1
|
Lummertz Magenis M, Souza de Marcos P, Paganini Damiani A, Ricardo Cantareli da Silva A, Martins Longaretti L, Bahia Franca I, Da Silva J, Rodrigues Boeck C, Moraes de Andrade V. Genotoxic effects of caffeine in female mice exposed during pregnancy and lactation period and their offspring. JOURNAL OF ENVIRONMENTAL SCIENCE AND HEALTH. PART C, TOXICOLOGY AND CARCINOGENESIS 2023; 41:36-60. [PMID: 37243358 DOI: 10.1080/26896583.2023.2213613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Caffeine is a widely consumed substance, and there is a discussion about its effects when ingested by women during pregnancy and lactation. We aimed to identify the genotoxic effects of caffeine in female mice that consumed it during pregnancy and lactation periods and its consequences in their offspring. Thirty-six couples of Swiss mice received water or caffeine (0.3 and 1.0 mg/mL) treatment during pregnancy and lactation. The male and female offspring were divided into 12 groups according to the treatment administered to the female mice. Genotoxicity was assessed using the comet assay and the micronucleus test. Both doses of caffeine showed genotoxic effects in pregnant and lactating mice groups compared to groups not administered caffeine. In relation to offspring, it can be observed that females and males of the offspring had low weight in early life. In both female and male offspring, genotoxicity was detected in the blood, liver, and kidney tissues. Thus, from the present study, we can suggest that the caffeine consumed by female mice during the periods of pregnancy and lactation led to genotoxic effects in their offspring.
Collapse
Affiliation(s)
- Marina Lummertz Magenis
- Laboratory of Translational Biomedicine, Graduate Program of Health Sciences, University of Southern Santa Catarina, Criciúma, Santa Catarina, Brazil
| | - Pamela Souza de Marcos
- Laboratory of Translational Biomedicine, Graduate Program of Health Sciences, University of Southern Santa Catarina, Criciúma, Santa Catarina, Brazil
| | - Adriani Paganini Damiani
- Laboratory of Translational Biomedicine, Graduate Program of Health Sciences, University of Southern Santa Catarina, Criciúma, Santa Catarina, Brazil
| | - Anderson Ricardo Cantareli da Silva
- Laboratory of Translational Biomedicine, Graduate Program of Health Sciences, University of Southern Santa Catarina, Criciúma, Santa Catarina, Brazil
| | - Luiza Martins Longaretti
- Laboratory of Translational Biomedicine, Graduate Program of Health Sciences, University of Southern Santa Catarina, Criciúma, Santa Catarina, Brazil
| | - Ive Bahia Franca
- Laboratory of Translational Biomedicine, Graduate Program of Health Sciences, University of Southern Santa Catarina, Criciúma, Santa Catarina, Brazil
| | - Juliana Da Silva
- Laboratory of Genetic Toxicology, Lutheran University of Brazil, Canoas, Rio Grande do Sul, Brazil
- La Salle University, Canoas, Rio Grande do Sul, Brazil
| | - Carina Rodrigues Boeck
- Graduate Program in Nanosciences, Master Degree in Health and Life Science, Franciscan University, Santa Maria, Rio Grande do Sul, Brazil
| | - Vanessa Moraes de Andrade
- Laboratory of Translational Biomedicine, Graduate Program of Health Sciences, University of Southern Santa Catarina, Criciúma, Santa Catarina, Brazil
| |
Collapse
|
2
|
Magenis ML, Damiani AP, Franca IB, de Marcos PS, Effting PS, Muller AP, de Bem Silveira G, Borges Correa MEA, Medeiros EB, Silveira PCL, Budni J, Boeck CR, de Andrade VM. Behavioral, genetic and biochemical changes in the brain of the offspring of female mice treated with caffeine during pregnancy and lactation. Reprod Toxicol 2022; 112:119-135. [PMID: 35868513 DOI: 10.1016/j.reprotox.2022.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 07/07/2022] [Accepted: 07/16/2022] [Indexed: 11/17/2022]
Abstract
The intrauterine environment is a critical location for exposure to exogenous and endogenous factors that trigger metabolic changes through fetal programming. Among the external factors, chemical compounds stand out, which include caffeine, since its consumption is common among women, including during pregnancy. Thereby, the aim of the present study was to evaluate the behavioral, genetic, and biochemical parameters in the offspring of female mice treated with caffeine during pregnancy and lactation. Swiss female mice (60 days old) received tap water or caffeine at 0.3 or 1.0 mg/mL during copulation (7 days), pregnancy (21 days) and lactation (21 days). After the end of the lactation period, the offspring were divided into groups (water, caffeine 0.3 or 1.0 mg/mL) with 20 animals (10 animals aged 30 days and 10 animals aged 60 days per group per sex). Initially, the offspring were submitted to behavioral tasks and then euthanized for genetic and biochemical analysis in the brain (cortex, striatum, and hippocampus). Behavioral changes in memory, depression, and anxiety were observed in the offspring: 30-day-old female offspring at 1.0 mg /mL dose presented anxiogenic behavior and male offspring the 0.3 mg/mL dose at 30 days of age did not alter long-term memory. Furthermore, an increase in DNA damage and oxidative stress in the brain were observed in the offspring of both sexes. Furthermore, there were changes in Ape-1, BAX, and Bcl-2 in the female offspring hippocampus at 30 days of life. Thus, with this study, we can suggest genotoxicity, oxidative stress, and behavioral changes caused by caffeine during pregnancy and lactation in the offspring that were not treated directly, but received through their mothers; thus, it is important to raise awareness regarding caffeine consumption among pregnant and lactating females.
Collapse
Affiliation(s)
- Marina Lummertz Magenis
- Laboratory of Translational Biomedicine, Graduate Program of Health Sciences, University of Southern Santa Catarina - UNESC, Criciúma, SC, Brazil
| | - Adriani Paganini Damiani
- Laboratory of Translational Biomedicine, Graduate Program of Health Sciences, University of Southern Santa Catarina - UNESC, Criciúma, SC, Brazil
| | - Ive Bahia Franca
- Laboratory of Translational Biomedicine, Graduate Program of Health Sciences, University of Southern Santa Catarina - UNESC, Criciúma, SC, Brazil
| | - Pamela Souza de Marcos
- Laboratory of Translational Biomedicine, Graduate Program of Health Sciences, University of Southern Santa Catarina - UNESC, Criciúma, SC, Brazil
| | - Pauline Souza Effting
- Laboratory of Translational Biomedicine, Graduate Program of Health Sciences, University of Southern Santa Catarina - UNESC, Criciúma, SC, Brazil
| | - Alexandre Pastoris Muller
- Laboratory of Translational Biomedicine, Graduate Program of Health Sciences, University of Southern Santa Catarina - UNESC, Criciúma, SC, Brazil
| | - Gustavo de Bem Silveira
- Laboratory of Experimental Pathophysiology, Graduate Program of Health Sciences, University of Southern Santa Catarina - UNESC, Criciúma, SC, Brazil
| | - Maria Eduarda Anastácio Borges Correa
- Laboratory of Experimental Pathophysiology, Graduate Program of Health Sciences, University of Southern Santa Catarina - UNESC, Criciúma, SC, Brazil
| | - Eduarda Behenck Medeiros
- Laboratory of Experimental Neurology, Graduate Program of Health Sciences, University of Southern Santa Catarina - UNESC, Criciúma, SC, Brazil
| | - Paulo Cesar Lock Silveira
- Laboratory of Experimental Pathophysiology, Graduate Program of Health Sciences, University of Southern Santa Catarina - UNESC, Criciúma, SC, Brazil
| | - Josiane Budni
- Laboratory of Experimental Neurology, Graduate Program of Health Sciences, University of Southern Santa Catarina - UNESC, Criciúma, SC, Brazil
| | - Carina Rodrigues Boeck
- Graduate Program in Nanosciences, Franciscan University Center - UNIFRA, Santa Maria, RS, Brazil
| | - Vanessa Moraes de Andrade
- Laboratory of Translational Biomedicine, Graduate Program of Health Sciences, University of Southern Santa Catarina - UNESC, Criciúma, SC, Brazil.
| |
Collapse
|
3
|
Ramos AC, de Mattos Hungria F, Camerini BA, Suiama MA, Calzavara MB. Potential beneficial effects of caffeine administration in the neonatal period of an animal model of schizophrenia. Behav Brain Res 2020; 391:112674. [PMID: 32417274 DOI: 10.1016/j.bbr.2020.112674] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 04/03/2020] [Accepted: 04/24/2020] [Indexed: 12/17/2022]
Abstract
Obstetric complications, like maternal hypertension and neonatal hypoxia, disrupt brain development, leading to psychiatry disorders later in life, like schizophrenia. The exact mechanisms behind this risk are not yet well known. Spontaneously hypertensive rats (SHR) are a well-established model to study neurodevelopment of schizophrenia since they exhibit behavioral alterations mimicking schizophrenia that can be improved with antipsychotic drugs. SHR mothers are hypertensive, and the SHR offspring develop in preeclampsia-like conditions. Hypoxic conditions increase levels of adenosine, which play an important role in brain development. The enhanced levels of adenosine at birth could be related to the future development of schizophrenia. To investigate this hypothesis adenosine levels of brain neonatal Wistar rats and SHR were quantified. After that, caffeine, an antagonist of adenosinergic system, was administrated on PND (postnatal day) 7 (neurodevelopmental age similar to a human at delivery) and rats were observed at adolescent and adult ages. We also investigated the acute effects of caffeine at adolescent and adult ages. SHR control adolescent and adult groups presented behavioral deficits like hyperlocomotion, deficit in social interaction (SI), and contextual fear conditioning (CFC). In SHR, neonatal caffeine treatment on PND 7 normalized hyperlocomotion, improved SI, and CFC observed at adolescent period and adult ages, showing a beneficial effect on schizophrenia-like behaviors. Wistar rats neonatally treated with caffeine exhibited hyperlocomotion, deficit in SI and CFC when observed at adolescent and adult ages. Acutely caffeine treatment administrated at adolescent and adult ages increased locomotion and decreased SI time of Wistar rats and impair CFC in adult Wistars. No effects were observed in SHR. In conclusion, caffeine can be suggested as a useful drug to prevent behavioral deficits observed in this animal model of prenatal hypoxia-induced schizophrenia profile when specifically administered on PND 7.
Collapse
Affiliation(s)
- Aline Camargo Ramos
- Department of Psychiatry, Universidade Federal De São Paulo, São Paulo, SP, Brazil
| | | | | | - Mayra Akimi Suiama
- Department of Pharmacology, Universidade Federal De São Paulo, São Paulo, SP, Brazil
| | - Mariana Bendlin Calzavara
- Department of Psychiatry, Universidade Federal De São Paulo, São Paulo, SP, Brazil; School of Medicine from Faculdade Israelita De Ciências Da Saúde Albert Einstein, São Paulo, SP, Brazil.
| |
Collapse
|
4
|
Prenatal caffeine exposure induces down-regulation of the protein kinase A/ryanodine receptor/large-conductance Ca2+-activated K+ pathway in the cerebral arteries of old offspring rats. J Hypertens 2020; 38:679-691. [DOI: 10.1097/hjh.0000000000002303] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
5
|
Qian J, Chen Q, Ward SM, Duan E, Zhang Y. Impacts of Caffeine during Pregnancy. Trends Endocrinol Metab 2020; 31:218-227. [PMID: 31818639 PMCID: PMC7035149 DOI: 10.1016/j.tem.2019.11.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 11/10/2019] [Accepted: 11/12/2019] [Indexed: 12/19/2022]
Abstract
Epidemiological studies have revealed that caffeine consumption during pregnancy is associated with adverse gestational outcomes, yet the underlying mechanisms remain obscure. Recent animal studies with physiologically relevant dosages have begun to dissect adverse effects of caffeine during pregnancy with respect to oviduct contractility, embryo development, uterine receptivity, and placentation that jointly contribute to pregnancy complications. Interestingly, caffeine's effects are highly variable between individual animals under well-controlled experimental settings, suggesting the possibility of epigenetic regulation of these phenotypes, in addition to genetic variants. Moreover, caffeine exposure during sensitive windows of pregnancy may induce epigenetic changes in the developing fetus or even the germ cells to cause adult-onset diseases in subsequent generations. We discuss these research frontiers in light of emerging data.
Collapse
Affiliation(s)
- Jingjing Qian
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Qi Chen
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521, USA
| | - Sean M Ward
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, NV 89557, USA
| | - Enkui Duan
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China.
| | - Ying Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA.
| |
Collapse
|
6
|
Qian J, Zhang Y, Qu Y, Zhang L, Shi J, Zhang X, Liu S, Kim BH, Hwang SJ, Zhou T, Chen Q, Ward SM, Duan E, Zhang Y. Caffeine consumption during early pregnancy impairs oviductal embryo transport, embryonic development and uterine receptivity in mice. Biol Reprod 2019; 99:1266-1275. [PMID: 29982366 DOI: 10.1093/biolre/ioy155] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 07/03/2018] [Indexed: 12/14/2022] Open
Abstract
Caffeine consumption has been widely used as a central nervous system stimulant. Epidemiological studies, however, have suggested that maternal caffeine exposure during pregnancy is associated with increased abnormalities, including decreased fertility, delayed conception, early spontaneous abortions, and low birth weight. The mechanisms underlying the negative outcomes of caffeine consumption, particularly during early pregnancy, remain unclear. In present study, we found that pregnant mice treated with moderate (5 mg/kg) or high (30 mg/kg) dosage of caffeine (intraperitoneally or orally) during preimplantation resulted in retention of early embryos in the oviduct, defective embryonic development, and impaired embryo implantation. Transferring normal blastocysts into the uteri of caffeine-treated pseudopregnant females also showed abnormal embryo implantation, thus indicating impaired uterine receptivity by caffeine administration. The remaining embryos that managed to implant after caffeine treatment also showed increased embryo resorption rate and abnormal development at mid-term stage, and decreased weight at birth. In addition to a dose-dependent effect, significant variations between individual mice under the same caffeine dosage were also observed, suggesting different sensitivities to caffeine, similar to that observed in human populations. Collectively, our data revealed that caffeine exposure during early pregnancy impaired oviductal embryo transport, embryonic development, and uterine receptivity, which are responsible for abnormal implantation and pregnancy loss. The study raises the concern of caffeine consumption during early stages of pregnancy.
Collapse
Affiliation(s)
- Jingjing Qian
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yunfang Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, Nevada, USA.,University of Chinese Academy of Sciences, Beijing, China
| | - Yongcun Qu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Liwen Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Junchao Shi
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, Nevada, USA
| | - Xudong Zhang
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, Nevada, USA
| | - Shichao Liu
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, Nevada, USA
| | - Bo Hyun Kim
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, Nevada, USA
| | - Sung Jin Hwang
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, Nevada, USA
| | - Tong Zhou
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, Nevada, USA
| | - Qi Chen
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, Nevada, USA
| | - Sean M Ward
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, Nevada, USA
| | - Enkui Duan
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Ying Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
7
|
Abu-Sa'da OS, Armstrong EA, Scott O, Shaw O, Nguyen AT, Shen K, Cheung PY, Baker G, Yager JY. The Effect of Caffeine on the Neuropathological and Neurobehavioral Outcome in the Newborn Rat. J Caffeine Adenosine Res 2018. [DOI: 10.1089/caff.2017.0036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Omar S. Abu-Sa'da
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
- Department of Neonatology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Edward A. Armstrong
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
- Faculty of Medicine and Dentistry, Centre for Neuroscience, University of Alberta, Edmonton, Canada
| | - Ori Scott
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Oriana Shaw
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Antoinette T. Nguyen
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
- Faculty of Medicine and Dentistry, Centre for Neuroscience, University of Alberta, Edmonton, Canada
| | - Keqin Shen
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
- Department of Pediatric Neurosciences, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Po-Yin Cheung
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
- Department of Neonatology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Glen Baker
- Department of Psychiatry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Jerome Y. Yager
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
- Faculty of Medicine and Dentistry, Centre for Neuroscience, University of Alberta, Edmonton, Canada
- Department of Pediatric Neurosciences, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
- Department of Psychiatry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| |
Collapse
|
8
|
Transient Disruption of Adenosine Signaling During Embryogenesis Triggers a Pro-epileptic Phenotype in Adult Zebrafish. Mol Neurobiol 2018; 55:6547-6557. [PMID: 29327202 DOI: 10.1007/s12035-017-0850-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 12/19/2017] [Indexed: 10/18/2022]
Abstract
Adenosinergic signaling has important effects on brain function, anatomy, and physiology in both late and early stages of development. Exposure to caffeine, a non-specific blocker of adenosine receptor, has been indicated as a developmental risk factor. Disruption of adenosinergic signaling during early stages of development can change the normal neural network formation and possibly lead to an increase in susceptibility to seizures. In this work, morpholinos (MO) temporarily blocked the translation of adenosine receptor transcripts, adora1, adora2aa, and adora2ab, during the embryonic phase of zebrafish. It was observed that the block of adora2aa and adora2aa + adora2ab transcripts increased the mortality rate and caused high rate of malformations. To test the susceptibility of MO adora1, MO adora2aa, MO adora2ab, and MO adora2aa + adora2ab animals to seizure, pentylenetetrazole (10 mM) was used as a convulsant agent in larval and adult stages of zebrafish development. Although no MO promoted significant differences in latency time to reach the seizures stages in 7-day-old larvae, during the adult stage, all MO animals showed a decrease in the latency time to reach stages III, IV, and V of seizure. These results indicated that transient interventions in the adenosinergic signaling through high affinity adenosine receptors during embryonic development promote strong outcomes on survival and morphology. Additionally, long-term effects on neural development can lead to permanent impairment on neural signaling resulting in increased susceptibility to seizure.
Collapse
|
9
|
Long-term consequences of disrupting adenosine signaling during embryonic development. Mol Aspects Med 2017; 55:110-117. [PMID: 28202385 DOI: 10.1016/j.mam.2017.02.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 01/27/2017] [Accepted: 02/03/2017] [Indexed: 12/16/2022]
Abstract
There is growing evidence that disruption in the prenatal environment can have long-lasting effects on an individual's health in adulthood. Research on the fetal programming of adult diseases, including cardiovascular disease, focuses on epi-mutations, which alter the normal pattern of epigenetic factors such as DNA methylation, miRNA expression, or chromatin modification, rather than traditional genetic alteration. Thus, understanding how in utero chemical exposures alter epigenetics and lead to adult disease is of considerable public health concern. Few signaling molecules have the potential to influence the developing mammal as the nucleoside adenosine. Adenosine levels increase rapidly with tissue hypoxia and inflammation. Adenosine antagonists including the methlyxanthines caffeine and theophylline are widely consumed during pregnancy. The receptors that transduce adenosine action are the A1, A2a, A2b, and A3 adenosine receptors (ARs). We examined the long-term effects of in utero disruption of adenosine signaling on cardiac gene expression, morphology, and function in adult offspring. One substance that fetuses are frequently exposed to is caffeine, which is a non-selective adenosine receptor antagonist. Over the past several years, we examined the role of adenosine signaling during embryogenesis and cardiac development. We discovered that in utero alteration in adenosine action leads to adverse effects on embryonic and adult murine hearts. We find that cardiac A1ARs protect the embryo from in utero hypoxic stress, a condition that causes an increase in adenosine levels. After birth in mice, we observed that in utero caffeine exposure leads to abnormal cardiac function and morphology in adults, including an impaired response to β-adrenergic stimulation. Recently, we observed that in utero caffeine exposure induces transgenerational effects on cardiac morphology, function, and gene expression. Our findings indicate that the effects of altered adenosine signaling are dependent on signaling through the A1ARs and timing of disruption. In addition, the long-term effects of altered adenosine signaling appear to be mediated by alterations in DNA methylation, an epigenetic process critical for normal development.
Collapse
|
10
|
Fang X, Poulsen RR, Rivkees SA, Wendler CC. In Utero Caffeine Exposure Induces Transgenerational Effects on the Adult Heart. Sci Rep 2016; 6:34106. [PMID: 27677355 PMCID: PMC5039698 DOI: 10.1038/srep34106] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 09/01/2016] [Indexed: 12/28/2022] Open
Abstract
Each year millions of pregnant woman are exposed to caffeine, which acts to antagonize adenosine action. The long-term consequences of this exposure on the developing fetus are largely unknown, although in animal models we have found adverse effects on cardiac function. To assess if these effects are transmitted transgenerationally, we exposed pregnant mice to caffeine equivalent to 2–4 cups of coffee at two embryonic stages. Embryos (F1 generation) exposed to caffeine early from embryonic (E) day 6.5–9.5 developed a phenotype similar to dilated cardiomyopathy by 1 year of age. Embryos exposed to caffeine later (E10.5–13.5) were not affected. We next examined the F2 generation and F3 generation of mice exposed to caffeine from E10.5–13.5, as this coincides with germ cell development. These F2 generation adult mice developed a cardiac phenotype similar to hypertrophic cardiomyopathy. The F3 generation exhibited morphological changes in adult hearts, including increased mass. This report shows that in utero caffeine exposure has long-term effects into adulthood and that prenatal caffeine exposure can exert adverse transgenerational effects on adult cardiac function.
Collapse
Affiliation(s)
- Xiefan Fang
- Child Health Research Institute, Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Ryan R Poulsen
- Child Health Research Institute, Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Scott A Rivkees
- Child Health Research Institute, Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Christopher C Wendler
- Child Health Research Institute, Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
11
|
Sarmah S, Chism GW, Vaughan MA, Muralidharan P, Marrs JA, Marrs KA. Using Zebrafish to Implement a Course-Based Undergraduate Research Experience to Study Teratogenesis in Two Biology Laboratory Courses. Zebrafish 2016; 13:293-304. [PMID: 26829498 PMCID: PMC5911693 DOI: 10.1089/zeb.2015.1107] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
A course-based undergraduate research experience (CURE) spanning three semesters was introduced into freshman and sophomore biology classes, with the hypothesis that participation in a CURE affects skills in research, communication, and collaboration, which may help students persist in science. Student research projects were centered on the hypothesis that nicotine and caffeine exposure during early development affects gastrulation and heart development in zebrafish. First, freshmen generated original data showing distinct effects of embryonic nicotine and caffeine exposure on zebrafish heart development and function. Next, Cell Biology laboratory students continued the CURE studies and identified novel teratogenic effects of nicotine and caffeine during gastrulation. Finally, new freshmen continued the CURE research, examining additional toxicant effects on development. Students designed new protocols, made measurements, presented results, and generated high-quality preliminary data that were studied in successive semesters. By implementing this project, the CURE extended faculty research and provided a scalable model to address national goals to involve more undergraduates in authentic scientific research. In addition, student survey results support the hypothesis that CUREs provide significant gains in student ability to (1) design experiments, (2) analyze data, and (3) make scientific presentations, translating into high student satisfaction and enhanced learning.
Collapse
Affiliation(s)
- Swapnalee Sarmah
- Department of Biology, Indiana University-Purdue University Indianapolis , Indianapolis, Indiana
| | - Grady W Chism
- Department of Biology, Indiana University-Purdue University Indianapolis , Indianapolis, Indiana
| | - Martin A Vaughan
- Department of Biology, Indiana University-Purdue University Indianapolis , Indianapolis, Indiana
| | - Pooja Muralidharan
- Department of Biology, Indiana University-Purdue University Indianapolis , Indianapolis, Indiana
| | - Jim A Marrs
- Department of Biology, Indiana University-Purdue University Indianapolis , Indianapolis, Indiana
| | - Kathleen A Marrs
- Department of Biology, Indiana University-Purdue University Indianapolis , Indianapolis, Indiana
| |
Collapse
|
12
|
Ahir BK, Pratten MK. The impact of caffeine on connexin expression in the embryonic chick cardiomyocyte micromass culture system. J Appl Toxicol 2016; 36:903-13. [PMID: 26304238 DOI: 10.1002/jat.3219] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2014] [Revised: 07/03/2015] [Accepted: 07/06/2015] [Indexed: 12/19/2022]
Abstract
Cardiomyocytes are electrically coupled by gap junctions, defined as clusters of low-resistance multisubunit transmembrane channels composed of connexins (Cxs). The expression of Cx40, Cx43 and Cx45, which are present in cardiomyocytes, is known to be developmentally regulated. This study investigates the premise that alterations in gap junction proteins are one of the mechanisms by which teratogens may act. Specifically, those molecules known to be teratogenic in humans could cause their effects via disruption of cell-to-cell communication pathways, resulting in an inability to co-ordinate tissue development. Caffeine significantly inhibited contractile activity at concentrations above and including 1500 μm (P < 0.05), while not affecting cell viability and total protein, in the embryonic chick cardiomyocyte micromass culture system. The effects of caffeine on key cardiac gap junction protein (Cx40, Cx43 and Cx45) expression were analysed using immunocytochemistry and in-cell Western blotting. The results indicated that caffeine altered the expression pattern of Cx40, Cx43 and Cx45 at non-cytotoxic concentrations (≥2000 μm), i.e., at concentrations that did not affect total cell protein and cell viability. In addition the effects of caffeine on cardiomyocyte formation and function (contractile activity score) were correlated with modulation of Cxs (Cx40, Cx43 and Cx45) expression, at above and including 2000 μm caffeine concentrations (P < 0.05). These experiments provide evidence that embryonic chick cardiomyocyte micromass culture may be a useful in vitro method for mechanistic studies of perturbation of embryonic heart development. Copyright © 2015 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Bhavesh K Ahir
- National Center for Computational Toxicology (B205-01), US Environmental Protection Agency, Research Triangle Park, NC, USA
| | - Margaret K Pratten
- School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, UK
| |
Collapse
|
13
|
Li N, Li Y, Gao Q, Li D, Tang J, Sun M, Zhang P, Liu B, Mao C, Xu Z. Chronic fetal exposure to caffeine altered resistance vessel functions via RyRs-BKCa down-regulation in rat offspring. Sci Rep 2015; 5:13225. [PMID: 26277840 PMCID: PMC4642531 DOI: 10.1038/srep13225] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 07/21/2015] [Indexed: 01/10/2023] Open
Abstract
Caffeine modifies vascular/cardiac contractility. Embryonic exposure to caffeine altered cardiac functions in offspring. This study determined chronic influence of prenatal caffeine on vessel functions in offspring. Pregnant Sprague-Dawley rats (5-month-old) were exposed to high dose of caffeine, their offspring (5-month-old) were tested for vascular functions in mesenteric arteries (MA) and ion channel activities in smooth muscle cells. Prenatal exposure to caffeine increased pressor responses and vasoconstrictions to phenylephrine, accompanied by enhanced membrane depolarization. Large conductance Ca2+-activated K+ (BKCa) channels in buffering phenylephrine-induced vasoconstrictions was decreased, whole cell BKCa currents and spontaneous transient outward currents (STOCs) were decreased. Single channel recordings revealed reduced voltage/Ca2+ sensitivity of BKCa channels. BKCa α-subunit expression was unchanged, BKCa β1-subunit and sensitivity of BKCa to tamoxifen were reduced in the caffeine offspring as altered biophysical properties of BKCa in the MA. Simultaneous [Ca2+]i fluorescence and vasoconstriction testing showed reduced Ca2+, leading to diminished BKCa activation via ryanodine receptor Ca2+ release channels (RyRs), causing enhanced vascular tone. Reduced RyR1 was greater than that of RyR3. The results suggest that the altered STOCs activity in the caffeine offspring could attribute to down-regulation of RyRs-BKCa, providing new information for further understanding increased risks of hypertension in developmental origins.
Collapse
Affiliation(s)
- Na Li
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Yongmei Li
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Qinqin Gao
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Dawei Li
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Jiaqi Tang
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Miao Sun
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Pengjie Zhang
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Bailin Liu
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Caiping Mao
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Zhice Xu
- 1] Institute for Fetology, First Hospital of Soochow University, Suzhou, China [2] Center for Perinatal Biology, Loma Linda University, California, USA
| |
Collapse
|
14
|
Iglesias I, Albasanz JL, Martín M. Effect of Caffeine Chronically Consumed During Pregnancy on Adenosine A 1 and A 2A Receptors Signaling in Both Maternal and Fetal Heart from Wistar Rats. JOURNAL OF CAFFEINE RESEARCH 2014; 4:115-126. [PMID: 25538864 DOI: 10.1089/jcr.2014.0010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Background: Caffeine is the most widely consumed psychoactive substance in the world, even during pregnancy. Its stimulatory effects are mainly due to antagonism of adenosine actions by blocking adenosine A1 and A2A receptors. Previous studies have shown that caffeine can cross the placenta and therefore modulate these receptors not only in the fetal brain but also in the heart. Methods: In the present work, the effect of caffeine chronically consumed during pregnancy on A1 and A2A receptors in Wistar rat heart, from both mothers and their fetuses, were studied using radioligand binding, Western-blotting, and adenylyl cyclase activity assays, as well as reverse transcription polymerase chain reaction. Results: Caffeine did not significantly alter A1R neither at protein nor at gene expression level in both the maternal and fetal heart. On the contrary, A2AR significantly decreased in the maternal heart, although mRNA was not affected. Gi and Gs proteins were also preserved. Finally, A1R-mediated inhibition of adenylyl cyclase activity did not change in the maternal heart, but A2AR mediated stimulation of this enzymatic activity significantly decreased according to the detected loss of this receptor. Conclusions: Opposite to the downregulation and desensitization of the A1R/AC pathway previously reported in the brain, these results show that this pathway is not affected in rat heart after caffeine exposure during pregnancy. In addition, A2AR is downregulated and desensitized in the maternal heart, suggesting a differential modulation of these receptor-mediated pathways by caffeine.
Collapse
Affiliation(s)
- Inmaculada Iglesias
- Facultad de Ciencias y Tecnologías Químicas, Universidad de Castilla-La Mancha , Ciudad Real, Spain
| | - Jose Luis Albasanz
- Facultad de Ciencias y Tecnologías Químicas, Universidad de Castilla-La Mancha , Ciudad Real, Spain . ; Facultad de Medicina de Ciudad Real; Departamento de Química Inorgánica, Orgánica y Bioquímica, Universidad de Castilla-La Mancha , Ciudad Real, Spain
| | - Mairena Martín
- Facultad de Ciencias y Tecnologías Químicas, Universidad de Castilla-La Mancha , Ciudad Real, Spain . ; Facultad de Medicina de Ciudad Real; Departamento de Química Inorgánica, Orgánica y Bioquímica, Universidad de Castilla-La Mancha , Ciudad Real, Spain
| |
Collapse
|
15
|
Fang X, Mei W, Barbazuk WB, Rivkees SA, Wendler CC. Caffeine exposure alters cardiac gene expression in embryonic cardiomyocytes. Am J Physiol Regul Integr Comp Physiol 2014; 307:R1471-87. [PMID: 25354728 DOI: 10.1152/ajpregu.00307.2014] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Previous studies demonstrated that in utero caffeine treatment at embryonic day (E) 8.5 alters DNA methylation patterns, gene expression, and cardiac function in adult mice. To provide insight into the mechanisms, we examined cardiac gene and microRNA (miRNA) expression in cardiomyocytes shortly after exposure to physiologically relevant doses of caffeine. In HL-1 and primary embryonic cardiomyocytes, caffeine treatment for 48 h significantly altered the expression of cardiac structural genes (Myh6, Myh7, Myh7b, Tnni3), hormonal genes (Anp and BnP), cardiac transcription factors (Gata4, Mef2c, Mef2d, Nfatc1), and microRNAs (miRNAs; miR208a, miR208b, miR499). In addition, expressions of these genes were significantly altered in embryonic hearts exposed to in utero caffeine. For in utero experiments, pregnant CD-1 dams were treated with 20-60 mg/kg of caffeine, which resulted in maternal circulation levels of 37.3-65.3 μM 2 h after treatment. RNA sequencing was performed on embryonic ventricles treated with vehicle or 20 mg/kg of caffeine daily from E6.5-9.5. Differential expression (DE) analysis revealed that 124 genes and 849 transcripts were significantly altered, and differential exon usage (DEU) analysis identified 597 exons that were changed in response to prenatal caffeine exposure. Among the DE genes identified by RNA sequencing were several cardiac structural genes and genes that control DNA methylation and histone modification. Pathway analysis revealed that pathways related to cardiovascular development and diseases were significantly affected by caffeine. In addition, global cardiac DNA methylation was reduced in caffeine-treated cardiomyocytes. Collectively, these data demonstrate that caffeine exposure alters gene expression and DNA methylation in embryonic cardiomyocytes.
Collapse
Affiliation(s)
- Xiefan Fang
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, Florida; and
| | - Wenbin Mei
- Department of Biology, University of Florida, Gainesville, Florida
| | | | - Scott A Rivkees
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, Florida; and
| | - Christopher C Wendler
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, Florida; and
| |
Collapse
|
16
|
Dayanim S, Lopez B, Maisonet TM, Grewal S, Londhe VA. Caffeine induces alveolar apoptosis in the hyperoxia-exposed developing mouse lung. Pediatr Res 2014; 75:395-402. [PMID: 24321990 PMCID: PMC3943688 DOI: 10.1038/pr.2013.233] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2012] [Accepted: 05/29/2013] [Indexed: 12/20/2022]
Abstract
BACKGROUND Caffeine is a nonspecific adenosine receptor antagonist used in premature neonates to treat apnea of prematurity. While its use may reduce the incidence of bronchopulmonary dysplasia (BPD), the precise mechanisms remain unknown. Evidence of increased adenosine levels are noted in chronic lung diseases including tracheal aspirates of infants with BPD. Utilizing a well-characterized newborn mouse model of alveolar hypoplasia, we hypothesized that hyperoxia-induced alveolar inflammation and hypoplasia is associated with alterations in the adenosine signaling pathway. METHODS Newborn murine pups were exposed to a 14-d period of hyperoxia and daily caffeine administration followed by a 14-d recovery period in room air. Lungs were collected at both time points for bronchoalveolar lavage (BAL) analysis as well as histopathology and mRNA and protein expression. RESULTS Caffeine treatment increased inflammation and worsened alveolar hypoplasia in hyperoxia-exposed newborn mice. These changes were associated with decreased alveolar type II (ATII) cell numbers, increased cell apoptosis, and decreased expression of A2A receptors. Following discontinuation of caffeine and hyperoxia, lung histology returned to baseline levels comparable to hyperoxia exposure alone. CONCLUSION Results of this study suggest a potentially adverse role of caffeine on alveolar development in a murine model of hyperoxia-induced alveolar hypoplasia.
Collapse
Affiliation(s)
- Sara Dayanim
- Department of Pediatrics, Division of Neonatology and Developmental Biology, Neonatal Research Center, David Geffen School of Medicine at University of California, Los Angeles, CA 90095
| | - Benjamin Lopez
- Department of Pediatrics, Division of Neonatology and Developmental Biology, Neonatal Research Center, David Geffen School of Medicine at University of California, Los Angeles, CA 90095
| | - Tiffany M. Maisonet
- Department of Pediatrics, Division of Neonatology and Developmental Biology, Neonatal Research Center, David Geffen School of Medicine at University of California, Los Angeles, CA 90095
| | - Sungat Grewal
- Department of Pediatrics, Division of Neonatology and Developmental Biology, Neonatal Research Center, David Geffen School of Medicine at University of California, Los Angeles, CA 90095
| | - Vedang A. Londhe
- Department of Pediatrics, Division of Neonatology and Developmental Biology, Neonatal Research Center, David Geffen School of Medicine at University of California, Los Angeles, CA 90095
| |
Collapse
|
17
|
Weldy CS, Liu Y, Liggitt HD, Chin MT. In utero exposure to diesel exhaust air pollution promotes adverse intrauterine conditions, resulting in weight gain, altered blood pressure, and increased susceptibility to heart failure in adult mice. PLoS One 2014; 9:e88582. [PMID: 24533117 PMCID: PMC3922927 DOI: 10.1371/journal.pone.0088582] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2013] [Accepted: 01/07/2014] [Indexed: 12/15/2022] Open
Abstract
Exposure to fine particulate air pollution (PM2.5) is strongly associated with cardiovascular morbidity and mortality. Exposure to PM2.5 during pregnancy promotes reduced birthweight, and the associated adverse intrauterine conditions may also promote adult risk of cardiovascular disease. Here, we investigated the potential for in utero exposure to diesel exhaust (DE) air pollution, a major source of urban PM2.5, to promote adverse intrauterine conditions and influence adult susceptibility to disease. We exposed pregnant female C57Bl/6J mice to DE (≈300 µg/m3 PM2.5, 6 hrs/day, 5 days/week) from embryonic day (E) 0.5 to 17.5. At E17.5 embryos were collected for gravimetric analysis and assessed for evidence of resorption. Placental tissues underwent pathological examination to assess the extent of injury, inflammatory cell infiltration, and oxidative stress. In addition, some dams that were exposed to DE were allowed to give birth to pups and raise offspring in filtered air (FA) conditions. At 10-weeks of age, body weight and blood pressure were measured. At 12-weeks of age, cardiac function was assessed by echocardiography. Susceptibility to pressure overload-induced heart failure was then determined after transverse aortic constriction surgery. We found that in utero exposure to DE increases embryo resorption, and promotes placental hemorrhage, focal necrosis, compaction of labyrinth vascular spaces, inflammatory cell infiltration and oxidative stress. In addition, we observed that in utero DE exposure increased body weight, but counterintuitively reduced blood pressure without any changes in baseline cardiac function in adult male mice. Importantly, we observed these mice to have increased susceptibility to pressure-overload induced heart failure, suggesting this in utero exposure to DE ‘reprograms’ the heart to a heightened susceptibility to failure. These observations provide important data to suggest that developmental exposure to air pollution may strongly influence adult susceptibility to cardiovascular disease.
Collapse
Affiliation(s)
- Chad S Weldy
- Division of Cardiology, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, United States of America ; Department of Pathology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Yonggang Liu
- Division of Cardiology, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - H Denny Liggitt
- Department of Comparative Medicine, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Michael T Chin
- Division of Cardiology, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, United States of America ; Department of Pathology, University of Washington School of Medicine, Seattle, Washington, United States of America
| |
Collapse
|
18
|
Cavalcante FS, Aiceles V, Moraes DDFS, Alves-Pereira JL, Faria TS, Ramos CDF. The testis of the mice C57/BL6 offspring in adulthood have alterations due to maternal caffeine consumption. Acta Cir Bras 2014; 29:16-23. [PMID: 24474173 DOI: 10.1590/s0102-86502014000100003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Accepted: 12/11/2013] [Indexed: 11/21/2022] Open
Abstract
PURPOSE To investigate the effects of the maternal caffeine consumption during pregnancy to adult male testis mice offspring. METHODS Twenty pregnant mice were divided into control group (c) and caffeine group (cf). dams received daily saline or 20 mg/kg of caffeine subcutaneously. Male offspring were monitored daily until 13th week. The testis were used to evaluate both the proliferation (pcna) and apoptosis (bax); leptin receptor (ob-r); aromatase; follicle stimulating hormone (fshr), luteinizing hormone (lhr) and androgen receptors (ar); steroidogenic acute regulatory protein (star); vascular endothelial growth factor (vegf) and estrogen receptors (erα and erβ) by western blotting. Serum concentrations of testosterone, estradiol and leptin were measured. RESULTS There was a significant reduction in food intake and the body mass gain (p<0.05) in the cf ; pcna (p=0.01), fshr (p=0.02), star (p=0.0007), vegf (p=0.009), ar (p=0.03) in the cf. while an increase were note in bax (p=0.01), ob-r (p=0.02), lhr (p=0.04) and in the aromatase (p=0.03) in the cf. only erα and erβ were not changed by maternal caffeine. The serum testosterone levels in the cf offspring were 90% lower than in the c offspring (p=0.04). CONCLUSION Maternal caffeine consumption has a role and alters the testis of the offspring in adulthood.
Collapse
|
19
|
Buscariollo DL, Fang X, Greenwood V, Xue H, Rivkees SA, Wendler CC. Embryonic caffeine exposure acts via A1 adenosine receptors to alter adult cardiac function and DNA methylation in mice. PLoS One 2014; 9:e87547. [PMID: 24475304 PMCID: PMC3903656 DOI: 10.1371/journal.pone.0087547] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Accepted: 12/31/2013] [Indexed: 12/28/2022] Open
Abstract
Evidence indicates that disruption of normal prenatal development influences an individual's risk of developing obesity and cardiovascular disease as an adult. Thus, understanding how in utero exposure to chemical agents leads to increased susceptibility to adult diseases is a critical health related issue. Our aim was to determine whether adenosine A1 receptors (A1ARs) mediate the long-term effects of in utero caffeine exposure on cardiac function and whether these long-term effects are the result of changes in DNA methylation patterns in adult hearts. Pregnant A1AR knockout mice were treated with caffeine (20 mg/kg) or vehicle (0.09% NaCl) i.p. at embryonic day 8.5. This caffeine treatment results in serum levels equivalent to the consumption of 2–4 cups of coffee in humans. After dams gave birth, offspring were examined at 8–10 weeks of age. A1AR+/+ offspring treated in utero with caffeine were 10% heavier than vehicle controls. Using echocardiography, we observed altered cardiac function and morphology in adult mice exposed to caffeine in utero. Caffeine treatment decreased cardiac output by 11% and increased left ventricular wall thickness by 29% during diastole. Using DNA methylation arrays, we identified altered DNA methylation patterns in A1AR+/+ caffeine treated hearts, including 7719 differentially methylated regions (DMRs) within the genome and an overall decrease in DNA methylation of 26%. Analysis of genes associated with DMRs revealed that many are associated with cardiac hypertrophy. These data demonstrate that A1ARs mediate in utero caffeine effects on cardiac function and growth and that caffeine exposure leads to changes in DNA methylation.
Collapse
Affiliation(s)
- Daniela L. Buscariollo
- Memorial Sloan-Kettering Cancer Center, New York City, New York, United States of America
| | - Xiefan Fang
- Department of Pediatrics, University of Florida College of Medicine, Gainesville, Florida, United States of America
| | - Victoria Greenwood
- University of Connecticut, Storrs, Connecticut, United States of America
| | - Huiling Xue
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Scott A. Rivkees
- Department of Pediatrics, University of Florida College of Medicine, Gainesville, Florida, United States of America
| | - Christopher C. Wendler
- Department of Pediatrics, University of Florida College of Medicine, Gainesville, Florida, United States of America
- * E-mail:
| |
Collapse
|
20
|
Wang D, Meng J, Gao H, Xu K, Xiao R, Zhong Y, Luo X, Yao P, Yan H, Liu L. Evaluation of reproductive and developmental toxicities of Pu-erh black tea (Camellia sinensis var. assamica) extract in Sprague Dawley rats. JOURNAL OF ETHNOPHARMACOLOGY 2013; 148:190-198. [PMID: 23602733 DOI: 10.1016/j.jep.2013.04.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2012] [Revised: 03/18/2013] [Accepted: 04/08/2013] [Indexed: 06/02/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Pu-erh black tea, which is obtained by first parching crude green tea leaves and followed by secondary fermentation with microorganisms, has been believed to be beneficial beverages for health in PR China. But its potential toxicity when administered at a high dose as concentrated extract has not been completely investigated. AIM OF THE STUDY The present study was aimed at evaluating potential reproductive and developmental toxicities of Pu-erh black tea extract (BTE) in Sprague Dawley rats. MATERIALS AND METHODS Growing rats were given BTE by gavage at levels of 0, 200, 700 and 2500mg/kg/day as the F0 generation in reproductive toxicity study. Additionally, BTE was administered to mate female rats from gestation day 0.5 through 19.5 at the doses of 0, 200, 700 and 2500mg/kg/day to evaluate the developmental toxicity. RESULTS In the reproductive toxicity study, only 2500mg/kg/day BTE reduced the body weight gain and altered the relative organ weights including testes, prostata and ovary both for F0 parents and F1 offspring compared to the controls. High dose of BTE (2500mg/kg/day) administration caused developmental disturbances in embryo-to-foetus period including resorbed embryos, decreased embryo size and skeletal anomalies. CONCLUSION In conclusion, the no-observed-adverse-effect level of BTE is 700mg/kg/day both for reproductive toxicity and developmental toxicities.
Collapse
Affiliation(s)
- Di Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science & Technology, 13 Hangkong Road, Wuhan 430030, PR China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Maternal caffeine administration leads to adverse effects on adult mice offspring. Eur J Nutr 2013; 52:1891-900. [PMID: 23291721 DOI: 10.1007/s00394-012-0490-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2012] [Accepted: 12/19/2012] [Indexed: 01/14/2023]
Abstract
PURPOSE This study aimed to evaluate the role of caffeine chronic administration during gestation of C57BL/6 mice on cardiac remodeling and the expression of components of the renin-angiotensin system (RAS) in male offspring as adults. METHODS Pregnant C57BL/6 female mice were divided into two groups (n = 10): Control group (C), dams were injected with the vehicle only (saline 0.9% NaCl); Caffeine group (CF), dams received daily a subcutaneous injection of 20 mg/kg of caffeine/day (1 mg/mL saline). Pups had free access to standard chow since weaning to 3 months of age, when they were killed. RESULTS CF group showed increased energy expenditure (+7%) with consequent reduction in body mass (BM) gain (-18%), increased blood pressure (+48%), and higher heart rate (+10%) than C group. The ratio between LV mass/BM was greater (+10%), with bigger cardiomyocytes (+40%), and reduced vascularization (-25%) in CF group than in C group. In the LV, the expression of angiotensin-converting enzyme (+30%), Angiotensin II (AngII) (+60%), AngII receptor (ATR)-1 (+77%) were higher, and the expression of ATR-2 was lower (-46%; P < 0.05) in CF group than in C group. In the kidney, the expressions of renin (+128%) and ATR-1 (+88%) were higher in CF group than in C group. CONCLUSIONS Chronic administration of caffeine to pregnant dams led to persistent activation of local RAS in the kidney and heart of the offspring, which, in turn, leads to high BP and adverse cardiac remodeling. These findings highlight the urge to encourage pregnant women to avoid food or medicines containing caffeine.
Collapse
|
22
|
Rivkees SA, Wendler CC. Regulation of cardiovascular development by adenosine and adenosine-mediated embryo protection. Arterioscler Thromb Vasc Biol 2012; 32:851-5. [PMID: 22423036 DOI: 10.1161/atvbaha.111.226811] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Few signaling molecules have as much potential to influence the developing mammal as the nucleoside adenosine. Adenosine levels increase rapidly with tissue hypoxia and inflammation. Adenosine antagonists include the methylxanthines caffeine and theophylline. The receptors that transduce adenosine action are the A1, A2a, A2b, and A3 adenosine receptors (A1AR, A2aAR, A2bAR, and A3AR). We examined how adenosine acts via A1ARs to influence embryo development. Transgenic mice were studied along with embryo cultures. Embryos lacking A1ARs were markedly growth retarded following intrauterine hypoxia exposure. Studies of mice selectively lacking A1AR in the heart identify the heart as a key site of adenosine's embryo-protective effects. Studies of isolated embryos showed that adenosine plays a key role in modulating embryo cardiac function, especially in the setting of hypoxia. When pregnant mice were treated during embryogenesis with the adenosine antagonist caffeine, adult mice had abnormal heart function. Adenosine acts via A1ARs to play an essential role in protecting the embryo against intrauterine stress, and adenosine antagonists, including caffeine, may be an unwelcome exposure for the embryo.
Collapse
Affiliation(s)
- Scott A Rivkees
- Department of Pediatrics, Yale Child Health Research Center, Yale University School of Medicine, New Haven CT, USA.
| | | |
Collapse
|
23
|
Abstract
This review discusses epidemiology and laboratory studies on the effects of prenatal methylxanthine administration on some systems developing organisms. They are mainly absorbed from coffee, tea and cocoa products such as cola beverages and chocolate bars. Prenatal methylxanthine exposure can induce several unfavourables changes in the developing organism, which are persistent even in later phases of life. Based on results obtained from animal studies, the effect on embryogenesis is not only poorly understood but also controversial. It is therefore important to study interspecies differences as results may differ depending on animals used and administration methods.
Collapse
|
24
|
Buscariollo DL, Breuer GA, Wendler CC, Rivkees SA. Caffeine acts via A1 adenosine receptors to disrupt embryonic cardiac function. PLoS One 2011; 6:e28296. [PMID: 22164264 PMCID: PMC3229565 DOI: 10.1371/journal.pone.0028296] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2011] [Accepted: 11/04/2011] [Indexed: 12/29/2022] Open
Abstract
Background Evidence suggests that adenosine acts via cardiac A1 adenosine receptors (A1ARs) to protect embryos against hypoxia. During embryogenesis, A1ARs are the dominant regulator of heart rate, and A1AR activation reduces heart rate. Adenosine action is inhibited by caffeine, which is widely consumed during pregnancy. In this study, we tested the hypothesis that caffeine influences developing embryos by altering cardiac function. Methodology/Principal Findings Effects of caffeine and adenosine receptor-selective antagonists on heart rate were studied in vitro using whole murine embryos at E9.5 and isolated hearts at E12.5. Embryos were examined in room air (21% O2) or hypoxic (2% O2) conditions. Hypoxia decreased heart rates of E9.5 embryos by 15.8% and in E12.5 isolated hearts by 27.1%. In room air, caffeine (200 µM) had no effect on E9.5 heart rates; however, caffeine increased heart rates at E12.5 by 37.7%. Caffeine abolished hypoxia-mediated bradycardia at E9.5 and blunted hypoxia-mediated bradycardia at E12.5. Real-time PCR analysis of RNA from isolated E9.5 and E12.5 hearts showed that A1AR and A2aAR genes were expressed at both ages. Treatment with adenosine receptor-selective antagonists revealed that SCH-58261 (A2aAR-specific antagonist) had no affects on heart function, whereas DPCPX (A1AR-specific antagonist) had effects similar to caffeine treatment at E9.5 and E12.5. At E12.5, embryonic hearts lacking A1AR expression (A1AR−/−) had elevated heart rates compared to A1AR+/− littermates, A1AR−/− heart rates failed to decrease to levels comparable to those of controls. Caffeine did not significantly affect heart rates of A1AR−/− embryos. Conclusions/Significance These data show that caffeine alters embryonic cardiac function and disrupts the normal cardiac response to hypoxia through blockade of A1AR action. Our results raise concern for caffeine exposure during embryogenesis, particularly in pregnancies with increased risk of embryonic hypoxia.
Collapse
Affiliation(s)
- Daniela L. Buscariollo
- Section of Developmental Endocrinology and Biology, Yale Child Health Research Center, Department of Pediatrics, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Gregory A. Breuer
- Section of Developmental Endocrinology and Biology, Yale Child Health Research Center, Department of Pediatrics, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Christopher C. Wendler
- Section of Developmental Endocrinology and Biology, Yale Child Health Research Center, Department of Pediatrics, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Scott A. Rivkees
- Section of Developmental Endocrinology and Biology, Yale Child Health Research Center, Department of Pediatrics, Yale University School of Medicine, New Haven, Connecticut, United States of America
- * E-mail:
| |
Collapse
|
25
|
An inflammatory pathway to apnea and autonomic dysregulation. Respir Physiol Neurobiol 2011; 178:449-57. [DOI: 10.1016/j.resp.2011.06.026] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2011] [Revised: 06/29/2011] [Accepted: 06/29/2011] [Indexed: 01/04/2023]
|
26
|
Koos BJ. Adenosine A₂a receptors and O₂ sensing in development. Am J Physiol Regul Integr Comp Physiol 2011; 301:R601-22. [PMID: 21677265 DOI: 10.1152/ajpregu.00664.2010] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Reduced mitochondrial oxidative phosphorylation, via activation of adenylate kinase and the resulting exponential rise in the cellular AMP/ATP ratio, appears to be a critical factor underlying O₂ sensing in many chemoreceptive tissues in mammals. The elevated AMP/ATP ratio, in turn, activates key enzymes that are involved in physiologic adjustments that tend to balance ATP supply and demand. An example is the conversion of AMP to adenosine via 5'-nucleotidase and the resulting activation of adenosine A(₂A) receptors, which are involved in acute oxygen sensing by both carotid bodies and the brain. In fetal sheep, A(₂A) receptors associated with carotid bodies trigger hypoxic cardiovascular chemoreflexes, while central A(₂A) receptors mediate hypoxic inhibition of breathing and rapid eye movements. A(₂A) receptors are also involved in hypoxic regulation of fetal endocrine systems, metabolism, and vascular tone. In developing lambs, A(₂A) receptors play virtually no role in O₂ sensing by the carotid bodies, but brain A(₂A) receptors remain critically involved in the roll-off ventilatory response to hypoxia. In adult mammals, A(₂A) receptors have been implicated in O₂ sensing by carotid glomus cells, while central A(₂A) receptors likely blunt hypoxic hyperventilation. In conclusion, A(₂A) receptors are crucially involved in the transduction mechanisms of O₂ sensing in fetal carotid bodies and brains. Postnatally, central A(₂A) receptors remain key mediators of hypoxic respiratory depression, but they are less critical for O₂ sensing in carotid chemoreceptors, particularly in developing lambs.
Collapse
Affiliation(s)
- Brian J Koos
- Department of Obstetrics and Gynecology; Brain Research Institute, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California, USA.
| |
Collapse
|
27
|
Rivkees SA, Wendler CC. Adverse and protective influences of adenosine on the newborn and embryo: implications for preterm white matter injury and embryo protection. Pediatr Res 2011; 69:271-8. [PMID: 21228731 PMCID: PMC3100210 DOI: 10.1203/pdr.0b013e31820efbcf] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Few signaling molecules have the potential to influence the developing mammal as the nucleoside adenosine. Adenosine levels increase rapidly with tissue hypoxia and inflammation. Adenosine antagonists include the methylxanthines caffeine and theophylline. The receptors that transduce adenosine action are the A1, A2a, A2b, and A3 adenosine receptors (ARs). In the postnatal period, A1AR activation may contribute to white matter injury in the preterm infant by altering oligodendrocyte (OL) development. In models of perinatal brain injury, caffeine is neuroprotective against periventricular white matter injury (PWMI) and hypoxic-ischemic encephalopathy (HIE). Supporting the notion that blockade of adenosine action is of benefit in the premature infant, caffeine reduces the incidence of bronchopulmonary dysplasia and CP in clinical studies. In comparison with the adverse effects on the postnatal brain, adenosine acts via A1ARs to play an essential role in protecting the embryo from hypoxia. Embryo protective effects are blocked by caffeine, and caffeine intake during early pregnancy increases the risk of miscarriage and fetal growth retardation. Adenosine and adenosine antagonists play important modulatory roles during mammalian development. The protective and deleterious effects of adenosine depend on the time of exposure and target sites of action.
Collapse
MESH Headings
- Adenosine/antagonists & inhibitors
- Adenosine/pharmacology
- Adenosine/therapeutic use
- Animals
- Caffeine/pharmacology
- Embryo, Mammalian/drug effects
- Embryo, Mammalian/physiology
- Female
- Humans
- Hypoxia-Ischemia, Brain/drug therapy
- Infant, Newborn
- Infant, Premature
- Leukomalacia, Periventricular/drug therapy
- Nerve Fibers, Myelinated/drug effects
- Nerve Fibers, Myelinated/pathology
- Neuroprotective Agents/pharmacology
- Neuroprotective Agents/therapeutic use
- Oligodendroglia/drug effects
- Oligodendroglia/pathology
- Pregnancy
- Protein Isoforms/genetics
- Protein Isoforms/metabolism
- Purinergic P1 Receptor Antagonists/pharmacology
- Receptors, Purinergic P1/genetics
- Receptors, Purinergic P1/metabolism
- Respiration/drug effects
Collapse
Affiliation(s)
- Scott A Rivkees
- Department of Pediatrics, Yale Child Health Research Center, Yale University School of Medicine, New Haven, Connecticut 06520, USA.
| | | |
Collapse
|
28
|
Conde A, Figueiredo B, Tendais I, Teixeira C, Costa R, Pacheco A, Rodrigues MC, Nogueira R. Mother's anxiety and depression and associated risk factors during early pregnancy: effects on fetal growth and activity at 20-22 weeks of gestation. J Psychosom Obstet Gynaecol 2010; 31:70-82. [PMID: 20236029 DOI: 10.3109/01674821003681464] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
To examine effects of mother's anxiety and depression and associated risk factors during early pregnancy on fetal growth and activity. Repeated measures of mother's anxiety (State-Anxiety Inventory (STAI-S)) and depression (Edinburgh Postnatal Depression Scale (EPDS)) and related socio demographics and substance consumption were obtained at the 1st and 2nd pregnancy trimesters, and fetus' (N = 147) biometric data and behavior was recorded during ultrasound examination at 20-22 weeks of gestation. Higher anxiety symptoms were associated to both lower fetal growth and higher fetal activity. While lower education, primiparity, adolescent motherhood, and tobacco consumption predicted lower fetal growth, coffee intake predicted lower fetal activity. Vulnerability of fetal development to mother's psychological symptoms as well as to other sociodemographic and substance consumption risk factors during early and mid pregnancy is suggested.
Collapse
Affiliation(s)
- Ana Conde
- School of Psychology, University of Minho, Braga, Portugal
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Wendler CC, Poulsen RR, Ghatpande S, Greene RW, Rivkees SA. Identification of the heart as the critical site of adenosine mediated embryo protection. BMC DEVELOPMENTAL BIOLOGY 2010; 10:57. [PMID: 20509906 PMCID: PMC2890593 DOI: 10.1186/1471-213x-10-57] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2010] [Accepted: 05/28/2010] [Indexed: 11/10/2022]
Abstract
BACKGROUND Our understanding of the mechanisms that protect the developing embryo from intrauterine stress is limited. Recently, adenosine has been demonstrated to play a critical role in protecting the embryo against hypoxia via adenosine A1 receptors (A1ARs), which are expressed in the heart, nervous system, and other sites during development. However, the sites of A1AR action that mediate embryo protection are not known. To determine if the heart is a key site of adenosine-mediated embryo protection, A1ARs were selectively deleted in the embryonic heart using a Cre-LoxP system in which the alpha-myosin heavy chain promoter drives Cre-recombinase expression and excision of the A1AR gene from cardiomyocytes. RESULTS With increasing exposure of maternal hypoxia (10% O2) from 48-96 hours beginning at embryonic day (E) 8.5, embryo viability decreased in the cardiac-A1AR deleted embryos. 48 hours of hypoxia reduced embryonic viability by 49% in embryos exposed from E10.5-12.5 but no effect on viability was observed in younger embryos exposed to hypoxia from E8.5-10.5. After 72 hours of hypoxia, 57.8% of the cardiac-A1AR deleted embryos were either dead or re-absorbed compared to 13.7% of control littermates and after 96 hours 81.6% of cardiac-A1AR deleted embryos were dead or re-absorbed. After 72 hours of hypoxia, cardiac size was reduced significantly more in the cardiac-A1AR deleted hearts compared to controls. Gene expression analysis revealed clusters of genes that are regulated by both hypoxia and A1AR expression. CONCLUSIONS These data identify the embryonic heart as the critical site where adenosine acts to protect the embryo against hypoxia. As such these studies identify a previously unrecognized mechanism of embryo protection.
Collapse
Affiliation(s)
- Christopher C Wendler
- Department of Pediatrics, Section of Developmental Endocrinology and Biology, Yale Child Health Research Center, Yale University School of Medicine, New Haven, CT 06520 USA.
| | | | | | | | | |
Collapse
|