1
|
Ur Rahman Shah SA, Hao Y, Tang B, Ahmad M, He D, Nabi G, Zheng J, Wan X, Wang C, Wang K. The association of seasonal dietary shift with fecal metabolome and microbiota in the captive Yangtze finless porpoise (Neophocaena asiaeorientalis asiaeorientalis). ENVIRONMENTAL RESEARCH 2025; 271:121082. [PMID: 39929417 DOI: 10.1016/j.envres.2025.121082] [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: 09/17/2024] [Revised: 02/06/2025] [Accepted: 02/07/2025] [Indexed: 02/18/2025]
Abstract
The gut microbiota can act as a buffer against changes in energy and food availability and adapt plastically to fluctuations in the host's diet. However, it is unknown how changes in the gut microbiome with the seasons impact microbial metabolism and the accessibility of nutrients to hosts. The study utilized 16S rRNA and UHPLC-MS/MS approaches to examine seasonal fecal metabolome variations in the captive Yangtze finless porpoises (YFPs) to determine if these variations are linked to nutrient intake or gut microbiome composition changes. The YFPs were mostly fed a frozen and live fish diet, with different food intakes yearly. We found that gut microbial diversity remained constant, but community structure varied seasonally. Firmicutes and Cyanobacteria were higher in winter, Actinobacteria in spring and fall, and proteobacteria in summer. The genus Paeniclostridium was significantly higher in the spring season, Romboutsia and Clostridium_sensu_stricto_13 were significantly higher in the summer, while Terrisporobacter and Macrococcus were significantly higher in the fall group. The study reported that seasonal dietary variation significantly impacted the fecal metabolome by affecting the metabolism, including energy, amino acid, carbohydrate, and nucleotide metabolism of the captive YFP. Moreover, significant correlations between metabolome and microbiome were found, and these correlations may indicate that the captive YFP has adapted to cope with dietary variations and enhance energy acquisition. These findings improve our knowledge of the link between microbiota, diet, metabolites, and the physiology of the host and suggest that gut microbial populations may adapt continuously to changes in diet.
Collapse
Affiliation(s)
- Syed Ata Ur Rahman Shah
- Key Laboratory of Aquatic Biodiversity and Conservation, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China; University of Chinese Academy of Sciences, Beijing, China
| | - Yujiang Hao
- Key Laboratory of Aquatic Biodiversity and Conservation, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China; National Aquatic Biological Resource Center, NABRC, Wuhan, 430072, China.
| | - Bin Tang
- Key Laboratory of Aquatic Biodiversity and Conservation, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China; University of Chinese Academy of Sciences, Beijing, China; National Aquatic Biological Resource Center, NABRC, Wuhan, 430072, China
| | - Maaz Ahmad
- Key Laboratory of Aquatic Biodiversity and Conservation, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China; University of Chinese Academy of Sciences, Beijing, China
| | - Dekui He
- Key Laboratory of Aquatic Biodiversity and Conservation, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China; National Aquatic Biological Resource Center, NABRC, Wuhan, 430072, China
| | - Ghulam Nabi
- Department of Zoology, Institute of Molecular Biology and Biotechnology, University of Lahore, Pakistan
| | - Jinsong Zheng
- Key Laboratory of Aquatic Biodiversity and Conservation, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China; National Aquatic Biological Resource Center, NABRC, Wuhan, 430072, China
| | - Xiaoling Wan
- Key Laboratory of Aquatic Biodiversity and Conservation, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China; National Aquatic Biological Resource Center, NABRC, Wuhan, 430072, China
| | - Chaoqun Wang
- Key Laboratory of Aquatic Biodiversity and Conservation, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China; National Aquatic Biological Resource Center, NABRC, Wuhan, 430072, China
| | - Kexiong Wang
- Key Laboratory of Aquatic Biodiversity and Conservation, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China; National Aquatic Biological Resource Center, NABRC, Wuhan, 430072, China
| |
Collapse
|
2
|
Saadh MJ, Ahmed HH, Kareem RA, Sanghvi G, Ganesan S, Agarwal M, Kaur P, Taher WM, Alwan M, Jawad MJ, Hamad AK. Short-chain fatty acids in Huntington's disease: Mechanisms of action and their therapeutic implications. Pharmacol Biochem Behav 2025; 249:173972. [PMID: 39983928 DOI: 10.1016/j.pbb.2025.173972] [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: 12/03/2024] [Revised: 02/10/2025] [Accepted: 02/14/2025] [Indexed: 02/23/2025]
Abstract
Huntington's disease (HD) is a progressive neurodegenerative disorder characterized by motor dysfunction, cognitive decline, and emotional instability, primarily resulting from the abnormal accumulation of mutant huntingtin protein. Growing research highlights the role of intestinal microbiota and their metabolites, particularly short-chain fatty acids (SCFAs), in modulating HD progression. SCFAs, including acetate, propionate, and butyrate, are produced by gut bacteria through dietary fiber fermentation and are recognized for their neuroprotective properties. Evidence suggests that SCFAs regulate neuroinflammation, neuronal communication, and metabolic functions within the central nervous system (CNS). In HD, these compounds may support neuronal health, reduce oxidative stress, and enhance blood-brain barrier (BBB) integrity. Their mechanisms of action involve binding to G-protein-coupled receptors (GPCRs) and modulating gene expression through epigenetic pathways, underscoring their therapeutic potential. This analysis examines the significance of SCFAs in HD, emphasizing the gut-brain axis and the benefits of dietary interventions aimed at modifying gut microbiota composition and promoting SCFA production. Further research into these pathways may pave the way for novel HD management strategies and improved therapeutic outcomes.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman 11831, Jordan.
| | | | | | - Gaurav Sanghvi
- Marwadi University Research Center, Department of Microbiology, Faculty of Science, Marwadi University, Rajkot 360003, Gujarat, India
| | - Subbulakshmi Ganesan
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Mohit Agarwal
- Department of Pharmaceutical Chemistry, NIMS Institute of Pharmacy, NIMS University, Rajasthan, Jaipur,302131, India
| | - Parjinder Kaur
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali 140307, Punjab, India
| | - Waam Mohammed Taher
- College of Nursing, National University of Science and Technology, Dhi Qar, Iraq
| | | | | | | |
Collapse
|
3
|
Rong Y, Zhang G, Ye W, Qi L, Hao X, Li X, Zhang W, Chao Y, Gu S. Uncovering the Effects and Molecular Mechanisms of Shaoyao Decoction Against Colorectal Cancer Using Network Pharmacology Analysis Coupled With Experimental Validation and Gut Microbiota Analysis. Cancer Med 2025; 14:e70813. [PMID: 40119640 PMCID: PMC11928771 DOI: 10.1002/cam4.70813] [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: 08/19/2024] [Revised: 03/12/2025] [Accepted: 03/13/2025] [Indexed: 03/24/2025] Open
Abstract
BACKGROUND Chronic gut inflammation and dysbiosis contribute significantly to colorectal cancer (CRC) development. Shaoyao decoction (SYD) is a well-established Chinese medicine prescription. Besides ameliorating CRC via anti-inflammatory effects, SYD modulates gut microbiota (GM) to improve inflammatory responses in ulcerative colitis (UC). However, whether and how SYD suppresses CRC by regulating GM remains largely unknown. METHODS SD rats were orally administered SYD for 7 days to obtain medicated serum. We utilized liquid chromatography-mass spectrometry (LC-MS) analysis, GeneCards, DisGeNET, and SwissTargetPrediction databases to analyze blank and SYD-medicated rat serum, comparing the findings with those of SYD aqueous extract in previous studies to identify SYD circulating compounds/components with predictable target genes. Using network pharmacology, the potential active compounds and corresponding hub genes associated with modulating GM to suppress CRC were selected for molecular docking. In vivo experiments, a CRC transplantation tumor model was established in BALB/c mice using CT26 cells, with SYD gavage for 14 days. To investigate the mechanism of SYD-regulated GM against CRC, HE and IHC staining, Western blotting, and 16S rRNA sequencing were employed. RESULTS LC-MS identified 26 SYD compounds with computationally predicted target genes. Network pharmacology prioritized 13 compounds targeting 8 inflammation/immunity-related genes (IL-17/TNF pathways), validated by molecular docking. In vivo experiments, SYD dose-dependently suppressed tumor growth (p < 0.05, medium/high doses), as confirmed by HE staining and IHC analysis of Ki-67. Notably, SYD potentially delayed CRC liver metastasis and alleviated hepatic injury in tumor-bearing mice. Western blotting demonstrated SYD's inhibition of the IL-17/TNF/NF-κB axis, aligning with computational predictions. 16S rRNA sequencing revealed SYD-enriched Akkermansia and GM structural shifts, mechanistically linking microbiota remodeling to anti-tumor efficacy. CONCLUSIONS SYD combats CRC via dual modulation of IL-17/TNF/NF-κB signaling and GM ecosystems (e.g., Akkermansia enrichment). This microbiota-immune crosstalk positions SYD as a potential adjunct to conventional therapies, particularly for CRC patients with dysbiosis.
Collapse
Affiliation(s)
- Yaojun Rong
- Shenzhen Bao'an Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Guiyu Zhang
- Shenzhen Bao'an Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Wenhao Ye
- The Seventh Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Linhua Qi
- Shenzhen Bao'an Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Xiaojiang Hao
- Shenzhen Bao'an Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Xiaolin Li
- Shenzhen Bao'an Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Wuhong Zhang
- Shenzhen Bao'an Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Yangfa Chao
- Shenzhen Bao'an Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Shaodong Gu
- Shenzhen Bao'an Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| |
Collapse
|
4
|
García G, Carlin M, Cano RDJ. Holobiome Harmony: Linking Environmental Sustainability, Agriculture, and Human Health for a Thriving Planet and One Health. Microorganisms 2025; 13:514. [PMID: 40142407 PMCID: PMC11945859 DOI: 10.3390/microorganisms13030514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 02/14/2025] [Accepted: 02/21/2025] [Indexed: 03/28/2025] Open
Abstract
The holobiome is an interconnected network of microbial ecosystems spanning soil, plants, animals, humans, and the environment. Microbial interactions drive nutrient cycling, pathogen suppression, and climate regulation. Soil microbiomes facilitate carbon sequestration and enhance soil fertility, while marine microbiomes contribute to carbon capture and climate stability. However, industrial agriculture, extensive herbicide use, antibiotic overuse, and climate change threaten microbial diversity, leading to ecosystem and health disruptions. Probiotic interventions help to restore microbial balance. In human health, probiotics support gut microbiota diversity, reduce inflammation, and regulate metabolism. In agriculture, soil probiotics enhance microbial diversity, improve nutrient cycling, and degrade contaminants, increasing crop yields and soil health. Case studies show that microbial inoculants effectively remediate degraded soils and enhance nutrient uptake. Artificial intelligence is transforming microbiome research by enabling predictive modeling, precision probiotic design, and microbial consortia optimization. Interdisciplinary collaboration and supportive policies are essential for restoring microbial equilibria, ensuring ecosystem resilience, and promoting long-term sustainability. The integration of artificial intelligence, clinical research, and sustainable practices is crucial for advancing holobiome science. The holobiome framework underscores the need for interdisciplinary collaboration to address global challenges, bridging environmental sustainability, agriculture, and public health for a resilient future.
Collapse
Affiliation(s)
- Gissel García
- Pathology Department, Hospital Hermanos Ameijeiras, La Habana 10400, Cuba;
| | | | - Raul de Jesus Cano
- Biological Sciences Department, California Polytechnic State University, San Luis Obispo, CA 93407, USA
- Chauvell, LLC, San Luis Obispo, CA 93401, USA
| |
Collapse
|
5
|
Samrit T, Osotprasit S, Chaiwichien A, Suksomboon P, Chansap S, Suthisintong T, Changklungmoa N, Kueakhai P. Microbial effects of cold-pressed Sacha inchi oil supplementation in rats. PLoS One 2025; 20:e0319066. [PMID: 39977445 PMCID: PMC11841868 DOI: 10.1371/journal.pone.0319066] [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: 06/20/2024] [Accepted: 01/26/2025] [Indexed: 02/22/2025] Open
Abstract
Oil supplements have various benefits for metabolism, particularly Sacha inchi oil (SI), which is rich in polyunsaturated fatty acids (PUFAs) such as ω-3 and fat-soluble vitamins. However, the impacts of oil supplements on gut health remain unclear. The aim of this study was to compare the effects of an SI supplement with those of lard oil (LO), known for its high saturated fatty acid content, and a normal diet on gut health in male Sprague Dawley rats for 12 consecutive weeks. Fecal DNA was used to assess gut microbiota diversity and species abundance, diversity, and function prediction. Colon tissue from each rat was examined for colon crypt depth and histology. Rats administered the LO supplement exhibited higher dysbiosis than those administered the SI supplement, with the LO supplement influencing the relative abundance of various bacteria at the genus level. A KEGG analysis was conducted to examine the effects on metabolic pathways, revealing that the SI supplement promoted carbohydrate metabolism while reducing immune system activity. In contrast, the LO supplement increased replication, repair, and translation activities. A histological analysis of the colon tissues showed no significant alterations in crypt depth or lesions in all groups, indicating that neither supplement induced adverse structural changes in the gut. The results of this study suggest that SI supplementation modulates the gut microbiota, thereby enhancing gut health and metabolic function.
Collapse
Affiliation(s)
- Tepparit Samrit
- Food Bioactive Compounds Research Unit and Faculty of Allied Health Sciences, Burapha University, Chonburi, Thailand
| | - Supawadee Osotprasit
- Food Bioactive Compounds Research Unit and Faculty of Allied Health Sciences, Burapha University, Chonburi, Thailand
| | - Athit Chaiwichien
- Food Bioactive Compounds Research Unit and Faculty of Allied Health Sciences, Burapha University, Chonburi, Thailand
| | - Phawiya Suksomboon
- Food Bioactive Compounds Research Unit and Faculty of Allied Health Sciences, Burapha University, Chonburi, Thailand
| | - Supanan Chansap
- Food Bioactive Compounds Research Unit and Faculty of Allied Health Sciences, Burapha University, Chonburi, Thailand
| | - Thitikul Suthisintong
- Food Bioactive Compounds Research Unit and Faculty of Allied Health Sciences, Burapha University, Chonburi, Thailand
| | - Narin Changklungmoa
- Food Bioactive Compounds Research Unit and Faculty of Allied Health Sciences, Burapha University, Chonburi, Thailand
| | - Pornanan Kueakhai
- Food Bioactive Compounds Research Unit and Faculty of Allied Health Sciences, Burapha University, Chonburi, Thailand
| |
Collapse
|
6
|
Wada T, Senokuchi T, Shi Y, Furusho T, Morita Y, Sarie M, Hanatani S, Fukuda K, Ishii N, Matsumura T, Fujiwara Y, Komohara Y, Araki E, Kubota N. Orally administrated acetate inhibits atherosclerosis progression through AMPK activation via GPR43 in plaque macrophages. Atherosclerosis 2025; 401:119088. [PMID: 39705906 DOI: 10.1016/j.atherosclerosis.2024.119088] [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: 08/15/2023] [Revised: 12/05/2024] [Accepted: 12/10/2024] [Indexed: 12/23/2024]
Abstract
BACKGROUND AND AIMS Oral administration of acetic acid, a short-chain fatty acid, has been shown to efficiently reduce obesity and insulin resistance in both experimental animals and humans. The anti-atherosclerotic effect of acetate is expected owing to its anti-inflammatory and anti-oxidative stress characteristics; however, this remains to be fully understood. METHODS For 12 weeks, apolipoprotein E-deficient mice were administered 0.6 % sodium acetate water or vehicle water. Plaque formation and progression were investigated using histological analysis of dissected aortic root sections. Flow cytometry and gene expression analyses were employed to assess plaque macrophage characteristics and functional states. In vitro tests were performed on mouse peritoneal primary macrophages and bone marrow-derived macrophages isolated from wild-type or GPR43-deficient mice. RESULTS Atherosclerotic plaque formation was inhibited in acetate-treated ApoE-deficient mice, and AMPK activation was directly validated in plaque macrophages. Acetate inhibited macrophage proliferation, reactive oxygen species production, and pro-inflammatory molecule expression, all of which were reversed by AMPK inhibition. Bone marrow transplantation study revealed the role of GPR43-mediated AMPK activation by acetic acid in anti-atherosclerotic effect. CONCLUSIONS Oral acetate administration suppressed arteriosclerosis formation and progression in ApoE-deficient mice. Acetate inhibited macrophage proliferation, inflammatory cytokine release, and reactive oxygen species production via GPR43-mediated AMPK activation in macrophages, ameliorating plaque formation and progression.
Collapse
Affiliation(s)
- Toshiaki Wada
- Department of Metabolic Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Takafumi Senokuchi
- Department of Metabolic Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan.
| | - Yudan Shi
- Department of Metabolic Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Tatsuya Furusho
- Department of Metabolic Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Yutaro Morita
- Department of Metabolism, Amakusa Medical Center, Kumamoto, Japan
| | - Maeda Sarie
- Department of Metabolic Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Satoko Hanatani
- Department of Metabolic Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Kazuki Fukuda
- Department of Metabolic Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Norio Ishii
- Department of Metabolism, Kumamoto City Hospital, Kumamoto, Japan
| | - Takeshi Matsumura
- Department of Metabolic Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Yukio Fujiwara
- Department of Cell Pathology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Yoshihiro Komohara
- Department of Cell Pathology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Eiichi Araki
- Department of Metabolic Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan; Research Center for Health and Sports Science, Kumamoto Health Science University, Kumamoto, Japan; Kikuchi Medical Association Hospital, Kumamoto, Japan
| | - Naoto Kubota
- Department of Metabolic Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
7
|
Wang T, Holscher HD, Maslov S, Hu FB, Weiss ST, Liu YY. Predicting metabolite response to dietary intervention using deep learning. Nat Commun 2025; 16:815. [PMID: 39827177 PMCID: PMC11742956 DOI: 10.1038/s41467-025-56165-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 01/10/2025] [Indexed: 01/22/2025] Open
Abstract
Due to highly personalized biological and lifestyle characteristics, different individuals may have different metabolite responses to specific foods and nutrients. In particular, the gut microbiota, a collection of trillions of microorganisms living in the gastrointestinal tract, is highly personalized and plays a key role in the metabolite responses to foods and nutrients. Accurately predicting metabolite responses to dietary interventions based on individuals' gut microbial compositions holds great promise for precision nutrition. Existing prediction methods are typically limited to traditional machine learning models. Deep learning methods dedicated to such tasks are still lacking. Here we develop a method McMLP (Metabolite response predictor using coupled Multilayer Perceptrons) to fill in this gap. We provide clear evidence that McMLP outperforms existing methods on both synthetic data generated by the microbial consumer-resource model and real data obtained from six dietary intervention studies. Furthermore, we perform sensitivity analysis of McMLP to infer the tripartite food-microbe-metabolite interactions, which are then validated using the ground-truth (or literature evidence) for synthetic (or real) data, respectively. The presented tool has the potential to inform the design of microbiota-based personalized dietary strategies to achieve precision nutrition.
Collapse
Affiliation(s)
- Tong Wang
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Hannah D Holscher
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Center for Artificial Intelligence and Modeling, The Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Sergei Maslov
- Center for Artificial Intelligence and Modeling, The Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Frank B Hu
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
| | - Scott T Weiss
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Yang-Yu Liu
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
- Center for Artificial Intelligence and Modeling, The Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.
| |
Collapse
|
8
|
Wang X, Cui J, Gu Z, Guo L, Liu R, Guo Y, Qin N, Yang Y. Aged garlic oligosaccharides modulate host metabolism and gut microbiota to alleviate high-fat and high-cholesterol diet-induced atherosclerosis in ApoE -/- mice. Food Chem 2025; 463:141409. [PMID: 39326312 DOI: 10.1016/j.foodchem.2024.141409] [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: 08/08/2024] [Revised: 09/11/2024] [Accepted: 09/22/2024] [Indexed: 09/28/2024]
Abstract
Atherosclerosis (AS) is a cardiovascular disease caused by excessive accumulation of lipids in arterial walls. In this study, we developed an AS model in ApoE-/- mice using a high-fat, high-cholesterol diet and investigated the anti-AS mechanism of aged garlic oligosaccharides (AGOs) by focusing on the gut microbiota. Results revealed that AGOs exhibited significant anti-AS effects, reduced trimethylamine N-oxide levels from 349.9 to 189.2 ng/mL, and reduced aortic lipid deposition from 31.7 % to 9.5 %. AGOs significantly increased the levels of short-chain fatty acids in feces, in which acetic, propionic, and butyric acids were increased from 1.580, 0.364, and 0.469 mg/g to 2.233, 0.774, and 0.881 mg/g, respectively. An analysis of the gut microbiota indicated that AGOs restored alpha and beta diversity, decreased the Firmicutes/Bacteroidetes ratio, and promoted the dominance of the genus Akkermansia. A metagenomic analysis revealed that AGOs alleviated AS through the ABC transporter pathway and the lipopolysaccharide biosynthesis pathway.
Collapse
Affiliation(s)
- Xiaomin Wang
- Institute of Pharmaceutical and Food Engineering, Shanxi University of Chinese Medicine, Yuci 030619, China
| | - Jianglu Cui
- Institute of Pharmaceutical and Food Engineering, Shanxi University of Chinese Medicine, Yuci 030619, China
| | - Ziyao Gu
- Institute of Pharmaceutical and Food Engineering, Shanxi University of Chinese Medicine, Yuci 030619, China
| | - Lili Guo
- Institute of Pharmaceutical and Food Engineering, Shanxi University of Chinese Medicine, Yuci 030619, China
| | - Rui Liu
- Institute of Pharmaceutical and Food Engineering, Shanxi University of Chinese Medicine, Yuci 030619, China
| | - Yu Guo
- Shanxi Agricultural Products Quality and Safety Center, Taiyuan 030006, China
| | - Nan Qin
- Institute of Pharmaceutical and Food Engineering, Shanxi University of Chinese Medicine, Yuci 030619, China.
| | - Yukun Yang
- School of Life Science, Xinghuacun College (Shanxi Institute of Brewing Technology and Industry), Shanxi University, Taiyuan 030006, China.
| |
Collapse
|
9
|
Aljumaah RS, Salama AAK, Abdelrahman MM, Ayadi M, Caja G, Alshaikh MA, Al-Badwi MA, Matar AM. Exploring the Role of Salt Supplementation on Milk Composition, Fatty Acids, and Insulin Response in Lactating Camels. Vet Sci 2025; 12:22. [PMID: 39852897 PMCID: PMC11768427 DOI: 10.3390/vetsci12010022] [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: 11/24/2024] [Revised: 12/31/2024] [Accepted: 01/03/2025] [Indexed: 01/26/2025] Open
Abstract
Camel milk is a valuable food source with unique nutritional properties and potential health benefits. This study investigated the influence of high dietary salt on milk composition and fatty acid (FA) profile as well as insulin regulation in dairy camels. Twelve multiparous female camels were used in a crossover design with two treatments: control concentrate (CON; 1.3% salt) and high-salt concentrate (SAL; 3.9% salt). Each period lasted 3 weeks, with camels switched between treatments in the second period. The measured variables included milk yield, milk composition, blood metabolites, and insulin levels in blood and milk. The SAL group exhibited higher (p < 0.01) water consumption. Nevertheless, milk yield and composition (fat, protein, lactose) remained unaffected. Notably, SAL camels had elevated blood insulin levels (p < 0.05) compared to the CON group, suggesting enhanced pancreatic activity possibly driven by osmotic balance changes. Milk FA profiles revealed a reduction in unsaturated fatty acids (UFA, p < 0.04), particularly monounsaturated (MUFA, p < 0.05) and odd-chain fatty acids (OCFA, p < 0.05). Furthermore, lipid quality indices such as the atherogenic index (p < 0.01) and the hypocholesterolemic/hypercholesterolemic FA ratio (h/H, p < 0.01) indicated a less favorable milk fat profile in the SAL group. These findings suggest that while moderate salt supplementation may not negatively impact milk yield in dairy camels, it alters both metabolic and milk fat composition variables, with potential implications for the nutritional quality of milk.
Collapse
Affiliation(s)
- Riyadh S. Aljumaah
- Department of Animal Production, College of Food and Agriculture Sciences, King Saud University, P.O. Box 2460, Riyadh 11451, Saudi Arabia; (R.S.A.); (M.M.A.); (M.A.); (M.A.A.); (M.A.A.-B.)
| | - Ahmed A. K. Salama
- Group of Research in Ruminants (G2R), Department of Animal and Food Sciences, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; (A.A.K.S.); (G.C.)
| | - Mutassim M. Abdelrahman
- Department of Animal Production, College of Food and Agriculture Sciences, King Saud University, P.O. Box 2460, Riyadh 11451, Saudi Arabia; (R.S.A.); (M.M.A.); (M.A.); (M.A.A.); (M.A.A.-B.)
| | - Moez Ayadi
- Department of Animal Production, College of Food and Agriculture Sciences, King Saud University, P.O. Box 2460, Riyadh 11451, Saudi Arabia; (R.S.A.); (M.M.A.); (M.A.); (M.A.A.); (M.A.A.-B.)
- Department of Animal Biotechnology, Higher Institute of Biotechnology of Beja, University of Jendouba, Beja 9000, Tunisia
| | - Gerardo Caja
- Group of Research in Ruminants (G2R), Department of Animal and Food Sciences, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; (A.A.K.S.); (G.C.)
| | - Mohammed A. Alshaikh
- Department of Animal Production, College of Food and Agriculture Sciences, King Saud University, P.O. Box 2460, Riyadh 11451, Saudi Arabia; (R.S.A.); (M.M.A.); (M.A.); (M.A.A.); (M.A.A.-B.)
| | - Mohammed A. Al-Badwi
- Department of Animal Production, College of Food and Agriculture Sciences, King Saud University, P.O. Box 2460, Riyadh 11451, Saudi Arabia; (R.S.A.); (M.M.A.); (M.A.); (M.A.A.); (M.A.A.-B.)
| | - Abdulkareem M. Matar
- Department of Animal Production, College of Food and Agriculture Sciences, King Saud University, P.O. Box 2460, Riyadh 11451, Saudi Arabia; (R.S.A.); (M.M.A.); (M.A.); (M.A.A.); (M.A.A.-B.)
| |
Collapse
|
10
|
Zhan W, Li R, Xu X. The relationship between low-carbohydrate diet score, dietary macronutrient intake, and rheumatoid arthritis: results from NHANES 2011-2016. Clin Rheumatol 2025; 44:171-182. [PMID: 39680261 DOI: 10.1007/s10067-024-07269-9] [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: 09/03/2024] [Revised: 11/21/2024] [Accepted: 12/06/2024] [Indexed: 12/17/2024]
Abstract
BACKGROUND This study sought to determine if dietary macronutrient consumption and the low-carbohydrate diet (LCD) score were linked to rheumatoid arthritis (RA). METHODS Participants ≥ 20 years were analyzed from the National Health and Nutrition Examination Survey (NHANES) 2011-2016. LCD score was calculated by summing the 11 quantiles values of the percentages of energy derived from carbohydrate, protein, and fat. Weighted logistic regression, eXtreme Gradient Boosting (XGBoost), and Light Gradient Boosting Machine (LightGBM) models were used to explore the relationship between LCD score, dietary macronutrient intake, and RA. Propensity score matching (PSM) were applied for sensitivity analysis. RESULTS Ultimately, 8118 participants (RA: 499, without RA: 7619) were analyzed. After fully adjusting for confounders, a negative association was found between the LCD score and the presence of RA [OR (95% CI), 0.97 (0.96, 0.99)]. A higher LCD score was also negatively associated with a lower likelihood of RA based on a categorical model. Among macronutrients, participants in the third and fourth quartiles had significantly increased odds of RA compared with the lowest carbohydrate intake. Regarding protein intake, individuals in the highest quartile of percentage of energy from protein had a 46% lower presence of RA compared with the lowest reference group. The relative importance of the LCD score on RA was determined based on XGBoost and LightGBM models. Moreover, the association between the LCD score, dietary macronutrient intake, and RA presence remained substantial after PSM. CONCLUSIONS LCD score was negatively associated with odds of RA in US adults. Moreover, a correlation was found between a lower likelihood of RA and high protein, and low carbohydrate consumption. Key Points • A significant negative association was found between LCD score and RA presence. • Machine learning models revealed the LCD score was a significant predictor of the presence of RA. • Low carbohydrate intake and high protein intake were correlated with a lower odds of RA.
Collapse
Affiliation(s)
- Wenqiang Zhan
- Qingpu District Center for Disease Control and Prevention, Shanghai, 201799, China
| | - Ruiqiang Li
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Key Laboratory of Environment and Human Health, Hebei Medical University, Shijiazhuang, China
| | - Xingxing Xu
- Qingpu District Center for Disease Control and Prevention, Shanghai, 201799, China.
| |
Collapse
|
11
|
An Y, He L, Xu X, Piao M, Wang B, Liu T, Cao H. Gut microbiota in post-acute COVID-19 syndrome: not the end of the story. Front Microbiol 2024; 15:1500890. [PMID: 39777148 PMCID: PMC11703812 DOI: 10.3389/fmicb.2024.1500890] [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: 09/24/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
The coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), has led to major global health concern. However, the focus on immediate effects was assumed as the tip of iceberg due to the symptoms following acute infection, which was defined as post-acute COVID-19 syndrome (PACS). Gut microbiota alterations even after disease resolution and the gastrointestinal symptoms are the key features of PACS. Gut microbiota and derived metabolites disorders may play a crucial role in inflammatory and immune response after SARS-CoV-2 infection through the gut-lung axis. Diet is one of the modifiable factors closely related to gut microbiota and COVID-19. In this review, we described the reciprocal crosstalk between gut and lung, highlighting the participation of diet and gut microbiota in and after COVID-19 by destroying the gut barrier, perturbing the metabolism and regulating the immune system. Therefore, bolstering beneficial species by dietary supplements, probiotics or prebiotics and fecal microbiota transplantation (FMT) may be a novel avenue for COVID-19 and PACS prevention. This review provides a better understanding of the association between gut microbiota and the long-term consequences of COVID-19, which indicates modulating gut dysbiosis may be a potentiality for addressing this multifaceted condition.
Collapse
Affiliation(s)
| | | | | | | | | | - Tianyu Liu
- Tianjin Key Laboratory of Digestive Diseases, Department of Gastroenterology and Hepatology, Tianjin Institute of Digestive Diseases, National Key Clinical Specialty, General Hospital, Tianjin Medical University, Tianjin, China
| | - Hailong Cao
- Tianjin Key Laboratory of Digestive Diseases, Department of Gastroenterology and Hepatology, Tianjin Institute of Digestive Diseases, National Key Clinical Specialty, General Hospital, Tianjin Medical University, Tianjin, China
| |
Collapse
|
12
|
Altamura S, Lombardi F, Palumbo P, Cinque B, Ferri C, Del Pinto R, Pietropaoli D. The Evolving Role of Neutrophils and Neutrophil Extracellular Traps (NETs) in Obesity and Related Diseases: Recent Insights and Advances. Int J Mol Sci 2024; 25:13633. [PMID: 39769394 PMCID: PMC11727698 DOI: 10.3390/ijms252413633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 12/16/2024] [Accepted: 12/18/2024] [Indexed: 01/03/2025] Open
Abstract
Obesity is a chronic, multifactorial disease characterized by persistent low-grade tissue and systemic inflammation. Fat accumulation in adipose tissue (AT) leads to stress and dysfunctional adipocytes, along with the infiltration of immune cells, which initiates and sustains inflammation. Neutrophils are the first immune cells to infiltrate AT during high-fat diet (HFD)-induced obesity. Emerging evidence suggests that the formation and release of neutrophil extracellular traps (NETs) play a significant role in the progression of obesity and related diseases. Additionally, obesity is associated with an imbalance in gut microbiota and increased intestinal barrier permeability, resulting in the translocation of live bacteria, bacterial deoxyribonucleic acid (DNA), lipopolysaccharides (LPS), and pro-inflammatory cytokines into the bloodstream and AT, thereby contributing to metabolic inflammation. Recent research has also shown that short-chain fatty acids (SCFAs), produced by gut microbiota, can influence various functions of neutrophils, including their activation, migration, and the generation of inflammatory mediators. This review comprehensively summarizes recent advancements in understanding the role of neutrophils and NET formation in the pathophysiology of obesity and related disorders while also focusing on updated potential therapeutic approaches targeting NETs based on studies conducted in humans and animal models.
Collapse
Affiliation(s)
- Serena Altamura
- Department of Life, Health & Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (S.A.); (F.L.); (P.P.); (B.C.); (C.F.); (R.D.P.)
- Prevention and Translational Research—Dental Clinic, Center of Oral Diseases, 67100 L’Aquila, Italy
| | - Francesca Lombardi
- Department of Life, Health & Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (S.A.); (F.L.); (P.P.); (B.C.); (C.F.); (R.D.P.)
| | - Paola Palumbo
- Department of Life, Health & Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (S.A.); (F.L.); (P.P.); (B.C.); (C.F.); (R.D.P.)
| | - Benedetta Cinque
- Department of Life, Health & Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (S.A.); (F.L.); (P.P.); (B.C.); (C.F.); (R.D.P.)
| | - Claudio Ferri
- Department of Life, Health & Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (S.A.); (F.L.); (P.P.); (B.C.); (C.F.); (R.D.P.)
- Unit of Internal Medicine and Nephrology, San Salvatore Hospital, Center for Hypertension and Cardiovascular Prevention, 67100 L’Aquila, Italy
| | - Rita Del Pinto
- Department of Life, Health & Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (S.A.); (F.L.); (P.P.); (B.C.); (C.F.); (R.D.P.)
- Unit of Internal Medicine and Nephrology, San Salvatore Hospital, Center for Hypertension and Cardiovascular Prevention, 67100 L’Aquila, Italy
| | - Davide Pietropaoli
- Department of Life, Health & Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (S.A.); (F.L.); (P.P.); (B.C.); (C.F.); (R.D.P.)
- Prevention and Translational Research—Dental Clinic, Center of Oral Diseases, 67100 L’Aquila, Italy
| |
Collapse
|
13
|
Tang MY, Xie H, Tao JT, Zhang C, Luo YH, Zhang C, Peng SQ, Xie LX, Lv WB, Zhang C, Huang L. Pathophysiological relevance and therapeutic outlook of GPR43 in atherosclerosis. Biochem Cell Biol 2024; 102:418-429. [PMID: 39013204 DOI: 10.1139/bcb-2024-0053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2024] Open
Abstract
Atherosclerosis (AS) is an inflammatory arterial disorder that occurs due to the deposition of the excessive lipoprotein under the artery intima, mainly including low-density lipoprotein and other apolipoprotein B-containing lipoproteins. G protein-coupled receptors (GPCRs) play a crucial role in transmitting signals in physiological and pathophysiological conditions. GPCRs recognize inflammatory mediators, thereby serving as important players during chronic inflammatory processes. It has been demonstrated that free fatty acids can function as ligands for various GPCRs, such as free fatty acid receptor (FFAR)1/GPR40, FFAR2/GPR43, FFAR3/GPR41, FFAR4/GPR120, and the lipid metabolite binding glucose-dependent insulinotropic receptor (GPR119). This review discusses GPR43 and its ligands in the pathogenesis of AS, especially focusing on its distinct role in regulating chronic vascular inflammation, inhibiting oxidative stress, ameliorating endothelial dysfunction and improving dyslipidemia. It is hoped that this review may provide guidance for further studies aimed at GPR43 as a promising target for drug development in the prevention and therapy of AS.
Collapse
Affiliation(s)
- Mu-Yao Tang
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
- Departments of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Hao Xie
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
- Departments of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Jin-Tao Tao
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
- Departments of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Chun Zhang
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
- Departments of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Yao-Hua Luo
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
- Departments of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Cong Zhang
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
- Departments of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Si-Qin Peng
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
- Departments of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Lin-Xi Xie
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
- Departments of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Wen-Bo Lv
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
- Departments of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Chi Zhang
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Liang Huang
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
| |
Collapse
|
14
|
Cao L, Wang X, Ma X, Xu M, Li J. Potential of natural products and gut microbiome in tumor immunotherapy. Chin Med 2024; 19:161. [PMID: 39567970 PMCID: PMC11580227 DOI: 10.1186/s13020-024-01032-7] [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: 06/22/2024] [Accepted: 11/01/2024] [Indexed: 11/22/2024] Open
Abstract
Immunotherapy is a novel treatment approach for malignant tumors, which has opened a new journey of anti-tumor therapy. Although some patients will show a positive response to immunotherapy, unfortunately, most patients and cancer types do not achieve an ideal response to immunotherapy. Therefore, it is urgent to search for the pathogenesis of sensitized immunotherapy. This review indicates that Fusobacterium nucleatum, Coprobacillus cateniformis, Akkermansia muciniphila, Bifidobacterium, among others, as well as intestinal microbial metabolites are closely associated with resistance to anti-tumor immunotherapy. While natural products of pectin, inulin, jujube, anthocyanins, ginseng polysaccharides, diosgenin, camu-camu, and Inonotus hispidus (Bull).Fr. P. Karst, Icariside I, Safflower yellow, Ganoderma lucidum, and Ginsenoside Rk3, and other Chinese native medicinal compound prescriptions to boost their efficacy of anti-tumor immunotherapy through the regulation of microbiota and microbiota metabolites. However, current research mainly focuses on intestinal, liver, and lung cancer. In the future, natural products could be a viable option for treating malignant tumors, such as pancreatic, esophageal, and gastric malignancies, via sensitizing immunotherapy. Besides, the application characteristics of different types, sources and efficacy of natural products in different immune resistance scenarios also need to be further clarified through the development of future immunotherapy-related studies.
Collapse
Affiliation(s)
- Luchang Cao
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No.5, Beixian'ge Street, Xicheng District, Beijing, China
| | - Xinmiao Wang
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No.5, Beixian'ge Street, Xicheng District, Beijing, China
| | - Xinyi Ma
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No.5, Beixian'ge Street, Xicheng District, Beijing, China
| | - Manman Xu
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No.5, Beixian'ge Street, Xicheng District, Beijing, China
| | - Jie Li
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No.5, Beixian'ge Street, Xicheng District, Beijing, China.
| |
Collapse
|
15
|
Armstrong Suthahar SS, Nettersheim FS, Alimadadi A, Wang E, Billitti M, Resto-Trujillo N, Roy P, Hedrick CC, Ley K, Orecchioni M. Olfr2-positive macrophages originate from monocytes proliferate in situ and present a pro-inflammatory foamy-like phenotype. Cardiovasc Res 2024; 120:1577-1589. [PMID: 39229899 DOI: 10.1093/cvr/cvae153] [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: 02/27/2024] [Revised: 04/25/2024] [Accepted: 06/13/2024] [Indexed: 09/05/2024] Open
Abstract
AIMS Olfactory receptor 2 (Olfr2) has been identified in a minimum of 30% of vascular macrophages, and its depletion was shown to reduce atherosclerosis progression. Mononuclear phagocytes, including monocytes and macrophages within the vessel wall, are major players in atherosclerosis. Single-cell RNA sequencing studies revealed that atherosclerotic artery walls encompass several monocytes and vascular macrophages, defining at least nine distinct subsets potentially serving diverse functions in disease progression. This study investigates the functional phenotype and ontogeny of Olfr2-expressing vascular macrophages in atherosclerosis. METHODS AND RESULTS Olfr2+ macrophages rapidly increase in Apoe-/- mice's aorta when fed a Western diet (WD). Mass cytometry showed that Olfr2+ cells are clustered within the CD64 high population and enriched for CD11c and Ccr2 markers. Olfr2+ macrophages express many pro-inflammatory cytokines, including Il1b, Il6, Il12, and Il23, and chemokines, including Ccl5, Cx3cl1, Cxcl9, and Ccl22. By extracting differentially expressed genes from bulk RNA sequencing (RNA-seq) of Olfr2+ vs. Olfr2- macrophages, we defined a signature that significantly mapped to single-cell data of plaque myeloid cells, including monocytes, subendothelial MacAir, and Trem2Gpnmb foamy macrophages. By adoptive transfer experiments, we identified that Olfr2 competent monocytes from CD45.1Apoe-/-Olfr2+/+ mice transferred into CD45.2Apoe-/-Olfr2-/- recipient mice fed WD for 12 weeks, accumulate in the atherosclerotic aorta wall already at 72 h, and differentiate in macrophages. Olfr2+ macrophages showed significantly increased BrdU incorporation compared to Olfr2- macrophages. Flow cytometry confirmed that at least 50% of aortic Olfr2+ macrophages are positive for BODIPY staining and have increased expression of both tumour necrosis factor and interleukin 6 compared to Olfr2- macrophages. Gene set enrichment analysis of the Olfr2+ macrophage signature revealed a similar enrichment pattern in human atherosclerotic plaques, particularly within foamy/TREM2hi-Mφ and monocytes. CONCLUSIONS In summary, we conclude that Olfr2+ macrophages in the aorta originate from monocytes and can accumulate at the early stages of disease progression. These cells can undergo differentiation into MacAir and Trem2Gpnmb foamy macrophages, exhibiting proliferative and pro-inflammatory potentials. This dynamic behaviour positions them as key influencers in shaping the myeloid landscape within the atherosclerotic plaque.
Collapse
Affiliation(s)
| | - Felix Sebastian Nettersheim
- Division of Inflammation Biology, La Jolla Institute for Immunology, 9420 Athena Cir, La Jolla, CA 92037, USA
| | - Ahmad Alimadadi
- Division of Inflammation Biology, La Jolla Institute for Immunology, 9420 Athena Cir, La Jolla, CA 92037, USA
- Immunology Center of Georgia, Augusta University, 1410 Laney Walker Blvd, Augusta, GA 30912, USA
| | - Erpei Wang
- Division of Inflammation Biology, La Jolla Institute for Immunology, 9420 Athena Cir, La Jolla, CA 92037, USA
| | - Monica Billitti
- Division of Inflammation Biology, La Jolla Institute for Immunology, 9420 Athena Cir, La Jolla, CA 92037, USA
| | - Natalya Resto-Trujillo
- Immunology Center of Georgia, Augusta University, 1410 Laney Walker Blvd, Augusta, GA 30912, USA
| | - Payel Roy
- Division of Inflammation Biology, La Jolla Institute for Immunology, 9420 Athena Cir, La Jolla, CA 92037, USA
- Immunology Center of Georgia, Augusta University, 1410 Laney Walker Blvd, Augusta, GA 30912, USA
| | - Catherine C Hedrick
- Immunology Center of Georgia, Augusta University, 1410 Laney Walker Blvd, Augusta, GA 30912, USA
- Department of Medicine, Augusta University, 1120 15th St BA 8412, Augusta, GA 30912, USA
| | - Klaus Ley
- Immunology Center of Georgia, Augusta University, 1410 Laney Walker Blvd, Augusta, GA 30912, USA
- Department of Physiology, Augusta University, 1462 Laney Walker Blvd, Augusta, GA 30912, USA
| | - Marco Orecchioni
- Immunology Center of Georgia, Augusta University, 1410 Laney Walker Blvd, Augusta, GA 30912, USA
- Department of Pharmacology & Toxicology, Augusta University, 1459 Laney Walker Blvd, Augusta, GA 30901, USA
| |
Collapse
|
16
|
de Oliveira DP, Todorov SD, Fabi JP. Exploring the Prebiotic Potentials of Hydrolyzed Pectins: Mechanisms of Action and Gut Microbiota Modulation. Nutrients 2024; 16:3689. [PMID: 39519522 PMCID: PMC11547739 DOI: 10.3390/nu16213689] [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: 09/25/2024] [Revised: 10/20/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
The intestinal microbiota is a complex ecosystem where the microbial community (including bacteria) can metabolize available substrates via metabolic pathways specific to each species, often related in symbiotic relations. As a consequence of using available substrates and microbial growth, specific beneficial metabolites can be produced. When this reflects the health benefits for the host, these substrates can be categorized as prebiotics. Given that most prebiotic candidates must have a low molecular weight to be further metabolized by the microbiota, the role in the preliminary biological pretreatment is crucial. To provide proper substrates to the intestinal microbiota, a strategy could be to decrease the complexity of polysaccharides and reduce the levels of polymerization to low molecular weight for the target molecules, driving better solubilization and the consequent metabolic use by intestinal bacteria. When high molecular weight pectin is degraded (partially depolymerized), its solubility increases, thereby improving its utilization by gut microbiota. With regards to application, prebiotics have well-documented advantages when applied as food additives, as they improve gut health and can enhance drug effects, all shown by in vitro, in vivo, and clinical trials. In this review, we aim to provide systematic evidence for the mechanisms of action and the modulation of gut microbiota by the pectin-derived oligosaccharides produced by decreasing overall molecular weight after physical and/or chemical treatments and to compare with other types of prebiotics.
Collapse
Affiliation(s)
- Débora Preceliano de Oliveira
- Department of Food Science and Experimental Nutrition, School of Pharmaceutical Sciences, University of São Paulo, São Paulo 05508-000, SP, Brazil;
- Food Research Center (FoRC), CEPID-FAPESP (Research, Innovation and Dissemination Centers, São Paulo Research Foundation), São Paulo 05508-080, SP, Brazil;
| | - Svetoslav Dimitrov Todorov
- Food Research Center (FoRC), CEPID-FAPESP (Research, Innovation and Dissemination Centers, São Paulo Research Foundation), São Paulo 05508-080, SP, Brazil;
- ProBacLab, Department of Food Science and Experimental Nutrition, School of Pharmaceutical Sciences, University of São Paulo, São Paulo 05508-000, SP, Brazil
- Food Research Center (FoRC), CEPIX-USP, University of São Paulo, São Paulo 05508-080, SP, Brazil
| | - João Paulo Fabi
- Department of Food Science and Experimental Nutrition, School of Pharmaceutical Sciences, University of São Paulo, São Paulo 05508-000, SP, Brazil;
- Food Research Center (FoRC), CEPID-FAPESP (Research, Innovation and Dissemination Centers, São Paulo Research Foundation), São Paulo 05508-080, SP, Brazil;
| |
Collapse
|
17
|
Zhang K, Zeng Y, Li J, Huang Y, Zhang N, Gong Y, Xiao K, Chen J, Chen T, Qiu H, Lei S, Yan F, Lang C, Duan X, Dong X. Inulin alleviates atherosclerosis through improving lipid metabolism, inflammation, and gut microbiota in ApoE-knockout mice: the short-chain is more efficacious. Front Pharmacol 2024; 15:1445528. [PMID: 39449970 PMCID: PMC11499155 DOI: 10.3389/fphar.2024.1445528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 09/23/2024] [Indexed: 10/26/2024] Open
Abstract
Introduction Atherosclerosis (AS) is considered the underlying cause of many diseases, particularly cardiovascular and cerebrovascular diseases. Inulin, a type of fructan, has shown potential in improving atherosclerosis, although there are conflicting findings. It is hypothesized that the polymerization degree of inulin may largely influence its therapeutic effectiveness. Therefore, this study aimed to investigate the effects and mechanisms of short-chain and long-chain inulin in AS. Methods ApoE-/- mice fed a high fat diet (HFD) were used to establish an atherosclerosis model. These mice received daily oral administration of either short-chain or long-chain inulin for 12 weeks. Plasma lipid metabolism-related indices were measured using biochemical analysis, and plasma immunological indices were analyzed via ELISA. The aorta, aortic root regions, liver tissue, adipose tissue, and colon tissue were examined through various staining techniques, including ORO staining, hematoxylin and eosin staining, Alcian blue staining, and immunofluorescent or immunohistochemical assays. Microbiome analysis was conducted in the cecal content. Results The results indicated that both short-chain and long-chain inulin substantially reduced the formation of atherosclerotic plaques. Inulin also improved plasma lipid concentrations and hepatic lipid metabolism, and partially alleviated both localized (atherosclerotic lesions) and systemic inflammation. Short-chain inulin was more effective than long-chain inulin in reducing atherosclerotic plaques formation, enhancing lipid metabolism and reducing inflammation. Additionally, both types of inulin showed similar effectiveness in enhancing intestinal epithelial barrier integrity, gut microbiota composition and functionality. Conclusion These findings suggest that inulin has a protective role against atherosclerosis by enhancing lipid metabolism, reducing inflammation, and improving intestinal barrier and gut microbiota. As a dietary intervention, short-chain inulin is more effective than long-chain inulin, offering clinical implications for using inulin as a therapeutic agent for atherosclerosis.
Collapse
Affiliation(s)
- Kun Zhang
- Chongqing University Three Gorges Hospital, Chongqing Municipality Clinical Research Center for Geriatric Diseases, Chongqing, China
- School of Medicine, Chongqing University, Chongqing, China
| | - Yu Zeng
- Chongqing Academy of Animal Sciences, Chongqing, China
| | - Jiawei Li
- Chongqing University Three Gorges Hospital, Chongqing Municipality Clinical Research Center for Geriatric Diseases, Chongqing, China
- School of Medicine, Chongqing University, Chongqing, China
| | - Yingchun Huang
- Department of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Nan Zhang
- Department of General Surgery, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Yue Gong
- College of Life Sciences, Anhui Agricultural University, Hefei, China
| | - Kaihu Xiao
- Chongqing University Three Gorges Hospital, Chongqing Municipality Clinical Research Center for Geriatric Diseases, Chongqing, China
- School of Medicine, Chongqing University, Chongqing, China
| | - Jian Chen
- Chongqing University Three Gorges Hospital, Chongqing Municipality Clinical Research Center for Geriatric Diseases, Chongqing, China
- School of Medicine, Chongqing University, Chongqing, China
| | - Tiantian Chen
- Department of Biochemistry and Molecular Biology, Chongqing Medical University, Chongqing, China
| | - Haomin Qiu
- Department of Biochemistry and Molecular Biology, Chongqing Medical University, Chongqing, China
| | - Sisi Lei
- Department of Biochemistry and Molecular Biology, Chongqing Medical University, Chongqing, China
| | - Fei Yan
- Chongqing University Three Gorges Hospital, Chongqing Municipality Clinical Research Center for Geriatric Diseases, Chongqing, China
- School of Medicine, Chongqing University, Chongqing, China
| | - Chunhui Lang
- Chongqing University Three Gorges Hospital, Chongqing Municipality Clinical Research Center for Geriatric Diseases, Chongqing, China
- School of Medicine, Chongqing University, Chongqing, China
| | - Xudong Duan
- Department of Biochemistry and Molecular Biology, Chongqing Medical University, Chongqing, China
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, China
| | - Xianwen Dong
- Chongqing Academy of Animal Sciences, Chongqing, China
| |
Collapse
|
18
|
Yadav S, Sapra L, Srivastava RK. Polysaccharides to postbiotics: Nurturing bone health via modulating "gut-immune axis". Int J Biol Macromol 2024; 278:134655. [PMID: 39128750 DOI: 10.1016/j.ijbiomac.2024.134655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 08/06/2024] [Accepted: 08/08/2024] [Indexed: 08/13/2024]
Abstract
The increasing prevalence of individuals affected by bone pathologies globally has sparked catastrophic concerns. Ankylosing spondylitis, osteoporosis, rheumatoid arthritis, osteoarthritis, and fractures alone impact an estimated 1.71 billion people worldwide. The gut microbiota plays a crucial role in interacting with the host through the synthesis of a diverse range of metabolites called gut-associated metabolites (GAMs), which originate from external dietary substrates or endogenous host compounds. Many metabolic disorders have been linked to alterations in the gut microbiota's activity and composition. The development of metabolic illnesses has been linked to certain microbiota-derived metabolites, such as branched-chain amino acids, bile acids, short-chain fatty acids, tryptophan, trimethylamine N-oxide, and indole derivatives. Moreover, the modulation of gut microbiota through biotics (prebiotics, probiotics and postbiotics) presents a promising avenue for therapeutic intervention. Biotics selectively promote the growth of beneficial gut bacteria, thereby enhancing the production of GAMs with potential beneficial effects on bone metabolism. Understanding the intricate interplay between GAMs, and bone-associated genes through molecular informatics holds significant promise for early diagnosis, prognosis, and novel treatment strategies for various bone disorders.
Collapse
Affiliation(s)
- Sumedha Yadav
- Translational Immunology, Osteoimmunology & Immunoporosis Lab (TIOIL), Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Leena Sapra
- Translational Immunology, Osteoimmunology & Immunoporosis Lab (TIOIL), Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Rupesh K Srivastava
- Translational Immunology, Osteoimmunology & Immunoporosis Lab (TIOIL), Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India.
| |
Collapse
|
19
|
Lee YH, Lee JH, Jeon SM, Park IK, Jang HB, Kim SA, Park SD, Shim JJ, Hong SS, Lee JH. The Effect of Organic Vegetable Mixed Juice on Blood Circulation and Intestine Flora: Randomized, Double-Blinded, Placebo-Controlled Clinical Trial. Diseases 2024; 12:223. [PMID: 39329892 PMCID: PMC11431145 DOI: 10.3390/diseases12090223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 09/09/2024] [Accepted: 09/11/2024] [Indexed: 09/28/2024] Open
Abstract
Epidemiological evidence suggests that fruit and vegetable intake significantly positively affects cardiovascular health. Since vegetable juice is more accessible than raw vegetables, it attracts attention as a health functional food for circulatory diseases. Therefore, this study measured blood lipids, antioxidants, blood circulation indicators, and changes in the microbiome to confirm the effect of organic vegetable mixed juice (OVJ) on improving blood circulation. This 4-week, randomized, double-blinded, placebo-controlled study involved adult men and women with borderline total cholesterol (TC) and low-density lipoprotein (LDL) levels. As a result, blood lipid profile indicators, such as TC, triglycerides, LDL cholesterol, and apolipoprotein B, decreased (p < 0.05) in the OVJ group compared with those in the placebo group. Additionally, the antioxidant biomarker superoxide dismutase increased (p < 0.05). In contrast, systolic and diastolic blood viscosities, as blood circulation-related biomarkers, decreased (p < 0.05) in the OVJ group compared with those in the placebo group. After the intervention, a fecal microbiome analysis confirmed differences due to changes in the intestinal microbiome composition between the OVJ and placebo groups. In conclusion, our research results confirmed that consuming OVJ improves blood circulation by affecting the blood lipid profile, antioxidant enzymes, and microbiome changes.
Collapse
Affiliation(s)
- Yun-Ha Lee
- R&BD Center, Hy Co., Ltd., 22, Giheungdanji-ro 24beon-gil, Giheung-gu, Yongin-si 17086, Republic of Korea; (Y.-H.L.); (J.-H.L.); (S.-M.J.); (I.-K.P.); (H.-B.J.); (S.-A.K.); (S.-D.P.); (J.-J.S.)
| | - Jae-Ho Lee
- R&BD Center, Hy Co., Ltd., 22, Giheungdanji-ro 24beon-gil, Giheung-gu, Yongin-si 17086, Republic of Korea; (Y.-H.L.); (J.-H.L.); (S.-M.J.); (I.-K.P.); (H.-B.J.); (S.-A.K.); (S.-D.P.); (J.-J.S.)
| | - Soo-Min Jeon
- R&BD Center, Hy Co., Ltd., 22, Giheungdanji-ro 24beon-gil, Giheung-gu, Yongin-si 17086, Republic of Korea; (Y.-H.L.); (J.-H.L.); (S.-M.J.); (I.-K.P.); (H.-B.J.); (S.-A.K.); (S.-D.P.); (J.-J.S.)
| | - Il-Kyu Park
- R&BD Center, Hy Co., Ltd., 22, Giheungdanji-ro 24beon-gil, Giheung-gu, Yongin-si 17086, Republic of Korea; (Y.-H.L.); (J.-H.L.); (S.-M.J.); (I.-K.P.); (H.-B.J.); (S.-A.K.); (S.-D.P.); (J.-J.S.)
| | - Hyun-Bin Jang
- R&BD Center, Hy Co., Ltd., 22, Giheungdanji-ro 24beon-gil, Giheung-gu, Yongin-si 17086, Republic of Korea; (Y.-H.L.); (J.-H.L.); (S.-M.J.); (I.-K.P.); (H.-B.J.); (S.-A.K.); (S.-D.P.); (J.-J.S.)
| | - Soo-A Kim
- R&BD Center, Hy Co., Ltd., 22, Giheungdanji-ro 24beon-gil, Giheung-gu, Yongin-si 17086, Republic of Korea; (Y.-H.L.); (J.-H.L.); (S.-M.J.); (I.-K.P.); (H.-B.J.); (S.-A.K.); (S.-D.P.); (J.-J.S.)
| | - Soo-Dong Park
- R&BD Center, Hy Co., Ltd., 22, Giheungdanji-ro 24beon-gil, Giheung-gu, Yongin-si 17086, Republic of Korea; (Y.-H.L.); (J.-H.L.); (S.-M.J.); (I.-K.P.); (H.-B.J.); (S.-A.K.); (S.-D.P.); (J.-J.S.)
| | - Jae-Jung Shim
- R&BD Center, Hy Co., Ltd., 22, Giheungdanji-ro 24beon-gil, Giheung-gu, Yongin-si 17086, Republic of Korea; (Y.-H.L.); (J.-H.L.); (S.-M.J.); (I.-K.P.); (H.-B.J.); (S.-A.K.); (S.-D.P.); (J.-J.S.)
| | - Seong-Soo Hong
- Department of Gastroenterology, Vievis Namuh Hospital, 627, Nonhyeon-ro, Gangnam-gu, Seoul 06117, Republic of Korea
| | - Jae-Hwan Lee
- R&BD Center, Hy Co., Ltd., 22, Giheungdanji-ro 24beon-gil, Giheung-gu, Yongin-si 17086, Republic of Korea; (Y.-H.L.); (J.-H.L.); (S.-M.J.); (I.-K.P.); (H.-B.J.); (S.-A.K.); (S.-D.P.); (J.-J.S.)
| |
Collapse
|
20
|
Wang T, Holscher HD, Maslov S, Hu FB, Weiss ST, Liu YY. Predicting metabolite response to dietary intervention using deep learning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.03.14.532589. [PMID: 36993761 PMCID: PMC10054958 DOI: 10.1101/2023.03.14.532589] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/29/2023]
Abstract
Due to highly personalized biological and lifestyle characteristics, different individuals may have different metabolite responses to specific foods and nutrients. In particular, the gut microbiota, a collection of trillions of microorganisms living in the gastrointestinal tract, is highly personalized and plays a key role in the metabolite responses to foods and nutrients. Accurately predicting metabolite responses to dietary interventions based on individuals' gut microbial compositions holds great promise for precision nutrition. Existing prediction methods are typically limited to traditional machine learning models. Deep learning methods dedicated to such tasks are still lacking. Here we develop a method McMLP (Metabolite response predictor using coupled Multilayer Perceptrons) to fill in this gap. We provide clear evidence that McMLP outperforms existing methods on both synthetic data generated by the microbial consumer-resource model and real data obtained from six dietary intervention studies. Furthermore, we perform sensitivity analysis of McMLP to infer the tripartite food-microbe-metabolite interactions, which are then validated using the ground-truth (or literature evidence) for synthetic (or real) data, respectively. The presented tool has the potential to inform the design of microbiota-based personalized dietary strategies to achieve precision nutrition.
Collapse
Affiliation(s)
- Tong Wang
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Hannah D. Holscher
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Center for Artificial Intelligence and Modeling, The Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Sergei Maslov
- Center for Artificial Intelligence and Modeling, The Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Frank B. Hu
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Scott T. Weiss
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Yang-Yu Liu
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Center for Artificial Intelligence and Modeling, The Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| |
Collapse
|
21
|
Xun P, Zhuang S, Yao H, Su J, Yang Y, Shu H, Yu W, Lin H. Effects of Sodium Acetate Supplementation on Growth, Hematologic and Plasma Biochemical Parameter, Lipid Deposition, and Intestinal Health of Juvenile Golden Pompano Trachinotus ovatus Fed High-Lipid Diets. AQUACULTURE NUTRITION 2024; 2024:7904141. [PMID: 39555552 PMCID: PMC11401687 DOI: 10.1155/2024/7904141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 07/11/2024] [Accepted: 08/08/2024] [Indexed: 11/19/2024]
Abstract
Experimental diets were formulated including the suitable lipid level (10%, PC), the high-lipid level (16%, HL), and HL containing sodium acetate diets (HS). Three diets were fed golden pompano (Trachinotus ovatus) (initial body weight: 12.88 ± 0.03 g) for 8 weeks. The results showed HL diets significantly increased hepatosomatic index (HSI) and abdominal fat percentage (ASF), aggravated liver lipid deposition, and caused blood metabolic disorder and liver damage (P < 0.05). Moreover, the fish fed HL diets significantly decreased intestinal villus number (VN) and muscular layer thickness (MLT) (P < 0.05), accompanied with an increased trend in the relative abundance of intestinal pathogenic bacteria such as Mycoplasma and Photobacterium. However, the fish fed HS diets significantly decreased the HSI and AFP, relieved hepatic lipid deposition, improved blood and liver metabolism, and intestinal morphology in comparison to the fish fed HL diets (P < 0.05). More importantly, sodium acetate addition improved intestinal microbiota by inhibiting the proportion of pathogens (Mycoplasma and Vibrio) and increasing the abundance of probiotics (Bacteroidales_S24-7_group_norank, Cetobacterium, Bacteroides, and Lachnospiraceae_NK4A136_group). Furthermore, there was a strong correlation between these bacteria (Mycoplasma, Vibrio, Lachnospiraceae_NK4A136_group, Bacteroidales_S24-7_group_norank, Bacteroides, and Cetobacterium) and main physiological indices. In conclusion, sodium acetate improved blood performance, alleviated hepatic lipid deposition induced by HL diets, and boosted the growth and intestinal health for golden pompano.
Collapse
Affiliation(s)
- Pengwei Xun
- School of FisheriesXinyang Agriculture and Forestry University, Xinyang 464000, China
| | - Siling Zhuang
- School of Life ScienceGuangzhou University, Guangzhou 510006, China
| | - Handong Yao
- School of FisheriesXinyang Agriculture and Forestry University, Xinyang 464000, China
| | - Jinhao Su
- School of FisheriesXinyang Agriculture and Forestry University, Xinyang 464000, China
| | - Yukai Yang
- Key Laboratory of South China Sea Fishery Resources Exploitation and UtilizationMinistry of Agriculture and Rural AffairsSouth China Sea Fisheries Research InstituteChinese Academy of Fishery Sciences, Guangzhou 510300, China
- Shenzhen Base of South China Sea Fisheries Research InstituteChinese Academy of Fishery Sciences, Shenzhen 518121, China
| | - Hu Shu
- School of Life ScienceGuangzhou University, Guangzhou 510006, China
| | - Wei Yu
- Key Laboratory of South China Sea Fishery Resources Exploitation and UtilizationMinistry of Agriculture and Rural AffairsSouth China Sea Fisheries Research InstituteChinese Academy of Fishery Sciences, Guangzhou 510300, China
- Shenzhen Base of South China Sea Fisheries Research InstituteChinese Academy of Fishery Sciences, Shenzhen 518121, China
- Tropical Fisheries Research and Development CenterSouth China Sea Fisheries Research InstituteChinese Academy of Fishery Sciences, Sanya 572018, China
| | - Heizhao Lin
- Key Laboratory of South China Sea Fishery Resources Exploitation and UtilizationMinistry of Agriculture and Rural AffairsSouth China Sea Fisheries Research InstituteChinese Academy of Fishery Sciences, Guangzhou 510300, China
- Shenzhen Base of South China Sea Fisheries Research InstituteChinese Academy of Fishery Sciences, Shenzhen 518121, China
| |
Collapse
|
22
|
Safdar M, Ullah M, Hamayun S, Wahab A, Khan SU, Abdikakhorovich SA, Haq ZU, Mehreen A, Naeem M, Mustopa AZ, Hasan N. Microbiome miracles and their pioneering advances and future frontiers in cardiovascular disease. Curr Probl Cardiol 2024; 49:102686. [PMID: 38830479 DOI: 10.1016/j.cpcardiol.2024.102686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 05/28/2024] [Indexed: 06/05/2024]
Abstract
Cardiovascular diseases (CVDs) represent a significant global health challenge, underscoring the need for innovative approaches to prevention and treatment. Recent years have seen a surge in interest in unraveling the complex relationship between the gut microbiome and cardiovascular health. This article delves into current research on the composition, diversity, and impact of the gut microbiome on CVD development. Recent advancements have elucidated the profound influence of the gut microbiome on disease progression, particularly through key mediators like Trimethylamine-N-oxide (TMAO) and other microbial metabolites. Understanding these mechanisms reveals promising therapeutic targets, including interventions aimed at modulating the gut microbiome's interaction with the immune system and its contribution to endothelial dysfunction. Harnessing this understanding, personalized medicine strategies tailored to individuals' gut microbiome profiles offer innovative avenues for reducing cardiovascular risk. As research in this field continues to evolve, there is vast potential for transformative advancements in cardiovascular medicine, paving the way for precision prevention and treatment strategies to address this global health challenge.
Collapse
Affiliation(s)
- Mishal Safdar
- Department of Biological Sciences, National University of Medical Sciences (NUMS), Rawalpindi, Punjab, Pakistan
| | - Muneeb Ullah
- College of Pharmacy, Pusan National University, Busandaehak-ro 63 beon-gil 2, Geumjeong-gu, Busan 46241, Republic of Korea; Department of Pharmacy, Kohat University of Science and Technology, Kohat, 26000, Khyber Pakhtunkhwa, Pakistan
| | - Shah Hamayun
- Department of Cardiology, Pakistan Institute of Medical Sciences (PIMS), Islamabad, 04485 Punjab, Pakistan
| | - Abdul Wahab
- Department of Pharmacy, Kohat University of Science and Technology, Kohat, 26000, Khyber Pakhtunkhwa, Pakistan
| | - Shahid Ullah Khan
- Department of Biochemistry, Women Medical and Dental College, Khyber Medical University, Abbottabad, 22080, Khyber Pakhtunkhwa, Pakistan
| | | | - Zia Ul Haq
- Department of Public Health, Institute of Public Health Sciences, Khyber Medical University, Peshawar 25120, Pakistan
| | - Aqsa Mehreen
- Department of Biological Sciences, National University of Medical Sciences (NUMS), Rawalpindi, Punjab, Pakistan
| | - Muhammad Naeem
- Department of Biological Sciences, National University of Medical Sciences (NUMS), Rawalpindi, Punjab, Pakistan
| | - Apon Zaenal Mustopa
- Research Center for Genetic Engineering, National Research, and Innovation Agency (BRIN), Bogor 16911, Indonesia
| | - Nurhasni Hasan
- Faculty of Pharmacy, Universitas Hasanuddin, Jl. Perintis Kemerdekaan Km 10, Makassar 90245, Republic of Indonesia.
| |
Collapse
|
23
|
Trivedi A, Bose D, Moffat K, Pearson E, Walsh D, Cohen D, Skupsky J, Chao L, Golier J, Janulewicz P, Sullivan K, Krengel M, Tuteja A, Klimas N, Chatterjee S. Gulf War Illness Is Associated with Host Gut Microbiome Dysbiosis and Is Linked to Altered Species Abundance in Veterans from the BBRAIN Cohort. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2024; 21:1102. [PMID: 39200711 PMCID: PMC11354743 DOI: 10.3390/ijerph21081102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 08/09/2024] [Accepted: 08/13/2024] [Indexed: 09/02/2024]
Abstract
Gulf War Illness (GWI) is a debilitating condition marked by chronic fatigue, cognitive problems, pain, and gastrointestinal (GI) complaints in veterans who were deployed to the 1990-1991 Gulf War. Fatigue, GI complaints, and other chronic symptoms continue to persist more than 30 years post-deployment. Several potential mechanisms for the persistent illness have been identified and our prior pilot study linked an altered gut microbiome with the disorder. This study further validates and builds on our prior preliminary findings of host gut microbiome dysbiosis in veterans with GWI. Using stool samples and Multidimensional Fatigue Inventory (MFI) data from 89 GW veteran participants (63 GWI cases and 26 controls) from the Boston biorepository, recruitment, and integrative network (BBRAIN) for Gulf War Illness, we found that the host gut bacterial signature of veterans with GWI showed significantly different Bray-Curtis beta diversity than control veterans. Specifically, a higher Firmicutes to Bacteroidetes ratio, decrease in Akkermansia sp., Bacteroides thetaiotamicron, Bacteroides fragilis, and Lachnospiraceae genera and increase in Blautia, Streptococcus, Klebsiella, and Clostridium genera, that are associated with gut, immune, and brain health, were shown. Further, using MaAsLin and Boruta algorithms, Coprococcus and Eisenbergiella were identified as important predictors of GWI with an area under the curve ROC predictive value of 74.8%. Higher self-reported MFI scores in veterans with GWI were also significantly associated with an altered gut bacterial diversity and species abundance of Lachnospiraceae and Blautia. These results suggest potential therapeutic targets for veterans with GWI that target the gut microbiome and specific symptoms of the illness.
Collapse
Affiliation(s)
- Ayushi Trivedi
- Environmental Health and Disease Laboratory, Department of Environmental and Occupational Health, Program in Public Health, Susan and Henry Samueli College of Health Sciences, University of California, Irvine, CA 92697, USA; (A.T.); (D.B.)
| | - Dipro Bose
- Environmental Health and Disease Laboratory, Department of Environmental and Occupational Health, Program in Public Health, Susan and Henry Samueli College of Health Sciences, University of California, Irvine, CA 92697, USA; (A.T.); (D.B.)
| | - Kelly Moffat
- CosmosID, Germantown, MD 20874, USA; (K.M.); (D.W.)
| | | | - Dana Walsh
- CosmosID, Germantown, MD 20874, USA; (K.M.); (D.W.)
| | - Devra Cohen
- Miami VA Healthcare System, Miami, FL 33125, USA;
- Institute for Neuro-Immune Medicine, Nova Southeastern University, Fort Lauderdale, FL 33314, USA;
| | - Jonathan Skupsky
- VA Research and Development, VA Long Beach Health Care, Long Beach, CA 90822, USA;
| | - Linda Chao
- San Francisco Veterans Affairs Health Care System, San Francisco, CA 94121, USA
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA 94143, USA
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, CA 94143, USA
| | - Julia Golier
- J. Peters VA Medical Center, Bronx, NY 10468, USA;
- Psychiatry Department, Icahn School of Medicine at Mount Sinai, 1428 Madison Ave, New York, NY 10029, USA
| | - Patricia Janulewicz
- Department of Environmental Health, Boston University School of Public Health, 715 Albany St. T4W, Boston, MA 02130, USA; (P.J.)
| | - Kimberly Sullivan
- Department of Environmental Health, Boston University School of Public Health, 715 Albany St. T4W, Boston, MA 02130, USA; (P.J.)
| | - Maxine Krengel
- Department of Neurology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02130, USA;
| | - Ashok Tuteja
- Division of Gastroenterology, School of Medicine, University of Utah, Salt Lake City, UT 84132, USA;
| | - Nancy Klimas
- Institute for Neuro-Immune Medicine, Nova Southeastern University, Fort Lauderdale, FL 33314, USA;
- Geriatric Research and Education Clinical Center, Miami VA Heathcare System, Miami, FL 33125, USA
| | - Saurabh Chatterjee
- Environmental Health and Disease Laboratory, Department of Environmental and Occupational Health, Program in Public Health, Susan and Henry Samueli College of Health Sciences, University of California, Irvine, CA 92697, USA; (A.T.); (D.B.)
- Institute for Neuro-Immune Medicine, Nova Southeastern University, Fort Lauderdale, FL 33314, USA;
- Department of Medicine, Infectious Disease, UCI School of Medicine, Irvine, CA 92697, USA
| |
Collapse
|
24
|
Bruun CF, Haldor Hansen T, Vinberg M, Kessing LV, Coello K. Associations between short-chain fatty acid levels and mood disorder symptoms: a systematic review. Nutr Neurosci 2024; 27:899-912. [PMID: 37976103 DOI: 10.1080/1028415x.2023.2277970] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2023]
Abstract
Background: Available evidence points to a possible role of Short Chain Fatty Acids (SCFAs) in mood disorders. This is the first systematic review to map the associations between SCFA levels and mood disorder symptoms.Methods: Following the PRISMA guidelines, the databases PubMed, Embase, and PsycINFO were searched for studies that assessed SCFA levels in human populations with mood disorder symptoms, or animal models of mood disorder. Risk of bias was assessed by the Strengthening of Reporting of Observational Studies in Epidemiology (STROBE) checklist.Results: 19 studies were included and could be divided into animal (n=8) and human studies (n=11), with the animal studies including 166 animals and 100 controls, and the human studies including 662 participants and 330 controls. The studies were characterized by heterogeneity and methodological challenges on multiple parameters, limiting the validity and transferability of findings. Notably, only two of the clinical studies assessed the presence of mood disorder with diagnostic criteria, and no studies of mania or bipolar disorder met the inclusion criteria.Discussion: Despite significant methodological limitations, associations between SCFA levels and depressive symptoms were reported in most of the studies. However, the direction of these associations and the specific SCFAs identified varied. The quantification of SCFA levels in mood disorders is an emerging yet sparsely studied research field. Although there is some evidence suggesting a link between SCFAs and depressive symptoms, the directionality of effects and mechanisms are unclear and the relation to manic symptoms is uninvestigated.
Collapse
Affiliation(s)
- Caroline Fussing Bruun
- Copenhagen Affective Disorder Research Center (CADIC), Psychiatric Center Copenhagen, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Tue Haldor Hansen
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Copenhagen, Denmark
| | - Maj Vinberg
- Copenhagen Affective Disorder Research Center (CADIC), Psychiatric Center Copenhagen, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Psychiatric Center Northern Zealand, Hilleroed, Denmark
| | - Lars Vedel Kessing
- Copenhagen Affective Disorder Research Center (CADIC), Psychiatric Center Copenhagen, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Klara Coello
- Copenhagen Affective Disorder Research Center (CADIC), Psychiatric Center Copenhagen, Copenhagen, Denmark
| |
Collapse
|
25
|
Cho YS, Han K, Xu J, Moon JJ. Novel strategies for modulating the gut microbiome for cancer therapy. Adv Drug Deliv Rev 2024; 210:115332. [PMID: 38759702 PMCID: PMC11268941 DOI: 10.1016/j.addr.2024.115332] [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: 01/29/2024] [Revised: 05/08/2024] [Accepted: 05/13/2024] [Indexed: 05/19/2024]
Abstract
Recent advancements in genomics, transcriptomics, and metabolomics have significantly advanced our understanding of the human gut microbiome and its impact on the efficacy and toxicity of anti-cancer therapeutics, including chemotherapy, immunotherapy, and radiotherapy. In particular, prebiotics, probiotics, and postbiotics are recognized for their unique properties in modulating the gut microbiota, maintaining the intestinal barrier, and regulating immune cells, thus emerging as new cancer treatment modalities. However, clinical translation of microbiome-based therapy is still in its early stages, facing challenges to overcome physicochemical and biological barriers of the gastrointestinal tract, enhance target-specific delivery, and improve drug bioavailability. This review aims to highlight the impact of prebiotics, probiotics, and postbiotics on the gut microbiome and their efficacy as cancer treatment modalities. Additionally, we summarize recent innovative engineering strategies designed to overcome challenges associated with oral administration of anti-cancer treatments. Moreover, we will explore the potential benefits of engineered gut microbiome-modulating approaches in ameliorating the side effects of immunotherapy and chemotherapy.
Collapse
Affiliation(s)
- Young Seok Cho
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Kai Han
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 21009, China; Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 21009, China
| | - Jin Xu
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - James J Moon
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
26
|
Festus ID, Spilberg J, Young ME, Cain S, Khoshnevis S, Smolensky MH, Zaheer F, Descalzi G, Martino TA. Pioneering new frontiers in circadian medicine chronotherapies for cardiovascular health. Trends Endocrinol Metab 2024; 35:607-623. [PMID: 38458859 DOI: 10.1016/j.tem.2024.02.011] [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: 12/21/2023] [Revised: 02/08/2024] [Accepted: 02/12/2024] [Indexed: 03/10/2024]
Abstract
Cardiovascular disease (CVD) is a global health concern. Circadian medicine improves cardiovascular care by aligning treatments with our body's daily rhythms and their underlying cellular circadian mechanisms. Time-based therapies, or chronotherapies, show special promise in clinical cardiology. They optimize treatment schedules for better outcomes with fewer side effects by recognizing the profound influence of rhythmic body cycles. In this review, we focus on three chronotherapy areas (medication, light, and meal timing) with potential to enhance cardiovascular care. We also highlight pioneering research in the new field of rest, the gut microbiome, novel chronotherapies for hypertension, pain management, and small molecules that targeting the circadian mechanism.
Collapse
Affiliation(s)
- Ifene David Festus
- Centre for Cardiovascular Investigations, University of Guelph; Guelph, Ontario, Canada; Department of Biomedical Sciences, University of Guelph; Guelph, Ontario, Canada
| | - Jeri Spilberg
- Centre for Cardiovascular Investigations, University of Guelph; Guelph, Ontario, Canada; Department of Biomedical Sciences, University of Guelph; Guelph, Ontario, Canada
| | - Martin E Young
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Sean Cain
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Melbourne, Victoria, Australia
| | - Sepideh Khoshnevis
- Department of Biomedical Engineering, Cockrell School of Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Michael H Smolensky
- Department of Biomedical Engineering, Cockrell School of Engineering, The University of Texas at Austin, Austin, TX, USA; Department of Internal Medicine, Division of Cardiology, McGovern School of Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Fariya Zaheer
- Department of Biomedical Sciences, University of Guelph; Guelph, Ontario, Canada
| | - Giannina Descalzi
- Department of Biomedical Sciences, University of Guelph; Guelph, Ontario, Canada
| | - Tami A Martino
- Centre for Cardiovascular Investigations, University of Guelph; Guelph, Ontario, Canada; Department of Biomedical Sciences, University of Guelph; Guelph, Ontario, Canada.
| |
Collapse
|
27
|
Shi K, Jiao Y, Yang L, Yuan G, Jia J. New insights into the roles of olfactory receptors in cardiovascular disease. Mol Cell Biochem 2024; 479:1615-1626. [PMID: 38761351 DOI: 10.1007/s11010-024-05024-x] [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: 02/21/2024] [Accepted: 04/26/2024] [Indexed: 05/20/2024]
Abstract
Olfactory receptors (ORs) are G protein coupled receptors (GPCRs) with seven transmembrane domains that bind to specific exogenous chemical ligands and transduce intracellular signals. They constitute the largest gene family in the human genome. They are expressed in the epithelial cells of the olfactory organs and in the non-olfactory tissues such as the liver, kidney, heart, lung, pancreas, intestines, muscle, testis, placenta, cerebral cortex, and skin. They play important roles in the normal physiological and pathophysiological mechanisms. Recent evidence has highlighted a close association between ORs and several metabolic diseases. Cardiovascular diseases (CVDs) are the leading cause of morbidity and mortality globally. Furthermore, ORs play an essential role in the development and functional regulation of the cardiovascular system and are implicated in the pathophysiological mechanisms of CVDs, including atherosclerosis (AS), heart failure (HF), aneurysms, and hypertension (HTN). This review describes the specific mechanistic roles of ORs in the CVDs, and highlights the future clinical application prospects of ORs in the diagnosis, treatment, and prevention of the CVDs.
Collapse
Affiliation(s)
- Kangru Shi
- Department of Endocrinology and Metabolissm, The Affiliated Hospital of Jiangsu University, Institute of Endocrine and Metabolic Diseases, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yang Jiao
- Department of Endocrinology and Metabolissm, The Affiliated Hospital of Jiangsu University, Institute of Endocrine and Metabolic Diseases, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Ling Yang
- Department of Endocrinology and Metabolissm, The Affiliated Hospital of Jiangsu University, Institute of Endocrine and Metabolic Diseases, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Guoyue Yuan
- Department of Endocrinology and Metabolissm, The Affiliated Hospital of Jiangsu University, Institute of Endocrine and Metabolic Diseases, Jiangsu University, Zhenjiang, Jiangsu, China.
| | - Jue Jia
- Department of Endocrinology and Metabolissm, The Affiliated Hospital of Jiangsu University, Institute of Endocrine and Metabolic Diseases, Jiangsu University, Zhenjiang, Jiangsu, China.
| |
Collapse
|
28
|
Ronen D, Rokach Y, Abedat S, Qadan A, Daana S, Amir O, Asleh R. Human Gut Microbiota in Cardiovascular Disease. Compr Physiol 2024; 14:5449-5490. [PMID: 39109979 DOI: 10.1002/cphy.c230012] [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: 01/03/2025]
Abstract
The gut ecosystem, termed microbiota, is composed of bacteria, archaea, viruses, protozoa, and fungi and is estimated to outnumber human cells. Microbiota can affect the host by multiple mechanisms, including the synthesis of metabolites and toxins, modulating inflammation and interaction with other organisms. Advances in understanding commensal organisms' effect on human conditions have also elucidated the importance of this community for cardiovascular disease (CVD). This effect is driven by both direct CV effects and conditions known to increase CV risk, such as obesity, diabetes mellitus (DM), hypertension, and renal and liver diseases. Cardioactive metabolites, such as trimethylamine N -oxide (TMAO), short-chain fatty acids (SCFA), lipopolysaccharides, bile acids, and uremic toxins, can affect atherosclerosis, platelet activation, and inflammation, resulting in increased CV incidence. Interestingly, this interaction is bidirectional with microbiota affected by multiple host conditions including diet, bile acid secretion, and multiple diseases affecting the gut barrier. This interdependence makes manipulating microbiota an attractive option to reduce CV risk. Indeed, evolving data suggest that the benefits observed from low red meat and Mediterranean diet consumption can be explained, at least partially, by the changes that these diets may have on the gut microbiota. In this article, we depict the current epidemiological and mechanistic understanding of the role of microbiota and CVD. Finally, we discuss the potential therapeutic approaches aimed at manipulating gut microbiota to improve CV outcomes. © 2024 American Physiological Society. Compr Physiol 14:5449-5490, 2024.
Collapse
Affiliation(s)
- Daniel Ronen
- Cardiovascular Research Center, Heart Institute, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Yair Rokach
- Cardiovascular Research Center, Heart Institute, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Suzan Abedat
- Cardiovascular Research Center, Heart Institute, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Abed Qadan
- Cardiovascular Research Center, Heart Institute, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Samar Daana
- Cardiovascular Research Center, Heart Institute, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Offer Amir
- Cardiovascular Research Center, Heart Institute, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Rabea Asleh
- Cardiovascular Research Center, Heart Institute, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
29
|
Nanaware PP, Khan ZN, Clement CC, Shetty M, Mota I, Seltzer ES, Dzieciatkowska M, Gamboni F, D'Alessandro A, Ng C, Nagayama M, Lichti CF, Soni RK, Jacob B Geri, Matei I, Lyden D, Longman R, Lu TT, Wan X, Unanue ER, Stern LJ, Santambrogio L. Role of the afferent lymph as an immunological conduit to analyze tissue antigenic and inflammatory load. Cell Rep 2024; 43:114311. [PMID: 38848214 PMCID: PMC11233987 DOI: 10.1016/j.celrep.2024.114311] [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: 10/16/2023] [Revised: 04/03/2024] [Accepted: 05/16/2024] [Indexed: 06/09/2024] Open
Abstract
The lymphatic fluid is the conduit by which part of the tissue "omics" is transported to the draining lymph node for immunosurveillance. Following cannulation of the pre-nodal cervical and mesenteric afferent lymphatics, herein we investigate the lymph proteomic composition, uncovering that its composition varies according to the tissue of origin. Tissue specificity is also reflected in the dendritic cell-major histocompatibility complex class II-eluted immunopeptidome harvested from the cervical and mesenteric nodes. Following inflammatory disruption of the gut barrier, the lymph antigenic and inflammatory loads are analyzed in both mice and subjects with inflammatory bowel diseases. Gastrointestinal tissue damage reflects the lymph inflammatory and damage-associated molecular pattern signatures, microbiome-derived by-products, and immunomodulatory molecules, including metabolites of the gut-brain axis, mapped in the afferent mesenteric lymph. Our data point to the relevance of the lymphatic fluid to probe the tissue-specific antigenic and inflammatory load transported to the draining lymph node for immunosurveillance.
Collapse
Affiliation(s)
- Padma P Nanaware
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, USA; Department of Pathology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Zohaib N Khan
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Cristina C Clement
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Madhur Shetty
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Ines Mota
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Ethan S Seltzer
- Pediatric Rheumatology and Autoimmunity and Inflammation Program, Hospital for Special Surgery Research Institute, New York NY 100021, USA
| | - Monika Dzieciatkowska
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Fabia Gamboni
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Charles Ng
- Department of Pathology and Laboratory Medicine, New York-Presbyterian Hospital and Weill Cornell Medicine, New York, NY 10065, USA
| | - Manabu Nagayama
- Division of Gastroenterology and Hepatology, New York-Presbyterian Hospital and Weill Cornell Medicine, New York, NY 10065, USA
| | - Cheryl F Lichti
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Rajesh K Soni
- Proteomics and Macromolecular Crystallography Shared Resource, Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York 10032, NY, USA
| | - Jacob B Geri
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics and Cell and Developmental Biology, Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY 10065, USA
| | - Irina Matei
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics and Cell and Developmental Biology, Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY 10065, USA; Sandra and Edward Meyer Cancer Center, New York, NY 10065, USA
| | - David Lyden
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics and Cell and Developmental Biology, Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY 10065, USA; Sandra and Edward Meyer Cancer Center, New York, NY 10065, USA
| | - Randy Longman
- Division of Gastroenterology and Hepatology, New York-Presbyterian Hospital and Weill Cornell Medicine, New York, NY 10065, USA
| | - Theresa T Lu
- Pediatric Rheumatology and Autoimmunity and Inflammation Program, Hospital for Special Surgery Research Institute, New York NY 100021, USA
| | - Xiaoxiao Wan
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Emil R Unanue
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Lawrence J Stern
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Laura Santambrogio
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, USA; Sandra and Edward Meyer Cancer Center, New York, NY 10065, USA; Caryl and Israel Englander Institute for Precision Medicine, New York, NY 10065, USA.
| |
Collapse
|
30
|
Zhang Y, Zhen S, Xu H, Sun S, Wang Z, Li M, Zou L, Zhang Y, Zhao Y, Cui Y, Han J. Vitamin C alleviates rheumatoid arthritis by modulating gut microbiota balance. Biosci Trends 2024; 18:187-194. [PMID: 38599880 DOI: 10.5582/bst.2024.01037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2024]
Abstract
Rheumatoid arthritis (RA) is a systemic autoimmune disease characterized by chronic and symmetric in-flammation. Our previous research revealed an imbalance in the gut flora of RA patients and showed that certain gut microbiota can accelerate RA progression by enhancing vitamin C degradation. However, it is unclear whether vitamin C supplementation could improve the gut microbiota to prevent the development of arthritis by interfering with the gut-joint axis. In this work, we aimed to evaluate the effects of vitamin C in regulating the gut microbiota and to elucidate its potential role in the onset and progression of RA in a mouse model, thus providing a basis for the development of new intervention strategies and treatments for RA. In this study, collagen-induced arthritis (CIA) mouse models, biochemical, histological and 16S rRNA microbiological methods were used to investigate the role and possible mechanism of vitamin C in rheumatoid arthritis. The results showed that treatment of CIA mice with vitamin C effectively rescued the gut mi-crobiota imbalance and suppressed the inflammatory response associated with RA, and effectively alleviated arthritis symptoms in mice in which levels of the pro-inflammatory cytokines IL-6 and TNF-α were specifi-cally reduced. In conclusion, our results demonstrate the potential of vitamin C as a potential therapeutic choice for RA.
Collapse
Affiliation(s)
- Yanjie Zhang
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji' nan, Shandong, China
- NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Ji'nan, Shandong, China
- Key Lab for Rare & Uncommon Diseases of Shandong Province, Ji'nan, Shandong, China
| | - Sibin Zhen
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji' nan, Shandong, China
- NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Ji'nan, Shandong, China
- Key Lab for Rare & Uncommon Diseases of Shandong Province, Ji'nan, Shandong, China
| | - Hao Xu
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji' nan, Shandong, China
- NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Ji'nan, Shandong, China
- Key Lab for Rare & Uncommon Diseases of Shandong Province, Ji'nan, Shandong, China
| | - Songfang Sun
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji' nan, Shandong, China
- NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Ji'nan, Shandong, China
- Key Lab for Rare & Uncommon Diseases of Shandong Province, Ji'nan, Shandong, China
| | - Ziwei Wang
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji' nan, Shandong, China
- NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Ji'nan, Shandong, China
- Key Lab for Rare & Uncommon Diseases of Shandong Province, Ji'nan, Shandong, China
| | - Mian Li
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji' nan, Shandong, China
- NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Ji'nan, Shandong, China
- Key Lab for Rare & Uncommon Diseases of Shandong Province, Ji'nan, Shandong, China
| | - Liang Zou
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji' nan, Shandong, China
- Bone Biomechanics Engineering Laboratory of Shandong Province, Neck-Shoulder and Lumbocrural Pain Hospital of Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, China
| | - Yangyang Zhang
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji' nan, Shandong, China
- NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Ji'nan, Shandong, China
- Key Lab for Rare & Uncommon Diseases of Shandong Province, Ji'nan, Shandong, China
| | - Yan Zhao
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji' nan, Shandong, China
- NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Ji'nan, Shandong, China
- Key Lab for Rare & Uncommon Diseases of Shandong Province, Ji'nan, Shandong, China
| | - Yazhou Cui
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji' nan, Shandong, China
- NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Ji'nan, Shandong, China
- Key Lab for Rare & Uncommon Diseases of Shandong Province, Ji'nan, Shandong, China
| | - Jinxiang Han
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji' nan, Shandong, China
- NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Ji'nan, Shandong, China
- Key Lab for Rare & Uncommon Diseases of Shandong Province, Ji'nan, Shandong, China
| |
Collapse
|
31
|
Ahmad A, Mahmood N, Raza MA, Mushtaq Z, Saeed F, Afzaal M, Hussain M, Amjad HW, Al-Awadi HM. Gut microbiota and their derivatives in the progression of colorectal cancer: Mechanisms of action, genome and epigenome contributions. Heliyon 2024; 10:e29495. [PMID: 38655310 PMCID: PMC11035079 DOI: 10.1016/j.heliyon.2024.e29495] [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: 05/08/2023] [Revised: 04/08/2024] [Accepted: 04/09/2024] [Indexed: 04/26/2024] Open
Abstract
Gut microbiota interacts with host epithelial cells and regulates many physiological functions such as genetics, epigenetics, metabolism of nutrients, and immune functions. Dietary factors may also be involved in the etiology of colorectal cancer (CRC), especially when an unhealthy diet is consumed with excess calorie intake and bad practices like smoking or consuming a great deal of alcohol. Bacteria including Fusobacterium nucleatum, Enterotoxigenic Bacteroides fragilis (ETBF), and Escherichia coli (E. coli) actively participate in the carcinogenesis of CRC. Gastrointestinal tract with chronic inflammation and immunocompromised patients are at high risk for CRC progression. Further, the gut microbiota is also involved in Geno-toxicity by producing toxins like colibactin and cytolethal distending toxin (CDT) which cause damage to double-stranded DNA. Specific microRNAs can act as either tumor suppressors or oncogenes depending on the cellular environment in which they are expressed. The current review mainly highlights the role of gut microbiota in CRC, the mechanisms of several factors in carcinogenesis, and the role of particular microbes in colorectal neoplasia.
Collapse
Affiliation(s)
- Awais Ahmad
- Department of Food Science, Government College University Faisalabad, Faisalabad, Pakistan
| | - Nasir Mahmood
- Department of Zoology, University of Central Punjab Bahawalpur, Bahawalpur, Pakistan
| | - Muhammad Ahtisham Raza
- Department of Food Science, Government College University Faisalabad, Faisalabad, Pakistan
| | - Zarina Mushtaq
- Department of Food Science, Government College University Faisalabad, Faisalabad, Pakistan
| | - Farhan Saeed
- Department of Food Science, Government College University Faisalabad, Faisalabad, Pakistan
| | - Muhammad Afzaal
- Department of Food Science, Government College University Faisalabad, Faisalabad, Pakistan
| | - Muzzamal Hussain
- Department of Food Science, Government College University Faisalabad, Faisalabad, Pakistan
| | - Hafiz Wasiqe Amjad
- International Medical School, Jinggangshan University, Ji'an, Jiangxi, China
| | | |
Collapse
|
32
|
Tan DSY, Akelew Y, Snelson M, Nguyen J, O’Sullivan KM. Unravelling the Link between the Gut Microbiome and Autoimmune Kidney Diseases: A Potential New Therapeutic Approach. Int J Mol Sci 2024; 25:4817. [PMID: 38732038 PMCID: PMC11084259 DOI: 10.3390/ijms25094817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 04/13/2024] [Accepted: 04/18/2024] [Indexed: 05/13/2024] Open
Abstract
The gut microbiota and short chain fatty acids (SCFA) have been associated with immune regulation and autoimmune diseases. Autoimmune kidney diseases arise from a loss of tolerance to antigens, often with unclear triggers. In this review, we explore the role of the gut microbiome and how disease, diet, and therapy can alter the gut microbiota consortium. Perturbations in the gut microbiota may systemically induce the translocation of microbiota-derived inflammatory molecules such as liposaccharide (LPS) and other toxins by penetrating the gut epithelial barrier. Once in the blood stream, these pro-inflammatory mediators activate immune cells, which release pro-inflammatory molecules, many of which are antigens in autoimmune diseases. The ratio of gut bacteria Bacteroidetes/Firmicutes is associated with worse outcomes in multiple autoimmune kidney diseases including lupus nephritis, MPO-ANCA vasculitis, and Goodpasture's syndrome. Therapies that enhance SCFA-producing bacteria in the gut have powerful therapeutic potential. Dietary fiber is fermented by gut bacteria which in turn release SCFAs that protect the gut barrier, as well as modulating immune responses towards a tolerogenic anti-inflammatory state. Herein, we describe where the current field of research is and the strategies to harness the gut microbiome as potential therapy.
Collapse
Affiliation(s)
- Diana Shu Yee Tan
- Department of Medicine, Centre for Inflammatory Diseases, Monash University, Clayton, VIC 3168, Australia; (D.S.Y.T.); (Y.A.)
| | - Yibeltal Akelew
- Department of Medicine, Centre for Inflammatory Diseases, Monash University, Clayton, VIC 3168, Australia; (D.S.Y.T.); (Y.A.)
| | - Matthew Snelson
- School of Biological Science, Monash University, Clayton, VIC 3168, Australia;
| | - Jenny Nguyen
- The Alfred Centre, School of Translational Medicine, Monash University, Melbourne, VIC 3004, Australia
| | - Kim Maree O’Sullivan
- Department of Medicine, Centre for Inflammatory Diseases, Monash University, Clayton, VIC 3168, Australia; (D.S.Y.T.); (Y.A.)
| |
Collapse
|
33
|
Kwon Y, Cho KH, Ma S, Ko H, Hong GH, Lee SY, Park KY, Chung JA, Jeong SJ. Supplementation of Heat-Treated Lactiplantibacillus plantarum nF1 Changes the Production of Short-Chain Fatty Acids in Healthy Infants. J Nutr Metab 2024; 2024:5558566. [PMID: 38623309 PMCID: PMC11018375 DOI: 10.1155/2024/5558566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 03/11/2024] [Accepted: 03/27/2024] [Indexed: 04/17/2024] Open
Abstract
Background Imbalance of the gut microbiome and decrease in the number of short-chain fatty acid (SCFA)-producing bacteria often affect human health by altering intestinal and immune homeostasis. The use of probiotics has been shown to be an attractive method to modulate gut microbiota to prevent or treat intestinal dysbiosis. Likewise, this study aimed to determine whether the oral consumption of heat-treated Lactiplantibacillus plantarum nF1 (HLp-nF1) induces changes in the gut environment in healthy infants by measuring changes in fecal SCFAs. Methods The study enrolled 43 infants aged under 2 months, with 30 infants in the HLp-nF1 group receiving HLp-nF1 orally (2.5 × 1010 cells/g/pack, daily dose of two packs) for 8 weeks. The fecal samples were collected and the questionnaires were administered at weeks 0 and 8. Results The concentrations of the total SCFAs, acetate, propionate, and butyrate significantly increased following HLp-nF1 supplementation (P < 0.0001, P < 0.0001, P < 0.0001, and P=0.028, respectively). Conclusions Supplementation of HLp-nF1 has a positive effect on SCFA production and could be a potentially useful and straightforward method to manipulate SCFA formation.
Collapse
Affiliation(s)
- Yoowon Kwon
- Department of Pediatrics, Chungnam National University Sejong Hospital, Chungnam National University School of Medicine, Sejong, Republic of Korea
| | - Kee Hyun Cho
- Department of Pediatrics, Kangwon National University Hospital, Kangwon National University School of Medicine, Chuncheon, Republic of Korea
| | - Sangbae Ma
- AiBiotics Co Ltd, Changwon, Republic of Korea
| | - Hyelyun Ko
- AiBiotics Co Ltd, Changwon, Republic of Korea
| | | | | | - Kun-Young Park
- IMMUNOBIOTECH Corp, Seoul, Republic of Korea
- School of Integrated Medicine, CHA University, Seongnam, Republic of Korea
| | - Jin A. Chung
- Department of Pediatrics, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Republic of Korea
| | - Su Jin Jeong
- Department of Pediatrics, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Republic of Korea
| |
Collapse
|
34
|
Wu H, Ma W, Wang Y, Wang Y, Sun X, Zheng Q. Gut microbiome-metabolites axis: A friend or foe to colorectal cancer progression. Biomed Pharmacother 2024; 173:116410. [PMID: 38460373 DOI: 10.1016/j.biopha.2024.116410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 03/06/2024] [Indexed: 03/11/2024] Open
Abstract
An expanding corpus of research robustly substantiates the complex interrelation between gut microbiota and the onset, progression, and metastasis of colorectal cancer. Investigations in both animal models and human subjects have consistently underscored the role of gut bacteria in a variety of metabolic activities, driven by dietary intake. These activities include amino acid metabolism, carbohydrate fermentation, and the generation and regulation of bile acids. These metabolic derivatives, in turn, have been identified as significant contributors to the progression of colorectal cancer. This thorough review meticulously explores the dynamic interaction between gut bacteria and metabolites derived from the breakdown of amino acids, fatty acid metabolism, and bile acid synthesis. Notably, bile acids have been recognized for their potential carcinogenic properties, which may expedite tumor development. Extensive research has revealed a reciprocal influence of gut microbiota on the intricate spectrum of colorectal cancer pathologies. Furthermore, strategies to modulate gut microbiota, such as dietary modifications or probiotic supplementation, may offer promising avenues for both the prevention and adjunctive treatment of colorectal cancer. Nevertheless, additional research is imperative to corroborate these findings and enhance our comprehension of the underlying mechanisms in colorectal cancer development.
Collapse
Affiliation(s)
- Hao Wu
- Department of Immunology, Basic Medicine College, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, PR China
| | - Wenmeng Ma
- Department of Immunology, Basic Medicine College, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, PR China
| | - Yiyao Wang
- Department of Immunology, Basic Medicine College, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, PR China
| | - Yuanyuan Wang
- Department of anesthesiology, The Fourth Affiliated Hospital, China Medical University, Shenyang, Liaoning Province, PR China
| | - Xun Sun
- Department of Immunology, Basic Medicine College, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, PR China.
| | - Qianqian Zheng
- Department of Pathophysiology, Basic Medicine College, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, PR China.
| |
Collapse
|
35
|
Kong H, Yang J, Wang X, Mamat N, Xie G, Zhang J, Zhao H, Li J. The combination of Brassica rapa L. polysaccharides and cisplatin enhances the anti liver cancer effect and improves intestinal microbiota and metabolic disorders. Int J Biol Macromol 2024; 265:130706. [PMID: 38458274 DOI: 10.1016/j.ijbiomac.2024.130706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 02/24/2024] [Accepted: 03/05/2024] [Indexed: 03/10/2024]
Abstract
Polysaccharides are commonly used as low-toxicity anticancer active substances to enhance the chemotherapeutic effect of cisplatin and reduce toxicity. Brassica rapa L. polysaccharides have been shown to have hepatoprotective effects; however, their anticancer effects in combination with cisplatin and their mechanisms have not been reported. An acidic polysaccharide from Brassica rapa L. (BRCPe) using hydroalcohol precipitation-assisted sonication was Characterized. The effects of BRCPe combined with cisplatin treatment on tumor growth in hepatocellular carcinoma mouse model were investigated. The impact of the combined treatment on the composition of intestinal flora, levels of short-chain fatty acids and endogenous metabolites in tumor mice were analyzed based on macrogenomic and metabolomic data Our results showed that the BRCPe combined with low-dose Cisplatin group showed better inhibitory activity against hepatocellular carcinoma cell growth in terms of tumor volume, tumor weight, and tumor suppression rate compared with the BRCPe and Cisplation alone group, and reduced the side effects of cisplatin-induced body weight loss, immune deficiency, and liver injury. Furthermore, BRCPe combined with cisplatin was found to induce apoptosis in hepatocellular carcinoma cell through the activation of the caspase cascade reaction. In addition, the intervention of BRCPe were observed to modulate the composition, structure and functional structure of intestinal flora affected by cisplatin. Notably, Lachnospiraceae bacteria, Lactobacillus murinus, Muribaculaceae, and Clostridiales bacteria were identified as significant contributors to microbial species involved in metabolic pathways. Moreover, BRCPe effectively regulate the metabolic disorders in cisplatin-induced hepatocellular carcinoma mice. In conclusion, BRCPe could potentially function as an adjuvant or dietary supplement to augment the effectiveness of cisplatin chemotherapy through the preservation of a more efficient intestinal microenvironmental homeostasis.
Collapse
Affiliation(s)
- Hanrui Kong
- Xinjiang Key Laboratory of Special Species Conservation and Regulatory Biology, Key Laboratory of Special Environment Biodiversity Application and Regulation in Xinjiang, College of Life Science, Xinjiang Normal University, Urumqi 830054, China
| | - Jun Yang
- Xinjiang Key Laboratory of Special Species Conservation and Regulatory Biology, Key Laboratory of Special Environment Biodiversity Application and Regulation in Xinjiang, College of Life Science, Xinjiang Normal University, Urumqi 830054, China
| | - Xiaojing Wang
- Xinjiang Key Laboratory of Special Species Conservation and Regulatory Biology, Key Laboratory of Special Environment Biodiversity Application and Regulation in Xinjiang, College of Life Science, Xinjiang Normal University, Urumqi 830054, China
| | - Nuramina Mamat
- Xinjiang Key Laboratory of Special Species Conservation and Regulatory Biology, Key Laboratory of Special Environment Biodiversity Application and Regulation in Xinjiang, College of Life Science, Xinjiang Normal University, Urumqi 830054, China
| | - Guoxuan Xie
- Xinjiang Key Laboratory of Special Species Conservation and Regulatory Biology, Key Laboratory of Special Environment Biodiversity Application and Regulation in Xinjiang, College of Life Science, Xinjiang Normal University, Urumqi 830054, China
| | - Jing Zhang
- Xinjiang Key Laboratory of Special Species Conservation and Regulatory Biology, Key Laboratory of Special Environment Biodiversity Application and Regulation in Xinjiang, College of Life Science, Xinjiang Normal University, Urumqi 830054, China
| | - Huixin Zhao
- Xinjiang Key Laboratory of Special Species Conservation and Regulatory Biology, Key Laboratory of Special Environment Biodiversity Application and Regulation in Xinjiang, College of Life Science, Xinjiang Normal University, Urumqi 830054, China.
| | - Jinyu Li
- Xinjiang Key Laboratory of Special Species Conservation and Regulatory Biology, Key Laboratory of Special Environment Biodiversity Application and Regulation in Xinjiang, College of Life Science, Xinjiang Normal University, Urumqi 830054, China.
| |
Collapse
|
36
|
Wu J, Chen N, Grau E, Johnson L, Liu Y, Li C, Scott PA, Kim C, Sun D, Kaplan HJ, Shao H. Short chain fatty acids inhibit corneal inflammatory responses to TLR ligands via the ocular G-protein coupled receptor 43. Ocul Surf 2024; 32:48-57. [PMID: 38224777 PMCID: PMC11056309 DOI: 10.1016/j.jtos.2024.01.005] [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: 09/09/2023] [Revised: 11/22/2023] [Accepted: 01/12/2024] [Indexed: 01/17/2024]
Abstract
PURPOSE Short chain fatty acids (SCFAs) produced by gut microbiota are known to play primary roles in gut homeostasis by immunomodulation partially through G-protein coupled receptors (GPR) 43. Using mouse models of TLR ligand induced keratitis, we investigated whether SCFAs and GPR43 play any regulatory roles in the pathogenesis of inflammatory responses in the eye. METHODS Both human and mouse eyes were labeled with a specific antibody for GPR43 and imaged by a laser scanning confocal microscope. Corneal cups from naïve C57BL/6J (B6) and GPR43 knockout (KO) mice were stimulated with TLR ligands in the presence or absence of sodium butyrate overnight and then processed for RT-PCR assay for expression of GPR43 and cytokines. Keratitis was induced by Poly I:C in wild type (WT) B6, GPR43KO and chimeric mice and the disease severity was evaluated by the corneal fluorescein staining test, and infiltrating cell staining and calculating in corneal whole mount. RESULTS GPR43 is expressed in both human and mouse eyes and the expression is bidirectionally regulated by TLR ligands and butyrate. Butyrate significantly inhibited inflammation caused by several TLR ligands such as Poly I:C, Flagellin, and CpG-ODN (TLR-3, 5 and 9 agonists, respectively) in WT, but not GPR43KO, mice. Butyrate inhibition of TLR-induced keratitis is mediated by the GPR43 expressed in tissue but not hematopoietic, cells. CONCLUSIONS This is the first report to demonstrate of the protective effect of SCFAs on microbial keratitis, and the dynamic expression and anti-inflammatory function of GPR43 in the eye. SCFAs can modulate inflammation and immunity in the eye through GPR43.
Collapse
Affiliation(s)
- Jun Wu
- Department of Ophthalmology and Visual Sciences, Kentucky Lions Eye Center, University of Louisville, School of Medicine, Louisville, KY, USA
| | - Nu Chen
- Department of Ophthalmology and Visual Sciences, Kentucky Lions Eye Center, University of Louisville, School of Medicine, Louisville, KY, USA
| | - Elizabeth Grau
- Department of Ophthalmology and Visual Sciences, Kentucky Lions Eye Center, University of Louisville, School of Medicine, Louisville, KY, USA
| | - Luke Johnson
- Department of Ophthalmology and Visual Sciences, Kentucky Lions Eye Center, University of Louisville, School of Medicine, Louisville, KY, USA
| | - Yongqing Liu
- Department of Medicine-oncology, University of Louisville, School of Medicine, Louisville, KY, USA
| | - Chi Li
- Department of Medicine-oncology, University of Louisville, School of Medicine, Louisville, KY, USA
| | - Patrick A Scott
- Department of Ophthalmology and Visual Sciences, Kentucky Lions Eye Center, University of Louisville, School of Medicine, Louisville, KY, USA
| | - Chang Kim
- Department of Pathology, Mary H. Weiser Food Allergy Center, 4025 Biomedical Science Research Building, 109 Zina Pitcher Place, Ann Arbor, MI, USA
| | - Deming Sun
- Doheny Eye Institute & Department Ophthalmology, David Geffen School of Medicine/UCLA, Los Angeles, CA, USA
| | - Henry J Kaplan
- Department of Ophthalmology and Biochemistry & Molecular Biology, St. Louis University School of Medicine, St. Louis, Missouri, USA
| | - Hui Shao
- Department of Ophthalmology and Visual Sciences, Kentucky Lions Eye Center, University of Louisville, School of Medicine, Louisville, KY, USA.
| |
Collapse
|
37
|
Yokoyama Y, Ichiki T, Yamakawa T, Tsuji Y, Kuronuma K, Takahashi S, Narimatsu E, Katanuma A, Nakase H. Gut microbiota and metabolites in patients with COVID-19 are altered by the type of SARS-CoV-2 variant. Front Microbiol 2024; 15:1358530. [PMID: 38505560 PMCID: PMC10948395 DOI: 10.3389/fmicb.2024.1358530] [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: 12/19/2023] [Accepted: 02/21/2024] [Indexed: 03/21/2024] Open
Abstract
Introduction Patients with COVID-19 have dysbiosis of the intestinal microbiota with altered metabolites in the stool. However, it remains unclear whether the differences among SARS-CoV-2 variants lead to differences in intestinal microbiota and metabolites. Thus, we compared the microbiome and metabolome changes for each SARS-CoV-2 variant in patients with COVID-19. Materials and methods We conducted a multicenter observational study of patients with COVID-19 and performed fecal microbiome, metabolome, and calprotectin analyses and compared the results among the different SARS-CoV-2 variants. Results Twenty-one patients with COVID-19 were enrolled and stratified according to the SARS-CoV-2 strain: six with the Alpha, 10 with the Delta, and five with the Omicron variant. Fecal microbiome analysis showed that α-diversity was reduced in the order of the Omicron, Delta, and Alpha variants (p = 0.07). Linear discriminant analysis revealed differences in the abundance of short-chain fatty acid-producing gut microbiota for each SARS-CoV-2 variant. Fecal metabolome analysis showed that the Omicron and Delta variants had markedly reduced propionic and lactic acid levels compared to the Alpha strain (p < 0.05). Conclusion The intestinal microbiota of patients with COVID-19 varies depending on the SARS-CoV-2 variant. Dysbiosis of the intestinal microbiota due to differences in SARS-CoV-2 variants causes a decrease in intestinal short-chain fatty acids.
Collapse
Affiliation(s)
- Yoshihiro Yokoyama
- Department of Gastroenterology and Hepatology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Tomoko Ichiki
- Department of General Medicine, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Tsukasa Yamakawa
- Department of Gastroenterology and Hepatology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Yoshihisa Tsuji
- Department of General Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Koji Kuronuma
- Department of Respiratory Medicine and Allergology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Satoshi Takahashi
- Department of Infection Control and Laboratory Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Eichi Narimatsu
- Department of Intensive Care Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Akio Katanuma
- Center for Gastroenterology, Teine-Keijinkai Hospital, Sapporo, Japan
| | - Hiroshi Nakase
- Department of Gastroenterology and Hepatology, Sapporo Medical University School of Medicine, Sapporo, Japan
| |
Collapse
|
38
|
Satheesh Babu AK, Petersen C, Paz HA, Iglesias-Carres L, Li Y, Zhong Y, Neilson AP, Wankhade UD, Anandh Babu PV. Gut Microbiota Depletion Using Antibiotics to Investigate Diet-Derived Microbial Metabolites: An Efficient Strategy. Mol Nutr Food Res 2024; 68:e2300386. [PMID: 38054624 DOI: 10.1002/mnfr.202300386] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 09/07/2023] [Indexed: 12/07/2023]
Abstract
SCOPE Gut microbiota depletion using antibiotics in drinking water is a valuable tool to investigate the role of gut microbes and microbial metabolites in health and disease. However, there are challenges associated with this model. Animals avoid drinking water because of the antibiotic bitterness, which affects their metabolic health. The present study develops an efficient strategy to deplete gut microbes without affecting metabolic parameters. METHODS AND RESULTS Male C57BL/6J mice (7 weeks old) are fed a control (C) or high-fat (HF) diet. Subgroups of C and HF mice receive an antibiotic cocktail in drinking water (CA and HA). The antibiotic dosage is gradually increased so that the animals adapt to the taste of antibiotics. Metabolic parameters, gut microbiome, and microbial metabolites are assessed after 12 weeks treatment. Culture methods and 16s rRNA amplification confirm the depletion of gut microbes in antibiotic groups (CA and HA). Further, antibiotic treatment does not alter metabolic parameters (body weight, body fat, lean body mass, blood glucose, and glucose/insulin tolerance), whereas it suppresses the production of diet-derived microbial metabolites (trimethylamine and trimethylamine-N-oxide). CONCLUSION This strategy effectively depletes gut microbes and suppresses the production of microbial metabolites in mice without affecting their metabolic health.
Collapse
Affiliation(s)
| | - Chrissa Petersen
- Department of Nutrition and Integrative Physiology, College of Health, University of Utah, Salt Lake City, UT, 84112, USA
| | - Henry A Paz
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, 72205, AR, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Lisard Iglesias-Carres
- Plants for Human Health Institute, Department of Food, Bioprocessing and Nutrition Sciences, North Carolina State University, Kannapolis, NC, 28081, USA
| | - Ying Li
- Department of Nutrition and Integrative Physiology, College of Health, University of Utah, Salt Lake City, UT, 84112, USA
| | - Ying Zhong
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, 72205, AR, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Andrew P Neilson
- Plants for Human Health Institute, Department of Food, Bioprocessing and Nutrition Sciences, North Carolina State University, Kannapolis, NC, 28081, USA
| | - Umesh D Wankhade
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, 72205, AR, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Pon Velayutham Anandh Babu
- Department of Nutrition and Integrative Physiology, College of Health, University of Utah, Salt Lake City, UT, 84112, USA
| |
Collapse
|
39
|
Rodrigues SG, van der Merwe S, Krag A, Wiest R. Gut-liver axis: Pathophysiological concepts and medical perspective in chronic liver diseases. Semin Immunol 2024; 71:101859. [PMID: 38219459 DOI: 10.1016/j.smim.2023.101859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 10/11/2023] [Accepted: 12/04/2023] [Indexed: 01/16/2024]
Affiliation(s)
- Susana G Rodrigues
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Switzerland
| | - Schalk van der Merwe
- Department of Gastroenterology and Hepatology, University hospital Gasthuisberg, University of Leuven, Belgium
| | - Aleksander Krag
- Institute of Clinical Research, University of Southern Denmark, Odense, Denmark; Centre for Liver Research, Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark, University of Southern Denmark, Odense, Denmark
| | - Reiner Wiest
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Switzerland.
| |
Collapse
|
40
|
McEvoy CT, Jennings A, Steves CJ, Macgregor A, Spector T, Cassidy A. Diet patterns and cognitive performance in a UK Female Twin Registry (TwinsUK). Alzheimers Res Ther 2024; 16:17. [PMID: 38263271 PMCID: PMC10804649 DOI: 10.1186/s13195-024-01387-x] [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: 07/27/2023] [Accepted: 01/04/2024] [Indexed: 01/25/2024]
Abstract
BACKGROUND Plant-based diets may provide protection against cognitive decline and Alzheimer's disease, but observational data have not been consistent. Previous studies include early life confounding from socioeconomic conditions and genetics that are known to influence both cognitive performance and diet behaviour. This study investigated associations between Mediterranean (MED) diet and MIND diets and cognitive performance accounting for shared genotype and early-life environmental exposures in female twins. METHODS Diet scores were examined in 509 female twins enrolled in TwinsUK study. The Cambridge Neuropsychological Test Automated Battery was used to assess cognition at baseline and 10 years later (in n = 275). A co-twin case-control study for discordant monozygotic (MZ) twins examined effects of diet on cognitive performance independent of genetic factors. Differences in relative abundance of taxa at 10-year follow-up were explored in subsamples. RESULTS Each 1-point increase in MIND or MED diet score was associated with 1.75 (95% CI: - 2.96, - 0.54, p = 0.005 and q = 0.11) and 1.67 (95% CI: - 2.71, - 0.65, p = 0.002 and q = 0.02) fewer respective errors in paired-associates learning. Within each MZ pair, the twin with the high diet score had better preservation in spatial span especially for MED diet (p = 0.02). There were no differences between diet scores and 10-year change in the other cognitive tests. MIND diet adherence was associated with higher relative abundance of Ruminococcaceae UCG-010 (0.30% (95% CI 0.17, 0.62), q = 0.05) which was also associated with less decline in global cognition over 10 years (0.22 (95% CI 0.06, 0.39), p = 0.01). CONCLUSIONS MIND or MED diets could help to preserve some cognitive abilities in midlife, particularly episodic and visuospatial working memory. Effects may be mediated by high dietary fibre content and increased abundance of short-chain fatty acid producing gut bacteria. Longer follow-up with repeated measures of cognition will determine whether diet can influence changes in cognition occurring in older age.
Collapse
Affiliation(s)
- Claire T McEvoy
- The Institute for Global Food Security, Queen's University Belfast, Belfast, Northern Ireland, UK.
- The Global Brain Health Institute, University of California San Francisco, San Francisco, CA, USA.
- The Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland.
- Centre for Public Health, Institute of Clinical Sciences B, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, Northern Ireland, BT12 6BJ, UK.
| | - Amy Jennings
- The Institute for Global Food Security, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Claire J Steves
- Department of Twin Research & Genetic Epidemiology, King's College London, St Thomas' Campus, London, UK
| | | | - Tim Spector
- Department of Twin Research & Genetic Epidemiology, King's College London, St Thomas' Campus, London, UK
| | - Aedin Cassidy
- The Institute for Global Food Security, Queen's University Belfast, Belfast, Northern Ireland, UK
| |
Collapse
|
41
|
Cheng J, Zhou J. Unraveling the gut health puzzle: exploring the mechanisms of butyrate and the potential of High-Amylose Maize Starch Butyrate (HAMSB) in alleviating colorectal disturbances. Front Nutr 2024; 11:1285169. [PMID: 38304546 PMCID: PMC10830644 DOI: 10.3389/fnut.2024.1285169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 01/02/2024] [Indexed: 02/03/2024] Open
Abstract
Colorectal disturbances encompass a variety of disorders that impact the colon and rectum, such as colitis and colon cancer. Butyrate, a short-chain fatty acid, plays a pivotal role in supporting gut health by nourishing colonocytes, promoting barrier function, modulating inflammation, and fostering a balanced microbiome. Increasing colorectal butyrate concentration may serve as a critical strategy to improve colon function and reduce the risk of colorectal disturbances. Butyrylated high-amylose maize starch (HAMSB) is an edible ingredient that efficiently delivers butyrate to the colon. HAMSB is developed by esterifying a high-amylose starch backbone with butyric anhydride. With a degree of substitution of 0.25, each hydroxy group of HAMSB is substituted by a butyryl group in every four D-glucopyranosyl units. In humans, the digestibility of HAMSB is 68% (w/w), and 60% butyrate molecules attached to the starch backbone is absorbed by the colon. One clinical trial yielded two publications, which showed that HAMSB significantly reduced rectal O6-methyl-guanine adducts and epithelial proliferation induced by the high protein diet. Fecal microbial profiles were assessed in three clinical trials, showing that HAMSB supplementation was consistently linked to increased abundance of Parabacteroides distasonis. In animal studies, HAMSB was effective in reducing the risk of diet- or AOM-induced colon cancer by reducing genetic damage, but the mechanisms differed. HAMSB functioned through affecting cecal ammonia levels by modulating colon pH in diet-induced cancer, while it ameliorated chemical-induced colon cancer through downregulating miR19b and miR92a expressions and subsequently activating the caspase-dependent apoptosis. Furthermore, animal studies showed that HAMSB improved colitis via regulating the gut immune modulation by inhibiting histone deacetylase and activating G protein-coupled receptors, but its role in bacteria-induced colon colitis requires further investigation. In conclusion, HAMSB is a food ingredient that may deliver butyrate to the colon to support colon health. Further clinical trials are warranted to validate earlier findings and determine the minimum effective dose of HAMSB.
Collapse
Affiliation(s)
- Junrui Cheng
- Global Scientific and Regulatory Department, Ingredion Incorporated, Bridgewater, NJ, United States
| | - Jing Zhou
- Global Scientific and Regulatory Department, Ingredion Incorporated, Bridgewater, NJ, United States
| |
Collapse
|
42
|
Caparrós-Martín JA, Maher P, Ward NC, Saladié M, Agudelo-Romero P, Stick SM, Chan DC, Watts GF, O’Gara F. An Analysis of the Gut Microbiota and Related Metabolites following PCSK9 Inhibition in Statin-Treated Patients with Elevated Levels of Lipoprotein(a). Microorganisms 2024; 12:170. [PMID: 38257996 PMCID: PMC10818477 DOI: 10.3390/microorganisms12010170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 12/26/2023] [Accepted: 01/08/2024] [Indexed: 01/24/2024] Open
Abstract
BACKGROUND Atherosclerotic cardiovascular disease (ASCVD) is a leading cause of global mortality, often associated with high blood levels of LDL cholesterol (LDL-c). Medications like statins and PCSK9 inhibitors, are used to manage LDL-c levels and reduce ASCVD risk. Recent findings connect the gut microbiota and its metabolites to ASCVD development. We showed that statins modulate the gut microbiota including the production of microbial metabolites involved in the regulation of cholesterol metabolism such as short chain fatty acids (SCFAs) and bile acids (BAs). Whether this pleiotropic effect of statins is associated with their antimicrobial properties or it is secondary to the modulation of cholesterol metabolism in the host is unknown. In this observational study, we evaluated whether alirocumab, a PCSK9 inhibitor administered subcutaneously, alters the stool-associated microbiota and the profiles of SCFAs and BAs. METHODS We used stool and plasma collected from patients enrolled in a single-sequence study using alirocumab. Microbial DNA was extracted from stool, and the bacterial component of the gut microbiota profiled following an amplicon sequencing strategy targeting the V3-V4 region of the 16S rRNA gene. Bile acids and SCFAs were profiled and quantified in stool and plasma using mass spectrometry. RESULTS Treatment with alirocumab did not alter bacterial alpha (Shannon index, p = 0.74) or beta diversity (PERMANOVA, p = 0.89) in feces. Similarly, circulating levels of SCFAs (mean difference (95% confidence interval (CI)), 8.12 [-7.15-23.36] µM, p = 0.25) and BAs (mean difference (95% CI), 0.04 [-0.11-0.19] log10(nmol mg-1 feces), p = 0.56) were equivalent regardless of PCSK9 inhibition. Alirocumab therapy was associated with increased concentration of BAs in feces (mean difference (95% CI), 0.20 [0.05-0.34] log10(nmol mg-1 feces), p = 0.01). CONCLUSION In statin-treated patients, the use of alirocumab to inhibit PCSK9 leads to elevated levels of fecal BAs without altering the bacterial population of the gut microbiota. The association of alirocumab with increased fecal BA concentration suggests an additional mechanism for the cholesterol-lowering effect of PCSK9 inhibition.
Collapse
Affiliation(s)
- Jose A. Caparrós-Martín
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, Perth, WA 6009, Australia
- Curtin Health Innovation Research Institute (CHIRI), Curtin University, Perth, WA 6102, Australia
| | - Patrice Maher
- Curtin Health Innovation Research Institute (CHIRI), Curtin University, Perth, WA 6102, Australia
| | - Natalie C. Ward
- Dobney Hypertension Centre, Medical School, The University of Western Australia, Perth, WA 6009, Australia
| | - Montserrat Saladié
- Curtin Health Innovation Research Institute (CHIRI), Curtin University, Perth, WA 6102, Australia
| | - Patricia Agudelo-Romero
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, Perth, WA 6009, Australia
- The University of Western Australia, Perth, WA 6009, Australia
| | - Stephen M. Stick
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, Perth, WA 6009, Australia
- The University of Western Australia, Perth, WA 6009, Australia
- Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, WA 6008, Australia
| | - Dick C. Chan
- Medical School, Faculty of Health and Medical Sciences, The University of Western Australia, Perth, WA 6009, Australia
| | - Gerald F. Watts
- Medical School, Faculty of Health and Medical Sciences, The University of Western Australia, Perth, WA 6009, Australia
- Cardiometabolic Service, Departments of Cardiology and Internal Medicine, Royal Perth Hospital, Perth, WA 6000, Australia
| | - Fergal O’Gara
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, Perth, WA 6009, Australia
- Curtin Health Innovation Research Institute (CHIRI), Curtin University, Perth, WA 6102, Australia
- BIOMERIT Research Centre, School of Microbiology, University College Cork, T12 XF62 Cork, Ireland
| |
Collapse
|
43
|
Zhang K, Wang Y, Cui X, Wang W, Li Y. Features of Metabolite Changes in Disease Evolution in Cholecystolithiasis. Dig Dis Sci 2024; 69:275-288. [PMID: 37943386 PMCID: PMC10787879 DOI: 10.1007/s10620-023-08134-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 09/28/2023] [Indexed: 11/10/2023]
Abstract
BACKGROUND Cholecystolithiasis is defined as a disease caused by complex and changeable factors. Advanced age, female sex, and a hypercaloric diet rich in carbohydrates and poor in fiber, together with obesity and genetic factors, are the main factors that may predispose people to choledocholithiasis. However, serum biomarkers for the rapid diagnosis of choledocholithiasis remain unclear. AIMS This study was designed to explore the pathogenesis of cholecystolithiasis and identify the possible metabolic and lipidomic biomarkers for the diagnosis of the disease. METHODS Using UHPLC-MS/MS and GC-MS, we detected the serum of 28 cholecystolithiasis patients and 19 controls. Statistical analysis of multiple variables included Principal Component Analysis (PCA). Visualization of differential metabolites was performed using volcano plots. The screened differential metabolites were further analyzed using clustering heatmaps. The quality of the model was assessed using random forests. RESULTS In this study, dramatically altered lipid homeostasis was detected in cholecystolithiasis group. In addition, the levels of short-chain fatty acids and amino acids were noticeably changed in patients with cholecystolithiasis. They detected higher levels of FFA.18.1, FFA.20.1, LPC16.0, and LPC20.1, but lower levels of 1-Methyl-L-histidine and 4-Hydroxyproline. In addition, glycine and L-Tyrosine were higher in choledocholithiasis group. Analyses of metabolic serum in affected patients have the potential to develop an integrated metabolite-based biomarker model that can facilitate the early diagnosis and treatment of the disease. CONCLUSION Our results highlight the value of integrating lipid, amino acid, and short-chain fatty acid to explore the pathophysiology of cholecystolithiasis disease, and consequently, improve clinical decision-making.
Collapse
Affiliation(s)
- Kun Zhang
- Shanghai Biotree Biotech Co. Ltd., Shanghai, China
- Institute of Basic Medical Sciences, The Second Hospital of Shandong University, Shandong, 250033, China
| | - Yongzheng Wang
- Department of Interventional, The Second Hospital of Shandong University, Shandong, 250033, China
| | - Xiaoxuan Cui
- Shanghai Biotree Biotech Co. Ltd., Shanghai, China
| | - Wei Wang
- Department of Interventional, The Second Hospital of Shandong University, Shandong, 250033, China.
| | - Yuliang Li
- Department of Interventional, The Second Hospital of Shandong University, Shandong, 250033, China
| |
Collapse
|
44
|
Bleich RM, Li C, Sun S, Ahn JH, Dogan B, Barlogio CJ, Broberg CA, Franks AR, Bulik-Sullivan E, Carroll IM, Simpson KW, Fodor AA, Arthur JC. A consortia of clinical E. coli strains with distinct in vitro adherent/invasive properties establish their own co-colonization niche and shape the intestinal microbiota in inflammation-susceptible mice. MICROBIOME 2023; 11:277. [PMID: 38124090 PMCID: PMC10731797 DOI: 10.1186/s40168-023-01710-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 10/26/2023] [Indexed: 12/23/2023]
Abstract
BACKGROUND Inflammatory bowel disease (IBD) patients experience recurrent episodes of intestinal inflammation and often follow an unpredictable disease course. Mucosal colonization with adherent-invasive Escherichia coli (AIEC) are believed to perpetuate intestinal inflammation. However, it remains unclear if the 24-year-old AIEC in vitro definition fully predicts mucosal colonization in vivo. To fill this gap, we have developed a novel molecular barcoding approach to distinguish strain variants in the gut and have integrated this approach to explore mucosal colonization of distinct patient-derived E. coli isolates in gnotobiotic mouse models of colitis. RESULTS Germ-free inflammation-susceptible interleukin-10-deficient (Il10-/-) and inflammation-resistant WT mice were colonized with a consortium of AIEC and non-AIEC strains, then given a murine fecal transplant to provide niche competition. E. coli strains isolated from human intestinal tissue were each marked with a unique molecular barcode that permits identification and quantification by barcode-targeted sequencing. 16S rRNA sequencing was used to evaluate the microbiome response to E. coli colonization. Our data reveal that specific AIEC and non-AIEC strains reproducibly colonize the intestinal mucosa of WT and Il10-/- mice. These E. coli expand in Il10-/- mice during inflammation and induce compositional dysbiosis to the microbiome in an inflammation-dependent manner. In turn, specific microbes co-evolve in inflamed mice, potentially diversifying E. coli colonization patterns. We observed no selectivity in E. coli colonization patterns in the fecal contents, indicating minimal selective pressure in this niche from host-microbe and interbacterial interactions. Because select AIEC and non-AIEC strains colonize the mucosa, this suggests the in vitro AIEC definition may not fully predict in vivo colonization potential. Further comparison of seven E. coli genomes pinpointed unique genomic features contained only in highly colonizing strains (two AIEC and two non-AIEC). Those colonization-associated features may convey metabolic advantages (e.g., iron acquisition and carbohydrate consumption) to promote efficient mucosal colonization. CONCLUSIONS Our findings establish the in vivo mucosal colonizer, not necessarily AIEC, as a principal dysbiosis driver through crosstalk with host and associated microbes. Furthermore, we highlight the utility of high-throughput screens to decode the in vivo colonization dynamics of patient-derived bacteria in murine models. Video Abstract.
Collapse
Affiliation(s)
- Rachel M Bleich
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Biology, Appalachian State University, Boone, NC, USA
| | - Chuang Li
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Shan Sun
- College of Computing and Informatics, University of North Carolina at Charlotte, Charlotte, NC, USA
| | - Ju-Hyun Ahn
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Belgin Dogan
- Department of Clinical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY, USA
| | - Cassandra J Barlogio
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Christopher A Broberg
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Adrienne R Franks
- Center for Gastrointestinal Biology & Disease, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Emily Bulik-Sullivan
- Center for Gastrointestinal Biology & Disease, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ian M Carroll
- Center for Gastrointestinal Biology & Disease, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kenneth W Simpson
- Department of Clinical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY, USA
| | - Anthony A Fodor
- College of Computing and Informatics, University of North Carolina at Charlotte, Charlotte, NC, USA
| | - Janelle C Arthur
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Center for Gastrointestinal Biology & Disease, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
45
|
Tang X, de Vos P. Structure-function effects of different pectin chemistries and its impact on the gastrointestinal immune barrier system. Crit Rev Food Sci Nutr 2023; 65:1201-1215. [PMID: 38095591 DOI: 10.1080/10408398.2023.2290230] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
The gastrointestinal immune system is crucial for overall health, safeguarding the human body against harmful substances and pathogens. One key player in this defense is dietary fiber pectin, which supports the gut's immune barrier and fosters beneficial gut bacteria. Pectin's composition, including degree of methylation (DM), RG-I, and neutral sugar content, influences its health benefits. This review assesses how pectin composition impacts the gastrointestinal immune barrier and what advantages specific chemistries of pectin has for metabolic, cardiovascular, and immune health. We delve into recent findings regarding pectin's interactions with the immune system, including receptors like TLRs and galectin 3. Pectin is shown to fortify mucosal and epithelial layers, but the specific effects are structure dependent. Additionally, we explore potential strategies for enhancing the gut immune barrier function. Understanding how distinct pectin chemistries affect the gastrointestinal immune system is vital for developing preventive and therapeutic solutions for conditions related to microbiota imbalances and immune issues. Ultimately, this review offers insights into strategies to boost the gut immune barrier's effectiveness, fostering better overall health by using specific pectins in the diet.
Collapse
Affiliation(s)
- X Tang
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - P de Vos
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
46
|
Zhang J, Wu G, Tang Y, Liu H, Ge X, Peng R, Cao J, Tu D, Su B, Jin S, Jiang G, Zhang C, Bai D. Causal associations between gut microbiota and primary biliary cholangitis: a bidirectional two-sample Mendelian randomization study. Front Microbiol 2023; 14:1273024. [PMID: 38033598 PMCID: PMC10684913 DOI: 10.3389/fmicb.2023.1273024] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Accepted: 10/31/2023] [Indexed: 12/02/2023] Open
Abstract
Background Previous studies have suggested an association between gut microbiota and primary biliary cholangitis (PBC). Nonetheless, the causal relationship between gut microbiota and PBC risk remains unclear. Methods A bidirectional two-sample Mendelian Randomization (MR) study was employed using summary statistical data for gut microbiota and PBC from the MiBioGen consortium and Genome-Wide Association Studies (GWAS) database to investigate causal relationships between 211 gut microbiota and PBC risk. Inverse variance weighted (IVW) method was the primary analytical approach to assess causality, and the pleiotropy and heterogeneity tests were employed to verify the robustness of the findings. Additionally, we performed reverse MR analyses to investigate the possibility of the reverse causal association. Results The IVW method identified five gut microbiota that demonstrated associations with the risk of PBC. Order Selenomonadales [odds ratio (OR) 2.13, 95% confidence interval (CI) 1.10-4.14, p = 0.03], Order Bifidobacteriales (OR 1.58, 95% CI 1.07-2.33, p = 0.02), and Genus Lachnospiraceae_UCG_004 (OR 1.64, 95%CI 1.06-2.55, p = 0.03) were correlated with a higher risk of PBC, while Family Peptostreptococcaceae (OR 0.65, 95%CI 0.43-0.98, p = 0.04) and Family Ruminococcaceae (OR 0.33, 95%CI 0.15-0.72, p = 0.01) had a protective effect on PBC. The reverse MR analysis demonstrated no statistically significant relationship between PBC and these five specific gut microbial taxa. Conclusion This study revealed that there was a causal relationship between specific gut microbiota taxa and PBC, which may provide novel perspectives and a theoretical basis for the clinical prevention, diagnosis, and treatment of PBC.
Collapse
Affiliation(s)
- Jiahao Zhang
- Department of Hepatobiliary Surgery, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, China
- The Yangzhou School of Clinical Medicine of Dalian Medical University, Dalian, Liaoning, China
| | - Gefeng Wu
- Department of Hepatobiliary Surgery, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, China
- The Yangzhou School of Clinical Medicine of Dalian Medical University, Dalian, Liaoning, China
| | - Yuhong Tang
- Department of Hepatobiliary Surgery, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, China
| | - Huanxiang Liu
- Department of Hepatobiliary Surgery, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, China
| | - Xinyu Ge
- Department of Hepatobiliary Surgery, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, China
- The Yangzhou School of Clinical Medicine of Dalian Medical University, Dalian, Liaoning, China
| | - Rui Peng
- Department of Hepatobiliary Surgery, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, China
| | - Jun Cao
- Department of Hepatobiliary Surgery, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, China
| | - Daoyuan Tu
- Department of Hepatobiliary Surgery, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, China
| | - Bingbing Su
- Department of Hepatobiliary Surgery, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, China
| | - Shengjie Jin
- Department of Hepatobiliary Surgery, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, China
| | - Guoqing Jiang
- Department of Hepatobiliary Surgery, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, China
| | - Chi Zhang
- Department of Hepatobiliary Surgery, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, China
| | - Dousheng Bai
- Department of Hepatobiliary Surgery, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, China
| |
Collapse
|
47
|
Cai Y, Yang Q, Yu Y, Yang F, Bai R, Fan X. Efficacy and underlying mechanisms of berberine against lipid metabolic diseases: a review. Front Pharmacol 2023; 14:1283784. [PMID: 38034996 PMCID: PMC10684937 DOI: 10.3389/fphar.2023.1283784] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 11/03/2023] [Indexed: 12/02/2023] Open
Abstract
Lipid-lowering therapy is an important tool for the treatment of lipid metabolic diseases, which are increasing in prevalence. However, the failure of conventional lipid-lowering drugs to achieve the desired efficacy in some patients, and the side-effects of these drug regimens, highlight the urgent need for novel lipid-lowering drugs. The liver and intestine are important in the production and removal of endogenous and exogenous lipids, respectively, and have an important impact on circulating lipid levels. Elevated circulating lipids predisposes an individual to lipid deposition in the vascular wall, affecting vascular function. Berberine (BBR) modulates liver lipid production and clearance by regulating cellular targets such as cluster of differentiation 36 (CD36), acetyl-CoA carboxylase (ACC), microsomal triglyceride transfer protein (MTTP), scavenger receptor class B type 1 (SR-BI), low-density lipoprotein receptor (LDLR), and ATP-binding cassette transporter A1 (ABCA1). It influences intestinal lipid synthesis and metabolism by modulating gut microbiota composition and metabolism. Finally, BBR maintains vascular function by targeting proteins such as endothelial nitric oxide synthase (eNOS) and lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1). This paper elucidates and summarizes the pharmacological mechanisms of berberine in lipid metabolic diseases from a multi-organ (liver, intestine, and vascular system) and multi-target perspective.
Collapse
Affiliation(s)
- Yajie Cai
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qiaoning Yang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- NMPA Key Laboratory for Clinical Research and Evaluation of Traditional Chinese Medicine, Beijing, China
| | - Yanqiao Yu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Department of Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Furong Yang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ruina Bai
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaodi Fan
- Institute of Basic Medical Sciences, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
- Key Laboratory of Pharmacology of Chinese Materia Medica, Beijing, China
| |
Collapse
|
48
|
Lan Y, Pan S, Chen B, Zhou F, Yang F, Chao S, Hua Y, Liu H. The relationship between gut microbiota, short-chain fatty acids, and glucolipid metabolism in pregnant women with large for gestational age infants. J Appl Microbiol 2023; 134:lxad240. [PMID: 37883533 DOI: 10.1093/jambio/lxad240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 10/02/2023] [Accepted: 10/21/2023] [Indexed: 10/28/2023]
Abstract
AIM To elucidate the association between gut microbiota, short-chain fatty acids (SCFAs), and glucolipid metabolism in women with large for gestational age (LGA) infants. METHODS AND RESULTS A single-center, observational prospective cohort study was performed at a tertiary hospital in Wenzhou, China. Normal pregnant women were divided into LGA group and appropriate for gestational age (AGA) group according to the neonatal birth weight. Fecal samples were collected from each subject before delivery for the analysis of gut microbiota composition (GMC) and SCFAs. Blood samples were obtained at 24-28 weeks of gestation age to measure fasting blood glucose and fasting insulin levels, as well as just before delivery to assess serum triglycerides, total cholesterol, high-density lipoprotein (HDL), and low-density lipoprotein. The GMC exhibited differences at various taxonomic levels. Within the Firmicutes phylum, genus Lactobacillus, genus Clostridium, species Lactobacillus agil, and species Lactobacillus salivarius were enriched in the LGA group. Microbispora at genus level, Microbispora rosea at species level belonging to the Actinobacteria phylum, Neisseriales at order level, Bartonellaceae at family level, Paracoccus aminovorans, and Methylobacterium at genus level from the Proteobacteria phylum were more abundant in the LGA group. In contrast, within the Bacteroidetes phylum, Prevotella at genus level and Parabacteroides distasonis at species level were enriched in the AGA group. Although there were few differences observed in SCFA levels and most glucolipid metabolism indicators between the two groups, the serum HDL level was significantly lower in the LGA group compared to the AGA group. No significant relevance among GMC, SCFAs, and glucolipid metabolism indicators was found in the LGA group or in the AGA group. CONCLUSIONS Multiple different taxa, especially phylum Firmicutes, genus Prevotella, and genus Clostridium, might play an important role in excessive fetal growth, and LGA might be associated with the lower serum HDL level.
Collapse
Affiliation(s)
- Yehui Lan
- Department of Obstetrics and Gynecology and General Surgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Shuangjia Pan
- Department of Obstetrics and Gynecology and General Surgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Baoyi Chen
- Department of Obstetrics and Gynecology and General Surgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Feifei Zhou
- Department of Obstetrics and Gynecology, The Wenzhou Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou 325027, China
| | - Fan Yang
- Key Laboratory of Cell Engineering in Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi 563000,China
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200240, China
- Research Center for Lin He Academician New Medicine, Institutes for Shanghai Pudong Decoding Life, Shanghai 2000240, China
| | - Shan Chao
- Research Center for Lin He Academician New Medicine, Institutes for Shanghai Pudong Decoding Life, Shanghai 2000240, China
| | - Ying Hua
- Department of Obstetrics and Gynecology and General Surgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Haibin Liu
- Department of Obstetrics and Gynecology and General Surgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| |
Collapse
|
49
|
Mu J, Lin Q, Liang Y. An update on the effects of food-derived active peptides on the intestinal microecology. Crit Rev Food Sci Nutr 2023; 63:11625-11639. [PMID: 35791779 DOI: 10.1080/10408398.2022.2094889] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
The intestinal microecology is a research hotspot, and neologisms related to the gut such as gut-brain axis, gut-lung axis, gut-bone axis, gut-skin axis, gut-renal axis, and gut-liver axis have emerged from recent research. Meticulous investigation has discovered that food-derived active peptides (FDAPs) are bioactive substances that optimize the structure of the gut microbiota to improve human health. However, few reviews have summarized and emphasized the nutritional value of FDAPs and their mechanisms of action in regulating the composition of the gut microbiota. We aim to provide an update on the latest research on FDAPs by comparing, summarizing, and discussing the potential food sources of FDAPs, their physiological functions, and regulatory effects on the intestinal microecology. The key findings are that few studies have analyzed the potential mechanisms and molecular pathways through which FDAPs maintain intestinal microecological homeostasis. We found that an imbalance in the ratio of Bacteroidetes and Firmicutes in the gut microbiota and abnormal production of short-chain fatty acids are key to the occurrence and development of various diseases. This review provides theoretical support for future comprehensive research on the digestion, distribution, metabolism, and excretion of FDAPs and the mechanisms underlying the interactions between FDAPs and the intestinal microecology.
Collapse
Affiliation(s)
- Jianfei Mu
- Molecular Nutrition Branch, National Engineering Research Center of Rice and By-Product Deep Processing/College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, Hunan, China
| | - Qinlu Lin
- Molecular Nutrition Branch, National Engineering Research Center of Rice and By-Product Deep Processing/College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, Hunan, China
| | - Ying Liang
- Molecular Nutrition Branch, National Engineering Research Center of Rice and By-Product Deep Processing/College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, Hunan, China
| |
Collapse
|
50
|
Shin Y, Han S, Kwon J, Ju S, Choi TG, Kang I, Kim SS. Roles of Short-Chain Fatty Acids in Inflammatory Bowel Disease. Nutrients 2023; 15:4466. [PMID: 37892541 PMCID: PMC10609902 DOI: 10.3390/nu15204466] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 10/19/2023] [Accepted: 10/19/2023] [Indexed: 10/29/2023] Open
Abstract
The gut microbiome is a diverse bacterial community in the human gastrointestinal tract that plays important roles in a variety of biological processes. Short-chain fatty acids (SCFA) are produced through fermentation of dietary fiber. Certain microbes in the gut are responsible for producing SCFAs such as acetate, propionate and butyrate. An imbalance in gut microbiome diversity can lead to metabolic disorders and inflammation-related diseases. Changes in SCFA levels and associated microbiota were observed in IBD, suggesting an association between SCFAs and disease. The gut microbiota and SCFAs affect reactive oxygen species (ROS) associated with IBD. Gut microbes and SCFAs are closely related to IBD, and it is important to study them further.
Collapse
Affiliation(s)
- Yoonhwa Shin
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (Y.S.); (S.H.); (J.K.); (S.J.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea;
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Sunhee Han
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (Y.S.); (S.H.); (J.K.); (S.J.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea;
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Juhui Kwon
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (Y.S.); (S.H.); (J.K.); (S.J.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea;
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Songhyun Ju
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (Y.S.); (S.H.); (J.K.); (S.J.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea;
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Tae Gyu Choi
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea;
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Insug Kang
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (Y.S.); (S.H.); (J.K.); (S.J.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea;
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Sung Soo Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (Y.S.); (S.H.); (J.K.); (S.J.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea;
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|