/18/2023]
Abstract
Plant-derived polyphenols are naturally occurring compounds widely distributed in plants. They have received greater attention in the food and pharmaceutical industries due to their potential health benefits, reducing the risk of some chronic diseases due to their antioxidant, anti-inflammatory, anticancer, cardioprotective, and neuro-action properties. Polyphenolic compounds orally administered can be used as adjuvants in several treatments but with restricted uses due to chemical instability. The review discusses the different structural compositions of polyphenols and their influence on chemical stability. Despite the potential and wide applications, there is a need to improve the delivery of polyphenolics to target the human intestine without massive chemical modifications. Oral administration of polyphenols is unfeasible due to instability, low bioaccessibility, and limited bioavailability. Nano-delivery systems based on polysaccharides (starch, pectin, chitosan, and cellulose) have been identified as a viable option for oral ingestion, potentiate biological effects, and direct-controlled delivery in specific tissues. The time and dose can be individualized for specific diseases, such as intestinal cancer. This review will address the mechanisms by which polysaccharides-based nanostructured systems can protect against degradation and enhance intestinal permeation, oral bioavailability, and the potential application of polysaccharides as nanocarriers for the controlled and targeted delivery of polyphenolic compounds.
Collapse
Affiliation(s)
- Thiécla Katiane Osvaldt Rosales
- Department of Food Science and Experimental Nutrition, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, SP, Brazil
- Instituto de Pesquisa Energéticas e Nucleares - IPEN, São Paulo, SP, Brazil
| | | | | | - João Paulo Fabi
- Department of Food Science and Experimental Nutrition, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, SP, Brazil
- Food Research Center (FoRC), CEPID-FAPESP (Research, Innovation and Dissemination Centers, São Paulo Research Foundation), São Paulo, SP, Brazil
- Food and Nutrition Research Center (NAPAN), University of São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
2
|
Meng J, Qiu C, Lu C, He X, Zhao X. A new crystalline daidzein-piperazine salt with enhanced solubility, permeability, and bioavailability. Front Pharmacol 2024; 15:1385637. [PMID: 39104399 PMCID: PMC11298695 DOI: 10.3389/fphar.2024.1385637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 06/26/2024] [Indexed: 08/07/2024] Open
Abstract
To overcome the poor solubility, permeability, and bioavailability of the plant isoflavone daidzein (DAI), a novel salt of DAI with anhydrous piperazine (PIP) was obtained based on cocrystallization strategy. The new salt DAI-PIP was characterized by powder X-ray diffraction (PXRD), differential scanning calorimetry (DSC), thermogravimetric analysis (TGA), Fourier-transform infrared (FT-IR) spectroscopy, and optical microscopy. The results showed that the maximum apparent solubility (Smax) of DAI-PIP increased by 7.27-fold and 1000-fold compared to DAI in pH 6.8 buffer and water, respectively. The peak apparent permeability coefficient (P app ) of DAI-PIP in the Caco-2 cell model was 30.57 ± 1.08 × 10-6 cm/s, which was 34.08% higher than that of DAI. Additionally, compared to DAI, the maximum plasma concentration (Cmax) value of DAI-PIP in beagle dogs was approximately 4.3 times higher, and the area under the concentration-time curve (AUC0-24) was approximately 2.4 times higher. This study provides a new strategy to enhance the dissolution performance and bioavailability of flavonoid drugs, laying a foundation for expanding their clinical applications.
Collapse
Affiliation(s)
| | | | | | - Xin He
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, China
| | - Xinghua Zhao
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, China
| |
Collapse
|
3
|
Liu Y, Luo J, Peng L, Zhang Q, Rong X, Luo Y, Li J. Flavonoids: Potential therapeutic agents for cardiovascular disease. Heliyon 2024; 10:e32563. [PMID: 38975137 PMCID: PMC11225753 DOI: 10.1016/j.heliyon.2024.e32563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 06/05/2024] [Accepted: 06/05/2024] [Indexed: 07/09/2024] Open
Abstract
Flavonoids are found in the roots, stems, leaves, and fruits of many plant taxa. They are related to plant growth and development, pigment formation, and protection against environmental stress. Flavonoids function as antioxidants and exert anti-inflammatory effects in the cardiovascular system by modulating classical inflammatory response pathways, such as the TLR4-NF-ĸB, PI3K-AKT, and Nrf2/HO-1 signalling pathways. There is increasing evidence for the therapeutic effects of flavonoids on hypertension, atherosclerosis, and other diseases. The potential clinical value of flavonoids for diseases of the cardiovascular system has been widely explored. For example, studies have evaluated the roles of flavonoids in the regulation of blood pressure via endothelium-dependent and non-endothelium-dependent pathways and in the regulation of myocardial systolic and diastolic functions by influencing calcium homeostasis and smooth muscle-related protein expression. Flavonoids also have hypoglycaemic, hypolipidemic, anti-platelet, autophagy, and antibacterial effects. In this paper, the role and mechanism of flavonoids in cardiovascular diseases were reviewed in order to provide reference for the clinical application of flavonoids in the future.
Collapse
Affiliation(s)
- Yingxue Liu
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jing Luo
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Lin Peng
- Department of Bone and Joint Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Qi Zhang
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Xi Rong
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yuhao Luo
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jiafu Li
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Southwest Medical University, China
| |
Collapse
|
4
|
Stevanoska M, Folz J, Beekmann K, Aichinger G. Physiologically based kinetic (PBK) modeling as a new approach methodology (NAM) for predicting systemic levels of gut microbial metabolites. Toxicol Lett 2024; 396:94-102. [PMID: 38685289 DOI: 10.1016/j.toxlet.2024.04.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 04/18/2024] [Accepted: 04/26/2024] [Indexed: 05/02/2024]
Abstract
There is a clear need to develop new approach methodologies (NAMs) that combine in vitro and in silico testing to reduce and replace animal use in chemical risk assessment. Physiologically based kinetic (PBK) models are gaining popularity as NAMs in toxico/pharmacokinetics, but their coverage of complex metabolic pathways occurring in the gut are incomplete. Chemical modification of xenobiotics by the gut microbiome plays a critical role in the host response, for example, by prolonging exposure to harmful metabolites, but there is not a comprehensive approach to quantify this impact on human health. There are examples of PBK models that have implemented gut microbial biotransformation of xenobiotics with the gut as a dedicated metabolic compartment. However, the integration of microbial metabolism and parameterization of PBK models is not standardized and has only been applied to a few chemical transformations. A challenge in this area is the measurement of microbial metabolic kinetics, for which different fermentation approaches are used. Without a standardized method to measure gut microbial metabolism ex vivo/in vitro, the kinetic constants obtained will lead to conflicting conclusions drawn from model predictions. Nevertheless, there are specific cases where PBK models accurately predict systemic concentrations of gut microbial metabolites, offering potential solutions to the challenges outlined above. This review focuses on models that integrate gut microbial bioconversions and use ex vivo/in vitro methods to quantify metabolic constants that accurately represent in vivo conditions.
Collapse
Affiliation(s)
- Maja Stevanoska
- Laboratory of Toxicology, Institute of Food, Nutrition and Health (IFNH), Department of Health Sciences and Technology, ETH Zürich, Switzerland
| | - Jacob Folz
- Laboratory of Toxicology, Institute of Food, Nutrition and Health (IFNH), Department of Health Sciences and Technology, ETH Zürich, Switzerland
| | - Karsten Beekmann
- Wageningen Food Safety Research (WFSR), Wageningen University and Research, the Netherlands
| | - Georg Aichinger
- Laboratory of Toxicology, Institute of Food, Nutrition and Health (IFNH), Department of Health Sciences and Technology, ETH Zürich, Switzerland.
| |
Collapse
|
5
|
Shete VS, Telange DR, Mahajan NM, Pethe AM, Mahapatra DK. Development of phospholipon®90H complex nanocarrier with enhanced oral bioavailability and anti-inflammatory potential of genistein. Drug Deliv 2023; 30:2162158. [PMID: 36587626 PMCID: PMC9809365 DOI: 10.1080/10717544.2022.2162158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 12/19/2022] [Indexed: 01/03/2023] Open
Abstract
Genistein (GEN), an isoflavonoid, offers multifunctional biological activities. However, its poor oral bioavailability, aqueous solubility, extensive metabolism, and short half-life restricted its clinical use. Therefore, the Phospholipon®90H complex of genistein (GPLC) was prepared to enhance its biopharmaceutical properties and anti-inflammatory activity. GPLC was characterized by employing particle size and zeta potential, Fourier transforms infrared spectrophotometry, differential scanning calorimetry, powder x-ray diffractometry, proton nuclear magnetic resonance, aqueous solubility, in vitro dissolution, ex vivo permeation, oral bioavailability and in vivo anti-inflammatory activity. The complex showed high entrapment of GEN (∼97.88% w/w) within the Phospholipon®90H matrix. Particle size and zeta potential studies confirmed the small particle size with the modest stability of GPLC. The characterization analysis supported the formation of GPLC through the participation of hydrogen bonding between GEN and Phospholipon®90H. GPLC significantly enhanced the aqueous solubility (∼2-fold) compared to GEN. Dissolution studies revealed that GPLC drastically improved the GEN dissolution rate compared to GEN. Likewise, the complex improved the permeation rate across the membrane compared to GEN. GPLC formulation significantly enhanced the oral bioavailability of GEN via improving its Cmax, tmax, AUC, half-life and mean residence time within the blood circulation compared to GEN. The GPLC (∼20 mg/kg, p.o.) remarkably inhibited the increase in paw edema up to 5 h, compared to GEN and diclofenac. Results suggest that the Phospholipon®90 complex is a superior and promising carrier for enhancing the biopharmaceutical parameters of GEN and other bioactive with similar properties.
Collapse
Affiliation(s)
- Vaishnavi S. Shete
- Datta Meghe College of Pharmacy, Datta Meghe Institute of Medical Sciences (Deemed to be University), Wardha, Maharashtra, India
| | - Darshan R. Telange
- Datta Meghe College of Pharmacy, Datta Meghe Institute of Medical Sciences (Deemed to be University), Wardha, Maharashtra, India
| | | | - Anil M. Pethe
- Datta Meghe College of Pharmacy, Datta Meghe Institute of Medical Sciences (Deemed to be University), Wardha, Maharashtra, India
| | | |
Collapse
|
6
|
Garbiec E, Rosiak N, Zalewski P, Tajber L, Cielecka-Piontek J. Genistein Co-Amorphous Systems with Amino Acids: An Investigation into Enhanced Solubility and Biological Activity. Pharmaceutics 2023; 15:2653. [PMID: 38139995 PMCID: PMC10747361 DOI: 10.3390/pharmaceutics15122653] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 11/07/2023] [Accepted: 11/15/2023] [Indexed: 12/24/2023] Open
Abstract
Genistein, an isoflavone known for its antioxidant and antidiabetic effects, suffers from the drawback of low solubility. To overcome this limitation, co-amorphous systems were synthesized by incorporating amino acids that were chosen through computational methods. The confirmation of the amorphous state of lysine and arginine-containing systems was ascertained by X-ray powder diffraction. Subsequently, the characterization of these systems was extended by employing thermo-gravimetry, differential scanning calorimetry, Fourier-transform infrared spectroscopy, and scanning electron microscopy. The investigation also included an assessment of the physical stability of the samples during storage. The apparent solubility of the systems was studied in an aqueous medium. To evaluate the in vitro permeability through the gastrointestinal tract, the parallel artificial membrane permeability assay was employed. The biological properties of the systems were assessed with regard to their antioxidant activity using 2,2-diphenyl-1-picrylhydrazyl and cupric ion-reducing antioxidant capacity assays, as well as their ability to inhibit α-glucosidase. The systems' glass transition temperatures were determined, and their homogeneity confirmed via differential scanning calorimetry analysis, while Fourier-transform infrared spectroscopy analysis provided data on molecular interactions. Stability was maintained for the entire 6-month storage duration. The co-amorphous system containing lysine displayed the most pronounced apparent solubility improvement, as well as a significant enhancement in antioxidant activity. Notably, both systems demonstrated superior α-glucosidase inhibition relative to acarbose, a standard drug for managing type 2 diabetes. The results indicate that co-amorphous systems with lysine and arginine have the potential to significantly enhance the solubility and biological activity of genistein.
Collapse
Affiliation(s)
- Ewa Garbiec
- Department of Pharmacognosy and Biomaterials, Poznan University of Medical Sciences, 3 Rokietnicka St., 60-806 Poznan, Poland; (E.G.); (N.R.); (P.Z.)
| | - Natalia Rosiak
- Department of Pharmacognosy and Biomaterials, Poznan University of Medical Sciences, 3 Rokietnicka St., 60-806 Poznan, Poland; (E.G.); (N.R.); (P.Z.)
| | - Przemysław Zalewski
- Department of Pharmacognosy and Biomaterials, Poznan University of Medical Sciences, 3 Rokietnicka St., 60-806 Poznan, Poland; (E.G.); (N.R.); (P.Z.)
| | - Lidia Tajber
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, University of Dublin, D02 PN40 Dublin, Ireland;
| | - Judyta Cielecka-Piontek
- Department of Pharmacognosy and Biomaterials, Poznan University of Medical Sciences, 3 Rokietnicka St., 60-806 Poznan, Poland; (E.G.); (N.R.); (P.Z.)
| |
Collapse
|
7
|
Berga M, Logviss K, Lauberte L, Paulausks A, Mohylyuk V. Flavonoids in the Spotlight: Bridging the Gap between Physicochemical Properties and Formulation Strategies. Pharmaceuticals (Basel) 2023; 16:1407. [PMID: 37895878 PMCID: PMC10610233 DOI: 10.3390/ph16101407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 09/27/2023] [Accepted: 09/30/2023] [Indexed: 10/29/2023] Open
Abstract
Flavonoids are hydroxylated polyphenols that are widely distributed in plants with diverse health benefits. Despite their popularity, the bioavailability of flavonoids is often overlooked, impacting their efficacy and the comparison of products. The study discusses the bioavailability-related physicochemical properties of flavonoids, with a focus on the poorly soluble compounds commonly found in dietary supplements and herbal products. This review sums up the values of pKa, log P, solubility, permeability, and melting temperature of flavonoids. Experimental and calculated data were compiled for various flavonoid subclasses, revealing variations in their physicochemical properties. The investigation highlights the challenges posed by poorly soluble flavonoids and underscores the need for enabling formulation approaches to enhance their bioavailability and therapeutic potential. Compared to aglycones, flavonoid glycosides (with sugar moieties) tend to be more hydrophilic. Most of the reviewed aglycones and glycosides exhibit relatively low log P and high melting points, making them "brick dust" candidates. To improve solubility and absorption, strategies like size reduction, the potential use of solid dispersions and carriers, as well as lipid-based formulations have been discussed.
Collapse
Affiliation(s)
| | | | | | | | - Valentyn Mohylyuk
- Laboratory of Finished Dosage Forms, Faculty of Pharmacy, Riga Stradiņš University, LV-1007 Riga, Latvia
| |
Collapse
|
8
|
Crosstalk between Resveratrol and Gut Barrier: A Review. Int J Mol Sci 2022; 23:ijms232315279. [PMID: 36499603 PMCID: PMC9739931 DOI: 10.3390/ijms232315279] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 12/01/2022] [Accepted: 12/01/2022] [Indexed: 12/08/2022] Open
Abstract
The plant-based nutraceuticals are receiving increasing interest in recent time. The high attraction to the phytochemicals is associated with their anti-inflammatory and antioxidant activities, which can lead to reduced risk of the development of cardiovascular and other non-communicable diseases. One of the most disseminated groups of plant bioactives are phenolic compounds. It was recently hypothesized that phenolic compounds can have the ability to improve the functioning of the gut barrier. The available studies showed that one of the polyphenols, resveratrol, has great potential to improve the integrity of the gut barrier. Very promising results have been obtained with in vitro and animal models. Still, more clinical trials must be performed to evaluate the effect of resveratrol on the gut barrier, especially in individuals with increased intestinal permeability. Moreover, the interplay between phenolic compounds, intestinal microbiota and gut barrier should be carefully evaluated in the future. Therefore, this review offers an overview of the current knowledge about the interaction between polyphenols with a special emphasis on resveratrol and the gut barrier, summarizes the available methods to evaluate the intestinal permeability, discusses the current research gaps and proposes the directions for future studies in this research area.
Collapse
|
9
|
Magni G, Riboldi B, Petroni K, Ceruti S. Flavonoids bridging the gut and the brain: intestinal metabolic fate, and direct or indirect effects of natural supporters against neuroinflammation and neurodegeneration. Biochem Pharmacol 2022; 205:115257. [PMID: 36179933 DOI: 10.1016/j.bcp.2022.115257] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 09/14/2022] [Accepted: 09/14/2022] [Indexed: 11/02/2022]
Abstract
In recent years, experimental evidence suggested a possible role of the gut microbiota in the onset and development of several neurodegenerative disorders, such as AD and PD, MS and pain. Flavonoids, including anthocyanins, EGCG, the flavonol quercetin, and isoflavones, are plant polyphenolic secondary metabolites that have shown therapeutic potential for the treatment of various pathological conditions, including neurodegenerative diseases. This is due to their antioxidant and anti-inflammatory properties, despite their low bioavailability which often limits their use in clinical practice. In more recent years it has been demonstrated that flavonoids are metabolized by specific bacterial strains in the gut to produce their active metabolites. On the other way round, both naturally-occurring flavonoids and their metabolites promote or limit the proliferation of specific bacterial strains, thus profoundly affecting the composition of the gut microbiota which in turn modifies its ability to further metabolize flavonoids. Thus, understanding the best way of acting on this virtuous circle is of utmost importance to develop innovative approaches to many brain disorders. In this review, we summarize some of the most recent advances in preclinical and clinical research on the neuroinflammatory and neuroprotective effects of flavonoids on AD, PD, MS and pain, with a specific focus on their mechanisms of action including possible interactions with the gut microbiota, to emphasize the potential exploitation of dietary flavonoids as adjuvants in the treatment of these pathological conditions.
Collapse
Affiliation(s)
- Giulia Magni
- Department of Pharmacological and Biomolecular Sciences - Università degli Studi di Milano - via Balzaretti, 9 - 20133 MILAN (Italy)
| | - Benedetta Riboldi
- Department of Pharmacological and Biomolecular Sciences - Università degli Studi di Milano - via Balzaretti, 9 - 20133 MILAN (Italy)
| | - Katia Petroni
- Department of Biosciences - Università degli Studi di Milano - via Celoria, 26 - 20133 MILAN (Italy)
| | - Stefania Ceruti
- Department of Pharmacological and Biomolecular Sciences - Università degli Studi di Milano - via Balzaretti, 9 - 20133 MILAN (Italy).
| |
Collapse
|
10
|
Genistein: Therapeutic and Preventive Effects, Mechanisms, and Clinical Application in Digestive Tract Tumor. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:5957378. [PMID: 35815271 PMCID: PMC9259214 DOI: 10.1155/2022/5957378] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 05/28/2022] [Indexed: 12/30/2022]
Abstract
Genistein is one of the numerous recognized isoflavones that may be found in a variety of soybeans and soy products, including tofu and tofu products. The chemical name for genistein is 4', 5, 7-trihydroxyisoflavone, and it is found in plants. In recent years, the scientific world has become more interested in genistein because of its possible therapeutic effects on many forms of cancer. It has been widely investigated for its anticancer properties. The discovery of genistein's mechanism of action indicates its potential for apoptosis induction and cell cycle arrest in gastrointestinal cancer, especially gastric and colorectal cancer. Genistein's pharmacological activities as determined by the experimental studies presented in this review lend support to its use in the treatment of gastrointestinal cancer; however, additional research is needed in the future to determine its efficacy, safety, and the potential for using nanotechnology to increase bioavailability and therapeutic efficacy.
Collapse
|
11
|
Li R, Robinson M, Ding X, Geetha T, Al-Nakkash L, Broderick TL, Babu JR. Genistein: A focus on several neurodegenerative diseases. J Food Biochem 2022; 46:e14155. [PMID: 35460092 DOI: 10.1111/jfbc.14155] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 03/15/2022] [Accepted: 03/17/2022] [Indexed: 12/14/2022]
Abstract
Neurodegenerative diseases are caused by the progressive loss of function or structure of nerve cells in the central nervous system. The most common neurodegenerative diseases include Alzheimer's disease, Huntington's disease, motor neuron disease, and Parkinson's disease. Although the physical or mental symptoms of neurodegenerative disease may be relieved by various treatment combinations, there are currently no strategies to directly slow or prevent neurodegeneration. Given the demographic evidence of a rapidly growing aging population and the associated prevalence of these common neurodegenerative diseases, it is paramount to develop safe and effective ways to protect against neurodegenerative diseases. Most neurodegenerative diseases share some common etiologies such as oxidative stress, neuroinflammation, and mitochondrial dysfunction. Genistein is an isoflavone found in soy products that have been shown to exhibit antioxidant, anti-inflammation, and estrogenic properties. Increasing evidence indicates the protective potential of genistein in neurodegenerative disorders. In this review, we aim to provide an overview of the role that genistein plays in delaying the development of neurodegenerative disease. PRACTICAL APPLICATIONS: Genistein is a naturally occurring isoflavone found mainly in soybean, but also green peas, legumes, and peanuts. Genistein is found to pass through the blood-brain barrier and possess a neuroprotective effect. In this review, we discuss studies in support of these actions and the underlying biological mechanisms. Together, these data indicate that genistein may hold neuroprotective effects in either delaying the onset or relieving the symptoms of neurodegenerative disease.
Collapse
Affiliation(s)
- Rongzi Li
- Department of Nutrition, Dietetics, and Hospitality Management, Auburn University, Auburn, Alabama, USA
| | - Megan Robinson
- Department of Nutrition, Dietetics, and Hospitality Management, Auburn University, Auburn, Alabama, USA
| | - Xiaowen Ding
- Department of Nutrition, Dietetics, and Hospitality Management, Auburn University, Auburn, Alabama, USA
| | - Thangiah Geetha
- Department of Nutrition, Dietetics, and Hospitality Management, Auburn University, Auburn, Alabama, USA
- Boshell Metabolic Diseases and Diabetes Program, Auburn University, Auburn, Alabama, USA
| | - Layla Al-Nakkash
- Department of Physiology, College of Graduate Studies, Midwestern University, Glendale, Arizona, USA
| | - Tom L Broderick
- Department of Physiology, Laboratory of Diabetes and Exercise Metabolism College of Graduate Studies, Midwestern University, Glendale, Arizona, USA
| | - Jeganathan Ramesh Babu
- Department of Nutrition, Dietetics, and Hospitality Management, Auburn University, Auburn, Alabama, USA
- Boshell Metabolic Diseases and Diabetes Program, Auburn University, Auburn, Alabama, USA
| |
Collapse
|
12
|
Genistein, a tool for geroscience. Mech Ageing Dev 2022; 204:111665. [DOI: 10.1016/j.mad.2022.111665] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/04/2022] [Accepted: 03/15/2022] [Indexed: 12/12/2022]
|
13
|
Kim MS, Jung YS, Jang D, Cho CH, Lee SH, Han NS, Kim DO. Antioxidant capacity of 12 major soybean isoflavones and their bioavailability under simulated digestion and in human intestinal Caco-2 cells. Food Chem 2022; 374:131493. [PMID: 34802809 DOI: 10.1016/j.foodchem.2021.131493] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 10/23/2021] [Accepted: 10/26/2021] [Indexed: 12/22/2022]
Abstract
Soy isoflavones (SIs) show various health benefits, such as antioxidant and estrogenic effects. It is important to understand the bioaccessibility and bioavailability of SIs due to the close relation to their bioactivities. In this study, the antioxidant capacity, bioaccessibility, and bioavailability of 12 SIs were evaluated using radical-scavenging methods, simulations of human digestion, and Caco-2 cells in Transwell, respectively. All SIs were stable (91.1-99.2%) under gastric digestion conditions compared with the control (100%), whereas acetyl and malonyl conjugates were unstable (38.5% and 65.5%, respectively) under small intestinal digestion conditions. SI aglycones showed higher permeability (7-15 times) and cellular accumulation (8.8 times) than their glucosides. A small amount of SI conjugates was intact in the cell and in the basolateral side of each Transwell. These results suggest that SI conjugates, especially malonyl and acetyl forms, have incidental bioactivity after being metabolized to aglycones inside the cell.
Collapse
Affiliation(s)
- Mi-Seon Kim
- Graduate School of Biotechnology, Kyung Hee University, Yongin 17104, Republic of Korea
| | - Young Sung Jung
- Department of Food Science and Biotechnology, Kyung Hee University, Yongin 17104, Republic of Korea
| | - Davin Jang
- Graduate School of Biotechnology, Kyung Hee University, Yongin 17104, Republic of Korea
| | - Chi Heung Cho
- Division of Functional Food Research, Korea Food Research Institute, Wanju 55365, Republic of Korea
| | - Sang-Hoon Lee
- Division of Functional Food Research, Korea Food Research Institute, Wanju 55365, Republic of Korea
| | - Nam Soo Han
- Brain Korea 21 Center for Bio-Health Industry, Department of Food Science and Biotechnology, Chungbuk National University, Chungbuk 28644, Republic of Korea
| | - Dae-Ok Kim
- Graduate School of Biotechnology, Kyung Hee University, Yongin 17104, Republic of Korea; Department of Food Science and Biotechnology, Kyung Hee University, Yongin 17104, Republic of Korea.
| |
Collapse
|
14
|
Zuñiga-Martínez BS, Domínguez-Avila JA, Wall-Medrano A, Ayala-Zavala JF, Hernández-Paredes J, Salazar-López NJ, Villegas-Ochoa MA, González-Aguilar GA. Avocado paste from industrial byproducts as an unconventional source of bioactive compounds: characterization, in vitro digestion and in silico interactions of its main phenolics with cholesterol. JOURNAL OF FOOD MEASUREMENT AND CHARACTERIZATION 2021. [DOI: 10.1007/s11694-021-01117-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
15
|
Kitaguchi T, Mizota T, Ito M, Ohno K, Kobayashi K, Ogawa I, Qiu S, Iwao T, Hanioka N, Tanaka M, Matsunaga T. Simultaneous evaluation of membrane permeability and UDP-glucuronosyltransferase-mediated metabolism of food-derived compounds using human induced pluripotent stem cell-derived small intestinal epithelial cells. Drug Metab Dispos 2021; 50:17-23. [PMID: 34670778 DOI: 10.1124/dmd.121.000605] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 10/18/2021] [Indexed: 11/22/2022] Open
Abstract
Pharmacokinetic prediction after oral ingestion is important for quantitative risk assessment of food-derived compounds. To evaluate the utility of human intestinal absorption prediction, we compared the membrane permeability and metabolic activities of human induced pluripotent stem cell-derived small intestinal epithelial cells (hiPSC-SIECs) with Caco-2 cells or human primary enterocytes (hPECs). We found that membrane permeability in hiPSC-SIECs had better predictivity than that in Caco-2 cells against 21 drugs with known human intestinal availability (r = 0.830 and 0.401, respectively). Membrane permeability in hiPSC-SIECs was only 0.019-0.25-fold as compared with that in Caco-2 cells for 7 in 15 food-derived compounds, primarily those which were reported to undergo glucuronidation metabolism. The metabolic rates of the glucuronide conjugate were similar or higher in hiPSC-SIECs as compared with hPECs, while lower in Caco-2 cells. Expression levels of UDP-glucuronosyltransferase (UGT) isoform mRNA in hiPSC-SIECs were similar or higher as compared with hPECs. Therefore, hiPSC-SIECs could be a useful tool for predicting human intestinal absorption, in order to simultaneously evaluate membrane permeability and UGT-mediated metabolism. Significance Statement Gastrointestinal absorption is an important step for predicting the internal exposure of food-derived compounds. This research revealed that human induced pluripotent stem cell-derived small intestinal cells (hiPSC-SIECs) had better predictivity of intestinal availability than Caco-2 cells; furthermore, the metabolic rates of UGT substrates of hiPSC-SIECs were closer those of human primary enterocytes than those of Caco-2 cells. Therefore, hiPSC-SIECs could be a useful tool for predicting human intestinal absorption to simultaneously evaluate membrane permeability and UGT-mediated metabolism.
Collapse
Affiliation(s)
- Takashi Kitaguchi
- Global Food Safety Institute, Nissin Foods Holdings Co., Ltd., Japan
| | - Taisei Mizota
- Global Food Safety Institute, Nissin Foods Holdings Co., Ltd., Japan
| | - Mina Ito
- Global Food Safety Institute, Nissin Foods Holdings Co., Ltd., Japan
| | - Katsutoshi Ohno
- Global Food Safety Institute, Nissin Foods Holdings Co., Ltd., Japan
| | | | - Isamu Ogawa
- Department of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Nagoya City University, Japan
| | - Shimeng Qiu
- Department of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Nagoya City University, Japan
| | | | - Nobumitsu Hanioka
- Department of Health Pharmacy, Yokohama University of Pharmacy, Japan
| | - Mitsuru Tanaka
- Global Food Safety Institute, Nissin Foods Holdings Co., Ltd., Japan
| | - Tamihide Matsunaga
- Department of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Nagoya City University, Japan
| |
Collapse
|
16
|
Botet-Carreras A, Tamames-Tabar C, Salles F, Rojas S, Imbuluzqueta E, Lana H, Blanco-Prieto MJ, Horcajada P. Improving the genistein oral bioavailability via its formulation into the metal-organic framework MIL-100(Fe). J Mater Chem B 2021; 9:2233-2239. [PMID: 33596280 DOI: 10.1039/d0tb02804e] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Despite the interesting chemopreventive, antioxidant and antiangiogenic effects of the natural bioflavonoid genistein (GEN), its low aqueous solubility and bioavailability make it necessary to administer it using a suitable drug carrier system. Nanometric porous metal-organic frameworks (nanoMOFs) are appealing systems for drug delivery. Particularly, mesoporous MIL-100(Fe) possesses a variety of interesting features related to its composition and structure, which make it an excellent candidate to be used as a drug nanocarrier (highly porous, biocompatible, can be synthesized as homogenous and stable nanoparticles (NPs), etc.). In this study, GEN was entrapped via simple impregnation in MIL-100 NPs achieving remarkable drug loading (27.1 wt%). A combination of experimental and computing techniques was used to achieve a deep understanding of the encapsulation of GEN in MIL-100 nanoMOF. Subsequently, GEN delivery studies were carried out under simulated physiological conditions, showing on the whole a sustained GEN release for 3 days. Initial pharmacokinetic and biodistribution studies were also carried out upon the oral administration of the GEN@MIL-100 NPs in a mouse model, evidencing a higher bioavailability and showing that this oral nanoformulation appears to be very promising. To the best of our knowledge, the GEN-loaded MIL-100 will be the first antitumor oral formulation based on nanoMOFs studied in vivo, and paves the way to the efficient delivery of nontoxic antitumorals via a convenient oral route.
Collapse
Affiliation(s)
- Adrià Botet-Carreras
- Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Navarra, Irunlarrea 1, 31008 Pamplona, Spain. and Institut Lavoisier, UMR CNRS 8180, Université de Versailles Saint-Quentin-en-Yvelines, 45 Avenue des Etats-Unis, 78035 Versailles Cedex, France
| | - Cristina Tamames-Tabar
- Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Navarra, Irunlarrea 1, 31008 Pamplona, Spain. and Institut Lavoisier, UMR CNRS 8180, Université de Versailles Saint-Quentin-en-Yvelines, 45 Avenue des Etats-Unis, 78035 Versailles Cedex, France
| | - Fabrice Salles
- ICGM, CNRS, Univ. Montpellier, ENSCM, Montpellier, France
| | - Sara Rojas
- IMDEA Energy, Avda. Ramón de la Sagra 3, 28035 Móstoles, Madrid, Spain.
| | - Edurne Imbuluzqueta
- Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Navarra, Irunlarrea 1, 31008 Pamplona, Spain.
| | - Hugo Lana
- Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Navarra, Irunlarrea 1, 31008 Pamplona, Spain.
| | - María José Blanco-Prieto
- Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Navarra, Irunlarrea 1, 31008 Pamplona, Spain.
| | - Patricia Horcajada
- Institut Lavoisier, UMR CNRS 8180, Université de Versailles Saint-Quentin-en-Yvelines, 45 Avenue des Etats-Unis, 78035 Versailles Cedex, France and IMDEA Energy, Avda. Ramón de la Sagra 3, 28035 Móstoles, Madrid, Spain.
| |
Collapse
|
17
|
An Overview on Dietary Polyphenols and Their Biopharmaceutical Classification System (BCS). Int J Mol Sci 2021; 22:ijms22115514. [PMID: 34073709 PMCID: PMC8197262 DOI: 10.3390/ijms22115514] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 05/18/2021] [Accepted: 05/20/2021] [Indexed: 12/12/2022] Open
Abstract
Polyphenols are natural organic compounds produced by plants, acting as antioxidants by reacting with ROS. These compounds are widely consumed in daily diet and many studies report several benefits to human health thanks to their bioavailability in humans. However, the digestion process of phenolic compounds is still not completely clear. Moreover, bioavailability is dependent on the metabolic phase of these compounds. The LogP value can be managed as a simplified measure of the lipophilicity of a substance ingested within the human body, which affects resultant absorption. The biopharmaceutical classification system (BCS), a method used to classify drugs intended for gastrointestinal absorption, correlates the solubility and permeability of the drug with both the rate and extent of oral absorption. BCS may be helpful to measure the bioactive constituents of foods, such as polyphenols, in order to understand their nutraceutical potential. There are many literature studies that focus on permeability, absorption, and bioavailability of polyphenols and their resultant metabolic byproducts, but there is still confusion about their respective LogP values and BCS classification. This review will provide an overview of the information regarding 10 dietarypolyphenols (ferulic acid, chlorogenic acid, rutin, quercetin, apigenin, cirsimaritin, daidzein, resveratrol, ellagic acid, and curcumin) and their association with the BCS classification.
Collapse
|
18
|
Guo TL, Chen Y, Xu HS, McDonough CM, Huang G. Gut microbiome in neuroendocrine and neuroimmune interactions: The case of genistein. Toxicol Appl Pharmacol 2020; 402:115130. [PMID: 32673657 DOI: 10.1016/j.taap.2020.115130] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 07/01/2020] [Accepted: 07/04/2020] [Indexed: 12/12/2022]
Abstract
The healthy and diverse microbes living in our gut provide numerous benefits to our health. It is increasingly recognized that the gut microbiome affects the host's neurobehavioral state through production of metabolites, modulation of intestinal immunity (e.g., cytokines) and other mechanisms (e.g., gut neuropeptides). By sending the sensed information (e.g., metabolic and immunologic mediators) about the state of the inner organs to the brain via afferent fibers, the vagus nerve maintains one of the connections between the brain and GI tract, and oversees many critical bodily functions (e.g., mood, immune response, digestion and heart rate). The microbiota-gut-brain axis is a bidirectional communication between the gut, its microbiome, and the nervous system. In the present review, the roles of microbiome in neuroendocrine and neuroimmune interactions have been discussed using naturally occurring isoflavones, particularly the phytoestrogen genistein, as there are sex differences in the interactions among the microbiome, hormones, immunity and disease susceptibility. A deep understanding of the mechanisms underlying the interactions among the endocrine modulators, brain, endocrine glands, gut immune cells, vagus nerve, enteric nervous system and gut microbiome will provide important knowledges that may ultimately lead to treatment and prevention of debilitating disorders characterized by deficits of microbiome-neuroendocrine-neuroimmune relationships.
Collapse
Affiliation(s)
- Tai L Guo
- Department of Veterinary Biosciences and Diagnostic Imaging, College of Veterinary Medicine, University of Georgia, Athens, GA, USA.
| | - Yingjia Chen
- Department of Veterinary Biosciences and Diagnostic Imaging, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | - Hannah Shibo Xu
- Department of Veterinary Biosciences and Diagnostic Imaging, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | - Callie M McDonough
- Department of Veterinary Biosciences and Diagnostic Imaging, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | | |
Collapse
|
19
|
Structure-bioavailability relationship study of genistein derivatives with antiproliferative activity on human cancer cell. J Pharm Biomed Anal 2020; 185:113216. [PMID: 32155543 DOI: 10.1016/j.jpba.2020.113216] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 02/24/2020] [Accepted: 02/26/2020] [Indexed: 11/21/2022]
Abstract
The present study assesses the in vitro and in vivo bioavailability of genistein derivatives, hydroxyalkyl- and glycosyl alkyl ethers (glycoconjugates). Studies were carried out using compounds that exhibit higher in vitro antiproliferative activity in comparison with the parent isoflavone. Based on in vitro experiments using the Parallel Artificial Membrane Permeability Assay (PAMPA) and the Caco-2 cell monolayer permeability model, we found that modification of the isoflavone structure by O-alkylation improved bioavailability in comparison to genistein. Additionally, the structure of the substituent and its position on genistein influenced the type of mechanism involved in the transport of compounds through biological membranes. The PAMPA assay showed that the structure of glycoconjugates had a significant influence on the passive transport of the genistein synthetic derivatives through a biological membrane. Preferentially the glycoconjugates containing O-glycosidic bond were transported and the transport rate decreased as the carbon linker increased. For glycoconjugates, determination of their transport and metabolism through the Caco-2 membrane was not possible due to interaction with the membrane surface, probably by the change of compound structure caused by contact with the cells or degradation in medium. The intestinal absorption and metabolism of genistein and three derivatives, Ram-3, Ram'-3 and Ram-C-4α (Fig. 1), were tested in vivo in rats. We found that in comparison to genistein, glycoconjugates were metabolized more slowly and to a lesser extent. As part of the in vivo research, we performed analysis of compound levels in plasma samples after enzymatic hydrolysis, but in the collected samples, analytes were not observed. We hypothesize that glycoconjugates compounds bind plasma proteins and were removed from the sample. In conclusion, we show that O-functionalization of the natural, biologically active isoflavone genistein can affect biological activity, bioavailability, and the rate of compound metabolism. The position of the substituent, the length of the linker and the structure of sugar moieties provides a tool for the optimization of the derivative's biological properties.
Collapse
|
20
|
Kawahara I, Nishikawa S, Yamamoto A, Kono Y, Fujita T. The Impact of Breast Cancer Resistance Protein (BCRP/ABCG2) on Drug Transport Across Caco-2 Cell Monolayers. Drug Metab Dispos 2020; 48:491-498. [PMID: 32193356 DOI: 10.1124/dmd.119.088674] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 03/06/2020] [Indexed: 01/16/2023] Open
Abstract
Breast cancer resistance protein (BCRP) is expressed on the apical membrane of small intestinal epithelial cells and functions as an efflux pump with broad substrate recognition. Therefore, quantitative evaluation of the contribution of BCRP to the intestinal permeability of new chemical entities is very important in drug research and development. In this study, we assessed the BCRP-mediated efflux of several model drugs in Caco-2 cells using WK-X-34 as a dual inhibitor of P-glycoprotein (P-gp) and BCRP and LY335979 as a selective inhibitor of P-gp. The permeability of daidzein was high with an apparent permeability coefficient for apical-to-basal transport (P AB) of 20.3 × 10-6 cm/s. In addition, its efflux ratio (ER) was 1.55, indicating that the contribution of BCRP to its transport is minimal. Estrone-3-sulfate and ciprofloxacin showed relatively higher ER values (>2.0), whereas their BCRP-related absorptive quotient (AQ BCRP) was 0.21 and 0.3, respectively. These results indicate that BCRP does not play a major role in regulating the permeability of estrone-3-sulfate and ciprofloxacin in Caco-2 cells. Nitrofurantoin showed a P AB of 1.8 × 10-6 cm/s, and its ER was 7.6. However, the AQ BCRP was 0.37, suggesting minimal contribution of BCRP to nitrofurantoin transport in Caco-2 cells. In contrast, topotecan, SN-38, and sulfasalazine had low P AB values (0.81, 1.13, and 0.19 × 10-6 cm/s, respectively), and each AQ BCRP was above 0.6, indicating that BCRP significantly contributes to the transport of these compounds in Caco-2 cells. In conclusion, Caco-2 cells are useful to accurately estimate the contribution of BCRP to intestinal drug absorption. SIGNIFICANCE STATEMENT: We performed an in vitro assessment of the contribution of breast cancer resistance protein (BCRP) to the transport of BCRP and/or P-glycoprotein (P-gp) substrates across Caco-2 cell monolayers using absorptive quotient, which has been proposed to represent the contribution of drug efflux transporters to the net efflux. The present study demonstrates that the combined use of a BCRP/P-gp dual inhibitor and a P-gp selective inhibitor is useful to estimate the impact of BCRP and P-gp on the permeability of tested compounds in Caco-2 cells.
Collapse
Affiliation(s)
- Iichiro Kawahara
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Kyoto, Japan (I.K., S.N., A.Y.) and Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, Ritsumeikan University, Shiga, Japan (Y.K., T.F.)
| | - Satoyo Nishikawa
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Kyoto, Japan (I.K., S.N., A.Y.) and Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, Ritsumeikan University, Shiga, Japan (Y.K., T.F.)
| | - Akira Yamamoto
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Kyoto, Japan (I.K., S.N., A.Y.) and Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, Ritsumeikan University, Shiga, Japan (Y.K., T.F.)
| | - Yusuke Kono
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Kyoto, Japan (I.K., S.N., A.Y.) and Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, Ritsumeikan University, Shiga, Japan (Y.K., T.F.)
| | - Takuya Fujita
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Kyoto, Japan (I.K., S.N., A.Y.) and Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, Ritsumeikan University, Shiga, Japan (Y.K., T.F.)
| |
Collapse
|
21
|
Wang Q, Spenkelink B, Boonpawa R, Rietjens IMCM, Beekmann K. Use of Physiologically Based Kinetic Modeling to Predict Rat Gut Microbial Metabolism of the Isoflavone Daidzein to S-Equol and Its Consequences for ERα Activation. Mol Nutr Food Res 2020; 64:e1900912. [PMID: 32027771 PMCID: PMC7154660 DOI: 10.1002/mnfr.201900912] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 11/06/2019] [Indexed: 12/13/2022]
Abstract
SCOPE To predict gut microbial metabolism of xenobiotics and the resulting plasma concentrations of metabolites formed, an in vitro-in silico-based testing strategy is developed using the isoflavone daidzein and its gut microbial metabolite S-equol as model compounds. METHODS AND RESULTS Anaerobic rat fecal incubations are optimized and performed to derive the apparent maximum velocities (Vmax ) and Michaelis-Menten constants (Km ) for gut microbial conversion of daidzein to dihydrodaidzein, S-equol, and O-desmethylangolensin, which are input as parameters for a physiologically based kinetic (PBK) model. The inclusion of gut microbiota in the PBK model allows prediction of S-equol concentrations and slightly reduced predicted maximal daidzein concentrations from 2.19 to 2.16 µm. The resulting predicted concentrations of daidzein and S-equol are comparable to in vivo concentrations reported. CONCLUSION The optimized in vitro approach to quantify kinetics for gut microbial conversions, and the newly developed PBK model for rats that includes gut microbial metabolism, provide a unique tool to predict the in vivo consequences of daidzein microbial metabolism for systemic exposure of the host to daidzein and its metabolite S-equol. The predictions reveal a dominant role for daidzein in ERα-mediated estrogenicity despite the higher estrogenic potency of its microbial metabolite S-equol.
Collapse
Affiliation(s)
- Qianrui Wang
- Division of ToxicologyWageningen University and ResearchWageningen6708 WEThe Netherlands
| | - Bert Spenkelink
- Division of ToxicologyWageningen University and ResearchWageningen6708 WEThe Netherlands
| | - Rungnapa Boonpawa
- Faculty of Natural Resources and Agro‐IndustryKasetsart University Chalermphrakiat Sakon Nakhon Province CampusSakon Nakhon47000Thailand
| | | | - Karsten Beekmann
- Division of ToxicologyWageningen University and ResearchWageningen6708 WEThe Netherlands
- Present address:
Wageningen Food Safety ResearchP. O. Box 2306700 AEWageningenThe Netherlands
| |
Collapse
|
22
|
Fujitani M, Mizushige T, Bhattarai K, Adhikari S, Ishikawa J, Kishida T. Dietary daidzein induces accumulation of S-equol in enterohepatic circulation to far higher levels than that of daidzein in female rats with and without ovariectomy. Biomed Res 2019; 40:97-105. [PMID: 31231095 DOI: 10.2220/biomedres.40.97] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
We previously found that daidzein decreased food intake in female rats. To understand the mechanism of anorectic action of dietary daidzein, it is necessary to determine distributions of daidzein and S-equol, a metabolite of intestinal bacterial conversion from daidzein, in the body. In the present study, we measured the concentrations of daidzein and S-equol in serum and bile in sham-operated and ovariectomized female rats fed a diet containing 150 mg/kg daidzein for 7 days. Dietary daidzein increased serum and bile concentrations of S-equol to far higher levels than those of daidzein. S-equol concentration was more than several hundred fold-higher in bile than in serum, regardless of ovariectomy. Moreover, to investigate whether accumulation of S-equol is facilitated by efficient enterohepatic circulation during continuous intake of daidzein and S-equol, female rats were fed diet containing daidzein or S-equol (both 150 mg/kg), or control diet for 1, 2, 3, or 5 days. Dietary daidzein significantly increased serum and bile concentrations of S-equol in a time-dependent manner, but not those of daidzein. These results indicated that substantial proportion of dietary daidzein was converted to S-equol, which underwent efficient enterohepatic circulation and predominantly accumulated there.
Collapse
Affiliation(s)
| | - Takafumi Mizushige
- Department of Applied Biological Chemistry, Faculty of Agriculture, Utsunomiya University
| | - Keshab Bhattarai
- Graduate School of Agriculture, Ehime University.,The United Graduate School of Agricultural Sciences, Ehime University
| | | | - Junji Ishikawa
- Central Research and Development Laboratory, FANCL Co. Ltd
| | - Taro Kishida
- Graduate School of Agriculture, Ehime University.,The United Graduate School of Agricultural Sciences, Ehime University
| |
Collapse
|
23
|
Inui TA, Yasuda M, Hirano S, Ikeuchi Y, Kogiso H, Inui T, Marunaka Y, Nakahari T. Daidzein-Stimulated Increase in the Ciliary Beating Amplitude via an [Cl -] i Decrease in Ciliated Human Nasal Epithelial Cells. Int J Mol Sci 2018; 19:ijms19123754. [PMID: 30486295 PMCID: PMC6321431 DOI: 10.3390/ijms19123754] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 11/22/2018] [Accepted: 11/22/2018] [Indexed: 01/26/2023] Open
Abstract
The effects of the isoflavone daidzein on the ciliary beat distance (CBD, which is a parameter assessing the amplitude of ciliary beating) and the ciliary beat frequency (CBF) were examined in ciliated human nasal epithelial cells (cHNECs) in primary culture. Daidzein decreased [Cl−]i and enhanced CBD in cHNECs. The CBD increase that was stimulated by daidzein was mimicked by Cl−-free NO3− solution and bumetanide (an inhibitor of Na+/K+/2Cl− cotransport), both of which decreased [Cl−]i. Moreover, the CBD increase was inhibited by 5-Nitro-2-(3-phenylpropylamino)benzoic acid (NPPB, a Cl− channel blocker), which increased [Cl−]i. CBF was also decreased by NPPB. The rate of [Cl−]i decrease evoked by Cl−-free NO3− solution was enhanced by daidzein. These results suggest that daidzein activates Cl− channels in cHNECs. Moreover, daidzein enhanced the microbead transport driven by beating cilia in the cell sheet of cHNECs, suggesting that an increase in CBD enhances ciliary transport. An [Cl−]i decrease enhanced CBD, but not CBF, in cHNECs at 37 °C, although it enhanced both at 25 °C. Intracellular Cl− affects both CBD and CBF in a temperature-dependent manner. In conclusion, daidzein, which activates Cl− channels to decrease [Cl−]i, stimulated CBD increase in cHNECs at 37 °C. CBD is a crucial factor that can increase ciliary transport in the airways under physiological conditions.
Collapse
Affiliation(s)
- Taka-Aki Inui
- Department of Molecular Cell Physiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan.
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan.
| | - Makoto Yasuda
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan.
| | - Shigeru Hirano
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan.
| | - Yukiko Ikeuchi
- Department of Molecular Cell Physiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan.
| | - Haruka Kogiso
- Department of Molecular Cell Physiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan.
| | - Toshio Inui
- Research Center for Drug Discovery and Pharmaceutical Development Science, Research Organization of Science and Technology, BKC, Ritsumeikan University, Kusatsu 525-8577, Japan.
- Saisei Mirai Clinics, Moriguchi 570-0012, Japan.
| | - Yoshinori Marunaka
- Department of Molecular Cell Physiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan.
- Research Center for Drug Discovery and Pharmaceutical Development Science, Research Organization of Science and Technology, BKC, Ritsumeikan University, Kusatsu 525-8577, Japan.
- Research Institute for Clinical Physiology, Kyoto Industrial Health Association, Kyoto 604-8472, Japan.
| | - Takashi Nakahari
- Research Center for Drug Discovery and Pharmaceutical Development Science, Research Organization of Science and Technology, BKC, Ritsumeikan University, Kusatsu 525-8577, Japan.
| |
Collapse
|
24
|
Domínguez-Avila JA, Wall-Medrano A, Velderrain-Rodríguez GR, Chen CYO, Salazar-López NJ, Robles-Sánchez M, González-Aguilar GA. Gastrointestinal interactions, absorption, splanchnic metabolism and pharmacokinetics of orally ingested phenolic compounds. Food Funct 2018; 8:15-38. [PMID: 28074953 DOI: 10.1039/c6fo01475e] [Citation(s) in RCA: 111] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The positive health effects of phenolic compounds (PCs) have been extensively reported in the literature. An understanding of their bioaccessibility and bioavailability is essential for the elucidation of their health benefits. Before reaching circulation and exerting bioactions in target tissues, numerous interactions take place before and during digestion with either the plant or host's macromolecules that directly impact the organism and modulate their own bioaccessibility and bioavailability. The present work is focused on the gastrointestinal (GI) interactions that are relevant to the absorption and metabolism of PCs and how these interactions impact their pharmacokinetic profiles. Non-digestible cell wall components (fiber) interact intimately with PCs and delay their absorption in the small intestine, instead carrying them to the large intestine. PCs not bound to fiber interact with digestible nutrients in the bolus where they interfere with the digestion and absorption of proteins, carbohydrates, lipids, cholesterol, bile salts and micronutrients through the inhibition of digestive enzymes and enterocyte transporters and the disruption of micelle formation. PCs internalized by enterocytes may reach circulation (through transcellular or paracellular transport), be effluxed back into the lumen (P-glycoprotein, P-gp) or be metabolized by phase I and phase II enzymes. Some PCs can inhibit P-gp or phase I/II enzymes, which can potentially lead to drug-nutrient interactions. The absorption and pharmacokinetic parameters are modified by all of the interactions within the digestive tract and by the presence of other PCs. Undesirable interactions have promoted the development of nanotechnological approaches to promote the bioaccessibility, bioavailability, and bioefficacy of PCs.
Collapse
Affiliation(s)
- J Abraham Domínguez-Avila
- Coordinación de Tecnología de Alimentos de Origen Vegetal, Centro de Investigación en Alimentación y Desarrollo, A.C. Carretera a la Victoria Km 0.6. C.P. 83304, Hermosillo, Sonora, Mexico.
| | - Abraham Wall-Medrano
- Departamento de Ciencias Químico-Biológicas, Instituto de Ciencias Biomédicas, Universidad Autónoma de Ciudad Juárez, Anillo Envolvente del Pronaf y Estocolmo s/n, CP 32310, Cd. Juárez, Chihuahua, Mexico.
| | - Gustavo R Velderrain-Rodríguez
- Coordinación de Tecnología de Alimentos de Origen Vegetal, Centro de Investigación en Alimentación y Desarrollo, A.C. Carretera a la Victoria Km 0.6. C.P. 83304, Hermosillo, Sonora, Mexico.
| | - C-Y Oliver Chen
- Antioxidants Research Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, 711 Washington Street, Boston, Massachusetts 02111, USA.
| | - Norma Julieta Salazar-López
- Departamento de Investigación y Posgrado en Alimentos, Universidad de Sonora, Blvd. Luis Encinas y Rosales S/N Col. Centro, C.P. 83000, Hermosillo, Sonora, Mexico.
| | - Maribel Robles-Sánchez
- Departamento de Investigación y Posgrado en Alimentos, Universidad de Sonora, Blvd. Luis Encinas y Rosales S/N Col. Centro, C.P. 83000, Hermosillo, Sonora, Mexico.
| | - Gustavo A González-Aguilar
- Coordinación de Tecnología de Alimentos de Origen Vegetal, Centro de Investigación en Alimentación y Desarrollo, A.C. Carretera a la Victoria Km 0.6. C.P. 83304, Hermosillo, Sonora, Mexico.
| |
Collapse
|
25
|
Yuan ZW, Li YZ, Liu ZQ, Feng SL, Zhou H, Liu CX, Liu L, Xie Y. Role of tangeretin as a potential bioavailability enhancer for silybin: Pharmacokinetic and pharmacological studies. Pharmacol Res 2018; 128:153-166. [DOI: 10.1016/j.phrs.2017.09.019] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 09/05/2017] [Accepted: 09/24/2017] [Indexed: 01/23/2023]
|
26
|
Maresca M, Pinton P, Ajandouz EH, Menard S, Ferrier L, Oswald IP. Overview and Comparison of Intestinal Organotypic Models, Intestinal Cells, and Intestinal Explants Used for Toxicity Studies. Curr Top Microbiol Immunol 2018; 430:247-264. [PMID: 30259111 DOI: 10.1007/82_2018_142] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The intestine is a complex organ formed of different types of cell distributed in different layers of tissue. To minimize animal experiments, for decades, researchers have been trying to develop in vitro/ex vivo systems able to mimic the cellular diversity naturally found in the gut. Such models not only help our understanding of the gut physiology but also of intestinal toxicity. This review describes the different systems used to evaluate the effects of drugs/contaminants on intestinal functions and compares their advantages and limitations. The comparison showed that the organotypic model is the best available model to perform intestinal toxicity studies, including on human tissues.
Collapse
Affiliation(s)
- Marc Maresca
- Aix Marseille Univ, CNRS, Centrale Marseille, iSm2, Marseille, France
| | - Philippe Pinton
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRA, ENVT, INP-Purpan, UPS, Toulouse, France
| | | | - Sandrine Menard
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRA, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Laurent Ferrier
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRA, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Isabelle P Oswald
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRA, ENVT, INP-Purpan, UPS, Toulouse, France.
| |
Collapse
|
27
|
Eaton AD, Zimmermann C, Delaney B, Hurley BP. Primary human polarized small intestinal epithelial barriers respond differently to a hazardous and an innocuous protein. Food Chem Toxicol 2017; 106:70-77. [PMID: 28533127 DOI: 10.1016/j.fct.2017.05.038] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 05/17/2017] [Accepted: 05/18/2017] [Indexed: 02/08/2023]
Abstract
An experimental platform employing human derived intestinal epithelial cell (IEC) line monolayers grown on permeable Transwell® filters was previously investigated to differentiate between hazardous and innocuous proteins. This approach was effective at distinguishing these types of proteins and perturbation of monolayer integrity, particularly transepithelial electrical resistance (TEER), was the most sensitive indicator. In the current report, in vitro indicators of monolayer integrity, cytotoxicity, and inflammation were evaluated using primary (non-transformed) human polarized small intestinal epithelial barriers cultured on Transwell® filters to compare effects of a hazardous protein (Clostridium difficile Toxin A [ToxA]) and an innocuous protein (bovine serum albumin [BSA]). ToxA exerted a reproducible decrease on barrier integrity at doses comparable to those producing effects observed from cell line-derived IEC monolayers, with TEER being the most sensitive indicator. In contrast, BSA, tested at concentrations substantially higher than ToxA, did not cause changes in any of the tested variables. These results demonstrate a similarity in response to certain proteins between cell line-derived polarized IEC models and a primary human polarized small intestinal epithelial barrier model, thereby reinforcing the potential usefulness of cell line-derived polarized IECs as a valid experimental platform to differentiate between hazardous and non-hazardous proteins.
Collapse
Affiliation(s)
- A D Eaton
- Department of Pediatrics, Mucosal Immunology & Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | | | | - B P Hurley
- Department of Pediatrics, Mucosal Immunology & Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
28
|
Hurley BP, Eaton AD, Zimmermann C, Delaney B. Polarized monolayer cultures of human intestinal epithelial cell lines exposed to intractable proteins - In vitro hazard identification studies. Food Chem Toxicol 2016; 98:262-268. [DOI: 10.1016/j.fct.2016.11.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Revised: 11/04/2016] [Accepted: 11/05/2016] [Indexed: 12/01/2022]
|
29
|
Hurley BP, Pirzai W, Eaton AD, Harper M, Roper J, Zimmermann C, Ladics GS, Layton RJ, Delaney B. An experimental platform using human intestinal epithelial cell lines to differentiate between hazardous and non-hazardous proteins. Food Chem Toxicol 2016; 92:75-87. [DOI: 10.1016/j.fct.2016.04.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 04/01/2016] [Accepted: 04/04/2016] [Indexed: 10/22/2022]
|
30
|
de Oliveira MR. Evidence for genistein as a mitochondriotropic molecule. Mitochondrion 2016; 29:35-44. [PMID: 27223841 DOI: 10.1016/j.mito.2016.05.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 05/13/2016] [Accepted: 05/16/2016] [Indexed: 12/19/2022]
Abstract
Genistein (4',5,7-trihydroxyisoflavone; C15H10O5), an isoflavone, has been investigated as an anti-cancer agent due to its ability to trigger cell death (both intrinsic and extrinsic apoptotic pathways) in different cancer cells in vitro and in vivo. Furthermore, genistein has been viewed as a mitochondriotropic molecule due to the direct effects this isoflavone induces in mitochondria, such as modulation of enzymatic activity of components of the oxidative phosphorylation system. Apoptosis triggering may also be mediated by genistein through activation of the mitochondria-dependent pathway by a mechanism associated with mitochondrial dysfunction (i.e., disruption of the mitochondrial membrane potential - MMP, release of cytochrome c, activation of the apoptosome, among others). Efforts have been made in order to elucidate how genistein coordinate these biochemical phenomena. Nonetheless, some areas of the mitochondria-associated research (mitochondrial biogenesis, redox biology of mitochondria, and mitochondria-associated bioenergetic parameters) need to be explored regarding the role of genistein as a mitochondria-targeted agent. This is a pharmacologically relevant issue due to the possibility of using genistein as a mitochondria-targeted drug in cases of cancer, neurodegeneration, cardiovascular, and endocrine disease, for example. The present review aims to describe, compare, and discuss relevant data about the effects of genistein upon mitochondria.
Collapse
Affiliation(s)
- Marcos Roberto de Oliveira
- Programa de Pós-Graduação em Química (PPGQ), Departamento de Química (DQ), Instituto de Ciências Exatas e da Terra (ICET), Universidade Federal de Mato Grosso (UFMT), Av. Fernando Corrêa da Costa, 2367, CEP 78060-900 Cuiabá, MT, Brasil.
| |
Collapse
|
31
|
Risk assessment for peri- and post-menopausal women taking food supplements containing isolated isoflavones. EFSA J 2015. [DOI: 10.2903/j.efsa.2015.4246] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
|
32
|
Spagnuolo C, Russo GL, Orhan IE, Habtemariam S, Daglia M, Sureda A, Nabavi SF, Devi KP, Loizzo MR, Tundis R, Nabavi SM. Genistein and cancer: current status, challenges, and future directions. Adv Nutr 2015; 6:408-19. [PMID: 26178025 PMCID: PMC4496735 DOI: 10.3945/an.114.008052] [Citation(s) in RCA: 323] [Impact Index Per Article: 35.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Primary prevention through lifestyle interventions is a cost-effective alternative for preventing a large burden of chronic and degenerative diseases, including cancer, which is one of the leading causes of morbidity and mortality worldwide. In the past decade, epidemiologic and preclinical evidence suggested that polyphenolic phytochemicals present in many plant foods possess chemopreventive properties against several cancer forms. Thus, there has been increasing interest in the potential cancer chemopreventive agents obtained from natural sources, such as polyphenols, that may represent a new, affordable approach to curb the increasing burden of cancer throughout the world. Several epidemiologic studies showed a relation between a soy-rich diet and cancer prevention, which was attributed to the presence of a phenolic compound, genistein, present in soy-based foods. Genistein acts as a chemotherapeutic agent against different types of cancer, mainly by altering apoptosis, the cell cycle, and angiogenesis and inhibiting metastasis. Targeting caspases, B cell lymphoma 2 (Bcl-2)-associated X protein (Bax), Bcl-2, kinesin-like protein 20A (KIF20A), extracellular signal-regulated kinase 1/2 (ERK1/2), nuclear transcription factor κB (NF-κB), mitogen-activated protein kinase (MAPK), inhibitor of NF-κB (IκB), Wingless and integration 1 β-catenin (Wnt/β-catenin), and phosphoinositide 3 kinase/Akt (PI3K/Akt) signaling pathways may act as the molecular mechanisms of the anticancer, therapeutic effects of genistein. Genistein also shows synergistic behavior with well-known anticancer drugs, such as adriamycin, docetaxel, and tamoxifen, suggesting a potential role in combination therapy. This review critically analyzes the available literature on the therapeutic role of genistein on different types of cancer, focusing on its chemical features, plant food sources, bioavailability, and safety.
Collapse
Affiliation(s)
- Carmela Spagnuolo
- Institute of Food Sciences, National Research Council, Avellino, Italy
| | - Gian Luigi Russo
- Institute of Food Sciences, National Research Council, Avellino, Italy;
| | - Ilkay Erdogan Orhan
- Department of Pharmacognosy, Faculty of Pharmacy, Gazi University, Ankara, Turkey
| | - Solomon Habtemariam
- Pharmacognosy Research Laboratories, Medway School of Science, University of Greenwich, Chatham-Maritime, United Kingdom
| | - Maria Daglia
- Department of Drug Sciences, Medicinal Chemistry and Pharmaceutical Technology Section, University of Pavia, Pavia, Italy
| | - Antoni Sureda
- Research Group on Community Nutrition and Oxidative Stress and CIBERobn (Physiopathology of Obesity and Nutrition), University of Balearic Islands, Palma de Mallorca, Spain
| | - Seyed Fazel Nabavi
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Kasi Pandima Devi
- Department of Biotechnology, Alagappa University, Karaikudi, Tamil Nadu, India; and
| | - Monica Rosa Loizzo
- Department of Pharmacy, Health, and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Rosa Tundis
- Department of Pharmacy, Health, and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Seyed Mohammad Nabavi
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran;
| |
Collapse
|
33
|
Tamura T, Ozawa M, Kobayashi S, Watanabe H, Arai S, Mura K. Inhibitory Effect of Oligomeric Polyphenols from Peanut-skin on Sugar Digestion Enzymes and Glucose Transport. FOOD SCIENCE AND TECHNOLOGY RESEARCH 2015. [DOI: 10.3136/fstr.21.111] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Affiliation(s)
- Tomoko Tamura
- Department of Nutritional Science, Faculty of Applied Bioscience, Tokyo University of Agriculture
| | | | - Shoko Kobayashi
- Research Center for Food Safety, Graduate School of Agricultural and Life Sciences, The University of Tokyo
| | - Hirohito Watanabe
- Department of Life Science and Department of Agricultural Chemistry, Meiji University
| | - Soichi Arai
- Department of Nutritional Science, Faculty of Applied Bioscience, Tokyo University of Agriculture
| | - Kiyoshi Mura
- Department of Nutritional Science, Faculty of Applied Bioscience, Tokyo University of Agriculture
| |
Collapse
|
34
|
Bircsak KM, Aleksunes LM. Interaction of Isoflavones with the BCRP/ABCG2 Drug Transporter. Curr Drug Metab 2015; 16:124-40. [PMID: 26179608 PMCID: PMC4713194 DOI: 10.2174/138920021602150713114921] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Revised: 03/03/2015] [Accepted: 04/08/2015] [Indexed: 12/15/2022]
Abstract
This review will provide a comprehensive overview of the interactions between dietary isoflavones and the ATP-binding cassette (ABC) G2 efflux transporter, which is also named the breast cancer resistance protein (BCRP). Expressed in a variety of organs including the liver, kidneys, intestine, and placenta, BCRP mediates the disposition and excretion of numerous endogenous chemicals and xenobiotics. Isoflavones are a class of naturallyoccurring compounds that are found at high concentrations in commonly consumed foods and dietary supplements. A number of isoflavones, including genistein and daidzein and their metabolites, interact with BCRP as substrates, inhibitors, and/or modulators of gene expression. To date, a variety of model systems have been employed to study the ability of isoflavones to serve as substrates and inhibitors of BCRP; these include whole cells, inverted plasma membrane vesicles, in situ organ perfusion, as well as in vivo rodent and sheep models. Evidence suggests that BCRP plays a role in mediating the disposition of isoflavones and in particular, their conjugated forms. Furthermore, as inhibitors, these compounds may aid in reversing multidrug resistance and sensitizing cancer cells to chemotherapeutic drugs. This review will also highlight the consequences of altered BCRP expression and/or function on the pharmacokinetics and toxicity of chemicals following isoflavone exposure.
Collapse
Affiliation(s)
| | - Lauren M Aleksunes
- Dept. of Pharmacology and Toxicology, Rutgers University, 170 Frelinghuysen Rd. Piscataway, NJ 08854, USA.
| |
Collapse
|
35
|
Study on the mechanism of intestinal absorption of epimedins a, B and C in the Caco-2 cell model. Molecules 2014; 19:686-98. [PMID: 24402200 PMCID: PMC6270917 DOI: 10.3390/molecules19010686] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Revised: 12/27/2013] [Accepted: 12/30/2013] [Indexed: 11/17/2022] Open
Abstract
Epimedium spp. is commonly used in Traditional Chinese Medicine. Epimedins A, B, and C are three major bioactive flavonoids found in Epimedium spp. that share similar chemical structures. In this study, the intestinal absorption mechanism of these three compounds was investigated using the Caco-2 cell monolayer model in both the apical-to-basolateral (A-B) and the basolateral-to-apical (B-A) direction. The absorption permeability (PAB) of epimedins A, B, and C were extremely low and increased as the concentration of the epimedins increased from 5 to 20 μM, but, at 40 μM, the PAB values were reduced. Meanwhile, the amount of transported compounds increased in a time-dependent manner. The PAB of epimedins A and C were significantly increased and efflux ratios decreased in the presence of verapamil (an inhibitor of P-glycoprotein) and dipyridamole (an inhibitor of breast cancer resistance protein) while, in the presence of MK571 (an inhibitor of multidrug resistance proteins), the absorption of epimedins A and C did not change significantly, indicating that P-gp and BCRP might be involved in the transport of epimedins A and C. The PAB of epimedin B significantly increased while its secretory permeability (PBA) significantly decreased in the presence of dipyridamole, indicating that BCRP might be involved in the transport of epimedin B. No obvious changes in the transport of epimedin B were observed in the presence of verapamil and MK571. In summary, our results clearly demonstrate, for the first time, that poor bioavailability of these three prenylated flavonoids is the result of poor intrinsic permeability and efflux by apical efflux transporters.
Collapse
|