1
|
Liu C, Tan M, Zhao L, Gai M, Zhou T, Yu C, Zhao Z. Anticancer activity of Weizmannia coagulans MZY531on H22 tumor-bearing mice by regulating inflammation, autophagy-dependent apoptosis, and gut microbiota. Sci Rep 2025; 15:8250. [PMID: 40065050 PMCID: PMC11894224 DOI: 10.1038/s41598-025-92825-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 03/03/2025] [Indexed: 03/14/2025] Open
Abstract
Increasing studies have shown that the efficacy of Weizmannia coagulans in treating various cancers. We recently identified W. coagulans MZY531 with potent cell anti-proliferation and exhibiting apoptosis induction activities against the mouse H22 hepatocellular carcinoma cell line.However, the anti-cancer effect of W. coagulans MZY531 against liver cancer in vivo has not been verified. The objective of this study was to assess the anti-hepatoma effect of W. coagulans MZY531 on H22 tumor-bearing mice and the underlying mechanism. The results demonstrated that W. coagulans MZY531 reduced the weight and size of the tumor in comparison to the model group. The levels of serum pro-inflammatory cytokines, including IL-1β, IL-6, IL-2 and TNF-α were suppressed by W. coagulans MZY531 administration. Immunofluorescence and TUNEL analyses demonstrated that W. coagulans MZY531 significantly increased the number of cleaved caspase-3 cells and induced apoptosis in tumor tissues. Importantly, W. coagulans MZY531 activated the AMPK/mTOR autophagy-dependent apoptosis pathway, and regulated the TLR4/MyD88/TRAF-6/NF-κB and JAK2/STAT3 inflammatory signaling pathways through mechanisms. Additionally, Fecal analysis demonstrated the capacity of W. coagulans MZY531 to remodel the gut microbiota of hepatocellular carcinoma-infected mice. Collectively, this experimental finding suggested that W. coagulans MZY531 exhibited prominent anticancer activities in vivo at least partly via reducing inflammation, inducing autophagy-dependent apoptosis, and regulating gut microbiota in H22 tumor-bearing mice.
Collapse
Affiliation(s)
- Chunhong Liu
- College of Special Education, Changchun University, Changchun, 130022, People's Republic of China
| | - Mengyao Tan
- College of Special Education, Changchun University, Changchun, 130022, People's Republic of China
| | - Lijun Zhao
- College of Special Education, Changchun University, Changchun, 130022, People's Republic of China
| | - Meichen Gai
- International Medical Department of Guang'anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, 100053, People's Republic of China
| | - Tingting Zhou
- College of Traditional Chinese Medicine, Changchun University of Traditional Chinese Medicine, Changchun, 130117, People's Republic of China
| | - Caixin Yu
- College of Special Education, Changchun University, Changchun, 130022, People's Republic of China
| | - Zhongwei Zhao
- College of Special Education, Changchun University, Changchun, 130022, People's Republic of China.
| |
Collapse
|
2
|
Yang W, Li T, An S, Chen R, Zhao Y, Cui J, Zhang M, Lu J, Tian Y, Bao L, Zhao P. Ligilactobacillus salivarius LZZAY01 accelerated autophagy and apoptosis in colon cancer cells and improved gut microbiota in CAC mice. Microbiol Spectr 2025; 13:e0186124. [PMID: 39792005 PMCID: PMC11792455 DOI: 10.1128/spectrum.01861-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Accepted: 12/04/2024] [Indexed: 01/12/2025] Open
Abstract
Colorectal cancer (CRC) is one of the malignant tumors globally, with high morbidity and mortality rates. The mainstay treatment of CRC includes surgery, radiotherapy, and chemotherapy. However, these treatments are associated with a high recurrence rate, poor prognosis, and highly toxic side effects. The probiotics have the potential to prevent CRC, and they display a favorable safety performance. Probiotics could provide a potential strategy to prevent and treat CRC. The impact of LZZAY01 on cancer cell lines CT-26, HCT-116, and SW-620 was evaluated by conducting cytotoxicity and clonogenicity tests. A model of colitis-associated cancer (CAC) was established in C57BL/6j mice following induction with AOM/DSS. The levels of autophagy and apoptosis proteins, tight junction proteins, and inflammatory factors were detected by western blotting, immunofluorescence assay, and enzyme-linked immunosorbent assay. High-throughput sequencing of gut 16S rRNA was performed to analyze the abundance and diversity of the gut microbiome. LZZAY01, a new strain of Ligilactobacillus salivarius, was certified by an evolutionary tree and average nucleotide identity. LZZAY01 enhanced autophagy and apoptosis in CT-26, HCT-116, and SW-620 cell lines. It preserved the integrity of the intestinal barrier by regulating the tight junction protein ZO-1 and claudin-1. The tumor necrosis factor-α and interleukin-6 were reduced by LZZAY01. The abundance and diversity of the intestinal microbiota were enhanced, especially the beneficial bacterial species maintaining the balance of the intestinal flora such as Bifidobacterium and Lactobacillus. L. salivarius LZZAY01 improved CAC via suppressing the growth of colon cancer cells, promoting autophagy and apoptosis, enhancing intestinal tight junctions, reducing intestinal barrier degradation, modifying the gut microbiota abundance, and decreasing inflammatory reactions.IMPORTANCEAlthough similar probiotics have been shown to have anticancer potential in colorectal cancer (CRC), there is a paucity of research related to the preventive function of probiotics against CRC. And there are fewer studies about the mechanism of probiotics' preventive effects on CRC. The regulation of tumor cell proliferation and apoptosis by the active ingredients of probiotics may be one of the mechanisms of their prevention of CRC. In this study, we explored the effects of L. salivarius LZZAY01 on autophagy and apoptosis of colon cancer cells in vitro and in vivo and proposed a possible mechanism for the prevention of CRC by probiotics.
Collapse
Affiliation(s)
- Wenhong Yang
- Laboratory of Microbiology and Immunology, School of Basic Medical Science, Inner Mongolia Medical University, Hohhot, China
| | - Tao Li
- College of Animal Science and Technology, Tarim University, Alar, China
| | - Shixiang An
- Laboratory of Microbiology and Immunology, School of Basic Medical Science, Inner Mongolia Medical University, Hohhot, China
| | - Rong Chen
- College of Animal Science and Technology, Tarim University, Alar, China
| | - Yuxin Zhao
- Department of Anesthesiology, Inner Mongolia Chest Hospital (The Fourth Hospital), Hohhot, China
| | - Jiaxian Cui
- Laboratory of Microbiology and Immunology, School of Basic Medical Science, Inner Mongolia Medical University, Hohhot, China
| | - Mingyu Zhang
- Laboratory of Microbiology and Immunology, School of Basic Medical Science, Inner Mongolia Medical University, Hohhot, China
| | - Jingkun Lu
- Laboratory of Microbiology and Immunology, School of Basic Medical Science, Inner Mongolia Medical University, Hohhot, China
| | - Yunpeng Tian
- Laboratory of Microbiology and Immunology, School of Basic Medical Science, Inner Mongolia Medical University, Hohhot, China
| | - Lili Bao
- Laboratory of Microbiology and Immunology, School of Basic Medical Science, Inner Mongolia Medical University, Hohhot, China
| | - Pengwei Zhao
- Laboratory of Microbiology and Immunology, School of Basic Medical Science, Inner Mongolia Medical University, Hohhot, China
| |
Collapse
|
3
|
Garavaglia B, Vallino L, Ferraresi A, Amoruso A, Pane M, Isidoro C. Probiotic-Derived Metabolites from Lactiplantibacillus plantarum OC01 Reprogram Tumor-Associated Macrophages to an Inflammatory Anti-Tumoral Phenotype: Impact on Colorectal Cancer Cell Proliferation and Migration. Biomedicines 2025; 13:339. [PMID: 40002754 PMCID: PMC11853712 DOI: 10.3390/biomedicines13020339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 01/29/2025] [Accepted: 01/30/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Tumor-associated macrophages (TAMs) are key players in the colorectal cancer (CRC) tumor microenvironment (TME), representing the most abundant immune cells within it. The interplay between the intestinal microbiota, macrophages, and cancer cells significantly impacts tumor progression by driving macrophage polarization. Particularly, the polarization into the pro-tumoral M2-like TAM phenotype promotes the extracellular matrix remodeling, cancer cell proliferation, metastasis, immune suppression, and therapy resistance. Probiotic metabolites can disrupt this crosstalk, possibly reverting the TAM polarization toward a pro-inflammatory anti-tumoral phenotype, thus potentially benefiting the intestinal mucosa and opposing CRC progression. Previously, we showed that Lactiplantibacillus plantarum OC01 metabolites counter interleukin (IL)-6-induced CRC proliferation and migration. Methods: Here, we explore how probiotics affect CRC secretome and how this influences TAM polarization, which then impacts CRC malignancy. Results: The conditioning medium (CM) from CRC cells indeed promoted the polarization of macrophage toward the M2-like phenotype, whereas the CM from CRC pre-treated with L. plantarum OC01 metabolites induced a pro-inflammatory macrophage phenotype, characterized by NLRP3 inflammasome activation and reactive oxygen species (ROS) production, and by decreased expression of the M2 phenotype markers CD206 and CD163. Consistently, the expression of tumor growth factor (TGF)-β, a promoter of M2 macrophage polarization, was reduced in CRC cells treated with L. plantarum OC01. The pro-inflammatory macrophages inhibited CRC proliferation and migration. Conclusions: Overall, our study highlights the potential of metabolites from L. plantarum OC01 to reprogram the metabolism in cancer cells and thus reshape the TME by shifting TAMs toward a more inflammatory and anti-tumoral phenotype, emphasizing the promise of probiotics in advancing novel therapeutic approaches for CRC.
Collapse
Affiliation(s)
- Beatrice Garavaglia
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale, Via P. Solaroli 17, 28100 Novara, Italy; (B.G.); (L.V.); (A.F.)
| | - Letizia Vallino
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale, Via P. Solaroli 17, 28100 Novara, Italy; (B.G.); (L.V.); (A.F.)
| | - Alessandra Ferraresi
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale, Via P. Solaroli 17, 28100 Novara, Italy; (B.G.); (L.V.); (A.F.)
| | - Angela Amoruso
- Probiotical S.p.A., Via E. Mattei, 3, 28100 Novara, Italy; (A.A.); (M.P.)
| | - Marco Pane
- Probiotical S.p.A., Via E. Mattei, 3, 28100 Novara, Italy; (A.A.); (M.P.)
| | - Ciro Isidoro
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale, Via P. Solaroli 17, 28100 Novara, Italy; (B.G.); (L.V.); (A.F.)
| |
Collapse
|
4
|
Shen Y, Gao Y, Yang G, Zhao Z, Zhao Y, Gao L, Li S. Anti-colorectal cancer effect of total minor ginsenosides produced by lactobacilli transformation of major ginsenosides by inducing apoptosis and regulating gut microbiota. Front Pharmacol 2025; 15:1496346. [PMID: 39845805 PMCID: PMC11750747 DOI: 10.3389/fphar.2024.1496346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Accepted: 12/13/2024] [Indexed: 01/24/2025] Open
Abstract
Objective Minor ginsenosides have demonstrated promising anticancer effects in previous reports. Total minor ginsenosides (TMG) were obtained through the fermentation of major ginsenosides with Lactiplantibacillus plantarum, and potential anticancer effects of TMGs on the mouse colon cancer cell line CT26.WT, in vitro and in vivo, were investigated. Materials and Methods We employed the Cell Counting Kit-8 (CCK-8), TdT-mediated dUTP nick end labeling (TUNEL), and Western blot analysis in vitro to explore the anti-proliferative and pro-apoptotic functions of TMG in CT26.WT cells. In vivo, a xenograft model was established by subcutaneously injecting mice with CT26.WT cells and administering a dose of 100 mg/kg/day TMG to the tumor-bearing mice. The level of apoptosis and expression of various proteins in the tumor tissues were detected by immunohistochemistry and Western blot. High-throughput 16S rRNA sequencing was used to determine the alterations in the gut microbiota. Results In vitro studies demonstrated that TMG significantly inhibited proliferation and promoted apoptosis in CT26.WT cells. Interestingly, TMG induced apoptosis in CT26.WT cells by affecting the Bax/Bcl-2/caspase-3 pathway. Furthermore, the result of the transplanted tumor model indicated that TMG substantially enhanced the activities of Bax and caspase-3, reduced the activity of Bcl-2, and suppressed the expression of Raf/MEK/ERK protein levels. Fecal analysis revealed that TMG reconstructed the gut microbiota in colorectal cancer-affected mice by augmenting the abundance of the advantageous bacterium Lactobacillus and decreasing the abundance of the harmful bacterium Proteus. Conclusion TMG can exhibit potent anti-colorectal cancer effects through diverse apoptotic mechanisms, with their mode of action closely related to the regulation of gut microbiota.
Collapse
Affiliation(s)
- Yunjiao Shen
- Institute of Agro-food Technology, Jilin Academy of Agricultural Sciences (Northeast Agricultural Research Center of China), Changchun, China
- School of Chinese Materia Medica, State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yansong Gao
- Institute of Agro-food Technology, Jilin Academy of Agricultural Sciences (Northeast Agricultural Research Center of China), Changchun, China
| | - Ge Yang
- Institute of Agro-food Technology, Jilin Academy of Agricultural Sciences (Northeast Agricultural Research Center of China), Changchun, China
| | - Zijian Zhao
- Institute of Agro-food Technology, Jilin Academy of Agricultural Sciences (Northeast Agricultural Research Center of China), Changchun, China
| | - Yujuan Zhao
- Institute of Agro-food Technology, Jilin Academy of Agricultural Sciences (Northeast Agricultural Research Center of China), Changchun, China
| | - Lei Gao
- Institute of Agro-food Technology, Jilin Academy of Agricultural Sciences (Northeast Agricultural Research Center of China), Changchun, China
| | - Shengyu Li
- Institute of Agro-food Technology, Jilin Academy of Agricultural Sciences (Northeast Agricultural Research Center of China), Changchun, China
| |
Collapse
|
5
|
Dadgar-Zankbar L, Mokhtaryan M, Bafandeh E, Javanmard Z, Asadollahi P, Darbandi T, Afifirad R, Dashtbin S, Darbandi A, Ghanavati R. Microbiome and bladder cancer: the role of probiotics in treatment. Future Microbiol 2025; 20:73-90. [PMID: 39445447 PMCID: PMC11974345 DOI: 10.1080/17460913.2024.2414671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 10/07/2024] [Indexed: 10/25/2024] Open
Abstract
Bladder cancer (BCa) remains a significant global health challenge, with increasing interest in the role of the bladder microbiome in its pathogenesis, progression and treatment outcomes. The complex relationship between bladder cancer and the microbiome, as well as the potential impact of probiotics on treatment effectiveness, is currently under investigation. Research suggests that the microbiota may influence BCa recurrence prevention and enhance the efficacy of the Bacillus Calmette-Guérin (BCG) vaccine. Recent studies reveal differences in the bladder microbiome between individuals without bladder cancer and those with the disease. In the healthy bladder, Streptococcus and Lactobacillus are consistently identified as the most prevalent genera. However, in men, the predominant bacterial genera are Staphylococcus, Corynebacterium and Streptococcus, while in women with bladder cancer, Gardnerella and Lactobacillus are dominant. Probiotics, particularly Lactobacillus spp., can exhibit anti-tumor properties by competing with pathogenic strains involved in carcinogenesis or by producing regulatory substances. They regulate cancer signaling, induce apoptosis, inhibit mutagenic activity, downregulate oncogene expression, induce autophagy, inhibit kinases, reactivate tumor suppressors and prevent metastasis. These mechanisms have shown promising results in both preclinical and some clinical studies.
Collapse
Affiliation(s)
- Leila Dadgar-Zankbar
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Mokhtaryan
- Department of Internal Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Elnaz Bafandeh
- Molecular Microbiology Research Center, Shahed University, Tehran, Iran
| | - Zahra Javanmard
- Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Parisa Asadollahi
- Microbiology Department, Faculty of Medicine, Ilam University of Medical Sciences, Ilam, Iran
| | - Taleih Darbandi
- Department of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Roghayeh Afifirad
- Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Shirin Dashtbin
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Atieh Darbandi
- Molecular Microbiology Research Center, Shahed University, Tehran, Iran
| | | |
Collapse
|
6
|
Stachelska MA, Karpiński P, Kruszewski B. Health-Promoting and Functional Properties of Fermented Milk Beverages with Probiotic Bacteria in the Prevention of Civilization Diseases. Nutrients 2024; 17:9. [PMID: 39796443 PMCID: PMC11722897 DOI: 10.3390/nu17010009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 12/19/2024] [Accepted: 12/23/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND/OBJECTIVES There is scattered information in the scientific literature regarding the characterization of probiotic bacteria found in fermented milk beverages and the beneficial effects of probiotic bacteria on human health. Our objective was to gather the available information on the use of probiotic bacteria in the prevention of civilization diseases, with a special focus on the prevention of obesity, diabetes, and cancer. METHODS We carried out a literature review including the following keywords, either individually or collectively: lactic acid bacteria; probiotic bacteria; obesity; lactose intolerance; diabetes; cancer protection; civilization diseases; intestinal microbiota; intestinal pathogens. RESULTS This review summarizes the current state of knowledge on the use of probiotic bacteria in the prevention of civilization diseases. Probiotic bacteria are a set of living microorganisms that, when administered in adequate amounts, exert a beneficial effect on the health of the host and allow for the renewal of the correct quantitative and qualitative composition of the microbiota. Probiotic bacteria favorably modify the composition of the intestinal microbiota, inhibit the development of intestinal pathogens, prevent constipation, strengthen the immune system, and reduce symptoms of lactose intolerance. As fermented milk beverages are an excellent source of probiotic bacteria, their regular consumption can be a strong point in the prevention of various types of civilization diseases. CONCLUSIONS The presence of lactic acid bacteria, including probiotic bacteria in fermented milk beverages, reduces the incidence of obesity and diabetes and serves as a tool in the prevention of cancer diseases.
Collapse
Affiliation(s)
| | - Piotr Karpiński
- Faculty of Health Sciences, University of Lomza, Akademicka 14, 18-400 Łomża, Poland;
| | - Bartosz Kruszewski
- Department of Food Technology and Assessment, Institute of Food Sciences, Warsaw University of Life Sciences—SGGW, Nowoursynowska 159 C, 02-776 Warsaw, Poland
| |
Collapse
|
7
|
Ismael M, Huang M, Zhong Q. The Bacteriocins Produced by Lactic Acid Bacteria and the Promising Applications in Promoting Gastrointestinal Health. Foods 2024; 13:3887. [PMID: 39682959 DOI: 10.3390/foods13233887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 11/25/2024] [Accepted: 11/29/2024] [Indexed: 12/18/2024] Open
Abstract
Bacteriocins produced by lactic acid bacteria (LAB) are promising bioactive peptides. Intriguingly, bacteriocins have health benefits to the host and may be applied safely in the food industry as bio-preservatives or as therapeutic interventions preventing intestinal diseases. In recent years, finding a safe alternative approach to conventional treatments to promote gut health is a scientific hotspot. Therefore, this review aimed to give insight into the promising applications of LAB-bacteriocins in preventing intestinal diseases, such as colonic cancer, Helicobacter pylori infections, multidrug-resistant infection-associated colitis, viral gastroenteritis, inflammatory bowel disease, and obesity disorders. Moreover, we highlighted the recent research on bacteriocins promoting gastrointestinal health. The review also provided insights into the proposed mechanisms, challenges and opportunities, trends and prospects. In addition, a SWOT analysis was conducted on the potential applications. Based on properties, biosafety, and health functions of LAB-bacteriocins, we conclude that the future applications of LAB-bacteriocins are promising in promoting gastrointestinal health. Further in vivo trials are needed to confirm these potential effects of LAB-bacteriocins interventions.
Collapse
Affiliation(s)
- Mohamedelfatieh Ismael
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Mingxin Huang
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Qingping Zhong
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| |
Collapse
|
8
|
Chen HH, Luo CW, Chen YL, Chiang JY, Huang CR, Wang YT, Chen CH, Guo J, Yip HK. Probiotic-facilitated cytokine-induced killer cells suppress peritoneal carcinomatosis and liver metastasis in colorectal cancer cells. Int J Biol Sci 2024; 20:6162-6180. [PMID: 39664585 PMCID: PMC11628340 DOI: 10.7150/ijbs.101051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 10/19/2024] [Indexed: 12/13/2024] Open
Abstract
Background: This study tested the hypothesis that combined therapy with probiotics and cytokine-induced killer (CIK) cells was superior to merely one on suppressing the peritoneal carcinomatosis and liver metastasis of colorectal cancer (CRC) cells in nude mice. Methods and Results: The in vitro study revealed that in HCT 116/SW620 CRC cell lines, cell viability, proliferation, colony formation, migratory ability, wound healing, and protein expression of PD-L1 and FAK were significantly and comparably suppressed and that apoptosis was significantly and comparably increased by probiotics and CIK cells, and these effects were further significantly enhanced by combined probiotics + CIK cell therapy (all p<0.001). Nude mice were categorized into Groups 1 (SC), 2 (HCT 116), 3 (HCT 116 + probiotics), 4 (HCT 116 + CIK cells), and 5 (HCT 116 + probiotics + CIK cells). CRC cells were intraperitoneally implanted into Groups 2 to 5, and the animals were euthanized by Day 28. The results demonstrated that the abdominal dissemination of CRC cells, tumor numbers, tumor weights, liver weights, liver necrosis areas and the expression of γ-H2AX/PD-L1/FAK in harvested liver tumors were lowest in Group 1, highest in Group 2, and significantly and progressively decreased in Groups 3 to 5 (all p<0.0001). The protein expression levels of apoptotic and DNA damage biomarkers (Bax/c-caspase 3/c-PARP/γ-H2AX), a metastatic biomarker (FAK) and three tumor proliferation and survival signaling biomarkers (JAK-STAT1, PI3K/Akt/m-TOR and Ras/Raf/MEK/ERK) exhibited identical patterns to that of a tumor immune escape biomarker (PD-L1) among the groups (all p<0.0001). Conclusion: The combination of probiotics and CIK cells was superior to either therapy alone in suppressing CRC cell growth, proliferation, liver metastasis and survival, mainly through downregulating cell proliferation and survival signaling pathways.
Collapse
Affiliation(s)
- Hong-Hwa Chen
- Division of Colorectal Surgery, Department of Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
| | - Chi-Wen Luo
- Division of Breast Oncology and Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung City 807, Taiwan
- Department of Cosmetic Science and Institute of Cosmetic Science, Chia Nan University of Pharmacy and Science, Tainan 717, Taiwan
| | - Yi-Ling Chen
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
| | - John Y. Chiang
- Department of Computer Science and Engineering, National Sun Yat-Sen University, Kaohsiung 80424, Taiwan
| | - Chi-Ruei Huang
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
- Center for Shockwave Medicine and Tissue Engineering, Kaohsiung Chang Gung Memorial Hospital Kaohsiung 833401, Taiwan
| | - Yi-Ting Wang
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
| | - Chih-Hung Chen
- Divisions of General Medicine, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
| | - Jun Guo
- Department of Cardiology, The First Affiliated Hospital, Jinan University, Guangzhou 510632, China
| | - Hon-Kan Yip
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
- Center for Shockwave Medicine and Tissue Engineering, Kaohsiung Chang Gung Memorial Hospital Kaohsiung 833401, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 40402, Taiwan
| |
Collapse
|
9
|
Jiang TQ, Wang H, Cheng WX, Xie C. Modulation of host N6-methyladenosine modification by gut microbiota in colorectal cancer. World J Gastroenterol 2024; 30:4175-4193. [PMID: 39493326 PMCID: PMC11525875 DOI: 10.3748/wjg.v30.i38.4175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 08/29/2024] [Accepted: 09/12/2024] [Indexed: 09/29/2024] Open
Abstract
As a research hotspot in the field of molecular biology, N6-methyladenosine (m6A) modification has made progress in the treatment of colorectal cancer (CRC), leukemia and other cancers. Numerous studies have demonstrated that the tumour microenvironment (TME) regulates the level of m6A modification in the host and activates a series of complex epigenetic signalling pathways through interactions with CRC cells, thus affecting the progression and prognosis of CRC. However, with the diversity in the composition of TME factors, this action is reciprocal and complex. Encouragingly, some studies have experimentally revealed that the intestinal flora can alter CRC cell proliferation by directly acting on m6A and thereby altering CRC cell proliferation. This review summarizes the data, supporting the idea that the intestinal flora can influence host m6A levels through pathways such as methyl donor metabolism and thus affect the progression of CRC. We also review the role of m6A modification in the diagnosis, treatment, and prognostic assessment of CRC and discuss the current status, limitations, and potential clinical value of m6A modification in this field. We propose that additional in-depth research on m6A alterations in CRC patients and their TME-related targeted therapeutic issues will lead to better therapeutic outcomes for CRC patients.
Collapse
Affiliation(s)
- Tian-Qi Jiang
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, China
- The First Clinical Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Hao Wang
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, China
- The First Clinical Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Wang-XinJun Cheng
- Queen Mary College, Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Chuan Xie
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, China
| |
Collapse
|
10
|
Peng S, Guo C, Zhang X, Bu X, Li X, Cui H, Duan Z. Kinetic study of the thermal inactivation of Weizmannia coagulans during food thermal processing. Heliyon 2024; 10:e36977. [PMID: 39286079 PMCID: PMC11402908 DOI: 10.1016/j.heliyon.2024.e36977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 08/25/2024] [Accepted: 08/26/2024] [Indexed: 09/19/2024] Open
Abstract
Weizmannia coagulans has attracted attention due to its remarkable health benefits for human, but the dynamic changes of its viable bacteria during thermal processing have been less reported. In this study, a predictive model for the survival of Weissmanella coagulans during thermal processing of food was developed and validated during the processing of coffee, tea, instant noodles, calcium milk biscuits, muffin cake and steamed buns. The kinetics of heat inactivation activities of Weizmannia coagulans VHProbi C08 and Weizmannia coagulans GBI-30, 6086 at 85, 95, 105, 110 and 115 °C were investigated, and their coefficients of determination were greater than 0.91 and 0.87, and the root-mean-square errors were less than 0.64 and 0.43, respectively. The z-values of VHProbi C08 and GBI-30, 6086 were obtained by Bigelow model fitting as 36.1 °C and 36.9 °C, respectively. The developed prediction model was applied to the thermal processing of six food products and the measured values were all within ±0.5 Log10 (CFU/mL) of the predicted values, indicating high prediction accuracy. The model predicts the survival of Weissmanella coagulans simply by obtaining the initial number of viable bacteria and the change in temperature. These suggested that the model can be used as an effective tool to evaluate the stability of Weizmannia coagulans in food thermal processing.
Collapse
Affiliation(s)
- Shudong Peng
- School of Food Science and Engineering, South China University of Technology, Guangzhou, China
| | - Chaoqun Guo
- Qingdao Vland Biotech Inc. Nutrition & Health Technology Center, Qingdao, China
| | - Xiaoyuan Zhang
- School of Food Science and Engineering, South China University of Technology, Guangzhou, China
| | - Xinping Bu
- Qingdao Vland Biotech Inc. Nutrition & Health Technology Center, Qingdao, China
| | - Xinping Li
- Qingdao Vland Biotech Inc. Nutrition & Health Technology Center, Qingdao, China
| | - Hongchang Cui
- Qingdao Vland Biotech Inc. Nutrition & Health Technology Center, Qingdao, China
| | - Zhi Duan
- Qingdao Vland Biotech Inc. Nutrition & Health Technology Center, Qingdao, China
| |
Collapse
|
11
|
Pyo Y, Kwon KH, Jung YJ. Probiotic Functions in Fermented Foods: Anti-Viral, Immunomodulatory, and Anti-Cancer Benefits. Foods 2024; 13:2386. [PMID: 39123577 PMCID: PMC11311591 DOI: 10.3390/foods13152386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 06/30/2024] [Accepted: 07/05/2024] [Indexed: 08/12/2024] Open
Abstract
Fermented foods can provide many benefits to our health. These foods are created by the action of microorganisms and help support our digestive health and immune system. Fermented foods include yogurt, kimchi, pickles, kefir, beer, wine, and more. Fermented foods contain probiotics, lactic acid bacteria (LAB), yeast, organic acids, ethanol, or antimicrobial compounds, which help balance the gut microbiome and improve digestive health. Fermented foods can also benefit your overall health by increasing the diversity of your gut microbiome and reducing inflammation. By routinely consuming fermented foods with these benefits, we can continue to improve our health. Probiotics from fermented foods are beneficial strains of bacteria that are safe for human health and constitute an important component of human health, even for children and the elderly. Probiotics can have a positive impact on your health, especially by helping to balance your gut microbiome and improve digestive health. Probiotics can also boost your immune system and reduce inflammation, which can benefit your overall health. Probiotics, which can be consumed in the diet or in supplement form, are found in many different types of foods and beverages. Research is continuing to investigate the health effects of probiotics and how they can be utilized. The potential mechanisms of probiotics include anti-cancer activity, preventing and treating immune system-related diseases, and slowing the development of Alzheimer's disease and Huntington's disease. This is due to the gut-brain axis of probiotics, which provides a range of health benefits beyond the digestive and gastrointestinal systems. Probiotics reduce tumor necrosis factor-α and interleukins through the nuclear factor-kappa B and mitogen-activated protein kinase pathways. They have been shown to protect against colon cancer and colitis by interfering with the adhesion of harmful bacteria in the gut. This article is based on clinical and review studies identified in the electronic databases PubMed, Web of Science, Embase, and Google Scholar, and a systematic review of clinical studies was performed.
Collapse
Affiliation(s)
- Yeonhee Pyo
- Department of Beauty Cosmetics, College of Biomedical and Health Science, Konkuk University, Chungju 27478, Republic of Korea
| | - Ki Han Kwon
- College of General Education, Kookmin University, Seoul 02707, Republic of Korea;
| | - Yeon Ja Jung
- Department of Beauty Cosmetics, College of Biomedical and Health Science, Konkuk University, Chungju 27478, Republic of Korea
| |
Collapse
|
12
|
Wang Y, Fleishman JS, Li T, Li Y, Ren Z, Chen J, Ding M. Pharmacological therapy of metabolic dysfunction-associated steatotic liver disease-driven hepatocellular carcinoma. Front Pharmacol 2024; 14:1336216. [PMID: 38313077 PMCID: PMC10834746 DOI: 10.3389/fphar.2023.1336216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 12/31/2023] [Indexed: 02/06/2024] Open
Abstract
In light of a global rise in the number of patients with type 2 diabetes mellitus (T2DM) and obesity, non-alcoholic fatty liver disease (NAFLD), now known as metabolic dysfunction-associated fatty liver disease (MAFLD) or metabolic dysfunction-associated steatotic liver disease (MASLD), has become the leading cause of hepatocellular carcinoma (HCC), with the annual occurrence of MASLD-driven HCC expected to increase by 45%-130% by 2030. Although MASLD has become a serious major public health threat globally, the exact molecular mechanisms mediating MASLD-driven HCC remain an open problem, necessitating future investigation. Meanwhile, emerging studies are focusing on the utility of bioactive compounds to halt the progression of MASLD to MASLD-driven HCC. In this review, we first briefly review the recent progress of the possible mechanisms of pathogenesis and progression for MASLD-driven HCC. We then discuss the application of bioactive compounds to mitigate MASLD-driven HCC through different modulatory mechanisms encompassing anti-inflammatory, lipid metabolic, and gut microbial pathways, providing valuable information for future treatment and prevention of MASLD-driven HCC. Nonetheless, clinical research exploring the effectiveness of herbal medicines in the treatment of MASLD-driven HCC is still warranted.
Collapse
Affiliation(s)
- Yumin Wang
- Department of Respiratory and Critical Care Medicine, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, China
| | - Joshua S. Fleishman
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY, United States
| | - Tongda Li
- Department of Traditional Chinese Medicine, Beijing Geriatric Hospital, Beijing, China
| | - Yulin Li
- Department of Respiratory and Critical Care Medicine, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, China
| | - Zhao Ren
- Department of Pharmacy, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, China
| | - Jichao Chen
- Department of Respiratory and Critical Care Medicine, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, China
| | - Mingchao Ding
- Department of Peripheral Vascular Intervention, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, China
| |
Collapse
|