www.lifeextension.com Open in urlscan Pro
2a02:26f0:6c00::210:ba89  Public Scan

URL: https://www.lifeextension.com/protocols/metabolic-health/metabolic-detoxification
Submission: On December 08 via api from US — Scanned from DE

Form analysis 0 forms found in the DOM

Text Content

12/8/2021

Search Loading
Hello,
 * Account Options
 * LE Dollars
 * AutoShip & Save
 * Order History & Reorder
 * Wish Lists
 * Shipping Addresses
 * Sign Out

0
Sign In/Join
0
Additional Product(s) in Your Cart


Order By Item# Find My Order

View My Cart Check Out
#1 Rated
Catalog/Internet Merchantt

 * Products
 * Health Basics
 * Science & Research
 * About Us
 * Contact Us

 * Vitamins & Supplements
   
   
   NEED HELP SELECTING WHAT'S BEST FOR YOU?
   
   
   Supplement Quizzes
   Shop Vitamins & Supplements
   Featured:
   EssentialsBest SellersNew & Reformulated
   Sale:
   Super Sale
   Shop By Type:
   Amino AcidsArthroMaxBone RestoreBooks & MediaCarnitineCognitexCoQ10Curcumin /
   TurmericDigestive EnzymesFish Oils & OmegasGeroprotect
   Hormones (DHEA)Life Extension MixLetter Vitamins
   BCDEK
   MagnesiumMelatoninMineralsMultivitaminsPre & ProbioticsResveratrol
   Shop By Health Concern:
   Active Lifestyle & FitnessAnti-Aging & LongevityBone HealthBrain
   HealthDigestive HealthEye HealthGeneral Health & WellnessGlucose Management /
   Blood SugarHeart HealthHormone BalanceImmune Support
   Inflammation ManagementJoint HealthKidney, Bladder, Urinary HealthLiver
   Health / DetoxificationMood SupportNerve Health & Comfort SupportSexual
   HealthSleepStress ManagementThyroid / Adrenal
 * Diet & Lifestyle
   
   Active Lifestyle & FitnessEnergy ManagementFood & DrinkMen's HealthPet
   CareProteinWeight ManagementWellness CodeWomen's Health
 * Skin & Personal Care
   
   Skin Care
   Anti-AgingCleansers / ExfoliatorsComplexionMoisturizersSkin ProtectionSkin
   TighteningWrinkles
   Hair & NailsOral Care
 * Lab Testing
   
   
   
   About Lab Testing
   
   
   All Lab Tests A-Z
   Best Selling Lab Tests:
   Chemistry Panel & Complete Blood Count (CBC)Female Basic Hormone PanelFemale
   PanelMale Basic Hormone PanelMale PanelThyroid PanelVitamin DWeight Loss
   Comprehensive Panel
   Lab Test Categories:
   Blood Clotting and CirculationBlood
   SugarBoneCardiovascularCholesterolDigestionHealthy WeightHormonesImmune
   System
   Inflammation ManagementKidney / UrinaryLab Picks from Life
   ExtensionLiverMen's TestingNeurologicalNutritionThyroid / AdrenalWomen's
   Testing
 * On Sale
 * A-Z

 * All Health Basics
   
   
   
   About Health Basics
   
   
   Featured:
   Anti-Aging & LongevityBrain HealthEnergy ManagementGeneral Health &
   WellnessHeart HealthInflammation ManagementWeight Management
   Active Lifestyle & FitnessAnti-Aging & LongevityBone HealthBrain
   HealthDigestive HealthEnergy ManagementEye HealthGeneral Health &
   WellnessGlucose Management / Blood Sugar
   Heart HealthHormonal Health / BalanceImmune SupportInflammation
   ManagementJoint HealthKidney, Bladder, Urinary HealthLiver Health /
   DetoxificationMen's HealthMood Support
   Nerve Health & Comfort SupportSexual HealthSkin CareSleepStress
   ManagementThyroid / AdrenalWomen's HealthWeight Management
 * Wellness Guide
 * Supplement Quizzes
 * eBooks
 * Wellness Workshop
 * Videos & Podcasts

 * About Science & Research
 * Health Protocols
 * Magazine
 * Health News
 * Science News

 * About Life Extension
   
   Welcome to Life ExtensionWhy Choose LE?Our PromiseOur ValuesOur HistoryOur
   People
 * Life Extension Perks
   
   RewardsPremierAutoShip & SaveWellness SpecialistsInnovative Doctors
 * Our Formulations
   
   Scientific ResearchProduct DevelopmentClinical ResearchSafeguarding Your
   Health
 * Media Center
   
   Media ResourcesPress ReleasesLE Health HubAuthors & Editorial Reviewers
 * Health & Wellness Market

 * Customer Service
 * Wellness Specialists
 * Health & Wellness Market


 * Science & Research
 * Health Protocols A-Z
 * Metabolic Detoxification


METABOLIC DETOXIFICATION


METABOLIC DETOXIFICATION

Contributor(s): Dr. Shayna Sandhaus, PhD

Table of Contents

 1. Overview
 2. Introduction
 3. Toxin and Toxicant Exposure
 4. Overview of Xenobiotic Metabolism
 5. The Three Phases of Detoxification
 6. Balance of Phase I and Phase II Reactions
 7. Additional Aspects of the Detoxification Process
 8. Nutrients
 9. References


1 OVERVIEW


SUMMARY AND QUICK FACTS FOR METABOLIC DETOXIFICATION

 * The body has many elegant mechanisms for eliminating potentially harmful
   compounds. Metabolic detoxification refers to the pathways by which the body
   processes and removes harmful compounds.
 * In this protocol, learn about toxic compounds and strategies for minimizing
   exposure to those toxins. Discover natural interventions that, when combined
   with healthy lifestyle practices, can help the body maintain optimal function
   of its detoxification pathway.
 * Sulforaphane is a healthful compound found in cruciferous vegetables like
   broccoli and bok choy. Supplementation with sulforaphane has been shown, in
   several studies, to promote the body’s natural detoxification process.


WHAT IS METABOLIC DETOXIFICATION?

Metabolic detoxification, within the context of this protocol, is the pathway by
which the body processes unwanted chemicals for elimination. The body
metabolizes xenobiotics (foreign chemicals) and unnecessary endobiotics
(endogenously produced chemicals) so they can be excreted.

Excess hormones, environmental toxicants, and prescription drugs are all cleared
via the same enzymatic detoxification systems. Metabolic detoxification is
therefore essential for protecting the body from environmental factors and
maintaining internal homeostasis.

Natural interventions, such as B vitamins and sulforaphane, may help support
detoxification pathways by promoting the activity of essential enzymes.

Note: The body is generally very effective at detoxifying itself. Providing the
body with essential vitamins, minerals, and other nutrients is the best way to
ensure proper detoxification. Many diets and trends that claim to “detox” the
body are pushing the boundaries of scientific plausibility and may even be
harmful.


HOW DOES THE BODY DETOXIFY ITSELF?

The metabolic detoxification process is comprised of three essential steps:

 * Phase I—enzymatic transformation:
    * Purpose is to chemically transform compounds from lipid-soluble into more
      water-soluble
    * Generally carried out by cytochrome P450 (CYP) enzymes

 * Phase II—enzymatic conjugation:
    * Purpose is to further increase water solubility and decrease reactivity of
      phase I products
    * Generally carried out by UDP-glucuronlytransferases (UGTs), glutathione
      S-transferases (GSTs), and sulfotransferases (SULTs)

 * Phase III—transport:
    * Purpose is to excrete water-soluble compounds from the cell
    * Generally carried out by ATP-binding cassette (ABC) transporters

Note: Products of phase I are often more toxic than the original compounds.
Phase II reactions neutralize the products to decrease their toxicity. Several
factors, including dietary influences, smoking and consuming alcohol, advanced
age, and certain diseases may cause phase II enzymes to become overwhelmed
(leading to toxicity, such as seen in acetaminophen overdose).


HOW CAN YOU MINIMIZE EXPOSURE TO TOXINS/TOXICANTS?

 * Choose cleaning products that are free of volatile organic compounds
 * Choose wood, tiles, or other alternatives for flooring instead of carpet
 * Use bisphenol A (BPA)-free or phthalate-free plastic containers; avoid
   warming food in plastic
 * Choose organic produce when possible
 * Wash fruits and vegetables before consuming or remove peel
 * Limit intake of processed foods
 * Cook foods at lower temperatures; avoid charring


WHAT NATURAL INTERVENTIONS MAY HELP THE BODY DETOXIFY?

 * Vitamins. Deficiencies in vitamins A, B2, B3, B9 (folate), C, and E are
   linked with decreased phase I activity and can slow the metabolism of certain
   drugs. B vitamins are also particularly important as cofactors in phase II
   reactions.
 * Minerals. Deficiencies in iron, calcium, copper, zinc, magnesium, and
   selenium have been shown to reduce phase I enzymatic activity.
 * Methionine and cysteine. The reduced glutathione for GST conjugation requires
   adequate dietary sulfur-containing amino acids (methionine or cysteine) for
   activity.
 * Flavonoids. Several flavonoids have demonstrated mild inhibition of multiple
   CYPs in animal models (which can help when phase II enzymes are overloaded);
   these include genistein, diadzein, and equol from soy, and theaflavins from
   black tea.
 * Green tea extracts. Tannins from green tea can increase CYP activity in vivo,
   but also increase phase II activity (GST and UGT).
 * Sulforaphane. Sulforaphane, an isothiocyanate found in broccoli and other
   cruciferous vegetables, is among the most potent natural inducers of phase II
   detoxification enzymes.
 * D-limonene. D-limonene has some chemopreventive activity due to its induction
   of phase I and II enzymes. In rats, D-limonene has been shown to increase
   total CYP activity, intestinal UGT activity, and liver GST and UGT activity.
 * N-acetyl cysteine (NAC). NAC can provide sulfur for glutathione production.
   It is effective at reducing oxidative stress, particularly due to heavy metal
   toxicity.
 * Milk thistle. The milk thistle derivative silymarin promotes detoxification
   by several complementary mechanisms. It can act as an antioxidant to lower
   the liver oxidative stress associated with toxin metabolism, which conserves
   cellular glutathione levels.
 * Artichoke. Artichoke and other plants and vegetables can stimulate bile flow,
   which is essential for toxin excretion.
 * A variety of natural products have been shown in vitro or cell culture to
   directly increase activity of phase II enzymes; these include
   epigallocatechin gallate (EGCG), resveratrol, curcumin and its metabolite
   tetrahydrocurcumin, alpha lipoic acid, alpha tocopherol, lycopene, gingko
   biloba, allyl sulfides from garlic, among others.
 * Other natural interventions that may be helpful for detoxification include
   calcium-D-glucarate, chlorophyllin, probiotics, and derivatives of quercetin.


2 INTRODUCTION

Detoxification ("detox") has broad connotations ranging from the spiritual to
the scientific, and has been used to describe practices and protocols that
embrace both complementary (fasting, colonic cleaning) and conventional
(chelation or antitoxin therapy) schools of medical thought—as well as some that
push the boundaries of scientific plausibility (such as ionic foot
detoxification).

In the context of human biochemistry (and this protocol), detoxification can be
described with much more precision; here it refers to a specific metabolic
pathway, active throughout the human body, that processes unwanted chemicals for
elimination. This pathway (which will be referred to as metabolic
detoxification) involves a series of enzymatic reactions that neutralize and
solubilize toxins, and transport them to secretory organs (like the liver or
kidneys), so that they can be excreted from the body. This type of
detoxification is sometimes called xenobiotic metabolism, because it is the
primary mechanism for ridding the body of xenobiotics (foreign chemicals);
however, detoxification reactions are frequently used to prepare unneeded
endobiotics (endogenously-produced chemicals) for excretion from the body.

Excess hormones, vitamins, inflammatory molecules, and signaling compounds,
amongst others, are typically eliminated from the body by the same enzymatic
detoxification systems that protect the body from environmental toxins, or clear
prescription drugs from circulation. Metabolic detoxification reactions,
therefore, are not only important for protection from the environment, but
central to homeostatic balance in the body.

This protocol describes nutritional approaches for general optimization of
metabolic detoxification; it is designed to provide a foundation for proper
function of this critical system. Specific health concerns may require
supplementary detoxification "intervention" protocols (such as "Heavy Metal
Detoxification" or "Alcohol: Reducing the Risks").


3 TOXIN AND TOXICANT EXPOSURE

Toxins are poisonous compounds produced by living organisms; sometimes the term
"biotoxin" is used to emphasize the biological origin of these compounds.
Man-made chemical compounds with toxic potential are more properly called
toxicants. Toxins and toxicants can exert their detrimental effects on health in
a number of ways. Some broadly act as mutagens or carcinogens (causing DNA
damage or mutations, which can lead to cancer), others can disrupt specific
metabolic pathways (which can lead to dysfunction of particular biological
systems such as the nervous system, liver, or kidneys).

The diet is a major source of toxin exposure. Toxins can find their way into the
diet by several routes, notably contamination by microorganisms, man-made
toxicants (including pesticides, residues from food processing, prescription
drugs and industrial wastes), or less frequently, contamination by toxins from
other "non-food" plant sources.1,2 Some of the toxic heavy metals (lead,
mercury, cadmium, chromium), while not "man-made," have been
released/redistributed into the environment at potentially dangerous levels by
man, and can find their way into the diet as well. Microbial toxins, secreted by
bacteria and fungi, can be ingested along with contaminated or improperly
prepared food.

Even the method of food preparation has the potential for converting
naturally-occurring food constituents into toxins.3 For instance, high
temperatures can convert nitrogen-containing compounds in meats and cereal
products into the potent mutagens benzopyrene and acrylamide, respectively.
Smoked fish and cheeses contain precursors to toxins called N-nitroso compounds
(NOCs), which become mutagenic when metabolized by colonic bacteria.

Outside of the diet, respiratory exposure to volatile organic compounds (VOCs)
is a common risk which has been associated with several adverse health effects,
including kidney damage, immunological problems, hormonal imbalances, blood
disorders, and increased rates of asthma and bronchitis.4

One of the greatest sources of non-dietary toxicant exposure is the air in the
home.5 Building materials (such as floor and wall coverings, particle board,
adhesives, and paints) can "off-gas" releasing several toxicants that can be
detected in humans.6 For example, a toxic benzene derivative commonly used in
disinfectants and deodorizers was detected in 98% of adults in the Environmental
Protection Agency's (EPA) "TEAM" study.7 In another EPA study, three additional
toxic solvents were present in 100% of human tissue samples tested across the
country.8

Newly built or remodeled buildings can have substantial amounts of chemical
"off-gassing," giving rise to what has been called "sick building syndrome."9
Carpet is an especially big offender, potentially releasing several neurotoxins;
in testing of over 400 carpet samples, neurotoxins were present in more than 90%
of the samples, quantitatively sufficient in some samples to cause death in
mice.10 Ironically, shortly after the TEAM report, 71 ill employees evacuated
the new EPA headquarters in Washington DC complaining claiming health problems,
which were eventually attributed to the 27,000 sq. feet of new carpet.11

Carpets also trap environmental toxins; the "Non-Occupational Pesticide Exposure
Study" (NOPES) found an average of 12 pesticide residues per carpet sampled, and
determined that this route of exposure likely provides infants and toddlers with
nearly all of their non-dietary exposure to the notorious pesticides DDT,
aldrin, atrazine, and carbaryl.12


AVOIDING TOXIN/TOXICANT EXPOSURE

While it is not possible to completely eliminate toxin/toxicant exposure from
all sources, there are ways to minimize it:

 * Limit the introduction of VOCs in the home by using VOC-free cleaning
   products, low-VOC paints, and choosing throw rugs instead of new carpeting13;
 * Store food in bisphenol A (BPA)-free or phthalate-free containers, and avoid
   reheating foods in plastic containers;
 * Look for organic produce, which is grown without pesticides, and will contain
   less residue than conventionally-produced fruits and vegetables (although be
   aware that organic produce isn't necessarily "pesticide free")14;
 * Washing fruits or vegetables can decrease some pesticide residue, although it
   is not effective against all pesticide types,15 and commercial fruit and
   vegetable wash solutions may not be any more effective than water alone.16
   Peeling skins off of produce may help to further lower pesticide levels;
 * Limit intake of processed foods. Even ones that are free of synthetic
   preservatives may contain detectable amounts of toxic compounds that were
   introduced (by chemical transformation) during processing. For example,
   numerous toxins are produced by the high temperatures used to manufacture
   some processed food ingredients.17
 * Although the risk of acute toxicity from undercooking meat (food poisoning)
   is likely a greater risk than toxin exposure from overcooking it, there are
   ways to reduce toxin production during meat preparation: avoid direct
   exposure of meat to open flame or hot metal surfaces; cook meat at or below
   250°F via stewing, braising, crockpot cooking (slow food preparation methods
   that utilize liquid); turn meat often during cooking, avoid prolonged cooking
   time at high temperatures, and refrain from consuming charred portions.18


4 OVERVIEW OF XENOBIOTIC METABOLISM

The driving force in the evolution of sophisticated metabolic detoxification
systems was actually fairly straight forward and dependent on the ability of
water to act as a "solvent" to dissolve substances.

Since cellular membranes are primarily lipid based and impermeable to most water
soluble (scientifically: "polar") substances, the transport of water-soluble
compounds into a cell requires specialized transport proteins. By placing the
appropriate transport proteins on the cell membrane, a cell will only allow
desirable water-soluble molecules to enter, and will prevent entry of
water-soluble toxins. This same paradigm also applies when the cell needs to
excrete unwanted water-soluble compounds (like cellular wastes); they exit the
cell by a similar mechanism.

In contrast to water-soluble compounds, the lipid cell membrane presents little
barrier to lipid-soluble compounds, which can freely pass through it.
Potentially damaging lipid-soluble toxins can therefore gain free access to
cellular interiors, and are much more difficult to remove.

The metabolic detoxification systems address this problem by converting
lipid-soluble toxins into inactive water-soluble metabolites. The
"solubilization" of a toxin is accomplished by enzymes which attach (conjugate)
additional water-soluble molecules to the lipid-soluble toxin at specific
attachment points. If the toxin does not contain any of these attachment points,
they are first added by a separate set of enzymes which chemically transform the
toxin to include these molecular "handles." Following the solubilization
reactions, the chemically-modified toxin is transported out of the cell and
excreted.

These three steps or phases of removing undesirable or harmful lipid-soluble
compounds are performed by three sets of cellular proteins or enzymes, called
the phase I (transformation) and phase II (conjugation) enzymes, and the phase
III (transport) proteins.

Phase I, II, and III metabolisms have different biochemical requirements and
respond to different metabolic signals, but must work in unison for proper
removal of unwanted xenobiotics (such as toxins or drugs) or endobiotics (such
as excess hormones). Enzymes of the phase I, II, and III pathways have several
characteristics that make them well suited for their important roles.19 Unlike
most other enzymes, detoxification enzymes; can react with many different
compounds broadening the number of toxins a single enzyme can metabolize; are
more concentrated in areas of the body that are most directly exposed to the
environment (like the liver, intestines, or lungs); are inducible, meaning that
their synthesis can be increased in response to toxin exposure.

The liver is the primary detoxification organ; it filters blood coming directly
from the intestines and prepares toxins for excretion from the body. Significant
amounts of detoxification also occur in the intestine, kidney, lungs, and brain,
with phase I, II, and III reactions occurring throughout the rest of the body to
a lesser degree.


5 THE THREE PHASES OF DETOXIFICATION


PHASE I DETOXIFICATION: ENZYMATIC TRANSFORMATION

Under most circumstances, phase I enzymes begin the detoxification process by
chemically transforming lipid soluble compounds into water soluble compounds in
preparation for phase II detoxification. The bulk of the phase I transformation
reactions are performed by a family of enzymes called the cytochrome P450s
(CYPs).

CYP enzymes are relatively non-specific, each has the potential to recognize and
modify countless different toxins; after all, a mere 57 human CYPs must be able
to detoxify any potential toxin that enters the body.20 However, the cost of
this versatility is speed; CYPs metabolize toxins very slowly compared to other
enzymes. For instance, compare the predominant CYP3A4, which metabolizes 1‒20
molecules per second,21 to superoxide dismutase (SOD), which metabolizes over a
million molecules per second. Major sites of detoxification overcome the slower
speed by producing large amounts of CYPs; CYPs may represent up to 5% of total
liver proteins, and similar large concentrations can be found in the intestines.
CYPs are amongst the most well studied and best characterized detoxification
proteins due to their role in the metabolism of prescription drugs, and to their
role in metabolizing endogenous biochemicals (for example, aromatase, which
transforms testosterone to estradiol, is a CYP).22

Several other enzymes contribute to the phase I process as well, notably the
flavin monooxygenases (FMOs; responsible for the detoxification of nicotine from
cigarette smoke); alcohol and aldehyde dehydrogenases (which metabolize drinking
alcohol); and monoamine oxidases (MAO's; which break down serotonin, dopamine,
and epinephrine in neurons and are targets of several older antidepressant
drugs).23


PHASE II DETOXIFICATION: ENZYMATIC CONJUGATION

Following phase I transformation, the original lipid-soluble toxin has been
converted into a more water-soluble form. However, this reactive intermediate is
still unsuitable for immediate elimination from the cell for a couple of
reasons: 1) phase I reactions are not sufficient to make the toxin water-soluble
enough to complete the entire excretion pathway; and 2) in many cases, products
from the phase I reactions have been rendered more reactive then the original
toxins, which makes them potentially more destructive than they once were. Both
of these shortcomings are addressed by the activities of the phase II enzymes,
which modify phase I products to both increase their solubility and reduce their
toxicity. The activation of the phase II enzymes is responsible for the
anti-mutagenic and anti-carcinogenic properties of the metabolic detoxification
systems; it is widely accepted that phase II enzymes protect against chemical
carcinogenesis, especially during the initiation phase of cancers.24

At the genetic level, the production of most phase II enzymes is controlled by a
protein called nuclear factor erythroid-derived 2 (Nrf2), a master regulator of
antioxidant response.25 Under normal cellular conditions, Nrf2 resides in the
cytoplasm (the liquid inside cells within which the cells components are
contained) of the cell in an inactive state.26 However, the presence of
oxidative stress (triggered by metabolism of toxins by CYPs) activates Nrf2,
allowing it to travel to the cell nucleus.27 In the cell nucleus, Nrf2 turns on
the genes of many antioxidant proteins, including the phase II enzymes.28 In
this way, Nrf2 "senses" oxidative stress or the presence of toxins in the cell,
and allows the cell to mount an appropriate response. Nrf2 regulates the
activity of genes involved in the synthesis and activation of important
detoxification molecules including glutathione and superoxide dismutase (SOD).
It also plays an important role in initiating heavy metal detoxification, and
the recycling of CoQ10, a potent antioxidant.29-31

Certain dietary constituents (including sulforaphane from broccoli and
xanthohumol from hops) may also directly activate Nrf2 and stimulate antioxidant
enzyme activity; this may partially explain their beneficial effects on
detoxification.32

There are several families of phase II enzymes that differ significantly in
their activities and biochemistry. In several cases, phase II enzymes exhibit
redundancy—a particular xenobiotic or endobiotic can be detoxified by more than
one phase II enzyme.

UDP-glucuronosyltransferases. UDP-glucuronosyltransferases (UGTs) catalyze
glucuronidation reactions, the attachment of glucuronic acid to toxins to render
them less reactive and more water-soluble. There are several different UGTs that
are distributed throughout the body, with the liver being the major location. In
humans, many xenobiotics, environmental toxicants, and 40‒70% of clinical drugs
are metabolized by UGTs.33 The plasticizer bisphenol A34 and benzopyrene (from
cooked meats)35 are two notable examples of UGT substrates (a substrate is a
molecule upon which an enzyme acts). Intestinal UGTs may affect oral
bioavailability of several drugs and dietary supplements, and may be responsible
for chemoprevention in this tissue.36

Glutathione S-transferases. Glutathione S-transferases (GSTs) catalyze the
transfer of glutathione (a significant cellular antioxidant) to phase I
products. GSTs play a major role in the metabolism of several endobiotics,
including steroids, thyroid hormone, fat-soluble vitamins, bile acids, bilirubin
and prostaglandins.37 GSTs can also function as antioxidant enzymes, detoxifying
free radicals38 and oxidized lipids or DNA.39 GSTs are soluble enzymes that are
ubiquitous in nature and in humans, forming about 4% of the soluble protein in
the human liver and present in several other tissues (including brain, heart,
lung, intestines, kidney, pancreas, lens, skeletal muscle, prostate, spleen and
testes).40,41 Products of GST conjugation can be excreted via bile, or can
travel to the kidneys where they are further processed and eliminated in urine.

Sulfotransferases. Sulfotransferases (SULTs) attach sulfates from a sulfur donor
to endo- or xenobiotic acceptor molecules. This reaction is important both in
detoxification reactions, as well as normal biosynthesis (the addition of
sulfate to chondroitin and heparin, for example, is catalyzed by specific
SULTs).42 SULTs play a major role in drug and xenobiotic detoxification, and the
metabolism of several endogenous molecules (including steroids, thyroid and
adrenal hormones, serotonin, retinol, ascorbate and vitamin D).43 SULTs in the
placenta, uterus, and prostate are thought to play a role in the regulation of
androgen levels.44 In contrast to other phase II enzymes, SULTs can convert a
number of procarcinogens (such as heterocyclic amines from cooked meats) into
highly reactive intermediates which may act as chemical carcinogens and
mutagens.45

While the UGTs, GSTs, and SULTs catalyze the bulk of human detoxification
reactions, several other phase II enzymes contribute to the process to a lesser,
but still important extent, including:

 * Methyltransferase enzymes. Methyltransferase enzymes catalyze methylation
   reactions using S-adenosyl-L-methionine (SAMe) as a substrate. Catechol
   O-methyltransferase (COMT) is a major pathway for eliminating excess
   catecholamine neurotransmitters (such as adrenaline or dopamine). Methylation
   reactions are one of the few phase II reactions that decrease water
   solubility46;
 * Arylamine N-acetyltransferases (NATs). NATs detoxify carcinogenic aromatic
   amines and heterocyclic amines47;
 * Amino acid conjugating enzymes. Acyl-CoA synthetase and acyl-CoA amino acid
   N-acyltransferases attach amino acids (most commonly glycine or glutamine) to
   xenobiotics. The food preservative benzoic acid is one example of a toxin
   metabolized by amino acid conjugation.48


PHASE III DETOXIFICATION: TRANSPORT

Phase III transporters are present in many tissues, including the liver,
intestines, kidneys, and brain, where they can provide a barrier against
xenobiotic entry, or a mechanism for actively moving xenobiotics and endobiotics
in and out of cells.49 Since water-soluble compounds require specific
transporters to move in and out of cells, phase III transporters are necessary
to excrete the newly formed phase II products out of the cell. Phase III
transporters belong to a family of proteins called the ABC transporters (for
ATP-binding cassette),50 because they require chemical energy, in the form of
ATP, to actively pump toxins through the cell membrane and out of the cell.51
They are sometimes called the multidrug resistance proteins (MRPs), because
drug-resistant cancer cells use them as protection against chemotherapy drugs.52

In the liver, phase III transporters move glutathione, sulfate, and glucuronide
conjugates out of cells into the bile for elimination. In the kidney and
intestine, phase III transporters can remove xenobiotics from the blood for
excretion from the body.53


6 BALANCE OF PHASE I AND PHASE II REACTIONS

The products of phase I metabolism are potentially more toxic than the original
molecules, which does not present a problem if the phase II enzymes are
functioning at a rate to rapidly neutralize the phase I products as they are
formed. This, however, is not always the case. Factors which increase the ratio
of phase I to phase II activity can upset this delicate balance, producing
harmful metabolites faster than they can be detoxified, and increasing the risk
of cellular damage. Some of the factors include: diet (some foods and
supplements increase phase I enzyme activity), smoking and alcohol consumption
(both induce phase I), age (which can decrease phase II UGT, GST, and SULT
activity), sex (premenopausal women show 30‒40% more phase I CYP3A4 activity
than men or postmenopausal women), disease, and genetics.54

An illustrative (and unfortunately common) example of the consequences of phase
I/phase II imbalance is toxicity caused by overdose of the analgesic
acetaminophen (paracetamol)—the active ingredient in Tylenol. Acetaminophen
toxicity is the most common cause of liver failure in the United States.55 With
a normal therapeutic dose of acetaminophen, the drug is predominantly detoxified
by the phase II UGT and SULT enzymes. A small amount of the drug is detoxified
by a third mechanism: it is first transformed into the toxic metabolite NAPQI
(N-acetyl-p-benzoquinoneimine) by phase I CYP enzymes; and this intermediate is
detoxified by conjugation with glutathione using the phase II enzyme GST.

During acetaminophen overdose, the UGT and SULT enzymes become quickly
overwhelmed. Proportionally more of the drug undergoes the third detoxification
mechanism (transformation to NAPQI and conjugation by GST). Eventually, activity
of the phase II GST enzyme slows as glutathione stores become depleted,56 and
NAPQI is produced faster than it can be detoxified. Rising levels of NAPQI in
the liver cause widespread damage, including lipid peroxidation, inactivation of
cellular proteins, and disruption of DNA metabolism.57 Treatment for
acetaminophen overdose involves the timely replenishment of glutathione stores
through administration of the precursor amino acids for glutathione synthesis
(most commonly N-acetyl cysteine).58


7 ADDITIONAL ASPECTS OF THE DETOXIFICATION PROCESS

Several other mechanisms work in concert with the phase I, II, and III enzyme
systems to improve their efficiency or extend their functionality. While not
officially characterized as part of xenobiotic metabolism, they are nonetheless
important for reducing or mitigating toxin exposure.


BILE SECRETION

Bile secretion is a critical digestive process for the absorption of dietary
fats and fat-soluble nutrients, but also functions as the major mechanism for
moving conjugated toxins out of the liver and into the intestines, where they
can be eliminated.


ANTIOXIDATION

Antioxidation is a necessary protective measure against the harsh phase I
oxidation reactions, which frequently produce free-radical byproducts. The
production of antioxidant enzymes, many of which are under the same genetic
regulation (by Nrf2) as the phase II enzymes, is important for minimizing this
free-radical damage.


HEAVY METAL TOXICITY

Heavy metal toxicity can lead to oxidative damage by direct generation of free
radical species and depletion of antioxidant reserves.59 Mercury, arsenic, and
lead, for example, effectively inactivate the glutathione molecule so it is
unavailable as an antioxidant or as a substrate for xenobiotic detoxification60;
lead can also reduce the activity of the enzymes of that recycle glutathione.61
One method for heavy metal removal is their chelation by the cellular proteins
metallothioneins (MTs), which have a high capacity to bind various reactive
metal ions, such as zinc, cadmium, mercury, copper, lead, nickel, cobalt, iron,
gold, and silver.62 One molecule of MT can bind 7–9 zinc or cadmium ions (or any
combination of these two), up to 12 copper ions, and up to 18 mercury ions.63
Cellular stress (particularly oxidative stress), turns up MT production, which,
like the phase II enzymes, is stimulated by the activity of Nrf2.64


PREVENTION OF ABSORPTION

Prevention of absorption through trapping of potential toxins (such as surface
adhesion to another molecule in the gut, like activated charcoal or kaolin
clay)65 is an effective means of mitigating exposure; this mechanism has the
requirement of some dietary adsorbent to be taken while the toxin is in transit
in the GI tract. Uptake of potential toxins and their detoxification by
beneficial colonic microflora could have a similar effect.


WHAT YOU NEED TO KNOW ABOUT METABOLIC DETOXIFICATION

 * Detoxification is the metabolic process of removing unwanted lipid-soluble
   compounds from the body.
 * These "unwanted" compounds can be foreign (such as an environmental
   toxicants) or endogenous (toxins; such as excess hormone) in nature.
 * Detoxification reactions occur throughout the body, with the liver being the
   predominant detoxifying organ.
 * Detoxification reactions follow three steps or "phases" that have the
   ultimate goal of converting the toxin into an inert, water-soluble form for
   excretion:

Phase I reactions transform the toxin into a chemical form that can be
metabolized by the phase II enzymes. Phase I reactions are performed primarily
by the cytochrome P450 enzymes.

Phase II reactions conjugate (attach) the toxin to other water-soluble
substances to increase its solubility. Each of the different types of phase II
enzymes catalyzes a different type of conjugation reaction.

 * UDP-glucuronosyltransferases (UGTs) catalyze the glucuronidation of most
   clinical drugs, and several environmental toxins
 * Glutathione-S-transferases (GSTs) conjugate toxins with the antioxidant
   glutathione; they can also directly detoxify free radicals
 * Sulfotransferases (SULTs) catalyze sulfonation reactions; they may also be
   important for controlling sex hormone levels

Other types of phase II reactions that are used less frequently include
methylation and amino acid conjugation reactions.

Phase III detoxification involves the transport of the transformed, conjugated
toxin into or out of cells. Different phase III transport proteins work in
concert to shuttle toxins from different parts of the body into bile or urine
for excretion.

Following detoxification reactions, the toxins are removed from the body by
excretion:

A) Products of liver detoxification often leave the body by being secreted into
the intestines in bile, but can sometimes be transported into the bloodstream
for processing by the kidneys.

B) The cells that line the intestines can detoxify toxins as they are absorbed,
and release them back into the intestinal lumen.

C) The kidneys can filter and further process toxins from circulation, excreting
them from the body as urine.


8 NUTRIENTS

Given the sheer number of diverse enzymes and transport proteins involved in
metabolic detoxification and its related pathways, it is no surprise that
detoxification depends on, and is sensitive to, a large number of dietary
factors.

Macronutrient and micronutrient intake influences phase I and II systems.
Protein deficiency decreases CYP metabolism, while high protein diets increase
it.66 The opposite effects are observed for carbohydrates; the effects of lipids
on CYP metabolism are unclear. Efficient phase I reactions require sufficiency
in several micronutrients; deficiencies in vitamins A, B2 and B3, folate, C, E,
iron, calcium, copper, zinc, magnesium, selenium have all been shown to decrease
the activities of one or more phase I enzymes, or slow the transformation of
specific drugs.67

The diverse set of phase II enzymes require an equally diverse set of essential
nutrients, especially B vitamins, as cofactors.

The reduced glutathione for GST conjugation depends on adequate dietary
sulfur-containing amino acids (methionine or cysteine), vitamin B6 for the
conversion of methionine to cysteine, as well vitamins B2 and B3 for the
activity of glutathione reductase, which recycles oxidized glutathione.

The methylation reactions use SAMe as a substrate; which, in turn, is
synthesized through folate- and vitamin B12-dependent enzymatic reactions.

The conjugation reactions of the NAT's and amino acid acyltransferases use the
cofactor acetyl-coenzyme A (acetyl-CoA), which is synthesized from vitamin B5,
using enzymes that themselves depend on multiple B vitamins.

Several phase II reactions require the energy molecule ATP in some fashion. For
example, the chemical cofactors for the phase II methylation, sulfonation,
glucuronidation, and glutathione conjugation reactions are all made using ATP;
these ATP mediated reactions are magnesium-dependent.

Flavonoids have been extensively studied in vitro and in animal models for their
ability to lower the activity of CYPs, and increase phase II enzyme activities
(except for SULTs, which they tend to inhibit).68 The inhibition of CYP activity
by naringenin (the principle flavonoid in grapefruit) has been well documented
in humans69; hence the recommendation to avoid grapefruit when taking
prescription drugs. Other flavonoids that have demonstrated mild inhibition of
multiple CYPs in animal models include genistein, diadzein, and equol from
soy,70,71 and theaflavins from black tea.72

Green tea extracts and the quercetin derivatives isoquercetin and rutin are an
exception to most other flavonoids; green tea tannins can increase CYP activity
in vivo,73 but also increase phase II activity (GST and UGT). Similarly, the
quercetin derivatives were demonstrated to increase intestinal and liver CYPs in
rats; quercetin had no effect on CYPs in this experiment.74

Nrf2 activators. A wide variety of dietary components have been shown in vitro
or cell culture to activate Nrf2 and directly increase activity of phase II
enzymes; these include epigallocatechin gallate (EGCG),75 resveratrol,76
curcumin77 and its metabolite tetrahydrocurcumin, which has greater phase II
activity,78 cinnamaldehyde,79 caffeic acid phenyethyl ester, alpha lipoic
acid,80 alpha tocopherol,81 lycopene,82 apple polyphenols (chlorogenic acid and
phloridzin),83 gingko biloba,84 chalcone,85 capsaicin,86 hydroxytyrosol from
olives,87 allyl sulfides from garlic,88 chlorophyllin,89 and xanthohumols from
hops.90 The beneficial effects of these phytochemicals have been demonstrated in
numerous animal and human studies, particularly their chemopreventative and
antioxidant abilities; these effects may be explained by their indirect
stimulation of antioxidant enzyme production and phase II detoxification through
Nrf2 signalling.91

Isothiocyanates derived from glucosinolates are reactive sulfur compounds with
potent chemopreventive properties; the prototypical member is sulforaphane, a
constituent of broccoli that is the subject of several human cancer trials.

Isothiocyanates such as sulforaphane and indoles such as indole-3-carbinol (I3C)
are among the most potent natural inducers of phase II detoxification enzymes.92
Sulforaphane and a derivative of I3C both directly activate Nrf2,93 which
increases the production of several protective enzymes, including GSTs, UGTs,
glutamate-cysteine ligase (which synthesizes glutathione), and NQO1.94 I3C
derivatives are also strong inducers of many phase I and II enzymes, and thus
are among the most well studied phytochemicals for detoxification, as well as
cancer prevention.95-99

Compounds from the Japanese horseradish Wasabi japonica,100,101 and benzyl
isothiocyanate (BITC)102 from cruciferous vegetables similarly stimulate phase
II enzyme activity via Nrf2 activation. Both sulforaphane and HITC also lower
CYP activity.103

Sulfur constituents from garlic are inhibitors of various CYPs,104 andinduce GST
and NQO1 activity in gastrointestinal tissues in rats.105 By activating Nrf2,
components in garlic were able to reverse the depletion of antioxidant enzymes
caused by a toxic metal compound in the livers of laboratory rats.106

D-limonene (from citrus oil) has been investigated for anticancer activity in
uncontrolled human trials and animal studies with some success107; part of this
chemopreventive activity is due to the induction of phase I and phase II
enzymes. In rats, D-limonene has been shown to increase total CYP activity,108
intestinal UGT activity109 and liver GST and UGT activity.110,111

Calcium D-glucarate is present in many fruits and vegetables, and can be
produced in small amounts in humans.112 When activated in the gut, it functions
as an inhibitor of beta-glucuronidase, an enzyme produced by colonic bacteria
and intestinal cells. In the intestines, beta-glucuronidase removes
(deconjugates) glucuronic acid from neutralized toxins—essentially reversing the
reaction catalyzed by UGTs. Deconjugation reverts the toxin to its previous
dangerous form, and allows it to be reabsorbed. Elevated beta-glucuronidase
activity has been associated with increased cancer risk.113

Chlorophyllin is a chlorophyll derivative114 that inhibits CYP activity,115 and
stimulates GST activity in cell culture and rodent models.116 The unique
chemical structures of chlorophyllin and chlorophyll enable them to bind and
"trap" toxins in the gut preventing their absorption. In animal models,
chlorophyllin and chlorophyll lower the bioavailability and accelerate the
excretion of several environmental carcinogens.117-119 Toxin trapping may partly
explain the results of a human trial of residents of Qidong, China, an area with
a high incidence of liver cancer due to exposure to aflatoxin (a toxin produced
by species of the fungus Aspergillus). Among the 180 people who took 100 mg of
chlorophyllin three times daily, urinary levels of DNA-aflatoxin conjugates (a
marker for DNA mutation) went down 55% compared to untreated people.120

Probiotics. Certain strains of probiotic bacteria may minimize toxin exposure by
trapping and metabolizing xenobiotics or heavy metals.121 Examples include the
detoxification of aflatoxin and patulin (two toxins produced by Aspergillus, a
type of mold),122 the metabolism of heterocyclic amines and
dimethylhydrazine,123 and the binding of lead and cadmium.124 Additionally, the
production of the short chain fatty acid butyrate by lactic acid bacteria (from
the fermentation of dietary fiber) has been shown to stimulate GST production in
intestinal cell culture; this may also contribute to some of the
anticarcinogenic properties of dietary fiber.125

N-acetyl cysteine can provide an alternative source of sulfur for glutathione
production. It is a free radical scavenger on its own, effective at reducing
oxidative stress, particularly due to heavy metal toxicity.126 Because it can
directly replenish glutathione stores, NAC is more effective than methionine at
preventing liver damage,127 and is the current treatment for acetaminophen
toxicity. It is an effective treatment for acute liver failure due to
non-acetaminophen drug toxicity as well.128

Milk thistle (Silybum marianum), the most well-researched plant in the treatment
of liver disease,129 contains a mixture of several related polyphenolic
compounds called silymarin. Silymarin promotes detoxification by several
complementary mechanisms. The antioxidant capacity of silymarin can lower the
liver oxidative stress associated with toxin metabolism, particularly lipid
peroxidation,130 which has the effect of conserving cellular glutathione
levels.131 Like NAC, silymarin can protect against acetaminophen toxicity
(possibly by the similar mechanism of preserving glutathione levels). Silymarin,
however, may be a more effective antidote than NAC for acetaminophen toxicity if
the treatment is delayed (in an animal model, it was effective when administered
up to 24 hours after overdose).132

Phase III transporters, while important for removing toxins from healthy cells,
can also decrease the effectiveness of pharmaceutical therapies by increasing
their clearance. This can be especially problematic with chemotherapy drugs, to
which phase III transporters enable cancer cells to become resistant. Therefore,
stimulation of phase III activity may not always be desirable.

Dietary factors can have differing effects on phase III transporters. For
example, apple polyphenols133 and sulforaphane (at levels equivalent to about
two servings of broccoli)134 both stimulate the activity of the phase III
proteins. In contrast, the curcumin metabolite tetrahydrocurcumin decreases the
activity of the phase III transporters in human cervical carcinoma and breast
cancer cell lines.135 Resveratrol decreases phase III protein synthesis which
prevented acute myeloid leukemia cells from becoming resistant to the
chemotherapy drug doxorubicin in cell culture.136 Silibinin, the chief
constituent of milk thistle,137 is also a phase III inhibitor, both in vitro and
in vivo.138

Bile flow. As a major carrier of toxins from the body, proper bile flow is a
critical final step in the metabolic detoxification process. Impairment of bile
flow (cholestasis), resulting from dysfunction within the liver or blockage of
the bile duct, can result in the buildup of liver toxins and liver injury.
Cholestasis can also be the result of the detoxification process itself; there
is increasing evidence that the detoxification and excretion of clinical drugs
into the bile can produce cholestatic liver disease.139 Artichoke has been used
for centuries in folk medicine as a liver protectant and to stimulate bile flow
(choleresis), and is the best-studied herbal choleretic agent.

Artichoke contains several antioxidants that can protect against oxidative liver
damage, as well as caffeoylquinic acids, which have been shown to stimulate bile
flow in animal models.140 Caffeoylquinic acids may also be responsible for the
choleretic properties of yarrow,141,142 fennel,143 and dandelion.144
Andrographis, garlic, cumin, ginger, ajowan (carom seed), and curry and mustard
leaf have also been shown to stimulate bile flow or bile acid production in
rodent models.145-148


DISCLAIMER AND SAFETY INFORMATION

This information (and any accompanying material) is not intended to replace the
attention or advice of a physician or other qualified health care professional.
Anyone who wishes to embark on any dietary, drug, exercise, or other lifestyle
change intended to prevent or treat a specific disease or condition should first
consult with and seek clearance from a physician or other qualified health care
professional. Pregnant women in particular should seek the advice of a physician
before using any protocol listed on this website. The protocols described on
this website are for adults only, unless otherwise specified. Product labels may
contain important safety information and the most recent product information
provided by the product manufacturers should be carefully reviewed prior to use
to verify the dose, administration, and contraindications. National, state, and
local laws may vary regarding the use and application of many of the therapies
discussed. The reader assumes the risk of any injuries. The authors and
publishers, their affiliates and assigns are not liable for any injury and/or
damage to persons arising from this protocol and expressly disclaim
responsibility for any adverse effects resulting from the use of the information
contained herein.

The protocols raise many issues that are subject to change as new data emerge.
None of our suggested protocol regimens can guarantee health benefits. Life
Extension has not performed independent verification of the data contained in
the referenced materials, and expressly disclaims responsibility for any error
in the literature.


REFERENCES

 1.   Prakash AS, Pereira TN, Reilly PE, Seawright AA. Pyrrolizidine alkaloids
      in human diet. Mutat Res 1999;443(1-2):53-67
 2.   Borchers A, Teuber SS, Keen CL, Gershwin M. Food safety. Clin Rev Allergy
      Immunol 2010;39 (2) : 95-141
 3.   Ferguson LR, Philpott M. Nutrition and mutagenesis. Annu Rev Nutr
      2008;28:313-29
 4.   Crinnion MJ. Environmental Medicine, Part 2 – Health Effects of and
      Protection from Ubiquitous Airborne Solvent Exposure. Altern Med Rev
      2000;5 (2) : 133-143
 5.   Nielsen GD, Larsen ST, Olsen O, et al. Do indoor chemicals promote
      development of airway allergy? Indoor Air 2007;17 (3) : 236-55
 6.   Wallace LA, Pellizzari ED, Hartwell TD, et al. Personal exposure,
      indoor-outdoor relation- ships, and breath levels of toxic air pollutants
      measured for 355 persons in New Jersey. EPA 0589.
 7.   Hill RH Jr, Ashley DL, Head SL, et al. p- Dichlorobenzene exposure among
      1,000 adults in the United States. Arch Environ Health 1995;50:277-280.
 8.   Broad scan analysis of the FY82 national human adipose tissue survey
      specimens. EPA Office of Toxic Substances. EPA 560/5-86- 035.
 9.   Ruhl RA, Chang CC, Halpern GM, Gershwin ME. The sick building syndrome.
      II. Assess- ment and regulation of indoor air quality. J Asthma
      1993;30:297-308.
 10.  Duehring C. Carpet, EPA stalls and industry hedges while consumers remain
      at risk. Informed Consent 1993;1:6-32
 11.  Crinnion MJ. Environmental Medicine, Part 2 – Health Effects of and
      Protection from Ubiquitous Airborne Solvent Exposure. Altern Med Rev
      2000;5 (2) : 133-143
 12.  Whitemore RW, Immerman FW, Camann DE, et al. Non-occupational exposures to
      pesticides for residents of two U.S. cities. Arch Environ Contam Toxicol
      1994;26:47-59.
 13.  Crinnion MJ. Environmental Medicine, Part 2 – Health Effects of and
      Protection from Ubiquitous Airborne Solvent Exposure. Altern Med Rev
      2000;5 (2) : 133-143
 14.  Crinnion MJ. Environmental Medicine, Part 4: Pesticides – Biologically
      Persistent and Ubiquitous Toxins. Altern Med Rev 2000;5 (5) : 432-447
 15.  Štěpán R. , Tichá J. , Hajšlová J. , Kovalczu, T. and Kocourek V. Baby
      food production chain: pesticide residues in fresh apples and products.
      Food Addit Contam 2005; 22 (12):1231-42
 16.  Krieger RI, Brutsche-Keiper P, Crosby HR, Krieger AD. Reduction of
      pesticide residues of fruit using water only or Plus Fit Fruit and
      Vegetable Wash. Bull Environ Contam Toxicol 2003;70(2): 213-8
 17.  Borchers A, Teuber SS, Keen CL, Gershwin M. Food safety. Clin Rev Allergy
      Immunol 2010;39 (2) : 95-141
 18.  Knize MG, Felton JS. Formation and human risk of carcinogenic heterocyclic
      amines formed from natural precursors in meat. Nutrition Reviews 2005;
      63(5):158–165.
 19.  Jakoby WB and Ziegler DM. The enzymes of detoxication. J Biol Chem
      1990;265(34):20715-8
 20.  Redlich G, Zanger UM, Riedmaier S, et al. Distinction between human
      cytochrome P450 (CYP) isoforms and identification of new phosphorylation
      sites by mass spectrometry. J Proteome Res 2008; 7 (11):4678-88
 21.  Dai D, Tang J, Rose R, et al. Identification of variants of CYP3A4 and
      characterization of their abilities to metabolize testosterone and
      chlorpyrifos. J Pharmacol Exp Ther 2001;299 (3):825-31
 22.  Lardone MC, Castillo P, et al. P450-aromatase activity and expression in
      human testicular tissues with severe spermatogenic failure. Int J Androl.
      2010 Aug 1;33(4):650-60. Epub 2009 Nov 3.
 23.  Johnson R. Physiology of the gastrointestinal tract, Volume 1 - Page 1827.
      (2006) : 2000
 24.  Nakamura Y, Miyamoto M, Murakami A et al. A phase II detoxification enzyme
      inducer from lemongrass: identification of citral and involvement of
      electrophilic reaction in the enzyme induction* 1. Biochemical and …
      (2003)
 25.  Moi P, Chan K, Asunis I, Cao A, Kan YW. Isolation of NF-E2-related factor
      2 (Nrf2), a NF-E2-like basic leucine zipper transcriptional activator that
      binds to the tandem NF-E2/AP1 repeat of the beta-globin locus control
      region. Proc Natl Acad Sci USA 1994;91 (21) : 9926-30
 26.  Kobayashi A, Kang MI, Okawa H, et al. Oxidative stress sensor Keap1
      functions as an adaptor for Cul3-based E3 ligase to regulate proteasomal
      degradation of Nrf2. Mol Cell Biol 2004;24 (16) : 7130-9
 27.  Motohashi H and Yamamoto M. Nrf2-Keap1 defines a physiologically important
      stress response mechanism. Trends Mol Med 2004;10 (11) : 549-57
 28.  Jung KA and Kwak MK. The Nrf2 system as a potential target for the
      development of indirect antioxidants. Molecules 2010;15 (10) : 7266-91
 29.  Landi L, Fiorentini D, Galli MC, Segura-Aguilar J, Beyer RE. DT-Diaphorase
      maintains the reduced state of ubiquinones in lipid vesicles thereby
      promoting their antioxidant function. Free Radic Biol Med 1997;22 (1-2) :
      329-35
 30.  Itoh K, Chiba T, Takahashi S, et al. An Nrf2/small Maf heterodimer
      mediates the induction of phase II detoxifying enzyme genes through
      antioxidant response elements. Biochem Biophys Res Commun 1997;236 (2) :
      313-22
 31.  Klaassen CD and Slitt AL. Regulation of hepatic transporters by xenobiotic
      receptors. Curr Drug Metab 2005;6 (4) : 309-28
 32.  Dinkova-Kostova AT, Holtzclaw WD, Cole RN et al. Direct evidence that
      sulfhydryl groups of Keap1 are the sensors regulating induction of phase 2
      enzymes that protect against carcinogens and oxidants. Proc Natl Acad Sci
      USA 2002;99 (18) : 11908-13
 33.  Jancova P, Anzenbacher P, Anzenbacherova E. Phase II drug metabolizing
      enzymes. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2010;154 (2)
      : 103-16
 34.  Mazur CS, Kenneke JF, Hess-Wilson JK, Lipscomb JC. Differences between
      human and rat intestinal and hepatic bisphenol A glucuronidation and the
      influence of alamethicin on in vitro kinetic measurements. Drug Metab
      Dispos 2010; 38 (12): 2232-8
 35.  Tukey and Strassburg. Human UDP-glucuronosyltransferases: metabolism,
      expression, and disease. Annu Rev Pharmacol Toxicol 2000; 40 pp. 581-616
 36.  Van der Logt EM, Roelofs HM, van Lieshout EM, Nagengast FM, Peters WH.
      Effects of dietary anticarcinogens and nonsteroidal anti-inflammatory
      drugs on rat gastrointestinal UDP-glucuronosyltransferases. Anticancer Res
      2004;24 (2B) : 843-9
 37.  van Bladeren PJ. Glutathione conjugation as a bioactivation reaction. Chem
      Biol Interact 2000;129 (1-2) : 61-76
 38.  Sheehan D, Meade G, Foley VM, Dowd CA. Structure, function and evolution
      of glutathione transferases: implications for classification of
      non-mammalian members of an ancient enzyme superfamily. Biochem J 2001;360
      (Pt 1) : 1-16
 39.  Ketterer B. Glutathione S-transferases and prevention of cellular free
      radical damage. Free Radic Res 1998;28 (6) : 647-58
 40.  van Bladeren PJ. Glutathione conjugation as a bioactivation reaction. Chem
      Biol Interact 2000;129 (1-2) : 61-76
 41.  Hayes JD and Strange RC. Glutathione S-transferase polymorphisms and their
      biological consequences. Pharmacology 2000;61 (3) : 154-66
 42.  Habuchi O. Diversity and functions of glycosaminoglycan sulfotransferases.
      Biochim Biophys Acta 2000;1474 (2) : 115-27
 43.  Glatt H and Meinl W. Pharmacogenetics of soluble sulfotransferases
      (SULTs). Naunyn Schmiedebergs Arch Pharmacol 2004;369 (1) : 55-68
 44.  Coleman. Human Drug Metabolism: An Introduction. (2010) pp. 360
 45.  Jancova P, Anzenbacher P, Anzenbacherova E. Phase II drug metabolizing
      enzymes. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2010;154 (2)
      : 103-16
 46.  Jancova P, Anzenbacher P, Anzenbacherova E. Phase II drug metabolizing
      enzymes. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2010;154 (2)
      : 103-16
 47.  Mulder GJ. Conjugation reactions in drug metabolism: an integrated
      approach : substrates, co-substrates, enzymes and their interactions in
      vivo and in vitro. Taylor and Francis, 1990. 413 pages
 48.  Hodgson. A Textbook of Modern Toxicology. (2010) pp. 672
 49.  Yang YM, Noh K, Han CY, Kim SG Transactivation of genes encoding for phase
      II enzymes and phase III transporters by phytochemical antioxidants.
      Molecules 2010;15 (9) : 6332-48
 50.  Keppler D. Multidrug resistance proteins (MRPs, ABCCs): importance for
      pathophysiology and drug therapy. Handb Exp Pharmacol 2011;201 : 299-323
 51.  Mizuno, N.; Niwa, T.; Yotsumoto, Y.; Sugiyama, Y. Impact of drug
      transporter studies on drug discovery and development. Pharmacol. Rev.
      2003, 55, 425-461.
 52.  Klaassen C and Lu H. Xenobiotic Transporters: Ascribing Function from Gene
      Knockout and Mutation Studies. Toxicological Sciences 2008;101 (2) :
      186-196
 53.  Klaassen C and Lu H. Xenobiotic Transporters: Ascribing Function from Gene
      Knockout and Mutation Studies. Toxicological Sciences 2008;101 (2) :
      186-196
 54.  Liska DJ. The Detoxification Enzyme Systems. Altern Med Rev 1998;3 (3) :
      187-198
 55.  Larson et al. Acetaminophen-induced acute liver failure: results of a
      United States multicenter, prospective study. Hepatology 2005;42 (6) :
      1364-72
 56.  Moyer AM, Fridley BL, Jenkins GD et al. Acetaminophen-NAPQI
      Hepatotoxicity: A Cell Line Model System Genome-Wide Association Study.
      Toxicol Sci 2011;120 (1) : 33-41
 57.  Bessems JG, Vermeulen NP. Paracetamol (acetaminophen)-induced toxicity:
      molecular and biochemical mechanisms, analogues and protective approaches.
      Crit Rev Toxicol 2001; 31 (1): 55-138
 58.  Lauterburg BH, Corcoran GB, Mitchell JR. Mechanism of action of
      N-acetylcysteine in the protection against the hepatotoxicity of
      acetaminophen in rats in vivo. J Clin Invest 1983; 71 (4): 980-91
 59.  Ercal N, Gurer-Orhan H, Aykin-Burns N. Toxic metals and oxidative stress
      part I: mechanisms involved in metal-induced oxidative damage. Curr Top
      Med Chem 2001;1 (6) : 529-39
 60.  Costa M. In vitro assessment of the toxicity of metal compounds.
      Biological Trace Element Research 1984;
 61.  Patrick L. Lead. Altern Med Rev 2006;11 (2) : 114-127
 62.  Nordberg M. Metallothioneins: historical development and overview. Met
      Ions Life Sci 2009;
 63.  Sabolić I, Breljak D, Skarica M, Herak-Kramberger CM. Role of
      metallothionein in cadmium traffic and toxicity in kidneys and other
      mammalian organs. Biometals 2010);23 (5) : 897-926
 64.  Nordberg M. Metallothioneins: historical review and state of knowledge.
      Talanta 1998;
 65.  Phillips TD, Lemke SL, Grant PG. Characterization of clay-based
      enterosorbents for the prevention of aflatoxicosis. Adv Exp Med Biol
      2002;504 : 157-71
 66.  Guengerich FP. Influence of nutrients and other dietary materials on
      cytochrome P-450 enzymes. Am J Clin Nutr 1995;61 (3 Suppl) : 651S-658S
 67.  Guengerich FP. Influence of nutrients and other dietary materials on
      cytochrome P-450 enzymes. Am J Clin Nutr 1995;61 (3 Suppl) : 651S-658S
 68.  Moon YJ, Wang X, Morris ME. Dietary flavonoids: effects on xenobiotic and
      carcinogen metabolism. Toxicol In Vitro 2006;20 (2) : 187-210
 69.  Fuhr, U., Klittich, K., Staib, A.H., Inhibitory effect of grapefruit juice
      and its bitter principal, naringenin, on CYP1A2 dependent metabolism of
      caffeine in man. Br. J. Clin. Pharmacol. 1993; 35, 431–436.
 70.  Helsby, N.A., Chipman, J.K., Gescher, A., Kerr, D., Inhibition of mouse
      and human CYP 1A- and 2E1-dependent substrate metabolism by the
      isoflavonoids genistein and equol. Food Chem. Toxicol. 1998;36, 375–382.
 71.  Yang YM, Noh K, Han CY, Kim SG Transactivation of genes encoding for phase
      II enzymes and phase III transporters by phytochemical antioxidants.
      Molecules 2010;15 (9) : 6332-48
 72.  Catterall F et al. Hepatic and intestinal cytochrome P450 and conjugase
      activities in rats treated with black tea theafulvins and theaflavins.
      Food Chem Toxicol. 2003 Aug;41(8):1141-7.
 73.  Liu, T.T., Liang, N.S., Li, Y., Yang, F., Lu, Y., Meng, Z.Q., Zhang, L.S.
      Effects of long-term tea polyphenols consumption on hepatic microsomal
      drug-metabolizing enzymes and liver function in Wistar rats. World J.
      Gastroenterol. 2003;9, 2742–2744.
 74.  Křížková J, Burdová K, Stiborová M, Křen V, Hodek P. The effects of
      selected flavonoids on cytochromes P450 in rat liver and small intestine.
      Interdiscip Toxicol 2009;2 (3) : 201-4
 75.  Yuan JH, Li YQ, Yang XY. Inhibition of epigallocatechin gallate on or-
      thotopic colon cancer by upregulating the Nrf2-UGT1A signal path- way in
      nude mice. Pharmacology 2007; 80: 269 – 78
 76.  Hsieh TC, Lu X, Wang Z, Wu JM. Induction of quinone reductase NQO1 by
      resveratrol in human K562 cells involves the antioxidant response element
      ARE and is accompanied by nuclear translocation of tran-scription factor
      Nrf2. Med Chem 2006; 2: 275 – 85
 77.  Nayak S and Sashidhar RB. Metabolic intervention of aflatoxin B1 toxicity
      by curcumin. J Ethnopharmacol 2010;127 (3) : 641-4
 78.  Osawa T. Nephroprotective and hepatoprotective effects of curcuminoids.
      Adv Exp Med Biol 2007;595 : 407-23
 79.  Liao BC, Hsieh CW, Liu YC, Tzeng TT, Sun YW, Wung BS. Cinnamaldehyde
      inhibits the tumor necrosis factor-alpha-induced expression of cell
      adhesion molecules in endothelial cells by suppressing NF-kap- paB
      activation: Effects upon IkappaB and Nrf2. Toxicol Appl Pharmacol 2008;
      229: 161 – 71
 80.  Lii CK, Liu KL, Cheng YP et al. Sulforaphane and alpha-lipoic acid
      upregulate the expression of the pi class of glutathione S-transferase
      through c-jun and Nrf2 activation. J Nutrition 2010;140 (5) : 885-92
 81.  Feng Z, Liu Z, Li X, et al. α-Tocopherol is an effective Phase II enzyme
      inducer: protective effects on acrolein-induced oxidative stress and
      mitochondrial dysfunction in human retinal pigment epithelial cells. J
      Nutr Biochem 2010;21 (12) : 1222-31
 82.  Wang H and Leung LK. The carotenoid lycopene differentially regulates
      phase I and II enzymes in dimethylbenz[a]anthracene-induced MCF-7 cells.
      Nutrition 2010;26 (11-12) : 1181-7
 83.  Veeriah S, Miene C, Habermann N et al. Apple polyphenols modulate
      expression of selected genes related to toxicological defence and stress
      response in human colon adenoma cells. Int J Cancer 2008;122 (12) :
      2647-55
 84.  Liu XP, Goldring CE, Wang HY, Copple IM, Kitteringham NR, Park BK. Extract
      of Ginkgo biloba induces glutathione-S-transferase subunit-P1 in vitro.
      Phytomedicine 2009; 16(5):451–455
 85.  Liu YC, Hsieh CW, Wu CC, Wung BS. Chalcone inhibits the activation of
      NF-kappaB and STAT3 in endothelial cells via endogenous electrophile. Life
      Sci 2007; 80: 1420 – 30
 86.  Joung EJ, Li MH, Lee HG, Somparn N, Jung YS, Na HK et al. Capsaicin in-
      duces heme oxygenase-1 expression in HepG2 cells via activation of
      PI3K-Nrf2 signaling: NAD(P)H:quinone oxidoreductase as a potential target.
      Antioxid Redox Signal 2007; 9: 2087 – 98
 87.  Zhu L, Liu Z, Feng Z et al. Hydroxytyrosol protects against oxidative
      damage by simultaneous activation of mitochondrial biogenesis and phase II
      detoxifying enzyme systems in retinal pigment epithelial cells. J Nutr
      Biochem 2010;21 (11) : 1089-98
 88.  Gong P, Hu B, Cederbaum AI. Diallyl sulfide induces heme oxygenase-1
      through MAPK pathway. Arch Biochem Biophys 2004; 432: 252 – 60
 89.  Zhang Y, Guan L, Wang X, Wen T, Xing J, Zhao J. Protection of chloro-
      phyllin against oxidative damage by inducing HO-1 and NQO1 ex- pression
      mediated by PI3K/Akt and Nrf2. Free Radic Res 2008; 42: 362–71
 90.  Dietz BM, Kang YH, Liu G et al. Xanthohumol isolated from Humulus lupulus
      Inhibits menadione-induced DNA damage through induction of quinone
      reductase. Chem Res Toxicol 2005;18 (8) : 1296-305
 91.  Surh YJ, Kundu JK, Na HK. Nrf2 as a master redox switch in turning on the
      cellular signaling involved in the induction of cytoprotective genes by
      some chemopreventive phytochemicals. Planta Med 2008;74 (13) : 1526-39
 92.  Sulforaphane Glucosinolate monograph. Altern Med Rev 2010;15 (4) : 352-362
 93.  Dinkova-Kostova AT, Holtzclaw WD, Cole RN et al. Direct evidence that
      sulfhydryl groups of Keap1 are the sensors regulating induction of phase 2
      enzymes that protect against carcinogens and oxidants. Proc Natl Acad Sci
      USA 2002;99 (18) : 11908-13
 94.  Mulcahy RT, Wartman MA, Bailey HH, Gipp JJ. Constitutive and
      beta-naphthal- one-induced expression of the human gamma-glutamylcysteine
      synthetase heavy subunit gene is regulated by a distral antioxidant
      response element/ TRE sequence. J Biol Chem 1997;272:7445-7454
 95.  Ociepa-Zawal M et al. The effect of indole-3-carbinol on the expression of
      CYP1A1, CYP1B1 and AhR genes and proliferation of MCF-7 cells. Acta
      Biochim Pol. 2007;54(1):113-7.
 96.  Katchamart S and Williams DE. Indole-3-carbinol modulation of hepatic
      monooxygenases CYP1A1, CYP1A2 and FMO1 in guinea pig, mouse and rabbit.
      Comp Biochem Physiol C Toxicol Pharmacol. 2001 Aug;129(4):377-84.
 97.  Ebert B et al. Induction of phase-1 metabolizing enzymes by oltipraz,
      flavone and indole-3-carbinol enhance the formation and transport of
      benzo[a]pyrene sulfate conjugates in intestinal Caco-2 cells. Toxicol
      Lett. 2005 Aug 14;158(2):140-51.
 98.  Bradlow HL. Review. Indole-3-carbinol as a chemoprotective agent in breast
      and prostate cancer. In Vivo. 2008 Jul-Aug;22(4):441-5.
 99.  Nho CW and Jeffery E. The synergistic upregulation of phase II
      detoxification enzymes by glucosinolate breakdown products in cruciferous
      vegetables. Toxicol Appl Pharmacol. 2001 Jul 15;174(2):146-52.
 100. Morimitsu Y, Hayashi K, Nakagawa Y et al. Antiplatelet and anticancer
      isothiocyanates in Japanese domestic horseradish, Wasabi. Mech Ageing Dev
      2000;116 (2-3) : 125-34
 101. Hasegawa K, Miwa S, Tsutsumiuchi K, Miwa J. Allyl isothiocyanate that
      induces GST and UGT expression confers oxidative stress resistance on C.
      elegans, as demonstrated by nematode biosensor. PLoS ONE 2010;5 (2) :
      e9267
 102. Y. Nakamura, Y. Morimitsu, T. Uzu, H. Ohigashi, A. Murakami, Y. Naito, Y.
      Nakagawa, T. Osawa, K. Uchida, A glutathione S-transferase inducer from
      papaya: rapid screening, identification and structure–activity
      relationship of isothiocyanates
 103. Zhou C, Poulton EJ, Grün F et al. The dietary isothiocyanate sulforaphane
      is an antagonist of the human steroid and xenobiotic nuclear receptor. Mol
      Pharmacol 2007;71 (1) : 220-9
 104. Zhou SF, Xue CC, Yu XQ, Wang G. Metabolic activation of herbal and dietary
      constituents and its clinical and toxicological implications: an update.
      Curr Drug Metab 2007;8 (6) : 526-53
 105. Munday, R. and Munday, C. M. (1999). Low dises of diallyl disulfide a com-
      pound derived from garlic increase tissue activities of quinone reductase
      and glutathione transferase in the gastrointestinal tract of the rat. Nutr
      Cancer. 34:42–48.
 106. Kalayarasan S, Sriram N, Sureshkumar A, Sudhandiran G. Chromium
      (VI)-induced oxidative stress and apoptosis is reduced by garlic and its
      derivative S-allylcysteine through the activation of Nrf2 in the
      hepatocytes of Wistar rats. J Appl Toxicol 2008;28 (7) : 908-19
 107. Sun J. D-Limonene: Safety and Clinical Applications. Altern Med Rev
      2007;12 (3) : 249-264
 108. Crowell PL. Prevention and therapy of cancer by dietary monoterpenes. J
      Nutrition 1999
 109. Van der Logt EM, Roelofs HM, van Lieshout EM, Nagengast FM, Peters WH.
      Effects of dietary anticarcinogens and nonsteroidal anti-inflammatory
      drugs on rat gastrointestinal UDP-glucuronosyltransferases. Anticancer Res
      2004;24 (2B) : 843-9
 110. Elegbede JA, Maltzman TH, Elson CE, Gould MN. Effects of anticarcinogenic
      monoterpenes on phase II hepatic metabolizing enzymes. Carcinogenesis
      1993;14 (6) : 1221-3
 111. Nakamura et al. A phase II detoxification enzyme inducer from lemongrass:
      identification of citral and involvement of electrophilic reaction in the
      enzyme induction* 1. Biochemical and … 2003;
 112. Calcium-D-Glucarate Monograph. Altern Med Rev 2002;7 (4) : 336-340
 113. Zoltaszek R et al. [The biological role of D-glucaric acid and its
      derivatives: potential use in medicine]. Postepy Hig Med Dosw (Online).
      2008 Sep 5;62:451-62.
 114. Ferruzzi MG Digestion, absorption, and cancer preventative activity of
      dietary chlorophyll derivatives. Nutrition Research 2007;
 115. Yun CH, Jeong HG, Jhoun JW, Guengerich FP. Non-specific inhibition of
      cytochrome P450 activities by chlorophyllin in human and rat liver
      microsomes. Carcinogenesis 1995;16:1437 - 40.
 116. Fahey JW, Stephenson KK, Dinkova-Kostova AT, Egner PA, Kensler TW, Talalay
      P. Chlorophyll, chlorophyllin and related tetrapyrroles are significant
      inducers of mammalian phase 2 cytoprotective genes.Carcinogenesis
      2005;26:1247 - 55.
 117. Morita K, Matsueda T, Iida T, Hasegawa T. Chlorella accelerates dioxin
      excretion in rats. J Nutr 1999;129:1731 - 6.
 118. Natsume Y, Satsu H, Kitamura K, Okamoto N, Shimizu M. Assessment system
      for dioxin absorption in the small intestine and prevention of its
      absorption by food factors. Biofactors 2004; 21(1-4):375 - 7.
 119. Versantvoort CHM, Oomen AG, Van de Kamp E, Rompelberg CJM, Sips AJAM.
      Applicability of an in vitro digestion model in assessing the
      bioaccessibility of mycotoxins from food. Food Chem Toxicol 2005;43:31 -
      40.
 120. Egner PA, Wang JB, Zhu YR, Zhang BC, Wu Y, Zhang QN, et al. Chlorophyllin
      intervention reduces aflatoxin-DNA adducts in individuals at high risk for
      liver cancer. Proc Natl Acad Sci 2001;98(25): 14601 - 6.
 121. Resta SC. Effects of probiotics and commensals on intestinal epithelial
      physiology: implications for nutrient handling. J Physiol (Lond) 2009;587
      (17) : 4169-74
 122. Topcu A, Bulat T, Wishah R, Boyaci IH. Detoxification of aflatoxin B1 and
      patulin by Enterococcus faecium strains. Int J Food Microbiol 2010;139 (3)
      : 202-5
 123. Nowak A and Libudzisz Z. Ability of probiotic Lactobacillus casei DN
      114001 to bind or/and metabolise heterocyclic aromatic amines in vitro.
      Eur J Nutr 2009;48 (7) : 419-27
 124. Ibrahim F, Halttunen T, Tahvonen R, Salminen S. Probiotic bacteria as
      potential detoxification tools: assessing their heavy metal binding
      isotherms. Can J Microbiol 2006;52 (9) : 877-85
 125. Pool-Zobel B, Veeriah S, Böhmer FD. Modulation of xenobiotic metabolising
      enzymes by anticarcinogens -- focus on glutathione S-transferases and
      their role as targets of dietary chemoprevention in colorectal
      carcinogenesis. Mutat Res 2005;591 (1-2) : 74-92
 126. Yedjou CG, Tchounwou CK, Haile S, Edwards F, Tchounwou PBl.
      N-acetyl-cysteine protects against DNA damage associated with lead
      toxicity in HepG2 cells. Ethn Dis 2010;20 (1 Suppl 1) : S1-101-3
 127. Alsalim W and Fadel M. Towards evidence based emergency medicine: best
      BETs from the Manchester Royal Infirmary. Oral methionine compared with
      intravenous n-acetyl cysteine for paracetamol overdose. Emerg Med J
      2003;20 (4) : 366-7
 128. Ghabril M, Chalasani N, Björnsson E. Drug-induced liver injury: a clinical
      update. Curr Opin Gastroenterol 2010;26 (3) : 222-6
 129. Abenavoli L, Capasso R, Milic N, Capasso F. Milk thistle in liver
      diseases: past, present, future. Phytother Res 2010; 24 (10): 1423-32
 130. Bosisio E, Benelli C, Pirola O, et al. Effect of the flavanolignans of
      Silybum marianum L. on lipid peroxidation in rat liver microsomes and
      freshly isolated hepatocytes. Pharmacol Res 1992;25:147-154.
 131. Campos R, Garido A, Guerra R, et al. Silybin dihemisuccinate protects
      against glutathione depletion and lipid peroxidation induced by
      acetaminophen on rat liver. Planta Med 1989;55:417-419.
 132. Hau DK, Wong RS, Cheng GY et al. Novel use of silymarin as delayed therapy
      for acetaminophen-induced acute hepatic injury. Forsch Komplementmed 2010;
      17 (4): 209-13
 133. Veeriah S, Miene C, Habermann N et al. Apple polyphenols modulate
      expression of selected genes related to toxicological defence and stress
      response in human colon adenoma cells. Int J Cancer 2008;122 (12) :
      2647-55
 134. Harris KE and Jeffery EH. Sulforaphane and erucin increase MRP1 and MRP2
      in human carcinoma cell lines. J Nutr Biochem 2008;19 (4) : 246-54
 135. Limtrakul P, Chearwae W, Shukla S, Phisalphong C, Ambudkar SV. Modulation
      of function of three ABC drug transporters, P-glycoprotein (ABCB1),
      mitoxantrone resistance protein (ABCG2) and multidrug resistance protein 1
      (ABCC1) by tetrahydrocurcumin, a major metabolite of curcumin. Mol Cell
      Biochem 2007;296 (1-2) : 85-95
 136. Kweon, S.H.; Song, J.H.; Kim, T.S. Resveratrol-mediated reversal of
      doxorubicin resistance in acute myeloid leukemia cells via downregulation
      of MRP1 expression. Biochem. Biophys. Res. Commun. 2010, 395, 104-110.
 137. Saller R, Meier R, Brignoli R: The use of silymarin in the treatment of
      liver diseases. Drugs 2001;61:2035–2063.
 138. Lee CK and Choi JS. Effects of silibinin, inhibitor of CYP3A4 and
      P-glycoprotein in vitro, on the pharmacokinetics of paclitaxel after oral
      and intravenous administration in rats. Pharmacology 2010; 85 (6): 350-6
 139. Padda MS, Sanchez M, Akhtar AJ, Boyer JL. Drug-induced cholestasis.
      Hepatology 2011; 53 (4): 1377-87
 140. Speroni E, Cervellati R, Govoni P, Guizzardi S, Renzulli C, Guerra MC.
      Efficacy of different Cynara scolymus preparations on liver complaints. J
      Ethnopharmacol 2003; 86 (2-3): 203-11
 141. Benedek B, Geisz N, Jäger W, Thalhammer T, Kopp B. Choleretic effects of
      yarrow (Achillea millefolium s.l.) in the isolated perfused rat liver.
      Phytomedicine 2006; 13 (9-10): 702-6
 142. Benedek B and Kopp B. Achillea millefolium L. s.l. revisited: recent
      findings confirm the traditional use. Wien Med Wochenschr 2007; 157
      (13-14): 312-4
 143. Krizman M, Baricevic D, Prosek M Determination of phenolic compounds in
      fennel by HPLC and HPLC-MS using a monolithic reversed-phase column. J
      Pharm Biomed Anal 2007; 43 (2): 481-5
 144. Schütz K, Carle R, Schieber A Taraxacum--a review on its phytochemical and
      pharmacological profile. J Ethnopharmacol 2006; 107 (3): 313-23
 145. Platel K and Srinlvasan K. Stimulatory influence of select spices on bile
      secretion in rats. Nutr Res 2000; 20 (10): 1493-1503
 146. Shukla B, Visen PK, Patnaik GK, Dhawan BN. Choleretic effect of
      andrographolide in rats and guinea pigs. Planta Med 1992; 58 (2): 146-9
 147. Khan BA, Abraham A, Leelamma S. Murraya koenigii and Brassica
      juncea--alterations on lipid profile in 1-2 dimethyl hydrazine induced
      colon carcinogenesis. Invest New Drugs 1996; 14 (4): 365-9
 148. Yamahara J, Miki K, Chisaka T et al. Cholagogic effect of ginger and its
      active constituents. J Ethnopharmacol 1985; 13 (2): 217-25



The importance of a healthy liver

Learn more

RELATED LAB TESTING

Gamma Glutamyl Transferase (GGT) Blood Test

Hepatitis Panel (A, B, C), Acute Blood Test

Hepatitis C Virus Antibody Blood Test

RELATED MAGAZINE ARTICLES

Fighting Cancer Metastasis and Heavy Metal Toxicities With Modified Citrus
Pectin

Protecting Your DNA from Lethal Mutations

N-Acetylcysteine





Top
 * Affiliate Program
 * Become a Reseller
 * Careers
 * Website Map
 * California Privacy

 * Shipping Information
 * Terms of Use
 * GDPR Privacy Notice
 * Cookie Statement
 * Privacy

 * About Us
 * Contact Us
 * FAQs
 * Blog
 * Website Accessibility Statement

 * California Privacy Contact Form


Life Extension does not provide medical advice, diagnosis, or treatment. All
Contents Copyright ©2021 Life Extension. All rights reserved.

†2021 Consumer Satisfaction, Rated #1 Catalog/Internet Merchant.
Ratings based on results of the 2021 and earlier ConsumerLab.com surveys of
supplement users.
More information at www.ConsumerLab.com/survey.

These statements have not been evaluated by the Food and Drug Administration.
These products are not intended to diagnose, treat, cure, or prevent any
disease.


SUCCESS! ITEM(S) ADDED TO CART

×



FAILURE TO ADD TO CART

X
An error on our end has caused the item you were trying to add to your cart to
fail, please try again or call 1-800-678-8989
Continue Shopping View Cart / Checkout
×



You need a Frames Capable browser to view this content.