• Non ci sono risultati.

25 Obesity and Asthma

N/A
N/A
Protected

Academic year: 2022

Condividi "25 Obesity and Asthma"

Copied!
9
0
0

Testo completo

(1)

Obesity and Asthma

Elisabeth Luder

25

From: Nutrition and Health: Adipose Tissue and Adipokines in Health and Disease Edited by: G. Fantuzzi and T. Mazzone © Humana Press Inc., Totowa, NJ

341 Abstract

Obesity and asthma are both common complex traits responsible for substantial morbidity in the developed world. The consistency of the relationship between obesity and asthma, the temporal and dose-response association, and the correlation of obesity with intermediate phenotypes for asthma suggest that the obesity–asthma link is causal. With few exceptions, the existing epidemiological studies show a consistent positive association of obesity with both incidence and prevalence of asthma in children and adults, with the effect being greater in females than in males. Obesity pre- cedes and predicts the development of asthma, not the other way around, and the effect persists after controlling for diet and physical activity. The dose–response relationship is demonstrated by the finding that the greater the obesity the greater the effect on asthma. Studies are showing improve- ments in asthma in subjects who lose weight. From these observations, the main research issues at present relate to the actual biological mechanisms by which obesity influences the asthma pheno- type. The specific areas requiring further investigation are: direct effect of obesity on mechanical functioning of the lung; changes in the immune or inflammatory responses directly or through genetic mechanism; sex-specific influences relating to hormones; and the influence of maternal diet on fetal programming. At present, we still do not have a good understanding of the precise relation- ship between obesity and asthma.

Key Words: Obesity; asthma; lung; inflammation; genes; hormones; diet.

1. INTRODUCTION

Obesity and asthma are both chronic conditions affecting millions worldwide. Over the past two decades there has been a rapid increase in the prevalence of both these con- ditions (1,2). In the United States, in 2002, 65% of adults were overweight, with a body mass index (BMI) higher than 25, compared with 46% in 1980; in addition, 16% of children, age 6 to 19 yr had a BMI higher than 25, compared with 5% in 1980 (3). From 1980 to 1996, there was an increase in self-reported asthma prevalence of 74% (4).

Given the parallel increase in obesity and asthma, it is not surprising that the prevalence and incidence of asthma and its related symptoms have been associated with BMI and obesity as described in a number of epidemiological studies for adults and children. In this chapter, the definition of asthma, association between obesity and asthma as described in cross-sectional and longitudinal studies, and causal hypotheses of obesity and asthma will be addressed.

(2)

2. DEFINITION AND EPIDEMIOLOGY OF ASTHMA

Asthma is defined by episodic airflow obstruction, increased airway responsiveness, and airway inflammation characterized by infiltration with eosinophils and T-lympho- cytes, particularly CD4+T-lymphocytes that express T-helper (Th) cell type 2 cytokines such as interleukin (IL)-4, IL-5, and IL-13. The histopathological appearance of the airways includes denudation of the airway epithelium, thickening of the basement membrane, mucus production, and airway smooth muscle hypertrophy (5). Although asthma is a chronic—often lifelong—disease that affects humans of all ages, the onset of the disease occurs primarily in early childhood. Fifty percent of all male asthma cases are diagnosed by age 3, and 50% of all female cases are diagnosed by age 8 (6). This increase of asthma in early childhood has been most marked in minority populations, particularly African Americans and Puerto Rican Hispanics (7).

3. DEFINITION OF OBESITY

A number of methods have been proposed to describe increases in body weight. The most widely used measurement is the BMI, calculated as weight/height2 (kg/m2).

Among adult subjects overweight is defined as a BMI of 25 to 29.9 kg/m2, and obesity as a BMI of at least 30 kg/m2. Different ethnicities, such as Caucasians and Chinese, have highly contrasting distributions of body weight and height. Males and females differ in their BMI distribution. Age is an obvious modifier of body weight and height.

Therefore, age- and sex-specific definitions for different ethnic groups must be applied when comparing effects in various study populations. Growth charts from the Centers for Disease Control and Prevention (CDC) include age- and sex-specific BMI reference values for children and adolescents aged 2 to 20 yr (8). However, BMI does not account for body frame and proportion of muscle mass. This limitation is particularly relevant in pediatric studies because of the effects of maturation and growth of lean muscle mass, fat mass, and hydration status. Furthermore, muscle mass increases with higher activity level, and fat mass values are higher among females than males and vary across ethnic groups (9). Therefore, other measures of body weight have been applied, such as assess- ment of body fat by skinfold thickness, dual-energy X-ray absorptiometry (DEXA) evaluation or bioelectrical impedance. However, large comparative studies on skinfold, DEXA, and bioelectrical impedance variables with BMI are missing to date.

4. ASSOCIATION BETWEEN OBESITY AND ASTHMA

Conflicting results of various studies investigating the potential association between obesity and asthma may be attributable to different study design—cross-sectional or longitudinal—or effect modifiers.

4.1. Cross-Sectional Studies

The cross-sectional diagnosis of asthma has been associated with obesity in both children (10–13) and adults (14–16). In most studies, the definition of asthma was based on a doctor diagnosis of asthma, partly including airway hyper-responsiveness (AHR), but in some studies only symptoms such as wheeze or asthma attacks were assessed.

Several of these studies noted a relationship only in women, but not in men (17,18).

(3)

However, findings are not consistent, and associations between obesity and AHR were even stronger in males than females (19). AHR may reflect airway inflammation but not asthma itself, as AHR is a feature rather loosely associated with a doctor’s diagnosis of asthma in population-based studies. Effect modification by sex, however, may also be based on differences in the shape of the relationship. This notion is supported by a survey demonstrating that the association between BMI and asthma differed only in the lowest weight category. Among women a monotonic association was seen, whereas in men a U-shaped relation was found, but both extremes of weight were associated with higher prevalence of asthma (20).

Cross-sectional studies may be prone to bias because they do not allow an assessment of the timing of the exposure in relation to the occurrence of asthma. Thus the relation- ship between BMI and asthma may reflect an asthmatic patient’s predisposition to gain weight because of reduced exercise tolerance rather than causal association between a high BMI and the inception of asthma. Therefore results from prospective studies may help to better interpret the findings (9).

4.2. Prospective Studies

Nearly all prospective studies demonstrate a positive association between BMI and the development of asthma and AHR, respectively (19,21,22). In these studies weight gain occurred before the new onset of asthma or asthma symptoms, suggesting a true relation between both conditions. In some studies stratification by sex either by study design (21,22) or in the analyses revealed sex-specific effects. Camargo et al. (21) reported a significant association between overweight development and new-onset asthma in the US Nurses’ Health Study, whereas Litonjua et al. (22) detected a U-shaped relation with AHR at high and low BMI in the Normative Aging Study, including only males. In a study of 135,000 Norwegians aged 14 to 60 yr who were fol- lowed on average for 21 yr, the risk of asthma increased steadily with an increase in BMI. For men the risk of asthma increased starting with a BMI of 20 and in women with a BMI of 22. In men, the risk of asthma increased by 10% and for women by 7% with each unit of increased BMI between 25 and 30 (23). A population of 10,597 adult twins, initially free of asthma, was followed for 9 yr. Obese men with a BMI v 30 had a sig- nificantly increased risk of developing asthma when compared to those with a BMI of 20 to 24.99 (OR = 3.47). More men were obese than women, and the association between BMI and asthma was not significant in women (24).

In a longitudinal population-based birth cohort study of 781 children, in boys and girls the presence of obesity during the prepubertal period and early onset of puberty were significant and independent risk factors for persistent asthma after puberty (25).

Among 3,792 participants aged 7 to 18 yr in the Children’s Health Study who were asthma-free at enrollment and were followed for 5 yr, the risk of asthma development increased among overweight and obese boys but not girls (OR = 2.06 vs OR = 1.25) (26). Among 9828 children aged 6 to 14 yr examined annually over a follow-up time of 5 yr in six US cities, an increased risk of new asthma diagnosis in girls was associated with higher BMI at entry into the study and greater increase in BMI during follow-up.

Boys with the largest and smallest annual change in BMI also had an increased risk of asthma. For boys and girls, extremes of annual BMI growth rates increased the risk of asthma (27). As addressed in the following section, in these children, overweight or

(4)

extreme leanness may represent a combination of in utero and postnatal influences on growth and development that increase the risk of airway mechanical dysfunction or inflammation (28,29).

5. CAUSAL HYPOTHESES

In the association between obesity and asthma, potential misclassifications of wheeze and asthma must be considered. Obstructive sleep apnea or hypoventilation are frequent among obese patients (5,9). However, it seems unlikely that these symptoms are mis- classified as asthma in longitudinal surveys. As previously stated, all the existing prospective epidemiological studies show a consistent positive association of obesity with both incidence and prevalence of asthma in children and adults. Obesity precedes asthma and predicts the development of asthma and the effect persists after controlling for diet and physical activity. The dose–response relationship is demonstrated by the finding that the greater the obesity, the greater the observed effect on asthma (10–12,21).

The effects of obesity seem greater for asthma and airway responsiveness than they do for other allergy phenotypes, although these effects have not been assessed as fre- quently. From these observations, studies suggest that obesity has the potential to affect airway function through a variety of agencies, including mechanical functioning of the lung, changes in immune or inflammatory responses directly or through genetic mecha- nisms, sex-specific influences relating to hormones, lung development, and the influ- ence of maternal diet on fetal programming (28,29). These are many possibilities, but they are not mutually exclusive, and the dominant mechanism among them is yet to be identified.

5.1. Effects of Lung Mechanics

Obesity leads to decreased lung tidal volume as well as decreased functional residual capacity. These volume changes result in reduced smooth muscle stretch or latching.

Consequently, the ability to respond to a physiological stress such as exercise is hampered by small tidal breaths, which alters smooth muscle contraction, worsening the respira- tory condition. Normal smooth muscle has an intrinsic rate of excitation and contrac- tion called the cycling rate. In obese people, lower cycling rates of the airway smooth muscles and thus decreased functional capacity result from the conversion of rapidly cycling actin–myosin crossbridges to slowly cycling latch bridges (28,30). The exact dose–effect relationship between the amount and distribution of body fat and the mechanical changes remains unknown and is an area for further research.

5.2. Comorbidities

Obesity may lead to asthma not directly, but through its role in other disease processes. For example, obesity increases the risk of both gastroesophageal reflux dis- ease (GERD) and sleep-disordered breathing (SDR). An increased prevalence of asthma has been observed in subjects with each of these conditions; furthermore, subjects under- going surgical induced weight loss showed improvements not only in asthma but also in GERD and sleep apnea. Consequently, there has been speculation that obesity leads to asthma through its effects on these other conditions. Two recent studies have examined the interrelationships between these conditions. Multivariate logistic regression in data

(5)

from more than 16,000 participants in the European Community Respiratory Health Survey demonstrated that the relationship between obesity and the onset of asthma was unaffected by adjustment for GERD or habitual snoring (31). Similarly, Sulit et al.

demonstrated that adjustment for SDB and asthma did not substantially alter the associ- ation between obesity and asthma (32). Taken together, these data indicate that the increased risk of asthma in the obese is independent of GERD and SDB.

5.3. Chronic Systemic Inflammation

There is increasing evidence that obesity is a proinflammatory state (33). Initial stud- ies have focused mainly on the association of obesity and tumor necrosis factor (TNF), IL-6, IL-1G, and C-reactive protein. IL-6 and TNF are constitutively expressed by adipocytes and correlate with total fat mass. TNF is increased in asthma, and it increases further with allergen exposure. Thus, the TNF inflammatory pathway is common to both obesity and asthma, and it is plausible that it is upregulated by the presence of both conditions (28,29).

Recent research shows that in obese humans, even in the absence of any overt inflam- matory insult, there is chronic, low-grade systemic inflammation characterized by increas- ed circulating leukocytes and increased serum concentration of cytokines, cytokine receptors, chemokines, and acute-phase proteins (34). Similar results are obtained in obese mice. The origin of this inflammation appears to be, at least in part, the adipose tissue itself, because expression of a variety of inflammatory genes is upregulated in adipose tissue from obese humans or mice. The cellular source of some of these factors appear to be macrophages that infiltrate adipose tissue (35,36). Systemic inflammatory markers in humans correlate with the presence of diseases common to obesity, includ- ing type 2 diabetes and atherosclerosis, suggesting that the inflammation is functionally important. Obese Cpefatmice display innate airway hyper-responsiveness, as well as increased airway responsiveness and inflammation following ozone (O3) exposure.

These increased effects of O3appear to be independent of changes in lung volume or lung mass, suggesting that obesity augments the airway response to O3in mice (34).

Adiponectin is one of the most abundant gene products in adipose tissue. In con- trast with many of the other adipokines, the levels of which rise in obesity, plasma adiponectin levels are decreased in obesity, and levels increase following weight loss.

The predominant metabolic effects of adiponectin are in the liver and in skeletal muscle and include increased glucose uptake, inhibition of gluconeogenesis, and increased fatty acid oxidation (37). Adiponectin also has anti-inflammatory proper- ties. Pertaining to asthma, adiponectin inhibits proliferation and migration of cultured vascular smooth muscle cells induced by mitogens (38). It will be important to deter- mine whether adiponectin has similar effects on airway smooth muscles (ASM), espe- cially because both the AdipoR1 and AdipoR2 receptors are expressed in cultured human ASM cells. In this context, it should be noted that increased ASM mass is a feature of human asthma, and modeling studies have shown that increased muscle mass alone can account for a large part of the AHR of asthma. Taken together, the anti-inflammatory effects of adiponectin and the possibility that adiponectin may have antimitogenic effects on ASM suggest that the decreased serum concentration of adiponectin observed in the obese may contribute to the propensity toward AHR in this population (29).

(6)

There are also limited data showing greater systemic inflammation in obese vs lean asthmatics, as measured by serum amyloid A, fibrinogen, and C-reactive protein (39).

Such changes are to be expected because levels of these acute-phase proteins are also elevated in nonasthmatic obese versus lean subjects. However, in some cases, obese asthmatics had higher serum acute-phase proteins than lean asthmatics even after cor- rection for BMI, suggesting that systemic as well as airway inflammation exists in asthma (29).

5.4. Genetics

The possibility that genes known to be important in asthma may also be important in obesity is among the most interesting areas of research in this field. Because genes tend to be pleiotropic, it is biologically plausible that genes important in one complex trait could be important in another. Linkage analysis has identified several linkage peaks with chromosomal regions that are shared for obesity and asthma phenotypes (28).

Chromosomal areas of 5q, 6p, 11q, and 12q all contain regions with loci common to both complex phenotypes. Chromosome 5q contains the G2-adrenergic receptor gene ADRB2 and the glucocorticoid receptor gene NR3C1. Furthermore, ADRB2 encodes a receptor that influences sympathetic nervous system activity, which is important in con- trolling both airway tone and metabolic rate. The glucocorticoid receptor is involved in modulating inflammation important in both diseases. Chromosome 6p, which contains the HLA gene cluster and TNF, influences the immune and inflammatory response important in both these conditions. Chromosome 11q13 contains UCP2, UCP3, and the gene encoding the low-affinity immunoglobulin E receptor FCJRB. The uncoupling proteins (encoded by UCP2 and UCP3) influence metabolic rate but have no known function in asthma. The low-affinity immunoglobulin E receptor is part of the T-helper type 2 inflammatory response, which is increased in asthma and has not been assessed for modification by obesity. Chromosome 12q contains the inflammatory cytokine genes STAT6, IGF1, IL1A, and LTA4H. As already noted, inflammation is a feature common to both obesity and asthma. Research needs to be conducted to relate specific genetic polymorphisms in these and other loci to the effects of the obesity phenotype on asthma (28,40,41).

5.5. Female Sex Steroid Hormones

The association between obesity and asthma has been particularly strong in adult women and postpubertal girls, suggesting that female sex hormones may be contribut- ing to the increased risk of asthma in obesity (5,28,41). Aromatase, the enzyme respon- sible for converting androgens to estrogens, is found in adipose tissue. Therefore, it is reasonable to hypothesize that obesity increases estrogen and is associated with early menarche (42), and the risk of developing asthma is particularly strong in girls with early menarche (25). The two different estrogen receptors (ERs), ERF and ERG, are expressed in adipose tissue. In general, estrogen leads to an increase in basal metabolic rate as well as increased ambulatory activity and decreased activity of lipoprotein lipase in laboratory animals. Mice genetically deficient in ERF, as well as those lacking in aro- matase, are obese; however, oophorectomized ERF mice have much less fat than intact ERF mice, suggesting that the estrogen signaling through ERG may promote fat depo- sition. Although there is some literature on the effect of estrogen on airway responsiveness

(7)

in animal models, it is unclear how estrogen might impact the development of asthma, but both estrogen and progesterone have been shown to increase IL-4 and IL-13 in peripheral blood mononuclear cells (5,28,41), and there may be other effects on immune or inflammatory cells. Pertaining to sex differences, in mice, O3induced injury and inflammation of the lungs was enhanced in both male and female Cpefatmice when compared to their respective gender-matched controls, indicating, at least in obese mice, that gender had no impact augmenting response to O3(34).

5.6. Developmental Effects, Physical Activity, and Diet

Asthma is primarily a disease of early childhood, with 90% of all cases being diag- nosed by age 6. There is increasing evidence that prenatal, neonatal, and early childhood events affect the subsequent development of both asthma and obesity (28,41). Although physical activity has not been shown to diminish the relationship between obesity and asthma in prospective epidemiological studies, physical activity of the mother during pregnancy may be important to the development of the sympathetic nervous system (SNS) in utero (43). For example, activation of brown adipose tissue, which is regulated by the SNS, is important in increasing thermogenesis and basal metabolism through acti- vation of uncoupling proteins. All three types of G-adrenergic receptors are expressed in adipose tissue and hence are relevant to this physiological effect (41,43). Other environ- mental factors acting during pregnancy, such as maternal diet, maternal stress, and ambi- ent temperature, may also affect fetal SNS development. Asthma and obesity share the possibility of decreased or defective SNS activity that may contribute to both disease phenotypes (28,41,43).

A variety of dietary factors have been linked to asthma prevalence in adults and chil- dren. Specifically, antioxidant vitamins C and E, carotene, riboflavin, and pyridoxine may have important effects, with greater intake being associated with enhanced immune function, reduced asthma symptoms, less eczema, and higher lung function (44). Cross- sectional studies have demonstrated a reduced risk of asthma in relation to a high intake of fruits, vegetables, whole-grain products, and fish (44,45). A cross-sectional preva- lence study of 1312 children (mean age = 11.4 yr) showed that frequent consumption of

“fast food type of meals” had a dose-dependent association with asthma symptoms (46).

The benefit of diet for asthma and obesity may be achieved from the combined nutri- tional value in particular foods, from the interaction of foods, or the combined effect of foods in a balanced diet throughout life (44,45). Weight loss achieved through diet inter- vention has been shown to improve lung function and asthma symptoms (47).

Another dietary factor worth greater attention is the omega-3 fatty acids, for which emerging data suggest a protective effect on asthma development in childhood (41,44, 45,48). Unfortunately, most of the work on diet and its effect on asthma has been done in adults or in children after the diagnosis of asthma has already been made. The ideal time in the lifecycle to assess the effects of diet is in the pregnant mother, in whom the effects of total caloric intake and dietary constituents can be measured and their effects on birthweight, obesity, and asthma can be assessed. Barker et al. (49) have proposed that many chronic diseases arise from adaptations the fetus makes when it is undernour- ished. The prototypical example of the relationship of fetal development to both asthma and obesity is the Dutch winter famine of 1944–1945. Women exposed during early and mid-pregnancy to the severe nutritional limitations imposed by the famine had offspring

(8)

of reduced birth size (28,49). Lower lung function and increased risk of death from obstructive airways disease as adults was increased in those exposed to famine in early and mid-gestation, but not in late gestation (49). Interestingly, in follow-up studies, the prevalence of obesity was higher in 19-yr-old men exposed to famine during early to mid-gestation (50), and maternal malnutrition during early gestation was associated with higher BMI and waist circumference in 50-yr-old women but not in men (51).

Pembrey et al. (52) described in a recent study that food and tobacco consumption may have sex-specific, male line transgenerational effects on health and that these transmis- sions are mediated by the sex chromosomes, X and Y.

With respect to asthma, Raby et al. (53) reported a strong relationship between low- normal gestational age and asthma symptoms at age 6 yr. Shaheen and coworkers (54) reported that impaired fetal growth is a risk facture for adult asthma. Low birth weight is associated with lower adult lung function (49) and small lung size is a known risk factor for asthma, likely because small lung size results in small airway caliber (27).

Animal models of obesity may provide clues about whether or how obesity affects lung development. Cpefat mice, which become obese more slowly than lean mice, have normal lung mass at 14 to 16 wk of age, at which time they weigh about 50% more than lean controls. However, there are changes in the pressure–volume curve of their lungs, suggesting that lung development has been affected by the obesity. It is possible that obesity affects lung anatomy, airway branching structure, the nature or distribution of connective tissue, the production of surfactant, or the innervation of the tracheobronchial tree (34). Ultimately, all fetal programming phenomena must have their basis in the altered expression of genes or epigenetic states. Interactions of the in utero environment with fetal genes may thus also contribute to the development of obesity and asthma (28).

6. CONCLUSIONS

There is a significant temporal relationship between alterations in body mass and asthma. The relationship is probably multifactorial and the potential independent influ- ences of biomechanics, inflammation, genetics, and sex-specific effects demonstrate the complex interactions in the complex traits of obesity and asthma. The likelihood of additional direct, interactive, or otherwise related contributions of physical activity, diet, and in utero development to the relationship between obesity and asthma further strengthens this notion. Although complex, this relationship has much to teach about how the environment and genes interact to produce disease phenotypes, and great insight can be gained by considering this potential interrelationship in a developmental context. That there are so many theoretical hypotheses underlying this relationship only enhances the intrigue related to suspected causality.

REFERENCES

1. World Health Organization. Obesity: prevention and managing the global epidemic. WHO Technical Report Series 894. Geneva, Switzerland: 2000.

2. Masoli M, Fabian D, Holt S, et al. Allergy 2004;59;469–478.

3. Hedley AA, Ogden CL, Johnson CL, et al. JAMA 2004;291:2847–2850.

4. Mannino DM, Homa DM, Akinbami L, et al. MMWR CDC Surveill Summ 2000;51:1–13.

5. Weiss ST, Shore S. Am J Respir Crit Care Med 2004;169:963–968.

6. Yunginger JW, Reed CE, O’Connell EJ, et al. Am Rev Respir Dis 1992;146:888–894.

(9)

7. The National Heart, Lung, and Blood Institute Working Group. Chest 1995;108:1380–1392.

8. Kuczmarsiki RJ, Ogden CL, Guo SS, et al. Vital Health Stat 2002;11:1–190.

9. Schaub B, von Mutius E. Curr Opin Allergy Clin Immunol 2005;5:185–193.

10. Luder E, Melnik TA, DiMaio M. J Pediatr 1998;132:699–703.

11. von Mutius E, Schwartz J, Neas LM, et al. Thorax 2001;56:835–838.

12. Belamarich PF, Luder E, Kattan M, et al. Pediatrics 2000;106:1436–1441.

13. Romieu I, Mannino DM, Redd SC, et al. Pediatr Pulmonol 2004;38:31–42.

14. Celedon JC, Palmer LJ, Litonjua AA, et al. Am J Respir Crit Care Med 2001;164:1835–1840.

15. Schachter LM, Salome CM, Peat JK, et al. Thorax 2001;56:4–8.

16. Jarvis D, Chinn S, Potts J, et al. Clin Exp Allergy 2002;32:831–837.

17. Chen Y, Dales R, Krewski D, et al. Am J Epidemiol 1999;150:255–262.

18. Del-Rio-Navarro BE, Fanghanel G, Berber A, et al. J Investig Allergol Clin Immunol 2003;13: 118–123.

19. Chinn S, Jarvis D, Burney P. Thorax 2002;57:1028–1033.

20. Luder E, Ehrlich RI, Lou WY, et al. Respir Med 2004;98:29–37.

21. Camargo CA Jr, Weiss ST, Zhang S, et al. Arch Intern Med 1999;159:2582–2588.

22. Litonjua AA, Sparrow D, Celedon JC, et al. Thorax 2002;57:581–585.

23. Nystad W, Meyer HE, Nafstad P, et al. Am J Epidemiol 2004;160:969–976.

24. Huovinen E, Kaprio J, Koskenvuo M. Respir Med 2003;97:273–280.

25. Guerra S, Wright AL, Morgan WJ, et al. Am J Respir Crit Care Med 2004;170:78– 85.

26. Gilliland FD, Berhane K, Islam T, et al. Am J Epidemiol 2003;158:406–415.

27. Gold DR, Damokosh AI, Dockery DW, et al. Pediatr Pulmonol 2003;36:514–521.

28. Tantisira KG, Weiss ST. Thorax 2001;56(Suppl.2):ii64–ii73.

29. Shore SA, Johnston RA. Pharmacol Ther 2006;110:83–110.

30. Fredberg JJ, Inouye D, Miller B, et al. Am J Respir Crit Care Med 1997;156:1752–1759.

31. Hampel H, Abraham NS, El-Serag HB. Ann Intern Med 2005;143:199–211.

32. Sulit LG, Storfer-Isser A, Rosen CL, et al. Am J Respir Crit Care Med 2005;171:659–664.

33. Visser M, Bouter LM, McQuillan GM, et al. JAMA 1999;282:2131–2135.

34. Johnston RA, Theman TA, Shore SA. Am J Physiol Regul Integr Comp Physiol 2006;290:R126–R133.

35. Weisberg SP, McCann D, Desai M, et al. J Clin Invest 2003;112:1796–1808.

36. Xu H, Barnes GT, Yang Q, et al. J Clin Invest 2003;112:1821–1830.

37. Berg AH, Combs TP, Du X, et al. Nat Med 2001;7:947–953.

38. Arita Y, Kihara S, Ouchi N, et al. Circulation 2002;105:2893–2898.

39. Jousilahti P, Salomaa V, Hakala K, et al. Ann Allergy Asthma Immunol 2002;89:381–385.

40. Weiss ST, Raby BA. Hum Mol Genet 2004;13(Spec No 1):R83–R89.

41. Weiss ST. Nat Immunol 2005;6:537–539.

42. Castro-Rodriguez JA, Holberg CJ, et al. Am J Respir Crit Care Med 2001;163:1344–1349.

43. Young JB, Morrison SF. Diabetes Care 1998;21:B156–B160.

44. McKeever TM, Britton J. Am J Respir Crit Care Med 2004;170:725–729.

45. Tabak C, Wijga AH, de Meer G, et al. Thorax 2005 in press.

46. Wickens K, Barry D, Friezema A, et al. Allergy 2005;60:1537–1541.

47. Stenius-Aarniala B, Poussa T, Kvarnstrom J, et al. BMJ 2000;320:827–832.

48. Oddy WH, de Klerk NH, Kendall GE, et al. J Asthma 2004;41:319–326.

49. Barker DJ, Godfrey KM, Fall C, et al. BMJ 1991;303:671–675.

50. Ravelli GP, Stein ZA, Susser MW. N Engl J Med 1976;295:349–353.

51. Ravelli ACJ, von der Meulen JHP, Osmond C, et al. J Clin Nutr 1999;70:811–816.

52. Pembrey ME, Bygren LO, Kaati G, and The ALSPAC Study Team. Eur J Hum Genet, 2006;14:159–166.

53. Raby BA, Celedon JC, Litonjua AA, et al. Pediatrics 2004;114:e327–e332.

54. Shaheen SO, Sterne JA, Montgomery SM, et al. Thorax 1999;54:396–402.

Riferimenti

Documenti correlati

Mean PI values were lower in PCA than MCA and ACA, suggesting a prefer- ential blood flow redistribution towards the posterior part of the brain however, when translating these

3 when discussing signaling in CLUE, the basic use case behind the CLUE protocol en- visages the presence of two endpoints, each acting as both a Media Provider (MP) and a

Dal confronto tra la generazione degli hedging in esame, come si evince da Figura (7.11), si osserva che anche in questo caso la strategia di copertura prodotta con il metodo standard

radial intensity profile in the disk of DG Tau (Fig. 2 ) shows characteristics in common with the three other disks observed at high angular resolution with ALMA (DM Tau, Loomis et

A CFA model was specified, imposing equality constrains on loadings, error variances and means for subsets of items belonging to the same theoretical domain (Anxiety,

Ipotizzando che ogni Sua skill (presente nell'elenco allegato alla mail) porti con sé una percentuale che rispecchia il Suo grado di padronanza di tale abilità,

Le analisi trascrittomiche microarray, RNAseq, qRT-PCR, che hanno accompagnato il progetto di sequenziamento, hanno inoltre evidenziato una sovra-regolazione di diversi geni

Each measure, thus, able to intervene in a natural way in terms of perturbation on the sections/routes due to the inclusion of the fixed or mobile (construction) road yards as