Skip to main content
Top
Published in: BMC Cancer 1/2023

Open Access 01-12-2023 | Cytokines | Research

Notch3 signaling promotes colorectal tumor growth by enhancing immunosuppressive cells infiltration in the microenvironment

Authors: Kai Huang, Wenwu Luo, Jinmei Fang, Changjun Yu, Guangjie Liu, Xiaodong Yuan, Yun Liu, Wenyong Wu

Published in: BMC Cancer | Issue 1/2023

Login to get access

Abstract

Background

Macrophage infiltration in the tumor microenvironment participates in the regulation of tumor progression. Previous studies have found that Notch signaling pathway is involved in regulating the progression of colorectal cancer (CRC), however, the specific mechanism is still unclear.

Methods

The correlation between Notch signaling pathway and macrophage infiltration was investigated in TCGA database and verified in clinical samples of patients with CRC using immunohistochemistry. Gene Set Enrichment Analysis was used to find out genes related to Notch3 expression. Colony formation assay, and flow cytometry were utilized to test tumor growth and immune cell infiltration in vitro and in vivo.

Results

Using bioinformatics analysis and clinical sample validation, we found that Notch3 was highly expressed in colon tumor tissues compared to adjacent normal tissues, and it participated in regulating the recruitment of macrophages to the tumor microenvironment. Furthermore, we found that the Notch3 expression was positively correlated with the expression of macrophage recruitment-related cytokines in colon tumor tissues. Finally, we demonstrated that depletion of Notch3 had no significant effect on the growth of colon tumor cells in vitro, while, attenuated the growth of colon cancer tumors in vivo. Simultaneous, immunosuppressive cells, macrophages and myeloid-derived suppressor cell (MDSC) infiltration were dramatically reduced in the tumor microenvironment.

Conclusion

Our study illustrated that Notch3 could facilitate the progression of CRC by increasing the infiltration of macrophages and MDSCs to promote the immunosuppressive tumor microenvironment. Targeting Notch3 specifically is a potentially effective treatment for CRC.
Appendix
Available only for authorised users
Literature
1.
go back to reference Rawla P, Sunkara T, Barsouk A. Epidemiology of colorectal cancer: incidence, mortality, survival, and risk factors. Przeglad gastroenterologiczny. 2019;14(2):89–103. Rawla P, Sunkara T, Barsouk A. Epidemiology of colorectal cancer: incidence, mortality, survival, and risk factors. Przeglad gastroenterologiczny. 2019;14(2):89–103.
2.
go back to reference Siegel RL, Miller KD, Fedewa SA, Ahnen DJ, Meester RGS, Barzi A, et al. Colorectal cancer statistics, 2017. CA Cancer J Clin. 2017;67(3):177–93.CrossRef Siegel RL, Miller KD, Fedewa SA, Ahnen DJ, Meester RGS, Barzi A, et al. Colorectal cancer statistics, 2017. CA Cancer J Clin. 2017;67(3):177–93.CrossRef
3.
go back to reference Müller MF, Ibrahim AE, Arends MJ. Molecular pathological classification of colorectal cancer. Virchows Arch. 2016;469(2):125–34.CrossRef Müller MF, Ibrahim AE, Arends MJ. Molecular pathological classification of colorectal cancer. Virchows Arch. 2016;469(2):125–34.CrossRef
4.
go back to reference Demitrack ES, Samuelson LC. Notch regulation of gastrointestinal stem cells. J Physiol. 2016;594(17):4791–803.CrossRef Demitrack ES, Samuelson LC. Notch regulation of gastrointestinal stem cells. J Physiol. 2016;594(17):4791–803.CrossRef
5.
go back to reference Sonoshita M, Aoki M, Fuwa H, Aoki K, Hosogi H, Sakai Y, et al. Suppression of colon cancer metastasis by Aes through inhibition of Notch signaling. Cancer Cell. 2011;19(1):125–37.CrossRef Sonoshita M, Aoki M, Fuwa H, Aoki K, Hosogi H, Sakai Y, et al. Suppression of colon cancer metastasis by Aes through inhibition of Notch signaling. Cancer Cell. 2011;19(1):125–37.CrossRef
6.
go back to reference Suman S, Das TP, Ankem MK, Damodaran C. Targeting Notch signaling in colorectal Cancer. Curr Colorectal Cancer Rep. 2014;10(4):411–6.CrossRef Suman S, Das TP, Ankem MK, Damodaran C. Targeting Notch signaling in colorectal Cancer. Curr Colorectal Cancer Rep. 2014;10(4):411–6.CrossRef
7.
go back to reference Serafin V, Persano L, Moserle L, Esposito G, Ghisi M, Curtarello M, et al. Notch3 signalling promotes tumour growth in colorectal cancer. J Pathol. 2011;224(4):448–60.CrossRef Serafin V, Persano L, Moserle L, Esposito G, Ghisi M, Curtarello M, et al. Notch3 signalling promotes tumour growth in colorectal cancer. J Pathol. 2011;224(4):448–60.CrossRef
8.
go back to reference Liu K, Wang J-F, Zhan Y, Kong D-L, Wang C. Prognosis model of colorectal cancer patients based on NOTCH3, KMT2C , and CREBBP mutations. J Gastrointest Oncol. 2021;12(1):79–88.CrossRef Liu K, Wang J-F, Zhan Y, Kong D-L, Wang C. Prognosis model of colorectal cancer patients based on NOTCH3, KMT2C , and CREBBP mutations. J Gastrointest Oncol. 2021;12(1):79–88.CrossRef
9.
go back to reference López-López S, Monsalve EM, Romero de Ávila MJ, González-Gómez J, Hernández de León N, Ruiz-Marcos F, et al. NOTCH3 signaling is essential for NF-κB activation in TLR-activated macrophages. Sci Rep. 2020;10(1):14839.CrossRef López-López S, Monsalve EM, Romero de Ávila MJ, González-Gómez J, Hernández de León N, Ruiz-Marcos F, et al. NOTCH3 signaling is essential for NF-κB activation in TLR-activated macrophages. Sci Rep. 2020;10(1):14839.CrossRef
10.
go back to reference Wang H, Tian T, Zhang J. Tumor-associated macrophages (TAMs) in colorectal Cancer (CRC): from mechanism to therapy and prognosis. Int J Mol Sci. 2021;22(16):8470.CrossRef Wang H, Tian T, Zhang J. Tumor-associated macrophages (TAMs) in colorectal Cancer (CRC): from mechanism to therapy and prognosis. Int J Mol Sci. 2021;22(16):8470.CrossRef
11.
go back to reference Li C, Tang Z, Zhang W, Ye Z, Liu F. GEPIA2021: integrating multiple deconvolution-based analysis into GEPIA. Nucleic Acids Res. 2021;49(W1):W242–6.CrossRef Li C, Tang Z, Zhang W, Ye Z, Liu F. GEPIA2021: integrating multiple deconvolution-based analysis into GEPIA. Nucleic Acids Res. 2021;49(W1):W242–6.CrossRef
12.
go back to reference Yuan X, Huang L, Luo W, Zhao Y, Nashan B, Yu F, et al. Diagnostic and prognostic significances of SOX9 in Thymic epithelial tumor. Front Oncol. 2021;11:708735.CrossRef Yuan X, Huang L, Luo W, Zhao Y, Nashan B, Yu F, et al. Diagnostic and prognostic significances of SOX9 in Thymic epithelial tumor. Front Oncol. 2021;11:708735.CrossRef
13.
go back to reference Li Z, Razavi P, Li Q, Toy W, Liu B, Ping C, et al. Loss of the FAT1 tumor suppressor promotes resistance to CDK4/6 inhibitors via the hippo pathway. Cancer Cell. 2018;34(6):893–905 e8.CrossRef Li Z, Razavi P, Li Q, Toy W, Liu B, Ping C, et al. Loss of the FAT1 tumor suppressor promotes resistance to CDK4/6 inhibitors via the hippo pathway. Cancer Cell. 2018;34(6):893–905 e8.CrossRef
14.
go back to reference Ru B, Wong CN, Tong Y, Zhong JY, Zhong SSW, Wu WC, et al. TISIDB: an integrated repository portal for tumor-immune system interactions. Bioinformatics. 2019;35(20):4200–2.CrossRef Ru B, Wong CN, Tong Y, Zhong JY, Zhong SSW, Wu WC, et al. TISIDB: an integrated repository portal for tumor-immune system interactions. Bioinformatics. 2019;35(20):4200–2.CrossRef
15.
go back to reference Li T, Fu J, Zeng Z, Cohen D, Li J, Chen Q, et al. TIMER2.0 for analysis of tumor-infiltrating immune cells. Nucleic Acids Res. 2020;48(W1):W509–14.CrossRef Li T, Fu J, Zeng Z, Cohen D, Li J, Chen Q, et al. TIMER2.0 for analysis of tumor-infiltrating immune cells. Nucleic Acids Res. 2020;48(W1):W509–14.CrossRef
16.
go back to reference Huang K, Chen S, Xie R, Jiang P, Yu C, Fang J, et al. Identification of three predictors of gastric cancer progression and prognosis. FEBS Open Bio. 2020;10(9):1891–9.CrossRef Huang K, Chen S, Xie R, Jiang P, Yu C, Fang J, et al. Identification of three predictors of gastric cancer progression and prognosis. FEBS Open Bio. 2020;10(9):1891–9.CrossRef
17.
go back to reference Li C, Song J, Guo Z, Gong Y, Zhang T, Huang J, et al. EZH2 inhibitors suppress colorectal Cancer by regulating macrophage polarization in the tumor microenvironment. Front Immunol. 2022;13:857808.CrossRef Li C, Song J, Guo Z, Gong Y, Zhang T, Huang J, et al. EZH2 inhibitors suppress colorectal Cancer by regulating macrophage polarization in the tumor microenvironment. Front Immunol. 2022;13:857808.CrossRef
18.
go back to reference Aburjania Z, Jang S, Whitt J, Jaskula-Stzul R, Chen H, Rose JB. The role of Notch3 in Cancer. Oncologist. 2018;23(8):900–11.CrossRef Aburjania Z, Jang S, Whitt J, Jaskula-Stzul R, Chen H, Rose JB. The role of Notch3 in Cancer. Oncologist. 2018;23(8):900–11.CrossRef
19.
go back to reference Ozawa T, Kazama S, Akiyoshi T, Murono K, Yoneyama S, Tanaka T, et al. Nuclear Notch3 expression is associated with tumor recurrence in patients with stage II and III colorectal cancer. Ann Surg Oncol. 2014;21(8):2650–8.CrossRef Ozawa T, Kazama S, Akiyoshi T, Murono K, Yoneyama S, Tanaka T, et al. Nuclear Notch3 expression is associated with tumor recurrence in patients with stage II and III colorectal cancer. Ann Surg Oncol. 2014;21(8):2650–8.CrossRef
20.
go back to reference Varga J, Nicolas A, Petrocelli V, Pesic M, Mahmoud A, Michels BE, et al. AKT-dependent NOTCH3 activation drives tumor progression in a model of mesenchymal colorectal cancer. J Exp Med. 2020;217(10):e2019151.CrossRef Varga J, Nicolas A, Petrocelli V, Pesic M, Mahmoud A, Michels BE, et al. AKT-dependent NOTCH3 activation drives tumor progression in a model of mesenchymal colorectal cancer. J Exp Med. 2020;217(10):e2019151.CrossRef
21.
go back to reference Pastò A, Serafin V, Pilotto G, Lago C, Bellio C, Trusolino L, et al. NOTCH3 signaling regulates MUSASHI-1 expression in metastatic colorectal cancer cells. Cancer Res. 2014;74(7):2106–18.CrossRef Pastò A, Serafin V, Pilotto G, Lago C, Bellio C, Trusolino L, et al. NOTCH3 signaling regulates MUSASHI-1 expression in metastatic colorectal cancer cells. Cancer Res. 2014;74(7):2106–18.CrossRef
22.
go back to reference Koch U, Radtke F. A third Notch in colorectal cancer progression and metastasis. J Exp Med. 2020;217(10):e20201017.CrossRef Koch U, Radtke F. A third Notch in colorectal cancer progression and metastasis. J Exp Med. 2020;217(10):e20201017.CrossRef
23.
go back to reference Chen Y, Song Y, Du W, Gong L, Chang H, Zou Z. Tumor-associated macrophages: an accomplice in solid tumor progression. J Biomed Sci. 2019;26(1):78.CrossRef Chen Y, Song Y, Du W, Gong L, Chang H, Zou Z. Tumor-associated macrophages: an accomplice in solid tumor progression. J Biomed Sci. 2019;26(1):78.CrossRef
24.
go back to reference Trimaglio G, Tilkin-Mariamé AF, Feliu V, Lauzéral-Vizcaino F, Tosolini M, Valle C, et al. Colon-specific immune microenvironment regulates cancer progression versus rejection. Oncoimmunology. 2020;9(1):1790125.CrossRef Trimaglio G, Tilkin-Mariamé AF, Feliu V, Lauzéral-Vizcaino F, Tosolini M, Valle C, et al. Colon-specific immune microenvironment regulates cancer progression versus rejection. Oncoimmunology. 2020;9(1):1790125.CrossRef
25.
go back to reference Ålgars A, Kemppinen L, Fair-Mäkelä R, Mustonen H, Haglund C, Jalkanen S. Stage I-IV colorectal Cancer prognosis can be predicted by type and number of Intratumoral macrophages and CLEVER-1(+) vessel density. Cancers (Basel). 2021;13(23):5988.CrossRef Ålgars A, Kemppinen L, Fair-Mäkelä R, Mustonen H, Haglund C, Jalkanen S. Stage I-IV colorectal Cancer prognosis can be predicted by type and number of Intratumoral macrophages and CLEVER-1(+) vessel density. Cancers (Basel). 2021;13(23):5988.CrossRef
26.
go back to reference Yin Y, Yao S, Hu Y, Feng Y, Li M, Bian Z, et al. The immune-microenvironment confers Chemoresistance of colorectal Cancer through macrophage-derived IL6. Clin Cancer Res. 2017;23(23):7375–87.CrossRef Yin Y, Yao S, Hu Y, Feng Y, Li M, Bian Z, et al. The immune-microenvironment confers Chemoresistance of colorectal Cancer through macrophage-derived IL6. Clin Cancer Res. 2017;23(23):7375–87.CrossRef
27.
go back to reference Colombo M, Mirandola L, Chiriva-Internati M, Basile A, Locati M, Lesma E, et al. Cancer cells exploit Notch signaling to redefine a supportive cytokine milieu. Front Immunol. 2018;9:1823.CrossRef Colombo M, Mirandola L, Chiriva-Internati M, Basile A, Locati M, Lesma E, et al. Cancer cells exploit Notch signaling to redefine a supportive cytokine milieu. Front Immunol. 2018;9:1823.CrossRef
28.
go back to reference Ye YC, Zhao JL, Lu YT, Gao CC, Yang Y, Liang SQ, et al. NOTCH signaling via WNT regulates the proliferation of alternative, CCR2-independent tumor-associated macrophages in hepatocellular carcinoma. Cancer Res. 2019;79(16):4160–72.CrossRef Ye YC, Zhao JL, Lu YT, Gao CC, Yang Y, Liang SQ, et al. NOTCH signaling via WNT regulates the proliferation of alternative, CCR2-independent tumor-associated macrophages in hepatocellular carcinoma. Cancer Res. 2019;79(16):4160–72.CrossRef
29.
go back to reference Hossain F, Majumder S, Ucar DA, Rodriguez PC, Golde TE, Minter LM, et al. Notch signaling in myeloid cells as a regulator of tumor immune responses. Front Immunol. 2018;9:1288.CrossRef Hossain F, Majumder S, Ucar DA, Rodriguez PC, Golde TE, Minter LM, et al. Notch signaling in myeloid cells as a regulator of tumor immune responses. Front Immunol. 2018;9:1288.CrossRef
30.
go back to reference Cui Y, Li Q, Li W, Wang Y, Lv F, Shi X, et al. NOTCH3 is a prognostic factor and is correlated with immune tolerance in gastric Cancer. Front Oncol. 2020;10:574937.CrossRef Cui Y, Li Q, Li W, Wang Y, Lv F, Shi X, et al. NOTCH3 is a prognostic factor and is correlated with immune tolerance in gastric Cancer. Front Oncol. 2020;10:574937.CrossRef
31.
go back to reference Gallo G, Vescio G, De Paola G, Sammarco G. Therapeutic targets and tumor microenvironment in colorectal Cancer. J Clin Med. 2021;10(11):2295.CrossRef Gallo G, Vescio G, De Paola G, Sammarco G. Therapeutic targets and tumor microenvironment in colorectal Cancer. J Clin Med. 2021;10(11):2295.CrossRef
32.
go back to reference Lin W, Xu D, Austin CD, Caplazi P, Senger K, Sun Y, et al. Function of CSF1 and IL34 in macrophage homeostasis, inflammation, and Cancer. Front Immunol. 2019;10:2019.CrossRef Lin W, Xu D, Austin CD, Caplazi P, Senger K, Sun Y, et al. Function of CSF1 and IL34 in macrophage homeostasis, inflammation, and Cancer. Front Immunol. 2019;10:2019.CrossRef
33.
go back to reference Huang YH, Cai K, Xu PP, Wang L, Huang CX, Fang Y, et al. CREBBP/EP300 mutations promoted tumor progression in diffuse large B-cell lymphoma through altering tumor-associated macrophage polarization via FBXW7-NOTCH-CCL2/CSF1 axis. Signal Transduct Target Ther. 2021;6(1):10.CrossRef Huang YH, Cai K, Xu PP, Wang L, Huang CX, Fang Y, et al. CREBBP/EP300 mutations promoted tumor progression in diffuse large B-cell lymphoma through altering tumor-associated macrophage polarization via FBXW7-NOTCH-CCL2/CSF1 axis. Signal Transduct Target Ther. 2021;6(1):10.CrossRef
34.
go back to reference Pathria P, Louis TL, Varner JA. Targeting tumor-associated macrophages in Cancer. Trends Immunol. 2019;40(4):310–27.CrossRef Pathria P, Louis TL, Varner JA. Targeting tumor-associated macrophages in Cancer. Trends Immunol. 2019;40(4):310–27.CrossRef
35.
go back to reference Chun E, Lavoie S, Michaud M, Gallini CA, Kim J, Soucy G, et al. CCL2 promotes colorectal carcinogenesis by enhancing Polymorphonuclear myeloid-derived suppressor cell population and function. Cell Rep. 2015;12(2):244–57.CrossRef Chun E, Lavoie S, Michaud M, Gallini CA, Kim J, Soucy G, et al. CCL2 promotes colorectal carcinogenesis by enhancing Polymorphonuclear myeloid-derived suppressor cell population and function. Cell Rep. 2015;12(2):244–57.CrossRef
36.
go back to reference Shen Q, Cohen B, Zheng W, Rahbar R, Martin B, Murakami K, et al. Notch shapes the innate Immunophenotype in breast Cancer. Cancer Discov. 2017;7(11):1320–35.CrossRef Shen Q, Cohen B, Zheng W, Rahbar R, Martin B, Murakami K, et al. Notch shapes the innate Immunophenotype in breast Cancer. Cancer Discov. 2017;7(11):1320–35.CrossRef
37.
go back to reference Khare T, Bissonnette M, Khare S. CXCL12-CXCR4/CXCR7 Axis in Colorectal Cancer: Therapeutic Target in Preclinical and Clinical Studies. Int J Mol Sci. 2021;22(14):7371. Khare T, Bissonnette M, Khare S. CXCL12-CXCR4/CXCR7 Axis in Colorectal Cancer: Therapeutic Target in Preclinical and Clinical Studies. Int J Mol Sci. 2021;22(14):7371.
38.
go back to reference Yu X, Wang D, Wang X, Sun S, Zhang Y, Wang S, et al. CXCL12/CXCR4 promotes inflammation-driven colorectal cancer progression through activation of RhoA signaling by sponging miR-133a-3p. J Exp Clin Cancer Res. 2019;38(1):32.CrossRef Yu X, Wang D, Wang X, Sun S, Zhang Y, Wang S, et al. CXCL12/CXCR4 promotes inflammation-driven colorectal cancer progression through activation of RhoA signaling by sponging miR-133a-3p. J Exp Clin Cancer Res. 2019;38(1):32.CrossRef
39.
go back to reference Zboralski D, Hoehlig K, Eulberg D, Frömming A, Vater A. Increasing tumor-infiltrating T cells through inhibition of CXCL12 with NOX-A12 synergizes with PD-1 blockade. Cancer Immunol Res. 2017;5(11):950–6.CrossRef Zboralski D, Hoehlig K, Eulberg D, Frömming A, Vater A. Increasing tumor-infiltrating T cells through inhibition of CXCL12 with NOX-A12 synergizes with PD-1 blockade. Cancer Immunol Res. 2017;5(11):950–6.CrossRef
40.
go back to reference Jackstadt R, van Hooff SR, Leach JD, Cortes-Lavaud X, Lohuis JO, Ridgway RA, et al. Epithelial NOTCH signaling rewires the tumor microenvironment of colorectal Cancer to drive poor-prognosis subtypes and metastasis. Cancer Cell. 2019;36(3):319–336.e7.CrossRef Jackstadt R, van Hooff SR, Leach JD, Cortes-Lavaud X, Lohuis JO, Ridgway RA, et al. Epithelial NOTCH signaling rewires the tumor microenvironment of colorectal Cancer to drive poor-prognosis subtypes and metastasis. Cancer Cell. 2019;36(3):319–336.e7.CrossRef
41.
go back to reference Vinson KE, George DC, Fender AW, Bertrand FE, Sigounas G. The Notch pathway in colorectal cancer. Int J Cancer. 2016;138(8):1835–42.CrossRef Vinson KE, George DC, Fender AW, Bertrand FE, Sigounas G. The Notch pathway in colorectal cancer. Int J Cancer. 2016;138(8):1835–42.CrossRef
42.
go back to reference Wang F, Huang C, Long J, Zhao ZB, Ma HQ, Yao XQ, et al. Notch signaling mutations increase intra-tumor chemokine expression and predict response to immunotherapy in colorectal cancer. BMC Cancer. 2022;22(1):933.CrossRef Wang F, Huang C, Long J, Zhao ZB, Ma HQ, Yao XQ, et al. Notch signaling mutations increase intra-tumor chemokine expression and predict response to immunotherapy in colorectal cancer. BMC Cancer. 2022;22(1):933.CrossRef
43.
go back to reference Tyagi A, Sharma AK, Damodaran C. A Review on Notch Signaling and Colorectal Cancer Cells. Cells. 2020;9(6):1549.CrossRef Tyagi A, Sharma AK, Damodaran C. A Review on Notch Signaling and Colorectal Cancer Cells. Cells. 2020;9(6):1549.CrossRef
44.
go back to reference Jo MK, Moon CM, Kim EJ, Kwon JH, Fei X, Kim SE, et al. Suppressive effect of α-mangostin for cancer stem cells in colorectal cancer via the Notch pathway. BMC Cancer. 2022;22(1):341.CrossRef Jo MK, Moon CM, Kim EJ, Kwon JH, Fei X, Kim SE, et al. Suppressive effect of α-mangostin for cancer stem cells in colorectal cancer via the Notch pathway. BMC Cancer. 2022;22(1):341.CrossRef
Metadata
Title
Notch3 signaling promotes colorectal tumor growth by enhancing immunosuppressive cells infiltration in the microenvironment
Authors
Kai Huang
Wenwu Luo
Jinmei Fang
Changjun Yu
Guangjie Liu
Xiaodong Yuan
Yun Liu
Wenyong Wu
Publication date
01-12-2023
Publisher
BioMed Central
Published in
BMC Cancer / Issue 1/2023
Electronic ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-023-10526-w

Other articles of this Issue 1/2023

BMC Cancer 1/2023 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine