Bioavailability of Neutraceuticals Nano

Embed Size (px)

Citation preview

  • 7/23/2019 Bioavailability of Neutraceuticals Nano

    1/8

  • 7/23/2019 Bioavailability of Neutraceuticals Nano

    2/8

    R:ConciseReviews

    Nanoencapsulation of nutraceuticals . . .

    option is to encapsulate the functional ingredients using food-

    grade or generally recognized as safe (GRAS) materials that

    can exhibit controlled-release behavior. Materials that can fulfill

    these requirements include polysaccharides of plant (for example,

    pectin, starch, gum Arabic, carrageenan, and so on) or microbial

    (that is, Xanthan gum, dextran) origin, food proteins (for exam-

    ple, soy proteins, casein, gelatin, oat proteins, whey proteins, and

    so on), emulsifiers, such as lecithin, Tweens, Spans, sugar esters,

    monoglycerides, and so on.

    Encapsulation and controlled-release of active food ingredientsare important applications in food and nutrition that can be at-

    tained with nanotechnological approaches. Target delivery of nu-

    trients to cells and cellular compartments was highlighted in the

    Experimental Biology 2009 symposium entitled Nanotechnology

    Research: Applications in Nutritional Sciences as an example of re-

    search area in food and nutrition with nanotech enhancement po-

    tential (Srinivas and others 2009). Although many different delivery

    systems are now available to delivery bioactive components in nu-

    traceuticals and functional foods (McClements and others 2009),

    clearin vitroor in vivoevidences of their biological efficacies are

    still limited. In this review article, we introduce some typical food-

    grade delivery platforms with demonstrated biological efficacy that

    can be used by food and beverage industry to deliver functionalingredients (that is, nutraceuticals). Formulation strategies and ex-

    amples of the biological efficacy of encapsulated nutraceuticals are

    also provided.

    Nanoemulsions-Based Delivery Systems

    Administration of active phytochemical components into thehuman body requires the use of an appropriate vehicle forbringing an effective amount of the active component intact to

    the desired site in the body. The desired site varies and it may be

    the blood stream, organs, and cells, and so on. Majority of phyto-

    chemicals, such as polyphenols and carotenoids, are either poorly

    soluble or lipophilic compounds. It is known that the delivery of

    these phytochemicals is significantly influenced by their physico-

    chemical properties, such as water solubility, partition coefficient,

    lipophilicity, and crystallinity, and so on. Active components that

    poorly dissolve in oil or water pose a problem as to the route for

    their administration, transport, and reaching their targets, result-

    ing in a poor oral bioavailability. Constructing an appropriate ve-

    hicle and the desired efficient formulation possess a challenge to

    dietary supplement researchers. To overcome instability, poor wa-

    ter solubility, and to enhance the bioavailability of nutraceuticals,

    one option to entrap the compound of interest into a food matrix

    is to use nanoemulsion. Nanoemulsions are a class of extremely

    small droplet emulsions that appear to be transparent or translu-

    cent with a bluish coloration. They contain continuous phase, dis-

    persed phase and emulsion stabilizer, the emulsifier or called the

    surfactant. They are usually in the range 50 to 200 nm but much

    smaller than the range (from 1 to 100 m) for conventional emul-

    sions (Solans and others 2005). Since an emulsifier molecule size

    is typically 2 nm long, a micelle, that is, a surfactant molecule ag-

    gregate in water is typically 5 nm or more in diameter. When oil

    phase moleculesenterthe micellar core, theaggregates getswollen,

    sometimes to a large extent, to produce a spherical object whose

    size canreach 100 nm or more. Compared with conventionalmeth-

    ods, such as co-solvent addition, micronizing/milling, spray dry-

    ing, and salt formation, the use of lipid based delivery systems,

    such as micro/nanoemulsions and micelles, offers many advan-

    tages: (i) high kinetic or thermodynamic stability, which provides

    significantly better stability over unstable dispersions, such as con-

    ventional emulsions and suspensions; (ii) either hydrophilic or

    lipophilic phytochemicals can be incorporated into the same na-

    noemulsions; and (iii) because of the small droplet sizes, phyto-

    chemicals can be transported through the cell membranes much

    more easily, resulting in an increased phytochemical concentration

    in plasma and bioavailability.

    It should be pointed out that nanoemulsions are nano-scaled

    emulsions, in which the size of the dispersed oil droplets is below a

    few 100 nanometers. On the other hand, conventional microemul-

    sions, which are thermodynamically stable, optically transparent,

    and have droplet sizes smaller than 100 nm, are often also callednanoemulsions. Thermodynamically stable microemulsion sys-

    tems were reviewed extensively in other articles (Lawrence and

    Rees 2000; Narang and others 2007; Gupta and Moulik 2008). How-

    ever, there are not many food-grade microemulsion systems avail-

    able. In addition, they often require high oil or emulsifier contents,

    or the use of organic cosolvents like ethanol. Therefore, one has to

    balance the benefits brought by the use of bioactives and poten-

    tial side effects (that is, obesity, cardiovascular diseases, and so on)

    caused by the use of high amount of lipids.

    Nanoemulsion preparationandcharacterizationNanoemulsion stability implies a high interfacial tension, and

    thus a considerable surface energy (which is the surface tensiontimes the surface area). Although many nanoemulsions are ther-

    modynamically unstable systems, because of their characteristic

    size, they may possess high kinetic stability. Nanoemulsions do not

    cream (or sediment) because the Brownian motion is larger than

    the small creaming rate induced by gravity. The internal phases of

    nanoemulsions supply an excellent reservoir for phytochemicals

    that need protection and transportation. The nanosizes of emul-

    sions enhance not only stability of the emulsions, but also the

    bioavailability of the encapsulated phytochemicals.

    Generally speaking, nanoemulsions can be prepared through

    either high or low energy emulsifications. Figure 1 shows the

    general high-energy process to prepare kinetically stable na-

    noemulsions. High-energy emulsification methods include highshear homogenization, high-pressure homogenization, microflu-

    idization, ultrasonic homogenization (Solans and others 2005), and

    electrified coaxial liquid jets (Loscertales and others 2002). It should

    be pointed out that, although ultrasonic homogenization and elec-

    trified coaxial liquidjets could also be used to form nanoemulsions,

    they are currently limited in laboratory use and have not been

    used in large batch production (Solans and others 2005; Mason

    and others 2007). High-energy methods are effective in reducing

    droplet sizes, but may not be suitable for some unstable molecules,

    such as proteins or peptides. Alternatively, low-energy emulsifica-

    tion methods, such as phase inversion temperature (PIT) method,

    which uses the changes in solubility of polyoxyethylene-type

    Water + oil + emulsifier

    Stir

    High-speed

    homogenization

    High-pressure

    homogenization

    Nanoemulsion

    Water + oil + emulsifier

    Stir

    High-speed

    homogenization

    High-pressure

    homogenization

    Nanoemulsion

    Figure 1 --- The generalhigh-energy process to preparekinetically stablenanoemulsions.

    Vol. 75, Nr. 1, 2010JOURNAL OF FOOD SCIENCE R51

  • 7/23/2019 Bioavailability of Neutraceuticals Nano

    3/8

    R:ConciseReviews

    Nanoencapsulation of nutraceuticals . . .

    nonionic surfactants with temperature (Shinoda and Saito 1968;

    Rang and Miller 1999), colloidosomes (Dinsmore and others 2002),

    cubosomes (Spicer 2004), and microfluidic channels (Xu and others

    2005), can also be used to prepare nanoemulsions.

    The formulation of nanoemulsions may be different accordingto

    theoils and emulsifiers used. At the same time, bioactives dissolved

    in the dispersed oil phase may also influence the formulation. To

    determine the right emulsion formulation for the specific phyto-

    chemicals, a common practice is to construct pseudoternary phase

    diagrams with large one-phase regions. A phase diagram is usuallysuggested to determine the formulation by interplaying water, oils,

    emulsifiers, and/or cosolvents as well as coemulsifiers of various

    HLB values at different mass ratios. The coemulsifiers are also am-

    phiphilic, which have an affinity for both oil and water phases, and

    partition to certain degrees into the interfacial layer of the emul-

    sions. Compositional variables can also be studied as a function

    of temperature, shear speed, and pressure, but majority of emul-

    sions were studied under ambient conditions with vortexing or

    magnetic stirring. Optimization methods, such as response surface

    methodology, are used to obtain smaller and more uniform (lower

    polydispersity) emulsion systems (Yuan and others 2008). For food

    applications, it is common that 4 or more components are involved

    to form isotropic regions in the pseudo-ternary diagrams, wherea corner typically represents a mixture of 2 or more components

    such as emulsifier/coemulsifier, water/cosolvent/phytochemicals,

    or oil/cosolvent/phytochemicals (Garti 2003). The construction of

    precise phase diagrams can be very time consuming and labor-

    intensive, and the phase boundary is especially different to deter-

    mine. A general strategy is to prepare a series of pseudo binary

    mixtures, and then titrate with the 3rd component. The mixtures

    are evaluated 24 h after each addition. It should be noted that

    many food-grade emulsifiers can only produce limited regions of

    nanoemulsions.

    The structure and physical properties of nanoemulsions can

    be characterized by the combination of a wide variety of

    techniques. For example, the macroscopic properties, such as

    viscosity/viscoelasticity, conductivity, and interfacial tension can

    be characterized by rheometer, conductivity meter, and pendant

    drop tensiometer, respectively (Boonme and others 2006). The size

    andshapeof the emulsion droplets were routinelycharacterized by

    static and dynamic light scattering techniques (McClements 2005).

    Themajordrawback of light scattering techniques is that dilution of

    emulsion samples is usually necessary to reduce multiple scatter-

    ing and interdroplet interactions. The dilution process may mod-

    ify the structure and composition of the pseudoternary phases of

    the nanoemulsions. The structure of the different pseudoternary

    phases can be investigated by small-angle X-ray scattering (SAXS),

    small-angle neutron scattering (SANS), and microscopy like cryo-

    transmission electron microscopy (TEM) (Spicer and others 2001;

    Borne and others 2002; Boonme and others 2006).

    Antioxidant efficiencies of phytochemicals in nanoemulsions

    are often determined by the distributions of antioxidants in the

    oil, water, or oil/water interfacial regions. The distributions of an-

    tioxidants are described by 2 partition constants, one between the

    oil/interfacial regions and the other between aqueous/interfacial

    regions. The 2 partition constants are obtained by fitting 2 sets of

    observed rate constants versus emulsifier concentration data de-termined using electrochemical method for the reaction of the an-

    tioxidant with an arenediazonium ion probe on the basis of the

    well-established pseudophase model (Gunaseelanand others 2004;

    Romsted andZhang 2004). Oil distributions between the oil andin-

    terfacial regions can also be determined by diffusivities measured

    by pulse field gradient spin echo NMR, PGSE-NMR (Nyden and So-

    derman 1995; Soderman and Nyden 1999), where the experiments

    are usually performed by varying the gradient strength along Z-axis

    (Gz) while keeping the gradient pulse duration () andthe diffusion

    delays () constant. The echo intensity,I(GZ), decay as the value of

    Gzincreasing is given by Eq. 1:

    I(Gz) = I(0)exp[D2

    2

    G2

    z( /3)] (1)

    whereDis the self-diffusion coefficient (diffusivity) of the species

    responsible of the spin-echo decay,I(0) is the echo intensity in the

    absence of any pulse gradient and is the gyromagnetic constant

    of the observed nucleus. The applied field gradient strength (G) is

    calibrated before each experiment. PGSE-NMR allows the simulta-

    neous and rapid determination of the self diffusion coefficients of

    many components existing in the nanoemulsions. Figure 2A shows

    the 1H NMRspectra of a typical O/Wnanoemulsion (a), pure oil(b),

    and pure water (c), while Figure 2B shows the processed 2D diffu-

    sion spectra of the same O/W nanoemulsion as Figure 2A.

    Bioavailability of nanoemulsion-encapsulatedphytochemicals

    Bioavailability is defined as a measurement of the extent of

    a therapeutically active component that reaches the systemic

    circulation and is available at the site of action (http://en.

    wikipedia.org/wiki/Bioavailability). It is one of the key pharma-

    cokinetic properties of a phytochemical or drug. Phytochemicals

    with health benefits, such as plant polyphenols (that is, curcumin,

    resveratrol, epigallocatechin gallate, and so on) and carotenoids

    (that is, lycopene, -carotene, lutein, zeaxanthin, and so on),

    have received much attention from the scientific community,

    Figure 2 --- (A). The 1H NMR spectra ofa typical O/W nanoemulsion (a), pureoil (b), and pure water (c); (B). Theprocessed 2D diffusion spectra of thesame O/W nanoemulsion asFigure 2(A).

    R52 JOURNAL OF FOOD SCIENCEVol. 75, Nr. 1, 2010

  • 7/23/2019 Bioavailability of Neutraceuticals Nano

    4/8

    R:ConciseReviews

    Nanoencapsulation of nutraceuticals . . .

    consumers, and food manufacturers because they can be used to

    lower blood pressure, reduce cancer risk factors, regulate diges-

    tive tract system, strengthen immune systems, regulate growth, reg-

    ulate sugar concentration in blood, lower cholesterol levels, and

    serve as antioxidant agents (Hagerman 1992; Wildman 2001). Al-

    though the use of polyphenols in capsules and tablets is abun-

    dant, their biological effects are frequently diminished or even lost

    due to incomplete absorption and first-pass metabolism. Manach

    andothers (2005)reviewedthe oral bioavailability of 97 polyphenol

    compounds and showed that proanthocyanidins, galloylated teacatechins, curcumin, and the anthocyanins are the least absorbed

    pholyphenols. Until now, most of the researches are focused on the

    improved solubilities of oil-soluble lycopene (Spernath and others

    2002), phytosterols (Garti and others 2005), -3 fatty acids (Mc-

    Clements and Decker 2000), coenzyme Q10 (Xia and others 2007;

    Xia and others 2009), and so on. The research on the in vivostud-

    ies of nanoencapsulated polyphenols is still limited. Following is a

    summary of recent advances in the biological efficacies of one of

    the most challenged polyphenols-curcumin.

    Curcumin, chemically named 1,7-bis(4-hydroxy-3-

    methoxyphenyl)-1,6- heptadiene-3,5-dione, is a polyphenol

    Figure 3 --- (A). Chemical structure of curcumin; (B). Chem-ical structure of dibenzoyl methane (DBM).

    extracted from the rhizomes of turmeric (Curcuma longa). The

    chemical structure of curcumin is shown in Figure 3A (Khanna

    1999). Curcumin has very strong yellow color and can be used as

    a natural food color. Curcumin has exhibited antioxidant (Sharma

    1976), antiinflammatory (Srimal and Dhawan 1973), antimicrobial

    (Kim and others 2003), and anticarcinogenic (Miller and others

    2008) activities. Various cell/animal models and human studies

    have demonstrated the preventive or therapeutic functions of cur-

    cumin (Kuttan and others 1985; Hsu and Cheng 2007; Miller and

    others 2008). It inhibits breast, bladder, prostate, or leukemia can-cer in cell culture. Curcumin affects arachidonic acid metabolism,

    inhibits COX and LOX, and induces apoptosis by activating apop-

    tosis signaling (Hong and others 2004). It also blocks many cell

    proliferation signaling pathways, such as MAP kinase pathway,

    AKT pathway, and mTOR pathways (Joe and others 2004; Duvoix

    and others 2005; Howitz and Sinclair 2008). However, low bioavail-

    ability is a general problem for oral administration of curcumin.

    Oral intake of curcumin inhibited chemically induced esophagus,

    fore-stomach, and colon cancer, but had negligible effect on lung

    or breast cancer in mice due to the low circulation concentration in

    blood (Huang and others 1998). Hsu and Cheng (2007) showed in

    phase I clinical trial that curcumin only had efficacy on the tissues

    (colorectum, oral mucosa and skin) exposed to drug directly, buthad no clear effect on other chronic inflammation or cancer.

    Several delivery systems have been studied to increase the

    bioavailability of curcumin, including nanoparticles, liposomes,

    and micelles (Anand and others 2007). Nanoparticle formula-

    tions have been developed and showed increased solubility and

    promisingin vitroresults, but no in vivooral administration re-

    sult is available yet (Bisht and others 2007; Tiyaboonchai and

    others 2007). Liposome encapsulated curcumin has shown en-

    hanced bioavailability and inhibition to pancreatic and colorec-

    tal cancer in vitroand in vivo (intravenous) (Li and others 2005;

    Li and others 2007). Small molecular-weight surfactants, such as

    cetyltrimethylammonium bromide (CTAB) (Iwunze 2004; Wang

    and others 2006; Leung and others 2008), as well as synthetic

    amphiphilic polymers, such as PEO-b-PCL [poly(ethylene glycol)-

    block-poly(caprolactone)] (Ma and others 2008) and methoxy

    poly(ethylene glycol)-palmitate (Sahu and others 2008a), were also

    reported to form polymer micelles to encapsulate curcumin. The

    improved solubilization capacity and loading efficiency were re-

    ported. Curcumin-phospholipid complex demonstrated about 2-

    fold increase in plasma concentration in rat after oral intake (Liu

    and others 2006). Yet, no therapeutic activity has been reported

    with orally administrated curcumin. In addition, organic solvent

    Figure 4 --- Photographic images ofcurcumin nanoemulsions (A),nanodispersion (B), and watersolution (C) (Yu and Huang 2010).

    Vol. 75, Nr. 1, 2010JOURNAL OF FOOD SCIENCE R53

  • 7/23/2019 Bioavailability of Neutraceuticals Nano

    5/8

  • 7/23/2019 Bioavailability of Neutraceuticals Nano

    6/8

    R:ConciseReviews

    Nanoencapsulation of nutraceuticals . . .

    droplets. Compared with conventional micron-sized emulsions,

    nanoemulsions have much smaller droplet sizes, thus have much

    larger surface areas. Such large surface area increases the acces-

    sibility of different lipases and co-lipases and endogenous sur-

    factants, such as bile salt, cholesterol, and phospholipids. They

    may all facilitate the solubilization of lipophilic compound and in-

    crease the absorption rate. The 2ndfactor is the lipid component in

    the formulation. Digested by various lipases in the gastrointestinal

    tract, triglyceride is degraded into diglyceride, monoglyceride, and

    free fatty acids, all of which are amphiphilic and could contributeto solubilization of lipophilic bioactives (Porter and others 2007).

    Other Nutraceutical Delivery Systems

    M icelles are formed by amphiphiles, which have both hy-drophilic and hydrophobic functional groups. When anamphiphile exceeds a certain concentration, the so-called critical

    micelle concentration (CMC) in aqueous solution,a numberof am-

    phiphiles spontaneously assemble to form structured complexes

    called micelles. The hydrophilic region forms the shell of micelles,

    while hydrophobic region forms the cores, where lipophilic bioac-

    tives are trapped. Different from most of the emulsion systems,

    micelles are thermodynamically stable, with no exogenous energy

    input required.Micellar encapsulation system is relative simple compared to

    nanoemulsion: it comprises aqueous solution, amphiphilc com-

    pound (either of low molecular weight or high molecular weight)

    with concentration above CMC, and encapsulated bioactives. Al-

    though the formulation is simpler, the loading process in micellar

    encapsulation could be tricky and affect the encapsulation capac-

    ity and efficiency. At least 4 methods are commonly used in the lit-

    erature to encapsulate bioactives: (i) solvent dialysis: amphiphiles

    and bioactives are dissolved in a common watermiscible solvent

    and then dialyzed against water or aqueous solution to remove the

    solvent; (ii) solvent evaporation: amphiphiles and bioactives are

    dissolved in a common volatile solvent and then brought into an

    aqueous solution. Subsequently, the solvent is removed by evap-

    oration; (iii) co-precipitation: amphiphiles and bioactives are dis-

    solved in a common solvent and then solvent is evaporated to

    form an AB mixture, to which the aqueous solution is added;

    and (iv) emulsification: amphiphiles are dissolved in aqueous so-

    lution, while bioactives are dissolved in water-immiscible volatile

    organic solvent andthen brought into theaqueous solution to form

    emulsion. Subsequently, the organic solvent is evaporated. Among

    these 4 approaches, (i) and (iv) were compared, and it was found

    that emulsification method generated higher encapsulation yields

    (Kwon and others 1997).

    Biopolymer micelles are now a fast growing area in the deliv-

    ery of nutraceuticals or drugs because they prolong their blood cir-

    culation time. Recently, a research in the authors group showed

    hydrophobically modified starch (HMS) was able to form mi-

    celles in aqueous solution (Yu and Huang 2010). Additionally,

    the solubility of curcumin in HMS micelles was increased almost

    1700-fold compared with that in pure water. Furthermore, the

    bioactivity of encapsulated curcumin was tested on an in vitro

    anti-cancer model and it was found that encapsulated curcumin

    demonstrated a significantly higher anti-cancer activity than free

    curcumin (Yu and Huang 2010). It is noteworthy that not all mi-

    cellar formulation could enhance the delivery of encapsulated

    bioactives. Taking curcumin-encapsulation formulations as an ex-

    ample, micelles formed by poly(ethylene oxide)-b-poly(epsilon-

    caprolactone) and methoxy poly(ethylene glycol)-palmitate did not

    increase the delivery of curcumin to tested cancer cells (Ma and

    others 2008; Sahu and others 2008a). On the contrary, curcumin

    Figure 7 --- Nanodispersions of -carotene in the aqueoussolutions of a novel chitosan-based amphiphile, octanoyl-chitosan-polyethylene glycol monomethyl ether.

    encapsulated in HMS micelles and in casein micelles displays

    enhanced bioactivity, which gives rise to a hypothesis that natural

    components may facilitate the cellular uptake of micelles (Sahu and

    others 2008b; Yu and Huang 2010).

    Very recently, a novel chitosan-based amphiphile, octanoyl-

    chitosan-polyethylene glycol monomethyl ether (acylChitoMPEG),

    has been synthesized using both hydrophobic octanoyl and

    hydrophilic polyethylene glycol monomethyl ether (MPEG) substi-

    tutions. The synthesized acylChitoMPEG exhibited good solubil-

    ity in either aqueous solution or common organic solvents such

    as ethanol, acetone, and CHCl3. Cytotoxicity results showed that

    acylChitoMPEG exhibited negligible cytotoxicity even at the con-

    centration as high as 1 mg/mL. The most attractive feature of this

    group of chitosan-based amphiphiles is the ability to tune the HLB

    values by controlling the amount of hydrophobic octanoyl or hy-

    drophilic PEG moieties, andcan be used to encapsulate wide range

    of nutraceuticals or drugs. Figure 7 shows the photographic images

    of-carotene of different concentrations in modified chitosan mi-celles (Huang and others 2009b). Although micelles have been a

    good candidate to encapsulate nutraceuticals and exhibit some en-

    hanced bioactivities, the bioactivities of micelle formulations are

    usually tested in cell culture assay (as shown previously) or by in-

    travenous (i.v.) injection into experimental animals (Jones and Ler-

    oux 1999). Experiments with oral administration of phytochemical

    micelles are still very rare, and more research needs to be done.

    There are many other delivery platforms that can also be used

    to delivery phytochemicals and micronutrients, such as solid

    lipid nanoparticles, multiple emulsions, protein/polysaccharide

    complexes-based multilayer emulsions, soluble complexes, and

    complex coacervates. Their structural design principles have been

    reviewed by McClements and others (2009) and will not be dupli-cated here.

    Conclusions

    Nanoemulsions-based delivery systems have been proved tobe one of the best platforms to enhance the oral bioavail-ability and biological efficacies (that is, antiinflammation, anti-

    cancer, and so on) of different phytochemicals. They are especially

    appealing to food industry because there are many food-grade

    lipids and emulsifiers available. They are simpler and easier to pre-

    pare compared with other lipid-based delivery systems, such as

    multiple emulsions and solid lipid nanoparticles. In addition, hu-

    man body has different lipases ready to digest these lipids, there-

    fore, the potential toxicity of these phytochemical nanoemulsions

    may be minimal. Similarly, polymer micelles also show promise to

    Vol. 75, Nr. 1, 2010JOURNAL OF FOOD SCIENCE R55

  • 7/23/2019 Bioavailability of Neutraceuticals Nano

    7/8

    R:ConciseReviews

    Nanoencapsulation of nutraceuticals . . .

    improve the water dispersibility of many crystalline phytochemi-

    cals, such as -carotene and curcumin, and also show improved

    in vitroanti-cancer activity. During the past decade, many efforts

    have been devoted to the design and development of different nu-

    traceutical delivery systems, and significant progresses have been

    made. A wide variety of delivery systems with different structures

    are now available, and their design principles are quite clear now.

    For the majority of the delivery systems, the in vivobiological ef-

    ficacies of encapsulated phytochemicals remain largely unknown.

    Many questions still remain unanswered. For example, why na-noencapsulated phytochemicals have better oral bioavailability?

    How are the cellular signal transduction pathways different for

    nanoencapsulated phytochemicals compared with the nonencap-

    sulated forms? Are the nanoencapsulated phytochemicals toxic?

    Therefore, more efforts should be devoted to the development of

    novel value-added food-grade or GRAS materials from biomass, as

    well as the understanding of the potential impacts of these nanoen-

    capsulated nutraceuticals to human body and environment to ad-

    dress the public concerns.

    AcknowledgmentThis study was supported by U.S. Dept. of Agriculture National Re-

    search Initiative Program (#2009-35603-05071).

    ReferencesAnand P, Kunnumakkara AB, Newman RA, Aggarwal BB. 2007. Bioavailability of cur-

    cumin: problems and promises. Mol Pharma 4(6):80718.Baeumner A. 2004. Nanosensors identify pathogens in food. Food Technol 58:515.Bisht S, Feldmann G, Soni S, Ravi R, Karikar C, Maitra A, Maitra A. 2007. Polymeric

    nanoparticle-encapsulated curcumin (nanocurcumin): a novel strategy for hu-man cancer therapy. J Nanobiotechnol 5:3.

    Boonme P, Krauel K, Graf A, Rades T, Junyaprasert VB. 2006. Characterization of mi-croemulsion structures in the pseudoternary phase diagram of isopropyl palmi-tate/water/Brij 97:1-butanol. AAPS Pharm Sci Technol 7(2):16.

    Borne J, Nylander T, Khan A. 2002. Effect of lipase on monoolein-based cubic phasedispersion (cubosomes) and vesicles. J Phys Chem B 106:10492500.

    Chen H, Weiss J, Shahidi F. 2006. Nanotechnology in nutraceuticals and functionalfoods. Food Technol 60(3):306.

    Dinsmore AD, Hsu MF, Nikolaides MG, Manuel M, Bausch AR, Weitz DA. 2002. Col-

    loidosomes: self-assembled, selectively permeable capsules composed of colloidalparticles. Science 298:10069.

    Duvoix A, Blasius R, Delhalle S, Schnekenburger M, Morceau F, Henry E, Dicato M,Diederich M. 2005. Chemopreventive and therapeutic effects of curcumin. CancerLett 223(2):18190.

    Garti N. 2003. Microemulsions as microreactors for food applications. J Colloid Inter-face Sci 8:197211.

    Garti N, Spernath A, Aserin A, Lutz R. 2005. Nano-sized self-assemblies of nonionicsurfactants as solubilization reservoirs and microreactors for food systems. SoftMatter 1:20618.

    Gunaseelan K, Bravo-Daz C, Gonzalez-Romero E, Romsted LS. 2004. Determiningpartition constants of polar organic molecules between the oil/interfacial and wa-ter/interfacial regions in emulsions: a combined electrochemical kinetic and spec-trometry method. Langmuir 20:304755.

    Gupta S, Moulik SP. 2008. Biocompatible microemulsions and their prospective usesin drug delivery. J Pharm Sci 97(1):2245.

    Hagerman AE. 1992. Tannin-protein interactions. In: Ho CT, Lee CY, Huang MT,editors. Phenolic compounds in food and their effects on health I. Analysis, oc-currence, and chemistry. ACS symposium series 506. American Chemical Society:

    Washington, DC. p 23647.Hong J, Bose M, Ju J, Ryu J-H, Chen X, Sang S, Lee M-J, Yang CS. 2004. Modulationof arachidonic acid metabolism by curcumin and related -diketone derivatives:effects on cytosolic phospholipase A2, cyclooxygenases and 5-lipoxygenase. Car-cinogenesis 25(9):16719.

    Howitz KT, Sinclair DA. 2008. Xenohormesis: sensing the chemical cues of otherspecies. Cell 133(3):38791.

    Hsu CH, Cheng AL. 2007. Clinical studies with curcumin. Adv Exp Med Biol595:47180.

    Huang M-T, Newmark HL, Frankel K. 1998. Inhibitory effects of curcumin on tumori-genesis in mice. J Cellular Biochem 67(27):2634.

    Huang Q, Given P, Qian M. 2009a. Micro/nano encapsulation of active food ingredi-ents. Vol 1007. Washington, DC: Oxford Univ. Press.

    Huang YP, Yu HL, Guo L, Huang QR. 2009b. Preparation, characterization, and appli-cations of octanoyl- chitosan-polyethylene glycol monomethyl ether amphiphile.The 238th ACS National Meeting; 2009 August 16-20; Washington, DC.

    Iwunze MO.2004. Binding and distribution characteristics of curcumin solubilized inCTAB micelle. J Mol Liq 111(1-3):1615.

    Jiang Y. 2009. Micro- and nano-encapsulation and controlled-release of phenoliccomponents and others food ingredient. [Dphil dissertation]. New Brunswick, N.J.:

    Rutgers Univ.

    Joe B, Vijaykumar M, Lokesh BR. 2004. Biological propertiesof curcumin-cellularandmolecular mechanisms of action. Crit Rev Food Sci Nutr 44(2):97111.

    Jones M-C, Leroux J-C. 1999. Polymeric micelles - a new generation of colloidal drugcarriers. Eur J Pharm Biopharm 48(2):10111.

    Khanna NM. 1999. Turmeric: natures precious gift. Curr Sci 76:13516.Kim MK, Choi GJ, Lee HS. 2003. Fungicidal property ofCurcuma longaL.rhizome-

    derived curcumin against phytopathogenic fungi in a greenhouse. J Agric FoodChem 51(6):157881.

    Kuttan R, Bhanumathy P, Nirmala K, George MC. 1985. Potential anticancer activityof turmeric (Curcuma longa). Cancer Lett 29(2):197202.

    Kwon G, Naito M, YokoyamaM, Okano T, Sakurai Y,Kataoka K.1997.Blockcopolymermicelles for drug delivery: loading and release of doxorubicin. J Control Release48(2-3):195201.

    LawrenceMJ, Rees GD.2000.Microemulsion-based media as novel drug delivery sys-tems. Adv Drug Deliver Rev 45(1):89121.

    LeeJY, Wang XY,Ruengruglikit C, Huang QR. 2007. Nanotechnology in food materialsresearch. In: Aguilera JM, Lillford PJ, editors. Topics in food materials science. New

    York City, N.Y.: Springer. p 12344.Leung MHA, Colangelo H, Kee TW. 2008. Encapsulation of curcumin in cationic mi-

    celles suppresses alkaline hydrolysis. Langm uir 24(11):56725.Li L, Braiteh FS, Kurzrock R. 2005. Liposome-encapsulatied curcumin: invitro and

    invivo effects on proliferation, apoptosis, signaling, and angiogenesis. Cancer104(6):132231.

    Li L, Ahmed B, Mehta K, Kurzrock R. 2007. Liposomal curcumin with and withoutoxaliplatin: effects on cell growth, apoptosis and angiogenesis in colorectal cancer.Mol Cancer Ther 6(4):127682.

    Lin W, Hong J-L, Shen G, Wu RT, Wang Y-W, Huang Q, Heimbach T, Kong A-N.2009. Pharmacokinetics of dietary cancer chemopreventive compound dibenzoyl-methanein therats andimpacts of nanoemulsion andgenetic knockoutof Nrf2 onits disposition. Biopharm Drug Dispos. In press.

    Liu A, Lou H, Zhao L, Fan P. 2006. Validated LC/MS/MS assay for curcumin andtetrahydrocurcumin in rat plasma and application to pharmacokinetic study ofphospholipid complex of curcumin. J Pharm Biomed 40(3):7207.

    Loscertales IG, Barrero A, Guerrero I, Cortijo R, Marquez M, Ganan-Calvo AM. 2002.Micro/nano encapsulation via electrified coaxial liquid jets. Science 295:16958.

    Ma ZS, Haddadi A, Molavi O, Lavasanifar A, Lai R, Samuel J. 2008. Micelles ofpoly(ethylene oxide)-b-poly(epsilon-caprolactone) as vehicles for the solubiliza-tion, stabilization, and controlled delivery of curcumin. J Biomed Mater Res A86(2):30010.

    Manach C, Williamson G, Morand C, Scalbert A, Remesy C. 2005. Bioavailability andbioefficacy of polyphenols in humans. I. Review of 97 bioavailability studies. Am JClin Nutr 81(Suppl):23042.

    Mason TG, Wilking JN, Meleson K, Chang CB, Graves SM. 2006. Nanoemulsions: for-mation, structure, and physical properties. J Phys Condes Matter 18:R63566.

    McClements DJ. 2005. Food emulsions: principles, practices, and techniques. BocaRaton, Fla.: CRC Press.

    McClements DJ, Decker EA. 2000. Lipid oxidation in oil-in-water emulsions: impactof molecular environment on chemical reactions in heterogeneous food systems. JFood Sci 65(8):127082.

    McClements DJ, Decker EA, Park Y, Weiss J. 2009. Structural design principles fordelivery of bioactive components in nutraceuticals and functional foods. Crit RevFood Sci 49:577606.

    Miller M, Chen S, Woodliff J, Kansra S. 2008. Curcumin (diferuloylmethane) inhibitscell proliferatin, induces apoptosis, and decreases hormone level and secretion inpituitary tumor cells. Endocrinology 149(8):415876.

    Moraru CI, Panchapakesan CP, Huang Q, Takhistov P, Liu S, Kokini JL. 2003. Nan-otechnology: a new frontier in food science. Food Technol 57:249.

    Moraru C, Huang QR, Takhistov P, Dogan H, Kokini J. 2009. Food nanotechnology:current developments and future prospects. In: Barbosa-Canovas GV, Mortimer A,Colonna P, Lineback D, Spiess W, Buckle K, editors. Global issues in food scienceand technology. Academic Press. p 36999.

    Narang AS, Delmarre D, Gao D. 2007. Stable drug encapsulation in micelles and mi-croemulsions. Int J Pharm 345(1-2):925.

    Nyden M, Soderman O. 1995. Structures and emulsification failure in the microemul-sion phase in the didodecyldimethylammonium sulfate/hydrocarbon/water sys-tem. A self-diffusion NMR study. Langmuir 11:153745.

    Pegg RB, Shahidi F. 2007. Encapsulation, stabilization, and controlled release of foodingredientsand bioactives.In: RahmanMS, editor. Handbook of foodpreservation.2nd ed. Boca Raton, Fla.: CRC Press. p 50968.

    Porter CJH, Trevaskis NL, Charman WN. 2007. Lipids and lipid-based formulations:optimizing the oral delivery of lipophilic drugs. Nat Rev Drug Discov 6(3):23148.

    Rang MJ, Miller CA. 1999. Spontaneous emulsification of oils containing hydro-carbon, nonionic surfactant, and oleyl alcohol. J Colloid Interface Sci 209:17992.

    Romsted LS, Zhang J. 2004. Determining antioxidant distributions in model food sys-tems: development of a new kinetic method based on the pseudophase model inmicelles and opaque emulsions. Prog Coll Pol Sci 123:1827.

    Sahu A, Bora U, Kasoju N, Goswami P. 2008a. Synthesis of novel biodegradable andself-assembling methoxy poly(ethylene glycol)-palmitatenanocarrier forcurcumindelivery to cancer cells. Acta Biomaterialia 4(6):175261.

    Sahu A, Kasoju N, Bora U. 2008b. Fluorescence study of the curcumincasein micellecomplexation and its application as a drug nanocarrier to cancer cells. Biomacro-molecules 9(10):290512.

    Sharma OP. 1976. Antioxidant activity of curcumin and related com pounds. BiochemPharmacol 25(15):18112.

    Shibamoto T, Kanazawa K, Shahidi F, Ho C-T. 2008. Functional food and health: anoverview. In: Shibamoto T, Kanazawa K, Shahidi F, Ho C-T, editors. ACS sympo-sium series 993. Functional food and health. Washington, DC: Oxford Univ. Press.p 16.

    Shinoda K, Saito H. 1968. The effect of temperature on the phase equilibria and thetypes of dispersions of the ternary system composed of water, cyclohexane, and

    nonionic surfactant. J Colloid Interf Sci 26(1):704.

    R56 JOURNAL OF FOOD SCIENCEVol. 75, Nr. 1, 2010

  • 7/23/2019 Bioavailability of Neutraceuticals Nano

    8/8

    R:ConciseReviews

    Nanoencapsulation of nutraceuticals . . .

    Soderman O, Nyden M. 1999. NMR in microemulsions. NMR translational diffusionstudies of a model microemulsion. Colloid Surface A 158:27380.

    Solans C, Izquierdo P, Nolla J, Azemar N, Garcia-Celma MJ. 2005. Nano-emulsions.Curr Opin Colloid In 10(34):10210.

    Spernath A, Yaghmur A, AserinA, Hoffman RE,GartiN. 2002. Food-grade microemul-sions based on nonionic emulsifiers: media to enhance lycopene solubilization. J

    Agric Food Chem 50:691722.Spicer P. 2004. Bicontinuous cubic liquid crystals nanostrucu red particles. In: Nalwa

    H, editor. Encyclopaedia of nanoscience and nanotechnology. New York: MarcelDekker. p 88192.

    Spicer PT, Hayden KL, Lynch ML, Ofori-Boateng A, Burns JL. 2001. A novel pro-cess for producing cubic liquid crystalline nanoparticles (cubosomes). Langmuir17:574856.

    Srimal RC, Dhawan BN. 1973. Pharmacology of diferuloyl methane (curcumin), anon-steroidal anti-inflammatory agent. J Pharm Pharmacol 25(6):44752.

    Srinivas PR, Philbert M, Vu T, Huang QR, Kokini JL, Saltos E, Chen HD, Peterson C,Friedl K, McDade-Ngutter C, Hubbard V, Starke-Reed P, Miller N, Betz J, Dwyer J,Milner J, RossS. 2009.Nanotechnologyresearch: applications in nutrition sciences.J Nutr. In press.

    Tiyaboonchai W, Tungpradit W, Plianbangchang P. 2007. Formulation and char-acterization of curcuminoids loaded solid lipid nanoparticles. Int J Pharm337(12):299306.

    Vaclavik VA, Christian EW. 2008. Essentials of food science. New York: Springer. 46 p.Wang F, Wu X, Liu SF, Jia Z, Yang JH. 2006. The sensitive fluorimetric method for

    the determination of curcumin using the enhancement of mixed micelle. J Fluo-res 16(1):539.

    Wang XY, Jiang Y, Wang YW, Huang MT, Ho CT , Huang QR. 2008. Enhancing anti-inflammation activity of curcumin through O/W nanoemulsions. Food Chem108(2):41924.

    Weiss J, Takhistov P, McClements DJ. 2006. Functional materials in food nanotech-nology. J Food Sci 71(9):10716.

    Wildman REC. 2001. Handbook of nutraceuticals and funct ional foods. Boca Roton,Fla.: CRC Press.

    Xia SQ, Xu SY, Zhang XM, Zhong F. 2007. Effect of coenzyme Q10 incorpora-tion on the characteristics of nanoliposomes. J Phys Chem B 111(9): 22007.

    Xia SQ, Xu SY, Zhang XM, Zhong F, Wang Z. 2009. Nanoliposomes mediate coenzyme

    Q10 transport and accumulation across humanintestinal Caco-2 cell monolayer.J Agric Food Chem 57(17):798996.

    Xu QY, Nakajima M, Binks BP. 2005. Preparation of particle-stabilized oil-in-water emulsions with the microchannel emulsification method. Colloids Surf A262(13):94100.

    Yu H, Huang QR. 2010. Enhancedin vitroanti-cancer activity of curcumin encapsu-lated in hydrophobically modified starch. Food Chem 119:66974.

    Yuan Y, Gao Y, Mao L, Zhao J. 2008. Optimisation of conditions for the preparation of[beta]-carotene nanoemulsions using response surface methodology. Food Chem107(3):13006.

    Vol. 75, Nr. 1, 2010JOURNAL OF FOOD SCIENCE R57