Open access peer-reviewed chapter

Helicobacter pylori Infection and Diabetes Mellitus

Written By

Saeda Haj, Michal Raviv and Khitam Muhsen

Submitted: 29 May 2015 Reviewed: 21 April 2016 Published: 07 July 2016

DOI: 10.5772/63826

From the Edited Volume

Extradigestive Manifestations of Helicobacter Pylori Infection - An Overview

Edited by Bruna Maria Roesler

Chapter metrics overview

1,973 Chapter Downloads

View Full Metrics

Abstract

Helicobacter pylori colonizes the stomach and causes chronic gastritis, which most often remains asymptomatic. However, in a small proportion of infected persons, it causes peptic ulcers and gastric cancer. We reviewed recent evidence of the association between H. pylori infection and diabetes mellitus (DM). Numerous studies have shown a positive association between H. pylori infection and DM, however, findings are still conflicting. Such a link is biologically plausible, given the importance of the stomach in the homeostasis of systems outside the digestive tract; however, the mechanisms by which H. pylori might affect the risk of DM are not clear. Current knowledge indicates that H. pylori infection can affect the regulation of ghrelin and leptin, two hormones that play central roles in energy homeostasis in humans. Yet, methodological limitations are present in studies that addressed the relationships of H. pylori infection with DM and with possible risk factors for DM, including inadequate control of confounders. The important question of whether H. pylori eradication might be beneficial for glycemic control in diabetic patients is still unresolved. Future well-designed studies are needed to address these research questions, which are of clinical and great public health significance.

Keywords

  • Helicobacter pylori
  • diabetes mellitus
  • epidemiology

1. Introduction

Helicobacter pylori is a gram-negative bacterium that colonizes the stomach and causes persistent infection. The infection is typically acquired in the first few years of life [13]. The associated risk factors of H. pylori infection include living in crowded households, low socioeconomic conditions and infected family members [46]. The infection is common worldwide with highest prevalence rates reaching 80–90% in developing countries and underprivileged communities [7], while a much lower prevalence of 20–50% is recorded in developed countries [7].

H. pylori infection has two phases: an acute phase and a chronic course. Acute H. pylori infection is rarely diagnosed. Following establishment of the infection, chronic gastritis develops; however, most infected people remain asymptomatic and only 10–20% of them develop peptic disease during their lifetime [7]. H. pylori causes gastric and duodenal ulcers, and in rare occasions distal gastric cancer and mucosa-associated lymphoid tissue (MALT) lymphoma [7]. These diseases are the main indications to test and treat H. pylori infection [8], in addition to unexplained iron deficiency anemia (IDA) and idiopathic thrombocytopenic purpura [8]. Although H. pylori infection is acquired in childhood, peptic ulcer disease typically occurs in adulthood.

Following H. pylori colonization, rigorous local and systemic immune responses develop. However, these do not clear the infection but rather contribute to the damage of the gastric mucosa [7, 9, 10]. H. pylori simulates the innate immune response, as well as humoral and cell-mediated immune responses [9, 10]. The predominant human T cell response is the T-helper 1 mediated response, which is associated with releasing proinflammatory cytokines and activation of phagocytes [9, 10]. H. pylori also induces Th2 and T-regulatory (Tregs) responses [9, 10]. The importance of Treg response is in both controlling inflammation and promoting the persistence of the infection [9, 10].

H. pylori-associated gastric pathology develops over time in a progressive manner [1113], and the damage to gastric mucosa can be observed even in asymptomatic persons [14]. Today it is clear that host (e.g., age, genetic susceptibility), agent (virulence antigens) and environment-related factors are important in the development of H. pylori-associated gastroduodenal diseases [9]. For example, host genetic polymorphisms that lead to increased release of proinflammatory cytokines are associated with increased gastric cancer risk [9]. Pathogenesis is dependent on a Th1-acquired immune response and on hormonal changes including hypergastrinemia [9]. Regarding pathogen virulence factors, most H. pylori strains carry the cag pathogenicity island that encodes for a type IV secretory apparatus, which allows translocation of cytotoxin-associated gene A (CagA) protein into the host cell. This, together with the vacuolating cytotoxin (VacA), plays a major role in the pathogenesis of gastroduodenal diseases [7, 9, 1517]. Novel H. pylori antigens have been identified recently [18], some of which were found to be associated with atrophic gastritis and gastric cancer risk such as GroEL [18], Helicobacter cysteine-rich protein (HcpC) [19], outer membrane protein (Omp) and others [20, 21].

Several studies have shown associations between H. pylori infection and various extragastric diseases [22]. H. pylori infection was positively linked with adulthood chronic diseases such as cardiovascular disease [2325], dementia [2628], insulin resistance and diabetes mellitus (DM) [22, 29, 30]. The mechanisms of such associations are not fully understood, and it is not clear whether such associations are causal or not. This chapter will focus on the association between H. pylori infection and DM.

Advertisement

2. H. pylori infection, changes in gastric physiology and metabolic hemostasis

Although the role of H. pylori infection in the pathogenesis of gastroduodenal diseases [7, 9, 17, 31] is well established, its impeding effects on metabolic homeostasis and DM are not clear. The stomach plays a major role in the homeostasis of systems outside the digestive tract. Therefore, the link between H. pylori-chronic gastritis and metabolic homeostasis and DM seems biologically plausible.

H. pylori-induced inflammation and its severity affect gastric physiology. For example, H. pylori leads to hormonal changes in the stomach, such as reduced production of somatostatin and hypergastrinemia [9]. H. pylori-gastritis also alters the secretion of gastric acid [32, 33]; increased secretion of gastric acid is associated with antral-predominant phenotype and increased risk of duodenal ulcers [9, 10]. H. pylori infection can reduce gastric acid production, and this is typically associated with corpus-predominant gastritis and increased likelihood of gastric ulcer and gastric adenocarcinoma [9, 10]. Moreover, H. pylori infection is associated with reduced gastric ascorbic acid levels [34]. H. pylori affects the levels of pepsinogen (PG) I and PGII; proenzymes of the digestive enzyme pepsin. PGI is secreted from cells in the corpus and PGII is also secreted from cells in the antrum and duodenum [35, 36]. About 1% of PGs can be found in the serum. Serum PGI and PGII are increased in H. pylori infected vs. uninfected individuals, and higher levels are found in more severe gastritis. As the severity of gastritis progresses and corpus atrophic lesions appear, the PGI level decreases, while the PGII level remains stable; the result is a decrease in the PGI:PGII ratio [37, 38]. These markers have clinical significance, and they predict various gastric pathologies [16, 3740].

In addition, H. pylori infection can affect the regulation of ghrelin and leptin [4147], two hormones that play central roles in energy homeostasis [48]. Ghrelin reduces energy expenditure and promotes weight gain [4850], while leptin decreases appetite and increases energy expenditure [48]. Both hormones are secreted by the epithelial cells in the stomach [48, 51]. The relationship between H. pylori and these hormones appears to be complex. While several studies reported no association between H. pylori infection and circulating leptin [43, 45, 46, 5254] and ghrelin levels [45, 52, 54], others found lower levels of one or the two hormones in H. pylori infected vs. uninfected individuals [41, 42, 44]. There also appears to be differences in gastric mucosa levels of these hormones, according to H. pylori infection [41, 42, 47, 5254]. Moreover, H. pylori eradication seems to affect these hormones as well [41, 43, 45, 47, 52] (Table 1).

Study Exposure Ghrelin Leptin
Circulating levels Gastric mucosa levels Circulating levels Gastric mucosa levels
Isomoto et al. [44] H. pylori infection ND ND
H. pylori eradication NS NS ND ND
Chuang et al. [46] H. pylori infection Males: ↓
Females: NS
ND NS ND
Jun et al. [52] H. pylori infection NS NS NS
Nishi et al. [53] H. pylori infection ND ND NS
H. pylori eradication ND ND NS
Francois et al. [45] H. pylori infection NS ND NS ND
H. pylori eradication Pre-meal: NS
Post meal: ↑
ND ND
Azuma et al. [47] H. pylori infection ND ND
H. pylori eradication ND ND NS
Jang et al. [54] H. pylori eradication NS NS ND
Roper et al. [41] H. pylori infection NS Fundic: NS
Antral: NS
Gastric juice:↑
Fundic: NS
Antral:↓
Breidert et al. [43] H. pylori infection ND ND NS Antrum: NS
Corpus:↑

Table 1.

Selected studies that addressed associations of H. pylori infection and H. pylori eradication with ghrelin and leptin levels

NS, no significant difference; ND, not determined;, ↑ increase;, ↓ decrease.


Altogether, these studies suggest that H. pylori can alter gastric physiology, which can in turn affect metabolic homeostasis and the risk of DM.

Advertisement

3. H. pylori infection and diabetes mellitus

DM refers to a group of metabolic disorders that manifest with hyperglycemia. DM is classified based on the pathogenic course that results in hyperglycemia, with two broad categories designated as type 1 DM (T1DM) and type 2 DM (T2DM). T1DM is the result of interaction among genetic, environmental and immunological factors that eventually leads to destruction of beta cells in the pancreas and complete or near-complete insulin deficiency. T2DM consists of various disorders with variable levels of insulin resistance, impaired insulin secretion and increased glucose production. T1DM usually occurs in childhood and adolescence, and comprises 5–10% of all DM cases [55]. T2DM typically develops in adulthood and is responsible for the majority (90–95%) of DM cases [55].

DM is a major public health problem [5660], causing an enormous burden to patients and their families, as well as to health care systems. The prevalence of T2DM is increasing globally [5660] due to increases in life expectancy and obesity [56, 58]. It is estimated that 240 million people have T2DM, and that in 2025 about 380 million will have the disease, while 418 million will have impaired glucose tolerance (IGT) [56]. The burden of DM is amplified given its significant macro and microvascular complications (such as cardiovascular disease, kidney disease), in addition to peripheral neuropathy [55].

There are well-established risk factors for T2DM [6167], including sociodemographic factors [64, 68, 69], lifestyle factors (e.g., obesity, physical inactivity, poor diet [6167]) and high glucose levels reflecting IGT [65, 66]. Changes in diet (i.e., higher consumption of whole grain products and exchanging unsaturated fat for saturated fat), and in particular physical activity and avoidance of obesity, can prevent T2DM through changes in body fat and other mechanisms [61, 67, 7072]. These may reduce the incidence of DM by 28–59% [72]. Such interventions are also important for better control of diabetes [70, 73]. Current evidence suggests that there must be additional factors besides lifestyle that contribute to the occurrence of DM.

In addition to the association mentioned above, between H. pylori infection and ghrelin and leptin [41, 45, 7480], associations have been reported of H. pylori infection with glycated hemoglobin levels (Hb1Ac) [81], as well as with disturbances in metabolic homeostasis including insulin resistance; the latter according to a recent literature review and a systematic review [22, 82]. These findings support the postulation that H. pylori infection may be involved in the etiology of the emerging pandemic of obesity and DM, and in diabetes-related complications.

Associations of H. pylori infection with DM incidence [30, 83, 85] have been reported. Recent meta-analyses showed a significant 1.7 to 2-fold higher prevalence of H. pylori infection in persons with T2DM vs. non-diabetic individuals [84, 85]. In some of the studies that reported a positive association between H. pylori infection and DM [30, 8688], the association became non-statistically significant after adjustment for potential confounders such as age and socioeconomic status [87, 88]. Other studies reported no significant association between H. pylori and DM [8992], or a significant association only in persons with BMI>25 [81] (Table 2). Several studies did not control adequately for socioeconomic status and for traditional risk factors of DM, such as obesity and physical inactivity. Furthermore, most of the evidence is based on small-scale hospital-based case–control studies, in which the source population, selection of control population and representativeness of the sample were not fully described. For these reasons, inference and generalizability of findings from such studies should be done with caution. On the other hand, recent well-designed studies show convincing evidence of the potential involvement of H. pylori infection in the occurrence of DM, and possibly in IGT. A large population-based follow-up investigation of elderly persons has demonstrated a significant two-fold increased risk of DM in H. pylori infected vs. uninfected persons, even after controlling for possible confounders, while such an association was not observed for other pathogens [30]. A large well-designed and thoroughly analyzed survey that utilized nationwide data (N~13,000) from the United States indicated no significant association between H. pylori infection and self-reported diabetes. However, among individuals with BMI>25 kg/m2 who were assessed in the 1999–2000 National Health & Nutrition Examination Survey (NHANES), DM was more prevalent among those who were H. pylori seropositive than those who were H. pylori-seronegative[81] (Table 2). Moreover, that study showed that H. pylori infected persons, especially those infected with CagA strains, had significantly elevated mean HbA1c levels compared with those who were H. pylori seronegative [81].

Study Study population Study design Hp detection Outcome Findings Adjusting for confounders
Jeon et al. [30]
California
N=782 diabetes free individuals at baseline
Age >60 years
Prospective cohort Serum IgG
by ELISA
DM Adjusted HR 2.69 (95% CI: 1.10–6.60) Sex, education, smoking, cholesterol, DBP, HSV-1
Hsieh et al. [86]
Taiwan
N=903 Hp infected patients aged 57.16±11.64 years
N=1167 uninfected patients aged 56.57±13.34 years
Cross-sectional Gastric biopsy: culture, histology and rapid urease test T2DM OR 1.67 (95% CI: 1.19–2.35)
Chen and Blaser [81]
USA
Data from NHANES III
N=7417 age ≥18 years
NHANES 1999–2000
N=6072 age ≥3 years
Cross-sectional Serum IgG
by ELISA
DM NHANES 1999–2000:
Adjusted OR: 1.30 (95% CI: 0.94–1.80)
BMI>25
OR (1.43; 95% CI: 1.00–2.03)
NHANES III:
Adjusted OR: 0.99 (95% CI: 0.80–1.23)
Age, sex, race, BMI, smoking, education
El-Eshmawy et al. [111]
Egypt
N=162 T1DM patients aged 19.35±2.6 years
N=80 healthy subjects aged 19.76±2.76 years
Case-control Serum IgG and IgA by ELISA T1DM OR 3.67 (95% CI: 2.07–6.55) Matching by age, sex, SES
Longo-Mbenza et al. [91]
Democratic Republic of the Congo
N=128 patients with Hp infection aged 53.4±12.9 years
N=77 uninfected patients aged 52.5±16.6 years
Prospective cohort
Follow-up 9.6±0.8 years
Serum IgG
by ELISA
DM OR: 0.97 (95% CI: 0.35–2.86)
Xia et al. [89]
Australia
N=49 T1DM and N=380 T2DM (aged 60.7±13.3 years)
N=170 non-diabetic controls aged 60.4± 11.3 years
Case-control Serum IgG
by ELISA
T1DM
T2DM
Overall 0.94 (95% CI: 0.65–1.39)
T2DM: OR: 1.03 (95% CI: 0.71–1.52)
T1DM: OR: 0.40 (95% CI: 0.15–0.94)
Demir et al. [90]
Turkey
N=141 T2DM patients aged 52±8.2 years
N=142 non-diabetic subjects aged 51±9.3 years
Case-control Gastric biopsy: rapid urease test and histology T2DM OR: 1.15 (95% CI: 0.71–1.85)
Colombo
et al. [112]
Italy
N=138 T1DM patients aged 12.0±3.4 years
N=138 controls aged 12.2±2.0 years
Case-control Serum IgG and IgA by ELISA T1DM OR: 0.87 (95% CI: 0.52–1.46) Matching by age
Cenerelli
et al. [92]
Italy
N=30 T2DM patients aged 55.7±9.7 years
N=43 controls aged 51.2±11.3 years
Case-control UBT T2DM OR: 1.06 (95% CI: 0.41–2.76)
Dore
et al. [87]
N=145 T1DM and N=240 T2DM
N=506 controls
(ages 12–75 years)
Case-control Serum IgG
by ELISA
T1DM
T2DM
T1DM: 0.59 (95%CI: 0.40–0.87)
T2DM: 2.08 (95%CI: 1.52–2.85)
In stratified analysis by age group, SES, the differences were not significant
Lutsey et al. [88] N= 1000 ages 45–84 years Cross-sectional Serum IgG
by ELISA
DM Crude OR: 1.65 (95%CI: 1.16–2.34)
Adjusted OR: 1.12 (0.78–1.62)
Age, sex, rate, education and site

Table 2.

Selected epidemiological studies that examined an association between H. pylori infection and diabetes mellitus

BMI, body mass index; CI, confidence intervals; DM, diabetes mellitus; DBP, diastolic blood pressure; ELISA, enzyme-linked immunosorbent assay; Hp, Helicobacter pylori; HR, hazard ratio; HSV-1, Herpes simplex virus 1; IgA, immunoglobulin A; IgG, immunoglobulin G; NHANES, National Health & Nutrition Examination Survey; OR, odd ratio: SES, socioeconomic status; T1DM, type 1 diabetes mellitus; T2DM, type 2 diabetes mellitus; UBT, urea breath test.


Advertisement

4. H. pylori infection and glycemic control among diabetic patients

Given the observed associations between H. pylori infection and various metabolic and glycemic measures, the question arises of whether H. pylori infection and/or H. pylori eradication can affect glycemic control in diabetic patients. If indeed H. pylori infection plays a role in glycemic control, H. pylori eradication might be beneficial to diabetic patients. A recent meta-analysis that included 14 observational studies involving 1781 diabetic patients (both T1DM and T2DM) showed no significant difference in mean HbA1c values among H. pylori infected individuals compared with those uninfected; mean difference 0.19% (95% CI: −0.18 to 0.46), (Pv=0.16) [93]. In contrast, another meta-analysis involving 11 studies and 513 patients reported significantly higher HbA1c values among H. pylori-infected diabetic persons than among uninfected ones: weighted mean difference 0.43 (95% CI: 0.07–0.79), (Pv=0.02) [94]. The discrepancy in results between the two meta-analyses can be explained by differences in their criteria of study selection, which determined the number and quality of the studies analyzed.

Table 3.

Helicobacter pylori eradication and glycemic control in diabetic patients

The question of whether H. pylori eradication can improve glycemic control was assessed in a limited number of observational studies, most of them were small scale [95101] (Table 3). Findings from these studies were conflicting, ranging from no difference, to small non-significant or borderline improvements from baseline to up to 2-years after eradication [95101] and to a significant decrease from baseline, in HbA1c at 3 months after H. pylori eradication [95]. A pooled analysis of two studies that compared mean differences in HbA1c between diabetic individuals who had undergone successful H. pylori eradication and those whose H. pylori eradication therapy had failed, showed no significant difference between the groups [94]. The optimal study design to examine the effect of H. pylori eradication therapy on glycemic control is a randomized controlled trial with intention-to-treat analysis, in which diabetic patients are assigned to either an H. pylori eradication group or a placebo control group. However, to-date such trials are lacking, and the current evidence is based on observational studies, which are evidently prone to biases and confounders. Therefore, the question of whether H. pylori infection affects glycemic control in diabetic patients remains unresolved.

Advertisement

5. H. pylori infection and metabolic syndrome

Metabolic syndrome is a cluster of metabolic risk factors that are associated with increased risk for atherosclerotic cardiovascular disease, T2DM and their complications. These factors include atherogenic dyslipidemia (elevated triglycerides and apolipoprotein B, increases small low-density lipoproteins [LDL], and low concentration of high-density lipoproteins [HDL]), elevated blood pressure and elevated fasting glucose levels known as impaired fasting glucose (IFG) or prediabetes [102, 103], which lead to a prothrombotic and proinflammatory state. The main risk factors for metabolic syndrome include obesity, mainly abdominal obesity and insulin resistance [103], as well as aging, physical inactivity and diet rich with saturated fat and cholesterol [103].

Recent studies have tested the hypothesis of a positive association between H. pylori and metabolic syndrome [22, 104106]. While the underlying mechanisms remain to be determined, the inflammatory response to infection and secretion of cytokines such as tumor necrosis factor alpha (TNF-α) and interleukin-1 (IL-1), IL-6 and IL-8 likely play a role in the postulated association. Additionally, H. pylori-induced atrophic gastritis, which develops with aging, reduces the levels of vitamin B12 and folate, which increase homocysteine levels, a known risk factor for insulin resistance [104].

The evidence from epidemiological studies on the association between H. pylori infection and metabolic syndrome has been evolving over the past few years.

A recent large cross-sectional study conducted among 3578 persons aged 18–64 years from Taiwan has demonstrated that H. pylori infected persons (according to urea breath test [UBT]) had a significantly increased prevalence of metabolic syndrome than uninfected persons; 12.4 vs. 7.4% (Pv<0.001) in men and 7.4 vs. 2.5% in women (Pv<0.001) [105]. In this study, metabolic syndrome was defined based on National Cholesterol Education Program (NCEP) Adult Treatment Panel (ATP) III Criteria, which were adjusted to the Taiwanese population [105]. The observed positive associations between H. pylori infection and metabolic syndrome were attenuated in multivariable analyses, while adjusting for confounders such as age, smoking and alcohol drinking; adjusted odds ratio (OR) 1.91 (95% CI: 1.03–3.53) in women, while in men the association was not statistically significant: adjusted OR: 1.38 (95% CI: 0.97–1.95) [105].

A population-based study conducted among adults aged 25 years or over in Iran also reported a 1.5-fold significantly increased prevalence of metabolic syndrome (according to NCEP-ATP-III criteria) among H. pylori (based on serum IgG detection) infected men and women compared with uninfected ones [107]. The same study reported positive associations in relation to exposure to other infectious agents as well such as Chlamydia pneumoniae, Herpes simplex virus 1 (HSV-1) and Cytomegalovirus (CMV) [107]. From this study, it is not clear whether the results were adjusted for confounders, and which ones [107].

Gunji et al. [106], in a well-designed study carried out among 5488 Japanese men (mean age 47± 5 years) and 1906 women (mean age 46±4 years), demonstrated a significant positive relationship between H. pylori seropositivity (according to the presence of IgG antibodies) and metabolic syndrome (based on the Japanese diagnostic criteria); adjusted OR: 1.39 (95% CI: 1.18–1.62) Pv<0.001 [106]. This association was independent of known risk factors for metabolic syndrome namely age, sex, diet and smoking [106].

While there is a growing compelling evidence from large epidemiological studies supporting the existence of a positive association between H. pylori infection and metabolic syndrome, other studies reported no signification association [108] or reported small magnitude association measures [109]. Therefore, the question of whether H. pylori infection is associated with metabolic syndrome, although biologically plausible, remains to be determined, as well as the source of variation among the studies in their findings. Multi-national studies employing similar clinical, epidemiological and diagnostic protocols and methods will be needed to assess true population-to-population variations.

Advertisement

6. Conclusions and future directions

Current evidence is conflicting regarding the question of whether H. pylori may be associated with an increased risk of DM, metabolic syndrome and poor glycemic control. Although an association between H. pylori infection and DM is biologically plausible [110], the nature of such an association is not yet understood. This is due, in part, to important methodological limitations apparent in studies that addressed the relationship between H. pylori infection and DM, including inadequate control for socioeconomic status and for known DM risk factors. Moreover, most studies focused on DM, and less on the reversible conditions of IGT, and IFG. Understanding the role in this association of pathogen-related factors, i.e., virulence antigens such as CagA and VacA is still limited. In addition, it is not clear which biological mechanisms may contribute to the postulated excess risk of DM and/or metabolic syndrome in H. pylori infected persons compared with uninfected ones. Importantly, it is not yet clear whether H. pylori eradication may be beneficial for glycemic control in diabetic patients. Randomized placebo-controlled trials assessing such research questions are lacking.

Addressing these research questions is of great public health and clinical significance given the high prevalence of H. pylori infection and significant burden of DM. If H. pylori infection is truly involved in the etiology of DM, even to a small magnitude (i.e., small relative risks), the public health impact is expected to be great, given the high prevalence of the infection.

References

  1. 1. Muhsen K, Jurban M, Goren S, Cohen D. Incidence, age of acquisition and risk factors of Helicobacter pylori infection among Israeli Arab Infants. J Trop Pediatr 2012; 58(3): 208–13.
  2. 2. Rothenbacher D, Inceoglu J, Bode G, Brenner H. Acquisition of Helicobacter pylori infection in a high-risk population occurs within the first 2 years of life. J Pediatr 2000; 136(6): 744–8.
  3. 3. Torres J, Perez-Perez G, Goodman KJ, et al. A comprehensive review of the natural history of Helicobacter pylori infection in children. Arch Med Res 2000; 31(5): 431–69.
  4. 4. Muhsen K, Athamna A, Athamna M, Spungin-Bialik A, Cohen D. Prevalence and risk factors of Helicobacter pylori infection among healthy 3- to 5-year-old Israeli Arab children. Epidemiol Infect 2006; 134(5): 990–6.
  5. 5. Muhsen K, Athamna A, Bialik A, Alpert G, Cohen D. Presence of Helicobacter pylori in a sibling is associated with a long-term increased risk of H. pylori infection in Israeli Arab children. Helicobacter 2010; 15(2): 108–13.
  6. 6. Weyermann M, Adler G, Brenner H, Rothenbacher D. The mother as source of Helicobacter pylori infection. Epidemiology 2006; 17(3): 332–4.
  7. 7. Suerbaum S, Michetti P. Helicobacter pylori infection. N Engl J Med 2002; 347(15): 1175–86.
  8. 8. Malfertheiner P, Megraud F, O'Morain CA, et al. Management of Helicobacter pylori infection—the Maastricht IV/Florence Consensus Report. Gut 2012; 61(5): 646–64.
  9. 9. Atherton JC. The pathogenesis of Helicobacter pylori-induced gastro-duodenal diseases. Ann Rev Pathol 2006; 1: 63–96.
  10. 10. Atherton JC, Blaser MJ. Coadaptation of Helicobacter pylori and humans: ancient history, modern implications. J Clin Invest 2009; 119(9): 2475–87.
  11. 11. Kuipers EJ, Uyterlinde AM, Pena AS, et al. Long-term sequelae of Helicobacter pylori gastritis. Lancet 1995; 345(8964): 1525–8.
  12. 12. Valle J, Kekki M, Sipponen P, Ihamaki T, Siurala M. Long-term course and consequences of Helicobacter pylori gastritis. Results of a 32-year follow-up study. Scand J Gastroenterol 1996; 31(6): 546–50.
  13. 13. Correa P, Haenszel W, Cuello C, et al. Gastric precancerous process in a high risk population: cohort follow-up. Cancer Res 1990; 50(15): 4737–40.
  14. 14. Ganga-Zandzou PS, Michaud L, Vincent P, et al. Natural outcome of Helicobacter pylori infection in asymptomatic children: a two-year follow-up study. Pediatrics 1999; 104(2 Pt 1): 216–21.
  15. 15. Monack DM, Mueller A, Falkow S. Persistent bacterial infections: the interface of the pathogen and the host immune system. Na Rev Microbiol 2004; 2(9): 747–65.
  16. 16. Nomura AM, Kolonel LN, Miki K, et al. Helicobacter pylori, pepsinogen, and gastric adenocarcinoma in Hawaii. J Infect Dis 2005; 191(12): 2075–81.
  17. 17. Peek RM, Jr., Blaser MJ. Pathophysiology of Helicobacter pylori-induced gastritis and peptic ulcer disease. Am J Med 1997; 102(2): 200–7.
  18. 18. Gao L, Michel A, Weck MN, Arndt V, Pawlita M, Brenner H. Helicobacter pylori infection and gastric cancer risk: evaluation of 15 H. pylori proteins determined by novel multiplex serology. Cancer Res 2009; 69(15): 6164–70.
  19. 19. Gao L, Weck MN, Michel A, Pawlita M, Brenner H. Association between chronic atrophic gastritis and serum antibodies to 15 Helicobacter pylori proteins measured by multiplex serology. Cancer Res 2009; 69(7): 2973–80.
  20. 20. Epplein M, Zheng W, Xiang YB, et al. Prospective study of Helicobacter pylori biomarkers for gastric cancer risk among Chinese men. Cancer Epidem Biomar 2012; 21(12): 2185–92.
  21. 21. Epplein M, Zheng W, Li HL, et al. Diet, Helicobacter pylori strain-specific infection, and gastric cancer risk among Chinese men. Nutr Cancer 2014; 66(4): 550–7.
  22. 22. Franceschi F, Gasbarrini A, Polyzos SA, Kountouras J. Extragastric diseases and Helicobacter pylori. Helicobacter 2015; 20(Suppl 1): 40–6.
  23. 23. Lai CY, Yang TY, Lin CL, Kao CH. Helicobacter pylori infection and the risk of acute coronary syndrome: a nationwide retrospective cohort study. Eur J Clin Microbiol Infect Dis 2015; 34(1): 69–74.
  24. 24. Liu J, Wang F, Shi SL. Helicobacter pylori infection increase the risk of myocardial infarction: a meta-analysis of 26 studies involving more than 20,000 participants. Helicobacter 2015; 20(3): 176–83.
  25. 25. Shmuely H, Wattad M, Solodky A, Yahav J, Samra Z, Zafrir N. Association of Helicobacter pylori with coronary artery disease and myocardial infarction assessed by myocardial perfusion imaging. Isr Med Assoc J 2014; 16(6): 341–6.
  26. 26. Kountouras J, Tsolaki M, Boziki M, et al. Association between Helicobacter pylori infection and mild cognitive impairment. Eur J Neurol 2007; 14(9): 976–82.
  27. 27. Kountouras J, Tsolaki M, Gavalas E, et al. Relationship between Helicobacter pylori infection and Alzheimer disease. Neurology 2006; 66(6): 938–40.
  28. 28. Huang WS, Yang TY, Shen WC, Lin CL, Lin MC, Kao CH. Association between Helicobacter pylori infection and dementia. J Clin Neurosci 2014; 21(8): 1355–8.
  29. 29. Wang F, Liu J, Lv Z. Association of Helicobacter pylori infection with diabetes mellitus and diabetic nephropathy: a meta-analysis of 39 studies involving more than 20,000 participants. Scand J Infect Dis 2013; 45(12): 930–8.
  30. 30. Jeon CY, Haan MN, Cheng C, et al. Helicobacter pylori infection is associated with an increased rate of diabetes. Diabetes Care 2012; 35(3): 520–5.
  31. 31. Cover TL, Blaser MJ. Helicobacter pylori in health and disease. Gastroenterology 2009; 136(6): 1863–73.
  32. 32. Calam J, Gibbons A, Healey ZV, Bliss P, Arebi N. How does Helicobacter pylori cause mucosal damage? Its effect on acid and gastrin physiology. Gastroenterology 1997; 113(6): S43–9.
  33. 33. Sipponen P, Kekki M, Seppala K, Siurala M. The relationships between chronic gastritis and gastric acid secretion. Aliment Pharmacol Ther 1996; 10(Suppl 1): 103–18.
  34. 34. Zhang ZW, Patchett SE, Perrett D, Katelaris PH, Domizio P, Farthing MJG. The relation between gastric vitamin C concentrations, mucosal histology, and CagA seropositivity in the human stomach. Gut 1998; 43(3): 322–6.
  35. 35. Samloff IM. Cellular localization of group I pepsinogens in human gastric mucosa by immunofluorescence. Gastroenterology 1971; 61(2): 185–8.
  36. 36. Samloff IM, Liebman WM. Cellular localization of the group II pepsinogens in human stomach and duodenum by immunofluorescence. Gastroenterology 1973; 65(1): 36–42.
  37. 37. Miki K, Urita Y. Using serum pepsinogens wisely in a clinical practice. J Dig Dis 2007; 8(1): 8–14.
  38. 38. Graham DY, Nurgalieva ZZ, El-Zimaity HM, et al. Noninvasive versus histologic detection of gastric atrophy in a Hispanic population in North America. Clin Gastroenterol Hepatol 2006; 4(3): 306–14.
  39. 39. Song HJ, Jang SJ, Yun SC, et al. Low levels of Pepsinogen I and Pepsinogen I/II ratio are valuable serologic markers for predicting extensive gastric corpus atrophy in patients undergoing endoscopic mucosectomy. Gut Liver 2010; 4(4): 475–80.
  40. 40. He CY, Sun LP, Gong YH, Xu Q, Dong NN, Yuan Y. Serum pepsinogen II: a neglected but useful biomarker to differentiate between diseased and normal stomachs. J Gastroenterol Hepatol 2011; 26(6): 1039–46.
  41. 41. Roper J, Francois F, Shue PL, et al. Leptin and ghrelin in relation to Helicobacter pylori status in adult males. J Clin Endocrinol Metab 2008; 93(6): 2350–7.
  42. 42. Nweneka CV, Prentice AM. Helicobacter pylori infection and circulating ghrelin levels—a systematic review. BMC Gastroenterol 2011; 11: 7.
  43. 43. Breidert M, Miehlke S, Glasow A, et al. Leptin and its receptor in normal human gastric mucosa and in Helicobacter pylori-associated gastritis. Scand J Gastroenterol 1999; 34(10): 954–61.
  44. 44. Isomoto H, Ueno H, Nishi Y, Wen CY, Nakazato M, Kohno S. Impact of Helicobacter pylori infection on ghrelin and various neuroendocrine hormones in plasma. World J Gastroenterol 2005; 11(11): 1644–8.
  45. 45. Francois F, Roper J, Joseph N, et al. The effect of H. pylori eradication on meal-associated changes in plasma ghrelin and leptin. BMC Gastroenterol 2011; 11: 37.
  46. 46. Chuang CH, Sheu BS, Yang HB, et al. Gender difference of circulating ghrelin and leptin concentrations in chronic Helicobacter pylori infection. Helicobacter 2009; 14(1): 54–60.
  47. 47. Azuma T, Suto H, Ito Y, et al. Gastric leptin and Helicobacter pylori infection. Gut 2001; 49(3): 324–9.
  48. 48. Cummings DE, Overduin J. Gastrointestinal regulation of food intake. J Clin Invest 2007; 117(1): 13–23.
  49. 49. Kojima M, Hosoda H, Date Y, Nakazato M, Matsuo H, Kangawa K. Ghrelin is a growth-hormone-releasing acylated peptide from stomach. Nature 1999; 402(6762): 656–60.
  50. 50. Nakazato M, Murakami N, Date Y, et al. A role for ghrelin in the central regulation of feeding. Nature 2001; 409(6817): 194–8.
  51. 51. Bado A, Levasseur S, Attoub S, et al. The stomach is a source of leptin. Nature 1998; 394(6695): 790–3.
  52. 52. Jun DW, Lee OY, Lee YY, Choi HS, Kim TH, Yoon BC. Correlation between gastrointestinal symptoms and gastric leptin and ghrelin expression in patients with gastritis. Digest Dis Sci 2007; 52(10): 2866–72.
  53. 53. Nishi Y, Isomoto H, Uotani S, et al. Enhanced production of leptin in gastric fundic mucosa with Helicobacter pylori infection. World J Gastroenterol 2005; 11(5): 695–9.
  54. 54. Jang EJ, Park SW, Park JS, et al. The influence of the eradication of Helicobacter pylori on gastric ghrelin, appetite, and body mass index in patients with peptic ulcer disease. J Gastroenterol Hepatol 2008; 23(Suppl 2): S278–85.
  55. 55. Powers AC. Harrison’s Principles of Internal Medicine, 18e. In: Longo DL, Fauci AS, Kasper DL, Hauser SL, Jameson JL, Loscalzo J, editors. Chapter 344, Diabetes Mellitus. 18e ed: The McGraw-Hill Companies, Inc.; 2013.
  56. 56. van Dieren S, Beulens JW, van der Schouw YT, Grobbee DE, Neal B. The global burden of diabetes and its complications: an emerging pandemic. Eur J Cardiovasc Prev Rehabil 2010; 17(Suppl 1): S3–8.
  57. 57. Monesi L, Baviera M, Marzona I, et al. Prevalence, incidence and mortality of diagnosed diabetes: evidence from an Italian population-based study. Diabet Med 2012; 29(3): 385–92.
  58. 58. Astrup A. Healthy lifestyles in Europe: prevention of obesity and type II diabetes by diet and physical activity. Public Health Nutr 2001; 4(2B): 499–515.
  59. 59. Joshi SR, Saboo B, Vadivale M, et al. Prevalence of Diagnosed and Undiagnosed Diabetes and Hypertension in India-Results from the Screening India's Twin Epidemic (SITE) Study. Diabetes Technol The 2012; 14(1): 8–15.
  60. 60. Gujral UP, Pradeepa R, Weber MB, Narayan KM, Mohan V. Type 2 diabetes in South Asians: similarities and differences with white Caucasian and other populations. Ann N Y Acad Sci 2013; 1281: 51–63.
  61. 61. Steyn NP, Mann J, Bennett PH, et al. Diet, nutrition and the prevention of type 2 diabetes. Public Health Nutr 2004; 7(1A): 147–65.
  62. 62. Hu FB, Li TY, Colditz GA, Willett WC, Manson JE. Television watching and other sedentary behaviors in relation to risk of obesity and type 2 diabetes mellitus in women. JAMA 2003; 289(14): 1785–91.
  63. 63. Hu FB, Manson JE, Stampfer MJ, et al. Diet, lifestyle, and the risk of type 2 diabetes mellitus in women. N Engl J Med 2001; 345(11): 790–7.
  64. 64. Medalie JH, Papier CM, Goldbourt U, Herman JB. Major factors in the development of diabetes mellitus in 10,000 men. Arch Intern Med 1975; 135(6): 811–7.
  65. 65. Beaty TH, Neel JV, Fajans SS. Identifying risk factors for diabetes in first degree relatives of non-insulin dependent diabetic patients. Am J Epidemiol1982; 115(3): 380–97.
  66. 66. Kadowaki T, Miyake Y, Hagura R, et al. Risk factors for worsening to diabetes in subjects with impaired glucose tolerance. Diabetologia 1984; 26(1): 44–9.
  67. 67. van Dam RM. The epidemiology of lifestyle and risk for type 2 diabetes. Eur J Epidemiol 2003; 18(12): 1115–25.
  68. 68. Espelt A, Arriola L, Borrell C, Larranaga I, Sandin M, Escolar-Pujolar A. Socioeconomic position and type 2 diabetes mellitus in Europe 1999–2009: a panorama of inequalities. Current Diabetes Rev 2011; 7(3): 148–58.
  69. 69. Agardh E, Allebeck P, Hallqvist J, Moradi T, Sidorchuk A. Type 2 diabetes incidence and socio-economic position: a systematic review and meta-analysis. Int J Epidemiol 2011; 40(3): 804–18.
  70. 70. Eriksson KF, Lindgarde F. Prevention of type 2 (non-insulin-dependent) diabetes mellitus by diet and physical exercise. The 6-year Malmo feasibility study. Diabetologia 1991; 34(12): 891–8.
  71. 71. Ryan DH, Diabetes Prevention Program Research G. Diet and exercise in the prevention of diabetes. Int J Clin Pract Supplement 2003; (134): 28–35.
  72. 72. Walker KZ, O'Dea K, Gomez M, Girgis S, Colagiuri R. Diet and exercise in the prevention of diabetes. J Hum Nutr Diet 2010; 23(4): 344–52.
  73. 73. Sukala WR, Page R, Cheema BS. Exercise training in high-risk ethnic populations with type 2 diabetes: a systematic review of clinical trials. Diabetes Res Clin Pr 2012; 97(2): 206–16.
  74. 74. Weigt J, Malfertheiner P. Influence of Helicobacter pylori on gastric regulation of food intake. Curr Opin Clin Nutr Metab Care 2009; 12(5): 522–5.
  75. 75. Isomoto H, Ueno H, Saenko VA, et al. Impact of Helicobacter pylori infection on gastric and plasma ghrelin dynamics in humans. Am J Gastroenterol 2005; 100(8): 1711–20.
  76. 76. Liew PL, Lee WJ, Lee YC, Chen WY. Gastric ghrelin expression associated with Helicobacter pylori infection and chronic gastritis in obese patients. Obes Surg 2006; 16(5): 612–9.
  77. 77. Cummings DE, Purnell JQ, Frayo RS, Schmidova K, Wisse BE, Weigle DS. A preprandial rise in plasma ghrelin levels suggests a role in meal initiation in humans. Diabetes 2001; 50(8): 1714–9.
  78. 78. Shintani M, Ogawa Y, Ebihara K, et al. Ghrelin, an endogenous growth hormone secretagogue, is a novel orexigenic peptide that antagonizes leptin action through the activation of hypothalamic neuropeptide Y/Y1 receptor pathway. Diabetes 2001; 50(2): 227–32.
  79. 79. Wolf G. Leptin: the weight-reducing plasma protein encoded by the obese gene. Nutr Rev 1996; 54(3): 91–3.
  80. 80. Halaas JL, Gajiwala KS, Maffei M, et al. Weight-reducing effects of the plasma protein encoded by the obese gene. Science 1995; 269(5223): 543–6.
  81. 81. Chen Y, Blaser MJ. Association between gastric Helicobacter pylori colonization and glycated hemoglobin levels. J Infect Dis 2012; 205(8): 1195–202.
  82. 82. Polyzos SA, Kountouras J, Zavos C, Deretzi G. The association between Helicobacter pylori infection and insulin resistance: a systematic review. Helicobacter 2011; 16(2): 79–88.
  83. 83. Bener A, Micallef R, Afifi M, Derbala M, Al-Mulla HM, Usmani MA. Association between type 2 diabetes mellitus and Helicobacter pylori infection. Turk J Gastroenterol 2007; 18(4): 225–9.
  84. 84. Zhou X, Zhang C, Wu J, Zhang G. Association between Helicobacter pylori infection and diabetes mellitus: a meta-analysis of observational studies. Diabetes Res Clin Pract 2013; 99(2): 200–8.
  85. 85. Wang F, Liu J, Lv Z. Association of Helicobacter pylori infection with diabetes mellitus and diabetic nephropathy: a meta-analysis of 39 studies involving more than 20,000 participants. Scand J Infect Dis 2013; 45(12): 930–8.
  86. 86. Hsieh MC, Wang SSW, Hsieh YT, Kuo FC, Soon MS, Wu DC. Helicobacter pylori infection associated with high HbA1c and type 2 diabetes. Eur J Clin Invest 2013; 43(9): 949–56.
  87. 87. Dore MP, Bilotta M, Malaty HM, et al. Diabetes mellitus and Helicobacter pylori infection. Nutrition 2000; 16(6): 407–10.
  88. 88. Lutsey PL, Pankow JS, Bertoni AG, Szklo M, Folsom AR. Serological evidence of infections and type 2 diabetes: the MultiEthnic Study of Atherosclerosis. Diabet Med 2009; 26(2): 149–52.
  89. 89. Xia HH, Talley NJ, Kam EP, Young LJ, Hammer J, Horowitz M. Helicobacter pylori infection is not associated with diabetes mellitus, nor with upper gastrointestinal symptoms in diabetes mellitus. Am J Gastroenterol 2001; 96(4): 1039–46.
  90. 90. Demir M, Gokturk HS, Ozturk NA, Kulaksizoglu M, Serin E, Yilmaz U. Helicobacter pylori prevalence in diabetes mellitus patients with dyspeptic symptoms and its relationship to glycemic control and late complications. Dig Dis Sci 2008; 53(10): 2646–9.
  91. 91. Longo-Mbenza B, Nsenga JN, Mokondjimobe E, et al. Helicobacter pylori infection is identified as a cardiovascular risk factor in Central Africans. VascHealth Risk manag 2012; 6: 455–61.
  92. 92. Cenerelli S, Bonazzi P, Galeazzi R, et al. Helicobacter pylori masks differences in homocysteine plasma levels between controls and type 2 diabetic patients. Eur J Clin Invest 2002; 32(3): 158–62.
  93. 93. Horikawa C, Kodama S, Fujihara K, et al. Association of Helicobacter pylori infection with glycemic control in patients with diabetes: a meta-analysis. J Diabetes Res 2014.
  94. 94. Dai YN, Yu WL, Zhu HT, Ding JX, Yu CH, Li YM. Is Helicobacter pylori infection associated with glycemic control in diabetics? World J Gastroenterol 2015; 21(17): 5407–16.
  95. 95. Zojaji H, Ataei E, Sherafat SJ, Ghobakhlou M, Fatemi SR. The effect of the treatment of helicobacter pylori infection on the glycemic control in type 2 diabetes mellitus. Gastroenterol Hepatol 2013; 6(1): 36–40.
  96. 96. Wada Y, Hamamoto Y, Kawasaki Y, et al. The eradication of Helicobacter pylori does not affect glycemic control in Japanese subjects with type 2 diabetes. Jap Clin Med 2013; 4: 41–3.
  97. 97. Vafaeimanesh J, Rajabzadeh R, Ahmadi A, et al. Effect of Helicobacter pylori eradication on glycaemia control in patients with type 2 diabetes mellitus and comparison of two therapeutic regimens. Arab J Gastroenterol 2013; 14(2): 55–8.
  98. 98. Akanuma M, Yanai A, Sakamoto K, et al. Influence of Helicobacter pylori eradication on the management of type 2 diabetes. Hepato-Gastroenterology 2012; 59(114): 641–5.
  99. 99. Candelli M, Rigante D, Marietti G, et al. Helicobacter pylori eradication rate and glycemic control in young patients with type 1 diabetes. J Pediatr Gastroenterol Nutr 2004; 38(4): 422–5.
  100. 100. Begue RE, Gomez R, Compton T, Vargas A. Effect of Helicobacter pylori eradication in the glycemia of children with type 1 diabetes: a preliminary study. South Med J 2002; 95(8): 842–5.
  101. 101. de Luis DA, Cordero JM, Caballero C, et al. Effect of the treatment of Helicobacter pylori infection on gastric emptying and its influence on the glycaemic control in type 1 diabetes mellitus. DiabResClin Pract 2001; 52(1): 1–9.
  102. 102. Grundy SM, Cleeman JI, Daniels SR, et al. Diagnosis and management of the metabolic syndrome: an American Heart Association/National Heart, Lung, and Blood Institute scientific statement. Curr Opin Cardiol 2006; 21(1): 1–6.
  103. 103. Grundy SM, Cleeman JI, Daniels SR, et al. Diagnosis and management of the metabolic syndrome: an American Heart Association/National Heart, Lung, and Blood Institute Scientific Statement. Circulation 2005; 112(17): 2735–52.
  104. 104. Polyzos SA, Kountouras J. Novel advances in the association between helicobacter pylori infection, metabolic syndrome, and related morbidity. Helicobacter 2015; 20(6): 405–9.
  105. 105. Chen TP, Hung HF, Chen MK, et al. Helicobacter pylori infection is positively associated with metabolic syndrome in Taiwanese adults: a cross-sectional study. Helicobacter 2015; 20(3): 184–91.
  106. 106. Gunji T, Matsuhashi N, Sato H, et al. Helicobacter pylori infection is significantly associated with metabolic syndrome in the Japanese population. Am J Gastroenterol 2008; 103(12): 3005–10.
  107. 107. Nabipour I, Vahdat K, Jafari SM, Pazoki R, Sanjdideh Z. The association of metabolic syndrome and Chlamydia pneumoniae, Helicobacter pylori, cytomegalovirus, and herpes simplex virus type 1: The Persian Gulf Healthy Heart Study. Cardiovascr Diabetol 2006; 5.
  108. 108. Naja F, Nasreddine L, Hwalla N, et al. Association of H. pylori infection with insulin resistance and metabolic syndrome among Lebanese adults. Helicobacter 2012; 17(6): 444–51.
  109. 109. Shin DW, Kwon HT, Kang JM, et al. Association between metabolic syndrome and Helicobacter pylori infection diagnosed by histologic status and serological status. J Clin Gastroenterol 2012; 46(10): 840–5.
  110. 110. He C, Yang Z, Lu NH. Helicobacter pylori infection and diabetes: is it a myth or fact? World J Gastroenterol 2014; 20(16): 4607–17.
  111. 111. El-Eshmawy MM, El-Hawary AK, Abdel Gawad SS, El-Baiomy AA. Helicobacter pylori infection might be responsible for the interconnection between type 1 diabetes and autoimmune thyroiditis. Diabetol Metab Syndr 2011; 3(1).
  112. 112. Colombo C TP, Meloni GF, Marinaro AM, Ogana A, Meloni T. Seroprevalence of Helicobacter pylori in children with type 1 diabetes mellitus in Sardinia. Diabetes Nutr Metab 2002; 15: 91–5.

Written By

Saeda Haj, Michal Raviv and Khitam Muhsen

Submitted: 29 May 2015 Reviewed: 21 April 2016 Published: 07 July 2016