1
|
Gupta MK, Srivastava R. Gut Microbiome Interventions: From Dysbiosis to Next-Generation Probiotics (NGPs) for Disease Management. Probiotics Antimicrob Proteins 2025:10.1007/s12602-025-10582-7. [PMID: 40434505 DOI: 10.1007/s12602-025-10582-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/06/2025] [Indexed: 05/29/2025]
Abstract
The gut microbiome, sometimes referred to as the "second brain," the "lost organ," the "identification card of the individual," and the "fingerprint of the host," possesses diverse traits and functions that influence health. The impact of gut commensal bacteria on health, as opposed to environmental pathogenic factors, has generated increasing interest in recent years, culminating in a substantial body of study. Research indicates that dysbiosis of the intestinal microbiota is commonly observed in chronic inflammatory diseases, including colitis, obesity/metabolic syndrome, diabetes mellitus, liver infections, allergic conditions, cardiovascular diseases, COVID-19, cancers, and neurodegenerative disorders. The International Scientific Association for Probiotics and Prebiotics has recently refined the theory of complementary and synergistic synbiotics. In recent years, the field of microbiome research has been significantly advanced by technological developments such as massive culturomics, gnotobiotics, metabolomics, parallel DNA sequencing, and RNA sequencing. This review article examined the potential next generation probiotics (NGPs) and explored some of them, Faecalibacterium prausnitzii, Bacteroides thetaiotaomicron, Akkermansia muciniphila, Parabacteroides goldsteinii, Bacteroides fragilis, Eubacterium hallii, Roseburia intestinalis, Christensenella minuta, Prevotella copri, and Oscillospira guilliermondii. In addition to these useful probiotic strains, psychobiotics, members of the families of Lactobacilli, Streptococci, Bifidobacteria, Escherichia, and Enterococci, have extended applicability in the use for neurodevelopmental and neurodegenerative disorders. The article also reviewed current trends and limitations in NGPs to enhance our comprehensive understanding of key concepts associated with the consumption of probiotics and proposed necessary initiatives for researchers to engage in collaborative translational research as future therapeutic solutions.
Collapse
Affiliation(s)
- Mandeep Kumar Gupta
- Moradabad Educational Trust Group of Institutions Faculty of Pharmacy, Moradabad, 244001, Uttar Pradesh, India.
| | - Rajnish Srivastava
- Chitkara University School of Pharmacy, Chitkara University, Baddi, 174103, Himachal Pradesh, India
| |
Collapse
|
2
|
Wu H, Liu J, Zhang XH, Jin S, Li P, Liu H, Zhao L, Wang J, Zhao S, Tian HD, Lai JR, Hao Y, Liu GR, Hou K, Yan M, Liu SL, Pang D. The combination of flaxseed lignans and PD-1/ PD-L1 inhibitor inhibits breast cancer growth via modulating gut microbiome and host immunity. Drug Resist Updat 2025; 80:101222. [PMID: 40048957 DOI: 10.1016/j.drup.2025.101222] [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: 12/29/2024] [Revised: 02/22/2025] [Accepted: 02/22/2025] [Indexed: 03/16/2025]
Abstract
BACKGROUND Patients with breast cancer (BC) who benefit from the PD-1/PD-L1 inhibitor (PDi) is limited, necessitating novel strategies to improve immunotherapy efficacy of BC. Here we aimed to investigate the inhibitory effects of flaxseed lignans (FL) on the biological behaviors of BC and evaluate the roles of FL in enhancing the anticancer effects of PDi. METHODS HPLC was used to detect the content of enterolactone (ENL), the bacterial transformation product of FL. Transcript sequencing was performed and identified CD38 as a downstream target gene of ENL. CD38-overexpressing cells were constructed and cell proliferation, colony formation, wound healing and transwell assays were used to assess the function of ENL/CD38 axis on BC cells in vitro. Multiplexed immunohistochemistry (mIHC) and CyTOF were used to detect the changes of the tumor immune microenvironment (TIM). 16S rDNA sequencing was used to explore the changes of gut microbiota in mice. A series of in vivo experiments were conducted to investigate the anticancer effects and mechanisms of FL and PDi. RESULTS FL was converted to ENL by gut microbiota and FL administration inhibited the progression of BC. ENL inhibited the malignant behaviors of BC by downregulating CD38, a key gene associated with immunosuppression and PD-1/PD-L1 blockade resistance. The mIHC assay revealed that FL administration enhanced CD3+, CD4+ and CD8+ cells and reduced F4/80+ cells in TIM. CyTOF confirmed the regulatory effects of FL and FL in combination with PDi (FLcPDi) on TIM. In addition, 16S rDNA analysis demonstrated that FLcPDi treatment significantly elevated the abundance of Akkermansia and, importantly, Akkermansia administration enhanced the response to PDi in mice treated with antibiotics. CONCLUSIONS The FL/ENL/CD38 axis inhibited BC progression. FL enhanced the anticancer effects of PDi by modulating gut microbiota and host immunity.
Collapse
Affiliation(s)
- Hao Wu
- Heilongjiang Clinical Research Center for Breast Cancer, Harbin Medical University Cancer Hospital, Harbin, China; Genomics Research Center, State Key Laboratory of Biomedicine-Pharmaceutics of China, College of Pharmacy, Harbin Medical University, Harbin, China.
| | - Jiena Liu
- Heilongjiang Clinical Research Center for Breast Cancer, Harbin Medical University Cancer Hospital, Harbin, China; Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China.
| | - Xing-Hua Zhang
- Genomics Research Center, State Key Laboratory of Biomedicine-Pharmaceutics of China, College of Pharmacy, Harbin Medical University, Harbin, China; Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province, Harbin Medical University, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin 150081, China
| | - Shengye Jin
- Heilongjiang Clinical Research Center for Breast Cancer, Harbin Medical University Cancer Hospital, Harbin, China; Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Ping Li
- The Third Affiliated Hospital of Beijing University of Chinese Medicine, Beijing, China
| | - Huidi Liu
- Genomics Research Center, State Key Laboratory of Biomedicine-Pharmaceutics of China, College of Pharmacy, Harbin Medical University, Harbin, China; Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province, Harbin Medical University, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin 150081, China; Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada
| | - Liuying Zhao
- Heilongjiang Clinical Research Center for Breast Cancer, Harbin Medical University Cancer Hospital, Harbin, China; Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Jianyu Wang
- Heilongjiang Clinical Research Center for Breast Cancer, Harbin Medical University Cancer Hospital, Harbin, China; Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Shilu Zhao
- Heilongjiang Clinical Research Center for Breast Cancer, Harbin Medical University Cancer Hospital, Harbin, China; Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Hong-Da Tian
- Genomics Research Center, State Key Laboratory of Biomedicine-Pharmaceutics of China, College of Pharmacy, Harbin Medical University, Harbin, China; Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province, Harbin Medical University, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin 150081, China
| | - Jin-Ru Lai
- Genomics Research Center, State Key Laboratory of Biomedicine-Pharmaceutics of China, College of Pharmacy, Harbin Medical University, Harbin, China; Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province, Harbin Medical University, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin 150081, China
| | - Yi Hao
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Gui-Rong Liu
- Genomics Research Center, State Key Laboratory of Biomedicine-Pharmaceutics of China, College of Pharmacy, Harbin Medical University, Harbin, China; Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province, Harbin Medical University, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin 150081, China
| | - Kaijian Hou
- School of Public Health, Shantou University, Shantou, China; Longhu People's Hospital, Shantou, China.
| | - Meisi Yan
- Department of Pathology, Harbin Medical University, Harbin, China.
| | - Shu-Lin Liu
- Genomics Research Center, State Key Laboratory of Biomedicine-Pharmaceutics of China, College of Pharmacy, Harbin Medical University, Harbin, China; Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province, Harbin Medical University, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin 150081, China; Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, AB, Canada.
| | - Da Pang
- Heilongjiang Clinical Research Center for Breast Cancer, Harbin Medical University Cancer Hospital, Harbin, China; Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China.
| |
Collapse
|
3
|
Alum EU, Uti DE, Ugwu OPC, Alum BN, Edeh FO, Ainebyoona C. Unveiling the microbial orchestra: exploring the role of microbiota in cancer development and treatment. Discov Oncol 2025; 16:646. [PMID: 40304829 PMCID: PMC12044139 DOI: 10.1007/s12672-025-02352-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 04/09/2025] [Indexed: 05/02/2025] Open
Abstract
The human microbiota comprises a diverse microbial ecosystem that significantly impacts health and disease. Among its components, the gut microbiota plays a crucial role in regulating metabolic, immunologic, and inflammatory responses. Dysbiosis, an imbalance in microbial composition, has been linked to carcinogenesis through mechanisms such as chronic inflammation, metabolic disturbances, epigenetic modifications, and immune system dysregulation. Additionally, dysbiosis influences the efficacy and toxicity of cancer therapies. Given these associations, there is growing interest in leveraging the microbiota as a biomarker for cancer detection and outcome prediction. Notably, distinct microbial signatures have been identified across various cancer types, suggesting their potential as diagnostic markers. Furthermore, modulation of the microbiota presents a promising avenue for improving cancer treatment outcomes through strategies such as antibiotics, prebiotics, probiotics, fecal microbiota transplantation, dietary interventions, small-molecule inhibitors, and phage therapy. To explore these relationships, we conducted a comprehensive literature review using Web of Science, Scopus, PubMed, MEDLINE, Embase, and Google Scholar as our primary online databases, focusing on indexed peer-reviewed articles up to the present year. This review aims to elucidate the role of dysbiosis in cancer development, examine the molecular mechanisms involved, and assess the impact of microbiota on cancer therapies. Additionally, we highlight microbiota-based therapeutic strategies and discuss their potential applications in cancer management. A deeper understanding of the intricate interplay between the microbiota and cancer may pave the way for novel approaches to cancer prevention, early detection, and treatment optimization.
Collapse
Affiliation(s)
- Esther Ugo Alum
- Department of Research and Publications, Kampala International University, Main Campus, P. O. Box 20000, Kampala, Uganda.
| | - Daniel Ejim Uti
- Department of Research and Publications, Kampala International University, Main Campus, P. O. Box 20000, Kampala, Uganda.
- Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, Federal University of Health Sciences, Otukpo, Benue State, Nigeria.
| | - Okechukwu Paul-Chima Ugwu
- Department of Research and Publications, Kampala International University, Main Campus, P. O. Box 20000, Kampala, Uganda
| | - Benedict Nnachi Alum
- Department of Research and Publications, Kampala International University, Main Campus, P. O. Box 20000, Kampala, Uganda
| | - Friday Ogbu Edeh
- College of Economics and Management, Kampala International University, Kampala, Uganda
| | - Christine Ainebyoona
- Faculty of Business and Management Sciences, Kampala International University, Kampala, Uganda
| |
Collapse
|
4
|
Zhu J, Qin S, Gu R, Ji S, Wu G, Gu K. Amuc_1434 From Akkermansia muciniphila Enhances CD8+ T Cell-Mediated Anti-Tumor Immunity by Suppressing PD-L1 in Colorectal Cancer. FASEB J 2025; 39:e70540. [PMID: 40231387 DOI: 10.1096/fj.202403295rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 03/21/2025] [Accepted: 04/04/2025] [Indexed: 04/16/2025]
Abstract
Colorectal cancer (CRC) shows a limited response to programmed death-ligand 1 (PD-L1) immunotherapies. Akkermansia muciniphila (AKK) may enhance tumor immunity. This study examines how its Outer Membrane Vesicles (OMVs) and Amuc_1434 influence PD-L1 expression and CD8+ T cell activity in CRC. OMVs were isolated and their characteristics were examined through transmission electron microscopy and Western blotting. PD-L1 expression was quantified via Western blot, while CD8+ T cell proliferation was measured using flow cytometry. Cytokine production (interferon-gamma (IFN-γ) and interleukin-2 (IL-2)) was evaluated using ELISA. A CRC mouse model was employed to examine its impact on tumor growth and immune cell infiltration. In CRC cells, treatment with AKK-derived OMVs (AKK-OMVs) significantly downregulated PD-L1 expression (p < 0.05) and markedly increased CD8+ T cell proliferation and the levels of IFN-γ and IL-2 (p < 0.01). Amuc_1434 was identified as the key protein mediating these effects. In vivo, AKK-OMVs treatment substantially reduced tumor volume (p < 0.01) and significantly enhanced CD8+ T cell infiltration into the tumor microenvironment (p < 0.01). Additionally, AKK-OMVs-treated mice showed increased expression of immune activation markers within the tumor tissue, further indicating enhanced antitumor immunity. This study reveals that AKK-OMVs, particularly those containing Amuc_1434, can modulate PD-L1 expression and potentiate CD8+ T cell-mediated antitumor immunity in CRC. These findings suggest a novel approach to overcoming resistance to immune checkpoint inhibitors in CRC.
Collapse
Affiliation(s)
- Jiahao Zhu
- Department of Radiotherapy and Oncology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, People's Republic of China
| | - Shaolei Qin
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, People's Republic of China
| | - Ruike Gu
- Department of Rehabilitation Medical, Suzhou Rehabilitation Hospital (Suzhou Municipal Hospital Rehabilitation Medical Center), Suzhou, Jiangsu, People's Republic of China
| | - Shengjun Ji
- Department of Radiotherapy and Oncology, The Affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, People's Republic of China
| | - Gang Wu
- Department of Radiotherapy and Oncology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, People's Republic of China
| | - Ke Gu
- Department of Radiotherapy and Oncology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, People's Republic of China
| |
Collapse
|
5
|
Zhao X, Chen C, Qiu H, Liu J, Shao N, Guo M, Jiang Y, Zhao J, Xu L. The landscape of ATF3 in tumors: Metabolism, expression regulation, therapy approach, and open concerns. Pharmacol Res 2025; 214:107666. [PMID: 39978658 DOI: 10.1016/j.phrs.2025.107666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 02/09/2025] [Accepted: 02/17/2025] [Indexed: 02/22/2025]
Abstract
Cellular stress response is a pivotal process in tumor development and therapy. Activating transcription factor 3 (ATF3), a representative stress-responsive protein, plays pleiotropic roles in various biological processes. Over the past decade, studies have described not only the general role of ATF3 in tumor metabolism but also the complexity of ATF3 expression regulation and its associated modifications, including phosphorylation, ubiquitination, SUMOylation, and NEDDylation. Interestingly, beyond being a transcription factor, ATF3 can act as a modifier to control the ubiquitination of target molecules, such as p53, to exert its function in tumors. These advances in uncovering ATF3 biological function have yielded new insights into the cellular stress response during tumor development and will be instrumental in developing novel interventions. In this review, we update the role of ATF3 as a nexus in amino acid metabolism, lipid metabolism, glycometabolism, and other metabolic pathways in tumors; delineate the underlying mechanisms involving DNA level regulation, epigenetic regulation, and post-translational modifications of ATF3; and summarize the progression of tumor mono/combination therapies related to ATF3. In particular, we discuss the challenges that need to be addressed to provide a new conceptual framework for further understanding the potential therapeutic value of ATF3 in ongoing clinical trials.
Collapse
Affiliation(s)
- Xu Zhao
- Medical College, Guizhou University, Guiyang, Guizhou Province 550025, China; Key Laboratory for Cancer Prevention and Treatment of Guizhou Province, Zunyi, Guizhou 563000, China; Department of Immunology, Zunyi Medical University, Guizhou 563000, China
| | - Chao Chen
- Medical College, Guizhou University, Guiyang, Guizhou Province 550025, China; Key Laboratory for Cancer Prevention and Treatment of Guizhou Province, Zunyi, Guizhou 563000, China; Department of Immunology, Zunyi Medical University, Guizhou 563000, China
| | - Hui Qiu
- Key Laboratory for Cancer Prevention and Treatment of Guizhou Province, Zunyi, Guizhou 563000, China; Department of Immunology, Zunyi Medical University, Guizhou 563000, China
| | - Jing Liu
- Key Laboratory for Cancer Prevention and Treatment of Guizhou Province, Zunyi, Guizhou 563000, China; Department of Immunology, Zunyi Medical University, Guizhou 563000, China
| | - Nan Shao
- Key Laboratory for Cancer Prevention and Treatment of Guizhou Province, Zunyi, Guizhou 563000, China; Department of Immunology, Zunyi Medical University, Guizhou 563000, China
| | - Mengmeng Guo
- Key Laboratory for Cancer Prevention and Treatment of Guizhou Province, Zunyi, Guizhou 563000, China; Department of Immunology, Zunyi Medical University, Guizhou 563000, China
| | - Yuanye Jiang
- Department of Gastroenterology, Putuo hospital, Shanghai University of Tradtional Chinese Medicine, Shanghai 200062, China.
| | - Juanjuan Zhao
- Key Laboratory for Cancer Prevention and Treatment of Guizhou Province, Zunyi, Guizhou 563000, China; Department of Immunology, Zunyi Medical University, Guizhou 563000, China.
| | - Lin Xu
- Medical College, Guizhou University, Guiyang, Guizhou Province 550025, China; Key Laboratory for Cancer Prevention and Treatment of Guizhou Province, Zunyi, Guizhou 563000, China; Department of Immunology, Zunyi Medical University, Guizhou 563000, China; Innovation Center for Tissue Damage Repair, Ministry of Education, Zunyi, Guizhou 563000, China.
| |
Collapse
|
6
|
Zhao Z, Hu Z, Li L. Cardiac energy metabolic disorder and gut microbiota imbalance: a study on the therapeutic potential of Shenfu Injection in rats with heart failure. Front Microbiol 2025; 16:1509548. [PMID: 40071211 PMCID: PMC11895768 DOI: 10.3389/fmicb.2025.1509548] [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: 10/11/2024] [Accepted: 02/11/2025] [Indexed: 03/14/2025] Open
Abstract
Objective To investigate the relationship between heart failure (HF) and gut microbiota-mediated energy metabolism, and to explore the role of Shenfu Injection in this process. Materials and methods In this study, Adriamycin-induced chronic heart failure (CHF) rat model was used and randomly divided into the blank control group (Normal, n = 9), HF control group (Model, n = 12), Shenfu Injection treatment group (SFI, n = 9), and positive drug control group (TMZ, n = 9). The changes in gut microbiota structure were analyzed by 16S rRNA high-throughput sequencing, the content of short-chain fatty acids (SCFAs) was detected by targeted metabolomics technology, and cardiac function and energy metabolism-related indicators were evaluated. Results Myocardial energy metabolism in HF rats was disordered, characterized by reduced fatty acid oxidation, enhanced anaerobic glycolysis of glucose, mitochondrial damage, and decreased ATP content; The gut microbiota of HF rats was imbalanced, with a reduction in beneficial bacteria, an increase in conditional pathogenic bacteria, and impaired intestinal barrier function; Both Shenfu Injection and trimetazidine improved myocardial energy metabolism and cardiac function, but Shenfu Injection was more significant in regulating gut microbiota and improving intestinal health; The production of SCFAs from the gut microbiota of HF rats increased, which may be closely related to myocardial energy metabolism; SCFAs-producing bacteria Akkermansia and Blautia played a key role in the development of HF, and their abundance was positively correlated with SCFAs content. Conclusion Shenfu Injection in treating HF may improve myocardial energy metabolism and intestinal health by regulating gut microbiota, especially the abundance of SCFAs-producing bacteria Akkermansia and Blautia, thereby exerting therapeutic effects. This provides theoretical support for treatment strategies based on gut microbiota.
Collapse
Affiliation(s)
- Zhenyu Zhao
- Provincial Key Laboratory of TCM Diagnostics, Hunan University of Chinese Medicine, Changsha, China
| | - Zhixi Hu
- Institute of TCM Diagnosis, Hunan University of Chinese Medicine, Changsha, China
| | - Lin Li
- Institute of TCM Diagnosis, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
7
|
Zhang Q, Peng L, Zhang Q, Guo J, Yu N, Yang J, Zuo W. Oral Akkermansia muciniphila Biomimetic Nanotherapeutics for Ulcerative Colitis Targeted Treatment by Repairing Intestinal Epithelial Barrier and Restoring Redox Homeostasis. ACS APPLIED MATERIALS & INTERFACES 2025; 17:5942-5954. [PMID: 39817352 DOI: 10.1021/acsami.4c18301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2025]
Abstract
The structural disruption of intestinal barrier and excessive reactive oxygen/nitrogen species (RONS) generation are two intertwined factors that drive the occurrence and development of ulcerative colitis (UC). Synchronously restoring the intestinal barrier and mitigating excess RONS is a promising strategy for UC management, but its treatment outcomes are still hindered by low drug accumulation and retention in colonic lesions. Inspired by intestine colonizing bacterium, we developed a mucoadhesive probiotic Akkermansia muciniphila-mimic entinostat-loaded hollow mesopores prussian blue (HMPB) nanotherapeutic (AM@HMPB@E) for UC-targeted therapy via repairing intestinal barrier and scavenging RONS. After oral administration, the negatively charged AM@HMPB@E specifically bind to the positively charged inflamed colon lesions via electrostatic interactions and Akkermansia muciniphila membrane-mediated bioadhesion mechanism. Subsequently, the superoxide dismutase (SOD)-, and catalase (CAT)-like HMPB eliminated RONS, thereby alleviating RONS-mediated inflammation and intestinal epithelial damage. Meanwhile, the UC-site locally released entinostat could repair the damaged intestinal epithelial barrier by inhibiting intestinal endothelial cell apoptosis and up-regulating the expression of tight junctions. Both in vitro and in vivo results shown that AM@HMPB@E not only exhibited an exceptional retention in the colitis site but also demonstrated superior therapeutic efficacy compared to the first-line drug sulfasalazine, as evidenced by the longer colon, less rectal bleeding and body weight loss. Collectively, our findings highlight the clinical application prospects of this synchronous nanotherapeutic strategy for UC treatment, offering a paradigm for the rational design of oral nanomedicine.
Collapse
Affiliation(s)
- Qiqi Zhang
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, No. 1160 Shengli South Street, Yinchuan 750004, PR China
| | - Li Peng
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, No. 1160 Shengli South Street, Yinchuan 750004, PR China
- Department of Pharmacy, General Hospital of Ningxia Medical University, No. 804 Shengli South Street, Yinchuan 750004, PR China
| | - Qian Zhang
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, No. 1160 Shengli South Street, Yinchuan 750004, PR China
| | - Jueshuo Guo
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, No. 1160 Shengli South Street, Yinchuan 750004, PR China
| | - Na Yu
- Department of Preparation Center, General Hospital of Ningxia Medical University, No. 804 Shengli South Street, Yinchuan 750004, PR China
| | - Jianhong Yang
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, No. 1160 Shengli South Street, Yinchuan 750004, PR China
- Department of Key Laboratory of Protection, Development and Utilization of Medicinal Resources in Liupanshan Area, Ministry of Education, School of Pharmacy, Ningxia Medical University, No. 1160 Shengli South Street, Yinchuan 750004, PR China
| | - Wenbao Zuo
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, No. 1160 Shengli South Street, Yinchuan 750004, PR China
- Department of Key Laboratory of Protection, Development and Utilization of Medicinal Resources in Liupanshan Area, Ministry of Education, School of Pharmacy, Ningxia Medical University, No. 1160 Shengli South Street, Yinchuan 750004, PR China
| |
Collapse
|
8
|
Lu W, Zha B, Lyu J, LingHu C, Chen J, Deng S, Zhang X, Li L, Wang G. Whole-genome sequencing and genomic analysis of four Akkermansia strains newly isolated from human feces. Front Microbiol 2024; 15:1500886. [PMID: 39736996 PMCID: PMC11683593 DOI: 10.3389/fmicb.2024.1500886] [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: 09/24/2024] [Accepted: 11/25/2024] [Indexed: 01/01/2025] Open
Abstract
Background Numerous studies have demonstrated that Akkermansia is closely associated with human health. These bacteria colonize the mucus layer of the gastrointestinal tract and utilize mucin as their sole source of carbon and nitrogen. Akkermansia spp. exhibit potential as probiotics under specific conditions. However, the gene accumulation curve derived from pan-genome analysis suggests that the genome of Akkermansia strains remains open. Consequently, current genome mining efforts are insufficient to fully capture the intraspecific and interspecific characteristics of Akkermansia, necessitating continuous exploration of the genomic and phenotypic diversity of new isolates. Methods Based on this finding, we sequenced, assembled, and functionally annotated the whole genomes of four new human isolates from our laboratory: AKK-HX001, AKK-HX002, AKK-HX003, and AKK-HX004. Results Phylogenetic analysis revealed that all four isolates belonged to the AmII phylogroup, whereas the type strain DSM 22959 is classified within the AmI phylogroup. Moreover, 2,184 shared homologous genes were identified among the four isolates. Functional annotation using the COG, KEGG, and CAZy databases indicated that the functional genes of the four isolates were primarily associated with metabolism. Two antibiotic resistance genes were identified in AKK-HX001 and AKK-HX002, while three resistance genes were detected in AKK-HX003 and AKK-HX004. Additionally, each of the four isolates possessed two virulence genes and three pathogenicity genes, none of which were associated with pathogenicity. The prediction of mobile genetic elements indicated unequal distributions of GIs among the isolates, and a complete CRISPR system was identified in all isolates except AKK-HX003. Two annotated regions of secondary metabolite biosynthesis genes, both belonging to Terpene, were detected using the antiSMASH online tool. Conclusion These findings indicate that the four Akkermansia isolates, which belong to a phylogroup distinct from the model strain DSM 22959, exhibit lower genetic risk and may serve as potential probiotic resources for future research.
Collapse
Affiliation(s)
- Wenjing Lu
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Biqing Zha
- Jiujiang Center for Disease Control and Prevention, Jiujiang, China
| | - Jie Lyu
- Microbiome Research and Application Center, BYHEALTH Institute of Nutrition and Health, Guangzhou, China
| | - Chenxi LingHu
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Jing Chen
- Chengdu East New District Public Health Center, Chengdu, China
| | - Sisi Deng
- Hunan Provincial Center for Disease Control and Prevention, Changsha, China
| | - Xiangling Zhang
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Liang Li
- Microbiome Research and Application Center, BYHEALTH Institute of Nutrition and Health, Guangzhou, China
| | - Guoqing Wang
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
9
|
Wang L, Hu J, Li K, Zhao Y, Zhu M. Advancements in gene editing technologies for probiotic-enabled disease therapy. iScience 2024; 27:110791. [PMID: 39286511 PMCID: PMC11403445 DOI: 10.1016/j.isci.2024.110791] [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] [Indexed: 09/19/2024] Open
Abstract
Probiotics typically refer to microorganisms that have been identified for their health benefits, and they are added to foods or supplements to promote the health of the host. A growing number of probiotic strains have been identified lately and developed into valuable regulatory pharmaceuticals for nutritional and medical applications. Gene editing technologies play a crucial role in addressing the need for safe and therapeutic probiotics in disease treatment. These technologies offer valuable assistance in comprehending the underlying mechanisms of probiotic bioactivity and in the development of advanced probiotics. This review aims to offer a comprehensive overview of gene editing technologies applied in the engineering of both traditional and next-generation probiotics. It further explores the potential for on-demand production of customized products derived from enhanced probiotics, with a particular emphasis on the future of gene editing in the development of live biotherapeutics.
Collapse
Affiliation(s)
- Lixuan Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, China, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jing Hu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, China, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Kun Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, China, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuliang Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, China, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Motao Zhu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, China, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
10
|
Montipó S, Menegussi EB, Fontana RC, Camassola M. Strategies for producing probiotic biomass and postbiotics from Akkermansia muciniphila in submerged cultivations incorporating prebiotic sources. World J Microbiol Biotechnol 2024; 40:314. [PMID: 39249571 DOI: 10.1007/s11274-024-04129-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 09/02/2024] [Indexed: 09/10/2024]
Abstract
This research propounds an innovative technology focused on sustainability to increase the biomass yield of Akkermansia muciniphila, the next-generation probiotic, using prebiotic sources to replace or reduce animal mucin levels. A series of experimental design approaches were developed aiming to optimize the growth of Akkermansiamuciniphila by incorporating extracts of green leafy vegetables and edible mushroom into the cultivation media. Experiments using kale extract (KE), Brassica oleracea L., associated with lyophilized mushroom extract (LME) of Pleurotus ostreatus were the most promising, highlighting the assays with 0.376% KE and 0.423% LME or 1.05% KE and 0.5% LME, in which 3.5 × 1010 CFU (Colony Forming Units) mL- 1 was achieved - higher than in experiments in optimized synthetic media. Such results enhance the potential of using KE and LME not only as mucin substitutes, but also as a source to increase Akkermansia muciniphila biomass yields and release short-chain fatty acids. The work is relevant to the food and pharmaceutical industries in the preparation of the probiotic ingredient.
Collapse
Affiliation(s)
- Sheila Montipó
- Biotechnology Institute, University of Caxias do Sul, Caxias do Sul, 95070-560, Brazil.
| | | | | | - Marli Camassola
- Biotechnology Institute, University of Caxias do Sul, Caxias do Sul, 95070-560, Brazil
| |
Collapse
|
11
|
Rowaiye A, Ibeanu GC, Bur D, Nnadi S, Mgbeke OE, Morikwe U. Gut microbiota alteration - Cancer relationships and synbiotic roles in cancer therapies. THE MICROBE 2024; 4:100096. [DOI: 10.1016/j.microb.2024.100096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
12
|
Li K, Ding W, Li X, Gao H, Wang S, Li T, Zhao H, Zhang S. Intestinal Akkermansia muciniphila is Beneficial to Functional Recovery Following Ischemic Stroke. J Neuroimmune Pharmacol 2024; 19:43. [PMID: 39141019 DOI: 10.1007/s11481-024-10146-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 08/05/2024] [Indexed: 08/15/2024]
Abstract
Recent studies have demonstrated the interaction between gut microbiota and brain on ischemic stroke, but the roles of gut microbiota in the pathophysiology of ischemic stroke remain largely unclear. In this study, we detected a significant increase of intestinal Akkermansia muciniphila (AKK) following ischemic stroke by a rose bengal photothrombosis model. To investigate the function and mechanism of AKK on ischemic stroke, we performed the AKK administration prior to stroke surgery. The results showed that mice treated with AKK gained significantly higher body weight and behaved better than those in PBS group at 3 days after ischemic stroke. Consistently, AKK administration remarkably decreased the infarct volumes as well as the density of degenerating neurons and apoptotic cells after ischemic stroke. Notably, AKK is a potential therapeutic target in immune-related disorders connected to the microbiota, and inflammation is crucially involved in the pathophysiological process of ischemic stroke. For the determination of underlying mechanisms of this protective effect, we investigated whether there are associations between AKK and neuroinflammation following ischemic stroke. The results suggested that AKK administration significantly reduced the activation of astrocytes and microglia but up-regulated multiple anti-inflammatory factors following ischemic stroke. Therefore, our study highlighted the beneficial roles of intestinal AKK on ischemic stroke and provided a new perspective for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Kemin Li
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Sciences, Lanzhou University, No. 222 South Tianshui Road, Lanzhou, Gansu Province, 730000, China
| | - Wancong Ding
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Sciences, Lanzhou University, No. 222 South Tianshui Road, Lanzhou, Gansu Province, 730000, China
| | - Xinrui Li
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Sciences, Lanzhou University, No. 222 South Tianshui Road, Lanzhou, Gansu Province, 730000, China
| | - Hao Gao
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Sciences, Lanzhou University, No. 222 South Tianshui Road, Lanzhou, Gansu Province, 730000, China
| | - Shuang Wang
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Sciences, Lanzhou University, No. 222 South Tianshui Road, Lanzhou, Gansu Province, 730000, China
| | - Ting Li
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Sciences, Lanzhou University, No. 222 South Tianshui Road, Lanzhou, Gansu Province, 730000, China
| | - Haiyu Zhao
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Sciences, Lanzhou University, No. 222 South Tianshui Road, Lanzhou, Gansu Province, 730000, China.
| | - Shengxiang Zhang
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Sciences, Lanzhou University, No. 222 South Tianshui Road, Lanzhou, Gansu Province, 730000, China.
| |
Collapse
|
13
|
Yu Y, Li L, Yang Q, Xue J, Wang B, Xie M, Shangguan W, Zhu Z, Wu P. Akkermansia muciniphila Metabolite Inosine Inhibits Castration Resistance in Prostate Cancer. Microorganisms 2024; 12:1653. [PMID: 39203495 PMCID: PMC11356635 DOI: 10.3390/microorganisms12081653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 07/26/2024] [Accepted: 08/05/2024] [Indexed: 09/03/2024] Open
Abstract
Prostate cancer (PCa) is initially sensitive to androgen deprivation therapy (ADT) but ultimately develops resistance and progresses to castration-resistant prostate cancer (CRPC) with a poor prognosis. This study indicated that some PCa patients and mice were more sensitive to ADT and entered CRPC later, which was related to the gut microbiota, especially the enrichment of Akkermansia muciniphila (AKK). Untargeted metabolomics analysis found that serum inosine level was upregulated in the treatment-sensitive group and significantly correlated with AKK. Furthermore, we revealed that intestinal permeability and serum lipopolysaccharide (LPS) levels increased in treatment-resistant mice. LPS stimulated the upregulation of p-NF-κB p65 and AR in tumors. Supplementing AKK metabolite inosine could alleviate intestinal barrier damage and reduce serum LPS level, ultimately inhibiting castration resistance via the LPS/NF-κB/AR axis. Finally, we constructed a predictive model for CRPC combining gut microbiota and clinical information (AUC = 0.729). This study revealed the potential mechanism of gut microbiota on CRPC and provided potential therapeutic targets and prognostic indicators.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Peng Wu
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; (Y.Y.); (L.L.); (Q.Y.); (J.X.); (B.W.); (M.X.); (W.S.); (Z.Z.)
| |
Collapse
|
14
|
Lv N, Wang C, Zhou H, Ma X, Yu X, Ren D. Genome- and Toxicology-Based Safety Assessment of Probiotic Akkermansia muciniphila ONE Isolated from Humans. Foods 2024; 13:1979. [PMID: 38998485 PMCID: PMC11241434 DOI: 10.3390/foods13131979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 06/15/2024] [Accepted: 06/18/2024] [Indexed: 07/14/2024] Open
Abstract
In this study, the genome of Akkermansia muciniphila ONE (designated AKK ONE) was sequenced, assembled, and analyzed. In addition, the safety of this strain was further evaluated by toxicological studies. The results showed that the AKK ONE genome is contained on a single chromosome with a total length of 2,817,524 bp and an average GC content of 55.48%. In total, 2411, 1131, 1168, 1745, and 1402 genes were annotated to the NR, GO, KEGG, COG, and SwissProt database, respectively. Potential resistance genes, adeF, tetW, ANT(3″)-IIa, and aadA1 were detected. AKK ONE was sensitive to ampicillin, ceftriaxone, cefotaxime, meropenem, tetracycline, and chloramphenicol and resistant to moxifloxacin. No potential virulence-related genes were detected. The PathogenFinder database analysis showed that AKK ONE was a non-potential human pathogen. This strain had good gastroenteric fluid tolerance and a weak ability to colonize the gut. No test item-related adverse effects were observed in the acute and subchronic toxicity test. AKK ONE did not display mutagenic activity either. This strain did not change the hematological and clinical biochemical parameters of mice. The weights of the organs were not affected by AKK ONE treatment. These results support that AKK ONE is safe for use as a probiotic at a dose of 8.28 × 109 CFU/kg bw/day.
Collapse
Affiliation(s)
- Na Lv
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, China; (N.L.); (C.W.); (H.Z.)
| | - Caiping Wang
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, China; (N.L.); (C.W.); (H.Z.)
| | - Hongtao Zhou
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, China; (N.L.); (C.W.); (H.Z.)
| | - Xin Ma
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China; (X.M.); (X.Y.)
| | - Xueping Yu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China; (X.M.); (X.Y.)
| | - Dayong Ren
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, China; (N.L.); (C.W.); (H.Z.)
| |
Collapse
|
15
|
Yan Y, Dai T, Guo M, Zhao X, Chen C, Zhou Y, Qin M, Xu L, Zhao J. A review of non-classical MAPK family member, MAPK4: A pivotal player in cancer development and therapeutic intervention. Int J Biol Macromol 2024; 271:132686. [PMID: 38801852 DOI: 10.1016/j.ijbiomac.2024.132686] [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: 02/23/2024] [Revised: 05/17/2024] [Accepted: 05/24/2024] [Indexed: 05/29/2024]
Abstract
Mitogen-Activated Protein Kinases (MAPKs) are serine/threonine protein kinases that play a crucial role in transmitting extracellular signals to the intracellular environment, influencing a wide range of cellular processes including proliferation, differentiation, apoptosis, metabolic activities, immune function and stress response. MAPK4, a non-classical MAPK, is frequently overexpressed in various malignancies, including prostate, breast, cervix, thyroid, and gliomas. It orchestrates cell proliferation, migration, and apoptosis via the AKT/mTOR and/or PDK1 signaling pathways, thus facilitating tumor cell growth. Furthermore, MAPK4 expression is closely associated with the effectiveness of specific inhibitors like PI3K and PARP1, and also correlate with the survival rates of cancer patients. Increasing evidence highlights MAPK4's involvement in the tumor microenvironment, modulating immune response and inflammation-related diseases. This review comprehensively explores the structure, function, and oncogenic role of MAPK4, providing a deeper understanding of its activation and mechanisms of action in tumorigenesis, which might be helpful for the development of innovative therapeutic strategies for cancer management.
Collapse
Affiliation(s)
- Yaping Yan
- Department of Immunology, Zunyi Medical University, Guizhou 563000, China; Key Laboratory of Gene Detection and Treatment of Guizhou province, Zunyi 563000, China
| | - Tengkun Dai
- Department of Immunology, Zunyi Medical University, Guizhou 563000, China; Key Laboratory of Gene Detection and Treatment of Guizhou province, Zunyi 563000, China
| | - Mengmeng Guo
- Department of Immunology, Zunyi Medical University, Guizhou 563000, China; Key Laboratory of Gene Detection and Treatment of Guizhou province, Zunyi 563000, China
| | - Xu Zhao
- Key Laboratory of Gene Detection and Treatment of Guizhou province, Zunyi 563000, China; School of Medicine, Guizhou University, Guiyang 550025, Guizhou, China
| | - Chao Chen
- Key Laboratory of Gene Detection and Treatment of Guizhou province, Zunyi 563000, China; School of Medicine, Guizhou University, Guiyang 550025, Guizhou, China
| | - Ya Zhou
- Key Laboratory of Gene Detection and Treatment of Guizhou province, Zunyi 563000, China; Department of Medical physics, Zunyi Medical University, Guizhou 563000, China
| | - Ming Qin
- Department of Immunology, Zunyi Medical University, Guizhou 563000, China; Key Laboratory of Gene Detection and Treatment of Guizhou province, Zunyi 563000, China
| | - Lin Xu
- Department of Immunology, Zunyi Medical University, Guizhou 563000, China; Key Laboratory of Gene Detection and Treatment of Guizhou province, Zunyi 563000, China.
| | - Juanjuan Zhao
- Department of Immunology, Zunyi Medical University, Guizhou 563000, China; Key Laboratory of Gene Detection and Treatment of Guizhou province, Zunyi 563000, China.
| |
Collapse
|
16
|
Xiang X, Li Q, Wan J, Chen C, Guo M, He Z, Wang D, Zhao X, Xu L. The role of amino acid metabolism in autoimmune hepatitis. Biomed Pharmacother 2024; 173:116452. [PMID: 38503235 DOI: 10.1016/j.biopha.2024.116452] [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: 12/23/2023] [Revised: 03/04/2024] [Accepted: 03/15/2024] [Indexed: 03/21/2024] Open
Abstract
Autoimmune hepatitis (AIH) is an inflammatory chronic liver disease with persistent and recurrent immune-mediated liver injury. The exact cause of AIH is still not fully understood, but it is believed to be primarily due to an abnormal activation of the immune system, leading to autoimmune injury caused by the breakdown of autoimmune tolerance. Although the pathogenesis of AIH remains unclear, recent studies have shown that abnormalities in amino acid metabolism play significant roles in its development. These abnormalities in amino acid metabolism can lead to remodeling of metabolic processes, activation of signaling pathways, and immune responses, which may present new opportunities for clinical intervention in AIH. In this paper, we first briefly outline the recent progress of clinically relevant research on AIH, focusing on the role of specific amino acid metabolism (including glutamine, cysteine, tryptophan, branched-chain amino acids, etc.) and their associated metabolites, as well as related pathways, in the development of AIH. Furthermore, we discuss the scientific issues that remain to be resolved regarding amino acid metabolism, AIH development and related clinical interventions, with the aim of contributing to the future development of amino acid metabolism-based as a new target for the clinical diagnosis and treatment of AIH.
Collapse
Affiliation(s)
- Xiaorong Xiang
- Nanshan Class, Zunyi Medical University, Zunyi 563000, China; Guizhou Key Laboratory of Gene Detection and Therapy, Zunyi 563000, China
| | - Qihong Li
- Guizhou Key Laboratory of Gene Detection and Therapy, Zunyi 563000, China
| | - Jiajia Wan
- Guizhou Key Laboratory of Gene Detection and Therapy, Zunyi 563000, China
| | - Chao Chen
- Guizhou Key Laboratory of Gene Detection and Therapy, Zunyi 563000, China
| | - Mengmeng Guo
- Guizhou Key Laboratory of Gene Detection and Therapy, Zunyi 563000, China
| | - Zhixu He
- Innovation Center for Tissue Damage Repair, Ministry of Education, Zunyi, Guizhou 563000, China
| | - Donghong Wang
- Department of Obstetrics and Gynecology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, China.
| | - Xu Zhao
- Medical College of Guizhou University, Guiyang 550025, China.
| | - Lin Xu
- Guizhou Key Laboratory of Gene Detection and Therapy, Zunyi 563000, China; Innovation Center for Tissue Damage Repair, Ministry of Education, Zunyi, Guizhou 563000, China.
| |
Collapse
|
17
|
Elisia I, Yeung M, Kowalski S, Shyp T, Tee J, Hollman S, Wong A, King J, Dyer R, Sorensen PH, Krystal G. A ketogenic diet rich in fish oil is superior to other fats in preventing NNK-induced lung cancer in A/J mice. Sci Rep 2024; 14:5610. [PMID: 38453966 PMCID: PMC10920871 DOI: 10.1038/s41598-024-55167-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 02/21/2024] [Indexed: 03/09/2024] Open
Abstract
Given that ketogenic diets (KDs) are extremely high in dietary fat, we compared different fats in KDs to determine which was the best for cancer prevention. Specifically, we compared a Western and a 15% carbohydrate diet to seven different KDs, containing either Western fats or fats enriched in medium chain fatty acids (MCTs), milk fat (MF), palm oil (PO), olive oil (OO), corn oil (CO) or fish oil (FO) for their ability to reduce nicotine-derived nitrosamine ketone (NNK)-induced lung cancer in mice. While all the KDs tested were more effective at reducing lung nodules than the Western or 15% carbohydrate diet, the FO-KD was most effective at reducing lung nodules. Correlating with this, mice on the FO-KD had low blood glucose and the highest β-hydroxybutyrate level, lowest liver fatty acid synthase/carnitine palmitoyl-1a ratio and a dramatic increase in fecal Akkermansia. We found no liver damage induced by the FO-KD, while the ratio of total cholesterol/HDL was unchanged on the different diets. We conclude that a FO-KD is superior to KDs enriched in other fats in reducing NNK-induced lung cancer, perhaps by being the most effective at skewing whole-body metabolism from a dependence on glucose to fats as an energy source.
Collapse
Affiliation(s)
- Ingrid Elisia
- The Terry Fox Laboratory, BC Cancer Research Centre, 675 West 10th Avenue, Vancouver, BC, V5Z 1L3, Canada
| | - Michelle Yeung
- The Terry Fox Laboratory, BC Cancer Research Centre, 675 West 10th Avenue, Vancouver, BC, V5Z 1L3, Canada
| | - Sara Kowalski
- The Terry Fox Laboratory, BC Cancer Research Centre, 675 West 10th Avenue, Vancouver, BC, V5Z 1L3, Canada
| | - Taras Shyp
- Department of Molecular Oncology, BC Cancer, Vancouver, BC, V5Z 1L3, Canada
| | - Jason Tee
- The Terry Fox Laboratory, BC Cancer Research Centre, 675 West 10th Avenue, Vancouver, BC, V5Z 1L3, Canada
| | - Serena Hollman
- The Terry Fox Laboratory, BC Cancer Research Centre, 675 West 10th Avenue, Vancouver, BC, V5Z 1L3, Canada
| | - Amy Wong
- The Terry Fox Laboratory, BC Cancer Research Centre, 675 West 10th Avenue, Vancouver, BC, V5Z 1L3, Canada
| | - Janette King
- Analytical Core for Metabolomics and Nutrition, BC Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Roger Dyer
- Analytical Core for Metabolomics and Nutrition, BC Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Poul H Sorensen
- Department of Molecular Oncology, BC Cancer, Vancouver, BC, V5Z 1L3, Canada
| | - Gerald Krystal
- The Terry Fox Laboratory, BC Cancer Research Centre, 675 West 10th Avenue, Vancouver, BC, V5Z 1L3, Canada.
| |
Collapse
|
18
|
Yu Z, Xiaojia L, Wei Z, Jian Z, Aiting W, Jing W, Lin Y, Bangwei C, Dan Y. Baicalin circumvents anti-PD-1 resistance by regulating the gut microbiota metabolite short-chain fatty acids. Pharmacol Res 2024; 199:107033. [PMID: 38061593 DOI: 10.1016/j.phrs.2023.107033] [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: 10/05/2023] [Revised: 11/20/2023] [Accepted: 12/04/2023] [Indexed: 01/13/2024]
Abstract
Baicalin is a small molecule medication used to treat hepatitis. Our research group discovered that administering baicalin orally to mice following fecal microbiota transplantation from patients resistant to ICIs supported anti-PD-1 activity. However, the precise mechanisms behind this effect are presently unknown. In this present study, ATB-treated C57BL/6 J mice received FMT from patients with advanced NSCLC amenable to αPD-1. Additionally, subcutaneous LLC cells were injected into the mice. Baicalin oral gavage and αPD-1 injection were administered to the mice on days 3 and 9 after tumour inoculation. 16 S rRNA, metabolomics, and flow cytometry were utilized to clarify the mechanisms of baicalin's relief of immunosuppression. The results indicated that oral administration of baicalin enriched bacteria such as Akkermansia and Clostridia_UCG-014, resulted in an increase in SCFAs, which improved the ratio of PD-1+ (CD8+ T cell/Treg) and promoted the levels of IFN-γ+ CD8+ T cells and TNF-α+ CD8+ T cells within the tumour microenvironment. In conclusion, baicalin regulates the metabolites of the gut microbiota to improve the PD-1+ (CD8+ T cell/Treg) balance and circumvent anti-PD-1 resistance. This is achieved through the regulation of short-chain fatty acids.
Collapse
Affiliation(s)
- Zhang Yu
- Beijing Institute of Clinical Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Liu Xiaojia
- Beijing Institute of Clinical Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Zhao Wei
- Beijing Institute of Clinical Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Zhang Jian
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Wang Aiting
- Beijing Institute of Clinical Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Wang Jing
- Department of Oncology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Yang Lin
- National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Cao Bangwei
- Department of Oncology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China.
| | - Yan Dan
- Beijing Institute of Clinical Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China.
| |
Collapse
|