1
|
Chen Y, Fang H, Chen H, Liu X, Zhao J, Stanton C, Ross RP, Chen W, Yang B. Bifidobacterium inhibits the progression of colorectal tumorigenesis in mice through fatty acid isomerization and gut microbiota modulation. Gut Microbes 2025; 17:2464945. [PMID: 39924893 PMCID: PMC11812354 DOI: 10.1080/19490976.2025.2464945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 01/14/2025] [Accepted: 01/21/2025] [Indexed: 02/11/2025] Open
Abstract
Colorectal cancer (CRC) represents the third most common cancer worldwide. Consequently, there is an urgent need to identify novel preventive and therapeutic strategies for CRC. This study aimed to screen for beneficial bacteria that have a preventive effect on CRC and to elucidate the potential mechanisms. Initially, we compared gut bacteria and bacterial metabolites of healthy volunteers and CRC patients, which demonstrated that intestinal conjugated linoleic acid (CLA), butyric acid, and Bifidobacterium in CRC patients were significantly lower than those in healthy volunteers, and these indicators were significantly negatively correlated with CRC. Next, spontaneous CRC mouse model were conducted to explore the effect of supplemental CLA-producing Bifidobacterium on CRC. Supplementation of mice with CLA-producing Bifidobacterium breve CCFM683 and B. pseudocatenulatum MY40C significantly prevented CRC. Moreover, molecular approaches demonstrated that CLA and the CLA-producing gene, bbi, were the key metabolites and genes for CCFM683 to prevent CRC. Inhibitor intervention results showed that PPAR-γ was the key receptor for preventing CRC. CCFM683 inhibited the NF-κB signaling pathway, up-regulated MUC2, Claudin-1, and ZO-1, and promoted tumor cell apoptosis via the CLA-PPAR-γ axis. Additionally, fecal microbiota transplantation (FMT) and metagenomic analysis showed that CCFM683 up-regulated Odoribacter splanchnicus through CLA production, which then prevented CRC by producing butyric acid, up-regulating TJ proteins, regulating cytokines, and regulating gut microbiota. These results will contribute to the clinical trials of Bifidobacterium and the theoretical research and development of CRC dietary products.
Collapse
Affiliation(s)
- Yang Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Huiting Fang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Haiqin Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Xiaoming Liu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Catherine Stanton
- International Joint Research Laboratory for Maternal-Infant Microbiota and Health, Jiangnan University, Wuxi, Jiangsu, China
- Teagasc Food Research Centre, Moorepark, Fermoy, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - R. Paul Ross
- International Joint Research Laboratory for Maternal-Infant Microbiota and Health, Jiangnan University, Wuxi, Jiangsu, China
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Wei Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Bo Yang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- International Joint Research Laboratory for Maternal-Infant Microbiota and Health, Jiangnan University, Wuxi, Jiangsu, China
| |
Collapse
|
2
|
Fan F, Guo R, Pan K, Xu H, Chu X. Mucus and mucin: changes in the mucus barrier in disease states. Tissue Barriers 2025:2499752. [PMID: 40338015 DOI: 10.1080/21688370.2025.2499752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 04/16/2025] [Accepted: 04/21/2025] [Indexed: 05/09/2025] Open
Abstract
In this review we discuss mucus, the viscoelastic secretion from goblet or mucous producing cells that covers and protects all non-keratinized wet epithelial surfaces. In addition to the surface of organs directly contacting with the external environment such as the eyes, this layer provides protection to the underlying gastrointestinal, respiratory and female reproductive tracts by trapping pathogens, irritants, environmental fine particles and potentially harmful foreign substances. Mucins, the primary structural components of mucus, form structurally different mucus layers at different sites in a process regulated by a variety of factors. Currently, more and more studies have shown that the mucus barrier is not only closely related to various intestinal mucus diseases, but also involved in the occurrence and development of various airway diseases and mucus-related diseases, thus it may become a new target for the treatment of various related diseases in the future. Since the dysfunction of the mucous layer is closely related to various pathological processes, in-depth understanding of its molecular mechanism and physiological role is of great theoretical and practical significance for disease prevention and treatment. Here, we discuss different aspects of the mucus layer by focusing on its chemical composition, synthetic pathways, and some of the characteristics of the mucus layer in physiological and pathological situations.
Collapse
Affiliation(s)
- Fangfang Fan
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Ruihan Guo
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Kun Pan
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Hongye Xu
- Quality Assurance department, Tongling Institutes for Food and Drug Control, Tongling, China
| | - Xiaoqin Chu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Hefei, China
- Engineering Technology Research Center of Modern Pharmaceutical Preparation, Hefei, Anhui Province, China
| |
Collapse
|
3
|
He F, Zheng Y, Elsabagh M, Fan K, Zha X, Zhang B, Wang M, Zhang H. Gut microbiota modulate intestinal inflammation by endoplasmic reticulum stress-autophagy-cell death signaling axis. J Anim Sci Biotechnol 2025; 16:63. [PMID: 40312439 PMCID: PMC12046778 DOI: 10.1186/s40104-025-01196-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 03/17/2025] [Indexed: 05/03/2025] Open
Abstract
The intestinal tract, a complex organ responsible for nutrient absorption and digestion, relies heavily on a balanced gut microbiome to maintain its integrity. Disruptions to this delicate microbial ecosystem can lead to intestinal inflammation, a hallmark of inflammatory bowel disease (IBD). While the role of the gut microbiome in IBD is increasingly recognized, the underlying mechanisms, particularly those involving endoplasmic reticulum (ER) stress, autophagy, and cell death, remain incompletely understood. ER stress, a cellular response to various stressors, can trigger inflammation and cell death. Autophagy, a cellular degradation process, can either alleviate or exacerbate ER stress-induced inflammation, depending on the specific context. The gut microbiome can influence both ER stress and autophagy pathways, further complicating the interplay between these processes. This review delves into the intricate relationship between ER stress, autophagy, and the gut microbiome in the context of intestinal inflammation. By exploring the molecular mechanisms underlying these interactions, we aim to provide a comprehensive theoretical framework for developing novel therapeutic strategies for IBD. A deeper understanding of the ER stress-autophagy axis, the gut microbial-ER stress axis, and the gut microbial-autophagy axis may pave the way for targeted interventions to restore intestinal health and mitigate the impact of IBD.
Collapse
Affiliation(s)
- Feiyang He
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, P. R. China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, 225009, P. R. China
- Key Laboratory of Fujian Universities Preventive Veterinary Medicine and Biotechnology, Longyan University, Longyan, 364012, P. R. China
| | - Yi Zheng
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, P. R. China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, 225009, P. R. China
| | - Mabrouk Elsabagh
- Department of Animal Production and Technology, Faculty of Agricultural Sciences and Technologies, Niğde Ömermer Halisdemir University, Nigde, 51240, Turkey
| | - Kewei Fan
- Key Laboratory of Fujian Universities Preventive Veterinary Medicine and Biotechnology, Longyan University, Longyan, 364012, P. R. China
| | - Xia Zha
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, P. R. China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, 225009, P. R. China
| | - Bei Zhang
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, P. R. China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, 225009, P. R. China
| | - Mengzhi Wang
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, P. R. China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, 225009, P. R. China
- State Key Laboratory of Sheep Genetic Improvement and Healthy Production, Xinjiang Academy of Agricultural Reclamation Science, Shihezi, 832000, P. R. China
| | - Hao Zhang
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, P. R. China.
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, 225009, P. R. China.
| |
Collapse
|
4
|
Jin S, Wang H, Gong H, Guo L, Zhang H, Zhang J, Chang Q, Li J, Zhang R, Bao J. Music intervention mitigates LPS-induced gut barrier disruption and immune stress in broilers via TLR4/NF-κB regulation. Poult Sci 2025; 104:105189. [PMID: 40294553 PMCID: PMC12059385 DOI: 10.1016/j.psj.2025.105189] [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: 11/11/2024] [Revised: 04/02/2025] [Accepted: 04/18/2025] [Indexed: 04/30/2025] Open
Abstract
Immune stress induced by harsh environment in intensive farming can impair broiler intestinal health. Although music as an environmental intervention can alleviate short-term stress injury, its long-term regulatory mechanism on intestinal inflammation has not been clarified. In this study, we investigated the effects of a music-enriched environment on growth performance, intestinal barrier function, and inflammatory responses in lipopolysaccharide (LPS)-induced immunostressed broilers. AA broilers were randomly divided into four groups: control group (CON), music-enriched environment group (MUC), LPS-induced immune stress group (LPS) and music-enriched environment + LPS group (MUC+LPS). On the 14th, 16th and 18th days, the LPS and MUC+LPS groups were injected intraperitoneally with 500 μg of LPS to construct an immune stress model, and the CON and MUC groups were injected with an equal amount of saline. On day 28, the birds were sacrificed to detect the indicators associated with intestinal barrier and inflammation. The LPS group showed a significant decrease in performance from 14 to 28 days, with elevated serum levels of CORT, ACTH, DAO, and d-LA, and a decrease in the activity of intestinal mucosal SOD/GSH-Px, and impaired gut morphology. impaired; music remission significantly alleviated the decline in production performance, reduced the levels of stress hormones and markers of intestinal barrier damage, while elevating jejuno-ileal GSH-Px activity and improving intestinal morphology. Significant inflammatory gene expression characteristics were observed in jejunum and ileum tissues after LPS injection: upregulation of TLR4, NF-κB, TNF-α, IL-1β, and IL-6, and significant suppression of jejunal IL-10 expression. Notably, IL-10 and IFN-γ expression in the ileum did not show statistical differences. Inflammation-related gene expression showed an overall down-regulation trend after the music intervention, but was still significantly different from the control group. Music intervention on the regulation of jejunal MYD88 and ileal TNF-α - the LPS group did not show statistically significant differences in the expression of these two key inflammatory nodes with the LPS+MUS group. Mechanistic studies have shown that LPS triggers an oxidative stress cascade through activation of the TLR4/NF-κB signaling axis, leading to disruption of intestinal barrier integrity. In contrast, music exposure exerts a protective effect through a dual mechanism: on the one hand, it helps to enhance the expression of the tight junction protein ZO-1/Occludin to repair the physical barrier; on the other hand, it inhibits the activation of the TLR4/NF-κB pathway, which can effectively alleviate LPS-induced immunopathological damage.
Collapse
Affiliation(s)
- Shengzi Jin
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, PR China
| | - Haowen Wang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, PR China
| | - Haiyue Gong
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, PR China
| | - Lu Guo
- Department of Basic Medical Sciences, Heilongjiang Nursing College, Harbin, Heilongjiang 150086, China
| | - Haoran Zhang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, PR China
| | - Jiaqi Zhang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, PR China
| | - Qingqing Chang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, PR China
| | - Jianhong Li
- College of Life Science, Northeast Agricultural University, Harbin 150030, PR China
| | - Runxiang Zhang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, PR China.
| | - Jun Bao
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, PR China
| |
Collapse
|
5
|
Mercado GAG, Fang TJ, Lee LY, Santos MKD. Laryngeal mucinous adenocarcinoma in a 45-year-old woman: a case report. Ann Med Surg (Lond) 2025; 87:2362-2366. [PMID: 40212139 PMCID: PMC11981391 DOI: 10.1097/ms9.0000000000002942] [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: 08/27/2024] [Accepted: 01/05/2025] [Indexed: 04/13/2025] Open
Abstract
Introduction and importance This paper emphasizes on an infrequent case of primary laryngeal mucinous adenocarcinoma. Despite partial laryngectomies being a traditional surgical technique, it is not frequently used in the treatment of laryngeal lesions. We would like to highlight the efficacy of open partial laryngectomy as a viable treatment option in cases of nonsquamous cell carcinoma of the larynx. The use of traditional and modern surgical techniques can aid in the preservation of voice and swallowing functions. This case report has been reported in line with the SCARE Criteria [Sohrabi C, Mathew G, Maria N, Kerwan A, Franchi T, Agha RA. The SCARE 2023 guideline: updating consensus Surgical CAse REport (SCARE) guidelines. Int J Surg Lond Engl. 2023;109(5):1136]. Presentation of the case This paper focuses on a 45-year-old female who consulted at a tertiary private hospital. She had no known medical comorbidities or family history of cancer who complained on persistent globus sensation and frequent throat clearing. Her symptoms subjectively were similar to acid reflux but on flexible endoscopy a laryngeal lesion was seen. The initial impression with the additional information gathered from a neck CT scan was that of a cystic lesion. During surgery, no cystic component was seen; tissue samples were sent for biopsy. The biopsy results were signed out as a case of mucinous adenocarcinoma. Given the age and medical status of the patient, an organ preservation surgery was done. Post operatively the patient was able to maintain all swallowing and phonatory capabilities. Clinical discussion There are currently two reported cases of primary laryngeal mucinous adenocarcinoma. Both cases were located in the supraglottic region presenting with dysphonia and dysphagia. Due to the limited number of cases, there is currently no standardized surgical or medical treatment for the management primary laryngeal mucinous adenocarcinomas. Conclusion Mucinous adenocarcinoma of the larynx is a rare disease usually found as distant metastatic lesions. Primary laryngeal mucinous adenocarcinoma is even more uncommon. To balance the oncological outcomes and life quality is important. We reported a primary laryngeal mucinous laryngeal adenocarcinoma after combined transoral transcervical partial laryngectomy that with good functional and oncological outcomes.
Collapse
Affiliation(s)
- Gabrielle Angela G. Mercado
- Department of Otolaryngology Head and Neck Surgery, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Department of Otolaryngology Head and Neck Surgery, St. Luke’s Medical Center, Manila, Philippines
| | - Tuan-Jen Fang
- Department of Otolaryngology Head and Neck Surgery, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Li-Yu Lee
- Department of Pathology, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Marissa Krizelda D. Santos
- Department of Pathology, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Department of Pathology, Chinese General Hospital and Medical Center, Manila, Philippines
| |
Collapse
|
6
|
Zhu Z, Hua Y, Wu J, Mei J. Elevated Levels of MUC and JADE1 Predict Poor Prognosis of Patients with Gastric Cancer. Cancer Manag Res 2025; 17:577-587. [PMID: 40098804 PMCID: PMC11912898 DOI: 10.2147/cmar.s493015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 02/08/2025] [Indexed: 03/19/2025] Open
Abstract
Objective This study aimed to investigate the relationship between the expression of mucin (MUC) and JADE family PHD finger factor 1 (JADE1) and Helicobacter pylori (HP) infection as well as depth of tumor invasion in gastric cancer. Methods According to the results of immunohistochemical staining, 132 gastric cancer patients diagnosed and treated in our hospital from March 2018 to May 2019 were divided into MUC2 negative group (n=43), MUC2 positive group (n=89), JADE1 negative group (n=36) and JADE1 positive group (n=96). The relationship between MUC2 and JADE1 expression and clinicopathological features of gastric cancer was analyzed. The diagnostic value of MUC2 and JADE1 alone or in combination in gastric cancer was analyzed using ROC curve. Results The MUC2 and JADE1 expressions in gastric cancer tissues was increased (P<0.05). MUC2 and JADE1 expressions were related to different tumor size, differentiation degree, HP infection, lymph node metastasis, depth of tumor invasion and Lauren classification (P<0.05). Kaplan-Meier survival analysis showed that the survival rate of patients with negative expression of MUC2 and JADE1 was significantly lower than that of patients with positive expression of MUC2 and JADE1 (P<0.05). The area under the curve, sensitivity and specificity of MUC2 alone, JADE1 alone and the two combined in detection of gastric cancer was 0.774, 72.46% and 80.03%, 0.796, 82.14% and 76.48%, and 0.918, 91.34% and 89.57%, respectively. Conclusion The expressions of MUC2 and JADE1 in gastric cancer tissues were significantly increased, and their expressions were associated with tumor size, differentiation degree, HP infection, lymph node metastasis, depth of tumor infiltration, Lauren's staging. The combined detection of the two has a high value in the diagnosis of gastric cancer. Analysis of the relationship between MUC2 and JADE1 expression and HP infection is helpful for clinical medical staff to effectively evaluate the condition of patients.
Collapse
Affiliation(s)
- Zhaowei Zhu
- Graduate School, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, People’s Republic of China
- Department of General Surgery, The General Surgery of Lanxi People’s Hospital, Jinhua, Zhejiang, 321100, People’s Republic of China
| | - Yanming Hua
- Department of General Surgery, The General Surgery of Lanxi People’s Hospital, Jinhua, Zhejiang, 321100, People’s Republic of China
| | - Jianta Wu
- Department of General Surgery, The General Surgery of Lanxi People’s Hospital, Jinhua, Zhejiang, 321100, People’s Republic of China
| | - Jianfeng Mei
- Department of General Surgery, The General Surgery of Lanxi People’s Hospital, Jinhua, Zhejiang, 321100, People’s Republic of China
| |
Collapse
|
7
|
Mou R, Cui XY, Luo YS, Cheng Y, Luo QY, Zhang ZF, Wu WL, Li JF, Zhang K. Adult Hymenolepis nana and its excretory-secretory products elicit mouse immune responses via tuft/IL-13 and FOXM1 signaling pathways. Parasit Vectors 2025; 18:100. [PMID: 40069907 PMCID: PMC11899370 DOI: 10.1186/s13071-025-06719-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 02/09/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Hosts typically elicit diverse immune responses to the infection of various parasitic worms, with intestinal epithelial cells playing pivotal roles in detecting parasite invasion. Hymenolepis nana (H. nana) is a zoonotic parasitic worm that resides in the host's intestine. The contribution and underlying mechanisms of tuft cell-mediated immune reactions against H. nana remain unexplored. METHODS This study endeavors to examine the immune responses in the mouse intestine elicited by the adult H. nana and its excretory-secretory products (ESP). Ileal tissue alteration was detected using hematoxylin and eosin (H&E) staining, changes in the number of intestinal stem cells, goblet cells, tuft cells, and Paneth cells were detected by immunohistochemistry (IHC), immunofluorescence (IF), etc., and changes in the expression of type 2 cytokines and FOXM1 were detected by Western blotting (WB) or real-time quantitative polymerase chain reaction (RT-qPCR). RESULTS The presence of adult H. nana and its ESP enhanced the number of tuft cells and goblet cells while fostering the production of type 2 cytokines. Furthermore, the surge in Paneth cells and FOXM1 triggered by H. nana aids in maintaining intestinal stem cells homeostasis and proliferation. Notably, the FOXM1 inhibitor RCM-1 dampened intestinal stem cells differentiation and type 2 cytokines secretion, potentially impeding the host's capacity to eliminate H. nana. CONCLUSIONS The adult H. nana and its ESP stimulate the immune responses in mice through tuft/interleukin (IL)-13 and FOXM1 signaling pathways and promote the elimination of H. nana from the host through the differentiation of intestinal stem cells into tuft cells, goblet cells, and Paneth cells, as well as the activation of type 2 immune responses. Meanwhile, RCM-1 inhibits the immune responses to H. nana in mice, thus affecting the excretion of H. nana by host.
Collapse
Affiliation(s)
- Rong Mou
- The Guizhou Key Laboratory of Microbio and Infectious Disease Prevention & Control/The Key and Characteristic Laboratory of Modern Pathogenicity Biology, Departments of Parasitology & Histology and Embryology, School of Basic Medical Sciences, Guizhou Medical University, Room 220, E-1 Building, Ankang Avenue No. 6, Guiyang, 561113, China
| | - Xuan-Yin Cui
- The Guizhou Key Laboratory of Microbio and Infectious Disease Prevention & Control/The Key and Characteristic Laboratory of Modern Pathogenicity Biology, Departments of Parasitology & Histology and Embryology, School of Basic Medical Sciences, Guizhou Medical University, Room 220, E-1 Building, Ankang Avenue No. 6, Guiyang, 561113, China
| | - Yu-Si Luo
- Emergency ICU, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
- Department of Emergency, Liupanshui Hospital of the Affiliated Hospital of Guizhou Medical University, Liupanshui, 553000, China
| | - Yi Cheng
- The Guizhou Key Laboratory of Microbio and Infectious Disease Prevention & Control/The Key and Characteristic Laboratory of Modern Pathogenicity Biology, Departments of Parasitology & Histology and Embryology, School of Basic Medical Sciences, Guizhou Medical University, Room 220, E-1 Building, Ankang Avenue No. 6, Guiyang, 561113, China
| | - Qing-Yuan Luo
- Center for Tissue Engineering and Stem Cell Research, Guizhou Medical University, Guiyang, 561113, China
| | - Zhen-Fen Zhang
- The Guizhou Key Laboratory of Microbio and Infectious Disease Prevention & Control/The Key and Characteristic Laboratory of Modern Pathogenicity Biology, Departments of Parasitology & Histology and Embryology, School of Basic Medical Sciences, Guizhou Medical University, Room 220, E-1 Building, Ankang Avenue No. 6, Guiyang, 561113, China
| | - Wen-Lan Wu
- The Guizhou Key Laboratory of Microbio and Infectious Disease Prevention & Control/The Key and Characteristic Laboratory of Modern Pathogenicity Biology, Departments of Parasitology & Histology and Embryology, School of Basic Medical Sciences, Guizhou Medical University, Room 220, E-1 Building, Ankang Avenue No. 6, Guiyang, 561113, China
| | - Jin-Fu Li
- The Guizhou Key Laboratory of Microbio and Infectious Disease Prevention & Control/The Key and Characteristic Laboratory of Modern Pathogenicity Biology, Departments of Parasitology & Histology and Embryology, School of Basic Medical Sciences, Guizhou Medical University, Room 220, E-1 Building, Ankang Avenue No. 6, Guiyang, 561113, China
| | - Ke Zhang
- The Guizhou Key Laboratory of Microbio and Infectious Disease Prevention & Control/The Key and Characteristic Laboratory of Modern Pathogenicity Biology, Departments of Parasitology & Histology and Embryology, School of Basic Medical Sciences, Guizhou Medical University, Room 220, E-1 Building, Ankang Avenue No. 6, Guiyang, 561113, China.
| |
Collapse
|
8
|
Li S, Wu T, Wu J, Chen W, Zhang D. Recognizing the biological barriers and pathophysiological characteristics of the gastrointestinal tract for the design and application of nanotherapeutics. Drug Deliv 2024; 31:2415580. [PMID: 39404464 PMCID: PMC11485891 DOI: 10.1080/10717544.2024.2415580] [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: 05/07/2024] [Revised: 10/04/2024] [Accepted: 10/07/2024] [Indexed: 10/19/2024] Open
Abstract
The gastrointestinal tract (GIT) is an important and complex system by which humans to digest food and absorb nutrients. The GIT is vulnerable to diseases, which may led to discomfort or even death in humans. Therapeutics for GIT disease treatment face multiple biological barriers, which significantly decrease the efficacy of therapeutics. Recognizing the biological barriers and pathophysiological characteristics of GIT may be helpful to design innovative therapeutics. Nanotherapeutics, which have special targeting and controlled therapeutic release profiles, have been widely used for the treatment of GIT diseases. Herein, we provide a comprehensive review of the biological barrier and pathophysiological characteristics of GIT, which may aid in the design of promising nanotherapeutics for GIT disease treatment. Furthermore, several typical diseases of the upper and lower digestive tracts, such as Helicobacter pylori infection and inflammatory bowel disease, were selected to investigate the application of nanotherapeutics for GIT disease treatment.
Collapse
Affiliation(s)
- Shan Li
- Department of Chemistry, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing, China
- Department of Gastroenterology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
- Army 953 Hospital, Shigatse Branch of Xinqiao Hospital, Army Medical University (Third Military Medical University), Shigatse, Tibet Autonomous Region, China
| | - Tianyu Wu
- Department of Gastroenterology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Jingfeng Wu
- Department of Gastroenterology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Wensheng Chen
- Department of Gastroenterology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Dinglin Zhang
- Department of Chemistry, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing, China
| |
Collapse
|
9
|
Chen Y, Ma W, Zhao J, Stanton C, Ross RP, Zhang H, Chen W, Yang B. Lactobacillus plantarum Ameliorates Colorectal Cancer by Ameliorating the Intestinal Barrier through the CLA-PPAR-γ Axis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:19766-19785. [PMID: 39186442 DOI: 10.1021/acs.jafc.4c02824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/28/2024]
Abstract
Colorectal cancer (CRC) is the third-largest cancer worldwide. Lactobacillus can regulate the intestinal barrier and gut microbiota. However, the mechanisms of Lactobacillus that alleviate CRC remained unknown. This study aimed to explore the regulatory effect of Lactobacillus plantarum on CRC and its potential mechanism. CCFM8661 treatment significantly ameliorated CRC compared with phosphate-buffered solution (PBS) treatment in ApcMin/+ mice. In addition, conjugated linoleic acid (CLA) was proved to be the key metabolite for CCFM8661 in ameliorating CRC by molecular biology techniques. Peroxisome proliferator-activated receptor γ (PPAR-γ) was proved to be the key receptor in ameliorating CRC by inhibitor intervention experiments. Moreover, supplementation with CCFM8661 ameliorated CRC by producing CLA to inhibit NF-κB pathway and pro-inflammatory cytokines, up-regulate ZO-1, Claudin-1, and MUC2, and promote tumor cell apoptosis in a PPAR-γ-dependent manner. Metagenomic analysis showed that CCFM8661 treatment significantly increased Odoribacter splanchnicus, which could ameliorate CRC by repairing the intestinal barrier. Clinical results showed that intestinal CLA, butyric acid, PPAR-γ, and Lactobacillus were significantly decreased in CRC patients, and these indicators were significantly negatively correlated with CRC. CCFM8661 alleviated CRC by ameliorating the intestinal barrier through the CLA-PPAR-γ axis. These results will promote the development of dietary probiotic supplements for CRC.
Collapse
Affiliation(s)
- Yang Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, China
- Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Hubei University of Technology, Wuhan 430068, Hubei, China
| | - Weiwei Ma
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin 150000, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, China
| | - Catherine Stanton
- International Joint Research Laboratory for Maternal-Infant Microbiota and Health, Jiangnan University, Wuxi 214122, Jiangsu, China
- Teagasc Food Research Centre, Moorepark, Fermoy, Cork P61 C996, Ireland
- APC Microbiome Ireland, University College Cork, Cork T12 K8AF, Ireland
| | - R Paul Ross
- International Joint Research Laboratory for Maternal-Infant Microbiota and Health, Jiangnan University, Wuxi 214122, Jiangsu, China
- Teagasc Food Research Centre, Moorepark, Fermoy, Cork P61 C996, Ireland
- APC Microbiome Ireland, University College Cork, Cork T12 K8AF, Ireland
| | - Hao Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, Jiangsu, China
| | - Wei Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, Jiangsu, China
| | - Bo Yang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, China
- International Joint Research Laboratory for Maternal-Infant Microbiota and Health, Jiangnan University, Wuxi 214122, Jiangsu, China
| |
Collapse
|
10
|
Pang X, Zhou B, Wu J, Mo Q, Yang L, Liu T, Jin G, Zhang L, Liu X, Xu X, Wang B, Cao H. Lacticaseibacillus rhamnosus GG alleviates sleep deprivation-induced intestinal barrier dysfunction and neuroinflammation in mice. Food Funct 2024; 15:8740-8758. [PMID: 39101469 DOI: 10.1039/d4fo00244j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/06/2024]
Abstract
Consuming probiotic products is a solution that people are willing to choose to augment health. As a global health hazard, sleep deprivation (SD) can cause both physical and mental diseases. The present study investigated the protective effects of Lacticaseibacillus rhamnosus GG (LGG), a widely used probiotic, on a SD mouse model. Here, it has been shown that SD induced intestinal damage in mice, while LGG supplementation attenuated disruption of the intestinal barrier and enhanced the antioxidant capacity. Microbiome analysis revealed that SD caused dysbiosis in the gut microbiota, characterized by increased levels of Clostridium XlVa, Alistipes, and Desulfovibrio, as well as decreased levels of Ruminococcus, which were partially ameliorated by LGG. Moreover, SD resulted in elevated pro-inflammatory cytokine concentrations in both the intestine and the brain, while LGG provided protection in both organs. LGG supplementation significantly improved locomotor activity in SD mice. Although heat-killed LGG showed some protective effects in SD mice, its overall efficacy was inferior to that of live LGG. In terms of mechanism, it was found that AG1478, an inhibitor of the epidermal growth factor receptor (EGFR) tyrosine kinase, could diminish the protective effects of LGG. In conclusion, LGG demonstrated the ability to alleviate SD-induced intestinal barrier dysfunction through EGFR activation and alleviate neuroinflammation.
Collapse
Affiliation(s)
- Xiaoqi Pang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Anshan Road No. 154, Heping District, Tianjin, 300052, China.
| | - Bingqian Zhou
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Anshan Road No. 154, Heping District, Tianjin, 300052, China.
| | - Jingyi Wu
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Anshan Road No. 154, Heping District, Tianjin, 300052, China.
| | - Qi Mo
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Anshan Road No. 154, Heping District, Tianjin, 300052, China.
| | - Lijiao Yang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Anshan Road No. 154, Heping District, Tianjin, 300052, China.
| | - Tiaotiao Liu
- School of Biomedical Engineering and Technology, Tianjin Medical University, Tianjin, 300070, China
| | - Ge Jin
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Anshan Road No. 154, Heping District, Tianjin, 300052, China.
| | - Lan Zhang
- Department of Geriatrics, General Hospital, Tianjin Medical University, Tianjin, 300052, China
| | - Xiang Liu
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Anshan Road No. 154, Heping District, Tianjin, 300052, China.
| | - Xin Xu
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Anshan Road No. 154, Heping District, Tianjin, 300052, China.
| | - Bangmao Wang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Anshan Road No. 154, Heping District, Tianjin, 300052, China.
| | - Hailong Cao
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Anshan Road No. 154, Heping District, Tianjin, 300052, China.
| |
Collapse
|
11
|
Chen Z, Chen J, Lin D, Kang H, Luo Y, Wang X, Wang L, Liu D. Forming Single-Cell-Derived Colon Cancer Organoid Arrays on a Microfluidic Chip for High Throughput Tumor Heterogeneity Analysis. ACS Biomater Sci Eng 2024; 10:5265-5273. [PMID: 39087916 DOI: 10.1021/acsbiomaterials.4c00727] [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] [Indexed: 08/02/2024]
Abstract
Single-cell-derived tumor organoids (STOs) possess a distinct genetic background, making them valuable tools for demonstrating tumor heterogeneity. In order to fulfill the high throughput demands of STO assays, we have developed a microfluidic chip containing 30 000 microwells, which is dedicated to a single cell culture approach for selective expansion and differential induction of cancer stem cells. The microwells are coated with a hydrophilic copolymer to eliminate cell adhesion, and the cell culture is supported by poly(ethylene glycol) (PEG) to establish a nonadhesive culture environment. By utilizing an input cell density of 7 × 103·mL-1, it is possible to construct a 4000 single cell culture system through stochastic cell occupation. We demonstrate that the addition of 15% PEG10000 in the cell culture medium effectively prevents cell loss while facilitating tumor stem cell expansion. As were demonstrated by HCT116, HT29, and SW480 colon cancer cells, the microfluidic approach achieved a STO formation rate of ∼20%, resulting in over 800 STOs generated from a single culture. Comprehensive analysis through histomorphology, immunohistochemistry, drug response evaluation, assessment of cell invasion, and biomarker detection reveals the heterogeneity among individual STOs. Specifically, the smaller STOs exhibited higher invasion and drug resistance capabilities compared with the larger ones. The developed microfluidic approach effectively facilitates STO formation and offers promising prospects for investigating tumor heterogeneity, as well as conducting personalized therapy-focused drug screening.
Collapse
Affiliation(s)
- Zihe Chen
- Department of Laboratory Medicine, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, China
| | - Jueming Chen
- Department of Laboratory Medicine, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, China
| | - Dongguo Lin
- Department of Laboratory Medicine, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, China
- Guangdong Engineering Technology Research Center of Microfluidic Chip Medical Diagnosis, Guangzhou 510180, China
| | - Hui Kang
- Department of Pathology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Yanzhang Luo
- Department of Laboratory Medicine, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, China
| | - Xiaogang Wang
- Department of Laboratory Medicine, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, China
| | - Lihui Wang
- Department of Pathology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Dayu Liu
- Department of Laboratory Medicine, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, China
- Guangdong Engineering Technology Research Center of Microfluidic Chip Medical Diagnosis, Guangzhou 510180, China
| |
Collapse
|
12
|
Chen G, Sun H, Chen Y, Wang L, Song O, Zhang J, Li D, Liu X, Feng L. Perineural Invasion in Cervical Cancer: A Hidden Trail for Metastasis. Diagnostics (Basel) 2024; 14:1517. [PMID: 39061654 PMCID: PMC11275432 DOI: 10.3390/diagnostics14141517] [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: 05/06/2024] [Revised: 06/20/2024] [Accepted: 06/21/2024] [Indexed: 07/28/2024] Open
Abstract
Perineural invasion (PNI), the neoplastic invasion of nerves, is an often overlooked pathological phenomenon in cervical cancer that is associated with poor clinical outcomes. The occurrence of PNI in cervical cancer patients has limited the promotion of Type C1 surgery. Preoperative prediction of the PNI can help identify suitable patients for Type C1 surgery. However, there is a lack of appropriate preoperative diagnostic methods for PNI, and its pathogenesis remains largely unknown. Here, we dissect the neural innervation of the cervix, analyze the molecular mechanisms underlying the occurrence of PNI, and explore suitable preoperative diagnostic methods for PNI to advance the identification and treatment of this ominous cancer phenotype.
Collapse
Affiliation(s)
- Guoqiang Chen
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
- Department of Gynecology, The People’s Hospital of Baoan Shenzhen, The Second Affiliated Hospital of Shenzhen University, Shenzhen 518101, China
| | - Hao Sun
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Yunxia Chen
- Department of Gynecology, The People’s Hospital of Baoan Shenzhen, The Second Affiliated Hospital of Shenzhen University, Shenzhen 518101, China
| | - Li Wang
- Department of Gynecology, The People’s Hospital of Baoan Shenzhen, The Second Affiliated Hospital of Shenzhen University, Shenzhen 518101, China
| | - Ouyi Song
- Department of Gynecology, The People’s Hospital of Baoan Shenzhen, The Second Affiliated Hospital of Shenzhen University, Shenzhen 518101, China
| | - Jili Zhang
- Department of Gynecology, The People’s Hospital of Baoan Shenzhen, The Second Affiliated Hospital of Shenzhen University, Shenzhen 518101, China
| | - Dazhi Li
- Department of Gynecology, The People’s Hospital of Baoan Shenzhen, The Second Affiliated Hospital of Shenzhen University, Shenzhen 518101, China
| | - Xiaojun Liu
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Lixia Feng
- Department of Gynecology, The People’s Hospital of Baoan Shenzhen, The Second Affiliated Hospital of Shenzhen University, Shenzhen 518101, China
| |
Collapse
|
13
|
Yang J, Xiao Y, Zhao N, Pei G, Sun Y, Sun X, Yu K, Miao C, Liu R, Lv J, Chu H, Zhou L, Wang B, Yao Z, Wang Q. PIM1-HDAC2 axis modulates intestinal homeostasis through epigenetic modification. Acta Pharm Sin B 2024; 14:3049-3067. [PMID: 39027246 PMCID: PMC11252454 DOI: 10.1016/j.apsb.2024.04.017] [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: 10/11/2023] [Revised: 03/07/2024] [Accepted: 03/14/2024] [Indexed: 07/20/2024] Open
Abstract
The mucosal barrier is crucial for intestinal homeostasis, and goblet cells are essential for maintaining the mucosal barrier integrity. The proviral integration site for Moloney murine leukemia virus-1 (PIM1) kinase regulates multiple cellular functions, but its role in intestinal homeostasis during colitis is unknown. Here, we demonstrate that PIM1 is prominently elevated in the colonic epithelia of both ulcerative colitis patients and murine models, in the presence of intestinal microbiota. Epithelial PIM1 leads to decreased goblet cells, thus impairing resistance to colitis and colitis-associated colorectal cancer (CAC) in mice. Mechanistically, PIM1 modulates goblet cell differentiation through the Wnt and Notch signaling pathways. Interestingly, PIM1 interacts with histone deacetylase 2 (HDAC2) and downregulates its level via phosphorylation, thereby altering the epigenetic profiles of Wnt signaling pathway genes. Collectively, these findings investigate the unknown function of the PIM1-HDAC2 axis in goblet cell differentiation and ulcerative colitis/CAC pathogenesis, which points to the potential for PIM1-targeted therapies of ulcerative colitis and CAC.
Collapse
Affiliation(s)
- Jianming Yang
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Institute of Immunology, State Key Laboratory of Experimental Hematology, the Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Immunology, School of Basic Medical Sciences, Tianjin Institute of Urology, the Second Hospital of Tianjin Medical University, Tianjin Key Laboratory of Cellular and Molecular Immunology, Tianjin Medical University, Tianjin 300070, China
| | - Yawen Xiao
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Institute of Immunology, State Key Laboratory of Experimental Hematology, the Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Immunology, School of Basic Medical Sciences, Tianjin Institute of Urology, the Second Hospital of Tianjin Medical University, Tianjin Key Laboratory of Cellular and Molecular Immunology, Tianjin Medical University, Tianjin 300070, China
| | - Ningning Zhao
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Institute of Immunology, State Key Laboratory of Experimental Hematology, the Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Immunology, School of Basic Medical Sciences, Tianjin Institute of Urology, the Second Hospital of Tianjin Medical University, Tianjin Key Laboratory of Cellular and Molecular Immunology, Tianjin Medical University, Tianjin 300070, China
| | - Geng Pei
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center of Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center of Cancer, Tianjin 30060, China
| | - Yan Sun
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center of Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center of Cancer, Tianjin 30060, China
| | - Xinyu Sun
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Institute of Immunology, State Key Laboratory of Experimental Hematology, the Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Immunology, School of Basic Medical Sciences, Tianjin Institute of Urology, the Second Hospital of Tianjin Medical University, Tianjin Key Laboratory of Cellular and Molecular Immunology, Tianjin Medical University, Tianjin 300070, China
| | - Kaiyuan Yu
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Institute of Immunology, State Key Laboratory of Experimental Hematology, the Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Immunology, School of Basic Medical Sciences, Tianjin Institute of Urology, the Second Hospital of Tianjin Medical University, Tianjin Key Laboratory of Cellular and Molecular Immunology, Tianjin Medical University, Tianjin 300070, China
| | - Chunhui Miao
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Institute of Immunology, State Key Laboratory of Experimental Hematology, the Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Immunology, School of Basic Medical Sciences, Tianjin Institute of Urology, the Second Hospital of Tianjin Medical University, Tianjin Key Laboratory of Cellular and Molecular Immunology, Tianjin Medical University, Tianjin 300070, China
| | - Ran Liu
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Institute of Immunology, State Key Laboratory of Experimental Hematology, the Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Immunology, School of Basic Medical Sciences, Tianjin Institute of Urology, the Second Hospital of Tianjin Medical University, Tianjin Key Laboratory of Cellular and Molecular Immunology, Tianjin Medical University, Tianjin 300070, China
| | - Junqiang Lv
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Institute of Immunology, State Key Laboratory of Experimental Hematology, the Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Immunology, School of Basic Medical Sciences, Tianjin Institute of Urology, the Second Hospital of Tianjin Medical University, Tianjin Key Laboratory of Cellular and Molecular Immunology, Tianjin Medical University, Tianjin 300070, China
| | - Hongyu Chu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin 300070, China
| | - Lu Zhou
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin 300070, China
| | - Bangmao Wang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin 300070, China
| | - Zhi Yao
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Institute of Immunology, State Key Laboratory of Experimental Hematology, the Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Immunology, School of Basic Medical Sciences, Tianjin Institute of Urology, the Second Hospital of Tianjin Medical University, Tianjin Key Laboratory of Cellular and Molecular Immunology, Tianjin Medical University, Tianjin 300070, China
| | - Quan Wang
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Institute of Immunology, State Key Laboratory of Experimental Hematology, the Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Immunology, School of Basic Medical Sciences, Tianjin Institute of Urology, the Second Hospital of Tianjin Medical University, Tianjin Key Laboratory of Cellular and Molecular Immunology, Tianjin Medical University, Tianjin 300070, China
| |
Collapse
|
14
|
Liu D, Li C, Cao T, Lv X, Yue Y, Li S, Cheng Y, Liu F, Huo G, Li B. Bifidobacterium longum K5 Prevents Enterohaemorrhagic Escherichia coli O157:H7 Infection in Mice through the Modulation of the Gut Microbiota. Nutrients 2024; 16:1164. [PMID: 38674854 PMCID: PMC11053520 DOI: 10.3390/nu16081164] [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: 03/21/2024] [Revised: 04/11/2024] [Accepted: 04/11/2024] [Indexed: 04/28/2024] Open
Abstract
Enterohemorrhagic Escherichia coli (EHEC) serotype O157:H7 is a commonly encountered foodborne pathogen that can cause hemorrhagic enteritis and lead to hemolytic uremic syndrome (HUS) in severe cases. Bifidobacterium is a beneficial bacterium that naturally exists in the human gut and plays a vital role in maintaining a healthy balance in the gut microbiota. This study investigated the protective effects of B. longum K5 in a mouse model of EHEC O157:H7 infection. The results indicated that pretreatment with B. longum K5 mitigated the clinical symptoms of EHEC O157:H7 infection and attenuated the increase in myeloperoxidase (MPO) activity in the colon of the mice. In comparison to the model group, elevated serum D-lactic acid concentrations and diamine oxidase (DAO) levels were prevented in the K5-EHEC group of mice. The reduced mRNA expression of tight junction proteins (ZO-1, Occludin, and Claudin-1) and mucin MUC2, as well as the elevated expression of virulence factors Stx1A and Stx2A, was alleviated in the colon of both the K5-PBS and K5-EHEC groups. Additionally, the increase in the inflammatory cytokine levels of TNF-α and IL-1β was inhibited and the production of IL-4 and IL-10 was promoted in the K5-EHEC group compared with the model group. B. longum K5 significantly prevented the reduction in the abundance and diversity of mouse gut microorganisms induced by EHEC O157:H7 infection, including blocking the decrease in the relative abundance of Roseburia, Lactobacillus, and Oscillibacter. Meanwhile, the intervention with B. longum K5 promoted the production of acetic acid and butyric acid in the gut. This study provides insights into the use of B. longum K5 for developing probiotic formulations to prevent intestinal diseases caused by pathogenic bacterial infections.
Collapse
Affiliation(s)
- Deyu Liu
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China; (D.L.); (C.L.); (T.C.); (X.L.); (Y.Y.); (S.L.); (Y.C.); (F.L.); (B.L.)
- Food College, Northeast Agricultural University, Harbin 150030, China
| | - Chunyan Li
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China; (D.L.); (C.L.); (T.C.); (X.L.); (Y.Y.); (S.L.); (Y.C.); (F.L.); (B.L.)
- Food College, Northeast Agricultural University, Harbin 150030, China
| | - Ting Cao
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China; (D.L.); (C.L.); (T.C.); (X.L.); (Y.Y.); (S.L.); (Y.C.); (F.L.); (B.L.)
- Food College, Northeast Agricultural University, Harbin 150030, China
| | - Xiuli Lv
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China; (D.L.); (C.L.); (T.C.); (X.L.); (Y.Y.); (S.L.); (Y.C.); (F.L.); (B.L.)
- Food College, Northeast Agricultural University, Harbin 150030, China
| | - Yingxue Yue
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China; (D.L.); (C.L.); (T.C.); (X.L.); (Y.Y.); (S.L.); (Y.C.); (F.L.); (B.L.)
- Food College, Northeast Agricultural University, Harbin 150030, China
| | - Shuang Li
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China; (D.L.); (C.L.); (T.C.); (X.L.); (Y.Y.); (S.L.); (Y.C.); (F.L.); (B.L.)
- Food College, Northeast Agricultural University, Harbin 150030, China
| | - Yang Cheng
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China; (D.L.); (C.L.); (T.C.); (X.L.); (Y.Y.); (S.L.); (Y.C.); (F.L.); (B.L.)
- Food College, Northeast Agricultural University, Harbin 150030, China
| | - Fei Liu
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China; (D.L.); (C.L.); (T.C.); (X.L.); (Y.Y.); (S.L.); (Y.C.); (F.L.); (B.L.)
- Food College, Northeast Agricultural University, Harbin 150030, China
| | - Guicheng Huo
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China; (D.L.); (C.L.); (T.C.); (X.L.); (Y.Y.); (S.L.); (Y.C.); (F.L.); (B.L.)
- Food College, Northeast Agricultural University, Harbin 150030, China
| | - Bailiang Li
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China; (D.L.); (C.L.); (T.C.); (X.L.); (Y.Y.); (S.L.); (Y.C.); (F.L.); (B.L.)
- Food College, Northeast Agricultural University, Harbin 150030, China
| |
Collapse
|
15
|
Man Y, Xin D, Ji Y, Liu Y, Kou L, Jiang L. Identification and validation of a novel six-gene signature based on mucinous adenocarcinoma-related gene molecular typing in colorectal cancer. Discov Oncol 2024; 15:63. [PMID: 38443703 PMCID: PMC10914658 DOI: 10.1007/s12672-024-00916-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 02/28/2024] [Indexed: 03/07/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Colorectal mucinous adenocarcinoma (MAC) is a particular pathological type that has yet to be thoroughly studied. This study aims to investigate the characteristics of colorectal MAC-related genes in colorectal cancer (CRC), explore the role of MAC-related genes in accurately classifying CRC, and further construct a prognostic signature. METHODS CRC samples were collected from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO). MAC-related differentially expressed genes (DEGs) were analyzed in TCGA samples. Based on colorectal MAC-related genes, TCGA CRC samples were molecularly typed by the non-negative matrix factorization (NMF). According to the molecular subtype characteristics, the RiskScore signature was constructed through univariate Cox, the least absolute shrinkage and selection operator (LASSO), and multivariate Cox regression analyses. Clinical significance in CRC of the RiskScore signature was analyzed. A nomogram was further built based on the RiskScore signature. RESULTS From the colorectal MAC-related genes, three distinct molecular subtypes were identified. A RiskScore signature composed of six CRC subtype-related genes (CALB1, MMP1, HOXC6, ZIC2, SFTA2, and HYAL1) was constructed. Patients with high-RiskScores had the worse prognoses. RiskScores led to differences in gene mutation characteristics, antitumor drug sensitivity, and tumor microenvironment of CRC. A nomogram based on the signature was developed to predict the one-, three-, and five-year survival of CRC patients. CONCLUSION MAC-related genes were able to classify CRC. A RiskScore signature based on the colorectal MAC-related molecular subtype was constructed, which had important clinical significance for guiding the accurate stratification of CRC patients.
Collapse
Affiliation(s)
- Yuxin Man
- Department of Medical Oncology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Dao Xin
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yang Ji
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yang Liu
- Department of Medical Oncology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Lingna Kou
- Department of Medical Oncology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Lingxi Jiang
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China.
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
16
|
Hoshimoto A, Tatsuguchi A, Yamada T, Kuriyama S, Hamakubo R, Nishimoto T, Omori J, Akimoto N, Gudis K, Mitsui K, Tanaka S, Fujimori S, Hatori T, Shimizu A, Iwakiri K. Relationship Between Immunophenotypes, Genetic Profiles, and Clinicopathologic Characteristics in Small Bowel Adenocarcinoma. Am J Surg Pathol 2024; 48:127-139. [PMID: 38062562 PMCID: PMC10786444 DOI: 10.1097/pas.0000000000002161] [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] [Indexed: 01/14/2024]
Abstract
Small bowel adenocarcinoma (SBA) is rare, and scant data exist regarding its molecular and clinicopathologic characteristics. This study aimed to clarify the correlation between immunophenotypes, DNA mismatch repair status, genomic profiling, and clinicopathologic characteristics in patients with SBA. We examined 68 surgical resections from patients with primary SBA for immunohistochemical analyses of CK7, CK20, CD10, CDX2, MUC1, MUC2, MUC4, MUC5AC, and MUC6 expression as well as mismatch repair status. Genomic profiling was performed on 30 cases using targeted next-generation sequencing. Tumor mucin phenotypes were classified as gastric, intestinal, gastrointestinal, or null based on MUC2, MUC5AC, MUC6, and CD10 immunostaining. The expression of these proteins was categorized into 3 classifications according to their relationship to: (1) tumor location: CK7/CK20, MUC4, and MUC6; (2) histologic type: mucinous adenocarcinoma was positive for MUC2 and negative for MUC6; and (3) TNM stage: CD10 was downregulated, whereas MUC1 was upregulated in advanced TNM stages. CDX2 was a specific marker for SBA generally expressed in the small intestine. MUC1 and MUC4 expression was significantly associated with worse prognosis. MUC2 expression correlated with better prognosis, except for mucinous adenocarcinoma. Although the difference was not statistically significant, gastric-type tumors were more frequently located in the duodenum and were absent in the ileum. APC and CTNNB1 mutations were not found in the gastric-type tumors. The SBA immunophenotype correlated with tumor location, biological behavior, and genomic alterations. Our results suggest that the molecular pathway involved in carcinogenesis of gastric-type SBA differs from that of intestinal-type SBA.
Collapse
Affiliation(s)
| | | | - Takeshi Yamada
- Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Nippon Medical School, Tokyo
| | - Sho Kuriyama
- Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Nippon Medical School, Tokyo
| | | | | | | | | | | | | | | | | | - Tsutomu Hatori
- Department of Pathology, Nippon Medical School, Chiba Hokusoh Hospital, Chiba, Japan
| | | | | |
Collapse
|
17
|
Yu H, Qin XK, Yin KW, Li ZM, Ni ED, Yang JM, Liu XH, Zhou AJ, Li SJ, Gao TM, Li Y, Li JM. EphB6 deficiency in intestinal neurons promotes tumor growth in colorectal cancer by neurotransmitter GABA signaling. Carcinogenesis 2023; 44:682-694. [PMID: 37294054 DOI: 10.1093/carcin/bgad041] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 05/24/2023] [Accepted: 06/08/2023] [Indexed: 06/10/2023] Open
Abstract
EphB6 belongs to the receptor tyrosine kinase, whose low expression is associated with shorter survival of colorectal cancer (CRC) patients. But the role and mechanism of EphB6 in the progression of CRC need further study. In addition, EphB6 was mainly expressed in intestinal neurons. But how EphB6 is involved in functions of intestinal neurons has not been known. In our study, we constructed a mouse xenograft model of CRC by injecting CMT93 cells into the rectum of EphB6-deficient mice. We found that the deletion of EphB6 in mice promoted tumor growth of CMT93 cells in a xenograft model of CRC, which was independent of changes in the gut microbiota. Interestingly, inhibition of intestinal neurons by injecting botulinum toxin A into rectum of EphB6-deficient mice could eliminate the promotive effect of EphB6 deficiency on tumor growth in the xenograft model of CRC. Mechanically, the deletion of EphB6 in mice promoted the tumor growth in CRC by increasing GABA in the tumor microenvironment. Furthermore, EphB6 deficiency in mice increased the expression of synaptosomal-associated protein 25 in the intestinal myenteric plexus, which mediated the release of GABA. Our study concluded that EphB6 knockout in mice promotes tumor growth of CMT93 cells in a xenograft model of CRC by modulating GABA release. Our study found a new regulating mechanism of EphB6 on the tumor progression in CRC that is dependent on intestinal neurons.
Collapse
Affiliation(s)
- Hao Yu
- Department of Pathology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, People's Republic of China
| | - Xiao-Kang Qin
- Department of Pathology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, People's Republic of China
| | - Kai-Wen Yin
- Department of Pathology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, People's Republic of China
| | - Zi-Ming Li
- State Key Laboratory of Organ Failure Research, Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders Collaborative Innovation Center for Brain Science, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - En-De Ni
- Department of Pathology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, People's Republic of China
| | - Jian-Ming Yang
- State Key Laboratory of Organ Failure Research, Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders Collaborative Innovation Center for Brain Science, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Xun-Hua Liu
- Department of Pathology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, People's Republic of China
| | - Ai-Jun Zhou
- Department of Pathology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, People's Republic of China
| | - Shu-Ji Li
- State Key Laboratory of Organ Failure Research, Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders Collaborative Innovation Center for Brain Science, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Tian-Ming Gao
- State Key Laboratory of Organ Failure Research, Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders Collaborative Innovation Center for Brain Science, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Ying Li
- Department of Pathology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, People's Republic of China
| | - Jian-Ming Li
- Department of Pathology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, People's Republic of China
- Department of Pathology, Soochow University Medical School, Suzhou 215123, People's Republic of China
| |
Collapse
|
18
|
Zhang Q, Chen M, Xu F, Wu W, Luo X, Wang Y, Li J, Cui X, Tan Y, Li Z, Lin Y, Zhang H, Wang W. One-pot preparation of bi-functional POSS-based hybrid monolith via photo-initiated polymerization for isolation of extracellular vesicles. Anal Chim Acta 2023; 1279:341785. [PMID: 37827681 DOI: 10.1016/j.aca.2023.341785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 08/16/2023] [Accepted: 09/05/2023] [Indexed: 10/14/2023]
Abstract
Extracellular vesicles (EVs) are important participants in numerous pathophysiological processes, and could be used as valuable biomarkers to detect and monitor various diseases. However, facile EV isolation methods are the essential and preliminary issue for their downstream analysis and function investigation. In this work, a polyhedral oligomeric silsesquioxanes (POSS) based hybrid monolith combined metal affinity chromatography (MAC) and distearoyl phospholipid ethanolamine (DSPE) function was developed via photo-initiated thiol-ene polymerization. This synthesis process was facile, simple and convenient, and the obtained hybrid monolith could be applied to efficiently isolate EVs from bio-samples by taking advantages of the specific bond of Ti4+ and phosphate groups on the phospholipid membrane of EVs and the synergistic effect of DSPE insertion. Meanwhile, the eluted EVs could maintain their structural integrity and biological activity, suggesting they could be used for downstream application. Furthermore, 75 up-regulated proteins and 56 down-regulated proteins were identified by comparing the urinary EVs of colorectal cancer (CRC) patients and healthy donors, and these proteins might be used as potential biomarkers for early screening of CRC. These results demonstrated that this hybrid monolith could be used as a simple and convenient tool for isolating EVs from bio-samples and for wider applications in biomarker discovery.
Collapse
Affiliation(s)
- Qi Zhang
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Mengxi Chen
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Fang Xu
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Wen Wu
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Xintong Luo
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Ying Wang
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China; Taichang Liuhe People's Hospital, Suzhou, 215431, China
| | - Jiaxi Li
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Xuanhao Cui
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Yujia Tan
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Zhi Li
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Yujie Lin
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Haiyang Zhang
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China.
| | - Weipeng Wang
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China.
| |
Collapse
|
19
|
Huang Z, Wu H, Fan J, Mei Q, Fu Y, Yin N, Xu B, Luo S, Li B, Ni J, Huang C, Hu J, Zeng Y. Colonic mucin-2 attenuates acute necrotizing pancreatitis in rats by modulating intestinal homeostasis. FASEB J 2023; 37:e22994. [PMID: 37249555 DOI: 10.1096/fj.202201998r] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 04/25/2023] [Accepted: 05/12/2023] [Indexed: 05/31/2023]
Abstract
Mucin-2 (MUC2) secreted by goblet cells participates in the intestinal barrier, but its mechanism in acute necrotizing pancreatitis (ANP) remains unclear. In acute pancreatitis (AP) patients, the functions of goblet cells (MUC2, FCGBP, CLCA1, and TFF3) decreased, and MUC2 was negatively correlated with AP severity. ANP rats treated with pilocarpine (PILO) (PILO+ANP rats) to deplete MUC2 showed more serious pancreatic and colonic injuries, goblet cell dysfunction, gut dysbiosis, and bacterial translocation than those of ANP rats. GC-MS analysis of feces showed that PILO+ANP rats had lower levels of butyric acid, isobutyric acid, isovaleric acid, and hexanoic acid than those of ANP rats. The expression of MUC2 was associated with colonic injury and gut dysbiosis. All these phenomena could be relieved, and goblet cell functions were also partially reversed by MUC2 supplementation in ANP rats. TNF-α-treated colonoids had exacerbated goblet cell dysfunction. MUC2 expression was negatively correlated with the levels of pro-inflammatory cytokines (IL-1β and IL-6) (p < .05) and positively related to the expression of tight junction proteins (Claudin 1, Occludin, and ZO1) (p < .05). Downregulating MUC2 by siRNA increased the levels of the pro-inflammatory cytokines in colonoids. MUC2 might maintain intestinal homeostasis to alleviate ANP.
Collapse
Affiliation(s)
- Zehua Huang
- Shanghai Key Laboratory of Pancreatic Disease, Shanghai JiaoTong University School of Medicine, Shanghai, China
- Department of Gastroenterology, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Huimin Wu
- Department of Gastroenterology, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Junjie Fan
- Department of Gastroenterology, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Qixiang Mei
- Department of Gastroenterology, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Yang Fu
- Shanghai Key Laboratory of Pancreatic Disease, Shanghai JiaoTong University School of Medicine, Shanghai, China
- Department of Gastroenterology, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Nuoming Yin
- Shanghai Key Laboratory of Pancreatic Disease, Shanghai JiaoTong University School of Medicine, Shanghai, China
- Department of Gastroenterology, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Binqiang Xu
- Shanghai Key Laboratory of Pancreatic Disease, Shanghai JiaoTong University School of Medicine, Shanghai, China
- Department of Gastroenterology, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Shengzheng Luo
- Department of Gastroenterology, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Baiwen Li
- Department of Gastroenterology, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Jianbo Ni
- Department of Gastroenterology, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Chunlan Huang
- Department of Gastroenterology, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Junjie Hu
- Department of Gastroenterology, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Yue Zeng
- Shanghai Key Laboratory of Pancreatic Disease, Shanghai JiaoTong University School of Medicine, Shanghai, China
- Department of Gastroenterology, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| |
Collapse
|
20
|
Zhou R, Li L, Xi S, Zhang Y, Liu Z, Zeng D, Sun H, Wu J, Wang L, Shi M, Bin J, Liao Y, Liao W. Expression pattern of secretory-cell-related transcriptional signatures in colon adenocarcinomas defines tumor microenvironment characteristics and correlates with clinical outcomes. Mol Oncol 2023; 17:499-517. [PMID: 36349418 PMCID: PMC9980301 DOI: 10.1002/1878-0261.13338] [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: 07/21/2022] [Revised: 10/21/2022] [Accepted: 11/07/2022] [Indexed: 11/11/2022] Open
Abstract
Despite the connection of secretory cells to distinct mucus-containing colon cancer histological subtypes and the interaction of secretory cells with immune cells in the pathogenesis of intestinal inflammatory diseases, whether the secretory cell signatures are associated with tumor microenvironment (TME) heterogeneity and can aid in colon cancer patient classification have not been investigated. Here, by performing the principal component analysis and consensus clustering analysis, we identified four distinct expression patterns based on secretory cell signatures which were significantly associated with different clinical behaviors, TME landscape, pathway activation, genomic mutations, and DNA methylation characteristics. Subsequently, a 'SCS score' model was constructed. The high SCS score indicated a pattern of 'secretory cell subtype 2', which was characterized by stromal infiltration and activation, and predicted poor prognosis and low sensitivity to fluorouracil-based chemotherapy and immunotherapy, but high sensitivity to PI3K catalytic subunit inhibitors. In conclusion, our study comprehensively uncovered the tumor heterogeneity related to secretory cell signature expression patterns. Moreover, the SCS score can supplement routine histopathological assessments to guide personalized therapeutic strategies in colon cancer patients.
Collapse
Affiliation(s)
- Rui Zhou
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Lingbo Li
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shaoyan Xi
- Department of Pathology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yue Zhang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhihong Liu
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Dongqiang Zeng
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Huiying Sun
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Jianhua Wu
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Ling Wang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Min Shi
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Jianping Bin
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yulin Liao
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wangjun Liao
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| |
Collapse
|
21
|
Hu S, Wei P, Li W, Liu Q, Chen S, Hu C, Guo X, Ma X, Zeng J, Zhang Y. Pharmacological effects of berberine on models of ulcerative colitis: A meta-analysis and systematic review of animal studies. Front Pharmacol 2022; 13:937029. [PMID: 36147325 PMCID: PMC9486070 DOI: 10.3389/fphar.2022.937029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 08/08/2022] [Indexed: 12/09/2022] Open
Abstract
Berberine (BBR) is the main active constituent of the Rhizoma coptidis (Huanglian) and has multiple biological activities. Although current evidence suggests that the BBR has a multi-target effect in ulcerative colitis (UC), its action and mechanism are unclear. The purpose of this meta-analysis was to assess the pharmacological effects and potential mechanisms of BBR in UC models. Studies were searched from four databases (PubMed, Embase, Web of Science, and Cochrane Library) until March 2022. Standardized mean difference (SMD) and 95% confidence intervals (CI) were used for the adjudication of outcomes. Stata 15.0 software was used for statistical analysis. Twenty-eight publications and 29 studies involving 508 animals were included in the meta-analysis. The results showed that BBR reduced disease activity index (DAI) scores, alleviated UC-induced colon length (CL) loss, prevented weight loss, and reduced histological colitis score (HCS). Mechanistically, BBR was found to reduce myeloperoxidase (MPO) activity and malondialdehyde (MDA) levels, reduce levels of pro-inflammatory factors interleukin-1β (IL-1β), interleukin 6 (IL-6), tumor necrosis factor α (TNF-α), interferon-γ (IFN-γ) and mRNA expression of interleukin 17, increase levels of anti-inflammatory factor interleukin 10 (IL-10), and to increase levels of tight junction protein zonula occludens-1 (ZO-1) and occludin, which may involve antioxidant, anti-apoptotic, neuromodulation, anti-fibrotic, anti-inflammatory, barrier protection, and flora regulation aspects. However, additional attention should be paid to these outcomes due to the heterogeneity and methodological quality of the studies.
Collapse
Affiliation(s)
- Shuangyuan Hu
- Department of Gastroenterology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Pengfei Wei
- Department of Gastroenterology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wei Li
- Department of Gastroenterology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qingsong Liu
- Department of Gastroenterology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shuanglan Chen
- Department of Gastroenterology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Caiyu Hu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaochuan Guo
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiao Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Xiao Ma, ; Jinhao Zeng, ; Yi Zhang,
| | - Jinhao Zeng
- Department of Geriatrics, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Xiao Ma, ; Jinhao Zeng, ; Yi Zhang,
| | - Yi Zhang
- Department of Gastroenterology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Xiao Ma, ; Jinhao Zeng, ; Yi Zhang,
| |
Collapse
|
22
|
Autophagy and EMT in cancer and metastasis: Who controls whom? Biochim Biophys Acta Mol Basis Dis 2022; 1868:166431. [PMID: 35533903 DOI: 10.1016/j.bbadis.2022.166431] [Citation(s) in RCA: 79] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/10/2022] [Accepted: 05/02/2022] [Indexed: 02/06/2023]
Abstract
Metastasis consists of hallmark events, including Epithelial-Mesenchymal Transition (EMT), angiogenesis, initiation of inflammatory tumor microenvironment, and malfunctions in apoptosis. Autophagy is known to play a pivotal role in the metastatic process. Autophagy has pulled researchers towards it in recent times because of its dual role in the maintenance of cancer cells. Evidence states that cells undergoing EMT need autophagy in order to survive during migration and dissemination. Additionally, it orchestrates EMT markers in certain cancers. On the other side of the coin, autophagy plays an oncosuppressive role in impeding early metastasis. This review aims to project the interrelationship between autophagy and EMT. Targeting EMT via autophagy as a useful strategy is discussed in this review. Furthermore, for the first time, we have covered the possible reciprocating roles of EMT and autophagy and its consequences in cancer metastasis.
Collapse
|
23
|
Dai YJ, Liu WB, Abasubong KP, Zhang DD, Li XF, Xiao K, Wang X, Jiang GZ. The Mechanism of Lipopolysaccharide Escaping the Intestinal Barrier in Megalobrama amblycephala Fed a High-Fat Diet. Front Nutr 2022; 9:853409. [PMID: 35464002 PMCID: PMC9023073 DOI: 10.3389/fnut.2022.853409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 03/07/2022] [Indexed: 11/21/2022] Open
Abstract
With the popularity of western food characterized by excessive fat and sugars, obesity has currently been a public health issue. Low-grade chronic inflammation accompanied by obesity increases the risk of multiple epidemics such as diabetes, cancer and cardiovascular diseases. Here, we show that feeding Megalobrama amblycephala with a high-fat diet (HFD) drives obesity-related chronic inflammation and the penetration of lipopolysaccharide (LPS). Interference with antibiotics inhibits the produce of LPS and this alleviates the sustained release of pro-inflammatory factors induced by HFD. LPS penetration is attributed to weakened intestinal mucus barrier after high-fat exposure. Mechanically, the consumption of HFD inhibits the secretion of mucin 2 (MUC2) due to the induction of endoplasmic reticulum stress mediated by the inositol-requiring enzyme 1 (IRE1) /X box-binding protein 1 (XBP1) pathway in goblet cells. Furthermore, excessive lipid exacerbates the leakage of LPS across the intestinal epithelial cell barrier via the transcellular pathway. Mechanically, lipid increases the internalization of LPS in intestinal epithelial cells depending on the activation of fatty acid translocase (FAT/CD36). These results demonstrate that HFD causes the penetration of LPS due to the weakened intestinal mucosal barrier and the assistance of CD36.
Collapse
|
24
|
Zhu L, Miao B, Dymerska D, Kuswik M, Bueno-Martínez E, Sanoguera-Miralles L, Velasco EA, Paramasivam N, Schlesner M, Kumar A, Yuan Y, Lubinski J, Bandapalli OR, Hemminki K, Försti A. Germline Variants of CYBA and TRPM4 Predispose to Familial Colorectal Cancer. Cancers (Basel) 2022; 14:cancers14030670. [PMID: 35158942 PMCID: PMC8833488 DOI: 10.3390/cancers14030670] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/17/2022] [Accepted: 01/26/2022] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Whole-genome sequencing and bioinformatics analysis on unique colorectal cancer families revealed two attractive candidate predisposition genes, CYBA and TRPM4, each with a loss-of-function variant. Supported by our functional studies, we suggest that the two gene defects mechanistically involve intestinal barrier integrity through reactive oxygen species and mucus biology, which converges in chronic bowel inflammation, a known risk factor for colorectal cancer. Abstract Familial colorectal cancer (CRC) is only partially explained by known germline predisposing genes. We performed whole-genome sequencing in 15 Polish families of many affected individuals, without mutations in known CRC predisposing genes. We focused on loss-of-function variants and functionally characterized them. We identified a frameshift variant in the CYBA gene (c.246delC) in one family and a splice site variant in the TRPM4 gene (c.25–1 G > T) in another family. While both variants were absent or extremely rare in gene variant databases, we identified four additional Polish familial CRC cases and two healthy elderly individuals with the CYBA variant (odds ratio 2.46, 95% confidence interval 0.48–12.69). Both variants led to a premature stop codon and to a truncated protein. Functional characterization of the variants showed that knockdown of CYBA or TRPM4 depressed generation of reactive oxygen species (ROS) in LS174T and HT-29 cell lines. Knockdown of TRPM4 resulted in decreased MUC2 protein production. CYBA encodes a component in the NADPH oxidase system which generates ROS and controls, e.g., bacterial colonization in the gut. Germline CYBA variants are associated with early onset inflammatory bowel disease, supported with experimental evidence on loss of intestinal mucus barrier function due to ROS deficiency. TRPM4 encodes a calcium-activated ion channel, which, in a human colonic cancer cell line, controls calcium-mediated secretion of MUC2, a major component of intestinal mucus barrier. We suggest that the gene defects in CYBA and TRPM4 mechanistically involve intestinal barrier integrity through ROS and mucus biology, which converges in chronic bowel inflammation.
Collapse
Affiliation(s)
- Lizhen Zhu
- Division of Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, D-69120 Heidelberg, Germany; (L.Z.); (B.M.); (A.K.); (A.F.)
- Department of Medical Oncology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China;
| | - Beiping Miao
- Division of Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, D-69120 Heidelberg, Germany; (L.Z.); (B.M.); (A.K.); (A.F.)
- Hopp Children’s Cancer Center (KiTZ), D-69120 Heidelberg, Germany
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), D-69120 Heidelberg, Germany
| | - Dagmara Dymerska
- Department of Genetics and Pathology, Hereditary Cancer Center, Pomeranian Medical University, Unii Lubelskiej 1, 71-252 Szczecin, Poland; (D.D.); (M.K.); (J.L.)
| | - Magdalena Kuswik
- Department of Genetics and Pathology, Hereditary Cancer Center, Pomeranian Medical University, Unii Lubelskiej 1, 71-252 Szczecin, Poland; (D.D.); (M.K.); (J.L.)
| | - Elena Bueno-Martínez
- Splicing and Genetic Susceptibility to Cancer, Instituto de Biología y Genética Molecular (CSIC-UVa), 47003 Valladolid, Spain; (E.B.-M.); (L.S.-M.); (E.A.V.)
| | - Lara Sanoguera-Miralles
- Splicing and Genetic Susceptibility to Cancer, Instituto de Biología y Genética Molecular (CSIC-UVa), 47003 Valladolid, Spain; (E.B.-M.); (L.S.-M.); (E.A.V.)
| | - Eladio A. Velasco
- Splicing and Genetic Susceptibility to Cancer, Instituto de Biología y Genética Molecular (CSIC-UVa), 47003 Valladolid, Spain; (E.B.-M.); (L.S.-M.); (E.A.V.)
| | - Nagarajan Paramasivam
- Computational Oncology, Molecular Diagnostics Program, National Center for Tumor Diseases (NCT), D-69120 Heidelberg, Germany;
| | - Matthias Schlesner
- Bioinformatics and Omics Data Analytics, German Cancer Research Center (DKFZ), D-69120 Heidelberg, Germany;
| | - Abhishek Kumar
- Division of Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, D-69120 Heidelberg, Germany; (L.Z.); (B.M.); (A.K.); (A.F.)
- Institute of Bioinformatics, International Technology Park, Bengaluru 560066, India
- Manipal Academy of Higher Education (MAHE), Manipal 576104, India
| | - Ying Yuan
- Department of Medical Oncology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China;
| | - Jan Lubinski
- Department of Genetics and Pathology, Hereditary Cancer Center, Pomeranian Medical University, Unii Lubelskiej 1, 71-252 Szczecin, Poland; (D.D.); (M.K.); (J.L.)
| | - Obul Reddy Bandapalli
- Division of Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, D-69120 Heidelberg, Germany; (L.Z.); (B.M.); (A.K.); (A.F.)
- Hopp Children’s Cancer Center (KiTZ), D-69120 Heidelberg, Germany
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), D-69120 Heidelberg, Germany
- Medical Faculty Heidelberg, Heidelberg University, D-69120 Heidelberg, Germany
- Correspondence: (O.R.B.); (K.H.)
| | - Kari Hemminki
- Division of Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, D-69120 Heidelberg, Germany; (L.Z.); (B.M.); (A.K.); (A.F.)
- Faculty of Medicine and Biomedical Center in Pilsen, Charles University in Prague, 30605 Pilsen, Czech Republic
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), D-69120 Heidelberg, Germany
- Correspondence: (O.R.B.); (K.H.)
| | - Asta Försti
- Division of Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, D-69120 Heidelberg, Germany; (L.Z.); (B.M.); (A.K.); (A.F.)
- Hopp Children’s Cancer Center (KiTZ), D-69120 Heidelberg, Germany
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), D-69120 Heidelberg, Germany
| |
Collapse
|
25
|
The Role of Gut Microbiota in Tumor Immunotherapy. J Immunol Res 2021; 2021:5061570. [PMID: 34485534 PMCID: PMC8413023 DOI: 10.1155/2021/5061570] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 08/10/2021] [Indexed: 12/12/2022] Open
Abstract
Tumor immunotherapy is the fourth therapy after surgery, chemotherapy, and radiotherapy. It has made great breakthroughs in the treatment of some epithelial tumors and hematological tumors. However, its adverse reactions are common or even more serious, and the response rate in some solid tumors is not satisfactory. With the maturity of genomics and metabolomics technologies, the effect of intestinal microbiota in tumor development and treatment has gradually been recognized. The microbiota may affect tumor immunity by regulating the host immune system and tumor microenvironment. Some bacteria help fight tumors by activating immunity, while some bacteria mediate immunosuppression to help cancer cells escape from the immune system. More and more studies have revealed that the effects and complications of tumor immunotherapy are related to the composition of the gut microbiota. The composition of the intestinal microbiota that is sensitive to treatment or prone to adverse reactions has certain characteristics. These characteristics may be used as biomarkers to predict the prognosis of immunotherapy and may also be developed as “immune potentiators” to assist immunotherapy. Some clinical and preclinical studies have proved that microbial intervention, including microbial transplantation, can improve the sensitivity of immunotherapy or reduce adverse reactions to a certain extent. With the development of gene editing technology and nanotechnology, the design and development of engineered bacteria that contribute to immunotherapy has become a new research hotspot. Based on the relationship between the intestinal microbiota and immunotherapy, the correct mining of microbial information and the development of reasonable and feasible microbial intervention methods are expected to optimize tumor immunotherapy to a large extent and bring new breakthroughs in tumor treatment.
Collapse
|