1
|
Wang Y, Dou W, Qian X, Chen H, Zhang Y, Yang L, Wu Y, Xu X. Advancements in the study of short-chain fatty acids and their therapeutic effects on atherosclerosis. Life Sci 2025; 369:123528. [PMID: 40049368 DOI: 10.1016/j.lfs.2025.123528] [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: 12/01/2024] [Revised: 02/15/2025] [Accepted: 03/02/2025] [Indexed: 03/09/2025]
Abstract
Atherosclerosis (AS) remains a leading cause of cardiovascular disease and mortality globally. This chronic condition is characterized by inflammation, lipid accumulation, and the deposition of cellular components within arterial walls. Emerging evidence has highlighted the multifaceted therapeutic potential of short-chain fatty acids (SCFAs) in mitigating AS progression. SCFAs have demonstrated anti-inflammatory properties and the ability to regulate immune responses, metabolic pathways, vascular integrity, and intestinal barrier function in animal models of AS. Consequently, SCFAs have garnered significant attention as a promising approach for the prevention and treatment of AS. However, further clinical trials and studies are necessary to fully elucidate the underlying mechanisms and effects of SCFAs. Additionally, different types of SCFAs may exert distinct impacts, necessitating more in-depth investigation into their specific roles and mechanisms. This review provides an overview of the diverse cellular mechanisms contributing to AS formation, as well as a discussion of the significance of SCFAs in AS pathogenesis and their multifaceted therapeutic potential. Nonetheless, additional research is warranted to comprehensively understand and harness the potential of various SCFAs in the context of AS.
Collapse
Affiliation(s)
- Yongsen Wang
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Taiping Street 25, Luzhou, Sichuan 646000, PR China; Department of Hepatobiliary Pancreatic and Splcnic Surgery, Luzhou People's Hospital, Luzhou, Sichuan 646000, PR China; Department of Vascular and Breast Surgery, The Third Hospital of Mianyang, Sichuan Mental Health Center, Mianyang, Sichuan 621000, PR China
| | - Wei Dou
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Taiping Street 25, Luzhou, Sichuan 646000, PR China
| | - Xin Qian
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Taiping Street 25, Luzhou, Sichuan 646000, PR China
| | - Hao Chen
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Taiping Street 25, Luzhou, Sichuan 646000, PR China
| | - Yi Zhang
- Department of Vascular and Breast Surgery, The Third Hospital of Mianyang, Sichuan Mental Health Center, Mianyang, Sichuan 621000, PR China
| | - Liu Yang
- Department of Hepatobiliary Pancreatic and Splcnic Surgery, Luzhou People's Hospital, Luzhou, Sichuan 646000, PR China
| | - Ya Wu
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Taiping Street 25, Luzhou, Sichuan 646000, PR China
| | - Xiongfei Xu
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Taiping Street 25, Luzhou, Sichuan 646000, PR China.
| |
Collapse
|
2
|
Tao H, Geng J, Bai L, Su D, Zhao Y, Xu G, Zhang M. Regulation of innate lymphoid cell by microbial metabolites. J Mol Med (Berl) 2025:10.1007/s00109-025-02530-3. [PMID: 40128460 DOI: 10.1007/s00109-025-02530-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 02/27/2025] [Accepted: 03/10/2025] [Indexed: 03/26/2025]
Abstract
Innate lymphoid cells (ILCs) are a unique category of immune cell that lack antigen-specific receptors yet possess the capacity to detect signals from the surrounding tissue. The majority of ILCs reside in the lymphoid and mucosal tissues, maintaining close associations with the microbiota. Beyond the contributions of accessory cells and adaptive immune cells, accumulating studies demonstrate that microbial metabolites serve a crucial role in mediating the relationship between ILCs and the microbiota. In this review, we highlight and summarize the roles of microbial metabolites from different sources in modulating ILC subsets, proposing these metabolites as potential therapeutic mechanisms in ILC-mediated diseases.
Collapse
Affiliation(s)
- Hongji Tao
- Department of Gastroenterology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, China
| | - Jingjing Geng
- Division of Gastroenterology and Hepatology; Shanghai Institute of Digestive Disease; NHC Key Laboratory of Digestive Diseases; State Key Laboratory for Oncogenes and Related Genes; Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 145 Middle Shandong Road, Shanghai, 200001, China
| | - Long Bai
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Dan Su
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, 14853, USA
| | - Yu Zhao
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, 60637, USA
| | - Guifang Xu
- Department of Gastroenterology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, China.
| | - Mingming Zhang
- Division of Gastroenterology and Hepatology; Shanghai Institute of Digestive Disease; NHC Key Laboratory of Digestive Diseases; State Key Laboratory for Oncogenes and Related Genes; Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 145 Middle Shandong Road, Shanghai, 200001, China.
| |
Collapse
|
3
|
Li S, Duan Y, Luo S, Zhou F, Wu Q, Lu Z. Short-chain fatty acids and cancer. Trends Cancer 2025; 11:154-168. [PMID: 39638744 DOI: 10.1016/j.trecan.2024.11.003] [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: 10/10/2024] [Revised: 11/04/2024] [Accepted: 11/04/2024] [Indexed: 12/07/2024]
Abstract
Short-chain fatty acids (SCFAs), derived from the diet and the microbiota, serve as crucial links between the diet, gut microbiota, metabolism, immunity, and cancer. They function as energy sources through β-oxidation and regulate macromolecular synthesis, G protein-coupled receptor (GPCR) and histone deacetylase (HDAC) activities, protein modifications, signaling pathways, and gene expression in cells within the tumor microenvironment, particularly in tumor and immune cells. The critical role of SCFAs in maintaining normal homeostasis and influencing tumor progression highlights the potential of targeting SCFA-mediated cellular processes for cancer prevention and treatment.
Collapse
Affiliation(s)
- Shan Li
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310029, China; Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, Zhejiang 310029, China
| | - Yixin Duan
- Department of Oncology, Cancer Institute of The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266071, China
| | - Shudi Luo
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310029, China; Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, Zhejiang 310029, China
| | - Fangxin Zhou
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310029, China; Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, Zhejiang 310029, China
| | - Qingang Wu
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310029, China; Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, Zhejiang 310029, China
| | - Zhimin Lu
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310029, China; Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, Zhejiang 310029, China.
| |
Collapse
|
4
|
Chen W, Ma Q, Li Y, Wei L, Zhang Z, Khan A, Khan MZ, Wang C. Butyrate Supplementation Improves Intestinal Health and Growth Performance in Livestock: A Review. Biomolecules 2025; 15:85. [PMID: 39858479 PMCID: PMC11763988 DOI: 10.3390/biom15010085] [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: 11/20/2024] [Revised: 12/30/2024] [Accepted: 01/06/2025] [Indexed: 01/27/2025] Open
Abstract
Butyrate supplementation has gained considerable attention for its potential benefits in livestock, particularly concerning intestinal health and growth performance. This review synthesizes recent research on the diverse roles of butyrate, across various livestock species. As a short-chain fatty acid, butyrate is known for enhancing intestinal development, improving immune function, and modulating microbial diversity. Studies indicate that butyrate supports gut barrier integrity, reduces inflammation, and optimizes feed efficiency, especially during the critical weaning and post-weaning periods in calves, piglets, and lambs. Supplementation with butyrate in livestock has been shown to increase average daily gain (ADG), improve gut microbiota balance, promote growth, enhance gut health, boost antioxidant capacity, and reduce diarrhea. Additionally, butyrate plays a role in the epigenetic regulation of gene expression through histone acetylation, influencing tissue development and immune modulation. Its anti-inflammatory and antioxidant effects have been demonstrated across various species, positioning butyrate as a potential therapeutic agent in animal nutrition. This review suggests that optimizing butyrate supplementation strategies to meet the specific needs of each species may yield additional benefits, establishing butyrate as an important dietary additive for enhancing growth performance and health in livestock.
Collapse
Affiliation(s)
- Wenting Chen
- School of Agricultural Science and Engineering, Liaocheng University, Liaocheng 252000, China
| | - Qingshan Ma
- School of Agricultural Science and Engineering, Liaocheng University, Liaocheng 252000, China
| | - Yan Li
- School of Agricultural Science and Engineering, Liaocheng University, Liaocheng 252000, China
| | - Lin Wei
- School of Agricultural Science and Engineering, Liaocheng University, Liaocheng 252000, China
| | - Zhenwei Zhang
- School of Agricultural Science and Engineering, Liaocheng University, Liaocheng 252000, China
| | - Adnan Khan
- Animal Genomics Laboratory, School of Agriculture and Food Science, University College Dublin, D04 V1W8 Dublin, Ireland;
| | - Muhammad Zahoor Khan
- School of Agricultural Science and Engineering, Liaocheng University, Liaocheng 252000, China
| | - Changfa Wang
- School of Agricultural Science and Engineering, Liaocheng University, Liaocheng 252000, China
| |
Collapse
|
5
|
Xia L, Li C, Zhao J, Sun Q, Mao X. Rebalancing immune homeostasis in combating disease: The impact of medicine food homology plants and gut microbiome. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 136:156150. [PMID: 39740376 DOI: 10.1016/j.phymed.2024.156150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 09/27/2024] [Accepted: 10/10/2024] [Indexed: 01/02/2025]
Abstract
BACKGROUND Gut microbiota plays an important role in multiple human physiological processes and an imbalance in it, including the species, abundance, and metabolites can lead to diseases. These enteric microorganisms modulate immune homeostasis by presenting a myriad of antigenic determinants and microbial metabolites. Medicinal and food homologous (MFH) plants, edible herbal materials for both medicine and food, are important parts of Traditional Chinese Medicine (TCM). MFH plants have drawn much attention due to their strong biological activity and low toxicity. However, the interplay of MFH and gut microbiota in rebalancing the immune homeostasis in combating diseases needs systematic illumination. PURPOSE The review discusses the interaction between MFH and gut microbiota, including the effect of MFH on the major group of gut microbiota and the metabolic effect of gut microbiota on MFH. Moreover, how gut microbiota influences the immune system in terms of innate and adaptive immunity is addressed. Finally, the immunoregulatory mechanisms of MFH in regulation of host pathophysiology via gut microbiota are summarized. METHODS Literature was searched, analyzed, and collected using databases, including PubMed, Web of Science, and Google Scholar using relevant keywords. The obtained articles were screened and summarized by the research content of MFH and gut microbiota in immune regulation. RESULTS The review demonstrates the interaction between MFH and gut microbiota in disease prevention and treatment. Not only do the intestinal microorganisms and intestinal mucosa constitute an important immune barrier of the human body, but also lymphoid tissue and diffused immune cells within the mucosa participate in the response of innate immunity and adaptive immunity. MFH modulates immune regulation by affecting intestinal flora, helps maintain the balance of the immune system and interfere with the occurrence and development of a broad category of diseases. CONCLUSION Being absorbed from the gastrointestinal tract, MFH can have profound effects on gut microbiota. In turn, the gut microbiota also actively participate in the bioconversion of complex constituents from MFH, which could further influence their physiological and pharmacological properties. The review deepens the understanding of the relationship among MFH, gut microbiota, immune system, and human diseases and further promotes the progression of additional relevant research.
Collapse
Affiliation(s)
- Lu Xia
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China
| | - Chuangen Li
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China
| | - Jia Zhao
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong SAR, China
| | - Quancai Sun
- Department of Health, Nutrition, and Food sciences, Florida State University, Tallahassee, USA
| | - Xiaowen Mao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China.
| |
Collapse
|
6
|
Hamada M, Nishiyama K, Nomura R, Akitomo T, Mitsuhata C, Yura Y, Nakano K, Matsumoto-Nakano M, Uzawa N, Inaba H. Clinical relationships between the intratumoral microbiome and risk factors for head and neck cancer. Heliyon 2024; 10:e39284. [PMID: 39497974 PMCID: PMC11533578 DOI: 10.1016/j.heliyon.2024.e39284] [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: 07/11/2024] [Revised: 10/09/2024] [Accepted: 10/10/2024] [Indexed: 11/07/2024] Open
Abstract
A bioinformatic analysis is a promising approach to understand the relationship between the vast tumor microbiome and cancer development. In the present study, we studied the relationships between the intratumoral microbiome and classical clinical risk factors using bioinformatics analysis of the Cancer Genome Atlas (TCGA) and the Cancer Microbiome Atlas (TCMA) datasets. We used TCMA database and investigated the abundance of microbes at the genus level in solid normal tissue (n = 22) and the primary tumors of patients with head and neck squamous cell carcinoma (HNSCC) (n = 154) and identified three major tumor microbiomes, Fusobacterium, Prevotella, and Streptococcus. The tissue level of Fusobacterium was higher in primary tumors than in solid normal tissue. However, univariate and multivariate analyses of these 3 microbes showed no significant effects on patient survival. We then extracted 43, 55, or 59 genes that were differentially expressed between the over and under the median groups for Fusobacterium, Prevotella, or Streptococcus using the criteria of >2.5, >1.5, or >2.0 fold and p < 0.05 in the Mann-Whitney U test. The results of a pathway analysis revealed the association of Fusobacterium- and Streptococcus-related genes with the IL-17 signaling pathway and Staphylococcus aureus infection, while Prevotella-associated pathways were not extracted. A protein-protein interaction analysis revealed a dense network in the order of Fusobacterium, Streptococcus, and Prevotella. An investigation of the relationships between the intratumoral microbiome and classical clinical risk factors showed that high levels of Fusobacterium were associated with a good prognosis in the absence of alcohol consumption and smoking, while high levels of Streptococcus were associated with a poor prognosis in the absence of alcohol consumption. In conclusion, intratumoral Fusobacterium and Streptococcus may affect the prognosis of patients with HNSCC, and their effects on HNSCC are modulated by the impact of drinking and smoking.
Collapse
Affiliation(s)
- Masakazu Hamada
- Department of Oral & Maxillofacial Oncology and Surgery, Osaka University Graduate School of Dentistry, 1-8 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Kyoko Nishiyama
- Department of Oral & Maxillofacial Oncology and Surgery, Osaka University Graduate School of Dentistry, 1-8 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Ryota Nomura
- Department of Pediatric Dentistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan
| | - Tatsuya Akitomo
- Department of Pediatric Dentistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan
| | - Chieko Mitsuhata
- Department of Pediatric Dentistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan
| | - Yoshiaki Yura
- Department of Oral & Maxillofacial Oncology and Surgery, Osaka University Graduate School of Dentistry, 1-8 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Kazuhiko Nakano
- Department of Pediatric Dentistry, Osaka University Graduate School of Dentistry, 1-8 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Michiyo Matsumoto-Nakano
- Department of Pediatric Dentistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8525, Japan
| | - Narikazu Uzawa
- Department of Oral & Maxillofacial Oncology and Surgery, Osaka University Graduate School of Dentistry, 1-8 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Hiroaki Inaba
- Department of Pediatric Dentistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8525, Japan
- Department of Dental Hygiene, Kyoto Koka Woman's College, 38, Kuzuno-cho, Nishikyogoku, Ukyo-ku, Kyoto-shi, 615-0882, Japan
| |
Collapse
|
7
|
Baraniuk JN, Eaton-Fitch N, Marshall-Gradisnik S. Meta-analysis of natural killer cell cytotoxicity in myalgic encephalomyelitis/chronic fatigue syndrome. Front Immunol 2024; 15:1440643. [PMID: 39483457 PMCID: PMC11524851 DOI: 10.3389/fimmu.2024.1440643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 09/09/2024] [Indexed: 11/03/2024] Open
Abstract
Reduced natural killer (NK) cell cytotoxicity is the most consistent immune finding in myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS). Meta-analysis of the published literature determined the effect size of the decrement in ME/CFS. Databases were screened for papers comparing NK cell cytotoxicity in ME/CFS and healthy controls. A total of 28 papers and 55 effector:target cell ratio (E:T) data points were collected. Cytotoxicity in ME/CFS was significantly reduced to about half of healthy control levels, with an overall Hedges' g of 0.96 (0.75-1.18). Heterogeneity was high but was explained by the range of E:T ratios, different methods, and potential outliers. The outcomes confirm reproducible NK cell dysfunction in ME/CFS and will guide studies using the NK cell model system for pathomechanistic investigations. Systematic review registration https://www.crd.york.ac.uk/prospero/, identifier CRD42024542140.
Collapse
Affiliation(s)
- James N. Baraniuk
- Department of Medicine, Georgetown University, Washington, DC, United States
- National Centre for Neuroimmunology and Emerging Diseases, Griffith University, Gold Coast, QLD, Australia
| | - Natalie Eaton-Fitch
- National Centre for Neuroimmunology and Emerging Diseases, Griffith University, Gold Coast, QLD, Australia
| | - Sonya Marshall-Gradisnik
- National Centre for Neuroimmunology and Emerging Diseases, Griffith University, Gold Coast, QLD, Australia
| |
Collapse
|
8
|
Shyanti RK, Greggs J, Malik S, Mishra M. Gut dysbiosis impacts the immune system and promotes prostate cancer. Immunol Lett 2024; 268:106883. [PMID: 38852888 PMCID: PMC11249078 DOI: 10.1016/j.imlet.2024.106883] [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: 05/02/2024] [Revised: 05/31/2024] [Accepted: 06/07/2024] [Indexed: 06/11/2024]
Abstract
The gut microbiota is a system of microorganisms in the human gastrointestinal (GI) system, consisting of trillions of microorganisms residing in epithelial surfaces of the body. Gut microbiota are exposed to various external and internal factors and form a unique gut-associated immunity maintained through a balancing act among diverse groups of microorganisms. The role of microbiota in dysbiosis of the gut in aiding prostate cancer development has created an urgency for extending research toward comprehension and preventative measures. The gut microbiota varies among persons based on diet, race, genetic background, and geographic location. Bacteriome, mainly, has been linked to GI complications, metabolism, weight gain, and high blood sugar. Studies have shown that manipulating the microbiome (bacteriome, virome, and mycobiome) through the dietary intake of phytochemicals positively influences physical and emotional health, preventing and delaying diseases caused by microbiota. In this review, we discuss the wealth of knowledge about the GI tract and factors associated with dysbiosis-mediated compromised gut immunity. This review also focuses on the relationship of dysbiosis to prostate cancer, the impact of microbial metabolites short-chain fatty acids (SCFAs) on host health, and the phytochemicals improving health while inhibiting prostate cancer.
Collapse
Affiliation(s)
- Ritis K Shyanti
- Cancer Biology Research and Training Program, Department of Biological Sciences, Alabama State University, AL 36104, USA
| | - Jazmyn Greggs
- Cancer Biology Research and Training Program, Department of Biological Sciences, Alabama State University, AL 36104, USA
| | - Shalie Malik
- Department of Zoology, University of Lucknow, Lucknow, Uttar Pradesh 226007, India
| | - Manoj Mishra
- Cancer Biology Research and Training Program, Department of Biological Sciences, Alabama State University, AL 36104, USA.
| |
Collapse
|
9
|
Garrison EC, Brown AMV, Salazar MM, Barr B, Moustaid-Moussa N, Gollahon LS. Microbiome Taxonomic and Functional Differences in C3H/HeJ Mice Fed a Long-Term High-Fat Diet with Beef Protein ± Ammonium Hydroxide Supplementation. Nutrients 2024; 16:1613. [PMID: 38892546 PMCID: PMC11174526 DOI: 10.3390/nu16111613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/17/2024] [Accepted: 05/21/2024] [Indexed: 06/21/2024] Open
Abstract
Studies have suggested that alkalinized foods may reduce the effects of the acidogenic Western diet in promoting obesity, metabolic syndrome, type 2 diabetes, cancer, and coronary heart disease. Indeed, a recent study in mice fed a high-fat diet containing dietary beef supplemented with ammonium hydroxide showed improvement in a suite of metabolic outcomes. However, the effects of dietary protein ammonium supplementation on the microbiome remain unknown. In this study, the effects of ammonium supplementation on beef protein towards microbiome taxa and function in a high-fat diet were analyzed. Fecal microbiomes were characterized using a shotgun metagenomic approach for 16-month-old male and female mice after long-term diet treatments. The results for ammoniated diets showed that several bacteria known to be associated with health benefits increased significantly, including Romboutsia, Oscillospiraceae, and Lactococcus cremoris. The beneficial mucin-degrader Akkermansia was especially abundant, with a high prevalence (~86%) in females. Concurrently, the phyla Actinomycetota (Actinobacteria) and Bacteroidota (Bacteroidetes) were significantly reduced. While sex was a confounding factor affecting microbiome responses to ammonium supplementation in dietary protein, it is worth noting that several putatively beneficial microbiome functions increased with ammonium supplementation, such as glycine betaine transport, xenobiotic detoxification, enhanced defense, and others. Conversely, many disease-associated microbiome functions reduced. Importantly, modifying protein pH alone via ammonium supplementation induced beneficial microbiota changes. Taken together, these results suggest that ammonium-supplemented proteins may mediate some negative microbiome-associated effects of high-fat/Western diets.
Collapse
Affiliation(s)
- Emily C. Garrison
- Department of Biological Sciences, Texas Tech University, 2500 Broadway, Lubbock, TX 79409, USA; (E.C.G.); (A.M.V.B.); (M.M.S.); (B.B.)
| | - Amanda M. V. Brown
- Department of Biological Sciences, Texas Tech University, 2500 Broadway, Lubbock, TX 79409, USA; (E.C.G.); (A.M.V.B.); (M.M.S.); (B.B.)
| | - McKinlee M. Salazar
- Department of Biological Sciences, Texas Tech University, 2500 Broadway, Lubbock, TX 79409, USA; (E.C.G.); (A.M.V.B.); (M.M.S.); (B.B.)
| | - Benjamin Barr
- Department of Biological Sciences, Texas Tech University, 2500 Broadway, Lubbock, TX 79409, USA; (E.C.G.); (A.M.V.B.); (M.M.S.); (B.B.)
| | - Naima Moustaid-Moussa
- Department of Nutritional Sciences, Texas Tech University, 2500 Broadway, Lubbock, TX 79409, USA;
- Obesity Research Institute, Texas Tech University, 2500 Broadway, Lubbock, TX 79409, USA
| | - Lauren S. Gollahon
- Department of Biological Sciences, Texas Tech University, 2500 Broadway, Lubbock, TX 79409, USA; (E.C.G.); (A.M.V.B.); (M.M.S.); (B.B.)
- Obesity Research Institute, Texas Tech University, 2500 Broadway, Lubbock, TX 79409, USA
| |
Collapse
|
10
|
Apaza CJ, Días M, García Tejedor A, Boscá L, Laparra Llopis JM. Contribution of Nucleotide-Binding Oligomerization Domain-like (NOD) Receptors to the Immune and Metabolic Health. Biomedicines 2024; 12:341. [PMID: 38397943 PMCID: PMC10886542 DOI: 10.3390/biomedicines12020341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 01/24/2024] [Accepted: 01/30/2024] [Indexed: 02/25/2024] Open
Abstract
Nucleotide-binding oligomerization domain-like (NOD) receptors rely on the interface between immunity and metabolism. Dietary factors constitute critical players in the activation of innate immunity and modulation of the gut microbiota. The latter have been involved in worsening or improving the control and promotion of diseases such as obesity, type 2 diabetes, metabolic syndrome, diseases known as non-communicable metabolic diseases (NCDs), and the risk of developing cancer. Intracellular NODs play key coordinated actions with innate immune 'Toll-like' receptors leading to a diverse array of gene expressions that initiate inflammatory and immune responses. There has been an improvement in the understanding of the molecular and genetic implications of these receptors in, among others, such aspects as resting energy expenditure, insulin resistance, and cell proliferation. Genetic factors and polymorphisms of the receptors are determinants of the risk and severity of NCDs and cancer, and it is conceivable that dietary factors may have significant differential consequences depending on them. Host factors are difficult to influence, while environmental factors are predominant and approachable with a preventive and/or therapeutic intention in obesity, T2D, and cancer. However, beyond the recognition of the activation of NODs by peptidoglycan as its prototypical agonist, the underlying molecular response(s) and its consequences on these diseases remain ill-defined. Metabolic (re)programming is a hallmark of NCDs and cancer in which nutritional strategies might play a key role in preventing the unprecedented expansion of these diseases. A better understanding of the participation and effects of immunonutritional dietary ingredients can boost integrative knowledge fostering interdisciplinary science between nutritional precision and personalized medicine against cancer. This review summarizes the current evidence concerning the relationship(s) and consequences of NODs on immune and metabolic health.
Collapse
Affiliation(s)
- César Jeri Apaza
- Molecular Immunonutrition Group, Madrid Institute for Advanced Studies in Food (IMDEA Food), Ctra Cantoblanco, 8, 28049 Madrid, Spain;
| | - Marisol Días
- Center of Biological Enginneering (CEB), Iberian Nantotechnology Laboratory (INL), University of Minho, 4715-330 Braga, Portugal;
| | - Aurora García Tejedor
- Bioactivity and Nutritional Immunology Group (BIOINUT), Faculty of Health Sciences, Universidad Internacional de Valencia (VIU), Pintor Sorolla 21, 46002 Valencia, Spain;
| | - Lisardo Boscá
- Instituto de Investigaciones Biomédicas Alberto Sols-Morreale (CSIC-UAM), Arturo Duperier 4, 28029 Madrid, Spain;
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Melchor Fernández Almagro 6, 28029 Madrid, Spain
| | - José Moisés Laparra Llopis
- Molecular Immunonutrition Group, Madrid Institute for Advanced Studies in Food (IMDEA Food), Ctra Cantoblanco, 8, 28049 Madrid, Spain;
| |
Collapse
|
11
|
Yeo K, Li R, Wu F, Bouras G, Mai LTH, Smith E, Wormald PJ, Valentine R, Psaltis AJ, Vreugde S, Fenix K. Identification of consensus head and neck cancer-associated microbiota signatures: a systematic review and meta-analysis of 16S rRNA and The Cancer Microbiome Atlas datasets. J Med Microbiol 2024; 73. [PMID: 38299619 DOI: 10.1099/jmm.0.001799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2024] Open
Abstract
Introduction. Multiple reports have attempted to describe the tumour microbiota in head and neck cancer (HNSC).Gap statement. However, these have failed to produce a consistent microbiota signature, which may undermine understanding the importance of bacterial-mediated effects in HNSC.Aim. The aim of this study is to consolidate these datasets and identify a consensus microbiota signature in HNSC.Methodology. We analysed 12 published HNSC 16S rRNA microbial datasets collected from cancer, cancer-adjacent and non-cancer tissues to generate a consensus microbiota signature. These signatures were then validated using The Cancer Microbiome Atlas (TCMA) database and correlated with the tumour microenvironment phenotypes and patient's clinical outcome.Results. We identified a consensus microbial signature at the genus level to differentiate between HNSC sample types, with cancer and cancer-adjacent tissues sharing more similarity than non-cancer tissues. Univariate analysis on 16S rRNA datasets identified significant differences in the abundance of 34 bacterial genera among the tissue types. Paired cancer and cancer-adjacent tissue analyses in 16S rRNA and TCMA datasets identified increased abundance in Fusobacterium in cancer tissues and decreased abundance of Atopobium, Rothia and Actinomyces in cancer-adjacent tissues. Furthermore, these bacteria were associated with different tumour microenvironment phenotypes. Notably, high Fusobacterium signature was associated with high neutrophil (r=0.37, P<0.0001), angiogenesis (r=0.38, P<0.0001) and granulocyte signatures (r=0.38, P<0.0001) and better overall patient survival [continuous: HR 0.8482, 95 % confidence interval (CI) 0.7758-0.9273, P=0.0003].Conclusion. Our meta-analysis demonstrates a consensus microbiota signature for HNSC, highlighting its potential importance in this disease.
Collapse
Affiliation(s)
- Kenny Yeo
- Discipline of Surgery, Adelaide Medical School, The University of Adelaide, Adelaide SA, 5000, Australia
- Department of Surgery- Otolaryngology Head and Neck Surgery, The University of Adelaide and the Basil Hetzel Institute for Translational Health Research, Central Adelaide Local Health Network, Woodville South SA, 5011, Australia
| | - Runhao Li
- Discipline of Surgery, Adelaide Medical School, The University of Adelaide, Adelaide SA, 5000, Australia
- Department of Haematology and Oncology, Basil Hetzel Institute for Translational Health Research and The Queen Elizabeth Hospital, Central Adelaide Local Health Network, Woodville South SA, 5011, Australia
| | - Fangmeinuo Wu
- Discipline of Surgery, Adelaide Medical School, The University of Adelaide, Adelaide SA, 5000, Australia
- Department of Haematology and Oncology, Basil Hetzel Institute for Translational Health Research and The Queen Elizabeth Hospital, Central Adelaide Local Health Network, Woodville South SA, 5011, Australia
| | - George Bouras
- Department of Surgery- Otolaryngology Head and Neck Surgery, The University of Adelaide and the Basil Hetzel Institute for Translational Health Research, Central Adelaide Local Health Network, Woodville South SA, 5011, Australia
| | - Linh T H Mai
- Discipline of Surgery, Adelaide Medical School, The University of Adelaide, Adelaide SA, 5000, Australia
- Department of Surgery- Otolaryngology Head and Neck Surgery, The University of Adelaide and the Basil Hetzel Institute for Translational Health Research, Central Adelaide Local Health Network, Woodville South SA, 5011, Australia
| | - Eric Smith
- Discipline of Surgery, Adelaide Medical School, The University of Adelaide, Adelaide SA, 5000, Australia
- Department of Haematology and Oncology, Basil Hetzel Institute for Translational Health Research and The Queen Elizabeth Hospital, Central Adelaide Local Health Network, Woodville South SA, 5011, Australia
| | - Peter-John Wormald
- Discipline of Surgery, Adelaide Medical School, The University of Adelaide, Adelaide SA, 5000, Australia
- Department of Surgery- Otolaryngology Head and Neck Surgery, The University of Adelaide and the Basil Hetzel Institute for Translational Health Research, Central Adelaide Local Health Network, Woodville South SA, 5011, Australia
| | - Rowan Valentine
- Department of Surgery- Otolaryngology Head and Neck Surgery, The University of Adelaide and the Basil Hetzel Institute for Translational Health Research, Central Adelaide Local Health Network, Woodville South SA, 5011, Australia
| | - Alkis James Psaltis
- Discipline of Surgery, Adelaide Medical School, The University of Adelaide, Adelaide SA, 5000, Australia
- Department of Surgery- Otolaryngology Head and Neck Surgery, The University of Adelaide and the Basil Hetzel Institute for Translational Health Research, Central Adelaide Local Health Network, Woodville South SA, 5011, Australia
| | - Sarah Vreugde
- Discipline of Surgery, Adelaide Medical School, The University of Adelaide, Adelaide SA, 5000, Australia
- Department of Surgery- Otolaryngology Head and Neck Surgery, The University of Adelaide and the Basil Hetzel Institute for Translational Health Research, Central Adelaide Local Health Network, Woodville South SA, 5011, Australia
| | - Kevin Fenix
- Discipline of Surgery, Adelaide Medical School, The University of Adelaide, Adelaide SA, 5000, Australia
- Department of Surgery- Otolaryngology Head and Neck Surgery, The University of Adelaide and the Basil Hetzel Institute for Translational Health Research, Central Adelaide Local Health Network, Woodville South SA, 5011, Australia
| |
Collapse
|
12
|
Pan W, Tao T, Qiu Y, Zhu X, Zhou X. Natural killer cells at the forefront of cancer immunotherapy with immune potency, genetic engineering, and nanotechnology. Crit Rev Oncol Hematol 2024; 193:104231. [PMID: 38070841 DOI: 10.1016/j.critrevonc.2023.104231] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 11/22/2023] [Accepted: 12/05/2023] [Indexed: 01/07/2024] Open
Abstract
Natural killer (NK) cells are vital components of the human immune system, acting as innate lymphocytes and playing a crucial role in immune surveillance. Their unique ability to independently eliminate target cells without antigen contact or antibodies has sparked interest in immunological research. This review examines recent NK cell developments and applications, encompassing immune functions, interactions with target cells, genetic engineering techniques, pharmaceutical interventions, and implications in cancers. Insights into NK cell regulation emerge, with a focus on promising genetic engineering like CAR-engineered NK cells, enhancing specificity against tumors. Immune checkpoint inhibitors also enhance NK cells' potential in cancer therapy. Nanotechnology's emergence as a tool for targeted drug delivery to improve NK cell therapies is explored. In conclusion, NK cells are pivotal in immunity, holding exciting potential in cancer immunotherapy. Ongoing research promises novel therapeutic strategies, advancing immunotherapy and medical interventions.
Collapse
Affiliation(s)
- Weiyi Pan
- Department of Immunology, School of Medicine, Nantong University, Nantong, China; School of Public Health, Southern Medical University, Guangzhou, China
| | - Tao Tao
- Department of Gastroenterology, Zibo Central Hospital, Zibo, China
| | - Yishu Qiu
- Department of Biology, College of Arts and Science, New York University, New York, USA
| | - Xiao Zhu
- Computational Systems Biology Lab (CSBL), The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China.
| | - Xiaorong Zhou
- Department of Immunology, School of Medicine, Nantong University, Nantong, China.
| |
Collapse
|
13
|
Zeng Z, Huang Z, Yue W, Nawaz S, Chen X, Liu J. Lactobacillus plantarum modulate gut microbiota and intestinal immunity in cyclophosphamide-treated mice model. Biomed Pharmacother 2023; 169:115812. [PMID: 37979376 DOI: 10.1016/j.biopha.2023.115812] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 10/22/2023] [Accepted: 10/26/2023] [Indexed: 11/20/2023] Open
Abstract
Gut microbiota (GM) contributes to the production of immune-regulatory molecules and cytokines. However, our understanding regarding intricate relationship between Lactobacillus plantarum and GM on regulation of immune function remained limited. To investigate the effect of Lactobacillus plantarum on an immunosuppressed mouse model, we employed cyclophosphamide treatment and conducted various analysis including H&E (hematoxylin-eosin staining), immunohistochemistry, 16S rRNA gene sequencing, and RT-PCR. Our results demonstrated that the administration of Lactobacillus plantarum had significant immunoenhancing effects in the immune-suppressed mice, as evidenced by the restoration of functional expression of specific immune markers in the spleen and an increase in the number of goblet cells in intestine (P < 0.05). Microbial taxonomic analysis revealed alterations in the gut microbiota composition, characterized by a decrease in the richness of Firmicutes and an increase in the proportion of Verrucomicrobia and Actinobacteria following cyclophosphamide treatment. Furthermore, cyclophosphamide treatment significantly suppressed the mRNA expression of inflammatory cytokines (P < 0.05), which were subsequently restored after administration of Lactobacillus plantarum. These observations provide valuable insights into the complex interplay between probiotics, gut microbiota, and immune system functioning.
Collapse
Affiliation(s)
- Zhibo Zeng
- Institute of Animal Husbandry and Veterinary Medicine, Fujian Academy of Agricultural Sciences/Fujian Key Laboratory of Animal Genetics and Breeding, Fuzhou 350013, China; Institute of Agricultural Sciences, ETH Zurich, Universitaetstrasse 2, 8092 Zurich, Switzerland
| | - Zonghao Huang
- Institute of Animal Husbandry and Veterinary Medicine, Fujian Academy of Agricultural Sciences/Fujian Key Laboratory of Animal Genetics and Breeding, Fuzhou 350013, China
| | - Wen Yue
- Institute of Animal Husbandry and Veterinary Medicine, Fujian Academy of Agricultural Sciences/Fujian Key Laboratory of Animal Genetics and Breeding, Fuzhou 350013, China
| | - Shah Nawaz
- Institute of Agricultural Sciences, ETH Zurich, Universitaetstrasse 2, 8092 Zurich, Switzerland
| | - Xinzhu Chen
- Institute of Animal Husbandry and Veterinary Medicine, Fujian Academy of Agricultural Sciences/Fujian Key Laboratory of Animal Genetics and Breeding, Fuzhou 350013, China.
| | - Jing Liu
- Institute of Animal Husbandry and Veterinary Medicine, Fujian Academy of Agricultural Sciences/Fujian Key Laboratory of Animal Genetics and Breeding, Fuzhou 350013, China.
| |
Collapse
|
14
|
Anderson G. Melatonin, BAG-1 and cortisol circadian interactions in tumor pathogenesis and patterned immune responses. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2023; 4:962-993. [PMID: 37970210 PMCID: PMC10645470 DOI: 10.37349/etat.2023.00176] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Accepted: 08/07/2023] [Indexed: 11/17/2023] Open
Abstract
A dysregulated circadian rhythm is significantly associated with cancer risk, as is aging. Both aging and circadian dysregulation show suppressed pineal melatonin, which is indicated in many studies to be linked to cancer risk and progression. Another independently investigated aspect of the circadian rhythm is the cortisol awakening response (CAR), which is linked to stress-associated hypothalamus-pituitary-adrenal (HPA) axis activation. CAR and HPA axis activity are primarily mediated via activation of the glucocorticoid receptor (GR), which drives patterned gene expression via binding to the promotors of glucocorticoid response element (GRE)-expressing genes. Recent data shows that the GR can be prevented from nuclear translocation by the B cell lymphoma-2 (Bcl-2)-associated athanogene 1 (BAG-1), which translocates the GR to mitochondria, where it can have diverse effects. Melatonin also suppresses GR nuclear translocation by maintaining the GR in a complex with heat shock protein 90 (Hsp90). Melatonin, directly and/or epigenetically, can upregulate BAG-1, suggesting that the dramatic 10-fold decrease in pineal melatonin from adolescence to the ninth decade of life will attenuate the capacity of night-time melatonin to modulate the effects of the early morning CAR. The interactions of pineal melatonin/BAG-1/Hsp90 with the CAR are proposed to underpin how aging and circadian dysregulation are associated with cancer risk. This may be mediated via differential effects of melatonin/BAG-1/Hsp90/GR in different cells of microenvironments across the body, from which tumors emerge. This provides a model of cancer pathogenesis that better integrates previously disparate bodies of data, including how immune cells are regulated by cancer cells in the tumor microenvironment, at least partly via the cancer cell regulation of the tryptophan-melatonin pathway. This has a number of future research and treatment implications.
Collapse
|
15
|
Mandal RK, Schmidt NW. Mechanistic insights into the interaction between the host gut microbiome and malaria. PLoS Pathog 2023; 19:e1011665. [PMID: 37824458 PMCID: PMC10569623 DOI: 10.1371/journal.ppat.1011665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2023] Open
Abstract
Malaria is a devastating infectious disease and significant global health burden caused by the bite of a Plasmodium-infected female Anopheles mosquito. Gut microbiota was recently discovered as a risk factor of severe malaria. This review entails the recent advances on the impact of gut microbiota composition on malaria severity and consequence of malaria infection on gut microbiota in mammalian hosts. Additionally, this review provides mechanistic insight into interactions that might occur between gut microbiota and host immunity which in turn can modulate malaria severity. Finally, approaches to modulate gut microbiota composition are discussed. We anticipate this review will facilitate novel hypotheses to move the malaria-gut microbiome field forward.
Collapse
Affiliation(s)
- Rabindra K. Mandal
- Ryan White Center for Pediatric Infectious Diseases and Global Health, Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indiana, United States of America
| | - Nathan W. Schmidt
- Ryan White Center for Pediatric Infectious Diseases and Global Health, Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indiana, United States of America
| |
Collapse
|
16
|
Abstract
Cardiometabolic disease comprises cardiovascular and metabolic dysfunction and underlies the leading causes of morbidity and mortality, both within the United States and worldwide. Commensal microbiota are implicated in the development of cardiometabolic disease. Evidence suggests that the microbiome is relatively variable during infancy and early childhood, becoming more fixed in later childhood and adulthood. Effects of microbiota, both during early development, and in later life, may induce changes in host metabolism that modulate risk mechanisms and predispose toward the development of cardiometabolic disease. In this review, we summarize the factors that influence gut microbiome composition and function during early life and explore how changes in microbiota and microbial metabolism influence host metabolism and cardiometabolic risk throughout life. We highlight limitations in current methodology and approaches and outline state-of-the-art advances, which are improving research and building toward refined diagnosis and treatment options in microbiome-targeted therapies.
Collapse
Affiliation(s)
- Curtis L Gabriel
- Division of Gastroenterology, Hepatology and Nutrition (C.L.G.), Vanderbilt University Medical Center, Nashville
- Tennessee Center for AIDS Research (C.L.G.), Vanderbilt University Medical Center, Nashville
| | - Jane F Ferguson
- Division of Cardiovascular Medicine (J.F.F.), Vanderbilt University Medical Center, Nashville
- Vanderbilt Microbiome Innovation Center (J.F.F.), Vanderbilt University Medical Center, Nashville
- Vanderbilt Institute for Infection, Immunology, and Inflammation (J.F.F.), Vanderbilt University Medical Center, Nashville
| |
Collapse
|
17
|
Liu XF, Shao JH, Liao YT, Wang LN, Jia Y, Dong PJ, Liu ZZ, He DD, Li C, Zhang X. Regulation of short-chain fatty acids in the immune system. Front Immunol 2023; 14:1186892. [PMID: 37215145 PMCID: PMC10196242 DOI: 10.3389/fimmu.2023.1186892] [Citation(s) in RCA: 60] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 04/24/2023] [Indexed: 05/24/2023] Open
Abstract
A growing body of research suggests that short-chain fatty acids (SCFAs), metabolites produced by intestinal symbiotic bacteria that ferment dietary fibers (DFs), play a crucial role in the health status of symbiotes. SCFAs act on a variety of cell types to regulate important biological processes, including host metabolism, intestinal function, and immune function. SCFAs also affect the function and fate of immune cells. This finding provides a new concept in immune metabolism and a better understanding of the regulatory role of SCFAs in the immune system, which impacts the prevention and treatment of disease. The mechanism by which SCFAs induce or regulate the immune response is becoming increasingly clear. This review summarizes the different mechanisms through which SCFAs act in cells. According to the latest research, the regulatory role of SCFAs in the innate immune system, including in NLRP3 inflammasomes, receptors of TLR family members, neutrophils, macrophages, natural killer cells, eosinophils, basophils and innate lymphocyte subsets, is emphasized. The regulatory role of SCFAs in the adaptive immune system, including in T-cell subsets, B cells, and plasma cells, is also highlighted. In addition, we discuss the role that SCFAs play in regulating allergic airway inflammation, colitis, and osteoporosis by influencing the immune system. These findings provide evidence for determining treatment options based on metabolic regulation.
Collapse
Affiliation(s)
- Xiao-feng Liu
- Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Wuxi, China
| | - Jia-hao Shao
- Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Wuxi, China
| | - Yi-Tao Liao
- Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Wuxi, China
| | - Li-Ning Wang
- School of Chinese Medicine, School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Yuan Jia
- Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Wuxi, China
| | - Peng-jun Dong
- Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Wuxi, China
| | - Zhi-zhong Liu
- Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Wuxi, China
| | - Dan-dan He
- Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Wuxi, China
| | - Chao Li
- Department of Spine, Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Wuxi, China
| | - Xian Zhang
- Department of Spine, Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Wuxi, China
| |
Collapse
|
18
|
Lin CP, Huang PH, Chen CY, Tzeng IS, Wu MY, Chen JS, Chen JW, Lin SJ. Tributyrin Intake Attenuates Angiotensin II-Induced Abdominal Aortic Aneurysm in LDLR-/- Mice. Int J Mol Sci 2023; 24:ijms24098008. [PMID: 37175712 PMCID: PMC10178859 DOI: 10.3390/ijms24098008] [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: 12/31/2022] [Revised: 03/31/2023] [Accepted: 04/19/2023] [Indexed: 05/15/2023] Open
Abstract
Abdominal aortic aneurysm (AAA) is a multifactorial cardiovascular disease with a high risk of death, and it occurs in the infrarenal aorta with vascular dilatation. High blood pressure acts on the aortic wall, resulting in rupture and causing life-threatening intra-abdominal hemorrhage. Vascular smooth muscle cell (VSMC) dysregulation and extracellular matrix (ECM) degradation, especially elastin breaks, contribute to structural changes in the aortic wall. The pathogenesis of AAA includes the occurrence of oxidative stress, inflammatory cell infiltration, elastic fiber fragmentation, VSMC apoptosis, and phenotypic transformation. Tributyrin (TB) is decomposed by intestinal lipase and has a function similar to that of butyrate. Whether TB has a protective effect against AAA remains uncertain. In the present study, we established an AAA murine model by angiotensin II (AngII) induction in low-density lipoprotein receptor knockout (LDLR-/-) mice and investigated the effects of orally administered TB on the AAA size, ratio of macrophage infiltration, levels of matrix metalloproteinase (MMP) expression, and epigenetic regulation. TB attenuates AngII-induced AAA size and decreases elastin fragmentation, macrophage infiltration, and MMP expression in the medial layer of the aorta and reduces the levels of SBP (systolic blood pressure, p < 0.001) and MMP-2 (p < 0.02) in the serum. TB reduces the AngII-stimulated expression levels of MMP2 (p < 0.05), MMP9 (p < 0.05), MMP12, and MMP14 in human aortic smooth muscle cells (HASMCs). Moreover, TB and valproic acid (VPA), a histone deacetylase (HDAC) inhibitor, suppress AngII receptor type 1 (AT1R, p < 0.05) activation and increase the expression of acetyl histone H3 by HDAC activity inhibition (p < 0.05). Our findings suggest that TB exerts its protective effect by suppressing the activation of HDAC to attenuate the AngII-induced AT1R signaling cascade.
Collapse
Affiliation(s)
- Chih-Pei Lin
- Department of Laboratory Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 23142, Taiwan
- Division of Clinical Pathology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 23142, Taiwan
- Department of Laboratory Medicine and Biotechnology, College of Medicine, Tzu Chi University, Hualien 97004, Taiwan
| | - Po-Hsun Huang
- Department of Critical Medicine, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- Cardiovascular Research Center, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Chi-Yu Chen
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| | - I-Shiang Tzeng
- Department of Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 23142, Taiwan
| | - Meng-Yu Wu
- Department of Emergency Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 23142, Taiwan
- Department of Emergency Medicine, School of Medicine, Tzu Chi University, Hualien 97004, Taiwan
| | - Jia-Shiong Chen
- Cardiovascular Research Center, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Jaw-Wen Chen
- Division of Cardiology & Healthcare and Management Center, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- Institute of Pharmacology, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Shing-Jong Lin
- Division of Cardiology & Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- Taipei Heart Institute, Taipei Medical University, Taipei 110301, Taiwan
- Division of Cardiology, Heart Center, Cheng-Hsin General Hospital, Taipei 11220, Taiwan
| |
Collapse
|