Phthalate exposure associated with self-reported diabetes among Mexican women.

BACKGROUND Phthalates are ubiquitous industrial chemicals used as plasticizers in plastics made of polyvinyl chloride (PVC) to confer flexibility and durability. They are also present in products used for personal-care, industry and in medical devices. Phthalates have been associated with several adverse health effects, and recently it has been proposed that exposure to phthalates, could have an effect on metabolic homeostasis. This exploratory cross-sectional study evaluated the possible association between phthalate exposure and self-reported diabetes among adult Mexican women. METHODS As part of an on-going case-control study for breast cancer, only controls were selected, which constituted 221 healthy women matched by age (±5 years) and place of residence with the cases. Women with diabetes were identified by self-report. Urinary concentrations of nine phthalate metabolites were measured by online solid phase extraction coupled to high performance liquid chromatography-isotope-dilution tandem mass spectrometry. RESULTS Participants with diabetes had significantly higher concentrations of di(2-ethylhexyl) pththalate (DEHP) metabolites: mono(2-ethyl-5-hydroxyhexyl) phthalate (MEHHP), mono(2-ethyl-5-oxohexyl) phthalate (MEOHP) and mono(2-ethyl-5-carboxypentyl) phthalate (MECPP) but lower levels of monobenzyl phthalate (MBzP) a metabolite of benzylbutyl phthalate, compared to participants without diabetes. A marginally significant positive associations with diabetes status were observed over tertiles with MEHHP (OR(T3 vs. T1)=2.66; 95% CI: 0.97-7.33; p for trend=0.063) and MEOHP (OR(T3 vs. T1)=2.27; 95% CI; 0.90-5.75; P for trend=0.079) even after adjusting for important confounders. CONCLUSIONS The results suggest that levels of some phthalates may play a role in the genesis of diabetes.

[1]  Antonia M. Calafat,et al.  Exposure to Phthalates and Breast Cancer Risk in Northern Mexico , 2009, Environmental health perspectives.

[2]  D. Waxman,et al.  Activation of PPARalpha and PPARgamma by environmental phthalate monoesters. , 2003, Toxicological sciences : an official journal of the Society of Toxicology.

[3]  A. Calafat,et al.  Altered Semen Quality in Relation to Urinary Concentrations of Phthalate Monoester and Oxidative Metabolites , 2006, Epidemiology.

[4]  Toxicological Profile for Di ( 2-Ethylhexyl ) Phthalate ( DEHP ) Draft for Public Comment December 2019 , 2009 .

[5]  D. Waxman,et al.  Activation of PPARα and PPARγ by Environmental Phthalate Monoesters , 2003 .

[6]  P. Aschner Metabolic syndrome as a risk factor for diabetes , 2010, Expert review of cardiovascular therapy.

[7]  Richard Donnelly,et al.  Diagnosis and Classification of Diabetes , 2010 .

[8]  C. R. Dhanya,et al.  Changes in some hormones by low doses of di (2-ethyl hexyl) phthalate (DEHP), a commonly used plasticizer in PVC blood storage bags & medical tubing. , 2004, The Indian journal of medical research.

[9]  Béatrice Desvergne,et al.  PPAR-mediated activity of phthalates: A link to the obesity epidemic? , 2009, Molecular and Cellular Endocrinology.

[10]  A. Calafat,et al.  Temporal variability in urinary concentrations of phthalate metabolites, phytoestrogens and phenols among minority children in the United States. , 2008, Environmental research.

[11]  J. V. Vanden Heuvel,et al.  Activation of mouse and human peroxisome proliferator-activated receptors (PPARs) by phthalate monoesters. , 2004, Toxicological sciences : an official journal of the Society of Toxicology.

[12]  Bernhard Liebl,et al.  Occurrence and daily variation of phthalate metabolites in the urine of an adult population. , 2007, International journal of hygiene and environmental health.

[13]  J. Sepúlveda,et al.  [Methodology of the National Seroepidemiologic Survey, Mexico]. , 1992, Salud publica de Mexico.

[14]  D. Baird,et al.  Reproducibility of urinary phthalate metabolites in first morning urine samples. , 2002, Environmental health perspectives.

[15]  A. Calafat,et al.  Intra- and inter-individual variability of urinary phthalate metabolite concentrations in Hmong women of reproductive age , 2010, Journal of Exposure Science and Environmental Epidemiology.

[16]  Ken Takahashi,et al.  Decreased Serum Free Testosterone in Workers Exposed to High Levels of Di-n-butyl Phthalate (DBP) and Di-2-ethylhexyl Phthalate (DEHP): A Cross-Sectional Study in China , 2006, Environmental health perspectives.

[17]  Jorma Toppari,et al.  Human Breast Milk Contamination with Phthalates and Alterations of Endogenous Reproductive Hormones in Infants Three Months of Age , 2005, Environmental health perspectives.

[18]  A. Calafat,et al.  Determination of 16 phthalate metabolites in urine using automated sample preparation and on-line preconcentration/high-performance liquid chromatography/tandem mass spectrometry. , 2005, Analytical chemistry.

[19]  G. Gutiérrez,et al.  Metodología de la encuesta nacional seroepidemiológica, México , 1992 .

[20]  G. Rossi,et al.  Diagnosis and Classification of Diabetes Mellitus The information that follows is based largely on the reports of the Expert Committee on the Diagnosis and Classification of Diabetes (Diabetes Care 20:1183–1197, 1997, and Diabetes Care 26:3160–3167, 2003). , 2008, Diabetes Care.

[21]  Antonia M. Calafat,et al.  Temporal Variability of Urinary Phthalate Metabolite Levels in Men of Reproductive Age , 2004, Environmental health perspectives.

[22]  A. Calafat,et al.  PHTHALATES AND HUMAN HEALTH , 2005, Occupational and Environmental Medicine.

[23]  Aurélien Grosdidier,et al.  The Endocrine Disruptor Monoethyl-hexyl-phthalate Is a Selective Peroxisome Proliferator-activated Receptor γ Modulator That Promotes Adipogenesis* , 2007, Journal of Biological Chemistry.

[24]  Shanna H Swan,et al.  Environmental phthalate exposure in relation to reproductive outcomes and other health endpoints in humans. , 2008, Environmental research.

[25]  L. Aerts,et al.  Lifetime consequences of abnormal fetal pancreatic development , 2003, The Journal of physiology.

[26]  M. Dalgaard,et al.  Impact of diisobutyl phthalate and other PPAR agonists on steroidogenesis and plasma insulin and leptin levels in fetal rats. , 2008, Toxicology.

[27]  Elizabeth E Hatch,et al.  Association of urinary phthalate metabolite concentrations with body mass index and waist circumference: a cross-sectional study of NHANES data, 1999–2002 , 2008, Environmental health : a global access science source.

[28]  F. Grün,et al.  Perturbed nuclear receptor signaling by environmental obesogens as emerging factors in the obesity crisis , 2007, Reviews in Endocrine and Metabolic Disorders.

[29]  C. Casals-Casas,et al.  Interference of pollutants with PPARs: endocrine disruption meets metabolism , 2008, International Journal of Obesity.

[30]  S. Swan,et al.  Concentrations of Urinary Phthalate Metabolites Are Associated with Increased Waist Circumference and Insulin Resistance in Adult U.S. Males , 2007, Environmental health perspectives.

[31]  J. Auwerx PPARγ, the ultimate thrifty gene , 1999, Diabetologia.

[32]  K. Balasubramanian,et al.  Diethylhexyl phthalate impairs insulin binding and glucose oxidation in Chang liver cells. , 2007, Toxicology in vitro : an international journal published in association with BIBRA.

[33]  Malcolm Williams,et al.  Toxicological profile for diethyl phthalate , 1995 .

[34]  Johan Auwerx,et al.  The Pollutant Diethylhexyl Phthalate Regulates Hepatic Energy Metabolism via Species-Specific PPARα-Dependent Mechanisms , 2009, Environmental health perspectives.

[35]  Frederica P. Perera,et al.  Characterization of Phthalate Exposure among Pregnant Women Assessed by Repeat Air and Urine Samples , 2008, Environmental health perspectives.

[36]  Samuel P. Caudill,et al.  Urinary Creatinine Concentrations in the U.S. Population: Implications for Urinary Biologic Monitoring Measurements , 2004, Environmental health perspectives.

[37]  K. Channer,et al.  Testosterone replacement therapy improves insulin resistance, glycaemic control, visceral adiposity and hypercholesterolaemia in hypogonadal men with type 2 diabetes. , 2006, European journal of endocrinology.