Skip to main content
  • Research article
  • Open access
  • Published:

Most colitis associated carcinomas lack expression of LGR5: a preliminary study with implications for unique pathways of carcinogenesis compared to sporadic colorectal carcinoma

Abstract

Background

Leucine-rich repeat-containing G-protein-coupled receptor 5 (LGR5), a component of the Wnt receptor complex, is thought to lineage label gastric and intestinal stem cells. LGR5 expression is increased in colorectal carcinoma (CRC) compared to normal tissue. Colitis associated colorectal adenocarcinoma (CAC) often shows distinct morphologic and molecular phenotypes compared to sporadic cases. However, the expression profile of LGR5, and by extension the potential role of an intestinal stem cell phenotype, has not been well described in a series of human CAC.

Method

RNA in situ hybridization (ISH) for LGR5 expression on 30 CACs (12 cases with conventional morphology and 18 cases with non-conventional type morphology) from 29 inflammatory bowel disease (IBD) patients was performed and compared the expression profile to a control group of 10 sporadic CRCs. Immunohistochemistry for beta-catenin and SATB2 was performed on the 30 CACs.

Result

LGR5 was positive in 30% (9/30) of CAC cases and 90% (9/10) of sporadic CRCs (p = 0.002). A large majority (89%) of LGR5 positive CACs were of the conventional histologic type, and conventional type CAC showed a significantly higher LGR5 score (median 3.0; interquartile range 1.75–3.25) than non-conventional type CAC (median 1.5; interquartile range 1.00–2.00) (p = 0.034). CAC with conventional morphology did have a lower level of LGR5 expression than sporadic CRC. Sporadic CRCs showed a significantly higher LGR5 level score than non-conventional type CACs (p < 0.001). Nuclear translocation of beta-catenin was strongly associated with LGR5 expression (p = 0.003), however no significant association was identified between SATB2 expression and LGR5 expression status in CACs.

Conclusion

These findings suggest that the wider spectrum of tumor morphology in CAC may be associated with absence of a LGR5-expressing intestinal stem cell phenotype.

Peer Review reports

Background

Inflammatory bowel disease (IBD) is a chronic relapsing–remitting disorder of the gastrointestinal tract, and for patients with long-standing colonic inflammation, colorectal adenocarcinoma (CRC) is a recognized complication, leading to routine periodic surveillance of patients with colitis for dysplasia and carcinoma. One meta-analysis estimated that the risk of colitis associated carcinoma (CAC) in patients with IBD is 2% by 10 years after initial diagnosis, and increases to 8% at 20 years and 18% at 30 years after colitis onset [1]. However more recent figures are lower than this, likely the results of better treatment reducing the risk [2, 3].

Leucine-rich repeat-containing G-protein-coupled receptor 5 (LGR5), a wingless-type mouse mammary tumor virus integration site family (Wnt) target gene that functions as a receptor for Wnt agonist R-spondins (RSPOs). It was identified as a selective marker of crypt base columnar cells, and these LGR5-positive crypt base columnar cells were demonstrated to be self-renewing, multipotent adult intestinal stem cells [4]. Increased LGR5 expression in human sporadic colorectal adenomas and cancers [5,6,7,8] and in murine models [9, 10] have been well described, however, the LGR5 expression profile has not been described in a series of CAC in human.

Recent studies have highlighted genetic, morphologic, and immunohistochemical differences between CAC and sporadic CRC, and suggested that chronic inflammation may lead to unique genomic changes that increase the risk for CAC [11, 12]. Carcinogenic mutations probably first start accumulating in the long-lived stem cell lineages of the crypt, as the lifespan of non-stem cells is too short for them to acquire the necessary mutations before being shed.

To our knowledge, a comparison of LGR5 expression in CAC and sporadic CRC has not been performed. Here, a series of CAC cases was compared with sporadic CRC for morphologic features and the expression of LGR5.

Methods

Study approval was obtained from the Research Ethics Board at Mount Sinai Hospital.

Thirty cases of surgically resected colitis associated primary colorectal adenocarcinoma from 29 patients between 2011 and 2016 were retrieved from the surgical pathology archives at Sinai Health System. Patients who underwent neoadjuvant therapy were excluded. 10 cases of surgically resected primary CRC from patients without inflammatory bowel disease or any known hereditary cancer syndrome, hence forth termed sporadic CRC were identified as a control group. All H&E sections were reviewed at a multiheaded microscope by four gastrointestinal pathologists (MI, HO, RR, and JC) and each invasive carcinoma was subclassified by consensus into one of five morphologic subtypes: conventional, mucinous, serrated, low grade tubuloglandular (LGTG) and others [13].

One representative paraffin block of tumor was selected in each case for RNA in situ hybridization (ISH). Detection of LGR5 mRNA was performed using the RNAscope® kit (Advanced Cell Diagnostics, Hayward, CA, USA) according to the manufacturer’s instructions using unstained sample tissue slides. Briefly, tissue sections were pretreated by heating and protease application prior to hybridization with an LGR5-specific probe. A detailed procedure has been described in an earlier publication [14].

Formalin fixation -paraffin embedding and preservation of formalin-fixed paraffin-embedded (FFPE) tissues were performed according to College of American pathologists protocol (https://documents.cap.org/documents/practical-guide-specimen-handling.pdf). RNA-ISH was performed immediately after cutting sections from FFPE blocks. Positive staining was indicated by brown punctate dots in the nucleus and/or cytoplasm. The expression level of LGR5 was quantified according to the five-grade scoring system previously described [8]: 0 = no staining or less than one dot per cell; 1 = 1 to 3 dots per cell; 2 = 4 to 10 dots per cell and no or very few dot clusters; 3 = > 10 dots per cell and < 10% positive cells overall; and 4 = > 10 dots per cell and > 10% positive cells with dot clusters. For a binary analysis, LGR5 status was considered positive if the ISH score was > 2. (Fig. 1) [15] Immunohistochemical staining was performed using commercially available antibodies with the immuno-enzyme polymer method (Novolink Polymer Detection Systems for beta-catenin, Leica, Germany or Histofine Simple Stain MAX PO Multi for SATB2, Nichirei Biosciences, Tokyo, Japan) with 3, 3′-diaminobenzidone as the chromogen. The primary antibodies were used in accordance with the manufacturers’ instructions: beta-catenin (clone: EP35, EPITOMICS, Burlingame, California USA) and SATB2 (clone: EPNCIR130A, abcam, Cambridge, UK). Beta-catenin nuclear staining was classified as negative or scattered; no or very few scattered positive cells without any clusters, focal; positive cells clustered in focal areas, diffuse; positive cells distributed diffusely, homogeneously or heterogeneously in accordance with the previous study [16]. The extent of staining for SATB2 was scored semiquantitatively (no staining; < 5%; 5–25%; 26–50%; 51–75%; and 76–100%), and the maximum intensity was graded as negative, weak, moderate, or strong. For binary analyses, cases with 5% or more tumor cells showing moderate or strong intensity were considered positive [13].

Fig. 1
figure 1

LGR5 expression level was quantified according to the five-grade scoring system: score 2; a few dots per cell and no or very few dot; LGR5 negative (a), score 3; > 10 dots per cell and < 10% positive cells overall; LGR5 positive, (b, c), and score 4; > 10 dots per cell and > 10% positive cells; LGR5 positive (d)

Two of the authors (MI and JC) reviewed the RNA-ISH stains, and two of the authors (MI and HO) reviewed the immunohistochemical stains at a multiheaded microscope and reached a consensus score for each case.

Statistics

Chi-square test or Fisher exact tests were used to characterize the relationship between categorical variables. Kruskal-Wallis chi-squared test and Mann–Whitney U-test were used for comparisons between LGR5 expression level scores. Staining scores are non-parametric and are thus expressed as a median score with the interquartile range. Differences were considered to be significant at p < 0.05. All statistical analyses were performed with EZR (Saitama Medical Center, Jichi Medical University, Saitama, Japan), which is a graphical user interface for R (The R Foundation for Statistical Computing, Vienna, Austria).

Results

The 29 patients with CAC included 18 with ulcerative colitis and 11 with Crohn’s disease. One patient with ulcerative colitis had synchronous carcinomas for a total of 30 tumors evaluated. A summary of the clinicopathologic features of the CAC cohort and sporadic controls is presented in Table 1. The median age at time of resection for patients with CAC was 54 years (range 26–76), significantly younger than sporadic cases (median age 67 years; range 40–95) (p = 0.017). CACs had a wider spectrum of tumor morphologies compared to sporadic CRCs. Only twelve of 30 (40%) CACs had conventional morphology compared to 80% of sporadic cases. None of sporadic cases were classified as LGTG. The one CAC case subclassified as “other” showed fetal enteric and hepatoid differentiation.

Table 1 Clinicopathologic features of study group patients

Adjacent non-neoplastic colonic mucosa was examined as an internal control for LGR5 expression. In all cases, there were measurable brown dots in the base of crypts in control areas. LGR5 was positive in 30% (9/30) of CAC cases (median 2.0, interquartile range 1.00–3.00) and 90% (9/10) of sporadic CRCs (median 4.0, interquartile range 3.00–4.00) (p = 0.002). In CACs, the distribution of LGR5 expression was heterogenous in the overlying mucosa and in some cases, overlying or adjacent dysplasia showed higher expression than the invasive component. An abrupt transition to LGR5 expression was identified in the CAC or dysplasia component in some cases. (Fig. 2) A large majority (89%) of LGR5 positive CAC cases were of the conventional histologic type, while morphologies were much more variable in the LGR5 negative CAC cases. (Fig. 2) In conventional type CACs, eight of 12 cases (67%) were LGR5 positive (median 3.0; interquartile range 1.75–3.25) and in contrast, only one of 18 (6%) non-conventional type CACs (one with serrated morphology) was LGR5 positive (median 1.5; interquartile range 1.00–2.00) (p < 0.001). Conventional type CAC showed a significantly higher level of LGR5 expression than non-conventional type CAC (p = 0.034). Conventional type CACs showed a lower level of LGR5 expression than sporadic CRCs, but this did not reach statistical significance (p = 0.221), whereas, expression level of LGR5 in non-conventional type CACs was significantly lower compared to sporadic CRCs (p < 0.001). (Fig. 3).

Fig. 2
figure 2

LGR5 expression in colitis associated adenocarcinoma (CAC). CAC with conventional morphology (a) often showed diffuse and strong LGR5 expression (b and c: high power view; score 4), however a few invasive glands adjacent to the LGR5 positive area were LGR5 negative. No morphological differences between these areas was apparent (d). An LGR5 positive invasive component (e) showed nuclear translocation of beta-catenin (f). A high grade mucinous CAC had adjacent dysplasia with focal adenomatous morphology (g). The mucinous component was negative for LGR5 (h); however, nuclear translocation of beta catenin was identified (i). Adjacent dysplasia with adenomatous morphology showed strong LGR5 expression (l) and nuclear expression of beta catenin (m)

Fig. 3
figure 3

Expression level of LGR5 in control sporadic colorectal adenocarcinoma, conventional type colitis associated adenocarcinoma and non-conventional type colitis associated adenocarcinoma. *P = 0.034, ***P < 0.001

In CACs, LGR5 expression was associated with nuclear beta catenin expression, but not with SATB2 expression; only two of 21 (10%) LGR5 negative CACs showed diffuse nuclear beta-catenin expression, while 6 of 9 (67%) LGR5 positive CACs showed diffuse nuclear-beta catenin expression (p = 0.003). Eleven of 30 CACs (37%) retained SATB2 expression and the expression rate was 33 and 44% in LGR5 positive CACs and LGR5 negative CACs, respectively (p = 0.687).

LGR5 positive CACs were more likely to be located in the rectum (p = 0.032). There was no significant difference in pT stage, pN stage, IBD phenotype, age, or sex between LGR5 positive CACs and negative CACs. (Table 2).

Table 2 Clinicopathologic Features of LGR5 positive CACs and LGR5 negative CACs

Discussion

Here, we evaluated the LGR5 expression profile of 30 CAC cases and 10 sporadic CRC cases and identified that LGR5 was less frequently expressed in CACs than in sporadic CRCs. Further, we found CAC with non-conventional morphology showed significantly lower LGR5 expression levels and nuclear translocation of beta-catenin than the conventional subtype of CAC.

LGR5 has been considered one of the most reliable crypt stem cell markers of CRCs, particularly those that arise through the APC-mutant pathway of tumorigenesis [9]. LGR5 is reportedly overexpressed in human colorectal adenomas and cancers by immunohistochemistry [6, 17, 18] and ISH [15], and Martin et al. showed that the mRNA levels of LGR5 expressed in human CRCs is 10-fold expanded than normal intestinal crypt [19]. Jang et al. recently showed that LGR5 mRNA positivity is observed in 68% of 788 human CRCs and suggested positive correlation with the chromosomal-instability pathway (characterized by left-sided location and nuclear β-catenin expression; representing an abnormal Wnt signal activation) and negative correlation with MSI, CIMP-high, and BRAF mutations [15]. In accordance with these findings, we found positive LGR5 expression in 90% of our sporadic CRC control group. However, only 30% of CAC were LGR5 positive. The latter finding is in agreement with the previous study by Yasuda et al. reporting significantly less expression of reliable intestinal stem cell markers such as CD133 [20], OCT4 [21] and NANOG [22] in CAC than sporadic CRC [23]. The molecular pathogenesis of CAC is different than that of sporadic CRCs with genomic changes that appear directly linked to the effects of ongoing inflammation and repeated mucosal injury in the IBD [11, 12]. In keeping with this, we recently reported CAC tends to show non-conventional tumor morphology and loose intestinal markers such as SATB2 and CDX2, and show aberrant gastric mucin expression [13], Ishibashi et al. described the expression profile of Atonal homolog-1 (ATOH1), a master transcription factor of the secretory lineage of intestinal epithelial cells, which retain their potential to revert to intestinal stem cells [24,25,26] in colitic mucosa. They identified both ATOH1 and LGR5 double positive tumor cells as well as LGR5 single positive tumor cells, suggesting that colitic tumors are mosaic and consisted of a heterogenous population of tumor stem cell [27]. Our data also support this interpretation given the heterogeneous distribution of LGR5 expression in the dysplastic and invasive components of some cases. While the exact mechanism of decreased LGR5 expression in CAC is uncertain, these data demonstrate that CACs have a unique expression profile of intestinal stem cell markers.

We found that despite only 40% of CACs in our cohort showing a conventional morphology, 89% of LGR5 positive CAC were conventional in appearance. These also tended to be located in the rectum and associated with nuclear beta-catenin staining. This raises the possibility that the minority of CACs in our cohort that showed conventional morphology might have arisen through the conventional APC pathway. However, as the majority of tumors had non-conventional morphology and were also LGR5 negative, our data provide support to other recent studies demonstrating that most CAC cases do not arise through the conventional APC pathway [12, 28]. Although it did not reach statistical significance, conventional type CACs trended toward a lower level of LGR5 expression compared to the control group of sporadic CRCs. Thus, even in tumors arising through more conventional molecular pathways in the setting of colitis, LGR5 expression might be affected by ongoing inflammation and repeated mucosal injury. This hypothesis is supported by other studies showing that Lgr5+ stem cells are highly sensitive to epithelial injury induced by radiation or colitis [27, 29, 30].

Our results in human cancers do not entirely align with previous data from animal models of colitis. Kim et al. reported increased Lgr5 expression in an AOM / DSS - colitis mouse model. They showed that Lgr5 expression is gradually increased as tumors developed with repeated colitis and reported all dysplastic lesions and cancers showed high Lgr5 expression [10]. This discrepancy could be due to differences between human CAC and the murine model. Interestingly, the figure showing tumor morphology in their report shows a more conventional morphology, implying that the murine model may simulate the carcinogenesis pathway in the conventional type of human CAC.

Data regarding any potential prognostic significance of LGR5 expression in sporadic CRC are mixed. Recent studies using RNA-ISH for LGR5 evaluation reported that LGR5 expression is an independent predictor of favorable outcome in CRCs [15, 31]. On the contrary, a previous meta-analyses of immunohistochemical studies of LGR5 expression showed that high LGR5 expression is associated with shorter overall survival and disease free survival [32, 33].

Kazama et al. described the immunohistochemical expression profile of LGR5 in ulcerative colitis cases and showed increased LGR5 expression in dysplasia and CAC [34]. This result is contrary to our present study, but may be due to the different methods of LGR5 detection, as the reliability of antibodies against LGR5 remains uncertain [35, 36], and most recent studies have used ISH to detect LGR5 expression.

One limitation of our cohort is the limited number of cases and the unavailability of clinical follow up data as well as further molecular analysis. Additional studies using a larger cohort will be needed to determine the relationship between LGR5 expression and clinical prognosis in human CACs.

Conclusion

Unlike sporadic CRC, LGR5 expression, along with nuclear beta-catenin expression is infrequent in CAC, particularly those with non-conventional morphology. It is more commonly expressed in CAC with conventional morphology and in tumors located in the rectum, indicating that a limited subset of CACs may arise through the conventional APC pathway. While the precise mechanisms for the low frequency of LGR5 expression in CAC remains undefined, the wider spectrum of tumor morphology in CAC may be associated with absence of a LGR5-expressing intestinal stem cell phenotype.

Availability of data and materials

All data generated and analyzed during the current study are available from the corresponding author on reasonable request.

Abbreviations

LGR5:

Leucine-rich repeat-containing G-protein-coupled receptor 5

CRC:

Colorectal adenocarcinoma

CAC:

Colitis associated colorectal adenocarcinoma

LGTG:

Low grade tubuloglandular

References

  1. Eaden JA, Abrams KR, Mayberry JF. The risk of colorectal cancer in ulcerative colitis: a meta-analysis. Gut. 2001;48(4):526–35.

    Article  CAS  Google Scholar 

  2. Lutgens MW, van Oijen MG, van der Heijden GJ, Vleggaar FP, Siersema PD, Oldenburg B. Declining risk of colorectal cancer in inflammatory bowel disease: an updated meta-analysis of population-based cohort studies. Inflamm Bowel Dis. 2013;19(4):789–99.

    Article  Google Scholar 

  3. Olen O, Erichsen R, Sachs MC, Pedersen L, Halfvarson J, Askling J, Ekbom A, Sorensen HT, Ludvigsson JF. Colorectal cancer in ulcerative colitis: a Scandinavian population-based cohort study. Lancet (London, England). 2020;395(10218):123–31.

    Article  Google Scholar 

  4. Barker N, van Es JH, Kuipers J, Kujala P, van den Born M, Cozijnsen M, Haegebarth A, Korving J, Begthel H, Peters PJ, et al. Identification of stem cells in small intestine and colon by marker gene Lgr5. Nature. 2007;449(7165):1003–7.

    Article  CAS  Google Scholar 

  5. Al-Kharusi MR, Smartt HJ, Greenhough A, Collard TJ, Emery ED, Williams AC, Paraskeva C. LGR5 promotes survival in human colorectal adenoma cells and is upregulated by PGE2: implications for targeting adenoma stem cells with NSAIDs. Carcinogenesis. 2013;34(5):1150–7.

    Article  CAS  Google Scholar 

  6. Fan XS, Wu HY, Yu HP, Zhou Q, Zhang YF, Huang Q. Expression of Lgr5 in human colorectal carcinogenesis and its potential correlation with beta-catenin. Int J Color Dis. 2010;25(5):583–90.

    Article  Google Scholar 

  7. Uchida H, Yamazaki K, Fukuma M, Yamada T, Hayashida T, Hasegawa H, Kitajima M, Kitagawa Y, Sakamoto M. Overexpression of leucine-rich repeat-containing G protein-coupled receptor 5 in colorectal cancer. Cancer Sci. 2010;101(7):1731–7.

    Article  CAS  Google Scholar 

  8. Baker AM, Graham TA, Elia G, Wright NA, Rodriguez-Justo M. Characterization of LGR5 stem cells in colorectal adenomas and carcinomas. Sci Rep. 2015;5:8654.

    Article  CAS  Google Scholar 

  9. Barker N, Ridgway RA, van Es JH, van de Wetering M, Begthel H, van den Born M, Danenberg E, Clarke AR, Sansom OJ, Clevers H. Crypt stem cells as the cells-of-origin of intestinal cancer. Nature. 2009;457(7229):608–11.

    Article  CAS  Google Scholar 

  10. Kim HS, Lee C, Kim WH, Maeng YH, Jang BG. Expression profile of intestinal stem cell markers in colitis-associated carcinogenesis. Sci Rep. 2017;7(1):6533.

    Article  Google Scholar 

  11. Robles AI, Traverso G, Zhang M, Roberts NJ, Khan MA, Joseph C, Lauwers GY, Selaru FM, Popoli M, Pittman ME, et al. Whole-exome sequencing analyses of inflammatory bowel disease-associated colorectal cancers. Gastroenterology. 2016;150(4):931–43.

    Article  CAS  Google Scholar 

  12. Yaeger R, Shah MA, Miller VA, Kelsen JR, Wang K, Heins ZJ, Ross JS, He Y, Sanford E, Yantiss RK, et al. Genomic alterations observed in colitis-associated cancers are distinct from those found in sporadic colorectal cancers and vary by type of inflammatory bowel disease. Gastroenterology. 2016;151(2):278–287.e276.

    Article  CAS  Google Scholar 

  13. Iwaya M, Ota H, Tateishi Y, Nakajima T, Riddell R, Conner JR. Colitis-associated colorectal adenocarcinomas are frequently associated with non-intestinal mucin profiles and loss of SATB2 expression. Modern Pathol. 2019;32:884.

    Article  CAS  Google Scholar 

  14. Ukpo OC, Flanagan JJ, Ma XJ, Luo Y, Thorstad WL, Lewis JS Jr. High-risk human papillomavirus E6/E7 mRNA detection by a novel in situ hybridization assay strongly correlates with p16 expression and patient outcomes in oropharyngeal squamous cell carcinoma. Am J Surg Pathol. 2011;35(9):1343–50.

    Article  Google Scholar 

  15. Jang BG, Kim HS, Chang WY, Bae JM, Kim WH, Kang GH. Expression profile of LGR5 and its prognostic significance in colorectal Cancer progression. Am J Pathol. 2018;188(10):2236–50.

    Article  CAS  Google Scholar 

  16. Kobayashi M, Honma T, Matsuda Y, Suzuki Y, Narisawa R, Ajioka Y, Asakura H. Nuclear translocation of beta-catenin in colorectal cancer. Br J Cancer. 2000;82(10):1689–93.

    Article  CAS  Google Scholar 

  17. Becker L, Huang Q, Mashimo H. Immunostaining of Lgr5, an intestinal stem cell marker, in normal and premalignant human gastrointestinal tissue. TheScientificWorldJournal. 2008;8:1168–76.

    Article  CAS  Google Scholar 

  18. Zheng Z, Yu H, Huang Q, Wu H, Fu Y, Shi J, Wang T, Fan X. Heterogeneous expression of Lgr5 as a risk factor for focal invasion and distant metastasis of colorectal carcinoma. Oncotarget. 2018;9(53):30025–33.

    Article  Google Scholar 

  19. Martin ML, Zeng Z, Adileh M, Jacobo A, Li C, Vakiani E, Hua G, Zhang L, Haimovitz-Friedman A, Fuks Z, et al. Logarithmic expansion of LGR5(+) cells in human colorectal cancer. Cell Signal. 2018;42:97–105.

    Article  CAS  Google Scholar 

  20. Horst D, Kriegl L, Engel J, Kirchner T, Jung A. CD133 expression is an independent prognostic marker for low survival in colorectal cancer. Br J Cancer. 2008;99(8):1285–9.

    Article  CAS  Google Scholar 

  21. Tai MH, Chang CC, Kiupel M, Webster JD, Olson LK, Trosko JE. Oct4 expression in adult human stem cells: evidence in support of the stem cell theory of carcinogenesis. Carcinogenesis. 2005;26(2):495–502.

    Article  CAS  Google Scholar 

  22. Wang J, Levasseur DN, Orkin SH. Requirement of Nanog dimerization for stem cell self-renewal and pluripotency. Proc Natl Acad Sci U S A. 2008;105(17):6326–31.

    Article  CAS  Google Scholar 

  23. Yasuda H, Tanaka K, Okita Y, Araki T, Saigusa S, Toiyama Y, Yokoe T, Yoshiyama S, Kawamoto A, Inoue Y, et al. CD133, OCT4, and NANOG in ulcerative colitis-associated colorectal cancer. Oncol Lett. 2011;2(6):1065–71.

    Article  CAS  Google Scholar 

  24. Buczacki SJ, Zecchini HI, Nicholson AM, Russell R, Vermeulen L, Kemp R, Winton DJ. Intestinal label-retaining cells are secretory precursors expressing Lgr5. Nature. 2013;495(7439):65–9.

    Article  CAS  Google Scholar 

  25. Kim TH, Saadatpour A, Guo G, Saxena M, Cavazza A, Desai N, Jadhav U, Jiang L, Rivera MN, Orkin SH, et al. Single-cell transcript profiles reveal multilineage priming in early progenitors derived from Lgr5(+) intestinal stem cells. Cell Rep. 2016;16(8):2053–60.

    Article  CAS  Google Scholar 

  26. van Es JH, Sato T, van de Wetering M, Lyubimova A, Yee Nee AN, Gregorieff A, Sasaki N, Zeinstra L, van den Born M, Korving J, et al. Dll1+ secretory progenitor cells revert to stem cells upon crypt damage. Nat Cell Biol. 2012;14(10):1099–104.

    Article  Google Scholar 

  27. Ishibashi F, Shimizu H, Nakata T, Fujii S, Suzuki K, Kawamoto A, Anzai S, Kuno R, Nagata S, Ito G, et al. Contribution of ATOH1(+) cells to the homeostasis, repair, and tumorigenesis of the colonic epithelium. Stem Cell Rep. 2018;10(1):27–42.

    Article  CAS  Google Scholar 

  28. Hartman DJ, Binion DG, Regueiro MD, Miller C, Herbst C, Pai RK. Distinct Histopathologic and molecular alterations in inflammatory bowel disease-associated intestinal adenocarcinoma: c-MYC amplification is common and associated with mucinous/signet ring cell differentiation. Inflamm Bowel Dis. 2018;24(8):1780–90.

    Article  Google Scholar 

  29. Asfaha S, Hayakawa Y, Muley A, Stokes S, Graham TA, Ericksen RE, Westphalen CB, von Burstin J, Mastracci TL, Worthley DL, et al. Krt19(+)/Lgr5(−) cells are Radioresistant Cancer-initiating stem cells in the Colon and Intestine. Cell Stem Cell. 2015;16(6):627–38.

    Article  CAS  Google Scholar 

  30. Yan KS, Chia LA, Li X, Ootani A, Su J, Lee JY, Su N, Luo Y, Heilshorn SC, Amieva MR, et al. The intestinal stem cell markers Bmi1 and Lgr5 identify two functionally distinct populations. Proc Natl Acad Sci U S A. 2012;109(2):466–71.

    Article  CAS  Google Scholar 

  31. Sato K, Uehara T, Iwaya M, Nakajima T, Miyagawa Y, Suga T, Ota H, Tanaka E. Correlation of clinicopathological features and LGR5 expression in colon adenocarcinoma. Ann Diagn Pathol. 2019;40:161–5.

    Article  Google Scholar 

  32. Chen Q, Zhang X, Li WM, Ji YQ, Cao HZ, Zheng P. Prognostic value of LGR5 in colorectal cancer: a meta-analysis. PLoS One. 2014;9(9):e107013.

    Article  Google Scholar 

  33. Jiang Y, Li W, He X, Zhang H, Jiang F, Chen Z. Lgr5 expression is a valuable prognostic factor for colorectal cancer: evidence from a meta-analysis. BMC Cancer. 2016;16:12.

    Article  Google Scholar 

  34. Kazama S, Kishikawa J, Tanaka T, Hata K, Kawai K, Nozawa H, Ishihara S. Immunohistochemical expression of CD133 and LGR5 in ulcerative colitis-associated colorectal cancer and dysplasia. In vivo (Athens, Greece). 2019;33(4):1279–84.

    CAS  Google Scholar 

  35. Barker N. Adult intestinal stem cells: critical drivers of epithelial homeostasis and regeneration. Nat Rev Mol Cell Biol. 2014;15(1):19–33.

    Article  CAS  Google Scholar 

  36. Kemper K, Prasetyanti PR, De Lau W, Rodermond H, Clevers H, Medema JP. Monoclonal antibodies against Lgr5 identify human colorectal cancer stem cells. Stem cells (Dayton, Ohio). 2012;30(11):2378–86.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

Not Applicable.

Disclosure

All authors have no financial relationships relevant to this publication.

Funding

The authors did not receive any funding.

Author information

Authors and Affiliations

Authors

Contributions

Each author has participated sufficiently in the work to take public responsibility for appropriate portions of the content: MI contributed to the conception and design of the study. MI, HO and JC analyzed histopathological features and drafted the manuscript. RR provided valuable advice and suggestions as a histopathologic consultant. TN and TU contributed to part of the RNA-ISH study. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Mai Iwaya.

Ethics declarations

Ethics approval and consent to participate

Study approval was obtained from the Research Ethics Board at Mount Sinai Hospital. The investigation was conducted in compliance with the Helsinki Declaration.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests” in this section.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Iwaya, M., Ota, H., Nakajima, T. et al. Most colitis associated carcinomas lack expression of LGR5: a preliminary study with implications for unique pathways of carcinogenesis compared to sporadic colorectal carcinoma. BMC Cancer 21, 119 (2021). https://doi.org/10.1186/s12885-021-07835-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12885-021-07835-3

Keywords