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

Open Access 01-12-2023 | Respiratory Microbiota | Research

Lactobacillus casei combined with Lactobacillus reuteri alleviate pancreatic cancer by inhibiting TLR4 to promote macrophage M1 polarization and regulate gut microbial homeostasis

Authors: Zemin Zhu, Bo Yi, Zikai Tang, Xun Chen, Ming Li, Tao Xu, Zhijian Zhao, Caixi Tang

Published in: BMC Cancer | Issue 1/2023

Login to get access

Abstract

Background

Pancreatic cancer is a highly lethal disease with no effective treatments. Lactobacillus casei (L. casei) and Lactobacillus reuteri (L. reuteri) exhibited therapeutic effects on several cancers, but their roles in pancreatic cancer are unknown. This study aims to explore how L. casei & L. reuteri influence pancreatic cancer and the underlying mechanisms.

Methods

Pancreatic cancer cells were treated with L. casei & L. reuteri and co-cultured with macrophages in a transwell system in vitro. Pancreatic cancer xenograft model was established and L. casei & L. reuteri was used to treat mice in vivo. MTT, CCK-8 assay or immunohistochemical staining were used to determine the proliferation of pancreatic cancer cells or tumor tissues. Transwell assay was applied to test the migration and invasion of pancreatic cells. RT-qPCR was utilized to assess TLR4 and MyD88 expressions in pancreatic cells or tumor tissues. WB, immunofluorescence staining, or flow cytometry was used to evaluate the M1/M2 polarization of macrophages. Besides, the composition of gut microbiota of tumor-bearing mice was determined by 16 S rRNA sequencing, and ultra-high performance liquid chromatography-mass spectrometry (UPLC-MS) untargeted metabolomics was used to evaluate the metabolic profiles of feces.

Results

L. casei & L. reuteri inhibited the proliferation, migration, invasion of pancreatic cancer cells and pancreatic cancer cell-induced M2 polarization of macrophages by suppressing TLR4. Meanwhile, L. casei & L. reuteri repressed pancreatic cancer growth and promoted M1 macrophage polarization. Besides, L. casei & L. reuteri reduced fecal Alloprevotella and increased fecal azelate and glutamate in nude mice, while TLR4 inhibitor TAK-242 increased Clostridia UCG-014, azelate, uridine, methionine sulfoxide, oxypurinol, and decreased glyceryl monoester in the feces of pancreatic tumor-bearing mice. Fecal oxypurinol and glyceryl monoester levels were positively or negatively associated with gut Clostridia UCG-014 abundance, respectively.

Conclusion

L. casei & L. reuteri alleviate pancreatic cancer by inhibiting TLR4 to promote macrophage M1 polarization and regulate gut microbial homeostasis.
Appendix
Available only for authorised users
Literature
1.
go back to reference Collaborators GBDPC. The global, regional, and national burden of Pancreatic cancer and its attributable risk factors in 195 countries and territories, 1990–2017: a systematic analysis for the global burden of Disease Study 2017. Lancet Gastroenterol Hepatol. 2019;4(12):934–47.CrossRef Collaborators GBDPC. The global, regional, and national burden of Pancreatic cancer and its attributable risk factors in 195 countries and territories, 1990–2017: a systematic analysis for the global burden of Disease Study 2017. Lancet Gastroenterol Hepatol. 2019;4(12):934–47.CrossRef
2.
go back to reference Neoptolemos JP, et al. Therapeutic developments in Pancreatic cancer: current and future perspectives. Nat Rev Gastroenterol Hepatol. 2018;15(6):333–48.PubMedCrossRef Neoptolemos JP, et al. Therapeutic developments in Pancreatic cancer: current and future perspectives. Nat Rev Gastroenterol Hepatol. 2018;15(6):333–48.PubMedCrossRef
3.
5.
go back to reference Huang X, et al. Regulation of Tumor microenvironment for Pancreatic cancer therapy. Biomaterials. 2021;270:120680.PubMedCrossRef Huang X, et al. Regulation of Tumor microenvironment for Pancreatic cancer therapy. Biomaterials. 2021;270:120680.PubMedCrossRef
6.
go back to reference Farajzadeh Valilou S, et al. The role of inflammatory cytokines and Tumor associated macrophages (TAMs) in microenvironment of Pancreatic cancer. Cytokine Growth Factor Rev. 2018;39:46–61.PubMedCrossRef Farajzadeh Valilou S, et al. The role of inflammatory cytokines and Tumor associated macrophages (TAMs) in microenvironment of Pancreatic cancer. Cytokine Growth Factor Rev. 2018;39:46–61.PubMedCrossRef
7.
go back to reference Yang J, et al. Macrophages in Pancreatic cancer: an immunometabolic perspective. Cancer Lett. 2021;498:188–200.PubMedCrossRef Yang J, et al. Macrophages in Pancreatic cancer: an immunometabolic perspective. Cancer Lett. 2021;498:188–200.PubMedCrossRef
8.
go back to reference Hu H, et al. The M2 phenotype of tumor-associated macrophages in the stroma confers a poor prognosis in Pancreatic cancer. Tumour Biol. 2016;37(7):8657–64.PubMedCrossRef Hu H, et al. The M2 phenotype of tumor-associated macrophages in the stroma confers a poor prognosis in Pancreatic cancer. Tumour Biol. 2016;37(7):8657–64.PubMedCrossRef
9.
go back to reference Laursen MF, Bahl MI, Licht TR. Settlers of our inner surface - factors shaping the gut microbiota from birth to toddlerhood. FEMS Microbiol Rev, 2021. 45(4). Laursen MF, Bahl MI, Licht TR. Settlers of our inner surface - factors shaping the gut microbiota from birth to toddlerhood. FEMS Microbiol Rev, 2021. 45(4).
10.
go back to reference Cheng WY, Wu CY, Yu J. The role of gut microbiota in cancer treatment: friend or foe? Gut. 2020;69(10):1867–76.PubMedCrossRef Cheng WY, Wu CY, Yu J. The role of gut microbiota in cancer treatment: friend or foe? Gut. 2020;69(10):1867–76.PubMedCrossRef
11.
go back to reference Mendez R, et al. Microbial dysbiosis and polyamine metabolism as predictive markers for early detection of Pancreatic cancer. Carcinogenesis. 2020;41(5):561–70.PubMedCrossRef Mendez R, et al. Microbial dysbiosis and polyamine metabolism as predictive markers for early detection of Pancreatic cancer. Carcinogenesis. 2020;41(5):561–70.PubMedCrossRef
12.
13.
go back to reference Leal-Lopes C et al. Roles of Commensal Microbiota in Pancreas Homeostasis and Pancreatic Pathologies J Diabetes Res, 2015. 2015: p. 284680. Leal-Lopes C et al. Roles of Commensal Microbiota in Pancreas Homeostasis and Pancreatic Pathologies J Diabetes Res, 2015. 2015: p. 284680.
15.
go back to reference Panebianco C et al. Engineered resistant-starch (ERS) Diet shapes Colon Microbiota Profile in parallel with the retardation of Tumor Growth in In Vitro and in vivo Pancreatic Cancer models. Nutrients, 2017. 9(4). Panebianco C et al. Engineered resistant-starch (ERS) Diet shapes Colon Microbiota Profile in parallel with the retardation of Tumor Growth in In Vitro and in vivo Pancreatic Cancer models. Nutrients, 2017. 9(4).
16.
go back to reference Sethi V, et al. Gut microbiota promotes Tumor Growth in mice by modulating Immune Response. Gastroenterology. 2018;155(1):33–37e6.PubMedCrossRef Sethi V, et al. Gut microbiota promotes Tumor Growth in mice by modulating Immune Response. Gastroenterology. 2018;155(1):33–37e6.PubMedCrossRef
17.
go back to reference Dong J, et al. Bidirectional and dynamic interaction between the microbiota and therapeutic resistance in Pancreatic cancer. Biochim Biophys Acta Rev Cancer. 2021;1875(1):188484.PubMedCrossRef Dong J, et al. Bidirectional and dynamic interaction between the microbiota and therapeutic resistance in Pancreatic cancer. Biochim Biophys Acta Rev Cancer. 2021;1875(1):188484.PubMedCrossRef
18.
20.
go back to reference Legesse Bedada T, et al. Probiotics for cancer alternative prevention and treatment. Biomed Pharmacother. 2020;129:110409.PubMedCrossRef Legesse Bedada T, et al. Probiotics for cancer alternative prevention and treatment. Biomed Pharmacother. 2020;129:110409.PubMedCrossRef
21.
go back to reference Zhu H, et al. The probiotic L. Casei Zhang slows the progression of acute and chronic Kidney Disease. Cell Metab. 2021;33(10):1926–42. e8.PubMedCrossRef Zhu H, et al. The probiotic L. Casei Zhang slows the progression of acute and chronic Kidney Disease. Cell Metab. 2021;33(10):1926–42. e8.PubMedCrossRef
22.
go back to reference Pan H, et al. A single bacterium restores the microbiome dysbiosis to protect bones from destruction in a rat model of rheumatoid arthritis. Microbiome. 2019;7(1):107.PubMedPubMedCentralCrossRef Pan H, et al. A single bacterium restores the microbiome dysbiosis to protect bones from destruction in a rat model of rheumatoid arthritis. Microbiome. 2019;7(1):107.PubMedPubMedCentralCrossRef
23.
go back to reference Kolodziej M, Szajewska H. Lactobacillus reuteri DSM 17938 in the prevention of antibiotic-associated Diarrhoea in children: a randomized clinical trial. Clin Microbiol Infect. 2019;25(6):699–704.PubMedCrossRef Kolodziej M, Szajewska H. Lactobacillus reuteri DSM 17938 in the prevention of antibiotic-associated Diarrhoea in children: a randomized clinical trial. Clin Microbiol Infect. 2019;25(6):699–704.PubMedCrossRef
24.
go back to reference Nilsson AG, et al. Lactobacillus reuteri reduces bone loss in older women with low bone mineral density: a randomized, placebo-controlled, double-blind, clinical trial. J Intern Med. 2018;284(3):307–17.PubMedCrossRef Nilsson AG, et al. Lactobacillus reuteri reduces bone loss in older women with low bone mineral density: a randomized, placebo-controlled, double-blind, clinical trial. J Intern Med. 2018;284(3):307–17.PubMedCrossRef
25.
go back to reference Hu C et al. Gut microbiota-derived short-chain fatty acids regulate group 3 innate lymphoid cells in HCC Hepatology, 2022. Hu C et al. Gut microbiota-derived short-chain fatty acids regulate group 3 innate lymphoid cells in HCC Hepatology, 2022.
27.
go back to reference Bell HN, et al. Reuterin in the healthy gut microbiome suppresses Colorectal cancer growth through altering redox balance. Cancer Cell. 2022;40(2):185–200e6.PubMedCrossRef Bell HN, et al. Reuterin in the healthy gut microbiome suppresses Colorectal cancer growth through altering redox balance. Cancer Cell. 2022;40(2):185–200e6.PubMedCrossRef
28.
go back to reference Samanta S. Potential impacts of Prebiotics and Probiotics on Cancer Prevention. Anticancer Agents Med Chem. 2022;22(4):605–28.PubMedCrossRef Samanta S. Potential impacts of Prebiotics and Probiotics on Cancer Prevention. Anticancer Agents Med Chem. 2022;22(4):605–28.PubMedCrossRef
29.
go back to reference Riaz Rajoka MS, et al. Anticancer potential against cervix cancer (HeLa) cell line of probiotic Lactobacillus casei and Lactobacillus paracasei strains isolated from human breast milk. Food Funct. 2018;9(5):2705–15.PubMedCrossRef Riaz Rajoka MS, et al. Anticancer potential against cervix cancer (HeLa) cell line of probiotic Lactobacillus casei and Lactobacillus paracasei strains isolated from human breast milk. Food Funct. 2018;9(5):2705–15.PubMedCrossRef
30.
go back to reference Rasouli BS, et al. In vitro activity of probiotic Lactobacillus reuteri against gastric cancer progression by downregulation of urokinase plasminogen activator/urokinase plasminogen activator receptor gene expression. J Cancer Res Ther. 2017;13(2):246–51.PubMedCrossRef Rasouli BS, et al. In vitro activity of probiotic Lactobacillus reuteri against gastric cancer progression by downregulation of urokinase plasminogen activator/urokinase plasminogen activator receptor gene expression. J Cancer Res Ther. 2017;13(2):246–51.PubMedCrossRef
32.
go back to reference Chen SM, et al. The synergistic Tumor growth-inhibitory effect of probiotic Lactobacillus on transgenic mouse model of Pancreatic cancer treated with gemcitabine. Sci Rep. 2020;10(1):20319.PubMedPubMedCentralCrossRef Chen SM, et al. The synergistic Tumor growth-inhibitory effect of probiotic Lactobacillus on transgenic mouse model of Pancreatic cancer treated with gemcitabine. Sci Rep. 2020;10(1):20319.PubMedPubMedCentralCrossRef
33.
go back to reference Lupi LA, et al. The role of toll-like receptor 4 signaling pathway in ovarian, cervical, and endometrial cancers. Life Sci. 2020;247:117435.PubMedCrossRef Lupi LA, et al. The role of toll-like receptor 4 signaling pathway in ovarian, cervical, and endometrial cancers. Life Sci. 2020;247:117435.PubMedCrossRef
34.
go back to reference Chen CY, Kao CL, Liu CM. The Cancer Prevention, anti-inflammatory and Anti-oxidation of Bioactive Phytochemicals Targeting the TLR4 Signaling Pathway. Int J Mol Sci, 2018. 19(9). Chen CY, Kao CL, Liu CM. The Cancer Prevention, anti-inflammatory and Anti-oxidation of Bioactive Phytochemicals Targeting the TLR4 Signaling Pathway. Int J Mol Sci, 2018. 19(9).
35.
go back to reference Grimmig T et al. Toll Like Receptor 2, 4, and 9 Signaling Promotes Autoregulative Tumor Cell Growth and VEGF/PDGF Expression in Human Pancreatic Cancer Int J Mol Sci, 2016. 17(12). Grimmig T et al. Toll Like Receptor 2, 4, and 9 Signaling Promotes Autoregulative Tumor Cell Growth and VEGF/PDGF Expression in Human Pancreatic Cancer Int J Mol Sci, 2016. 17(12).
36.
go back to reference Sun Y, et al. Toll-like receptor 4 promotes angiogenesis in Pancreatic cancer via PI3K/AKT signaling. Exp Cell Res. 2016;347(2):274–82.PubMedCrossRef Sun Y, et al. Toll-like receptor 4 promotes angiogenesis in Pancreatic cancer via PI3K/AKT signaling. Exp Cell Res. 2016;347(2):274–82.PubMedCrossRef
37.
go back to reference Yin H, et al. Gut-derived lipopolysaccharide remodels tumoral microenvironment and synergizes with PD-L1 checkpoint blockade via TLR4/MyD88/AKT/NF-kappaB pathway in Pancreatic cancer. Cell Death Dis. 2021;12(11):1033.PubMedPubMedCentralCrossRef Yin H, et al. Gut-derived lipopolysaccharide remodels tumoral microenvironment and synergizes with PD-L1 checkpoint blockade via TLR4/MyD88/AKT/NF-kappaB pathway in Pancreatic cancer. Cell Death Dis. 2021;12(11):1033.PubMedPubMedCentralCrossRef
38.
go back to reference Topcu KSB, et al. Investigation of the effects of the toll-like receptor 4 pathway on immune checkpoint vista in Pancreatic cancer. Invest New Drugs. 2022;40(3):519–28.PubMedCrossRef Topcu KSB, et al. Investigation of the effects of the toll-like receptor 4 pathway on immune checkpoint vista in Pancreatic cancer. Invest New Drugs. 2022;40(3):519–28.PubMedCrossRef
39.
go back to reference Michaud D, et al. Cell receptor signaling and protein kinase D2 support Regulatory B cell function in Pancreatic Cancer. Front Immunol. 2021;12:745873.PubMedCrossRef Michaud D, et al. Cell receptor signaling and protein kinase D2 support Regulatory B cell function in Pancreatic Cancer. Front Immunol. 2021;12:745873.PubMedCrossRef
40.
go back to reference Gao J, Liang Y, Wang L. Shap Polarization Tumor-Associated Macrophages Cancer Immunotherapy Front Immunol. 2022;13:888713. Gao J, Liang Y, Wang L. Shap Polarization Tumor-Associated Macrophages Cancer Immunotherapy Front Immunol. 2022;13:888713.
41.
go back to reference Wang X, et al. Hypoxic tumor-derived exosomal miR-301a mediates M2 macrophage polarization via PTEN/PI3Kgamma to promote Pancreatic Cancer Metastasis. Cancer Res. 2018;78(16):4586–98.PubMedCrossRef Wang X, et al. Hypoxic tumor-derived exosomal miR-301a mediates M2 macrophage polarization via PTEN/PI3Kgamma to promote Pancreatic Cancer Metastasis. Cancer Res. 2018;78(16):4586–98.PubMedCrossRef
42.
go back to reference He Z et al. Exosome-derived FGD5-AS1 promotes tumor-associated macrophage M2 polarization-mediated Pancreatic cancer cell proliferation and Metastasis. Cancer Lett, 2022: p. 215751. He Z et al. Exosome-derived FGD5-AS1 promotes tumor-associated macrophage M2 polarization-mediated Pancreatic cancer cell proliferation and Metastasis. Cancer Lett, 2022: p. 215751.
43.
go back to reference Alexander AF, et al. Single-cell secretion analysis reveals a dual role for IL-10 in restraining and resolving the TLR4-induced inflammatory response. Cell Rep. 2021;36(12):109728.PubMedPubMedCentralCrossRef Alexander AF, et al. Single-cell secretion analysis reveals a dual role for IL-10 in restraining and resolving the TLR4-induced inflammatory response. Cell Rep. 2021;36(12):109728.PubMedPubMedCentralCrossRef
44.
go back to reference Adolph TE, et al. Pancreas-Microbiota Cross talk in Health and Disease. Annu Rev Nutr. 2019;39:249–66.PubMedCrossRef Adolph TE, et al. Pancreas-Microbiota Cross talk in Health and Disease. Annu Rev Nutr. 2019;39:249–66.PubMedCrossRef
45.
go back to reference Zhang L, et al. The oral Microbiota May have influence on Oral Cancer. Front Cell Infect Microbiol. 2019;9:476.PubMedCrossRef Zhang L, et al. The oral Microbiota May have influence on Oral Cancer. Front Cell Infect Microbiol. 2019;9:476.PubMedCrossRef
46.
go back to reference Zeng B, et al. The Oral cancer microbiome contains Tumor space-specific and clinicopathology-specific bacteria. Front Cell Infect Microbiol. 2022;12:942328.PubMedPubMedCentralCrossRef Zeng B, et al. The Oral cancer microbiome contains Tumor space-specific and clinicopathology-specific bacteria. Front Cell Infect Microbiol. 2022;12:942328.PubMedPubMedCentralCrossRef
48.
go back to reference Wang C, et al. Saccharomyces boulardii alleviates ulcerative Colitis carcinogenesis in mice by reducing TNF-alpha and IL-6 levels and functions and by rebalancing intestinal microbiota. BMC Microbiol. 2019;19(1):246.PubMedPubMedCentralCrossRef Wang C, et al. Saccharomyces boulardii alleviates ulcerative Colitis carcinogenesis in mice by reducing TNF-alpha and IL-6 levels and functions and by rebalancing intestinal microbiota. BMC Microbiol. 2019;19(1):246.PubMedPubMedCentralCrossRef
49.
go back to reference Manosroi A, et al. Characteristics and anti-proliferative activity of azelaic acid and its derivatives entrapped in bilayer vesicles in cancer cell lines. J Drug Target. 2007;15(5):334–41.PubMedCrossRef Manosroi A, et al. Characteristics and anti-proliferative activity of azelaic acid and its derivatives entrapped in bilayer vesicles in cancer cell lines. J Drug Target. 2007;15(5):334–41.PubMedCrossRef
52.
go back to reference Capurso G et al. Probiotics and severe acute pancreatitis J Clin Gastroenterol, 2008. 42 Suppl 3 Pt 1: p. S148-51. Capurso G et al. Probiotics and severe acute pancreatitis J Clin Gastroenterol, 2008. 42 Suppl 3 Pt 1: p. S148-51.
Metadata
Title
Lactobacillus casei combined with Lactobacillus reuteri alleviate pancreatic cancer by inhibiting TLR4 to promote macrophage M1 polarization and regulate gut microbial homeostasis
Authors
Zemin Zhu
Bo Yi
Zikai Tang
Xun Chen
Ming Li
Tao Xu
Zhijian Zhao
Caixi Tang
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/s12885-023-11557-z

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