Download as doc, pdf, or txt
Download as doc, pdf, or txt
You are on page 1of 45

Return to top

The rhizome (root) of turmeric ( Curcuma longa Linn.) has long been used in traditional Asian
medicine to treat gastrointestinal upset, arthritic pain, and "low energy." Laboratory and animal
research has demonstrated anti-inflammatory, antioxidant, and anti-cancer properties of turmeric
and its constituent curcumin. Preliminary human evidence, albeit poor quality, suggests possible
efficacy in the management of dyspepsia (heartburn), hyperlipidemia (high cholesterol), and
scabies (when used on the skin).

SynonymsReturn to top

Amomoum curcuma, anlatone (constituent), ar-tumerone, CUR, Curcuma , Curcuma aromatica ,


Curcuma aromatica salisbury, Curcuma domestica , Curcuma domestica valet, Curcuma
longa , Curcuma longa Linn, Curcuma longa rhizoma, curcuma oil, curcumin,
diferuloylmethane, E zhu, Gelbwurzel, gurkemeje, haldi, Haridra, Indian saffron, Indian yellow
root, jiang huang, kunir, kunyit, Kurkumawurzelstock, kyoo, NT, number ten, Olena, radix zedoaria
longa, rhizome de curcuma, safran des Indes, sesquiterpenoids, shati, tumeric, turmeric oil,
turmeric root, tumerone (constituent), Ukon, yellowroot, zedoary, Zingiberaceae (family),
zingiberene, Zitterwurzel.

EvidenceReturn to top

These uses have been tested in humans or animals. Safety and effectiveness have not always
been proven. Some of these conditions are potentially serious, and should be evaluated by a
qualified healthcare provider.

Uses based on scientific evidence Grade*

Blood clot prevention


Early research suggests that turmeric may prevent the
C
formation of blood clots. However, more research is needed
before turmeric can be recommended for these conditions.

Cancer
Several early animal and laboratory studies report anti-cancer
(colon, skin, breast) properties of curcumin. Many mechanisms
have been considered, including antioxidant activity, anti-
C
angiogenesis (prevention of new blood vessel growth), and
direct effects on cancer cells. Currently it remains unclear if
turmeric or curcumin has a role in preventing or treating human
cancers. There are several ongoing studies in this area.

Cognitive function
Curcumin has been shown to have antioxidant and anti-
inflammatory properties and to reduce beta-amyloid and plaque
C
burden in lab studies. However, there is currently not enough
evidence to suggest the use of curcumin for cognitive
performance.

Dyspepsia (heartburn) C
Turmeric has been traditionally used to treat stomach problems
(such as indigestion from a fatty meal). There is preliminary
evidence that turmeric may offer some relief from these
stomach problems. However, at high doses or with prolonged
use, turmeric may actually irritate or upset the stomach.
Reliable human research is necessary before a
recommendation can be made.

Gallstone prevention/bile flow stimulant


It has been said that there are fewer people with gallstones in
India, which is sometimes credited to turmeric in the diet. Early
studies report that curcumin, a chemical in turmeric, may C
decrease the occurrence of gallstones. However, reliable
human studies are lacking in this area. The use of turmeric may
be inadvisable in patients with active gallstones.

High cholesterol
Early studies suggest that turmeric may lower levels of low-
density lipoprotein ("bad cholesterol") and total cholesterol in C
the blood. Better human studies are needed before a
recommendation can be made.

HIV/AIDS
Several laboratory studies suggest that curcumin, a component
C
of turmeric, may have activity against HIV. However, reliable
human studies are lacking in this area.

Inflammation
Laboratory and animal studies show anti-inflammatory activity
C
of turmeric and its constituent curcumin. Reliable human
research is lacking.

Irritable bowel syndrome (IBS)


Irritable bowel syndrome (IBS) is a common functional disorder
for which there are limited reliable medical treatments. One
C
study investigated the effects of Curcuma xanthorriza on IBS
and found that treatment did not show any therapeutic benefit
over placebo. More studies are needed to verify these findings.

Liver protection
In traditional Indian Ayurvedic medicine, turmeric has been
used to tone the liver. Early research suggests that turmeric C
may have a protective effect on the liver, but more research is
needed before any recommendations can be made.

Oral leukoplakia
Results from lab and animal studies suggest turmeric may have
C
anticancer effects. Large, well-designed human studies are
needed before a recommendation can be made.

Osteoarthritis
Turmeric has been used historically to treat rheumatic
conditions. Laboratory and animal studies show anti-
C
inflammatory activity of turmeric and its constituent curcumin,
which may be beneficial in people with osteoarthritis. Reliable
human research is lacking.

Peptic ulcer disease (stomach ulcer) C


Turmeric has been used historically to treat stomach and
duodenal ulcers. However, at high doses or with prolonged use,
turmeric may actually further irritate or upset the stomach.
Currently, there is not enough human evidence to make a firm
recommendation.

Rheumatoid arthritis
Turmeric has been used historically to treat rheumatic
conditions and based on animal research may reduce C
inflammation. Reliable human studies are necessary before a
recommendation can be made in this area.

Scabies
Historically, turmeric has been used on the skin to treat chronic
skin ulcers and scabies. It has also been used in combination
C
with the leaves of the herb Azadirachta indica ADR or "neem."
More research is necessary before a firm recommendation can
be made.

Uveitis (eye inflammation)


Laboratory and animal studies show anti-inflammatory activity
of turmeric and its constituent curcumin. A poorly designed
C
human study suggests a possible benefit of curcumin in the
treatment of uveitis. Reliable human research is necessary
before a firm conclusion can be drawn.

Viral infection
Evidence suggests that turmeric may help treat viral infections.
However, there is not enough human evidence in this area. C
Well-designed trials are needed to determine if these claims are
true.
*Key to grades
A: Strong scientific evidence for this use;
B: Good scientific evidence for this use;
C: Unclear scientific evidence for this use;
D: Fair scientific evidence against this use;
F: Strong scientific evidence against this use.

Grading rationale

Uses based on tradition or theory


The below uses are based on tradition or scientific theories. They often have not been thoroughly
tested in humans, and safety and effectiveness have not always been proven. Some of these
conditions are potentially serious, and should be evaluated by a qualified healthcare provider.

Abdominal bloating, Alzheimer's disease, antibacterial, antifungal, antimicrobial, antispasmodic,


anti-venom, appetite stimulant, asthma, boils, breast milk stimulant, bruises, cataracts,
chemoprotective, contraception, cough, cystic fibrosis, diabetes, diarrhea, dizziness, epilepsy,
flavoring agent, gas, gonorrhea, heart damage from doxorubicin (Adriamycin®, Doxil®),
Helicobacter pylori infection, hepatitis, high blood pressure, histological dye, human
papillomavirus (HPV), hypoglycemic agent (blood sugar lowering), infections (methicillin-resistant
Staphylococcus aureus), insect bites, insect repellent, jaundice, kidney disease, kidney stones,
leprosy, liver damage from toxins/drugs, male fertility, menstrual pain, menstrual period
problems/lack of menstrual period, multidrug resistance, neurodegenerative disorders, pain,
parasites, ringworm, scarring, scleroderma, weight reduction.
DosingReturn to top

The below doses are based on scientific research, publications, traditional use, or expert opinion.
Many herbs and supplements have not been thoroughly tested, and safety and effectiveness may
not be proven. Brands may be made differently, with variable ingredients, even within the same
brand. The below doses may not apply to all products. You should read product labels, and
discuss doses with a qualified healthcare provider before starting therapy.

Adults (over 18 years old)


Doses used range from 450 milligrams of curcumin capsules to 3 grams of turmeric root daily,
divided into several doses, taken by mouth. As a tea, 1 to 1.5 grams of dried root may be steeped
in 150 milliliters of water for 15 minutes and taken twice daily. Average dietary intake of turmeric
in the Indian population may range between 2 to 2.5 grams, corresponding to 60 to 200
milligrams of curcumin daily. A dose of 0.6 milliliters of turmeric oil has been taken three times
daily for one month and a dose of 1 milliliter in three divided doses has been taken for two
months.
One reported method for treating scabies is to cover affected areas once daily with a paste
consisting of a 4:1 mixture of Azadirachta indica ADR ("neem") to turmeric, for up to 15 days.
Scabies should be treated under the supervision of a qualified healthcare professional.
Children (under 18 years old)
There is no proven or safe medicinal dose of turmeric in children.

SafetyReturn to top

The U.S. Food and Drug Administration does not strictly regulate herbs and supplements. There
is no guarantee of strength, purity or safety of products, and effects may vary. You should always
read product labels. If you have a medical condition, or are taking other drugs, herbs, or
supplements, you should speak with a qualified healthcare provider before starting a new
therapy. Consult a healthcare provider immediately if you experience side effects.

Allergies
Allergic reactions to turmeric may occur, including contact dermatitis (an itchy rash) after skin or
scalp exposure. People with allergies to plants in the Curcuma genus are more likely to have an
allergic reaction to turmeric. Use cautiously in patients allergic to turmeric or any of its
constituents (including curcumin), to yellow food colorings, or to plants in the Zingiberaceae
(ginger) family.
Side Effects and Warnings
Turmeric may cause an upset stomach, especially in high doses or if given over a long period of
time. Heartburn has been reported in patients being treated for stomach ulcers. Since turmeric is
sometimes used for the treatment of heartburn or ulcers, caution may be necessary in some
patients. Nausea and diarrhea have also been reported.
Based on laboratory and animal studies, turmeric may increase the risk of bleeding. Caution is
advised in patients with bleeding disorders or taking drugs that may increase the risk of bleeding.
Dosing adjustments may be necessary. Turmeric should be stopped prior to scheduled surgery.
Limited animal studies show that a component of turmeric, curcumin, may increase liver function
tests. However, one human study reports that turmeric has no effect on these tests. Turmeric or
curcumin may cause gallbladder squeezing (contraction) and may not be advised in patients with
gallstones. In animal studies, hair loss (alopecia) and lowering of blood pressure have been
reported. In theory, turmeric may weaken the immune system and should be used cautiously in
patients with immune system deficiencies.
Turmeric should be used with caution in people with diabetes or hypoglycemia or people taking
drugs or supplements that lower blood sugar.
Pregnancy and Breastfeeding
Historically, turmeric has been considered safe when used as a spice in foods during pregnancy
and breastfeeding. However, turmeric has been found to cause uterine stimulation and to
stimulate menstrual flow and caution is therefore warranted during pregnancy. Animal studies
have not found turmeric taken by mouth to cause abnormal fetal development.

InteractionsReturn to top

Most herbs and supplements have not been thoroughly tested for interactions with other herbs,
supplements, drugs, or foods. The interactions listed below are based on reports in scientific
publications, laboratory experiments, or traditional use. You should always read product labels. If
you have a medical condition, or are taking other drugs, herbs, or supplements, you should
speak with a qualified healthcare provider before starting a new therapy.

Interactions with Drugs


Based on laboratory and animal studies, turmeric may inhibit platelets in the blood and increase
the risk of bleeding caused by other drugs. Some examples include aspirin, anticoagulants
("blood thinners") such as warfarin (Coumadin®) or heparin, anti-platelet drugs such as
clopidogrel (Plavix®), and non-steroidal anti-inflammatory drugs such as ibuprofen (Motrin®,
Advil®) or naproxen (Naprosyn®, Aleve® ).
Based on animal data, turmeric may lower blood sugar and therefore may have additive effects
with diabetes medications.
In animals, turmeric protects against stomach ulcers caused by non-steroidal anti-inflammatory
drugs (NSAIDs) such as indomethacin (Indocin®) and against heart damage caused by the
chemotherapy drug doxorubicin (Adriamycin®).
Turmeric may lower blood pressure levels and may have an additive effect if taken with drugs that
also lower blood pressure.
Turmeric may lower blood levels of low-density lipoprotein (LDL or "bad" cholesterol) and
increase high-density lipoprotein (HDL or "good" cholesterol). Thus, turmeric may increase the
effects of cholesterol-lowering drugs such as lovastatin (Mevacor®) or atorvastatin (Lipitor®).
Based on animal studies, turmeric may interfere with the way the body processes certain drugs
using the liver's "cytochrome P450" enzyme system. As a result, the levels of these drugs may be
increased in the blood and may cause increased effects or potentially serious adverse reactions.
Patients using any medications should check the package insert and speak with a healthcare
professional or pharmacist about possible interactions.
When taken with indomethacin or reserpine, turmeric may help reduce the number of stomach
and intestinal ulcers normally caused by these drugs. However, when taken in larger doses or
when used for long periods of time, turmeric itself can cause ulcers.
Interactions with Herbs and Dietary Supplements
Based on animal studies, turmeric may increase the risk of bleeding when taken with herbs and
supplements that are believed to increase the risk of bleeding. Multiple cases of bleeding have
been reported with the use of Ginkgo biloba , some cases with garlic, and fewer cases with saw
palmetto.
Based on animal data, turmeric may lower blood sugar. Individuals taking other herbs or
supplements or diabetes medications should speak with a healthcare professional before starting
turmeric.
Turmeric may lower blood levels of low-density lipoprotein (LDL or "bad" cholesterol) and
increase high-density lipoprotein (HDL or "good" cholesterol). Thus, turmeric may increase the
effects of cholesterol-lowering herbs or supplements such as fish oil, garlic, guggul, or niacin.
Based on animal studies, turmeric may interfere with the way the body processes certain herbs or
supplements using the liver's "cytochrome P450" enzyme system. As a result, the levels of other
herbs or supplements may become too high in the blood. It may also alter the effects that other
herbs or supplements possibly have on the P450 system.
Turmeric may lower blood pressure and may therefore have an additive effect if taken with herbs
that also lower blood pressure.
Methodology Return to top

This information is based on a professional level monograph edited and peer-reviewed by


contributors to the Natural Standard Research Collaboration (www.naturalstandard.com): Winnie
Abramson, ND (Natural Standard Research Collaboration); E-P Barrette, MD (Case Western
Reserve University School of Medicine); Ethan Basch, MD (Memorial Sloan-Kettering Cancer
Center); Michael Bodock, RPh (Massachusetts General Hospital); Heather Boon, B.Sc.Phm, PhD
(University of Toronto); Dawn Costa, BA, BS (Natural Standard Research Collaboration); Sadaf
Hashmi, MD (Johns Hopkins School of Hygiene and Public Health); Jenna Hollenstein, MS, RD
(Natural Standard Research Collaboration); George Papaliodis, MD (Massachusetts Eye and Ear
Infirmary); Michael Smith, MRPharmS, ND (Canadian College of Naturopathic Medicine); Shaina
Tanguay-Colucci, BS (Natural Standard Research Collaboration); Catherine Ulbricht, PharmD
(Massachusetts General Hospital); Mamta Vora (Northeastern University); Wendy Weissner, BA
(Natural Standard Research Collaboration).

Methodology details

Selected references Return to top

1. Aggarwal BB, Kumar A, Bharti AC. Anticancer potential of curcumin: preclinical and clinical studies. Anticancer
Res 2003;23(1A):363-398.
2. Brinkhaus B, Hentschel C, Von Keudell C, et al. Herbal medicine with curcuma and fumitory in the treatment of
irritable bowel syndrome: a randomized, placebo-controlled, double-blind clinical trial. Scand J Gastroenterol
2005 Aug;40(8):936-43.
3. Deodhar SD, Sethi R, Srimal RC. Preliminary study on antirheumatic activity of curcumin (diferuloyl methane).
Indian J Med Res 1980;71:632-634.
4. Egan ME, Pearson M, Weiner S, et al. Curcumin, a major constituent of turmeric, corrects cystic fibrosis
defects. Science 4-23-2004;304(5670):600-602.
5. Kositchaiwat C, Kositchaiwat S, Havanondha J. Curcuma longa Linn. in the treatment of gastric ulcer
comparison to liquid antacid: a controlled clinical trial. J Med Assoc Thai 1993;76(11):601-605.
6. Kulkarni RR, Patki PS, Jog VP, et al. Treatment of osteoarthritis with a herbomineral formulation: a double-blind,
placebo-controlled, cross-over study. J Ethnopharmacol 1991;33(1-2):91-95.
7. Limtrakul P, Anuchapreeda S, Buddhasukh D. Modulation of human multidrug-resistance MDR-1 gene by
natural curcuminoids. BMC Cancer 4-17-2004;4(1):13.
8. Ng TP, Chiam PC, Lee T, et al. Curry consumption and cognitive function in the elderly. Am J Epidemiol 2006
1;164(9):898-906.
9. Nishiyama T, Mae T, Kishida H, et al. Curcuminoids and sesquiterpenoids in turmeric (Curcuma longa L.)
suppress an increase in blood glucose level in type 2 diabetic KK-Ay mice. J Agric Food Chem 2-23-
2005;53(4):959-963.
10. Prusty BK, Das BC. Constitutive activation of transcription factor AP-1 in cervical cancer and suppression of
human papillomavirus (HPV) transcription and AP-1 activity in HeLa cells by curcumin. Int J Cancer 3-1-
2005;113(6):951-960.
11. Rithaporn T, Monga M, Rajasekaran M. Curcumin: a potential vaginal contraceptive. Contraception
2003;68(3):219-223.
12. Taher MM, Lammering G, Hershey C, et al. Curcumin inhibits ultraviolet light induced human immunodeficiency
virus gene expression. Mol Cell Biochem 2003;254(1-2):289-297.
13. Thamlikitkul V, Bunyapraphatsara N, Dechatiwongse T, et al. Randomized double blind study of Curcuma
domestica Val. for dyspepsia. J Med Assoc Thai 1989;72(11):613-620.
14. Tilak JC, Banerjee M, Mohan H, et al. Antioxidant availability of turmeric in relation to its medicinal and culinary
uses. Phytother.Res 2004;18(10):798-804.
15. Van Dau N, Ngoc Ham N, Huy Khac D, et al. The effects of a traditional drug, turmeric (Curcuma longa), and
placebo on the healing of duodenal ulcer. Phytomed 1998;5(1):29-34.

Elemental speciation in human health risk


assessment, Environmental Health Criteria 234 (World
Health Organization, Geneva) 2007. 238 pages. Price:
CHF 30.00/US $ 30.00; in developing countries: CHF
21.00/US$ 18.90
ISBN 92-4-157234-5
This book is based on a draft prepared by WHO
Task Group on Environmental Health Criteria for
Elemental Speciation in Human Health Risk
Assessment under the supervision of Dr Inge
Mangelsdorf of Fraunhofer Institute, Hanover,
Germany and Dr A. Aitio, International Programme on
Chemical Safety, WHO, Geneva, Switzerland, held on
November 15-18, 2005 at Fraunhofer Institute of
Toxicology and Experimental Medicine, Hanover,
Germany.
The book is directed at risk assessors and regulators,
to emphasize the importance of consideration of
speciation which is hardly taken into consideration as
a part of most hazard and risk assessments. Further, the
book points at the significance of analysis of speciation
of elements to increase knowledge on the effect of
speciation on mode of action and also to increase
understanding of health effects. Speciation is the
evaluation of chemical form and distribution of the
elements, which play an important role in their toxicity
and bioavailability. The areas of speciation analysis
have undergone a continued evolution and development
for the last two decades. The rapid development of
inorganic instrumental analysis mainly driven by the
development of atomic spectrometry enabled the
analysts to look into the role of trace elements in
different areas, such as health and environment,
geochemistry and material sciences, to name some. This
book provides a comprehensive description of the major
areas involved in elemental speciation with the
Book Reviews
Indian J Med Res 127, April 2008, pp 417-420
417
following chapters: structural aspects of speciation;
analytical techniques and methodology; bioaccessibility
and bioavailability; toxicokinetics and biological
monitoring; molecular and cellular mechanisms of
metal toxicity; and health effects. In addition, individual
chapters are devoted to elemental speciation and
mechanism of actions of most of the elements
(chromium, manganese, iron, zinc, cobalt, nickel,
copper, arsenic, selenium, cadmium, mercury and lead)
in detail. Apart from this, role of tin, barium, palladium,
platinum, thallium is also discussed in health effects
chapter. The book contains an important chapter
entitled, “Structural aspects of speciation” where it
clearly explained electronic and oxidation states,
nuclear (isotopic) composition, inorganic compounds
and complexes, organometallic compounds, organic and
macromolecular complexes of various elemental
species. The book successfully pointed out that the
distribution, mobility and biological availability of
chemical elements depends not simply on their
concentration but, critically, on the chemical and
physical association which they undergo in natural
systems. However, the book does not point out some
of the error sources in analysis of speciation, e.g.,
incomplete extraction from solid samples, degradation
reactions, matrix-dependent recovery of extraction, lack
of adequate internal standards, matrix effects, poor
sensitivity of analytical techniques, etc. Actually the
most difficult area of speciation remains the provision
of a truly representative sample and the ability to
transfer this to a suitable measurement technique whilst
ensuring that the speciation profile remains intact.
Fundamental understanding of trace element
behaviour, the realistic formulation of historical
perspectives of trace element contamination, an
assessment of environmental transformation processes
and a thorough appraisal of environment-related health
problems and diseases are well documented in this book.
418 INDIAN J MED RES, APRIL 2008
The book explains the toxicokinetics and
toxicodynamics of elements influenced by carriers,
valence state and isotopes, particle size, element ligands,
organic versus inorganic element species and
biotransformations with resultant health effects.
The book also reflects on understanding of some
of the most important forms of an element, the
transformation between forms and various
consequences in terms of risk assessment, toxicity or
biological activity. Different elemental species might
not only differ quantitatively in their characteristics
(e.g., toxicity) but also qualitatively (toxic versus
essential). While Cr (III) compounds do have some
positive biological activity and are therefore considered
to be essential, Cr (VI) compounds are carcinogenic.
Inorganic As (III) trioxide (As2O3) is also a carcinogenic
but it was found to be effective in the treatment of certain
forms of leukaemia. The methylated forms of arsenic
are found to be far less toxic than the inorganic arsenic
compounds. Methylated mercury, by contrast, is much
more toxic than inorganic mercury. The book
successfully highlighted molecular mechanisms of
metal toxicity and carcinogenesis in respective chapters.
Chronic exposure of many heavy metals and metalderivatives
is associated with an increased risk of cancer,
although the mechanisms of tumour genesis are largely
unknown. Major areas of focus in this book include
exposure assessment and biomarker identification, role
of ROS or oxidative stress in carcinogenesis,
mechanisms of metal-induced DNA damage, metal
signaling, metal-protein interactions and the effects of
different elemental species on immune system.
In conclusion, this is an excellent reference book
and presents a comprehensive insight on the
toxicological evaluation of elemental species and their
intended usage pattern and possible human exposure,
and provides an extensive bibliography. The text is
clearly written and presents a vast amount of sound
technical information. This book should be useful to
all toxicologists, scientists, medical practitioners
involved in regulatory agencies and is a good guide to
those who are engaged in basic medical research on
heavy metals or trace elements.
Kusal K. Das
Environmental Health Research Unit
Department of Physiology
Al Ameen Medical College
Bijapur 586 108, India
e-mail: kusaldas@yahoo.com
Turmeric: The salt of the orients is the spice of life,
Kamala Krishnaswamy (Allied Publishers Private
Limited, New Delhi) 2007. 248 pages. Price: Rs.350/-
ISBN 81-8424-126-7
Spices have been used since times immemorial for
culinary purposes but the discovery of their
physiological properties and therapeutic potential is
much more recent. Several spices had been used in
traditional systems of medicine, mainly for preventive
purposes. The most important among them undoubtedly
is turmeric (rhizome of Curcuma longa Linn). About
30 species of Curcuma have been described and several
of these are used for food colouring and flavouring but
C. longa is most extensively used. Turmeric occupies
an unique place among the spices since besides being
used for culinary and medicinal use it is also employed
extensively as a cosmetic, colouring agent and
preservative.
The medicinal use of turmeric is mentioned even
among the Vedas and elaborated in classical Ayurvedic
texts like the Charaka Samhita. A systematic
experimental and clinical evaluation of its medicinal
properties, however, has been done largely from the
middle of the last century. Major attention has centered
round curcumin and related curcuminoids which
constitute 2-5 per cent of the biomass of the rhizome
and are responsible for its characteristic yellow colour.
Curcumin free extracts also exhibit biological activity.
There have been several reviews on pharmacology
and chemistry of C. longa including a monograph in
WHO Monographs on Selected Medicinal Plants,
Volume I (1999). The present book is perhaps the first
book to comprehensively cover all aspects of the plant,
including the historical development, botany, chemistry,
pharmacology and uses in medical and other fields.
The opening chapter has reviewed the traditional
uses vis-à-vis modern concepts of functional foods and
nutraceuticals. It has highlighted the need to provide
evidence information to support its traditional uses. It
provides justification for turmeric being considered the
world’s ‘most important herb’ today and lists about 20
medicinal uses in countries from all parts of the world
even though the plant is a native of tropical and
subtropical regions. The chapter also illustrates the wide
variation in chemical composition of various varieties
of turmeric. Thus, the curcumin content varies from
2.8 (Krishna) to 9.3 per cent (Roma, Suroma), oleoresins
from 3.8 (Krishna) to 16.2 per cent (IISR Pratibha) and
essential oil from 2 (Krishna) to 7 per cent (Suvarna,
Sudarsana). These may be responsible partly for the
varying results obtained with the crude extracts by
different investigators.
The next chapter describes the chemical and
nutritional composition of turmeric. The chemical
description mainly covers curcumin and some other
curcuminoids. An important omission is any mention
of the pharmacologically active peptide turmerin. The
yield of this 5-KDa water soluble cyclic peptide is 0.1
per cent and it contains 40 amino acid residues.
The remaining chapters of the book review the
major pharmacological activities, mainly of
curcuminoids , and related clinical and pharmacokinetic
studies. The activities reviewed include antiinflammatory,
anti-carcinogenic anti-mutagenic, antioxidant,
anti-atherosclerotic and some other minor
effects. The experimental data have been reviewed in
chapters 3-11. The basic mechanisms underlying the
pathological processes like inflammation,
carcinogenesis, atherosclerosis, etc., have been
reviewed in adequate detail thereby helping to locate
the possible sites of action of curcumin, its analogues
and metabolites.
Curcuminoids act on multiple sites in each of these
conditions and some of these sites are common. These
include kinins like PKC and tyrosine kinase, TNF, NFkappa
B, cyclic D, protein API, STAT family agents,
etc. They play important role in mutagenesis, tumour
initiation, transformation, progression and metastasis.
Pathogenic changes in vasculature are involved in
different ways in growth and differentiation of tumours,
wound healing, immune responses and thrombotic
episodes. Curcumin suppresses proliferation of normal,
transformed and malignant cells by modulating growth
factors and induces apoptosis in a variety of tumour
and other cells. The mechanisms include mitochondrial
dependent and independent pathways. An important
observation evident from the review is the potentiation
of effects of anti-cancer drugs like cis-platinum by
curcumin.
The prostaglandins are growth factors for
inflammation and angiogenesis and perform a house
keeping function. Curcumin modulates their activity by
acting on the arachidonic acid metabolism. Other
mechanisms involved in its anti-inflammatory activity
include effect on lysosomal enzymes and membranes,
inhibition of adhesion molecules and cytokine
production by inhibition of NF-Kappa B target genes.
The mechanism of other effects like hypolipidaemic,
wound healing, anti-infective activity, etc., has also been
similarly reviewed.
The author has reviewed the extensive data
exhaustively but has not commented on the procedures
used or the results obtained. Some of the studies have
employed very high amounts of the test substances like
5 per cent in the diet and up to 5 mg/ml in some in vitro
studies. Results of such studies must be interpreted with
caution.
The clinical studies have been reviewed in a
separate chapter and their inadequacies, and in many
cases poor design of the trial, have been clearly brought
out. It may have been better if the clinical studies had
been given along with experimental studies on the
condition and permitted better correlation. Still the
clinical data provide enough evidence for
chemopreventive and therapeutic potential of
curcuminoids in malignancy and several chronic
conditions.
Chronic disorders including degenerative
(atherosclerosis, Alzheimer’s disease), proliferative
(cancer) or inflammatory (rheumatoid arthritis,
chronic infections) conditions share cellular/
subcellular biochemical, molecular regulatory and
pathological events including abnormal redox sites,
cytokines, apoptotic mechanisms, etc. The book
elegantly brings out how the effect of curcuminoids
on basic modulators of these processes is responsible
for their beneficial effects in such diverse conditions.
It helps to identify potential targets on which future
work may help in developing novel chemical entities
for the management of these chronic conditions. It
also highlights the therapeutic potential of the
polypeptide tumerin which has not received adequate
attention. Similarly data on bioavailability studies
suggest the possibility of improving it by combining
with piperine.
The studies reviewed in the book also clearly
demonstrate that the safety and excellent tolerance of
even large doses of curcuminoids and turmeric extracts
enhance their utility in preventive health programmes
and as nutraceuticals. The author aptly points out (p
237) the paucity of in vivo bioefficacy data in terms of
their concentration and synergistic or additive effects
with other bioactive compounds. She has also
emphasized (p 298) the impact food-based approach
for enhancing the intake of such phytochemicals may
have on the onset and progression of several chronic
diseases. The approach may also provide means of
BOOK REVIEW 419
‘improving and prolonging the success of standard
therapies.
The book is extensively documented and covers
references of papers published up to 2004. There are
many patents on the curcuminoids and analogues, etc.,
but they have not been so well covered. The book has
been profusely illustrated with black and white
photographs, bar diagrams, etc. It would have been
better to provide coloured photograph, for example, of
the plant (Fig. 1) or the tissues (Fig. 6.1) The legends
of some of the figures (e.g. Fig. 7.3, 14.4) could have
been more explicit. Similarly the splitting of some of
the figures (e.g., 4.3) and tables (e.g., 5.3) should have
been avoided.
These minor aberrations, however, detract very little
from the merits of a timely and authoritative document
on biomedical aspects of turmeric. This well produced
monograph will be a useful reference book not only for
research workers and clinicians but also those involved
in economic utilization of turmeric and its value added
products in pharmaceuticals, cosmetics, etc.
B.N. Dhawan
3, Ramakrishan Marg
Faizabad Road
Lucknow 226 001, India
e-mail: bndhawan@hotmail.com
Influenza and its global public health significance,
T. M. Mathew & T. Mathew, editors (Thejma Publishers,
West Orange, NJ, USA) 2006. 203 pages. Price: $ 33.99
ISBN: 0-9727597-5-1
This book published in 2006 is aimed for the public,
and government authorities dealing with the influenza
epidemic, researchers, scientists working on the
treatment and control of influenza during the impending
threat of influenza pandemic. It provides a
comprehensive overview of epidemic and pandemic
influenza. The detailed information about all the facts
related to influenza has been elaborated chapter-wise
precisely making comprehensive overview of epidemic
and is user friendly and ready referenced. It provides
an educational and reference tool for students as well
as a practical guide to all those involved in the activities
related to influenza.
Overall, the book contains 13 chapters. The chapter
on history of influenza pandemics describes each
pandemic nicely in a tabular form which can be helpful
in studying the strain pattern. The structure of the virus
has been explained in detail but a diagrammatic
representation of the virus would have contributed more,
making it more understandable, particularly for the
students.
Chapter on influenza C highlights its growth
properties, which are different from influenza A & B as
well as the factors influencing it. Chapter on avian
influenza focuses on all the outbreaks and describes
the receptor specificity of influenza viruses which helps
in human transmission. Also, the different perspectives
of avian influenza in canines, equines and swine have
been explained in different chapters.
Diagnosis of avian influenza and all the
significant investigations have been outlined nicely
and briefly. The author also describes the experience
of infection and isolation of influenza virus in her
family during the 1972 - 1973 epidemics in Madras
(now Chennai). The interesting fact is that infection
was brought to the house by her husband who
contracted the disease through his friends who had
respiratory illness while the author did not get
respiratory infection in spite of working with the
respiratory viruses.
The steps to be followed for the outbreak
investigation starting from descriptive epidemiology,
the diagnosis with clinical features of the illness,
determining the aetiology, etc. are described. Also
included is the case study on influenza like outbreak,
which can help in better understanding and analysis of
the subject, particularly for the students.
In short, it is a handy book but no new information
has been added, there is repetition of the material in
different chapters.
Shashi Khare
Department of Microbiology
National Institute of Communicable
Diseases
22 Sham Nath Marg
Delhi 110 054, India
420 INDIAN J MED RES, APRIL 2008
©2009.Al Ameen Charitable Fund Trust, Bangalore 73
SHORT COMMUNICATION Al Ame en J Med S c i (2 00 9 )2 (1 ) :7 3 -7 7
Studies on Arsenic Toxicity in Male Rat Gonads and its
Protection by High Dietary Protein Supplementation
Sanjit Mukherjee and Prabir K.Mukhopadhyay*
Department of Physiology, Presidency College, Kolkata-700 073, India
Abstract: Arsenic was given orally to rats as arsenic tri oxide, 3mg /kg body wt/day
in a
single dose for 28 consecutive days. This treatment in male Wistar rats caused
increase in
seminiferous tubular luminal size coupled with reduced accumulation of
spermatozoa, and
signs of necrotic changes with disarray in cellular organization. Other significant
changes
were decrease in sperm count, viability and motility (p<0.001). On high protein diet
(containing pea 37gm/100 gm of diet and casein 9gm/100gm of diet)
supplementation along
with same arsenic exposure caused partial restoration of normalcy. All these sperm
physiological changes and altered gonadal features, both histomorphometric and
histological
observations, were found significantly ameliorated. Results of this study propose that
high
protein diet supplementation may be effective to recovery from the toxic effect of
arsenic on
male gonad of rat.
Introduction
It has become evident that increasing human activities have modified the
global cycle
of heavy metals and metalloids, including the toxic non-essential elements
like
arsenic (As) [1]. Among these metals, arsenic exhibits a complex metabolism
and is
possibly the most abundant and potential carcinogen [2]. Arsenic is present
in the
nature in stable form as As 5+ species, and As 3+species. An analysis of 25000
tube
wells in West Bengal reveals that the average As concentration reaches to
0.3mg/lt of
water, and in some places even the concentration reaches up to 3mg/lt of
water,
where 0.05mg/lt is the permissible limit for drinking water as per WHO [3]. In
the
process of arsenic metabolism, inorganic arsenic is methylated to
monomethyl
arsonic acid (MMA) and finally to dimethyl arsinic acid (DMA) followed by a
renal
excretion. In this process of biomethylation, constant depletion of methyl
causes
DNA hypomethylation, and thus generates mutation followed by
carcinogenesis [2].
Arsenic affects the mitochondrial enzymes, impairs the cellular respiration
and
causes cellular toxicity. It can also substitute phosphate intermediates, which
could
theoretically slow down the rate of metabolism and interrupt the production
of
energy [4]. Male infertility is reflected by low sperm count, low sperm motility
and
bad quality of sperms (4). Sodium arsenite has been found to have an
inhibitory
effect on the activity of testicular steroidogenic enzyme ∆5­3β­hydroxysteroid
dehydrogenase (∆5­3β­HSD) and 17ß-hydroxysteroid dehydrogenase (17ß-
HSD)
and to reduce the weight of testes and accessory sex glands [4] in rat. High
Arsenic
level may suppress the sensitivity of gonadotroph cells to GnRH as well as
gonadotropin secretion by elevating plasma levels of glucocorticoids. These
ultimately lead to the development of gonadal toxicity [4-5]. In recent
studies, dietary
proteins have been found to have antioxidant activities [6-8]. Wheat
(Puccinia
graminis tritici) and pea (Pisum sativum) are both good sources of dietary
plant
protein, while casein is an animal protein. The antioxidant activities of pea,
wheat,
Al Ame en J Med S c i Volume2, No.1, 2009 Mukherjee S & Mukhopadhyay PK
©2009. Al Ameen Charitable Fund Trust, Bangalore 74
and casein has been studied using different liposomal models and the results
show a
minimization in lipid peroxidation, thus preventing the damage produced by
the free
radicals [6]. Our present study has been done to test the causal relationship
between
arsenic generated oxidative stress and testicular cell damage using rat as a
model
animal and further to examine whether dietary high protein interventions
may be an
effective strategy of detoxification that may help in preventing the disorders
induced
by arsenic.
Materials and Methods:
Animal selection and drug treatment: Normal (Wistar) male rats weighing
between
120gm—140gm were selected for our experiment. Animals were maintained
in an
environmentally controlled animal house (temperature 24 ± 3°C) and in a 12 h
light/dark schedule with free access to water. For experiments, rats were
randomly
selected into three groups consisting six rats in each: group A, control; group
B,
arsenic-treated; and group C, arsenic + pea + casein-diet supplemented. The
animals
of groups A and B were provided with a control diet composed of 71%
carbohydrate,
18% protein, 7% fat and 4% salt mixture and vitamins [9]. For chronic oral
exposure
to arsenic, a dose was selected (3 mg/kg body wt/day), which is within the
range of
LD50 of a 70-kg body wt human (1–4 mg/kg) and lesser than one-thirteenth of
LD50
value of rats (40 mg/kg) [6]. Accordingly, animals of groups B, and C were
orally
treated with aqueous solution of arsenic trioxide, 3 mg/kg body wt/day for 28
days.
The animals of group C, in addition, were supplemented with pea (37 g/100 g
of
diet), which contributed 8.5% protein, and casein (9 g/100 g of diet) which
contributed additional 9% protein in the formulation of a high protein (27%)
diet [6].
To overcome the impact of any altered food intake, control (group A) animals
were
pair-fed with other experimental groups B, and C.
1. Preparation of permanent slide for histological study of testes: Testes from
all
groups of animals were dissected out and were Bouin’s-fixed. Paraffin blocks
were
prepared, and 4–5 ∝m thin sections were cut with a high precision microtome
(IEC
Microtome, USA) and routine microscopic slides were prepared. For staining,
standard Haematoxylin/Eosin method was followed to study both the
histomorphometric and histological alterations.
2. Sperm viability count: Immediately after the sacrifice the cauda portion of
epididymis was cut. The cauda was kept in 1 ml diluent [10]. This was kept
for 5
mins at 37oC. It was then taken out, and an incision was given through the
cauda and
the sperms were dispersed in the fluid. 40µl of this spermatozoal suspension
was
transferred to an eppendorf. This was stained with EosinY and Nigrosin (40µl
each).
This was mixed gently and 25µl from the mixture was taken on a grease free
slide
and a smear was drawn and dried. The number of viable and non-viable
sperms was
counted under light microscope.
3. Sperm count: From the dispersed spermatozoal suspension 25µl was
charged on
a Neubauer Haemocytometer [11] and the numbers of sperms were counted
and
calculated using WBC chambers.
4. Sperm motility count: The cauda epididymis from all three groups was
obtained
as mentioned earlier, and each cauda was kept in 1ml PBS 0.2 M, pH 7.4 at
37°C
[12]. 25µl of this was taken on a clean slide covered with a cover slip and was
Al Ame en J Med S c i Volume2, No.1, 2009 Mukherjee S & Mukhopadhyay PK
©2009. Al Ameen Charitable Fund Trust, Bangalore 75
observed under a microscope. Total numbers of motile sperms per 100
sperms were
counted. The readings were taken at the beginning of 1st hour, 2nd hour and
3rd hour
of the experiment (the temperature of the spermatozoal suspension was
maintained at
37 o C throughout the process).
5. Statistical Analysis: The data were expressed as mean ± SEM (Standard
error of
mean). For statistical analysis, the quantitative data of each parameter from
the
different groups were analyzed by Student’s “t” test. The mean ± SEM was
calculated for each group and the corresponding level of significance was
calculated.
Results:
Histological analysis: The H/E stained histological sections of arsenic treated
testes
showed increase in luminal areas associated with reduced accumulation of
spermatozoa, signs of necrotic changes with
disarray in cellular
organization (Fig
2) compared to that
of control (Fig 1).
Protein supple
mented rat testes
showed the partial
recovery compared
to the treated group
(Fig 3). Recovery
includes an accumulation of increased spermatozoa in the luminal areas
along with
partial amelioration of arsenic induced changes.
Analysis of spermatozoal status :
1. Sperm count: The Table 1 shows that there is
a reduction in the number of matured
spermatozoa in case of treated group compared to
that of the control(p<0.001). The values from
high protein supplemented group, reveals an
increased (p>0.001) sperm count towards normal.
All the values are presented in table1. Results are
expressed as mean ±SEM (Standard error of the
mean) (n=6).
2. Sperm viability count: The Table1 shows that there is a reduction in the
number
of viable sperms in cases of treated group (p<0.001) and this decrease in
viability is
minimized (p>0.001) in the high protein diet supplemented group. Results
are
expressed in table 1 following the same statistical analysis as mentioned
earlier.
3. Sperm motility count: In case of treated group the decrease in motility was
observed in studies done in the 1st, 2nd and 3rd hours compared to control. This
decrease was around 10% as observed in three consecutive hours;
administration of
protein diet minimized the decrease in motility as reflected in the values
nearer to
that of control. The mean ± SEM (Standard error of mean) was calculated for
each
Fig: 1:- Section of control testis
showing normal features (a) x10 (b)
x40
Fig: 2: - Section of treated testis showing
an increase in luminal area, reduced
spermatozoal mass, disintegrated cell and
nuclear membranes and disorganized
cellular orientation.(a) x10 (b) x 40
1a 1b
2a 2b
Fig 3: - Section of protein diet
supplemented testis showing features
towards normalcy.(a) x10 (b) x 40
3a 3b
Al Ame en J Med S c i Volume2, No.1, 2009 Mukherjee S & Mukhopadhyay PK
©2009. Al Ameen Charitable Fund Trust, Bangalore 76
group and the corresponding level of significance was calculated by using the
previous statistical analysis.
Table: - 1. Table presenting the effect of protein diet supplementation on the
arsenic
trioxide induced changes in spermatozoal status in rat: -
Discussion
Increase in the luminal areas of the seminiferous tubules associated with
decreased
spermatozoal mass might be due to low levels of gonadotropins in arsenic
treated
rats, and these low levels are responsible for the decreased production of
steroidogenic enzymes [4]. It has been established that arsenic
administration leads to
decrease in ovarian steroidogenic enzymes synthesis [4]. Thus the low levels
of
gonadotropins and possibly testosterone might be responsible for the
decrease in the
spermatozoal mass in the lumen. Decrease in epididymal spermatozoal
number
provides support towards this histological observation. Arsenic causes lipid
peroxidation by generation of reactive oxygen species (ROS) [13-14]. This
peroxidation may cause rupture of cell as well as nuclear membrane. This
might be
responsible for the observed necrosis and disarray in cellular organization in
histological section (40 X) Fig-2b. Evidence suggests that arsenic induces free
radical formation and thus the generated reactive oxygen species (ROS) react
with
the polyunsaturatedfattyacid (PUFA) rich spermatozoa, specially the mid
spermatozoa and results in peroxidation which finally leads to destruction in
spermatozoa causing reduced motility and viability [4]. Pea plus casein diet
supplementation along with arsenic treatment reveals that the decrease in
sperm
count, motility and viability due to toxic effects of arsenic is minimized. As a
possible mechanism it could be stated that either pea or casein or both have
a
recovery role on arsenic tri oxide mediated toxicity by inducing an antioxidant
effect
against the oxidative stress [15]. The antioxidant properties of milk casein
have been
established [15]. Studies also indicate that casein phosphopeptides (CPP) and
casein
hydrolysate bind the peroxidant and thus lipid peroxidation is suppressed [15-
17].
Studies for finding the antioxidant mechanisms of pea and wheat have also
been
done [5]. The pea or pea and casein supplement is effective in reducing the
Parameters
Studied
Control
groupA
Treated
groupB
Percent
decrease
%
Significance
Level
between
A&B
Protein diet
supplemented
groupC
Percentage
restored
%
Significance
Level
between
B&C
Mean±SEM Mean±SEM Mean±SEM
1) Sperm
count
(10^6/cauda)
135.14±5.87
122.24± 5.7
9.5
P<0.001
131.92± 5.31
75
P>0.001
2) Sperm
viability (%)
27.16±3.42
14.7± 1.54
46
P<0.001
22.63 ± 3.93
64
P>0.001
3) Sperm
motility (%)
1st hr 97.66 ±0.71 89.16 ± 2.8 8.7 P<0.001 94 ± 1.11 57 P>0.001
2nd hr 81 ±1.01 73 ± 3.2 98 P<0.001 74.5 ± 0.71 19 P>0.001
3rd hr 61.33 ±2.7 54.66 ± 4.5 10.3 P<0.001 54.8 ± 2.98 2 P<0.20
Al Ame en J Med S c i Volume2, No.1, 2009 Mukherjee S & Mukhopadhyay PK
©2009. Al Ameen Charitable Fund Trust, Bangalore 77
production of nitric oxide and MDA which could markedly increase the activity
of
the antioxidant enzymes. This could not only overcome the oxidative stress
caused
by arsenic but also suppresses the ROS generation from other sources. [6].
Studies on
arsenic trioxide induced toxicity on male gonad reveal a good deal of changes
in
histology of seminiferous tubule, associated with decreased spermatozoal
mass.
Sperm count, viability, and motility are seen to be affected and the possible
mechanisms behind these changes have been discussed. Supplementation of
specific
proteins with the normal diet causes significant recovery from all these toxic
effects.
In summary we have demonstrated that the pea and casein by virtue of
having
antioxidant properties are responsible for restoration of normal gonadal
status when
supplemented with simultaneous arsenic exposure.
Reference
1. Clarkson, T; Envioron. Health Prospect, 1995, 103, 9—12.
2. Roy, P; Saha, A Metabolism and toxicity of arsenic: A human carcinogen Current Sc. Vol 82
No.1,
Jan 2002, Pg-38—45.
3. Mandal,BK, ;Roychowdhury,T, Samanta,G , BasuGK, ChowdhuryPP, Chanda,CR (1996)
Current
science 70, 976—986
4. Sarkar M, Ray Chaudhuri G, Chattopadhyay A, Biswas NM; Effect of sodium arsenite on
spermatogenesis, plasma gonadotrophins and testosterone in rats. Indian Asian J Androl 2003
Mar; 5: 27-31
5. Kreiger DT, Porlow MJ, Gibson MJ, Davis TF , Brain grafts reverse hypogonadism of
gonadotropin releasing hormone deficiency. Nature (1982) 298,468-471
6. Mukherjee,S, Das,D Darbar D and Mitra*,C Dietary intervention affects arsenic-generated
nitric
oxide and reactive oxygen intermediate toxicity in islet cells of rats Current Science, vol. 85,
no. 6,
25 September 2003 786-793.
7. S.Maiti and A. K. Chatterjee; Effects on levels of glutathione and some related enzymes in
tissues
after an acute arsenic exposure in rats and their relationship to dietary protein deficiency. Arch
Toxicol 2001 Nov;75(9):531-7
8. P. Y. Y. Wong and D. D. Kitts* Chemistry of Buttermilk Solid Antioxidant Activity J. Dairy Sci.
2003, 86:1541-1547.
9. Chatterjee, A. K., Jamdar, S. C. and Ghosh, B. B., Effect of riboflavin deficiency on
incorporation
in vivo of [14C] amino acid into liver proteins of rats. Br. J. Nutr., 1970, 24, 635–640.
10. Jequier A.M., Male Infertility. A guide for clinician. Blackwell Pg: 64—65.
11. Jequier A.M. Male Infertility. A guide for clinician. Blackwell Pg: 55—61.
12. Jequier A.M. Male Infertility. A guide for clinician. Blackwell Pg: 61—63.
13. Xu Y, Wang Y, Zheng Q, Li X, Li B, Jin Y, Sun X, Sun G. Association of oxidative stress with
arsenic methylation in chronic arsenic-exposed children and adults. Toxicol Appl Pharmacol.
2008
Jul 1. (Epub ahead of print).
14. Osbaldo Ramos, Leticia Carrizales, Leticia Yáñez, Jesús Mejía, Lilia Batres,
deogracias Ortíz, and Fernando Díaz-Barriga : Arsenic Increased Lipid Peroxidation in Rat
Tissues
by a Mechanism Independent of Glutathione Levels. Environmental Health Perspectives 103,
Supplement 1, February 1995.
15. Cervato G, Cazzola R, Cestaro B. Int. Studies on the antioxidant activity of milk caseins. J
Food
Sci Nutr. 1999 Jul; 50(4):291-6.
16. Taylor MJ, Richardson T. Antioxidant activity of skim milk: effect of sonication. J Dairy Sci.
1980
Nov; 63(11):1938-42.
17. Diaz M, Decker EA.: Antioxidant mechanisms of caseinophosphopeptides and casein
hydrolysates
and their application in ground beef. J Agric Food Chem. 2004 Dec 29; 52(26):8208-13.
Address for correspondence: Dr. Prabir Kumar Mukhopadhyay, Department of Physiology, Presidency
College,
86/1, College street, Kolkata- 700073. West Bengal, India.
Comparative toxicity of trivalent and pentavalent inorganic and methylated arsenicals in rat and human cells
Styblo, M | Del Razo, LM | Vega, L | Germolec, DR | LeCluyse, EL | Hamilton, GA | Reed, W | Wang, C | Cullen, WR |
Thomas, DJ
Archives of Toxicology [Arch. Toxicol.]. Vol. 74, no. 6, pp. 289-299. 18 Aug 2000.

Biomethylation is considered a major detoxification pathway for inorganic arsenicals (iAs). According to the postulated
metabolic scheme, the methylation of iAs yields methylated metabolites in which arsenic is present in both pentavalent
and trivalent forms. Pentavalent mono- and dimethylated arsenicals are less acutely toxic than iAs. However, little is
known about the toxicity of trivalent methylated species. In the work reported here the toxicities of iAs and trivalent and
pentavalent methylated arsenicals were examined in cultured human cells derived from tissues that are considered a
major site for iAs methylation (liver) or targets for carcinogenic effects associated with exposure to iAs (skin, urinary
bladder, and lung). To characterize the role of methylation in the protection against toxicity of arsenicals, the capacities of
cells to produce methylated metabolites were also examined. In addition to human cells, primary rat hepatocytes were
used as methylating controls. Among the arsenicals examined, trivalent monomethylated species were the most cytotoxic
in all cell types. Trivalent dimethylated arsenicals were at least as cytotoxic as trivalent iAs (arsenite) for most cell types.
Pentavalent arsenicals were significantly less cytotoxic than their trivalent analogs. Among the cell types examined,
primary rat hepatocytes exhibited the greatest methylation capacity for iAs followed by primary human hepatocytes,
epidermal keratinocytes, and bronchial epithelial cells. Cells derived from human bladder did not methylate iAs. There
was no apparent correlation between susceptibility of cells to arsenic toxicity and their capacity to methylate iAs. These
results suggest that (1) trivalent methylated arsenicals, intermediary products of arsenic methylation, may significantly
contribute to the adverse effects associated with exposure to iAs, and (2) high methylation capacity does not protect cells
from the acute toxicity of trivalent arsenicals.

Descriptors: Article Subject Terms Arsenic | Liver | Lung | Methylation | Skin | Urinary bladder

Abstract

The effects of arsenic and ethanol interaction on blood, liver and serum biochemical indices, and arsenic
concentration in soft tissues of rats were investigated to determine the influence of these substances in
inducing susceptibility to arsenic poisoning. Arsenic, intraperitoneally (100 ppm, once, daily), ethanol in
drinking water (10%), or the combination were administered for a period of 6 weeks. Both the
chemicals had some additive effects in marginally elevating blood zinc protoporphyrin. Glutathione
(GSH) concentrations of blood and liver were reduced by both arsenic and ethanol; however, there was
a more pronounced depletion of hepatic GSH concentration in animals coexposed to arsenic and
ethanol. Combined arsenic plus ethanol exposure led to significantly more elevated activities of serum
transaminases than in animals administered arsenic or ethanol alone. Histopathological alterations in
kidneys and liver occurred following arsenic exposure. Arsenic plus ethanol produced more pronounced
liver lesions, whereas kidney changes were the same as with arsenic alone. The concentrations of
arsenic in kidney and liver were higher in rats exposed to arsenic plus ethanol. The results suggest that
animals exposed to arsenic plus ethanol are more vulnerable to arsenic toxicity.

Subscriptions

Performing your original search, arsenic toxicity in rats, within the ACS Publications
collection will retrieve 218 results. Go to results
Prev. Article Next Article Table of Contents
Research Article
Insecticide-Rodenticide Toxicology, The Acute Oral Toxicity in Rats of Several Diet-
Arsenic Trioxide Mixtures
First Page
Hi-Res PDF[233 KB]
E. W. Packman, D. D. Abbott, J. W. E. Harrison
J. Agric. Food Chem., 1961, 9 (4), pp 271–272
DOI: 10.1021/jf60116a008
Publication Date: July 1961

Note: In lieu of an abstract, this is the article's first page.


Click to decrease image sizeX

View: Hi-Res PDF

History
Published In Issue July, 1961
Recommend & Share
Related Content
Other ACS content by these authors:
E. W. Packman
D. D. Abbott
J. W. E. Harrison
JOURNALS
A
Accounts of Chemical Research
ACS Applied Materials & Interfaces
ACS Chemical Biology
ACS Chemical Neuroscience
ACS Nano
Analytical Chemistry
- I&EC Analytical Edition
B
Biochemistry
Bioconjugate Chemistry
Biomacromolecules
Biotechnology Progress
C
Chemical Research in Toxicology
Chemical Reviews
Chemistry of Materials
Crystal Growth & Design
E
Energy & Fuels
Environmental Science & Technology
I
Industrial & Engineering Chemistry Research
- Journal of I&EC
- I&EC
- I&EC Fundamentals
- I&EC Process Design and Development
- I&EC Product Research and Development
- Product R&D
Inorganic Chemistry
J
Journal of the American Chemical Society
Journal of Agricultural and Food Chemistry
Journal of Chemical & Engineering Data
- I&EC Chemical & Engineering Data Series
Journal of Chemical Information and Modeling
- Journal of Chemical Documentation
- Journal of Chemical Information and Computer Sciences
Journal of Chemical Theory and Computation
Journal of Combinatorial Chemistry
Journal of Medicinal Chemistry
Journal of Natural Products
The Journal of Organic Chemistry
The Journal of Physical Chemistry A
The Journal of Physical Chemistry B
The Journal of Physical Chemistry C
The Journal of Physical Chemistry Letters
- The Journal of Physical Chemistry
- The Journal of Physical and Colloid Chemistry
Journal of Proteome Research
L
Langmuir
M
Macromolecules
Molecular Pharmaceutics
N
Nano Letters
O
Organic Letters
Organic Process Research & Development
Organometallics
BOOKS
ACS Division Proceedings Online
Reagent Chemicals
ACS Style Guide
MAGAZINE
Chemical & Engineering News
COMMUNITIES
ACS Chemical Biology
ACS Nanotation
ACS Network
C&ENtral Science
DIRECTORIES & DATABASES
Chemical Abstracts Service (CAS)
Directory of Graduate Research (DGRweb)
Copyright © 2009 American Chemical Society
1155 Sixteenth Street N.W., Washington, DC 20036
Journals A–Z| Books| Authors & Reviewers| Librarians| ACS Members| Help ACS
Publications is a partner of:
Technology Partner - Atypon Systems, Inc.

Lead-, cadmium-, and arsenic-induced DNA damage in rat


germinal cells.
Full Abstract

Toxic agents can interfere with the male reproductive system at many targets. One of the major
unresolved questions concerning male infertility is identification of its molecular origins. Clinical and
animal studies indicate that abnormalities of spermatogenesis result from exposure to three toxic
metals (lead acetate, cadmium chloride, and arsenic trioxide), but the effects on primary spermatocyte
DNA of the male rat after chronic exposure to these metals have not been identified. The aims of this
study were to analyze, in three independent experiments, the DNA damage induced by lead (Pb),
cadmium (Cd), and arsenic (As) in rat germinal cells during three time periods, and to determine the
relationship between DNA damage and blood Pb, blood Cd, and urine As levels. For lead acetate and
cadmium chloride experiments, blood was collected by cardiac puncture, while for arsenic trioxide a
24-h urine sample was collected. Afterward, the animals were sacrificed by decapitation. Pachytene
spermatocytes from rat testes were purified by trypsin digestion followed by centrifugal elutriation. After
establishment of cell purity and viability, DNA damage (tail length) was measured employing a single
cell gel/comet assay. Significant DNA damage was found in primary spermatocytes from rats with
chronic exposure (13 weeks) to toxic metals. In conclusion, these findings indicate that exposure to
toxic metals affects primary spermatocyte DNA and are suggestive of possible direct testicular toxicity.
27. Blair PC, Thompson MB, Bechtold M, Wilson RE, Moorman MP, Fowler BA.
Evidence for oxidative damage to red blood cells in mice induced by arsine gas.
Toxicology 63:25-34 (1990).

28. Yamanaka K, Hasegawa A, Sawamura R, Okada S. Dimethylated arsenics


induce DNA strand breaks in lung via the production of active oxygen in mice.
Biochem Biophys Res Commun 165:43-50 (1989).

29. Keyse SM, Tyrrell RM. Heme oxygenase is the major 32-kDa stress protein
induced in human skin fibroblasts by UVA radiation, hydrogen peroxide, and
sodium arsenite. Proc Natl Acad Sci USA 86:99-103 (1989).

30. Shannon RL, Strayer, DS. Arsenic-induced skin toxicity. Human Toxicol 8:99-
104 (1989).

Last Update: September 30, 1998


|

ARSENIC Toxicity

Significant exposure to arsenic occurs through both anthropogenic and natural


sources. Arsenic is released into the air by volcanoes and is a natural
contaminant of some deep-water wells. Occupational exposure to arsenic is
common in the smelting industry (in which arsenic is a byproduct of ores
containing lead, gold, zinc, cobalt, and nickel) and is increasing in the
microelectronics industry (in which gallium arsenide is responsible). Low-level
arsenic exposure continues to take place in the general population (as do some
cases of high-dose poisoning) through the commercial use of inorganic arsenic
compounds in common products such as wood preservatives, pesticides,
herbicides, fungicides, and paints; through the consumption of foods and the
smoking of tobacco treated with arsenic-containing pesticides; and through the
burning of fossil fuels in which arsenic is a contaminant. Arsenic was also a major
ingredient of Fowler's solution and continues to be found in some folk remedies.
METABOLISM

The toxicity of an arsenic-containing compound depends on its valence state


(zero-valent, trivalent, or pentavalent), its form (inorganic or organic), and the
physical aspects governing its absorption and elimination. In general, inorganic
arsenic is more toxic than organic arsenic, and trivalent arsenite is more toxic
than pentavalent and zero-valent arsenic. The normal intake of arsenic by adults
occurs primarily through ingestion and averages around 50 ug/d (range, 8 to 104
ug/d). Most (around 64 percent) of this amount is accounted for by organic
arsenic from fish, seafood, and algae; the specific arsenic compounds obtained
from these sources are arsenobentaine and arsenocholine, which are relatively
nontoxic and are rapidly excreted in unchanged form in the urine. After
absorption, inorganic arsenic accumulates in the liver, spleen, kidneys, lungs,
and gastrointestinal tract. It is then rapidly cleared from these sites but leaves a
residue in keratin-rich tissues such as skin, hair, and nails. Arsenite (+5)
undergoes biomethylation in the liver to the less toxic metabolites methylarsenic
acid and dimethylarsenic acid; biomethylation can quickly become saturated,
however, and the result is the deposition of increasing doses of inorganic arsenic
in soft tissues. Arsenic, particularly in its trivalent form, inhibits critical sulfhydryl-
containing enzymes. In the pentavalent form, the competitive substitution of
arsenic for phosphate can lead to rapid hydrolysis of the high-energy bonds in
compounds such as ATP.

CLINICAL FEATURES

Acute arsenic poisoning from ingestion results in increased permeability of small


blood vessels and inflammation and necrosis of the intestinal mucosa; these
changes manifest as hemorrhagic gastroenteritis, fluid loss, and hypotension.
Delayed cardiomyopathy accompanied by electrocardiographic abnormalities
may develop. Symptoms include nausea, vomiting, diarrhea, abdominal pain,
delirium, coma, and seizures. A garlicky odor may be detectable on the breath.
Acute tubular necrosis and hemolysis may develop. The reported lethal dose
of arsenic ranges from 120 to 200 mg in adults and is 2 mg/kg in children. Arsine
gas causes severe hemolysis within 3 to 4 h of exposure and can lead to acute
tubular necrosis and renal failure.

In chronic arsenic poisoning, the onset of symptoms comes at 2 to 8 weeks.


Typical findings are skin and nail changes, such as hyperkeratosis,
hyperpigmentation, exfoliative dermatitis, and Mees' lines (transverse white
striae of the fingernails); sensory and motor polyneuritis manifesting as
numbness and tingling in a "stocking-glove" distribution, distal weakness, and
quadriplegia; and inflammation of the respiratory mucosa. Epidemiologic
evidence has linked chronic consumption of water containing arsenic at
concentrations in the range of 10 to 1820 ppb with vasospasm and peripheral
vascular insufficiency culminating in "blackfoot disease," a gangrenous
condition affecting the extremities. Chronic arsenic exposure has also been
associated with a greatly elevated risk of skin cancer and possibly of cancers
of the lung, liver (angiosarcoma), bladder, kidney, and colon.

LABORATORY FINDINGS

When acute arsenic poisoning is suspected, an x-ray of the abdomen may reveal
ingested arsenic, which is radiopaque. The serum arsenic level may exceed 0.9
umol/L (7 ug/dL); however, arsenic is rapidly cleared from the blood.
Electrocardiographic findings may include QRS complex broadening, QT
prolongation, ST-segment depression, T-wave flattening, and multifocal
ventricular tachycardia. Urinary arsenic should be measured in 24-h specimens
collected after 48 h of abstinence from seafood ingestion; normally, levels of total
urinary arsenic excretion are less than 0.67 umol/d (50 ug/d). Arsenic may be
detected in the hair and nails for months after exposure. Abnormal liver function,
anemia, leukocytosis or leukopenia, proteinuria, and hematuria may be detected.
Electromyography may reveal features similar to those of Guillain-Barre
syndrome.

TREATMENT

Vomiting should be induced with ipecac in the alert patient with acute arsenic
ingestion.
Gastric lavage may be useful; activated charcoal with a cathartic (such as
sorbitol) may be tried.
Aggressive therapy with intravenous fluid and electrolyte replacement in an
intensive-care setting may be life-saving.
Dimercaprol is the chelating agent of choice and is administered
intramuscularly at an initial dose of 3 to 5 mg/kg on the following schedule:
every 4 h for 2 days, every 6 h on the third day, and every 12 h thereafter for 10
days. (An oral chelating agent may be substituted.) Succimer is sometimes an
effective alternative, particularly if adverse reactions to dimercaprol develop
(such as nausea, vomiting, headache, increased blood pressure, and
convulsions). In cases of renal failure, doses should be adjusted carefully, and
hemodialysis may be needed to remove the chelating agent-arsenic complex.
Arsine gas poisoning should be treated supportively with the goals of
maintaining renal function and circulating red-cell mass.

Arsenic toxicity
From Wikipedia, the free encyclopedia

Jump to: navigation, search


Arsenic and many of its compounds are especially potent poisons. Arsenic
disrupts ATP production through several mechanisms. At the level of the citric
acid cycle, arsenic inhibits pyruvate dehydrogenase and by competing with
phosphate it uncouples oxidative phosphorylation, thus inhibiting energy-linked
reduction of NAD+, mitochondrial respiration, and ATP synthesis. Hydrogen
peroxide production is also increased, which might form reactive oxygen species
and oxidative stress. These metabolic interferences lead to death from multi-
system organ failure (see arsenic poisoning) probably from necrotic cell death,
not apoptosis. A post mortem reveals brick red colored mucosa, due to severe
hemorrhage. Although arsenic causes toxicity, it can also play a protective role.[1]

[edit] Exposure

Exposure to arsenic can occur from the environment and food consumption.[2] As
a component of geologic formations, arsenic can be washed out into the ground
water. Human activities such as mining and ore smelting can contribute to this
process. Moreover, certain prokaryotes can metabolize arsenic in order to
generate energy. This may enhance the process of solubilization from the solid to
the aqueous phase. [3]

[edit] Kinetics

The two forms of arsenic, reduced (trivalent As (III)) and oxidized (pentavalent
As(V)), can be absorbed, and accumulated in tissues and body fluids.[2] In the
liver, the metabolism of arsenic involves enzymatic and non-enzymatic
methylation, the most frequently excreted metabolite (≥ 90%) in the urine of
mammals is dimethylarsinic acid (DMA(V)).[4]

In humans inorganic arsenic is reduced nonenzymatically from pentavalent to


trivalent state using glutathione (GSH) or its is mediated by enzymes.
Methylation occurs through methyltransferase enzymes. S-adenosylmethionine
(SAM) may serve as methyl donor. Various pathways are used, the principal
route being dependant on the current environment of the cell.[5] Resulting
metabolites are monomethylarsonous acid (MMA(III)) and dimethylarsinous acid
(DMA(III)).

These metabolites are less likely to react with tissue components than inorganic
arsenic (especially +3 As) and as they are easly excreted, methylation has been
regarded as a detoxification process. While reduction from +5 As to +3 As may
be considered as a bioactivation.[6] Another suggestion is that methylation might
be a detoxification if "As[III] intermediates are not permitted to accumulate"
because the pentavalent organoarsenics have a lower affinity to thiol groups than
inorganic pentavalent arsenics.[5] Gebel (2002) stated that methylation is a
detoxification through accelerated excretion. With regard to carcinogenicity it has
been suggested that methylation should be regarded as a toxification. (Kitchin
2001, Kenyon et al. 2001, Styblo et al. 2002)
Arsenic, especially +3 As, binds to single, but with higher affinity to vicinal
sulfhydryl groups, thus reacts with a variety of proteins and inhibits their activity.
It was also proposed that binding of arsenite at nonessential sites might
contribute to detoxification (Aposhian 1989). Arsenite inhibits members of the
disulfide oxidoreductase family like glutathione reductase (Rodríguez et al. 2005)
and thioredoxin reductase.[7]

The remaining non-excreted arsenic (≤ 10%) accumulates in cells, which over


time may lead to skin, bladder, kidney, liver, lung, and prostate cancers.[4] Other
forms of arsenic toxicity in humans have been observed in blood, bone marrow,
cardiac, central nervous system, gastrointestinal, gonadal, kidney, liver,
pancreatic, and skin tissues.[4]

[edit] Mechanism

Arsenite inhibits not only the formation of Acetyl-CoA but also the enzyme
succinic dehydrogenase. Arsenate can replace phosphate in many reactions. It is
able to form Glc-6-Arsenate in vitro; therefore it has been argued that hexokinase
could be inhibited (reported by Hughes 2002). (Eventually this may be a
mechanism leading to muscle weakness in chronic arsenic poisoning.) In the
glyceraldehyde-3-P-dehydrogenase reaction arsenate attacks the enzyme-bound
thioester. The formed 1-arseno-3-posphoglycerate is unstable and hydrolyzes
spontaneously. Thus, ATP formation in Glycolysis is inhibited while bypassing the
phosphoglycerate kinase reaction. (Moreover, the formation of 2,3-
bisphosphoglycerate in erythrocytes might be affected, followed by a higher
oxygen affinity of hemoglobin and subsequently enhanced cyanosis) As shown
by Gresser (1981), submitochondrial particles synthesize Adenosine-
5’-diphosphate-arsenate from ADP and arsenate in presence of succinate. Thus,
by a variety of mechanisms arsenate leads to an impairment of cell respiration
and subsequently diminished ATP formation. This is consistent with observed
ATP depletion of exposed cells and histopathological findings of mitochondrial
and cell swelling, glycogen depletion in liver cells and fatty change in liver, heart
and kidney.

Experiments demonstrated enhanced arterial thrombosis in a rat animal model,


elevations of serotonin levels, thromboxane A[2] and adhesion proteins in
platelets, while human platelets showed similar responses (Lee et al. 2002). The
effect on vascular endothelium may eventually be mediated by the arsenic-
induced formation of nitric oxide. It was demonstrated that +3 As concentrations
substantially lower than concentrations required for inhibition of the lysosomal
protease cathepsin L in B cell line TA3 were sufficient to trigger apoptosis in the
same B cell line, while the latter could be a mechanism mediating
immunosuppressive effects (Harrison et al. 2001).
[edit] Carcinogenicity

It is still a matter of debate whether DNA repair inhibition or alterations in the


status of DNA methylation are responsible for the carcinogenic potential of As. As
vicinal sulfhydryl groups are frequently found in DNA-binding proteins,
transcription factors and DNA-repair proteins, interaction of arsenic with these
molecules appears to be likely. However, in vitro, most purified DNA repair
enzymes are rather insensitive to As, but in cell culture, As produces a dose-
dependant decrease of DNA ligase activity. This might indicate that inhibition of
DNA repair is an indirect effect due to changes in cellular redox levels or
alterated signal transduction and consequent gene expression (Hu et al. 1998).
In spite of its carcinogenicity, the potential of arsenic to induce point mutations is
weak. If administered with point mutagens it enhances the frequency of
mutations in a syngergistic way (Gebel 2001).

Its comoutagenic effects may be explained by interference with base and


nucleotide excision repair, eventually through interaction with zinc finger
structures (Hartwig et al. 2002). DMA showed to effectuate DNA single stand
breaks resulting from inhibition of repair enzymes at levels of 5 to 100 mM in
human epithelial type II cells (Yamanaka 1997). (Significant DNA strand breaks
have also been observed by Bau et al. 2002)

+3 MMA and +3 DMA were also shown to be directly genotoxic by effectuating


scissions in supercoiled ΦX174 DNA (Mass et al. 2001). Increased arsenic
exposure is associated with an increased frequency of chromosomal aberrations
(recently shown in an epidemiologic stud by Maki-Paakkanen et al. 1998),
mikronuklei (Warner et al. 1994, Gonseblatt et al. 1997) and sister-chromatid
exchanges. An explanation for chromosomal aberrations is the sensitivity of the
protein tubulin and the mitotic spindle to arsenic. Histological observations
confirm effects on cellular integrity, shape and locomotion (reviewed by Bernstam
et al. 2000).

+3 DMA is able to form reactive oxygen species (ROS) by reaction with


molecular oxygen. Resulting metabolites are the dimethylarsenic radical and the
dimethylarsenic peroxyl radical (Yamanaka et al. 1990). Both +5 DMA and +3
DMA were shown to release iron from horse spleen as well as from human liver
ferritin if ascorbic acid was administered simultaneously. Thus, formation of ROS
can be promoted (Ahmad et al. 2000). Moreover, Arsenic could cause oxidative
stress by depleting the cell’s antioxidants, especially the ones containing thiol
groups. The accumulation of ROS like the cited above and hydroxyl radicals,
superoxide radicals and hydrogen peroxides causes aberrant gene expression at
low concentrations and lesions of lipids, proteins and DNA in higher
concentrations which eventually lead to cellular death. In a rat animal model,
urine levels of 8-hydroxy-2’-desoxyguanosine (as a biomarker of ROS DNA
damage) were measured after treatment with DMA. In comparison to control
levels, they turned out to be significantly increased (Yamanaka et al. 2001). This
theory is further supported by a cross-sectional study which found elevated mean
serum lipid peroxides (LPO) in the As exposed individuals which correlated with
blood levels of inorganic arsenic and methylated metabolites and inversely
correlated with nonprotein sulfhydryl (NPSH) levels in whole blood (Pi et al.
2002). Another study found an association of As levels in whole blood with the
level of reactive oxidants in plasma and an inverse relationship with plasma
antioxidants (Wu et al. 2001). A finding of the latter study indicates that
methylation might in fact be a detoxification pathway with regard to oxidative
stress: the results showed that the lower the As methylation capacity was, the
lower the level of plasma antioxidant capacity. As reviewed by Kitchin (2001), the
oxidative stress theory provides an explanation for the preferred tumor sites
connected with arsenic exposure. Considering that a high partial pressure of
oxygen is present in lungs and +3 DMA is excreted in gaseous state via the lungs
this seems to be a plausible mechanism for special vulnerability. The fact that
DMA is produced by methylation in the liver, excreted via the kidneys and latter
on stored in the bladder accounts for the other tumor localizations. Regarding
DNA methylation, some studies suggest interaction of As with methyltransferases
which leads to an inactivation of tumor suppressor genes through
hypermethylation, others state that hypomethylation might occur due to a lack of
SAM resulting in aberrant gene activation (Goering et al. 1999). An experiment
by Zhong et al. (2001) with arsenite-exposed human lung A549, kidney UOK123,
UOK109 and UOK121 cells isolated eight different DNA fragments by
methylation-sensitive arbitrarily primed PCR. It turned out that six of the
fragments were hyper- and two of them were hypomethylated. Higher levels of
DNA methltransferase mRNA and enzyme activity were found.

Kitchin (2001) proposed a model of altered growth factors which lead to cell
proliferation and thus to carcinogenesis. From observations it is known that
chronic low-dose arsenic poisoning can lead to increased tolerance to its acute
toxicity. MRP1-overexpressing lung tumor GLC4/Sb30 cells poorly accumulate
arsenite and arsenate. This is mediated through MRP-1 dependent efflux
(Vernhet et al. 2000). The efflux requires GSH, but no As-GSH complex formation
(Salerno et al. 2002). Resistant Cell lines have also been studied by Gebel 2001
and Brambila et al. 2002. Although a lot of mechanisms have been proposed, no
definite model can be given for the mechanisms of chronic arsenic poisoning.
The prevailing events of toxicity and carcinogenicity might be quite tissue-
specific. Current consensus on the mode of carcinogenesis is that it acts
primarily as a tumor promoter. Its co-carcinogenicity has been demonstrated in
several models. However, the finding of several studies that chronically arsenic-
exposed Andean populations (as most extremely exposed to UV-light) do not
develop skin cancer with chronic arsenic exposure, is puzzling (cited by Gebel
2000).[8]
[edit] Heat shock response

Another aspect is the similarity of arsenic effects to the heat shock response.
Short-term arsenic exposure has effects on signal transduction inducing heat
shock proteins with masses of 27,60,70,72,90,110 kDa as well as metallotionein,
ubiquitin, mitogen-activated [MAP] kinases, extracellular regulated kinase [ERK],
c-jun terminal kinases [JNK] and p38 (Bernstam 2000, Del Razo 2001). Via JNK
and p38 it activates c-fos, c-jun and egr-1 which are usually activated by growth
factors and cytokines (Cavigelli et al. 1996, Ludwig et al. 1998, Bernstam et al.
2000) The effects are largely dependant on the dosing regime and may be as
well inversed. As shown by some experiments reviewed by Del Razo (2001),
ROS induced by low levels of inorganic arsenic increase the transcription and the
activity of the activator protein 1 (AP-1) and the nuclear factor-kB (NF-kB)
(maybe enhanced by elevated MAPK levels), which results in c-fos/c-jun
activation, over-secretion of pro-inflammatory and growth promoting cytokines
stimulating cell proliferation (reviewed by Simeonova and Luster 2000).
Germolec et al. (1996) found an increased cytokine expression and cell
proliferation in skin biopsies from individuals chronically exposed to arsenic-
contaminated drinking water. Increased AP-1 and NF-kB obviously also result in
an up-regulation of mdm2 protein, which decreases p53 protein levels (Hamadeh
et al. 1999) Thus, taking into account p53’s function, a lack of it could cause a
faster accumulation of mutations contributing to carcinogenesis. However, high
levels of inorganic arsenic inhibit NF-kB activation and cell proliferation. An
experiment of Hu et al. (2002) demonstrated increased binding activity of AP-1
and NF-kB after acute (24 h) exposure to +3 sodium arsenite, whereas long-term
exposure (10–12 weeks) yielded the opposite result. The authors conclude that
the former may be interpreted as a defense response while the latter could lead
to carcinogenesis. As the contradicting findings and connected mechanistic
hypotheses indicate, there is a difference in acute and chronic effects of arsenic
on signal transduction which is not clearly understood yet.

[edit] Oxidative stress

Studies have demonstrated that the oxidative stress generated by arsenic may
disrupt the signal transduction pathways of the nuclear transcriptional factors
PPAR’s, AP-1, and NFκB,[4][9][10] as well as the pro-inflammatory cytokines IL-8
and TNF-α.[9][4][11][12][10][13][14][15] The interference of oxidative stress with signal
transduction pathways may affect physiological processes associated with cell
growth, metabolic syndrome X, glucose homeostasis, lipid metabolism, obesity,
insulin resistance, inflammation, and diabetes-2.[16][17][18] Recent scientific
evidence has elucidated the physiological roles of the PPAR’s in the ω-
hydroxylation of fatty acids and the inhibition of pro-inflammatory transcription
factors (NFκB and AP-1), pro-inflammatory cytokines (IL-1, -6, -8, -12, and TNF-
α), cell4 adhesion molecules (ICAM-1 and VCAM-1), inducible nitric oxide
synthase, proinflammatory nitric oxide (NO), and anti-apoptotic factors.[4][11][16][18][19]
Epidemiological studies have suggested a correlation between chronic
consumption of drinking water contaminated with arsenic and the incidence of
Type 2-diabetes.[4] The human liver after exposure to therapeutic drugs may
exhibit hepatic non-cirrhotic portal hypertension, fibrosis, and cirrhosis.[4]
However, the literature provides insufficient scientific evidence to show cause
and effect between arsenic and the onset of diabetes mellitus Type 2.[4]

[edit] References

1. ^ Klaassen, Curtis; Watkins, John (2003). Casarett and Doull's Essentials of


Toxicology. McGraw-Hill. pp. 512. ISBN 978-0071389143.
2. ^ a b Ueki, K.; Kondo, T.; Tseng, Y.-H.; Kahn, R. C. (2004). "Central role of
suppressors of cytokine signaling proteins in hepatic steatosis, insulin resistance,
and the metabolic syndrome in the mouse". Proceedings of the National Academy
of Sciences of the United States of America 101 (28): 10422–10427.
doi:10.1073/pnas.0402511101.
3. ^ "The ecology of arsenic". Science 300: 939–944. 2003.
doi:10.1126/science.1081903.
http://www.sciencemag.org/cgi/content/abstract/sci;300/5621/939?maxtoshow=&
HITS=10&hits=10&RESULTFORMAT=&fulltext=oremland+2003&searchid=1
&FIRSTINDEX=0&resourcetype=HWCIT.
4. ^ a b c d e f g h i Vigo, J. B., and J. T. Ellzey (2006). "Effects of Arsenic Toxicity at
the Cellular Level: A Review". Texas Journal of Microscopy 37 (2): 45–49.
5. ^ a b "A chemical hypothesis for arsenic methylation in mammals". Chem Biol
Interact 88 (2-3): 89–14. 1993. http://www.ncbi.nlm.nih.gov/pubmed/8403081.
6. ^ "Role of metabolism in arsenic toxicity". Pharmacol Toxicol 89 (1): 1–5. 2001.
http://www.ncbi.nlm.nih.gov/pubmed/11484904.
7. ^ Environmental and Occupational Medicine. Environmental and Occupational
Medicine. 2006. pp. 1014–1015.
http://books.google.com/books?id=H4Sv9XY296oC&pg=RA2-
PA1014&lpg=RA2-PA1014#PRA2-PA1014,M1.
8. ^ "Confounding variables in the environmental toxicology of arsenic". Toxicology
144: 155–1062. 2000. http://www.ncbi.nlm.nih.gov/pubmed/10781883.
9. ^ a b Hu Y., X. Jin, and E. Snow (2002). "Effect of arsenic on transcription factor
AP-1 and NF- K B". Toxicology Letters 133: 33. doi:10.1016/S0378-
4274(02)00083-8.
10. ^ a b Walton FS, Harmon AW, Paul DS, Drobná Z, Patel YM, Styblo M (August
2004). "Inhibition of insulin-dependent glucose uptake by trivalent arsenicals:
possible mechanism of arsenic-induced diabetes". Toxicol. Appl. Pharmacol. 198
(3): 424–33. doi:10.1016/j.taap.2003.10.026. PMID 15276423.
11. ^ a b Black PH (October 2003). "The inflammatory response is an integral part of
the stress response: Implications for atherosclerosis, insulin resistance, type II
diabetes and metabolic syndrome X". Brain Behav. Immun. 17 (5): 350–64.
doi:10.1016/S0889-1591(03)00048-5. PMID 12946657.
12. ^ Carey AL, Lamont B, Andrikopoulos S, Koukoulas I, Proietto J, Febbraio MA
(March 2003). "Interleukin-6 gene expression is increased in insulin-resistant rat
skeletal muscle following insulin stimulation". Biochem. Biophys. Res. Commun.
302 (4): 837–40. doi:10.1016/S0006-291X(03)00267-5. PMID 12646246.
13. ^ Dandona P, Aljada A, Bandyopadhyay A (January 2004). "Inflammation: the
link between insulin resistance, obesity and diabetes". Trends Immunol. 25 (1): 4–
7. doi:10.1016/j.it.2003.10.013. PMID 14698276.
http://linkinghub.elsevier.com/retrieve/pii/S1471490603003363.
14. ^ Fischer CP, Perstrup LB, Berntsen A, Eskildsen P, Pedersen BK (November
2005). "Elevated plasma interleukin-18 is a marker of insulin-resistance in type 2
diabetic and non-diabetic humans". Clin. Immunol. 117 (2): 152–60.
doi:10.1016/j.clim.2005.07.008. PMID 16112617.
15. ^ Gentry PR, Covington TR, Mann S, Shipp AM, Yager JW, Clewell HJ (January
2004). "Physiologically based pharmacokinetic modeling of arsenic in the
mouse". J. Toxicol. Environ. Health Part A 67 (1): 43–71.
doi:10.1080/15287390490253660. PMID 14668111.
16. ^ a b Kota BP, Huang TH, Roufogalis BD (February 2005). "An overview on
biological mechanisms of PPARs". Pharmacol. Res. 51 (2): 85–94.
doi:10.1016/j.phrs.2004.07.012. PMID 15629253.
17. ^ Luquet, S., C. Gaudel, D. Holst, J. Lopez-Soriano, C. Jehl-Pietri, A. Fredenrich
(2005). Biochimica et Biophysica Acta 1740: 313–317.
18. ^ a b Moraes, La; Piqueras, L; Bishop-Bailey, D (Jun 2006). "Peroxisome
proliferator-activated receptors and inflammation.". Pharmacology &
therapeutics 110 (3): 371–85. doi:10.1016/j.pharmthera.2005.08.007. PMID
16168490.
19. ^ Hara K, Okada T, Tobe K, et al. (April 2000). "The Pro12Ala polymorphism in
PPAR gamma2 may confer resistance to type 2 diabetes". Biochem. Biophys. Res.
Commun. 271 (1): 212–6. doi:10.1006/bbrc.2000.2605. PMID 10777704.

Paul B. Tchounwou1, Jose A. Centeno2 and


Anita K. Patlolla1

(1) Molecular Toxicology Research Laboratory, NIH-Center for Environmental Health, School of
Science and Technology, Jackson State University, Jackson, MS 39217, USA
(2) Environmental and Toxicologic Pathology Laboratory, Armed Forces Institute of Pathology,
Washington DC, 20306, USA
Abstract A comprehensive analysis of published data
indicates that arsenic exposure induces cardiovascular diseases,
developmental abnormalities, neurologic and neurobehavioral
disorders, diabetes, hearing loss, hematologic disorders, and
various types of cancer. Although exposure may occur via the
dermal, and parenteral routes, the main pathways of exposure
include ingestion, and inhalation. The severity of adverse health
effects is related to the chemical form of arsenic, and is also
time- and dose-dependent. Recent reports have pointed out that
arsenic poisoning appears to be one of the major public health
problems of pandemic nature. Acute and chronic exposure to
arsenic has been reported in several countries of the world where
a large proportion of drinking water (groundwater) is
contaminated with high concentrations of arsenic. Research has
also pointed significantly higher standardized mortality rates for
cancers of the bladder, kidney, skin, liver, and colon in many
areas of arsenic pollution. There is therefore a great need for
developing a comprehensive health risk assessment (RA) concept
that should be used by public health officials and environmental
managers for an effective management of the health effects
associated with arsenic exposure. With a special emphasis on
arsenic toxicity, mutagenesis, and carcinogenesis, this paper is
aimed at using the National Academy of Science''s RA framework
as a guide, for developing a RA paradigm for arsenic based on a
comprehensive analysis of the currently available scientific
information on its physical and chemical properties, production
and use, fate and transport, toxicokinetics, systemic and
carcinogenic health effects, regulatory and health guidelines,
analytical guidelines and treatment technologies.

arsenic - contamination - health effects - risk assessments and


management

Arsenic-cadmium interaction in rats: toxic effects


Title: in the heart and tissue metal shifts.
Yanez, L : Carrizales, L : Zanatta, M T : Mejia, J J :
Author:
Batres, L : Diaz Barriga, F
Citation: Toxicology. 1991 Apr 8; 67(2): 227-34
Abstract: Previously, we had shown that arsenic interacts with
cadmium in rats; our results showed that the toxicity
of a mixture of arsenic + cadmium cannot be
predicted by the toxic mechanisms of the individual
components. In this paper, we present further
evidence about the interaction of arsenic and
cadmium in rats. The results were: arsenic modified
the 24 h-LD50 value of cadmium more clearly than
cadmium did with the one of arsenic; based on the
LD50 values, the mixtures we studied were more
toxic than either metal alone. With single doses (As
10 mg/kg, Cd 2.6 mg/kg, and As 10 mg/kg + Cd 2.6
mg/kg) the mixture As + Cd was more toxic than
each metal. At these doses, cadmium significantly
induces the levels of glutathione, metallothionein,
and lipid peroxidation in heart tissue, as compared to
a saline group of rats. Arsenic incremented
glutathione and lipid peroxidation at higher values
than those obtained with cadmium. The mixture of
As + Cd behaved as arsenic in the induction of lipid
peroxidation and glutathione and like cadmium in
metallothionein induction. Finally, rats treated with
As + Cd had less Cd in liver than animals treated
only with cadmium, and more As in heart tissue than
rats treated only with arsenic. Our results give
further evidence about the arsenic-cadmium
interaction in rats, demonstrate the utility of
employing different biomarkers in the study of
chemical mixtures and indicate that heart tissue is
affected not only by the mixture of As + Cd, but also
by either metal alone.
Review None
References:

Concurrent exposure to lead, cadmium, and arsenic. Effects on toxicity and tissue
metal concentrations in the rat.

Mahaffey KR, Capar SG, Gladen BC, Fowler BA.

Male rats were exposed to dietary Pb (200 ppm), Cd (50 ppm), or As (50 ppm) as
arsenate either alone or in combination for 10 weeks using a 2 x 2 x 2 factorial
design. Cd and As reduced weight gain even when differences in food intake were
taken into account, and administration of both Cd and As depressed weight gain
more than did either metal alone. Pb did not adversely affect food consumption or
weight gain. Increased RBCs were observed after administration of Pb, Cd, or As, and
more cells were observed when two or three metals were concomitantly
administered. Despite increased numbers of circulating RBCs, hemoglobin and
hematocrit were reduced, especially with the Pb-Cd combination. Analysis of blood
chemistries showed normal ranges for blood urea nitrogen, creatinine, cholesterol,
calcium, albumin, total protein, and bilirubin. Uric acid was increased by Pb, but not
by Cd or As. SGOT activity was reduced by As alone. Serum alkaline phosphatase
was reduced by either As or Cd but not Pb. Combinations of As and Cd did not further
reduce the activity of this enzyme. Kidney weight and kidney weight/body weight
ratios were increased by Pb alone, but Cd or As alone or in combination had no
effect. Liver weight/body weight ratios were reduced in animals fed Cd. Kidney
histology showed predominantly Pb effects, i.e., intranuclear inclusion bodies and
cloudy swelling. Ultrastructural evaluation of kidneys from Pb-treated animals
disclosed nuclear inclusion bodies and mitochondrial swelling. Concurrent
administration of Cd reduced total mean bone and kidney Pb levels by 50% and 60%,
respectively, and this was associated with a decrease in kidney intranuclear
inclusions. Cd exposure also reduced renal, femur, and liver concentrations of Fe by
33%, 43%, and 63%, respectively, decreased femur Zn by 27%, but increased renal
Zn by 20%. Administration of As produced mild swelling of tubule cell mitochondria,
increased mean total renal Cu to 200% of control, and increased liver Fe by 44%.
Dietary Pb produced increased urinary excretion of ALA and coproporphyrin. Dietary
exposure to As caused increased urinary excretion of uroporphyrin and to a lesser
extent coproporphyrin, whereas dietary Cd caused no significant changes in urinary
levels of any of the porphyrins measured. Pb plus As produced an additive effect on
coproporphyrin excretion but not that of ALA or uroporphyrin. These studies indicate
that interactions between common toxic elements do occur and are characterized by
alterations in both tissue trace metal levels and toxicity.

Humans are exposed to a number of toxic elements in the environment; however,


most experiments with laboratory animals investigate only one toxic element. To
determine if concomitant exposure to lead (Pb), cadmium (Cd), and/or arsenic (As)
modified the changes produced by any one metal in various parameters of toxicity,
168 male, Sprague-Dawley, young adult rats were fed nutritionally adequate diets to
which had been added 0 or 200 ppm Pb as Pb acetate, or 50 ppm Cd as Cd chloride,
or 50 ppm As as sodium arsenate or arsanilic acid in a factorial design for a period of
10 weeks.At these concentrations, Cd and As reduced weight gain even when
differences in food intake were taken into account; administration of both Cd and As
depressed weight gain more than did either metal alone. Pb did not adversely affect
food consumption or weight gain. Increased numbers of red blood cells (RBCs) were
observed following administration of Pb, Cd, or As; usually more cells were observed
when two or three metals were administered, compared to individual metals. Despite
increasing numbers of circulating RBCs, hemoglobin and hematocrit were reduced,
especially with the Pb-Cd combination and the Cd-arsanilic acid combination. Specific
effects of Pb on heme synthesis were observed, including increased urinary excretion
of delta-aminolevulinic acid; this increase was reduced by the presence of dietary
cadmium.Analyses of blood showed values for the laboratory rat within normal
ranges for blood urea nitrogen, creatinine, cholesterol, calcium, albumin, total
protein, and bilirubin. Uric acid was increased by Pb, with little modification by
dietary Cd or As content. Serum glutamate-oxalate transaminase activity was
reduced by As. Serum alkaline phosphatase was greatly reduced by either As or Cd
but not Pb. Combinations of As and Cd did not further reduce the activity of this
enzyme. Kidney weight and kidney weight/body weight ratios were increased by Pb
alone, with no effects of Cd or As alone or as interactions. Liver weight/body weight
ratios were reduced in animals fed 50 ppm dietary Cd. Kidney histology shows
predominantly Pb effects, namely, intranuclear inclusion bodies and cloudy swelling.
Ultrastructural evaluation of kidneys from Pb-treated animals disclosed nuclear
inclusion bodies of the usual morphology and mitochondrial swelling. Concurrent
administration of Cd greatly minimized Pb effects on the kidney under conditions of
this experiment. Liver histology suggests an increased rate of cell turnover with
either As compound, but few specific changes.

PMID: 198203 [PubMed - indexed for MEDLINE]


PMCID: PMC1637428

Effect of sodium arsenite on spermatogenesis,


plasma gonadotrophins and testosterone in rats
Mahitosh Sarkar, Gargi Ray Chaudhuri, Aloke Chattopadhyay, Narendra Mohan
Biswas

Reproductive Physiology Unit, Dept. of Physiology, University Colleges of


Science and Technology, Calcutta University, Calcutta-700 009, India

Asian J Androl 2003 Mar; 5: 27-31

Keywords: arsenite; spermatogenesis; gonadotrophins; testosterone;


testis

Abstract

Aim: To investigate the effect of arsenic on spermatogenesis. Methods: Mature (4 months old)
Wistar rats were intraperitoneally administered sodium arsenite at doses of 4, 5 or 6 mg.kg-1.day-
1 for 26 days. Different varieties of germ cells at stage VII seminiferous epithelium cycle, namely,
type A spermatogonia (ASg), preleptotene spermatocytes (pLSc), midpachytene spermatocytes
(mPSc) and step 7 spermatids (7Sd) were quantitatively evaluated, along with radioimmunoassay
of plasma follicle-stimulating hormone (FSH), lutuneizing hormone (LH), testosterone and
assessment of the epididymal sperm count. Results: In the 5 and 6 mg/kg groups, there were
significant dose-dependent decreases in the accessory sex organ weights, epididymal sperm
count and plasma concentrations of LH, FSH and testosterone with massive degeneration of all
the germ cells at stage VII. The changes were insignificant in the 4 mg/kg group. Conclusion:
Arsenite has a suppressive influence on spermatogenesis and gonadotrophin and testosterone
release in rats.

1 Introduction

Arsenicals are used as herbicides, fungicides and rodenticides and may cause air, soil and water
pollution. Arsenical exposure through drinking water is common in many areas of the world [1-3].

Exposure to arsenic is associated with metabolic disorders, hypertrophy of adrenal glands [4] and
anemia [5]. A number of sulfhydryl containing proteins and enzyme systems have been found to
be altered by exposure to arsenite [6]. Arsenite affects mitochondrial enzymes and impairs tissue
respiration, which seem to be related to the cellular toxicity of arsenic [7]. Gonadal effects of
arsenic were first evaluated in mice, then in fishes [8-10]. Sodium arsenite has been found to
have an inhibitory effect on the activity of testicular steroidogenic enzymes, ∆5-3β-hydroxysteroid
dehydrogenase (∆5-3β-HSD) and 17β-hydroxysteroid dehydrogenase (17β-HSD) and to reduce
the weight of testes and accessory sex glands [11].

Most of the available data on arsenic reproductive toxicity indicate that the main concern is with
the developmental toxicity on the fetus [12]. Till date there is very few evidence on its male
reproductively effects [11,13]. The present study was designed to quantitative study the effect of
arsenite on spermatogenesis at stage VII of the seminiferous epithelial cycle and plasma
hormone concentrations, which has not been reported so far in the literature.

2 Materials and methods

2.1 Animals and treatment

Adult male Wistar rats weighing 160¡À10 g (120~140 days of age) were maintained in a 12 h light
and 12 h dark and 26 ¡æ~28 ¡æ animal house and standard laboratory chew and tap water were
available ad libitum. Sodium arsenite was purchased from Sigma (USA) and dissolved in sterile
distilled water.

Thirty-two rats were divided into 4 groups of 8 animals each. Three groups of animals were
injected with either 4, 5 or 6 mg.kg-1.day-1 sodium arsenite per 1 mL sterile distilled water for 26
days (Group II, III and IV, respectively). Animals of Group I were injected the same amount of
distilled water for 26 days and served as the controls. Treatment for 26 days was selected as the
duration of one seminiferous cycle is 13.2 days in Wistar rats.

On the 27th day between 08:00 to 10:00, blood samples were collected from the hepatic vein
under light ether anesthesia and after that the rats were killed following ethical procedure.
Heparinized plasma was prepared and stored at -20 ¡æ until hormone radioimmuno-assay.

2.2 Body and organ weights

The body weights were recorded on the first day before injection (initial) and the day of sacrifice
(final). The testicles and accessory sex organs were dissected out, trimmed off the attached
tissues and weighed. The relative weight of organs was expressed per 100g body weight. The left
testis of each rat was used for histological study and the right for estimation of elementary arsenic
content.

2.3 Histological study

Immediately after removal, the testis was fixed in Bouin's fluid and embedded in paraffin. Sections
of 5 µm thickness were taken from the mid portion of each testis and stained with periodic acid
Schiff (PAS)-hematoxylin and examined under a light microscope. Quantitative analysis of
spermatogenesis was carried out by counting the relative number of each variety of germ-cells at
stage VII of the seminiferous epithelium cycle, i.e. type-A spermatogonia (ASg), preleptotene
spermatocytes (pLSc), mid pachytene spermatocytes (mPSc) and step 7 spermatids (7Sd),
according to the method of Leblond and Clermont [14]. The nuclei of different germ cells (except
step 19 spermatids which cannot be enumerated precisely) were counted in 20 round tubules of
each rat. All the counts (crude counts) of the germ cells were corrected for differences in the
nuclear diameter by the formula of Abercrombie [15]: true count = (crude count ¡Á section
thickness)/ (section thickness - nuclear diameter of germ cell). The nuclear diameter of each
variety of germ cell was measured with a Leitz micrometer. The possibility of variable tubular
shrinkage in the sections of both arsenite and vehicle injected groups were eliminated by the
index of tubular shrinkage which was obtained from the average number of Sertoli cell nuclei
containing prominent nucleoli in the sections of the treated rats divided by that of the controls
[16]. Stage VII spermatogenesis was analyzed because this stage is highly susceptible to
testosterone deficiency [17] and also reflects the final stages of spermatid maturation and thus
provides evidence of spermatogenesis as a whole [18].

2.4 Sperm count


The sperm count was determined by counting in a haemocytometer. Sperm samples were
collected from the cauda epididymis. To minimize the count was repeated at least five times for
each rat.

2.5 Hormone assay

Plasma follicle-stimulating hormone (FSH) and lutuneizing hormone (LH) were measured by
radioimmunoassay (RIA) as described in the instructions provided with the kits (NIADDK, USA).
Carrier free 125I for hormone iodination was obtained from the Bhaba Atomic Research Centre,
Bombay, India. Pure rat FSH (NIADDK-r FSH-1-6) and LH (NIADDK-rLH-1-6) were iodinated
using the chloramine T (Sigma, USA) method of Greenwood et al [19]. The antisera to FSH and
LH were NIADDK anti-r FSH-S-11 and NIADDK-anti-rLH-S-9, and were used at a final dilution of
1:100000 and 1:150 000, respectively. Goat anti-rabbit g-globulin used as the second antibody
was obtained from the Indo Medix Inc., USA. The sensitivity of the assay was 2.0 µg/L for FSH
and 0.15 mg/L for LH. Each sample was assayed at two concentrations, each in duplicate at a
time. The intra assay coefficient of variation in each assay was 7.5 % for FSH and 6.0 % for LH.
Hormone concentrations were expressed in terms of NIH reference preparation RP-2.

Plasma testosterone was assayed according to Auletta et al [20]. Methodological loss was
monitored and accounted for by adding 1000 c. p. m. [1β, 2β3 - H (N)¡¡] testosterone (sp. act. 50.4
Ci/m mol; New England Nuclear, Boston, MA, USA) before extraction with diethyl ether. Samples
were assayed at two concentrations, each in duplicate. The antisera to testosterone was
purchased from the Endocrine Science, USA, and had a 44 % cross reactivity with
dihydrotestosterone. Free and bound testosterone were separated by using dextrancoated
charcoal. The recovery of plasma testosterone after ether extraction was estimated to be 88.5 %.
The sensitivity of the assay was 69.5 % pmol/L and the intraassay variance was 6.4 %. All
samples were measured in a single assay. Since chromatographic purification of the samples
was not performed, the testosterone values reported are the sum of testosterone and
dihydrotestosterone.

2.6 Testicular arsenic concentration

The flameless atomic absorption spectrophotometric technique [21] was used for the
determination of arsenic concentration. Animals were scarified 24h after the last arsenite injection
and one testis from each animal was collected and digested with a mixture of nitric acid, sulfuric
acid and perchloric acid (3:1:1). Values are expressed in µg of arsenic/g of testicular wet tissue.

2.7 Statistical Analysis

Data were expressed in mean¡ÀSEM. Statistical analysis was performed by analysis of variance
(ANOVA) followed by multiple comparison by two-tailed t-test.

3 Results

3.1 Body and organ weights

In all the treated groups, the body weight was not significantly different from that of the controls.
The relative weights of the testis, seminal vesicle and ventral prostate were significantly
decreased (P<0.05) after 5 mg or 6 mg treatment, but not after 4 mg of sodium arsenite treatment
(Table 1).

Table 1. Effect of sodium arsenite on body weight (g) and organ weights (mg % body weight) (mean¡ÀSEM, n=8) in rats. bP<0.05,
compared with controls. ANOVA followed by multiple comparison two-tailed t-test.
Body weight
Group Testes (pair) Seminal vesicle Ventral prostate
(g)
Control 190.85¡À18.21 1478.19¡À32.93 472.62¡À22.81 227.29¡À14.78
4 mg/kg 185.60¡À16.83 1434.65¡À36.75 450.87¡À29.38 212.42¡À12.79
5 mg/kg 181.81¡À15.39 1308.39¡À34.26b 349.34¡À20.52b 174.84¡À12.09b
6 mg/kg 187.30¡À16.45 1270.89¡À35.19b 322.16¡À18.98b 155.42¡À13.69b

3.2 Histological findings

Sodium arsenite treatment at the dose of 5 mg/kg significantly reduced the number of ASg, pLSc,
mPSc and 7Sd when compared with those of the controls. Six mg/kg caused a more prominent
spermatogenic arrest. No significant change was found in the cellular counts in the 4 mg/kg
treated group (Table 2). Theoretically, the ratio of mPSc:7Sd is 1: 4. This ratio was 1:2.76 and
1:2.57 after 5 and 6 mg/kg treatment, respectively; the ratio of the control group was 1:3.41. The
percentage spermatid degeneration (35.75 %) as calculated from the above ratio, was highly
significant after 6 mg of sodium arsenite treatment (Table 2).

Table 2. Effect of sodium arsenite on number of germ cells per tubular cross section at stage VII of the seminiferous epithelial
cycle in rats (mean¡ÀSEM, n=8). ASg = spermatogonia A; pLSc = preleptotene spermatocytes; mPSc = mid pachytene
spermatocytes; 7Sd = step 7 spermatid. bP<0.05, compared with controls. ANOVA followed by multiple comparison two-tailed t-
test.

Spermatogenesis pattern at stage VII 7Sd Effective


Group mPSc: 7Sd degeneration 7Sd
ASg pLSc mPSc 7Sd
(%) degeneration
Control 0.64¡À0.04 19.72¡À0.82 19.07¡À0.69 65.09¡À1.17 1:3.41 14.75 -
4 mg/kg 0.62¡À0.06 19.89¡À0.80 19.89¡À0.88 62.92¡À1.07 1:3.16 21.00 +6.25
5 mg/kg 0.52¡À0.04b 16.37¡À0.68b 14.62¡À0.74b 40.36¡À1.68b 1:2.76 31.00 +16.25
6 mg/kg 0.47¡À0.03b 15.98¡À0.73b 13.63¡À0.39b 35.09¡À1.92b 1:2.57 35.75 +21

3.3 Sperm count

The sperm count was significantly reduced in both the 5 and 6 mg/kg, but not in the 4 mg/kg
treated groups compared with the controls (Table 3).

Table 3. Effect of sodium arsenite on the sperm count and testicular arsenic content in rats (n=8). bP<0.05, compared with
controls.

Sperm count
Group Elementary arsenic (mg/g)
(106/cauda epididymis)

Control 135.75¡À8.39 0.98¡À0.27


4 mg/kg 122.87¡À7.28 2.68¡À0.42
5 mg/kg 84.81¡À6.33b 4.39¡À0.31b
6 mg/kg 65.09¡À7.32b 6.58¡À0.29b

3.4 Plasma hormonal levels


At the dose of 5 and 6 but not 4 mg/kg, the plasma levels of FSH and LH were significantly
decreased compared with the controls. The changes were more prominent in the 6 mg/kg group
(Figures 1 and 2).

Figure 1. Effect of sodium arsenite on plasma level of FSH. bP<0.05, compared with controls (n=8).

Figure 2. Effect of sodium arsenite on plasma level of LH. bP<0.05, compared with controls (n=8).

Plasma testosterone was significantly decreased (P<0.01) in rats of the 5 and 6 mg/kg group and
was more prominent in the latter compared with the controls. It was not significantly changed in
the 4 mg/kg group (Figure 3).

Figure 3. Effect of sodium arsenite on plasma level of testosterone. bP<0.05, compared with controls (n=8).¡¡

3.5 Testicular arsenic concentration

The arsenic concentration was significantly increased in the testes in all the treated animals in a
dose-dependent manner (Table 3).

4 Discussion

A significant decrease in the plasma FSH, LH and testosterone levels and degenerative changes
in testicular histology in arsenite treated rats are in agreement with previous findings of the
inhibitory effect of arsenite on the gonadal structure and function in mice [13] and fishes [8-10]
and the testicular ∆5-3β-HSD and 17β-HSD activities in rats [11]. As these enzymes are
gonadotrophin dependent [22], a decrease in their levels may reflect reduced pituitary
gonadotrophin secretion.

It was indicated that in parallel with the decrease in 7Sd and increased 7Sd degeneration, the
sperm count was markedly reduced. Earlier reports have revealed degenerative changes in the
testicular histology in fish and mice treated with arsenite [8, 13].

LH and FSH are required for the initiation and maintenance of spermatogenesis in prepubertal
and pubertal rats [23] and for quantitatively normal spermatogenesis in pubertal rats [17]. The
reduction of FSH and LH and a consequent reduction in testosterone production may, therefore,
be held responsible for this arsenite-induced changes in spermatogenesis. The reduction in the
number of ASg in arsenite treated rats is possibly due to the increased rate of degeneration of
Asg as FSH inhibits the normal degeneration of ASg and reduced FSH secretion may promote
ASg degeneration.

We found that higher doses of sodium arsenite increase adrenocortical activity and elevated
serum corticosterone level [4], which in turn may reduce the serum gonadotriphin and
testosterone levels [24]. Inhibitory effects of glucocorticoids on LH secretion have been reported
in cultured pituitary [25]. Glucocorticoids also directly suppress testosterone production and
secretion by decreasing the testicular LH receptors [26], resulting in the reduction of
spermatogenesis and sperm count.

It appears that the primary site of arsenic action may be on the brain or pituitary, however, a direct
action on the germ cells cannot be ruled out and further studies are required to clarify these
points.

References
[1] Nickson R, McArthur J, Burges W, Ahmed KM, Ravenserof P, Rahman M. Arsenic poisoning of
Bangladesh ground water. Nature 1998; 395: 338.
[2] Borzsonyl A, Bereczky A, Rudnai P, Csanady M, Horvath A. Epidemiological studies on human
subjects exposed to arsenic in drinking Water in Southern Hungary. Arch Toxicol 1992; 66: 77-8.
[3] Chatterjee A, Das D, Chatterjee D. The study of ground water contamination by arsenic in the
residental area of Behala, Calcutta, due to industrial pollution. Environ Pollution 1993; 80: 57-65.
[4] Biswas NM, Roy Chowdhury G, Sarkar M. Effect of sodium arsenite on adrenocortical
activities in male rats: dose-duration dependent responses. Med Sci Res 1994; 23: 153-4.
[5] Sarkar M, Ghosh D, Biswas HM, Biswas NM. Effect of sodium arsenite on haematology in
male albino rats. Ind J Physiol Allied Sc 1992; 46: 116-20.
[6] Robert EM, Judd ON. Water and soil pollutants. In : Klassen CD, Ambur MD, J Doull, editors,
Toxicology - The basic science of poison. 3rd edition. New York: Macmillan Publishing Company;
1986. p825.
[7] Brown MM, Rhyne BC, Boyer RA, Fowler BA. Intracellular effects of chronic arsenite
administration on renal proximal tubule cells. J Toxicol Environ Health 1976; 1: 505-14.
[8] Shukla JP, Pandey K. Impaired spermatogenesis in arsenic treated fresh water fish. Colisa
fasciatus (Bl & Sch). Toxicol Lett 1984; 21: 191-5.
[9] Shukla JP, Pandey K. Arsenic induced cellular and biochemical changes during the testicular
cycle of a fresh water perch. Colisa fasciatus (Bl & Sch). Cell Mol Biol 1984; 30: 227-31.
[10] Shukla J P, Pandey K. Toxicity and long term effect of arsenic on the gonadal protein
metabolism in a tropical fresh water fish. Colisa fasciatus (Bl & Sch). Acta Hydrochem Hydrobiol
1985; 13: 127-31.
[11] Sarkar M, Biswas NM, Ghosh D. Effect of sodium arsenite on testicular ∆5- 3β and 17β-
hydroxysteroid dehydrogenase activities in albino rats: dose and duration dependent response.
Med Sci Res 1991; 19: 789-90.
[12] Golub MS, Macintoch MS, Baumrind N. Development and reproductive toxicity of inorganic
arsenic: animal studies and human concerns. J Toxicol Environ Health B Crit Rev 1998; 1: 199-
241.
[13] Pant N, Kumar R, Murthy RC, Srivastava SP. Male reproductive effect of arsenic in mice.
Biometals 2001; 14 : 113-7.
[14] Leblond PC, Clermont Y. Definition of the stages of the seminiferous epithelium in the rat.
Ann N Y Acad Sci 1952; 55: 548-73.
[15] Abercrombie M. Estimation of nuclear population from microtome sections. Anat Rec 1946;
94: 239-47.
[16] Clermont Y, Morgentalor H. Quantitative study of spermatogenesis in hypophysectomized
rats. Endocrinology 1955; 57: 369-82.
[17] Russell LD, Alger LF, Naquin LG. Hormonal control of pubertal spermatogenesis.
Endocrinology 1987; 120: 1615-32.
[18] Clermont Y, Harvey SC. Effect of hormones on spermatogenesis in the rat. Ciba Foundation
Coll. Endocrinology 1967; 16: 173-96.
[19] Greenwood FC, Hunter WM, Glover J S. The preparation of 131I-labeled human growth
hormone of high specific activity. Biochem J 1963; 89: 114-23.
[20] Auletta FJ, Caldwell BV, Hamilton G. Androgen: testosterone and dihydrotestosterone. In:
Jaffe BM, Behrman HR, editors. Methods of Hormone Radioimmunoassay. New York: Academic
Press; 1974. p359-70.
[21] N¨¹rnberg HW. Processing Biological Samples for Metal Analysis. In: Brown SS, Savory J,
editors. International Union of Pure and Applied Chemistry; New York: Academic Press; 1983.
p31-44.
[22] Murono EP, Payne AH. Testicular maturation in the rat. In vivo effect of gonadotrophins on
steroidogenic enzymes in hypophysectomized immature rats. Biol Reprod 1979; 20: 911-6.
[23] Chowdhury AK. Dependence of testicular germ cells on hormones: A quantitative study in
hypophysectomized testosterone - treated rats. J Endocrinology 1979; 83: 331-40.
[24] Philips DM, Lakshmi V, Monder C. Corticosteroid 11b-dehydrogenase in rat testis.
Encocrinology 1989; 125: 209-16.
[25] D'Agostino JB, Valadka RJ, Schwartz NB. Differntial effect of in vitro glucocorticoids on LH
and FSH secretion: Dependence of sex of pituitary donor. Endocrinology 1990; 127: 891.
[26] Bombino TH, Hsueh AJW. Direct effect of glucocorticoids upon testicular luteinizing hormone
receptor and steroidogenesis in vivo and in vitro. Endocrinology 1989; 125 : 209-16.

Niraj Pant1 , Rakesh Kumar1 , Ramesh C. Murthy1 and


Satya P. Srivastava1

(1) Industrial Toxicology Research Centre, Post Box No 80, M.G. Marg, Lucknow-, 226001, India
Abstract Arsenic, a known human carcinogen, was given to
mice via drinking water as sodium arsenite at a dose 53.39,
133.47, 266.95 and 533.90 mol l for 35 days. A decrease in the
activity of 17 HSD along with increase in LDH, GT activity
were observed at 533.90 mol l. The observed sperm count,
motility and morphological abnormalities in sperm were similar to
control at lower dose levels. However at 533.90 mol l a
significant decrease in sperm count and motility along with
increase in abnormal sperm were noticed. Significant
accumulation of arsenic in testes and accessory sex organs may
be attributed to the arsenic binding to the tissues or greater
cellular uptake. No effects were observed on indices studied for
reproductive effects at 53.39 mol l arsenic close to which
human being are exposed through drinking water under the
present set of experimental conditions.

arsenic - drinking water - sperm - testes

You might also like