1
|
Pirolli NH, Raufman JP, Jay SM. Therapeutic Potential and Translational Challenges for Bacterial Extracellular Vesicles in Inflammatory Bowel Disease. Inflamm Bowel Dis 2025:izaf107. [PMID: 40357729 DOI: 10.1093/ibd/izaf107] [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: 02/19/2025] [Indexed: 05/15/2025]
Abstract
Despite the availability of numerous new immune-directed therapeutics, the major constituents of inflammatory bowel disease (IBD)-ulcerative colitis (UC) and Crohn's disease (CD)-continue to afflict millions worldwide, resulting in significant morbidity and long-term health risks. IBD results from a triad of immune, environmental (eg, gut microbiome), and genetic (including epigenetic) mechanisms, and therefore has been subject to a wide variety of therapeutic strategies. Among these, the administration of probiotics, particularly Gram-positive lactic acid bacteria (LAB), targeting both immune and environmental factors, has shown promising potential for efficacy in selected populations in early clinical trials. However, knowledge gaps and inconsistent efficacy currently prevent recommendations for the use of probiotics in larger IBD patient populations. The inconsistent efficacy of probiotics is likely due to variable cell viability and potency after administration, further exacerbated by IBD patient heterogeneity. Thus, an alternative to live probiotics for IBD has emerged in the form of bacterial extracellular vesicles (BEVs)-cell-secreted nanovesicles containing abundant bioactive cargo that, like live probiotics, can regulate immune and environmental factors but with fewer viability limitations and safety concerns. In this review, we summarize the work done to date establishing the potential of BEVs to provide the therapeutic benefits in IBD and discuss the hurdles BEVs must overcome to achieve clinical translation. We also consider future directions for BEV therapeutics, especially treatment potential for necrotizing enterocolitis (NEC), which shares similarities in pathophysiology with IBD.
Collapse
Affiliation(s)
- Nicholas H Pirolli
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
- Robert E. Fischell Institute for Biomedical Devices, University of Maryland, College Park, MD, USA
| | - Jean-Pierre Raufman
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, USA
- Biomedical Laboratory Research and Development Service, Veterans Affairs Maryland Healthcare System, Baltimore, MD, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland Medical Center, Baltimore, MD, USA
| | - Steven M Jay
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
- Robert E. Fischell Institute for Biomedical Devices, University of Maryland, College Park, MD, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland Medical Center, Baltimore, MD, USA
- Program in Molecular and Cell Biology, University of Maryland, College Park, MD 20742, USA
| |
Collapse
|
2
|
Lu Y, Chen L, Lin Y, Zhang Y, Wang Y, Yu W, Ren F, Guo H. Fatty acid metabolism: The crossroads in intestinal homeostasis and tumor. Metabolism 2025; 169:156273. [PMID: 40280478 DOI: 10.1016/j.metabol.2025.156273] [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: 03/07/2025] [Revised: 04/09/2025] [Accepted: 04/19/2025] [Indexed: 04/29/2025]
Abstract
Fatty acids (FAs) have various functions on cell regulation considering their abundant types and metabolic pathways. In addition, the relation between FA and other nutritional metabolism makes their functions more complex. As the first place for diet-derived FA metabolism, intestine is significantly influenced despite lack of clear conclusions due to the inconsistent findings. In this review, we discuss the regulation of fatty acid metabolism on the fate of intestinal stem cells in homeostasis and disorders, and also focus on the intestinal tumor development and treatment from the aspect of gut microbiota-epithelium-immune interaction. We summarize that the balances between FA oxidation and glycolysis, between oxidative phosphorylation and ketogenesis, between catabolism and anabolism, and the specific roles of individual FA types determine the diverse effects of intestinal FA metabolism in different cases. We hope this will inspire further dissection and suggest precise dietary/metabolic intervention for different demands related to intestinal health.
Collapse
Affiliation(s)
- Yao Lu
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Lining Chen
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Yingying Lin
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| | - Yafei Zhang
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Yuqi Wang
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Weiru Yu
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| | - Fazheng Ren
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China.
| | - Huiyuan Guo
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
3
|
Matsushita R, Sato K, Uchida K, Imi Y, Amano R, Kasahara N, Kitao Y, Oishi Y, Kawaai H, Tomimoto C, Hosokawa Y, Kishino S, Ogawa J, Hosooka T. A Gut Microbial Metabolite HYA Ameliorates Adipocyte Hypertrophy by Activating AMP-Activated Protein Kinase. Nutrients 2025; 17:1393. [PMID: 40284256 PMCID: PMC12030234 DOI: 10.3390/nu17081393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2025] [Revised: 04/15/2025] [Accepted: 04/19/2025] [Indexed: 04/29/2025] Open
Abstract
Background/Objectives: Metabolites produced by gut microbiota play an important role in the crosstalk between the gut and other organs. Although HYA (10-hydroxy-cis-12-octadecenoic acid), a linoleic acid metabolite produced by lactic acid bacteria represented by Lactobacillus, has been shown to exert physiological effects such as metabolic improvement and anti-inflammation in the host, its direct action on adipose tissue and the mechanism remains unknown. Methods: The effect of HYA administration on adipocyte size in mice fed a high-fat diet was examined. In 3T3-L1 mature adipocytes treated with HYA, the amount of intracellular lipid droplets was evaluated by Oil red O staining, gene expression by real-time qPCR, phosphorylation of AMP-activated protein kinase (AMPK) by immunoblotting, and intracellular Ca2+ concentration with calcium imaging. Results: Administration of HYA, but not linoleic acid, to obese mice fed a high-fat diet significantly reduced adipocyte size. To investigate whether the inhibition of adipocyte hypertrophy by HYA has a direct effect on adipocytes, 3T3-L1 adipocytes were treated with HYA, which significantly decreased the amount of intracellular lipid droplets in these cells. Gene expression analysis by real-time PCR showed decreased expression of genes related to lipogenesis such as FAS and ACC1, and increased expression of CPT1A, which is involved in fatty acid oxidation. Mechanistically, HYA was found to activate AMPK in adipocytes by increasing intracellular Ca2+ concentration. Conclusions: HYA suppresses adipocyte hypertrophy by activating AMPK in adipocytes. HYA may be a potential therapeutic for obesity and related metabolic disorders.
Collapse
Affiliation(s)
- Rino Matsushita
- Laboratory of Nutritional Physiology, Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, Suruga-ku, Shizuoka 422-8526, Japan (Y.O.)
| | - Kaori Sato
- Laboratory of Nutritional Physiology, Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, Suruga-ku, Shizuoka 422-8526, Japan (Y.O.)
| | - Kunitoshi Uchida
- Laboratory of Functional Physiology, Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, Suruga-ku, Shizuoka 422-8526, Japan
| | - Yukiko Imi
- Laboratory of Nutritional Physiology, Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, Suruga-ku, Shizuoka 422-8526, Japan (Y.O.)
| | - Reina Amano
- Laboratory of Nutritional Physiology, Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, Suruga-ku, Shizuoka 422-8526, Japan (Y.O.)
| | - Nanaho Kasahara
- Laboratory of Nutritional Physiology, Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, Suruga-ku, Shizuoka 422-8526, Japan (Y.O.)
| | - Yuki Kitao
- Laboratory of Nutritional Physiology, Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, Suruga-ku, Shizuoka 422-8526, Japan (Y.O.)
| | - Yuki Oishi
- Laboratory of Nutritional Physiology, Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, Suruga-ku, Shizuoka 422-8526, Japan (Y.O.)
| | - Honoka Kawaai
- Laboratory of Nutritional Physiology, Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, Suruga-ku, Shizuoka 422-8526, Japan (Y.O.)
| | | | - Yusei Hosokawa
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan;
| | - Shigenobu Kishino
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Sakyo-ku, Kyoto 606-8502, Japan; (S.K.)
| | - Jun Ogawa
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Sakyo-ku, Kyoto 606-8502, Japan; (S.K.)
| | - Tetsuya Hosooka
- Laboratory of Nutritional Physiology, Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, Suruga-ku, Shizuoka 422-8526, Japan (Y.O.)
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan;
| |
Collapse
|
4
|
Fang X, Zhang Y, Huang X, Miao R, Zhang Y, Tian J. Gut microbiome research: Revealing the pathological mechanisms and treatment strategies of type 2 diabetes. Diabetes Obes Metab 2025. [PMID: 40230225 DOI: 10.1111/dom.16387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 03/19/2025] [Accepted: 03/23/2025] [Indexed: 04/16/2025]
Abstract
The high prevalence and disability rate of type 2 diabetes (T2D) caused a huge social burden to the world. Currently, new mechanisms and therapeutic approaches that may affect this disease are being sought. With in-depth research on the pathogenesis of T2D and growing advances in microbiome sequencing technology, the association between T2D and gut microbiota has been confirmed. The gut microbiota participates in the regulation of inflammation, intestinal permeability, short-chain fatty acid metabolism, branched-chain amino acid metabolism and bile acid metabolism, thereby affecting host glucose and lipid metabolism. Interventions focusing on the gut microbiota are gaining traction as a promising approach to T2D management. For example, dietary intervention, prebiotics and probiotics, faecal microbiota transplant and phage therapy. Meticulous experimental design and choice of analytical methods are crucial for obtaining accurate and meaningful results from microbiome studies. How to design gut microbiome research in T2D and choose different machine learning methods for data analysis are extremely critical to achieve personalized precision medicine.
Collapse
Affiliation(s)
- Xinyi Fang
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate College, Beijing University of Chinese Medicine, Beijing, China
| | - Yanjiao Zhang
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xinyue Huang
- First Clinical Medical College, Changzhi Medical College, Shanxi, China
| | - Runyu Miao
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate College, Beijing University of Chinese Medicine, Beijing, China
| | - Yuxin Zhang
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jiaxing Tian
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
5
|
Yin F, Chen Y, Zhang H, Zhao H, Li X, Wang Z, Meng W, Zhao J, Tang L, Li Y, Li J, Wang X. Lactobacillus paracasei Expressing Porcine Trefoil Factor 3 and Epidermal Growth Factor: A Novel Approach for Superior Mucosal Repair. Vet Sci 2025; 12:365. [PMID: 40284867 PMCID: PMC12031595 DOI: 10.3390/vetsci12040365] [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: 02/23/2025] [Revised: 03/23/2025] [Accepted: 04/10/2025] [Indexed: 04/29/2025] Open
Abstract
Trefoil factor 3 (TFF3) and epidermal growth factor (EGF) exert a promotive effect on the functions of intestinal epithelial cells and offer protection to the intestinal mucosa. Lactobacillus paracasei can ameliorate intestinal mucosal damage. In this study, pPG-pTFF3/27-2, pPG-pEGF/27-2, and pPG-pTE/27-2 were constructed to express porcine TFF3, EGF, and a fusion protein (pTE). Functional assays showed they promoted Immortalized Porcine Enterocyte Cell line J2 (IPEC-J2) proliferation and migration, with pTE having a greater migratory effect. In dextran sulfate sodium (DSS)-induced colitis mice, oral administration of pPG-pTE/27-2 reduced colitis, improved mucosal integrity, increased the expression of tight-junction proteins and the serum level of Interleukin-10 (IL-10), and decreased the levels of pro-inflammatory Tumor Necrosis Factor-α (TNF-α), Interleukin-6 (IL-6), and Interleukin-1β (IL-1β). These results imply that recombinant L. paracasei 27-2 strains engineered to express pTFF3 and pEGF represent a promising approach for augmenting intestinal epithelial cell function and facilitating mucosal restitution, and they possess significant potential in the treatment of intestinal mucosal injury and inflammatory bowel disease (IBD).
Collapse
Affiliation(s)
- Fangjie Yin
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (F.Y.); (Y.C.); (H.Z.); (H.Z.); (X.L.); (L.T.); (Y.L.)
| | - Ying Chen
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (F.Y.); (Y.C.); (H.Z.); (H.Z.); (X.L.); (L.T.); (Y.L.)
| | - Huijun Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (F.Y.); (Y.C.); (H.Z.); (H.Z.); (X.L.); (L.T.); (Y.L.)
| | - Hongzhe Zhao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (F.Y.); (Y.C.); (H.Z.); (H.Z.); (X.L.); (L.T.); (Y.L.)
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin 150030, China
| | - Xuenan Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (F.Y.); (Y.C.); (H.Z.); (H.Z.); (X.L.); (L.T.); (Y.L.)
| | - Zi Wang
- Tongliao Institute of Animal Husbandry and Veterinary Science, Tongliao 028000, China;
| | - Weijing Meng
- Tongliao Agricultural and Animal Husbandry Development Center, Tongliao 028000, China;
| | - Jie Zhao
- Nanjing Dr. Vet Health Management Co., Ltd., Nanjing 210000, China;
| | - Lijie Tang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (F.Y.); (Y.C.); (H.Z.); (H.Z.); (X.L.); (L.T.); (Y.L.)
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin 150030, China
| | - Yijing Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (F.Y.); (Y.C.); (H.Z.); (H.Z.); (X.L.); (L.T.); (Y.L.)
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin 150030, China
| | - Jiaxuan Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (F.Y.); (Y.C.); (H.Z.); (H.Z.); (X.L.); (L.T.); (Y.L.)
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin 150030, China
| | - Xiaona Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (F.Y.); (Y.C.); (H.Z.); (H.Z.); (X.L.); (L.T.); (Y.L.)
- Tongliao Agricultural and Animal Husbandry Development Center, Tongliao 028000, China;
| |
Collapse
|
6
|
Pérez Escriva P, Correia Tavares Bernardino C, Letellier E. De-coding the complex role of microbial metabolites in cancer. Cell Rep 2025; 44:115358. [PMID: 40023841 DOI: 10.1016/j.celrep.2025.115358] [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: 08/09/2024] [Revised: 12/11/2024] [Accepted: 02/06/2025] [Indexed: 03/04/2025] Open
Abstract
The human microbiome, an intricate ecosystem of trillions of microbes residing across various body sites, significantly influences cancer, a leading cause of morbidity and mortality worldwide. Recent studies have illuminated the microbiome's pivotal role in cancer development, either through direct cellular interactions or by secreting bioactive compounds such as metabolites. Microbial metabolites contribute to cancer initiation through mechanisms such as DNA damage, epithelial barrier dysfunction, and chronic inflammation. Furthermore, microbial metabolites exert dual roles on cancer progression and response to therapy by modulating cellular metabolism, gene expression, and signaling pathways. Understanding these complex interactions is vital for devising new therapeutic strategies. This review highlights microbial metabolites as promising targets for cancer prevention and treatment, emphasizing their impact on therapy responses and underscoring the need for further research into their roles in metastasis and therapy resistance.
Collapse
Affiliation(s)
- Pau Pérez Escriva
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Catarina Correia Tavares Bernardino
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Elisabeth Letellier
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg.
| |
Collapse
|
7
|
Matsuo M, Takaoka S, Nakayama K, Nagira A, Goto H, Nakajima A. Fatty acids from cheese stimulate cholesterol efflux by ATP-binding cassette transporters. Biosci Biotechnol Biochem 2025; 89:573-585. [PMID: 39725457 DOI: 10.1093/bbb/zbae207] [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: 10/11/2024] [Accepted: 12/21/2024] [Indexed: 12/28/2024]
Abstract
It is essential to remove cholesterol from the body to suppress atherosclerosis progression. ABCA1 and ABCG1 transport cholesterol in peripheral cells, including macrophages, and function in the formation of high-density lipoprotein. ABCG5/ABCG8 functions in the efflux of cholesterol from the body. In this study, we investigated the effects of Camembert cheese extracts and ingredients on cholesterol transport via ABC transporters. Camembert cheese extracts were added to baby hamster kidney (BHK) cells expressing ABCA1, ABCG1, or ABCG5/ABCG8, and THP-1 cells expressing ABCA1 and ABCG1. Organic solvent extracts of Camembert cheese increased cholesterol efflux in THP-1 and BHK cells expressing ABCA1 or ABCG5/ABCG8. After fractionation of the extracts, palmitoleic acid was found to increase cholesterol efflux by ABCA1 and ABCG5/ABCG8, whereas 10-hydroxypalmitic acid increased it by ABCA1 and ABCG1. It is suggested that palmitoleic acid and 10-hydroxypalmitic acid in Camembert cheese may prevent the accumulation of excess cholesterol in cells by stimulating ABC transporters.
Collapse
MESH Headings
- Cholesterol/metabolism
- Animals
- Humans
- Cheese/analysis
- Cricetinae
- ATP Binding Cassette Transporter 1/metabolism
- ATP Binding Cassette Transporter 1/genetics
- Biological Transport/drug effects
- Fatty Acids, Monounsaturated/pharmacology
- ATP Binding Cassette Transporter, Subfamily G, Member 5/metabolism
- ATP Binding Cassette Transporter, Subfamily G, Member 5/genetics
- ATP Binding Cassette Transporter, Subfamily G, Member 8/metabolism
- ATP Binding Cassette Transporter, Subfamily G, Member 8/genetics
- ATP-Binding Cassette Transporters/metabolism
- Fatty Acids/pharmacology
- Cell Line
- ATP Binding Cassette Transporter, Subfamily G, Member 1/metabolism
- THP-1 Cells
Collapse
Affiliation(s)
- Michinori Matsuo
- Department of Food and Nutrition, Faculty of Home Economics, Kyoto Women's University, Kyoto, Japan
| | - Shiho Takaoka
- Department of Food and Nutrition, Faculty of Home Economics, Kyoto Women's University, Kyoto, Japan
| | - Kai Nakayama
- R&D Division, Meiji Co., Ltd., Hachioji, Tokyo, Japan
| | - Akika Nagira
- R&D Division, Meiji Co., Ltd., Hachioji, Tokyo, Japan
| | - Hirofumi Goto
- R&D Division, Meiji Co., Ltd., Hachioji, Tokyo, Japan
| | | |
Collapse
|
8
|
Park Y, Woo JM, Shin J, Chung M, Seo EJ, Lee SJ, Park JB. Unveiling the biological activities of the microbial long chain hydroxy fatty acids as dual agonists of GPR40 and GPR120. Food Chem 2025; 465:142010. [PMID: 39556901 DOI: 10.1016/j.foodchem.2024.142010] [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: 02/04/2024] [Revised: 09/10/2024] [Accepted: 11/09/2024] [Indexed: 11/20/2024]
Abstract
The physiological functions of various fatty acid-originating metabolites from foods and fermented products remained mostly untouched. Thereby, this study examined the biological activities of hydroxy fatty acids as agonists of G protein-coupled receptors (i.e., GPR40 and GPR120), which are derived from long-chain fatty acids (e.g., oleic acid and linoleic acid) by microbiota. Cell-based Ca2+ mobilization assays and in silico docking simulations revealed that not only the degree of unsaturation but also the number and position of hydroxyl groups played a key role in their agonist activities. For instance, 8,11-dihydroxyoctadec-9Z-enoic acid exhibited significantly greater Ca2+ response in the GPR40/GPR120-expressing cells as compared to the endogenous agonists (e.g., linoleic acid and docosahexaenoic acid), forming hydrogen bond interactions with residues in the ligand-binding pockets of receptors. This study will contribute to understanding the relationships between fatty acid structures and agonist activities.
Collapse
Affiliation(s)
- Yeeun Park
- Department of Food Science and Biotechnology, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Ji-Min Woo
- Department of Food Science and Biotechnology, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Jaeeun Shin
- Department of Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Myunghae Chung
- Department of Food Science and Biotechnology, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Eun-Ji Seo
- Department of Food Science and Biotechnology, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Sung-Joon Lee
- Department of Biotechnology, Korea University, Seoul 02841, Republic of Korea.
| | - Jin-Byung Park
- Department of Food Science and Biotechnology, Ewha Womans University, Seoul 03760, Republic of Korea.
| |
Collapse
|
9
|
Lialios P, Alimperti S. Role of E-cadherin in epithelial barrier dysfunction: implications for bacterial infection, inflammation, and disease pathogenesis. Front Cell Infect Microbiol 2025; 15:1506636. [PMID: 40007608 PMCID: PMC11850337 DOI: 10.3389/fcimb.2025.1506636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 01/15/2025] [Indexed: 02/27/2025] Open
Abstract
Epithelial barriers serve as critical defense lines against microbial infiltration and maintain tissue homeostasis. E-cadherin, an essential component of adherens junctions, has emerged as a pivotal molecule that secures epithelial homeostasis. Lately, its pleiotropic role beyond barrier function, including its involvement in immune responses, has become more evident. Herein, we delve into the intricate relationship between (dys)regulation of epithelial homeostasis and the versatile functionality of E-cadherin, describing complex mechanisms that underlie barrier integrity and disruption in disease pathogenesis such as bacterial infection and inflammation, among others. Clinical implications of E-cadherin perturbations in host pathophysiology are emphasized; downregulation, proteolytic phenomena, abnormal localization/signaling and aberrant immune reactions are linked with a broad spectrum of pathology beyond infectious diseases. Finally, potential therapeutic interventions that may harness E-cadherin to mitigate barrier-associated tissue damage are explored. Overall, this review highlights the crucial role of E-cadherin in systemic health, offering insights that could pave the way for strategies to reinforce/restore barrier integrity and treat related diseases.
Collapse
Affiliation(s)
- Peter Lialios
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC, United States
- Center for Biological and Biomedical Engineering, Georgetown University, Washington, DC, United States
| | - Stella Alimperti
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC, United States
- Center for Biological and Biomedical Engineering, Georgetown University, Washington, DC, United States
| |
Collapse
|
10
|
Yamamoto Y, Narumi K, Yamagishi N, Yonejima Y, Iseki K, Kobayashi M, Kanai Y. HYA ameliorated postprandial hyperglycemia in type 1 diabetes model rats with bolus insulin treatment. Acta Diabetol 2025:10.1007/s00592-025-02459-6. [PMID: 39899133 DOI: 10.1007/s00592-025-02459-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 01/24/2025] [Indexed: 02/04/2025]
Abstract
AIMS The oral administration of linoleic acid immediately before glucose tolerance test (OGTT) ameliorated postprandial hyperglycemia via GPR120 pathway in normal and type 1 diabetes (T1DM) rats. Linoleic acid could promote inflammatory mediators, but 10-hydroxy-cis-12-octadecenoic acid (HYA) converted from linoleic acid by Lactobacillus plantarum has higher GPR120 agonistic activity without promoting inflammatory mediators. This study examined whether the oral-administration of HYA immediately before OGTT also ameliorated the postprandial hyperglycemia in normal rats and T1DM rats injected with bolus insulin. METHODS Normal and T1DM male Sprague-Dawley rats received HYA immediately before OGTT. Other T1DM rats were given HYA and Humulin R immediately before OGTT. We measured the concentration of glucose, insulin, glucagon-like peptide 1 (GLP-1) and cholecystokinin in blood before and after OGTT. We also measured the amount of glucose in the gastric tract after OGTT, and the amount of uptake of methyl-α-D-glucopyranoside in CACO-2 cells. RESULTS Postprandial hyperglycemia was ameliorated by HYA in normal rats, and the postprandial blood glucose levels were slowly elevated by HYA in the T1DM model rats. HYA partially inhibited the uptake of methyl-α-D-glucopyranoside in CACO-2 cells. HYA slowed gastric motility and increased the plasma GLP-1 and cholecystokinin levels in normal rats. HYA also ameliorated the postprandial hyperglycemia in T1DM rats given bolus insulin. CONCLUSION Oral administration of HYA immediately before OGTT ameliorated postprandial hyperglycemia through inhibition of glucose absorption and slowing of gastric motility in normal rats. Furthermore, this beneficial effect of HYA was also revealed in T1DM rats injected with bolus insulin.
Collapse
Affiliation(s)
- Yuta Yamamoto
- Department of Anatomy and Cell Biology, Graduate School of Medical and Pharmaceutical Sciences, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8509, Japan.
| | - Katsuya Narumi
- Laboratory of Clinical Pharmaceutics and Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Naoko Yamagishi
- Department of Anatomy and Cell Biology, Graduate School of Medical and Pharmaceutical Sciences, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8509, Japan
| | | | - Ken Iseki
- Laboratory of Clinical Pharmaceutics and Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Masaki Kobayashi
- Laboratory of Clinical Pharmaceutics and Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Yoshimitsu Kanai
- Department of Anatomy and Cell Biology, Graduate School of Medical and Pharmaceutical Sciences, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8509, Japan
| |
Collapse
|
11
|
Chen A, Gong Y, Wu S, Du Y, Liu Z, Jiang Y, Li J, Miao YB. Navigating a challenging path: precision disease treatment with tailored oral nano-armor-probiotics. J Nanobiotechnology 2025; 23:72. [PMID: 39893419 PMCID: PMC11786591 DOI: 10.1186/s12951-025-03141-3] [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: 10/23/2024] [Accepted: 01/19/2025] [Indexed: 02/04/2025] Open
Abstract
Oral probiotics have significant potential for preventing and treating many diseases. Yet, their efficacy is often hindered by challenges related to survival and colonization within the gastrointestinal tract. Nanoparticles emerge as a transformative solution, offering robust protection and enhancing the stability and bioavailability of these probiotics. This review explores the innovative application of nanoparticle-armored engineered probiotics for precise disease treatment, specifically addressing the physiological barriers associated with oral administration. A comprehensive evaluation of various nano-armor probiotics and encapsulation methods is provided, carefully analyzing their respective merits and limitations, alongside strategies to enhance probiotic survival and achieve targeted delivery and colonization within the gastrointestinal tract. Furthermore, the review explores the potential clinical applications of nano-armored probiotics in precision therapeutics, critically addressing safety and regulatory considerations, and proposing the innovative concept of 'probiotic intestinal colonization with nano armor' for brain-targeted therapies. Ultimately, this review aspires to guide the advancement of nano-armored probiotic therapies, driving progress in precision medicine and paving the way for groundbreaking treatment modalities.
Collapse
Affiliation(s)
- Anmei Chen
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610041, China
- Department of Haematology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, No. 32, West Section 2, First Ring Road, Qingyang District, Chengdu, 610000, China
| | - Ying Gong
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610041, China
- Department of Haematology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, No. 32, West Section 2, First Ring Road, Qingyang District, Chengdu, 610000, China
| | - Shaoquan Wu
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610041, China
- Department of Haematology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, No. 32, West Section 2, First Ring Road, Qingyang District, Chengdu, 610000, China
| | - Ye Du
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610041, China
- Department of Haematology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, No. 32, West Section 2, First Ring Road, Qingyang District, Chengdu, 610000, China
| | - Zhijun Liu
- Urology Institute of Shenzhen University, The Third Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, 518000, China
| | - Yuhong Jiang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610041, China.
| | - Jiahong Li
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610041, China.
| | - Yang-Bao Miao
- Department of Haematology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, No. 32, West Section 2, First Ring Road, Qingyang District, Chengdu, 610000, China.
| |
Collapse
|
12
|
Wang YF, Chen CY, Lei L, Zhang Y. Regulation of the microglial polarization for alleviating neuroinflammation in the pathogenesis and therapeutics of major depressive disorder. Life Sci 2025; 362:123373. [PMID: 39756509 DOI: 10.1016/j.lfs.2025.123373] [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: 06/06/2024] [Revised: 12/18/2024] [Accepted: 01/02/2025] [Indexed: 01/07/2025]
Abstract
Major depressive disorder (MDD), as a multimodal neuropsychiatric and neurodegenerative illness with high prevalence and disability rates, has become a burden to world health and the economy that affects millions of individuals worldwide. Neuroinflammation, an atypical immune response occurring in the brain, is currently gaining more attention due to its association with MDD. Microglia, as immune sentinels, have a vital function in regulating neuroinflammatory reactions in the immune system of the central nervous system. From the perspective of steady-state branching states, they can transition phenotypes between two extremes, namely, M1 and M2 phenotypes are pro-inflammatory and anti-inflammatory, respectively. It has an intermediate transition state characterized by different transcriptional features and the release of inflammatory mediators. The timing regulation of inflammatory cytokine release is crucial for damage control and guiding microglia back to a steady state. The dysregulation can lead to exorbitant tissue injury and neuronal mortality, and targeting the cellular signaling pathway that serves as the regulatory basis for microglia is considered an essential pathway for treating MDD. However, the specific intervention targets and mechanisms of microglial activation pathways in neuroinflammation are still unclear. Therefore, the present review summarized and discussed various signaling pathways and effective intervention targets that trigger the activation of microglia from its branching state and emphasizes the mechanism of microglia-mediated neuroinflammation associated with MDD.
Collapse
Affiliation(s)
- Yu-Fei Wang
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Cong-Ya Chen
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Lan Lei
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yi Zhang
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China.
| |
Collapse
|
13
|
Revol-Cavalier J, Quaranta A, Newman JW, Brash AR, Hamberg M, Wheelock CE. The Octadecanoids: Synthesis and Bioactivity of 18-Carbon Oxygenated Fatty Acids in Mammals, Bacteria, and Fungi. Chem Rev 2025; 125:1-90. [PMID: 39680864 PMCID: PMC11719350 DOI: 10.1021/acs.chemrev.3c00520] [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] [Received: 07/21/2023] [Revised: 11/06/2024] [Accepted: 11/15/2024] [Indexed: 12/18/2024]
Abstract
The octadecanoids are a broad class of lipids consisting of the oxygenated products of 18-carbon fatty acids. Originally referring to production of the phytohormone jasmonic acid, the octadecanoid pathway has been expanded to include products of all 18-carbon fatty acids. Octadecanoids are formed biosynthetically in mammals via cyclooxygenase (COX), lipoxygenase (LOX), and cytochrome P450 (CYP) activity, as well as nonenzymatically by photo- and autoxidation mechanisms. While octadecanoids are well-known mediators in plants, their role in the regulation of mammalian biological processes has been generally neglected. However, there have been significant advancements in recognizing the importance of these compounds in mammals and their involvement in the mediation of inflammation, nociception, and cell proliferation, as well as in immuno- and tissue modulation, coagulation processes, hormone regulation, and skin barrier formation. More recently, the gut microbiome has been shown to be a significant source of octadecanoid biosynthesis, providing additional biosynthetic routes including hydratase activity (e.g., CLA-HY, FA-HY1, FA-HY2). In this review, we summarize the current field of octadecanoids, propose standardized nomenclature, provide details of octadecanoid preparation and measurement, summarize the phase-I metabolic pathway of octadecanoid formation in mammals, bacteria, and fungi, and describe their biological activity in relation to mammalian pathophysiology as well as their potential use as biomarkers of health and disease.
Collapse
Affiliation(s)
- Johanna Revol-Cavalier
- Unit
of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institutet, Stockholm SE-171 77, Sweden
- Larodan
Research Laboratory, Karolinska Institutet, Stockholm SE-171 77, Sweden
| | - Alessandro Quaranta
- Unit
of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institutet, Stockholm SE-171 77, Sweden
| | - John W. Newman
- Western
Human Nutrition Research Center, Agricultural
Research Service, USDA, Davis, California 95616, United States
- Department
of Nutrition, University of California, Davis, Davis, California 95616, United States
- West
Coast Metabolomics Center, Genome Center, University of California, Davis, Davis, California 95616, United States
| | - Alan R. Brash
- Department
of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Mats Hamberg
- Unit
of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institutet, Stockholm SE-171 77, Sweden
- Larodan
Research Laboratory, Karolinska Institutet, Stockholm SE-171 77, Sweden
| | - Craig E. Wheelock
- Unit
of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institutet, Stockholm SE-171 77, Sweden
- Department
of Respiratory Medicine and Allergy, Karolinska
University Hospital, Stockholm SE-141-86, Sweden
| |
Collapse
|
14
|
Wang JX, Chang SY, Jin ZY, Li D, Zhu J, Luo ZB, Han SZ, Kang JD, Quan LH. Lactobacillus reuteri-Enriched Eicosatrienoic Acid Regulates Glucose Homeostasis by Promoting GLP-1 Secretion to Protect Intestinal Barrier Integrity. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:393-408. [PMID: 39680859 DOI: 10.1021/acs.jafc.4c03818] [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: 12/18/2024]
Abstract
Lactobacillus reuteri is a well-known probiotic with beneficial effects, such as anti-insulin resistance, anti-inflammatory, and improvement of the intestinal barrier. However, the underlying mechanisms remain unclear. Here, we found that gavage of L. reuteri improved the intestinal barrier and glucose homeostasis in HFD-fed mice. Analysis of lipid metabolomics reveals a significant increase in eicosatrienoic acid (ETA) levels in mouse feces after L. reuteri gavage. We found that ETA maintain intestinal barrier integrity and improve glucose homeostasis by promoting GLP-1 secretion. Mechanistically, by using CD36 inhibitor in vivo and CD36 knockdown STC-1 cells in vitro, we elucidate that ETA activates intestinal CD36-activated PLC/IP3R/Ca2+ signaling to promote GLP-1 secretion. In vivo administration of GLP-1R inhibitor and in vitro intestinal organoid experiments demonstrate that GLP-1 upregulates the PI3K/AKT/HIF-1α pathway by GLP-1R and increases intestinal tight junction protein expressions, which in turn enhance the intestinal barrier integrity, reduce serum LPS level, attenuate inflammation in white adipose tissue (WAT), and ultimately improve glucose homeostasis in HFD and db/db mice. Our study elucidates for the first time the mechanism by which L. reuteri and its enriched metabolite ETA inhibit WAT inflammation by ameliorating the intestinal barrier, ultimately improving glucose homeostasis, and provides a new treatment strategy for T2D.
Collapse
Affiliation(s)
- Jun-Xia Wang
- Department of Animal Science, College of Agricultural, Yanbian University, Yanji 133002, China
| | - Shuang-Yan Chang
- Department of Animal Science, College of Agricultural, Yanbian University, Yanji 133002, China
- Jilin Provincial Key Laboratory of Transgenic Animal and Embryo Engineering, Yanbian University, Yanji 133002, China
| | - Zheng-Yun Jin
- Department of Animal Science, College of Agricultural, Yanbian University, Yanji 133002, China
| | - Dongxu Li
- Interdisciplinary Program of Biological Functional Molecules, College of Integration Science, Yanbian University, Yanji 133002, China
| | - Jun Zhu
- Interdisciplinary Program of Biological Functional Molecules, College of Integration Science, Yanbian University, Yanji 133002, China
| | - Zhao-Bo Luo
- Department of Animal Science, College of Agricultural, Yanbian University, Yanji 133002, China
- Jilin Provincial Key Laboratory of Transgenic Animal and Embryo Engineering, Yanbian University, Yanji 133002, China
| | - Sheng-Zhong Han
- Department of Animal Science, College of Agricultural, Yanbian University, Yanji 133002, China
- Jilin Provincial Key Laboratory of Transgenic Animal and Embryo Engineering, Yanbian University, Yanji 133002, China
| | - Jin-Dan Kang
- Department of Animal Science, College of Agricultural, Yanbian University, Yanji 133002, China
- Jilin Provincial Key Laboratory of Transgenic Animal and Embryo Engineering, Yanbian University, Yanji 133002, China
| | - Lin-Hu Quan
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji 133002, China
| |
Collapse
|
15
|
Lathram WA, Neff RJ, Zalla AN, Brien JD, Subramanian V, Radka CD. Dissecting the biophysical mechanisms of oleate hydratase association with membranes. Front Mol Biosci 2025; 11:1504373. [PMID: 39845901 PMCID: PMC11751051 DOI: 10.3389/fmolb.2024.1504373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 12/16/2024] [Indexed: 01/24/2025] Open
Abstract
This study investigates the dynamics of oleate hydratase (OhyA), a bacterial flavoenzyme from Staphylococcus aureus, and its interactions with lipid membranes, focusing on the factors influencing membrane binding and oligomerization. OhyA catalyzes the hydration of unsaturated fatty acids, playing a key role in bacterial pathogenesis by neutralizing host antimicrobial fatty acids. OhyA binds the membrane bilayer to access membrane-embedded substrates for catalysis, and structural studies have revealed that OhyA forms oligomers on membrane surfaces, stabilized by both protein-protein and protein-lipid interactions. Using fluorescence correlation spectroscopy (FCS), we examined the effects of membrane curvature and lipid availability on OhyA binding to phosphatidylglycerol unilamellar vesicles. Our results reveal that OhyA preferentially binds to vesicles with moderate curvature, while the presence of substrate fatty acids slightly enhanced the overall interaction despite reducing the binding affinity by 3- to 4-fold. Complementary phosphorus-31 (31P) NMR spectroscopy further demonstrated two distinct binding modes: a fast-exchange interaction at lower protein concentrations and a longer lasting interaction at higher protein concentrations, likely reflecting cooperative oligomerization. These findings highlight the reversible, non-stoichiometric nature of OhyA•membrane interactions, with dynamic binding behaviors influenced by protein concentration and lipid environment. This research provides new insights into the dynamic behavior of OhyA on bacterial membranes, highlighting that initial interactions are driven by lipid-mediated protein binding, while sustained interactions are primarily governed by the protein:lipid molar ratio rather than the formation of new, specific lipid-protein interactions. These findings advance our understanding of the biophysical principles underlying OhyA's role in bacterial membrane function and virulence.
Collapse
Affiliation(s)
- William A. Lathram
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, KY, United States
| | - Robert J. Neff
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, KY, United States
| | - Ashley N. Zalla
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, KY, United States
| | - James D. Brien
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, KY, United States
| | | | - Christopher D. Radka
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, KY, United States
| |
Collapse
|
16
|
Chen F, Zhao Y, Dai Y, Sun N, Gao X, Yin J, Zhou Z, Wu KJ. Chick Early Amniotic Fluid Alleviates Dextran-Sulfate-Sodium-Induced Colitis in Mice via T-Cell Receptor Pathway. Antioxidants (Basel) 2025; 14:51. [PMID: 39857385 PMCID: PMC11762673 DOI: 10.3390/antiox14010051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 12/25/2024] [Accepted: 12/31/2024] [Indexed: 01/27/2025] Open
Abstract
Ulcerative colitis (UC) is a chronic immune disease that is difficult to cure. We recently found that chick early amniotic fluid (ceAF) has notable anti-inflammatory and antioxidative properties, through its active components. This study demonstrates the potential of ceAF as a protective agent against UC. UPLC-MS mass spectrometry identified key components of ceAF, including various fatty acids and nucleosides. In vitro, ceAF improved viability in DSS-induced Caco-2 cells, reduced pro-inflammatory cytokines IL-1β and TNF-α, and increased the anti-inflammatory cytokine IL-10. It also upregulated the tight junction proteins ZO-1 and occludin. In DSS-induced UC mice, ceAF treatment alleviated weight loss, colon shortening, and disease activity, while improving histopathology, crypt depth, and colonic fibrosis. Mechanistically, ceAF's anti-inflammatory effects are mediated by inhibiting the overactivation of TCR signaling through the LCK/ZAP70/LAT pathway. Our findings suggest that ceAF could be a valuable nutritional intervention for UC, potentially enhancing existing functional foods aimed at managing this condition.
Collapse
Affiliation(s)
- Fan Chen
- Department of Psychiatry, The Affiliated Wuxi Mental Health Center of Jiangnan University, Wuxi 214151, China; (F.C.); (X.G.); (J.Y.)
- Wuxi School of Medicine, Jiangnan University, Wuxi 214082, China; (Y.Z.); (Y.D.); (N.S.)
| | - Yining Zhao
- Wuxi School of Medicine, Jiangnan University, Wuxi 214082, China; (Y.Z.); (Y.D.); (N.S.)
| | - Yanfa Dai
- Wuxi School of Medicine, Jiangnan University, Wuxi 214082, China; (Y.Z.); (Y.D.); (N.S.)
| | - Ning Sun
- Wuxi School of Medicine, Jiangnan University, Wuxi 214082, China; (Y.Z.); (Y.D.); (N.S.)
| | - Xuezheng Gao
- Department of Psychiatry, The Affiliated Wuxi Mental Health Center of Jiangnan University, Wuxi 214151, China; (F.C.); (X.G.); (J.Y.)
| | - Jiajun Yin
- Department of Psychiatry, The Affiliated Wuxi Mental Health Center of Jiangnan University, Wuxi 214151, China; (F.C.); (X.G.); (J.Y.)
| | - Zhenhe Zhou
- Department of Psychiatry, The Affiliated Wuxi Mental Health Center of Jiangnan University, Wuxi 214151, China; (F.C.); (X.G.); (J.Y.)
| | - Ke-jia Wu
- Department of Psychiatry, The Affiliated Wuxi Mental Health Center of Jiangnan University, Wuxi 214151, China; (F.C.); (X.G.); (J.Y.)
- Wuxi School of Medicine, Jiangnan University, Wuxi 214082, China; (Y.Z.); (Y.D.); (N.S.)
| |
Collapse
|
17
|
Jia D, Tian X, Chen Y, Liu J, Wang M, Hao Z, Wang C, Zhao D. Preparation of enzymatic hydrolysates of mulberry leaf flavonoids and investigation into its treatment and mechanism for zebrafish inflammatory bowel disease. FISH & SHELLFISH IMMUNOLOGY 2024; 154:109960. [PMID: 39393613 DOI: 10.1016/j.fsi.2024.109960] [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: 05/21/2024] [Revised: 09/24/2024] [Accepted: 10/09/2024] [Indexed: 10/13/2024]
Abstract
INTRODUCTION The incidence of inflammatory bowel disease (IBD) has been increasing year by year in recent years. Flavonoids are the main components of mulberry leaves exerting anti-inflammatory effects and have potential applications in drug screening for IBD treatment. Enzymatic hydrolysis can enhance the bioavailability and activity of flavonoid glycosides. No relevant reports on the potentiation of mulberry leaf flavonoids (MLF) after deglycosylation have been retrieved. METHODS An enzymatic method was used to prepare enzymatic hydrolysates of MLF (EMLF). The protective effect of EMLF on zebrafish with IBD was evaluated by observing zebrafish intestinal length and width, intestinal histopathological morphology, as well as neutrophil and goblet expression. Network pharmacology and metabolomics were performed to explore the mechanism of action of EMLF against IBD. Finally, the mechanism of EMLF against IBD was validated using Western Blot and real-time quantitative polymerase chain reaction (RT-qPCR). RESULTS The EMLF was successfully prepared by hydrolyzing MLF with snailase for the first time, and the anti-inflammatory effect of EMLF was clarified to be superior to that of MLF in IBD zebrafish. The network pharmacology and metabolomics studies showed that the mechanism of EMLF for IBD may be related to regulation of purine metabolic pathway. The expression levels of adenosine deaminase (ADA), critical targets in purine metabolism, were significantly down-regulated, while the expression of adenine phosphoribosyltransferase (APRT) and xanthine dehydrogenase (XDH) was significantly increased when compared with the TNBS group. CONCLUSION The results suggested that enzymatic deglycosylation is an effective measure to enhance the anti-inflammatory activity of MLF, which provides referable ideas and methods for the deep processing and utilization of natural drug resources.
Collapse
Affiliation(s)
- Dongsheng Jia
- Department of Pharmacognosy, School of Pharmacy, Hebei Medical University, 361 East Zhongshan Road, Shijiazhuang, Hebei, 050017, PR China; Institute of Cash Crop, Hebei Academy of Agriculture and Forestry Sciences, Shijiazhuang, Hebei, 050051, PR China
| | - Xi Tian
- Institute of Cash Crop, Hebei Academy of Agriculture and Forestry Sciences, Shijiazhuang, Hebei, 050051, PR China
| | - Yuting Chen
- Department of Pharmacognosy, School of Pharmacy, Hebei Medical University, 361 East Zhongshan Road, Shijiazhuang, Hebei, 050017, PR China
| | - Jie Liu
- Department of Pharmacognosy, School of Pharmacy, Hebei Medical University, 361 East Zhongshan Road, Shijiazhuang, Hebei, 050017, PR China
| | - Man Wang
- Department of Pharmacognosy, School of Pharmacy, Hebei Medical University, 361 East Zhongshan Road, Shijiazhuang, Hebei, 050017, PR China
| | - Zhangsen Hao
- Department of Pharmacognosy, School of Pharmacy, Hebei Medical University, 361 East Zhongshan Road, Shijiazhuang, Hebei, 050017, PR China
| | - Changshun Wang
- Department of Pharmacognosy, School of Pharmacy, Hebei Medical University, 361 East Zhongshan Road, Shijiazhuang, Hebei, 050017, PR China
| | - Ding Zhao
- Department of Pharmacognosy, School of Pharmacy, Hebei Medical University, 361 East Zhongshan Road, Shijiazhuang, Hebei, 050017, PR China.
| |
Collapse
|
18
|
Peng D, Wang Y, Yao Y, Yang Z, Wu S, Zeng K, Hu X, Zhao Y. Long-chain polyunsaturated fatty acids influence colorectal cancer progression via the interactions between the intestinal microflora and the macrophages. Mol Cell Biochem 2024; 479:2895-2906. [PMID: 38217838 DOI: 10.1007/s11010-023-04904-y] [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: 08/28/2023] [Accepted: 11/15/2023] [Indexed: 01/15/2024]
Abstract
The metabolism of long-chain polyunsaturated fatty acids (LCPUFAs) is closely associated with the risk and progression of colorectal cancer (CRC). This paper aims to investigate the role of LCPUFA in the crosstalk between intestinal microflora and macrophages, as well as the effects of these three parties on the progression of CRC. The metabolism and function of LCPUFA play important roles in regulating the composition of the human gut microflora and participating in the regulation of inflammation, ultimately affecting macrophage function and polarization, which is crucial in the tumor microenvironment. The effects of LCPUFA on cellular interactions between the two species can ultimately influence the progression of CRC. In this review, we explore the molecular mechanisms and clinical applications of LCPUFA in the interactions between intestinal microflora and intestinal macrophages, as well as its significance for CRC progression. Furthermore, we reveal the role of LCPUFA in the construction of the CRC microenvironment and explore the key nodes of the interactions between intestinal flora and intestinal macrophages in the environment. It provides potential targets for the metabolic diagnosis and treatment of CRC.
Collapse
Affiliation(s)
- Duo Peng
- Pathology Department of The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523713, China
- School of Medical Technology, Guangdong Medical University, Dongguan, 523808, China
| | - Yan Wang
- Pathology Department of The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523713, China
- Microbiology and Immunology Department, Guangdong Medical University, Dongguan, 523808, China
| | - Yunhong Yao
- Pathology Department of The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523713, China
- Pathology Department, Guangdong Medical University, Dongguan, 523808, China
| | - Zisha Yang
- School of Medical Technology, Guangdong Medical University, Dongguan, 523808, China
| | - Shuang Wu
- Pathology Department, Guangdong Medical University, Dongguan, 523808, China
| | - Kaijing Zeng
- Pathology Department, Guangdong Medical University, Dongguan, 523808, China
| | - Xinrong Hu
- Pathology Department of The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523713, China.
- Pathology Department, Guangdong Medical University, Dongguan, 523808, China.
| | - Yi Zhao
- Pathology Department of The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523713, China.
- Microbiology and Immunology Department, Guangdong Medical University, Dongguan, 523808, China.
- School of Medical Technology, Guangdong Medical University, Dongguan, 523808, China.
| |
Collapse
|
19
|
Pessina F, Casini I, Gamberucci A, Carullo G, Signorini C, Brizzi A, Aiello F, Aloisi AM, Pieretti S. Anti-Inflammatory and Antinociceptive Properties of the Quercetin-3-Oleate AV2, a Novel FFAR1 Partial Agonist. Int J Mol Sci 2024; 25:11635. [PMID: 39519187 PMCID: PMC11546106 DOI: 10.3390/ijms252111635] [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: 09/27/2024] [Revised: 10/24/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
Free fatty acid receptor 1 (FFAR1) has emerged as the most targeted isoform of the free fatty acid receptors because of its involvement in the modulation of energy balance and its potential role in the control of inflammatory and pain conditions. Quercetin-3-oleate (AV2), recognized as a new FFAR1 partial agonist, was investigated for its ability to modulate inflammation and nociception. Human immortal neuroblastoma SH and the murine macrophagic RAW 264.7 cells were used to evaluate cell viability, the potential cytoprotective activity, and the anti-inflammatory properties of AV2 in vitro. Paw edema, caused by zymosan-A, and the formalin test were used to assess the in vivo anti-inflammatory and antinociceptive effects in CD-1 mice. In vitro, AV2 was devoid of cytotoxicity, significantly reduced ROS in both cell types, and protected RAW 264.7 cells from lipopolysaccharide damage by reducing tumor necrosis factor-α production. Interestingly, AV2 induced a transient elevation of intracellular calcium that was reduced in cells, pre-incubated with the FFAR1 antagonist DC260126. In vivo, AV2 reduced formalin-induced nociception and zymosan A-induced paw edema, and both effects were reversed by the FFAR1 antagonist GW1100. In conclusion, these data strongly support the AV2-mediated antioxidant, anti-inflammatory, and antinociceptive activity. AV2 represents a promising molecule for the clinical management of inflammatory-related pain conditions.
Collapse
Affiliation(s)
- Federica Pessina
- Department of Molecular and Developmental Medicine, University of Siena, Via A. Moro 2, 53100 Siena, Italy; (A.G.); (C.S.)
| | - Ilenia Casini
- Department of Medicine, Surgery and Neuroscience, University of Siena, Via A. Moro 2, 53100 Siena, Italy; (I.C.); (A.M.A.)
| | - Alessandra Gamberucci
- Department of Molecular and Developmental Medicine, University of Siena, Via A. Moro 2, 53100 Siena, Italy; (A.G.); (C.S.)
| | - Gabriele Carullo
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via A. Moro 2, 53100 Siena, Italy;
| | - Cinzia Signorini
- Department of Molecular and Developmental Medicine, University of Siena, Via A. Moro 2, 53100 Siena, Italy; (A.G.); (C.S.)
| | - Antonella Brizzi
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via A. Moro 2, 53100 Siena, Italy;
| | - Francesca Aiello
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata, Italy;
| | - Anna Maria Aloisi
- Department of Medicine, Surgery and Neuroscience, University of Siena, Via A. Moro 2, 53100 Siena, Italy; (I.C.); (A.M.A.)
| | - Stefano Pieretti
- Istituto Superiore di Sanità, National Centre for Drug Research and Evaluation, Viale Regina Elena 299, 00161 Rome, Italy;
| |
Collapse
|
20
|
Heng YC, Wong GWJ, Kittelmann S. Expanding the biosynthesis spectrum of hydroxy fatty acids: unleashing the potential of novel bacterial fatty acid hydratases. BIOTECHNOLOGY FOR BIOFUELS AND BIOPRODUCTS 2024; 17:131. [PMID: 39456067 PMCID: PMC11515146 DOI: 10.1186/s13068-024-02578-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 10/18/2024] [Indexed: 10/28/2024]
Abstract
BACKGROUND Hydroxy fatty acids represent an emerging class of compounds with promising applications in the chemical, medicinal and functional food sectors. The challenges associated with their chemical synthesis have spurred exploration of biological synthesis as an alternative route, particularly through the use of fatty acid hydratases. Fatty acid hydratases catalyse the regioselective addition of a hydrogen atom and a hydroxyl group from a water molecule to the carbon-carbon cis-double bond of unsaturated fatty acids to form hydroxy fatty acids. Despite having been discovered in the early 1960s, previous research has primarily focused on characterizing single fatty acid hydratase variants with a limited range of substrates. Comprehensive studies that systematically examine and compare the characteristics of multiple variants of fatty acid hydratases are still lacking. RESULTS In this study, we employed an integrated bioinformatics workflow to identify 23 fatty acid hydratases and characterized their activities against nine unsaturated fatty acid substrates using whole-cell biotransformation assays. Additionally, we tested a dual-protein system involving two fatty acid hydratases of distinct regioselectivity and demonstrated its suitability in enhancing the biosynthesis of di-hydroxy fatty acids. CONCLUSIONS Our study demonstrates that fatty acid hydratases can be classified into three subtypes based on their regioselectivity and provides insights into their preferred substrate structures. These understandings pave ways for the design of optimal fatty acid hydratase variants and bioprocesses for the cost-efficient biosynthesis of hydroxy fatty acids.
Collapse
Affiliation(s)
- Yu Chyuan Heng
- Wilmar International Limited, 28 Biopolis Road, Singapore, 138568, Singapore.
| | - Garrett Wei Jie Wong
- Wilmar International Limited, 28 Biopolis Road, Singapore, 138568, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, 14 Medical Drive, Singapore, 117599, Singapore
| | - Sandra Kittelmann
- Wilmar International Limited, 28 Biopolis Road, Singapore, 138568, Singapore.
| |
Collapse
|
21
|
Van Hul M, Cani PD, Petitfils C, De Vos WM, Tilg H, El-Omar EM. What defines a healthy gut microbiome? Gut 2024; 73:1893-1908. [PMID: 39322314 PMCID: PMC11503168 DOI: 10.1136/gutjnl-2024-333378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 08/16/2024] [Indexed: 09/27/2024]
Abstract
The understanding that changes in microbiome composition can influence chronic human diseases and the efficiency of therapies has driven efforts to develop microbiota-centred therapies such as first and next generation probiotics, prebiotics and postbiotics, microbiota editing and faecal microbiota transplantation. Central to microbiome research is understanding how disease impacts microbiome composition and vice versa, yet there is a problematic issue with the term 'dysbiosis', which broadly links microbial imbalances to various chronic illnesses without precision or definition. Another significant issue in microbiome discussions is defining 'healthy individuals' to ascertain what characterises a healthy microbiome. This involves questioning who represents the healthiest segment of our population-whether it is those free from illnesses, athletes at peak performance, individuals living healthily through regular exercise and good nutrition or even elderly adults or centenarians who have been tested by time and achieved remarkable healthy longevity.This review advocates for delineating 'what defines a healthy microbiome?' by considering a broader range of factors related to human health and environmental influences on the microbiota. A healthy microbiome is undoubtedly linked to gut health. Nevertheless, it is very difficult to pinpoint a universally accepted definition of 'gut health' due to the complexities of measuring gut functionality besides the microbiota composition. We must take into account individual variabilities, the influence of diet, lifestyle, host and environmental factors. Moreover, the challenge in distinguishing causation from correlation between gut microbiome and overall health is presented.The review also highlights the resource-heavy nature of comprehensive gut health assessments, which hinders their practicality and broad application. Finally, we call for continued research and a nuanced approach to better understand the intricate and evolving concept of gut health, emphasising the need for more precise and inclusive definitions and methodologies in studying the microbiome.
Collapse
Affiliation(s)
- Matthias Van Hul
- Louvain Drug Research Institute (LDRI), Metabolism and Nutrition research group (MNUT), UCLouvain, Université catholique de Louvain, Brussels, Belgium
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO) department, WEL Research Institute, Wavre, Belgium
| | - Patrice D Cani
- Louvain Drug Research Institute (LDRI), Metabolism and Nutrition research group (MNUT), UCLouvain, Université catholique de Louvain, Brussels, Belgium
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO) department, WEL Research Institute, Wavre, Belgium
- Institute of Experimental and Clinical Research (IREC), UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Camille Petitfils
- Louvain Drug Research Institute (LDRI), Metabolism and Nutrition research group (MNUT), UCLouvain, Université catholique de Louvain, Brussels, Belgium
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO) department, WEL Research Institute, Wavre, Belgium
| | - Willem M De Vos
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
| | - Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medizinische Universitat Innsbruck, Innsbruck, Austria
| | - Emad M El-Omar
- Microbiome Research Centre, St George and Sutherland Clinical Campuses, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
22
|
Oldham ML, Zuhaib Qayyum M, Kalathur RC, Rock CO, Radka CD. Cryo-EM reconstruction of oleate hydratase bound to a phospholipid membrane bilayer. J Struct Biol 2024; 216:108116. [PMID: 39151742 PMCID: PMC11385989 DOI: 10.1016/j.jsb.2024.108116] [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: 04/14/2024] [Revised: 08/08/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024]
Abstract
Oleate hydratase (OhyA) is a bacterial peripheral membrane protein that catalyzes FAD-dependent water addition to membrane bilayer-embedded unsaturated fatty acids. The opportunistic pathogen Staphylococcus aureus uses OhyA to counteract the innate immune system and support colonization. Many Gram-positive and Gram-negative bacteria in the microbiome also encode OhyA. OhyA is a dimeric flavoenzyme whose carboxy terminus is identified as the membrane binding domain; however, understanding how OhyA binds to cellular membranes is not complete until the membrane-bound structure has been elucidated. All available OhyA structures depict the solution state of the protein outside its functional environment. Here, we employ liposomes to solve the cryo-electron microscopy structure of the functional unit: the OhyA•membrane complex. The protein maintains its structure upon membrane binding and slightly alters the curvature of the liposome surface. OhyA preferentially associates with 20-30 nm liposomes with multiple copies of OhyA dimers assembling on the liposome surface resulting in the formation of higher-order oligomers. Dimer assembly is cooperative and extends along a formed ridge of the liposome. We also solved an OhyA dimer of dimers structure that recapitulates the intermolecular interactions that stabilize the dimer assembly on the membrane bilayer as well as the crystal contacts in the lattice of the OhyA crystal structure. Our work enables visualization of the molecular trajectory of membrane binding for this important interfacial enzyme.
Collapse
Affiliation(s)
- Michael L Oldham
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - M Zuhaib Qayyum
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Ravi C Kalathur
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Charles O Rock
- Department of Host Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Christopher D Radka
- Department of Host Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA; Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, KY, 40536, USA.
| |
Collapse
|
23
|
Paquay S, Duraffourd J, Bury M, Heremans IP, Caligiore F, Gerin I, Stroobant V, Jacobs J, Pinon A, Graff J, Vertommen D, Van Schaftingen E, Dewulf JP, Bommer GT. ACAD10 and ACAD11 allow entry of 4-hydroxy fatty acids into β-oxidation. Cell Mol Life Sci 2024; 81:367. [PMID: 39174697 PMCID: PMC11342911 DOI: 10.1007/s00018-024-05397-8] [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: 04/27/2024] [Revised: 08/02/2024] [Accepted: 08/05/2024] [Indexed: 08/24/2024]
Abstract
Hydroxylated fatty acids are important intermediates in lipid metabolism and signaling. Surprisingly, the metabolism of 4-hydroxy fatty acids remains largely unexplored. We found that both ACAD10 and ACAD11 unite two enzymatic activities to introduce these metabolites into mitochondrial and peroxisomal β-oxidation, respectively. First, they phosphorylate 4-hydroxyacyl-CoAs via a kinase domain, followed by an elimination of the phosphate to form enoyl-CoAs catalyzed by an acyl-CoA dehydrogenase (ACAD) domain. Studies in knockout cell lines revealed that ACAD10 preferentially metabolizes shorter chain 4-hydroxy fatty acids than ACAD11 (i.e. 6 carbons versus 10 carbons). Yet, recombinant proteins showed comparable activity on the corresponding 4-hydroxyacyl-CoAs. This suggests that the localization of ACAD10 and ACAD11 to mitochondria and peroxisomes, respectively, might influence their physiological substrate spectrum. Interestingly, we observed that ACAD10 is cleaved internally during its maturation generating a C-terminal part consisting of the ACAD domain, and an N-terminal part comprising the kinase domain and a haloacid dehalogenase (HAD) domain. HAD domains often exhibit phosphatase activity, but negligible activity was observed in the case of ACAD10. Yet, inactivation of a presumptive key residue in this domain significantly increased the kinase activity, suggesting that this domain might have acquired a regulatory function to prevent accumulation of the phospho-hydroxyacyl-CoA intermediate. Taken together, our work reveals that 4-hydroxy fatty acids enter mitochondrial and peroxisomal fatty acid β-oxidation via two enzymes with an overlapping substrate repertoire.
Collapse
Affiliation(s)
- Stéphanie Paquay
- Metabolic Research Group, de Duve Institute & WELRI, Université Catholique de Louvain, 1200, Brussels, Belgium
- WELBIO Department, WEL Research Institute, avenue Pasteur, 6, 1300, Wavre, Belgium
- Department of Pediatric Neurology and Metabolic Diseases, Cliniques Universitaires St. Luc, Université Catholique de Louvain, 1200, Brussels, Belgium
| | - Julia Duraffourd
- Metabolic Research Group, de Duve Institute & WELRI, Université Catholique de Louvain, 1200, Brussels, Belgium
- WELBIO Department, WEL Research Institute, avenue Pasteur, 6, 1300, Wavre, Belgium
| | - Marina Bury
- Metabolic Research Group, de Duve Institute & WELRI, Université Catholique de Louvain, 1200, Brussels, Belgium
- WELBIO Department, WEL Research Institute, avenue Pasteur, 6, 1300, Wavre, Belgium
| | - Isaac P Heremans
- Metabolic Research Group, de Duve Institute & WELRI, Université Catholique de Louvain, 1200, Brussels, Belgium
- WELBIO Department, WEL Research Institute, avenue Pasteur, 6, 1300, Wavre, Belgium
| | - Francesco Caligiore
- Metabolic Research Group, de Duve Institute & WELRI, Université Catholique de Louvain, 1200, Brussels, Belgium
- WELBIO Department, WEL Research Institute, avenue Pasteur, 6, 1300, Wavre, Belgium
| | - Isabelle Gerin
- Metabolic Research Group, de Duve Institute & WELRI, Université Catholique de Louvain, 1200, Brussels, Belgium
- WELBIO Department, WEL Research Institute, avenue Pasteur, 6, 1300, Wavre, Belgium
| | | | - Jean Jacobs
- Metabolic Research Group, de Duve Institute & WELRI, Université Catholique de Louvain, 1200, Brussels, Belgium
- WELBIO Department, WEL Research Institute, avenue Pasteur, 6, 1300, Wavre, Belgium
| | - Aymeric Pinon
- Metabolic Research Group, de Duve Institute & WELRI, Université Catholique de Louvain, 1200, Brussels, Belgium
- WELBIO Department, WEL Research Institute, avenue Pasteur, 6, 1300, Wavre, Belgium
| | - Julie Graff
- Metabolic Research Group, de Duve Institute & WELRI, Université Catholique de Louvain, 1200, Brussels, Belgium
- WELBIO Department, WEL Research Institute, avenue Pasteur, 6, 1300, Wavre, Belgium
| | - Didier Vertommen
- Protein Phosphorylation Unit, de Duve Institute & MASSPROT Platform, Université Catholique de Louvain, 1200, Brussels, Belgium
| | - Emile Van Schaftingen
- Metabolic Research Group, de Duve Institute & WELRI, Université Catholique de Louvain, 1200, Brussels, Belgium
- WELBIO Department, WEL Research Institute, avenue Pasteur, 6, 1300, Wavre, Belgium
| | - Joseph P Dewulf
- Metabolic Research Group, de Duve Institute & WELRI, Université Catholique de Louvain, 1200, Brussels, Belgium
- WELBIO Department, WEL Research Institute, avenue Pasteur, 6, 1300, Wavre, Belgium
- Department of Laboratory Medicine, Cliniques Universitaires St. Luc, Université Catholique de Louvain, 1200, Brussels, Belgium
| | - Guido T Bommer
- Metabolic Research Group, de Duve Institute & WELRI, Université Catholique de Louvain, 1200, Brussels, Belgium.
- WELBIO Department, WEL Research Institute, avenue Pasteur, 6, 1300, Wavre, Belgium.
| |
Collapse
|
24
|
Navarré A, Nazareth T, Luz C, Meca G, Escrivá L. Characterization of lactic acid bacteria isolated from human breast milk and their bioactive metabolites with potential application as a probiotic food supplement. Food Funct 2024; 15:8087-8103. [PMID: 38989729 DOI: 10.1039/d4fo02171a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/12/2024]
Abstract
The probiotic properties of twenty-five lactic acid bacteria (LAB) isolated from human breast milk were investigated considering their resistance to gastrointestinal conditions and proteolytic activity. Seven LAB were identified and assessed for auto- and co-aggregation capacity, antibiotic resistance, and behavior during in vitro gastrointestinal digestion. Three Lacticaseibacillus strains were further evaluated for antifungal activity, metabolite production (HPLC-Q-TOF-MS/MS and GC-MS/MS) and proteolytic profiles (SDS-PAGE and HPLC-DAD) in fermented milk, whey, and soy beverage. All strains resisted in vitro gastrointestinal digestion with viable counts higher than 7.9 log10 CFU mL-1 after the colonic phase. Remarkable proteolytic activity was observed for 18/25 strains. Bacterial auto- and co-aggregation of 7 selected strains reached values up to 23 and 20%, respectively. L. rhamnosus B5H2, L. rhamnosus B9H2 and L. paracasei B10L2 inhibited P. verrucosum, F. verticillioides and F. graminearum fungal growth, highlighting L. rhamnosus B5H2. Several metabolites were identified, including antifungal compounds such as phenylacetic acid and 3-phenyllactic acid, and volatile organic compounds produced in fermented milk, whey, and soy beverage. SDS-PAGE demonstrated bacterial hydrolysis of the main milk (caseins) and soy (glycines and beta-conglycines) proteins, with no apparent hydrolysis of whey proteins. However, HPLC-DAD revealed alpha-lactoglobulin reduction up to 82% and 54% in milk and whey, respectively, with L. rhamnosus B5H2 showing the highest proteolytic activity. Overall, the three selected Lacticaseibacillus strains demonstrated probiotic capacity highlighting L. rhamnosus B5H2 with remarkable potential for generating bioactive metabolites and peptides which are capable of promoting human health.
Collapse
Affiliation(s)
- Abel Navarré
- Laboratory of Food Chemistry and Toxicology, Faculty of Pharmacy, University of Valencia, Av/Vicent A. Estellés, s/n 46100 Burjassot, Valencia, Spain.
| | - Tiago Nazareth
- Laboratory of Food Chemistry and Toxicology, Faculty of Pharmacy, University of Valencia, Av/Vicent A. Estellés, s/n 46100 Burjassot, Valencia, Spain.
| | - Carlos Luz
- Laboratory of Food Chemistry and Toxicology, Faculty of Pharmacy, University of Valencia, Av/Vicent A. Estellés, s/n 46100 Burjassot, Valencia, Spain.
| | - Giuseppe Meca
- Laboratory of Food Chemistry and Toxicology, Faculty of Pharmacy, University of Valencia, Av/Vicent A. Estellés, s/n 46100 Burjassot, Valencia, Spain.
| | - Laura Escrivá
- Laboratory of Food Chemistry and Toxicology, Faculty of Pharmacy, University of Valencia, Av/Vicent A. Estellés, s/n 46100 Burjassot, Valencia, Spain.
| |
Collapse
|
25
|
Ma M, Li Y, He Y, Li D, Niu H, Sun M, Miao X, Su Y, Zhang H, Hua M, Wang J. The Combination of Bacillus natto JLCC513 and Ginseng Soluble Dietary Fiber Attenuates Ulcerative Colitis by Modulating the LPS/TLR4/NF-κB Pathway and Gut Microbiota. J Microbiol Biotechnol 2024; 34:1287-1298. [PMID: 38783703 PMCID: PMC11239422 DOI: 10.4014/jmb.2402.02027] [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: 02/19/2024] [Revised: 04/22/2024] [Accepted: 04/25/2024] [Indexed: 05/25/2024]
Abstract
Ulcerative colitis (UC) is an inflammatory bowel disease (IBD) that is currently difficult to treat effectively. Both Bacillus natto (BN) and ginseng-soluble dietary fiber (GSDF) are anti-inflammatory and helps sustain the intestinal barrier. In this study, the protective effects and mechanism of the combination of B. natto JLCC513 and ginseng-soluble dietary fiber (BG) in DSS-induced UC mice were investigated. Intervention with BG worked better than taking BN or GSDF separately, as evidenced by improved disease activity index, colon length, and colon injury and significantly reduced the levels of oxidative and inflammatory factors (LPS, ILs, and TNF-α) in UC mice. Further mechanistic study revealed that BG protected the intestinal barrier integrity by maintaining the tight junction proteins (Occludin and Claudin1) and inhibited the LPS/TLR4/NF-κB pathway in UC mice. In addition, BG increased the abundance of beneficial bacteria such as Bacteroides and Turicibacter and reduced the abundance of harmful bacteria such as Allobaculum in the gut microbiota of UC mice. BG also significantly upregulated genes related to linoleic acid metabolism in the gut microbiota. These BG-induced changes in the gut microbiota of mice with UC were significantly correlated with changes in pathological indices. In conclusion, this study demonstrated that BG exerts protective effect against UC by regulating the LPS/TLR4/NF-κB pathway and the structure and metabolic function of gut microbiota. Thus, BG can be potentially used in intestinal health foods to treat UC.
Collapse
Affiliation(s)
- Mingyue Ma
- Agronomy of Food Science and Technology, Yanbian University, Yanji 133002, Jilin, P.R. China
- Institute of Agro-product Process, Jilin Academy of Agricultural Science (Northeast Agricultural Research Center of China), Changchun 130033, Jilin, P.R. China
| | - Yueqiao Li
- Institute of Agro-product Process, Jilin Academy of Agricultural Science (Northeast Agricultural Research Center of China), Changchun 130033, Jilin, P.R. China
| | - Yuguang He
- Institute of Agro-product Process, Jilin Academy of Agricultural Science (Northeast Agricultural Research Center of China), Changchun 130033, Jilin, P.R. China
| | - Da Li
- Institute of Agro-product Process, Jilin Academy of Agricultural Science (Northeast Agricultural Research Center of China), Changchun 130033, Jilin, P.R. China
| | - Honghong Niu
- Institute of Agro-product Process, Jilin Academy of Agricultural Science (Northeast Agricultural Research Center of China), Changchun 130033, Jilin, P.R. China
| | - Mubai Sun
- Institute of Agro-product Process, Jilin Academy of Agricultural Science (Northeast Agricultural Research Center of China), Changchun 130033, Jilin, P.R. China
| | - Xinyu Miao
- Institute of Agro-product Process, Jilin Academy of Agricultural Science (Northeast Agricultural Research Center of China), Changchun 130033, Jilin, P.R. China
| | - Ying Su
- Institute of Agro-product Process, Jilin Academy of Agricultural Science (Northeast Agricultural Research Center of China), Changchun 130033, Jilin, P.R. China
| | - Hua Zhang
- Agronomy of Food Science and Technology, Yanbian University, Yanji 133002, Jilin, P.R. China
| | - Mei Hua
- Institute of Agro-product Process, Jilin Academy of Agricultural Science (Northeast Agricultural Research Center of China), Changchun 130033, Jilin, P.R. China
| | - Jinghui Wang
- Institute of Agro-product Process, Jilin Academy of Agricultural Science (Northeast Agricultural Research Center of China), Changchun 130033, Jilin, P.R. China
| |
Collapse
|
26
|
Haneishi Y, Treppiccione L, Maurano F, Luongo D, Miyamoto J, Rossi M. High Fat Diet-Wheat Gliadin Interaction and its Implication for Obesity and Celiac Disease Onset: In Vivo Studies. Mol Nutr Food Res 2024; 68:e2300779. [PMID: 38632845 DOI: 10.1002/mnfr.202300779] [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: 11/03/2023] [Revised: 03/03/2024] [Indexed: 04/19/2024]
Abstract
The intestinal immune system plays a crucial role in obesity and insulin resistance. An altered intestinal immunity is associated with changes to the gut microbiota, barrier function, and tolerance to luminal antigens. Lipid metabolism and its unbalance can also contribute to acute and chronic inflammation in different conditions. In celiac disease (CD), the serum phospholipid profile in infants who developed CD is dramatically different when compared to that of infants at risk of CD not developing the disease. In a mouse model of gluten sensitivity, oral wheat gliadin challenge in connection with inhibition of the metabolism of arachidonic acid, an omega-6 polyunsaturated fatty acid, specifically induces the enteropathy. Recent evidence suggests that gluten may play a role also for development of life-style related diseases in populations on a high fat diet (HFD). However, the mechanisms behind these effects are not yet understood. Exploratory studies in mice feed HFD show that wheat gliadin consumption affects glucose and lipid metabolic homeostasis, alters the gut microbiota, and the immune cell profile in liver.
Collapse
Affiliation(s)
- Yuri Haneishi
- Department of Applied Biological Science, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Fuchu-shi, Tokyo, 183-8509, Japan
| | | | - Francesco Maurano
- Institute of Food Sciences, CNR, via Roma 64, Avellino, 83100, Italy
| | - Diomira Luongo
- Institute of Food Sciences, CNR, via Roma 64, Avellino, 83100, Italy
| | - Junki Miyamoto
- Department of Applied Biological Science, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Fuchu-shi, Tokyo, 183-8509, Japan
| | - Mauro Rossi
- Institute of Food Sciences, CNR, via Roma 64, Avellino, 83100, Italy
| |
Collapse
|
27
|
Chen L, Xu R, Zhu J. Lipidome isotope labelling of gut microbes (LILGM): A method of discovering endogenous microbial lipids. Talanta 2024; 271:125730. [PMID: 38310758 DOI: 10.1016/j.talanta.2024.125730] [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: 10/21/2023] [Revised: 01/22/2024] [Accepted: 01/28/2024] [Indexed: 02/06/2024]
Abstract
Lipidomics analysis of gut microbiome has become critical in recent surge of extensive human disease studies that investigate microbiome contributions. However, challenges remain in comprehending the origins of thousands of lipid species produced by the diverse microbes. Here, we proposed the development and utilization of a liquid chromatography-mass spectrometry-based approach, named lipidome isotope labelling of gut microbes (LILGM), which enables confident detection and identification of endogenous gut microbial lipidome via 13C/15N labeling strategy and high-resolution mass spectrometry. Our method leveraged in vitro microbial cultures and stable isotope-labeled 13C and 15N, allowing a reasonable degree of isotope incorporation into microbial lipids over short-term of inoculation. We then systematically detected the mass spectral patterns of 182 labeled lipid species by our in-house data analysis pipeline. Further bioinformatics analyses confidently identified biologically relevant microbial lipids from lipid classes such as diacylglycerols (DGs), fatty acids (FAs), phosphatidylglycerols (PGs), and phosphatidylethanolamines (PEs) that may have profound impacts to human physiology. Our study also demonstrated the application of LILGM by showcasing the confident detection of dysregulated microbial lipids post antibiotic perturbation. The debiased sparse partial correlation analysis provides insights into lipid metabolism intricacies. Overall, our method can provide unambiguous analyses to the endogenous microbial lipids in given biological context, and can also instantly reflect the lipidomic changes of gut microbes in response to environmental factors. We believe our LILGM approach has the potential to provide new body of knowledge by combining promising analytical approaches for sensitive and specific lipid detection to support functional microbiome studies.
Collapse
Affiliation(s)
- Li Chen
- Human Nutrition Program, Department of Human Sciences, The Ohio State University, Columbus, OH, 43210, USA; James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA
| | - Rui Xu
- Human Nutrition Program, Department of Human Sciences, The Ohio State University, Columbus, OH, 43210, USA
| | - Jiangjiang Zhu
- Human Nutrition Program, Department of Human Sciences, The Ohio State University, Columbus, OH, 43210, USA; James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA.
| |
Collapse
|
28
|
Ando M, Nagata K, Takeshita R, Ito N, Noguchi S, Minamikawa N, Kodama N, Yamamoto A, Yashiro T, Hachisu M, Ichihara G, Kishino S, Yamamoto M, Ogawa J, Nishiyama C. The gut lactic acid bacteria metabolite, 10-oxo- cis-6, trans-11-octadecadienoic acid, suppresses inflammatory bowel disease in mice by modulating the NRF2 pathway and GPCR-signaling. Front Immunol 2024; 15:1374425. [PMID: 38745644 PMCID: PMC11091332 DOI: 10.3389/fimmu.2024.1374425] [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: 01/22/2024] [Accepted: 04/05/2024] [Indexed: 05/16/2024] Open
Abstract
Various gut bacteria, including Lactobacillus plantarum, possess several enzymes that produce hydroxy fatty acids (FAs), oxo FAs, conjugated FAs, and partially saturated FAs from polyunsaturated FAs as secondary metabolites. Among these derivatives, we identified 10-oxo-cis-6,trans-11-octadecadienoic acid (γKetoC), a γ-linolenic acid (GLA)-derived enon FA, as the most effective immunomodulator, which inhibited the antigen-induced immunoactivation and LPS-induced production of inflammatory cytokines. The treatment with γKetoC significantly suppressed proliferation of CD4+ T cells, LPS-induced activation of bone marrow-derived dendritic cells (BMDCs), and LPS-induced IL-6 release from peritoneal cells, splenocytes, and CD11c+ cells isolated from the spleen. γKetoC also inhibited the release of inflammatory cytokines from BMDCs stimulated with poly-I:C, R-848, or CpG. Further in vitro experiments using an agonist of GPR40/120 suggested the involvement of these GPCRs in the effects of γKetoC on DCs. We also found that γKetoC stimulated the NRF2 pathway in DCs, and the suppressive effects of γKetoC and agonist of GPR40/120 on the release of IL-6 and IL-12 were reduced in Nrf2-/- BMDCs. We evaluated the role of NRF2 in the anti-inflammatory effects of γKetoC in a dextran sodium sulfate-induced colitis model. The oral administration of γKetoC significantly reduced body weight loss, improved stool scores, and attenuated atrophy of the colon, in wild-type C57BL/6 and Nrf2+/- mice with colitis. In contrast, the pathology of colitis was deteriorated in Nrf2-/- mice even with the administration of γKetoC. Collectively, the present results demonstrated the involvement of the NRF2 pathway and GPCRs in γKetoC-mediated anti-inflammatory responses.
Collapse
Affiliation(s)
- Miki Ando
- Department of Biological Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, Tokyo, Japan
| | - Kazuki Nagata
- Department of Biological Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, Tokyo, Japan
| | - Ryuki Takeshita
- Department of Biological Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, Tokyo, Japan
| | - Naoto Ito
- Department of Biological Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, Tokyo, Japan
| | - Sakura Noguchi
- Department of Biological Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, Tokyo, Japan
| | - Natsuki Minamikawa
- Department of Biological Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, Tokyo, Japan
| | - Naoki Kodama
- Department of Biological Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, Tokyo, Japan
| | - Asuka Yamamoto
- Department of Biological Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, Tokyo, Japan
| | - Takuya Yashiro
- Department of Biological Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, Tokyo, Japan
| | - Masakazu Hachisu
- Department of Biological Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, Tokyo, Japan
| | - Gaku Ichihara
- Department of Occupational and Environmental Health, Faculty of Pharmaceutical Science, Tokyo University of Science, Chiba, Japan
| | - Shigenobu Kishino
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Masayuki Yamamoto
- Department of Molecular Biochemistry, Tohoku University Tohoku Medical Megabank Organization, Sendai, Japan
| | - Jun Ogawa
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Chiharu Nishiyama
- Department of Biological Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, Tokyo, Japan
| |
Collapse
|
29
|
Mo C, Bi J, Li S, Lin Y, Yuan P, Liu Z, Jia B, Xu S. The influence and therapeutic effect of microbiota in systemic lupus erythematosus. Microbiol Res 2024; 281:127613. [PMID: 38232494 DOI: 10.1016/j.micres.2024.127613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 12/18/2023] [Accepted: 01/09/2024] [Indexed: 01/19/2024]
Abstract
Systemic erythematosus lupus (SLE) is an autoimmune disease involving multiple organs that poses a serious risk to the health and life of patients. A growing number of studies have shown that commensals from different parts of the body and exogenous pathogens are involved in SLE progression, causing barrier disruption and immune dysregulation through multiple mechanisms. However, they sometimes alleviate the symptoms of SLE. Many factors, such as genetic susceptibility, metabolism, impaired barriers, food, and sex hormones, are involved in SLE, and the microbiota drives the development of SLE either by depending on or interacting with these factors. Among these, the crosstalk between genetic susceptibility, metabolism, and microbiota is a hot topic of research and is expected to lay the groundwork for the amelioration of the mechanism, diagnosis, and treatment of SLE. Furthermore, the microbiota has great potential for the treatment of SLE. Ideally, personalised therapeutic approaches should be developed in combination with more specific diagnostic methods. Herein, we provide a comprehensive overview of the role and mechanism of microbiota in lupus of the intestine, oral cavity, skin, and kidney, as well as the therapeutic potential of the microbiota.
Collapse
Affiliation(s)
- Chuzi Mo
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Jiaming Bi
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Siwei Li
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Yunhe Lin
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Peiyan Yuan
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhongjun Liu
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China.
| | - Bo Jia
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China.
| | - Shuaimei Xu
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
30
|
Radka CD, Frank MW, Simmons TS, Johnson CN, Rosch JW, Rock CO. Staphylococcus aureus oleate hydratase produces ligands that activate host PPARα. Front Cell Infect Microbiol 2024; 14:1352810. [PMID: 38601738 PMCID: PMC11004285 DOI: 10.3389/fcimb.2024.1352810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 02/26/2024] [Indexed: 04/12/2024] Open
Abstract
Commensal gut bacteria use oleate hydratase to release a spectrum of hydroxylated fatty acids using host-derived unsaturated fatty acids. These compounds are thought to attenuate the immune response, but the underlying signaling mechanism(s) remain to be established. The pathogen Staphylococcus aureus also expresses an oleate hydratase and 10-hydroxyoctadecanoic acid (h18:0) is the most abundant oleate hydratase metabolite found at Staphylococcal skin infection sites. Here, we show h18:0 stimulates the transcription of a set of lipid metabolism genes associated with the activation of peroxisome proliferator activated receptor (PPAR) in the RAW 264.7 macrophage cell line and mouse primary bone marrow-derived macrophages. Cell-based transcriptional reporter assays show h18:0 selectively activates PPARα. Radiolabeling experiments with bone marrow-derived macrophages show [1-14C]h18:0 is not incorporated into cellular lipids, but is degraded by β-oxidation, and mass spectrometry detected shortened fragments of h18:0 released into the media. The catabolism of h18:0 was >10-fold lower in bone marrow-derived macrophages isolated from Ppara -/- knockout mice, and we recover 74-fold fewer S. aureus cells from the skin infection site of Ppara -/- knockout mice compared to wildtype mice. These data identify PPARα as a target for oleate hydratase-derived hydroxy fatty acids and support the existence of an oleate hydratase-PPARα signaling axis that functions to suppress the innate immune response to S. aureus.
Collapse
Affiliation(s)
- Christopher D. Radka
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, KY, United States
- Department of Host Microbe Interactions, St. Jude Children’s Research Hospital, Memphis, TN, United States
| | - Matthew W. Frank
- Department of Host Microbe Interactions, St. Jude Children’s Research Hospital, Memphis, TN, United States
| | - Tyler S. Simmons
- Department of Host Microbe Interactions, St. Jude Children’s Research Hospital, Memphis, TN, United States
| | - Cydney N. Johnson
- Department of Host Microbe Interactions, St. Jude Children’s Research Hospital, Memphis, TN, United States
| | - Jason W. Rosch
- Department of Host Microbe Interactions, St. Jude Children’s Research Hospital, Memphis, TN, United States
| | - Charles O. Rock
- Department of Host Microbe Interactions, St. Jude Children’s Research Hospital, Memphis, TN, United States
| |
Collapse
|
31
|
Salvadori M, Rosso G. Update on the gut microbiome in health and diseases. World J Methodol 2024; 14:89196. [PMID: 38577200 PMCID: PMC10989414 DOI: 10.5662/wjm.v14.i1.89196] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 12/18/2023] [Accepted: 01/27/2024] [Indexed: 03/07/2024] Open
Abstract
The Human Microbiome Project, Earth Microbiome Project, and next-generation sequencing have advanced novel genome association, host genetic linkages, and pathogen identification. The microbiome is the sum of the microbes, their genetic information, and their ecological niche. This study will describe how millions of bacteria in the gut affect the human body in health and disease. The gut microbiome changes in relation with age, with an increase in Bacteroidetes and Firmicutes. Host and environmental factors affecting the gut microbiome are diet, drugs, age, smoking, exercise, and host genetics. In addition, changes in the gut microbiome may affect the local gut immune system and systemic immune system. In this study, we discuss how the microbiome may affect the metabolism of healthy subjects or may affect the pathogenesis of metabolism-generating metabolic diseases. Due to the high number of publications on the argument, from a methodologically point of view, we decided to select the best papers published in referred journals in the last 3 years. Then we selected the previously published papers. The major goals of our study were to elucidate which microbiome and by which pathways are related to healthy and disease conditions.
Collapse
Affiliation(s)
- Maurizio Salvadori
- Department of Renal Transplantation, Careggi University Hospital, Florence 50139, Tuscany, Italy
| | - Giuseppina Rosso
- Division of Nephrology, San Giovanni di Dio Hospital, Florence 50143, Toscana, Italy
| |
Collapse
|
32
|
Salvadori M, Rosso G. Update on the reciprocal interference between immunosuppressive therapy and gut microbiota after kidney transplantation. World J Transplant 2024; 14:90194. [PMID: 38576749 PMCID: PMC10989467 DOI: 10.5500/wjt.v14.i1.90194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 12/22/2023] [Accepted: 12/29/2023] [Indexed: 03/15/2024] Open
Abstract
Gut microbiota is often modified after kidney transplantation. This principally happens in the first period after transplantation. Antibiotics and, most of all, immunosuppressive drugs are the main responsible. The relationship between immunosuppressive drugs and the gut microbiota is bilateral. From one side immunosuppressive drugs modify the gut microbiota, often generating dysbiosis; from the other side microbiota may interfere with the immunosuppressant pharmacokinetics, producing products more or less active with respect to the original drug. These phenomena have influence over the graft outcomes and clinical consequences as rejections, infections, diarrhea may be caused by the dysbiotic condition. Corticosteroids, calcineurin inhibitors such as tacrolimus and cyclosporine, mycophenolate mofetil and mTOR inhibitors are the immunosuppressive drugs whose effect on the gut microbiota is better known. In contrast is well known how the gut microbiota may interfere with glucocorticoids, which may be transformed into androgens. Tacrolimus may be transformed by micro biota into a product called M1 that is 15-fold less active with respect to tacrolimus. The pro-drug mycophenolate mofetil is normally transformed in mycophenolic acid that according the presence or not of microbes producing the enzyme glu curonidase, may be transformed into the inactive product.
Collapse
Affiliation(s)
- Maurizio Salvadori
- Department of Renal Transplantation, Careggi University Hospital, Florence 50139, Tuscany, Italy
| | - Giuseppina Rosso
- Division of Nephrology, San Giovanni di Dio Hospital, Florence 50143, Toscana, Italy
| |
Collapse
|
33
|
Muñoz-Alvarez KY, Gutiérrez-Aguilar R, Frigolet ME. Metabolic effects of milk fatty acids: A literature review. NUTR BULL 2024; 49:19-39. [PMID: 38226553 DOI: 10.1111/nbu.12657] [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: 08/30/2023] [Revised: 12/07/2023] [Accepted: 12/18/2023] [Indexed: 01/17/2024]
Abstract
Milk and dairy products are known to have a significant role in human development and tissue maintenance due to their high nutritional value. With the higher incidence of obesity and metabolic diseases, nutrition and public health authorities have recommended the intake of fat-free or low-fat dairy due to the saturated fatty acid content of whole-fat products and their effect on serum cholesterol levels. However, recent studies have questioned the association between milk fat consumption and cardiometabolic risk. This literature review aims to compile the scientific evidence of the metabolic effects of milk fatty acids in clinical and basic research studies, as well as their relationship with metabolic disorders and gut microbiota composition. Research shows that various milk fatty acids exert effects on metabolic alterations (obesity, type 2 diabetes and cardiovascular diseases) by modifying glucose homeostasis, inflammation and lipid profile-related factors. Additionally, recent studies have associated the consumption of milk fatty acids with the production of metabolites and the promotion of healthy gut microbiota. From mainly observational studies, evidence suggests that milk and dairy fatty acids are not directly linked to cardiometabolic risk, but further controlled research is necessary to clarify such findings and to assess whether dietary recommendations to choose low-fat dairy foods are necessary for the population for the prevention of obesity and cardiometabolic disease.
Collapse
Affiliation(s)
- Karla Y Muñoz-Alvarez
- Laboratorio de Investigación en Enfermedades Metabólicas: Obesidad y Diabetes, Hospital Infantil de México 'Federico Gómez' (HIMFG), Mexico City, Mexico
| | - Ruth Gutiérrez-Aguilar
- Laboratorio de Investigación en Enfermedades Metabólicas: Obesidad y Diabetes, Hospital Infantil de México 'Federico Gómez' (HIMFG), Mexico City, Mexico
- División de Investigación, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - María E Frigolet
- Laboratorio de Investigación en Enfermedades Metabólicas: Obesidad y Diabetes, Hospital Infantil de México 'Federico Gómez' (HIMFG), Mexico City, Mexico
| |
Collapse
|
34
|
Li J, Zhang Z, Liu H, Qu X, Yin X, Chen L, Guo N, Wang C, Zhang Z. Effects of continuous intravenous infusion with propofol on intestinal metabolites in rats. Biomed Rep 2024; 20:25. [PMID: 38169795 PMCID: PMC10758916 DOI: 10.3892/br.2023.1713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 05/18/2023] [Indexed: 01/05/2024] Open
Abstract
Microbial metabolites play an important role in regulating intestinal homeostasis and immune responses. Propofol is a common anesthetic in clinic, but it is not clear whether it affects intestinal metabolites in rats. Tail vein puncture was performed after adaptive feeding for 1 month in eight 2-month-old rats and they were given continuous intravenous infusion of propofol for 3 h. The feces of rats were divided into different groups based on time periods, with before and after anesthesia with propofol on days 1, 3 and 7 labeled as groups P, A1, A3 and A7, respectively. The effect of continuous intravenous infusion with propofol on rat fecal metabolites was determined using the non-targeted metabolomics technique gas chromatography coupled with a time-of-flight mass spectrometer analysis. The types and contents of metabolites in rat feces were changed after continuous intravenous infusion with propofol, but the changes were not statistically significant. The contents of the metabolites 3-hydroxyphenylacetic acid and palmitic acid increased from day 3 to 7, and it was shown that the two metabolites were positively correlated at a statistically significant level. Linoleic acid decreased to its lowest level on day 3, and it returned to pre-anesthesia level on day 7. At the same time, linoleic acid metabolism was a metabolic pathway that was co-enriched 7 days after infusion with propofol. Spearman correlation analysis showed that there was significant correlation between some differential metabolites and differential microorganisms. It was observed that zymosterol 1, cytosin and elaidic acid were negatively correlated with Alloprevotella in the A3 vs. P group. In the A7 vs. P group, cortexolone 3 and coprostan-3-one were positively correlated with Faecalibacterium, whilst aconitic acid was negatively correlated with it. In conclusion, the present study revealed statistically insignificant effects of continuous intravenous propofol on the intestinal metabolites in rats.
Collapse
Affiliation(s)
- Jiaying Li
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, P.R. China
| | - Zhongjie Zhang
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, P.R. China
| | - Hongyu Liu
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, P.R. China
| | - Xutong Qu
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, P.R. China
| | - Xueqing Yin
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, P.R. China
| | - Lu Chen
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, P.R. China
| | - Nana Guo
- Department of Critical Care Medicine, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, P.R. China
| | - Changsong Wang
- Department of Critical Care Medicine, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, P.R. China
| | - Zhaodi Zhang
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, P.R. China
| |
Collapse
|
35
|
Jing J, Ouyang L, Zhang H, Liang K, Ma R, Ge X, Tang T, Zhao S, Xue T, Shen J, Ma J, Li Z, Wu J, Yang Y, Zhao W, Zheng L, Qian Z, Sun S, Ge Y, Chen L, Li C, Yao B. Omega-3 polyunsaturated fatty acids and its metabolite 12-HEPE rescue busulfan disrupted spermatogenesis via target to GPR120. Cell Prolif 2024; 57:e13551. [PMID: 37743695 PMCID: PMC10849791 DOI: 10.1111/cpr.13551] [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] [Received: 11/25/2022] [Revised: 08/26/2023] [Accepted: 09/08/2023] [Indexed: 09/26/2023] Open
Abstract
Busulfan is an antineoplastic, which is always accompanied with the abnormal of spermatogonia self-renewal and differentiation. It has been demonstrated that the omega-3 polyunsaturated fatty acids (PUFAs) benefits mature spermatozoa. However, whether omega-3 can protect endogenous spermatogonia and the detailed mechanisms are still unclear. Evaluate of spermatogenesis function (in vivo) were examined by histopathological analysis, immunofluorescence staining, and western blotting. The levels of lipid metabolites in testicular tissue were determined via liquid chromatography. We investigated the effect of lipid metabolites on Sertoli cells provided paracrine factors to regulate spermatogonia proliferation and differentiation using co-culture system. In our study, we showed that omega-3 PUFAs significantly improved the process of sperm production and elevated the quantity of both undifferentiated Lin28+ spermatogonia and differentiated c-kit+ spermatogonia in a mouse model where spermatogenic function was disrupted by busulfan. Mass spectrometry revealed an increase in the levels of several omega-3 metabolites in the testes of mice fed with omega-3 PUFAs. The eicosapentaenoic acid metabolite 12-hydroxyeicosapentaenoic acid (12-HEPE) up-regulated bone morphogenic protein 4 (BMP4) expression through GPR120-ERK1/2 pathway activation in Sertoli cells and restored spermatogonia proliferation and differentiation. Our study provides evidence that omega-3 PUFAs metabolite 12-HEPE effectively protects spermatogonia and reveals that GPR120 might be a tractable pharmacological target for fertility in men received chemotherapy or severe spermatogenesis dysfunction.
Collapse
Affiliation(s)
- Jun Jing
- State Key Laboratory of Reproductive Medicine and Offspring HealthNanjing Medical UniversityNanjingChina
- Department of Reproductive Medicine, Affiliated Jinling HospitalNanjing Medical UniversityNanjingChina
- Department of Reproductive Medicine, Affiliated Jinling Hospital, Clinical School of Medical CollegeNanjing UniversityNanjingChina
| | - Lei Ouyang
- Department of Reproductive Medicine, Affiliated Jinling Hospital, The First School of Clinical MedicineSouthern Medical UniversityNanjingChina
| | - Hong Zhang
- Department of Reproductive Medicine, Affiliated Jinling Hospital, Clinical School of Medical CollegeNanjing UniversityNanjingChina
| | - Kuan Liang
- Department of Reproductive Medicine, Affiliated Jinling HospitalNanjing Medical UniversityNanjingChina
- Department of Reproductive Medicine, Affiliated Jinling Hospital, The First School of Clinical MedicineSouthern Medical UniversityNanjingChina
| | - Rujun Ma
- Department of Reproductive Medicine, Affiliated Jinling HospitalNanjing Medical UniversityNanjingChina
- Department of Reproductive Medicine, Affiliated Jinling Hospital, Clinical School of Medical CollegeNanjing UniversityNanjingChina
| | - Xie Ge
- Department of Reproductive Medicine, Affiliated Jinling HospitalNanjing Medical UniversityNanjingChina
- Department of Reproductive Medicine, Affiliated Jinling Hospital, Clinical School of Medical CollegeNanjing UniversityNanjingChina
| | - Ting Tang
- State Key Laboratory of Reproductive Medicine and Offspring HealthNanjing Medical UniversityNanjingChina
- Department of Reproductive Medicine, Affiliated Jinling HospitalNanjing Medical UniversityNanjingChina
- Department of Reproductive Medicine, Affiliated Jinling Hospital, Clinical School of Medical CollegeNanjing UniversityNanjingChina
| | - Shanmeizi Zhao
- School of Life ScienceNanjing Normal UniversityNanjingChina
| | - Tongmin Xue
- State Key Laboratory of Reproductive Medicine and Offspring HealthNanjing Medical UniversityNanjingChina
- Department of Reproductive Medicine, Affiliated Jinling HospitalNanjing Medical UniversityNanjingChina
- Reproductive Medical Center, Clinical Medical College (Northern Jiangsu People's Hospital)Yangzhou UniversityYangzhouChina
| | - Jiaming Shen
- Department of Reproductive Medicine, Affiliated Jinling Hospital, Clinical School of Medical CollegeNanjing UniversityNanjingChina
| | - Jinzhao Ma
- Department of Reproductive Medicine, Affiliated Jinling HospitalNanjing Medical UniversityNanjingChina
- Department of Reproductive Medicine, Affiliated Jinling Hospital, Clinical School of Medical CollegeNanjing UniversityNanjingChina
| | - Zhou Li
- Department of Reproductive Medicine, Affiliated Jinling Hospital, Clinical School of Medical CollegeNanjing UniversityNanjingChina
| | - Jing Wu
- Core Laboratory, Sir Run Run HospitalNanjing Medical UniversityNanjingChina
| | - Yang Yang
- Basic Medical Laboratory, Affiliated Jinling Hospital, Clinical School of Medical CollegeNanjing UniversityNanjingChina
| | - Wei Zhao
- Department of Reproductive Medicine, Affiliated Jinling HospitalNanjing Medical UniversityNanjingChina
- Department of Reproductive Medicine, Affiliated Jinling Hospital, Clinical School of Medical CollegeNanjing UniversityNanjingChina
| | - Lu Zheng
- Department of Reproductive Medicine, Affiliated Jinling Hospital, Clinical School of Medical CollegeNanjing UniversityNanjingChina
| | - Zhang Qian
- Department of Reproductive Medicine, Affiliated Jinling Hospital, Clinical School of Medical CollegeNanjing UniversityNanjingChina
| | - Shanshan Sun
- School of Life ScienceNanjing Normal UniversityNanjingChina
| | - Yifeng Ge
- Department of Reproductive Medicine, Affiliated Jinling HospitalNanjing Medical UniversityNanjingChina
- Department of Reproductive Medicine, Affiliated Jinling Hospital, Clinical School of Medical CollegeNanjing UniversityNanjingChina
| | - Li Chen
- Department of Reproductive Medicine, Affiliated Jinling HospitalNanjing Medical UniversityNanjingChina
- Department of Reproductive Medicine, Affiliated Jinling Hospital, Clinical School of Medical CollegeNanjing UniversityNanjingChina
| | - Chaojun Li
- State Key Laboratory of Reproductive Medicine and Offspring HealthNanjing Medical UniversityNanjingChina
| | - Bing Yao
- State Key Laboratory of Reproductive Medicine and Offspring HealthNanjing Medical UniversityNanjingChina
- Department of Reproductive Medicine, Affiliated Jinling HospitalNanjing Medical UniversityNanjingChina
- Department of Reproductive Medicine, Affiliated Jinling Hospital, Clinical School of Medical CollegeNanjing UniversityNanjingChina
- Department of Reproductive Medicine, Affiliated Jinling Hospital, The First School of Clinical MedicineSouthern Medical UniversityNanjingChina
- School of Life ScienceNanjing Normal UniversityNanjingChina
| |
Collapse
|
36
|
Zhang H, Xie Y, Cao F, Song X. Gut microbiota-derived fatty acid and sterol metabolites: biotransformation and immunomodulatory functions. Gut Microbes 2024; 16:2382336. [PMID: 39046079 PMCID: PMC11271093 DOI: 10.1080/19490976.2024.2382336] [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: 12/28/2023] [Revised: 05/26/2024] [Accepted: 07/16/2024] [Indexed: 07/25/2024] Open
Abstract
Commensal microorganisms in the human gut produce numerous metabolites by using small molecules derived from the host or diet as precursors. Host or dietary lipid molecules are involved in energy metabolism and maintaining the structural integrity of cell membranes. Notably, gut microbes can convert these lipids into bioactive signaling molecules through their biotransformation and synthesis pathways. These microbiota-derived lipid metabolites can affect host physiology by influencing the body's immune and metabolic processes. This review aims to summarize recent advances in the microbial transformation and host immunomodulatory functions of these lipid metabolites, with a special focus on fatty acids and steroids produced by our gut microbiota.
Collapse
Affiliation(s)
- Haohao Zhang
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Yadong Xie
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Fei Cao
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Xinyang Song
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
37
|
Yan Z, Yin B, Wang Y, Ni Z, Feng J, Yang Q, Li X, Zhu H, Dou Y. Therapeutic mechanism of Liangxue-Guyuan-Yishen decoction on intestinal stem cells and tight junction proteins in gastrointestinal acute radiation syndrome rats. JOURNAL OF RADIATION RESEARCH 2023; 64:880-892. [PMID: 37697698 PMCID: PMC10665307 DOI: 10.1093/jrr/rrad065] [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: 02/23/2023] [Revised: 05/04/2023] [Accepted: 08/19/2023] [Indexed: 09/13/2023]
Abstract
On the basis of the previous research, the Traditional Chinese Medicine theory was used to improve the drug composition for gastrointestinal acute radiation syndrome (GI-ARS). The purpose of this study was to study the therapeutic mechanism of Liangxue-Guyuan-Yishen decoction (LGYD) on GI-ARS and to provide a new scheme for the treatment of radiation injury. Here, we investigated the effects of LGYD on intestinal stem cells (ISCs) in a GI-ARS rat model. Rat health and survival and the protective efficacy of LGYD on the intestines were analyzed. The active principles in LGYD were detected using liquid chromatography-mass spectrometry (LC-MS). ISC proliferation, intestinal epithelial tight junction (TJ) protein expression and regulatory pathways were explored using immunohistochemistry, western blotting (WB) and reverse transcription quantitative polymerase chain reaction (RT-qPCR), respectively. Involvement of the WNT and MEK/ERK pathways in intestinal recovery was screened using network pharmacology analysis and validated by WB and RT-qPCR. LGYD administration significantly improved health and survival in GI-ARS rats. Pathological analysis showed that LGYD ameliorated radiation-induced intestinal injury and significantly promoted LGR5+ stem cell regeneration in the intestinal crypts, upregulated TJ protein, and accelerated crypt reconstruction in the irradiated rats. LC-MS revealed ≥13 constituents that might contribute to LGYD's protective effects. Collectively, LGYD can promote crypt cell proliferation and ISCs after radiation damage, the above effect may be related to WNT and MEK/ERK pathway.
Collapse
Affiliation(s)
- Ziqiao Yan
- Department of Traditional Chinese Medicine, The First Medical Center of Chinese People’s Liberation Army (PLA) General Hospital, Fuxing Road 28th, Haidian District, Beijing, 10038, China
- Chinese PLA Medical School, Chinese People’s Liberation Army (PLA) General Hospital, Fuxing Road 28th, Haidian District, Beijing, 10038, China
| | - Bofeng Yin
- Department of Experimental Hematology and Biochemistry, Beijing Institute of Radiation Medicine, Taiping Road 27th, Haidian District, Beijing, 10039, China
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Taiping Road 27th, Haidian District, Beijing, 10039, China
| | - Yuguo Wang
- Department of Traditional Chinese Medicine, The Sixth Medical Center of Chinese People’s Liberation Army (PLA) General Hospital, Fucheng Road 6th, Haidian District, Beijing, 10037, China
| | - Zhexin Ni
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Taiping Road 27th, Haidian District, Beijing, 10039, China
| | - Jian Feng
- Department of Traditional Chinese Medicine, The Chinese People’s Liberation Army (PLA) 96604 Hospital, Jingningnan Road 72th, Chengguan District, Lanzhou, 730030, China
| | - Qianyu Yang
- Graduate School of Hebei University of Chinese Medicine, Xinshinan Road 326th, Qiaoxi District, Shijiazhuang, Hebei, 050090, China
| | - Xiao Li
- Chinese PLA Medical School, Chinese People’s Liberation Army (PLA) General Hospital, Fuxing Road 28th, Haidian District, Beijing, 10038, China
| | - Heng Zhu
- Department of Experimental Hematology and Biochemistry, Beijing Institute of Radiation Medicine, Taiping Road 27th, Haidian District, Beijing, 10039, China
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Taiping Road 27th, Haidian District, Beijing, 10039, China
- Beijing Institute of Basic Medical Sciences, Taiping Road 27th, Haidian District, Beijing, 10039, China
- Graduate School of Anhui Medical University, Meishan Road 69th, Shushan District, Hefei, Anhui, 230022, China
| | - Yongqi Dou
- Department of Traditional Chinese Medicine, The First Medical Center of Chinese People’s Liberation Army (PLA) General Hospital, Fuxing Road 28th, Haidian District, Beijing, 10038, China
- Chinese PLA Medical School, Chinese People’s Liberation Army (PLA) General Hospital, Fuxing Road 28th, Haidian District, Beijing, 10038, China
| |
Collapse
|
38
|
Uehira Y, Ueno H, Miyamoto J, Kimura I, Ishizawa Y, Iijima H, Muroga S, Fujita T, Sakai S, Samukawa Y, Tanaka Y, Murayama S, Sakoda H, Nakazato M. Impact of the lipase inhibitor orlistat on the human gut microbiota. Obes Res Clin Pract 2023; 17:411-420. [PMID: 37679239 DOI: 10.1016/j.orcp.2023.08.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 08/18/2023] [Accepted: 08/24/2023] [Indexed: 09/09/2023]
Abstract
Orlistat, an anti-obesity agent, inhibits the metabolism and absorption of dietary fat by inactivating pancreatic lipase in the gut. The effect of orlistat on the gut microbiota of Japanese individuals with obesity is unknown. This study aimed to explore the effects of orlistat on the gut microbiota and fatty acid metabolism of Japanese individuals with obesity. Fourteen subjects with visceral fat obesity (waist circumference ≥85 cm) took orlistat orally at a dose of 60 mg, 3 times a day for 8 weeks. Body weight; waist circumference; visceral fat area; levels of short-chain fatty acids, gut microbiota, fatty acid metabolites in the feces, and gastrointestinal hormones; and adverse events were evaluated. Body weight, waist circumference, and blood leptin concentrations were significantly lower after orlistat treatment (mean ± standard deviation, 77.8 ± 9.1 kg; 91.9 ± 8.7 cm; and 4546 ± 3211 pg/mL, respectively) compared with before treatment (79.4 ± 9.0 kg; 94.4 ± 8.0 cm; and 5881 ± 3526 pg/mL, respectively). Significant increases in fecal levels of fatty acid metabolites (10-hydroxy-cis-12-octadecenoic acid, 10-oxo-cis-12-octadecenoic acid, and 10-oxo-trans-11-octadecenoic acid) were detected. Meanwhile, no significant changes were found in abdominal computed tomography parameters, blood marker levels, or short-chain fatty acid levels in the feces. Gut microbiota analysis revealed that some study subjects had decreased abundance of Firmicutes, increased abundance of Bacteroidetes, and increased α-diversity indices (Chao1 and ACE) after 8 weeks of treatment. The levels of Lactobacillus genus and Lactobacillus gasseri were significantly higher after 8 weeks of treatment. None of the subjects discontinued treatment or experienced severe adverse events. This study suggested that orlistat might alter gut microbiota composition and affect the body through fatty acid metabolites produced by the modified gut bacteria.
Collapse
Affiliation(s)
- Yudai Uehira
- Division of Neurology, Respirology, Endocrinology, and Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Kihara, Kiyotake, Miyazaki 889-1692, Japan
| | - Hiroaki Ueno
- Division of Neurology, Respirology, Endocrinology, and Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Kihara, Kiyotake, Miyazaki 889-1692, Japan
| | - Junki Miyamoto
- Department of Applied Biological Science, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Fuchu-shi, Tokyo 183-8509, Japan
| | - Ikuo Kimura
- Laboratory of Molecular Neurobiology, Graduate School of Biostudies, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Yohei Ishizawa
- DNA Chip Research Inc., Minato-ku, Tokyo 105-0022, Japan
| | - Hiroshi Iijima
- DNA Chip Research Inc., Minato-ku, Tokyo 105-0022, Japan
| | - Shota Muroga
- Taisho Pharmaceutical Co., Ltd., Toshima-ku, Tokyo 170-8633, Japan
| | - Toru Fujita
- Taisho Pharmaceutical Co., Ltd., Toshima-ku, Tokyo 170-8633, Japan
| | - Soichi Sakai
- Taisho Pharmaceutical Co., Ltd., Toshima-ku, Tokyo 170-8633, Japan
| | | | - Yuri Tanaka
- Department of Bioregulatory Sciences, Faculty of Medicine, University of Miyazaki, Kihara, Kiyotake, Miyazaki 889-1692, Japan
| | - Shinya Murayama
- Department of Bioregulatory Sciences, Faculty of Medicine, University of Miyazaki, Kihara, Kiyotake, Miyazaki 889-1692, Japan
| | - Hideyuki Sakoda
- Division of Neurology, Respirology, Endocrinology, and Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Kihara, Kiyotake, Miyazaki 889-1692, Japan; Department of Bioregulatory Sciences, Faculty of Medicine, University of Miyazaki, Kihara, Kiyotake, Miyazaki 889-1692, Japan
| | - Masamitsu Nakazato
- Division of Neurology, Respirology, Endocrinology, and Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Kihara, Kiyotake, Miyazaki 889-1692, Japan; Department of Bioregulatory Sciences, Faculty of Medicine, University of Miyazaki, Kihara, Kiyotake, Miyazaki 889-1692, Japan; Department of Inter-organ Communication Research Project, Frontier Science Research Center, University of Miyazaki, Kihara, Kiyotake, Miyazaki 889-1692, Japan; Institute for Protein Research, Osaka University, Osaka 565-0871, Japan.
| |
Collapse
|
39
|
Sahoo DK, Heilmann RM, Paital B, Patel A, Yadav VK, Wong D, Jergens AE. Oxidative stress, hormones, and effects of natural antioxidants on intestinal inflammation in inflammatory bowel disease. Front Endocrinol (Lausanne) 2023; 14:1217165. [PMID: 37701897 PMCID: PMC10493311 DOI: 10.3389/fendo.2023.1217165] [Citation(s) in RCA: 110] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 08/07/2023] [Indexed: 09/14/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic, relapsing gastrointestinal (GI) disorder characterized by intestinal inflammation. The etiology of IBD is multifactorial and results from a complex interplay between mucosal immunity, environmental factors, and host genetics. Future therapeutics for GI disorders, including IBD, that are driven by oxidative stress require a greater understanding of the cellular and molecular mechanisms mediated by reactive oxygen species (ROS). In the GI tract, oxidative stressors include infections and pro-inflammatory responses, which boost ROS generation by promoting the production of pro-inflammatory cytokines. Nuclear factor kappa B (NF-κB) and nuclear factor erythroid 2-related factor 2 (Nrf2) represent two important signaling pathways in intestinal immune cells that regulate numerous physiological processes, including anti-inflammatory and antioxidant activities. Natural antioxidant compounds exhibit ROS scavenging and increase antioxidant defense capacity to inhibit pro-oxidative enzymes, which may be useful in IBD treatment. In this review, we discuss various polyphenolic substances (such as resveratrol, curcumin, quercetin, green tea flavonoids, caffeic acid phenethyl ester, luteolin, xanthohumol, genistein, alpinetin, proanthocyanidins, anthocyanins, silymarin), phenolic compounds including thymol, alkaloids such as berberine, storage polysaccharides such as tamarind xyloglucan, and other phytochemicals represented by isothiocyanate sulforaphane and food/spices (such as ginger, flaxseed oil), as well as antioxidant hormones like melatonin that target cellular signaling pathways to reduce intestinal inflammation occurring with IBD.
Collapse
Affiliation(s)
- Dipak Kumar Sahoo
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| | - Romy M. Heilmann
- Department for Small Animals, Veterinary Teaching Hospital, College of Veterinary Medicine, University of Leipzig, Leipzig, SN, Germany
| | - Biswaranjan Paital
- Redox Regulation Laboratory, Department of Zoology, College of Basic Science and Humanities, Odisha University of Agriculture and Technology, Bhubaneswar, India
| | - Ashish Patel
- Department of Life Sciences, Hemchandracharya North Gujarat University, Patan, Gujarat, India
| | - Virendra Kumar Yadav
- Department of Life Sciences, Hemchandracharya North Gujarat University, Patan, Gujarat, India
| | - David Wong
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| | - Albert E. Jergens
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| |
Collapse
|
40
|
Huang C, Hao W, Wang X, Zhou R, Lin Q. Probiotics for the treatment of ulcerative colitis: a review of experimental research from 2018 to 2022. Front Microbiol 2023; 14:1211271. [PMID: 37485519 PMCID: PMC10358780 DOI: 10.3389/fmicb.2023.1211271] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 06/22/2023] [Indexed: 07/25/2023] Open
Abstract
Ulcerative colitis (UC) has become a worldwide public health problem, and the prevalence of the disease among children has been increasing. The pathogenesis of UC has not been elucidated, but dysbiosis of the gut microbiota is considered the main cause of chronic intestinal inflammation. This review focuses on the therapeutic effects of probiotics on UC and the potential mechanisms involved. In animal studies, probiotics have been shown to alleviate symptoms of UC, including weight loss, diarrhea, blood in the stool, and a shortened colon length, while also restoring intestinal microecological homeostasis, improving gut barrier function, modulating the intestinal immune response, and attenuating intestinal inflammation, thereby providing theoretical support for the development of probiotic-based microbial products as an adjunctive therapy for UC. However, the efficacy of probiotics is influenced by factors such as the bacterial strain, dose, and form. Hence, the mechanisms of action need to be investigated further. Relevant clinical trials are currently lacking, so the extension of animal experimental findings to clinical application requires a longer period of consideration for validation.
Collapse
Affiliation(s)
- Cuilan Huang
- Wuxi People’s Hospital Affiliated to Nanjing Medical University, Wuxi Children’s Hospital, Wuxi, China
| | - Wujuan Hao
- Department of Digestive, Affiliated Children’s Hospital of Jiangnan University, Wuxi, China
| | - Xuyang Wang
- Wuxi People’s Hospital Affiliated to Nanjing Medical University, Wuxi Children’s Hospital, Wuxi, China
| | - Renmin Zhou
- Department of Digestive, Affiliated Children’s Hospital of Jiangnan University, Wuxi, China
| | - Qiong Lin
- Wuxi People’s Hospital Affiliated to Nanjing Medical University, Wuxi Children’s Hospital, Wuxi, China
| |
Collapse
|
41
|
Guiducci L, Nicolini G, Forini F. Dietary Patterns, Gut Microbiota Remodeling, and Cardiometabolic Disease. Metabolites 2023; 13:760. [PMID: 37367916 DOI: 10.3390/metabo13060760] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 06/14/2023] [Accepted: 06/16/2023] [Indexed: 06/28/2023] Open
Abstract
The cardiovascular and metabolic disorders, collectively known as cardiometabolic disease (CMD), are high morbidity and mortality pathologies associated with lower quality of life and increasing health-care costs. The influence of the gut microbiota (GM) in dictating the interpersonal variability in CMD susceptibility, progression and treatment response is beginning to be deciphered, as is the mutualistic relation established between the GM and diet. In particular, dietary factors emerge as pivotal determinants shaping the architecture and function of resident microorganisms in the human gut. In turn, intestinal microbes influence the absorption, metabolism, and storage of ingested nutrients, with potentially profound effects on host physiology. Herein, we present an updated overview on major effects of dietary components on the GM, highlighting the beneficial and detrimental consequences of diet-microbiota crosstalk in the setting of CMD. We also discuss the promises and challenges of integrating microbiome data in dietary planning aimed at restraining CMD onset and progression with a more personalized nutritional approach.
Collapse
Affiliation(s)
- Letizia Guiducci
- CNR Institute of Clinical Physiology, Via Moruzzi 1, 56124 Pisa, Italy
| | | | - Francesca Forini
- CNR Institute of Clinical Physiology, Via Moruzzi 1, 56124 Pisa, Italy
| |
Collapse
|
42
|
Mimatsu H, Onoda A, Kazama T, Nishijima K, Shimoyama Y, Go S, Ueda K, Takahashi Y, Matsumoto T, Hayakawa M, Sato Y. Dedifferentiated fat cells administration ameliorates abnormal expressions of fatty acids metabolism-related protein expressions and intestinal tissue damage in experimental necrotizing enterocolitis. Sci Rep 2023; 13:8266. [PMID: 37217485 DOI: 10.1038/s41598-023-34156-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 04/25/2023] [Indexed: 05/24/2023] Open
Abstract
Neonatal necrotizing enterocolitis (NEC) is a serious disease of premature infants that necessitates intensive care and frequently results in life-threatening complications and high mortality. Dedifferentiated fat cells (DFATs) are mesenchymal stem cell-like cells derived from mature adipocytes. DFATs were intraperitoneally administrated to a rat NEC model, and the treatment effect and its mechanism were evaluated. The NEC model was created using rat pups hand fed with artificial milk, exposed to asphyxia and cold stress, and given oral lipopolysaccharides after cesarean section. The pups were sacrificed 96 h after birth for macroscopic histological examination and proteomics analysis. DFATs administration significantly improved the survival rate from 25.0 (vehicle group) to 60.6% (DFAT group) and revealed a significant reduction in macroscopical, histological, and apoptosis evaluation compared with the vehicle group. Additionally, the expression of C-C motif ligand 2 was significantly decreased, and that of interleukin-6 decreased in the DFAT group. DFAT administration ameliorated 93 proteins mainly related to proteins of fatty acid metabolism of the 436 proteins up-/down-regulated by NEC. DFATs improved mortality and restored damaged intestinal tissues in NEC, possibly by improving the abnormal expression of fatty acid-related proteins and reducing inflammation.
Collapse
Affiliation(s)
- Haruka Mimatsu
- Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, 65 Tsurumai-Cho Showa-Ku, Nagoya, 466-8550, Japan
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Atsuto Onoda
- Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, 65 Tsurumai-Cho Showa-Ku, Nagoya, 466-8550, Japan
- Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, Sanyo-Onoda, Yamaguchi, Japan
| | - Tomohiko Kazama
- Department of Functional Morphology, Division of Cell Regeneration and Transplantation, Nihon University School of Medicine, Tokyo, Japan
| | - Koji Nishijima
- Center for Perinatal, Maternal and Neonatal Medicine, Niigata University Medical and Dental Hospital, Niigata, Japan
| | - Yoshie Shimoyama
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shoji Go
- Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, 65 Tsurumai-Cho Showa-Ku, Nagoya, 466-8550, Japan
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kazuto Ueda
- Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, 65 Tsurumai-Cho Showa-Ku, Nagoya, 466-8550, Japan
| | - Yoshiyuki Takahashi
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Taro Matsumoto
- Department of Functional Morphology, Division of Cell Regeneration and Transplantation, Nihon University School of Medicine, Tokyo, Japan
| | - Masahiro Hayakawa
- Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, 65 Tsurumai-Cho Showa-Ku, Nagoya, 466-8550, Japan
| | - Yoshiaki Sato
- Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, 65 Tsurumai-Cho Showa-Ku, Nagoya, 466-8550, Japan.
| |
Collapse
|
43
|
Cuciniello R, Di Meo F, Filosa S, Crispi S, Bergamo P. The Antioxidant Effect of Dietary Bioactives Arises from the Interplay between the Physiology of the Host and the Gut Microbiota: Involvement of Short-Chain Fatty Acids. Antioxidants (Basel) 2023; 12:antiox12051073. [PMID: 37237938 DOI: 10.3390/antiox12051073] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/20/2023] [Accepted: 05/05/2023] [Indexed: 05/28/2023] Open
Abstract
The maintenance of redox homeostasis is associated with a healthy status while the disruption of this mechanism leads to the development of various pathological conditions. Bioactive molecules such as carbohydrates accessible to the microbiota (MACs), polyphenols, and polyunsaturated fatty acids (PUFAs) are food components best characterized for their beneficial effect on human health. In particular, increasing evidence suggests that their antioxidant ability is involved in the prevention of several human diseases. Some experimental data indicate that the activation of the nuclear factor 2-related erythroid 2 (Nrf2) pathway-the key mechanism in the maintenance of redox homeostasis-is involved in the beneficial effects exerted by the intake of PUFAs and polyphenols. However, it is known that the latter must be metabolized before becoming active and that the intestinal microbiota play a key role in the biotransformation of some ingested food components. In addition, recent studies, indicating the efficacy of the MACs, polyphenols, and PUFAs in increasing the microbial population with the ability to yield biologically active metabolites (e.g., polyphenol metabolites, short-chain fatty acids (SCFAs)), support the hypothesis that these factors are responsible for the antioxidant action on the physiology of the host. The underlying mechanisms through which MACs, polyphenols, and PUFAs might influence the redox status have not been fully elucidated, but based on the efficacy of SCFAs as Nrf2 activators, their contribution to the antioxidant efficacy of dietary bioactives cannot be excluded. In this review, we aimed to summarize the main mechanisms through which MACs, polyphenols, and PUFAs can modulate the host's redox homeostasis through their ability to directly or indirectly activate the Nrf2 pathway. We discuss their probiotic effects and the role played by the alteration of the metabolism/composition of the gut microbiota in the generation of potential Nrf2-ligands (e.g., SCFAs) in the host's redox homeostasis.
Collapse
Affiliation(s)
- Rossana Cuciniello
- Institute of Biosciences and BioResources-UOS Naples CNR, Via P. Castellino, 111-80131 Naples, Italy
- IRCCS Neuromed, 86077 Pozzilli, Italy
| | - Francesco Di Meo
- Institute of Biosciences and BioResources-UOS Naples CNR, Via P. Castellino, 111-80131 Naples, Italy
- Department of Medicine, Indiana University, Indianapolis, IN 46202, USA
| | - Stefania Filosa
- Institute of Biosciences and BioResources-UOS Naples CNR, Via P. Castellino, 111-80131 Naples, Italy
- IRCCS Neuromed, 86077 Pozzilli, Italy
| | - Stefania Crispi
- Institute of Biosciences and BioResources-UOS Naples CNR, Via P. Castellino, 111-80131 Naples, Italy
| | - Paolo Bergamo
- Institute of Biosciences and BioResources-UOS Naples CNR, Via P. Castellino, 111-80131 Naples, Italy
| |
Collapse
|
44
|
Haneishi Y, Furuya Y, Hasegawa M, Takemae H, Tanioka Y, Mizutani T, Rossi M, Miyamoto J. Polyunsaturated fatty acids-rich dietary lipid prevents high fat diet-induced obesity in mice. Sci Rep 2023; 13:5556. [PMID: 37019935 PMCID: PMC10076282 DOI: 10.1038/s41598-023-32851-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 04/03/2023] [Indexed: 04/07/2023] Open
Abstract
Diet is the primary factor affecting host nutrition and metabolism, with excess food intake, especially high-calorie diets, such as high-fat and high-sugar diets, causing an increased risk of obesity and related disorders. Obesity alters the gut microbial composition and reduces microbial diversity and causes changes in specific bacterial taxa. Dietary lipids can alter the gut microbial composition in obese mice. However, the regulation of gut microbiota and host energy homeostasis by different polyunsaturated fatty acids (PUFAs) in dietary lipids remains unknown. Here, we demonstrated that different PUFAs in dietary lipids improved host metabolism in high-fat diet (HFD)-induced obesity in mice. The intake of the different PUFA-enriched dietary lipids improved metabolism in HFD-induced obesity by regulating glucose tolerance and inhibiting colonic inflammation. Moreover, the gut microbial compositions were different among HFD and modified PUFA-enriched HFD-fed mice. Thus, we have identified a new mechanism underlying the function of different PUFAs in dietary lipids in regulating host energy homeostasis in obese conditions. Our findings shed light on the prevention and treatment of metabolic disorders by targeting the gut microbiota.
Collapse
Affiliation(s)
- Yuri Haneishi
- Department of Applied Biological Science, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Fuchu-shi, Tokyo, 183-8509, Japan
| | - Yuma Furuya
- Department of Applied Biological Science, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Fuchu-shi, Tokyo, 183-8509, Japan
| | - Mayu Hasegawa
- Department of Applied Biological Science, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Fuchu-shi, Tokyo, 183-8509, Japan
| | - Hitoshi Takemae
- Center for Infectious Diseases Epidemiology and Prevention Research: CEPiR, Tokyo University of Agriculture and Technology, Fuchu-shi, Tokyo, 183-8509, Japan
| | - Yuri Tanioka
- Department of International Food and Agricultural Science, Faculty of International Food and Agricultural Studies, Tokyo University of Agriculture, Setagaya-ku, Tokyo, 156-8502, Japan
| | - Tetsuya Mizutani
- Center for Infectious Diseases Epidemiology and Prevention Research: CEPiR, Tokyo University of Agriculture and Technology, Fuchu-shi, Tokyo, 183-8509, Japan
| | - Mauro Rossi
- Institute of Food Sciences, CNR, via Roma 64, 83100, Avellino, Italy
| | - Junki Miyamoto
- Department of Applied Biological Science, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Fuchu-shi, Tokyo, 183-8509, Japan.
| |
Collapse
|
45
|
Barrea L, Verde L, Auriemma RS, Vetrani C, Cataldi M, Frias-Toral E, Pugliese G, Camajani E, Savastano S, Colao A, Muscogiuri G. Probiotics and Prebiotics: Any Role in Menopause-Related Diseases? Curr Nutr Rep 2023; 12:83-97. [PMID: 36746877 PMCID: PMC9974675 DOI: 10.1007/s13668-023-00462-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/10/2023] [Indexed: 02/08/2023]
Abstract
PURPOSE OF REVIEW The aim of this review is to provide an overview of the menopause-related changes in microbiota and their role in the pathogenesis of menopause-related diseases. In addition, evidence on probiotic supplementation as a therapeutic strategy is discussed. RECENT FINDINGS The human microbiota is a complex community that lives in a mutualism relationship with the host. Menopause is associated with dysbiosis, and these changes in the composition of microbiota in different sites (gut, vaginal, and oral microbiota) might play a role in the pathogenesis of menopause-related diseases (i.e., osteoporosis, breast cancer, endometrial hyperplasia, periodontitis, and cardiometabolic diseases). The present review highlights the pivotal role of microbiota in postmenopausal women health, in particular it (a) may increase intestinal calcium absorption thus preventing osteoporosis, (b) is associated with reduced risk of breast cancer and type 1 endometrial hyperplasia, (c) reduces gingival inflammation and menopausal periodontitis, and (d) beneficially affects multiple cardiometabolic risk factors (i.e., obesity, inflammation, and blood glucose and lipid metabolism). However, whether oral probiotic supplementation might be used for the treatment of menopause-related dysbiosis requires further clarification.
Collapse
Affiliation(s)
- Luigi Barrea
- Dipartimento di Scienze Umanistiche, Centro Direzionale, Università Telematica Pegaso, Via Porzio, isola F2, 80143, Naples, Italy
- Centro Italiano per la cura e il Benessere del Paziente con Obesità (C.I.B.O), Department of Clinical Medicine and Surgery, Endocrinology Unit, University Medical School of Naples, Via Sergio Pansini 5, 80131, Naples, Italy
| | - Ludovica Verde
- Centro Italiano per la cura e il Benessere del Paziente con Obesità (C.I.B.O), Department of Clinical Medicine and Surgery, Endocrinology Unit, University Medical School of Naples, Via Sergio Pansini 5, 80131, Naples, Italy.
- Department of Public Health, University of Naples Federico II, Naples, Italy.
| | - Renata Simona Auriemma
- Dipartimento di Medicina Clinica e Chirurgia, Unità di Endocrinologia, Diabetologia e Andrologia, Università degli Studi di Napoli Federico II, Via Sergio Pansini 5, Naples, 80131, Italy
| | - Claudia Vetrani
- Centro Italiano per la cura e il Benessere del Paziente con Obesità (C.I.B.O), Department of Clinical Medicine and Surgery, Endocrinology Unit, University Medical School of Naples, Via Sergio Pansini 5, 80131, Naples, Italy
- Dipartimento di Medicina Clinica e Chirurgia, Unità di Endocrinologia, Diabetologia e Andrologia, Università degli Studi di Napoli Federico II, Via Sergio Pansini 5, Naples, 80131, Italy
| | - Mauro Cataldi
- Section of Pharmacology, Department of Neuroscience, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Evelyn Frias-Toral
- Universidad Católica Santiago de Guayaquil, Av. Pdte. Carlos Julio Arosemena Tola, Guayaquil, 090615, Ecuador
| | - Gabriella Pugliese
- Centro Italiano per la cura e il Benessere del Paziente con Obesità (C.I.B.O), Department of Clinical Medicine and Surgery, Endocrinology Unit, University Medical School of Naples, Via Sergio Pansini 5, 80131, Naples, Italy
- Dipartimento di Medicina Clinica e Chirurgia, Unità di Endocrinologia, Diabetologia e Andrologia, Università degli Studi di Napoli Federico II, Via Sergio Pansini 5, Naples, 80131, Italy
| | - Elisabetta Camajani
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166, Rome, Italy
| | - Silvia Savastano
- Centro Italiano per la cura e il Benessere del Paziente con Obesità (C.I.B.O), Department of Clinical Medicine and Surgery, Endocrinology Unit, University Medical School of Naples, Via Sergio Pansini 5, 80131, Naples, Italy
- Dipartimento di Medicina Clinica e Chirurgia, Unità di Endocrinologia, Diabetologia e Andrologia, Università degli Studi di Napoli Federico II, Via Sergio Pansini 5, Naples, 80131, Italy
| | - Annamaria Colao
- Centro Italiano per la cura e il Benessere del Paziente con Obesità (C.I.B.O), Department of Clinical Medicine and Surgery, Endocrinology Unit, University Medical School of Naples, Via Sergio Pansini 5, 80131, Naples, Italy
- Dipartimento di Medicina Clinica e Chirurgia, Unità di Endocrinologia, Diabetologia e Andrologia, Università degli Studi di Napoli Federico II, Via Sergio Pansini 5, Naples, 80131, Italy
- Cattedra Unesco "Educazione alla salute e allo sviluppo sostenibile", University Federico II, Naples, Italy
| | - Giovanna Muscogiuri
- Centro Italiano per la cura e il Benessere del Paziente con Obesità (C.I.B.O), Department of Clinical Medicine and Surgery, Endocrinology Unit, University Medical School of Naples, Via Sergio Pansini 5, 80131, Naples, Italy
- Dipartimento di Medicina Clinica e Chirurgia, Unità di Endocrinologia, Diabetologia e Andrologia, Università degli Studi di Napoli Federico II, Via Sergio Pansini 5, Naples, 80131, Italy
- Cattedra Unesco "Educazione alla salute e allo sviluppo sostenibile", University Federico II, Naples, Italy
| |
Collapse
|
46
|
Deandra FA, Ketherin K, Rachmasari R, Sulijaya B, Takahashi N. Probiotics and metabolites regulate the oral and gut microbiome composition as host modulation agents in periodontitis: A narrative review. Heliyon 2023; 9:e13475. [PMID: 36820037 PMCID: PMC9937986 DOI: 10.1016/j.heliyon.2023.e13475] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 01/25/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
Periodontitis is defined as an oral bacterial dysbiosis-induced persistent inflammation on dental supporting tissue resulting in periodontal tissue breakdown and alveolar bone destruction. The disease is initiated by the interaction between periodontopathogens and the host immune system. Its development and severity can be associated with several systemic diseases, such as cardiovascular disease (CVD), diabetes mellitus, and rheumatoid arthritis (RA). Moreover, the latest research has suggested that the oral and gut microbiome hypothesis lays the oral and systemic connection mechanism. Bacterial homeostasis and restoration in the oral cavity and intestine become therapeutics concepts. Concerning the treatment of periodontitis, a local inflammatory condition, prolonged systemic administration of antibiotics is no longer recommended due to bacterial resistance issues. Probiotics and several bioactive metabolites have been widely investigated to address the needs of host modulation therapy in periodontitis. Evidence suggests that the use of probiotics helps downregulate the inflammation process through the regulation of toll-like receptor 4 (TLR4) and the production of fatty acid, targeting reactive oxygen species (ROS). In brief, several herbals have anti-inflammatory properties by inhibiting pro-inflammatory cytokines and mediators, including mitogen-activated protein kinase (MAPK) and nuclear factor kappa B (NF-κB). Consistently, improvement of periodontal pocket depth (PPD) and gingival index (GI) was seen in a group given melatonin as an adjunct treatment. In all, this review will highlight host modulation agents regarding periodontitis therapy, plausible mechanisms on how probiotics and metabolites work on periodontal restoration, and their reported studies. Limitations given by published studies will be elaborated, while future directions will be proposed.
Collapse
Affiliation(s)
- Fathia Agzarine Deandra
- Postgraduate Program in Periodontology, Department of Periodontology, Universitas Indonesia, Jakarta, Indonesia
| | - Ketherin Ketherin
- Postgraduate Program in Periodontology, Department of Periodontology, Universitas Indonesia, Jakarta, Indonesia
| | - Rieska Rachmasari
- Postgraduate Program in Periodontology, Department of Periodontology, Universitas Indonesia, Jakarta, Indonesia
| | - Benso Sulijaya
- Department of Periodontology, Universitas Indonesia, Jakarta, Indonesia,Dental Division, Universitas Indonesia Hospital, Depok, West Java, Indonesia,Corresponding author. Department of Periodontology, Universitas Indonesia, Jakarta, Indonesia.
| | - Naoki Takahashi
- Division of Periodontology, Graduate School of Medical and Dental Sciences, Niigata University, Japan
| |
Collapse
|
47
|
Rinninella E, Tohumcu E, Raoul P, Fiorani M, Cintoni M, Mele MC, Cammarota G, Gasbarrini A, Ianiro G. The role of diet in shaping human gut microbiota. Best Pract Res Clin Gastroenterol 2023; 62-63:101828. [PMID: 37094913 DOI: 10.1016/j.bpg.2023.101828] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 02/13/2023] [Accepted: 02/15/2023] [Indexed: 04/26/2023]
Abstract
Gut microbiota plays a fundamental role within human health, and exerts key functions within the human body. Diet is one of the most powerful modulators of gut microbiota functions and composition. This complex interplay involves also the immune system and the intestinal barrier, highlighting the central role of diet in the pathogenesis and treatment of multiple diseases. In this review article we will paint the landscape of the effects of specific dietary nutrients, and of the detrimental or beneficial outcomes of different dietary patterns, on the composition of human gut microbiota. Moreover, we will discuss the potential application of diet as a therapeutic modulator of gut microbiota, including cutting-edge ways of exploitation, including the use of dietary components as adjuvants to promote microbial engraftment after fecal microbiota transplantation, or personalized nutritional approaches, targeted to the patient microbiome.
Collapse
Affiliation(s)
- Emanuele Rinninella
- Department of Medical and Surgical Sciences, Clinical Nutrition Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Ege Tohumcu
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy; Department of Medical and Surgical Sciences, Digestive Disease Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Pauline Raoul
- Department of Medical and Surgical Sciences, Clinical Nutrition Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Marcello Fiorani
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy; Department of Medical and Surgical Sciences, Digestive Disease Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Marco Cintoni
- Department of Medical and Surgical Sciences, Clinical Nutrition Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Maria Cristina Mele
- Department of Medical and Surgical Sciences, Clinical Nutrition Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giovanni Cammarota
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy; Department of Medical and Surgical Sciences, Digestive Disease Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Antonio Gasbarrini
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy; Department of Medical and Surgical Sciences, Digestive Disease Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Gianluca Ianiro
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy; Department of Medical and Surgical Sciences, Digestive Disease Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy.
| |
Collapse
|
48
|
Manoharan N, Parasuraman R, Jayamurali D, Govindarajulu SN. The therapeutic role of microbial metabolites in human health and diseases. RECENT ADVANCES AND FUTURE PERSPECTIVES OF MICROBIAL METABOLITES 2023:1-38. [DOI: 10.1016/b978-0-323-90113-0.00002-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
49
|
Quaranta A, Revol-Cavalier J, Wheelock CE. The octadecanoids: an emerging class of lipid mediators. Biochem Soc Trans 2022; 50:1569-1582. [PMID: 36454542 PMCID: PMC9788390 DOI: 10.1042/bst20210644] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 10/11/2022] [Accepted: 10/24/2022] [Indexed: 10/27/2023]
Abstract
Oxylipins are enzymatic and non-enzymatic metabolites of mono- or polyunsaturated fatty acids that encompass potent lipid mediators including the eicosanoids and docosanoids. Previously considered of low interest and often dismissed as 'just fat', octadecanoid oxylipins have only recently begun to be recognized as lipid mediators in humans. In the last few years, these compounds have been found to be involved in the mediation of multiple biological processes related to nociception, tissue modulation, cell proliferation, metabolic regulation, inflammation, and immune regulation. At the same time, the study of octadecanoids is hampered by a lack of standardization in the field, a paucity of analytical standards, and a lack of domain expertise. These issues have collectively limited the investigation of the biosynthesis and bioactivity of octadecanoids. Here, we present an overview of the primary enzymatic pathways for the oxidative metabolism of 18-carbon fatty acids in humans and of the current knowledge of the major biological activity of the resulting octadecanoids. We also propose a systematic nomenclature system based upon that used for the eicosanoids in order to avoid ambiguities and resolve multiple designations for the same octadecanoid. The aim of this review is to provide an initial framework for the field and to assist in its standardization as well as to increase awareness of this class of compounds in order to stimulate research into this interesting group of lipid mediators.
Collapse
Affiliation(s)
- Alessandro Quaranta
- Unit of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Johanna Revol-Cavalier
- Unit of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden
- Larodan Research Laboratory, Karolinska Institutet, 171 65 Stockholm, Sweden
| | - Craig E. Wheelock
- Unit of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden
- Department of Respiratory Medicine and Allergy, Karolinska University Hospital, 171 76 Stockholm, Sweden
- Gunma University Initiative for Advanced Research (GIAR), Gunma University, Maebashi, Gunma 371-8511, Japan
| |
Collapse
|
50
|
Lund PJ, Gates LA, Leboeuf M, Smith SA, Chau L, Lopes M, Friedman ES, Saiman Y, Kim MS, Shoffler CA, Petucci C, Allis CD, Wu GD, Garcia BA. Stable isotope tracing in vivo reveals a metabolic bridge linking the microbiota to host histone acetylation. Cell Rep 2022; 41:111809. [PMID: 36516747 PMCID: PMC9994635 DOI: 10.1016/j.celrep.2022.111809] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 03/09/2022] [Accepted: 11/17/2022] [Indexed: 12/14/2022] Open
Abstract
The gut microbiota influences acetylation on host histones by fermenting dietary fiber into butyrate. Although butyrate could promote histone acetylation by inhibiting histone deacetylases, it may also undergo oxidation to acetyl-coenzyme A (CoA), a necessary cofactor for histone acetyltransferases. Here, we find that epithelial cells from germ-free mice harbor a loss of histone H4 acetylation across the genome except at promoter regions. Using stable isotope tracing in vivo with 13C-labeled fiber, we demonstrate that the microbiota supplies carbon for histone acetylation. Subsequent metabolomic profiling revealed hundreds of labeled molecules and supported a microbial contribution to host fatty acid metabolism, which declined in response to colitis and correlated with reduced expression of genes involved in fatty acid oxidation. These results illuminate the flow of carbon from the diet to the host via the microbiota, disruptions to which may affect energy homeostasis in the distal gut and contribute to the development of colitis.
Collapse
Affiliation(s)
- Peder J Lund
- Department of Biochemistry and Biophysics, Penn Epigenetics Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Leah A Gates
- Laboratory of Chromatin Biology and Epigenetics, The Rockefeller University, New York, NY 10065, USA
| | - Marylene Leboeuf
- Laboratory of Chromatin Biology and Epigenetics, The Rockefeller University, New York, NY 10065, USA
| | - Sarah A Smith
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lillian Chau
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mariana Lopes
- Department of Biochemistry and Biophysics, Penn Epigenetics Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Elliot S Friedman
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yedidya Saiman
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Min Soo Kim
- Metabolomics Core, Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Clarissa A Shoffler
- Metabolomics Core, Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Christopher Petucci
- Metabolomics Core, Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - C David Allis
- Laboratory of Chromatin Biology and Epigenetics, The Rockefeller University, New York, NY 10065, USA
| | - Gary D Wu
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Benjamin A Garcia
- Department of Biochemistry and Biophysics, Penn Epigenetics Institute, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|