Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Int J Mol Sci ; 24(2)2023 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-36675277

RESUMEN

The pathogenesis of non-alcoholic fatty liver disease (NAFLD), the most prevalent chronic liver disease, is associated with zinc deficiency. Previous studies show zinc supplementation improves steatosis and glucose metabolism, but its therapeutic effects in patients with established NAFLD remain unclear. We developed an in vivo model to characterize the effects of zinc supplementation on high-fat diet (HFD) induced NAFLD and hypothesized that the established NAFLD would be attenuated by zinc supplementation. Male C57BL/6J mice were fed a control diet or HFD for 12 weeks. Mice were then further grouped into normal and zinc-supplemented diets for 8 additional weeks. Body composition and glucose tolerance were determined before and after zinc supplementation. At euthanasia, plasma and liver tissue were collected for characterization and downstream analysis. As expected, 12 weeks of HFD resulted in reduced glucose clearance and altered body composition. Eight weeks of subsequent zinc supplementation did not alter glucose handling, plasma transaminases, steatosis, or hepatic gene expression. Results from our model suggest 8-week zinc supplementation cannot reverse established NAFLD. The HFD may have caused NAFLD disease progression beyond rescue by an 8-week period of zinc supplementation. Future studies will address these limitations and provide insights into zinc as a therapeutic agent for established NAFLD.


Asunto(s)
Enfermedad del Hígado Graso no Alcohólico , Masculino , Ratones , Animales , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Enfermedad del Hígado Graso no Alcohólico/etiología , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Dieta Alta en Grasa/efectos adversos , Zinc/metabolismo , Ratones Endogámicos C57BL , Hígado/metabolismo , Suplementos Dietéticos , Glucosa/metabolismo , Modelos Animales de Enfermedad
2.
Toxicol Appl Pharmacol ; 436: 115855, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-34990729

RESUMEN

Nonalcoholic fatty liver disease (NAFLD) is a major global public health concern affecting more than 25% of the world's population. Although obesity and diabetes are major risk factors for NAFLD, they cannot account for all cases, indicating the importance of other factors such as environmental exposures. Cadmium (Cd) exposure is implicated in the development of NAFLD; however, the influence of early life, in utero Cd exposure on the development of diet-induced NAFLD is poorly understood. Therefore, we developed an in vivo, multiple-hit model to study the effect of whole-life, low dose Cd exposure on high fat diet (HFD)-induced NAFLD. Adult male and female C57BL/6 J mice fed normal diets (ND) were exposed to 0, 0.5 or 5 ppm Cd-containing drinking water for 14 weeks before breeding. At weaning, offspring were fed ND or HFD and continued on the same drinking water regimen as their parents for 24 weeks. Cd exposure at different concentrations differentially altered HFD-associated adverse health effects, including liver injury. HFD-induced increased body weight, decreased glucose tolerance. Liver injury and lipid deposition were exacerbated by 5 ppm Cd exposure but attenuated by 0.5 ppm Cd exposure. Further, HFD blunted the response of metallothionein, a major Cd detoxification protein, in mice exposed to 5 ppm Cd but enhanced the response in mice exposed to 0.5 ppm Cd, suggesting a possible mechanism for Cd alteration of HFD-induced NAFLD. These results confirm the multi-hit nature of NAFLD and show whole life, low dose Cd exposure alters HFD-induced NAFLD with outcomes dependent on Cd concentration.


Asunto(s)
Cadmio/efectos adversos , Hígado/efectos de los fármacos , Enfermedad del Hígado Graso no Alcohólico/inducido químicamente , Animales , Dieta Alta en Grasa/métodos , Modelos Animales de Enfermedad , Femenino , Metabolismo de los Lípidos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Estrés Oxidativo/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
3.
Ecotoxicol Environ Saf ; 234: 113373, 2022 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-35272187

RESUMEN

As an environmental pollutant, cadmium (Cd) has been widely reported to induce male infertility due to its gonadotoxicity. However, the specific mechanism of Cd-induced testicular damage remains unclear. We investigated whether Cd causes testicular injury through ferroptosis. Male C57BL/6 J mice were exposed to 0, 0.5, or 5 ppm Cd via drinking water, starting in utero, and continuing through 24 weeks post-weaning. The results showed that Cd accumulated in the testes in a dose-dependent manner. Cd exposure at a concentration of 5 ppm, but not 0.5 ppm, caused a mass loss and detachment of germ cells, as well as a decreased meiotic index and testis weight. Exposure to 5 ppm Cd caused iron accumulation, increased levels of malondialdehyde (MDA) and nitro tyrosine (3-NT), and decreased expression of Nrf2, HO-1 and SOD2. We also found that exposure to 5 ppm Cd significantly decreased the expression of SLC7A11, a marker of ferroptosis in mice, along with the expression of SLC40A1 mRNA and ferritin heavy chain (FTH) protein, whereas there was no obvious change in the mRNA expression of Tfrc, ZIP8, ZIP14, and NCOA4. These findings indicate that 5 ppm Cd exposure increased testicular ferroptosis, which may be attributed to the reduction of stored iron export.

4.
Toxicol Appl Pharmacol ; 403: 115161, 2020 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-32721433

RESUMEN

Cadmium is a ubiquitous, non-essential metal that has earned a spot on the World Health Organizations top 10 chemicals of major public health concern. The mechanisms of cadmium-induced adverse health outcomes, such as cardiovascular disease, renal toxicity and cancer, are well studied in adults. However, the implications for early life exposures to low-level cadmium leading to increased risk of developing diseases in adulthood remains elusive. Epidemiological investigation of the long term implications of cadmium-associated adverse birth outcomes are limited and studies do not extend into adulthood. This review will summarize the literature on the non-lethal, adverse health effects associated with prenatal and early life exposure to cadmium and the implications of these exposures in the development of diseases later in life. In addition, this review will highlight possible mechanisms responsible for these outcomes as well as address the inconsistencies in the literature. More recent studies have addressed sex as a biological variable, showing prenatal cadmium exposure elicits sex-specific outcomes that would otherwise be masked by pooling male and female data. Furthermore, researchers have begun to investigate the role of prenatal and early life cadmium exposures in the development of diet-induced diseases with evidence of altered essential metal homeostasis as a likely mechanism for cadmium-enhanced, diet-induced diseases. Although novel experimental models are beginning to be established to study the association between prenatal cadmium exposure and adverse health outcomes in adulthood, the studies are few, highlighting a major need for further investigation.


Asunto(s)
Cadmio/toxicidad , Contaminantes Ambientales/toxicidad , Efectos Tardíos de la Exposición Prenatal , Epigénesis Genética , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Humanos , Embarazo
5.
Chem Res Toxicol ; 32(6): 1070-1081, 2019 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-30912652

RESUMEN

Childhood obesity, which is prevalent in developed countries, is a metabolic risk factor for cardiovascular disease. Cadmium (Cd), a ubiquitous environmental toxic metal, also has deleterious effects on the cardiovascular system. However, the combined effects of a high-fat diet (HFD) and lifelong, low-dose Cd exposure on cardiac remodeling remain unclear. This study aims to determine the effects of combined HFD and Cd exposure on cardiac remodeling, as well as gender-specific differences in the response. C57BL/6J mice were exposed to Cd at a low dose (L-Cd, 0.5 ppm) or high dose (H-Cd, 5 ppm) via drinking water from conception to sacrifice. After being weaned, the offspring mice were fed with a HFD (42% kcal from fat) for an additional 10 weeks. H-Cd exposure significantly increased Cd accumulation in the hearts of both parents and their offspring; a HFD showed no added effects on cardiac Cd content. H-Cd exposure increased cardiac metallothionein protein levels only in female mice, regardless of dietary intake. Histological analysis revealed that H-Cd exposure combined with a HFD induced cardiac hypertrophy and fibrosis only in female mice. This was further supported by elevated expression of ANP and COL1A1 protein levels along with COL1A1, COL1A2, and COL3A1 mRNA levels. Profibrotic markers PAI-1, CTGF, and FN were also elevated in HFD/H-Cd-exposed female mice. Levels of the oxidative stress marker 3-NT significantly increased in the hearts of HFD-fed female mice, whereas Cd exposure showed no additional effects. Of all the antioxidant markers examined, levels of CAT significantly increased in mice fed a HFD, regardless of gender and Cd exposure. In summary, a HFD combined with lifelong, low-dose Cd exposure induces cardiac hypertrophy and fibrosis in female but not male mice, a response that is independent of oxidative stress.


Asunto(s)
Cadmio/administración & dosificación , Cadmio/toxicidad , Cardiomegalia/inducido químicamente , Dieta Alta en Grasa/efectos adversos , Fibrosis/inducido químicamente , Animales , Cardiomegalia/patología , Relación Dosis-Respuesta a Droga , Femenino , Fibrosis/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Estrés Oxidativo/efectos de los fármacos , Factores Sexuales , Factores de Tiempo
6.
Life Sci ; 315: 121385, 2023 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-36634865

RESUMEN

Cadmium is a hazardous metal with multiple organ toxicity that causes great harm to human health. Cadmium enters the human body through occupational exposure, diet, drinking water, breathing, and smoking. Cadmium accumulation in the human body is associated with increased risk of developing obesity, cardiovascular disease, diabetes, and metabolic syndrome (MetS). Cadmium uptake is enhanced during pregnancy and can cross the placenta affecting placental development and function. Subsequently, cadmium can pass to fetus, gathering in multiple organs such as the liver and pancreas. Early-life cadmium exposure can induce hepatic oxidative stress and pancreatic ß-cell dysfunction, resulting in insulin resistance and glucose metabolic dyshomeostasis in the offspring. Prenatal exposure to cadmium is also associated with increasing epigenetic effects on the offspring's multi-organ functions. However, whether and how maternal exposure to low-dose cadmium impacts the risks of developing type 2 diabetes (T2D) in the young and/or adult offspring remains unclear. This review collected available data to address the current evidence for the potential role of cadmium exposure, leading to insulin resistance and the development of T2D in offspring. However, this review reveals that underlying mechanisms linking prenatal cadmium exposure during pregnancy with T2D in offspring remain to be adequately investigated.


Asunto(s)
Diabetes Mellitus Tipo 2 , Resistencia a la Insulina , Síndrome Metabólico , Efectos Tardíos de la Exposición Prenatal , Adulto , Embarazo , Femenino , Humanos , Síndrome Metabólico/inducido químicamente , Síndrome Metabólico/complicaciones , Exposición Materna , Cadmio/toxicidad , Diabetes Mellitus Tipo 2/inducido químicamente , Diabetes Mellitus Tipo 2/complicaciones , Placenta/metabolismo , Efectos Tardíos de la Exposición Prenatal/metabolismo
7.
Alcohol Clin Exp Res (Hoboken) ; 47(1): 60-75, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36377258

RESUMEN

BACKGROUND: The prevalence of alcohol-associated liver disease (ALD), a subtype of fatty liver disease (FLD), continues to rise. ALD is a major cause of preventable death. Polychlorinated biphenyl (PCB) 126 is an environmentally relevant, dioxin-like pollutant whose negative metabolic effects have been well documented. In human and animal studies, PCB has been associated with the severity of nonalcoholic fatty liver disease (NAFLD). However, few studies have investigated whether exposures to environmental toxicants can worsen ALD. Thus, the objective of the current study was to develop an alcohol-plus-toxicant model to study how an environmental pollutant, PCB 126, impacts rodent ALD pathology. METHODS: Briefly, male C57BL/6J mice were exposed to 0.2 mg/kg PCB 126 or corn oil vehicle four days prior to ethanol feeding using the chronic-binge (10-plus-one) model. RESULTS: Concentrations of macromolecules, including hepatic lipids, carbohydrates, and protein (albumin) were impacted. Exposure to PCB 126 exacerbated hepatic steatosis and hepatomegaly in mice exposed to the chemical and fed an ethanol diet. Gene expression and the analysis of blood chemistry showed a potential net increase and retention of hepatic lipids and reductions in lipid oxidation and clearance capabilities. Depletion of glycogen and glucose was evident, which contributes to disease progression by generating systemic malnutrition. Granulocytic immune infiltrates were present but driven solely by ethanol feeding. Hepatic albumin gene expression and plasma levels were decreased by ~50% indicating a potential compromise of liver function. Finally, gene expression analyses indicated that the aryl hydrocarbon receptor and constitutive androstane receptor were activated by PCB 126 and ethanol, respectively. CONCLUSIONS: Various environmental toxicants are known to modify or enhance FLD in high-fat diet models. Findings from the present study suggest that they interact with other lifestyle factors such as alcohol consumption to reprogram intermediary metabolism resulting in exacerbated ethanol-associated systemic malnutrition in ALD.


Asunto(s)
Contaminantes Ambientales , Hepatopatías Alcohólicas , Desnutrición , Enfermedad del Hígado Graso no Alcohólico , Bifenilos Policlorados , Humanos , Masculino , Ratones , Animales , Bifenilos Policlorados/metabolismo , Bifenilos Policlorados/farmacología , Contaminantes Ambientales/metabolismo , Contaminantes Ambientales/farmacología , Roedores , Ratones Endogámicos C57BL , Hígado/metabolismo , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Hepatopatías Alcohólicas/metabolismo , Dieta Alta en Grasa , Etanol/farmacología , Lípidos/farmacología , Desnutrición/metabolismo , Desnutrición/patología
8.
Environ Int ; 158: 106877, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34547640

RESUMEN

Hexavalent chromium [Cr(VI)] is a global environmental pollutant that increases risk for several types of cancers and is increasingly being recognized as a neurotoxicant. Traditionally, the brain has been viewed as a largely post-mitotic organ due to its specialized composition of neurons, and consequently, clastogenic effects were not considered in neurotoxicology. Today, we understand the brain is composed of at least eight distinct cell types - most of which continue mitotic activity throughout lifespan. We have learned these dividing cells play essential roles in brain and body health. This review focuses on Cr(VI), a potent clastogen and known human carcinogen, as a potentially neurotoxic agent targeting mitotic cells of the brain. Despite its well-established role as a human carcinogen, Cr(VI) neurotoxicity studies have failed to find a significant link to brain cancers. In the few studies that did find a link, Cr(VI) was identified as a risk for gliomas. Instead, in the human brain, Cr(VI) appears to have more subtle deleterious effects that can impair childhood learning and attention development, olfactory function, social memory, and may contribute to motor neuron diseases. Studies of Cr(VI) neurotoxicity with animal and cell culture models have demonstrated elevated markers of oxidative damage and redox stress, with widespread neurodegeneration. One study showed mice exposed to Cr(VI)-laden tannery effluent exhibited longer periods of aggressive behavior toward an "intruder" mouse and took longer to recognize mice previously encountered, recapitulating the social memory deficits observed in humans. Here we conducted a critical review of the available literature on Cr(VI) neurotoxicity and synthesize the collective observations to thoroughly evaluate Cr(VI) neurotoxicity - much remains to be understood and recognized.


Asunto(s)
Cromo , Contaminantes Ambientales , Animales , Cromo/toxicidad , Daño del ADN , Contaminantes Ambientales/toxicidad , Ratones , Estrés Oxidativo
9.
Chem Biol Interact ; 353: 109797, 2022 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-34998821

RESUMEN

Although several studies have reported testicular impairments caused by cadmium (Cd) or obesity alone, the combined effect of Cd and obesity on the testes and its underlying mechanism remains unclear. We examined the combined effect of whole-life exposure to low-dose Cd started at preconception and post-weaning high-fat diet (HFD) on the testes of offspring mice. At weaning, male offspring parented with and without exposure to low-dose Cd were continued on the same drinking water regimen as their parents and fed with either a normal diet (ND) or HFD for 10 or 24 weeks. Whole-life exposure to Cd resulted in its accumulation in testes, and HFD induced obesity and lipid metabolism disorder. Exposure to Cd or HFD alone significantly decreased Johnsen scores, disrupted testicular structure, and increased germ cell apoptosis at both 10 and 24 weeks. However, co-exposure to Cd and HFD did not induce the toxic effects that were induced by either alone, as revealed by preserved testicular structure and spermatogenesis, lack of significant apoptosis, and increased cell proliferation. Mechanistically, the combined effects of low-dose Cd and HFD consumption were associated with the activation of the JAK/STAT pathway. These findings suggest that co-exposure to low-dose Cd and HFD did not cause Cd- or HFD-induced testicular injury, probably because of the activation of the JAK/STAT pathway to prevent germ cell apoptosis.


Asunto(s)
Cadmio/toxicidad , Dieta Alta en Grasa , Espermatogénesis/efectos de los fármacos , Testículo/fisiología , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Cadmio/análisis , Femenino , Células Germinativas/citología , Células Germinativas/metabolismo , Quinasas Janus/metabolismo , Metabolismo de los Lípidos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Estrés Oxidativo/efectos de los fármacos , Factores de Transcripción STAT/metabolismo , Transducción de Señal/efectos de los fármacos , Testículo/anatomía & histología , Testículo/química
10.
Sci Total Environ ; 809: 152176, 2022 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-34875320

RESUMEN

We previously showed the development of cardiac remodeling (hypertrophy or fibrosis) in mice with either post-weaning high-fat diet (HFD, 60% kcal fat) feeding or exposure to chronic low-dose cadmium. Here, we determined whether whole-life exposure to environmentally relevant, low-dose cadmium affects the susceptibility of offspring to post-weaning HFD-induced cardiac pathologies and function. Besides, we also determined whether these effects are sex-dependent. Male and female mice were exposed to cadmium-containing (0, 0.5, or 5 parts per million [ppm]) drinking water before breeding; the pregnant mice and dams with offspring continually drank the same cadmium-containing water. After weaning, the offspring were continued on the same regime as their parents and fed either a HFD or normal fat diet for 24 weeks. Cardiac function was examined with echocardiography. Cardiac tissues were used for the histopathological and biochemical (gene and protein expression by real-time PCR and Western blotting) assays. Results showed a dose-dependent cadmium accumulation in the hearts of male and female mice along with decreased cardiac zinc and copper levels only in female offspring. Exposure to 5 ppm, but not 0.5 ppm, cadmium significantly enhanced HFD cardiac effects only in female mice, shown by worsened cardiac systolic and diastolic dysfunction (ejection fraction, mitral E-to-annular e' ratio), increased fibrosis (collagen, fibronectin, collagen1A1), hypertrophy (cardiomyocyte size, atrial natriuretic peptide, ß-myosin heavy chain), and inflammation (intercellular adhesion molecule-1, tumor necrosis factor-α, plasminogen activator inhibitor type 1), compared to the HFD group. These synergistic effects were associated with activation of the p38 mitogen-activated protein kinases (MAPK) signaling pathway and increased oxidative stress, shown by 3-nitrotyrosine and malondialdehyde, along with decreased metallothionein expression. These results suggest that whole-life 5 ppm cadmium exposure significantly increases the susceptibility of female offspring to HFD-induced cardiac remodeling and dysfunction. The underlying mechanism and potential intervention will be further explored in the future.


Asunto(s)
Cadmio , Dieta Alta en Grasa , Animales , Cadmio/metabolismo , Cadmio/toxicidad , Dieta Alta en Grasa/efectos adversos , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Miocitos Cardíacos , Estrés Oxidativo , Embarazo , Caracteres Sexuales
11.
Sci Total Environ ; 818: 151848, 2022 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-34822883

RESUMEN

An increasing body of evidence implicates high levels of selenium intake in the development of diabetes, although prospective studies remain sparse. We conducted a nested case-control study of 622 diabetes incident cases and 622-age, sex, and follow-up time-matched controls in the prospective Jinchang cohort of 48,001 participants with a median of 5.8 years of follow-up. Inductively coupled plasma mass spectrometry (ICP-MS) was used to measure all 622 case-control pairs' baseline serum levels of selenium (Se), which were then categorized into quartiles based on the frequency distribution among the controls. Multivariable adjusted conditional logistic regression and restricted cubic splines (RCS) models were applied to evaluate independent odds ratios (OR) as estimates for relative risks (RR) of diabetes according to quartiles (Q) of selenium levels. Compared to the lowest quartile (Q1 as reference), significantly greater diabetes risks (with 95% confidence interval) were observed in Q3 (OR = 1.62, 1.17-2.35) and Q4 (OR = 1.79, 1.21-2.64). Sub-analyses showed these increased risks of diabetes by serum levels of Se. appeared to differ by sex, age, BMI status, history of hypertension, and dyslipidemia. Further, application of RSC models showed that serum Se levels between 95 and 120 µg/L were significantly and positively associated with diabetes risk whereas no apparent relation exists when Se levels were under 95 µg/L in this cohort population.


Asunto(s)
Diabetes Mellitus , Selenio , Estudios de Casos y Controles , Preescolar , Diabetes Mellitus/epidemiología , Humanos , Oportunidad Relativa , Estudios Prospectivos , Factores de Riesgo
12.
Oxid Med Cell Longev ; 2021: 1427787, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34876963

RESUMEN

Maternal exposure to cadmium causes obesity and metabolic changes in the offspring, including nonalcoholic fatty liver disease-like pathology. However, whether maternal cadmium exposure accelerates liver cancer in the offspring is unknown. This study investigated the impact of early-life exposure to cadmium on the incidence and potential mechanisms of hepatocellular carcinoma (HCC) in offspring subjected to postweaning HCC induction. HCC in C57BL/6J mice was induced by diethylnitrosamine (DEN) injection at weaning, followed by a long-term high-fat choline-deficient (HFCD) diet. Before weaning, liver cadmium levels were significantly higher in mice with early-life cadmium exposure than in those without cadmium exposure. However, by 26 and 29 weeks of age, hepatic cadmium fell to control levels, while a significant decrease was observed in copper and iron in the liver. Both male and female cadmium-exposed mice showed increased body weight compared to non-cadmium-treated mice. For females, early-life cadmium exposure also worsened insulin intolerance but did not significantly promote DEN/HFCD diet-induced liver tumors. In contrast, in male mice, early-life cadmium exposure enhanced liver cancer induction by DEN/HFCD with high incidence and larger liver tumors. The liver peritumor tissue of early-life cadmium-exposed mice exhibited greater inflammation and disruption of fatty acid metabolism, accompanied by higher malondialdehyde and lower esterified triglyceride levels compared to mice without cadmium exposure. These findings suggest that early-life exposure to low-dose cadmium accelerates liver cancer development induced by a DEN/HFCD in male mice, probably due to chronic lipotoxicity and inflammation caused by increased uptake but decreased consumption of fatty acids.


Asunto(s)
Cadmio/toxicidad , Dieta Alta en Grasa , Dietilnitrosamina/farmacología , Neoplasias Hepáticas/patología , Animales , Animales Recién Nacidos , Colina/metabolismo , Dieta Alta en Grasa/veterinaria , Modelos Animales de Enfermedad , Ácidos Grasos/metabolismo , Femenino , Hígado/patología , Neoplasias Hepáticas/inducido químicamente , Masculino , Malondialdehído/metabolismo , Ratones , Ratones Endogámicos C57BL , Triglicéridos/metabolismo
13.
Toxicol Rep ; 8: 718-723, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33889501

RESUMEN

The effects of exposure to the environmental toxicant cadmium, in combination with obesity, on the metal content in mouse testis were evaluated. Starting in utero and continuing through to 10 or 24 weeks post-weaning, male mice were exposed to cadmium (0, 0.5 or 5 ppm), and fed either a low (LFD) or high fat diet (HFD) post-weaning. Testicular levels of cadmium and essential metals were determined 10 and 24 weeks post-weaning by ICP-MS. Similar to what has been previously observed in the liver, kidney, heart and brain, significant levels of cadmium accumulated in the testis under all exposure conditions. Additionally, HFD-fed animals accumulated more cadmium than did their LFD-treated counterparts. Both treatments affected essential metal homeostasis in the testis. These findings suggest that cadmium and obesity may compromise the reproductive potential in the male mouse by disrupting essential metal levels.

14.
Mol Med Rep ; 22(2): 603-611, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32468027

RESUMEN

Diabetes mellitus (DM) is a growing health concern in society. Type 1 and type 2 DM are the two main types of diabetes; both types are chronic diseases that affect glucose metabolism in the body and the impaired regulation of glucose and lipid metabolism promotes the development and progression of DM. During the physiological metabolism process, the liver serves a unique role in glucose and lipid metabolism. The present article aimed to review the association between DM and glucose metabolism in the liver and discuss the changes of the following hepatic glucose fluxes: Gluconeogenesis, glucose/glucose 6­phosphate cycling, glycogenolysis, glycogenesis and the pentose phosphate pathway. Moreover, the incidence of fatty liver in DM was also investigated.


Asunto(s)
Diabetes Mellitus Tipo 1/complicaciones , Diabetes Mellitus Tipo 2/complicaciones , Glucosa/metabolismo , Metabolismo de los Lípidos , Hígado/metabolismo , Animales , Humanos , Insulina/metabolismo , Resistencia a la Insulina
15.
Sci Rep ; 10(1): 2609, 2020 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-32042093

RESUMEN

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

16.
Toxicol Appl Pharmacol ; 234(3): 293-9, 2009 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-19027772

RESUMEN

Hexavalent chromium Cr(VI) is a respiratory toxicant and carcinogen, with solubility playing an important role in its carcinogenic potential. Zinc chromate, a water insoluble or 'particulate' Cr(VI) compound, has been shown to be carcinogenic in epidemiology studies and to induce tumors in experimental animals, but its genotoxicity is poorly understood. Our study shows that zinc chromate induced concentration-dependent increases in cytotoxicity, chromosome damage and DNA double strand breaks in human lung cells. In response to zinc chromate-induced breaks, MRE11 expression was increased and ATM and ATR were phosphorylated, indicating that the DNA double strand break repair system was initiated in the cells. In addition, our data show that zinc chromate-induced double strand breaks were only observed in the G2/M phase population, with no significant amount of double strand breaks observed in G1 and S phase cells. These data will aid in understanding the mechanisms of zinc chromate toxicity and carcinogenesis.


Asunto(s)
Cromatos/toxicidad , Inestabilidad Cromosómica , Roturas del ADN de Doble Cadena , ADN/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Pulmón/efectos de los fármacos , Mutágenos/toxicidad , Compuestos de Zinc/toxicidad , Proteínas de la Ataxia Telangiectasia Mutada , Ciclo Celular/efectos de los fármacos , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Supervivencia Celular/efectos de los fármacos , ADN/metabolismo , Proteínas de Unión al ADN/metabolismo , Relación Dosis-Respuesta a Droga , Fibroblastos/metabolismo , Fibroblastos/patología , Humanos , Pulmón/metabolismo , Pulmón/patología , Proteína Homóloga de MRE11 , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Regulación hacia Arriba
17.
Chem Biol Interact ; 310: 108719, 2019 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-31238026

RESUMEN

Both obesity and arsenic exposure are global public health problems that are associated with increased risk of renal disease. The effect of whole-life exposure to environmentally relevant levels of arsenic within dietary high fat diet on renal pathogenesis were examined. In this study, C57BL/6 J mice were parentally exposed to 100 ppb arsenic before conception. After weaning, both male and female offspring were maintained on 100 ppb arsenic and fed either a normal (LFD) or high fat diet (HFD). At 10 and 24 weeks of age, the offspring were sacrificed and kidneys collected. Exposure to arsenic led to an increase body-weight in LFD diet-fed female but not male mice. This response was not observed in HFD-fed female mice; however male mice showed significant increases in body weight in both As- and non-treated animals. Histological analysis shows that arsenic exposure significantly increases HFD-induced glomerular area expansion, mesangial matrix accumulation and fibrosis compared to LFD control animals. HFD alone increases renal inflammation and fibrosis; reflected by increases in IL-1ß, ICAM-1 and fibronectin levels. Arsenic exposure significantly increases HFD-induced inflammatory and oxidative stress responses. In general, male mice have more severe responses than female mice to HFD or arsenic treatment. These results demonstrate that arsenic exposure causes sex-dependent alterations in HFD-induced kidney damage.


Asunto(s)
Arsénico/efectos adversos , Dieta Alta en Grasa/efectos adversos , Enfermedades Renales/etiología , Riñón/efectos de los fármacos , Animales , Arsénico/toxicidad , Peso Corporal/efectos de los fármacos , Inflamación/etiología , Riñón/lesiones , Enfermedades Renales/inducido químicamente , Enfermedades Renales/patología , Ratones , Ratones Endogámicos C57BL , Estrés Oxidativo/efectos de los fármacos , Factores Sexuales
18.
Sci Rep ; 9(1): 14675, 2019 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-31604971

RESUMEN

Exposure to the environmental toxicant cadmium (Cd) contributes to the development of obesity-associated diseases. Obesity is a risk factor for a spectrum of unhealthy conditions including systemic metabolic dyshomeostasis. In the present study, the effects of whole-life exposure to environmentally-relevant concentrations of Cd on systemic essential metal distribution in adult mice fed a high-fat diet (HFD) were examined. For these studies, male and female mice were exposed to Cd-containing drinking water for >2 weeks before breeding. Pregnant mice and dams with offspring were exposed to Cd-containing drinking water. After weaning, offspring were continuously exposed to the same Cd concentration as their parents, and divided into HFD and normal (low) fat diet (LFD) groups. At 10 and 24 weeks, mice were sacrificed and blood, liver, kidney and heart harvested for metal analyses. There were significant concentration dependent increases in Cd levels in offspring with kidney > liver > heart. Sex significantly affected Cd levels in kidney and liver, with female animals accumulating more metal than males. Mice fed the HFD showed > 2-fold increase in Cd levels in the three organs compared to similarly treated LFD mice. Cadmium significantly affected essential metals levels in blood, kidney and liver. Additionally, HFD affected essential metal levels in these three organs. These findings suggest that Cd interacts with HFD to affect essential metal homeostasis, a phenomenon that may contribute to the underlying mechanism responsible for the development of obesity-associated pathologies.


Asunto(s)
Riñón/química , Hígado/química , Metales/química , Miocardio/química , Obesidad/metabolismo , Animales , Cadmio/farmacología , Cadmio/toxicidad , Dieta con Restricción de Grasas , Dieta Alta en Grasa/efectos adversos , Femenino , Corazón/efectos de los fármacos , Riñón/efectos de los fármacos , Hígado/efectos de los fármacos , Masculino , Metales/aislamiento & purificación , Metales/metabolismo , Ratones , Miocardio/metabolismo , Obesidad/complicaciones , Obesidad/patología , Embarazo , Factores de Riesgo
19.
Syst Biol Reprod Med ; 64(6): 469-483, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29873257

RESUMEN

Exposure to environmental stressors during susceptible windows of development can result in negative health outcomes later in life, a concept known as the Developmental Origins of Health and Disease (DOHaD). There is a growing body of evidence that exposures to metals early in life (in utero and postnatal) increase the risk of developing adult diseases such as cancer, cardiovascular disease, non-alcoholic fatty liver disease, and diabetes. Of particular concern is exposure to the metalloid arsenic, a drinking water contaminant and worldwide health concern. Epidemiological studies of areas with high levels of arsenic in the drinking water, such as some regions in Chile and Bangladesh, indicate an association between in utero arsenic exposure and the development of adult diseases. Therefore, the need for experimental models to address the mechanism underlining early onset of adult diseases have emerged including the in utero and whole-life exposure models. This review will highlight the epidemiological events and subsequent novel experimental models implemented to study the impact of early life exposure to arsenic on the development of adult diseases. In addition, current research using these models will be discussed as well as possible underlying mechanism for the early onset of disease. Abbreviations: ALT: alanine aminotransferase; AMI: acute myocardial infarction; AST: aspartate aminotransferase; ATSDR: Agency for Toxic Substances and Disease Registry; CVD: cardiovascular disease; DMA: dimethylarsinate; DOHaD: Developmental Origins of Health and Disease; EPA: U.S. Environmental Protection Agency; ER-α: estrogen receptor alpha; HDL: high-density lipoprotein; HOMA-IR: homeostatic model assessment of insulin resistance; iAs: inorganic arsenic; LDL: low-density lipoprotein; MetS: metabolic syndrome; MMA: monomethylarsonate; NAFLD: non-alcoholic fatty liver disease; PND: postnatal day; ppb: parts per billion; ppm: parts per million; SAM: S-adenosylmethionine; USFDA: United States Food and Drug Administration.


Asunto(s)
Arsénico/toxicidad , Enfermedad Crónica , Enfermedad/etiología , Efectos Tardíos de la Exposición Prenatal , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Embarazo
20.
Aquat Toxicol ; 198: 149-157, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29547730

RESUMEN

Hexavalent chromium [Cr(VI)] is a marine pollution of concern as recent studies show it has a global distribution, with some regions showing high Cr concentrations in marine animal tissue, and it is extensively used. Leatherback sea turtles (Dermochelys coriacea) are an endangered marine species that may experience prolonged exposures to environmental contaminants including Cr(VI). Human activities have led to global Cr(VI) contamination of the marine environment. While Cr(VI) has been identified as a known human carcinogen, the health effects in marine species are poorly understood. In this study, we assessed the cytotoxic and genotoxic effects of particulate and soluble Cr(VI) in leatherback sea turtle lung cells. Both particulate and soluble Cr(VI) induced a concentration-dependent increase in cytotoxicity. Next, using a chromosome aberration assay, we assessed the genotoxic effects of Cr(VI) in leatherback sea turtle lung cells. Particulate and soluble Cr(VI) induced a concentration-dependent increase in clastogenicity in leatherback sea turtle lung cells. These data indicate that Cr(VI) may be a health concern for leatherback sea turtles and other long-lived marine species. Additionally, these data provide foundational support to use leatherback sea turtles as a valuable model species for monitoring the health effects of Cr(VI) in the environment and possibly as an indicator species to assess environmental human exposures and effects.


Asunto(s)
Cromo/toxicidad , Pulmón/patología , Pruebas de Mutagenicidad , Mutágenos/toxicidad , Material Particulado/toxicidad , Tortugas/metabolismo , Animales , Recuento de Células , Muerte Celular/efectos de los fármacos , Línea Celular , Cromatos/toxicidad , Cromo/análisis , Aberraciones Cromosómicas , Daño del ADN/efectos de los fármacos , Iones , Solubilidad , Contaminantes Químicos del Agua/toxicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA