Hostname: page-component-8448b6f56d-cfpbc Total loading time: 0 Render date: 2024-04-18T06:47:19.271Z Has data issue: false hasContentIssue false

Air pollution and your brain: what do you need to know right now

Published online by Cambridge University Press:  26 September 2014

Lilian Calderón-Garcidueñas*
Affiliation:
The Center for Structural and Functional Neurosciences, The University of Montana, Missoula, MT, USA
Ana Calderón-Garcidueñas
Affiliation:
Instituto de Medicina Forense, Universidad Veracruzana, Boca del Río, Veracruz, México
Ricardo Torres-Jardón
Affiliation:
Centro de Ciencias de la Atmósfera, Universidad Nacional Autónoma de México, Mexico City, Mexico
José Avila-Ramírez
Affiliation:
Hospital Médica Sur, México DF, México
Randy J. Kulesza
Affiliation:
Auditory Research Center, Lake Erie College of Osteopathic Medicine, Erie, PA, USA
Amedeo D. Angiulli
Affiliation:
Department of Neuroscience, Carleton University, Ottawa, ON, Canada
*
Correspondence to: Dr Lilian Calderón-Garcidueñas, The Center for Structural and Functional Neurosciences, The University of Montana, 32 Campus Drive, Skaggs Building 287, Missoula, MT 59812, USA. Email: lilian.calderon-garciduenas@umontana.edu
Rights & Permissions [Opens in a new window]

Abstract

Research links air pollution mostly to respiratory and cardiovascular disease. The effects of air pollution on the central nervous system (CNS) are not broadly recognized. Urban outdoor pollution is a global public health problem particularly severe in megacities and in underdeveloped countries, but large and small cities in the United States and the United Kingom are not spared. Fine and ultrafine particulate matter (UFPM) defined by aerodynamic diameter (<2.5-μm fine particles, PM2.5, and <100-nm UFPM) pose a special interest for the brain effects given the capability of very small particles to reach the brain. In adults, ambient pollution is associated to stroke and depression, whereas the emerging picture in children show significant systemic inflammation, immunodysregulation at systemic, intratechal and brain levels, neuroinflammation and brain oxidative stress, along with the main hallmarks of Alzheimer and Parkinson’s diseases: hyperphosphorilated tau, amyloid plaques and misfolded α-synuclein. Animal models exposed to particulate matter components show markers of both neuroinflammation and neurodegeneration. Epidemiological, cognitive, behavioral and mechanistic studies into the association between air pollution exposures and the development of CNS damage particularly in children are of pressing importance for public health and quality of life. Primary health providers have to include a complete prenatal and postnatal environmental and occupational history to indoor and outdoor toxic hazards and measures should be taken to prevent or reduce further exposures.

Type
Development
Copyright
© Cambridge University Press 2014 

Introduction

Millions of people around the world, 74 million in the United States and 24 million people living in the Mexico City Metropolitan Area (MCMA) are involuntarily inhaling ambient concentrations of particulate matter (PM) above safety standards. In the United States, millions of people in busy corridors from New York–New Jersey–Long Island to relatively small cities like Provo, UT are chronically exposed to fine PM year after year (Chen and Kan, Reference Chen and Kan2008). Well-documented air pollution events have been described in United Kingdom including the 1952 London episode with a high impact on public awareness of pollution (Bell et al., Reference Bell, Davis and Fletcher2004; Beelen et al., Reference Beelen, Raaschou-Nielsen, Stafoggia and Andersen2013; Langrish and Mills, Reference Langrish and Mills2014). Cohort longitudinal studies across Europe show 7% increase in natural cause mortality with each 5 μg/m3 in fine PM2.5 (Beelen et al., Reference Beelen, Raaschou-Nielsen, Stafoggia and Andersen2013; Langrish and Mills, Reference Langrish and Mills2014). According to a report by the European Topic Centre on Air and Climate Change (ETC/ACC), fine PM air pollution is associated with 455 000 premature deaths every year in the EU’s 27 member states, including 46 200 in the United Kingdom (ETC/ACC, 2009 report). Air pollution levels have been decreasing in developed countries, while in developing countries and countries in transition, the problem is decreasing at a very low rate or even worsening. MCMA is an example of a polluted megacity with over 40 000 industries, >4 million vehicles that consume more than 40 million liters of petroleum fuels per day and produce thousands of tons of pollutants (INE, 2011). Since MCMA lies in an elevated basin 2240 m above sea level, surrounded on three sides by mountain ridges, surface, as well as surface-based air temperature inversions occur frequently, trapping pollutant emissions close to the surface and aggravating the pollution (Bravo-Alvarez and Torres-Jardón, Reference Bravo-Alvarez and Torres-Jardón2002). MCMA geographical setting and the climatological characteristics along with the relatively little mobility of their residents allows for the opportunity of studying health effects associated to sustained yearlong exposures to concentrations of air pollutants above the current US National Ambient Air Quality Standards.

PM air pollution

Air pollution is a complex mixture of PM, gases, organic and inorganic compounds present in outdoor and indoor air. Urban outdoor pollution is a global public health problem (Molina and Molina, Reference Molina and Molina2004). PM defined by aerodynamic diameter (>2.5 to <10-μm coarse particles, PM10, <2.5-μm fine particles, PM2.5, and <100-nm ultrafine particulate matter (UFPM)) poses a special interest for the brain effects given the capability of fine and UFPM to reach the brain (Block and Calderón-Garcidueñas, Reference Block and Calderón-Garcidueñas2009). The major sources of fine and UFPM in the environment, that is, combustion sources from gas, oil, coal, industry and fires, metals and biological materials are common threat in urban and rural sites (Brook et al., Reference Brook, Rajagopala, Pope, Brook, Bhatnagar, Diez-Roux, Holguin, Hong, Luepker, Mittleman, Peters, Siscovick, Smith, Whitse and Kaufman2010). The smaller the particle, the larger the surface area, the better lung penetration and diffusion and major particle deposition in the respiratory tract and direct translocation into the brain. A significant threat not contemplated by health care workers are man-made particles <100 nm. Nano-sized materials are included in many consumer products and daily use indoor machines produce them in significant amounts, and thus humans are being exposed on daily bases (ie, tooth paste, flavor enhancers, food additives, food and drink containers including baby bottles and pacifiers, laser printers, etc.; Hagens et al., Reference Hagens, Oomen, de Jong, Cassee and Sips2007; Benn et al., Reference Benn, Cavanagh, Hristovski, Posner and Westerhoff2010; Bergin and Witzmann, Reference Bergin and Witzmann2013). Most people spend >90% of their time indoors, so indoor air pollution is very important: smoking, cooking, candle/incense burning, cleaning and use of plastics and conglomerates all contribute to indoor pollution (Habre et al., Reference Habre, Coull, Moshier, Godbold, Nath, Castro, Schachter, Rohr, Kattan, Spengler and Koutrakis2013). Poor air quality in schools and in-vehicle concentrations of pollutants are also of deep concern (Brown et al., Reference Brown, Sarnat and Koutrakis2012; Annesi-Maesano et al., Reference Annesi-Maesano, Baiz, Banerjee, Rudnai and Rive2013).

As in any city, air quality in MCMA is determined by the balance between pollutant emissions and the capacity of the geographical site to eliminate, disperse or concentrate those air pollutants. Despite MCMA rapid growth and development, air quality has improved during the past two decades. Nevertheless, residents remain exposed to concentrations of airborne pollutants exceeding ambient air quality standards, especially for PM and ozone, the two most important pollutants from the standpoint of public health (Brook et al., Reference Brook, Rajagopala, Pope, Brook, Bhatnagar, Diez-Roux, Holguin, Hong, Luepker, Mittleman, Peters, Siscovick, Smith, Whitse and Kaufman2010). The higher socioeconomic status (SES) MCMA population lives toward the south and west of the urban area with access to vegetation, water and better road networks. The industry is located primarily in the northeast and northwest, whereas the east side has been covered by large housing low SES developments in areas of difficult access and poor services. This distribution of the population, as well as the intensity and type of activities carried out, has great influence on the spatial distribution of emissions. According to the most recent emissions inventory for the MCMA (Secretaria de Medio Ambiente, Gobierno de Distrito Federal (SMA-GDF), 2008), the distribution of PM10 depends largely on the activity of vehicles on paved and unpaved streets and roads, and in some urban areas of the northwest, these emissions are added with those generated by heavy industries. The fractional composition of PM2.5 has been dominated by organic and black carbon, sulfate, nitrate, ammonium and crustal components, with site- and time-dependent variations (Aiken et al., Reference Aiken, Salcedo, Cubison, Huffman and DeCarlo2009). Particle species are typically categorized as ‘primary’ if they are emitted in the particle phase and ‘secondary’ if their precursors (volatile organic compounds, NO x , SO2, NH3 and others) are emitted in the gas-phase and subsequent chemical reactions bring them to the particle phase.

Diesel and gasoline exhausts emissions have been responsible for a significant fraction of the fine particle primary emissions in MCMA (Molina et al., Reference Molina, Madronich, Gaffney, Apel, de Foy, Fast, Ferrare, Herndon, Jimenez, Lamb, Osornio-Vargas, Russell, Schauer, Stevens, Volkamer and Zavala2010). Polycyclic aromatic hydrocarbons (PAH) are a family of species, some of which are highly mutagenic and carcinogenic, that are generally associated with black carbon as their emissions are largely from combustion sources (Valle-Hernández et al., Reference Valle-Hernández, Mugica-Alvarez, Salinas-Talavera, Amador-Muñoz, Murillo-Tovar, Villalobos-Pietrini and De Vizcaya-Ruíz2010).

Peak concentrations of PAHs in MCMA are reached during the morning rush hour and are of the order of 120 ngm−3, which is significantly higher than in United States (Marr et al., Reference Marr, Grogan, Wohrnschimmel, Molina, Molina, Smith and Garshick2004; Molina et al., Reference Molina, Madronich, Gaffney, Apel, de Foy, Fast, Ferrare, Herndon, Jimenez, Lamb, Osornio-Vargas, Russell, Schauer, Stevens, Volkamer and Zavala2010). High levels of anthropogenic metals including chromium (Cr), zinc (Zn), copper, lead (Pb), antimony, arsenic, tin and barium complete the toxic potential of fine PM (Molina et al., Reference Molina, Madronich, Gaffney, Apel, de Foy, Fast, Ferrare, Herndon, Jimenez, Lamb, Osornio-Vargas, Russell, Schauer, Stevens, Volkamer and Zavala2010). As expected, these metals exhibit strong temporal variations in concentration and are largely associated with industrial and mobile sources. Elements representing mostly road traffic, that is, Cr, Mn, Zn and Pb, are typically associated with engine emissions and abrasion of tires and brake pads. V and Ni are interpreted as tracers of long-range transport from the use of heavy fuel oil in the north of the basin (Querol et al., Reference Querol, Pey, Minguillón, Pérez, Alastuey, Viana, Moreno, Bernabé, Blanco, Cárdenas, Vega, Sosa, Escalona, Ruiz and Artiñano2008). Pb deserves a special mention. Before 1986, Pb was probably the most harmful pollutant in MCMA, associated with the exclusive use of leaded gasoline and resulting in Pb concentrations three times the air quality standard. In response to a very strong social pressure and to a growing international trend to control car emissions with catalytic converters, PEMEX, the federal Mexican oil company, was forced to reduce the gasoline content of tetra-ethyl-lead (Bravo-Alvarez and Torres-Jardón, Reference Bravo-Alvarez and Torres-Jardón2002) resulting in Pb concentrations∼2 μg/m3 in 1988 and reaching ∼0.5 μg/m3 by 1998. Although Pb is no longer an air pollutant problem, lipopolysaccharides (products of the outer membrane of Gram-negative bacteria) associated with PM, are detected in very high concentrations in southern Mexico City (Rosas-Pérez et al., Reference Rosas-Pérez, Serrano, Alfaro-Moreno, Baumgardner, Garcia-Cuellar, Martin del Campo, Raga, Castillejos, Colin and Osornio-Vargas2007). The grim scenario for MCMA residents is a sustained exposure to PM2.5 several hours per day above the current standards, every year, in such a way that residents are living in a very effective exposure chamber from conception to death. Is this scenario exclusive for Mexico City residents? The answer is no, Salt Lake City, Tacoma, San Francisco Bay, Los Angeles, Fairbanks, London, Greater Manchester and Oxfordshire residents to name a few have similar scenarios, albeit less severe (Bell et al., Reference Bell, Goldberg, Hogrefe, Kinney, Knowlton, Lynn, Rosenthal, Rosenzweig and Patz2007; Maheswarang et al., Reference Maheswarang, Pearson, Smeeton, Beevers, Campbell and Wolfe2010; Williams et al., Reference Williams, Ulrich, Larson, Wener, Wood, Chen-Levy, Campbell, Potter, McTiernan and Roos2011; Beelen et al., Reference Beelen, Raaschou-Nielsen, Stafoggia and Andersen2013; UK Air Data). On the other hand, millions of Shanghai residents are covered with heavy haze substantially increasing cardiovascular, respiratory and cerebrovascular morbidity and mortality (Xu et al., Reference Xu and Ye2013).

The impact of polluted air on an adult brain

The first important statement to be made is that age and disease status are key factors on the impact of air pollutants. Associations between stroke, ambient pollution and coal fumes were suggested in the 1980s (Knox, Reference Knox1981; Zhang et al., Reference Zhang, Yu and Zhou1988). Cigarette smoking as a stroke risk factor (and of course an excellent source of large amounts of PM) was published a decade later (Howard et al., Reference Howard, Wagenknecht, Cai, Cooper, Kraut and Toole1998). Ischemic stroke mortality and transient ischemic attacks relate to fine and UFPM exposures even at concentrations below the current standards (Hong et al., Reference Hong, Lee, Kim and Kwon2002; Kettunen et al., Reference Kettunen, Lanki, Tiittanen, Aaalto, Koskentalo, Kulmala, Salomaa and Pekkanen2007; Lisabeth et al., Reference Lisabeth, Escobar, Dvonch, Sanchez, Majersik, Brown, Smith and Morgenstern2008; Bedada et al., Reference Bedada, Smith, Tyrrell, Hirst and Agius2012; Leiva et al., Reference Leiva, Santibañez, Ibarra, Matus and Seguel2013) and very important from the clinical point of view, outdoor air pollution and proximity to high-traffic roadways impact stroke survival (Maheswarang et al., Reference Maheswarang, Pearson, Smeeton, Beevers, Campbell and Wolfe2010; Wilker et al., Reference Wilker, Mostofsky, Lue, Gold, Schwartz, Wellenius and Mittleman2013). The largest association between PM2.5 and ischemic stroke risk was seen with stroke due to large-artery atherosclerosis and small-vessel occlusion: diabetic patients are particularly at risk (O’Donnell et al., Reference O’Donnell, Fang, Mittleman, Kapral and Wellenius2011). Moreover, the associations involve not only urban pollutants but also dust storms and unfortunate events like 9/11 World Trade Center (Yang et al., Reference Yang, Chen, Chiu and Goggins2005; Brackbill et al., Reference Brackbill, Thorpe, DiGrande, Perrin, Sapp, Wu, Campolucci, Walker, Cone, Pulliam, Thalji, Farfel and Thomas2006). Although most of the literature associating stroke with air pollution emphasizes PM, ozone – a key photochemical pollutant affecting our populations in Mexico City, Los Angeles, San Joaquin Valley, Riverside, Sacramento, Baltimore, Dallas, south-east of England, etc. – is also a big player (Parrish, et al., Reference Parrish, Singh, Molina and Madronich2011; Carlsen et al., Reference Carlsen, Forsberg, Meister, Gislason and Oudin2013; Suissa et al., Reference Suissa, Fortier, Lachaud, Staccini and Mahagne2013). Depression and mood disorders have been associated with air pollution. Increases in PM10, PM2.5, NO2, CO, SO2, O3 and biomass burning are associated with depression in the elderly, and emotional symptoms and suicide attempts in younger subjects (Szyszkowicz et al., Reference Szyszkowicz, Willey, Grafstein, Rowe and Colman2010; Wisnivesky et al., Reference Wisnivesky, Teitelbaum, Todd, Boffetta, Crane, Crowley, de la Hoz, Dellenbaugh, Harrison, Herbert, Kim, Jeon, Kaplan, Katz, Levin, Luft, Markowitz, Moline, Ozbay, Pietrzak, Shapiro, Sharma, Skloot, Southwick, Stevenson, Udasin, Wallenstein and Landrigan2011; Banerjee et al., Reference Banerjee, Siddique, Dutta, Mukherjee and Ray2012; Lim et al., Reference Lim, Kim, Kim, Bae, Park and Hong2012). Exposure of animal models to ambient PM2.5 results in depressive responses and increase hippocampal pro-inflammatory cytokines (Fonken et al., Reference Fonken, Xu, Weil, Chen, Sun, Rajagopalan and Nelson2011), whereas prenatal nano-PM impacts neonatal neurons and adult behavior in mice (Davis et al., Reference Davis, Bortolato, Godar, Sander, Iwata, Pakbin, Shib, Berhane, McConnell, Sioutas, Finch, Morgan and Block2013).

The impact of polluted air on a developing brain

Clean air is fundamental for children’s health and well-being. Millions of children are showing an array of adverse short- and long-term health outcomes related to air pollution exposures. Widely recognized among pediatric health providers is the impact of intrauterine factors, parent–child interactions, cognitive stimulation, maternal SES during pregnancy and the child’s nutrition and exposure to complex learning stimuli, all vital for brain development (Calderón-Garcidueñas and Torres-Jardón, Reference Calderón-Garcidueñas and Torres-Jardón2012a). However, air pollution brain effects rooted in intrauterine life and childhood are not generally acknowledged. Air pollution is not broadly recognized in the context of children’s brain effects including the presence of neuroinflammation, cognitive deficits, structural brain alterations and neuropathological hallmarks of Alzheimer (AD) and Parkinson’s diseases (PD) (Calderón-Garcidueñas et al., Reference Calderón-Garcidueñas, Mora-Tiscareño, Fordham, Valencia-Salazar, Chung, Rodriguez-Alcaraz, Paredes, Variakojis, Villarreal-Calderón, Flores-Camacho, Antunez-Solis, Henríquez-Roldán and Hazucha2003; 2008Reference Calderón-Garcidueñas, Mora-Tiscareño, Ontiveros, Gómez-Garza, Barragán-Mejía, Broadway, Chapman, Valencia-Salazar, Jewells, Maronpot, Henríquez-Roldán, Pérez-Guillé, Torres-Jardón, Herrit, Brooks, Osnaya-Brizuela, Monroy, González-Maciel, Reynoso-Robles, Villarreal-Calderon, Solt and Englea; 2008Reference Calderón-Garcidueñas, Villarreal-Calderon, Valencia-Salazar, Henríquez-Roldán, Gutiérrez-Castrellón, Torres-Jardón, Osnaya-Brizuela, Romero, Torres-Jardón, Solt and Reedb; 2008Reference Calderón-Garcidueñas, Solt, Franco-Lira, Torres-Jardón, Nuse, Herritt, Villarreal-Calderón, Osnaya, Stone, García, Brooks, González-Maciel, Reynoso-Robles, Delgado-Chávez and Reedc; Reference Calderón-Garcidueñas, Macias-Parra, Hoffmann, Valencia-Salazar, Henríquez-Roldán, Monte, Barragán-Mejía, Villarreal-Calderon, Romero, Granada-Macías, Torres-Jardón, Medina-Cortina and Maronpot2009; Reference Calderón-Garcidueñas, Franco-Lira, Henríquez-Roldán, González-Maciel, Reynoso-Robles, Villarreal-Calderon, Herritt, Brooks, Keefe, Palacios-Moreno, Torres-Jardón, Medina-Cortina, Delgado-Chávez, Aiello-Mora, Maronpot and Doty2010; 2011a; 2011Reference Calderón-Garcidueñas, Engle, Mora-Tiscareño, Styner, Gomez-Garza, Zhu, Jewells, Torres-Jardón, Romero, Monroy-Acosta, González-González, Medina-Cortina and D'Angiullib; 2012Reference Calderón-Garcidueñas and Torres-Jardóna; 2012b; 2012Reference Calderón-Garcidueñas, Mora-Tiscareño, Styner, Gómez-Garza, Zhu, Torres-Jardón, Carlos, Solorio-López, Medina-Cortina, Kavanaugh and D’Angiullic; 2013a; 2013Reference Calderón-Garcidueñas, Cross, Franco-Lira, Aragón-Flores, Kavanaugh, Torres-Jardón, Chao, Thompson, Chang, Zhu and D’Angiullib).

A coherent pathway linking exposure to air pollution and brain damage starts with a chronic inflammatory process involving the respiratory upper and lower tracts, which result in a systemic inflammatory response with the production of inflammatory mediators capable of reaching the brain (Calderón-Garcidueñas et al., Reference Calderón-Garcidueñas, Mora-Tiscareño, Fordham, Valencia-Salazar, Chung, Rodriguez-Alcaraz, Paredes, Variakojis, Villarreal-Calderón, Flores-Camacho, Antunez-Solis, Henríquez-Roldán and Hazucha2003; Reference Ou, Hedley, Chung, Thach, Chau, Chan, Yang, Ho, Wong and Lam2008 Reference Calderón-Garcidueñas, Mora-Tiscareño, Ontiveros, Gómez-Garza, Barragán-Mejía, Broadway, Chapman, Valencia-Salazar, Jewells, Maronpot, Henríquez-Roldán, Pérez-Guillé, Torres-Jardón, Herrit, Brooks, Osnaya-Brizuela, Monroy, González-Maciel, Reynoso-Robles, Villarreal-Calderon, Solt and Englea; 2008Reference Calderón-Garcidueñas, Villarreal-Calderon, Valencia-Salazar, Henríquez-Roldán, Gutiérrez-Castrellón, Torres-Jardón, Osnaya-Brizuela, Romero, Torres-Jardón, Solt and Reedb; 2008Reference Calderón-Garcidueñas, Solt, Franco-Lira, Torres-Jardón, Nuse, Herritt, Villarreal-Calderón, Osnaya, Stone, García, Brooks, González-Maciel, Reynoso-Robles, Delgado-Chávez and Reedc; Reference Calderón-Garcidueñas, Macias-Parra, Hoffmann, Valencia-Salazar, Henríquez-Roldán, Monte, Barragán-Mejía, Villarreal-Calderon, Romero, Granada-Macías, Torres-Jardón, Medina-Cortina and Maronpot2009; 2011Reference Calderón-Garcidueñas, D’Angiulli, Kulesza, Torres-Jardón, Osnaya, Romero, Keefe, Herritt, Brooks, Avila-Ramirez, Delgado-Chávez, Medina-Cortina and González-Gonzáleza; 2011Reference Calderón-Garcidueñas, Engle, Mora-Tiscareño, Styner, Gomez-Garza, Zhu, Jewells, Torres-Jardón, Romero, Monroy-Acosta, González-González, Medina-Cortina and D'Angiullib; 2012Reference Calderón-Garcidueñas and Torres-Jardóna; 2012Reference Calderón-Garcidueñas, Kavanaugh, Block, D’Angiulli, Delgado-Chávez, Torres-Jardón, González-Maciel, Reynoso-Robles, Osnaya, Villarreal-Calderon, Guo, Hua, Zhu, Perry and Diazb; 2013Reference Calderón-Garcidueñas, Franco-Lira, Mora-Tiscareño, Medina-Cortina, Torres-Jardón and Kavanaugha; 2013Reference Calderón-Garcidueñas, Cross, Franco-Lira, Aragón-Flores, Kavanaugh, Torres-Jardón, Chao, Thompson, Chang, Zhu and D’Angiullib). Continuous expression of potent inflammatory mediators in the central nervous system (CNS) and the formation of reactive oxygen species are major findings in urban residents (Block and Calderón-Garcidueñas, Reference Block and Calderón-Garcidueñas2009). UFPM, particulate matter-associated lipopolysaccharides, and metal uptake could take place through olfactory neurons, cranial nerves such as the trigeminal and vagus, the systemic circulation and macrophage-like cells loaded with PM from the lungs (Calderón-Garcidueñas et al., Reference Calderón-Garcidueñas, Solt, Franco-Lira, Torres-Jardón, Nuse, Herritt, Villarreal-Calderón, Osnaya, Stone, García, Brooks, González-Maciel, Reynoso-Robles, Delgado-Chávez and Reed2008c; 2012Reference Calderón-Garcidueñas and Torres-Jardóna). Activation of the brain innate immune responses resulting from the interaction between circulating cytokines and constitutively expressed cytokine receptors located in brain endothelial cells is followed by activation of cells involved in adaptive immunity (Lampron et al., Reference Lampron, Elali and Rivest2013). Monocytes are the main innate immune response mediator cells, producing and secreting TNFα, interleukin-1β (IL-1β) and IL-6, which in turn recruit and increase the activity of other immune cells. Fine and UFPM could serve as the crucial trigger for a chain of events leading to endothelial activation, disruption of blood–brain barrier (BBB), altered response of the innate immune system, production of autoantibodies to cell junction and neural proteins, neuroinflammation and neurodegeneration. These early changes, amenable to intervention and viewed initially as a compensatory phenomenon by some groups including ours (Perry et al., Reference Perry, Nunomura, Hirai, Zhu, Pérez, Avila, Castellani, Atwood, Aliev, Sayre, Takeda and Smith2002; Castellani et al., Reference Castellani, Zhu, Zhu, Moreira, Perry and Smith2006; Calderón-Garcidueñas et al., 2012Reference Calderón-Garcidueñas and Torres-Jardóna; Reference Calderón-Garcidueñas, Franco-Lira, Mora-Tiscareño, Medina-Cortina, Torres-Jardón and Kavanaugh2013a) are likely critical to onset and progression of the neurodegenerative changes and the full clinical picture seen in AD patients (Castellani et al., Reference Castellani, Zhu, Zhu, Moreira, Perry and Smith2006).

There is growing recognition of the role of systemic and neural inflammation and the interplay between immunity, neurodegeneration and maladaptive activation of innate/adaptive immunity as key pathogenic phenomenon in AD (Blasko et al., Reference Blasko, Stampfer-Kountchev, Robatscher, Veerhuis, Eikelenboom and Grubeck-Loebenstein2004; Dik et al., Reference Dik, Jonker, Hack, Smit, Comijs and Eikelenboom2005; Finch and Morgan, Reference Finch and Morgan2007; Bonotis et al., Reference Bonotis, Krikki, Holeva, Aggouridaki, Costa and Baloyannis2008; Gomez-Ravetti and Moscato, Reference Gomez-Ravetti and Moscato2008; Cunningham et al., Reference Cunningham, Campion, Lunnon, Murray, Woods, Deacon, Rawlins and Perry2009; Keene et al., Reference Keene, Cudaback, Li, Montine and Montine2011). Likewise, change in immune status has been suggested as a plausible biological mechanism by which PM could cause adverse health effects (Eikelenboom et al., Reference Eikelenboom, van Exel, Veerhuis, Rozemuller, van Gool and Hoozemans2011). PM has the capability of crossing barriers, including the BBB resulting in neuroinflammation and intrinsically disordered neural proteins associated with neurodegenerative diseases (Campbell, Reference Campbell2004; Win-Shwe et al., Reference Win-Shwe, Yamamoto, Fujitani, Hirano and Fujimaki2008; Levesque et al., Reference Levesque, Surace, McDonald and Block2011).

The emerging picture reveals highly exposed urban children exhibit significant neuroinflammation and brain oxidative stress (Calderón-G arcidueñas et al., Reference Calderón-Garcidueñas, Solt, Franco-Lira, Torres-Jardón, Nuse, Herritt, Villarreal-Calderón, Osnaya, Stone, García, Brooks, González-Maciel, Reynoso-Robles, Delgado-Chávez and Reed2008c; 2012Reference Calderón-Garcidueñas and Torres-Jardóna; Reference Calderón-Garcidueñas, Franco-Lira, Mora-Tiscareño, Medina-Cortina, Torres-Jardón and Kavanaugh2013a; 2013Reference Calderón-Garcidueñas, Cross, Franco-Lira, Aragón-Flores, Kavanaugh, Torres-Jardón, Chao, Thompson, Chang, Zhu and D’Angiullib). In addition, these children have extensive abnormal white matter blood vessels, perivascular inflammation and a breakdown of the BBB (Calderón-Garcidueñas et al., Reference Calderón-Garcidueñas, Solt, Franco-Lira, Torres-Jardón, Nuse, Herritt, Villarreal-Calderón, Osnaya, Stone, García, Brooks, González-Maciel, Reynoso-Robles, Delgado-Chávez and Reed2008c). Fine tuning of immune-to-brain communication is crucial to neural networks appropriate functioning, thus our recent finding of autoantibodies to actin and occludin/zonulin in the presence of BBB compromise might represent one more factor contributing to initiation and/or pathogenesis of neurodegenerative changes. Current data support a role for air pollution in CNS damage and urban children shared mechanistic pathways potentially conducting to AD and PD (Calderón-Garcidueñas et al., Reference Calderón-Garcidueñas, Franco-Lira, Mora-Tiscareño, Medina-Cortina, Torres-Jardón and Kavanaugh2013a).

The short- and long-term effects of air pollution upon the CNS

We have discussed plausible mechanistic pathways causing CNS effects associated with sustained exposures of ambient pollutants during a lifetime, however, it should be clear that acute, subchronic or chronic exposures to air pollutants all have detrimental CNS effects. An example of acute and subchronic massive exposures was the World Trade Center 9/11-related environmental disaster (Bills et al., Reference Bills, Levy, Sharma, Charney, Herbert, Moline and Katz2008; Jordan et al., Reference Jordan, Stellman, Morabia, Miller-Archie, Alper, Laskaris, Brackbill and Cone2013; Ozbay et al., Reference Ozbay, Aud der Heyde, Reissman and Sharma2013). Massive exposure to a complex mixture of inhalable fine PM, nanoparticles (NPs) and toxic chemicals, resulted in persistent mental detrimental effects and evolution toward unknown brain health outcomes beyond posttraumatic stress disorder (Bills et al., Reference Bills, Levy, Sharma, Charney, Herbert, Moline and Katz2008; Jordan et al., Reference Jordan, Stellman, Morabia, Miller-Archie, Alper, Laskaris, Brackbill and Cone2013; Ozbay et al., Reference Ozbay, Aud der Heyde, Reissman and Sharma2013).

The olfactory bulb (OB) pathology needs special attention because large segments of the world population inhale toxic substances on daily basis that have the potential for harming the olfactory system and penetrating the brain via the olfactory epithelium (Calderón-Garcidueñas et al., Reference Calderón-Garcidueñas, Franco-Lira, Henríquez-Roldán, González-Maciel, Reynoso-Robles, Villarreal-Calderon, Herritt, Brooks, Keefe, Palacios-Moreno, Torres-Jardón, Medina-Cortina, Delgado-Chávez, Aiello-Mora, Maronpot and Doty2010). The issue is very important in the context of air pollution because olfactory dysfunction is among the earliest ‘pre-clinical’ features of AD and PD, occurring in ~90% of early onset cases (Wang et al., Reference Wang, Eslinger, Doty, Zimmerman, Grunfeld, Sun, Meadowcroft, Connor, Price, Smith and Yang2010; Doty, Reference Doty2012). Early olfactory deficits in MCMA young residents appear to be associated with the presence of β-amyloid, α- synuclein, PM in glomerular structures and the massive distortion of the OB organization (Calderón-Garcidueñas et al., Reference Calderón-Garcidueñas, Franco-Lira, Henríquez-Roldán, González-Maciel, Reynoso-Robles, Villarreal-Calderon, Herritt, Brooks, Keefe, Palacios-Moreno, Torres-Jardón, Medina-Cortina, Delgado-Chávez, Aiello-Mora, Maronpot and Doty2010). The central delayed brainstem auditory evoked potentials (BAEPs), auditory impairment and vestibular dysfunction in exposed children is likely related to the extensive brainstem inflammation with accumulation of β-amyloid and α-synuclein in key auditory and vestibular nuclei (Calderón-Garcidueñas et al., Reference Calderón-Garcidueñas, D’Angiulli, Kulesza, Torres-Jardón, Osnaya, Romero, Keefe, Herritt, Brooks, Avila-Ramirez, Delgado-Chávez, Medina-Cortina and González-González2011). Neurodegenerative changes in the dorsal motor nucleus of the vagus, the nucleus of the solitary tract, arcuate nucleus, raphe midline, and extra-raphe medial and lateral tegmental neurons (Calderón-Garcidueñas et al., Reference Calderón-Garcidueñas, D’Angiulli, Kulesza, Torres-Jardón, Osnaya, Romero, Keefe, Herritt, Brooks, Avila-Ramirez, Delgado-Chávez, Medina-Cortina and González-González2011) are similar to the PD stages I and II of Braak et al. (Reference Braak, Ghebremedhin, Rüb, Bratzke and Del Tredeci2004). The brainstem pathology we are observing in highly exposed children has strong links with both AD and PD (Braak et al., Reference Braak, Ghebremedhin, Rüb, Bratzke and Del Tredeci2004; Reference Braak, Thal, Ghebremedhin and Del Tredeci2011).

Recent studies have reported associations between prenatal and perinatal exposures to air pollutants and autism spectrum disorder (ASD) in children (van den Hazel et al., Reference van den Hazel, Zuurbier, Babisch, Bartonova, Bistrup, Bolte, Busby, Butter, Ceccatelli, Fucic, Hanke, Johansson, Kohlhuber, Leijs, Lundqvist, Moshammer, Naginiene, Preece, Ronchetti, Salines, Saunders, Schoeters, Stilianakis, ten Tusscher and Koppe2006; Larsson et al., Reference Larsson, Weiss, Janson, Sundell and Bornehag2009; Herbert, Reference Herbert2010; Volk et al., Reference Volk, Hertz-Picciotto, Delwiche, Lurmann and McConnell2011; Reference Volk, Lurmann, Penfold, Hertz-Picciotto and McConnell2013; Reference Volk, Kerin, Lurmann, Hertz-Picciotto, McConnell and Campbell2014; Becerra et al., Reference Becerra, Wilhelm, Olsen, Cockburn and Ritz2013; Roberts et al., Reference Roberts, Lyall, Hart, Laden, Just, Bobb, Koenen, Ascherio and Weisskoof2013; Visser et al., Reference Visser, Rommelse, Vink, Schrieken, Oosterling, van der Gaag and Buitelaar2013). ASD is likely caused by complex interactions between genetic and environmental factors. ASD is associated to air pollution exposures during pregnancy in US cities with significant air pollution from traffic sources (Becerra et al., Reference Becerra, Wilhelm, Olsen, Cockburn and Ritz2013). Activation of the maternal immune system during pregnancy and abnormal behavioral development in Rhesus monkey offspring brings up key questions regarding the role of air pollution and autism among vulnerable populations (Bauman et al., Reference Bauman, Iosif, Smith, Bregere, Amaral and Patterson2013). To complicate matters for clinicians in real-world settings, the changes in the diagnostic criteria for autism in DSM-5 is causing significant controversies and concerns among health providers without formal training in research instruments and complicated assessment processes (Hazen et al., Reference Hazen, McDougle and Volkmar2013).

Reflections from practice

This paper is the product of discussions among the authors, all of us familiar with MCMA air pollution. We are physicians, pathologists, neuropathologists, auditory system experts, toxicologists, psychologists and atmospheric researchers dealing with children, worried parents and school officials, looking at neurodegenerative pathologies in autopsy materials from seemingly healthy children dying in accidents and having significant trouble in explaining parents plausible biological pathways that will solve all their questions. Several concerns have been raised in this short review about the influence of air pollutants upon the adult and the developing brain that involve a wide spectrum of pathologies the clinician should take into account for short- and long-term CNS impact.

In adults, it is imperative to take care of chronic diseases that aggravate vascular, metabolic and structural brain changes, while advising the patient about tobacco and cigarette smoking and harmful occupations and hobbies. In children, the cognitive, auditory and vestibular effects have immediate early school negative effects, while the prefrontal lesions and the diffuse cortical neuroinflammation will be reflected in decreased career opportunities, negative social health outcomes, including increases in delinquent or criminal activity and violence and consequently a major negative impact on the economy in which those individuals reside. An issue everybody ignores is that the teens reduced capacity to block impulsive antisocial behavior that accompanies impaired fluid cognition is having a significant impact on society.

Next, we review more in detail the specific air pollution effects of brain impairment on cognition and behavior that support the practical reflections presented in this paper. Table 1 summarizes in schematic form selected studies examining neurocognitive/neurophysiological effects of air pollution in children, adolescents and young adult populations. The table shows the populations and the air pollutants studied, the tests and deficits found, other tests used and the city/cities/country where the study took place. In what follows we will be focusing mainly on the available findings concerning MCMA children and mechanistic studies on brain development in animal research literature.

Table 1 Selected studies examining neurocognitive/neurophysiological effects of air pollution in children, adolescents and young adult populations. The table shows the populations and the air pollutants studied, the tests and deficits found, other tests used and the city/cities/country where the study took place

The table shows the populations and the air pollutants studied, the tests and deficits found, other tests used and the city/cities/country where the study took place.

Making the links: detrimental brain effects and cognitive/behavioral functions

As we have already mentioned, depending on the pollutant component, doses, exposure protocol, age and gender, health status, etc., the detrimental effects range from endothelial dysfunction, BBB breakdown, dopaminergic neuronal damage, DNA damage, white matter lesions, neuroinflammation, formation of free radicals and oxidative stress, to the identification of early hallmarks of AD and PD (Campbell et al., Reference Bell, Davis and Fletcher2004; Fonken et al., Reference Fonken, Xu, Weil, Chen, Sun, Rajagopalan and Nelson2011; Levesque et al., Reference Levesque, Surace, McDonald and Block2011; Win-Shwe and Fujimaki, Reference Win-Shwe and Fujimaki2011; Wu et al., Reference Wu, Wang and Sun2011; Brun et al., Reference Brun, Carrière and Mabondzo2012; Guo et al., Reference Guo, Zhu, Guo, Li, Chen, Sang and Yao2012; Sharma and Sharma, Reference Sharma and Sharma2012; Trickler et al., Reference Trickler, Lantz and Schrand2012). The chronic effects of PM2.5 in mice result in spatial learning and memory deficits with neuroinflammation and hippocampal dendritic alterations (Fonken et al., Reference Fonken, Xu, Weil, Chen, Sun, Rajagopalan and Nelson2011). Metal accumulation is associated with memory dysfunction, especially if the metal is administered during the neonatal period, a clear example being iron loading in rodents resulting in impaired spatial memory and in long-term retention of object recognition deficient memory (Schröder et al., Reference Schröder, Silva-Figueiredo and Martins de Lima2013). Equally important in the context of urban environments is the massive exposure to NPs entering the body mostly through inhalation (Win-Shwe et al., Reference Win-Shwe and Fujimaki2011; Brun et al., Reference Brun, Carrière and Mabondzo2012). Gold and silver NPs have an effect on human embryonic neural precursor cell growth, suggesting a negative impact of NPs on the developing CNS (Söderstjerna et al., Reference Söderstjerna, Johansson, Klefbohm and Englund-Johansson2013). Intense oxidative damage and lipid, protein and DNA peroxidation are observed in mice after intranasal administration of TiO2 NPs (Ze et al., Reference Ze, Sheng, Zhao, Ze, Wang, Zhou, Liu, Yuan, Gui, Sang, Sun, Hong, Yu, Wang, Li and Hong2014). Ferromagnetic mineral magnetite (Fe(3)O(4)) NPs cause structural changes of microtubule and tau protein, essential in the memory mechanism and memory dysfunction (Dadras et al., Reference Dadras, Riazi, Afrasiabi, Naghshineh, Ghalandari and Mokhtari2013). Ozone, the main component of photochemical pollution in urban areas has been shown to alter adult neurogenesis and produce progressive hippocampal neurodegeneration along with memory deficits in rats exposed to low subchronic concentrations (Rivas-Arancibia et al., Reference Rivas-Arancibia, Guevara-Gúzman, Lopez-Vidal, Rodriguez-Martinez, Zanardo-Gomez, Angoa-Pérez and Raisman-Vozari2010).

Clinically healthy urban children from MCMA selected by stringent criteria exhibited structural, neurophysiological and cognitive detrimental effects compared with matched SES, gender, age and mother’s IQ low pollution exposed children (Calderón-Garcidueñas et al., 2008Reference Calderón-Garcidueñas, Mora-Tiscareño, Ontiveros, Gómez-Garza, Barragán-Mejía, Broadway, Chapman, Valencia-Salazar, Jewells, Maronpot, Henríquez-Roldán, Pérez-Guillé, Torres-Jardón, Herrit, Brooks, Osnaya-Brizuela, Monroy, González-Maciel, Reynoso-Robles, Villarreal-Calderon, Solt and Englea; Calderón-Garcidueñas et al., Reference Calderón-Garcidueñas, D’Angiulli, Kulesza, Torres-Jardón, Osnaya, Romero, Keefe, Herritt, Brooks, Avila-Ramirez, Delgado-Chávez, Medina-Cortina and González-González2011 Reference Calderón-Garcidueñas, D’Angiulli, Kulesza, Torres-Jardón, Osnaya, Romero, Keefe, Herritt, Brooks, Avila-Ramirez, Delgado-Chávez, Medina-Cortina and González-Gonzáleza). The cognitive deficits in MC children matched the magnetic resonance imaging (MRI) volumetric changes in their right parietal and bilateral temporal areas (Calderón-Garcidueñas et al., Reference Calderón-Garcidueñas, Kavanaugh, Block, D’Angiulli, Delgado-Chávez, Torres-Jardón, González-Maciel, Reynoso-Robles, Osnaya, Villarreal-Calderon, Guo, Hua, Zhu, Perry and Diaz2012b). Highly exposed children without white matter hyperintensities (WMH−) displayed the profile of classical pro-inflammatory defensive responses: high IL-12, production of powerful pro-inflammatory cytokines and low concentrations of key cytokines and chemokines associated with neuroprotection. Conversely, children with WMH+ exhibited a response involved in resolution of inflammation, immunoregulation and tissue remodeling. The WMH+ group responded to the air pollution-associated brain volumetric alterations with white and gray matter volume increases in temporal, parietal and frontal regions and better cognitive performance compared with the WMH− group. We conclude that complex modulation of cytokines and chemokines influences children’s CNS structural and volumetric responses and cognitive correlates resulting from environmental pollution exposures. Regardless of the presence of prefrontal WMH, MC children performed more poorly across a variety of cognitive tests, compared with control children.

We have identified a number of abnormalities also within the auditory brainstem nuclei in children exposed to severe air pollution. Specifically, we have observed that neuronal cell bodies within the medial superior olive (MSO) are significantly smaller and more round than those in age-matched control brains (Calderón-Garcidueñas et al., Reference Calderón-Garcidueñas, Engle, Mora-Tiscareño, Styner, Gomez-Garza, Zhu, Jewells, Torres-Jardón, Romero, Monroy-Acosta, González-González, Medina-Cortina and D'Angiulli2011b). We interpret this dysmorphology to indicate injury and dysfunction in the MSO. The MSO is the largest nucleus within the human superior olivary complex and has clear roles in localization of sound sources, encoding temporal features of sound and likely plays an important role in brainstem encoding of speech. Incidentally, similar morphological alterations were observed in autistic children (Kulesza and Mangunay, Reference Kulesza and Mangunay2008; Kulesza et al., Reference Kulesza, Lukose and Stevens2011). Confirming brainstem pathology, MCMA children showed clearly abnormal BAEPs with delays in wave III and wave V but no delay in wave I. These findings are consistent with delayed central conduction time of brainstem neural transmission, and increased risk for auditory and vestibular impairment (Calderón-Garcidueñas et al., Reference Calderón-Garcidueñas, Engle, Mora-Tiscareño, Styner, Gomez-Garza, Zhu, Jewells, Torres-Jardón, Romero, Monroy-Acosta, González-González, Medina-Cortina and D'Angiulli2011b). We are assessing the integrity of the auditory system in highly exposed children through a number of noninvasive techniques, such as BAEPs, otoacoustic emissions, speech recognition tasks and listening in background noise.

It is clear that air pollution exposed children experience a chronic, intense state of oxidative stress and exhibit an early brain imbalance in genes involved in inflammation, innate and adaptive immune responses, cell proliferation and apoptosis. Neuroinflammation, endothelial activation and the breakdown of the BBB contribute to cognitive impairment and pathogenesis and pathophysiology of neurodegenerative states (Roher et al., Reference Roher, Debbins, Malek-Ahmandi, Chen, Pipe, Maze., Belden, Maarout, Thiyyagura, Mo, Hunter, Kokjohn, Walker, Kruchowsky, Belohlavek, Sabbagh and Beach2012; Jian et al., Reference Jian, Yi-Fang, Qi, Xiai-Song and Gui-Yun2013). The associations between cognition and urban pollution has been established in cities like Boston, where black carbon – a marker for traffic PM – predicted decreased cognitive function across assessments of verbal and nonverbal intelligence and memory in nine-year-olds (Suglia et al., Reference Suglia, Gryparis, Wright, Schwartz and Wright2008).

A working framework for prevention and intervention

Although genetic factors play a key role in CNS responses (as evidenced by the acceleration of neurodegenerative pathology in children carrying an APOE 4 allele), studies such the above mentioned ones in Boston and others, sketch a complex scenario where air pollution and SES can influence neural development and cognition, as well as genetics, nutrition, access to a cognitively stimulating environment, thereby influencing and determining mental health, academic achievements and overall life performance (D’Angiulli et al., Reference D’Angiulli, Warburton, Dahinten and Hertzman2009; Siddique et al., Reference Siddique, Banerjee, Ray and Lahiri2011; Calderón-Garcidueñas & Torres-Jardón, Reference Calderón-Garcidueñas and Torres-Jardón2012a; Becerra et al., Reference Becerra, Wilhelm, Olsen, Cockburn and Ritz2013).

Thus, identification of children at risk for cognitive deficits, brain structural/volumetric and neurodegenerative accelerating changes should be prioritized in populations exposed to significant concentrations of air pollutants. There is growing public concern about the direct and indirect influences air pollution may have on several developmental outcomes such as school performance, behavioral changes and mood disorders in children and teens. Moreover, childhood aggression and teen delinquency are increasing in megacities, establishing early environmental health risk factors for violence prediction and prevention (Haynes et al., Reference Haynes, Chen, Ryan, Succop, Wright and Dietrich2011; Liu, Reference Liu2011) in populations at risk will be absolutely critical. New concerns involve the association between air pollution exposure and increased risk of attention deficit hyperactive disorder (Siddique et al., Reference Siddique, Banerjee, Ray and Lahiri2011) and autism (Becerra et al., Reference Becerra, Wilhelm, Olsen, Cockburn and Ritz2013) in young and older children.

Strong support for the need of neurocognitive screening comes from a growing psychological and epidemiological literature suggesting evidence of suboptimal cognitive functioning across the developmental span in clinically healthy children (Guxens and Sunyer, Reference Guxens and Sunyer2012; Calderón-Garcidueñas et al., Reference Calderón-Garcidueñas, Kavanaugh, Block, D’Angiulli, Delgado-Chávez, Torres-Jardón, González-Maciel, Reynoso-Robles, Osnaya, Villarreal-Calderon, Guo, Hua, Zhu, Perry and Diaz2012b). Importantly, a significant proportion of urban schools are situated near major traffic-related air pollution sources (Amram et al., Reference Amram, Abernethy, Brauer, Davies and Allen2011; Amato et al., Reference Amato, Rivas, Viana, Moreno, Bouso, Reche, Alvarez-Pedrerol, Alastuey, Sunyer and Querol2014), and cognitive outcomes may be partly associated with air pollution levels around schools (Mohai et al., Reference Mohai, Kweon, Lee and Ard2011).

Consistent with these observations, the National Institute of Environmental Health Sciences/National Institute of Health panel on outdoor air pollution indicated cognitive, neuropsychological (and possibly neuroimaging) screening of children as one of the priority target areas for future research advocating a multidisciplinary collaborative approach wherein brain-related cognitive (henceforth, neurocognitive) development testing would have a prominent role (Block et al., Reference Block, Elder, Auten, Bilbo, Chen, Chen, Cory-Slechta, Costa, Diaz-Sanchez, Dorman, Gold, Gray, Jeng, Kaufman, Kleinman, Kirshner, Lawler, Miller, Nadadur, Ritz, Semmens, Tonelli, Veronesi, Wright and Wright2012).

Hence, the use of standard neurocognitive tasks to screen clinically healthy children in schools or pediatric offices seems not only desirable but also highly beneficial. Because the primary goal would be to identify children at risk, possibly at the largest scale, the first step could be to screen entire schools in different neighborhoods with different air pollutant profiles. Multidisciplinary approaches for early risk identification could include using air pollution databases from available monitoring stations to gather: (1) Air pollutant profiles in selected geographic area; (2) exposures to traffic-related air pollutants at each child’s school and current residence with land use regression models that combine a geographic information system with ambient passive monitoring in the target area; (3) cumulative ambient exposures to fine PM2.5; (4) robust baseline information on the oxidative potential and metal content of PM found in the targeted regions; (5) cognitive screening first and, when applicable, more elaborate neurocognitive/neurophysiological follow-ups, which could include EEG/ERPs, BAEPs, MRI, f MRI and MRS.

The first goal in targeted areas will be to define the cohorts with the most risk for neurocognitive deficits based on traffic emissions, fixed sources of contaminants, profile of toxic pollutant components (ie, metals) and cumulative concentrations of fine PM. The initial studies should be followed by interventions aimed at breaking the cycle of air pollution, indoor air pollution, tobacco use, high body mass index, low fruit and vegetable intake and physical inactivity. Since health risk factors are more likely to have a toll on low SES children, identification of spatial concentrations of low SES pediatric populations at risk, should be prioritized (Forastiere et al., Reference Forastiere, Stafoggia, Tasco, Picciotto, Agabiti, Casaroni and Perucci2007; Ou et al., Reference Ou, Hedley, Chung, Thach, Chau, Chan, Yang, Ho, Wong and Lam2008).

According to the American Neuropsychiatric Association and the Shulman criteria (Malloy et al., Reference Malloy, Cummings, Coffey, Duffy, Fink, Lauterbach, Lovell, Royall and Salloway1997; Shulman, Reference Shulman2000), the ideal cognitive screening instrument should (i) be relatively brief to administer; (ii) be easy to score; (iii) well tolerated and accepted by participants, in our cases both child and parent; (iv) test all targeted cognitive domains; (v) be valid and reliable; and (vi) relatively independent of or controlling for age and education.

A possible battery of tests (easily translatable in many languages) for the initial screening of school-aged children is described in Table 2. The measures in this table have reference to previous neuroimaging findings implicating functional, maturational or structural correlates of frontal, parietal and temporal regions and/or neurocognitive tests applied to air pollution studies in children as seen on Table 1. Because several studies link risk of negative effects of air pollution to early development, from infancy to preschool, screening could start in daycare or during transition from kindergarten to first grade, for example using tools such as the Early Development Instrument (D’Angiulli et al., Reference D’Angiulli, Warburton, Dahinten and Hertzman2009). With advances in wireless neuroimaging technology, neurocognitive screening may even incorporate rapid EEG/ERP recording procedures.

Table 2 Suggested battery of neuropsychological and psychoeducational tests for the initial screening of school-aged children and teens exposed to urban air pollution

In summary, air pollution effects on the developing brain may vary along a continuum from minor subclinical deficits in cognitive functioning to significant cognitive deficits that are identified readily by parents and/or teachers. The detrimental effects may also worsen with the age of the child, thus selected neurocognitive tools ought to be useful for longitudinal studies, across educational backgrounds and expecting overlaps in the functional areas and tests affected. Complex cognitive responses that may be affected include attention and short-term memory, information processing speed and executive function, verbal abstraction and visuospatial and motor skills. We should also expect deficits in auditory and vestibular responses and sound localization, along with olfaction deficits. The diffuse nature of the neuroinflammation and the neurodegenerative changes observed in exposed children obligates us not to rely on a single study or measure but rather to employ a weight of evidence approach incorporating current clinical, neurophysiological, radiological and epidemiological research as well as the results of animal exposure studies to single pollutants/mixtures/or pollutant components. Inflammatory biomarkers play a key role in the identification of children with positive volumetric and cognitive responses to their lifelong pollutant exposures (Calderón-Garcidueñas et al., Reference Calderón-Garcidueñas, Kavanaugh, Block, D’Angiulli, Delgado-Chávez, Torres-Jardón, González-Maciel, Reynoso-Robles, Osnaya, Villarreal-Calderon, Guo, Hua, Zhu, Perry and Diaz2012b) and since neuroinflammation/vascular damage/neurodegeneration go hand in hand (Calderón-Garcidueñas et al., 2013Reference Calderón-Garcidueñas, Franco-Lira, Mora-Tiscareño, Medina-Cortina, Torres-Jardón and Kavanaugha and 2013Reference Calderón-Garcidueñas, Cross, Franco-Lira, Aragón-Flores, Kavanaugh, Torres-Jardón, Chao, Thompson, Chang, Zhu and D’Angiullib), definition of inflammatory/endothelial dysfunction biomarkers establishing an association between brain growth and cognition are urgently needed.

Of course, in light of the findings reviewed in the present paper, a pressing important question that jumps to mind is whether the prevalence of AD/PD is increased in MCMA? Unfortunately we do not have accurate records from health institutions. Mexico health care is covered by both private and government hospitals and institutes and there is no universal statistical information regarding AD and PD prevalence. Empirically, however, neurologists and general practitioners report a significant increase in the number of cases for both neurodegenerative diseases (Victor Esquivel MD, Neurologist, personal communication). Death certificates that would be a source of information, unfortunately fail to list AD or PD in the certificate, basically because families are reluctant to link their patients to a disease with genetic implications, and so only allowed for the writing of acute causes of death like pneumonia, but not the base disease. Therefore, the answer to the central issue of incidence of AD/PD in MCMA is very much an open empirical question for future research.

Our ultimate goal is to protect exposed children through multidimensional interventions yielding both impact and reach: cognitive (Diamond and Lee, Reference Diamond and Lee2011), family participation (Josephson, Reference Josephson2013) and modifiable lifestyle factors such as diet and micronutrient supply (Villarreal-Calderon et al., Reference Villarreal-Calderón, Torres-Jardon, Palacios-Moreno, Perez-Guille, Maronpot, Reed, Zhu and Calderón-Garciduenas2010; Calderón-Garcidueñas et al., Reference Calderón-Garcidueñas, Mora-Tiscareño, Styner, Gómez-Garza, Zhu, Torres-Jardón, Carlos, Solorio-López, Medina-Cortina, Kavanaugh and D’Angiulli2012c).

Air pollution brain effects on children and teens ought to be key public health targets.

Key points to remember

  • Risk for stroke and depression are associated with common ambient air pollutants, including fine and UFPM and ozone.

  • The stroke risk is present even at pollutant concentrations below the current standards, meaning there is a wide spectrum of susceptibility to pollutants likely related to factors such as the presence of chronic diseases, and genetic and nutrition variables.

  • Stroke is not a stroke, specific patient characteristics modify associations between air pollution and ischemic stroke (Villeneuve et al., Reference Villeneuve, Johnson, Pasichnvk, Lowes, Kirkland and Rowe2012).

  • Check for factors that will aggravate the neurological/psychiatric effects of air pollution such as diabetes, hypertension, infectious processes, residency in close proximity to a busy road, changes in occupation, etc.

  • Keep an eye on the air quality index of your city, record proximity of the patient to high density traffic (Mohai et al., Reference Mohai, Kweon, Lee and Ard2011; van Kempen et al., Reference Van Kempen, Fischer, Janssen, Houthuijis, van Kamp, Stansfeld and Cassee2012) or fixed sources of pollutants, check for acute events such as forest fires or the arrival of fumes from a distant area (Chen et al., Reference Chen, Villeneuve, Rowe, Liu and Stieb2013).

  • PM occupational exposures and tobacco are to be taken into consideration as important sources of pollutants.

  • The patient’s occupation and hobbies are important. Is she an outdoor person? Is he protected against toxic substances at work? Is there a significant source of PM or NPs at the office? Where is the patient from? A good clinical history is a must.

  • Parents and school officials should be aware of the high pollutant concentrations in their area and keep children indoors.

  • Identification of children at risk for cognitive deficits, brain structural/volumetric and neurodegenerative accelerating changes should be prioritized in populations exposed to significant concentrations of air pollutants.

  • Brain damage is cumulative and has deleterious effects on cognitive, emotional and behavioral areas.

  • Preventive measures against the harmful effects of environmental pollution should be referred to and applied as early as the conception time.

  • Finally, we must consider whether we want to pay the high price that involves ignoring the serious health impacts of environmental pollution.

Acknowledgment

Gratitude is due to all children and their families who have participated in our clinical studies.

Financial Support

None.

Conflicts of Interest

None.

Ethical Standards

The authors assert that all procedures contributing to this work comply with the ethical standards of the institutional guidelines based on the Helsinki Declaration of 1975, as revised in 2008.

References

Aiken, A.C., Salcedo, D., Cubison, M.J., Huffman, A. and DeCarlo, P.E. 2009: Mexico City aerosol analysis during MILAGRO using high resolution aerosol mass spectrometry at the urban supersite (T0) e part 1: fine particle composition and organic source apportionment. Atmospheric Chemistry and Physics 9, 66336653.Google Scholar
Amato, F., Rivas, I., Viana, M., Moreno, T., Bouso, L., Reche, C., Alvarez-Pedrerol, M., Alastuey, A., Sunyer, J., Querol, X. 2014: Sources of indoor and outdoor PM2.5 concentrations in primary schools. Science of the Total Environment 490C, 757765.Google Scholar
Amram, O., Abernethy, R., Brauer, M., Davies, H. and Allen, R.W. 2011: Proximity of public elementary schools to major roads in Canadian urban areas. International Journal of Health Geographics 10, 68.Google Scholar
Annesi-Maesano, J., Baiz, N., Banerjee, S., Rudnai, P. and Rive, S., SINPHONIE Group. 2013: Indoor air quality and sources in schools and related health effects. Journal of Toxicology and Environmental Health. Part B Critical Reviews 16, 491550.Google Scholar
Banerjee, M., Siddique, S., Dutta, A., Mukherjee, B. and Ray, M.R. 2012: Cooking with biomass increases the risk of depression in pre-menopausal women in India. Social Science & Medicine 75, 565572.Google Scholar
Bauman, M.D., Iosif, A.M., Smith, S.E., Bregere, C., Amaral, D.G. and Patterson, P.H. 2013: Activation of the maternal immune system during pregnancy alters behavioral development of Rhesus monkey offspring. Biological Psychiatry 75, 332341.Google Scholar
Becerra, T.A., Wilhelm, M., Olsen, J., Cockburn, M. and Ritz, B. 2013: Ambient air pollution and autism in Los Angeles county, California. Environmental Health Perspectives 121, 80386.Google Scholar
Bedada, G.B., Smith, C.J., Tyrrell, P.J., Hirst, A.A. and Agius, R. 2012: Short-term effects of ambient particulates and gaseous pollutants on the incidence of transient ischemic attack and minor stroke: a case-crossover study. Environmental Health. doi: 10.1186/1476-069X-11-77.Google Scholar
Beelen, R., Raaschou-Nielsen, O., Stafoggia, M., Andersen, Z.J. 2013: Effects of long-term exposure to air pollution on natural-cause mortality: an analysis of 22 European cohorts within the multicenter ESCAPE project. Lancet 383, 785795.Google Scholar
Bell, M., Davis, D.L. and Fletcher, T. 2004: A retrospective assessment of mortality from the London smog episode of 1952: the role of influenza and pollution. Environmental Health Perspectives 112, 68.Google Scholar
Bell, M.L., Goldberg, R., Hogrefe, C., Kinney, P.L., Knowlton, K., Lynn, B., Rosenthal, J., Rosenzweig, C. and Patz, J.A. 2007: Climate change, ambient ozone, and health in 50 US cities. Climatic Change 82, 6176.Google Scholar
Benn, T., Cavanagh, B., Hristovski, K., Posner, J.D. and Westerhoff, P. 2010: The release of nanosilver from consumer products used in the home. Journal of Environmental Quality 39, 18751882.Google Scholar
Bergin, I.L. and Witzmann, F.A. 2013: Nanoparticle toxicity by the gastrointestinal route: evidence and knowledge gaps. International Journal of Biomededical Nanoscience and Nanotechnology 3, 12.Google Scholar
Bills, C.B., Levy, N.A., Sharma, V., Charney, D.S., Herbert, R., Moline, J. and Katz, C.L. 2008: Mental health of workers and volunteers responding to events of 9/11: review of the literature. Mount Sinai Journal of Medicine 75, 115127.Google Scholar
Blasko, I., Stampfer-Kountchev, M., Robatscher, P., Veerhuis, R., Eikelenboom, P. and Grubeck-Loebenstein, B. 2004: How chronic inflammation can affect the brain and support the development of Alzheimer’s disease in old age: the role of microglia and astrocytes. Aging Cell 3, 169176.Google Scholar
Block, M.L. and Calderón-Garcidueñas, L. 2009: Air pollution: mechanisms of neuroinflammation and CNS disease. Trends in Neurosciences 32, 506516.Google Scholar
Block, M.L., Elder, A., Auten, R.L., Bilbo, S.D., Chen, H., Chen, J.C., Cory-Slechta, D.A., Costa, D., Diaz-Sanchez, D., Dorman, D.C., Gold, D.R., Gray, K., Jeng, H.A., Kaufman, J.D., Kleinman, M.T., Kirshner, A., Lawler, C., Miller, D.S., Nadadur, S.S., Ritz, B., Semmens, E.O., Tonelli, L.H., Veronesi, B., Wright, R.O., Wright, R.J. 2012: The outdoor air pollution and brain health workshop. Neurotoxicology 33, 972984.Google Scholar
Bonotis, K., Krikki, E., Holeva, V., Aggouridaki, C., Costa, V. and Baloyannis, S. 2008: Systemic immune aberrations in Alzheimer’s disease patients. Journal of Neuroimmunology 193, 183187.Google Scholar
Braak, H., Ghebremedhin, E., Rüb, U., Bratzke, H. and Del Tredeci, K. 2004: Stages in the development of Parkinson’s disease-related pathology. Cell and Tissue Research 318, 121134.Google Scholar
Braak, H., Thal, D.R., Ghebremedhin, E. and Del Tredeci, K. 2011: Stages of the pathological process in Alzheimer’s disease: age categories from 1 to 100 years. The Journal of Neuropathology & Experimental Neurology 70, 960969.Google Scholar
Brackbill, R.M., Thorpe, L.E., DiGrande, L., Perrin, M., Sapp, J.H. 2nd, Wu, D., Campolucci, S., Walker, D.J., Cone, J., Pulliam, P., Thalji, L., Farfel, M.R. and Thomas, P. 2006: Surveillance for World Trade Center disaster health effects among survivors of collapsed and damaged buildings. MMWR Surveillance Summeries 55, 118.Google Scholar
Bravo-Alvarez, H. and Torres-Jardón, R. 2002: Air pollution levels and trends in the México City metropolitan area. In Fenn, M.E., de Bauer, L.I. and Hernández-Tejeda, T., editors, Urban air pollution and forest: resources at risk in the Mexico City air basin. Ecological Studies, Chapter 6, Volume 156. New York: Springer-Verlag, 121159.Google Scholar
Brook, R., Rajagopala, S., Pope, A., Brook, J.R., Bhatnagar, A., Diez-Roux, A.V., Holguin, F., Hong, Y., Luepker, R.V., Mittleman, M.A., Peters, A., Siscovick, D., Smith, S.C., Whitse, L. and Kaufman, J. 2010: Particulate matter air pollution and cardiovascular disease: an update to the scientific statement from the American Heart Association. Circulation 12, 23312378.Google Scholar
Brown, K.W., Sarnat, J.A. and Koutrakis, K. 2012: Concentrations of PM2.5 mass and components in residential and non-residential indoor microenvironments: the Source and Composition of particulate exposure study. Journal of Exposure Science & Environmental Epidemiology 22, 161172.Google Scholar
Brun, E., Carrière, M. and Mabondzo, A. 2012: In vitro evidence of dysregulation of blood-brain barrier function after acute and repeated/long term exposure to TiO2 nanoparticles. Biomaterials 33, 886896.Google Scholar
Calderón-Garcidueñas, L., Mora-Tiscareño, A., Fordham, L.A., Valencia-Salazar, G., Chung, C.J., Rodriguez-Alcaraz, A., Paredes, R., Variakojis, D., Villarreal-Calderón, A., Flores-Camacho, L., Antunez-Solis, A., Henríquez-Roldán, C. and Hazucha, M.J. 2003: Respiratory damage in children exposed to urban pollution. Pediatric Pulmonology 36, 148161.Google Scholar
Calderón-Garcidueñas, L., Mora-Tiscareño, A., Ontiveros, E., Gómez-Garza, G., Barragán-Mejía, G., Broadway, J., Chapman, S., Valencia-Salazar, G., Jewells, V., Maronpot, R.R., Henríquez-Roldán, C., Pérez-Guillé, B., Torres-Jardón, R., Herrit, L., Brooks, D., Osnaya-Brizuela, N., Monroy, M.E., González-Maciel, A., Reynoso-Robles, R., Villarreal-Calderon, R., Solt, A.C. and Engle, R.W. 2008a: Air pollution, cognitive deficits and brain abnormalities: a pilot study with children and dogs. Brain and Cognition 68, 117127.Google Scholar
Calderón-Garcidueñas, L., Villarreal-Calderon, R., Valencia-Salazar, G., Henríquez-Roldán, C., Gutiérrez-Castrellón, P., Torres-Jardón, R., Osnaya-Brizuela, N., Romero, L., Torres-Jardón, R., Solt, A. and Reed, W. 2008b: Systemic inflammation, endothelial dysfunction, and activation in clinically healthy children exposed to air pollutants. Inhalation Toxicology 20, 499506.Google Scholar
Calderón-Garcidueñas, L., Solt, A., Franco-Lira, M., Torres-Jardón, R., Nuse, B., Herritt, L., Villarreal-Calderón, R., Osnaya, N., Stone, I., García, R., Brooks, D.M., González-Maciel, A., Reynoso-Robles, R., Delgado-Chávez, R. and Reed, W. 2008c: Long-term air pollution exposure is associated with neuroinflammation, an altered innate immune response, disruption of the blood-brain-barrier, ultrafine particle deposition, and accumulation of amyloid beta 42 and alpha synuclein in children and young adults. Toxicologic Pathology 36, 289310.Google Scholar
Calderón-Garcidueñas, L., Macias-Parra, M., Hoffmann, H.J., Valencia-Salazar, G., Henríquez-Roldán, C., Monte, O.C., Barragán-Mejía, G., Villarreal-Calderon, R., Romero, L., Granada-Macías, M., Torres-Jardón, R., Medina-Cortina, H. and Maronpot, R.R. 2009: Immunotoxicity and environment: immunodysregulation and systemic inflammation in children. Toxicologic Pathology 37, 161169.Google Scholar
Calderón-Garcidueñas, L., Franco-Lira, M., Henríquez-Roldán, C., González-Maciel, A., Reynoso-Robles, R., Villarreal-Calderon, R., Herritt, L., Brooks, D., Keefe, S., Palacios-Moreno, J., Torres-Jardón, R., Medina-Cortina, H., Delgado-Chávez, R., Aiello-Mora, M., Maronpot, R.R. and Doty, R.L. 2010: Urban air pollution: influences on olfactory function and pathology in exposed children and young adults. Experimental and Toxicologic Pathology 62, 91102.Google Scholar
Calderón-Garcidueñas, L., D’Angiulli, A., Kulesza, R.J., Torres-Jardón, R., Osnaya, N., Romero, L., Keefe, S., Herritt, L., Brooks, D.M., Avila-Ramirez, J., Delgado-Chávez, R., Medina-Cortina, H. and González-González, L.O. 2011a: Air pollution is associated with brainstem auditory nuclei pathology and delayed brainstem auditory evoked potentials. International Journal of Developmental Neuroscience 29, 365375.Google Scholar
Calderón-Garcidueñas, L., Engle, R., Mora-Tiscareño, A., Styner, M., Gomez-Garza, G., Zhu, H., Jewells, V., Torres-Jardón, R., Romero, L., Monroy-Acosta, M.E., González-González, L.O., Medina-Cortina, H. and D'Angiulli, A. 2011b: Exposure to severe urban air pollution influences cognitive outcomes, brain volume and systemic inflammation in clinically healthy children. Brain and Cognition 77, 345355.Google Scholar
Calderón-Garcidueñas, L. and Torres-Jardón, R. 2012a: Air pollution, socioeconomic status and children’s cognition in megacities: the Mexico City scenario. Frontiers in Psychology 3, 217.Google Scholar
Calderón-Garcidueñas, L., Kavanaugh, M., Block, M.L., D’Angiulli, A., Delgado-Chávez, R., Torres-Jardón, R., González-Maciel, A., Reynoso-Robles, R., Osnaya, N., Villarreal-Calderon, R., Guo, R., Hua, Z., Zhu, H., Perry, G. and Diaz, P. 2012b: Neuroinflammation, hyperphosphorilated tau, diffuse amyloid plaques and down-regulation of the cellular prion protein in air pollution exposed children and adults. Journal of Alzheimer’s Disease 28, 93107.Google Scholar
Calderón-Garcidueñas, L., Mora-Tiscareño, A., Styner, M., Gómez-Garza, G., Zhu, H., Torres-Jardón, R., Carlos, E., Solorio-López, E., Medina-Cortina, H., Kavanaugh, M. and D’Angiulli, A. 2012c: White matter hyperintensities, systemic inflammation, brain growth, and cognitive functions in children exposed to air pollution. Journal of Alzheimer’s Disease 31, 183191.Google Scholar
Calderón-Garcidueñas, L., Franco-Lira, M., Mora-Tiscareño, A., Medina-Cortina, H., Torres-Jardón, R. and Kavanaugh, M. 2013a: Early Alzheimer’s and Parkinson’s disease pathology in urban children: friend versus foe responses – it is time to face the evidence. Biomed Research International. doi: 10.1155/2013/161687.Google Scholar
Calderón-Garcidueñas, L., Cross, J.V., Franco-Lira, M., Aragón-Flores, M., Kavanaugh, M., Torres-Jardón, R., Chao, C.K., Thompson, C., Chang, J., Zhu, H. and D’Angiulli, A. 2013b: Brain immune interactions and air pollution: macrophage inhibitory factor (MIF), prion cellular protein (PrPc), interleukin-6 (IL6), interleukin 1 receptor antagonist (IL-1Ra) and interleukin-2 (IL2) in cerebrospinal fluid and MIF in serum differentiate urban children exposed to severe v low air pollution. Frontiers in Neuroscience 7, 183.Google Scholar
Campbell, A. 2004: Inflammation, neurodegenerative diseases and environmental exposures. Annals of the New York Academy of Sciences 1035, 117132.Google Scholar
Cappelli, M., Gray, C., Zemek, R., Cloutier, P., Kennedy, A., Glennie, E., Doucet, G., Lyons, J.S. 2012: The HEADS-ED: a rapid mental health screening tool for pediatric patients in the emergency department. Pediatrics 130, e321327.Google Scholar
Carlsen, H.K., Forsberg, B., Meister, K., Gislason, T. and Oudin, A. 2013: Ozone is associated with cardiopulmonary and stroke emergency hospital visits in Reykjavík, Iceland 2003-2009. Environmental Health. doi: 10.1186/1476-069X-12-28.Google Scholar
Castellani, R.J., Zhu, X., Zhu, X., Moreira, P.I., Perry, G. and Smith, M.A. 2006: Involvement of oxidative stress in Alzheimer disease. The Journal of Neuropathology & Experimental Neurology 65, 631641.Google Scholar
Clark, C., Crombie, R., Head, J., van Kamp, I., van Kempen, E. and Stansfeld, S.A. 2012: Does traffic-related air pollution explain associations of aircraft and road traffic noise exposure on children’s health and cognition? A secondary analysis of the United Kingdom sample from the RANCH project. American Journal of Epidemiology 176, 327337.Google Scholar
Cunningham, C., Campion, S., Lunnon, K., Murray, C.L., Woods, J.F., Deacon, R.M., Rawlins, J.N. and Perry, V.H. 2009: Systemic inflammation induces acute behavioral and cognitive changes and accelerates neurodegenerative disease. Biological Psychiatry 65, 304312.Google Scholar
Chen, B. and Kan, H. 2008: Air pollution and population health: a global challenge. Environmental Health and Preventive Medicine 13, 94101.Google Scholar
Chen, L., Villeneuve, P.J., Rowe, B.H., Liu, L. and Stieb, D.M. 2013: The Air Quality Health Index as a predictor of emergency department visits for ischemic stroke in Edmonton, Canada. Journal of Exposure Science & Environmental Epidemiology 24, 358364.Google Scholar
D’Angiulli, A., Warburton, W., Dahinten, S. and Hertzman, C. 2009: Population-level associations between preschool vulnerability and grade-four basic skills. PLoS One 4, e7692.Google Scholar
Dadras, A., Riazi, G.H., Afrasiabi, A., Naghshineh, A., Ghalandari, F. and Mokhtari, F. 2013: In vitro study of the alterations of brain tubulin structure and assembly affected by magnetite nanoparticles. Journal of Biological Inorganic Chemistry 18, 357369.Google Scholar
Davis, D.A., Bortolato, M., Godar, S.C., Sander, T.K., Iwata, N., Pakbin, P., Shib, J.C., Berhane, K., McConnell, R., Sioutas, C., Finch, C.E., Morgan, T.E. and Block, M. 2013: Prenatal exposure to urban air nanoparticles in mice causes altered neuronal differentiation and depression-like responses. PLoS One 8, e64128.Google Scholar
Diamond, A. and Lee, K. 2011: Interventions shown to aid executive function development in children 4 to 12 years old. Science 333, 959964.Google Scholar
Dik, M.G., Jonker, C., Hack, C.E., Smit, J.H., Comijs, H.C. and Eikelenboom, P. 2005: Serum inflammatory proteins and cognitive decline in older persons. Neurology 64, 13711377.Google Scholar
Doty, R.L. 2012: Olfactory dysfunction in Parkinson disease. Nature Reviews. Neurology 8, 329339.Google Scholar
Eikelenboom, P., van Exel, E., Veerhuis, R., Rozemuller, A.J., van Gool, W.A. and Hoozemans, J.J. 2011: Innate immunity and the etiology of late-onset Alzheimer’s disease. Neurodegenerative Disease 10, 271273.Google Scholar
Finch, C.E. and Morgan, T.E. 2007: Systemic inflammation, infection, ApoE alleles and Alzheimer disease: a position paper. Current Alzheimer Research 4, 185189.Google Scholar
Fonken, L.K., Xu, X., Weil, Z.M., Chen, G., Sun, Q., Rajagopalan, S. and Nelson, R.J. 2011: Air pollution impairs cognition, provokes depressive-like behaviors and alters hippocampal cytokine expression and morphology. Molecular Psychiatry 16, 987995.Google Scholar
Forastiere, F., Stafoggia, M., Tasco, C., Picciotto, S., Agabiti, N., Casaroni, G., Perucci, C.A. 2007: Socioeconomic status, particulate air pollution, and daily mortality: differential exposure. American Journal of Industrial Medicine 50, 208216.Google Scholar
Freire, C., Ramos, R., Puertas, R., Lopez-Espinoza, M.J., Julyez, J., Aguilera, I., Cruz, F., Fernandez, M.F., Sunyer, J. and Olea, N. 2010: Association of traffic-related air pollution with cognitive development in children. Journal of Epidemiology and Community Health 64, 223228.Google Scholar
Gomez-Ravetti, M. and Moscato, P. 2008: Identification of a 5-protein biomarker molecular signature for predicting Alzheimer’s disease. PLoS One 3, e3111.Google Scholar
Guo, L., Zhu, N., Guo, Z., Li, G.K., Chen, C., Sang, N. and Yao, Q.C. 2012: Particulate matter (PM10) exposure induces endothelial dysfunction and inflammation in rat brain. Journal of Hazardous Materials 213–214, 2837.Google Scholar
Guxens, M. and Sunyer, J. 2012: A review of epidemiological studies on neuropsychological effects of air pollution. Swiss Medical Weekly 141, w13322.Google Scholar
Habre, R., Coull, B., Moshier, E., Godbold, J., Nath, A., Castro, W., Schachter, N., Rohr, A., Kattan, M., Spengler, J. and Koutrakis, P. 2013: Sources of indoor air pollution in New York City residences of asthmatic children. Journal of Exposure Science & Environmental Epidemiology 24, 269278.Google Scholar
Hagens, W.I., Oomen, A.G., de Jong, W.H., Cassee, F.R. and Sips, A.J. 2007: What do we (need to) know about the kinetic properties of nanoparticles in the body? Regulatory Toxicology and Pharmacology 49, 217229.Google Scholar
Haynes, E.N., Chen, A., Ryan, P., Succop, P., Wright, J. and Dietrich, K.N. 2011: Exposure to airborne metals and particulate matter and risk for youth adjudicated for criminal activity. Environmental Research 111, 12431248.Google Scholar
Hazen, E.P., McDougle, C.J. and Volkmar, F.R. 2013: Changes in the diagnostic criteria for autism in DSM-5: controversies and concerns. The Journal of Clinical Psychiatry 74, 739740.Google Scholar
Herbert, M.R. 2010: Contributions of the environment and environmentally vulnerable physiology to autism spectrum disorders. Current Opinion in Neurology 23, 103110.Google Scholar
Hong, Y.C., Lee, J.T., Kim, H. and Kwon, H.J. 2002: Air pollution: a new risk factor in ischemic stroke mortality. Stroke 33, 21652169.Google Scholar
Howard, G., Wagenknecht, L.E., Cai, J., Cooper, L., Kraut, M.A. and Toole, J.F. 1998: Cigarette smoking and other risk factors for silent cerebral infarction in the general population. Stroke 29, 913917.Google Scholar
INE. 2011: Cuarto Almanaque de Datos y Tendencias de la Calidad del Aire en 20 Ciudades Mexicanas (2000–2009). first edition. Mexico D.F.: Instituto Nacional de Ecología.Google Scholar
Jian, H., Yi-Fang, W., Qi, L., Xiai-Song, H. and Gui-Yun, Z. 2013: Cerebral blood flow and metabolic changes in hippocampal regions of a modified rat model with chronic cerebral hypoperfusion. Acta Neurologica Belgica 113, 313317.Google Scholar
Jordan, H.T., Stellman, S.D., Morabia, A., Miller-Archie, S.A., Alper, H., Laskaris, Z., Brackbill, R.M. and Cone, J.E. 2013: Cardiovascular disease hospitalizations in relation to exposure to the September 11, 2001 World Trade Center disaster and posttraumatic stress disorder. Journal of American Heart Association 2, e000431.Google Scholar
Josephson, A.M. 2013: Family intervention as a developmental psychodynamic therapy. Child and Adolescent Psychiatric Clinics of North America 22, 241260.Google Scholar
Keene, C.D., Cudaback, E., Li, X., Montine, K.S. and Montine, T.J. 2011: Apolipoprotein E isoforms and regulation of the innate immune response in brain of patients with Alzheimer’s disease. Current Opinion in Neurobiology 21, 920928.Google Scholar
Kettunen, J., Lanki, T., Tiittanen, P., Aaalto, P.P., Koskentalo, T., Kulmala, M., Salomaa, V. and Pekkanen, J. 2007: Associations of fine and ultrafine particulate air pollution with stroke mortality in an area of low air pollution levels. Stroke 38, 918922.Google Scholar
Knox, E.G. 1981: Metereological associations of cerebrovascular disease mortality in England and Wales. Journal of Epidemiology Community Health 35, 220223.Google Scholar
Kulesza, R.J. and Mangunay, K. 2008: Morphological features of the medial superior olive in autism. Brain Research 1200, 132137.Google Scholar
Kulesza, R.J. Jr, Lukose, R. and Stevens, L.V. 2011: Malformation of the human superior olive in autistic spectrum disorders. Brain Research 1367, 360371.Google Scholar
Lampron, A., Elali, A. and Rivest, S. 2013: Innate immunity in the CNS: redefining the relationship between the CNS and its environment. Neuron 78, 214232.Google Scholar
Langrish, J.P. and Mills, N.L. 2014: Air pollution and mortality in Europe. Lancet 383, 758760.Google Scholar
Larsson, M., Weiss, B., Janson, S., Sundell, J. and Bornehag, C.G. 2009: Associations between indoor environmental factors and parental-reported autistic spectrum disorders in children 6–8 years of age. Neurotoxicology 30, 822831.Google Scholar
Leiva, G.M.A., Santibañez, D.A., Ibarra, E.S., Matus, C.P. and Seguel, R. 2013: A five-year study of particulate matter (PM2.5) and cerebrovascular diseases. Environmental Pollution 181, 16.Google Scholar
Levesque, S., Surace, M.J., McDonald, J. and Block, M.L. 2011: Air pollution and the brain: subchronic diesel exhaust exposure causes neuroinflammation and elevates early markers of neurodegenerative disease. Journal of Neuroinflammation 24, 105.Google Scholar
Lim, Y.H., Kim, H., Kim, J.H., Bae, S., Park, H.Y. and Hong, Y.C. 2012: Air pollution and symptoms of depression in elderly adults. Environmental Health Perspectives 120, 10231028.Google Scholar
Lisabeth, L.D., Escobar, J.D., Dvonch, J.T., Sanchez, B.N., Majersik, J.J., Brown, D.L., Smith, M.A. and Morgenstern, L.B. 2008: Ambient air pollution and risk for ischemic stroke and transient ischemic attack. Annals of Neurology 64, 5359.Google Scholar
Liu, J. 2011: Early risk factors for violence: conceptualization, review of the evidence and implications. Agression and Violent Behavior 16, 6373.Google Scholar
Maheswarang, R., Pearson, T., Smeeton, N.C., Beevers, S.D., Campbell, M.J. and Wolfe, C.D. 2010: Impact of outdoor air pollution on survival after stroke: population-based cohort study. Stroke 41, 869877.Google Scholar
Malloy, P.F., Cummings, J.L., Coffey, C.E., Duffy, J., Fink, M., Lauterbach, E.C., Lovell, M., Royall, D., Salloway, S. 1997: Cognitive screening instruments in neuropsychiatry: a report of the committee of research of the American Neuropsychiatric Association. Journal of Neuropsychiatry and Clinical Neurosciences 9, 189197.Google Scholar
Marr, L.C., Grogan, L.A., Wohrnschimmel, H., Molina, L.T., Molina, M.J., Smith, T.J. and Garshick, E. 2004: Vehicle traffic as a source of particulate polycyclic aromatic hydrocarbon exposure in Mexico City. Environmental Science and Technology 38, 25842592.Google Scholar
Mohai, P., Kweon, B.S., Lee, S. and Ard, K. 2011: Air pollution around schools is linked to poorer student health and academic performance. Health Affairs 30, 852862.Google Scholar
Molina, J.M., Molina, L.T. 2004: Megacities and atmospheric pollution. Journal of Air & Waste Management Association 54, 644680.Google Scholar
Molina, L.T., Madronich, S.J., Gaffney, J.S., Apel, E., de Foy, B., Fast, J., Ferrare, R., Herndon, S., Jimenez, J.L., Lamb, B., Osornio-Vargas, A.R., Russell, P., Schauer, J.J., Stevens, P.S., Volkamer, R. and Zavala, M. 2010: An overview of the MILAGRO 2006 Campaign: Mexico City emissions and their transport and transformation. Atmospheric Chemistry and Physics 10, 86978760.Google Scholar
O’Donnell, M.J., Fang, J., Mittleman, M.A., Kapral, M.K. and Wellenius, G.A., Investigators of the Registry of Canadian Stroke Network. 2011: Fine particulate air pollution (PM2.5) and the risk of acute ischemic stroke. Epidemiology 22, 422431.Google Scholar
Ou, C.Q., Hedley, A.J., Chung, R.Y., Thach, T.Q., Chau, Y.K., Chan, K.P., Yang, L., Ho, S.Y., Wong, C.M., Lam, T.H. 2008: Socioeconomic disparities in air pollution-associated mortality. Environmental Research 107, 237244.Google Scholar
Ozbay, F., Aud der Heyde, T., Reissman, D. and Sharma, V. 2013: The enduring mental health impact of the September 11th terrorist attacks: challenges and lessons learned. The Psychiatric Clinics of North America 36, 417429.Google Scholar
Parrish, D.D., Singh, H.B., Molina, L. and Madronich, S. 2011: Air quality progress in North American megacities: a review. Atmospheric Environment 45, 70157025.Google Scholar
Perry, G., Nunomura, A., Hirai, K., Zhu, X., Pérez, M., Avila, J., Castellani, R.J., Atwood, C.S., Aliev, G., Sayre, L.M., Takeda, A. and Smith, M.A. 2002: Is oxidative damage the fundamental pathogenic mechanism of Alzheimer’s and other neurodegenerative diseases? Free Radical Biology & Medicine 33, 14751479.Google Scholar
Querol, X., Pey, J., Minguillón, M.C., Pérez, N., Alastuey, A., Viana, M., Moreno, T., Bernabé, R.M., Blanco, S., Cárdenas, B., Vega, E., Sosa, G., Escalona, S., Ruiz, H. and Artiñano, B. 2008: PM speciation and sources in Mexico during the MILAGRO-2006 Campaign. Atmospheric Chemistry and Physics 8, 111128.Google Scholar
Rivas-Arancibia, S., Guevara-Gúzman, R., Lopez-Vidal, Y., Rodriguez-Martinez, E., Zanardo-Gomez, M., Angoa-Pérez, M., Raisman-Vozari, R. 2010: Oxidative stress caused by ozone exposure induces loss of brain repair in hippocampus of adult rats. Toxicological Sciences 113, 187197.Google Scholar
Roberts, A.L., Lyall, K., Hart, J.E., Laden, F., Just, A.C., Bobb, J.F., Koenen, K.C., Ascherio, A. and Weisskoof, M.G. 2013: Perinatal air pollutant exposures and autism spectrum disorder in the children of nurses’ health study II participants. Environmental Health Perspectives 121, 978984.Google Scholar
Roher, A.E., Debbins, J.P., Malek-Ahmandi, M., Chen, K., Pipe, J.G., Maze., S., Belden, C., Maarout, C.L., Thiyyagura, P., Mo, H., Hunter, J.M., Kokjohn, T.A., Walker, D.J., Kruchowsky, J.C., Belohlavek, M., Sabbagh, M.N., Beach, T.G. 2012: Cerebral blood flow in Alzheimer’s disease. Vascular Health & Risk Management 8, 599608.Google Scholar
Rosas-Pérez, I., Serrano, J., Alfaro-Moreno, E., Baumgardner, D., Garcia-Cuellar, C., Martin del Campo, J.M., Raga, G.B., Castillejos, M., Colin, R.D. and Osornio-Vargas, A.R. 2007: Relations between PM10 composition and cell toxicity: a multivariate and graphical Approach. Chemosphere 67, 12181228.Google Scholar
Schröder, N., Silva-Figueiredo, L. and Martins de Lima, M.N. 2013: Role of brain iron accumulation in cognitive dysfunction: evidence from animal models and human studies. Journal of Alzheimer’s Disease 34, 797812.Google Scholar
Sharma, H.S. and Sharma, A. 2012: Neurotoxicity of engineered nanoparticles from metals. CNS & Neurological Disorders 11, 6580.Google Scholar
Shulman, K.I. 2000: Clock-drawing: is it the ideal cognitive screening test? International Journal of Geriatric Psychiatry 15, 548561.Google Scholar
Siddique, S., Banerjee, M., Ray, M.R. and Lahiri, T. 2011: Attention-deficit hyperactivity disorder in children chronically exposed to high level of vehicular pollution. European Journal of Pediatrics 170, 923929.Google Scholar
Secretaria de Medio Ambiente, Gobierno de Distrito Federal (SMA-GDF). 2008: Inventario de Emisiones de la Atmósfera. Zona Metropolitana del Valle de México 2006, Secretaria de Medio Ambiente, Gobierno de Distrito Federal, Mexoico D.F.Google Scholar
Suglia, S.F., Gryparis, A., Wright, R.O., Schwartz, J. and Wright, R.J. 2008: Association of black carbon with cognition among children in a prospective birth cohort study. American Journal of Epidemiology 167, 280286.Google Scholar
Suissa, L., Fortier, M., Lachaud, S., Staccini, P. and Mahagne, M.H. 2013: Ozone air pollution and ischemic stroke occurrence: a case-crossover study in Nice, France. BMJ Open 3, e004060.Google Scholar
Söderstjerna, E., Johansson, F., Klefbohm, B. and Englund-Johansson, U. 2013: Gold and silver nanoparticles affect the growth characteristics of human embryonic neural precursor cells. PLoS One 8, e58211.Google Scholar
Szyszkowicz, M., Willey, J.B., Grafstein, E., Rowe, B.H. and Colman, I. 2010: Air pollution and emergency department visits for suicide attempts in Vancouver, Canada. Environmental Health Insights 4, 7986.Google Scholar
Trickler, W.J., Lantz, S.M. and Schrand, A.M. 2012: Effects of copper nanoparticles on rat cerebral microvessel endothelial cells. Nanomedicine 7, 835846.Google Scholar
U.S. Environmental Protection Agency. 2006: Particulate Matter (PM-2.5). Standard Nonattainment Areas . U.S. Environmental Protection Agency.Google Scholar
Valle-Hernández, B.L., Mugica-Alvarez, V., Salinas-Talavera, E., Amador-Muñoz, O., Murillo-Tovar, M.A., Villalobos-Pietrini, R. and De Vizcaya-Ruíz, A. 2010: Temporal variation of nitro-polycyclic aromatic hydrocarbons in PM10 and PM2.5 collected in Northern Mexico City. The Science of the Total Environment 408, 54295438.Google Scholar
van den Hazel, P., Zuurbier, M., Babisch, W., Bartonova, A., Bistrup, M.L., Bolte, G., Busby, C., Butter, M., Ceccatelli, S., Fucic, A., Hanke, W., Johansson, C., Kohlhuber, M., Leijs, M., Lundqvist, C., Moshammer, H., Naginiene, R., Preece, A., Ronchetti, R., Salines, G., Saunders, M., Schoeters, G., Stilianakis, N., ten Tusscher, G. and Koppe, J.G. 2006: Today’s epidemics in children: possible relations to environmental pollution and suggested preventive measures. Acta Paediatrica Supplement 95, 1825.Google Scholar
Van Kempen, E., Fischer, P., Janssen, N., Houthuijis, D., van Kamp, I., Stansfeld, S., Cassee, F. 2012: Neurobehavioral effects of exposure to traffic-related air pollution and transportation noise in primary school children. Environmental Research 115, 1825.Google Scholar
Villarreal-Calderón, R., Torres-Jardon, R., Palacios-Moreno, J., Perez-Guille, B., Maronpot, R.R., Reed, W., Zhu, H., Calderón-Garciduenas, L. 2010: Urban air pollution targets the dorsal vagal complex and dark chocolate offers neuroprotection. International Journal of Toxicology 29, 604615.Google Scholar
Villeneuve, P.J., Johnson, J.Y., Pasichnvk, D., Lowes, J., Kirkland, S. and Rowe, B.H. 2012: Short term effects of ambient air pollution on stroke: who is most vulnerable? The Science of the Total Environment 430, 193201.Google Scholar
Visser, J.C., Rommelse, N., Vink, L., Schrieken, M., Oosterling, I.J., van der Gaag, R.J. and Buitelaar, J.K. 2013: Narrowly versus broadly defined autism spectrum disorders: differences in pre and perinatal risk factors. Journal of Autism Developmental Disorders 43, 15051516.Google Scholar
Volk, H.E., Hertz-Picciotto, I., Delwiche, L., Lurmann, F. and McConnell, R. 2011: Residential proximity to freeways and autism in the CHARGE study. Environmental Health Perspectives 119, 873877.Google Scholar
Volk, H.E., Lurmann, F., Penfold, B., Hertz-Picciotto, I. and McConnell, R. 2013: Traffic-related air pollution, particulate matter and autism. JAMA Psychiatry 70, 7177.Google Scholar
Volk, H.E., Kerin, T., Lurmann, F., Hertz-Picciotto, I., McConnell, R. and Campbell, D.B. 2014: Autism spectrum disorder: interaction of air pollution with the MET receptor tyrosine kinase gene. Epidemiology 25, 4447.Google Scholar
Wang, S., Zhang, J., Zeng, X., Zeng, Y., Wang, S. and Chen, S. 2009: Association of traffic-related air pollution with children’s neurobehavioral functions in Quanzhou, China. Environmental Health Perspectives 117, 16121618.Google Scholar
Wang, J., Eslinger, P.J., Doty, R.L., Zimmerman, E.K., Grunfeld, R., Sun, X., Meadowcroft, M.D., Connor, J.R., Price, J.L., Smith, M.B. and Yang, Q.X. 2010: Olfactory deficit detected by fMRI in early Alzheimer’s disease. Brain Research 1357, 184194.Google Scholar
Wilker, E.H., Mostofsky, E., Lue, S.H., Gold, D., Schwartz, J., Wellenius, G.A. and Mittleman, M.A. 2013: Residential proximity to high-traffic roadways and poststroke mortality. Journal of Stroke and Cerebrovascular Diseases 22, e366e372.Google Scholar
Williams, L., Ulrich, C.M., Larson, T., Wener, M.H., Wood, B., Chen-Levy, Z., Campbell, P.T., Potter, J., McTiernan, A. and Roos, A.J. 2011: Fine particulate matter PM2.5 air pollution and immune status among women in the Seattle area. Archives of Environmental Health 66, 155164.Google Scholar
Win-Shwe, T.T., Yamamoto, S., Fujitani, Y., Hirano, S. and Fujimaki, H. 2008: Spatial learning and memory function-related gene expression in the hippocampus of mouse exposed to nanoparticle-rich diesel exhaust. Neurotoxicology 29, 940947.Google Scholar
Win-Shwe, T.T. and Fujimaki, H. 2011: Nanoparticles and neurotoxicity. International Journal of Molecular Sciences 12, 62676280.Google Scholar
Wisnivesky, J.P., Teitelbaum, S.L., Todd, A.C., Boffetta, P., Crane, M., Crowley, L., de la Hoz, R.E., Dellenbaugh, C., Harrison, D., Herbert, R., Kim, H., Jeon, Y., Kaplan, J., Katz, C., Levin, S., Luft, B., Markowitz, S., Moline, J.M., Ozbay, F., Pietrzak, R.H., Shapiro, M., Sharma, V., Skloot, G., Southwick, S., Stevenson, L.A., Udasin, I., Wallenstein, S. and Landrigan, P.J. 2011: Persistence of multiple illnesses in World Trade center rescue and recovery workers: a cohort study. Lancet 378, 888897.Google Scholar
Wu, J., Wang, C. and Sun, J. 2011: Neurotoxicity of silica nanoparticles: brain localization and dopaminergic neurons damage pathways. ACS Nano 5, 44764489.Google Scholar
Xu, P. and Ye, X. 2013: Haze air pollution and health in China. Lancet 49, 217229.Google Scholar
Yang, C.Y., Chen, Y.S., Chiu, H.F. and Goggins, W.B. 2005: Effects of Asian dust storm events on daily stroke admissions in Taipei, Taiwan. Environ Res 99, 7984.Google Scholar
Ze, Y., Sheng, L., Zhao, X., Ze, X., Wang, X., Zhou, Q., Liu, J., Yuan, Y., Gui, S., Sang, X., Sun, Q., Hong, J., Yu, X., Wang, L., Li, B., Hong, F. 2014: Neurotoxic characteristics of spatial recognition damage of the hippocampus in mice following subchronic peroral exposure to TiO2 nanoparticles. Journal of Hazardous Materials 264, 219229.Google Scholar
Zhang, Z.F., Yu, S.Z. and Zhou, G.D. 1988: Indoor air pollution of coal fumes as a risk factor of stroke, Shanghai. American Journal of Public Health 78, 975977.Google Scholar
Figure 0

Table 1 Selected studies examining neurocognitive/neurophysiological effects of air pollution in children, adolescents and young adult populations. The table shows the populations and the air pollutants studied, the tests and deficits found, other tests used and the city/cities/country where the study took place

Figure 1

Table 2 Suggested battery of neuropsychological and psychoeducational tests for the initial screening of school-aged children and teens exposed to urban air pollution