You are on page 1of 50

Organic Acids

Chapter 6

Organic Acids

173
174
TABLE 6-1. Summary of Organic Acids in Urine
Chapter 6

Name Potential Intervention Metabolic Pathway


Fatty Acid Oxidation Adipate H L-carnitine, 250 mg TID; B2, 100 mg BID;
Suberate H B5, 500 mg BID; Choline, 100 mg TID; Fatty acid oxidation
Ethylmalonate H C, 1000 mg TID; CoQ10, 150 mg/day

Carbohydrate Metabolism Pyruvate H B-complex, 1 capsule TID; Aerobic/anaerobic energy


B1, 100 mg TID; Lipoic acid, 100 mg TID production
Lactate H Coenzyme Q10, 50 mg TID
β-Hydroxybutyrate H Chromium picolinate, 200 mcg BID Glucose utilization

Energy Production Citrate H Arginine, 1-3 gm/day


(Citric Acid Cycle) L Aspartic acid, 500 mg
cis-Aconitate H Lipoate, 25 mg/kg/day
Isocitrate H Magnesium, 400 mg; Manganese, 20 mg Citric Acid Cycle
α-Ketoglutarate L α-KG, 300 mg; Arginine, 1000 mg; Glutamine, 1-5 g Intermediates
H B-complex, 1 TID; Lipoic acid 100 mg
Succinate L Isoleucine, 1000 mg TID; Valine, 1000 mg TID
H Magnesium, 500 mg, CoQ10, 50 mg TID
Fumarate L Tyrosine, 1000 mg BID; Phenylalanine, 500 mg BID
Malate H B3, 100 mg TID
Hydroxymethylglutarate H/L CoQ10, 50 mg TID CoQ10 synthesis

B-Complex Vitamin Markers α-Ketoisovalerate H


α-Ketoisocaproate H B-complex, 1 capsule TID + 100 mg lipoic acid Amino acid catabolism
α-Keto-β-methylvalerate H
Xanthurenate H B6, 100 mg Tryptophan catabolism
β-Hydroxyisovalerate H Biotin, 5 mg BID; Magnesium, 100 mg BID Isoleucine catabolism

Methylation Cofactors Methylmalonate H B12, 1000 mcg TID Valine catabolism


Formiminoglutamate H Folic Acid, 1000 mcg Histidine catabolism

Oxidative Damage and p-Hydroxyphenyllactate H Vitamin C up to 100 mg/kg Prooxidant and carcinogen
Antioxidants 8-Hydroxy-2`-deoxyguanosine H Antioxidants, esp. Vit. C & polypheurls DNA oxidative damage

Detoxification Indicators 2-Methylhippurate H Avoidance of xylene; Glycine, 2-5 g/day Hepatic conjugation
Orotate H Arginine, 1-3 gm/day; α-KG, 300 mg TID; Ammonia clearance,
Aspartic acid, 500 mg BID; Magnesum, 300 mg Pyrimidine synthesis
Glucarate H Glycine, GSH, NAC, 500-5000 mg/day Detox. liver enzyme induction
α-Keto-β-methylvalerate H
Xanthurenate H B6, 100 mg Tryptophan catabolism
β-Hydroxyisovalerate H Biotin, 5 mg BID; Magnesium, 100 mg BID Isoleucine catabolism

Methylation Cofactors Methylmalonate H B12, 1000 mcg TID Valine catabolism


Formiminoglutamate H Folic Acid, 1000 mcg Histidine catabolism

Oxidative Damage and p-Hydroxyphenyllactate H Vitamin C up to 100 mg/kg Prooxidant and carcinogen
Antioxidants 8-Hydroxy-2`-deoxyguanosine H Antioxidants, esp. Vit. C & polypheurls DNA oxidative damage
TABLE 6-1. Cont.

Detoxification Indicators 2-Methylhippurate H Avoidance of xylene; Glycine, 2-5 g/day Hepatic conjugation
Orotate H Arginine, 1-3 gm/day; α-KG, 300 mg TID; Ammonia clearance,
Aspartic acid, 500 mg BID; Magnesum, 300 mg Pyrimidine synthesis
Glucarate H Glycine, GSH, NAC, 500-5000 mg/day Detox. liver enzyme induction
α-Hydroxybutyrate H NAC, 1000 mg; Glutathione, 2-5 g/day Glutathione demand
Pyroglutamate H NAC, 1000 mg, Glutathione, 300 mg Renal amino acid recovery-
Sulfate Taurine, 500 mg BID; Glutathione, 300 mg Detox and anti-oxidant functions

Neurotransmitter Metabolism Vanilmandelate L/H Tyrosine, 1000 mg BID-TID, between meals Epinephrine, Norepinephrine
Contraindicated for patients taking MAO inhibitors catabolism
Homovanillate L/H DOPA catabolism
5-Hydroxindoleacetate L/H 5-Hydroxytryptophan, 100 mg TID Serotonin catabolism
Kynurenate H B6, 100mg; Mg, 300 mg Kynurenin pathway
Quinolinate H Magnesum, 300 mg Serotonin catabolism

Dysbiosis Markers
(Products of Abnormal Benzoate H (Glycine, 1 gm + Pantothenate, 100 mg) - TID Hepatic Phase II conjugation
Gut Microflora) Hippurate H
Phenylacetate H
Phenylpropionate H These compounds reflect intestinal overgrowth,
p-Cresol H usually accompanied by microbial hyperpermeability. Bacterial
p-Hydroxybenzoate H Glutamine, 10-20 gm daily and free-form amino
p-Hydroxyphenylacetate H acids to normalize gut permeability. Numerous interferences in energy
Tricarballylate H Digestive aids (betaine, enzymes, bile) pathways and cellular control
Indican H Take appropriate steps to ensure favorable mechanisms
D-Lactate H gut microflora population.
Dihydroxyphenylpropionate H Yeast
D-Arabinitol H
Organic Acids

175
Chapter 6
FIGURE 6-1. Urinary Markers of Nutrients Involved in Central Energy Pathways
Note: Vitamin and mineral requirements for cofactors are shown in black boxes. Elevations of metabolites before
these steps indicate functional deficit of the nutrients.

Digestion FATS CARBOHYDRATES PROTEINS


and
Assimilation
STAGE I

Fatty Acids, Glycerol Glucose & Other sugars Amino Acids


Cholesterol

Keto Acids
Carnitine Pyruvate Lactate
B1, B2, B3, B5, Lipoate B1, B2, B3, B5, Lipoate

Intermediary Acetyl CoA


Metabolism

Aspartate Oxaloacetate Citrate


Citric Acid Cycle
STAGE II

B3 cis-Aconitate
++ Glu
Malate Cysteine, Fe
His
Isocitrate Arg
Tyr Pro
Fumarate Gln
Phe B3,Mg, Mn
B2
-ketoglutarate
Succinate B1, B2, B3, B5, Lipoate
Ile
Mg Succinyl-CoA Val
Met

Electron Transport NADH


and
Oxidative
STAGE III

NADH
Phosphorylation Dehydrogenase

Coenzyme Q10
ADP + Pi

Cytochromes
Hydroxymethyl- ATP Energy for muscle and
glutarate nerve function and for
O2 building new tissue
H2O

176
Organic Acids

Introduction
The logical sequence of nutrient categories followed throughout
the previous four chapters is broken at this point in the text. Unlike
amino acids and fatty acids, the category of compounds called organic
acids contains no essential nutrients. Instead of tests that measure
nutrient concentrations, abnormal concentrations of organic acids in
urine provide functional markers for the metabolic effects of vitamin
inadequacies, toxic exposure, neuroendocrine activity, and intestinal
bacterial overgrowth. The ultimate tool for laboratory evaluations
in nutritional medicine is a simple, sensitive test that can reveal
evidence of functional inadequacy of specific nutrients. The promise
of such a tool is found in profiling of organic acids in urine.
All bodily functions are powered by the release of chemical
energy. Each day, the energy content of the food for an average person
could raise the temperature of about seven gallons of water to the
boiling point. The energy is released through a process of controlled
oxidation where chemical bonds are broken and energy is released.
Fats, carbohydrates, and amino acids are converted into carboxylic
acids before they flow on to the final conversion to carbon dioxide
(Figure 6-1). The organic acids that are formed as intermediates in
this process are normally absent from urine or present at very low
concentrations. When specific reactions are blocked due to the
absence of sufficient enzyme or cofactor, the intermediates that
precede the blocked step accumulate and spill into urine. Overt
nutrient deficiencies are one reason for metabolic inefficiency.
Variations in enzyme structure that lead to decreased cofactor
binding are another major cause of nutrient deficiencies. Individuals
with faulty enzyme binding can have increased nutrient needs that
will not be revealed by measures of vitamin concentrations in blood.
Various nutrient-related abnormalities that might appear on a typical
quantitative report of organic acids in urine are summarized in Table
6-1. The supplementary nutrient amounts are given as guides for
starting points to improve clinical outcomes for adults. Maple syrup
urine disease provides an example of this concept.
The oxidation of carbon skeletons from amino acids is a major
energy production pathway in which an early step is catalyzed by the
enzyme, branched chain keto acid dehydrogenase. Variants of maple
syrup urine disease are due to degrees of impairment of this enzyme
(Table 6-2). Genetic variants in which the enzyme is not expressed
result in severe clinical consequences in infancy. Other conditions
may develop in adults with mild levels of functional impairment due
to a high requirement for thiamin [1].
Urinary organic acid analysis for metabolic profiling has
traditionally been used for detection of neonatal inborn errors of
metabolism. Since the reporting of isovaleric acidemia in 1966, there
has been a rapidly growing list of disorders resulting in elevated
excretion of metabolic intermediates [2, 3]. The application of the
testing to assess special nutrient requirements of individuals is
177
Chapter 6

TABLE 6-2. Maple Syrup Urine Disease

Variant Organic Acid Clinical Presentation


Severe α-ketoisocaproate Infant death or prominent
α-ketoisovalerate neurologic abnormalities and
α-keto-β-methylvalerate profound mental retardation
Mild Same as "Severe" Irritability, ataxia, lethargy,
developmental retardation
Thiamin-responsive Same as "Severe" Same signs and symptoms
(10-4000 mg/day) (lower levels in urine) as above that appear only
with the stress of infection,
surgery, etc. Bio-chemical
abnormalities clear and
return as thiamin is
administered and withdrawn

discussed in a variety of sources [3-7]. Organic acid profiling has


also been useful in identification of the source of toxicants from the
environment [8] and the gut [9].
The term “organic acid” refers to a broad class of compounds used
in fundamental metabolic processes of the body. Chemically, organic
acids share the common features of water solubility, acidity, and
ninhydrin-negativity (no primary or secondary amines). The term is
generally considered to include all carboxylic acids, with or without
keto, hydroxyl, or other non-amino functional groups, but does not
include most amino acids. Some nitrogen-containing compounds
are included, such as pyroglutamate, or amino conjugates like
hippurate (benzoylglycine). Short chain fatty acids are also contained
in this group. With adjustments of the method for extraction of the
compounds from urine, the analysis may include neutral compounds
that possess no acid group, in which case the naming of the entire set
as “organic acids” is somewhat inaccurate.
Morning urine has become widely accepted for primary study of
metabolic disorders. Urination is generally the first post-sleep activity
because the cortisol-stimulated, anabolic sleep period requires the
metabolic, energy-yielding pathways that produce water. Prior to
waking, the cortisol peaks and then drops, initiating a shift to greater
utilization of catabolic pathways. For these reasons, the first morning
urine usually contains a greater array of metabolic by-products than at
any other time of the day and is relatively free of the metabolic effects
of strenuous muscle activity. In one methodology organic compounds
are extracted from the overnight urine sample and analyzed by gas
chromatography with mass spectrometric detection (GC-MS). The
basic methodologies are well known [10], but improvements are
regularly reported [11]. Most recently, significant advances have
come from the use of liquid chromatography with tandem mass
spectrometric detection (LC-MS/MS) [12]. This technology allows
more reliable analyte recovery due to greatly simplified sample
preparation. This method does not require organic solvent extraction.
It also provides a shorter run time and greater sample throughput, so
the very complex analysis can be performed at a reasonable cost.

178
Organic Acids
The information content of the profile is high, but the
interpretation is simplified by keeping in mind that the data supplies
answers to five basic questions of clinical relevance:
(1) Is mitochondrial energy production adversely affected?
(2) Are functional nutrient deficiencies present?
(3) Are symptoms related to excessive growth of bacteria and
fungi in the gut?
(4) Is there a high toxin load and is this adversely affecting
detoxification capacity?
(5) Are antioxidant nutrients protecting against oxidative stress?
Each of several compounds reported in the typical profiling of
organic acids in urine will be discussed briefly to indicate why they
are related to these clinical questions with key references cited.

Fatty Acid Metabolism Markers


Adipate, Suberate, and Ethylmalonate
Carnitine is a non-essential metabolic cofactor that can be
synthesized from the essential amino acid, L-lysine. However, because
of dietary or digestive limitations of available lysine or increased
carnitine demand, carnitine requirement may exceed capacity for
synthesis in some individuals. Therefore carnitine is in the category
of conditionally essential dietary components.
Need for carnitine is indicated when any of the dicarboxylic acids
adipate, suberate, and ethylmalonate are elevated [13]. Carnitine is
required as a carrier for the transport of fatty acids from the cytosol
into the mitochondria for beta-oxidation (Figure 6-2). This is the
dominant pathway used to derive energy from fat in most cells. When
carnitine is inadequate to keep up with demand, the degradation of
fatty acids takes place through an alternate, less efficient pathway
known as “omega-oxidation.” Adipate and suberate are products of
incomplete oxidation in the omega-oxidation pathway.

FIGURE 6-2. Fatty Acid Metabolism Markers

Fatty Acids

Carnitine

Fattyacyl Carnitine
ω-Oxidation
β-Oxidation

Adipate CO2 + H20 + ATP


Suberate

179
Chapter 6
Failure in the formation or oxidation of butyrylcarnitine can lead
to higher rates of butyrate carboxylation forming ethylmalonate.
The enzyme that controls the butyrate oxidative pathway is short-
chain acyl-CoA dehydrogenase (SCAD). Its activity may be lowered
by genetic mutations or inhibition by dietary toxin. A variant allele
(A625) resulting from a guanine to adenine polymorphism occurs in
homozygous and in heterozygous form in 7 and 34.8% respectively
of the general population. The A625 allele confers susceptibility to
the development of ethylmalonic aciduria [14]. Ingestion of the ackee
plant results in SCAD inhibition due to the presence of the toxin,
hypoglycin. This dietary-related illness has been called Jamaican
vomiting illness because of the common use of ackee in that country.
It has also been reported in the United States from ingestion of
canned ackee [15]. Ethylmalonate excretion may also be stimulated
by isoleucine loading, indicating that it may be an intermediate
produced in the isoleucine catabolic pathway [16].
All three of the compounds will be elevated in severe carnitine
insufficiency. The patterns of elevations in milder carnitine
insufficiency vary. Symptoms include periodic mild weakness,
nausea, fatigue, hypoglycemia, “sweaty feet” odor, and recurrent
infections. Abnormalities are frequently found in children with
attention deficit disorders. Patients may also exhibit a Reyes-like
syndrome in dicarboxylic aciduria, which has been associated
with various metabolic toxins from viral infections that affect
mitochondrial function. Patients with low carnitine respond well to
therapeutic riboflavin [17]. The biochemical relationship between
these two nutrients can explain this effect. Carnitine is required to
allow fatty acids to enter into mitochondria, but fatty acids cannot
undergo oxidative metabolism without riboflavin coenzymes. The
use of aspirin can change the interpretation of results for the three
marker compounds because salicylic acid is an inhibitor of fatty
acid β-oxidation and may lead to elevated markers [18]. The β-
oxidation enzymes involved also respond to environmental toxin
exposure with altered lipid metabolism that can lead to impaired
immune responsiveness and mitochondrial DNA damage [19].
Supplementation of carnitine and riboflavin is indicated when
adipate, suberate, or ethylmalonate are elevated [20].

Carbohydrate Metabolism Markers


Pyruvate and Lactate
Abnormalities of urinary excretion of pyruvate and lactate provide
useful insight to basic metabolic factors due to their position in the
energy production process. Pyruvate is the anaerobic breakdown
product of glucose. Its further conversion to acetyl-CoA requires the
pyruvate dehydrogenase enzyme complex. Pyruvate dehydrogenase
requires cofactors derived from thiamin, riboflavin, niacin, lipoic
acid, and pantothenic acid. Elevated levels of pyruvate may reflect
failure of the enzyme due to a functional need for increased B
180
Organic Acids
vitamins, particularly thiamin [21] and pantothenic acid [22]. Metabolic Acidosis:
Pyruvate elevation in the brain accompanies the encephalopathy • Increased levels of
associated with alcohol use in the thiamin-responsive Wernicke- organic acids due to
their increased
Korsakoff syndrome [23]. Dietary supplementation with pyruvate production or failure
has been found to prevent fatty liver caused by chronic alcohol to be removed
ingestion in laboratory animals [24]. The beneficial effect of pyruvate
• Narrowly defined as
is explained as a stimulation of glucogenesis, which results in either diabetic or
inhibition of fatty acid synthesis. lactic acidosis but
Lactate, also known as lactic acid, is a principal product of more broadly
includes greatly
glucose oxidation in skeletal muscle. Lactate accumulates when there elevated levels of
is a block in the final oxidative phosphorylation stage of energy any organic acids in
production. Such a block results in inactivation of the citric acid cycle. body fluids
Because lactate is transported to the liver and converted to pyruvate,
they have similar responses to nutrient deficiencies. Coenzyme Q10
supplementation is effective when lactate is elevated and energy
production is interrupted [25-27]. Elevated lactate, therefore, is a
sign of potential coenzyme Q10 insufficiency. Increased lactate is
a common acidotic condition that can be caused by a variety of
metabolic problems. A case of lactic acidosis and marked hepatic
steatosis has been normalized with administration of 50 mg of
riboflavin per day [28]. Both lactate and pyruvate may be elevated
by alcohol consumption prior to specimen collection [29]. This
effect may be due to the niacin-depleting effect of alcohol because
the conversion of lactate to pyruvate (Figure 6-3) and the further
oxidation of pyruvate are NAD-dependent. FIGURE 6-3. Lactate
and Pyruvate Inter-
After glucose loading, lactate and pyruvate levels are significantly conversion with
increased in patients with type 2 diabetes. Short-term treatment with Transfer of Electrons
600mg of lipoic acid orally prevented the hyperglycemia-induced to NAD
increments of lactate and pyruvate [30]. This effect was mediated
through increased activity of the pyruvate dehydrogenase enzyme, H3C-CH-CO2-
indicating that the coenzyme role of lipoate was inadequate without I
OH
the supplemental lipoic acid.
Lactate
Lactic acidosis with increased excretion of hydroxybutyrate
and fumarate were biochemical signs of the metabolic disturbance
found in a 42-year-old woman who presented with a 10-year NAD+ NADH
history of ophthalmoplegia, malabsorption resulting in chronic
malnutrition, muscle atrophy, and polyneuropathy [31, 32]. The H3C-C-CO2-
underlying biochemical lesion in this mitochondrial, multi-system II
O
disorder was found to be genetically defective cytochrome C oxidase Pyruvate
(complex IV of the respiratory chain). This case illustrates the
potential for graded metabolic effects from genetic polymorphic
expressions. Interestingly, this defect did not manifest until adulthood.
Compensatory mechanisms maintained nominal function until the
genetic weakness became manifest, which was most likely due to
accumulation of environmental insult and dietary inadequacy. As
we age, metabolic systems function less efficiently. How many of our
“degenerative diseases” are genetic weaknesses manifesting as our
systems become less efficient?
Most individuals carry mild enzyme defects that manifest as
181
Chapter 6
symptoms only when metabolic stressors exceed the ability to
maintain normal function. The age of onset and the degree and
duration of functional loss depend on an individual’s ability to
maintain organ reserve. As we proceed to describe the scientific
reports of specific organic acidurias, it is good to consider this central
axiom: any report of a case or small group of patients with a serious
disorder showing severe organic acidurias is expected in less severe
forms in larger numbers of individuals. Those with the milder forms
may maintain normal function until adulthood.

ß-Hydroxybutyrate
Increased β-hydroxybutyrate is a textbook “ketone body” feature of
metabolic acidosis due to failure of glucose utilization as with diabetes.
Ketone body production increases in diabetes because the oxidation
of free fatty acids is stimulated, and excess acetyl-CoA is converted
α-Hydroxybutyrate
- to the four-carbon acids, β-hydroxybutyrate. This acetyl-CoA spill-
H3C-CH2-CH-CO2
I over phenomenon occurs because the control of ATP production
OH from fatty acids cannot be regulated as well as from carbohydrate
oxidation. Individuals with normal blood glucose response to insulin
do not produce high concentrations of ketone bodies because their
β-Hydroxybutyrate
production of energy from glucose is well controlled. Elevations of
H3C-CH-CH2-CO2
-
β-hydroxybutyrate in an overnight urine collection may indicate
I
OH inefficient utilization or mobilization of glucose [33]. In these cases,
chromium and vanadium supplementation may support carbo-
hydrate utilization by improving the action of insulin [34-36]. Defects
in cytochrome oxidase enzymes of the electron transport system are
another reason for elevated hydroxybutyrate [31]. Excessive fatigue
on exertion is the most common symptom associated with ketosis.

Central Energy Pathway Markers


Citrate, Isocitrate and cis-Aconitate
The Krebs or citric acid cycle (CAC) not only is the final common
pathway of energy release from food components but is also the source
of basic structural or anabolic molecules that are drawn away from
the cycle to support organ maintenance and neurological function.
Therefore, the CAC serves both anabolic and catabolic functions
of the body, representing the “crossroads” of food conversion and
utilization (Figure 6-1).
Conversions of the CAC intermediates are under the control of
enzymes that often require vitamin-derived cofactors and minerals
for their function. Abnormal spilling of CAC intermediates in urine
indicates mitochondrial inefficiencies in energy production. This
fundamental pathway of energy flow is critical for all organ systems.
Elevated excretion of the compounds involved in the cycle can
explain the biochemical basis of excessive fatigue and weakness and
guide ways to improve energy production by supplying B complex
vitamins. Since the compounds are related in a cyclic manner, a
182
Organic Acids
block at any step can cause accumulation of multiple compounds
that precede that step. In cytochrome C oxidase deficiency, there
is inefficient utilization of NADH, the primary product of the CAC,
resulting in citrate, malate, fumarate, and α-ketoglutarate elevations
[42]. Coenzyme Q10 deficiency can also result in elevated CAC
intermediates (see HMG below). Toxic effects of drugs like gentamicin
are, in part, due to loss of mitochondrial integrity. Such drugs inhibit
isocitrate dehydrogenase, the enzyme that converts cis-aconitate
isocitrate. Lipoic acid, used at 25 mg/kg/day, protects against such
cytotoxic effects. Thus, elevated isocitrate and citrate are signs of
increased need for lipoic acid. Isocitrate dehydrogenase activity
also requires the presence of magnesium and manganese ions as
cofactors.
Citrate is frequently the most abundant organic acid in urine.
The range of normal values is broad because citrate excretion varies
with changes in renal metabolism and acid-base disturbances [43].
The anion properties of citrate are used to balance increased positive
ammonia ion excretion. Citrate elevation is a marker of ammonia
accumulation due to arginine deficiency [44, 45]. Orotate elevation
is an additional, sensitive marker for this condition (see below).
Low levels of these three compounds may occur due to the
multiple reactions that utilize CAC intermediates for biosynthesis.
Aspartic acid is a preferred substrate for the refilling or “anaplerotic”
pathway that restores CAC levels. By a single transamination step,
aspartate is converted into oxaloacetate, the immediate precursor of
citrate. The reaction yields a net gain of cycle intermediates, a result
that cannot be achieved from the flow of acetyl-CoA (Figure 6-1).

Alpha-Ketoglutarate
Alpha ketoglutarate (α-KG), also known as 2-ketoglutarate or 2-
oxoglutarate, is formed by oxidation of isocitrate, and it is further
oxidized in an energy-releasing step that requires an enzyme system
similar to the one described for pyruvate oxidation above. The
same five B vitamins are necessary for the action of this enzyme,
so elevations of α-KG signal potential B-complex deficiencies. Low
activity of this enzyme has been shown to produce elevated excretion
of α-KG [46].
The compounds that make up the CAC can be derived from
amino acids as discussed above for aspartic acid. This would explain
the energy-boosting effect people often report when they take free-
form amino acid supplements. The effect is due to the conversion of
specific amino acids directly into depleted CAC intermediates needed
for the energy-producing cycle. α-Ketoglutarate also is formed in the
catabolic breakdown of glutamic acid, histidine, arginine, proline,
and glutamine. The fatigue-reducing effect of supplementation of
aspartate salts and ketoglutaric acid has been attributed to such a
mechanism [47, 48]. The importance of α-KG in cell energetics is
shown by the effectiveness of oral α-KG in cyanide poisoning where
the availability of oxygen is the primary deficit [49].
183
Chapter 6

Levels of α-KG can also serve to mark an aspect of the carbohydrate


and fat metabolic relationship. When metabolic conditions, such
as insulin intolerance, act to stimulate fatty acid synthesis, the
rising levels of palmitoyl-CoA cause an effective inhibition of
glutamate dehydrogenase. Since the dominant pathway for α-KG
formation is production from glutamate by the activity of glutamate
dehydrogenase, mitochondrial α-KG is depleted [50]. To state the
corollary, low α-KG is a marker for up-regulated fatty acid synthesis,
increased palmitic acid in plasma and cell membranes, and increased
serum triglycerides.

Succinate
Succinate cannot play its role in cellular energy production via
the citric acid cycle when coenzyme Q10 (CoQ10) is inadequate.
Clinical signs of CoQ10 and riboflavin deficiency include fatigue,
lassitude, and myocardial and neurological degeneration. CoQ10
depletion can be tissue specific. For example, a 4-year-old boy who
presented with progressive muscle weakness, seizures, and cerebellar
syndrome had greatly reduced muscle CoQ10 without corresponding
CoQ10 deficits in lymphoblasts or skin fibroblasts [27]. His muscle
mitochondria had severely restricted ability to utilize succinate.
Elevated succinate excretion is a marker for deficiency of CoQ10
and riboflavin. The reaction in which succinate is converted into
fumarate depends on the presence of flavin adenine dinucleotide
(FAD) derived from riboflavin. Riboflavin administration has been
shown to produce dramatic regression of neurological impairment
in cases of deficits in the complex II respiratory chain succinate
dehydrogenase enzyme (Figure 6-4) that forms the first link in the
transport of electrons from succinate to oxygen [17, 51].
When succinate levels drop below normal, leucine and isoleucine
are effective precursors that are converted into succinate to assure
functioning of the CAC. Since this conversion requires adequacy of
vitamin B12, functional adequacy of B12 should be assured when
amino acids are used to raise succinate. Individuals with inherited
disorders in which the final steps of amino acid conversion to
succinate are blocked exhibit serious neurologic symptoms [52].
FIGURE 6-4. Coenzyme Q10 Function Coenzyme Q10 controls the rate of citric acid
cycle processing in the oxidation of succinate to fumarate. Reduced coenzyme Q10 (HQ10)
accepts hydrogen atoms one at a time from reduced FAD (FADH2).

Citrate Mitochondrial Membrane

COMPLEX II

(Riboflavin)
Succinate
FAD
HQ10
Fumarate FADH2
Cytochromes
Malate
Q10

184
Organic Acids

Fumarate and Malate


The removal of electrons from succinate in the reaction shown in
Figure 6-4 generates fumarate. Fumarate is then converted to malate.
Because of the sequential nature of these reactions in the mitochondrial
matrix, fumarate and malate elevations serve as additional markers
to substantiate the extent of metabolic interference from coenzyme
Q10 deficiency or cytochrome oxidase defects [31]. Elevated malate
also could be the result of stimulation of fatty acid synthesis. The
fatty acid synthesis product palmitoyl-CoA is known to inhibit the
malate dehydrogenase enzyme [50]. This sign of metabolic resistance
to the oxidation of stored fat is seen in patients on low fat, high
carbohydrate diets or with the hyperinsulinism known as metabolic
syndrome.
Low fumarate and malate concentrations in urine are a sign that
the refilling reactions for the CAC are not keeping up with losses of
intermediates to other pathways. Amino acid catabolism supplies a
major portion of compounds that re-supply of the CAC (Figure 6-1).
The addition of tyrosine or phenylalanine can help supply fumarate
when this intermediate is low.

FIGURE 6-5. Coenzyme Q10 Pathways Elevated urinary markers can reveal blocks
in the biosynthetic pathway (HMG) or in the mitochondrial utilization of coenzyme Q10
(succinate, fumarate, malate).

Carbohydrates Pyruvate

Acetyl-CoA

Hydroxymethylglutarate
Spilling in urine
Citrate
A due to blocks at
A and B
Malate
Cholesterol

Coenzyme Q10
α -KG
Fumarate

Electron Carriers Succinate

B
= Metabolic block
Coenzyme Q10
A = HMG CoA reductase inhibitors
O2
H20 B = CoQ10 deficiency

Hydroxymethylglutarate
Hydroxymethylglutarate (HMG) is the metabolic precursor of
both cholesterol and coenzyme Q10 (CoQ10) (Figure 6-5). Low levels
of HMG may reflect inadequate synthesis and possible deficiency of
CoQ10. Drugs that are used to lower serum cholesterol by inhibiting
185
Chapter 6
the HMG-CoA reductase simultaneously inhibit the endogenous
synthesis of CoQ10 and cause accumulation of the HMG intermediate
in the pathway [53]. Therefore, high levels of HMG reveal inhibition
in the synthesis of CoQ10. CoQ10 is utilized in the mitochondrial
oxidative phosphorylation pathway for ATP synthesis and is a
potent antioxidant. It has been used extensively as a cardiovascular
protective agent [54]. Although it may seem paradoxical, cases where
simultaneous low serum CoQ10 levels were found with low urinary
HMG suggest that CoQ10 inadequacy may be associated with both
low and high levels of HMG. The rationale for this association is
that a block earlier in the pathway inhibits the formation of HMG
producing low levels in urine, while a block subsequent to the
formation of HMG produces high levels.

B-Complex Vitamin Markers


All of the compounds that we have placed in this category are
metabolic intermediates in the degradation of amino acids (Figure
6-6). Once again, vitamin insufficiencies result in slower rates of the
required reactions. Because of the specificity of individual enzymes
utilizing the vitamin-derived coenzymes, discrete patterns of elevated
excretion of specific intermediates signal functional deficiencies of
individual vitamins.

FIGURE 6-6. B-Complex Vitamin Markers Elevations of specific compounds


appearing in urine indicate deficiencies of nutrients required in their catabolic pathways.
Deficiencies of the B-complex vitamins cause blocked pathways shown as:

Compound Appearing
In Urine
Nutrient
Involvement

Valine α-KETOISOVALERATE B-Complex


Transamination Leucine α-KETOISOCAPROATE
Isoleucine α-KETO-β-METHYLVALERATE
CO2 + H2O

Amino Acid
METHYLMALONATE
Catabolism B12
β-HYDROXYISOVALERATE Biotin

Alpha-Ketoisovalerate, Alpha-Ketoisocaproate, and


Alpha-Keto-Beta-Methylvalerate
When hepatic or small intestinal enzymes remove the amino
groups from the branched chain amino acids valine, leucine,
and isoleucine, the products formed are α-ketoisovalerate, α-keto-
isocaproate, and α-keto-β-methylvalerate, respectively. In most
current biochemical literature, these compounds are named 2-oxo
acids, rather than α-keto acids. In medical discussions, the older
Greek letter designation still prevails.
These compounds have been discussed previously. Although
that discussion noted the specific response to thiamin, the reactions
186
Organic Acids
require all of the B-complex cofactors: vitamins B1, B2, B3, B5, and
lipoic acid. Elevations of the branched chain keto acids provide a
functional assessment of the sufficiency of these vitamins, especially
thiamin [10].
Patients taking the hypolipidemic agent, clofibrate, may have
a diminished sensitivity for these markers of vitamin deficiency,
because the drug stimulates their metabolism by the branched chain
keto acid dehydrogenase complex [55].

Xanthurenate
Xanthurenic acid is the first of several tryptophan metabolites
that will be discussed. Vitamin B6 insufficiency leads to elevated
excretion of xanthurenate [56].
The liver regulates tryptophan levels in plasma and brain through
the kynurenin pathway that is initiated by the hepatic enzyme,
tryptophan-2,3-dioxygenase. In extrahepatic tissues, including brain
and macrophages, kynurenin is formed from tryptophan by a second
enzyme called indoleamine-2,3-dioxygenase (IDO). In either case, the
kynurenin is subsequently converted into kynurenic, xanthurenic,
and quinolinic acids (Figure 6-7).
There are important consequences for the tissue-specific gateways
for initiation of kynurenin synthesis. In general, however, the
pathway in which kynurenic, xanthurenic, and quinolinic acids are
formed from L-tryptophan serves the following functions:
1. Clearance of excess L-tryptophan
2. Maintenance of nicotinic acid levels
3. Regulation of peripheral pain perception by CNS neurons
4. Enhancing macrophage defense functions

Dietary tryptophan is an essential amino acid that often is the


rate-limiting amino acid in biosynthetic reactions. Therefore, there is
little need for the tryptophan clearance function in most individuals
except following high protein meals. When an L-tryptophan load
is received, the kynurenin pathway should become very active in
the liver. The extra metabolic load is the basis of using a 3-5 gm L-
tryptophan challenge to increase the sensitivity for detecting marginal
vitamin B6 status [57]. Although the excretion of both kynurenic and
xanthurenic acids are increased when vitamin B6 is insufficient to
maintain this function, xanthurenate is the principal product that
appears. This is due to the further metabolism of kynurenate to citric
acid cycle intermediates in the liver.
Elevated xanthurenate may be found even when there is no
challenge with extra tryptophan. In such cases, the insufficiency
of vitamin B6 is sufficiently great to interfere with even the normal
conversion of dietary tryptophan. When piglets were used to study
vitamin B6 deficiency, urinary levels of kynurenate and xanthurenate
were elevated early in vitamin B6 deficiency and they continued to
rise as the deficiency worsened [58].

187
Chapter 6
FIGURE 6-7. The Serotonin and Kynurenin Tryptophan Pathways
Hepatic tryptophan oxidase clears excess L-tryptophan via the kynurenin pathway. In
extrahepatic tissues, tryptophan may be converted into serotonin or, by the action of IDO,
into kynurenin, kynurenic, and quinolinic acids that interact with NMDA receptors.

One productive outcome of the kynurenin pathway is the


production of nicotinic acid for NAD+ and NADP+ formation. Thus,
dietary tryptophan can be used to form nicotinic acid, sparing the
need for dietary niacin. If dietary tryptophan is inadequate, then
dietary niacin is required. However, even if the supply of tryptophan
is in excess, niacin cannot be formed in the absence of vitamin B6.
The kynurenine pathway offers an excellent example of the complex
interplay of essential nutrients. As we discuss below, this pathway
also provides critical regulation of nervous system function. Estrogen
hormones inhibit the initial conversion of tryptophan to kynurenine,
offering an explanation of the twofold excess of females over males
in many outbreaks of the niacin deficiency disease, pellagra [59].
Further discussion of kynurenin pathway products is found below
under the topic, kynurenate and quinolinate.

ß-Hydroxyisovalerate
For many years, students of nutrition were taught that there is
no evidence of biotin deficiency in adult humans [60]. The discovery
of biotin production by some organisms that inhabit the human gut
added impetus to the attitude that, although biotin is clearly not
made in human tissues, biotin deficiency was of little concern. It
was thought that only certain disorders of infancy, such as Leiner’s
disease (desquamative erythroderma) and other forms of seborrheic
dermatitis, are clinical manifestations of biotin deficiency. Recent
findings are shifting the clinical outlook for biotin deficiency in
adults.
188
Organic Acids
The biochemical function of biotin-requiring enzymes is the
insertion of carboxyl groups to allow modification of metabolic
intermediates. The catabolism of the amino acid leucine is a high-
flux process that offers a biochemical marker of biotin deficiency.
The product formed after the first three steps of the pathway, β-
methylcrotonyl-CoA, requires a biotin-dependent carboxylation to
allow the flow to continue. This compound accumulates if biotin
is deficient, and its hydrated product, β-hydroxyisovalerate, spills
in urine. β-Hydroxyisovaleric aciduria appears early in people who
are made biotin deficient by consuming the biotin-binding protein,
avidin. After starting avidin administration, elevated β-hydroxy-
isovalerate appears at the third day while serum biotin concentrations
remain in the normal range until the tenth day (Figure 6-8). The
effects are completely reversible.
Heritable disorders of biotin metabolism lead to the condition
called multiple carboxylase deficiency, in which the activities of
enzymes that have absolute requirements for biotin to carry out
carboxylation reactions are deficient [61]. β-Hydroxyisovalerate
is a compound that is elevated in biotin deficiency and multiple
carboxylase insufficiency [62]. Biotin deficiencies of various degrees
have been shown to develop in normal pregnancies [63] and
in patients on long-term anticonvulsant therapy [64]. When β-
hydroxyisovalerate was used to assess biotin in pregnant women,
biotin status was found to decrease during pregnancy, and 9 out of
13 women studied were biotin depleted even in early pregnancy [65].
Symptoms of biotin deficiency include alopecia, skin rash, Candida
dermatitis, unusual odor to the urine, immune deficiencies, and
muscle weakness.
The enzymes that use biotin as a cofactor are called carboxylases,
because they use carbon dioxide to insert carboxyl groups into
substrates. Biotin from food or from intestinal microbial synthesis
is absorbed in the upper small intestine and transported to tissues
bound to several blood proteins. Cellular biotin must be incorporated
into the carboxylase enzymes by the action of other enzymes called
synthases (Figure 6-9). If these enzymes are not fully active, higher
biotin concentrations can increase enzymatic activity and enhance
the reaction rate. The carboxylase enzymes have critical roles in
major pathways for the utilization of energy from amino acids (where
β-hydroxyisovalerate is formed), the synthesis of fatty acids for cell
membrane replacement, and the maintenance of blood glucose via
gluconeogenesis. At cell death, the biotin may be recovered if there
is sufficient activity of the enzyme biotinidase, which acts on the
biotin-peptide fragments called “biocytin.” All five of the major
factors (boxed text) in Figure 6-9 contribute to the maintenance of
active carboxylase enzymes. Biotin deficiency can be caused by lack
of biotin-rich foods or genetic variations in the enzymes shown in
Figure 6-8. Antibiotic overuse can contribute to biotin insufficiency
by lowering the population of biotin-producing organisms and
favoring the overgrowth of non-biotin producing species.
189
Chapter 6

FIGURE 6-8. Human Biotin Deficiency and β-Hydroxyisovalerate Excretion

Adapted from [51]

80 800

70 700

Urinary β-Hydroxyisovaleric acid


60 600

50 500
(nmol/ 24 h)
Plama Biotin

(umol/24 h)
40 400

30 300

20 200

10 100

0 0

0 3 7 10 14 17 20
Normal Range for Biotin Normal Range for β-Hydroxyisovaleric acid

A case involving biotinidase deficiency illustrates the use of biotin


replacement. A two-year-old male patient presented with ataxia,
seizures, and a history of normal development for 19 months, and
then experienced loss of language and developmental milestones and
progressive gait disturbances [66]. Upon physical examination, the
patient was obtunded with eczematous dermatitis, truncal ataxia, and
a fine volitional tremor on the arms. Symptoms of ataxia, tremors,
loss of language development, gait disturbances, and eczematous
dermatitis cleared only after increasing biotin intake to 30 mg/day.
The data presented in Table 6-3 are from this report. After one year
of high dose biotin, the symptoms did not return and there was no
further progression of hearing loss but some attention disorder with
hyperactivity.
In addition to increased β-hydroxyisovalerate, elevations of
lactate and alanine in urine and accumulations of odd-chain fatty
acids in plasma or red cell membranes have also been found in biotin
deficiency [61].

TABLE 6-3. Biotin Doses for 2 yr. Old Male with Biotinidase Deficiency

Biotin Dose Clearing Signs/Symptoms

6 mg Truncal ataxia
Convulsions
Alertness

20 mg Organic acids
Leukocyte carboxylase

30 mg Dermatitis (Returned at 25 mg)

190
Organic Acids
FIGURE 6-9. Defects of Biotin Pathways

O
N N
Dietary Biotin
HOOC S
Small Intestinal Microbial Synthesis
Cellular biotin

Biotinidase
Carboxylase Synthase(s)
Loss in Urine
O
N N
O
Peptide -N-OC S N N
Biocytin in blood
Enzyme -N-OC S

Proteases Carboxylases

CARBOHYDRATES
PROTEINS Gluconeogenesis
Amino acid catabolism LIPIDS Blood glucose
Cell energy Fatty acid synthesis
New tissue membranes

Methylation Pathway Markers


Methylmalonate
Methylmalonyl-CoA is produced in the multi-step process of
breaking down the amino acid valine into energy-yielding products.
The intermediate is converted into succinic acid using a vitamin
B12-dependent enzyme, methylmalonyl CoA mutase (see Chapter
2). The lack of vitamin B12 impairs this conversion as well as the
one in which methionine is recovered from homocysteine, leading
to accumulation of both homocysteine and methylmalonate. Such
biochemical impairment helps explain the fatigue resulting from
vitamin B12 deficiency and the rapid improvement of symptoms
vitamin B12-deficient patients experience with administration of
intramuscular vitamin B12.
Vitamin B12 deficiency has been demonstrated to be present in
over 12% of the free-living elderly population in the United States
[67] and is especially common in the early stages of HIV infection
[68].

Formiminoglutamate
Formiminoglutamic acid (FIGLU) is an intermediate in the
deamination of the amino acid histidine. Folic acid is the cofactor
required by the enzyme formiminotransferase that converts FIGLU
to glutamic acid (Figure 6-10). Folate accepts the formimino group
from FIGLU to yield N5-formimino-THF. The conversion of FIGLU
to glutamate can be used to determine folate sufficiency. FIGLU
excretion increases in folate deficiency, especially when histidine is
administered as an oral load [69]. Up to 15 gm of oral histidine has
been used as a FIGLU metabolic challenge for adults. Additionally,

191
Chapter 6

FIGURE 6-10. Folate Requirement in Formiminoglutamate Metabolism

L-Histidine Marker of functional folate,


independent of other single
carbon transfer factors
Formiminoglutamic acid

Glutamate Formiminotransferase

Folate

Glutamic acid

FIGLU excretion has been used to measure the influence of drugs or


alcohol on functional folate status [70] [71].
Functions of folate include amino acid metabolism, cell division
and replication, and nucleic acid synthesis, purine and pyrimidine
synthesis in particular. Consequently, deficiency of folate can lead to
inhibition of DNA synthesis, impaired cell division, and alterations
in protein synthesis. Mild folate deficiency is characterized by
low plasma folate and hypersegmentation of polymorphonuclear
leukocytes. After about four months of low folate intake, red blood
cell folate concentrations diminish [72]. High urinary FIGLU appears
about 90 days after a period of insufficient folate availability.
Since its function is to accept a single carbon group from the
substrate in the FIGLU reaction, the simple folic acid form of the
cofactor is needed. Another form of folate, 5-methylfolate, functions as
a methyl-donor in the methionine-homocysteine pathway. Deficiency
of this form of folate results in high levels of homocysteine. Elevated
homocysteine increases the risk of ischemic heart disease, stroke,
and deep vein thrombosis [73]. Differences in binding affinity and
in the forms of folate make it possible to have elevated FIGLU with
normal homocysteine. The FIGLU pathway can be thought of as
folate loading, while the homocysteine pathway is folate unloading.
Addition of methyl groups in the homocysteine pathway requires
loaded (methyl-) folate, while removal of formimino groups in the
FIGLU pathway require only folic acid. Therefore, simple folic acid
supplementation (not methyl forms) is needed to normalize FIGLU
elevations.
A serum folate concentration of less than 0.3 ng/ml is evidence
of deficiency, but false normal values are frequently found due to
the response of serum levels to transient dietary intake changes that
do not correspond to tissue repletion of the nutrient. The period
of deficiency that must pass before erythrocyte folate drops below
normal levels is longer than that resulting in hyperhomocysteinemia.
Changes in leukocyte folate levels occur at the same rate as
erythrocyte, leukocyte folate thus is not a valuable early marker for
deficiency. Combination assays of the methylation pathway markers
FIGLU, methylmalonate, and homocysteine have been proposed as
the most sensitive and cost effective means for routine diagnosis of
megaloblastic anemia [74].
192
Organic Acids
TABLE 6-4. Differentiation B12, Folate, and B6 Deficiency
Elevated levels of metabolic markers are denoted by a  symbol. Combinations
of metabolic markers allow the differentiation of folate, methylfolate, vitamin B12,
vitamin B6, or methyl donor insufficiencies, any of which may contribute to high
homocysteine.

Methylmalonate

Homocysteine
Xanthurenate
Scenario

FIGLU

Vitamin Deficiency Indication


1  N N , N Folate - Not methylfolate
2 N  N , N B12
3 N N  , N B6
4 N N N  Methylfolate or Methyl donors (betaine, etc.)
5  N N  Folate (and Methyl donors) or Methylfolate
6 N  N  B12
7     Folate, B12, B6, and Methyl donors

Homocysteinemia is a sensitive marker of folate inadequacy at


the biochemical level. However, homocysteinemia is not necessarily
due to folate deficiency. Serum folate can be used as a confirmatory
test. A differential diagnosis of vitamin B12, folate, and vitamin B6
deficiencies can be made with simultaneous assay of methylmalonate,
formiminoglutamate, and homocysteine (Table 6-4). If only methyl-
malonate is elevated (Scenario 1), only B12 supplementation is
needed. In Scenario 5, folate is indicated and though B12 is not
needed according to the normal methylmalonate, the homocysteine
elevation may be signaling a need for methyl donor compounds
like betaine or dimethylglycine. If all of the markers are found to be
elevated (Scenario 7), then the full spectrum of cofactors and methyl
donating nutrients should be considered.
Folate deficiency can occur for a number of reasons. Since
humans cannot synthesize folate and it must be provided by the
diet, poor dietary intake of folate-rich foods or excessive intake of
processed foods increases the chances of insufficiency. Impaired
absorption, inadequate utilization due to a metabolic block, increased
demands, increased excretion, or increased destruction of folate,
all contribute to folate insufficiency. Prescription drugs, lack of
specific nutrients (see below), genetic enzyme defects, many diseases,
and alcoholism are factors that can influence the likelihood of a
folate deficiency. Methotrexate (MTX) is a competitive inhibitor of
dihydrofolate (FH2) and prevents recycling of folate to its usable
THF form. Oral contraceptive use has been shown to impair folate
metabolism as measured by serum folate, erythrocyte folate and
FIGLU [75]. Vitamin B12 deficiency results in reduced folate uptake
across gut and other cell walls, and reduced cell retention. Ascorbate
protects folate from oxidative destruction, thus low dietary vitamin C
influences folate status. Chronic alcohol consumption impairs folate

193
Chapter 6
coenzymes, and, in folate deficiency, causes possible malabsorption
of enterohepatically-circulated folates [76].
A wide variety of clinical conditions can suggest folate evaluation.
Extra folate may be needed to improve endothelial function and
lower risk of heart disease [77, 78]. Women with inadequate folate
have a 53% higher risk of breast cancer, and folate adequacy reduces
the increased breast cancer risk from alcoholic beverage consumption
[79, 80]. Patients with small cell carcinoma of the lung, who were
losing weight, had elevated FIGLU. It was demonstrated that these
individuals had high demands for one-carbon units supplied by
folate [81]. Cigarette smoking-associated increase in pancreatic
cancer is reduced when folate function is assured [82]. Smoking
seems also to be associated with elevated homocysteine levels in
patients with early atherosclerotic development [83]. Patients with
Crohn’s diseases should be evaluated for folate adequacy to control
homocysteine-associated thrombotic events [84]. Demands for folate
increase with gestation [85]. Supplementation of folate in women
of childbearing years not only prevents neural tube defects, but also
prevents megaloblastic anemia, which can complicate pregnancies.

Neurotransmitter Metabolism Markers


Vanilmandelate and Homovanillate
Vanilmandelate, also known as vanilmandelic or vanillyl-
mandelic acid (VMA), is the main metabolite of the catecholamines,
epinephrine, and norepinephrine. Homovanillate (HVA) is the
main metabolite of dopamine that appears in urine (Figure 6-11).
Low urinary levels of VMA and HVA have been associated with low
CNS levels of these neurotransmitters [86]. Symptoms associated
with this condition include depression, sleep disturbances, anxiety,
and fatigue. Treatments aimed at improving protein digestion
and supplementation of the amino acid precursor (L-tyrosine) can
normalize neurotransmitter levels in the CNS [87]. The relationship
of tyrosine to epinephrine also gives rise to the antihypertensive role
of tyrosine [88].

FIGURE 6-11. Neurotransmitter Metabolism Markers

Neurotransmitter
COMPOUND APPEARING Essential
Biosynthesis
IN URINE Nutrient
Involvement
Epinephrine VANILMANDELATE
Norepinephrine
Tyrosine Phenylalanine
Dopamine HOMOVANILLATE
Tryptophan Serotonin 5-HYDROXYINDOLACETATE Tryptophan

194
Organic Acids
Elevated levels of VMA and HVA signal an increased rate of
synthesis and degradation in normal tissue or abnormal production
by tumor tissue. Neuroblastic tumors frequently cause a profound
elevation in VMA, which may be expressed as the VMA/HVA ratio
[89]. The incidence of elevated values appears to increase as a
function of tumor size, and small tumors are not likely to result in
positive urinary measurements [90]. Elevation of VMA along with
specific elevation of cardiac troponin I are, together, precursors to
myocardial injury [91].
In the absence of such disease processes, increased catecholamine
synthesis results from the synergism of pituitary adrenocorticotropic
hormone and adrenal cortisol. This constitutes the widely recognized
chronic stress response that manifests as heightened sympathetic
reactions to stress.

5-Hydroxyindoleacetate
Catabolic breakdown of serotonin leads to excretion of 5-
hydroxyindoleacetate (5-HIA) (Figure 6-11). Abnormal high levels
of this metabolite result from the use of serotonin-specific re-uptake
inhibitor (SSRI) drugs or increased release of serotonin from any of
three primary sites: central nervous system, intestinal argentaffin
cells, or platelets. Carcinoid tumors composed of chromaffin tissue
also can release large amounts of serotonin. Because of the magnitude
of total body serotonin synthesis, increased rates of serotonin
turnover indicated by elevated urinary 5-HIA can lead to depletion of
the essential amino acid precursor, L-tryptophan. A very high 5-HIA
result calls attention to potential deficiency of tryptophan. Plasma
amino acid analysis provides a useful, direct measure of this amino
acid (see Chapter 4 – Amino Acids).
Serotonin is required for control of gut motility as it activates
smooth muscle activity. Inadequate production of serotonin leads
to constipation. Low levels of serotonin have also been associated
with depression, fatigue, insomnia, suicide, and attention deficit and
behavioral disorders. Ethanol consumption also causes lowered 5-HIA
excretion, because the biochemical pathway that catalyzes serotonin
metabolism is extremely sensitive to ethanol [92]. When 5-HIA levels
are low, increased consumption of foods high in tryptophan including
turkey, bananas, low fat milk, lentils, and eggs can minimize the need
for oral tryptophan or 5-hydroxytryptophan.

Kynurenate and Quinolinate


The kynurenin pathway has been discussed above under the
xanthurenate topic with regard to evaluating vitamin B6 status.
There are two other products of that pathway that have quite a
different meaning with regard to the clinical application of organic
acids in urine testing. The kynurenin pathway in interferon-gamma
stimulated macrophages may be a mechanism of host defense against
intracellular pathogens (Figure 6-12) [93].
Kynurenic acid (KYNA) and quinolinic acid (QUIN) have well-
195
Chapter 6

FIGURE 6-12. Functions of the Kynurenic Pathway

Chemical communication from immune


Hepatic disposal of excess L-trytophan system to brain for activation
and production of nictinate of sensory perceptiveness

Liver Macrophage +
L-Tryptophan
L-Tryptophan Interferron γ

Kynurenine
+
Kynurenine

Quinolinate Inflammatory response


Quinolinate
NMDA agonist
Sleep disturbance (adults)

Nicotinic acid or
other oxidation products

documented effects in the central nervous system. KYNA is an


endogenous ligand that antagonizes all types of ionotropic glutamate
receptors. It has preferential affinity for the glycine-binding site of
the N-methyl-D-aspartate (NMDA) receptor. Intrathecal kynurenic
acid has antinociceptive effects and it interacts with endomorphin-
1 in rats to decrease pain perception [94]. Other studies suggest
that elevated KYNA may represent a pathophysiological condition
analogous to that seen in schizophrenic patients [95]. Because of the
activity of pyridoxal-5-phosphate in this pathway (discussed above),
these effects may mediate the clinical uses of vitamin B6 in treatment
of neurological disorders [96-99]. Even transient deficits of B6 may
cause significant shifts in brain physiology.
QUIN production is a link between the immune system and the
brain. Chronic stimulation of the immune response causes release of
interferon gamma by macrophages. Interferon gamma (IFN-γ) induces
increased production of the enzyme, indoleamine-2,3-dioxygenase,
that starts the pathway of tryptophan conversion to QUIN (Figure
6-7). Since the gut is a primary point of chronic inflammatory signal
induction via interferon gamma, there is reason to suspect that QUIN
elevation may indicate both inflammatory bowel conditions and
neuronal degeneration. Within the brain, the hippocampus is an
area rich in NMDA receptors and it is very sensitive to the neurotoxic
effects of QUIN.
Because of the tight, positive association between QUIN and IFN-
γ, immune stimulation and increased QUIN production may be a
key feature in the progression of events that lead to chronic fatigue
syndrome. An immunological dysfunction in patients with chronic
fatigue syndrome has been demonstrated by decreased monocyte
response to IFN-γ [100]. In one set of patients with chronic fatigue
syndrome, CD4 T lymphocytes have decreased interferon-gamma
production [101]. However, during acute and convalescent parvovirus
B19 infection circulating IFN-γ is associated with prolonged and
196
Organic Acids
chronic fatigue [102]. Quinolinate levels may allow discrimination of
the contribution of immune cell stimulation to chronic fatigue. Thus,
elevated QUIN may be an indication of the immune-mediated form
of chronic fatigue.
KYNA production via the kynurenin pathway is characteristically Inflammatory
altered in relapsing-onset multiple sclerosis. Patients in remission or diseases resulting in
not progressing for at least two months have lower rates of CSF KYNA quinolinate elevation
formation [103]. KYNA is a precursor to QUIN and is simultaneously [108]:
elevated by inflammatory responses. QUIN also interacts with Bacterial infection
ionotropic glutamate receptors that respond to pain and other Viral infection
peripheral signals. The neurons respond by activation of NMDA Fungal and parasitic
receptors. If they are over stimulated, the neurons can degenerate infections
with permanent loss of brain function. Their loss is associated with Menigitis
the effects of stroke and with the end stages of HIV infection. Studies Autoimmune diseases
have shown that QUIN has a role in the etiology of HIV-related Septicemia
neurologic dysfunction [104, 105]. Congenital
QUIN is a powerful agonist of the NMDA receptors, eliciting hyperammonemia
heightened response and greatly accelerated neuronal degeneration.
Kynurenic acid is antagonistic for NMDA activation, so it opposes the
neurodegenerative effect of QUIN. Therefore, the relative amounts
of the two metabolites should determine the rate of degeneration.
In inflammatory diseases, the ratio of QUIN:KYNA is usually far in
favor of neurotoxicity [108]. Efforts have been intensified to develop
kynurenic acid analogues as neuroprotectants for the treatment of
stroke and neurodegenerative disease. Any agent that can change the
balance of synthesis of kynurenic and quinolinic acids away from
the excitotoxin and towards the neuroprotectant, has anticonvulsant
and neuroprotective properties [109].
A final, useful outcome of the conversion of Trp to QUIN is the
production of nicotinic acid and its cofactor, NAD. This step requires
the enzyme quinolinic acid phosphoribosyl transferase for the linkage
of the adenosyl moiety of NAD. The transferase has low activity in
the brain, thus giving rise to increased QUIN as an unprocessed
intermediate. In the liver, QUIN may be largely processed to NAD.

Oxidative Damage and Antioxidant


Markers
p-Hydroxyphenyllactate
p-Hydroxyphenyllactate (HPLA) is a metabolite of tyrosine.
Elevated HPLA is associated with tumor growth [110] and leukemia.
HPLA is also a marker of hepatic encephalopathy in patients with
hepatic cirrhosis [111], and urinary levels may be elevated in some
inborn errors of metabolism [112, 113].
The methyl ester, methyl-p-hydroxyphenyllactate (MeHPLA),
is an important cell growth-inhibiting agent. Tumor cells contain
esterase activities that hydrolyze the compound to the free acid,
HPLA. Thus, the effects of elevated HPLA may be due to depletion
197
Chapter 6
of MeHPLA. MeHPLA binds strongly to nuclear sites, blocks estradiol
stimulation of uterine growth, and inhibits growth of human breast
cancer cells [114]. HPLA, on the other hand, binds more weakly to
the same nuclear site and does not block uterotropic responses to
estradiol or inhibit growth of breast cancer cells. Thus, HPLA is an
important regulator of normal and malignant cell growth, and it
appears to mediate the cancer promoting effects of estrogen. MeHPLA
is deficient in a variety of rat and mouse mammary tumors and
human breast cancer preparations, and this deficiency correlates
with the loss of regulatory control [114]. Other estradiol-potentiating
effects are mediated by toxins such as pertussis [115] and cholera
toxin [116]. These effects are worsened by high HPLA because of the
loss of regulatory control when MeHPLA is depleted.
Elevated levels of HPLA result in a dramatic decrease of ascorbic
acid (vitamin C) concentration in the liver, adrenal glands, and
blood [117]. High doses of ascorbic acid (100 mg/kg body weight
daily) were shown to arrest or significantly inhibit the excretion of
HPLA in patients with hemoblastoses and nephroblastoma [118].
Thus, high urinary HPLA signals an increased tissue growth response
that may be associated with neoplastic disease or increased growth
of normal tissue. The dramatic responses to ascorbic acid suggest its
involvement in mediating this level of cell growth. Tissue saturating
levels of vitamin C should be considered in patients with elevated
HPLA.

8-Hydroxy-2’-deoxyguanosine
When inflammatory markers like quinolinate or metabolic
enhancers like p-hydroxyphenyllactate are elevated, there is a need to
know the overall impact of increased production of reactive oxygen
species (ROS). Sustained inflammatory responses and increased cell
proliferations rates cause increased production of reactive oxygen
species. When local antioxidant protection fails to keep reactive
oxygen species in check, there are molecular consequences for every
class of structural molecule. 8-Hydroxy-2’-deoxyguanosine (8-OHdG)
is a product of oxidative damage to DNA [119]. 8-OHdG is formed in a
promutagenic DNA lesion reaction of oxygen radicals with guanosine
groups in DNA. It is recognized as a useful marker in estimating
DNA damage induced by oxidative stress. Oxidative DNA damage is
common in various forms of chronic liver disease ,suggesting a link
between chronic inflammation and hepatocarcinogenesis [119].
The antioxidant protection in young children was evaluated by
urinary 8-OHdG [120]. 8-OHdG formation is sufficiently sensitive to
reveal even mild chronic effects of ROS. The association of cancer
with chronic psychological stress and perceived overwork may be via
the formation of 8-OHdG [121]. Increases in 8-OHdG with cigarette
smoking is associated with aging and enhancement of oxidative
damage in human lung tissues [122]. 8-OHdG levels rise with age
in adults with mild hypercholesterolemia or/and mild hypertension
[123].
198
Organic Acids
Lower levels of antioxidants may predispose to oxidative stress,
which is manifested by higher levels of 8-OHdG. Levels of 8-OHdG
were significantly higher in atherosclerotic patients and vitamin C
levels were significantly lower. This oxidative stress may promote
and worsen atherosclerosis [124]. Exposure to organochlorines does
not result in elevated 8-OHdG, indicating that the genotropic effects
of this compound are exerted through mechanisms other than ROS
formation [125]. A similar conclusion may be reached regarding
moderate alcohol consumption, which seems to have the overall
effect of reducing DNA damage, as shown by the decrease in 8-OHdG
levels [126].
Diabetics tend to have higher urinary 8-OHdG excretion than
healthy controls and the levels are related to the severity of
tubulointerstitial lesions. Oxidative stress may contribute to the
progression of tubulointerstitial injury in patients with diabetic
nephropathy [127]. In a study of the analytical performance of 8-
OHdG measurements, no significant difference between the mean
group levels of 8-OHdG/creatinine in spot urine and in 24-h urine
was observed [128]. Thus, a first morning urine specimen is adequate
for 8-OHdG measurements.

Detoxification Markers
The compounds discussed here are also presented in Chapter 8,
which elucidates their relationship to other tests for detoxification.
They are organic acids that are significant markers of exogenous and
endogenous toxin accumulation. The ability of the laboratory to
measure glucarate is determined by the type of sample preparation
chosen. Glucarate is extremely water-soluble and does not move into
the organic solvent layer when methods requiring solvent extraction
are used. Newer methods utilizing LC-MS/MS technology may report
glucarate because no solvent extractions are performed.

Orotate
When there is insufficient capacity for detoxifying the load
of ammonia via the urea cycle, carbamoyl phosphate leaves the
mitochondria and stimulates the synthesis of orotate [129] (Figure 6-
13). Increased orotic acid (orotate) production is a sensitive indicator
of arginine deficiency. Most of the symptoms that develop following
arginine deprivation can largely be accounted for by a decreased
efficiency of ammonia detoxification and reduced formation of
nitric oxide. Increased orotic biosynthesis is observed with increasing
ammonia concentrations in rat, mouse, and human liver and is
reduced by in vitro arginine supplementation [44, 130]. In addition,
orotate requires magnesium for its metabolism. While a normal
level in a urinary organic acid panel may not necessarily indicate
magnesium sufficiency, high levels should alert one to a significant
possibility of intracellular magnesium insufficiency.
Oral glutamine supplementation at levels above 10 gm/day in
199
Chapter 6
adults can result in elevation of urinary orotate due to increased
ammonia generated from hepatic oxidative deamination reactions
[131].
FIGURE 6-14.
Metabolism of
Xylene to 2-Methylhippurate
2-Methylhippurate This compound is a by-product from the detoxification of the
common solvent, xylene. Xylene is oxidized via hepatic p450 oxidase
enzymes, and the 2-methylbenzoate product is conjugated with
Xylene
glycine to form 2-methylhippurate (Figure 6-14). Spray-painting
P450 workers show elevated methylhippurate, indicating recent exposure
to xylene [132]. Patient counseling in avoidance is indicated. Xylene
is a very common solvent found in paint thinners and building
2-Methylbenzoate
Glycine
products, fuel and exhaust fumes, and industrial degreasers and
solvents.

Glucarate
2-Methylhippurate
Glucaric acid is a by-product of the predominant liver hepatic
phase I detoxification reactions involving cytochrome P450 oxidation
of glucose to glucuronic acid. The further oxidation of glucuronic
acid produces glucarate. Hepatic output of glucarate is accurately
reflected by urinary levels, and glucarate excretion is an indicator of
overall hepatic detoxification function.
The clinical significance of endogenous glucarate production
should not be confused with the cancer protective role of oral
supplementation with glucarate salts. Beta-glucuronidase produced
by intestinal bacteria can increase the entero-hepatic circulation
of carcinogens. Oral D-glucarate is converted into the potent beta-
glucuronidase inhibitor D-glucaro-1,4-lactone under the influence
of stomach acid. Urinary glucarate is influenced by oral supple-
mentation (usually as calcium D-glucarate), because most absorbed
glucarate is cleared in urine [133].
The liver produces glucuronate for use in Phase II conjugation
reactions [134-136]. The best-known example is glucuronate
conjugation of the hemoglobin degradation product bilirubin as a
final preparation for urinary excretion. A great variety of drugs, food
components, and products of gut microbial metabolism are prepared
for excretion by glucuronidation (Table 6-5). Glucarate elevation
is an indication of enzyme induction due to such potentially toxic
exposures [137-139]. Any exposure that results in stimulation of
hepatic P-450 activity will result in increased excretion of glucarate.
Urinary D-glucaric acid, for example, is elevated in pesticide-exposed
groups [140]. Glucarate measurements have been advanced as useful
biomarkers to xenobiotic exposure, being particularly useful as a
screening tool for xenobiotic exposure in reproductive epidemiology
[141].
Long term exposure to environmental pollutants and continued
toxic load to the detoxifying systems may lead to oxidative stress, high
levels of P-450 activity, and reduced capacity for Phase II conjugation
reactions. Patients suffering from toxic burdens may experience a
200
Organic Acids
wide range of symptoms, among them fatigue, headaches, muscle
pain, mood disorders and poor exercise tolerance. Researchers
have reported that many chronic fatigue syndrome patients have
disordered liver detoxification ability and show signs of increased
toxic exposure [142].
Hepatic Phase II conjugation reactions convert fat-soluble
substances to water-soluble forms for elimination. The major Phase II
pathways generate mercaptan (glutathione), methyl, sulfate, glycine,
and glucuronide conjugates. Markers for all of these pathways may be
found in a profile of organic acids in urine (Table 6-6). Assessment of
the metabolic status of these major detoxification processes assists in
understanding the body’s capacity to detoxify foreign substances and
thereby, prevent long-term damage from their continued exposure.
Measurement of D-glucarate in urine serves as a specific biomarker
for glucuronidation [143]. Elevations in urinary glucarate specifically
suggest exposure to pesticides, herbicides, fungicides, petrochemicals,
alcohol and drugs.
Table 6-5. Compounds Removed from the Body by Glucuronidation

Polycyclic aromatic hydrocarbons (Benzo(a)pyrene, Benzanthracene, Naphthalene)


Various nitrosamines (cured meats, tobacco products, rubber products, pesticides)
Fungal toxins (aflatoxin)
Steroid hormones (estrogen, testosterone)
Heterocyclic amines (well-done, fried, or barbequed meats)
Pharmaceutical drugs (aspirin, lorazepam, digoxin, morphine)
Vitamins (A, E, D, K)

Alpha-Hydroxybutyrate
Myocardial tissue has high activities of the enzyme, alpha
hydroxybutyrate dehydrogenase (αHBD) that catalyzes oxidation
of the organic acid, alpha-hydroxybutyrate (αHB). The activity
of αHBD on the second day after a myocardial infarction is a
marker for estimates of infarct size and a measure of reperfusion
effectiveness [37]. The intense energy demand of cardiac muscle is
likely the reason for such high concentrations of αHBD in that tissue,
because αHB strongly inhibits mitochondrial energy metabolism
as measured by CO2 production [38]. Possibly the most widely
applicable concept for the interpretation of elevated urinary αHB is
increased cytoplasmic NADH2/NAD ratio. Elevation of this ratio has
been proposed as the explanation of high αHB excretion [39]. Any

Table 6-6. Organic Acids Markers of Phase II Detoxification


Phase II Conjugate Urinary Marker Compounds
Mercaptan, (glutathione) a-Hydroxybutyrate, Pyroglutamate, Sulfate
Methyl Methylmalonate, FIGLU
Sulfate Sulfate
Glycine Benzoate
Glucuronide Glucarate

201
Chapter 6
Figure 6-15. Formation of α-Hydroxybutyrate
When Methionine is Converted
into Glutathione

Methionine

Homocysteine
Serine

Cystathionine
α-Ketobutyrate
NADH
Cysteine NAD+
α-Hydroxybutyrate
Glutathione
conditions that result in high activity of carbohydrate oxidation
can raise the NADH2/NAD ratio, because the extramitochondrial
oxidative pathways tend to load reducing equivalents (-H) onto NAD.
Alcohol consumption and insulin-stimulated glucose uptake raise
this ratio.
Smoking, poor diet, and lack of exercise significantly inhibit the
activity of αHBD, suggesting that urinary elevation of α-hydroxy-
butyrate may be related to these factors [40]. High α-hydroxybutyrate
is also found during phases of increased lymphocyte destruction in
infectious diseases such as measles [41]. Elevated αHB is found in birth
asphyxia and in the inherited metabolic diseases such as “cerebral”
lactic acidosis, glutaric aciduria type II, dihydrolipoyl dehydrogenase
(E3) deficiency, and propionic acidemia [38]. All of the conditions
that have been associated with increased a-hydroxybutyrate excretion
may be related to increased rates of hapatic glutathione synthesis
from methionine (Figure 6-15).

FIGURE 6-16. Pyroglutamate and Glutathione Glutathione is used to recover


amino acids in the tubules of the kidney. A block in the ATP-required step is shown and the
resulting shunt pathway is represented by thick, grey arrows.
ADP + Pi
Glutathione

ATP
Kidney Glycine
Cys-Gly
Amino acids Cysteine γ-Glutamylcysteine

Pyroglutamate

Urine Glutamate

202
Organic Acids

FIGURE 6-17. Distribution of Pyroglutamate Results The cutoff of 80 ug/mg


creatinine is indicated by the shaded bar. A large number of individuals have some degree
of wasting of glutathione and some individuals are in a state of severe depletion, which can
effect detoxification and protection against free radical damage.

Urinary Pyroglutamate
500

450

400
(µg/mg creatinine)

350
Pyroglutamate

300

250

200

150

100

50
0

1567
1513
1459
1405
1351
1297
1243
1189
1135
1081
1027
973
919
865
811
757
703
649
595
541
487
433
379
325
271
217
163
109
55
1

Cases

Pyroglutamate
Small amounts of pyroglutamate are always present in overnight
urine because it is produced as an intermediate in a cycle used in the
active transport of amino acids in renal tubules [144]. This process
utilizes glutathione as a carrier. With each molecule of amino acid
recovered, glutathione is split and then reformed by an ATP-requiring
enzymatic step (Figure 6-16). When the cycle is impaired, the glutamic
acid portion of glutathione is converted to pyroglutamate, which is
excreted. This shunt pathway conserves amino acids at the expense of
glutathione. Up to one third of the glutathione circulating in blood
may be used in this amino acid recovery process. Figure 6-17 shows
actual results for urinary pyroglutamate in 1500 sequential cases
submitted to a clinical laboratory. Relative frequencies of moderate
to severe occurrences of elevated pyroglutamate are apparent from
inspection of the figure. N-acetylcysteine (NAC) is an effective oral
agent for rebuilding total body glutathione, and oral taurine spares
sulfur amino acids while providing an effective antioxidant.

Sulfate
The sulfation pathway is used in Phase II liver detoxification
for biotransformation of many drugs, steroid hormones, phenolic
compounds, and other compounds. The addition of a sulfate group
increases water solubility of hydrophobic compounds in preparation
for their excretion in urine (see Chapter 8). The ratio of sulfate
to creatinine has been used to assess the body’s reserve of sulfur-
containing compounds (especially glutathione) used in Phase II
pathways [145]. When the ratio of sulfate to creatinine is low, these
203
Chapter 6
stores need replenishment. Glutathione administration with oral
cysteine, taurine, and salts of sulfate are used in combinations to
replenish sulfur pathways and restore the hepatic supply of inorganic
sulfate [146].
Since both pyroglutamate and sulfate are markers of sulfur
compound depletion, explanation of the interpretation of various
combinations of abnormalities is needed. Severe depletion of organic
sulfur sources will cause simultaneous high pyroglutamate and
low sulfate excretion. High pyroglutamate with normal sulfate
indicates inadequate organic sulfur sources for production of cysteine
required for glutathione synthesis. Only organic sulfur in the form
of compounds such as N-acetylcysteine or methionine will restore
normal glutathione levels. Normal urinary pyroglutamate with
low sulfate levels can occur in individuals with impaired sulfate
activation. In these cases, rapid replenishment of hepatic sulfate may
Origins of Dysbiosis be accomplished with either sulfur donors like N-acetylcysteine or
inorganic sulfate such as sodium sulfate [145].
• Widespread use of
antacids

• Prescription drugs
Intestinal Dysbiosis Markers
that block stomach
acid production Intestinal dysbiosis markers are discussed in Chapter 7 since they
• Repeated use of are related to gastrointestinal function. These organic acids provide
antibiotics an entirely different type of information from that discussed so far.
They are reported as a part of the full profiling of organic acids in
• Food allergies and
sensitivities urine, because the specimen set up and analytical methods are the
same as for the other organic acids. In addition to their coverage
• Use of NSAIDs
in Chapter 7, we will review their interpretation here in order to
• Consumption of complete the coverage of organic acids.
infected foods The abnormal overgrowth of unfavorable microflora in the small

FIGURE 6-18. Dysbiosis Markers Amino acids and sugars that are not assimilated can
be used by gut microbes for growth. The specific compounds that are formed depend on
the starting material and the species of organism present. In additon to bacterial formation
from phenylalanine, dietary intake of benzoate may add to urinary levels of benzoate and
hippurate. The other compounds are of microbial origin.

COMPOUND APPEARING
IN URINE Actions

BENZOATE & HIPPURATE


Phenylalanine PHENYLACETATE (Antibiotics)
PHENYLPROPIONATE Glutamine

Tyrosine p-CRESOL Lactobacillii and


Bacteria & p-HYDROXYBENZOATE other Pre-biotics
p-HYDROXYPHENYLACETATE
Giardia
Fiber, FOS, and
Sugars TRICARBALLYLATE
other Pro-biotics

Clostridia Sp. DIHYDROXYPHENYLPROPIONATE S. boulardii

β-KETOGLUTARATE Antifungals
Sugars Yeast & D-ARABINITOL Anti-fermentive diet
Fungi

204
Organic Acids
and large intestine is sometimes referred to as “gut dysbiosis” in order
to distinguish this clinical condition from that of infection. Dysbiosis
has been related to a wide variety of symptoms due to pathogenic
toxins produced by the populations of microflora. Some compounds
that appear in urine are unique metabolic products of the microbes
that inhabit the lumen of gut (Figure 6-18). The compounds have FIGURE 6-19.
a wide range of relative toxicities, with cresol at the upper extreme Conversion of
and hippurate and benzoate at the lower end. Among the factors Tyrosine to p-Cresol
by Gut Microbes
that lead to dysbiosis, importance of mealtime habits should not
be overlooked because of the power to restore intestinal health by Tyrosine
allowing normal stomach emptying rate and adequate flow of gastric
P. vulgaris
and pancreaticobiliary fluids. The condition of microbial overgrowth C. dificile
is usually chronic, and the toxic products can affect multiple
organs. p-Hydroxyphenylacetate
Amino acids and sugars that are not assimilated can be used by
gut microbes for growth. The specific compounds that are formed C. dificile
L. ?*
depend on the starting material and the species of organism present.
In addition to bacterial formation from phenylalanine, dietary intake
p-Cresol
of benzoate may add to urinary levels of benzoate and hippurate. The
other compounds are of microbial origin. *Lactobacillus species other
than L. acidophilus, leichmanii,
The high predictive value of urinary markers has been established delbrukii, plantarum, brevis,
minutis, or fermentus.
from several lines of investigation. No false negative results and
only 2% false positive results for small bowel disease and bacterial
overgrowth syndrome were found in a study of p-hydroxy-
phenylacetate in 360 acutely ill infants and children [147]. Three of
the patients who tested positive for this compound were diagnosed
with Giardia lamblia infection.

Hydroxyphenylacetate
No other species has a digestive tract exactly like humans. The
one that has the closest resemblance is swine. Studies in newly
weaned pigs [148] have revealed microbes that carry out tyrosine

FIGURE 6-20. Response of HPA to Treatment The dramatic lowering of HPA


after treatments commensurate with six different clinical presentations demonstrates the
removal of microbial overgrowth in every case, whether the cause was lactose intolerance
or giardiasis.

600
Jejunal Web
Urinary p-Hydroxyphenylacetate

500
Lactose Intolerance

400
Giardia Iamblia

300
Septicemia

200
Low Ig

100
Ileo-colic intersuception

0
At Diagnosis After Diagnosis

205
Chapter 6
degradation (Figure 6-19). Both the transamination to form p-
hydroxyphenylacetate (HPA) and the decarboxylation to p-cresol are
carried out by Clostridium difficile. Since Proteus vulgaris can do only
the first of these steps, HPA will increase in urine if Proteus vulgaris
is the predominant organism. When P. vulgaris is accompanied by
overgrowth of a newly identified strain of lactobacillus, however,
p-cresol will be the major product to accumulate. The lactobacillus
was not given species identification, but the characteristics eliminate
any of the species listed in the footnote of Figure 6-19. Such studies
illustrate the potential for more specific identifications based on
patterns of products appearing in urine.
HPA is elevated in a wide variety of conditions involving direct
intestinal pathology or digestive organ failure (Figure 6-20), which are
obvious candidates for dysbiosis. Although treatments vary greatly
depending on the nature of the disorder, the lowering of urinary HPA
reveals a normalized intestinal bacterial population. Some microbial
compounds are absorbed and enter the detoxification pathways of the
liver to be excreted as modified products that can serve as indicators of
gastrointestinal activities. For example, bacterial amines are converted
to piperidine, a sensitive biochemical index of gastrointestinal flora
changes in celiac disease [149]. Other compounds appear due to
genetic traits that affect how bacterial products are metabolized.
The anaerobic bacterial product, 3-phenylpropionate, for example,
is normally converted to common hippuric acid, but is excreted as
3-phenylpropionylglycine in individuals with a relatively common
inborn error of fatty acid oxidation [150]. Some compounds excreted
in these instances are not organic acids, so they must be analyzed in
separate assays to enhance the interpretation of origins for microbial
compounds in urine.

Benzoate and Hippurate


Bacterial deamination of the amino acid phenylalanine produces
benzoate, which is conjugated with glycine in the liver to form
hippurate. Benzoic acid is also a common food component. It is
used as a preservative in packaged foods such as pickles and lunch
meats, and it occurs naturally in cranberries. This should be taken
into account when interpreting elevated hippurate levels in urine.
Whether the source is dietary intake or jejunal bacterial metabolism,
benzoate is usually converted to hippurate by conjugation with
glycine in the liver. Glycine and pantothenic acid are the limiting
factors in this process. Therefore, elevated benzoate is a marker of
inadequate levels of these nutrients [151, 152].

Phenylacetate and Phenylpropionate


Intestinal bacterial action on phenylalanine also causes the
appearance of phenylacetate and phenylpropionate in urine.
Excretion of these compounds in urine is markedly increased after
the gastrointestinal tracts of germ-free rats are inoculated with
fecal microorganisms, indicating their microbial origin [9]. For
206
Organic Acids
individuals with normal, healthy intestinal function, phenylacetate
and phenylpropionate should not appear at more than background
concentrations in urine. They are products of unidentified, specific
strains of bacteria, marking a state of bacterial overgrowth when they
appear elevated in urine.

p-Cresol, p-Hydroxybenzoate,
and p-Hydroxyphenylacetate
Tyrosine from dietary protein is the parent compound from
which p-cresol, p-hydroxybenzoate, and p-hydroxyphenylacetate
are formed. The compounds are not products of normal human
metabolism, but are produced by bacteria and protozoa that can
populate the gut. Cresol has a chemical structure very similar to
phenol and is highly toxic. Cresol excretion is not affected by dietary
protein intake, suggesting that the bacteria responsible reside in
the lower portions of the small intestine and colon. They produce
cresol from intestinal secretions rather than from dietary sources
[153]. Almost all adult celiac disease patients excrete unusually high
amounts of p-cresol [154]. Due to the loss of renal function, uremic
patients accumulate cresol. The resultant increase in serum cresol can
be prevented by the use of oral sorbents [155], demonstrating that the
origin of the p-cresol is the bowel. Finely powdered, activated charcoal
is a widely available sorbent, but newer synthetic compounds such as
AST120 may be more effective.
Strains of Escherichia coli can produce p-hydroxybenzoate from
glucose [156]. Esters of p-hydroxybenzoate, called parabens, have
antibacterial activity [157], and they are part of the mechanism for
establishing bacterial dominance in intestinal populations.
p-Hydroxyphenylacetic aciduria has been found useful in detecting
small bowel disease associated with Giardia lamblia infestation, ileal
resection with blind loop, and other diseases of the small intestine
associated with anaerobic bacterial overgrowth [147]. Use of antibiotics
that act primarily against aerobic bacteria (such as neomycin) can
encourage the growth of protozoa and anaerobic bacteria that then
produce greater amounts of these compounds [158].

Indican
Bacteria in the upper bowel produce the enzymes that catalyze
the conversion of tryptophan to indole. Absorbed indole is converted
in the liver to indoxyl, which is then sulfated to allow for urinary
excretion. Indoxyl sulfate (also known as indican) can be measured
colorimetrically by conversion to colored oxidation products or
directly by liquid chromatography with a UV absorption or mass
spectrometric detector.
Because the upper bowel is sparsely populated with bacteria,
indican is present in urine at low levels in health. An elevated level of
urinary indican is an indication of upper bowel bacterial overgrowth.
Such an overgrowth was reported for some patients with adult celiac
disease [154]. Bacterial overgrowth was demonstrated with indican in
207
Chapter 6
8 out of 12 patients following jejuno-ileal bypass surgery [159].
Oral, unabsorbed antibiotics reduce indican excretion [160].
Indican excretion is also reduced when the gut is populated
with strains of lactobacillus at levels above 105 organisms/g [160].
L. salivarius, L. plantarum, and L. casei were more effective in
achieving reduced indican than were two strains of L. acidophilus.
In patients with cirrhosis of the liver, tryptophan loading can
produce neuropsychiatric manifestations due to intestinal bacterial
production of tryptophan metabolites [161]. The symptoms are
reduced by antibiotic therapy, demonstrating the bacterial origin of
the metabolites.
Indican testing can differentiate pancreatic insufficiency from
biliary stasis as the cause of steatorrhea (fatty stools). Patients with
steatorrhea due to pancreatic insufficiency show a rise of indican from
low values to above normal when they are treated with pancreatic
enzyme extract [162]. Urinary indican does not rise in patients with
steatorrhea not due to pancreatic insufficiency nor in the normal
subjects who receive pancreatic enzymes. This scenario demonstrates
how bacterial populations respond to increased concentrations of
luminal amino acids. Large shifts in bacterial populations induced by
the artificial sweetener, saccharin, have also been demonstrated by
changes in indican excretion [163].
No age adjustment for reference limits is necessary since excretion
is constant for young and elderly control subjects [164]. The test may
be performed after oral loading of 5 g tryptophan [165]. Including
elevations of other bacterial metabolites with that of indican as
criteria of abnormal bacterial colonization of the small intestine
reduces the number of false positives [166].
The interpretation of indican results is complicated by impaired
protein digestion, which increases the tryptophan available for
bacterial action. Even patients with normal intestinal bacterial
populations can show increased postprandial indican excretion when
they fail to digest dietary protein. The relationship between increased
indican and incomplete digestion might be utilized as a measure of
protein digestive adequacy. Indican evaluation has been used to assess
intestinal absorption of tryptophan in scleroderma [167]. Increased
urinary indican has been shown to correlate with enteric protein loss
[168]. Indican elevation has revealed that impaired protein digestion
and increased bacterial conversion of tryptophan is a complication of
cirrhosis of the liver [169]. Some degree of malabsorption was found
in 30% of an elderly population by combinations of indican with the
Shilling and other tests [170].

D-Lactate
When conditions for bacterial growth are highly favorable in
absorptive regions of the gut, the rate of production of organic
acids can become so high that the luminal pH drops dramatically.
Lactobacillus acidophilus is highly competitive under carbohydrate-
rich, acidic conditions and a major product of its growth is D-lactic
208
Organic Acids
acid. D-lactate entering portal circulation can undergo hepatic
conversion to carbon dioxide, but this pathway has limited capacity.
This limitation is in contrast to the extremely large capacity for
metabolism of the L-lactate isomer produced in skeletal muscle and
other tissues. With continued increases in intestinal output, rising
blood levels are reflected in urinary output of D-lactate [171].
Jejunoileostomy patients have the highest risk of developing D-
lactic acidosis and the accompanying encephalopathy [172, 173]. They
usually have some degree of carbohydrate malabsorption. Because of
the specificity and sensitivity of urinary D-lactate, the test has been
proposed for routine diagnosis of bacterial infections [174]. Elevated
levels of D-lactate were found in blood samples of 13 out of 470
randomly selected hospitalized patients [175]. Studies in cattle have
confirmed that increases in D-lactate following overloading of grain
in the diet corresponded to growth of lactobacilli rather than colliform
bacteria [176]. Enteric overgrowth of Lactobacillus acidophilus has also
been reported in a case of oral antibiotic-induced D-lactic acidosis
[177]. D-Lactic acidosis due to overgrowth of Lactobacillus plantarum
and Lactobacillus salivarius was reported in a child who developed
an unusual syndrome due to the resulting D-lactic encephalopathy
[178].
The phenomenon of D-lactic acidosis has been described as
turning sugar into acid in the gastrointestinal tract [179]. D-Lactate
is not the only organic acid produced from simple carbohydrates.
They are also turned into p-hydroxybenzoate as already discussed
and tricarballylate as discussed below, but those compounds are never
absorbed at rates that can produce the systemic effects found with D-
lactate. When D-lactate is elevated, supplementation with all species
of Lactobacillus is contraindicated, and steps to reduce bacterial
populations should be considered.

Tricarballylate
Tricarballylate (tricarb) is produced by a strain of aerobic bacteria
that quickly repopulates in the gut of germ-free animals [180]. As
its name implies, tricarb contains three carboxylic acid groups that
are ionized at physiological pH to give a small molecule with three
negative charges akin to the structure of the powerful chelating agent
EDTA. Magnesium is bound so tightly by tricarb that magnesium
deficiency results from overgrowth of tricarb-producing intestinal
bacteria in ruminants [181]. This condition, known as “grass tetany,”
is also accompanied by lower levels of calcium and zinc, all of which
can form divalent ion complexes with tricarb (Figure 6-21).
FIGURE 6-21. The Magnesium-tricarballylate Complex

CH
CH2 C
CH2
O O-
C C
O O- -O O
+ Mg+
209
Chapter 6

2, 3-Dihydroxyphenylpropionate (DHPP)
We have received numerous reports of patients with Clostridium
overgrowth confirmed by stool culture, where elevated levels of
DHPP have fallen to baseline with Flagyl, but were unaffected by
Nystatin. While other organisms may produce DHPP, Clostridia is
the most commonly encountered genera among those susceptible
to Flagyl. In vitro studies have confirmed the production of DHPP
from dietary quinolines by various species of Clostridia [182, 183].
Rats excrete DHPP when they are fed the naturally occurring
flavonoid hesperetin [184]. Depending on the species, clostridia
excrete various other organic acids as the end products of aromatic
amino acid metabolism [185]. Cytotoxic quinoid metabolites that
require glutathione conjugation for removal may be formed from
DHPP [186]. Various compounds closely related to DHPP also are
produced by the genus Clostridium [185]. In addition, DHPP has
been found as a product of Pseudomonas stutzeri isolated from sewage
by enrichment culture on quinoline [187, 188]. E. Coli produces an
enzyme to degrade DHPP, thus, helping to insure its survival in the
presence of intestinal Clostridial growth [189, 190].

D-Arabinitol
Among pathogenic yeasts and fungi, Candida spp. are of widest
clinical concern, because of their transmission by direct invasion of
the GI and GU tracts and their ability to rapidly overwhelm immune
responses in many hospitalized patients. Most species of candida
grow best on carbohydrate substrates. Activities of the enzymes
aldose reductase and xylitol dehydrogenase are induced in Candida
tenuis when the organism is grown on arabinose [191]. D-arabinitol
(DA) is a metabolite of most pathogenic Candida spp, in vitro as well
as in vivo. DA is a five-carbon sugar alcohol that can be assayed by
enzymatic analysis. The rate of arabinitol appearance in the body
from any source equals the urinary arabinitol excretion rate and

FIGURE 6-22. Nutrient Deficiency Due to Gut Fermentation

25
B1
Serum enzyme stimulation
or mineral concentration

20
B2
15 B6

10 Mg

Zn
5

0
Normals Fermentors
Ethanol=8.4 Ethanol=640

210
Organic Acids
is directly proportional to the concentration ratio of arabinitol to
creatinine in serum or urine [192].
Measuring serum D-arabinitol (DA) allows prompt diagnosis
of invasive candidiasis [193]. The somewhat more discriminating
elevated urine D-arabinitol/L-arabinitol (DA/LA) ratio has been
found to be a sensitive diagnostic marker for invasive candidiasis
in infants treated in neonatal intensive care units. Eight infants
with mucocutaneous candidiasis were given empiric antifungal
treatment ,but had negative cultures; five of these had repeatedly
elevated DA/LA ratios. Three infants with suspected and four with
confirmed invasive candidiasis had ratios to become normalized
during antifungal treatment [194]. The ratio of D- to L-arabinitol in
serum reveals the presence of disseminated candidiasis in immuno-
suppressed patients [195].
Immunocompromised patients with invasive candidiasis have
elevated DA/creatine ratios in urine. Positive DA results have been
obtained several days to weeks before positive blood cultures, and
the normalization of DA levels has been correlated with therapeutic
response in both humans and animals [195, 196].
The enzymatic method using D-arabinitol dehydrogenase is
precise (mean intra-assay coefficients of variation [CVs], 0.8%, and
mean interassay CVs, 1.6%) and it shows excellent recovery of added
DA (mean recovery rate, 101%) [197]. It also is noteworthy that there
are multiple negative impacts on nutrients because of dysbiosis.

TABLE 6-7. Upper Jejunal Bacterial Strains in Small Intestinal Bacterial


Overgrowth [98]

Antibiotic Sensitivity
% of Susceptible Strains
(See footnote for explanation)
Bacteria Prevalence (%) 1 2 3 4 5
Microaerophilic 100
Streptococcus 71 85 85 80 57 43
E. coli 69 63 76 76 ? ?
Straphylococcus 25 38 62 69 46 100
Micrococcus 22 75 75 75 87 50
Klebsiella 20 0 100 100 ? 100
Nisseria 16 100 100 100 100 30
Proteus 11 100 100 100 ? 100
Acinetobacter 9 0 0 0 ? 100
Enterobacter 7 0 0 0 ? 100

Anaerobic 93
Lactobacillus 75 89 100 ? 72 28
Bacteroides 29 16 100 16 42 63
Clostridium 25 90 100 ? 52 52
Veillonella 25 89 92 ? 13 61
Fusobacterium 13 80 100 ? 73 20
Peptostreptococcus 13 80 90 ? 60 5

1: Amoxicillin
2: Amoxicillin-Clavulanic Acid (Frequent digestive intolerance)
3: Cephalotin
4: Erythromycin
5: Trimethoprim-Sulfamethoxazol (Significant side effects)
211
Chapter 6
Lowered levels of vitamins B1, B2, B6, magnesium, and zinc are found
in patients with elevated nascent production of ethanol due to gut
fermentation [198] (Figure 6-22).

Actions to Consider for Elevated Dysbiosis


Markers
Although species identification is not possible from the limited
number of compounds currently detected, and no antibiotic
sensitivity testing can be done, the information available is
sufficient for clinical decisions about appropriate interventions.
The susceptibilities of various bacteria known to reside in the
upper jejunum to five antibiotics are shown in Table 6-7. Many
bacteriostatic herbal preparations and natural compounds inhibit
growth of bacterial and yeast. A summary of interventions commonly
used is presented in Table 6-8. Additional insight may be gained from
studies of the antibiotic sensitivities of organisms present in cases of
small intestinal bacterial overgrowth syndrome. Since the specimen
used for the data in Table 6-7 was taken in the upper jejunum [199],
the data represent only the proximal part of the small bowel where
microaerobic species are most likely to persist. The more distal regions
of the jejunum and the entire ileum are less favorable for aerobes.
Since the microbial compounds that appear in urine originate
in the lumen of the gut, oral sorbents that bind them and prevent
their absorption can be used. This approach is especially helpful
for those compounds of high toxicity like cresol. The absorbent,
AST-120 (3-7 gm/day), has been shown to effectively lower p-cresol
accumulation in uremic patients [155, 200]. Uremic coma and

TABLE 6-8. Intestinal Overgrowth Interventions

Class Examples

General Encourage high fiber diet, remove mucosal irritants such as


allergenic or IgG-positive foods, alcohol, and NSAIDs

Antibacterial Pharmaceutical: Amoxycillin/Clavulanic Acid


Natural: Goldenseal or other berberine-containing herbs, citrus
seed extract, garlic, uva ursi, aloe vera, glycyrrhiza, olive leaf
extract, garlic

Antifungal Pharmaceutical: Nystatin


Natural: See natural antibacterials plus oil of oregano, citrus seed
extract, garlic, uva ursi, undecylenic acid

Antiprotozoal Pharmaceutical: Flagyl


Natural: See natural antibacterials

Probiotic L. acidophilus, L. sporogenes, S. boulardii, favorable soil organisms

Prebiotic Fructo-oligosaccharide (FOS), increase use of raw and cooked


vegetables

Mucosal Glutamine, pantothenic acid, deglycyrrhizanated licorice, slippery


regeneration elm, oligopeptide preparations

212
Organic Acids
bleeding tendency is attributed to the accumulation of p-cresol, and
p-cresol is a cocarcinogen in the mouse skin test. The fact that an oral
sorbent lowers levels of cresol in uremia demonstrates that the gut is
a significant source of the toxin. Activated charcoal may be used for
this purpose, but there is greater tendency for constipation than with
AST-120.
It is worth noting that fasting is an effective way to reduce
microbial populations in the gut. If the resident microbes do not
receive a relatively constant infusion of substrate for growth, such as
carbohydrates or amino acids, they die. A discussion of the various
clinical ramifications of fasting is beyond the scope of this book.
Although many patients may be candidates for fasting [201], the
vast majority is reluctant to apply the discipline required to properly
execute severe food restriction.
Pharmaceutical and natural or herbal extract interventions are
summarized in Table 6-8. Regular oral dosing with organisms favorable
to the human gut (probiotics) and use of substrates that encourage their
growth such as dietary fiber and fructo-oligosaccharide (pre-biotics)
are generally implemented to achieve long term control of intestinal
microbes [202]. When aggressive intervention is warranted, use of
amoxicillin-clavulanic acid is a suitable candidate, because offending
microaerophilic bacteria are very susceptible to its antibacterial action
and the anaerobic populations decline as well, possibly due to the
more oxygen-rich environment produced by declining populations
of the oxygen-consuming species. The rising oxygen content can
have bactericidal effects on strictly anaerobic specie.

213
Chapter 6

References
[1] Sweetman, L. and J.C. Williams, Branched chain chain acyl-coenzyme A dehydrogenase. Pediatr
organic acidurias, in The Metabolic and Molecular Res, 1996. 39(6): p. 1059-66.
Bases of Inherited Disease, C. Scriver, et al., [15] McTague, J.A. and R. Forney, Jr., Jamaican
Editors. 1995, McGraw-Hill, Inc.: New York. vomiting sickness in Toledo, Ohio. Ann Emerg
p. 1387-1422. Med, 1994. 23(5): p. 1116-8.
[2] Tanake, K., et al., Gas-chromatographic method [16] Burlina, A.B., et al., A new syndrome with
of analysis for urinary organic acids. I. ethylmalonic aciduria and normal fatty acid
retention indices of 155 metabolically oxidation in fibroblasts. J Pediatr, 1994. 124(1):
important compounds. Clin Chem, 26((13)). p. 79-86.
1839-46, 1980. [17] Scholte, H.R., et al., Riboflavin-responsive
[3] Scriver, C., et al., The metabolic and molecular complex I deficiency. Biochim Biophys Acta,
bases of inherited disease. 7th ed. Vol. 1 - 3. 1271(1). 75-83, 1995.
1995, New York: McGraw-Hill. [18] Trauner, D.A., E. Horvath, and L.E. Davis,
[4] Scriver, C. and L. Rosenberg, Amino Acid Inhibition of fatty acid beta oxidation by
Metabolism and Its Disorders. Major problems influenza B virus and salicylic acid in mice:
in clinical pediatrics, ed. A. Schaffer. Vol. X. implications for Reye's syndrome. Neurology,
1973, Philadelphia: W. B. Saunders. 491. 38(2). 239-41, 1988.
[5] Nyhan, W., Abnormalities in amino acid [19] Wallace, D.C., Mitochondrial genetics: a
metabolism in clinical medicine. 1984, Norwald: paradigm for aging and degenerative diseases?
Appleton-Century-Crofts. 463. Science, 256(5057). 628-32, 1992.
[6] Lord, R. and J. Bralley, Organics in urine: [20] Bates, C.J., Liberation of 14CO2 from
Assessment of gut dysbiosis, nutrient [14C]adipic acid and [14C]octanoic acid by
deficiencies and toxemia. Nutr Pers, 20(4). 25- adult rats during riboflavin deficiency and its
31, 1997. reversal. Br J Nutr, 63(3). 553-62, 1990.
[7] Bralley, J. and R. Lord, Urinary organic acids [21] Naito, E., et al., Thiamine-responsive lactic
profiling, in Textbook of Natural Medicine, P.a. acidaemia: role of pyruvate dehydrogenase
Murray, Editor. 1998, Churchill Livingstone: complex. Eur J Pediatr, 157(8). 648-52, 1998.
Edinburgh. p. 229 - 237. [22] Gries, C.L. and M.L. Scott, The pathology of
[8] Ong, C.N., et al., Elevated levels of benzene- thiamin, riboflavin, pantothenic acid and
related compounds in the urine of cigarette niacin deficiencies in the chick. J Nutr, 102(10).
smokers. Int J Cancer, 59(2). 177-80, 1994. 1269-85, 1972.
[9] Goodwin, B.L., C.R. Ruthven, and M. Sandler, [23] Munujos, P., et al., Brain pyruvate oxidation in
Gut flora and the origin of some urinary experimental thiamin-deficiency encephalo-
aromatic phenolic compounds. Biochem pathy. Clin Chim Acta, 255(1). 13-25, 1996.
Pharmacol, 47(12). 2294-7, 1994. [24] Rao, G.A., D.E. Riley, and E.C. Larkin, Fatty
[10] Sweetman, L., Qualitative and quantitative liver caused by chronic alcohol ingestion is
analysis of organic acids in physiologic fluids prevented by dietary supplementation with
for diagnosis of the organic acidurias., in pyruvate or glycerol. Lipids, 19(8). 583-8, 1984.
Abnormalities in amino acid metabolism in [25] Chen, R.S., C.C. Huang, and N.S. Chu,
clinical medicine, W. Nyhan, Editor. 1984, Coenzyme Q10 treatment in mitochondrial
Appleton-Century-Crofts: Norwalk. p. 419-53. encephalomyopathies. Short-term double-
[11] Duez, P., A. Kumps, and Y. Mardens, GC-MS blind, crossover study. Eur Neurol, 37(4). 212-8,
profilng of urinary organic acids evaluated as a 1997.
quantitative method. Clin Chem, 42(10). 1609- [26] Sobreira, C., et al., Mitochondrial
1615, 1996. encephalomyopathy with coenzyme Q10
[12] Insinga, R.P., R.H. Laessig, and G.L. Hoffman, deficiency. Neurology, 48(5). 1238-43, 1997.
Newborn screening with tandem mass [27] Boitier, E., et al., A case of mitochondrial
spectrometry: examining its cost-effectiveness encephalomyopathy associated with a muscle
in the Wisconsin Newborn Screening Panel. coenzyme Q10 deficiency. J Neurol Sci, 156(1).
J Pediatr, 2002. 141(4): p. 524-31. 41-6, 1998.
[13] Feller, A.G. and D. Rudman, Role of carnitine [28] Fouty, B., F. Frerman, and R. Reves, Riboflavin
in human nutrition. J Nutr, 118(5). 541-547, to treat nucleoside analogue-induced lactic
1988. acidosis [letter]. Lancet, 352(9124). 291-2, 1998.
[14] Corydon, M.J., et al., Ethylmalonic aciduria is [29] Santos, J.L., et al., Nonsynergic effect of
associated with an amino acid variant of short ethanol and lead on heme metabolism in rats.
214
Organic Acids
Ecotoxicol Environ Saf, 43(1). 98-102, 1999. studies in a patient with cytochrome c oxidase
[30] Konrad, T., et al., alpha-Lipoic acid treatment deficiency. 39th ASMS Conf., 1991.
decreases serum lactate and pyruvate [43] Hamm, L.L. and E.E. Simon, Roles and
concentrations and improves glucose mechanisms of urinary buffer excretion. Am J
effectiveness in lean and obese patients with Physiol, 253(4 Pt 2). F595-605, 1987.
type 2 diabetes. Diabetes Care, 1999. 22(2): p. [44] Milner, J.A., Metabolic aberrations associated
280-7. with arginine deficiency. J Nutr, 115(4). 516-23,
[31] Bardosi, A., W. Cruetzfeldt, and et. al., Lactate, 1985.
hydroxybutyrate, and fumarate elevation in an [45] Prior, R.L., A. Zimber, and W.J. Visek, Citric,
adult with defective cytochrome c oxidase. Acta orotic, and other organic acids in rats injected
Neuropathol, 1987. 74(3): p. 248-58. with active or inactive urease. Am J Physiol,
[32] Bardosi, A., et al., Myo-, neuro-, gastrointestinal 228(3). 828-33, 1975.
encephalopathy (MNGIE syndrome) due to [46] al Aqeel, A., et al., A new patient with alpha-
partial deficiency of cytochrome-c-oxidase. A ketoglutaric aciduria and progressive
new mitochondrial multisystem disorder. Acta extrapyramidal tract disease. Brain Dev, 16. 33-
Neuropathol, 1987. 74(3): p. 248-58. 7, 1994.
[33] Oshida, Y., et al., Effect of insulin on [47] Shaw, D.e.a., Management of fatigue: A
intramuscular 3-hydroxybutyrate levels in physiological approach. Am J Med Sci. 243:758,
diabetic rats. Horm Metab Res, 30(2). 70-1, 1962.
1998. [48] Hicks, J., Treatment of fatigue in general
[34] Anderson, R.A., et al., Elevated intakes of practice: A double blind study. Clin Med. 85,
supplemental chromium improve glucose and 1964 (Jan.).
insulin variables in individuals with type 2 [49] Hume, A.S., et al., Antidotal efficacy of alpha-
diabetes. Diabetes, 46(11). 1786-91, 1997. ketoglutaric acid and sodium thiosulfate in
[35] Halberstam, M., et al., Oral vanadyl sulfate cyanide poisoning. J Toxicol Clin Toxicol, 33(6).
improves insulin sensitivity in NIDDM 721-4, 1995.
but not in obese nondiabetic subjects [50] Kawaguchi, A. and K. Bloch, Inhibition of
[published erratum appears in Diabetes 1996 glutamate dehydrogenase and malate
Sep;45(9):1285]. Diabetes, 45(5). 659-66, 1996. dehydrogenases by palmitoyl coenzyme A.
[36] Pandey, S.K., M.B. Anand-Srivastava, and A.K. J Biol Chem, 251(5). 1406-12, 1976.
Srivastava, Vanadyl sulfate-stimulated glycogen [51] Pinard, J.M., et al., Leigh syndrome and
synthesis is associated with activation of leukodystrophy due to partial succinate
phosphatidylinositol 3-kinase and is dehydrogenase deficiency: regression with
independent of insulin receptor tyrosine riboflavin. Arch Pediatr, 6(4). 421-6, 1999.
phosphorylation. Biochemistry, 37(19). 7006-14, [52] Nowaczyk, M.J., et al., Ethylmalonic and
1998. methylsuccinic aciduria in ethylmalonic
[37] Dissmann, R., T. Linderer, and R. Schroder, encephalopathy arise from abnormal isoleucine
Estimation of enzymatic infarct size: direct metabolism. Metabolism, 47(7). 836-9, 1998.
comparison of the marker enzymes creatine [53] Folkers, K., et al., Lovastatin decreases
kinase and alpha-hydroxybutyrate dehydro- coenzyme Q10 levels in humans. Proc Natl
genase. Am Heart J, 135(1). 1-9, 1998. Acad Sci U S A, 1990. 87(22): p. 8931-4.
[38] Silva, A.R., et al., Inhibition of in vitro CO2 [54] Mortensen, S.A., et al., Coenzyme Q10: clinical
production and lipid synthesis by 2-hydroxy- benefits with biochemical correlates suggesting
butyric acid in rat brain. Braz J Med Biol Res, a scientific breakthrough in the management
2001. 34(5): p. 627-31. of chronic heart failure. Int J Tissue React, 1990.
[39] Landaas, S., The formation of 2-hydroxybutyric 12(3): p. 155-62.
acid in experimental animals. Clin Chim Acta, [55] Zhao, Y., J. Jaskiewicz, and R.A. Harris, Effects
1975. 58(1): p. 23-32. of clofibric acid on the activity and activity
[40] Imaki, M., et al., Evaluation of the effects of state of the hepatic branched-chain 2-oxo acid
various factors on the serum alpha dehydrogenase complex. Biochem J, 285(Pt 1).
hydroxybutyrate dehydrogenase activity in 167-72, 1992.
young females. Appl Human Sci, 14(6). 297-302, [56] Tada, K., et al., Vitamin B6 dependent
1995. xanthurenic aciduria (the second report).
[41] Kano, K. and T. Ichimura, Increased alpha- Tohoku J Exp Med, 1968. 95(2): p. 107-14.
hydroxybutyrate dehydrogenase in serum from [57] Takeuchi, F., et al., Kynurenine metabolism
children with measles. Clin Chem, 38(5). 624-7, and xanthurenic acid formation in vitamin B6-
1992. deficient rat after tryptophan injection. J Nutr
[42] Matsumoto, I., et al., Urinary organic acid profile Sci Vitaminol (Tokyo), 1989. 35(2): p. 111-22.

215
Chapter 6
[58] Kosters, W.W. and M. Kirchgessner, Effect of evidence on causality from a meta-analysis.
varying vitamin B6 intake of early-weaned Bmj, 2002. 325(7374): p. 1202.
piglets on urinary xanthurenic and kynurenic [74] Marin, G.H., J. Tentoni, and G. Cicchetti,
acid excretion, serum transaminase activity and [Megaloblastic anemia: rapid and economical
urea concentration. Int J Vitam Nutr Res, 1976. study]. Sangre (Barc), 1997. 42(3): p. 235-8.
46(3): p. 373-80. [75] Shojania, A.M., Oral contraceptives: effect of
[59] Bender, D.A., et al., The effects of oestrogen folate and vitamin B12 metabolism. Can Med
administration on tryptophan metabolism Assoc J, 1982. 126(3): p. 244-7.
in rats and in menopausal women receiving [76] Blocker, D.E. and S.W. Thenen, Intestinal
hormone replacement therapy. Biochem absorption, liver uptake, and excretion of
Pharmacol, 1983. 32(5): p. 843-8. 3H-folic acid in folic acid-deficient, alcohol-
[60] Guthrie, H., Introductory Nutrition. Fourth ed. consuming nonhuman primates. Am J Clin
1979, St. Louis: C.V. Mosby. Nutr, 1987. 46(3): p. 503-10.
[61] Nyhan, W.L., Inborn errors of biotin [77] Title, L.M., et al., Effect of folic acid and
metabolism. Arch Dermatol, 1987. 123(12): p. antioxidant vitamins on endothelial
1696-1698a. dysfunction in patients with coronary artery
[62] Mock, N., et al., Increased urinary excretion of disease. J Am Coll Cardiol, 2000. 36(3): p. 758-
3-hydroxyisovaleric acid and decreased urinary 65.
excretion of biotin are sensitive early indicators [78] Loria, C.M., et al., Serum folate and cardio-
of decreased biotin status in experimental vascular disease mortality among US men and
biotin deficiency. Am J Clin Nutr, 1997. 65: p. women. Arch Intern Med, 2000. 160(21): p.
951-8. 3258-62.
[63] Dostalova, L., Vitamin status during [79] Shrubsole, M.J., et al., Dietary folate intake and
puerperium and lactation. Ann Nutr Metab, breast cancer risk: results from the Shanghai
1984. 28(6): p. 385-408. Breast Cancer Study. Cancer Res, 2001. 61(19):
[64] Krause, K.H., et al., Vitamin status in patients p. 7136-41.
on chronic anticonvulsant therapy. Int J Vitam [80] Sellers, T.A., et al., Dietary folate intake,
Nutr Res, 1982. 52(4): p. 375-85. alcohol, and risk of breast cancer in a
[65] Mock, D.M., et al., Biotin status assessed prospective study of postmenopausal women.
longitudinally in pregnant women. J Nutr, Epidemiology, 2001. 12(4): p. 420-8.
1997. 127(5): p. 710-6. [81] Sengelov, H., et al., Inter-relationships
[66] Riudor, E., et al., Requirement of high biotin between single carbon units’ metabolism and
doses in a case of biotinidase deficiency. J resting energy expenditure in weight-losing
Inherit Metab Dis, 1989. 12(3): p. 338-9. patients with small cell lung cancer. Effects of
[67] Lindenbaum, J., et al., Prevalence of cobalamin methionine supply and chemotherapy. Eur J
deficiency in the Framingham elderly Cancer, 1994. 30A(11): p. 1616-20.
population. Am J Clin Nutr, 60(1). 2-11, 1994. [82] Stolzenberg-Solomon, R.Z., et al., Dietary and
[68] Rule, S.A., et al., Serum vitamin B12 and other methyl-group availability factors and
transcobalamin levels in early HIV disease. Am pancreatic cancer risk in a cohort of male
J Hematol, 47(3). 167-71, 1994. smokers. Am J Epidemiol, 2001. 153(7): p. 680-
[69] Roon-Djordjevic, B.v. and S. Cerfontain-van, 7.
Urinary excretion of histidine metabolites as [83] Bergmark, C., et al., Hyperhomocysteinemia in
an indication for folic acid and vitamin B12 patients operated for lower extremity ischaemia
deficiency. Clin Chim Acta, 1972. 41: p. 55-65. below the age of 50--effect of smoking and
[70] Armstrong, P., et al., Nitrous oxide and extent of disease. Eur J Vasc Surg, 1993. 7(4): p.
formiminoglutamic acid: excretion in surgical 391-6.
patients and anaesthetists. Br J Anaesth, 1991. [84] Chowers, Y., et al., Increased levels of
66(2): p. 163-9. homocysteine in patients with Crohn’s disease
[71] Everman, B.W. and D.D. Koblin, Aging, chronic are related to folate levels. Am J Gastroenterol,
administration of ethanol, and acute exposure 2000. 95(12): p. 3498-502.
to nitrous oxide: effects on vitamin B12 and [85] Campbell, B.A., Megaloblastic anemia in
folate status in rats. Mech Aging Dev, 1992. pregnancy. Clin Obstet Gynecol, 1995. 38(3): p.
62(3): p. 229-43. 455-62.
[72] Hine, R.J., Folic acid: Contemporary clinical [86] Orgacka, H. and Z. Zbytniewski, [Excretion
perspective. Persp Appl Nutr, 1993. 93(1): p. 3- of vanillic acid and homovanillic acid and
14. tissue distribution of catecholamines and their
[73] Wald, D.S., M. Law, and J.K. Morris, metabolites in mice with various levels of
Homocysteine and cardiovascular disease: pigmentation]. Endokrynol Pol, 1991. 42(3): p.

216
Organic Acids
471-9. [101] Visser, J., et al., CD4 T lymphocytes from
[87] Braverman, E. and C. Pfeiffer, Tyrosine: The patients with chronic fatigue syndrome have
antidepressant, in The Healing Nutrients Within. decreased interferon-gamma production and
1987, Keats: New Canaan. p. 44-58. increased sensitivity to dexamethasone. J Infect
[88] Mauron, J., Tyrosine and hypertension. Bibl Dis, 1998. 177(2): p. 451-4.
Nutr Dieta, 1986. 38: p. 209-18. [102] Kerr, J.R., et al., Circulating tumor necrosis
[89] Akimaru, K., et al., Amplification of N-myc factor-alpha and interferon-gamma are
gene and increase of urinary VMA and HVA in detectable during acute and convalescent
patients with neuroblastic tumors. Nippon Ika parvovirus B19 infection and are associated
Daigaku Zasshi, 1994. 61(2): p. 148-53. with prolonged and chronic fatigue. J Gen
[90] Lucas, K., et al., Catecholamine metabolites Virol, 2001. 82(Pt 12): p. 3011-9.
in ganglioneuroma. Med Pediatr Oncol, 1994. [103] Rejdak, K., et al., Decreased level of kynurenic
22(4): p. 240-3. acid in cerebrospinal fluid of relapsing-onset
[91] Parekh, N., et al., Cardiac troponin I predicts multiple sclerosis patients. Neurosci Lett, 2002.
myocardial dysfunction in aneurysmal 331(1): p. 63.
subarachnoid hemorrhage [In Process Citation]. [104] Brouwers, P., et al., Quinolinic acid in
J Am Coll Cardiol, 2000. 36(4): p. 1328-35. the cerebrospinal fluid of children with
[92] Keung, W.M., et al., Volitional ethanol symptomatic human immunodeficiency virus
consumption affects overall serotonin type 1 disease: relationships to clinical status
metabolism in Syrian golden hamsters and therapeutic response. J Infect Dis, 1993.
(Mesocricetus auratus). Biochem Biophys Res 168(6): p. 1380-6.
Commun, 2000. 271(3): p. 823-30. [105] Achim, C.L., M.P. Heyes, and C.A. Wiley,
[93] Rabinoff, M., Short note: possible role of Quantitation of human immunodeficiency
macrophage metabolic products including virus, immune activation factors, and
quinolinic acid and neopterin in the quinolinic acid in AIDS brains. J Clin Invest,
pathogenesis of inflammatory brain diseases. 1993. 91(6): p. 2769-75.
Med Hypotheses, 1994. 42(2): p. 133-4. [106] Rejdak, R., et al., Changes of kynurenic acid
[94] Kekesi, G., et al., The antinociceptive effect of content in the rat and chicken retina during
intrathecal kynurenic acid and its interaction ontogeny. Graefes Arch Clin Exp Ophthalmol,
with endomorphin-1 in rats. Eur J Pharmacol, 2002. 240(8): p. 687-91.
2002. 445(1-2): p. 93-6. [107] Farmer, B.T., 2nd and D.A. Butterfield,
[95] Erhardt, S. and G. Engberg, Increased Quinolinic acid, an endogenous metabolite
phasic activity of dopaminergic neurones with neurotoxic properties, alters the physical
in the rat ventral tegmental area following state of membrane proteins in human
pharmacologically elevated levels of erythrocytes. Life Sci, 1984. 35(5): p. 501-9.
endogenous kynurenic acid. Acta Physiol Scand, [108] Heyes, M.P., et al., Quinolinic acid and
2002. 175(1): p. 45-53. kynurenine pathway metabolism in
[96] Pulkkinen, M.O., J. Salminen, and S. Virtanen, inflammatory and non-inflammatory
Serum vitamin B6 in pure pregnancy neurological disease. Brain, 1992. 115(Pt 5): p.
depression. Acta Obstet Gynecol Scand, 1978. 1249-73.
57(5): p. 471-2. [109] Stone, T.W., Kynurenic acid antagonists and
[97] Lelord, G., E. Callaway, and J.P. Muh, Clinical kynurenine pathway inhibitors. Expert Opin
and biological effects of high doses of vitamin Investig Drugs, 2001. 10(4): p. 633-45.
B6 and magnesium on autistic children. Acta [110] Ishiwata, K., et al., Metabolic studies with L-[1-
Vitaminol Enzymol, 1982. 4(1-2): p. 27-44. 14C]tyrosine for the investigation of a kinetic
[98] Santhosh-Kumar, C.R., et al., Are model to measure protein synthesis rates with
neuropsychiatric manifestations of folate, PET. J Nucl Med, 1988. 29(4): p. 524-9.
cobalamin and pyridoxine deficiency mediated [111] Muting, D., et al., Urinary p-
through imbalances in excitatory sulfur amino hydroxyphenyllactic acid as indicator of
acids? Med Hypotheses, 1994. 43(4): p. 239-44. hepatic encephalopathy in patients with
[99] Shor-Posner, G., et al., Impact of vitamin hepatic cirrhosis [letter]. Lancet, 1985. 2(8468):
B6 status on psychological distress in a p. 1365-6.
longitudinal study of HIV-1 infection. Int J [112] Mayatepek, E., C.K. Seppel, and G.F. Hoffmann,
Psychiatry Med, 1994. 24(3): p. 209-22. Increased urinary excretion of dicarboxylic
[100] Gupta, S. and B. Vayuvegula, A comprehensive acids and 4- hydroxyphenyllactic acid in
immunological analysis in chronic fatigue patients with Zellweger syndrome. Eur J Pediatr,
syndrome. Scand J Immunol, 1991. 33(3): p. 1995. 154(9): p. 755-6.
319-27. [113] Matsuo, M., et al., Citrullinaemia: an infantile

217
Chapter 6
form with p-hydroxyphenylpyruvic and p- breast cancer? Arch Environ Contam Toxicol,
hydroxyphenyllactic acidurias. J Inherit Metab 2001. 41(3): p. 386-95.
Dis, 1987. 10(3): p. 276. [126] Yoshida, R., et al., Moderate alcohol
[114] Markaverich, B.M., et al., Methyl p-hydroxy- consumption reduces urinary 8-
phenyllactate. An inhibitor of cell growth and hydroxydeoxyguanosine by inducing of uric
proliferation and an endogenous ligand for acid. Ind Health, 2001. 39(4): p. 322-9.
nuclear type-II binding sites. J Biol Chem, 1988. [127] Kanauchi, M., H. Nishioka, and T. Hashimoto,
263(15): p. 7203-10. Oxidative DNA damage and tubulointerstitial
[115] Markaverich, B.M., et al., Methyl p-hydroxy- injury in diabetic nephropathy. Nephron, 2002.
phenyllactate and nuclear type II binding 91(2): p. 327-9.
sites in malignant cells: metabolic fate and [128] Pilger, A., et al., Urinary excretion of 8-
mammary tumor growth. Cancer Res, 1990. hydroxy-2’-deoxyguanosine measured by
50(5): p. 1470-8. high-performance liquid chromatography
[116] Sheffield, L.G. and L.C. Kotolski, Epidermal with electrochemical detection. J Chromatogr B
growth factor modulates cholera toxin induced Analyt Technol Biomed Life Sci, 2002. 778(1-2):
mammary gland development. Endocr Res, p. 393-401.
1993. 19(4): p. 259-71. [129] Visek, W.J., Nitrogen-stimulated orotic acid
[117] Levchuk, A.A., et al., [Effect of the carcinogenic synthesis and nucleotide imbalance. Cancer Res,
tyrosine metabolite p-hydroxyphenyllactic 52(7 Suppl). 2082s-2084s, 1992.
acid on the ascorbic acid concentration in the [130] Milner, J.A. and W.J. Visek, Orotate, citrate,
organs and blood of mice]. Biull Eksp Biol Med, and urea excretion in rats fed various levels of
1986. 102(10): p. 462-3. arginine. Proc Soc Exp Biol Med, 147(3). 754-9,
[118] Baikova, V.N., et al., [Congenital disorders 1974.
of tyrosine metabolism and their correction [131] Nelson, J., et al., Regulation of orotic acid
in children with tumors]. Vopr Onkol, 1987. biosynthesis and excretion induced by oral
33(11): p. 42-8. glutamine administration in mice. Biochem Med
[119] Kitada, T., et al., In situ detection of oxidative Metab Biol, 49(3). 338-50, 1993.
DNA damage, 8-hydroxydeoxyguanosine, in [132] Triebig, G., K.H. Schaller, and D. Weltle,
chronic human liver disease. J Hepatol, 2001. Neurotoxicity of solvent mixtures in spray
35(5): p. 613-8. painters. I. Study design, workplace exposure,
[120] Stewart, R.J., et al., Antioxidant status of and questionnaire. Int Arch Occup Environ
young children: response to an antioxidant Health, 64(5). 353-9, 1992.
supplement. J Am Diet Assoc, 2002. 102(11): p. [133] Walaszek, Z., et al., Metabolism, uptake, and
1652-7. excretion of a D-glucaric acid salt and its
[121] Irie, M., et al., Relationships between perceived potential use in cancer prevention. Cancer
workload, stress and oxidative DNA damage. Detect Prev, 1997. 21(2): p. 178-90.
Int Arch Occup Environ Health, 2001. 74(2): p. [134] Mulder, G.J., Conjugation reactions in drug
153-7. metabolism: an integrated approach: substrates, co-
[122] Lee, H.C., et al., Concurrent increase of substrates, enzymes and their interactions in vivo
oxidative DNA damage and lipid peroxidation and in vitro. 1990, London; New York: Taylor &
together with mitochondrial DNA mutation Francis. x, p. 413.
in human lung tissues during aging--smoking [135] Timbrell, J., Principles of biochemical toxicology.
enhances oxidative stress on the aged tissues. 3rd ed. 2000, London ; New York: Taylor &
Arch Biochem Biophys, 1999. 362(2): p. 309-16. Francis. ix, p. 394.
[123] Kouda, K., et al., The relationship [136] Anders, M. and W. Dekant, eds. Conjugation-
of oxidative DNA damage marker 8- dependent carcinogenicity and toxicity of foreign
hydroxydeoxyguanosine and glycoxidative compounds. 1994, Academic Press: New York.
damage marker pentosidine. Clin Biochem, [137] Hunter, J. and et al., Urinary D-glucaric
2001. 34(3): p. 247-50. acid excretion as a test for hepatic enzyme
[124] Gackowski, D., et al., Further evidence that induction in man. Lancet, (March 20). 572-575,
oxidative stress may be a risk factor responsible 1971.
for the development of atherosclerosis. Free [138] Hunter, J., et al., Urinary D-glucaric acid
Radic Biol Med, 2001. 31(4): p. 542-7. excretion and total liver content of cytochrome
[125] Charles, M.J., et al., Organochlorines and P-450 in guinea-pigs: relationship during
8-hydroxy-2’-deoxyguanosine (8-OHdG) in enzyme induction and following inhibition of
cancerous and noncancerous breast tissue: do protein synthesis. Biochem Pharmacol, 22(6).
the data support the hypothesis that oxidative 743-7, 1973.
DNA damage caused by organochlorines affects [139] Sandle, L.N. and J.M. Braganza, An evaluation

218
Organic Acids
of the low-pH enzymatic assay of urinary D- Xenobiotica, 1993. 23(12): p. 1427-33.
glucaric acid, and its use as a marker of enzyme [153] Bures, J., et al., Excretion of phenol and p-
induction in exocrine pancreatic disease cresol in the urine in fasting obese individuals
[published erratum appears in Clin Chim Acta and in persons treated with total enteral
1990 May;188(3):279]. Clin Chim Acta, 162(3). nutrition. Cas Lek Cesk, 129(37). 1166-71,
245-56, 1987. 1990.
[140] Edwards, J.W. and B.G. Priestly, Effect of [154] Tamm, A.O., Biochemical activity of intestinal
occupational exposure to aldrin on urinary D- microflora in adult celiac disease. Nahrung,
glucaric acid, plasma dieldrin, and lymphocyte 28(6-7). 711-5, 1984.
sister chromatid exchange. Int Arch Occup [155] Niwa, T., et al., Suppressive effect of an oral
Environ Health, 66(4). 229-34, 1994. sorbent on the accumulation of p-cresol in the
[141] Hogue, C.J. and M.A. Brewster, The potential of serum of experimental uremic rats. Nephron,
exposure biomarkers in epidemiologic studies 65(1). 82-7, 1993.
of reproductive health. Environ Health Perspect, [156] Barker, J.L. and J.W. Frost, Microbial synthesis
90. 261-9, 1991. of p-hydroxybenzoic acid from glucose.
[142] Bland, J., J. Bralley, and S. Rigden. Management Biotechnol Bioeng, 2001. 76(4): p. 376-90.
of chronic fatigue symptoms by tailored [157] Zhivotnikova, N.V., [Changes in bioenergy
nutritional intervention using a program processes in rat liver mitochondria
designed to support hepatic detoxification. in after exposure to diglycidyl ether of p-
HealthComm. 1997. Gig Harbor, WA. hydroxybenzoic acid]. Gig Sanit, 1990(11): p.
[143] Marsh, C.A., Biosynthesis of D-glucaric acid in 20-2.
mammals: a free-radical mechanism? Carbohydr [158] Fellaman, J.H., N.R. Buist, and N.G. Kennaway,
Res, 1986. 153(1): p. 119-31. Pitfalls in metabolic studies: the origin of
[144] Chalmers, R. and A. Lawson, Organic acidurias urinary p-tyramine. Clin Biochem, 10(5). 168-
due to disorders in other metabolic pathways, 70, 1977.
in Organic Acids in Man. 1982, Chapman & [159] Powell-Jackson, P.R., et al., Intestinal bacterial
Hall: London. p. 350 - 381. metabolism of protein and bile acids: role in
[145] Levy, G., Sulfate conjugation in drug pathogenesis of hepatic disease after jejuno-
metabolism: role of inorganic sulfate. Fed Proc, ileal bypass surgery. Br J Surg, 1979. 66(11): p.
45(8). 2235-40, 1986. 772-5.
[146] Gregus, Z., et al., Effect of glutathione [160] Tohyama, K., et al., Effect of lactobacilli on
depletion on sulfate activation and sulfate ester urinary indican excretion in gnotobiotic rats
formation in rats. Biochem Pharmacol, 37(22). and in man. Microbiol Immunol, 1981. 25(2): p.
4307-12, 1988. 101-12.
[147] Chalmers, R.A., H.B. Valman, and M.M. [161] Yoshida, K. and C. Hirayama, Tryptophan
Liberman, Measurement of 4-hydroxyphenyl- metabolism in liver cirrhosis: influence of oral
acetic aciduria as a screening test for small- antibiotics on neuro-psychiatric symptoms.
bowel disease. Clin Chem, 25(10). 1791-4, 1979. Tohoku J Exp Med, 1984. 142(1): p. 35-41.
[148] Ward, L.A., et al., Isolation from swine feces of [162] Miloszewski, K., et al., Increase in urinary
a bacterium which decarboxylates indican excretion in pancreatic steatorrhoea
p-hydroxyphenylacetic acid to 4-methylphenol following replacement therapy. Scand J
(p-cresol). Appl Environ Microbiol, 53(1). 189-92, Gastroenterol, 1975. 10(5): p. 481-5.
1987. [163] Lawrie, C.A., A.G. Renwick, and J. Sims, The
[149] Lindblad, B.S., et al., Absorption of biological urinary excretion of bacterial amino-acid
amines of bacterial origin in normal and sick metabolites by rats fed saccharin in the diet.
infants. Ciba Found Symp, (70). 281-91, 1979. Food Chem Toxicol, 1985. 23(4-5): p. 445-50.
[150] Bennett, M.J., et al., When do gut flora in the [164] Kirkland, J.L., E. Vargas, and M. Lye, Indican
newborn produce 3-phenylpropionic acid? excretion in the elderly. Postgrad Med J, 1983.
Implications for early diagnosis of medium- 59(697): p. 717-9.
chain acyl-CoA dehydrogenase deficiency. Clin [165] Smith, D.F., Effects of age on serum tryptophan
Chem, 38(2). 278-81, 1992. and urine indican in adults given a tryptophan
[151] Quick, A., The study of benzoic acid load test. Eur J Drug Metab Pharmacokinet, 1982.
conjugation in the dog with a direct 7(1): p. 55-8.
quantitative method from hippuric acid. J Biol [166] Aarbakke, J. and H. Schjonsby, Value of urinary
Chem. 477-90, 1934. simple phenol and indican determinations in
[152] Temellini, A., et al., Conjugation of benzoic the diagnosis of the stagnant loop syndrome.
acid with glycine in human liver and kidney: Scand J Gastroenterol, 1976. 11(4): p. 409-14.
a study on the interindividual variability. [167] Stachow, A., S. Jablonska, and A.

219
Chapter 6
Skiendzielewska, Intestinal absorption of Microbial metabolism of quinoline and related
L-tryptophan in sclero-derma. Acta Derm compounds. II. Degradation of quinoline by
Venereol, 1976. 56(4): p. 257-64. Pseudomonas fluorescens 3, Pseudomonas
[168] Mayer, P. and W. Beeken, The role of urinary putida 86 and Rhodococcus spec. B1. Biol Chem
indican as a predictor of bacterial colonization Hoppe Seyler, 1989. 370(11): 1183-1189.
in the human jejunum. Am J Dig Dis, 1975. [183] Konishi, Y, Kobayashi S, Microbial metabolites
20(11): p. 1003-9. of ingested caffeic acid are absorbed by the
[169] Patney, N.L., et al., Urinary indican excretion monocarboxylic acid transporter (MCT) in
in cirrhosis of liver. J Assoc Physicians India, intestinal Caco-2 cell monolayers. J Agric Food
1976. 24(5): p. 291-5. Chem, 2004. 52(21): 6418-6424.
[170] Montgomery, R.D., et al., The aging gut: a [184] Kim, HK, Jeong TS, Lee MK, et al., Lipid-
study of intestinal absorption in relation to lowering efficacy of hesperetin metabolites in
nutrition in the elderly. Q J Med, 1978. 47(186): high-cholesterol fed rats. Clin Chim Acta, 2003.
p. 197-24. 327(1-2): 129-137.
[171] Hudson, M., R. Pocknee, and N.A. Mowat, D- [185] Elsden, SR, Hilton MG, Waller JM, The end
lactic acidosis in short bowel syndrome--an products of the metabolism of aromatic amino
examination of possible mechanisms. Q J Med, acids by Clostridia. Arch Microbiol, 1976.
1990. 74(274): p. 157-63. 107(3): 283-288.
[172] Dahlquist, N.R., et al., D-Lactic acidosis [186] Moridani, MY, Scobie H, Jamshidzadeh A,
and encephalopathy after jejunoileostomy: et al., Caffeic acid, chlorogenic acid, and
response to overfeeding and to fasting in dihydrocaffeic acid metabolism: glutathione
humans. Mayo Clin Proc, 1984. 59(3): p. 141-5. conjugate formation. Drug Metab Dispos, 2001.
[173] Halverson, J., A. Gale, and C. Lazarus, D-lactic 29(11): 1432-1439.
acidosis and other complications of intestinal [187] Shukla, OP, Microbial transformation of
bypass surgery. Arch Intern Med, 1984. 144(2): quinoline by a Pseudomonas sp. Appl Environ
p. 357-60. Microbiol, 1986. 51(6): 1332-1342.
[174] Smith, S.M., R.H. Eng, and F. Buccini, Use of D- [188] Shukla, OP, Microbiological degradation
lactic acid measurements in the diagnosis of of quinoline by Pseudomonas stutzeri: the
bacterial infections. J Infect Dis, 1986. 154(4): coumarin pathway of quinoline catabolism.
p. 658-64. Microbios, 1989. 59(238): 47-63.
[175] Thurn, J.R., et al., D-lactate encephalopathy. [189] Spence, EL, Kawamukai M, Sanvoisin J, et
Am J Med, 1985. 79(6): p. 717-21. al., Catechol dioxygenases from Escherichia
[176] Slyter, L.L. and T.S. Rumsey, Effect of coliform coli (MhpB) and Alcaligenes eutrophus (MpcI):
bacteria, feed deprivation, and pH on sequence analysis and biochemical properties
ruminal D-lactic acid production by steer or of a third family of extradiol dioxygenases. J
continuous-culture microbial populations Bacteriol, 1996. 178(17): 5249-5256.
changed from forage to concentrates. J Anim [190] Bugg, TD, Overproduction, purification and
Sci, 1991. 69(7): p. 3055-66. properties of 2,3-dihydroxyphenylpropionate
[177] Coronado, B.E., S.M. Opal, and D.C. Yoburn, 1,2-dioxygenase from Escherichia coli. Biochim
Antibiotic-induced D-lactic acidosis. Ann Intern Biophys Acta, 1993. 1202(2): 258-264.
Med, 1995. 122(11): p. 839-42. [191] Kern, M., et al., Induction of aldose reductase
[178] Caldarini, M.I., et al., Abnormal fecal flora in a and xylitol dehydrogenase activities in Candida
patient with short bowel syndrome. An in vitro tenuis CBS 4435. FEMS Microbiol Lett, 1997.
study on effect of pH on D-lactic acid pro- 149(1): p. 31-7.
duction. Dig Dis Sci, 1996. 41(8): p. 1649-52. [192] Wong, B., et al., The arabinitol appearance rate
[179] Halperin, M.L. and K.S. Kamel, D-lactic in laboratory animals and humans: estimation
acidosis: turning sugar into acids in the gastro- from the arabinitol/creatine ratio and relevance
intestinal tract. Kidney Int, 1996. 49(1): p. 1-8. to the diagnosis of candidiasis. J Infect Dis,
[180] McDevitt, J. and P. Goldman, Effect of the 1982. 146(3): p. 353-9.
intestinal flora on the urinary organic acid [193] Tokunaga, S., et al., Clinical significance of
profile of rats ingesting a chemically simplified measurement of serum D-arabinitol levels in
diet. Food Chem Toxicol, 29(2). 107-13, 1991. candiduria patients. Urol Int, 1992. 48(2): p. 195-
[181] Schwartz, R., M. Topley, and J.B. Russell, Effect 9.
of tricarballylic acid, a nonmetabolizable [194] Sigmundsdottir, G., B. Shristensson, et. al.
rumen fermentation product of trans-aconitic (2000). “Urine D-arabinitol/L-arabinitol ratio
acid, on Mg, Ca and Zn utilization of rats. J in diagnosis of invasive candidiasis in newborn
Nutr, 118(2). 183-8, 1988. infants.” J Clin Microbiol 38(8): 3039-42.
[182] Schwarz, G, Bauder R, Speer M, et al., [195] Roboz, J., Diagnosis and monitoring of dis-

220
Organic Acids
seminated candidiasis based on serum/urine D/
L-arabinitol ratios. Chirality, 1994. 6(2): p. 51-7.
[196] Christensson, B., G. Sigmundsdottir, and L.
Larsson, D-arabinitol--a marker for invasive
candidiasis. Med Mycol, 1999. 37(6): p. 391-6.
[197] Yeo, S.F., et al., A rapid, automated enzymatic
fluorometric assay for determination of D-
arabinitol in serum. J Clin Microbiol, 2000.
38(4): p. 1439-43.
[198] Eaton, K.K., et al., Abnormal gut fermation:
Laboratory studies reveal deficiency of B
vitamins, zinc, and magnesium. J Nutr Biochem,
4. 635-638, 1993.
[199] Bouhnik, Y., et al., Bacterial populations
contaminating the upper gut in patients
with small intestinal bacterial overgrowth
syndrome. Am J Gastroenterol, 94(5). 1327-31,
1999.
[200] Sanaka, T., et al., Therapeutic effects of oral
sorbent in undialyzed uremia. Am J Kidney
Dis, 12(2). 97-103, 1988.
[201] Furhman, J., Fasting and Eating for Health. 1995,
New York: St. Martin’s Griffin.
[202] Bouhnik, Y., et al., Effects of fructooligo-
saccharides ingestion on fecal bifidobacteria
and selected metabolic indexes of colon
carcinogenesis in healthy humans. Nutr Cancer,
26(1). 21-9, 1996.

221
222

You might also like