1
|
Jing Z, Yinhang W, Jian C, Zhanbo Q, Xinyue W, Shuwen H. Interaction between gut microbiota and T cell immunity in colorectal cancer. Autoimmun Rev 2025; 24:103807. [PMID: 40139455 DOI: 10.1016/j.autrev.2025.103807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 02/26/2025] [Accepted: 03/22/2025] [Indexed: 03/29/2025]
Abstract
This review delves into the complex and multi-layered mechanisms that govern the interaction between gut microbiota and T cells in the context of colorectal cancer (CRC), revealing a novel "microbiota-immune regulatory landscape" within the tumor microenvironment. As CRC progresses, the gut microbiota experiences a significant transformation in both its composition and metabolic patterns. On one hand, specific microbial entities within the gut microbiota can directly engage with T cells, functioning as "immunological triggers" that shape T-cell behavior. Simultaneously, microbial metabolites, such as short-chain fatty acids and bile acids, serve as "molecular regulators" that intricately govern T-cell function and differentiation, fine-tuning the immune response. On the other hand, the quorum-sensing mechanism, a recently recognized communication network among bacteria, also plays a pivotal role in orchestrating T-cell immunity. Additionally, the gut microbiota forms an intriguing connection with the neuro-immune regulatory axis, a largely unexplored "territory" in CRC research. Regarding treatment strategies, a diverse array of intervention approaches-including dietary modifications, the utilization of probiotics, bacteriophages, and targeted antibiotic therapies-offer promising prospects for restoring the equilibrium of the gut microbiota, thereby acting as "ecosystem renovators" that impede tumor initiation and progression. Nevertheless, the current research landscape in this field is fraught with challenges. These include significant variations in microbial composition, dietary preferences, and tumor microenvironments among individuals, a lack of large-scale cohort studies, and insufficient research that integrates tumor mutation analysis, gut microbiota investigations, and immune microenvironment evaluations. This review emphasizes the necessity for future research efforts to seamlessly incorporate multiple factors and utilize bioinformatics analysis to construct a more comprehensive "interactive map" of the gut microbiota-T cell relationship in CRC. The aim is to establish a solid theoretical basis for the development of highly effective and personalized treatment regimens, ultimately transforming the therapeutic approach to CRC.
Collapse
Affiliation(s)
- Zhuang Jing
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China; Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China; Zhejiang-France United Laboratory of Integrated Traditional Chinese and Modern Medicine in Colorectal Cancer, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China
| | - Wu Yinhang
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China; Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China; Zhejiang-France United Laboratory of Integrated Traditional Chinese and Modern Medicine in Colorectal Cancer, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China
| | - Chu Jian
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China; Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China; Zhejiang-France United Laboratory of Integrated Traditional Chinese and Modern Medicine in Colorectal Cancer, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China
| | - Qu Zhanbo
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China; Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China; Zhejiang-France United Laboratory of Integrated Traditional Chinese and Modern Medicine in Colorectal Cancer, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China
| | - Wu Xinyue
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China; Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China; Zhejiang-France United Laboratory of Integrated Traditional Chinese and Modern Medicine in Colorectal Cancer, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China
| | - Han Shuwen
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China; Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China; Zhejiang-France United Laboratory of Integrated Traditional Chinese and Modern Medicine in Colorectal Cancer, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China; ASIR (Institute - Association of intelligent systems and robotics), 14B rue Henri Sainte Claire Deville, 92500 Rueil-Malmaison, France.
| |
Collapse
|
2
|
Tao Y, Shen L, Wang P. Advances and perspectives on isolation, structural characterization, structure-property relationships, and pharmacological mechanisms of tumor immune glucans: A review. Int J Biol Macromol 2025:144621. [PMID: 40414394 DOI: 10.1016/j.ijbiomac.2025.144621] [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: 12/03/2024] [Revised: 05/14/2025] [Accepted: 05/22/2025] [Indexed: 05/27/2025]
Abstract
Tumor immune glucans (TIGs) are emerging as promising anticancer agents capable of enhancing immune responses and mitigating the side effects of chemotherapy. This review analyzes 59 TIGs published between 2015 and 2025, focusing on their isolation, structural characterization, structure-activity relationships, and pharmacological mechanisms. Literature searches were conducted in databases such as Web of Science, PubMed, Google Scholar, Embase, Cochrane Library, ClinicalTrials.gov and CNKI using keywords including "glucans", "tumor immunity", and "polysaccharide structure," covering studies in both English and Chinese. The review highlights the relationship between the structural features of TIGs and their antitumor activities.1,3-linked β-glucans surpassed 1,4-linked or 1,6-linked configurations in antitumor immunostimulatory activity. Elevated molecular weight enhances the structural robustness of TIGs chains, facilitating their selective recognition by cellular receptors and amplifying their anti-tumor immune responses. TIGs exert antitumor effects through mechanisms such as immunomodulation, inhibition of tumor immune escape, and remodeling of the tumor microenvironment, involving signaling pathways such as TLRs/NF-κB, JAK/STAT, MAPK, Bax/Bcl-2, PI3K/Akt, and VEGF/VEGFR. Despite their promising therapeutic potential, challenges remain, including incomplete structural characterization, insufficient IC50 value data, and a lack of reference drugs. Many studies focus on cytokine levels and cellular activity, but deeper molecular mechanistic insights are needed. Further research into the mechanisms of action and targets of TIGs may pave the way for new cancer treatment strategies.
Collapse
Affiliation(s)
- Yi Tao
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, PR China; Zhejiang Provincial Key Laboratory of TCM for Innovative R&D and Digital Intelligent Manufacturing of TCM Great Health Products, PR China.
| | - Lisha Shen
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, PR China; Zhejiang Provincial Key Laboratory of TCM for Innovative R&D and Digital Intelligent Manufacturing of TCM Great Health Products, PR China
| | - Ping Wang
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, PR China; Zhejiang Provincial Key Laboratory of TCM for Innovative R&D and Digital Intelligent Manufacturing of TCM Great Health Products, PR China.
| |
Collapse
|
3
|
Dong J, Du J, Liu R, Gao X, Wang Y, Ma L, Yang Y, Wu J, Yu J, Liu N. Depressive Disorder Affects TME and Hormonal Changes Promoting Tumour Deterioration Development. Immunology 2025. [PMID: 40341563 DOI: 10.1111/imm.13933] [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: 12/01/2024] [Revised: 04/04/2025] [Accepted: 04/07/2025] [Indexed: 05/10/2025] Open
Abstract
Cancer patients often suffer from depression, the presence of which promotes the deterioration of the cancer patient's condition and thus affects the patient's survival. However, the exact mechanisms underlying the relationship between depression and tumour progression remain unclear, and this complexity involves multi-system and multi-level interactions, with several key challenges remaining in current research. First, the extreme complexity of biological systems. Depression and tumors involve multiple pathways such as neuroendocrine, immune system, and metabolism, respectively, and there are nonlinear interactions between these pathways (e.g., HPA axis activation affects both immunosuppression and tumor angiogenesis), so it is difficult to isolate the predominant role of a single mechanism, and there are feedback loops (e.g., inflammatory factors (e.g., IL-6) can both induce depressive symptoms and promote tumor growth) form a "feedback loop between depression and tumors" that makes it difficult to determine the direction of causality. Second, the potential blind spot of mechanism research. There is insufficient direct evidence for the brain-tumor axis, and it is known that the vagus nerve or sympathetic nerves can directly modulate the tumor microenvironment (TME) (e.g., via β-adrenergic receptors), but there is a lack of technical support for in vivo imaging on how the CNS remotely affects tumors through the neural circuits; whereas depression-associated disturbances of the intestinal flora or in certain stages of tumor development (e.g., metastatic) or specific microenvironments (e.g., areas of hyper-infiltrating T-cells) may have long-term effects on the tumors, but such changes are difficult to capture in short-term experiments and cannot be precisely temporally resolved by existing technologies. However, there are limitations in current research methods. Existing studies have relied on mouse models of chronic stress (e.g., chronic unpredictable stress), but the "depression-like behaviour" of mice is fundamentally different from the clinical manifestations of depression in humans, and the TME (e.g., immune composition) is different from that of humans. Finally, for patients with cancer-associated depression, clinical treatment is usually a two-pronged strategy, but the combination of anticancer and antidepressant drugs has limitations, such as drug-drug interactions, safety issues, and the challenge of individualised treatment in clinical practice. Therefore, by elucidating the relationship between depression and tumour bidirectional effects, this review relatively clarifies how depression affects TME to promote tumour progression by influencing changes in immunosuppression, hormonal changes, glutamate/glutamate receptors, and intestinal flora. Further, some potential therapeutic strategies are proposed for the clinical treatment of this group of patients through the above pathological mechanism; at the same time, it was found that antidepressant drugs have potential antitumor activity, and their dual pharmacological effects may provide synergistic therapeutic benefits for patients with cancer-associated depressive disorders. This finding not only expands the choice of drugs for tumour therapy but also provides a new theoretical basis for comprehensive treatment strategies in the field of psycho-oncology.
Collapse
Affiliation(s)
- Jingjing Dong
- Department of Pharmacy, Ningxia Medical University, Yin Chuan, China
| | - Juan Du
- Department of Pharmacy, Ningxia Medical University, Yin Chuan, China
| | - Ruyun Liu
- Department of Pharmacy, Ningxia Medical University, Yin Chuan, China
| | - Xinghua Gao
- Center for New Drug Safety Evaluation and Research, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Yixiao Wang
- Department of Pharmacy, Ningxia Medical University, Yin Chuan, China
| | - Lin Ma
- Department of Pharmacy, Ningxia Medical University, Yin Chuan, China
| | - Yong Yang
- Center for New Drug Safety Evaluation and Research, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Jing Wu
- College of Basic Medicine, Ningxia Medical University, Yin Chuan, China
| | - Jianqiang Yu
- Department of Pharmacy, Ningxia Medical University, Yin Chuan, China
| | - Ning Liu
- Department of Pharmacy, Ningxia Medical University, Yin Chuan, China
| |
Collapse
|
4
|
Sun J, Song S, Liu J, Chen F, Li X, Wu G. Gut microbiota as a new target for anticancer therapy: from mechanism to means of regulation. NPJ Biofilms Microbiomes 2025; 11:43. [PMID: 40069181 PMCID: PMC11897378 DOI: 10.1038/s41522-025-00678-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 03/04/2025] [Indexed: 03/15/2025] Open
Abstract
In order to decipher the relationship between gut microbiota imbalance and cancer, this paper reviewed the role of intestinal microbiota in anticancer therapy and related mechanisms, discussed the current research status of gut microbiota as a biomarker of cancer, and finally summarized the reasonable means of regulating gut microbiota to assist cancer therapy. Overall, our study reveals that the gut microbiota can serve as a potential target for improving cancer management.
Collapse
Affiliation(s)
- Jiaao Sun
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Shiyan Song
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jiahua Liu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Feng Chen
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, China.
| | - Xiaorui Li
- Department of oncology, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China.
| | - Guangzhen Wu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, China.
| |
Collapse
|
5
|
Abbas M, Tangney M. The oncobiome; what, so what, now what? MICROBIOME RESEARCH REPORTS 2025; 4:16. [PMID: 40207280 PMCID: PMC11977386 DOI: 10.20517/mrr.2024.89] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 02/14/2025] [Accepted: 02/21/2025] [Indexed: 04/11/2025]
Abstract
Microbial communities inhabiting various body sites play critical roles in the initiation, progression, and treatment of cancer. The gut microbiota, a highly diverse microbial ecosystem, interacts with immune cells to modulate inflammation and immune surveillance, influencing cancer risk and therapeutic outcomes. Local tissue microbiota may impact the transition from premalignant states to malignancy. Characterization of the intratumoral microbiota increasingly reveals distinct microbiomes that may influence tumor growth, immune responses, and treatment efficacy. Various bacteria species have been reported to modulate cancer therapies through mechanisms such as altering drug metabolism and shaping the tumor microenvironment (TME). For instance, gut or intratumoral bacterial enzymatic activity can convert prodrugs into active forms, enhancing therapeutic effects or, conversely, inactivating small-molecule chemotherapeutics. Specific bacterial species have also been linked to improved responses to immunotherapy, underscoring the microbiome's role in treatment outcomes. Furthermore, unique microbial signatures in cancer patients, compared with healthy individuals, demonstrate the diagnostic potential of microbiota. Beyond the gut, tumor-associated and local microbiomes also affect therapy by influencing inflammation, tumor progression, and drug resistance. This review explores the multifaceted relationships between microbiomes and cancer, focusing on their roles in modulating the TME, immune activation, and treatment efficacy. The diagnostic and therapeutic potential of bacterial members of microbiota represents a promising avenue for advancing precision oncology and improving patient outcomes. By leveraging microbial biomarkers and interventions, new strategies can be developed to optimize cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Munawar Abbas
- APC Microbiome Ireland, University College Cork, Cork, T12 YT20, Ireland
- Cancer Research@UCC, University College Cork, Cork, T12 XF62, Ireland
| | - Mark Tangney
- APC Microbiome Ireland, University College Cork, Cork, T12 YT20, Ireland
- Cancer Research@UCC, University College Cork, Cork, T12 XF62, Ireland
| |
Collapse
|
6
|
Han S, Luo Y, Hu Z, Li X, Zhou Y, Luo F. Tumor Microenvironment Targeted by Polysaccharides in Cancer Prevention: Expanding Roles of Gut Microbiota and Metabolites. Mol Nutr Food Res 2025; 69:e202400750. [PMID: 39757562 DOI: 10.1002/mnfr.202400750] [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: 09/15/2024] [Revised: 10/31/2024] [Accepted: 12/02/2024] [Indexed: 01/07/2025]
Abstract
Since the development of immune checkpoint inhibitors (ICIs), immunotherapy has been widely used as a novel cancer treatment. However, the efficacy of tumor immunotherapy is largely dependent on the tumor microenvironment (TME). The high degree of heterogeneity within TME remains a major obstacle to acquire satisfactory therapeutic. Emerging studies suggest that gut microbiota is becoming an important regulator of TME. Polysaccharides as tumor immunotherapeutic agents or immune adjuvants not only exhibit antitumor activity by targeting gut microbiota, but also expand their role in the tumor immunotherapy by remodeling TME. To date, the mechanism by which polysaccharides targeting TME for tumor prevention via gut microbiota has not been deeply investigated. In this review, recent advances in the regulation of TME by polysaccharides through gut microbiota were systematically outlined, and the challenges and possible solutions in the clinical application of TME-targeted polysaccharides were discussed. Exploring the relationship between polysaccharides and TME from the perspective of gut microbiota may provide new ideas for the application of polysaccharides in tumor immunotherapy. This is a new area with major challenges that deserve further exploration.
Collapse
Affiliation(s)
- Shuai Han
- Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, Central South University of Forestry and Technology, Changsha, Hunan, China
- College of Tea and Food, Wuyi University, Wuyishan, Fujian, China
| | - Yi Luo
- Department of Gastroenterology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zuomin Hu
- Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, Central South University of Forestry and Technology, Changsha, Hunan, China
| | - Xinhua Li
- Department of Gastroenterology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yaping Zhou
- Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, Central South University of Forestry and Technology, Changsha, Hunan, China
| | - Feijun Luo
- Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, Central South University of Forestry and Technology, Changsha, Hunan, China
| |
Collapse
|
7
|
Li Y, Huang H, Xie H, Cao R, Li X, Huang F, Lin L, Chen L. Akkermansia muciniphila activates natural killer cells by suppressing the TGF-β signaling pathway in lung adenocarcinoma cells. Cytokine 2025; 186:156833. [PMID: 39700665 DOI: 10.1016/j.cyto.2024.156833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 12/09/2024] [Accepted: 12/09/2024] [Indexed: 12/21/2024]
Abstract
Lung adenocarcinoma (LUAD) stands out as a prevalent malignant tumor necessitating innovative strategies to enhance therapeutic outcomes. Akkermansia muciniphila (AKK) has emerged as intricately linked to tumor immunotherapy, yet its impact on natural killer (NK) cells, which play a crucial role in immunotherapy, remains unclear. This study aims to investigate the effects of AKK outer membrane proteins on NK cells in LUAD and elucidate potential associated molecular mechanisms. 16S rRNA sequencing was employed to analyze bacterial genera and their abundance in fecal samples from LUAD patients. Co-culturing of NK-92 cells with LUAD cells, with or without treatment of AKK outer membrane protein Amuc_1100, was conducted to investigate the mechanisms of AKK on LUAD. Additionally, a xenograft mouse model was established to validate the effects of AKK in an in vivo setting. The experimental findings indicated that LUAD patients with elevated AKK levels in their fecal samples demonstrated increased NK cell infiltration and reduced TGF-β levels. Treatment with Amuc_1100 elevated TNF-α and IL-15 cytokine levels, decreased TGF-β levels and proteins associated with TGF-β pathway, enhanced NK cell cytotoxicity, upregulated perforin and granzyme B expression, induced apoptosis and cell cycle arrest, thereby inhibiting cancer cell proliferation. Amuc_1100 also impeded tumor growth in vivo. In summary, these results suggest that AKK activates NK cells to target tumor cells by suppressing the TGF-β signaling pathway in LUAD cells, underscoring the potential of Akk as an effective immunotherapeutic agent in LUAD NK cell-directed therapies.
Collapse
Affiliation(s)
- Yong Li
- Department of Pulmonary and Critical Care Medicine, Fujian Medical University Union Hospital, Fuzhou, Fujian 350000, China
| | - Huiqin Huang
- Fujian Provincial Key Laboratory of Medical Testing, Fujian Academy Of Medical Sciences, Fuzhou, Fujian 350000, China
| | - Hang Xie
- Department of Interventional Radiology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350000, China
| | - Rongxiang Cao
- Department of Pulmonary and Critical Care Medicine, Fujian Medical University Union Hospital, Fuzhou, Fujian 350000, China
| | - Xiuling Li
- Department of Pulmonary and Critical Care Medicine, Fujian Medical University Union Hospital, Fuzhou, Fujian 350000, China
| | - Feijian Huang
- Department of Pulmonary and Critical Care Medicine, Fujian Medical University Union Hospital, Fuzhou, Fujian 350000, China
| | - Lu Lin
- Department of Pulmonary and Critical Care Medicine, Fujian Medical University Union Hospital, Fuzhou, Fujian 350000, China
| | - Limin Chen
- Department of Pulmonary and Critical Care Medicine, Fujian Medical University Union Hospital, Fuzhou, Fujian 350000, China.
| |
Collapse
|
8
|
Singh DD, Haque S, Kim Y, Han I, Yadav DK. Remodeling of tumour microenvironment: strategies to overcome therapeutic resistance and innovate immunoengineering in triple-negative breast cancer. Front Immunol 2024; 15:1455211. [PMID: 39720730 PMCID: PMC11666570 DOI: 10.3389/fimmu.2024.1455211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 10/31/2024] [Indexed: 12/26/2024] Open
Abstract
Triple-negative breast cancer (TNBC) stands as the most complex and daunting subtype of breast cancer affecting women globally. Regrettably, treatment options for TNBC remain limited due to its clinical complexity. However, immunotherapy has emerged as a promising avenue, showing success in developing effective therapies for advanced cases and improving patient outcomes. Improving TNBC treatments involves reducing side effects, minimizing systemic toxicity, and enhancing efficacy. Unlike traditional cancer immunotherapy, engineered nonmaterial's can precisely target TNBC, facilitating immune cell access, improving antigen presentation, and triggering lasting immune responses. Nanocarriers with enhanced sensitivity and specificity, specific cellular absorption, and low toxicity are gaining attention. Nanotechnology-driven immunoengineering strategies focus on targeted delivery systems using multifunctional molecules for precise tracking, diagnosis, and therapy in TNBC. This study delves into TNBC's tumour microenvironment (TME) remodeling, therapeutic resistance, and immunoengineering strategies using nanotechnology.
Collapse
Affiliation(s)
- Desh Deepak Singh
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, India
| | - Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan, Saudi Arabia
| | - Youngsun Kim
- Department of Obstetrics and Gynecology, Kyung Hee University Medical Center, Seoul, Republic of Korea
| | - Ihn Han
- Plasma Bioscience Research Center, Applied Plasma Medicine Center, Department of Electrical & Biological Physics, Kwangwoon University, Seoul, Republic of Korea
| | - Dharmendra Kumar Yadav
- Department of Biologics, College of Pharmacy, Hambakmoeiro 191, Yeonsu-gu, Incheon, Republic of Korea
| |
Collapse
|
9
|
Qasem HH, El-Sayed WM. The bacterial microbiome and cancer: development, diagnosis, treatment, and future directions. Clin Exp Med 2024; 25:12. [PMID: 39607612 PMCID: PMC11604675 DOI: 10.1007/s10238-024-01523-9] [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: 06/14/2024] [Accepted: 11/11/2024] [Indexed: 11/29/2024]
Abstract
The term "microbiome" refers to the collection of bacterial species that reside in the human body's tissues. Sometimes, it is used to refer to all microbial entities (bacteria, viruses, fungi, and others) which colonize the human body. It is now generally acknowledged that the microbiome plays a critical role in the host's physiological processes and general well-being. Changes in the structure and/or function of the microbiome (dysbiosis) are linked to the development of many diseases including cancer. The claim that because of their negatively charged membrane, cancer cells are more vulnerable to some bacteria than normal cells and that is how the link between these bacteria and cancer evolved has been refuted. Furthermore, the relationship between the microbiome and cancer is more evident in the emerging field of cancer immunotherapy. In this narrative review, we detailed the correlation between the presence/absence of specific bacterial species and the development, diagnosis, prognosis, and treatment of some types of cancer including colorectal, lung, breast, and prostate cancer. In addition, we discussed the mechanisms of microbiome-cancer interactions including genotoxin production, the role of free radicals, modification of signaling pathways in host cells, immune modulation, and modulation of drug metabolism by microbiome. Future directions and clinical application of microbiome in the early detection, prognosis, and treatment of cancer emphasizing on the role of fecal transplantation, probiotics, prebiotics, and microbiome biomarkers were also considered.
Collapse
Affiliation(s)
- Hasnaa H Qasem
- Department of Zoology, Faculty of Science, Ain Shams University, Abbassia, Cairo, 11566, Egypt
| | - Wael M El-Sayed
- Department of Zoology, Faculty of Science, Ain Shams University, Abbassia, Cairo, 11566, Egypt.
| |
Collapse
|
10
|
Roszkowska M. Multilevel Mechanisms of Cancer Drug Resistance. Int J Mol Sci 2024; 25:12402. [PMID: 39596466 PMCID: PMC11594576 DOI: 10.3390/ijms252212402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 11/14/2024] [Accepted: 11/17/2024] [Indexed: 11/28/2024] Open
Abstract
Cancer drug resistance represents one of the most significant challenges in oncology and manifests through multiple interconnected molecular and cellular mechanisms. Objective: To provide a comprehensive analysis of multilevel processes driving treatment resistance by integrating recent advances in understanding genetic, epigenetic, and microenvironmental factors. This is a systematic review of the recent literature focusing on the mechanisms of cancer drug resistance, including genomic studies, clinical trials, and experimental research. Key findings include the following: (1) Up to 63% of somatic mutations can be heterogeneous within individual tumors, contributing to resistance development; (2) cancer stem cells demonstrate enhanced DNA repair capacity and altered metabolic profiles; (3) the tumor microenvironment, including cancer-associated fibroblasts and immune cell populations, plays a crucial role in promoting resistance; and (4) selective pressure from radiotherapy drives the emergence of radioresistant phenotypes through multiple adaptive mechanisms. Understanding the complex interplay between various resistance mechanisms is essential for developing effective treatment strategies. Future therapeutic approaches should focus on combination strategies that target multiple resistance pathways simultaneously, guided by specific biomarkers.
Collapse
Affiliation(s)
- Malgorzata Roszkowska
- Department of Clinical Neuropsychology, Collegium Medicum, Nicolaus Copernicus University, 85-067 Bydgoszcz, Poland
| |
Collapse
|
11
|
Lin G, Tian F, Yu Q, Weng X, Yu N, Zhang F, Yi C, Ye J, Ye D. IL-17RA/CTSK axis mediates H. pylori-induced castration-resistant prostate cancer growth. Oncogene 2024; 43:3598-3616. [PMID: 39424989 DOI: 10.1038/s41388-024-03169-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 09/11/2024] [Accepted: 09/13/2024] [Indexed: 10/21/2024]
Abstract
In this investigation, we explored the molecular dynamics guiding the progression of castration-resistant prostate cancer (CRPC) influenced by Helicobacter pylori (H. pylori)-mediated M2 polarization of macrophages through the IL-17RA/CTSK/EMT axis. An 830-patient clinical trial categorized subjects into hormone-sensitive prostate cancer (HSPC) and CRPC groups. H. pylori infection, evaluated by ELISA, exhibited a higher incidence in CRPC patients, impacting overall survival (OS) and progression-free survival. In-depth in vitro and in vivo experiments, including 16S rDNA sequencing, immunohistochemical tests, and transcriptome analysis, unveiled that H. pylori promotes CRPC growth and metastasis by upregulating IL-17RA and CTSK, leading to enhanced EMT. Notably, M2 macrophages emerged as pivotal immune cells influencing CRPC progression. This study uncovers a novel pathway wherein H. pylori enrichment exacerbates CRPC by inducing macrophage M2 polarization, IL-17RA/CTSK expression, and EMT activation, shedding light on a previously unrecognized mechanism contributing to the growth and metastasis of CRPC.
Collapse
Affiliation(s)
- Guowen Lin
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Feng Tian
- Department Of Urology, Shanghai Eighth People's Hospital, Shanghai, 200235, China
| | - Qiwei Yu
- Department of Urology, Kunshan Hospital of Traditional Chinese Medicine, Suzhou, 215399, China
| | - Xiaoling Weng
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200001, China
| | - Nanhui Yu
- Department of Gastrointestinal Surgery, the Second Xiangya Hospital of Central South University, Changsha, 410011, China
| | - Feng Zhang
- Department Of Urology, Shanghai Eighth People's Hospital, Shanghai, 200235, China
| | - Chen Yi
- Department of Urology, Changsha Central Hospital Affiliated to University of South China, Changsha, 410000, China
| | - Jian Ye
- Department of Immuno-Oncology, Beckman Research Institute of the City of Hope, Duarte, CA, USA
| | - Dingwei Ye
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
12
|
Li Q, Geng S, Luo H, Wang W, Mo YQ, Luo Q, Wang L, Song GB, Sheng JP, Xu B. Signaling pathways involved in colorectal cancer: pathogenesis and targeted therapy. Signal Transduct Target Ther 2024; 9:266. [PMID: 39370455 PMCID: PMC11456611 DOI: 10.1038/s41392-024-01953-7] [Citation(s) in RCA: 54] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 07/25/2024] [Accepted: 08/16/2024] [Indexed: 10/08/2024] Open
Abstract
Colorectal cancer (CRC) remains one of the leading causes of cancer-related mortality worldwide. Its complexity is influenced by various signal transduction networks that govern cellular proliferation, survival, differentiation, and apoptosis. The pathogenesis of CRC is a testament to the dysregulation of these signaling cascades, which culminates in the malignant transformation of colonic epithelium. This review aims to dissect the foundational signaling mechanisms implicated in CRC, to elucidate the generalized principles underpinning neoplastic evolution and progression. We discuss the molecular hallmarks of CRC, including the genomic, epigenomic and microbial features of CRC to highlight the role of signal transduction in the orchestration of the tumorigenic process. Concurrently, we review the advent of targeted and immune therapies in CRC, assessing their impact on the current clinical landscape. The development of these therapies has been informed by a deepening understanding of oncogenic signaling, leading to the identification of key nodes within these networks that can be exploited pharmacologically. Furthermore, we explore the potential of integrating AI to enhance the precision of therapeutic targeting and patient stratification, emphasizing their role in personalized medicine. In summary, our review captures the dynamic interplay between aberrant signaling in CRC pathogenesis and the concerted efforts to counteract these changes through targeted therapeutic strategies, ultimately aiming to pave the way for improved prognosis and personalized treatment modalities in colorectal cancer.
Collapse
Affiliation(s)
- Qing Li
- The Shapingba Hospital, Chongqing University, Chongqing, China
- Chongqing Key Laboratory of Intelligent Oncology for Breast Cancer, Chongqing University Cancer Hospital and School of Medicine, Chongqing University, Chongqing, China
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Shan Geng
- Central Laboratory, The Affiliated Dazu Hospital of Chongqing Medical University, Chongqing, China
| | - Hao Luo
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
- Cancer Center, Daping Hospital, Army Medical University, Chongqing, China
| | - Wei Wang
- Chongqing Municipal Health and Health Committee, Chongqing, China
| | - Ya-Qi Mo
- Chongqing Key Laboratory of Intelligent Oncology for Breast Cancer, Chongqing University Cancer Hospital and School of Medicine, Chongqing University, Chongqing, China
| | - Qing Luo
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Lu Wang
- Chongqing Key Laboratory of Intelligent Oncology for Breast Cancer, Chongqing University Cancer Hospital and School of Medicine, Chongqing University, Chongqing, China
| | - Guan-Bin Song
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China.
| | - Jian-Peng Sheng
- College of Artificial Intelligence, Nanjing University of Aeronautics and Astronautics, Nanjing, China.
| | - Bo Xu
- Chongqing Key Laboratory of Intelligent Oncology for Breast Cancer, Chongqing University Cancer Hospital and School of Medicine, Chongqing University, Chongqing, China.
| |
Collapse
|
13
|
Liu D, Zhou J, Fu Q, Zhao Y, Wang P, Zheng Y, Cui M, Zhang H. A Bioinformatic Analysis of Gut Microbiota Related with Immune Cell Infiltration in Colorectal Cancer. Cancer Invest 2024; 42:491-499. [PMID: 38905519 DOI: 10.1080/07357907.2024.2368233] [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: 01/03/2023] [Accepted: 06/11/2024] [Indexed: 06/23/2024]
Abstract
OBJECTIVE The composition of microbiota which correlates with infiltrating immune cells and clinical signatures is not clarified in CRC. METHODS We applied 4 kinds of bioinformatic tools GSVA (version: 1.42.0), ESTIMATE (version: 1.0.13), CIBERSORT (version: 2.0), and immune-related genes. RESULTS We found that a total of 8 types of microbiotas appeared in the three immune correlation analyses. Among these microbiotas, significant enrichments in relative abundances associated with immune cell infiltration can be found for the dominant phyla Proteobacteria, Firmicutes, and Actinobacteria. Moreover, there existed correlations between some of the 8 microbiotas and clinical-related indicators. CONCLUSION We identified some novel microbiotas involved in immune regulation in CRC.
Collapse
Affiliation(s)
- Dan Liu
- Department of Ultrasound Imaging, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, China
| | - Jiang Zhou
- Department of Medicine, Tianjin Georigin Biology Co., Ltd, Tianjin, China
| | - Qiong Fu
- Department of Ultrasound Imaging, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, China
| | - Yuanzhu Zhao
- Department of Ultrasound Imaging, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, China
| | - Panpan Wang
- Department of Ultrasound Imaging, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, China
| | - Yang Zheng
- Department of Ultrasound Imaging, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, China
| | - Meihong Cui
- Department of Ultrasound Imaging, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, China
| | - Heng Zhang
- Department of Ultrasound Imaging, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, China
| |
Collapse
|
14
|
Liu Y, Zhang T, Pan K, Wei H. Mechanisms and therapeutic research progress in intestinal fibrosis. Front Med (Lausanne) 2024; 11:1368977. [PMID: 38947241 PMCID: PMC11211380 DOI: 10.3389/fmed.2024.1368977] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 06/05/2024] [Indexed: 07/02/2024] Open
Abstract
Intestinal fibrosis is a common complication of chronic intestinal diseases with the characteristics of fibroblast proliferation and extracellular matrix deposition after chronic inflammation, leading to lumen narrowing, structural and functional damage to the intestines, and life inconvenience for the patients. However, anti-inflammatory drugs are currently generally not effective in overcoming intestinal fibrosis making surgery the main treatment method. The development of intestinal fibrosis is a slow process and its onset may be the result of the combined action of inflammatory cells, local cytokines, and intestinal stromal cells. The aim of this study is to elucidate the pathogenesis [e.g., extracellular matrix (ECM), cytokines and chemokines, epithelial-mesenchymal transition (EMT), differentiation of fibroblast to myofibroblast and intestinal microbiota] underlying the development of intestinal fibrosis and to explore therapeutic advances (such as regulating ECM, cytokines, chemokines, EMT, differentiation of fibroblast to myofibroblast and targeting TGF-β) based on the pathogenesis in order to gain new insights into the prevention and treatment of intestinal fibrosis.
Collapse
Affiliation(s)
- Yanjiang Liu
- School of Basic Medical Sciences, Chengdu Medical College, Chengdu, China
| | - Tao Zhang
- School of Bioscience and Technology, Chengdu Medical College, Chengdu, China
| | - Kejian Pan
- School of Bioscience and Technology, Chengdu Medical College, Chengdu, China
| | - He Wei
- School of Bioscience and Technology, Chengdu Medical College, Chengdu, China
| |
Collapse
|
15
|
Acharjee A, Okyere D, Nath D, Nagar S, Gkoutos GV. Network dynamics and therapeutic aspects of mRNA and protein markers with the recurrence sites of pancreatic cancer. Heliyon 2024; 10:e31437. [PMID: 38803850 PMCID: PMC11128524 DOI: 10.1016/j.heliyon.2024.e31437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 05/11/2024] [Accepted: 05/15/2024] [Indexed: 05/29/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a deadly disease that typically manifests late patient presentation and poor outcomes. Furthermore, PDAC recurrence is a common challenge. Distinct patterns of PDAC recurrence have been associated with differential activation of immune pathway-related genes and specific inflammatory responses in their tumour microenvironment. However, the molecular associations between and within cellular components that underpin PDAC recurrence require further development, especially from a multi-omics integration perspective. In this study, we identified stable molecular associations across multiple PDAC recurrences and utilised integrative analytics to identify stable and novel associations via simultaneous feature selection. Spatial transcriptome and proteome datasets were used to perform univariate analysis, Spearman partial correlation analysis, and univariate analyses by Machine Learning methods, including regularised canonical correlation analysis and sparse partial least squares. Furthermore, networks were constructed for reported and new stable associations. Our findings revealed gene and protein associations across multiple PDAC recurrence groups, which can provide a better understanding of the multi-layer disease mechanisms that contribute to PDAC recurrence. These findings may help to provide novel association targets for clinical studies for constructing precision medicine and personalised surveillance tools for patients with PDAC recurrence.
Collapse
Affiliation(s)
- Animesh Acharjee
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
- MRC Health Data Research UK (HDR UK), Birmingham, United Kingdom
- Institute of Translational Medicine, University Hospitals Birmingham NHS, Foundation Trust, B15 2TT, United Kingdom
- Centre for Health Data Research, University of Birmingham, B15 2TT, United Kingdom
| | - Daniella Okyere
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Dipanwita Nath
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Shruti Nagar
- Eureka Tutorials, Muzaffarnagar, U.P., 251201, India
| | - Georgios V. Gkoutos
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
- MRC Health Data Research UK (HDR UK), Birmingham, United Kingdom
- Institute of Translational Medicine, University Hospitals Birmingham NHS, Foundation Trust, B15 2TT, United Kingdom
- Centre for Health Data Research, University of Birmingham, B15 2TT, United Kingdom
| |
Collapse
|
16
|
Shen J, Sun H, Chu J, Gong X, Liu X. Cervicovaginal microbiota: a promising direction for prevention and treatment in cervical cancer. Infect Agent Cancer 2024; 19:13. [PMID: 38641803 PMCID: PMC11027553 DOI: 10.1186/s13027-024-00573-8] [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: 02/05/2024] [Accepted: 03/18/2024] [Indexed: 04/21/2024] Open
Abstract
Cervical cancer is a common malignancy in women, with high incidence rate and mortality. Persistent infection of high-risk human papillomavirus (HPV) is the most important risk factor for cervical cancer and precancerous lesions. Cervicovaginal microbiota (CVM) plays an essential role in the defense of HPV infections and prevention of subsequent lesions. Dominance of Lactobacillus is the key of CVM homeostasis, which can be regulated by host, exogenous and endogenous factors. Dysbiosis of CVM, including altered microbial, metabolic, and immune signatures, can contribute to persist HPV infection, leading to cervical cancer. However, there is no evidence of the causality between CVM and cervical cancer, and the underlying mechanism remains unexplored. Considering the close correlation between CVM dysbiosis and persistent HPV infection, this review will overview CVM, its role in cervical cancer development and related mechanisms, and the prospects for therapeutic applications.
Collapse
Affiliation(s)
- Jie Shen
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), 200003, Shanghai, China
| | - Hao Sun
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), 200003, Shanghai, China
| | - Jing Chu
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), 200003, Shanghai, China
| | - Xiaodi Gong
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), 200003, Shanghai, China.
| | - Xiaojun Liu
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), 200003, Shanghai, China.
| |
Collapse
|
17
|
Xu J, Xia Q, Wu T, Shao Y, Wang Y, Jin N, Tian P, Wu L, Lu X. Prophylactic treatment with Bacteroides uniformis and Bifidobacterium bifidum counteracts hepatic NK cell immune tolerance in nonalcoholic steatohepatitis induced by high fat diet. Gut Microbes 2024; 16:2302065. [PMID: 38196273 PMCID: PMC10793665 DOI: 10.1080/19490976.2024.2302065] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 01/02/2024] [Indexed: 01/11/2024] Open
Abstract
Hepatic immunity is one of the driving forces for the development of nonalcoholic steatohepatitis (NASH), and targeting gut microbiota is believed to affect the hepatic immune constitution. Here, we aimed to investigate the hepatic immunological state in NASH, with a specific emphasis on natural killer (NK) cells. In addition, we aimed to identify the contributing species that target hepatic immunity to provide new directions and support the feasibility of immunotherapy for NASH. A possible NASH population was determined by combination of long-term severe fatty liver, metabolic disorders and increased serum CK18 to detect serum immune factors and gut microbiota. NASH was induced in mice fed a high-fat diet to verify the prophylactic effect of the functional species on the immunopathology and development of NASH. Hepatic immunologic state was examined, and the effector functions of NK cells were detected. Hepatic transcriptome, proteomic, and fecal metagenome were performed. We observed a statistical increase in serum IL-10 (p < 0.001) and non-statistical decrease in interferon-γ and IL-6 in NASH population, hinting at the possibility of immune tolerance. Fecal Bacteroides uniformis and Bifidobacterium bifidum were abundant in healthy population but depleted in NASH patients. In NASH mice, hepatic CD8+T cells, macrophages, and dendritic cells were increased (p < 0.01), and NK cells were inhibited, which were identified with decreased granzyme B (p < 0.05). Bacteroides uniformis and Bifidobacterium bifidum improved hepatic pathological and metabolic cues, increased hepatic NK cells and reduced macrophages (p < 0.05). Bacteroides uniformis also restored hepatic NK cell function, which was identified as increased CD107a (p < 0.05). Transcriptional and translational profiling revealed that the functional species might restore the function of hepatic NK cells through multiple pathways, such as reduction of inhibitory molecules in NK cells. Bacteroides uniformis and Bifidobacterium bifidum are novel prophylactics for NASH that restore the impaired function of hepatic NK cells.
Collapse
Affiliation(s)
- Jingyuan Xu
- Department of Gastroenterology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
- Department of Gastroenterology, the Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
- Department of Gastroenterology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Qiaoyun Xia
- Department of Gastroenterology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Ting Wu
- Department of Citizen Health, Community Health Service Center of Jinxi Town, Kunshan, China
| | - Yong Shao
- Department of Citizen Health, Community Health Service Center of Jinxi Town, Kunshan, China
| | - Yatao Wang
- Department of Gastroenterology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Nuyun Jin
- Department of Gastroenterology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Peiying Tian
- Department of Gastroenterology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Longyun Wu
- Department of Gastroenterology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Xiaolan Lu
- Department of Gastroenterology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| |
Collapse
|
18
|
Roy S, Ray D, Laha I, Choudhury L. Human Mycobiota and Its Role in Cancer Progression, Diagnostics and Therapeutics: A Link Lesser-Known. Cancer Invest 2024; 42:44-62. [PMID: 38186047 DOI: 10.1080/07357907.2024.2301733] [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: 03/16/2023] [Accepted: 01/01/2024] [Indexed: 01/09/2024]
Abstract
Although not as well studied as the bacterial component of the human microbiota, the commensal fungi or mycobiota play important roles in maintaining our health by augmenting our immune system. This mycobiota is also associated with various fatal diseases like opportunistic mycoses, and even cancer, with different cancers having respective type-specific mycobiota. The different fungal species which comprise these different intratumoral mycobiota play important roles in cancer progression. The aim of this review paper is to decipher the association between mycobiota and cancer, and shed light on new avenues in cancer diagnosis, and the development of new anti-cancer therapeutics.
Collapse
Affiliation(s)
- Souvik Roy
- Post-Graduate & Research Department of Biotechnology, St. Xavier's College (Autonomous), Kolkata, India
| | - Dhrisaj Ray
- 5th Year Integrated M. Sc. (5 year integrated) students, Post-Graduate & Research Department of Biotechnology, St. Xavier's College (Autonomous), Kolkata, India
| | - Ishani Laha
- 5th Year Integrated M. Sc. (5 year integrated) students, Post-Graduate & Research Department of Biotechnology, St. Xavier's College (Autonomous), Kolkata, India
| | - Lopamudra Choudhury
- State-Aided College Teacher, Department of Microbiology, Sarsuna College (Affiliated to Calcutta University), Kolkata, India
| |
Collapse
|
19
|
Qin YM, Sha J. Progress in understanding of relationship between intestinal microecology and pancreatic cancer. Shijie Huaren Xiaohua Zazhi 2023; 31:1001-1006. [DOI: 10.11569/wcjd.v31.i24.1001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 11/30/2023] [Accepted: 12/20/2023] [Indexed: 12/28/2023] Open
Abstract
In recent years, the association between the gut microbiota (GM) and pancreatic cancer (PC) has attracted extensive attention. Studies have shown that the oral, intestinal, and pancreatic microbiota of PC patients is different from that of healthy people, showing different characteristics. On this basis, the application of characteristic GM and its metabolites as biomarkers for early diagnosis and prognosis evaluation of PC holds great potential. Intestinal microecological therapy targeting the GM, such as probiotics and fecal microbiota transplantation, may affect the response to chemotherapy and immunotherapy by remodeling the tumor microenvironment, to improve the prognosis. In this paper, we review the role of the GM in PC development, early diagnosis, prognosis assessment, and treatment.
Collapse
Affiliation(s)
- Yu-Meng Qin
- Jingjiang People's Hospital, Taizhou 214500, Jiangsu Province, China
| | - Jie Sha
- Jingjiang People's Hospital, Taizhou 214500, Jiangsu Province, China
| |
Collapse
|
20
|
Abdelhamid A, Tuminello S, Ivic-Pavlicic T, Flores R, Taioli E. Antibiotic treatment and survival in non-small cell lung cancer patients receiving immunotherapy: a systematic review and meta-analysis. Transl Lung Cancer Res 2023; 12:2427-2439. [PMID: 38205205 PMCID: PMC10775008 DOI: 10.21037/tlcr-23-597] [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: 09/21/2023] [Accepted: 12/15/2023] [Indexed: 01/12/2024]
Abstract
Background In patients with non-small cell lung cancer (NSCLC), immune checkpoint inhibitors (ICIs) are an effective mode of treatment. Despite their efficacy, responses to ICIs have been shown to differ based on several factors; for example, antibiotic use prior to and/or during immunotherapy has been associated with lower survival in NSCLC patients. The objective of this study is to provide an updated review of the literature and to fill in important knowledge gaps by accounting for potential confounding in the relationship between ICIs and survival. Methods We performed a systematic review and meta-analysis on peer-reviewed studies that examined the effects of antibiotic use on overall survival (OS) and progression-free survival (PFS) in NSCLC patients treated with ICIs. We searched MEDLINE for studies published up to June 30th, 2023 that included NSCLC patients treated with anti-programmed cell death protein 1 (PD-1) or programmed death-ligand 1 (PD-L1) agents, who received antibiotics before and/or during immunotherapy, and included a control group who did not receive antibiotics and had available data on the associations between antibiotics and OS and PFS. We calculated aggregated crude OS and PFS for all studies, and only for studies that reported multivariable hazard ratios (HRs). Risk of bias was assessed using a funnel plot. All results were synthesized and displayed using the metaphor statistical package in R, version 4.2.1. Results Nineteen studies, conducted between 2017 and 2022, met the inclusion criteria, and included 2,932 patients with advanced and/or metastatic NSCLC. Compared to those who did not receive antibiotics, immunotherapy patients who did had a significantly reduced PFS (HR: 1.22, 95% CI: 1.03-1.44) and OS (HR: 1.56, 95% CI: 1.23-1.99). Adjusted HRs were even more pronounced (OS HRadj: 1.67, 95% CI: 1.23-2.27, PFS HRadj: 1.64, 95% CI: 1.16-2.32). Conclusions NSCLC patients treated with antibiotics have significantly lowered survival compared with patients not treated with antibiotics. These results support the hypothesis that antibiotic use in conjunction with ICI among NSCLC patients lowers survival. Limitations of this analysis include the use of studies available only on a single database, limiting the literature search to NSCLC patients, which may impact the generalizability of results to other cancer patient populations, and the inability to account for and adjust the estimates for the same variables (e.g., age, sex) across all studies. Nevertheless, our findings underscore the importance of taking antibiotic use into consideration when using ICIs to treat NSCLC and suggest that confounders should be taken into account when designing future similar studies.
Collapse
Affiliation(s)
- Arwa Abdelhamid
- Institute for Translational Epidemiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Stephanie Tuminello
- Department of Population Health, NYU Grossman School of Medicine, New York, NY, USA
| | - Tara Ivic-Pavlicic
- Institute for Translational Epidemiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Raja Flores
- Department of Thoracic Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Emanuela Taioli
- Institute for Translational Epidemiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Thoracic Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
21
|
Zha X, Su S, Wu D, Zhang P, Wei Y, Fan S, Huang Q, Peng X. The impact of gut microbiota changes on the intestinal mucus barrier in burned mice: a study using 16S rRNA and metagenomic sequencing. BURNS & TRAUMA 2023; 11:tkad056. [PMID: 38130728 PMCID: PMC10734567 DOI: 10.1093/burnst/tkad056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 10/29/2023] [Accepted: 11/06/2023] [Indexed: 12/23/2023]
Abstract
Background The gut microbiota is a complex ecosystem that plays a critical role in human health and disease. However, the relationship between gut microbiota and intestinal damage caused by burns is not well understood. The intestinal mucus layer is crucial for maintaining intestinal homeostasis and providing a physiological barrier against bacterial invasion. This study aims to investigate the impact of gut microbiota on the synthesis and degradation of intestinal mucus after burns and explore potential therapeutic targets for burn injury. Methods A modified histopathological grading system was employed to investigate the effects of burn injury on colon tissue and the intestinal mucus barrier in mice. Subsequently, 16S ribosomal RNA sequencing was used to analyze alterations in the gut microbiota at days 1-10 post-burn. Based on this, metagenomic sequencing was conducted on samples collected at days 1, 5 and 10 to investigate changes in mucus-related microbiota and explore potential underlying mechanisms. Results Our findings showed that the mucus barrier was disrupted and that bacterial translocation occurred on day 3 following burn injury in mice. Moreover, the gut microbiota in mice was significantly disrupted from days 1 to 3 following burn injury, but gradually recovered to normal as the disease progressed. Specifically, there was a marked increase in the abundance of symbiotic and pathogenic bacteria associated with mucin degradation on day 1 after burns, but the abundance returned to normal on day 5. Conversely, the abundance of probiotic bacteria associated with mucin synthesis changed in the opposite direction. Further analysis revealed that after a burn injury, bacteria capable of degrading mucus may utilize glycoside hydrolases, flagella and internalins to break down the mucus layer, while bacteria that synthesize mucus may help restore the mucus layer by promoting the production of short-chain fatty acids. Conclusions Burn injury leads to disruption of colonic mucus barrier and dysbiosis of gut microbiota. Some commensal and pathogenic bacteria may participate in mucin degradation via glycoside hydrolases, flagella, internalins, etc. Probiotics may provide short-chain fatty acids (particularly butyrate) as an energy source for stressed intestinal epithelial cells, promote mucin synthesis and accelerate repair of mucus layer.
Collapse
Affiliation(s)
- Xule Zha
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Sen Su
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Dan Wu
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Panyang Zhang
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Yan Wei
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Shijun Fan
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Qianying Huang
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Xi Peng
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Gaotanyan Street, Shapingba District, Chongqing, 400038, China
- State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| |
Collapse
|
22
|
Zhang J, Hu C, Zhang R, Xu J, Zhang Y, Yuan L, Zhang S, Pan S, Cao M, Qin J, Cheng X, Xu Z. The role of macrophages in gastric cancer. Front Immunol 2023; 14:1282176. [PMID: 38143746 PMCID: PMC10746385 DOI: 10.3389/fimmu.2023.1282176] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 11/24/2023] [Indexed: 12/26/2023] Open
Abstract
As one of the deadliest cancers of the gastrointestinal tract, there has been limited improvement in long-term survival rates for gastric cancer (GC) in recent decades. The poor prognosis is attributed to difficulties in early detection, minimal opportunity for radical resection and resistance to chemotherapy and radiation. Macrophages are among the most abundant infiltrating immune cells in the GC stroma. These cells engage in crosstalk with cancer cells, adipocytes and other stromal cells to regulate metabolic, inflammatory and immune status, generating an immunosuppressive tumour microenvironment (TME) and ultimately promoting tumour initiation and progression. In this review, we summarise recent advances in our understanding of the origin of macrophages and their types and polarisation in cancer and provide an overview of the role of macrophages in GC carcinogenesis and development and their interaction with the GC immune microenvironment and flora. In addition, we explore the role of macrophages in preclinical and clinical trials on drug resistance and in treatment of GC to assess their potential therapeutic value in this disease.
Collapse
Affiliation(s)
- Jiaqing Zhang
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Gastric Surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Can Hu
- Department of Gastric Surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, China
| | - Ruolan Zhang
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Gastric Surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Jingli Xu
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Gastric Surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Yanqiang Zhang
- Department of Gastric Surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, China
| | - Li Yuan
- Department of Gastric Surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, China
| | - Shengjie Zhang
- Department of Gastric Surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, China
| | - Siwei Pan
- Department of Gastric Surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, China
| | - Mengxuan Cao
- Department of Gastric Surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, China
| | - Jiangjiang Qin
- Department of Gastric Surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, China
| | - Xiangdong Cheng
- Department of Gastric Surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, China
| | - Zhiyuan Xu
- Department of Gastric Surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, China
| |
Collapse
|
23
|
Zhou D, Li Y. Gut microbiota and tumor-associated macrophages: potential in tumor diagnosis and treatment. Gut Microbes 2023; 15:2276314. [PMID: 37943609 PMCID: PMC10653702 DOI: 10.1080/19490976.2023.2276314] [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: 07/04/2023] [Accepted: 10/24/2023] [Indexed: 11/12/2023] Open
Abstract
Avoiding immune destruction and polymorphic microbiomes are two key hallmarks of cancer. The tumor microenvironment (TME) is essential for the development of solid tumors, and the function of tumor-associated macrophages (TAMs) in the TME is closely linked to tumor prognosis. Therefore, research on TAMs could improve the progression and control of certain tumor patients. Additionally, the intestinal flora plays a crucial role in metabolizing substances and maintaining a symbiotic relationship with the host through a complex network of interactions. Recent experimental and clinical studies have suggested a potential link between gut microbiome and TME, particularly in regulating TAMs. Understanding this association could improve the efficacy of tumor immunotherapy. This review highlights the regulatory role of intestinal flora on TAMs, with a focus on gut microbiota and their metabolites. The implications of this association for tumor diagnosis and treatment are also discussed, providing a promising avenue for future clinical treatment strategies.
Collapse
Affiliation(s)
- Dongqin Zhou
- The Second Affliated Hospital & Yuying Children's Hospital / The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Yongsheng Li
- The Second Affliated Hospital & Yuying Children's Hospital / The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, China
| |
Collapse
|
24
|
Zhang Y, Gao X, Gao S, Liu Y, Wang W, Feng Y, Pei L, Sun Z, Liu L, Wang C. Effect of gut flora mediated-bile acid metabolism on intestinal immune microenvironment. Immunology 2023; 170:301-318. [PMID: 37317655 DOI: 10.1111/imm.13672] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 05/28/2023] [Indexed: 06/16/2023] Open
Abstract
According to reports, gut microbiota and metabolites regulate the intestinal immune microenvironment. In recent years, an increasing number of studies reported that bile acids (BAs) of intestinal flora origin affect T helper cells and regulatory T cells (Treg cells). Th17 cells play a pro-inflammatory role and Treg cells usually act in an immunosuppressive role. In this review, we emphatically summarised the influence and corresponding mechanism of different configurations of lithocholic acid (LCA) and deoxycholic acid (DCA) on intestinal Th17 cells, Treg cells and intestinal immune microenvironment. The regulation of BAs receptors G protein-coupled bile acid receptor 1 (GPBAR1/TGR5) and farnesoid X receptor (FXR) on immune cells and intestinal environment are elaborated. Furthermore, the potential clinical applications above were also concluded in three aspects. The above will help researchers better understand the effects of gut flora on the intestinal immune microenvironment via BAs and contribute to the development of new targeted drugs.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Henan Institute of Interconnected Intelligent Health Management, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xueyan Gao
- Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Henan Institute of Interconnected Intelligent Health Management, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Shuochen Gao
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yang Liu
- Department of Radiotherapy, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Wenkang Wang
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yudi Feng
- Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Liping Pei
- Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zhenqiang Sun
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Lin Liu
- Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Henan Institute of Interconnected Intelligent Health Management, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Chengzeng Wang
- Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Henan Institute of Interconnected Intelligent Health Management, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
25
|
Ming-bin G, Ya-nan W, Yong-ting X, Min Z, Hao T, Lian-ping Q, Feng G. TCM syndrome differentiation in colorectal cancer patients assisted by differences in gut microbiota: An exploratory study. Heliyon 2023; 9:e21057. [PMID: 37928040 PMCID: PMC10623286 DOI: 10.1016/j.heliyon.2023.e21057] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 10/12/2023] [Accepted: 10/13/2023] [Indexed: 11/07/2023] Open
Abstract
Objective To explore the difference in gut microbiota between different traditional Chinese Medicine (TCM) syndromes in patients with colorectal cancer (CRC) and its internal relationship. Methods From June 2020 to August 2021, 109 colorectal cancer patients with a clear pathological diagnosis who had not yet undergone surgery or chemotherapy were classified according to the TCM syndrome classification, and the feces samples of 109 patients with preoperative colorectal cancer were collected. 16s rRNA gene sequencing was used to determine gut microbiota diversity and abundance in CRC patients with different TCM syndrome, and LEfSe analysis was made to screen different TCM syndrome for differential representative microbiota. Results 109 patients were divided into 5 syndromes by TCM syndrome classification, which were Liver and Kidney Yin Deficiency Syndrome (LKYDS, n = 19), Spleen Deficient Qi Stagnation Syndrome (SDQSS, n = 30), Stasis and Poison Obstruction Syndrome (SPOS, n = 17), Damp-Heat Syndrome (DHS, n = 30), Qi and Blood Deficiency Syndrome (QBDS, n = 13). Alpha diversity index showed significant differences among the five groups of TCM syndromes, with Shannon index being highest in the SDQSS group and lowest in the LKYDS (p = 0.003). ACE index being highest in the SDQSS group and lowest in the SPOS (p = 0.010). PD whole tree index being highest in the SDQSS group and lowest in the SPOS (p = 0.017). Similarly, beta diversity showed significant differences among the five groups of TCM syndromes, with principal coordinate analysis (PCo1 = 31.86 %, PCo2 = 5.62 %) showing separation and coincidence between the groups, and Adonis group differences showing coincidence between the QBDS-LKYDS (p = 0.702), QBDS-DHS (p = 0.133), and SDQSS-DHS (p = 0.260) groups. LEfSe analysis revealed that the representative microbiota of DHS patients was Dialister sp Marseille P5638 (LDA = 3.05, p<0.001), the representative microbiota of SPOS patients was Oscillospirales (LDA = 4.78, p = 0.029), the representative microbiota of SDQSS patients was Selenomonadaceae (LDA = 3.94, p = 0.003), the representative microbiota of LKYDS patients was Dialister (LDA = 4.19, p = 0.001), and the representative microbiota of QBDS patients was Akkermansia muciniphila (LDA = 4.23, p = 0.006). Conclusions There are significant differences in gut microbiota between different TCM syndromes in CRC patients. The five microbiota, Dialister sp Marseille P5638, Oscillospirales, Selenomonadaceae, Dialister, and Akkermansia muciniphila, may be differential markers of TCM syndrome in CRC and are expected to be one of the bases for accurate TCM syndrome differentiation of CRC.
Collapse
Affiliation(s)
- Gui Ming-bin
- Department of Colorectal & Anal surgery, The 940th Hospital of Joint Logistics support force of Chinese people's Liberation Army, Lanzhou 730050, China
| | - Wang Ya-nan
- Department of Colorectal & Anal surgery, The 940th Hospital of Joint Logistics support force of Chinese people's Liberation Army, Lanzhou 730050, China
| | - Xue Yong-ting
- Clinical College of Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, China
| | - Zou Min
- Department of Colorectal & Anal surgery, The 940th Hospital of Joint Logistics support force of Chinese people's Liberation Army, Lanzhou 730050, China
| | - Tu Hao
- Department of Colorectal & Anal surgery, The 940th Hospital of Joint Logistics support force of Chinese people's Liberation Army, Lanzhou 730050, China
| | - Qu Lian-ping
- Department of Colorectal & Anal surgery, The 940th Hospital of Joint Logistics support force of Chinese people's Liberation Army, Lanzhou 730050, China
| | - Gao Feng
- Department of Colorectal & Anal surgery, The 940th Hospital of Joint Logistics support force of Chinese people's Liberation Army, Lanzhou 730050, China
| |
Collapse
|
26
|
Zhao X, Zhao J, Li D, Yang H, Chen C, Qin M, Wen Z, He Z, Xu L. Akkermansia muciniphila: A potential target and pending issues for oncotherapy. Pharmacol Res 2023; 196:106916. [PMID: 37690533 DOI: 10.1016/j.phrs.2023.106916] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 09/02/2023] [Accepted: 09/07/2023] [Indexed: 09/12/2023]
Abstract
In the wake of the development of metagenomic, metabolomic, and metatranscriptomic approaches, the intricate interactions between the host and various microbes are now being progressively understood. Numerous studies have demonstrated evident changes in gut microbiota during the process of a variety of diseases, such as diabetes, obesity, aging, and cancers. Notably, gut microbiota is viewed as a potential source of novel therapeutics. Currently, Next-generation probiotics (NGPs) are gaining popularity as therapeutic agents that alter the gut microbiota and affect cancer development. Akkermansia muciniphila (A. muciniphila), a representative commensal bacterium, has received substantial attention over the past decade as a promising NGP. The components and metabolites of A. muciniphila can directly or indirectly affect tumorigenesis, in particular through its effects on antitumor immunosurveillance, including the stimulation of pattern recognition receptors (PRRs), which also leads to better outcomes in a variety of situations, including the prevention and curation of cancers. In this article, we systematically summarize the role of A. muciniphila in tumorigenesis (involving gastrointestinal and non-gastrointestinal cancers) and in tumor therapy. In particular, we carefully discuss some critical scientific issues that need to be solved for the future using A. muciniphila as a representative beneficial bacterium in tumor treatment, which might provide bright clues and assistance for the application of drugs targeting A. muciniphila in clinical oncotherapy.
Collapse
Affiliation(s)
- Xu Zhao
- Guizhou University Medical College, Guiyang 550025, Guizhou Province, China; Special Key Laboratory of Gene Detection &Therapy of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou 563000, China; Department of Immunology, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Juanjuan Zhao
- Special Key Laboratory of Gene Detection &Therapy of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou 563000, China; Department of Immunology, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Dongmei Li
- Special Key Laboratory of Gene Detection &Therapy of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou 563000, China; Department of Immunology, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Han Yang
- Special Key Laboratory of Gene Detection &Therapy of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou 563000, China; Department of Immunology, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Chao Chen
- Guizhou University Medical College, Guiyang 550025, Guizhou Province, China; Special Key Laboratory of Gene Detection &Therapy of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Ming Qin
- Special Key Laboratory of Gene Detection &Therapy of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Zhenke Wen
- Institutes of Biology and Medical Sciences, Soochow Univeristy, Jiangsu 215000, China
| | - Zhixu He
- Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, Zunyi, Guizhou 563000, China.
| | - Lin Xu
- Guizhou University Medical College, Guiyang 550025, Guizhou Province, China; Special Key Laboratory of Gene Detection &Therapy of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou 563000, China; Department of Immunology, Zunyi Medical University, Zunyi, Guizhou 563000, China; Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, Zunyi, Guizhou 563000, China.
| |
Collapse
|
27
|
Xinyuan T, Lei Y, Jianping S, Rongwei Z, Ruiwen S, Ye Z, Jing Z, Chunfang T, Hongwei C, Haibin G. Advances in the role of gut microbiota in the regulation of the tumor microenvironment (Review). Oncol Rep 2023; 50:181. [PMID: 37615187 PMCID: PMC10485805 DOI: 10.3892/or.2023.8618] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 08/01/2023] [Indexed: 08/25/2023] Open
Abstract
As a protector of human health, the gut microbiota plays an important role in the development of the immune system during childhood, and the regulation of dietary habits, metabolism and immune system during adulthood. Dysregulated gut flora is not pathogenic, but it can weaken the protective effect of the immune system and cause various diseases. The tumor microenvironment is a physiological environment formed during tumor growth, which provides nutrients and growth factors necessary for tumor growth. As an important factor affecting the tumor microenvironment, the intestinal microflora affects the development of tumors through the mechanisms of gut and microflora metabolites, gene toxins and signaling pathways. The present article aimed to review the components and mechanisms of action, clinical applications, and biological targets of gut microbiota in the regulation of the tumor microenvironment. The present review provides novel insights for the future use of intestinal flora, to regulate the tumor microenvironment, to intervene in the occurrence, development, treatment and prognosis of tumors.
Collapse
Affiliation(s)
- Tian Xinyuan
- School of Pharmacy, Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region 010107, P.R. China
| | - Yu Lei
- Department of Pharmacy, Traditional Chinese Medicine Hospital of Inner Mongolia Autonomous Region, Hohhot, Inner Mongolia Autonomous Region 010020, P.R. China
| | - Shi Jianping
- School of Traditional Chinese Medicine, Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region 010107, P.R. China
| | - Zhao Rongwei
- Department of Obstetrics and Gynecology, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region 010050, P.R. China
| | - Shi Ruiwen
- School of Pharmacy, Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region 010107, P.R. China
| | - Zhang Ye
- School of Pharmacy, Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region 010107, P.R. China
| | - Zhao Jing
- School of Pharmacy, Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region 010107, P.R. China
| | - Tian Chunfang
- Department of Oncology, Traditional Chinese Medicine Hospital of Inner Mongolia Autonomous Region, Hohhot, Inner Mongolia Autonomous Region 010020, P.R. China
| | - Cui Hongwei
- Department of Scientific Research, Peking University Cancer Hospital (Inner Mongolia Campus)/Affiliated Cancer Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region 010020, P.R. China
| | - Guan Haibin
- School of Pharmacy, Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region 010107, P.R. China
| |
Collapse
|
28
|
Kong X, Zhang Y, Xiang L, You Y, Duan Y, Zhao Y, Li S, Wu R, Zhang J, Zhou L, Duan L. Fusobacterium nucleatum-triggered neutrophil extracellular traps facilitate colorectal carcinoma progression. J Exp Clin Cancer Res 2023; 42:236. [PMID: 37684625 PMCID: PMC10492297 DOI: 10.1186/s13046-023-02817-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 08/29/2023] [Indexed: 09/10/2023] Open
Abstract
BACKGROUND Fusobacterium nucleatum (Fn) acts as a procarcinogenic bacterium in colorectal carcinoma (CRC) by regulating the inflammatory tumor microenvironment (TME). Neutrophil extracellular traps (NETs), which can be generated by persistent inflammation, have been recently considered to be significant contributors in promoting cancer progression. However, whether NETs are implicated in Fn-related carcinogenesis is still poorly characterized. Here, we explored the role of NETs in Fn-related CRC as well as their potential clinical significance. METHODS Fn was measured in tissue specimens and feces samples from CRC patients. The expression of NET markers were also detected in tissue specimens, freshly isolated neutrophils and blood serum from CRC patients, and the correlation of circulating NETs levels with Fn was evaluated. Cell-based experiments were conducted to investigate the mechanism by which Fn modulates NETs formation. In addition, we clarified the functional mechanism of Fn-induced NETs on the growth and metastasis of CRC in vitro and in vivo experiments. RESULTS Tissue and blood samples from CRC patients, particularly those from Fn-infected CRC patients, exhibited greater neutrophil infiltration and higher NETs levels. Fn infection induced abundant NETs production in in vitro studies. Subsequently, we demonstrated that Fn-induced NETs indirectly accelerated malignant tumor growth through angiopoiesis, and facilitated tumor metastasis, as manifested by epithelial-mesenchymal transition (EMT)-related cell migration, matrix metalloproteinase (MMP)-mediated basement membrane protein degradation, and trapping of CRC cells. Mechanistically, the Toll-like receptor (TLR4)-reactive oxygen species (ROS) signaling pathway and NOD-like receptor (NOD1/2)-dependent signaling were responsible for Fn-stimulated NETs formation. More importantly, circulating NETs combined with carcinoembryonic antigen (CEA) could predict CRC occurrence and metastasis, with areas under the ROC curves (AUCs) of 0.92 and 0.85, respectively. CONCLUSIONS Our findings indicated that Fn-induced NETs abundance by activating TLR4-ROS and NOD1/2 signalings in neutrophils facilitated CRC progression. The combination of circulating NETs and CEA was identified as a novel screening strategy for predicting CRC occurrence and metastasis.
Collapse
Affiliation(s)
- Xuehua Kong
- Department of Laboratory Medicine, Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Chongqing Medical University, No. 1 of Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, No.74 Linjiang Road, Yu Zhong District, Chongqing, 400010, China
| | - Yu Zhang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, No.74 Linjiang Road, Yu Zhong District, Chongqing, 400010, China
| | - Linwei Xiang
- Department of Laboratory Medicine, Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Chongqing Medical University, No. 1 of Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, No.74 Linjiang Road, Yu Zhong District, Chongqing, 400010, China
| | - Yan You
- Department of Pathology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Yaqian Duan
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, No.74 Linjiang Road, Yu Zhong District, Chongqing, 400010, China
| | - Yuqing Zhao
- Department of Laboratory Medicine, Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Chongqing Medical University, No. 1 of Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Shue Li
- Department of Academic Research, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Rui Wu
- Department of Laboratory Medicine, The First Affiliated Hospital of Chonqing Medical University, Chongqing, 400016, China
| | - Jiangbo Zhang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lan Zhou
- Department of Laboratory Medicine, Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Chongqing Medical University, No. 1 of Yixueyuan Road, Yuzhong District, Chongqing, 400016, China.
| | - Liang Duan
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, No.74 Linjiang Road, Yu Zhong District, Chongqing, 400010, China.
| |
Collapse
|
29
|
Zhou M, Tang Y, Xu W, Hao X, Li Y, Huang S, Xiang D, Wu J. Bacteria-based immunotherapy for cancer: a systematic review of preclinical studies. Front Immunol 2023; 14:1140463. [PMID: 37600773 PMCID: PMC10436994 DOI: 10.3389/fimmu.2023.1140463] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 03/30/2023] [Indexed: 08/22/2023] Open
Abstract
Immunotherapy has been emerging as a powerful strategy for cancer management. Recently, accumulating evidence has demonstrated that bacteria-based immunotherapy including naive bacteria, bacterial components, and bacterial derivatives, can modulate immune response via various cellular and molecular pathways. The key mechanisms of bacterial antitumor immunity include inducing immune cells to kill tumor cells directly or reverse the immunosuppressive microenvironment. Currently, bacterial antigens synthesized as vaccine candidates by bioengineering technology are novel antitumor immunotherapy. Especially the combination therapy of bacterial vaccine with conventional therapies may further achieve enhanced therapeutic benefits against cancers. However, the clinical translation of bacteria-based immunotherapy is limited for biosafety concerns and non-uniform production standards. In this review, we aim to summarize immunotherapy strategies based on advanced bacterial therapeutics and discuss their potential for cancer management, we will also propose approaches for optimizing bacteria-based immunotherapy for facilitating clinical translation.
Collapse
Affiliation(s)
- Min Zhou
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Yucheng Tang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Wenjie Xu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Xinyan Hao
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Yongjiang Li
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Si Huang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Daxiong Xiang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Junyong Wu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Changsha, China
| |
Collapse
|
30
|
Li J, Guo Y, Liu J, Guo F, Du L, Yang Y, Li X, Ma Y. Depicting the landscape of gut microbial-metabolic interaction and microbial-host immune heterogeneity in deficient and proficient DNA mismatch repair colorectal cancers. J Immunother Cancer 2023; 11:e007420. [PMID: 37597851 PMCID: PMC10441105 DOI: 10.1136/jitc-2023-007420] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/04/2023] [Indexed: 08/21/2023] Open
Abstract
BACKGROUND Accumulating evidence has indicated the role of gut microbiota in remodeling host immune signatures, but various interplays underlying colorectal cancers (CRC) with deficient DNA mismatch repair (dMMR) and proficient DNA mismatch repair (pMMR) remain poorly understood. This study aims to decipher the gut microbiome-host immune interactions between dMMR and pMMR CRC. METHOD We performed metagenomic sequencing and metabolomic analysis of fecal samples from a cohort encompassing 455 participants, including 21 dMMR CRC, 207 pMMR CRC, and 227 healthy controls. Among them, 50 tumor samples collected from 5 dMMR CRC and 45 pMMR CRC were conducted bulk RNA sequencing. RESULTS Pronounced microbiota and metabolic heterogeneity were identified with 211 dMMR-enriched species, such as Fusobacterium nucleatum and Akkermansia muciniphila, 2 dMMR-depleted species, such as Flavonifractor plautii, 13 dMMR-enriched metabolites, such as retinoic acid, and 77 dMMR-depleted metabolites, such as lactic acid, succinic acid, and 2,3-dihydroxyvaleric acid. F. plautii was enriched in pMMR CRC and it was positively associated with fatty acid degradation, which might account for the accumulation of dMMR-depleted metabolites classified as short chain organic acid (lactic acid, succinic acid, and 2,3-dihydroxyvaleric acid) in pMMR CRC. The microbial-metabolic association analysis revealed the characterization of pMMR CRC as the accumulation of lactate induced by the depletion of specific gut microbiota which was negatively associated with antitumor immune, whereas the nucleotide metabolism and peptide degradation mediated by dMMR-enriched species characterized dMMR CRC. MMR-specific metabolic landscapes were related to distinctive immune features, such as CD8+ T cells, dendritic cells and M2-like macrophages. CONCLUSIONS Our mutiomics results delineate a heterogeneous landscape of microbiome-host immune interactions within dMMR and pMMR CRC from aspects of bacterial communities, metabolic features, and correlation with immunocyte compartment, which infers the underlying mechanism of heterogeneous immune responses.
Collapse
Affiliation(s)
- Jinming Li
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yangyang Guo
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jianqiang Liu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Endoscopy, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Fanying Guo
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lutao Du
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong province, China
| | - Yongzhi Yang
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xinxiang Li
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yanlei Ma
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
31
|
Chai Y, Huang Z, Shen X, Lin T, Zhang Y, Feng X, Mao Q, Liang Y. Microbiota Regulates Pancreatic Cancer Carcinogenesis through Altered Immune Response. Microorganisms 2023; 11:1240. [PMID: 37317214 PMCID: PMC10221276 DOI: 10.3390/microorganisms11051240] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 05/02/2023] [Accepted: 05/04/2023] [Indexed: 06/16/2023] Open
Abstract
The microbiota is present in many parts of the human body and plays essential roles. The most typical case is the occurrence and development of cancer. Pancreatic cancer (PC), one of the most aggressive and lethal types of cancer, has recently attracted the attention of researchers. Recent research has revealed that the microbiota regulates PC carcinogenesis via an altered immune response. Specifically, the microbiota, in several sites, including the oral cavity, gastrointestinal tract, and pancreatic tissue, along with the numerous small molecules and metabolites it produces, influences cancer progression and treatment by activating oncogenic signaling, enhancing oncogenic metabolic pathways, altering cancer cell proliferation, and triggering chronic inflammation that suppresses tumor immunity. Diagnostics and treatments based on or in combination with the microbiota offer novel insights to improve efficiency compared with existing therapies.
Collapse
Affiliation(s)
- Yihan Chai
- Department of General Surgery, Zhejiang University School of Medicine, Sir Run Run Shaw Hospital, Hangzhou 310016, China
| | - Zhengze Huang
- Department of General Surgery, Zhejiang University School of Medicine, Sir Run Run Shaw Hospital, Hangzhou 310016, China
| | - Xuqiu Shen
- Department of General Surgery, Zhejiang University School of Medicine, Sir Run Run Shaw Hospital, Hangzhou 310016, China
| | - Tianyu Lin
- Department of General Surgery, Zhejiang University School of Medicine, Sir Run Run Shaw Hospital, Hangzhou 310016, China
| | - Yiyin Zhang
- Department of General Surgery, Zhejiang University School of Medicine, Sir Run Run Shaw Hospital, Hangzhou 310016, China
| | - Xu Feng
- Department of General Surgery, Zhejiang University School of Medicine, Sir Run Run Shaw Hospital, Hangzhou 310016, China
| | - Qijiang Mao
- Department of General Surgery, Zhejiang University School of Medicine, Sir Run Run Shaw Hospital, Hangzhou 310016, China
- Zhejiang Provincial Key Laboratory of Laparoscopic Technology, Hangzhou 310016, China
- Zhejiang Province Medical Research Center of Minimally Invasive Diagnosis and Treatment of Abdominal Diseases, Hangzhou 310028, China
| | - Yuelong Liang
- Department of General Surgery, Zhejiang University School of Medicine, Sir Run Run Shaw Hospital, Hangzhou 310016, China
- Zhejiang Province Medical Research Center of Minimally Invasive Diagnosis and Treatment of Abdominal Diseases, Hangzhou 310028, China
| |
Collapse
|
32
|
Gao C, Wang X, Yang B, Yuan W, Huang W, Wu G, Ma J. Synergistic Target of Intratumoral Microbiome and Tumor by Metronidazole-Fluorouridine Nanoparticles. ACS NANO 2023; 17:7335-7351. [PMID: 37036121 DOI: 10.1021/acsnano.2c11305] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Clinical and experimental evidence confirmed bacterial infiltration in a variety of tumors, which is related to the progression and therapeutic effects of the tumors. Although the administration of antibiotics inhibits the growth of bacteria inside the tumor, systemic distribution of antibiotics induces an imbalance of other microbiomes in the body, which in turn leads to the development of new diseases. To address this clinical challenge, we nanonized an antibiotic in this study. Metronidazole, an antibiotic against broad anaerobes, was linked to fluorouridine to form an amphiphilic small molecule, metronidazole-fluorouridine, which further autoassembled as metronidazole-fluorouridine nanoparticles (MTI-FDU) in a hydrophilic solution. The disulfide bond in the linker cleaves in response to high levels of glutathione (GSH) in the tumor microenvironment. The synergistic antitumor effect of MTI-FDU was observed in two animal models of gut cancer with intratumoral bacteria. Analysis revealed that metronidazole delivered by nanoparticles attacked bacteria inside the tumor, while it had minimal effect on gut microbial homeostasis. Further experiments at the cellular and molecular levels disclosed that MTI-FDU shaped the tumor immune microenvironment through clearance of bacteria and bacterial products. In conclusion, we achieved a synergistic antitumor effect by a dual target of both the intratumoral microbiome and tumor cells. Antibiotic-composed nanoparticles have a clinical advantage in the treatment of tumors with bacteria infiltration, which kill pro-tumor bacteria efficiently as well as keep a balanced microbiota of the patient.
Collapse
Affiliation(s)
- Chunxiao Gao
- Department of Biotherapy, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Graduate School of Peking Union Medical College, Beijing, 100730, People's Republic of China
- Department of Clinical Laboratory, Beijing Jishuitan Hospital, Beijing, 100035, People's Republic of China
| | - Xijun Wang
- Department of Biotherapy, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Graduate School of Peking Union Medical College, Beijing, 100730, People's Republic of China
| | - Bing Yang
- Department of Biotherapy, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Graduate School of Peking Union Medical College, Beijing, 100730, People's Republic of China
| | - Wei Yuan
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, People's Republic of China
| | - Wei Huang
- School of Chemistry and Chemical Engineering, State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, People's Republic of China
| | - Guoju Wu
- Department of General Surgery, Department of Gastrointestinal Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, People's Republic of China
| | - Jie Ma
- Department of Biotherapy, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Graduate School of Peking Union Medical College, Beijing, 100730, People's Republic of China
| |
Collapse
|
33
|
Zhang C, Chen Y, Chen S, Guan X, Zhong Y, Yang Q. Occurrence, risk assessment, and in vitro and in vivo toxicity of antibiotics in surface water in China. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 255:114817. [PMID: 36963185 DOI: 10.1016/j.ecoenv.2023.114817] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 03/09/2023] [Accepted: 03/18/2023] [Indexed: 06/18/2023]
Abstract
Antibiotics have been widely detected in the water environment and thus pose a potential threat to human health. Although antibiotics have health-promoting properties, whether and how they affect health at environmental concentrations remains uncharacterised. We detected antibiotics in surface water and groundwater in China. Sulfonamides (851 ng/L) and tetracyclines (1322 ng/L) showed the highest concentrations in surface water, while the highest concentration of sulfonamides detected in groundwater was 250 ng/L. We analysed the distribution of four classes of antibiotics (sulfonamides, tetracyclines, macrolides, and quinolones) and evaluated the associated health risks in the surface water of seven cities. We found that antibiotic pollution caused health risks to the 0-3-months age group, but not to other age groups. We further demonstrated that simulated long-term exposure to environmental concentrations of antibiotics had concentration-dependent toxic effects on L-02 hepatocytes, affected cell proliferation, and induced oxidative damage and DNA damage. Chronic exposure to mixed sulfonamides affected growth, caused liver damage, and reduced the abundance of intestinal flora in mice. Under exposure to antibiotics, the abundance of Helicobacter pylori in the gut flora significantly increased and posed a health risk to humans. These results indicated that exposure to antibiotics at environmental concentrations can cause oxidative damage and inflammation both in vitro and in vivo. These findings add to the body of basic data on the distribution of antibiotics in the water environment, and provide a scientific basis for the evaluation of antibiotic toxicity.
Collapse
Affiliation(s)
- Cheng Zhang
- School of Public Health, Guangzhou Medical University, Guangzhou 511436, China
| | - Yuyang Chen
- School of Anesthesiology, Southern Medical University, Guangzhou 510515, China
| | - Sili Chen
- School of Public Health, Guangzhou Medical University, Guangzhou 511436, China
| | - Xinchao Guan
- School of Public Health, Guangzhou Medical University, Guangzhou 511436, China
| | - Yi Zhong
- Guangzhou Center for Disease Control and Prevention, Guangzhou 510440, China; Institute of Public Health, Guangzhou Medical University, Guangzhou 511436, China
| | - Qiaoyuan Yang
- School of Public Health, Guangzhou Medical University, Guangzhou 511436, China; Institute of Public Health, Guangzhou Medical University, Guangzhou 511436, China.
| |
Collapse
|
34
|
Yu H, Li XX, Han X, Chen BX, Zhang XH, Gao S, Xu DQ, Wang Y, Gao ZK, Yu L, Zhu SL, Yao LC, Liu GR, Liu SL, Mu XQ. Fecal microbiota transplantation inhibits colorectal cancer progression: Reversing intestinal microbial dysbiosis to enhance anti-cancer immune responses. Front Microbiol 2023; 14:1126808. [PMID: 37143538 PMCID: PMC10151806 DOI: 10.3389/fmicb.2023.1126808] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 03/27/2023] [Indexed: 05/06/2023] Open
Abstract
Many lines of evidence demonstrate the associations of colorectal cancer (CRC) with intestinal microbial dysbiosis. Recent reports have suggested that maintaining the homeostasis of microbiota and host might be beneficial to CRC patients, but the underlying mechanisms remain unclear. In this study, we established a CRC mouse model of microbial dysbiosis and evaluated the effects of fecal microbiota transplantation (FMT) on CRC progression. Azomethane and dextran sodium sulfate were used to induce CRC and microbial dysbiosis in mice. Intestinal microbes from healthy mice were transferred to CRC mice by enema. The vastly disordered gut microbiota of CRC mice was largely reversed by FMT. Intestinal microbiota from normal mice effectively suppressed cancer progression as assessed by measuring the diameter and number of cancerous foci and significantly prolonged survival of the CRC mice. In the intestine of mice that had received FMT, there were massive infiltration of immune cells, including CD8+ T and CD49b+ NK, which is able to directly kill cancer cells. Moreover, the accumulation of immunosuppressive cells, Foxp3+ Treg cells, seen in the CRC mice was much reduced after FMT. Additionally, FMT regulated the expressions of inflammatory cytokines in CRC mice, including down-regulation of IL1a, IL6, IL12a, IL12b, IL17a, and elevation of IL10. These cytokines were positively correlated with Azospirillum_sp._47_25, Clostridium_sensu_stricto_1, the E. coli complex, Akkermansia, Turicibacter, and negatively correlated with Muribaculum, Anaeroplasma, Candidatus_Arthromitus, and Candidatus Saccharimonas. Furthermore, the repressed expressions of TGFb, STAT3 and elevated expressions of TNFa, IFNg, CXCR4 together promoted the anti-cancer efficacy. Their expressions were positively correlated with Odoribacter, Lachnospiraceae-UCG-006, Desulfovibrio, and negatively correlated with Alloprevotella, Ruminococcaceae UCG-014, Ruminiclostridium, Prevotellaceae UCG-001 and Oscillibacter. Our studies indicate that FMT inhibits the development of CRC by reversing gut microbial disorder, ameliorating excessive intestinal inflammation and cooperating with anti-cancer immune responses.
Collapse
Affiliation(s)
- Hao Yu
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical, University, Harbin, China
- HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, Heilongjiang, China
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, Heilongjiang, China
| | - Xing-Xiu Li
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical, University, Harbin, China
| | - Xing Han
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical, University, Harbin, China
- HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, Heilongjiang, China
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, Heilongjiang, China
| | - Bin-Xin Chen
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical, University, Harbin, China
| | - Xing-Hua Zhang
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical, University, Harbin, China
- HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, Heilongjiang, China
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, Heilongjiang, China
| | - Shan Gao
- Pathology Department, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Dan-Qi Xu
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical, University, Harbin, China
- HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, Heilongjiang, China
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, Heilongjiang, China
| | - Yao Wang
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical, University, Harbin, China
- HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, Heilongjiang, China
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, Heilongjiang, China
| | - Zhan-Kui Gao
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical, University, Harbin, China
- HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, Heilongjiang, China
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, Heilongjiang, China
| | - Lei Yu
- Department of Colorectal Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Song-Ling Zhu
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical, University, Harbin, China
- HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, Heilongjiang, China
| | - Li-Chen Yao
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical, University, Harbin, China
| | - Gui-Rong Liu
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical, University, Harbin, China
- HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, Heilongjiang, China
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, Heilongjiang, China
- *Correspondence: Xiao-Qin Mu, ; Shu-Lin Liu, ; Gui-Rong Liu,
| | - Shu-Lin Liu
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical, University, Harbin, China
- HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, Heilongjiang, China
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, Heilongjiang, China
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, AB, Canada
- *Correspondence: Xiao-Qin Mu, ; Shu-Lin Liu, ; Gui-Rong Liu,
| | - Xiao-Qin Mu
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical, University, Harbin, China
- HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, Heilongjiang, China
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, Heilongjiang, China
- *Correspondence: Xiao-Qin Mu, ; Shu-Lin Liu, ; Gui-Rong Liu,
| |
Collapse
|
35
|
Chang L, Qiu L, Lei N, Zhou J, Guo R, Gao F, Dong S, Chen M, Wu F, Qin B. Characterization of fecal microbiota in cervical cancer patients associated with tumor stage and prognosis. Front Cell Infect Microbiol 2023; 13:1145950. [PMID: 36909733 PMCID: PMC9995373 DOI: 10.3389/fcimb.2023.1145950] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 02/13/2023] [Indexed: 02/25/2023] Open
Abstract
Cervical cancer (CC) is the fourth most frequent malignancy among women worldwide, and its prevention and treatment are evolving rapidly. The gut microbiota has been reported to play a crucial role both in the preservation of homeostasis and the development of cervical cancer. In this study, we collected fecal samples to investigate the microbial signatures in cervical cancer patients compared with healthy controls using 16S rRNA sequencing analysis and metagenomic next-generation sequencing (mNGS) testing. Our findings demonstrated a substantial difference in the gut microbiota composition of cervical cancer patients and healthy controls. The disease and stage were most significantly negatively correlated with Ruminococcus 2, which might be considered a potential clinically relevant biomarker. Functions of differential microbiomes were also analyzed, indicating significant differences in metabolisms and biosynthesis between the two groups. These findings demonstrate that patients with cervical cancer have certain species of gut microbiota that are exclusive to them and particular species have the potential to be used in the prognosis of cervical cancer.
Collapse
Affiliation(s)
- Lei Chang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China
- *Correspondence: Lei Chang,
| | - Luojie Qiu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China
| | - Ningjing Lei
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Junying Zhou
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China
| | - Ruixia Guo
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China
| | - Feng Gao
- Department of Neuroimmunology, Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Henan Engineering Technology Research Center for Accurate Diagnosis Neuroimmunity, Zhengzhou, Henan, China
| | - Shiliang Dong
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Mengyu Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China
| | - Fengling Wu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China
| | - Bo Qin
- Translational Medical Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
36
|
Li T, Han L, Ma S, Lin W, Ba X, Yan J, Huang Y, Tu S, Qin K. Interaction of gut microbiota with the tumor microenvironment: A new strategy for antitumor treatment and traditional Chinese medicine in colorectal cancer. Front Mol Biosci 2023; 10:1140325. [PMID: 36950522 PMCID: PMC10025541 DOI: 10.3389/fmolb.2023.1140325] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 02/23/2023] [Indexed: 03/08/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most common malignancies worldwide and the second leading cause of cancer-related death. In recent years, the relationship between gut microbiota and CRC has attracted increasing attention from researchers. Studies reported that changes in the composition of gut microbiota, such as increase in the number of Fusobacterium nucleatum and Helicobacter hepaticus, impair the immune surveillance by affecting the intestinal mucosal immunity and increase the risk of tumor initiation and progression. The tumor microenvironment is the soil for tumor survival. Close contacts between gut microbiota and the tumor microenvironment may directly affect the progression of tumors and efficacy of antitumor drugs, thus influencing the prognosis of patients with CRC. Recently, many studies have shown that traditional Chinese medicine can safely and effectively improve the efficacy of antitumor drugs, potentially through remodeling of the tumor microenvironment by regulated gut microbiota. This article describes the effect of gut microbiota on the tumor microenvironment and possible mechanisms concerning the initiation and progression of CRC, and summarizes the potential role of traditional Chinese medicine.
Collapse
Affiliation(s)
- Tingting Li
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liang Han
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Simin Ma
- Department of Nosocomial Infection Management, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weiji Lin
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Ba
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiahui Yan
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ying Huang
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shenghao Tu
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kai Qin
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Kai Qin,
| |
Collapse
|
37
|
Li Z, Ke X, Zuo D, Wang Z, Fang F, Li B. New Insights into the Relationship between Gut Microbiota and Radiotherapy for Cancer. Nutrients 2022; 15:nu15010048. [PMID: 36615706 PMCID: PMC9824372 DOI: 10.3390/nu15010048] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 12/18/2022] [Accepted: 12/19/2022] [Indexed: 12/25/2022] Open
Abstract
Cancer is the second most common cause of death among humans in the world, and the threat that it presents to human health is becoming more and more serious. The mechanisms of cancer development have not yet been fully elucidated, and new therapies are changing with each passing day. Evidence from the literature has validated the finding that the composition and modification of gut microbiota play an important role in the development of many different types of cancer. The results also demonstrate that there is a bidirectional interaction between the gut microbiota and radiotherapy treatments for cancer. In a nutshell, the modifications of the gut microbiota caused by radiotherapy have an effect on tumor radiosensitivity and, as a result, affect the efficacy of radiotherapy and show a certain radiation toxicity, which leads to numerous side effects. What is of new research significance is that the "gut-organ axis" formed by the gut microbiota may be one of the most interesting potential mechanisms, although the relevant research is still very limited. In this review, we combine new insights into the relationship between the gut microbiota, cancer, and radiotherapy. Based on our current comprehensive understanding of this relationship, we give an overview of the new cancer treatments based on the gut microbiota.
Collapse
Affiliation(s)
- Zhipeng Li
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China
| | - Xiyang Ke
- Key Laboratory of Carcinogenesis and Translational Research, Department of Radiation Oncology, Peking University Cancer Hospital and Institute, Ministry of Education, Beijing 100142, China
| | - Dan Zuo
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China
| | - Zhicheng Wang
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China
| | - Fang Fang
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China
- School of Public Health, Jilin University, Changchun 130021, China
- Correspondence: ; Tel.: +86-431-85619455
| | - Bo Li
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China
- School of Public Health, Jilin University, Changchun 130021, China
| |
Collapse
|
38
|
Xia T, Wang N, Tang Y, Gao Y, Gao C, Hao J, Jiang Y, Wang X, Shan Z, Li J, Zhou H, Cui W, Qiao X, Tang L, Wang L, Li Y. Delivery of antigen to porcine dendritic cells by fusing antigen with porcine dendritic cells targeting peptide. Front Immunol 2022; 13:926279. [PMID: 36159835 PMCID: PMC9499840 DOI: 10.3389/fimmu.2022.926279] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 08/18/2022] [Indexed: 11/13/2022] Open
Abstract
Dendritic cells (DCs) are professional antigen-presenting cells that can recognize, capture, and process antigens. Fusing molecules targeting DCs with antigens can effectively improve the efficiency with which antigens are recognized and captured by DCs. This targeting strategy can be used for vaccine development to effectively improve the efficiency of antigen recognition and capture by DCs. The targeting sequence of porcine cytotoxic T-lymphocyte associated protein 4 (CTLA4), which binds porcine DCs, was identified in this study. Recombinant Lactobacillus reuteri (L. reuteri) expressing CTLA4-6aa (LYPPPY) and CTLA4-87aa fused to the porcine epidemic diarrhea virus (PEDV) protective antigen core neutralizing epitope (COE) were used to evaluate the ability of the two targeting motifs to bind the B7 molecule on DCs. Our results demonstrate that CTLA4-6aa could bind porcine DCs, and recombinant Lactobacillus expressing the CTLA4-6aa captured by porcine DCs was more efficient than those expressing CTLA4-87aa. In addition, the expression of DC markers, toll-like receptors, and cytokines was significantly higher in the 6aa-COE/L. reuteri-stimulated porcine DCs compared to DCs treated with 87aa-COE/L. reuteri (p<0.01) and recombinant Lactobacillus expressing CTLA4-6aa enhanced the ability of porcine DCs to activate T-cell proliferation. Our analysis of the protein structure revealed that CTLA4-87aa contains intramolecular hydrogen bonds, which may have weakened the intermolecular force between the residues on porcine CTLA4 and that on B7. In conclusion, recombinant Lactobacillus expressing CTLA4-6aa were more efficiently captured by porcine DCs and had a stronger ability to promote DC maturation and enhance T-cell proliferation. The LYPPPY motif is the optimal sequence for binding to porcine DCs. Piglets immunized with recombinant Lactobacillus showed that recombinant Lactobacillus expressing CTLA4-6aa induced significant levels of anti-PEDV-specific IgG and IgA antibody responses. Our study may promote research on DC-targeting strategies to enhance the effectiveness of porcine vaccines.
Collapse
Affiliation(s)
- Tian Xia
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Ning Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Yuqing Tang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Yueyi Gao
- Division of Viral Biologic Testing(I), China Institute of Veterinary Drug Control, Beijing, China
| | - Chong Gao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Jianhui Hao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Yanping Jiang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- China Ministry of Agriculture Key Laboratory of Animal Pathogen Biology, Northeastern Science Inspection Station, Harbin, China
| | - Xiaona Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- China Ministry of Agriculture Key Laboratory of Animal Pathogen Biology, Northeastern Science Inspection Station, Harbin, China
| | - Zhifu Shan
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- China Ministry of Agriculture Key Laboratory of Animal Pathogen Biology, Northeastern Science Inspection Station, Harbin, China
| | - Jiaxuan Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- China Ministry of Agriculture Key Laboratory of Animal Pathogen Biology, Northeastern Science Inspection Station, Harbin, China
| | - Han Zhou
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- China Ministry of Agriculture Key Laboratory of Animal Pathogen Biology, Northeastern Science Inspection Station, Harbin, China
| | - Wen Cui
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- China Ministry of Agriculture Key Laboratory of Animal Pathogen Biology, Northeastern Science Inspection Station, Harbin, China
| | - Xinyuan Qiao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- China Ministry of Agriculture Key Laboratory of Animal Pathogen Biology, Northeastern Science Inspection Station, Harbin, China
| | - Lijie Tang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- China Ministry of Agriculture Key Laboratory of Animal Pathogen Biology, Northeastern Science Inspection Station, Harbin, China
| | - Li Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- China Ministry of Agriculture Key Laboratory of Animal Pathogen Biology, Northeastern Science Inspection Station, Harbin, China
- *Correspondence: Yijing Li, ; Li Wang,
| | - Yijing Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- China Ministry of Agriculture Key Laboratory of Animal Pathogen Biology, Northeastern Science Inspection Station, Harbin, China
- *Correspondence: Yijing Li, ; Li Wang,
| |
Collapse
|
39
|
Zhu D, Jiang Y, Cao H, Yang J, Shu Y, Feng H, Yang X, Sun X, Shao M. Lactate: A regulator of immune microenvironment and a clinical prognosis indicator in colorectal cancer. Front Immunol 2022; 13:876195. [PMID: 36091047 PMCID: PMC9458902 DOI: 10.3389/fimmu.2022.876195] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 08/09/2022] [Indexed: 11/13/2022] Open
Abstract
Lactate can play an immunosuppressive role in the tumor microenvironment and promote tumor development by recruiting and inducing the activity of immunosuppressive cells and molecules. High lactate concentrations are important for tumor cell metastasis, angiogenesis, and treatment resistance. With the in-depth studies on tumor metabolism, lactate, one of the key factors involved in glycolysis, has been increasing emerged its characteristic clinical value in colorectal cancer (CRC). In this study, lactate genes were screened based on lactate metabolism pathways. Subsequently, the lactate subtypes were determined by clustering and analysis of the subtypes at all levels, including immune checkpoints, immune infiltration, and clinical characteristics, which revealed the biological significance of lactate metabolism in CRC. Subtype-based differential gene analysis resulted in a lactate score, which stratifies the prognosis of CRC. We discovered that 27 lactate genes and 61 lactate-phenotype genes are associated with immune cell infiltration and have a significant prognostic efficacy. The CRC patients were clustered into four subtypes and five clusters, based on lactate genes and lactate-phenotype genes, respectively. There are significant differences in survival time and activities of hallmark pathways, namely immune-related signatures and chemokines, among these subtypes and clusters. Particularly, cluster 2 and subtype 1 have significantly higher lactate scores than that of the others. In conclusion, lactate score is an independent prognostic factor for cancer that can be used as a clinical guide for predicting CRC progression and as an evaluation factor for the effect of immunotherapy in CRC.
Collapse
Affiliation(s)
- Daoqi Zhu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Yiping Jiang
- Department of Pharmacy, Zhuhai People’s Hospital (Zhuhai hospital affiliated with Jinan University), Zhuhai, China
| | - Huihui Cao
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
- Traditional Chinese Pharmacological, Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Jiabin Yang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Yuqi Shu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Haowei Feng
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Xiaoyu Yang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Xiaomin Sun
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
- *Correspondence: Meng Shao, ; Xiaomin Sun,
| | - Meng Shao
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
- *Correspondence: Meng Shao, ; Xiaomin Sun,
| |
Collapse
|
40
|
Local Breast Microbiota: A "New" Player on the Block. Cancers (Basel) 2022; 14:cancers14153811. [PMID: 35954474 PMCID: PMC9367283 DOI: 10.3390/cancers14153811] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/01/2022] [Accepted: 08/04/2022] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Microbiota plays a fundamental role in the induction, training and function of the human immune system. The interactions between microbiota and immune cells have consequences in several settings, namely in carcinogenesis but also in anticancer activity. Immunotherapy, already widely used in the treatment of several solid cancers, modulates the action of the immune system, promoting antitumour effects. Recently, there has been a growing interest in studying the microbiota composition as a possible modulator of the tumour microenvironment and consequently of the response to certain therapies such as immunotherapy. Abstract The tumour microenvironment (TME) comprises a complex ecosystem of different cell types, including immune cells, cells of the vasculature and lymphatic system, cancer-associated fibroblasts, pericytes, and adipocytes. Cancer proliferation, invasion, metastasis, drug resistance and immune escape are all influenced by the dynamic interaction between cancer cells and TME. Microbes, such as bacteria, fungi, viruses, archaea and protists, found within tumour tissues, constitute the intratumour microbiota, which is tumour type-specific and distinct among patients with different clinical outcomes. Growing evidence reveals a significant relevance of local microbiota in the colon, liver, breast, lung, oral cavity and pancreas carcinogenesis. Moreover, there is a growing interest in the tumour immune microenvironment (TIME) pointed out in several cross-sectional studies on the correlation between microbiota and TME. It is now known that microorganisms have the capacity to change the density and function of anticancer and suppressive immune cells, enabling the promotion of an inflammatory environment. As immunotherapy (such as immune checkpoint inhibitors) is becoming a promising therapy using TIME as a therapeutic target, the analysis and comprehension of local microbiota and its modulating strategies can help improve cancer treatments.
Collapse
|
41
|
Xu B, Wang X, Wang H, Cao L, Ge Y, Yuan B, Gao R, Li J. Efficacy and safety of herbal formulas with the function of gut microbiota regulation for gastric and colorectal cancer: A systematic review and meta-analysis. Front Cell Infect Microbiol 2022; 12:875225. [PMID: 35992176 PMCID: PMC9386000 DOI: 10.3389/fcimb.2022.875225] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 07/04/2022] [Indexed: 01/20/2023] Open
Abstract
Background Currently, gastric cancer (GC) and colorectal cancer (CRC) are the most common causes of cancer-related mortality worldwide. Gut microbiota is closely related to the occurrence of GC and CRC and the efficacy of chemotherapy. This study is aimed at evaluating the efficacy and safety of herbal formulas with the function of gut microbiota regulation (HFGMR) in the treatment of GC and CRC and to assess the quality of the synthesized evidence. Methods A comprehensive search was performed on eight electronic databases, PubMed, EMBASE, CENTRAL, Web of Science, Chinese Biomedical Literature Database, China National Knowledge Infrastructure, Wanfang database, Chinese Scientific Journals Database, and two registries, Chinese Clinical Trial Registry and ClinicalTrials.gov, from their initiation to January 2022. Randomized controlled trials (RCTs) studying the therapeutic effects of HFGMR were included. We used Stata 16 for data synthesis and Risk of Bias 2 (RoB 2) for methodological quality evaluation and assessed the quality of the synthesized evidence in the Grading of Recommendations, Assessment, Development and Evaluations (GRADE) approach. Results Fifty-three RCTs involving 4,478 patients were included. These trials involve seven herbal formulas that could regulate the gut microbiota of Bifidobacterium, Lactobacillus, Escherichia coli, Bacteroides, and Enterococcus faecalis. The meta-analysis results were subgrouped to three different stages in GC and CRC. 1) For the perioperative stage, HFGMR combined with conventional therapy could shorten the time to bowel sound recovery by 1.63 h [mean difference (MD) = -1.63, 95% confidence interval (CI) (-2.62, -0.65)], the time to first flatus by 9.69 h [MD = -9.69, 95% CI (-10.89, -8.48)], and the duration of hospitalization by 2.91 days [MD = -2.91, 95% CI (-4.01, -1.80)] in GC. There were no significant differences in outcomes of gastrointestinal function recovery and adverse events in CRC. 2) For postoperative patients, combined with adjuvant chemotherapy, HFGMR could decrease the incidence of diarrhea, nausea and vomiting, anorexia, and peripheral neurotoxicity in GC; boost Karnofsky performance status (KPS) improvement rate [risk ratio (RR) = 1.96, 95% CI (1.38, 2.79)]; and decrease the incidence of leucopenia and nausea and vomiting in CRC. 3) For advanced stage, HFGMR can significantly improve the objective response rate (ORR) [RR = 1.35, 95% CI (1.19~1.53)], disease control rate (DCR) [RR = 1.14, 95% CI (1.05~1.23)], and KPS improvement rate [RR = 1.56, 95% CI (1.17, 2.09)] and decrease the incidence of leucopenia, neutropenia, anemia, nausea and vomiting, diarrhea, and fatigue in GC. There were no significant differences in ORR [RR = 1.32, 95% CI (0.94~1.86)] and DCR [RR = 1.22, 95% CI (0.99~1.50)], but they can improve the KPS response rate [RR = 1.62, 95% CI (1.13, 2.32)] and decrease the incidence of myelosuppression, nausea and vomiting, diarrhea, and hepatic and renal dysfunction in CRC. Conclusion This study indicates that herbal formulas that could regulate the composition and proportion of gut microbiota have a positive effect in three stages (perioperative, postoperative, and advanced) of GC and CRC. They could promote the recovery of postoperative gastrointestinal function, increase tumor response, improve performance status, and reduce the incidence of adverse events. Herbal formulas exerted anti-cancer efficacy through multiple mechanisms and pathways; among them, the regulation of gut microbiota has not been paid enough attention. To further support the conclusion and better understand the role of gut microbiota in the treatment of GC and CRC, more rigorously designed, large-scale, and multicenter RCTs that focus on herbal formulas and gut microbiota are needed in the future.
Collapse
Affiliation(s)
- Bowen Xu
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Xinmiao Wang
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Heping Wang
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Luchang Cao
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuansha Ge
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Bo Yuan
- Department of Rheumatology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ruike Gao
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jie Li
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
42
|
Makaranka S, Scutt F, Frixou M, Wensley KE, Sharma R, Greenhowe J. The gut microbiome and melanoma: A review. Exp Dermatol 2022; 31:1292-1301. [PMID: 35793428 DOI: 10.1111/exd.14639] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/21/2022] [Accepted: 07/03/2022] [Indexed: 12/16/2022]
Abstract
Disturbances in the microbial ecosystem have been implemented in chronic inflammation, immune evasion and carcinogenesis, with certain microbes associated with the development of specific cancers. In recent times, the gut microbiome has been recognised as a potential novel player in the pathogenesis and treatment of malignant melanoma. It has been shown that the composition of gut microbiota in early-stage melanoma changes from in situ to invasive and then to metastatic disease. The gut bacterial and fungal profile has also been found to be significantly different in melanoma patients compared to controls. Multiple studies of immune checkpoint inhibitor (ICI) therapies have shown that the commensal microbiota may have an impact on anti-tumor immunity and therefore ICI response in cancer patients. When it comes to chemotherapy and radiotherapy treatments, studies demonstrate that gut microbiota are invaluable in the repair of radiation and chemotherapy-induced damage and therapeutic manipulation of gut microbiota can be an effective strategy to deal with side effects. Studies demonstrate the oncogenic and tumor-suppressive properties of the gut microbiome, which may play a role in the pathogenesis of melanoma. Despite this, investigations into specific interactions are still in its infancy, but starting to gain momentum as more significant and clinically relevant effects are emerging.
Collapse
Affiliation(s)
| | - Freya Scutt
- Department of Plastic Surgery, Aberdeen Royal Infirmary, Aberdeen, UK
| | - Mikaela Frixou
- Department of Gastroenterology, Aberdeen Royal Infirmary, Aberdeen, UK
| | | | - Ravi Sharma
- Department of Oncology, Aberdeen Royal Infirmary, Aberdeen, UK
| | | |
Collapse
|
43
|
Wu Q, Chen Y, Li Q, Chen J, Mo J, Jin M, Yang Q, Rizzello L, Tian X, Luo L. Time Rules the Efficacy of Immune Checkpoint Inhibitors in Photodynamic Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2200999. [PMID: 35470595 PMCID: PMC9313507 DOI: 10.1002/advs.202200999] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 04/02/2022] [Indexed: 06/14/2023]
Abstract
Lack of adequate effector T cells infiltrated in tumor is one of the main problems in the failure of immune checkpoint blockade therapy (ICBT). Photodynamic therapy (PDT) induced acute inflammation can sensitize tumors and activate T cells, thus assisting immune checkpoint inhibitors (ICI) against tumor growth and metastasis. T cells maturation and activation lag 3 to 7 days behind PDT. However, such timing in the combination therapy of ICI and PDT is commonly ignored in designing numerous multi-functional integrated nanomedicines. Herein, the authors illustrate that intervention timing of ICI after PDT affects the anti-tumor efficacy. A tumor-targeting nanomedicine is prepared by encapsulating indocyanine green into CD44 specifically binding material, a hyaluronic acid conjugated lipid poly(ethylene glycol). The PDT nanomedicine is designed to induce a robust immune response in tumor. The optimal group (Combo-STAR), ICI gave 5 days after PDT, significantly suppresses local tumor growth and eliminates metastasis. What should be highlighted is the time point of administration because if ICI is given too early, T cells are immature, otherwise, T cells are exhausted if ICI is given too late. This work presents theoretical guidance for raising awareness of intervention timing when augmenting ICBT with immune response inducers in clinic.
Collapse
Affiliation(s)
- Qinghua Wu
- College of Pharmaceutical SciencesSouthwest UniversityChongqing400715China
| | - Yang Chen
- College of Pharmaceutical SciencesSouthwest UniversityChongqing400715China
| | - Qing Li
- College of Pharmaceutical SciencesSouthwest UniversityChongqing400715China
| | - Junmeng Chen
- College of Pharmaceutical SciencesSouthwest UniversityChongqing400715China
| | - Junfeng Mo
- College of Pharmaceutical SciencesSouthwest UniversityChongqing400715China
| | - Ming Jin
- College of Pharmaceutical SciencesSouthwest UniversityChongqing400715China
| | - Qianzhan Yang
- Analytical Instruments DepartmentAnalytical Applications CenterShimadzu (China) Co., Ltd. Chongqing BranchChongqing404100China
| | - Loris Rizzello
- Department of Pharmaceutical SciencesUniversity of MilanMilan20133Italy
| | - Xiaohe Tian
- Huaxi MR Research Center (HMRRC)Department of RadiologyFunction & Molecular Imaging Key LabSichuan UniversityChengdu610041China
| | - Lei Luo
- College of Pharmaceutical SciencesSouthwest UniversityChongqing400715China
| |
Collapse
|
44
|
Zhang Y, Huo L, Wei Z, Tang Q, Sui H. Hotspots and Frontiers in Inflammatory Tumor Microenvironment Research: A Scientometric and Visualization Analysis. Front Pharmacol 2022; 13:862585. [PMID: 35370647 PMCID: PMC8968939 DOI: 10.3389/fphar.2022.862585] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 02/11/2022] [Indexed: 12/13/2022] Open
Abstract
Methods: Articles on inflammatory tumor microenvironment were retrieved from the Web of Science Core Collection, and the characteristics of the articles were analyzed by CiteSpace software. Background: The inflammatory tumor microenvironment is an essential feature of the tumor microenvironment. The way in which it promotes or inhibits tumor progression plays an important role in the outcome of a tumor treatment. This research aims to explore a scientific collaboration network, describe evolution of hotspots, and predict future trends through bibliometric analysis. Results: A total of 3,534 papers published by 390 institutions in 81 countries/regions were screened, and the annual quantity has been increasing rapidly in the past decades. United States was the leading country and has the most productive institutions in this field. The research topics were mainly focused on inflammation and immunity mediated by crucial factors as well as the mechanisms of angiogenesis. Additionally, the development and application of nanoparticles is currently a novel research frontier with bright prospect. Conclusion: The present scientometric study provides an overview of inflammatory tumor microenvironment research over the previous decades using quantitative and qualitative methods, and the findings of this study can provide references for researchers focusing on tumor treatment.
Collapse
Affiliation(s)
- Yuli Zhang
- Medical Experiment Center, Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of Traditional Chinese Medicine, Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Long Huo
- Department of Gastroenterology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhenzhen Wei
- Medical Experiment Center, Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qingfeng Tang
- Department of Clinical Laboratory, Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of Clinical Laboratory and Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hua Sui
- Medical Experiment Center, Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
45
|
Liu J, Yang D, Wang X, Asare PT, Zhang Q, Na L, Shao L. Gut Microbiota Targeted Approach in the Management of Chronic Liver Diseases. Front Cell Infect Microbiol 2022; 12:774335. [PMID: 35444959 PMCID: PMC9014089 DOI: 10.3389/fcimb.2022.774335] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Accepted: 02/21/2022] [Indexed: 12/12/2022] Open
Abstract
The liver is directly connected to the intestines through the portal vein, which enables the gut microbiota and gut-derived products to influence liver health. There is accumulating evidence of decreased gut flora diversity and alcohol sensitivity in patients with various chronic liver diseases, including non-alcoholic/alcoholic liver disease, chronic hepatitis virus infection, primary sclerosing cholangitis and liver cirrhosis. Increased intestinal mucosal permeability and decline in barrier function were also found in these patients. Followed by bacteria translocation and endotoxin uptake, these will lead to systemic inflammation. Specific microbiota and microbiota-derived metabolites are altered in various chronic liver diseases studies, but the complex interaction between the gut microbiota and liver is missing. This review article discussed the bidirectional relationship between the gut and the liver, and explained the mechanisms of how the gut microbiota ecosystem alteration affects the pathogenesis of chronic liver diseases. We presented gut-microbiota targeted interventions that could be the new promising method to manage chronic liver diseases.
Collapse
Affiliation(s)
- Jing Liu
- Department of Research, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital; The College of Medical Technology, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Dakai Yang
- Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Xiaojing Wang
- School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Paul Tetteh Asare
- Human and Animal Health Unit, Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Basel, Switzerland
| | - Qingwen Zhang
- Department of Research, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital; The College of Medical Technology, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Lixin Na
- Department of Research, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital; The College of Medical Technology, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Lei Shao
- School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai, China
- *Correspondence: Lei Shao,
| |
Collapse
|
46
|
Zanetta P, Ormelli M, Amoruso A, Pane M, Azzimonti B, Squarzanti DF. Probiotics as Potential Biological Immunomodulators in the Management of Oral Lichen Planus: What's New? Int J Mol Sci 2022; 23:ijms23073489. [PMID: 35408849 PMCID: PMC8998608 DOI: 10.3390/ijms23073489] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/17/2022] [Accepted: 03/21/2022] [Indexed: 02/04/2023] Open
Abstract
Oral lichen planus (OLP) is a T cell-mediated chronic inflammatory disorder with multifactorial aetiology and malignant transformation potential. Despite the treatments so far identified, new tailored and safe specific measures are needed. Recently, human microbiota imbalance has been linked to several immune-mediated diseases, opening new therapeutic perspectives for probiotics; besides their ability to directly interact with the host microbiota, they also display a strain-specific immune-modulatory effect. Thus, this non-systematic review aims to elucidate the molecular pathways underlying probiotic activity, mainly those of Lactobacilli and Bifidobacteria and their metabolites in OLP pathogenesis and malignant transformation, focusing on the most recent in vitro and in vivo research evidence. Findings related to their activity in other immune-mediated diseases are here included, suggesting a probiotic translational use in OLP. Probiotics show immune-modulatory and microbiota-balancing activities; they protect the host from pathogens, hamper an excessive effector T cell response, reduce nuclear factor-kappa B (NF-kB) signalling and basal keratinocytes abnormal apoptosis, shifting the mucosal response towards the production of anti-inflammatory cytokines, thus preventing uncontrolled damage. Therefore, probiotics could be a highly encouraging prevention and immunotherapeutic approach for a safer and more sustainable OLP management.
Collapse
Affiliation(s)
- Paola Zanetta
- Laboratory of Applied Microbiology, Department of Health Sciences (DiSS), Center for Translational Research on Allergic and Autoimmune Diseases (CAAD), School of Medicine, Università del Piemonte Orientale (UPO), Corso Trieste 15/A, 28100 Novara, Italy; (P.Z.); (M.O.)
| | - Margherita Ormelli
- Laboratory of Applied Microbiology, Department of Health Sciences (DiSS), Center for Translational Research on Allergic and Autoimmune Diseases (CAAD), School of Medicine, Università del Piemonte Orientale (UPO), Corso Trieste 15/A, 28100 Novara, Italy; (P.Z.); (M.O.)
| | - Angela Amoruso
- Probiotical Research Srl, Via Mattei 3, 28100 Novara, Italy; (A.A.); (M.P.)
| | - Marco Pane
- Probiotical Research Srl, Via Mattei 3, 28100 Novara, Italy; (A.A.); (M.P.)
| | - Barbara Azzimonti
- Laboratory of Applied Microbiology, Department of Health Sciences (DiSS), Center for Translational Research on Allergic and Autoimmune Diseases (CAAD), School of Medicine, Università del Piemonte Orientale (UPO), Corso Trieste 15/A, 28100 Novara, Italy; (P.Z.); (M.O.)
- Correspondence: (B.A.); (D.F.S.); Tel.: +39-0321-660-870 (B.A.)
| | - Diletta Francesca Squarzanti
- Laboratory of Applied Microbiology, Department of Health Sciences (DiSS), Center for Translational Research on Allergic and Autoimmune Diseases (CAAD), School of Medicine, Università del Piemonte Orientale (UPO), Corso Trieste 15/A, 28100 Novara, Italy; (P.Z.); (M.O.)
- Correspondence: (B.A.); (D.F.S.); Tel.: +39-0321-660-870 (B.A.)
| |
Collapse
|
47
|
Hu Q, Niu Y, Yang Y, Mao Q, Lu Y, Ran H, Zhang H, Li X, Gu H, Su Q. Polydextrose Alleviates Adipose Tissue Inflammation and Modulates the Gut Microbiota in High-Fat Diet-Fed Mice. Front Pharmacol 2022; 12:795483. [PMID: 35185543 PMCID: PMC8848743 DOI: 10.3389/fphar.2021.795483] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 12/22/2021] [Indexed: 12/13/2022] Open
Abstract
The soluble dietary fiber polydextrose (PDX) is a randomly linked glucose oligomer containing small amounts of sorbitol and citric acid and is widely used in the food industry. However, whether PDX can prevent and treat obesity in high-fat diet (HFD)-fed mice has not been directly investigated, and further studies are needed to better understand the complex interactions among PDX, adipose tissue inflammation and the gut microbiota. In the present study, PDX reduced body weight, fasting blood glucose (FBG), adipose tissue accumulation, adipocyte hypertrophy, serum total cholesterol (TC), low-density lipoprotein cholesterol (LDL-C) and high-density lipoprotein cholesterol (HDL-C) levels in HFD-fed mice. Moreover, PDX alleviated serum lipopolysaccharide (LPS) levels and macrophage infiltration in epididymal adipose tissue and resulted in macrophage polarization toward the M2 phenotype. Gut microbiota analysis revealed that PDX promoted the growth of beneficial microbes such as Bacteroides, Parabacteroides, Alloprevotella, Muribaculum, Akkermansia, Ruminococcaceae_UCG-014 and UBA1819 in obese mice, which were negatively correlated with subcutaneous fat, epididymal fat, body weight, FBG, serum TC, HDL-C, LDL-C and LPS levels. Our results indicates that PDX can prevent and treat obesity in HFD-fed mice, specifically in alleviating glucolipid metabolism disorders and adipose tissue inflammation, which may be mediated by modulating the structure of the gut microbiota. Therefore, PDX may become a promising nondrug therapy for obesity.
Collapse
Affiliation(s)
- Qiuyue Hu
- Department of Endocrinology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yixin Niu
- Department of Endocrinology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanxia Yang
- Department of Endocrinology, Xinhua Hospital Chongming Branch, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qianyun Mao
- Department of Endocrinology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yao Lu
- Department of Endocrinology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hui Ran
- Department of Endocrinology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongmei Zhang
- Department of Endocrinology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoyong Li
- Department of Endocrinology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongxia Gu
- Department of Endocrinology, Xinhua Hospital Chongming Branch, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qing Su
- Department of Endocrinology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
48
|
Niu H, Xing JH, Zou BS, Shi CW, Huang HB, Jiang YL, Wang JZ, Cao X, Wang N, Zeng Y, Yang WT, Yang GL, Wang CF. Immune Evaluation of Recombinant Lactobacillus plantarum With Surface Display of HA1-DCpep in Mice. Front Immunol 2021; 12:800965. [PMID: 34925386 PMCID: PMC8673267 DOI: 10.3389/fimmu.2021.800965] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Accepted: 11/04/2021] [Indexed: 11/13/2022] Open
Abstract
Avian influenza viruses can be efficiently transmitted through mucous membranes, and conventional vaccines are not effective in protecting against mucosal infection by influenza viruses. To induce multiple immune responses in an organism, we constructed a recombinant Lactobacillus plantarum expressing the influenza virus antigen HA1 with the adjuvant dendritic cell-targeting peptide (DCpep). The recombinant L. plantarum strains NC8Δ-pWCF-HA1 and NC8Δ-pWCF-HA1-DCpep were used to immunize mice via oral administration, and the humoral, cellular and mucosal immune responses were evaluated. In addition, the serum levels of specific antibodies and hemagglutination inhibition (HI) levels were also measured. Our results showed that recombinant L. plantarum activated dendritic cells in Peyer's patches (PPs), increased the numbers of CD4+IFN-γ+ and CD8+IFN-γ+ cells in the spleen and mesenteric lymph nodes (MLNs), and affected the ability of CD4+ and CD8+ cells to proliferate in the spleen and MLNs. Additionally, recombinant L. plantarum increased the number of B220+IgA+ cells in PPs and the level of IgA in the lungs and different intestinal segments. In addition, specific IgG, IgG1 and IgG2a antibodies were induced at high levels in the mice serum, specific IgA antibodies were induced at high levels in the mice feces, and HI potency was significantly increased. Thus, the recombinant L. plantarum strains NC8Δ-pWCF-HA1 and NC8Δ-pWCF-HA1-DCpep have potential as vaccine candidates for avian influenza virus.
Collapse
Affiliation(s)
- Hui Niu
- College of Veterinary Medicine, College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Key Laboratory of Animal Production and Product Quality Safety of the Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Jun-Hong Xing
- College of Veterinary Medicine, College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Key Laboratory of Animal Production and Product Quality Safety of the Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Bo-Shi Zou
- College of Veterinary Medicine, College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Key Laboratory of Animal Production and Product Quality Safety of the Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Chun-Wei Shi
- College of Veterinary Medicine, College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Key Laboratory of Animal Production and Product Quality Safety of the Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Hai-Bin Huang
- College of Veterinary Medicine, College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Key Laboratory of Animal Production and Product Quality Safety of the Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Yan-Long Jiang
- College of Veterinary Medicine, College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Key Laboratory of Animal Production and Product Quality Safety of the Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Jian-Zhong Wang
- College of Veterinary Medicine, College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Key Laboratory of Animal Production and Product Quality Safety of the Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Xin Cao
- College of Veterinary Medicine, College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Key Laboratory of Animal Production and Product Quality Safety of the Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Nan Wang
- College of Veterinary Medicine, College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Key Laboratory of Animal Production and Product Quality Safety of the Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Yan Zeng
- College of Veterinary Medicine, College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Key Laboratory of Animal Production and Product Quality Safety of the Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Wen-Tao Yang
- College of Veterinary Medicine, College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Key Laboratory of Animal Production and Product Quality Safety of the Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Gui-Lian Yang
- College of Veterinary Medicine, College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Key Laboratory of Animal Production and Product Quality Safety of the Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Chun-Feng Wang
- College of Veterinary Medicine, College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Key Laboratory of Animal Production and Product Quality Safety of the Ministry of Education, Jilin Agricultural University, Changchun, China
| |
Collapse
|