SciELO - Scientific Electronic Library Online

 
vol.14 issue2Feline asthma: pathophysiology, diagnosis and treatment author indexsubject indexarticles search
Home Pagealphabetic serial listing  

Services on Demand

Journal

Article

Indicators

  • Have no cited articlesCited by SciELO

Related links

Share


InVet

On-line version ISSN 1668-3498

InVet vol.14 no.2 Ciudad Autónoma de Buenos Aires July/Dec. 2012

 

ARTÍCULO DE INVESTIGACIÓN

Efecto del cadmio en órganos maternos de ratas y fetos en diferentes tiempos de gestación

 

Díaz, M.1; Quiroga, M.A.2; Landa, R.2; Giordano, A.2; Najle, R.1

1Departamento de Ciencias Biológicas,
2Centro de Investigación Veterinaria de Tandil (CIVETAN ) CONICET, Departamento de Fisiopatología. Facultad de Ciencias Veterinarias; Universidad Nacional del Centro de la Provincia de Buenos Aires (UNCPBA), Campus Universitario; Paraje Arroyo Seco s/n (7000) Tandil, ARGENTINA . Fax 54 0249 4439850

Correspondencia e-mail: Maria del Carmen Diaz maridel@vet.unicen.edu.ar

Recibido: 10/07/2012
Aceptado: 06/07/2013

 


Resumen

Para evaluar los efectos de la administración de cadmio (Cd) a diferentes tiempos de gestación, 24 ratas fueron preñadas bajo condiciones estandarizadas de crianza. Confirmada la preñez, las ratas fueron asignadas al azar a 4 tratamientos, que recibieron una dosis subcutánea de 10 mg Cd por kg PV a los días 7(GI); 9(GII) y 11(GIII) de preñez; o una dosis equivalente de solución fisiológica (GIV - control). El día 20 posconcepción, todas las ratas fueron sacrificadas. Se obtuvieron muestras de hígado, riñón, bazo, pulmón, fetos y placenta, para determinar la concentración tisular de Cd y para estudios histológicos. Los resultados fueron analizados estadísticamente (ANOVA y test t de Student). En los órganos de ratas de los grupos GI, GII and GIII, las concentraciones de Cd fueron significativamente más elevadas que en el control (p<0.05). En los grupos tratados con Cd, se observaron riñones con núcleos picnóticos en los túbulos de la corteza renal; hígado con infiltrado leucocítico multifocal, vacuolización celular en la zona centrolobulillar y cuerpos apoptóticos. En las placentas, se determinaron núcleos picnóticos, depósitos fibrinoides e infiltración de granulocitos. Se observó agenesia de extremidades, cola y cráneo en los fetos.

Palabras claves: Ratas; Cadmio toxicidad; Órganos maternos; Preñez; Fetos.

Effect of cadmium on maternal organs of rats and fetuses at different times of gestation

Summary

To evaluate the effects of the cadmium (Cd) administration at different gestation times, 24 rats were mated under standard rearing conditions. After pregnancy confirmation, rats were randomly assigned to 4 treatment groups. Experimental groups received a subcutaneous dose of 10 mg Cd per kg of body weight at the day: 7 Group I(GI); 9 Group II(GII) and 11 Group III(GIII) of pregnancy. The control group (Group IV- GIV) received an equivalent volume of saline solution. On day 20 post-conception, all rats were sacrificed. Samples of liver, kidney, spleen, lung, placenta and fetuses were collected to determine Cd concentration and for histological studies. The results were analyzed statistically (ANOVA and Student's t test). In the organs and fetuses of rats from GI; GII and GIII, Cd concentrations were significantly higher than in the control (P<0.05). In Cd-treated groups, were observed: kidneys with picnotic nuclei in tubules of the renal cortex; liver with multifocal leukocytic infiltrate, cellular vacuolization in the centrolobular zone; lungs with atelectasis and alveolar emphysema; placentas with picnotic nuclei, fibrinoid deposits and infiltration of granulocytes. Bone lesions were observed in fetuses, such as limb, tail and skull bones agenesis.

Key words: Rats; Cadmium toxicity; Maternal organs; Pregnancy; Fetuses.


 

Introduction

The exposition to heavy metals generates deleterious effects on the reproduction and prenatal development of man and animals. Among those metals considered as toxicant is cadmium (Cd) an element considered as nonessential for animal physiology.
Cadmium is widely distributed in the environment (air, soil, water)31 . It is not biodegradable and is present in the nature forming different oxides and salts (carbonates, sulphates, sulphites or chlorides)19 .
Modern agricultural and industrial practices have favored the exposure to Cd and other metals, since these elements are frequently found in insecticides, fungicides, phosphated fertilizers, paints, and batteries18, 23. Cadmium can also contaminate the air during the meeting process of rocks to extracting Zn, Cu, and Pb31.
Cadmium can cause damage to different tissues of many organs in mammals (kidney, liver, pancreas, ovaries, testicles, placenta and bones, among others)1, 20, 22, 25, 42.
The lung is a target organ for cadmium toxicity13, 38. It has been reported that exposure to cadmium chloride by inhalation induced lesions in the alveolar septum in rats11.
Data showed that Cd produced enhanced lipid peroxidation in the liver, heart and spleen32. Among the Cd teratogenic effects, it is highlighted the embryologic and fetal toxicities26. Studies performed in rats with 109Cd corroborated that Cd cross the placenta and accumulate in fetuses29. Embryos can be affected depending on the stage of development and time of exposure. In newborns, Cd toxicity seems to be dependant on other factors, such as the development to the hemato-encephalic barrier.
Among other possible sources of pollution, it can be cited contaminated food products such as vegetables, edible tissues of cattle (liver, kidney), fishes, seafood and mollusks20.
According to studies pe r formed by Kjëllstrom25 alterations determined by intoxication with Cd are very variables, determining acute or chronic effects that depend on factors such as the dose, time of exposition and the route of contamination38 .
Both direct and indirect exposition to Cd can be deleterious to human health. The effects of an indirect exposition can be also observed in the progeny. The Cd exposition previous to pregnancy or during it can affect the body weight of the offspring41 .
The mechanism of action of Cd on the fetus has not been elucidated yet. Proposed mechanisms are changes of metabolic processes in the maternal organism or disturbances in trophic functions of the placenta10.
The placenta of rats is similar to that of the woman, and it has been used as an animal model for studies of teratogenicity induced by Cd4, 10, 14, 41.
The objective of the present work was to evaluate the effects of Cd administration at different times of pregnancy, by reporting on Cd tissue concentrations and morphology of maternal organs, placenta and fetuses.

Material and methods

Animals

Twenty four (24) virgin female Wistar rats, 4 month-old and weighing 242.8 ± 22.3g (Laboratory Animal Rearing Facilities, Faculty of Veterinary Science, Universidad Nacional del Centro de la Provincia de Buenos Aires)5. Animals were housed in 12 plastic boxes in standard rearing conditions (temperature 22º ± 2ºC, relative humidity 50-60%, and a light period of 12 hours). Extruded feed and water ad libitum were provided during the experimental period.

Reproductive program

Rats were mated with 6-month-old males in a female: male rate of 2:1. Pregnancy was confirmed by the presence of spermatozoa in the vaginal fluid (day 0). Pregnant females were single housed and weighed on days 0 and 20 of pregnancy to determining the gestation status.

Treatments

Pregnant rats were assigned to 4 groups. Three groups received a subcutaneous dose equivalent to 10 mg of Cd+2 (as CdCl2.H2O) per kg of BW. The times of administration for the experimental groups were at the following pregnancy days: 7, Group I (GI); 9, Group II (GII); and 11, Group III (GIII). The fourth group was the control group, Group IV (GIV), which was administered an equal volume of saline solution.

Sample collection

On day 20 post-conception, all the gestating rats were sacrificed according to the methods established by the Animal Welfare Act of the Faculty of Veterinary Science22. In this particular case, an overdose of ethylic ether was used.
Samples of the following maternal organs were collected: liver, kidney, spleen, lung and placenta. A portion of the sampled organs was dried to constant weight to determining the Cd concentration by atomic absorption spectrophotometry (AAS).
Some of the analyzed organs (liver, kidney and placenta) were fixed with buffered formalin at a 10% concentration in PBS, for further histological studies.
Morphological determinations. Weight of each litter was determined, as well as the following lengths: total, head to tailhead (HTL), and head (HL). Placentas were weighed and diameters measured.
Determination of Cd concentration. Samples of maternal organs (liver, kidney, spleen and lung) placentas and fetuses were dried at 70ºC until a constant weight was achieved.
Approximately 50 mg of the samples were homogenized and digested with 0.5 mL of nitric acid 70% during 6 hours at room temperature and for 12 hours at 90ºC. Cd concentrations were determined by AAS, by using a GBC 906 equipment.
Histological examination. Samples of kidney, liver, lung, spleen, placenta and fetuses were fixed in 10% neutral buffered formalin, dehydrated with ethanol and xylene, embedded in paraffin, cutted into 5 μm slices and stained with Hematoxylin and Eosin for microscopic examination.
Samples of fetuses were fixed in ethanol 96º and their organs were removed. The skeletons of fetuses were examined by the Dawson alizarinred staining technique (Dawson technique modified by Barrow)7. Statistical analysis. Results of morphological measures and Cd tissue concentrations in the different organs of each group were analyzed by ANOVA and Student's t test. Differences with a p< 0.05 were considered as statistically significant and were included in tables that were constructed depicting the main values and the standard deviations for each experimental group.

Results

Morphological determinations

Weights and diameters of placentas from Cd-treated (groups I, II and III) and control animals (group IV) were similars; observed differences were statistically non-significant (p>0.05). (Table 1).

Table 1. Placental morphometric measures (mean ± S.E) of rats sacrificed on day 20 of pregnancy from different experimental groups (n=6).

Statistically non-significant differences were observed between treated groups versus control (p>0.05).

The size of the litter was lower only in G II compared to the control group. The mean gestational sac weight and the mean fetal weight were lower and the mean length, HTL and HL were shorter in treated animals than in the control group. However, differences were statistically non-significant (p>0.05) (Table 2).

Table 2. Fetal morphometric measures (mean ± S.E.) of rats sacrificed on day 20 of pregnancy from different experimental groups (n=6).

Litter: number of fetuses by rat; HTL: Head to Tail Length; HL: Head Length. Statistically non-significant differences were observed between treated groups versus control (p>0.05).

Cadmium tissue concentrations

Cadmium concentrations (mean ± SD), expressed as ppm DM in the tissues of the analyzed organs are presented in Table 3. In the organs of rats treated with Cd (GI; GII and GIII), significantly higher concentrations compared to those in the control group were observed (p<0.05). Cd concentrations differed between the analyzed organs, being in descendant order: kidney > liver > spleen > lung > placenta > fetuses.

Table 3. Cadmium concentration (ppm DM) (Mean ± DS) determined in the organs of rats, placenta and fetuses in the different experimental groups (n=6).

Rate Pl/ F: Cd concentrations relationship between placenta vs. fetuses. Differences between treated and control group are statistically significant at p< 0.05 (a).

Cd levels were higher in the different organs of G III, evidencing an inverse relation between Cd concentration and time from treatment to sacrifice. However, in the spleen, that tendency was not observed.

Histological examination

The main observations performed in the slices of kidneys, liver, lungs, spleen, fetuses and placenta belonging to animals of the different experimental groups are described:
Kidneys. In all the three Cd treated groups, tubular cells presented picnotic nuclei (Figure 1)


Figure 1.
Renal cortex from Group II rats (day 9). Arrows indicate picnotic nuclei (necrosis) of the tubular cells. (Hematoxyline-Eosin, (H&E) 40x)

In G II, tubules in the renal cortex showed a turbid tumefaction.
Liver. It was observed in Group I, multifocal leukocytic infiltrate, loss of cellular details, cellular vacuolization in the centrolobular zone (Figure 2); in other groups, congestion area, with multiple focuses of necrosis and leukocytic infiltrate.


Figure 2.
Liver slice from Group I rats (day 7). Arrows indicate the leukocytes infiltrate. (H &E, 40x)

Lungs. It was observed emphysema, atelectasis and congestion (Figure 3), epithelial desquamation of bronchioles in all treated groups and fibrinoid deposits in G III.


Figure 3.
Emphysema, atelectasis and congestion in lung. Group III (day 11). (H&E, 20x)

Spleen. Disorganization of the white pulp and hemosiderine deposits were observed in animals of groups I and II.
Placenta
. It showed trophoblast necrosis in G III (Figure 4), hemorrhage and picnotic nuclei of giant cells in the placenta of Group I.


Figure 4.
Necrosis of trophoblast in G III (H&E, 40x)

Fetuses. Some fetuses of the GI, GII and GIII presented incomplete ossification of the vertebrae , while other showed alterations of the architecture of the vertebral pericondrium and limbs (Figure 5, b1 y b2).


Figure 5.
Hinlimbs from (a) control and (b) contaminated fetuses. (a1 and b1, 20x stained with H&E; a2 and b2, 10x stained with Masson technique).

Using the Alizarin red technique it was observed lack of head bones and incomplete ossification of the vertebrae in some fetuses of G III. Also, some fetuses of GI, G II and G III presented bone alterations in the limbs (Figure 6).


Figure 6.
Different grades of ossification in limbs from fetuses of Control and Cd contaminated groups. (Allizarin technique)

Discussion

Cadmium is one of the heavy metals that can affect most of the organs, apparatus and systems of the mammal body20, 31.
Liver, kidney, lung and bones are the most affected organs9, 33.
The severity of these alterations depends, among other factors, upon the exposition route, dose, and time of exposition to the pollutant38, 46.
In gestating women exposed to high doses of Cd, placental and fetal alterations may occur.
Different administration routes have been proposed to perform intoxication experiences. Among them are the oral and the subcutaneous routes2.
The percentages of Cd absorbed differ depending on the route of exposition31. When the subcutaneous route is used, higher organic concentrations are achieved compared to those alter the oral administration. This can be explained because in the subcutaneous route there is no competition for absorption with other elements as could be expected at the intestinal cell lining46.
There is some controversy in determining the lethal dose, the LD50 and the minimum toxic dose for Cd30 . As a result of the bibliographic review, we decided to administer a dose equivalent to 10 mg/kg of body weight, which is similar to the 8 mg/kg dose used by Zhao et al. to produce in vivo teratogenic effects in rats49 .
Cadmium can produce placental alterations29, 36, 44. In our experimental model, Cd was administered to rats on days 7, 9 or 11 of pregnancy since in this species, on day 7, implantation is complete and the vitelin placenta begins to form; on day 9, the formation of the corio-vitelin placenta is complete, and, on day 11, the corio-alantoid placenta is developing4, 9, 10.
A high percentage (50 al 75%) of Cd deposited in the organism is found in the liver and kidneys8, 20, 48.
The concentrations determined in these organs in the present work are in agreement with those reported elsewhere 6,40. The deposition of the element produces hepatotoxicity and nephrotoxicity3, 6, 15, 42, 48 .
The pathogenesis of these alterations could be hypothesized as follow: the liver is the first organ to take most of the absorbed Cd1, 48, inducing the synthesis of intrahepatic metallothionein (MT-Cd) in order to avoid the Cd cytotoxic effect in that cells19, 29, 30, 32.
Hence, Cd reaches the kidneys where the MT-Cd complex is filtered. The epithelial cells lining the convoluted proximal tubules take by endocytosis a fraction of the filtered MT-Cd complex47.
This endocytosed fraction would be responsible for the cellular lesions in the kidneys.
It has been shown that the kidney is the most sensitive organ to Cd intoxication1, 20. It is characterized by glomerular lesion and cellular degeneration in the proximal convoluted tubules, these determining alterations in the renal function20.
The Cd values found in liver and kidneys of treated Cd groups would support the above described process. For example, in Group I, Cd was probably early deposited and later eliminated in a great percentage, since the time between treatment and sacrifice was greater that in the other groups.
Concentrations of Cd determined in the spleen and lung37 of animals treated with this heavy metal show increments that could be explained by the function accomplished by the studied organ. The results obtained in the present study are in agreement with those reported by other autor37.
The spleen, among other functions, acts as a blood reservoir and as the place for hemocatheresis,17 while the lungs acts as an important filter of the blood16, 37, and can retain little clots. It can also be considered as a blood reservoir, since it posses a great vascular net that can contain a great volume of blood at a given time.
In the spleen, a direct relationship between Cd concentration and time from treatment to sacrifice was established. It could be due to the incorporation of the heavy metal into the erythrocytes of treated animals.
In the lung, an inverse relationship between Cd concentration and time from treatment to sacrifice was determined. This is probably due to a higher renal clearance of the blood Cd in animals treated on day 7 of gestation (GI) than in the animals of the other experimental groups. Hence, the circulating Cd concentrations reaching the lungs would be low.
Concentrations of Cd in the placenta are directly related with the placental development stage27. The administration of Cd during the early gestation (GI and GII) determined that Cd passed the placental barrier and a greater percentage was later eliminated.
The placenta of animals in the GIII was more developed at the time of treatment, and Cd passed in a lesser extent and its tissue accumulation took place, which was responsible for the higher Cd concentrations observed2.
The greatest percentage of Cd accumulated in the kidneys is located presumably in the proximal tubules46. The histological findings produced by the toxic effect of Cd in the kidneys and reported in the present work are in agreement with those reported elsewhere24, 39.
The morphological alterations described here correspond to an acute/sub acute intoxication, since the experimental animals were sacrificed at 13, 11 and 9 days post-treatment (GI, GII and GIII, respectively).
The death of hepatocytes of Cd-intoxicated rats can be due to necrosis33.
The histopathological results obtained in the present study are similar to those reported in the reviewed bibliography12, 28, 43, 45. The severity of the morphological alterations would be directly correlated with the time elapsed between treatment and sacrifice of the experimental animals.
The administration of Cd to gestating rats produces necrosis of the placenta. Parizek35 and Padmanabhan34 observed that the administration of Cd on day 7 of gestation could determine severe placental malformations, since Cd could affect the ecthoplacental cone and the allantoids. The residual Cd in the maternal circulation could damage the placenta and pass through it causing teratogenesis29.
The Cd deposits can affect the placental function35. According to the results of the present study, the severity of the lesions is in inverse relation with the stage of placental development, as it is described for animals of the GI.
The mean weight of placenta from intoxicated females was lower than in the control group, which is in agreement with other assays12, 29 .
A direct correlation could be determined between the low weight and the size of fetuses and the low weight of placentas of rats treated with Cd (GI, II and III).
Different authors have reported the effects of Cd on the fetal ossification process during pregnancy in rats40 particularly in the vertebrae, ribs and metacarpal bones8.
Skeletal alterations observed in fetuses from G II and III, could be due to the characteristic embryogenesis of the rat, where the intraembryonic mesoderm extend on each side of the notochordal plate and specifically the beginning of somite formation takes place at the end of day 9 or during day 10 of pregnancy21.
The formation of the intra-embryonic mesoderm and its differentiation in regions is fundamental for the formation of cartilaginous structures, which will be the base for the skeletal system development21.
The administered dose of Cd determined increments in the tissue concentrations of the microelement and morphologic alterations in the studied organs, which are compatible with an acute/sub acute Cd intoxication27, 45.
The dosage regimen described in the present article can be used for studying morphological and functional alterations produced by Cd intoxication in rats.
Further research is required to confirm some of these results and to understand some of the mechanisms involved on the development of placental and fetal alterations induced by the cadmium administered during the gestation.

References

1. Agency for Toxic Substances and Disease Registry (ATSDR). Toxicological profile for cadmium. Atlanta, G.A. 1999.         [ Links ]

2. Ahokas, R.; Dilts, P.Cadmium uptake by the rat embryo as a function of gestational age. Am. J. Obstet. Gynecol.1979; 135:219- 222.         [ Links ]

3. Akesson, A.; Lundh, M.; Vahter, M.; et al. Tubular and glomerular kidney effects in Swedish women with low environmental cadmium exposure. Environ Health Perspect. 2005;113:1627-1631.         [ Links ]

4. Amoroso, E. Marshall's Phisiology of Reproduction.In Parkes, A.S, (eds). Placentation Placenta Haemochorialis. 1964; 243-276.         [ Links ]

5. Animal Welfare Act of the Faculty of Veterinary Science, Universidad Nacional del Centro de la Provincia de Buenos Aires 2002.         [ Links ]

6. Arnold, S.; Zarnke, R.; Lynn, T.; Chimonas, M.; Frank, A. Public health evaluation of cadmium concentrations in liver and kidney of moose (Alces alces) from four areas of Alaska. Sci Total Environ. 2006; 357:103-111.         [ Links ]

7. Barrow, P. Technical procedures in reproduction toxicology. Royal Soc Med Serv, London. 1990.         [ Links ]

8. Bhattacharyya, M. Cadmium osteotoxicity in experimental animals: Mechanisms and relationship to human exposures. Toxicology and Applied Pharmacology. 2009; 238: 258-265.         [ Links ]

9. Bhattacharyya, M.; Wilson, A.; Tajan. S.; Jonah, M. Molecular Biology and Toxicology of Metals. In Zalups, R.K., Koropatnick, J, editors. Biochemical pathways in cadmium toxicity. Taylor and Francis, London. 2000; 34-74.         [ Links ]

10. Björkman, N. Fine structure of the fetal-maternal area of exchange in the epitheliochorial and endotheliochorial types of placentation. Acta Anatómica. 1973; 86:1-22.         [ Links ]

11. Bus, S.;Vinegar, A.; Brooks, S. Biochemical and physiologic changes in lungs of rats exposed to a cadmium chloride aerosol. Am. Rev. Respir. Dis. 1978; 118: 573-580.         [ Links ]

12. Copius Peereboom-Stegeman, J.H.J.; van der Velde, W.J.; Dessing, J.W.M. Influence of cadmium on placental structure. Ecotoxicol Environ Safety. 1983; 7:79-86.         [ Links ]

13. Dechanet, C.; Anahory, T.; Mathieu Daude, J.; Quantin, X.; Reyftmann, L. et al. Effects of cigarette smoking on reproduction. Human Reproduction Update. 2011; 17: 76-9.         [ Links ]

14. Díaz, M.; Teruel, M.; García, V.; Catalano, R. Effects of Cadmium on placental and fetal parameters in Wistar rats. Revista Ciencias Morfológicas. 2006; 8: 5-11.         [ Links ]

15. Dudley, R.; Svoboda, D.; Klaassen, C. Acute exposure to cadmium causes severe liver injury in rats. Toxicology and Applied Pharmacology.1982; 65:302-313.         [ Links ]

16. Dukes, H.; Swenson, M. Fisiología de los animales domésticos. Ed. Aguilar.Tomo I, 1981, pp. 274.         [ Links ]

17. Fahim, M.; Nemmar, S.; Dhanasekaran, S.; Singh, M.; Shafiullah, J. et al. Acute Cadmium Exposure Causes Systemic and Thromboembolic Events in Mice. Physiological Research, 2012; 61 : 73-80.         [ Links ]

18. Franz, E.; Römkens, P.; van Raamsdonk, L.; van der Fels-Klerx, I. A chain modeling approach to estimate the impact of soil cadmium pollution on human dietary exposure. Journal of Food Protection. 2008; 71: 2504-2513.         [ Links ]

19. Goerin, P.; Klaassen, C. Resistance to cadmiuminduced hepatoxicity in immature rats. Toxicology and Applied Pharmacolog. 1984; 74: 321- 329.         [ Links ]

20. Goyer, R, Casarett and Doull´s: Toxicology.Toxics effects of metals, chapter 23. MacGraw-Hill Companies, Inc. New York 1996. pp. 699-702.         [ Links ]

21. Hebel, R. Anatomy and Embryology of the Laboratory Rat. BioMed Verlag editors.1976. 22. International Agency for Research on Cancer. Cadmium and certain cadmium compounds. IARC monographs, vol. 58, World Health Organization. Lyon., France, 1993; pp 119-146, 210-236.         [ Links ]

23. Kang, M.; Cho, S.; Lim, Y.; Seo, J.; Hong, Y. Effects of environmental cadmium exposure on liver function in adults. Occup Environ Med. 2013; 70:268-73. doi: 10.1136/oemed-2012-101063. Epub 2013 Jan 15.         [ Links ]

24. Kim, Y.; Choi, J.; Kim, J.; Park, Y. Changes in renal function in cadmium-intoxicated rats. Pharmacology and Toxicology. 1988; 63: 342-350.         [ Links ]

25. Kjëllstrom, T. Itai-itai disease. In Cadmium and Health, L Friberg y cols.editors Boca Raton. CRC Press. 1986.         [ Links ]

26. Kotsonis, F. Klaassen C. Toxicity and distribution of cadmium administered to rats at sublethal doses. Toxicology and Applied Pharmacology. 1977; 41: 667-680.         [ Links ]

27. Kuriwaki, J.; Muneko, N.; Honda, R. et al. Effects of cadmium exposure during pregnancy on trace elements in fetal rat liver and kidney. Toxicology Letters. 2005; 156: 369-376.         [ Links ]

28. Lag, M.; Westly, S.; Lerstad, T.; Bjørnsrud, C.; Refsnes, M.; Schwarze, P. Cadmium-induced apoptosis of primary epithelial lung cells: Involvement of Bax and p53, but not of oxidative stress. Cell Biology and Toxicology. 2002; 18:29-42.         [ Links ]

29. Levin, A.; Plautz, J.; di Sant'Agnese, P.; Miller, R. Cadmium: placental mechanisms of fetal toxicity. Placenta Supplement. 1981; 3: 303-318.         [ Links ]

30. Liu, J.; Liu,Y.; Klaassen, C. Metallothionein-null and wild-type mice show similar cadmium absorption and tissue distribution following oral cadmium administration. Toxicology and Applied Pharmacology. 2001; 175: 253-259.         [ Links ]

31. Mc Dowell, L. In: Minerals in Animal and Human Nutrition. Academic Press. .ISBN 0-12-483369-1. 1992; pp. 359-361.         [ Links ]

32. Nakamura, Y.; Ohba, K.; Suzuki, K. ; Ohta, H. Health effects of low-level cadmium intake and the role of metallothionein on cadmium-transport from mother rats to fetus. The Journal of Toxicological Sciences. 2012; 37:149-56.         [ Links ]

33. Nordberg, M.; Nordberg, G.; Kjellstrom, T. Kinetics and metabolism, in: Friberg L, Elinder C. and Kjellstrom T, editors. Cadmium and Health: A Toxicological and Epidemiological Appraisal, vol. I. CRC Press 1985; pp 103-178.         [ Links ]

34. Padmanabhan, R. The effect of Cadmium on Placental Structure and its Relation to Fetal Malformations in the mouse. Z. mikrosk.-anat.Forsch, Leipzig. 1986; 100: 419-427.         [ Links ]

35. Parize, J. The peculiar toxicity of cadmium during pregnancy: An experimental toxemia of pregnancy induced by cadmium salts. Journal of Reproduction and Fertility. 1965; 9: 111-112.         [ Links ]

36. Piasek, M.; Blanus, M.; Laskey, J. Placental cadmium and progesterone concentrations in cigarette smokers. Reproductive Toxicology. 2001; 15: 673-681. 37. Pirie, H.: Respiratory Diseases of Animals. Thompson Litho Ltd., East Kilbride editors. Scotland. 1988; pp. 15.         [ Links ]

38. Repetto, M. Toxicología Avanzada. Díaz de Santos, S.A editors . España. 1995. Capítulo 10. pp 393-418.         [ Links ]

39. Salvatori, F.; Talassia, C.; Salzgeberb, S.; Spinosa, H.; Bernardia, M. Embryotoxic and long-term effects of cadmium exposure during embryogenesis in rats. Neurotoxicology and Teratology. 2004; 26: 673-680.         [ Links ]

40. Satarug, S.; Baker, J.; Reilly, P.; Moore, M.; Williams, D. Cadmium levels in the lung, liver, kidney cortex, and urine samples from Australians without occupational exposure to metals. Arch Environ Health. 2002; 57: 69-77.         [ Links ]

41. Soares, M.; Chakraborty, M.; Karim Rumi, T.; Konno, S. Renaud, R. Rat placentation: An experimental model for investigating the hemochorial maternal-fetal interface. Placenta. 2012; 33: 233-243.         [ Links ]

42. Souza Arroyo, V.; Karina Martínez Flores, K.; Bucio Ortiz, K.; Luis Enrique Gómez, L. et al. Liver and Cadmium Toxicity. J Drug Metab Toxicol. 2012, S5http://dx.doi.org/10.4172/2157-7609.S5-001.         [ Links ]

43. Sudo, J. et al. Mechanism of nephrotoxicity induced by repeated administration of cadmium chloride in rats. Journal of Toxicology & Environmental Health. 1996, 48: 333-348.         [ Links ]

44. Turgut, S. et al. Influence of cadmium and copper on tissue element levels of pregnant rats. Central European Journal of Medicine. 2007, 2: 447-457.         [ Links ]

45. Yamano, T.; Shimizu, M.; Noda, T.Comparative effects of repeated administration of cadmium on kidney, spleen, thymus, and bone marrow in 2-, 4- and 8-month-old male wistar rats. Society of Toxicology. 1998; 46: 393-402.         [ Links ]

46. Zalups, R.; Ahmad, S.Molecular handling of cadmiun in transporting epithelia. Toxicology and Applied Pharmacology. 2003; 163-188.         [ Links ]

47. Zalups, R.; Gelein, R.; Cherian, M. Shifts in the dose-effect relationship for the nephropathy induced by cadmium-metallothionein in rats after a reduction of renal mass. Journal of Pharmacology and Experimental Therapeutics. 1992; 262: 1256-1266.         [ Links ]

48. Zalups, R. Evidence for basolateral uptake of cadmium in the kidneys of rats. Toxicology and Applied Pharmacology. 2000; 164: 15-23.         [ Links ]

49. Zhao, Shu- Fen et al. The evaluation of developmental toxicity of chemicals exposed occupationally using whole embryo culture. International Journal of Development Biology. 1997; 41: 275- 282.         [ Links ]

Creative Commons License All the contents of this journal, except where otherwise noted, is licensed under a Creative Commons Attribution License