Editorial Open Access
Copyright ©2011 Baishideng Publishing Group Co., Limited. All rights reserved.
World J Gastroenterol. Jul 7, 2011; 17(25): 2981-2986
Published online Jul 7, 2011. doi: 10.3748/wjg.v17.i25.2981
Effect of ageing on colonic mucosal regeneration
Ferenc Sipos, Katalin Leiszter, 2nd Department of Internal Medicine, Semmelweis University, 1088 Budapest, Hungary
Zsolt Tulassay, Molecular Medicine Research Unit, Hungarian Academy of Sciences, 1088 Budapest, Hungary
Author contributions: Sipos F and Leiszter K contributed to the writing and revising of this paper; Tulassay Z revised and edited the manuscript.
Correspondence to: Ferenc Sipos, MD, PhD, Cell Analysis Laboratory, 2nd Department of Internal Medicine, Semmelweis University, 1088 Budapest, Hungary. dr.siposf@gmail.com
Telephone: +36-20-4780752 Fax: +36-1-2660816
Received: February 18, 2011
Revised: April 1, 2011
Accepted: April 8, 2011
Published online: July 7, 2011

Abstract

The physiologic and pathologic cellular and molecular changes occurring with age in the human colon affect both the inflammatory process leading to mucosal injury and the regenerative capacity of the epithelium. On the one hand, age-related telomere shortening and inflamm-ageing may lead to the development of colonic inflammation, which results in epithelial damage. On the other hand, the altered migration and function of regenerative stem cells, the age-related methylation of mucosal healing-associated genes, together with the alterations of growth factor signaling with age, may be involved in delayed mucosal regeneration. The connections of these alterations to the process of ageing are not fully known. The understanding and custom-tailored modification of these mechanisms are of great clinical importance with regard to disease prevention and modern therapeutic strategies. Here, we aim to summarize the age-related microscopic and molecular changes of the human colon, as well as their role in altered mucosal healing.

Key Words: Colon, Mucosal repair, Ageing, Colorectal cancer, Telomere shortening, DNA methylation



INTRODUCTION

With the possible exception of stem cells and tumor cells, ageing is a nearly universal process that, through functional decline, leads to cell death and, eventually, death of the organism. The age-related molecular changes taking place in the human colon have not been exactly determined; moreover, the results of animal tests and human studies are sometimes contradictory.

Researchers have identified a variety of changes in colonic and rectal function associated with ageing. The total number of colonic myenteric neurons decreases with age in rats and in children, particularly during the first 4 years of life[1,2]. While noting an increase in the surface area of myenteric ganglia with age, Hanani et al[3] found that the proportion of ganglia with cavities and other structural abnormalities increases with age. Furthermore, a positive association between age and collagen content within myenteric ganglia has also been identified[2]. These changes in colonic innervation may have an impact on colonic motility[4-9].

As far as colonic epithelium is concerned, its renewal takes 4-5 d in humans[10]. The regulated balance of epithelial proliferation and apoptosis allows normal epithelial regeneration. Any deviation in epithelial cell kinetics may result in a loss of not only structural but also functional integrity. The imbalance of colonic epithelial renewal may lead to either ulcer or carcinoma development of the colonic mucosa.

The effect of ageing on colonic epithelial regeneration is not fully understood. In rat colon, crypt epithelial proliferation and apoptosis were found to be the most active in the 3rd week of life[11], which was thought to be in connection with the development of the gastrointestinal tract. The results of Xiao et al[12] are, however, contradictory; the number of proliferative epithelial cells was higher, while the rate of apoptosis was lower in older rats. The epithelial expression of the anti-apoptotic Bcl protein and of the pro-apoptotic Bak protein was also in accord with ageing: the former was high, while the latter was low in older rats. This phenomenon may explain the survival of genetically defective cells, hence the increasing incidence of colorectal cancer in the elderly. The age-related rise in cell proliferation is thought to be in part the result of enhanced transition from G1 to S phase as well as stimulated progression through the S phase of the cell cycle[13,14]. Inconsistencies in results may be due to different sampling locations and different ages of the animals, together with the effects of errors in sample proceedings, immunohistochemical methods and data evaluation[15].

Inflammatory bowel diseases (IBD), including ulcerative colitis (UC) and Crohn’s disease, are one of the major causes of epithelial destruction in the colon. It was recently demonstrated that the perceived differences seen in clinical practice between adults and children with UC are largely due to a decrease in histologic features of colitis in children less than 10 years of age[16]. As children approach adulthood, the degree of inflammation and microscopic architectural distortion seen becomes similar to that found in adults. Interestingly, in rectal biopsies of UC, no differences were found amongst all age groups.

Although the understanding of age-related physiological and pathological processes is of great clinical importance, data about the effect of ageing on the induction of mucosal inflammation and the age-related alterations of mucosal healing are scarce in scientific literature.

THE EFFECT OF AGEING ON INFLAMMATION INDUCTION
Telomere shortening and telomerase activity

Telomeres protect the ends of the chromosomes from end-to-end fusions, degradation and recombination. Telomere length decreases with age in most human tissues, including colon[17], and it has been hypothesized that short telomeres might partially explain the connection between cancer and ageing[18]. Telomeres shorten approximately 100 base pairs in each cell division because of their incomplete replication[19] and also as a consequence of oxidative damage[20]. When telomeres become critically short, and in the absence of efficient DNA repair mechanisms, anaphase bridges are formed during mitosis as a result of end-to-end chromosome fusions, which initiates breakage-fusion-bridge cycles. These cycles facilitate the accumulation of genetic changes and chromosomal instability; hence cells need to acquire mechanisms of telomere maintenance, which usually consist of telomerase reactivation. Telomerase prevents further accumulation of chromosomal instability and confers unlimited replicative potential to the cell[21]. It has been previously demonstrated that in UC, colorectal cancer progression is associated with shorter colonocyte telomeres, chromosomal instability and anaphase bridges[22]. It has also been observed that age-related telomere shortening is accelerated in UC[23]. It seems that there is a minimal viable length of colonocyte telomeres, consistent with data from both human cell lines[24] and a telomerase-deficient mouse model[25]. After reaching this critical length, colonocytes defective for DNA damage checkpoints can continue to proliferate but with increased chromosomal instability. Together with alterations in p53 and p16, which are frequent in the non-neoplastic epithelium of UC[10,26,27], this could be a common pathway of tumor progression in this chronic inflammatory disease.

Interestingly, decreased telomerase activity was observed not just in the non-neoplastic epithelium of severely active UC, but also in the non-affected normal mucosa[28,29]. This result suggests that telomerase deficiency may contribute to the pathogenesis of the disease. Moreover, the elevated epithelial telomerase expression found in mildly active UC[30] may help survival and immortalization of the genetically defective epithelial cells in long-standing, chronic inflammation, and thus create the basis for subsequent pathological cell proliferation.

Alterations of immune response

Ageing is associated with a progressive dysregulation of immune response. During ageing, adaptive immunity significantly declines, a phenomenon called immunosenescence, whereas innate immunity seems to be activated, which induces a characteristic pro-inflammatory profile. The latter is called inflamm-ageing[31,32].

Recently, a new subset of CD4+ T cells has been identified. The Th17 cells are distinct from the Th1 and Th2 cells, and secrete interleukin 17 (IL-17) and IL-22[33,34]. IL-17 has been shown to be a primary mediator in several autoimmune and inflammatory diseases, including IBD[35]. Ouyang et al[36] demonstrated that the induction of Th17 cytokines is significantly elevated in both aged humans and mice. In addition, they found that memory T cells are an important cell type for the induction of IL-17, and that the transfer of CD4+CD45Rbhi cells from aged mice induced more severe colitis in recombination-activating gene 1-deficient mice compared to cells from young mice. Their results suggest that ageing promotes an intrinsic predisposition towards the pathological Th17 immune response, which may explain the second peak (between 50-80 years of age) in the incidence of IBD which occurs in humans.

Ageing also results in alterations in the function of Toll-like receptors (TLRs), which have an important role in the pathogenesis of IBD[37]. Recent studies have begun to elucidate the consequences of ageing on TLR function in human cohorts and add to existing findings established in animal models. In general, these studies show that human TLR function is impaired in the context of ageing, and in addition there is evidence for inappropriate persistence of TLR activation in specific systems[38-41].

It has been shown in a mouse model that ageing and TLR2 deficiency have significant effects on the levels of pro-inflammatory cytokines, such as IL-10 and IFN-γ, which could potentially provide a microenvironment that favors the development of more severe colitis following epithelial damage[42]. It has also been reported that the level of trefoil factor 3 (TFF3), an important colonic protective and repair factor, decreases over time in mice, and is negatively regulated by TLR2 signaling[42]. Cytokines such as TNF and IL-1β negatively regulate TFF3 expression in an epithelial cell line by activation of NF-κB, which has been demonstrated to negatively regulate the transcription of TFF3[43,44]. These interactions among ageing-associated changes, TLR deficiency and TFF3 regulation may be of particular relevance in understanding the development of chronic intestinal inflammation and mucosal injury in the elderly.

Several TLR polymorphisms are also involved in the course of IBD[45-47], although it seems likely that large-scale population studies will be needed to clarify the role of key TLR polymorphisms in ageing-related alterations of IBD pathogenesis.

THE EFFECT OF AGEING ON COLONIC MUCOSAL HEALING
Stem cells

The luminal border of the colonic wall is lined by an epithelial monolayer which has several functions, such as water and electrolyte absorption, and it is also a barrier against luminal pathogens[48,49]. Due to the high turnover of shedding epithelial cells, their continuous replacement from the local stem cell pool is required even in healthy colon. Stem cells are located at the basal part of crypts; their progeny migrate towards the luminal surface and undergo terminal differentiation to secretory (Paneth, enteroendocrine and goblet cells) and absorptive (epithelial) cells[48-50]. In the case of tissue injury, such as in IBD or graft-versus-host disease (GVHD), the capacity of intestinal stem cells is not sufficient for the perfect tissue repair, hence the homing of bone marrow-derived multipotent cells is also essential for mucosal regeneration[48,51,52]. It is well known that circulating hematopoietic stem cells (HSCs) play an important role not just in hematopoietic homeostasis, but in the regeneration of solid-organ tissue, which has been certified by several studies[53]. Stem cells are long-lived cells; therefore, they can sustain several genetic and epigenetic changes during cellular senescence. There is evidence both for and against stem cell ageing, and publications are not in agreement with regard to quality and quantity alterations of stem cells in the course of ageing[54].

HSCs in older mice have decreased per-cell repopulating activity, self-renewal and homing abilities, myeloid skewing of differentiation, and increased apoptosis related to stress[55]. It was recently reported that the cyclin-dependent kinase inhibitor p16INK4a, the level of which was previously noted to increase in other cell types with age, accumulates and modulates specific age-associated HSC functions[56]. Notably, in the absence of p16INK4a, HSC repopulating defects and apoptosis were mitigated, improving the stress tolerance of cells and the survival of animals in successive transplants, in a stem-cell-autonomous tissue regeneration model. As p16INK4a is involved in colorectal carcinogenesis[57], it may be supposed to have a specific role in the regulation of the behavior of migrating stem cells of the human colon as well.

Stem cells are in close connection with their niche by means of mechanical and/or chemical processes. Based on the results of bone marrow transplantation studies, one can assume that stem cell function and life span depend on the recipient’s age, since increased post-transplant autoimmunity has been observed in the case of older recipients[54]. The age-related decrease of regenerative stem cell capacity, however, needs to be further studied.

The balance of cell proliferation and death ensures adequate epithelial regeneration. Although colonic epithelial stem cells can be distinguished from other epithelial cells only by morphology, several cellular markers may help the identification of both normal and cancer stem cells of the intestinal tract[58-60].

The effect of ageing on colonic epithelial regeneration and crypt-base stem cell function has come to the frontline of discovery recently. In a mouse model, a few apoptotic cells were seen around the stem cell position and this frequency did not alter with age. However, the apoptotic index within crypts was nearly twice as high in older mice after low dose gamma-irradiation and the number of surviving crypts decreased significantly faster after increasing the dose of irradiation; moreover, in the post-irradiation period the crypt regeneration was much slower in older animals[61]. It was shown that clonogenic cells are more radiosensitive in old mice, and that the growth of surviving crypts after injury was delayed in old mice even though the number of resident clonogenic cells were higher in older colonic crypts[62].

Methylation

In non-cancerous colonic mucosa, repeated injuries are likely to induce adaptive methylation changes that enable efficient wound healing and act against cancer development. The methylation-variable sites that are located in promoter or noncoding neutral regions have demonstrated gradual methylation changes associated with the ageing or long-term adaptation process[63]. The presence of transitional-CpG sites between the unmethylated promoters and nearby densely methylated retroelements has been proposed, in order to describe the complexity of variable methylation in gene control regions[64,65]. The transitional-CpG sites at the margin of the CpG islands and at the non-island CpG sites around the transcription start sites have been found to be either under- or over-methylated in a tissue-specific manner as well as to be methylated to various degrees in the same tissue type[65]. This result suggests that the methylation-variable sites nearest to the transcription start sites may serve as epigenetic markers for adaptive DNA methylation. The methylation-variable site of a strongly expressed tissue-specific gene can influence the expression of the nearby gene as well as its related genes. Therefore, the methylation-variable sites of the key colon-specific genes are expected to participate in both the discrete mucosal adaptation and the interactive changes of methylation patterns that lead to mucosal alterations. In the stomach, recent evidence suggests that mucosal injury induces adaptive changes in DNA methylation[66]. The ulcer-healing genes, such as trefoil factor 1 and 2, (TFF1, -2), cadherin 1 (CDH1), and peroxisome proliferator-activated receptor gamma (PPARG), were found to be concurrently methylated with other genes depending on the presence or absence of CpG islands in the normal mucosa of healthy individuals, while both the TFF2 and PPARG genes were frequently undermethylated in gastric ulcer patients.

Age-related methylation loci, such as estrogen receptor 1 (ESR1) and myogenic differentiation 1 (MYOD1), have also been highlighted[67-69]. These loci showed age-dependent methylation in normal colon mucosa, and this type of methylation is considered to serve as a functional link between ageing and cancer, possibly by deregulating the growth and differentiation of normal colonic epithelial cells and predisposing them to tumorous transformation. In addition, methylation of ESR1 in colon epithelium occurs more frequently in patients with UC who have neoplasia than in UC patients without neoplasia[70].

Differences in methylation levels of age-related methylation loci between the proximal and distal colon have also been described[71]. This phenomenon may have an impact not just in colorectal carcinogenesis, but in the understanding of the pathogenesis of UC.

Growth factor receptors

Gastrointestinal mucosal cell proliferation is known to be under the regulation of a number of nutritional and hormonal factors. It was reported that an age-related rise in gastric and colonic mucosal cell proliferation is accompanied by a marked rise in expression and activation of several tyrosine kinases, including epithelial growth factor receptor (EGFR)[72-74]. The age-related rise in EGFR activation in the mucosa of the gastrointestinal tract is not fully understood, but some recent data suggest this could be partly the result of loss of EGFR-related peptide (ERRP), a “negative regulator”[75]. It was recently reported that in Fischer-344 rats, ageing is associated with increased activation of EGFR in the colonic mucosa, as evidenced by a 30%-35% increase in the levels of tyrosine phosphorylated EGFR in the proximal and distal colon of aged (20-22 mo old) compared to young (4-6 mo old) rats. In contrast, the levels of ERRP in both regions of the colon of aged rats were decreased by 50%-60%, compared to their younger counterparts[76]. Expression of ERRP was also found to be high in benign human colon, stomach and pancreas, but low in the respective invasive adenocarcinomas[77,78].

Age-related decrease of EGFR signaling in cells of ectodermal origin has additionally been described[79]. This may cause delayed mucosal healing in the case of older individuals.

Hepatocyte-derived growth factor (HGF) is mainly produced by mesenchymal cells and acts on cells of epithelial origin which express the HGF receptor C-met (HGFR). The HGF-HGFR system is important in gut homeostasis, and has a crucial role in gastrointestinal wound healing[30]. Furthermore, this system has morphogenetic effects, and it also regulates the formation of epithelial tubular and gland structures[30,80]. It was demonstrated that the production of HGF by fibroblasts increased sharply after about 70% completion of their lifespan in culture, which is regulated at the transcriptional level[80]. The expression of HGFR may decrease with ageing as well[81], which may have consequences on tissue repair.

Revealing age-related alterations in the expression of growth factor receptors involved in colonic mucosal repair, and the better understanding of alterations in receptor signaling, may result in new therapeutic targets of colonic mucosal damage.

CONCLUSION

There are numerous signs of ageing in the human gastrointestinal tract, including the colon (Figure 1). Beyond macro- and microscopic alterations, some of these can be detected at the genetic, gene expression and/or epigenetic level. The connection between ageing and colonic mucosal regeneration has been reported by several studies, and their results may provide an insight into physiologic and pathologic mucosal healing in the context of ageing. An understanding of the influence of these age-related mechanisms may help to develop new therapeutic strategies for chronic inflammatory bowel diseases accompanied by mucosal damage.

Figure 1
Figure 1 The effect of ageing on factors resulting in colonic mucosal damage.
Footnotes

Peer reviewer: Amedeo Columbano, Professor, Dipartimento di Tossicologia, Sezione di Oncologia e Patologia Molecolare, Via Porcell 4, 09124 Cagliari, Italy

S- Editor Tian L L- Editor Logan S E- Editor Ma WH

References
1.  Wester T, O'Briain DS, Puri P. Notable postnatal alterations in the myenteric plexus of normal human bowel. Gut. 1999;44:666-674.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Gomes OA, de Souza RR, Liberti EA. A preliminary investigation of the effects of aging on the nerve cell number in the myenteric ganglia of the human colon. Gerontology. 1997;43:210-217.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Hanani M, Fellig Y, Udassin R, Freund HR. Age-related changes in the morphology of the myenteric plexus of the human colon. Auton Neurosci. 2004;113:71-78.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Di Lorenzo C, Flores AF, Hyman PE. Age-related changes in colon motility. J Pediatr. 1995;127:593-596.  [PubMed]  [DOI]  [Cited in This Article: ]
5.  Gabella G. Development and ageing of intestinal musculature and nerves: the guinea-pig taenia coli. J Neurocytol. 2001;30:733-766.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  McDougal JN, Miller MS, Burks TF, Kreulen DL. Age-related changes in colonic function in rats. Am J Physiol. 1984;247:G542-G546.  [PubMed]  [DOI]  [Cited in This Article: ]
7.  Graff J, Brinch K, Madsen JL. Gastrointestinal mean transit times in young and middle-aged healthy subjects. Clin Physiol. 2001;21:253-259.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Loening-Baucke V, Anuras S. Sigmoidal and rectal motility in healthy elderly. J Am Geriatr Soc. 1984;32:887-891.  [PubMed]  [DOI]  [Cited in This Article: ]
9.  Meier JM, Alavi A, Iruvuri S, Alzeair S, Parker R, Houseni M, Hernandez-Pampaloni M, Mong A, Torigian DA. Assessment of age-related changes in abdominal organ structure and function with computed tomography and positron emission tomography. Semin Nucl Med. 2007;37:154-172.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Sipos F, Molnár B, Zágoni T, Berczi L, Tulassay Z. Growth in epithelial cell proliferation and apoptosis correlates specifically to the inflammation activity of inflammatory bowel diseases: ulcerative colitis shows specific p53- and EGFR expression alterations. Dis Colon Rectum. 2005;48:775-786.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Mandir N, FitzGerald AJ, Goodlad RA. Differences in the effects of age on intestinal proliferation, crypt fission and apoptosis on the small intestine and the colon of the rat. Int J Exp Pathol. 2005;86:125-130.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Xiao ZQ, Moragoda L, Jaszewski R, Hatfield JA, Fligiel SE, Majumdar AP. Aging is associated with increased proliferation and decreased apoptosis in the colonic mucosa. Mech Ageing Dev. 2001;122:1849-1864.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Xiao ZQ, Yu Y, Khan A, Jaszewski R, Ehrinpreis MN, Majumdar AP. Induction of G(1) checkpoint in the gastric mucosa of aged rats. Am J Physiol. 1999;277:G929-G934.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Xiao ZQ, Jaszewski R, Majumdar AP. Aging enhances G(1) phase in the colonic mucosa of rats. Mech Ageing Dev. 2000;116:1-14.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Lee HM, Greeley GH Jr, Englander EW. Effects of aging on expression of genes involved in regulation of proliferation and apoptosis in the colonic epithelium. Mech Ageing Dev. 2000;115:139-155.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Robert ME, Tang L, Hao LM, Reyes-Mugica M. Patterns of inflammation in mucosal biopsies of ulcerative colitis: perceived differences in pediatric populations are limited to children younger than 10 years. Am J Surg Pathol. 2004;28:183-189.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  O'Sullivan J, Risques RA, Mandelson MT, Chen L, Brentnall TA, Bronner MP, Macmillan MP, Feng Z, Siebert JR, Potter JD. Telomere length in the colon declines with age: a relation to colorectal cancer? Cancer Epidemiol Biomarkers Prev. 2006;15:573-577.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Blasco MA. Telomeres and human disease: ageing, cancer and beyond. Nat Rev Genet. 2005;6:611-622.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Levy MZ, Allsopp RC, Futcher AB, Greider CW, Harley CB. Telomere end-replication problem and cell aging. J Mol Biol. 1992;225:951-960.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  von Zglinicki T. Oxidative stress shortens telomeres. Trends Biochem Sci. 2002;27:339-344.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Maser RS, DePinho RA. Connecting chromosomes, crisis, and cancer. Science. 2002;297:565-569.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  O'Sullivan JN, Bronner MP, Brentnall TA, Finley JC, Shen WT, Emerson S, Emond MJ, Gollahon KA, Moskovitz AH, Crispin DA. Chromosomal instability in ulcerative colitis is related to telomere shortening. Nat Genet. 2002;32:280-284.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  Risques RA, Lai LA, Brentnall TA, Li L, Feng Z, Gallaher J, Mandelson MT, Potter JD, Bronner MP, Rabinovitch PS. Ulcerative colitis is a disease of accelerated colon aging: evidence from telomere attrition and DNA damage. Gastroenterology. 2008;135:410-418.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Zou Y, Sfeir A, Gryaznov SM, Shay JW, Wright WE. Does a sentinel or a subset of short telomeres determine replicative senescence? Mol Biol Cell. 2004;15:3709-3718.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  Blasco MA, Lee HW, Hande MP, Samper E, Lansdorp PM, DePinho RA, Greider CW. Telomere shortening and tumor formation by mouse cells lacking telomerase RNA. Cell. 1997;91:25-34.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  Brentnall TA, Crispin DA, Rabinovitch PS, Haggitt RC, Rubin CE, Stevens AC, Burmer GC. Mutations in the p53 gene: an early marker of neoplastic progression in ulcerative colitis. Gastroenterology. 1994;107:369-378.  [PubMed]  [DOI]  [Cited in This Article: ]
27.  Furth EE, Gustafson KS, Dai CY, Gibson SL, Menard-Katcher P, Chen T, Koh J, Enders GH. Induction of the tumor-suppressor p16(INK4a) within regenerative epithelial crypts in ulcerative colitis. Neoplasia. 2006;8:429-436.  [PubMed]  [DOI]  [Cited in This Article: ]
28.  Usselmann B, Newbold M, Morris AG, Nwokolo CU. Deficiency of colonic telomerase in ulcerative colitis. Am J Gastroenterol. 2001;96:1106-1112.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  Kleideiter E, Friedrich U, Möhring A, Walker S, Höring E, Maier K, Fritz P, Thon KP, Klotz U. Telomerase activity in chronic inflammatory bowel disease. Dig Dis Sci. 2003;48:2328-2332.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Sipos F, Galamb O, Herszényi L, Molnár B, Solymosi N, Zágoni T, Berczi L, Tulassay Z. Elevated insulin-like growth factor 1 receptor, hepatocyte growth factor receptor and telomerase protein expression in mild ulcerative colitis. Scand J Gastroenterol. 2008;43:289-298.  [PubMed]  [DOI]  [Cited in This Article: ]
31.  Salminen A, Huuskonen J, Ojala J, Kauppinen A, Kaarniranta K, Suuronen T. Activation of innate immunity system during aging: NF-kB signaling is the molecular culprit of inflamm-aging. Ageing Res Rev. 2008;7:83-105.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Desai A, Grolleau-Julius A, Yung R. Leukocyte function in the aging immune system. J Leukoc Biol. 2010;87:1001-1009.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Bettelli E, Korn T, Kuchroo VK. Th17: the third member of the effector T cell trilogy. Curr Opin Immunol. 2007;19:652-657.  [PubMed]  [DOI]  [Cited in This Article: ]
34.  Weaver CT, Harrington LE, Mangan PR, Gavrieli M, Murphy KM. Th17: an effector CD4 T cell lineage with regulatory T cell ties. Immunity. 2006;24:677-688.  [PubMed]  [DOI]  [Cited in This Article: ]
35.  Korn T, Bettelli E, Oukka M, Kuchroo VK. IL-17 and Th17 Cells. Annu Rev Immunol. 2009;27:485-517.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  Ouyang X, Yang Z, Zhang R, Arnaboldi P, Lu G, Li Q, Wang W, Zhang B, Cui M, Zhang H. Potentiation of Th17 cytokines in aging process contributes to the development of colitis. Cell Immunol. 2011;266:208-217.  [PubMed]  [DOI]  [Cited in This Article: ]
37.  Cario E. Toll-like receptors in inflammatory bowel diseases: a decade later. Inflamm Bowel Dis. 2010;16:1583-1597.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  Shaw AC, Panda A, Joshi SR, Qian F, Allore HG, Montgomery RR. Dysregulation of human Toll-like receptor function in aging. Ageing Res Rev. 2010;Epub ahead of print.  [PubMed]  [DOI]  [Cited in This Article: ]
39.  Biswas A, Wilmanski J, Forsman H, Hrncir T, Hao L, Tlaskalova-Hogenova H, Kobayashi KS. Negative regulation of Toll-like receptor signaling plays an essential role in homeostasis of the intestine. Eur J Immunol. 2011;41:182-194.  [PubMed]  [DOI]  [Cited in This Article: ]
40.  van Duin D, Mohanty S, Thomas V, Ginter S, Montgomery RR, Fikrig E, Allore HG, Medzhitov R, Shaw AC. Age-associated defect in human TLR-1/2 function. J Immunol. 2007;178:970-975.  [PubMed]  [DOI]  [Cited in This Article: ]
41.  Kovacs EJ, Palmer JL, Fortin CF, Fülöp T Jr, Goldstein DR, Linton PJ. Aging and innate immunity in the mouse: impact of intrinsic and extrinsic factors. Trends Immunol. 2009;30:319-324.  [PubMed]  [DOI]  [Cited in This Article: ]
42.  Albert EJ, Marshall JS. Aging in the absence of TLR2 is associated with reduced IFN-gamma responses in the large intestine and increased severity of induced colitis. J Leukoc Biol. 2008;83:833-842.  [PubMed]  [DOI]  [Cited in This Article: ]
43.  Loncar MB, Al-azzeh ED, Sommer PS, Marinovic M, Schmehl K, Kruschewski M, Blin N, Stohwasser R, Gött P, Kayademir T. Tumour necrosis factor alpha and nuclear factor kappaB inhibit transcription of human TFF3 encoding a gastrointestinal healing peptide. Gut. 2003;52:1297-1303.  [PubMed]  [DOI]  [Cited in This Article: ]
44.  Dossinger V, Kayademir T, Blin N, Gött P. Down-regulation of TFF expression in gastrointestinal cell lines by cytokines and nuclear factors. Cell Physiol Biochem. 2002;12:197-206.  [PubMed]  [DOI]  [Cited in This Article: ]
45.  Pierik M, Joossens S, Van Steen K, Van Schuerbeek N, Vlietinck R, Rutgeerts P, Vermeire S. Toll-like receptor-1, -2, and -6 polymorphisms influence disease extension in inflammatory bowel diseases. Inflamm Bowel Dis. 2006;12:1-8.  [PubMed]  [DOI]  [Cited in This Article: ]
46.  Kathrani A, House A, Catchpole B, Murphy A, German A, Werling D, Allenspach K. Polymorphisms in the TLR4 and TLR5 gene are significantly associated with inflammatory bowel disease in German shepherd dogs. PLoS One. 2010;5:e15740.  [PubMed]  [DOI]  [Cited in This Article: ]
47.  Glas J, Konrad A, Schmechel S, Dambacher J, Seiderer J, Schroff F, Wetzke M, Roeske D, Török HP, Tonenchi L. The ATG16L1 gene variants rs2241879 and rs2241880 (T300A) are strongly associated with susceptibility to Crohn's disease in the German population. Am J Gastroenterol. 2008;103:682-691.  [PubMed]  [DOI]  [Cited in This Article: ]
48.  Matsumoto T, Okamoto R, Yajima T, Mori T, Okamoto S, Ikeda Y, Mukai M, Yamazaki M, Oshima S, Tsuchiya K. Increase of bone marrow-derived secretory lineage epithelial cells during regeneration in the human intestine. Gastroenterology. 2005;128:1851-1867.  [PubMed]  [DOI]  [Cited in This Article: ]
49.  van der Flier LG, Clevers H. Stem cells, self-renewal, and differentiation in the intestinal epithelium. Annu Rev Physiol. 2009;71:241-260.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Ricci-Vitiani L, Pagliuca A, Palio E, Zeuner A, De Maria R. Colon cancer stem cells. Gut. 2008;57:538-548.  [PubMed]  [DOI]  [Cited in This Article: ]
51.  Barry FP, Murphy JM. Mesenchymal stem cells: clinical applications and biological characterization. Int J Biochem Cell Biol. 2004;36:568-584.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  Okamoto R, Yajima T, Yamazaki M, Kanai T, Mukai M, Okamoto S, Ikeda Y, Hibi T, Inazawa J, Watanabe M. Damaged epithelia regenerated by bone marrow-derived cells in the human gastrointestinal tract. Nat Med. 2002;8:1011-1017.  [PubMed]  [DOI]  [Cited in This Article: ]
53.  Kørbling M, Estrov Z. Adult stem cells for tissue repair - a new therapeutic concept? N Engl J Med. 2003;349:570-582.  [PubMed]  [DOI]  [Cited in This Article: ]
54.  Van Zant G, Liang Y. The role of stem cells in aging. Exp Hematol. 2003;31:659-672.  [PubMed]  [DOI]  [Cited in This Article: ]
55.  Chen J, Astle CM, Harrison DE. Development and aging of primitive hematopoietic stem cells in BALB/cBy mice. Exp Hematol. 1999;27:928-935.  [PubMed]  [DOI]  [Cited in This Article: ]
56.  Janzen V, Forkert R, Fleming HE, Saito Y, Waring MT, Dombkowski DM, Cheng T, DePinho RA, Sharpless NE, Scadden DT. Stem-cell ageing modified by the cyclin-dependent kinase inhibitor p16INK4a. Nature. 2006;443:421-426.  [PubMed]  [DOI]  [Cited in This Article: ]
57.  Psofaki V, Kalogera C, Tzambouras N, Stephanou D, Tsianos E, Seferiadis K, Kolios G. Promoter methylation status of hMLH1, MGMT, and CDKN2A/p16 in colorectal adenomas. World J Gastroenterol. 2010;16:3553-3560.  [PubMed]  [DOI]  [Cited in This Article: ]
58.  Todaro M, Francipane MG, Medema JP, Stassi G. Colon cancer stem cells: promise of targeted therapy. Gastroenterology. 2010;138:2151-2162.  [PubMed]  [DOI]  [Cited in This Article: ]
59.  Papailiou J, Bramis KJ, Gazouli M, Theodoropoulos G. Stem cells in colon cancer. A new era in cancer theory begins. Int J Colorectal Dis. 2011;26:1-11.  [PubMed]  [DOI]  [Cited in This Article: ]
60.  Barker N, Clevers H. Leucine-rich repeat-containing G-protein-coupled receptors as markers of adult stem cells. Gastroenterology. 2010;138:1681-1696.  [PubMed]  [DOI]  [Cited in This Article: ]
61.  Kirkwood TB. Intrinsic ageing of gut epithelial stem cells. Mech Ageing Dev. 2004;125:911-915.  [PubMed]  [DOI]  [Cited in This Article: ]
62.  Martin K, Potten CS, Roberts SA, Kirkwood TB. Altered stem cell regeneration in irradiated intestinal crypts of senescent mice. J Cell Sci. 1998;111:2297-2303.  [PubMed]  [DOI]  [Cited in This Article: ]
63.  Yatabe Y, Tavaré S, Shibata D. Investigating stem cells in human colon by using methylation patterns. Proc Natl Acad Sci USA. 2001;98:10839-10844.  [PubMed]  [DOI]  [Cited in This Article: ]
64.  Turker MS. Gene silencing in mammalian cells and the spread of DNA methylation. Oncogene. 2002;21:5388-5393.  [PubMed]  [DOI]  [Cited in This Article: ]
65.  Kang MI, Rhyu MG, Kim YH, Jung YC, Hong SJ, Cho CS, Kim HS. The length of CpG islands is associated with the distribution of Alu and L1 retroelements. Genomics. 2006;87:580-590.  [PubMed]  [DOI]  [Cited in This Article: ]
66.  Hong SJ, Oh JH, Jung YC, Kim YH, Kim SJ, Kang SJ, Seo EJ, Choi SW, Kang MI, Rhyu MG. DNA methylation patterns of ulcer-healing genes associated with the normal gastric mucosa of gastric cancers. J Korean Med Sci. 2010;25:405-417.  [PubMed]  [DOI]  [Cited in This Article: ]
67.  Ahuja N, Li Q, Mohan AL, Baylin SB, Issa JP. Aging and DNA methylation in colorectal mucosa and cancer. Cancer Res. 1998;58:5489-5494.  [PubMed]  [DOI]  [Cited in This Article: ]
68.  Toyota M, Issa JP. CpG island methylator phenotypes in aging and cancer. Semin Cancer Biol. 1999;9:349-357.  [PubMed]  [DOI]  [Cited in This Article: ]
69.  Kawakami K, Ruszkiewicz A, Bennett G, Moore J, Grieu F, Watanabe G, Iacopetta B. DNA hypermethylation in the normal colonic mucosa of patients with colorectal cancer. Br J Cancer. 2006;94:593-598.  [PubMed]  [DOI]  [Cited in This Article: ]
70.  Tominaga K, Fujii S, Mukawa K, Fujita M, Ichikawa K, Tomita S, Imai Y, Kanke K, Ono Y, Terano A. Prediction of colorectal neoplasia by quantitative methylation analysis of estrogen receptor gene in nonneoplastic epithelium from patients with ulcerative colitis. Clin Cancer Res. 2005;11:8880-8885.  [PubMed]  [DOI]  [Cited in This Article: ]
71.  Horii J, Hiraoka S, Kato J, Harada K, Kuwaki K, Fujita H, Toyooka S, Yamamoto K. Age-related methylation in normal colon mucosa differs between the proximal and distal colon in patients who underwent colonoscopy. Clin Biochem. 2008;41:1440-1448.  [PubMed]  [DOI]  [Cited in This Article: ]
72.  Malecka-Panas E, Relan NK, Majumdar AP. Increased activation of EGF-receptor tyrosine kinase by EGF and TGF-alpha in the colonic mucosa of aged rats. J Gerontol A Biol Sci Med Sci. 1996;51:B60-B65.  [PubMed]  [DOI]  [Cited in This Article: ]
73.  Tureaud J, Sarkar FH, Fligiel SE, Kulkarni S, Jaszewski R, Reddy K, Yu Y, Majumdar AP. Increased expression of EGFR in gastric mucosa of aged rats. Am J Physiol. 1997;273:G389-G398.  [PubMed]  [DOI]  [Cited in This Article: ]
74.  Tran KT, Rusu SD, Satish L, Wells A. Aging-related attenuation of EGF receptor signaling is mediated in part by increased protein tyrosine phosphatase activity. Exp Cell Res. 2003;289:359-367.  [PubMed]  [DOI]  [Cited in This Article: ]
75.  Majumdar AP. Regulation of gastrointestinal mucosal growth during aging. J Physiol Pharmacol. 2003;54 Suppl 4:143-154.  [PubMed]  [DOI]  [Cited in This Article: ]
76.  Schmelz EM, Levi E, Du J, Xu H, Majumdar AP. Age-related loss of EGF-receptor related protein (ERRP) in the aging colon is a potential risk factor for colon cancer. Mech Ageing Dev. 2004;125:917-922.  [PubMed]  [DOI]  [Cited in This Article: ]
77.  Yu Y, Rishi AK, Turner JR, Liu D, Black ED, Moshier JA, Majumdar AP. Cloning of a novel EGFR-related peptide: a putative negative regulator of EGFR. Am J Physiol Cell Physiol. 2001;280:C1083-C1089.  [PubMed]  [DOI]  [Cited in This Article: ]
78.  Marciniak DJ, Moragoda L, Mohammad RM, Yu Y, Nagothu KK, Aboukameel A, Sarkar FH, Adsay VN, Rishi AK, Majumdar AP. Epidermal growth factor receptor-related protein: a potential therapeutic agent for colorectal cancer. Gastroenterology. 2003;124:1337-1347.  [PubMed]  [DOI]  [Cited in This Article: ]
79.  Enwere E, Shingo T, Gregg C, Fujikawa H, Ohta S, Weiss S. Aging results in reduced epidermal growth factor receptor signaling, diminished olfactory neurogenesis, and deficits in fine olfactory discrimination. J Neurosci. 2004;24:8354-8365.  [PubMed]  [DOI]  [Cited in This Article: ]
80.  Miyazaki M, Gohda E, Kaji K, Namba M. Increased hepatocyte growth factor production by aging human fibroblasts mainly due to autocrine stimulation by interleukin-1. Biochem Biophys Res Commun. 1998;246:255-260.  [PubMed]  [DOI]  [Cited in This Article: ]
81.  Barani AE, Durieux AC, Sabido O, Freyssenet D. Age-related changes in the mitotic and metabolic characteristics of muscle-derived cells. J Appl Physiol. 2003;95:2089-2098.  [PubMed]  [DOI]  [Cited in This Article: ]