1,108
Views
20
CrossRef citations to date
0
Altmetric
Editorial

Maternal stress and perinatal programming in the expression of atopy

&
Pages 535-538 | Published online: 10 Jan 2014

Biological hypersensitivity to environmental stimuli, a fundamental feature of atopy, contributes to a spectrum of costly pediatric disorders, including allergic rhinitis, atopic dermatitis and asthma. The increase in the prevalence of these disorders and the enormous costs involved in managing atopic patients have motivated efforts to identify early risk factors that may be amenable to intervention and prevention Citation[1–3].

Research continues to delineate early immunophenotypes and early airway response outcomes among children predisposed to chronic atopic disorders Citation[4]. The development of allergic sensitization, childhood asthma and other atopic disorders run in parallel if certain hereditary and/or perinatal environmental influences prevail, rather than being stepping stones in a progressive atopic march Citation[5]. Those with early (i.e., starting in the first year of life) sensitization to allergens are at greatest risk of subsequently developing chronic atopic disorders, airway inflammation and obstruction. Persistent reactivity throughout early childhood, high-level sensitization and polysensitization have also been more clearly linked to later atopic disease Citation[6,7]. Even in persistently sensitized children, the risk of developing atopic disorders is particularly increased by a positive parental history of asthma/atopy, with these effects being strongest for maternal history Citation[8]. This latter finding, together with other evidence, buttresses the concept of perinatal programing, which proposes that nongenetic factors acting early in life may permanently organize or imprint physiological systems.

The growing list of potential programing agents includes psychological stress. Evidence linking psychological stress to atopy suggests that early disruption of neuroimmunoregulatory processes are probably involved Citation[9]. Both primate and rodent models of prenatal stress and early adverse caregiving have helped us to understand the potential consequences of similar experiences in humans and their relevance to atopy Citation[10]. Early-life adversity shapes stress neurobiology, resulting in disturbed regulation of endocrine and autonomic processes (e.g., the hypothalamic–pituitary–adrenal [HPA] axis and the sympathetic–adrenal–medullary system). These disturbed patterns of stress regulation are hypothesized to subsequently modulate immune function, increasing susceptibility to asthma and related diseases. Maternal stress experienced in utero may influence programing of these key physiological systems in children Citation[10]. Stressors influence pathogenesis by causing dysregulated biobehavioral states (e.g., depression and post-traumatic stress disorder [PTSD]), which, in turn, program lasting effects on physiological processes that influence disease risk. Infants of mothers who have prenatally programed biobehavioral sequelae from stress may, thus, inherit biological vulnerabilities that alter reactivity to subsequent challenges Citation[11]. Likewise, nonoptimal early-childhood environments and care-giving experiences (e.g., maternal psychopathology or maternal insensitivity) may impact these processes Citation[12,13]. Stress-elicited disruption of these inter-related systems – autonomic, neuroendocrine and immune – may lead to increased vulnerability to allergic sensitization and atopic risk.

The HPA axis seems particularly susceptible to early programing. Prenatal stress has been associated with early and long-term developmental effects resulting, in part, from altered maternal and/or fetal glucocorticoid exposure. Maternal and fetal stress also stimulate placental secretion of corticotrophin-releasing hormone (CRH), which in turn is elevated in the neonatal circulation. Elevated CRH may stimulate the fetal HPA axis to amplify fetal glucocorticoid excess, as well as activate additional elements of the fetal stress response (i.e., catecholamines and neurotrophins), influencing the developing immune and autonomic nervous systems Citation[14]. For example, it has been proposed that alterations in stress-induced maternal cortisol levels may influence fetal immune system development and Th2 cell predominance, perhaps through a direct influence of stress hormones on cytokine production Citation[15]. While these in utero responses may be adaptive in the short term, being geared toward coping with anticipated environmental challenges, they may exact a toll in contributing to an increased risk of atopic diseases in later life.

In humans, both the HPA system and the autonomic nervous system show significant developmental changes in infancy, shaped by interactions with the environment. The HPA axis starts to become organized between 2 and 6 months of age Citation[16], and the autonomic nervous system demonstrates relative stability in resting measures by 6-12 months of age while stability in reactivity measures in response to challenge likely occurs later in development Citation[17]. The HPA axis in particular has been shown to be highly responsive to child–caregiver interactions, with sensitive caregiving programing the HPA axis to become an effective physiological regulator of stress, and insensitive caregiving, promoting hyper- or hyporeactive HPA responses Citation[18]. Several animal models, as well as human studies, also support the connection between caregiving experiences in early postnatal life and alterations of autonomic nervous system balance Citation[17,19,20]. Furthermore, children who have a history of sensitive caregiving are more likely to demonstrate optimal affective and behavioral strategies for coping with stress Citation[13]. Therefore, children with a history of supportive, sensitive caregiving in early development may be better able to cope with environmental stressors and, consequently, less likely to manifest disturbed HPA and autonomic reactivity that put them at risk for stress-related illnesses, such as asthma.

As yet, the specific pathways of an increased vulnerability to atopy in response to perinatal stress is largely unknown in humans. Data regarding the development of allergic sensitization in inner-city minority populations in the USA suggest that new models for understanding the developmental etiology of asthma and other atopic disorders are needed. There is a paradox between the prevalence of atopy in the US inner-city and the currently proposed hygiene hypothesis – if the hygiene hypothesis is correct, early exposure to microbial products in the inner city should markedly reduce the incidence of allergic sensitization and atopic diseases compared with other US locales Citation[3]. The converse, however, is the reality. In a cross-sectional study of 4164 children in the Third National Health and Nutrition Examination Survey (NHANES), African–American and Hispanic ethnicity was associated with an increased risk of sensitization to cockroaches, dust mites and mold Citation[21]. Stevenson and colleagues have found that allergic sensitization is prevalent in a lower socioeconomic status sample, even among children who do not have a family history of atopy Citation[22]. While impoverished households are more likely to be reservoirs for allergens in higher concentrations than more affluent settings, this does not completely explain the observed differences Citation[23]. Sensitization to multiple allergens is also more prevalent in urban homes, suggesting that some other factor may be enhancing response to allergens in these urban populations. Differential exposure to stress, and variability in stress responses, occurring in these early critical periods, may be factors that augment the likelihood of early atopy by enhancing the immune response to allergen exposure Citation[24]. One intriguing mechanism gaining support in the literature is environmental influences on epigenetic regulation and atopic risk Citation[25]. New data from behavioral studies link the social environment (stress and caregiving) with epigenetic programing. One line of evidence comes from studies of epigenetic changes during long-term potentiation and fear conditioning by Sweat’s group Citation[26], and another comes from a study of epigenetic programing related to maternal care Citation[27].

The nature of the stressor(s) that are likely to have a measurable impact on atopic risk is currently unknown. Emerging evidence suggests that exposure to trauma may be a particularly robust potentiator of the cascade of biological events that increase vulnerability to atopy and may help explain the increased risk found in low-income urban populations Citation[28]. The Diagnostic and Statistical Manual of Mental Health Disorders (DSM)-IV-TR defines a traumatic event as one that involves experiencing, being threatened with, witnessing or learning about death or serious injury to self or others and a resultant feeling of intense fear, helplessness or horror (criterion A traumatic event) Citation[29]. Low-income, urban, minority women experience interpersonal violence (e.g., child abuse, intimate partner violence, community violence) and other traumas over their lives at rates markedly above national samples in the USA Citation[30–32]. Numerous studies have demonstrated that victims of trauma experience a host of physiological, affective and behavioral responses, with PTSD and/or major depression emerging in a significant minority, often concomitantly Citation[33,34]. Therefore, not surprisingly, low-income and ethnic-minority mothers, particularly in the perinatal period, demonstrate elevated rates of PTSD and depression Citation[35,36]. These psychological conditions during pregnancy have been repeatedly linked to disruptions in the maternal–fetal HPA axis and, later, the infant HPA axis and autonomic reactivity Citation[11,37]. Furthermore, extensive literature documents associations between maternal traumatic stress, anxiety and depression and serious difficulties in maternal caregiving. Such caregiving difficulties have, in turn, been associated with disruptions in child endocrine, autonomic, affective and behavioral regulation Citation[13]. Consequently, trauma influences on perinatal maternal–child interactions may disrupt infant neuroimmune development through its impact on child stress neurobiology, setting the stage for the altered reactivity characteristic of atopy Citation[10].

Studies evaluating psychophysiological stress reactivity during critical periods of development, including gestation and early childhood, are needed, as they hold particular promise in elucidating specific pathways that increase vulnerability to atopy. Exploring links between maternal trauma and atopic risk may be particularly relevant in urban, high-risk US populations that are disproportionately burdened by both phenomena.

Financial & competing interests disclosure

RJ Wright is supported by R01HL080674 and M Bosquet Enlow is supported by K08MH074588. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

No writing assistance was utilized in the production of this manuscript.

References

  • Lewis-Jones S. Quality of life and childhood atopic dermatitis: the misery of living with childhood eczema. Int. J. Clin. Pract.60(8), 984–992 (2006).
  • Blaiss MS. Cognitive, social, and economic costs of allergic rhinitis. Allergy Asthma Proc.21(1), 7–13 (2000).
  • Gold DR, Wright RJ. Population disparities in asthma. Ann. Rev. Public Health26, 1–25 (2005).
  • Heaton T, Rowe J, Turner S et al. An immunoepidemiological approach to asthma: identification of in vitro T-cell response patterns associated with different wheezing phenotypes in children. Lancet365, 142–149 (2005).
  • Saglani S, Bush A. The early-life origins of asthma. Curr. Opin. Allergy Clin. Immunol.7(1), 83–90 (2007).
  • Johnke H, Norberg LA, Vach W, Host A, Andersen KE. Patterns of sensitization in infants and its relation to atopic dermatitis. Pediatr. Allergy Immunol.17, 591–600 (2006).
  • Panettieri RA, Covar R, Grant E, Hillyer EV, Bacharier L. Natural history of asthma: persistence versus progression – does the beginning predict the end? J. Allergy Clin. Immunol.121, 607–613 (2008).
  • Bjerg A, Hedman L, Perzanowski MS, Platts-Mills T, Lundback B, Ronmark E. Family history of asthma and atopy: in-depth analyses of the impact on asthma and wheeze in 7- to 8-year-old children. Pediatrics120(4), 741–748 (2007).
  • Wright RJ. Stress and atopic disorders. J. Allergy Clin. Immunol.116(6), 1301–1306 (2005).
  • Wright RJ. Prenatal maternal stress and early caregiving experiences: implications for childhood asthma risk. Paediatr. Perinat. Epidemiol.21(Suppl. 3), 8–14 (2007).
  • Yehuda R, Bierer LM. Transgenerational transmission of cortisol and PTSD risk. Progress Brain Res.167, 121–134 (2008).
  • Meaney MJ. Maternal care, gene expression, and the transmission of individual differences in stress reactivity across generations. Ann. Rev. Neurosci.24, 1161–1192 (2001).
  • Rifkin-Graboi A, Borelli JL, Bosquet M. Neurobiology of stress in infancy. In: Handbook of Infant Mental Health (3rd Edition). Zeanah CHJ (Ed.). Guilford, NY, USA (2008) (In Press).
  • Arck PC, Knackstedt MK, Blois SM. Current insights and future perspectives on neuro–endocrine-immune circuitry challenging pregnancy maintenance and fetal health. J. Reproduktionsmed. Endokrinol.3(2), 98–102 (2006).
  • von Hertzen LC. Maternal stress and T-cell differentiation of the developing immune system: possible implications for the development of asthma and atopy. J. Allergy Clin. Immunol.109(6), 923–928 (2002).
  • Gunnar MR, Donzella B. Social regulation of the cortisol levels in early human development. Psychoneuroendocrinology27, 199–220 (2002).
  • Alkon A, Lippert S, Vujan N, Rodriguez ME, Boyce WT, Eskenazi B. The ontogeny of autonomic measures in 6- and 12-month-old infants. Dev. Psychobiol.48(3), 197–208 (2006).
  • Lyons-Ruth K, Block DE. The disturbed caregiving system: relations among childhood trauma, maternal caregiving, and infant affect and attachment. Infant Ment. Health J.17, 257–275 (1996).
  • Herlenius E, Lagercrantz H. Development of neurotransmitter systems during critical periods. Exp. Neurol.190(Suppl. 1), S8–S21 (2004).
  • Pryce CR, Ruedi-Bettschen D, Dettling AC, Feldon J. Early life stress: long-term physiological impact in rodents and primates. News Physiol. Sci.17, 150–155 (2002).
  • Stevenson LA, Gergen PJ, Hoover DR, Rosenstreich D, Mannino DM, Matte TD. Sociodemographic correlates of indoor allergen sensitivity among United States children. J. Allergy Clin. Immunol.108(5), 747–752 (2001).
  • Stevenson MD, Selling S, Grube E et al. Aeroallergen sensitization in healthy children: racial and socioeconomic correlates. J. Pediatr.151, 187–191 (2007).
  • Shapiro GG, Stout JW. Childhood asthma in the United States: urban issues. Pediatr. Pulmonol.33(1), 47–55 (2002).
  • Peters JM, Franco Suglia S, Platts-Mills TAE, Hosen J, Wright RJ. Psychological stress modifies the influence of prenatal allergen exposure on cord blood IgE: the Boston ACCESS project. Am. J. Respir. Crit. Care Med.177, A231 (2008).
  • Miller RL, Ho S-M. Environmental epigentics and asthma: current concepts and call for studies. Am. J. Respir. Crit. Care Med.177, 567–573 (2008).
  • Levenson JM, Roth TL, Lubin FD et al. Evidence that DNA (cytosine-5) methytransferase regulates synaptic plasticity in the hippocampus. J. Biol. Chem.281(23), 15763–15773 (2006).
  • Meaney MJ, Szyf M. Maternal care as a model for experience-dependent chromatin plasticity? Trends Neurosci.28(9), 456–463 (2005).
  • van der Kolk BA. The complexity of adaptation to trauma: self-regulation, stimulus, discrimination, and characterological development. In: Traumatic Stress: The Effects of Overwhelming Experience on Mind, Body, and Society. van der Kolk BA, McFarlane AC, Weisaeth L (Eds). Guilford, NY, USA 182–213 (1996).
  • Diagnostic and Statisitcal Manual of Mental Disorders, Text Revision (DSM-IV-TR) American Psychiatric Press, Inc., Washington, DC, USA (2000).
  • Hien D, Bukszpan C. Interpersonal violence in a “normal” low-income control group. Women Health29(4), 1–15 (1999).
  • Clark C, Ryan L, Kawachi I, Jacobson Canner M, Berkman LF, Wright RJ. Witnessing community violence in residential neighborhoods: a mental health hazard for urban women. J. Urban Health85(1), 22–38 (2007).
  • Holman EA, Silver RC, Waitzkin H. Traumatic life events in primary care patients: a study in an ethnically diverse sample. Arch. Fam. Med.9(9), 802–810 (2000).
  • Breslau N, Kessler RC, Chilcoat HD, Schultz LR, Davis GC, Andreski P. Trauma and posttraumatic stress disorders in the community: the 1996 Detroit Area Survey of Trauma. Arch. Gen. Psychiatry55, 626–632 (1998).
  • Kessler RC, Sonnega A, Bromet E, Hughes M, Nelson CR. Posttraumatic stress disorder in the National Comorbidity Survey. Arch. Gen. Psychiatry52, 1048–1060 (1995).
  • Chaudron LH, Kitzman HJ, Peifer KL, Morrow S, Perez LM, Newman M. Prevalence of maternal depressive symptoms in low-income Hispanic women. J. Clin. Psychiatry66, 418–423 (2005).
  • Benoit C, Westfall R, Treloar AEB, Phillips R, Jansson SM. Social factors linked to postpartum depression: a mixed-methods longitudinal study. J. Mental Health16, 719–730 (2007).
  • Field T, Diego M, Hernandez-Reif M. Prenatal depression effects on the fetus and newborn: a review. Infant Behav. Dev.29, 445–455 (2006).

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.