Abstract
The amount of scientific research linking environmental exposures and childhood health outcomes continues to grow; yet few studies have teased out the mechanisms involved in environmentally-induced diseases. Cells can respond to environmental stressors in many ways: inducing oxidative stress/inflammation, changes in energy production and epigenetic alterations. Mitochondria, tiny organelles that each retains their own DNA, are exquisitely sensitive to environmental insults and are thought to be central players in these pathways. While it is intuitive that mitochondria play an important role in disease processes, given that every cell of our body is dependent on energy metabolism, it is less clear how environmental exposures impact mitochondrial mechanisms that may lead to enhanced risk of disease. Many of the effects of the environment are initiated in utero and integrating mitochondriomics into children’s environmental health studies is a critical priority. This review will highlight (i) the importance of exploring environmental mitochondriomics in children’s environmental health, (ii) why environmental mitochondriomics is well suited to biomarker development in this context, and (iii) how molecular and epigenetic changes in mitochondria and mitochondrial DNA (mtDNA) may reflect exposures linked to childhood health outcomes.
Keywords: environmental health, children, mitochondria, copy number, neurodevelopment, asthma, biomarkers, environmental exposure
Environmental Mitochondriomics and Child Health
In this review, we examine the current literature and expert opinions on environmental mitochondriomics (EM), a new area of research dedicated to discovering in mitochondria novel sensors and mediators of environmental effects. While it is intuitive that mitochondria play an important role in disease processes, given that every cell of our body is dependent on energy metabolism, it is less clear how environmental exposures impact mitochondrial mechanisms that may lead to enhanced risk of disease. This review will highlight why environmental mitochondriomics is well suited to biomarker development and how molecular and epigenetic changes in mitochondrial (mtDNA) may reflect exposures linked to childhood health outcomes. While there are many child health outcomes that could be explored with respect to environmental mitochondriomics given their mitochondrial underpinnings (Fleischman et al., 2009; Simoes et al., 2012; Flaquer et al., 2014; Knoll et al., 2013; Legido et al., 2013) (Fig. 1), we will focus on two of the most common health concerns in childhood which are supported by a solid foundation of research suggesting mitochondriomics is likely a promising biomarker platform to explore – asthma and neurodevelopment.
Figure 1.
Conceptual model linking environmental mitochondriomics to child health outcomes.
Asthma and Neurodevelopment: Model Outcomes for Mitochondriomics
Childhood Asthma and the Mitochondria
Asthma is the most common chronic inflammatory/respiratory disease among children worldwide, and its prevalence continues to rise (Anandan et al., 2010). While the origins of asthma are likely multi-factorial, the underlying mechanisms leading to reduced lung function and exaggerated airway responsiveness involve chronic airway inflammation associated with a cycle of injury, repair and remodeling (Holgate, 1997; Holt et al., 2005). Notably, airway inflammation and remodeling begin and progress even in the presymptomatic state in early childhood (Morgan et al., 2005; Pohunek et al., 2005). Furthermore, the fundamental cause of airway inflammation is aberrant and/or excessive immune responses to various environmental factors (Holt et al., 2005) and the most common cause of chronic airway inflammation in early childhood is arguably asthma.
Mitochondria are important to the inflammatory process for many reasons. Inflammation is triggered by exposure of the airway epithelial cells to oxidative stress, a process mediated by reactive oxygen species (ROS) (Riedl and Nel, 2008). Endogenous ROS are primarily a product of the mitochondria. If mitochondria are not functioning properly, this can lead to excess ROS ultimately boosting inflammation. It is possible an excessive amount of ROS can damage the mitochondria until apoptosis of airway epithelial cells occurs (Trian et al., 2007; Simoes et al., 2012). Further, mitochondria are key players in human eosinophil apoptosis and survival (Ilmarinen et al., 2014); eosinophils are cells of the innate immune system primarily involved in the allergic inflammatory response in chronic asthma (Felton et al., 2014).
The preferential importance of maternal history of atopy or asthma as a risk factor for childhood asthma is one of the most consistent and reproducible observations in asthma epidemiology (Martinez et al., 1995; Litonjua et al., 1998; Celedon et al., 2003; Tan et al., 2007; Barrett, 2008; Lim et al., 2010). The consistency of these relationships and their fairly early onset suggest a genetic influence that perhaps involves the mitochondrial genome, predominantly inherited from the mother (Giles et al., 1980). While the evidence demonstrating the presence of mitochondrial dysfunction in asthma is not abundant, there does appear to be enough literature supporting an association between mitochondrial dysfunction and atopy (Raby et al., 2007; Chodaczek et al., 2009), respiratory morbidity (Schmuczerova et al., 2009; Meert et al., 2012; Ostroukhova et al., 2012) and asthma risk (Heinzmann et al., 2003; Zifa et al., 2012; Flaquer et al., 2014).
Child Neurodevelopment and the Mitochondria
The developing fetus and infant are most vulnerable owing to the rapid differentiation of the central nervous system in utero and early life (Landrigan et al., 1999; Rice and Barone, 2000). Neurocognitive and behavioral deficits in childhood, including problems with learning, attention, conduct, depression and anxiety affect up to 20% of U.S. children, placing a burden on education and healthcare systems (Froehlich et al., 2007; Boulet et al., 2009; Montes et al., 2012). However, the potential loss of functioning early in life as a result of environmental factors may result in diminished academic and economic productivity that persists over the life span (Trasande et al., 2005). Weiss (2000) estimated that a U.S. population average decrease of 1% in IQ, caused by an environmental toxicant, would result in an annual cost of $50 billion in lost economic productivity and increased special educational needs amounting to a lifespan cost estimate in the trillions. Fewer than 25% of childhood neurodevelopmental disabilities have an identified cause, signifying the need to examine candidate neurotoxicants and programming mechanisms that may be amenable to intervention strategies.
The mitochondria play an important role in neurodevelopment. They regulate innate immune responses that can have broad consequences for brain development and susceptibility to damage (Hagberg et al., 2014). During fetal and postnatal brain development, new mitochondria are being formed and present mitochondria are growing rapidly (e.g. mitochondrial biogenesis) as the brain has a high demand for energy and thus neurons contain a large number of mitochondria. Mitochondrial biogenesis is highly regulated; proteins encoded by nuclear and mitochondrial DNA are needed (Scarpulla, 2011). Many mitochondrial functions and pathophysiological responses in the immature and adult brain vary. As reviewed by Hagberg et al. (2014), mitochondrial respiratory activity, and protein and respiratory enzyme content are lower in the immature brain compared to the adult brain. A role of mitochondria in neurogenesis and synaptic plasticity has been established (Vayssiere et al., 1992; Mattson et al., 2008; Streck et al., 2014) signifying their importance in normal neurodevelopment.
Mitochondrial dysfunction (MD) has been extensively researched with regard to autism spectrum disorders (ASDs) with several reviews highlighting the potential role of MD in ASDs (Palmieri and Persico, 2010; Rossignol and Frye, 2012; Legido et al., 2013). Several lines of evidence suggest that suboptimal mitochondrial energy metabolism (e.g. high lactate, increased lactate to pyruvate ratio, increased alanine and low carnitine) and electron transport chain (ETC) abnormalities are linked to ASDs (Oliveira et al., 2005; Correia et al., 2006; Weissman et al., 2008; Rossignol and Frye, 2012). Evidence of the first mtDNA mutations associated with ASDs was published in 1999 and 2000 (Sue et al., 1999; Graf et al., 2000), mtDNA mutations A3243G [often associated with mitochondrial encephalopathy with lactic acidosis (MELAS) and seizures] (Sue et al., 1999; Pons et al., 2004) and G8363A in mtDNA tRNAlys (Graf et al., 2000). Other mtDNA mutations and abnormalities (e.g. mtDNA copy number) have been identified in patients with ASD (Weissman et al., 2008; Giulivi et al., 2010; Legido et al., 2013; Napoli et al., 2013). Subsequently, progressive increases in mtDNA A3243G heteroplasmy (defined below in Molecular archive of past environments and cumulative risk), can lead to abrupt transcriptional reprogramming (Picard et al., 2014). Few studies have investigated the influence of mitochondrial dysfunction on other neurodevelopmental disorders; however, findings suggest mitochondrial respiratory chain defects are present in attention deficit hyperactivity disorder (Papa et al., 2000; Fagundes et al., 2007; Bradstreet et al. 2010) as well as other psychiatric disorders (Rezin et al., 2009; Scaglia, 2010). While research supports a role of mitochondrial dysfunction in child neurodevelopment, it is unclear whether MD is primary etiology or secondary to other causes such as environmental exposures.
Novel Biomarkers for Environmental Health Risk
Mitochondria: The Power House
Mitochondrial cell biology
Mitochondria play a central role in many cellular processes, notably cellular energy delivery through ATP. They consume oxygen (O2) and produce ATP by electron transport and oxidative phosphorylation taking place across the respiratory chain complexes located in the inner mitochondrial membrane (O’Connor and Adams, 2010). The mitochondrial genome also differs in many ways from the nuclear genome. First, mitochondria are the only organelles in human cells to possess their own genomes and are predominantly maternal transmission (Giles et al., 1980). Each human cell contains several hundred to >1000 mitochondria, each carrying 2–10 copies of mtDNA. The mitochondrial genome is double-stranded and circular containing approximately 16500 nucleotides; it encodes 13 peptides for oxidative phosphorylation, and 22 transfer ribonucleic acids (tRNAs) and 2 ribosomal RNAs (rRNAs) essential for protein synthesis within the mitochondria (Li et al., 2012). The mitochondrial genome contains fewer, yet evenly dispersed, CpG dinucleotides compared with nDNA and lacks retrotransposons (Byun and Baccarelli, 2014).
The importance of mitochondria in fetoplacental development
The placenta is a key regulator of fetal development as it facilitates nutrient supply to and waste removal from the fetus; alterations in placental function can mediate fetal programming (Wu et al., 2012). Placental metabolic activity throughout gestation is sustained by increasing mitochondrial activity (i.e. mitochondrial oxidative phosphorylation) and biogenesis (Bax and Bloxam, 1997; Leduc et al., 2010). Animal and human studies have shown that altered mitochondrial function affects subsequent fetal and placental development (Wakefield et al., 2011; Mandò et al., 2014). Conversely, maternal factors such as obesity (Mele et al., 2014) and malnutrition (Mayeur et al., 2013) have been shown to induce impaired placental mitochondrial function leading to fetoplacental growth deficits.
Susceptibility to environmental toxicants
Mitochondria are vulnerable to damage by ROS and during normal energy production they are the main source of endogenous ROS in most mammalian cells (Shaughnessy et al., 2014). When the production of ROS is greater than the inherent antioxidant defenses, a condition known as oxidative stress occurs (Burton and Jauniaux, 2011). Mitochondrial proteins and mtDNA are also vulnerable to exogenous insults leading to the alteration of mtDNA integrity, electron transport chain activity, modifying membrane potential, ion transport and apoptotic signaling (Meyer et al., 2013). Damaged mitochondria can produce 10 times more hydrogen peroxide than undamaged mitochondria (Grivennikova et al., 2010). All of these factors are involved in mitochondrial dysfunction.
Mitochondria also vary from tissue to tissue with different cell types being differentially sensitized to environmental toxicants. The placenta, for example, has a high reliance on mitochondrial function; this may mean that dysfunction could more easily result in cell death as a result of increased ROS production (Meyer et al., 2013). In addition, research suggests that mtDNA repair capacity and antioxidant defense mechanisms also vary by cell type, with brain tissue expressly having a low repair capacity (Szczesny et al., 2010) and neurons having low antioxidant defenses and/or high inherent oxidative stress (Crouch et al., 2007).
A large number of environmental toxicants may exert their effect through the production of ROS, oxidative stress and inflammation. Mitochondrial function can be altered by oxidative stress and inflammation (Lee and Wei, 2000). In turn, dysfunctional mitochondria produce additional oxidation that may sustain systemic oxidative stress (Lee and Wei, 2000) which may alter developmental programming of key regulatory systems affecting respiratory and neurodevelopment in children.
Molecular Archive of Past Environments and Cumulative Risk
Studies have identified a set of mtDNA-based molecular markers associated with mitochondrial dysfunction and oxidative stress; these include mtDNA oxidation (Lin et al., 2008), heteroplasmy (Sondheimer et al., 2011) and copy number (mtDNAcn) (Hou et al., 2010). In blood, mtDNA accumulates oxidative damage over time – as reflected in 8-hydroxy-2′-deoxyguanosine (8-OHdG) levels in mtDNA (Mecocci et al., 1998). To compensate for oxidation and damage, more copies of mtDNA are replicated, thus leading to increased mtDNAcn (Liu et al., 2006). Perhaps owing to its continuing exposure to mitochondrial ROS, mitochondrial DNA has a fairly high mutation rate. When a new mutation arises in the mtDNA of a cell, a mixed population of mtDNAs is generated; this is known as mitochondrial heteroplasmy (MH). MH is a common phenomenon. Baseline levels of MH are commonly detectable owing to the limited mtDNA protection from oxidative stress (Chinnery et al., 2012; Li et al., 2012). Baseline MH is hypothetically tolerated by the cell that carries multiple mitochondria each with multiple mtDNA copies. MH becomes symptomatic when it reaches a threshold affecting the cellular functioning presumably due to oxidative insults and replication errors (Golden and Melov, 2001; Graziewicz et al., 2002; Szibor and Holtz, 2003; Sondheimer et al., 2011). Therefore, the degree of mtDNAcn and/or heteroplasmy may be considered as a ’biological clock‘ that measures past exposure to oxidative stress.
Compared with mitochondrial DNA oxidative damage, copy number and mutations, which have been more widely studied in relation to environmental exposures and disease risk, mitochondrial epigenetics is still in its infancy but is fast growing. Mitochondrial epigenetics has been a point of contention in the literature. One of the first reports on mtDNA methylation was published in 1971 (Vanyushin et al., 1971). Many studies supported this finding in the years following (Kudriashova et al., 1976; Shock et al., 2011; Bellizzi et al., 2013) while others found no evidence of CpG methylation in human mtDNA (Hong et al., 2013). Given that oxidative stress in the nucleus has been shown to alter nuclear DNA methylation, numerous environmental toxicants may be potential candidates with important links to mitochondrial epigenetics. To date, no studies have investigated the association between mitochondrial epigenetics and asthma and/or neurodevelopment despite the plethora of research implicating mitochondrial mechanisms in the development of a large number of childhood conditions, such as asthma, obesity, insulin resistance, autism and attention problems (Fleischman et al., 2009; Bradstreet et al., 2010; Giulivi et al., 2010; Stephens et al., 2011; Meert et al., 2012; Anitha et al., 2013; Hernandez-Aguilera et al., 2013; Killeen et al., 2013; Oguzkan-Balci et al., 2013). Further, chronic diseases with mitochondrial underpinnings, such as asthma and various neurodevelopmental conditions, do not follow Mendelian inheritance. Therefore, despite the debate, there is increasing interest in studying the role of mitochondrial epigenetics in environmentally-induced diseases.
Mitochondrial markers such as RNA modification (i.e. RNA methylation) and miRNAs should also be explored (Barrey et al., 2011; Motorin and Helm, 2011; Byun and Baccarelli, 2014). Mitochondrial RNA accounts for 5% of total cellular RNA in most tissues, but can be as high as 30% in tissues with a high metabolic demand such as the heart (Mercer et al., 2011); however, additional work is needed in describing mitochondrial RNA/miRNAs and their role in regulating mitochondrial gene expression. Further, cellular reprogramming may begin with mitochondrial response (e.g. changes in mitochondrial copy number) and followed by changes in nDNA genes coding for mitochondrial proteins making the cross-talk between the mitochondrial and nuclear genome an important area of research (Shaughnessy et al., 2014). Regardless of the mitochondrial marker (s) one chooses to utilize, careful consideration should be given when designing epidemiological studies (Byun and Baccarelli, 2014). It has been suggested that integrating multiple mitochondrial endpoints would be the most efficient way to probe the extent of systemic oxidative stress for many clinical outcomes (Chan et al., 2013).
Mitochondrial Markers of Environmental Exposures
According to the World Health Organization’s Global Plan of Action for Children’s Health and the Environment, each year over 3 million children under the age of five die as a result of environmentally-related illnesses (WHO, 2009). Environmental mitochondriomics is in its infancy with respect to environmental health research with nearly all studies occurring after 2010; even more scarce are studies examining prenatal and early-life exposures (Table 1). The research detailed below ranges from in vitro/in vivo animal studies to large epidemiological cohort analyses; however, it is critical to recognize that many of the effects of the environment are initiated in utero and thus the need to integrate mitochondriomics into children’s environmental health.
Table 1.
Studies investigating the association between environmental exposures and mitochondrial markers
Exposure Category | Author | Study description | Study sample | Exposure (s) | Biomarker(s) | Significant Findings | Comment(s) |
---|---|---|---|---|---|---|---|
Air pollution | |||||||
Li et al. 2003 | In vitro | RAW 264.7 and BEAS-2B murine cell lines | PM10, PM2.5, UFPs | Mitochondria morphology | UFPs localized in mitochondria and induced structural damage | Cell lines mimic oxidative stress response in pulmonary alveolar macrophages and bronchial epithelial cells | |
Hou et al. 2010 | Brescia, Italy-Steel Plant | 63 healthy male workers | PM10, PM1 | mtDNAcn in blood | PM positively associated with mtDNAcn | Metal exposures examined (see below) | |
Janssen et al. 2012 | Genk, Belgium | 178 Newborns | PM10,NO2, distance to major road | mtDNAcn in placental tissue and cord blood | PM10 associated with 16.1% decrease in placental mtDNAcn; increase in distance to major road resulted in increased placental mtDNAcn | No association found for cord blood mtDNAcn | |
Hou et al. 2013 | Beijing, China | 120 men and women (60 truck drivers; 60 office workers) | PM2.5, Elemental Carbon (EC) | mtDNAcn in blood | Increased EC and PM10 exposure associated with decreased mtDNAcn in all participants | No association found with PM2.5 | |
Byun et al. 2013 | Brescia, Italy; Milan, Italy; Beijing, China | 40 male participants from each location (20 high/20 low exposed) | PM1 (Brescia), air benzene (Milan), and EC (Beijing) | mtDNA methylation in 3 regions (MT-TF, MT-RNR1, D-loop) and mtDNAcn in blood | High PM1 associated with higher MT-TF and MT-RNR1 methylation; MT-RNR1 methylation positively correlated with mtDNAcn; D-loop was not associated with PM1 exposure; No effects on mtDNA methylation from benzene or EC | Participants were steel workers (Brescia), gas-station attendants (Milan), and truck drivers (Beijing); PM1 was rich in metals | |
Carugno et al. 2012 | Genoa, Milan, and Cagliari Italy | 519 men and women (341 exposed, 178 referents) | Low-level air benzene | mtDNAcn in blood | In each city, benzene-exposed had higher mtDNAcn than referents; IQR increase in exposure associated with 10% increase in mtDNAcn across cities | Sub analysis (Milan participants only) showed mtDNAcn to be associated with LINE-1 hypomethylation and p15 hypermethylation | |
Pavanello et al. 2013 | Poland | 46 coke-oven workers; 44 matched controls (all men) | Polycyclic aromatic hydrocarbons (PAHs) | mtDNAcn in blood | High PAH exposure associated with higher mtDNAcn compared to controls | Matched on gender and ethnicity | |
Wittkopp et al. 2013 | Los Angeles, CA | 60 elderly men and women over 65 years | Traffic-related air pollution (TRAP)- PAH, BC, NOx, and CO | mtDNA haplogroups H & U | Increases in inflammation biomarkers (IL-6 and TNF-α) associated with higher TRAP exposure; associations were the strongest for haplogroups H | All participants had confirmed coronary artery disease and were non-smokers | |
Colicino et al. 2014 | United States, VA Normative Aging Study | 582 elderly men | Black Carbon (BC) | Nine mtDNA haplogroups categorized into 4 clusters | Largest effect of BC on cognitive impairment observed for haplogroups I, W, and X; moderate effect for J and T; no effect for H, V, K, or U haplogroups | mtDNA haplotypes may modify individual susceptibility to BC cognitive effects | |
Tobacco Smoke | |||||||
Hara et al. 2013 | In vitro | Human bronchial epithelial cells | Cigarette smoke extract (CSE) | Mitochondrial morphology and ROS production | CSE induced mitochondrial fragmentation and ROS production; this resulted in cellular senescence | Cellular senescence has been implicated in asthma development [Albrecht et al. 2014] | |
Ballinger et al. 1996 | In vivo | Human bronchoalveolar lavage tissues | Tissues from smokers and non-smokers | mtDNA damage and deletions | Smokers 5.6 times more mtDNA damage including a 4.9 kb mtDNA deletion | mtDNA damage assessed by Q-XLPCR | |
Westbrook et al. 2010 | In vivo | M. mulatta non-human primates | Low levels ETS (1 mg/m3) from gestation to 1 year | Mitochondrial function and mtDNA damage (mtDNAcn) in aortic tissues and blood | Perinatal ETS exposure associated with mitochondrial dysfunction and mtDNAcn in aortic tissues; mtDNA damage also observed in blood | Mitochondrial function assessed by cytochrome oxidase activity | |
Micale et al. 2013 | In vivo | Swiss H mice | CS exposure for 4 weeks starting at birth or 4 months of age | Mitochondrial potential/mass | Exposure to CS affected mitochondrial potential/mass in young mice | Control mice were sham-exposed | |
Aravamudan et al. 2014 | In vitro | Non-asthmatic human airway smooth muscle cells | CS exposure | Mitochondrial fragmentation and morphology | CSE induced fragmentation and damaged mitochondria via increased expression of mitochondrial fission protein dynamin-related protein (Drp1) and decreased fusion protein mitofusin (Mfn) | Mitochondrial fission/fusion proteins can influence ROS production, cell proliferation, and apotptosis | |
Metals | |||||||
Hou et al. 2010 | Brescia, Italy-Steel Plant | 63 healthy male workers | Airborne Metals: chromium, lead, arsenic, nickel, manganese | mtDNAcn in blood | Metal concentrations were not associated with MtDNAcn | Ambient air pollution exposures examined (see above) | |
Wang et al. 2009 | In vivo and human study (Shandong, China) | Wistar rats; Han Chinese pregnant women (n = 67) | Lead acetate exposure during gestation (rats) and blood lead levels | Mitochondrial structure/number | In the rat placenta mitochondria were swollen, decreased in number, endoplasmic reticula were distended and ribosomal number on membranes decreased; no abnormal structure was observed in human placenta | Damaged cytoplasmic organelles may interfere with oxygen exchange and nutrition at the maternal-fetal interface | |
Vergilio Cdos & de Melo 2013 | In vitro | Human hepatic carcinoma cells | Cadmium | Mitochondrial function | Cd treated cells displayed a progressive loss of mitochondrial function leading to apoptotic events | Disorganized endoplasmic reticula also observed | |
Kurochkin et al. 2011 | In vivo | Crassostrea virginica (eastern oyster) | Cadmium | Mitochondrial function | Cadmium exposure inhibited substrate oxidation and stimulated proton conductance across inner mitochondrial membrane | Cadmium effects were largely ROS independent | |
Hernandez et al. 2011 | In vitro | Neocortical and cerebellar granular neurons | Manganese | Mitochondrial function | Manganese exposure lead to mitochondrial homeostasis dysfunction | Ascorbate protected against Mn-induced neurotoxicity | |
Sharma et al. 2013 | In vivo | Male albino Wistar Rats | Aluminum lactate | Mitochondrial DNA oxidation, mtDNAcn, ROS levels, citrate synthase activity, and morphology in brain tissue | After 12 weeks of exposure, Hippocampus and Corpus striatum regions of the brain showed increased ROS levels, mtDNA oxidation and decreased citrate synthase activity and mtDNAcn. Mitochondria were swollen, exhibited a loss of cristae, and chromatin condensation | Decreases in mRNA levels of mitochondrial encoded subunits were observed as well as reduced expression of nuclear encoded subunits of the electron transport chain | |
Endocrine Disruptors | |||||||
Lin et al. 2013 | In vitro | INS-1 β cells | Bisphenol A (BPA) | Mitochondrial function and gene expression | After BPA exposure, ATP depletion, cytochrome c release, loss in mitochondrial mass and membrane potential, and altered expression of genes involved in mitochondrial function was observed | Cells also became progressively became apoptotic after BPA exposure | |
Kaur et al. 2014 | In vitro | Lymphoblastoid cells from autistic and control children | BPA | Mitochondrial function | BPA was associated with decreased mitochondrial membrane potential in both groups. Increases in mtDNAcn were observed in the unaffected siblings group | Control children were age-matched unaffected siblings | |
Posnack et al. 2012 | In vitro | Rat cardiomyocytes | Di (2-ethylhexyl) phthalate (DEHP) | Mitochondrial mass, gene expression | DEHP exposure resulted in the up-regulation of genes associated with mitochondrial import and an increase in mitochondrial mass | DEHP concentration comparable to clinical exposure (50–100 μg/mL, 72 hr) | |
Rosado-Berrios et al. 2011 | In vitro | Human TK6 lymphoblast cells | DEHP and monoethylhexyl phthalate (MEHP) | Mitochondrial membrane permeability, ROS generation | DEHP and MEHP were associated with increased ROS production and affected the mitochondrial membrane potential; MEHP is more toxic and promotes higher levels of ROS production | The IC50 at 24 hr was approximately 250 μM for both compounds. The longer the exposure time the lower the IC50’s | |
Chen et al. 2010 | In vitro | JAR cells | 2, 3, 7, 8-tetra-chlorodibenzo-p-dioxin (TCDD) | mtDNAcn, mtDNA mutations, ROS production | TCDD resulted in increased oxidative damage and mitochondrial dysfunction; decreases in mtDNAcn, ATP content, and increases in mtDNA deletions were also observed | JAR cells are trophoblast-like cells; placental trophoblast cells are important for maternal-fetal crosstalk[Du et al. 2014] | |
Psychosocial Stress | |||||||
Gimsa et al. 2009 | In vivo | C57BL/6 J mice | Elevated plus-maze test and social defeat | Two strains of mice with different mtDNA variations (AKR/J or FVB/N) | mt(FVB/N) mice showed increased anxiety after the stressor, reduced corticosterone response, and activation of serotonergic and dopaminergic neurotransmitter systems after repeated challenge | This study established a mouse model for studying the role of single nucleotide exchanges in mtDNA in the response to psychological stress | |
Chakravarty et al. 2013 | In vivo | Danio rerio (Zebrafish) | Chronic upredictable stress (CUS); CUS paradigm used daily, involving at least 2 stressors, for 15 days | Mitochondrial proteomics analyses using mass spectrometry | 18 proteins deregulated; 4 out of 18 were mitochondrial proteins (PHB2, SLC25A5, VDAC3, IDH2) | Stressors included restraint stress, social isolation, over-crowding, tank change, cold stress, heat stress, predator stress, dorsal body exposure, and alarm pheromone stress | |
Filipovic et al. 2011 | In vivo | Male Wistar rats | Acute, chronic, or combined stressors | Protein levels of p53 and cytochrome c in mitochondrial and cytosolic fractions; enzyme activity of MnSOD (oxidative stress) in brain tissue | Increased p53 and cytochrome c release into the cytoplasm was observed in the prefrontal cortex, but not the hippocampus, following combined stressors. Decreased mitochondrial MnSOD activity following combined stressors was observed in both brain structures. | Results suggest tissue specific response to stress in the brain | |
Gong et al. 2011 | In vivo | C57 male mice | Chronic mild stress (CMS) | Mitochondrial function and ultrastructure in the mouse brain | CMS inhibited mitochondrial respiration rates and dissipated mitochondrial membrane potential in the cortex, hypothalamus, and hippocampus; damaged mitochondrial structure also observed | Exposure to CMS was a paradigm developed to represent depression in an animal model | |
Song et al. 2009 | In vivo | Sprague-Dawley rats | Prenatal stress | Mitochondria 8-OH-dG levels | 8-OH-dG levels were elevated in female and male prenatal stressed offspring; effects were stronger in female-stressed group | Prenatal stress group exposed to restraint stress during pregnancy 3x daily for 45 min | |
Howerton and Bale 2014 | In vivo | C57BL/6 J male mice; 129S1/SvlmJ female mice | Early prenatal stress | Mitochondrial cytochrome c oxidase activity and gene expression analyses from the hypothalamus | Reduced mitochondrial function in EPS exposed males was observed and verified by cytochrome c oxidase activity assays | Stressed received during days 1–7 of gestation | |
Moller et al. 2013 | In vivo | Male Sprague-Dawley rats | Social isolation rearing (SIR) | Mitochondrial function via ATP release | SIR resulted in increased striatal and decreased frontal cortical accumulation of ATP | SIR in rats is a valid animal model of schizophrenia | |
Nutritional Factors | |||||||
Mortensen et al. 2014 | In vivo | Fischer F344 rats | High fructose diet | Mitochondrial function in brain | High fructose exposed off spring exhibited increased mitochondrial respiration and decreased phosphorylation efficiency | Diet fed during gestation and lactation | |
Ahn et al. 2014 | In vivo | Sprague-Dawley rats-dams treated with valproic acid | Ketogenic diet (KD) | Mitochondrial bioenergetics | Prenatal exposure to VPA altered mitochondrial respiration; this was restored with the KD diet | VPA treatment results in a rodent model of ASD; Pups fed a KD or standard diet for 10 days starting at postnatal day 21 | |
Jousse et al. 2014 | In vivo | Balb/c mice | Low protein diet (LPD) | Mitochondrial-related gene expression, and mtDNAcn in white and brown adipose tissue and skeletal muscle | Gene expression changes were observed in white adipose tissue in LPD mice as was increases in mtDNA content; increased mitochondrial function was also observed in skeletal muscle | LPD contains 10% of protein; control group contains 22% | |
Mitchell et al. 2009 | In vivo | Swiss mice | High, medium, or low protein diets | Mitochondrial function in the embryos | Embryos from HPD females had elevated levels of ROS and ADP concentrations (metabolic stress); embryos from LPD exhibited reduced mitochondrial clustering | ROS, mitochondrial membrane potential and mitochondrial calcium levels assayed | |
Feng et al. 2012 | In vivo | Prenatally stressed rats | Docosahexaenoic acid (DHA) | Mitochondrial metabolism in the hippocampus | Prenatal stress resulted in changes in mitochondrial complexes I–V and enhanced expression of proteins involved in mitochondrial fusion/fission | Rats exposed to restraint stress on days 14–20 of pregnancy | |
Burgueno et al. 2013 | In vivo | Rats | High fat diet (HFD) | mtDNAcn in the liver | HFD during pregnancy associated with decreased liver mtDNAcn | Dams fed HFD for an 18-week period |
Ambient air pollution
Overlapping research suggests a role for mitochondria in the toxicity of ambient air pollution, in part due to the central role that oxidative stress plays in air pollution toxicity (Peretz et al., 2007; Wright and Baccarelli, 2007; Allen et al., 2009; Lodovici and Bigagli, 2011; Wright and Brunst, 2013). Over a decade ago, Li et al. (2003) demonstrated in two murine cell lines that ultrafine particles localize in the mitochondria and induce major structural damage; however, it was not until recently that epidemiological studies started examining the relationship between ambient air pollution and mitochondrial dysfunction. Research now suggests that exposure to air pollution, including particulate matter, benzene, diesel exhaust particles and polycyclic aromatic hydrocarbons (PAHs), is associated with changes in mtDNA copy number in the placenta ( Janssen et al., 2012) and blood (Hou et al., 2010, 2013; Carugno et al., 2012; Byun et al., 2013; Pavanello et al., 2013; Pieters et al., 2013). Diesel exhaust particles and black carbon are also capable of inducing mitochondrial dysfunction in alveolar macrophages (Zhao et al., 2009). More recent work has demonstrated an effect of air pollutants on mtDNA methylation (Byun et al., 2013). Further, air pollutant exposure-related inflammation (Wittkopp et al., 2013) and cognition (Colicino et al., 2014) has been shown to be modified by an individual’s mitochondrial genetic background. These findings suggest mtDNA markers of oxidative damage, copy number, methylation and genetics may serve as novel biomarkers and/or causal mediators in air pollution research.
Tobacco smoke
It has been demonstrated that tobacco smoke is involved in mitochondrial fragmentation, defined as mitochondria shorter than 1 μm without fusion to other mitochondria, in human bronchial epithelial and airway smooth muscle cells and cell senescence (Hara et al., 2013; Aravamudan et al., 2014). Accelerated cellular senescence resulting from tobacco smoke exposure has been implicated in the pathogenesis of asthma (Albrecht et al., 2014). Earlier studies have shown that tobacco smoke can affect mitochondrial structure and function in alveolar macrophages of the lung (Ballinger et al., 1996) and mtDNA changes such as copy number and heteroplasmy in buccal cells (Tan et al., 2008). The effects of early-life tobacco smoke exposure on mitochondrial dysfunction have also been characterized by decreases in mitochondrial antioxidant capacity, mtDNA copy number and mitochondrial potential/mass (Westbrook et al., 2010; Micale et al., 2013). Given these findings, mitochondriomic changes may serve not only a mechanistic function in the pathway from tobacco smoke exposure to disease (e.g. respiratory outcomes), but also as a ’biomarker of exposure’.
Metals
In 2009, Wang et al. (2009) investigated the toxicity of lead exposure on the placenta and found that the placenta of rats that consumed 0.025% lead acetate during gestation were characterized by mitochondria that were swollen, showed distended endoplasmic reticula and a decreased ribosomal number on membranes. Cadmium, another toxic metal, has also been shown to cause mitochondrial dysfunction through reducing ATP production (Kurochkin et al., 2011) and altering mitochondria morphology (Vergilio Cdos and de Melo, 2013). The toxicity of exogenous zinc exposure has been shown to induce mitochondrial dysfunction, elevate the production of ROS and increase expression of adaptive and inflammatory genes (Wu et al., 2013; Zhao et al., 2013). Mercury, methylmercury and manganese have also been shown to induce mitochondrial dysfunction (Mori et al., 2007; Hernandez et al., 2011; Carocci et al., 2014). It is clear that metal exposures have previously been linked to mitochondrial markers of mitochondrial dysfunction; what is less clear is whether metal exposures specifically impact mitochondrial DNA markers such as copy number, heteroplasmy or epigenetics (Hou et al., 2010; Byun et al., 2013). While the amount of literature supporting the use of mtDNA markers for metal exposure is limited, studies have shown exposure to metal-rich particulate to be associated with mtDNA methylation (Byun et al., 2013) and aluminum exposure resulting in decreases in mtDNA copy number and oxidation (Sharma et al., 2013).
Endocrine disruptors (BPA, phthalates)
While environmental endocrine disruption in child health is a rapidly growing area of research, compared with the previously discussed environmental exposures, the literature on endocrine disruptors is less comprehensive with respect to mitochondrial dysfunction. Bisphenol A (BPA) and phthalates are commonly used products in plastics, both with endocrine-disrupting characteristics. BPA has been implicated in mitochondrial dysfunction, particularly by depleting ATP, decreasing mitochondrial mass and dysregulation of membrane potential (Lin et al., 2013) as well as increases in mtDNA copy number in lymphoblasts from children with autism and their unaffected siblings (Kaur et al., 2014). Phthalates have been shown to alter mitochondrial mass in cardiac cells (Posnack et al., 2012) and the mitochondrial membrane potential in immune cells (Rosado-Berrios et al., 2011). Other endocrine disruptors can decrease mtDNA copy number and ATP content, and increase mtDNA deletions in human trophoblast cells; trophoblasts are specialized cells of the placenta and play an important role in the maternal–fetal interface (Chen et al., 2010; Du et al., 2014).
Psychosocial stress
Increasing evidence suggests chronic social stress promotes oxidative stress and thus oxidative damage (Irie et al., 2000; Gidron et al., 2006; Aschbacher et al., 2013; Colaianna et al., 2013; Jorgensen, 2013). For example, women caring for a chronically ill child who endorsed greater perceived stress had higher systemic oxidative stress (Epel et al., 2004). Evidence that social stress and other physical environmental toxins (e.g. ambient air pollutants) may influence common physiological pathways (e.g. oxidative stress) suggests that while not a physical toxin, social stress may also impact mitochondrial functioning, a mechanism that is key to our body’s stress response (Manoli et al., 2007; Gimsa et al., 2009; Song et al., 2009; Filipovic et al., 2011; Gong et al., 2011; Chakravarty et al., 2013; Howerton and Bale, 2014). There are numerous animal studies to support this hypothesis, most pertaining to neurodevelopment. For instance, chronic stress in rats (immobilization for 6 h during 21 days) induces mitochondrial dysfunction in the brain, at least in part due to the overproduction of nitric oxide (NO) via inducible NO synthase (iNOS) (Madrigal et al., 2001). With respect to mtDNA biomarkers, mtDNA variations have been shown to influence anxiety-like behavior and hormone response to stress in mice and zebrafish (Gimsa et al., 2009; Chakravarty et al., 2013). Interestingly, prenatal stress has been linked to mtDNA oxidation and cognitive dysfunction among female rats (Song et al., 2009), which may shed light on gender differences. More recent animal studies also demonstrated associations between early life stress and mitochondrial changes linked to neurodevelopmental deficits (Moller et al., 2013). To date, child health studies of prenatal social stress, mitochondrial dysfunction, and asthma and/or neurodevelopment have not been conducted. The evidence for social stress affecting mitochondrial function in the human lung or placenta is also non-existent, and thus is an area in need of further investigation given the relationship between maternal stress, stress-mechanisms and childhood respiratory symptoms (Wright et al., 2013; Wright and Brunst, 2014).
Nutritional factors
Maternal and postnatal macronutrient consumption of fat, protein, sugar and carbohydrates as well as fatty acids such as docosahexaenoic acid (DHA) have been shown to alter mitochondrial function/content/methylation in multiple organs and tissues (Mitchell et al., 2009; Feng et al., 2012; Burgueno et al., 2013; Jia et al., 2013; Ahn et al., 2014; Jousse et al., 2014; Mortensen et al., 2014). Many studies also support a role for micronutrients such as vitamins B, C, E and polyphenols in regulating mitochondrial function (Depeint et al., 2006; Tannetta et al., 2008; Wang et al., 2014). In 2014, resveratrol, a natural polyphenol in various foods and beverages such as grapes, red wine, peanuts, chocolate and certain berries, was shown to exert antioxidative, anti-inflammatory activities (Birrell et al., 2005; Rahman et al., 2006), significantly decreasing apoptosis by increasing mitochondrial biogenesis, and reducing oxidative stress of regulatory T cells (Tregs) in mice fed a high-fat diet. This is particularly interesting due to the role of Tregs in allergy/asthma development (Nguyen et al., 2008; Robinson, 2009; Brunst et al., 2013). Vitamin B plays a key role in the cellar pathways for methyl transfers (e.g. methyl-donors) which are believed to be required for mitochondrial protein and nucleic acid synthesis (Depeint et al., 2006. Ketogenic diets (high-fat, adequate-protein, low-carbohydrate diets) are able to restore mitochondrial function in the prenatal valproic acid (VPA) rodent model of ASD that exhibits mitochondrial dysfunction (Ahn et al., 2014). Further, maternal DHA intake can prevent prenatal stress-induced impairment in learning and memory and the modulation of mitochondrial metabolism likely plays a key role in DHA protection (Feng et al., 2012). While the evidence suggests a critical role of the mitochondria linking maternal/postnatal diet to chronic disease and child health, to date, there have been no human studies to confirm the usefulness of mitochondrial markers in studies of early-life nutrition (e.g. prenatal or postnatal) and its effects on asthma and/or neurodevelopment.
Co-exposures and cumulative risk
In addition, understanding the potential synergistic effects of physical and social toxins will more completely inform children’s environmental health risk given the shared physiological mechanisms that are at play (Wright, 2009). For instance, children exposed to social stress (community violence) and ambient air pollution (NO2) are at a greater risk of developing asthma (Clougherty et al., 2007). Conversely, socially enriched environments may protect children from the toxic effects of other environmental hazards (Schneider et al., 2001; Laviola et al., 2008). This integrated framework, incorporating mitochondriomics, may have particular implications for prevention and intervention of neurodevelopmental and respiratory outcomes in high-risk children.
New Research and Future Directions
There are numerous opportunities for new research advancements in mitochondriomics from a basic science standpoint including metabolomics, improvement of high-throughput screening, 3D imaging and better sequencing for mtDNA (Shaughnessy et al., 2014). Focused investigations on the crosstalk between exposures and mitochondriomics in environmental health are also needed given mitochondria may represent an environmental target (Meyer et al., 2013). Specifically regarding children’s environmental health, a better understanding of how critical windows of susceptibility impact mitochondrial biomarkers are needed. For instance, increases in mtDNA copy number tend to compensate for mtDNA damage, but this differs by cell type and developmental stage creating potential windows of vulnerability (Carling et al., 2011). Of the studies outlined in this review, only two human studies, focused on children’s environmental health, incorporated mitochondriomics (Wang et al., 2009; Janssen et al., 2012); in total there were only 10 human studies conducted across all exposures examined with the majority being conducted with respect to air pollution. Clearly there is a need to examine the utility and usefulness of incorporating mitochondriomics in more environmental health studies. Not only are toxicological animal studies, which investigate other tissues/cell types including immune and airway epithelial cells to neurons and the placenta needed, but also studies conducted in large-scale pregnancy and birth cohorts.
For some exposures, such as ambient air pollution, it may be more difficult to eliminate exposure. A therapy that could remove mtDNA damage would be advantageous in some instances. For instance, a fertilized nucleus from an oocyte burdened with mtDNA mutations is injected into the cytoplasm of an enucleated donor oocyte carrying mutation free mtDNA; this is known as mitochondrial replacement (MR) and has been successful in producing healthy offspring (Sato et al., 2005; Amato et al., 2014). Could MR gene therapy help women with mitochondrial disease or excessive mitochondrial DNA damage or dysfunction as a result of environmental exposures have healthy children (Reinhardt et al., 2013)? The clinical application of MR is still in its infancy and has many challenges and raises some ethical issues (Amato et al., 2014; Koch, 2014); however, MR could be beneficial when prevention is not an option and thus holds many promises.
Conclusions
Mitochondrial markers have properties that make them exceptionally well suited for biomarker development, as they: (i) have been shown to be persistently altered in blood/tissues by environmental exposures (data derived from mostly animal studies); and (ii) can mark the presence of damaged blood mitochondria, a primary source of systemic oxidative stress to which the brain and lungs are particularly vulnerable. Further work is needed to determine the utility of mitochondrial markers with respect to children’s environmental health. However, it is possible that mitochondrial markers can serve as exposure biomarkers; biomarkers of child health outcomes (i.e. asthma/neurodevelopment); and/or biomarkers of child health outcomes specific to environmental exposure effects.
References
- Ahn Y, Narous M, Tobias R, Rho JM, Mychasiuk R. The ketogenic diet modifies social and metabolic alterations identified in the prenatal valproic acid model of autism spectrum disorder. Dev Neurosci. 2014;36:371–380. doi: 10.1159/000362645. [DOI] [PubMed] [Google Scholar]
- Albrecht E, Sillanpää E, Karrasch S, Alves AC, Codd V, Hovatta I, Buxton JL, Nelson CP, Broer L, Hägg S, Mangino M, Willemsen G, Surakka I, Ferreira MA, Amin N, Oostra BA, Bäckmand HM, Peltonen M, Sarna S, Rantanen T, Sipilä S, Korhonen T, Madden PA, Gieger C, Jörres RA, Heinrich J, Behr J, Huber RM, Peters A, Strauch K, Wichmann HE, Waldenberger M, Blakemore AI, de Geus EJ, Nyholt DR, Henders AK, Piirilä PL, Rissanen A, Magnusson PK, Viñuela A, Pietiläinen KH, Martin NG, Pedersen NL, Boomsma DI, Spector TD, van Duijn CM, Kaprio J, Samani NJ, Jarvelin MR, Schulz H. Telomere length in circulating leukocytes is associated with lung function and disease. Eur Respir J. 2014;43:983–992. doi: 10.1183/09031936.00046213. [DOI] [PubMed] [Google Scholar]
- Allen J, Trenga CA, Peretz A, Sullivan JH, Carlsten CC, Kaufman JD. Effect of diesel exhaust inhalation on antioxidant and oxidative stress responses in adults with metabolic syndrome. Inhal Toxicol. 2009;21:1061–1067. doi: 10.3109/08958370902721424. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Amato P, Tachibana M, Sparman M, Mitalipov S. Three-parent in vitro fertilization: Gene replacement for the prevention of inherited mitochondrial diseases. Fertil Steril. 2014;101:31–35. doi: 10.1016/j.fertnstert.2013.11.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Anandan C, Nurmatov U, van Schayck OC, Sheikh A. Is the prevalence of asthma declining? Systematic review of epidemiological studies. Allergy. 2010;65:152–167. doi: 10.1111/j.1398-9995.2009.02244.x. [DOI] [PubMed] [Google Scholar]
- Anitha A, Nakamura K, Thanseem I, Matsuzaki H, Miyachi T, Tsujii M, Iwata Y, Suzuki K, Sugiyama T, Mori N. Downregulation of the expression of mitochondrial electron transport complex genes in autism brains. Brain Pathol. 2013;23:294–302. doi: 10.1111/bpa.12002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Aravamudan B, Kiel A, Freeman M, Delmotte P, Thompson M, Vassallo R, Sieck GC, Pabelick CM, Prakash YS. Cigarette smoke-induced mitochondrial fragmentation and dysfunction in human airway smooth muscle. Am J Physiol Lung Cell Mol Physiol. 2014;306:L840–854. doi: 10.1152/ajplung.00155.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Aschbacher K, O’Donovan A, Wolkowitz OM, Dhabhar FS, Su Y, Epel E. Good stress, bad stress and oxidative stress: Insights from anticipatory cortisol reactivity. Psychoneuroendocrinology. 2013;38:1698–1708. doi: 10.1016/j.psyneuen.2013.02.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ballinger SW, Bouder TG, Davis GS, Judice SA, Nicklas JA, Albertini RJ. Mitochondrial genome damage associated with cigarette smoking. Cancer Res. 1996;56:5692–5697. [PubMed] [Google Scholar]
- Barrett EG. Maternal influence in the transmission of asthma susceptibility. Pulm Pharmacol Ther. 2008;21:474–484. doi: 10.1016/j.pupt.2007.06.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Barrey E, Saint-Auret G, Bonnamy B, Damas D, Boyer O, Gidrol X. Pre-microrna and mature microrna in human mitochondria. PLoS One. 2011;6:e20220. doi: 10.1371/journal.pone.0020220. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bax BE, Bloxam DL. Energy metabolism and glycolysis in human placental trophoblast cells during differentiation. Biochim Biophys Acta. 1997;1319:283–292. doi: 10.1016/s0005-2728(96)00169-7. [DOI] [PubMed] [Google Scholar]
- Bellizzi D, D’Aquila P, Scafone T, Giordano M, Riso V, Riccio A, Passarino G. The control region of mitochondrial DNA shows an unusual cpg and non-cpg methylation pattern. DNA Res. 2013;20:537–547. doi: 10.1093/dnares/dst029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Birrell MA, McCluskie K, Wong S, Donnelly LE, Barnes PJ, Belvisi MG. Resveratrol, an extract of red wine, inhibits lipopolysaccharide induced airway neutrophilia and inflammatory mediators through an nf-kappab-independent mechanism. FASEB J. 2005;19:840–841. doi: 10.1096/fj.04-2691fje. [DOI] [PubMed] [Google Scholar]
- Boulet SL, Boyle CA, Schieve LA. Health care use and health and functional impact of developmental disabilities among us children, 1997–2005. Arch Pediatr Adolesc Med. 2009;163:19–26. doi: 10.1001/archpediatrics.2008.506. [DOI] [PubMed] [Google Scholar]
- Bradstreet JJ, Smith S, Baral M, Rossignol DA. Biomarker-guided interventions of clinically relevant conditions associated with autism spectrum disorders and attention deficit hyperactivity disorder. Altern Med Rev. 2010;15:15–32. [PubMed] [Google Scholar]
- Brunst KJ, Leung YK, Ryan PH, Khurana Hershey GK, Levin L, Ji H, Lemasters GK, Ho SM. Forkhead box protein 3 (foxp3) hypermethylation is associated with diesel exhaust exposure and risk for childhood asthma. J Allergy Clin Immunol. 2013;131:592–594. e591–593. doi: 10.1016/j.jaci.2012.10.042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Burgueno AL, Cabrerizo R, Gonzales Mansilla N, Sookoian S, Pirola CJ. Maternal high-fat intake during pregnancy programs metabolic-syndrome-related phenotypes through liver mitochondrial DNA copy number and transcriptional activity of liver ppargc1a. J Nutr Biochem. 2013;24:6–13. doi: 10.1016/j.jnutbio.2011.12.008. [DOI] [PubMed] [Google Scholar]
- Burton GJ, Jauniaux E. Oxidative stress. Best Pract Res Clin Obstet Gynaecol. 2011;25:287–299. doi: 10.1016/j.bpobgyn.2010.10.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Byun HM, Panni T, Motta V, Hou L, Nordio F, Apostoli P, Bertazzi PA, Baccarelli AA. Effects of airborne pollutants on mitochondrial DNA methylation. Part Fibre Toxicol. 2013;10:18. doi: 10.1186/1743-8977-10-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Byun HM, Baccarelli AA. Environmental exposure and mitochondrial epigenetics: Study design and analytical challenges. Hum Genet. 2014;133:247–257. doi: 10.1007/s00439-013-1417-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Carling PJ, Cree LM, Chinnery PF. The implications of mitochondrial DNA copy number regulation during embryogenesis. Mitochondrion. 2011;11:686–692. doi: 10.1016/j.mito.2011.05.004. [DOI] [PubMed] [Google Scholar]
- Carocci A, Rovito N, Sinicropi MS, Genchi G. Mercury toxicity and neurodegenerative effects. Rev Environ Contam Toxicol. 2014;229:1–18. doi: 10.1007/978-3-319-03777-6_1. [DOI] [PubMed] [Google Scholar]
- Carugno M, Pesatori AC, Dioni L, Hoxha M, Bollati V, Albetti B, Byun HM, Bonzini M, Fustinoni S, Cocco P, Satta G, Zucca M, Merlo DF, Cipolla M, Bertazzi PA, Baccarelli A. Increased mitochondrial DNA copy number in occupations associated with low-dose benzene exposure. Environ Health Perspect. 2012;120:210–215. doi: 10.1289/ehp.1103979. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Celedon JC, Wright RJ, Litonjua AA, Sredl D, Ryan L, Weiss ST, Gold DR. Day care attendance in early life, maternal history of asthma, and asthma at the age of 6 years. Am J Respir Crit Care Med. 2003;167:1239–1243. doi: 10.1164/rccm.200209-1063OC. [DOI] [PubMed] [Google Scholar]
- Chakravarty S, Reddy BR, Sudhakar SR, Saxena S, Das T, Meghah V, Brahmendra Swamy CV, Kumar A, Idris MM. Chronic unpredictable stress (cus)-induced anxiety and related mood disorders in a zebrafish model: Altered brain proteome profile implicates mitochondrial dysfunction. PLoS One. 2013;8:e63302. doi: 10.1371/journal.pone.0063302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chan SW, Chevalier S, Aprikian A, Chen JZ. Simultaneous quantification of mitochondrial DNA damage and copy number in circulating blood: A sensitive approach to systemic oxidative stress. Biomed Res Int. 2013;2013:157547. doi: 10.1155/2013/157547. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen SC, Liao TL, Wei YH, Tzeng CR, Kao SH. Endocrine disruptor, dioxin (tcdd)-induced mitochondrial dysfunction and apoptosis in human trophoblast-like jar cells. Mol Hum Reprod. 2010;16:361–372. doi: 10.1093/molehr/gaq004. [DOI] [PubMed] [Google Scholar]
- Chinnery PF, Elliott HR, Hudson G, Samuels DC, Relton CL. Epigenetics, epidemiology and mitochondrial DNA diseases. Int J Epidemiol. 2012;41:177–187. doi: 10.1093/ije/dyr232. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chodaczek G, Bacsi A, Dharajiya N, Sur S, Hazra TK, Boldogh I. Ragweed pollen-mediated ige-independent release of biogenic amines from mast cells via induction of mitochondrial dysfunction. Mol Immunol. 2009;46:2505–2514. doi: 10.1016/j.molimm.2009.05.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Clougherty J, Levy JI, Kubzansky LD, Ryan B, Franco Suglia S, Jacobson Canner M, Wright RJ. The effects of traffic-related air pollution and exposure to violence on urban asthma etiology. Environ Health Perspect. 2007;115:1140–1146. doi: 10.1289/ehp.9863. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Colaianna M, Schiavone S, Zotti M, Tucci P, Morgese MG, Backdahl L, Holmdahl R, Krause KH, Cuomo V, Trabace L. Neuroendocrine profile in a rat model of psychosocial stress: Relation to oxidative stress. Antioxid Redox Signal. 2013;18:1385–1399. doi: 10.1089/ars.2012.4569. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Colicino E, Power MC, Cox DG, Weisskopf MG, Hou L, Alexeeff SE, Sanchez-Guerra M, Vokonas P, Spiro A, 3rd, Schwartz J, Baccarelli AA. Mitochondrial haplogroups modify the effect of black carbon on age-related cognitive impairment. Environ Health. 2014;13:42. doi: 10.1186/1476-069X-13-42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Correia C, Coutinho AM, Diogo L, Grazina M, Marques C, Miguel T, Ataíde A, Almeida J, Borges L, Oliveira C, Oliveira G, Vicente AM. Brief report: High frequency of biochemical markers for mitochondrial dysfunction in autism: No association with the mitochondrial aspartate/glutamate carrier slc25a12 gene. J Autism Dev Disord. 2006;36:1137–1140. doi: 10.1007/s10803-006-0138-6. [DOI] [PubMed] [Google Scholar]
- Crouch PJ, Cimdins K, Duce JA, Bush AI, Trounce IA. Mitochondria in aging and alzheimer’s disease. Rejuvenation Res. 2007;10:349–357. doi: 10.1089/rej.2007.0592. [DOI] [PubMed] [Google Scholar]
- Depeint F, Bruce WR, Shangari N, Mehta R, O’Brien PJ. Mitochondrial function and toxicity: Role of b vitamins on the one-carbon transfer pathways. Chem Biol Interact. 2006;163:113–132. doi: 10.1016/j.cbi.2006.05.010. [DOI] [PubMed] [Google Scholar]
- Du MR, Wang SC, Li DJ. The integrative roles of chemokines at the maternal-fetal interface in early pregnancy. Cell Mol Immunol. 2014;11:438–448. doi: 10.1038/cmi.2014.68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Epel ES, Blackburn EH, Lin J, Dhabhar FS, Adler NE, Morrow JD, Cawthon RM. Accelerated telomere shortening in response to life stress. Proc Natl Acad Sci U S A. 2004;101:17312–17315. doi: 10.1073/pnas.0407162101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fagundes AO, Rezin GT, Zanette F, Grandi E, Assis LC, Dal-Pizzol F, Quevedo J, Streck EL. Chronic administration of methylpheni-date activates mitochondrial respiratory chain in brain of young rats. Int J Dev Neurosci. 2007;25:47–51. doi: 10.1016/j.ijdevneu.2006.11.001. [DOI] [PubMed] [Google Scholar]
- Felton JM, Lucas CD, Rossi AG, Dransfield I. Eosinophils in the lung-modulating apoptosis and efferocytosis in airway inflammation. Front Immunol. 2014;5:302. doi: 10.3389/fimmu.2014.00302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Feng Z, Zou X, Jia H, Li X, Zhu Z, Liu X, Bucheli P, Ballevre 0, Hou Y, Zhang W, Wang J, Chen Y, Liu J. Maternal docosahexaenoic acid feeding protects against impairment of learning and memory and oxidative stress in prenatally stressed rats: Possible role of neuronal mitochondria metabolism. Antioxid Redox Signal. 2012;16:275–289. doi: 10.1089/ars.2010.3750. [DOI] [PubMed] [Google Scholar]
- Filipovic D, Zlatkovic J, Inta D, Bjelobaba I, Stojiljkovic M, Gass P. Chronic isolation stress predisposes the frontal cortex but not the hippocampus to the potentially detrimental release of cytochrome c from mitochondria and the activation of caspase-3. J Neurosci Res. 2011;89:1461–1470. doi: 10.1002/jnr.22687. [DOI] [PubMed] [Google Scholar]
- Flaquer A, Heinzmann A, Rospleszcz S, Mailaparambil B, Dietrich H, Strauch K, Grychtol R. Association study of mitochondrial genetic polymorphisms in asthmatic children. Mitochondrion. 2014;14:49–53. doi: 10.1016/j.mito.2013.11.002. [DOI] [PubMed] [Google Scholar]
- Fleischman A, Kron M, Systrom DM, Hrovat M, Grinspoon SK. Mitochondrial function and insulin resistance in overweight and normal-weight children. J Clin Endocrinol Metab. 2009;94:4923–4930. doi: 10.1210/jc.2009-1590. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Froehlich TE, Lanphear BP, Epstein JN, Barbaresi WJ, Katusic SK, Kahn RS. Prevalence, recognition, and treatment of attention-deficit/ hyperactivity disorder in a national sample of us children. Arch Pediatr Adolesc Med. 2007;161:857–864. doi: 10.1001/archpedi.161.9.857. [DOI] [PubMed] [Google Scholar]
- Gidron Y, Russ K, Tissarchondou H, Warner J. The relation between psychological factors and DNA-damage: A critical review. Biol Psychol. 2006;72:291–304. doi: 10.1016/j.biopsycho.2005.11.011. [DOI] [PubMed] [Google Scholar]
- Giles RE, Blanc H, Cann HM, Wallace DC. Maternal inheritance of human mitochondrial DNA. Proc Natl Acad Sci U S A. 1980;77:6715–6719. doi: 10.1073/pnas.77.11.6715. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gimsa U, Kanitz E, Otten W, Ibrahim SM. Behavior and stress reactivity in mouse strains with mitochondrial DNA variations. Ann N Y Acad Sci. 2009;1153:131–138. doi: 10.1111/j.1749-6632.2008.03960.x. [DOI] [PubMed] [Google Scholar]
- Giulivi C, Zhang YF, Omanska-Klusek A, Ross-Inta C, Wong S, Hertz-Picciotto I, Tassone F, Pessah IN. Mitochondrial dysfunction in autism. JAMA. 2010;304:2389–2396. doi: 10.1001/jama.2010.1706. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Golden TR, Melov S. Mitochondrial DNA mutations, oxidative stress, and aging. Mech Ageing Dev. 2001;122:1577–1589. doi: 10.1016/s0047-6374(01)00288-3. [DOI] [PubMed] [Google Scholar]
- Gong Y, Chai Y, Ding JH, Sun XL, Hu G. Chronic mild stress damages mitochondrial ultrastructure and function in mouse brain. Neurosci Lett. 2011;488:76–80. doi: 10.1016/j.neulet.2010.11.006. [DOI] [PubMed] [Google Scholar]
- Graf WD, Marin-Garcia J, Gao HG, Pizzo S, Naviaux RK, Markusic D, Barshop BA, Courchesne E, Haas RH. Autism associated with the mitochondrial DNA g8363a transfer rna(lys) mutation. J Child Neurol. 2000;15:357–361. doi: 10.1177/088307380001500601. [DOI] [PubMed] [Google Scholar]
- Graziewicz MA, Day BJ, Copeland WC. The mitochondrial DNA polymerase as a target of oxidative damage. Nucleic Acids Res. 2002;30:2817–2824. doi: 10.1093/nar/gkf392. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Grivennikova VG, Kareyeva AV, Vinogradov AD. What are the sources of hydrogen peroxide production by heart mitochondria? Biochim Biophys Acta. 2010;1797:939–944. doi: 10.1016/j.bbabio.2010.02.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hagberg H, Mallard C, Rousset CI, Thornton C. Mitochondria: Hub of injury responses in the developing brain. Lancet Neurol. 2014;13:217–232. doi: 10.1016/S1474-4422(13)70261-8. [DOI] [PubMed] [Google Scholar]
- Hara H, Araya J, Ito S, Kobayashi K, Takasaka N, Yoshii Y, Wakui H, Kojima J, Shimizu K, Numata T, Kawaishi M, Kamiya N, Odaka M, Morikawa T, Kaneko Y, Nakayama K, Kuwano K. Mitochondrial fragmentation in cigarette smoke-induced bronchial epithelial cell senescence. Am J Physiol Lung Cell Mol Physiol. 2013;305:L737–746. doi: 10.1152/ajplung.00146.2013. [DOI] [PubMed] [Google Scholar]
- Heinzmann A, Thoma C, Dietrich H, Deichmann KA. Identification of common polymorphisms in the mitochondrial genome. Allergy. 2003;58:830–831. doi: 10.1034/j.1398-9995.2003.00223.x. [DOI] [PubMed] [Google Scholar]
- Hernandez-Aguilera A, Rull A, Rodriguez-Gallego E, Riera-Borrull M, Luciano-Mateo F, Camps J, Menéndez JA, Joven J. Mitochondrial dysfunction: A basic mechanism in inflammation-related non-communicable diseases and therapeutic opportunities. Mediators Inflamm. 2013;2013:135698. doi: 10.1155/2013/135698. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hernandez RB, Farina M, Esposito BP, Souza-Pinto NC, Barbosa F, Jr, Sunol C. Mechanisms of manganese-induced neurotoxicity in primary neuronal cultures: The role of manganese speciation and cell type. Toxicol Sci. 2011;124:414–423. doi: 10.1093/toxsci/kfr234. [DOI] [PubMed] [Google Scholar]
- Holgate ST. Asthma: A dynamic disease of inflammation and repair. In: Chadwick DJ, Cardew G, editors. The Rising Trends in Asthma. John Wiley & Sons; West Sussex: 1997. pp. 5–34. [DOI] [PubMed] [Google Scholar]
- Holt PG, Upham JW, Sly PD. Contemporaneous maturation of immunologic and respiratory functions during early childhood: Implications for development of asthma prevention strategies. J Allergy Clin Immunol. 2005;116:16–24. doi: 10.1016/j.jaci.2005.04.017. quiz 25. [DOI] [PubMed] [Google Scholar]
- Hong EE, Okitsu CY, Smith AD, Hsieh CL. Regionally specific and genome-wide analyses conclusively demonstrate the absence of cpg methylation in human mitochondrial DNA. Mol Cell Biol. 2013;33:2683–2690. doi: 10.1128/MCB.00220-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hou L, Zhu ZZ, Zhang X, Nordio F, Bonzini M, Schwartz J, Hoxha M, Dioni L, Marinelli B, Pegoraro V, Apostoli P, Bertazzi PA, Baccarelli A. Airborne particulate matter and mitochondrial damage: A cross-sectional study. Environ Health. 2010;9:48. doi: 10.1186/1476-069X-9-48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hou L, Zhang X, Dioni L, Barretta F, Dou C, Zheng Y, Hoxha M, Bertazzi PA, Schwartz J, Wu S, Wang S, Baccarelli AA. Inhalable particulate matter and mitochondrial DNA copy number in highly exposed individuals in beijing, china: A repeated-measure study. Part Fibre Toxicol. 2013;10:17. doi: 10.1186/1743-8977-10-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Howerton CL, Bale TL. Targeted placental deletion of ogt recapitulates the prenatal stress phenotype including hypothalamic mitochondrial dysfunction. Proc Natl Acad Sci U S A. 2014;111:9639–9644. doi: 10.1073/pnas.1401203111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ilmarinen P, Moilanen E, Kankaanranta H. Mitochondria in the center of human eosinophil apoptosis and survival. Int J Mol Sci. 2014;15:3952–3969. doi: 10.3390/ijms15033952. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Irie M, Asami S, Nagata S, Miyata M, Kasai H. Classical conditioning of oxidative DNA damage in rats. Neurosci Lett. 2000;288:13–16. doi: 10.1016/s0304-3940(00)01194-0. [DOI] [PubMed] [Google Scholar]
- Janssen BG, Munters E, Pieters N, Smeets K, Cox B, Cuypers A, Fierens F, Penders J, Vangronsveld J, Gyselaers W, Nawrot TS. Placental mitochondrial DNA content and particulate air pollution during in utero life. Environ Health Perspect. 2012;120:1346–1352. doi: 10.1289/ehp.1104458. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jia Y, Li R, Cong R, Yang X, Sun Q, Parvizi N, Zhao R. Maternal low-protein diet affects epigenetic regulation of hepatic mitochondrial DNA transcription in a sex-specific manner in newborn piglets associated with gr binding to its promoter. PLoS One. 2013;8:e63855. doi: 10.1371/journal.pone.0063855. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jorgensen A. Oxidatively generated DNA/rna damage in psychological stress states. Dan Med J. 2013;60:B4685. [PubMed] [Google Scholar]
- Jousse C, Muranishi Y, Parry L, Montaurier C, Even P, Launay JM, Carraro V, Maurin AC, Averous J, Chaveroux C, Bruhat A, Mallet J, Morio B, Fafournoux P. Perinatal protein malnutrition affects mitochondrial function in adult and results in a resistance to high fat diet-induced obesity. PLoS One. 2014;9:e104896. doi: 10.1371/journal.pone.0104896. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kaur K, Chauhan V, Gu F, Chauhan A. Bisphenol a induces oxidative stress and mitochondrial dysfunction in lymphoblasts from children with autism and unaffected siblings. Free Radic Biol Med. 2014;76C:25–33. doi: 10.1016/j.freeradbiomed.2014.07.030. [DOI] [PubMed] [Google Scholar]
- Killeen PR, Russell VA, Sergeant JA. A behavioral neuroenergetics theory of adhd. Neurosci Biobehav Rev. 2013;37:625–657. doi: 10.1016/j.neubiorev.2013.02.011. [DOI] [PubMed] [Google Scholar]
- Knoll N, Jarick I, Volckmar AL, Klingenspor M, Illig T, Grallert H, Gieger C, Wichmann HE, Peters A, Hebebrand J, Scherag A, Hinney A. Gene set of nuclear-encoded mitochondrial regulators is enriched for common inherited variation in obesity. PLoS One. 2013;8:e55884. doi: 10.1371/journal.pone.0055884. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Koch L. Clinical genetics: Mitochondrial replacement techniques under the spotlight. Nat Rev Genet. 2014;15:516. doi: 10.1038/nrg3784. [DOI] [PubMed] [Google Scholar]
- Kudriashova IB, Kirnos MD, Vaniushin BF. DNA-methylase activities from animal mitochondria and nuclei: Different specificity of DNA methylation. Biokhimiia. 1976;41:1968–1977. [PubMed] [Google Scholar]
- Kurochkin IO, Etzkorn M, Buchwalter D, Leamy L, Sokolova IM. Top-down control analysis of the cadmium effects on molluscan mitochondria and the mechanisms of cadmium-induced mitochondrial dysfunction. Am J Physiol Regul Integr Comp Physiol. 2011;300:R21–31. doi: 10.1152/ajpregu.00279.2010. [DOI] [PubMed] [Google Scholar]
- Landrigan PL, Claudio L, Markowitz SB, Berkowitz GS, Brenner BL, Romero H, Wetmur JG, Matte TD, Gore AC, Godbold JH, Wolff MS. Pesticides and inner-city children: Exposures, risks, and prevention. Environ Health Perspect. 1999;107:431–437. doi: 10.1289/ehp.99107s3431. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Laviola G, Hannan AJ, Macri S, Solinas M, Jaber M. Effects of enriched environment on animal models of neurodegenerative diseases and psychiatric disorders. Neurobiol Dis. 2008;31:159–168. doi: 10.1016/j.nbd.2008.05.001. [DOI] [PubMed] [Google Scholar]
- Leduc L, Levy E, Bouity-Voubou M, Delvin E. Fetal programming of atherosclerosis: Possible role of the mitochondria. Eur J Obstet Gynecol Reprod Biol. 2010;149:127–130. doi: 10.1016/j.ejogrb.2009.12.005. [DOI] [PubMed] [Google Scholar]
- Lee HC, Wei YH. Mitochondrial role in life and death of the cell. J Biomed Sci. 2000;7:2–15. doi: 10.1007/BF02255913. [DOI] [PubMed] [Google Scholar]
- Legido A, Jethva R, Goldenthal MJ. Mitochondrial dysfunction in autism. Semin Pediatr Neurol. 2013;20:163–175. doi: 10.1016/j.spen.2013.10.008. [DOI] [PubMed] [Google Scholar]
- Li H, Liu D, Lu J, Bai Y. In: Advances in Mitochondrial Medicine, Advances in Experimental Medicine and Biology. Scatena R, Bottoni P, Giardina B, editors. Springer Science + Business Media; B.V. New York, NY: 2012. [Google Scholar]
- Li N, Sioutas C, Cho A, Schmitz D, Misra C, Sempf J, Wang M, Oberley T, Froines J, Nel A. Ultrafine particulate pollutants induce oxidative stress and mitochondrial damage. Environ Health Perspect. 2003;111:455–460. doi: 10.1289/ehp.6000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lim RH, Kobzik L, Dahl M. Risk for asthma in offspring of asthmatic mothers versus fathers: A meta-analysis. PLoS One. 2010;5:e10134. doi: 10.1371/journal.pone.0010134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lin CS, Wang LS, Tsai CM, Wei YH. Low copy number and low oxidative damage of mitochondrial DNA are associated with tumor progression in lung cancer tissues after neoadjuvant chemotherapy. Interact Cardiovasc Thorac Surg. 2008;7:954–958. doi: 10.1510/icvts.2008.177006. [DOI] [PubMed] [Google Scholar]
- Lin Y, Sun X, Qiu L, Wei J, Huang Q, Fang C, Ye T, Kang M, Shen H, Dong S. Exposure to bisphenol a induces dysfunction of insulin secretion and apoptosis through the damage of mitochondria in rat insulinoma (ins-1) cells. Cell Death Dis. 2013;4:e460. doi: 10.1038/cddis.2012.206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Litonjua AA, Carey VJ, Burge HA, Weiss ST, Gold DR. Parental history and the risk for childhood asthma. Does mother confer more risk than father? Am J Respir Crit Care Med. 1998;158:176–181. doi: 10.1164/ajrccm.158.1.9710014. [DOI] [PubMed] [Google Scholar]
- Liu CS, Cheng WL, Lee CF, Ma YS, Lin CY, Huang CC, Wei YH. Alteration in the copy number of mitochondrial DNA in leukocytes of patients with mitochondrial encephalomyopathies. Acta Neurol Scand. 2006;113:334–341. doi: 10.1111/j.1600-0404.2006.00586.x. [DOI] [PubMed] [Google Scholar]
- Lodovici M, Bigagli E. Oxidative stress and air pollution exposure. J Toxicol. 2011;2011:487074. doi: 10.1155/2011/487074. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Madrigal JL, Olivenza R, Moro MA, Lizasoain I, Lorenzo P, Rodrigo J, Leza JC. Glutathione depletion, lipid peroxidation and mitochondrial dysfunction are induced by chronic stress in rat brain. Neuropsychopharmacology. 2001;24:420–429. doi: 10.1016/S0893-133X(00)00208-6. [DOI] [PubMed] [Google Scholar]
- Mandò C, De Palma C, Stampalija T, Anelli GM, Figus M, Novielli C, Parisi F, Clementi E, Ferrazzi E, Cetin I. Placental mitochondrial content and function in intrauterine growth restriction and preeclampsia. Am J Physiol Endocrinol Metab. 2014;306:E404–413. doi: 10.1152/ajpendo.00426.2013. [DOI] [PubMed] [Google Scholar]
- Manoli I, Alesci S, Blackman MR, Su YA, Rennert OM, Chrousos GP. Mitochondria as key components of the stress response. Trends Endocrinol Metab. 2007;18:190–198. doi: 10.1016/j.tem.2007.04.004. [DOI] [PubMed] [Google Scholar]
- Martinez FD, Wright AL, Taussig LM, Holberg CJ, Halonen M, Morgan WJ. Asthma and wheezing in the first six years of life. The group health medical associates. N Engl J Med. 1995;332:133–138. doi: 10.1056/NEJM199501193320301. [DOI] [PubMed] [Google Scholar]
- Mattson MP, Gleichmann M, Cheng A. Mitochondria in neuroplasticity and neurological disorders. Neuron. 2008;60:748–766. doi: 10.1016/j.neuron.2008.10.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mayeur S, Lancel S, Theys N, Lukaszewski MA, Duban-Deweer S, Bastide B, Hachani J, Cecchelli R, Breton C, Gabory A, Storme L, Reusens B, Junien C, Vieau D, Lesage J. Maternal calorie restriction modulates placental mitochondrial biogenesis and bioenergetic efficiency: Putative involvement in fetoplacental growth defects in rats. Am J Physiol Endocrinol Metab. 2013;304:E14–22. doi: 10.1152/ajpendo.00332.2012. [DOI] [PubMed] [Google Scholar]
- Mecocci P, Polidori MC, Ingegni T, Cherubini A, Chionne F, Cecchetti R, Senin U. Oxidative damage to DNA in lymphocytes from ad patients. Neurology. 1998;51:1014–1017. doi: 10.1212/wnl.51.4.1014. [DOI] [PubMed] [Google Scholar]
- Meert KL, McCaulley L, Sarnaik AP. Mechanism of lactic acidosis in children with acute severe asthma. Pediatr Crit Care Med. 2012;13:28–31. doi: 10.1097/PCC.0b013e3182196aa2. [DOI] [PubMed] [Google Scholar]
- Mele J, Muralimanoharan S, Maloyan A, Myatt L. Impaired mitochondrial function in human placenta with increased maternal adiposity. Am J Physiol Endocrinol Metab. 2014;307:E419–425. doi: 10.1152/ajpendo.00025.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mercer TR, Neph S, Dinger ME, Crawford J, Smith MA, Shearwood AM, Haugen E, Bracken CP, Rackham O, Stamatoyannopoulos JA, Filipovska A, Mattick JS. The human mitochondrial transcriptome. Cell. 2011;146:645–658. doi: 10.1016/j.cell.2011.06.051. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Meyer JN, Leung MC, Rooney JP, Sendoel A, Hengartner MO, Kisby GE, Bess AS. Mitochondria as a target of environmental toxicants. Toxicol Sci. 2013;134:1–17. doi: 10.1093/toxsci/kft102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Micale RT, La Maestra S, Di Pietro A, Visalli G, Baluce B, Balansky R, Steele VE, De Flora S. Oxidative stress in the lung of mice exposed to cigarette smoke either early in life or in adulthood. Arch Toxicol. 2013;87:915–918. doi: 10.1007/s00204-012-0993-1. [DOI] [PubMed] [Google Scholar]
- Mitchell M, Schulz SL, Armstrong DT, Lane M. Metabolic and mitochondrial dysfunction in early mouse embryos following maternal dietary protein intervention. Biol Reprod. 2009;80:622–630. doi: 10.1095/biolreprod.108.072595. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Moller M, Du Preez JL, Viljoen FP, Berk M, Emsley R, Harvey BH. Social isolation rearing induces mitochondrial, immunological, neurochemical and behavioural deficits in rats, and is reversed by clozapine or n-acetyl cysteine. Brain Behav Immun. 2013;30:156–167. doi: 10.1016/j.bbi.2012.12.011. [DOI] [PubMed] [Google Scholar]
- Montes G, Lotyczewski BS, Halterman JS, Hightower AD. School readiness among children with behavior problems at entrance into kindergarten: Results from a us national study. Eur J Pediatr. 2012;171:541–548. doi: 10.1007/s00431-011-1605-4. [DOI] [PubMed] [Google Scholar]
- Morgan WJ, Stern DA, Sherrill DL, Guerra S, Holberg CJ, Guilbert TW, Taussig LM, Wright AL, Martinez FD. Outcome of asthma and wheezing in the first 6 years of life: Follow-up through adolescence. Am J Respir Crit Care Med. 2005;172:1253–1258. doi: 10.1164/rccm.200504-525OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mori N, Yasutake A, Hirayama K. Comparative study of activities in reactive oxygen species production/defense system in mitochondria of rat brain and liver, and their susceptibility to methylmercury toxicity. Arch Toxicol. 2007;81:769–776. doi: 10.1007/s00204-007-0209-2. [DOI] [PubMed] [Google Scholar]
- Mortensen OH, Larsen LH, Orstrup LK, Hansen LH, Grunnet N, Quistorff B. Developmental programming by high fructose decreases phosphorylation efficiency in aging offspring brain mitochondria, correlating with enhanced ucp5 expression. J Cereb Blood Flow Metab. 2014;34:1205–1211. doi: 10.1038/jcbfm.2014.72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Motorin Y, Helm M. Rna nucleotide methylation. Wiley Interdiscip Rev RNA. 2011;2:611–631. doi: 10.1002/wrna.79. [DOI] [PubMed] [Google Scholar]
- Napoli E, Wong S, Giulivi C. Evidence of reactive oxygen species-mediated damage to mitochondrial DNA in children with typical autism. Mol Autism. 2013;4:2. doi: 10.1186/2040-2392-4-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nguyen KD, Fohner A, Booker JD, Dong C, Krensky AM, Nadeau KC. Xcl1 enhances regulatory activities of cd4+ cd25(high) cd127(low/-) t cells in human allergic asthma. J Immunol. 2008;181:5386–5395. doi: 10.4049/jimmunol.181.8.5386. [DOI] [PMC free article] [PubMed] [Google Scholar]
- O’Connor CM, Adams JU. Essentials of Cell Biology. NPG Education; Cambridge, MA: 2010. [Google Scholar]
- Oguzkan-Balci S, Col-Araz N, Nacak M, Araz M, Sabanci H, Balat A, Pehlivan S. Mitochondrial uncoupling protein 2 (ucp2) gene polymorphisms are associated with childhood obesity and related metabolic disorders. J Pediatr Endocrinol Metab. 2013;26:277–283. doi: 10.1515/jpem-2012-0267. [DOI] [PubMed] [Google Scholar]
- Oliveira G, Diogo L, Grazina M, Garcia P, Ataide A, Marques C, Miguel T, Borges L, Vicente AM, Oliveira CR. Mitochondrial dysfunction in autism spectrum disorders: A population-based study. Dev Med Child Neurol. 2005;47:185–189. doi: 10.1017/s0012162205000332. [DOI] [PubMed] [Google Scholar]
- Ostroukhova M, Goplen N, Karim MZ, Michalec L, Guo L, Liang Q, Alam R. The role of low-level lactate production in airway inflammation in asthma. Am J Physiol Lung Cell Mol Physiol. 2012;302:L300–307. doi: 10.1152/ajplung.00221.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Palmieri L, Persico AM. Mitochondrial dysfunction in autism spectrum disorders: Cause or effect? Biochim Biophys Acta. 2010;1797:1130–1137. doi: 10.1016/j.bbabio.2010.04.018. [DOI] [PubMed] [Google Scholar]
- Papa M, Sellitti S, Sadile AG. Remodeling of neural networks in the anterior forebrain of an animal model of hyperactivity and attention deficits as monitored by molecular imaging probes. Neurosci Biobehav Rev. 2000;24:149–156. doi: 10.1016/s0149-7634(99)00052-4. [DOI] [PubMed] [Google Scholar]
- Pavanello S, Dioni L, Hoxha M, Fedeli U, Mielzynska-Svach D, Baccarelli AA. Mitochondrial DNA copy number and exposure to polycyclic aromatic hydrocarbons. Cancer Epidemiol Biomarkers Prev. 2013;22:1722–1729. doi: 10.1158/1055-9965.EPI-13-0118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Peretz A, Peck EC, Bammler TK, Beyer RP, Sullivan JH, Trenga CA, Srinouanprachnah S, Farin FM, Kaufman JD. Diesel exhaust inhalation and assessment of peripheral blood mononuclear cell gene transcription effects: An exploratory study of healthy human volunteers. Inhal Toxicol. 2007;19:1107–1119. doi: 10.1080/08958370701665384. [DOI] [PubMed] [Google Scholar]
- Picard M, Zhang J, Hancock S, Derbeneva O, Golhar R, Golik P, O’Hearn S, Levy S, Potluri P, Lvova M, Davila A, Lin CS, Perin JC, Rappaport EF, Hakonarson H, Trounce IA, Procaccio V, Wallace DC. Progressive increase in mtdna 3243a >g heteroplasmy causes abrupt transcriptional reprogramming. Proc Natl Acad Sci U S A. 2014;111:E4033–4042. doi: 10.1073/pnas.1414028111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pieters N, Koppen G, Smeets K, Napierska D, Plusquin M, De Prins S, Van De Weghe H, Nelen V, Cox B, Cuypers A, Hoet P, Schoeters G, Nawrot TS. Decreased mitochondrial DNA content in association with exposure to polycyclic aromatic hydrocarbons in house dust during winter-time: From a population enquiry to cell culture. PLoS One. 2013;8:e63208. doi: 10.1371/journal.pone.0063208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pohunek P, Warner JO, Turzikova J, Kudrmann J, Roche WR. Markers of eosinophilic inflammation and tissue re-modelling in children before clinically diagnosed bronchial asthma. Pediatr Allergy Immunol. 2005;16:43–51. doi: 10.1111/j.1399-3038.2005.00239.x. [DOI] [PubMed] [Google Scholar]
- Pons R, Andreu AL, Checcarelli N, Vila MR, Engelstad K, Sue CM, Shungu D, Haggerty R, de Vivo DC, DiMauro S. Mitochondrial DNA abnormalities and autistic spectrum disorders. J Pediatr. 2004;144:81–85. doi: 10.1016/j.jpeds.2003.10.023. [DOI] [PubMed] [Google Scholar]
- Posnack NG, Swift LM, Kay MW, Lee NH, Sarvazyan N. Phthalate exposure changes the metabolic profile of cardiac muscle cells. Environ Health Perspect. 2012;120:1243–1251. doi: 10.1289/ehp.1205056. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Raby BA, Klanderman B, Murphy A, Mazza S, Camargo CA, Jr, Silverman EK, Weiss ST. A common mitochondrial haplogroup is associated with elevated total serum ige levels. J Allergy Clin Immunol. 2007;120:351–358. doi: 10.1016/j.jaci.2007.05.029. [DOI] [PubMed] [Google Scholar]
- Rahman I, Biswas SK, Kirkham PA. Regulation of inflammation and redox signaling by dietary polyphenols. Biochem Pharmacol. 2006;72:1439–1452. doi: 10.1016/j.bcp.2006.07.004. [DOI] [PubMed] [Google Scholar]
- Reinhardt K, Dowling DK, Morrow EH. Medicine. Mitochondrial replacement, evolution, and the clinic. Science. 2013;341:1345–1346. doi: 10.1126/science.1237146. [DOI] [PubMed] [Google Scholar]
- Rezin GT, Amboni G, Zugno AI, Quevedo J, Streck EL. Mitochondrial dysfunction and psychiatric disorders. Neurochem Res. 2009;34:1021–1029. doi: 10.1007/s11064-008-9865-8. [DOI] [PubMed] [Google Scholar]
- Rice D, Barone S. Critical periods of vulnerability for the developing nervous system: Evidence from human and animal models. Environ Health Perspect. 2000;108:511–533. doi: 10.1289/ehp.00108s3511. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Riedl MA, Nel AE. Importance of oxidative stress in the pathogenesis and treatment of asthma. Curr Opin Allergy Clin Immunol. 2008;8:49–56. doi: 10.1097/ACI.0b013e3282f3d913. [DOI] [PubMed] [Google Scholar]
- Robinson DS. Regulatory t cells and asthma. Clin Exp Allergy. 2009;39:1314–1323. doi: 10.1111/j.1365-2222.2009.03301.x. [DOI] [PubMed] [Google Scholar]
- Rosado-Berrios CA, Velez C, Zayas B. Mitochondrial permeability and toxicity of diethylhexyl and monoethylhexyl phthalates on tk6 human lymphoblasts cells. Toxicol In Vitro. 2011;25:2010–2016. doi: 10.1016/j.tiv.2011.08.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rossignol DA, Frye RE. Mitochondrial dysfunction in autism spectrum disorders: A systematic review and meta-analysis. Mol Psychiatry. 2012;17:290–314. doi: 10.1038/mp.2010.136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sato I, Miyado M, Sunohara M. Nadh dehydrogenase activity and expression of mrna of complex i (nd1, 51kda, and 75kda) in heart mitochondria of klotho mouse. Okajimas Folia Anat Jpn. 2005;82:49–56. doi: 10.2535/ofaj.82.49. [DOI] [PubMed] [Google Scholar]
- Scaglia F. The role of mitochondrial dysfunction in psychiatric disease. Dev Disabil Res Rev. 2010;16:136–143. doi: 10.1002/ddrr.115. [DOI] [PubMed] [Google Scholar]
- Scarpulla RC. Metabolic control of mitochondrial biogenesis through the pgc-1 family regulatory network. Biochim Biophys Acta. 2011;1813:1269–1278. doi: 10.1016/j.bbamcr.2010.09.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schmuczerova J, Brdicka R, Dostal M, Sram RJ, Topinka J. Genetic variability of hvrii mtdna in cord blood and respiratory morbidity in children. Mutat Res. 2009;666:1–7. doi: 10.1016/j.mrfmmm.2009.03.002. [DOI] [PubMed] [Google Scholar]
- Schneider JS, Lee MH, Anderson DW, Zuck L, Lidsky TI. Enriched environment during development is protective against lead-induced neurotoxicity. Brain Res. 2001;896:48–55. doi: 10.1016/s0006-8993(00)03249-2. [DOI] [PubMed] [Google Scholar]
- Sharma DR, Sunkaria A, Wani WY, Sharma RK, Kandimalla RJ, Bal A, Gill KD. Aluminium induced oxidative stress results in decreased mitochondrial biogenesis via modulation of pgc-1alpha expression. Toxicol Appl Pharmacol. 2013;273:365–380. doi: 10.1016/j.taap.2013.09.012. [DOI] [PubMed] [Google Scholar]
- Shaughnessy DT, McAllister K, Worth L, Haugen AC, Meyer JN, Domann FE, Van Houten B, Mostoslavsky R, Bultman SJ, Baccarelli AA, Begley TJ, Sobol RW, Hirschey MD, Ideker T, Santos JH, Copeland WC, Tice RR, Balshaw DM, Tyson FL. Mitochondria, energetics, epigenetics, and cellular responses to stress. Environ Health Perspect. 2014;122:1271–1278. doi: 10.1289/ehp.1408418. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shock LS, Thakkar PV, Peterson EJ, Moran RG, Taylor SM. DNA methyltransferase 1, cytosine methylation, and cytosine hydroxymethylation in mammalian mitochondria. Proc Natl Acad Sci U S A. 2011;108:3630–3635. doi: 10.1073/pnas.1012311108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Simoes DC, Psarra AM, Mauad T, Pantou I, Roussos C, Sekeris CE, Gratziou C. Glucocorticoid and estrogen receptors are reduced in mitochondria of lung epithelial cells in asthma. PLoS One. 2012;7:e39183. doi: 10.1371/journal.pone.0039183. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sondheimer N, Glatz CE, Tirone JE, Deardorff MA, Krieger AM, Hakonarson H. Neutral mitochondrial heteroplasmy and the influence of aging. Hum Mol Genet. 2011;20:1653–1659. doi: 10.1093/hmg/ddr043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Song L, Zheng J, Li H, Jia N, Suo Z, Cai Q, Bai Z, Cheng D, Zhu Z. Pre-natal stress causes oxidative damage to mitochondrial DNA in hippocampus of offspring rats. Neurochem Res. 2009;34:739–745. doi: 10.1007/s11064-008-9838-y. [DOI] [PubMed] [Google Scholar]
- Stephens M, Ludgate M, Rees DA. Brown fat and obesity: The next big thing? Clin Endocrinol (Oxf ) 2011;74:661–670. doi: 10.1111/j.1365-2265.2011.04018.x. [DOI] [PubMed] [Google Scholar]
- Streck EL, Goncalves CL, Furlanetto CB, Scaini G, Dal-Pizzol F, Quevedo J. Mitochondria and the central nervous system: Searching for a pathophysiological basis of psychiatric disorders. Rev Bras Psiquiatr. 2014;36:156–167. doi: 10.1590/1516-4446-2013-1224. [DOI] [PubMed] [Google Scholar]
- Sue CM, Bruno C, Andreu AL, Cargan A, Mendell JR, Tsao CY, Luquette M, Paolicchi J, Shanske S, DiMauro S, De Vivo DC. Infantile encephalopathy associated with the melas a3243g mutation. J Pediatr. 1999;134:696–700. doi: 10.1016/s0022-3476(99)70283-0. [DOI] [PubMed] [Google Scholar]
- Szczesny B, Tann AW, Mitra S. Age- and tissue-specific changes in mitochondrial and nuclear DNA base excision repair activity in mice: Susceptibility of skeletal muscles to oxidative injury. Mech Ageing Dev. 2010;131:330–337. doi: 10.1016/j.mad.2010.03.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Szibor M, Holtz J. Mitochondrial ageing. Basic Res Cardiol. 2003;98:210–218. doi: 10.1007/s00395-003-0421-z. [DOI] [PubMed] [Google Scholar]
- Tan D, Goerlitz DS, Dumitrescu RG, Han D, Seillier-Moiseiwitsch F, Spernak SM, Orden RA, Chen J, Goldman R, Shields PG. Associations between cigarette smoking and mitochondrial DNA abnormalities in buccal cells. Carcinogenesis. 2008;29:1170–1177. doi: 10.1093/carcin/bgn034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tan Z, Randall G, Fan J, Camoretti-Mercado B, Brockman-Schneider R, Pan L, Solway J, Gern JE, Lemanske RF, Nicolae D, Ober C. Allele-specific targeting of micrornas to hla-g and risk of asthma. Am J Hum Genet. 2007;81:829–834. doi: 10.1086/521200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tannetta DS, Sargent IL, Linton EA, Redman CW. Vitamins c and e inhibit apoptosis of cultured human term placenta trophoblast. Placenta. 2008;29:680–690. doi: 10.1016/j.placenta.2008.04.009. [DOI] [PubMed] [Google Scholar]
- Trasande L, Landrigan PL, Schechter C. Public health and economic consequences of methyl mercury toxicity to the developing brain. Environ Health Perspect. 2005;113:590–596. doi: 10.1289/ehp.7743. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Trian T, Benard G, Begueret H, Rossignol R, Girodet PO, Ghosh D, Ousova O, Vernejoux JM, Marthan R, Tunon-de-Lara JM, Berger P. Bronchial smooth muscle remodeling involves calcium-dependent enhanced mitochondrial biogenesis in asthma. J Exp Med. 2007;204:3173–3181. doi: 10.1084/jem.20070956. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vanyushin BF, Kiryanov GI, Kudryashova IB, Belozersky AN. DNA-methylase in loach embryos (misgurnus fossilis) FEBS Lett. 1971;15:313–316. doi: 10.1016/0014-5793(71)80646-4. [DOI] [PubMed] [Google Scholar]
- Vayssiere JL, Cordeau-Lossouarn L, Larcher JC, Basseville M, Gros F, Croizat B. Participation of the mitochondrial genome in the differentiation of neuroblastoma cells. In Vitro Cell Dev Biol. 1992;28A:763–772. doi: 10.1007/BF02631065. [DOI] [PubMed] [Google Scholar]
- Vergilio Cdos S, de Melo EJ. Autophagy, apoptosis and organelle features during cell exposure to cadmium. Biocell. 2013;37:45–54. [PubMed] [Google Scholar]
- Wakefield SL, Lane M, Mitchell M. Impaired mitochondrial function in the preimplantation embryo perturbs fetal and placental development in the mouse. Biol Reprod. 2011;84:572–580. doi: 10.1095/biolreprod.110.087262. [DOI] [PubMed] [Google Scholar]
- Wang B, Sun J, Ma Y, Wu G, Tian Y, Shi Y, Le G. Resveratrol preserves mitochondrial function, stimulates mitochondrial biogenesis, and attenuates oxidative stress in regulatory t cells of mice fed a high-fat diet. J Food Sci. 2014;79:H1823–1831. doi: 10.1111/1750-3841.12555. [DOI] [PubMed] [Google Scholar]
- Wang YY, Sui KX, Li H, Ma HY. The effects of lead exposure on placental nf-kappab expression and the consequences for gestation. Reprod Toxicol. 2009;27:190–195. doi: 10.1016/j.reprotox.2008.12.006. [DOI] [PubMed] [Google Scholar]
- Weiss B. Vulnerability of children and the developing brain to neurotoxic hazards. Environ Health Perspect. 2000;108(Suppl 3):375–381. doi: 10.1289/ehp.00108s3375. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Weissman JR, Kelley RI, Bauman ML, Cohen BH, Murray KF, Mitchell RL, Kern RL, Natowicz MR. Mitochondrial disease in autism spectrum disorder patients: A cohort analysis. PLoS One. 2008;3:e3815. doi: 10.1371/journal.pone.0003815. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Westbrook DG, Anderson PG, Pinkerton KE, Ballinger SW. Perinatal tobacco smoke exposure increases vascular oxidative stress and mitochondrial damage in non-human primates. Cardiovasc Toxicol. 2010;10:216–226. doi: 10.1007/s12012-010-9085-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- WHO. World Health Organization Global Plan of Action for Children’s Health and the Environment (2010–2015) WHO; Geneva: 2009. [Google Scholar]
- Wittkopp S, Staimer N, Tjoa T, Gillen D, Daher N, Shafer M, Schauer JJ, Sioutas C, Delfino RJ. Mitochondrial genetic background modifies the relationship between traffic-related air pollution exposure and systemic biomarkers of inflammation. PLoS One. 2013;8:e64444. doi: 10.1371/journal.pone.0064444. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wright RJ. Moving towards making social toxins mainstream in children’s environmental health. Curr Opin Pediatr. 2009;21:222–229. doi: 10.1097/MOP.0b013e3283292629. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wright RJ, Brunst KJ. Programming of respiratory health in childhood: Influence of outdoor air pollution. Curr Opin Pediatr. 2013;25:232–239. doi: 10.1097/MOP.0b013e32835e78cc. [DOI] [PubMed] [Google Scholar]
- Wright RJ, Fisher K, Chiu YH, Wright RO, Fein R, Cohen S, Coull BA. Disrupted prenatal maternal cortisol, maternal obesity, and childhood wheeze insights into prenatal programming. Am J Respir Crit Care Med. 2013;187:1186–1193. doi: 10.1164/rccm.201208-1530OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wright RJ, Brunst KJ. Social determinants in children’s environmental health: The stress-health paradigm. In: Landrigan PJ, Etzel RA, editors. Textbook f Children’s Environmental Health. London: Oxford University Press; 2014. [Google Scholar]
- Wright RO, Baccarelli A. Metals and neurotoxicology. J Nutr. 2007;137:2809–2813. doi: 10.1093/jn/137.12.2809. [DOI] [PubMed] [Google Scholar]
- Wu G, Imhoff-Kunsch B, Girard AW. Biological mechanisms for nutritional regulation of maternal health and fetal development. Paediatr Perinat Epidemiol. 2012;26(Suppl 1):4–26. doi: 10.1111/j.1365-3016.2012.01291.x. [DOI] [PubMed] [Google Scholar]
- Wu W, Bromberg PA, Samet JM. Zinc ions as effectors of environmental oxidative lung injury. Free Radic Biol Med. 2013;65:57–69. doi: 10.1016/j.freeradbiomed.2013.05.048. [DOI] [PubMed] [Google Scholar]
- Zhao H, Ma JK, Barger MW, Mercer RR, Millecchia L, Schwegler-Berry D, Castranova V, Ma JY. Reactive oxygen species- and nitric oxide-mediated lung inflammation and mitochondrial dysfunction in wild-type and inos-deficient mice exposed to diesel exhaust particles. J Toxicol Environ Health A. 2009;72:560–570. doi: 10.1080/15287390802706330. [DOI] [PubMed] [Google Scholar]
- Zhao X, Wang S, Wu Y, You H, Lv L. Acute zno nanoparticles exposure induces developmental toxicity, oxidative stress and DNA damage in embryo-larval zebrafish. Aquat Toxicol. 2013;136–137:49–59. doi: 10.1016/j.aquatox.2013.03.019. [DOI] [PubMed] [Google Scholar]
- Zifa E, Daniil Z, Skoumi E, Stavrou M, Papadimitriou K, Terzenidou M, Kostikas K, Bagiatis V, Gourgoulianis KI, Mamuris Z. Mitochondrial genetic background plays a role in increasing risk to asthma. Mol Biol Rep. 2012;39:4697–4708. doi: 10.1007/s11033-011-1262-8. [DOI] [PubMed] [Google Scholar]