07.01.2013 Views

World Journal of Gastrointestinal Pathophysiology

World Journal of Gastrointestinal Pathophysiology

World Journal of Gastrointestinal Pathophysiology

SHOW MORE
SHOW LESS

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

Sherman AE et al . Sonic Hedgehog and gastric cancer<br />

notype found in Drosophila embryos with a mutation in<br />

the gene [5] . Three mammalian homologs have since been<br />

discovered: Shh, Ihh and Dhh. Ihh and Dhh have mostly<br />

tissue-specific expression while Shh has widespread expression<br />

<strong>of</strong> which deficiency leads to neural, limb growth<br />

and foregut defects [6] . Hedgehog is generated as a preprotein<br />

before its signal sequence is cleaved, yielding<br />

an approximately 45 kDa precursor. The precursor is<br />

internally cleaved and a cholesterol moiety is attached to<br />

a glycine residue on the nascent carboxy-terminus [7] . This<br />

covalent attachment <strong>of</strong> cholesterol is a unique modification<br />

among secreted ligands and is a requirement for<br />

tissue specificity and localization <strong>of</strong> the molecule. In vertebrates,<br />

Shh binds and inhibits Patched-1 (Ptc), consequently<br />

activating Smoothened (Smo), which transduces<br />

the Hedgehog signal into the cytoplasm, activating the<br />

Gli family <strong>of</strong> transcription factors [8,9] . Despite this information,<br />

the events leading to Gli activation, especially in<br />

the mammalian stomach, are poorly understood.<br />

Shh is a peptide morphogen produced in gastric<br />

epithelium; however, expression in specific cell types<br />

has been controversial. Recently, a study utilizing a Shh-<br />

LacZ reporter mouse line identified all major cell lineages<br />

<strong>of</strong> the corpus expressing Shh; these included surface<br />

pit, mucous neck, zymogenic and parietal cells [10] . Some<br />

stromal cells express signal transduction components<br />

in addition to Shh; these include the 12-transmembrane<br />

receptor, Ptc, Smo, 7-transmembrane receptor and Gli<br />

transcription factor family. The Hedgehog ligand activates<br />

Smo which leads to transcriptional activation <strong>of</strong> Gli1,<br />

Gli2 becoming an activator and Gli3 no longer acting as a<br />

repressor [11] . Therefore, Hedgehog signaling results in an<br />

increase <strong>of</strong> activator Gli and a decrease in repressor Gli.<br />

These events are crucial in the development and maintenance<br />

<strong>of</strong> multiple organs and the role <strong>of</strong> Shh as a key<br />

morphogen has been well documented [5,12] .<br />

Shh has important roles in the development <strong>of</strong> multiple<br />

organ systems including neuronal and gastrointestinal<br />

systems [13,14] . Shh in the mammalian stomach has been<br />

found to be hormonally regulated by gastrin and requires<br />

an acidic environment for proper processing [15] . The acidactivated<br />

pepsin A protease was found to mediate the<br />

processing <strong>of</strong> the 45kDa precursor molecule to the active<br />

Shh molecule. These findings indicate the importance <strong>of</strong><br />

the acidic gastric environment for maintenance <strong>of</strong> Shh in<br />

the stomach.<br />

Much <strong>of</strong> what we know about Shh’s role in mammals<br />

has come from studies in animal models. Shh-knockout<br />

mice display key features that support the importance <strong>of</strong><br />

proper Shh homeostasis for normal development and<br />

physiological function.<br />

Shh -/- mouse embryos displayed multiple gut abnormalities<br />

including overt gut malrotation and intestinal<br />

transformation <strong>of</strong> the stomach [16] . Additionally, neurons<br />

<strong>of</strong> the enteric nervous system underwent abnormal differentiation<br />

compared to wildtype. While this system provided<br />

evidence for the role <strong>of</strong> Hedgehog in the coordination<br />

<strong>of</strong> developmental genes, these mice die at or shortly<br />

WJGP|www.wjgnet.com<br />

after birth.<br />

In order to study the effects <strong>of</strong> Shh signaling in the<br />

stomach, Xiao et al [17] developed a mouse model expressing<br />

a parietal cell-specific deletion <strong>of</strong> Shh (HKCre/<br />

Shh KO ). This study found evidence <strong>of</strong> hypochlorhydria,<br />

hypergastrinemia and foveolar hyperplasia by 8 mo <strong>of</strong><br />

age. Additionally, a delay in the differentiation <strong>of</strong> zymogen<br />

cells, demonstrated by an increase in immature cells<br />

expressing markers for both mucous neck and zymogen<br />

cells, was documented in the stomach <strong>of</strong> HKCre/Shh KO<br />

mice [17] . The deletion <strong>of</strong> Shh from parietal cells led to<br />

alterations in stomach morphology and gastric cell differentiation;<br />

however, there was no evidence <strong>of</strong> parietal cell<br />

atrophy, a key step in the development <strong>of</strong> stomach cancer<br />

in Helicobacter pylori (H. pylori) infection [18,19] . The absence<br />

<strong>of</strong> parietal cell atrophy suggests that the loss <strong>of</strong> Shh expression<br />

alone is not sufficient to trigger the development<br />

<strong>of</strong> stomach cancer.<br />

H. PYLORI INFECTION AND SONIC<br />

HEDGEHOG<br />

H. pylori infection is globally widespread and a major<br />

cause <strong>of</strong> chronic atrophic gastritis with persistent infection<br />

in 50% <strong>of</strong> the global population [20,21] . While infection<br />

with H. pylori has been directly linked to the development<br />

<strong>of</strong> gastric cancer, the mechanism <strong>of</strong> tumorigenesis is still<br />

unclear. There are many factors that have been considered<br />

in Helicobacter-mediated carcinogenesis; however, the<br />

focus <strong>of</strong> this review will be to discuss its contribution by<br />

modulating Shh expression. H. pylori infection primarily<br />

contributes to gastric cancer development through two<br />

mechanisms: loss <strong>of</strong> Shh by destruction <strong>of</strong> parietal cells<br />

and induction <strong>of</strong> a chronic inflammatory gastric environment.<br />

H. pylori and the inflammatory response<br />

The ability <strong>of</strong> H. pylori to evade the primarily Th1-mediated<br />

host immune response contributes to the development<br />

<strong>of</strong> chronically elevated interleukin-1β (IL-1β) and<br />

other inflammatory cytokines in the stomach [10,22] . A study<br />

<strong>of</strong> H. pylori associated with chronic atrophic gastritis<br />

found upregulation <strong>of</strong> a gene encoding cysteine-rich protein<br />

A, which induces Th1 cytokines interferon (IFN)-γ<br />

and IL-12 [23] . Additionally, H. pylori-associated atrophic<br />

gastritis has been found to be more frequent in patients<br />

with pro-inflammatory polymorphisms <strong>of</strong> genes for IL-<br />

1β and tumor necrosis factor (TNF)-α [24,25] . Specifically, it<br />

has been shown that elevated IL-1β leads to the suppression<br />

<strong>of</strong> Shh, which is necessary for the growth and differentiation<br />

<strong>of</strong> the gastric mucosa during cell restitution<br />

during H. pylori infection [10,26] .<br />

Loss <strong>of</strong> Shh during chronic inflammation<br />

The loss <strong>of</strong> parietal cells through Helicobacter infection and<br />

subsequent autoimmunity has been well documented [22,27] .<br />

Molecular mimicry between the H + ,K + -ATPase local-<br />

104 December 15, 2011|Volume 2|Issue 6|

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!