573
Views
17
CrossRef citations to date
0
Altmetric
BASIC REVIEW

Immunological Effects of Tobacco Smoking in “Healthy” Smokers

Pages 48-58 | Published online: 02 Jul 2009

Abstract

The history of tobacco smoking is quite extensive having existed for many years. Over the past 10–15 years significant gains have been made in curbing exposure to tobacco smoke, but tobacco smoke continues to contribute to significant disease. The immunological effects of cigarette smoking have been evaluated and can provide important insight into mechanism of disease. With the extensive growth in elucidation of basic immune processes within healthy host, further research is possible to aide understanding of immune alterations associated with smoking. Specifically, in the area of cellular immunity in which over 30 different cytokines contributing various regulatory functions in immunity, there are many proteins and mechanisms that may be altered. This review will cover the current state of knowledge in the areas of humoral, cellular immunity including results of cytokine studies in healthy smokers.

INTRODUCTION

History of smoking

The Solanaceae family is a large, diverse and economically important family of shrubs and herbs comprising 94 genera and over 3,000 species. The Solanaceae family includes plants such as belladonna, the potato, tomato, eggplant, peppers, petunias, and the tobacco plant. Tobacco originated in the subtropical regions of North and South America. Best estimates place the origins of the tobacco plant around 6000 BCE and the use of tobacco smoke took place in 1 BCE by inhabitants of the Americas (Citation[1]).

Tobacco smoking is thought to have evolved from the use of smoke during religious ceremonies by priest for various effects including healing and therapeutic benefits. By the time of the arrival of the Spanish in Central America, the Mayan and Aztecs were smoking tobacco in a pipe for its pleasure or intoxicating effects. There are no findings to suggest Greek or Roman use of tobacco for smoking as it is currently known. The origin of tobacco in sub-tropical areas further supports the origin of tobacco smoking in the American continents. This includes Canada where exploration found evidence of Indians who smoked (Citation[1]).

By the early 1500s European explorers introduced smoking to Europe, and initiated cultivation of the plant. Tobacco became a lucrative crop based on increasing use in pipe and cigar smoking, as well as chewing tobacco and the use of snuff. A major advancement for the tobacco industry was in the discovery of the cigarette around 1865, but it was not until World War I that cigarette smoking rapidly grew (Citation[2]). Cigarette smoking reached peaked levels with improvements in curing tobacco, increased production and increased availability of cigarettes during the early 1900s. WW I veterans were provided free cigarettes and had one of the highest rates of tobacco use. Interested readers are encouraged to read the books by Corti and Lock for further history on tobacco smoking.

Over the recent years cigarette use in the United States (US) has decreased. In 2000, approximately 23.3% of adults in the US or 46.5 million adults were current smokers (range 13–30%) compared to 25% in 1993. The prevalence of smoking is highest among persons 18–24 years and 25–44 years of age (Citation[3]). The prevalence of tobacco smoking was estimated at 22.5% in 2002 with successive decreases to 21.6% and 20.9% in 2003 and 2004, respectively. Approximately one-half of adult males smoked in the mid-1960s, compared to less than a third of men in a 2005 report (Citation[4]).

Over 400,000 people die in the United States from cigarette smoking per year (Citation[5]). One in five deaths in the United States is smoking-related. Each year 278,000 men and 152,000 women die from smoking. Men who smoke increase their risk of lung cancer 22 times, and increase the risk of bronchitis and emphysema by nearly 10 times. Women who smoke increase their risk of dying from lung cancer by 12 times and they have more than a 10-fold increased risk of dying from bronchitis and emphysema as compared to non-smoking women.

The effects of cigarette smoking on heart disease are well documented. Cigarette smokers experience a 1.5-to 3-fold or greater risk for coronary heart disease (CHD) than do nonsmokers (Citation[6], Citation[7]). This increased risk is independent of other major risk factors for CHD. Each factor contributes about the same order of magnitude of risk for CHD. When one factor is present, the risk approximately doubles, with two factors the risk is fourfold greater; and when all three are present, the CHD risk is eightfold greater than when none of the three factors are present. The risk of CHD is dose-related when measured by the number of cigarettes smoked per day, and decreases with cessation of smoking (Citation[8]).

Smoking triples the risk of dying from heart disease in middle-age men and women, and may contribute to the inflammatory process seen in atherosclerotic cardiovascular disease. Prospective mortality studies involving over 20 million person-years show a 70% greater CHD death rate in smokers compared to non-smokers. Heavy smokers (two or more pack/day) have almost 200% CHD mortality rate compared to non-smokers (Citation[9]). As well as the risk of cancer and CHD associated with smoking, there are increased risks of other disorders including increased infections and asthma (Citation[14]).

It is well recognized that tobacco smoking causes significant disease and mortality. Therefore, it is very important to understand the pathogenesis associated with tobacco smoking, with particular emphasis on the immune pathogenesis given the large occurrences of infections and neoplasia in smokers.

Immunology of smoking

The immune system is an intricate network of various components and signals executed through various cells, proteins, and genetic signals. The signals contribute to amplification of the response to invaders and regulatory control of the overall immune reactions leading to neutralization and eradiation of foreign agents (toxin or biologic). Over the years details of the immune cells and various subpopulations, cellular components including receptors, cytokines, immunoglubulins, and major histocompatibility (MHC) components have been reported. This complex network may be stimulated, which may lead to immunity or an autoimmune process, or suppressed by a myriad of chemical and biological agents.

The mucosal surfaces are in direct contact with the external environment and a major site of antigenic and toxic exposure in smokers. It has been reported that tobacco smoke contains over 4,000 toxic chemicals, many carcinogens (Citation[15], Citation[16]), as well as the potent immuno-modulator, lipopolysaccharide (LPS) (Citation[17]). Toxins including endotoxin associated with tobacco smoke may impact inflammation contributing to coronary heart disease, as well as altering humoral and cell mediated immunity (Citation[18], Citation[19]). Understanding these effects prior to development of disease can be useful in understanding pathogenesis of the various pulmonary disorders and, perhaps, the overall systemic immune changes that occur in smokers.

This review focuses on and summarizes the results of studies of the immunologic response to tobacco smoking in healthy humans. The work done thus far will be organized based on the major systems within the immune response, namely humoral and cellular immunity.

HUMORAL IMMUNITY

Mucosal response

Secretory IgA (SIgA) is the main immunoglobulin isotype mediating humoral immunity in human secretions at luminal sites including oral, gastrointestinal, respiratory passages as well as in the eye. SIgA production is triggered after translocation of antigen from the lumen to mucosa-associated lymphoid tissue (Citation[20]). The presence of IgG antibodies at the mucosal surface may be protective and it has been show that IgG2 subtype predominates (Citation[21]). IgG in secretions is derived from serum or mucosa (Citation[22]). Studies show tobacco smoke impacts both the systemic and mucosal immunity with changes demonstrated in mucosal antibody production and systemic antibody production. This includes passive smoking, which increases the risk of respiratory disease (Citation[23], Citation[24]).

A summary of studies evaluating mucosal immune response is shown in . These studies generally suggest smokers have a decrease in salivary IgA (sIgA) levels, a form of SIgA (Citation[25], Citation[26], Citation[27], Citation[28], Citation[29], Citation[30]). This is especially seen in heavy (> 20 cigarettes/day) smokers (Citation[14]) and may represent an acute inflammatory effect which resolves once smoking is stopped (Citation[18]). Barton et al. found slightly higher salivary IgM in smokers, however, this is the only study to look for this finding (Citation[28]). Nicotine was not a cause of elevated sIgA in this study.

Griesel et al. determined sIgA levels in people who stopped smoking for at least 2 weeks (Citation[31]). Transient decrease in sIgA occurred followed by return to normal values within 2 weeks of stopping smoking. The level of sIgA in their study ranged from 3.5 mg/dL to 19.2 mg/dL. Evans et al controlled for social class, sex and age, and confirmed smoking to be an independent predictor of lower sIgA (Citation[32]).

Serum antibody response

All five major classes of serum antibodies have been investigated in healthy smokers. The study results were variable as shown in .

Table 1 Smoking effects on humoral immunity

Many studies evaluated IgE levels in smokers (Citation[33], Citation[34], Citation[35], Citation[36]). Increased total IgE was found in both smokers and passive smokers (Citation[37]). IgE elevation in smokers is particularly intriguing since IgE levels tend to decrease with age. Additionally, the increase in serum IgE in smokers is independent of allergic reactivity (Citation[38]). Miyake et al. confirmed elevation of total IgE, but did not find elevation upon exposure to environmental tobacco smoke (Citation[39]). Total IgE elevation was also reported in smokers by Court et al., and the authors noted an epidemiological pattern to elevated IgE levels that suggest a range for “usual” values (Citation[40]). While total IgE elevation was confirmed for smokers, specific IgE (to house dust mite) was not increased. It has also been reported that smoking modifies genetic enhancement of total serum IgE levels (Citation[41]). This is an intriguing report, but there are no other reports to confirm this finding.

Gender-related IgE response has been reported in smokers. A large study by Court 2002 evaluating more than 24,000 subjects found men have significantly higher IgE in both specific and non-specific IgE (Citation[40]). Sherril et al. did not find any sex difference to explain smoking effects on IgE levels (Citation[35]).

In 1980, Gerrard et al. showed lower serum levels of IgG and higher serum IgE in smokers compared to nonsmokers (Citation[42]). The elevated IgE finding was not confirmed in a subset of teachers. The levels of IgA, IgD and IgM were not different. However, another study by Moszczynski showed decrease IgA, IgM and IgG without a change in absolute number or percent of B-cells (Citation[43], Citation[44]).

The mechanism of change in serum IgG level is not known, but there may be a dose-response relationshipwith lower levels of IgG occurring in those with increased smoking (Citation[45]). Furthermore, nicotine was not observed to lead to a change in IgG level. Smoking may enhance underlying disease process in generation of disease specific immunoglobulin as seen in a study in asthmatics (Citation[46]).

Systemic antibody response and development of influenza after vaccination was studied. Cruijff et al. examined the relationship between cigarette smoking and the occurrence of influenza, efficacy of influenza vaccination and antibody response after 3 weeks and 5 months (Citation[47]). There was no relationship between smoking and either serological or clinical influenza. The risk of serological influenza was slightly, but not significantly, elevated in smokers compared to nonsmokers. The decline in serological titers after 5 months was similar for both smokers and nonsmokers. Thus, the authors conclude that smoking has no clinical or preventive risk of influenza in elderly. Smokers had an increased reactivity to influenza vaccine suggesting greater efficacy.

Finally, there were reports of elevated antigen specific immunoglobulins in smokers. This includes the elevation of specific immunoglobulin to a proteinase inhibitor (Citation[48]), and elevation of Chlamydia pneumoniae specific IgA elevation in smokers (Citation[49]). The elevation of the Ig specific anti-α 1 proteinase inhibitor was seen in patients with rheumatoid arthritis, was independent of the underlying disease and related to smoking status. The significance of these findings is not known.

In summary, studies results for secretory, specifically salivary IgA, are mixed. The majority of the studies show decreased in sIgA levels and one study demonstrating the decrease in sIgA as an acute response. The decrease in sIgA may be dose dependent as seen in Bennet et al., where the decrease occurred in heavy smokers (> 20 cigarettes/day). Clinical significance of sIgA change is not known, that is, whether this translates to increased risk of local or systemic infection. Overall, the studies evaluating serum immunoglobulins show a decrease in serum IgA and IgG with one study reporting decreased serum IgM. Serum IgG2 decrease has also been reported and may explain increased respiratory tract infections when considered with the decreased IgA (Citation[45], Citation[74]). Additionally, serum IgE level is increase in smokers. Last, no alteration in specific antibody immunity has been consistently shown.

CELLULAR IMMUNITY

The large number of toxins in cigarette smoke places smokers at risk for immune suppression and diseases including cancer and viral infections. This raises the question as to what are the changes in cell mediate immunity related to smoking. Are the smoke constituents stimulatory or inhibitory? Which cell lines are affected, what are the effects on regulatory cells and cytokines?

In 1998 Morrison found increased number of cells in the broncho-alveolar lavage fluid (BALF) from acute smoking (Citation[50]). The total number of cells recovered was 6.5 times greater in acute smokers compared to nonsmokers. The percentage of neutrophils in BALF from the acute smoking group compared to the nonsmoking group was also higher. Both macrophage and lymphocytes results did not differ between acute smokers and the nonsmoking group. Thus, acute inflammatory response occurs as a result of acute injury and is likely sustained by repeated insults to the lung with continued smoking. Yamaguchi et al. found decreased lymphocytes in the BALF of smokers compared to that of non-smokers, but this was not statistically significant (14 vs 10 × 103/ml fluid) (Citation[51]). Overall, the current literature search provides very little information on cell counts from BALF in smokers without clinical pulmonary disease.

There was one study providing information on peripheral blood lymphocytes in smokers and non-smokers. The study assessed various immunological parameters in 497 men in the U.S. Air Force (Citation[52]). Twenty-seven percent never smoked and 33% of the men were smoking at the time of the study. This study did not present data on the presence or absence of pulmonary disease in the smokers. Overall, active smoking increased all immune cell numbers including total lymphocytes, total T-cells, helper and suppressor T-cells, B-cells, and monocytes. The author did suggest age-specific ranges for these cell populations should be determined.

Effects on cytokines

Cytokines are a diverse family of soluble modulatory proteins and peptides which act as regulatory agents (Citation[53]). They are secreted by various cells in the body including white blood cells, macrophages, fibroblasts and epithelial cells. Cytokines enable and provide communication between cells and have various effects on cellular proliferation, growth, differentiation, homeostasis and other functions. list known cytokines, provides a summary of the source of each cytokine and general function of each class, and a summary of specific cytokines in selected cases.

Table 2 Cytokine families and members (Citation[53])

Interleukins are cytokines originally thought to be produced by white blood cells and key effector proteins in intercellular communication between cells of hematopoietic origin. As the specific identification and source of production became clear, the name “interleukin” did not adhere to original definitions. “Interleukins” will be used based on specific regulatory factor name and “cytokine” will be used to refer to the overall family of cytokines. The number of interleukins continues to grow, and there are now over 30 interleukins.

The lung is directly exposed to the environment and must protect itself from a host of insults including microbial, allergens and toxins. Protection of the lung occurs through various mechanisms including structural components, as well as cough reflex, mucociliary clearance, and the presence of both immune and non-immune cells that elicit protection and recruit cells to eradicate and neutralize noxious agents. Importantly, cytokines, based on their role in cellular recruitment, proliferation, regulation of growth factors, play a key role in the innate and adaptive immune response in the lung (Citation[54]).

The majority of the quantitative studies on cytokines in smokers were performed on BALF. Studies evaluating cytokine levels in sputum and peripheral blood are few in number (). The studies found for this review addressed cytokine production in COPD; however, control populations included “healthy smokers” and non-smokers. This review focuses on cytokines changes in healthy smokers.

Table 3 Summary of cytokine changes found in healthy smokers

Bronchoalveolar studies

In 1989 Yamaguchi reported decreased IL-1 production by alveolar macrophages (AM) from smokers (Citation[51]). The AM were obtained by brochoalveolar lavage, and then cultured. Both the AM cell culture supernatant and supernatant from LPS stimulated AM were evaluated by a bioassay for IL-1 activity. This is one of the earliest reports demonstrating a cytokine production defect in the pulmonary environment of smokers (decreased proportion of lymphocytes in BALF and reduced CD4/8 ratio was also reported). The decrease IL-1 was due to decreased “capacity” of AM cells and not due to inhibition by prostaglandins which was confirmed by using indomethacin. Furthermore, they confirmed results by using enzyme-linked immunosorbent assay for IL-1β.

IL-1 DNA haplotype determined from peripheral blood may be associated with a decline in lung function in smokers (Citation[55]). Joos et al. reported a decline in lung function in smokers associate with IL-1 gene haplotypes. A study by Karjalainen reported IL-1 haplotypes were significantly associated with the rate of decline of lung function in asthmatics, but not controls (Citation[56]). Thus, work by Karjalainen utilizing the same gene polymorphism and same methods in adult non-smoking asthmatics and non-smoking controls indicated the finding in the study by Joos is not limited to smokers.

Additionally, the rate of decline in lung function was the opposite in smoker as compared to asthmatic. The haplotype associated with rapid decline in pulmonary function in the Joos cohort was associated with slow decline in asthma in Karjalainen study; and the haplotype associated with slow decline in pulmonary function in Joos' study was associated with rapid decline in asthma in Karjalainen study. This does not negate the findings, but raises the interesting prospect of variation of cytokine and/or genetic response in different disease process which is modulated or expressed based on other as yet undetermined factors.

As seen in , the majority of studies on BALF and sputa focused on chemokines including IL-8, MCP-1 (CCL-2) and IL-18. This research was likely driven by:

  1. Knowledge of the role in recruitment and activation of neutrophils and monocytes in the airway by IL-8;

  2. Induction of IL-8 by TNF-α;

  3. Possible role of IL-6 in inhibiting inflammation.

McCrea et al. showed BALF in healthy smokers had lower IL-6, but no difference in IL-8 compared to nonsmokers. Furthermore, LPS stimulated macrophages released less TNF and IL-6 (Citation[57]). This was in the presence of smokers having significantly higher number of cells. Two smokers with the highest BALF cell counts had elevated IL-8. Kuschner also found increased number of cells, including macrophages (Citation[58]). The proinflammatory cytokine IL-8 was significantly elevated and its release demonstrated a dose response to cigarette smoking (up to 2 ppd). Secondly, epithelial cells may play role in cytokine release. Mikuniya found no difference in smokers compared to nonsmokers in levels of supernatant IL1β, TNF-α, and IL-8. However, stimulated macrophages had decreased IL-1ra and IL-6(Citation[59]). The decrease IL-1ra also occurred in unstimulated macrophages. Thus, the results appear to be mixed, but stimulatory assays for IL-1, IL-6 and TNF suggest decreased capacity to produce cytokines. Further studies are needed on both IL-1 and IL-8.

Ohta et al. also showed an increased in BALF and decrease in LPS stimulated macrophages IL-8 secretion. Peripheral blood mononuclear cells, stimulated and unstimulated, produced comparable levels of IL-8 (Citation[60]).

Kotani et al. evaluated BALF obtained from orthopedic surgical patients under anesthesia. This study provides useful information regarding gene expression in AM for IL-1β, IL-6, IL-8, IFN-γ, and TNF-α (Citation[61]). This study demonstrated decrease in pro-inflammatory cytokines and showed no difference for the regulatory cytokine IL-8. IL-6 production may require the presence of epithelial cells as well as IL-8 production; however, one cannot ignore the potential role of anesthesia. Additionally, decrease mRNA for all pro-inflammatory cytokines was seen in smokers, with the exception of no difference in IL-6 between groups or change from baseline.

Morrison et al. (Citation[62]) found the same results as McCrea for IL-8. There was no difference in the presence of IL-8, but an increase was seen in LPS stimulated BALF cells in acute smoking (within 1 hour of broncho-alveolar lavage), which further supports the idea of dose effect. A similar finding for the neutrophil chemo-attractants, growth-related oncogene-α and extractable nuclear antigen, was seen in the same study.

Soliman et al. added more issues (Citation[63]). A decreased level of IL-1 and IL-6 was seen in LPS stimulated AM. There was increased intracellular IL-1 however, and the study demonstrated a lack of secretion of IL-1 as previously reported by Brown et al. (Citation[64]). They further demonstrated that some smokers had an inhibitor to IL-6. Thus, in some smokers the antigenic IL-6 level was not different from nonsmokers, but bioactive IL-6 was clearly decreased. Hence, a mixed picture occurs that is possibly explained by changes in the regulatory pathway with production of an inhibitor to IL-6.

Last, Laan and colleagues evaluated IL-16 levels in BALF and peripheral blood lymphocytes (Citation[65]). It is a selective CD4+ T-cell chemoattractant and induces IL-2 receptor expression by airway epithelium and CD8+ T-cells. IL-16 was increased in smokers. This later finding is opposed to population based findings in smokers regarding decreased CD4 numbers and function peripherally.

Sputum studies

Sputa studies on the presence and cytokines in general have been few. Vernooy found increased IL-8 which was partially explained by increased presence of neutrophils (Citation[66]). The TNF-α level was not different between smokers and non-smokers. Soluble TNF receptor levels in sputa and peripheral blood were also investigated. TNF-receptors of epithelial origin were less and there was no difference in the TNF receptor of myeloid origin. The study report also presented results of peripheral blood TNF-receptor of epithelial origin, which did not show any difference between healthy smokers and smokers with COPD.

However, the soluble TNF-receptor level was significantly higher in plasma of COPD patients. It is difficult to draw a conclusion since no data on normal controls were presented. The data on plasma level showed no IL-8 or TNF-α peripherally in healthy smoker. This study was small containing 17 healthy smokers compared to 18 patients with COPD and variable smoking status. This study does highlight a differential effect on cytokine and receptor level in the local (lung) inflammatory site as compared to the systemic findings.

Schulz et al. evaluation of IL-8, TNF-alpha, and IFN-gamma did not show any difference between healthy smokers and controls from BALF, sputa, or primary bronchial epithelial cells (Citation[67]). IL-10 is an important regulatory cytokine that reduces inflammatory response/cytokine production by eosinophils. IL-10 was lower in smokers compared to nonsmokers. Further work showed this was not due to clearance and suggests decreased cytokine production (Citation[68]).

IL-18, originally called an IFN-γ inducing factor, is pro-inflammatory and secreted by airway epithelia and macrophages. While the McKay study was performed to evaluate asthmatics, data are presented for healthy smokers and nonsmokers (Citation[69]). IL-18 level was decreased in healthy smokers as compared to non-smokers. IL-18 mRNA expression in cells from sputum did not significantly differ between smokers and non-smokers and the level of IL-18 in healthy smokers may resemble that seen in stable asthmatics. This result is very intriguing and opens the door to many possibilities for the alteration in production of IL-18 ranging from defects in production to inhibition of production of IL-18. Perhaps there is some altered form of the cytokine leading to increased destruction or elimination.

Peripheral blood studies

There were a few studies evaluating cytokine changes in peripheral blood. A total of 4 studies were found in the literature. Majori et al. evaluated TH-1/TH-2 intracellular cytokine production in subjects with COPD, and also provided results for healthy smokers (Citation[70]). No difference in percentage or absolute numbers of CD3, CD19, CD23 cells, as well as CD4, CD8 cell populations was seen between the healthy smokers and those with overt disease. Cytokine production had to be stimulated in order to detect their presence. INF-γ and IL-4 were not spontaneously present. While the %CD4+ cells with IFN or IL-4 showed a difference between healthy smokers and those with COPD, a normal control population was not available to compare the results of the healthy smokers.

The findings by Miller et al. (Citation[78]) on peripheral blood immunoregulatory T-cells in smokers differed from the findings of Majori. Total T-cell numbers were not different across non-smokers, light (10–19 pack-years), moderate (20–49 pack-years) and heavy smokers (50–120 pack-years). There were less CD4, but increased CD8 lymphocytes. The Miller study was an early report and functional data for the lymphocytes are not available. However, this study suggests, as seen previously in other studies, the possibility of dose-effect phenomena of smoking. The subjects in this study were evaluated for serious illness, immunodeficiency and use of immunosuppressive drugs, however, no data were provided regarding the pulmonary status. Thus, it is not clear that there was no significant pulmonary obstructive disease. The reverse CD4/CD8 ratio was not seen in peripheral blood lymphocytes, but present in BAL cell population in a small study (n = 12 smokers) by Costabel (Citation[79]).

Byron et al. studied IL-4 and IFN-γ production in peripheral blood mononuclear cells (PBMC) using phytohemagglutinin stimulation (Citation[71]). Again, no difference was seen in the production of IFN-γ, but more IL-4 production was found in smokers compared to non-smokers, particularly the heavy smokers. Wirtz and colleagues showed decrease IL-6 and a trend for higher TNF among smokers whose leukocytes were stimulated with LPS (Citation[72]). However, this study only included men.

Immunopathology findings

Finally, there was one immunopathology study of airway subepithelium in smokers with and without COPD (Citation[80]). As seen in studies using peripheral blood and BAL, there were increased numbers of CD8 cells. This finding was only seen when comparing the smokers without COPD to those with COPD. There was no difference in CD8 cell counts when non-smokers were compared to either smoking group. There was no significant difference in tissue eosinophils, CD4, total T-cells or activated T-cells. Interestingly, there was a negative correlation between activated eosinophils as measure by presence of major basic protein and FEV1, and a positive correlation in pack-years and the presence of CD8 cells.

SUMMARY/CONCLUSION

The study results in both healthy and diseased smokers represent a beginning to building a pathogenetic picture of immune reactivity in tobacco smokers. First, the lung as a major and significant immune organ is demonstrated by studies summarized in this review. Second, with the introduction of a considerable number of toxins with local tissue damage and activation of local immune cells including macrophages, epithelial cells, fibroblast, and dendritic cells, both non-specific and specific immune mechanisms are initiated. A host of chemokines and interleukins are produced locally to mediate this activation and recruitment process including the production of inhibitory regulatory factors. Granulocytes and lymphocytes may recruit through mediators as well as through the activation of circulating immune cells within the local vascular and perivascular areas. The circulating activated inflammatory and immune cells can further amplify the immune response in the peripheral blood stream. Although not summarize here, non-specific proteases and reactive oxygen species are produced leading to neutralization of toxins and damage to local tissues as the inflammatory process persist or escalates.

The work on humoral immunologic changes appears clear pointing to a decrease in sIgA and, therefore, decreased airway protection from infection. The immune surveillance defect may be further heightened by the decreased in serum immunoglobulins of smokers, but the significance of peripheral blood immunoglobulin changes is not clear. Further research is needed. The association of quantitative antibody changes in smokers with infection is not clear as shown in Qvarfordt paper on chronic bronchitis. Tobacco smoking has the ability to enhance or suppress antibody response dependent upon the number of cigarettes and perhaps duration of smoking regardless of the number of cigarettes per day. Study populations need clarification particularly with regard to the extent of lung disease. The increase in non-specific IgE is interesting and may represent immunoglobulin class switch stimulated by LPS in tobacco smoke (Citation[81]). The increased IgE could play a role in mast cell mediator release in the overall inflammatory response.

Changes in cytokines represent areas of interest and importance. The changes in cytokine production in healthy smokers seem clear for IL-6, IL-1 and TNF, which generally decrease. These results suggest less inflammation and therefore less tissue damage in healthy smokers. IL-8 results are mixed. It would be useful and important to confirm these results as IL-6, IL-8, IL-1α, and IL-1β are key inflammatory mediators associated with tissue damage. Schulz et al. (Citation[67]) has shown increased constitutive and simulated release of IL-8 in patients with chronic obstructive pulmonary disease, but are there earlier points within the activation sequence of turning on the expression and/or secretion of IL-8 in healthy smokers? Are other inhibitory signal or proteins present as seen with IL-6 and what are the stimuli for these inhibitors? IL-13, a potent stimulator of B-cells especially in allergic individuals, may be present in smokers as an explanation for some of the immunoglobulin findings noted above.

Overall, a key question exists as to the inter-relationship of local pulmonary cytokines, whether found in sputa, BALF, or stimulated mononuclear cells obtained from the lung to that of peripheral blood cytokines remains. What are the sources of cytokines, how are they processed, what are the kinetics? What is/are the primary inflammatory stimulus (stimuli) in smoke? Many questions remain in this area and this may be of importance in understanding disease pathogenesis for infection, malignancy or atherosclerosis. For any toxin the primary treatment is avoidance. Large strides are being made in the United States on smoking education and abstinence, but significant numbers of smokers, many young smokers, remain.

REFERENCES

  • Corti C, England P. A History of Smoking. Bracken Books, London 1996
  • Lock S, Reynolds L A, Tansey E M. Ashes to Ashes: The History of Smoking and Health. Editions Rodopi B V, Amsterdam—Atlanta, GA 1998
  • Center for Disease Control. Cigarette Smoking Among Adults—United States, 2000. MMWR 2002; 51: 642–645
  • Center for Disease Control. Cigarette Smoking Among Adults—United States 2004. MMWR 2005; 54: 1122–1127
  • Center for Disease Control. Annual smoking-attributable mortality, years of potential life lost, and economic cost—United States, 1995–1999. MMWR 2002; 51: 300–303
  • Critchley J A, Capewell S. Mortality risk reduction associated with smoking cessation in patients with coronary heart disease: a systematic review. JAMA. 2003; 290: 86–97
  • U.S. Department of Health and Human Service. Reducing the Health Consequences of Smoking—25 Years of Progress: A Report of the Surgeon General. US Department of Health and Human Services, CDC, Atlanta, GA 1989, DHHS Pub. No. (CDC) 89-8411
  • Office on Smoking and Health. The Health Benefits of Smoking Cessation: A Report of the Surgeon General. US Office of the Surgeon General, Rockville, Md 1990
  • Center for Disease Control. Perspectives in Disease Prevention and Health Promotion Smoking and Cardiovascular Disease. MMWR 1984; 32: 677
  • Branden E, Koyi H, Gnarpe J, Gnarpe H, Tomlin G. Chronic Chlamydia pneumoniae infection is a risk factor for the development of COPD. Respiratory Medicine 2005; 99: 20–26
  • Nuorti J P, Butler J C, Farley M M, Harrison L H, et al. Cigarette smoking and invasive pneumococcal disease. NEJM 2000; 342: 681–89
  • Karl J D, Lebiush M, Rannon L. Cigarette smoking as a risk factor for epidemic A(H1N1) influenza in young men. N Engl J Med 1982; 307: 1042–6
  • Cohen S, Tyrrell D AJT, Russell M AH, Jarvis M J, Smith A P. Smoking, alcohol consumption, and susceptibility to the common cold. Am J Public Health 1993; 83: 1277–1283
  • Gilmour M I, Jaakkola M S, London S J, Nei A E, Rogers C A. How exposure to environmental tobacco smoke, outdoor air pollutants and increased pollen burdens influences the incidence of asthma. Environ Health Perspect 2006; 114: 627–633
  • Hoffman D, Hoffman I. The changing cigarette, 1950–1995. J Toxicol Environ Health 1997; 50: 307–364
  • Smith C J, Livingston S D, Doolittle D J. An international literature survey of IARC Group I carcinogens reported in mainstream cigarette smoke. Food Chem Toxicol 1997; 35: 1107–1130
  • Cerny E H, Levy R, Mauel J, Mpandi M, et al. Preclinical development of a vaccine against smoking. Onkologie 2002; 25: 406–411
  • Hasday J D, Bascom R, Costa J, Fitzgerald T, Dubin W. Bacterial endotoxin is an active component of cigarette smoke. Chest 1999; 115: 829–835
  • Hansson G K. Inflammation, atherosclerosis, and coronary artery disease. New Engl J Med 2005; 352: 1685–1695
  • Kraehenbuhl J P, Neutra M R. Epithelial M cells: differentiation and function. Ann Rev Cell Dev Biol 2000; 16: 301–332
  • Bouvet J, Decroix N, Pamonsinlapatham P. Stimulation of local antibody production: parenteral or mucosal vaccination. Trends Immunol 2002; 23: 209–213
  • Berneman A, et al. The specific patterns of human immunoglobin G antibodies in serum differ from those in autologous secretions. Infect Immun 1998; 66: 4163–4168
  • Kriz P, Bobak M, Kriz B. Parental smoking, socioeconomic factors, and risk of invasive meningococcal disease in children: a population based case-control study. Arch Dis Child 2000; 83: 117–121
  • Strachan D P, Cook D G. Parental smoking and allergic sensitization in children. Thorax 1998; 53: 117–123
  • Bennet K R, Reade P C. Salivary immunoglobulin A levels in normal subjects, tobacco smokers, and patients with minor aphthous ulceration. Oral Surg Med Oral Pathol 1982; 53: 461–465
  • Hersey P, Prendergast D, Edwards A. Effects of cigarette smoking on the immunes system. Med J Aust 1983; 2: 425–429
  • Jedrychowski W A, Maugeri U, Adanczyk B. Effect of smoking on serum immunoglobulins and cellular blood constituents in healthy individuals. G Ital Med Lav 1986; 8: 53–56
  • Barton J R, Riad M A, Gaze M N, Maran A GD, Feguson A. Mucosal immunodeficiency in smokers, and in patients with epithelial head and neck tumors. Gut 1990; 31: 378–382
  • Ussher M, West R, Evans P, Steptoe A, et al. Acute reduction in secretory immunoglobulin A following smoking cessation. Psychoneuroendocrinology 2004; 29: 1335–1340
  • Olson B L, McDonald M J, Jr, Gleason M J, Stookey G K, et al. Comparisons of various salivary parameters in smokers before and after the use of a nicotine-containing chewing gum. J Dent Res 1985; 64: 826–830
  • Griesel A G, Germishuys P J. Salivary immunoglobulin A levels of persons who have stopped smoking. Oral Surg Oral Med Oral Pathol Oral Radiol Endod 1999; 87: 170–173
  • Evans P, Ford G D, Ford G, Hucklebridge F, et al. Social Class, sex, and age differences in mucosal immunity in a large community sample. Brain Behav Immun 2000; 14: 41–48
  • Barbee R A, Halonen M, Kaltenborn W, Lebowitz M, Burrows B. A longitudinal study of serum IgE in a community cohort: correlations with age, sex smoking, and atopic status. J Allergy Clin Immunol 1987; 79: 919–927
  • Oryszczyn M P, Annesi I, Neukirch F, Dore M F, Kauffmann F. Relationships of total IgE level, skin prick test response, and smoking habits. Ann Allergy 1991; 67: 355–358
  • Sherill D L, Halonen M, Burrows B. Relationships between total serum IgE, atopy and smoking: a twenty-year follow-up analysis. J Allergy Clin Immunol 1994; 94: 954–962
  • Jarvis D, Luczynska C, Chinn S, Burney P. The association of age, gender and smoking with total IgE and specific IgE. Clin Exp Allergy 1995; 25: 1083–1091
  • Oryszczyn M P, Annesi-Mawsano I, Charpin D, Paty E, et al. Relationships of active and passive smoking to total IgE in adults of the epidemiological study of the genetics and environment of asthma, bronchial hyper-responsiveness, and atopy (EGEA). Am J Respir Crit Care Med 2000; 161: 1241–1246
  • Raherison C, Nejjari C, Marty M L, Filleui L, et al. IgE level and Phadiatop® in an elderly population from the PAQUID cohort: relationship to respiratory symptoms and smoking. Allergy 2004; 59: 940–945
  • Miyake Y, Miyamoto S, Ohya Y, Sasaki S, et al. Relationship between active and passive smoking and total serum IgE levels in Japanese women: baseline data from Osaka maternal and child health study. Int Arch Allergy Immunol 2004; 135: 221–228
  • Court C S, Cook D G, Stachan D P. The descriptive epidemiology of house dust mite-specific and total immunoglobin E in England using an notionally representative sample. Clin Exp Allergy 2002; 32: 1033–1041
  • Liu X, Beaty T H, Deindl P, Huang S K, et al. Association between total serum IgE levels and the 6 potentially functional variants within the genes IL-4, IL-13, and IL-4RA in German children: the German multicenter atopy study. J Allergy Clin Immunol 2003; 112: 382–388
  • Gerrard J W, Heiner D C, Ko C G, Mink J, et al. Immunoglobulin levels in smokers and non-smokers. Ann Allergy 1980; 44: 261–262
  • Moszczynski P, Moszczynski P, Jr, Slowinski S. Indicators of humoral immunity and acute phase reaction in cigarette smokers. Pol Arch Med Wewn 1990; 83: 194–199
  • Moszczynski P, Zabinski A, Moszczynski P, Jr, Rutowski J, et al. Immunological findings in cigarette smokers. Toxicol Lett 2001; 118: 121–127
  • Gyllen P, Andersson B A, Qvarfordt I. Smokeless tobacco or nicotine replacement therapy has no effect on serum immunoglobulin levels. Respir Med 2004; 98: 108–114
  • Mitsunobu F, Ashida K, Hosaki Y, Tsugeno H, et al. Influence of long term cigarette smoking on immunoglobulin E mediated allergy, pulmonary function and high-resolution computed tomography lung densitometry in elderly patients with asthma. Clin Exp Allergy 2004; 34: 59–64
  • Cruijff M, Thijs C, Govaert T, Aretz K, et al. The effect of smoking on influenza, influenza vaccination efficacy and on the antibody response to influenza vaccination. Vaccine 1999; 17: 426–432
  • Hutchinson D, O'Leary C, Nixon N B, Mattey D L. Serum complexes of immunoglobulin A-alpha1 proteinase inhibitor in rheumatoid arthritis: association with current cigarette smoking and disease activity. Clin Exp Rheumatol 2002; 20: 387–391
  • Von Hertzen L, Kaprio J, Koskenvuo M, Isoaho R, Saikku P. Humoral immune response to Chlamydia pneumoniae in twins discordant for smoking. J Intern Med 1998; 244: 227–234
  • Morrison D, Strieter R M, Donnelly S C, Burdick M D, et al. Neutrophil chemokines in bronchoalveolar lavage fluid and leukocyte conditioned medium fro nonsmokers and smokers. Eur Respir J 1998; 12: 1067–1072
  • Yamaguchi E, Okazaki N, Itoh A, Abe S, et al. Interleukin 1 production by alveolar macrophages is decreased in smokers. Am Rev Respir Dis 1989; 140: 397–402
  • Wolfe W H, Miner J C, Michalek J E. Immunological parameters in current and former US Air Force personnel. Vaccine 1993; 11: 545–547
  • Vilcek J. The cytokines: An overview. The Cytokine Handbook, Fourth edition, A G Thomson, M T Lotze. Elsevier Science, London 2003
  • Strieter R M, Belperio J A, Keane M P. Host innate defenses in the lung: the role of cytokines. Curr Opin Infect Dis 2003; 16: 193–198
  • Joos L, McIntyre L, Ruan J, Connett J E, et al. Association of IL-1β, and IL-1 receptor antagonist haplotypes with rate of decline in lung function in smokers. Thorax 2001; 56: 863–866
  • Karjalainen J, Julkkonen J, Hurme M, Joos L, Pare P K, Sandford A. IL-1 haplotypes and lung function decline. Thorax 2002; 57: 561–562
  • McCrea K A, Ensor J E, Nall K, Bleecker E R, Hasday J D. Altered Cytokine regulation in the lungs of cigarette smokers. Am J Respir Crit Care Med 1994; 150: 696–703
  • Kuschner W G, D'Alessandro A, Wong H, Blanc P K. Dose dependent cigarette smoking related inflammatory responses in healthy adults. Eur Respir J 1996; 9: 1989–1994
  • Mikuniya T, Nagai S, Tsutsumi T, Morita K, et al. Proinflammatory or regulatory cytokines released from BALF macrophages of healthy smokers. Respiration 1999; 66: 419–426
  • Ohta T, Yamashita N, Maruyama M, Sugiyama E, Kobayashi M. Cigarette smoking decreases ineerleukin-8 secretion by human alveolar macrophages. Respir Med 1998; 92: 922–927
  • Kotani N, Hashimoto H, Sessler D I, Yoshida H, et al. Smoking deceases alveolar macrophage function during anesthesia and surgery. Anesthesiology 2000; 92: 1268–1277
  • Morrison D, Strieter R M, Donnelly S C, Burdick M D, et al. Neutrophil chemokines in bronchoalveolar lavage fluid and leukocyte conditioned medium fro nonsmokers and smokers. Eur Respir J 1998; 12: 1067–1072
  • Soliman D M, Twigg H L, III. Cigarette smoking decreases bioactive interlieukin-6 secretion by alveolar macrophages. Am J Physiol Lung Cell Mol Physiol 1992; 63: L471–L478
  • Brown G P, Iwamoto G K, Monick M M, Gunninghake G W. Cigarette smoking deceases interleukin-1 release by human alveolar macrophages. Am J Physiol Cell Physiol 1989; 256: C260–C264
  • Laan M, Qvarfordt I, Riise G C, Andersson A A. Increased levels of interleukin-16 in the airways of tobacco smokers: relationship with peripheral blood T lymphocytes. Thorax 1999; 54: 911–916
  • Vernooy J H, Kucukayacan M, Jacobs J A, Chavannes N H, et al. Local and systemic inflammation in patients with chronic obstruction pulmonary disease. Am J Respir Crit Care Med 2002; 166: 1218–1224
  • Schulz C, Wolf K, Harth M, Kratzel K, et al. Expression and release of interleukin-8 by human bronchial epithelial cells from patients with chronic obstructive pulmonary disease, smokers and never smokers. Respiration 2003; 70: 254–261
  • Takanashi S, Hasegawa Y, Kanehira Y, Yamamoto K, et al. Interleukin-10 level in sputum is reduced in bronchial asthma, COPD and in smokers. Eur Respir J 1999; 14: 309–314
  • McKay A, Komai-Koma M, MacLeod K J, Campbell C C, et al. Interleukin-18 levels in induced sputum are reduced in asthmatic and normal smokers. Clin Exp Allergy 2004; 34: 904–910
  • Majori M, Corradi M, Caminati A, Cacciani G, et al. Predominate Th-1 cytokine pattern in peripheral blood from subjects with chronic obstructive pulmonary disease. J Allergy Clin Immunol 1999; 103: 458–462
  • Byron K A, Varigos G A, Wootton A M. IL-4 production is increased in cigarette smokers. Clin Exp Immunol 1994; 95: 333–336
  • Wirtz P H, von Kanel R, Kunz-Ebrecht S, Ehlert U, Fischer J E. Enhanced glucocorticoid sensitivity of cytokine release from circulating leukocytes stimulated with lipopolysaccharide in healthy male smokers. Brain Behav Immun 2004; 18: 536–543
  • Graswinckel J EM, ban der Velden U, van Winkelhoff A J, Hoek F J, et al. Plasma antibody levels in periodontitis patients and controls. J Clin Periodontol 2004; 31: 562–568
  • Qvarfordt I, Riise G C, Andersson B A, Larsson S. IgG subclasses in smokers with chronic bronchitis and recurrent exacerbations. Thorax 2001; 56: 445–449
  • Conti P, Kempuraj D, Frydas S, Kandere K, et al. IL-10 subfamily members: IL-19, IL-20, IL-22, IL-24 and IL-26. Immunol Lett 2003; 88: 171–174
  • Jordan W J, Eskdale J, Boniotto M, Lennon G P, et al. Human IL-19 regulates immunity through auto-induction of IL-19 and production of IL-10. Eu J Immunol 2005; 35: 1576–1582
  • Hagiwara E, Takahashi K, Okubo T, Ohno S, et al. Cigarette smoking depletes cells spontaneously secreting Th 1 cytokines in human airway. Cytokine 2001; 14: 121–126
  • Miller L G, Goldstein G, Murphy M, Ginns L C. Reversible alterations in immunogrgulatory T cells in smoking. Analysis by monoclonal antibodies and flow cytometry. Chest 1982; 82: 526–529
  • Costabel U, Bross K J, Reuter C, Ruhle K H, Matthys H. Alterations in immunoregulatory T cells subsets in cigarette smoker. A phenotypic analysis of bronchoaleveolar and blood lymphocytes. Chest 1986; 90: 39–44
  • Lams B EA, Lousa A R, Rees P J, Lee T H. Subepithelial immunopathology of the large airways in smokers with and without chronic obstructive pulmonary disease. Eur Respir J 2000; 15: 512–516
  • Snapper C M, Pecanha L MT, Levine A D, Mond J J. IgE class switching is critically dependent upon the nature of the B-cell activator, in addition to the presence of IL-4. J Immunol 1991; 147: 1163–1170

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.