1
|
Goyal A, Chopra V, Garg K, Sharma S. Mechanisms coupling the mTOR pathway to chronic obstructive pulmonary disease (COPD) pathogenesis. Cytokine Growth Factor Rev 2025; 82:55-69. [PMID: 39799015 DOI: 10.1016/j.cytogfr.2024.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 12/10/2024] [Accepted: 12/26/2024] [Indexed: 01/15/2025]
Abstract
Chronic Obstructive Pulmonary Disease (COPD) is a poorly reversible respiratory disorder distinguished by dyspnea, cough, expectoration and exacerbations due to abnormality of airways or emphysema. In this review, we consider the therapeutic potential of targeting Mammalian target of Rapamycin (mTOR) for treating COPD. The mTOR is a highly conserved serine-threonine protein kinase that integrates signals from growth factors and nutrients to control protein synthesis, lipid biogenesis and metabolism. Dysregulated mTOR pathway signaling due to genetic factors or cigarette smoking impairs autophagy, driving the buildup of abnormal cells and damaged proteins, resulting in inflammation and oxidative stress. Persistent mTOR activation also contributes to pulmonary vascular cell proliferation, facilitating the development of pulmonary resistance in COPD. Rapamycin, an inhibitor of mTOR, prevents the buildup of senescent cells in the lungs of COPD patients and inhibits the release of lung tissue-damaging proteases. mTOR also impacts the corticosteroid sensitivity in COPD patients by regulating the levels of histone deacetylases. The emerging role of gut-lung axis dysbiosis in the progression of COPD and its influence on mTOR further highlights the relevance of the mTOR pathway in COPD pathophysiology.
Collapse
Affiliation(s)
- Ankita Goyal
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, India
| | - Vishal Chopra
- Department of Pulmonary Medicine, Government Medical College, Patiala, India
| | - Kranti Garg
- Department of Pulmonary Medicine, Government Medical College, Patiala, India
| | - Siddharth Sharma
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, India.
| |
Collapse
|
2
|
Soga K, Takahashi M, Uno A, Sinada T, Oba K, Kawashima K, Tatewaki Y, Nakase T, Taki Y. Circuit training intervention for cognitive function, gut microbiota, and aging control: study protocol for a longitudinal, open-label randomized controlled trial. Trials 2025; 26:94. [PMID: 40102917 PMCID: PMC11917102 DOI: 10.1186/s13063-025-08807-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 03/03/2025] [Indexed: 03/20/2025] Open
Abstract
BACKGROUND Long-term exercise is increasingly considered an effective strategy to counteract cognitive decline associated with aging. Previous studies have indicated that circuit training exercises integrating aerobic and resistance modalities positively affect cognitive function. Furthermore, a growing body of evidence suggests that long-term exercise alters the gut microbiota, leading to an optimal environment for cognitive enhancement. Recent empirical evidence suggests that exercise plays a significant role in modulating aging-control factors at the protein level. Although the interaction between exercise and cognitive function is multifaceted, most studies have only examined a direct pathway from exercise to cognitive function. Therefore, this study aims to elucidate the effects of long-term circuit training on cognitive function through a comprehensive analysis of factors such as gut microbiota and proteins related to aging control. METHODS A total of fifty-one participants will be randomly assigned to either the circuit training or waitlist control group. The intervention group will participate in a circuit training program developed by Curves Japan Co., Ltd. two to three times weekly for 16 weeks. The control group will continue their usual daily routines without participating in any new active lifestyle program. The participants will undergo cognitive assessments at baseline and after the intervention. Fecal and blood samples for protein analysis will be collected before and after the intervention. The effect of exercise on cognition will be analyzed by comparing the measured outcomes before and after the intervention. The associations among these outcomes will be assessed using a linear mixed model and structural equation modeling approaches. DISCUSSION This study aims to provide the first insights into the comprehensive effects of exercise on cognitive function from the perspectives of gut microbiota and aging control. The findings are expected to contribute to improving brain health and combating age-related cognitive decline. Furthermore, the findings may help establish new guidelines for future studies on the relationship between exercise and cognitive function.
Collapse
Affiliation(s)
- Keishi Soga
- Smart Aging Research Center, Tohoku University, 4-1 Seiryo-Machi, Aoba-Ku, Sendai, 980-8575, Japan.
| | - Michio Takahashi
- Smart Aging Research Center, Tohoku University, 4-1 Seiryo-Machi, Aoba-Ku, Sendai, 980-8575, Japan
| | - Akari Uno
- Smart Aging Research Center, Tohoku University, 4-1 Seiryo-Machi, Aoba-Ku, Sendai, 980-8575, Japan
| | - Takamitsu Sinada
- Smart Aging Research Center, Tohoku University, 4-1 Seiryo-Machi, Aoba-Ku, Sendai, 980-8575, Japan
| | - Kentaro Oba
- Smart Aging Research Center, Tohoku University, 4-1 Seiryo-Machi, Aoba-Ku, Sendai, 980-8575, Japan
| | - Keisei Kawashima
- Department of Medical Sciences, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Yasuko Tatewaki
- Department of Aging Research and Geriatric Medicine, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Taizen Nakase
- Department of Aging Research and Geriatric Medicine, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Yasuyuki Taki
- Smart Aging Research Center, Tohoku University, 4-1 Seiryo-Machi, Aoba-Ku, Sendai, 980-8575, Japan
- Department of Aging Research and Geriatric Medicine, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| |
Collapse
|
3
|
Mahamud AGMSU, Tanvir IA, Kabir ME, Samonty I, Chowdhury MAH, Rahman MA. Gerobiotics: Exploring the Potential and Limitations of Repurposing Probiotics in Addressing Aging Hallmarks and Chronic Diseases. Probiotics Antimicrob Proteins 2025:10.1007/s12602-025-10501-w. [PMID: 40029460 DOI: 10.1007/s12602-025-10501-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/20/2025] [Indexed: 03/05/2025]
Abstract
As unhealthy aging continues to rise globally, there is a pressing need for effective strategies to promote healthy aging, extend health span, and address aging-related complications. Gerobiotics, an emerging concept in geroscience, offers a novel approach to repurposing selective probiotics, postbiotics, and parabiotics to modulate key aging processes and enhance systemic health. This review explores recent advancements in gerobiotics research, focusing on their role in targeting aging hallmarks, regulating longevity-associated pathways, and reducing risks of multiple age-related chronic conditions. Despite their promise, significant challenges remain, including optimizing formulations, ensuring safety and efficacy across diverse populations, and achieving successful clinical translation. Addressing these gaps through rigorous research, well-designed clinical trials, and advanced biotechnologies can establish gerobiotics as a transformative intervention for healthy aging and chronic disease prevention.
Collapse
Affiliation(s)
| | | | - Md Ehsanul Kabir
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53715, USA.
| | - Ismam Samonty
- Department of Agricultural Chemistry, Bangladesh Agricultural University, Mymensingh, 2202, Bangladesh
| | - Md Anamul Hasan Chowdhury
- Department of Food Safety and Regulatory Science, Chung-Ang University, Anseong-Si, Gyeonggi-Do, 17546, Republic of Korea
| | - Md Ashikur Rahman
- Department of Food Safety and Regulatory Science, Chung-Ang University, Anseong-Si, Gyeonggi-Do, 17546, Republic of Korea
| |
Collapse
|
4
|
Zhang L, Cai Y, Li L, Hu J, Jia C, Kuang X, Zhou Y, Lan Z, Liu C, Jiang F, Sun N, Zeng N. Analysis of global trends and hotspots of skin microbiome in acne: a bibliometric perspective. BioData Min 2025; 18:19. [PMID: 40033326 DOI: 10.1186/s13040-025-00433-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 02/13/2025] [Indexed: 03/05/2025] Open
Abstract
BACKGROUND Acne is a chronic inflammatory condition affecting the hair follicles and sebaceous glands. Recent research has revealed significant advances in the study of the acne skin microbiome. Systematic analysis of research trends and hotspots in the acne skin microbiome is lacking. This study utilized bibliometric methods to conduct in-depth research on the recognition structure of the acne skin microbiome, identifying hot trends and emerging topics. METHODS We performed a topic search to retrieve articles about skin microbiome in acne from the Web of Science Core Collection. Bibliometric research was conducted using CiteSpace, VOSviewer, and R language. RESULTS This study analyzed 757 articles from 1362 institutions in 68 countries, the United States leading the research efforts. Notably, Brigitte Dréno from the University of Nantes emerged as the most prolific author in this field, with 19 papers and 334 co-citations. The research output on the skin microbiome of acne continues to increase, with Experimental Dermatology being the journal with the highest number of published articles. The primary focus is investigating the skin microbiome's mechanisms in acne development and exploring treatment strategies. These findings have important implications for developing microbiome-targeted therapies, which could provide new, personalized treatment options for patients with acne. Emerging research hotspots include skincare, gut microbiome, and treatment. CONCLUSION The study's findings indicate a thriving research interest in the skin microbiome and its relationship to acne, focusing on acne treatment through the regulation of the skin microbiome balance. Currently, the development of skincare products targeting the regulation of the skin microbiome represents a research hotspot, reflecting the transition from basic scientific research to clinical practice.
Collapse
Affiliation(s)
- Lanfang Zhang
- Department of Dermatology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yuan Cai
- Department of Dermatology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Lin Li
- Department of Dermatology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jie Hu
- Department of Dermatology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Changsha Jia
- Department of Dermatology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Xu Kuang
- Department of Dermatology, Sinan Branch of Zunyi Medical University Affiliated Hospital, Tongren, China
| | - Yi Zhou
- Department of Dermatology, Dejiang Nation Hospital of TCM, Tongren, China
| | - Zhiai Lan
- Department of Dermatology, Dejiang Nation Hospital of TCM, Tongren, China
| | - Chunyan Liu
- Department of Dermatology, Dejiang Nation Hospital of TCM, Tongren, China
| | - Feng Jiang
- Department of Neonatology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, 200011, China.
| | - Nana Sun
- Department of Dermatology, Guizhou Province Cosmetic Plastic Surgery Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China.
| | - Ni Zeng
- Department of Dermatology, Guizhou Province Cosmetic Plastic Surgery Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China.
| |
Collapse
|
5
|
Dang H, Feng P, Zhang S, Peng L, Xing S, Li Y, Wen X, Zhou L, Goswami S, Xiao M, Barker N, Sansonetti P, Kundu P. Maternal gut microbiota influence stem cell function in offspring. Cell Stem Cell 2025; 32:246-262.e8. [PMID: 39667939 DOI: 10.1016/j.stem.2024.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 08/29/2024] [Accepted: 10/03/2024] [Indexed: 12/14/2024]
Abstract
The maternal microbiome influences child health. However, its impact on a given offspring's stem cells, which regulate development, remains poorly understood. To investigate the role of the maternal microbiome in conditioning the offspring's stem cells, we manipulated maternal microbiota using Akkermansia muciniphila. Different maternal microbiomes had distinct effects on proliferation and differentiation of neuronal and intestinal stem cells in the offspring, influencing their developmental trajectory, physiology, and long-term health. Transplantation of altered maternal microbiota into germ-free mice transmitted these stem cell phenotypes to the recipients' offspring. The progeny of germ-free mice selectively colonized with Akkermansia did not display these stem cell traits, emphasizing the importance of microbiome diversity. Metabolically more active maternal microbiomes enriched the levels of circulating short-chain fatty acids (SCFAs) and amino acids, leaving distinct transcriptomic imprints on the mTOR pathway of offsprings' stem cells. Blocking mTOR signaling during pregnancy eliminated the maternal-microbiome-mediated effects on stem cells. These results suggest a fundamental role of the maternal microbiome in programming offsprings' stem cells and represent a promising target for interventions.
Collapse
Affiliation(s)
- Haiyue Dang
- Laboratory for Microbiota-Host Interactions, The Center for Microbes, Development and Health, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100039, China
| | - Panpan Feng
- Laboratory for Microbiota-Host Interactions, The Center for Microbes, Development and Health, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100039, China
| | - Shuning Zhang
- Laboratory for Microbiota-Host Interactions, The Center for Microbes, Development and Health, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100039, China
| | - Lihua Peng
- Laboratory for Microbiota-Host Interactions, The Center for Microbes, Development and Health, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China
| | - Shuli Xing
- Laboratory for Microbiota-Host Interactions, The Center for Microbes, Development and Health, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yuchen Li
- Laboratory for Microbiota-Host Interactions, The Center for Microbes, Development and Health, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xiang Wen
- Laboratory for Microbiota-Host Interactions, The Center for Microbes, Development and Health, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China
| | - Liqiang Zhou
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200120, China
| | - Shyamal Goswami
- Laboratory for Microbiota-Host Interactions, The Center for Microbes, Development and Health, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China
| | - Mingbing Xiao
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Nick Barker
- Institute of Molecular and Cell Biology, Singapore and Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Philippe Sansonetti
- The Center for Microbes, Development and Health, Institut Pasteur of Shanghai-Chinese Academy of Sciences, Shanghai 200031, China
| | - Parag Kundu
- Laboratory for Microbiota-Host Interactions, The Center for Microbes, Development and Health, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100039, China.
| |
Collapse
|
6
|
Sabo MC, Mustafa S, Saha A, Oyaro B, Fiedler TL, Krueger M, Fuchs E, Mureithi M, Mandaliya K, Jaoko W, Richardson BA, Gharib SA, Fredricks DN, Shah JA, McClelland RS. Bacterial vaginosis is associated with transcriptomic changes but not higher concentrations of cervical leukocytes in a study of women at high risk for HIV acquisition. J Infect Dis 2025:jiaf049. [PMID: 39874304 DOI: 10.1093/infdis/jiaf049] [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: 10/18/2024] [Revised: 01/13/2025] [Accepted: 01/26/2025] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND The association between bacterial vaginosis (BV) and increased HIV acquisition risk may be related to concentrations of HIV-susceptible immune cells in the cervix. METHODS Participants (31 with BV and 30 with normal microbiota) underwent cervical biopsy at a single visit. Immune cells were quantified and sorted using flow cytometry (N=55), localization assessed by immunofluorescence (N=16), and function determined by bulk RNA sequencing (RNA-seq) of live CD45+ cells (N=21). RESULTS Linear regression analyses demonstrated no differences in mean log2 [cells/mg tissue] between women with BV vs normal microbiota for antigen presenting cell (APC) subtypes linked to HIV risk (including CD1a+HLA-DR+ Langerhans cells, CD11c+CD14+ dendritic cells [DCs], and CD11c+HLA-DR+ DCs) and CD4+ T cells. Women with BV had a higher median proportion of CD11c+HLA-DR+ APCs (out of total cells) in cervical epithelium (0.1% vs 0.0%; p=0.03 using Mann-Whitney testing). RNA-seq identified 1,032 differentially expressed genes (adjusted p-value <0.05) in CD45+ cells between women with BV vs normal microbiota. Women with BV demonstrated downregulation of pathways linked to translation, metabolism, cell stress, and immune signaling. CONCLUSIONS BV alters immune cell localization and function; future studies are needed to address how these changes may mediate HIV acquisition risk.
Collapse
Affiliation(s)
- Michelle C Sabo
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Salwa Mustafa
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Aparajita Saha
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Brenda Oyaro
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Tina L Fiedler
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Melissa Krueger
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Esther Fuchs
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, USA
| | - Marianne Mureithi
- Department of Medical Microbiology and Immunology, University of Nairobi, Nairobi, Kenya
| | - Kishor Mandaliya
- Institute of Tropical and Infectious Diseases, University of Nairobi, Nairobi, Kenya
| | - Walter Jaoko
- Department of Medical Microbiology and Immunology, University of Nairobi, Nairobi, Kenya
| | - Barbra A Richardson
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Global Health, University of Washington, Seattle, WA, USA
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Sina A Gharib
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - David N Fredricks
- Department of Medicine, University of Washington, Seattle, WA, USA
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Javeed A Shah
- Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Global Health, University of Washington, Seattle, WA, USA
- VA Puget Sound Healthcare System, Seattle, WA, USA
| | - R Scott McClelland
- Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Medical Microbiology and Immunology, University of Nairobi, Nairobi, Kenya
- Department of Global Health, University of Washington, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| |
Collapse
|
7
|
Wang J, Du J, Gou X, Huang Y, He J, Lu X, Xie M. Propyl acetate protects intestinal barrier during parenteral nutrition in mice and Caco-2 cells. JPEN J Parenter Enteral Nutr 2024; 48:917-926. [PMID: 39187914 DOI: 10.1002/jpen.2681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 08/04/2024] [Accepted: 08/08/2024] [Indexed: 08/28/2024]
Abstract
BACKGROUND Gut microbiota dysbiosis induces intestinal barrier damage during parenteral nutrition (PN). However, the underlying mechanisms remain unclear. This study aimed to investigate gut microbiota dysbiosis, luminal short-chain fatty acids, and autophagy in a mouse model and how these short-chain fatty acids regulate autophagy. METHODS Eight-week-old male specific-pathogen-free mice were randomly divided into a Chow group (standard diet and intravenous normal saline infusion) and a PN group (continuous infusion of PN nutrient solution) for 7 days. Caco-2 cells were also treated with intestinal rinse solutions from Chow and PN mouse models. RESULTS Compared with the Chow group, the PN group exhibited increased Proteobacteria and decreased Firmicutes, correlating with decreased propyl acetate. In the PN group, intestinal tissue exhibited elevated adenosine monophosphate-activated protein kinase (AMPK) phosphorylation, LC3II protein levels, and Atg3 and Atg7 messenger RNA levels. P62 protein levels were decreased, indicating an increase of autophagy flux in the PN group. In the Caco-2 cell model, cells treated with PN solution plus propyl acetate exhibited increased Claudin-1 and occluding along with decreased interleukin-6 and tumor necrosis factor α compared with those treated with PN solution alone. Propyl acetate addition inhibited the AMPK-mammalian target of rapamycin (mTOR) pathway, mitigating the excessive autophagy induced by the PN intestinal rinse solution in Caco-2 cells. CONCLUSION PN led to a significant reduction in propyl acetate levels in the intestine, excessive activation of autophagy, and barrier dysfunction. Propyl acetate inhibited excessive autophagy via the AMPK/mTOR signaling pathway and protected the intestinal barrier during PN.
Collapse
Affiliation(s)
- Jiwei Wang
- Department of General Surgery, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jing Du
- Department of General Surgery, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Xiaomei Gou
- Department of General Surgery, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yong Huang
- Department of General Surgery, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jixin He
- Department of General Surgery, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Xiaoyun Lu
- Department of General Surgery, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Ming Xie
- Department of General Surgery, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| |
Collapse
|
8
|
He S, Wang Z, Xia J, Jia H, Dai Q, Chen C, He F, Wang X, Zhou M. Dasabuvir alleviates 5-fluorouracil-induced intestinal injury through anti-senescence and anti-inflammatory. Sci Rep 2024; 14:15730. [PMID: 38977864 PMCID: PMC11231161 DOI: 10.1038/s41598-024-66771-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: 01/02/2024] [Accepted: 07/03/2024] [Indexed: 07/10/2024] Open
Abstract
5-Fluorouracil (5-Fu) is a basic drug that is used to treat colorectal cancer. Patients who receive 5-Fu chemotherapy often experience side effects that affect the digestive system, such as intestinal injury and diarrhoea, which significantly affect patient compliance with anticancer treatment and quality of life. Therefore, identifying approaches to treat or prevent these side effects is urgent. Dasabuvir (DSV) is a hepatitis C virus inhibitor, but its impact on 5-Fu-induced intestinal injury remains unknown. Our study investigated the effects of DSV on 5-Fu-induced intestinal injury in HUVECs, HIECs and male BALB/c mice. We found that 5-Fu caused intestinal damage by inducing senescence, increasing inflammatory factor expression, and generating oxidative stress. Compared with 5-Fu treatment alone, DSV inhibited senescence by reducing senescence-β-galactosidase (SA-β-gal) activity, the senescence-associated secretory phenotype (SASP, including IL-1, IL-6, and TNF-α) and senescence marker expression levels (p16, p21, and p53). Moreover, the anti-senescence effect of DSV was achieved by inhibiting the mTOR signaling pathway. DSV increased antioxidant enzyme levels and alleviated intestinal tissue injury in mice. In addition, DSV suppressed the 5-Fu-induced increase the diarrhoea scores and ameliorated the weight loss, food intake and water intake of the mice. Overall, this study indicated that DSV could be used to treat chemotherapy-induced intestinal damage.
Collapse
Affiliation(s)
- Siyue He
- School of Basic Medicine, Dali University, Dali, 671000, Yunnan, China
| | - Zhiwei Wang
- School of Basic Medicine, Dali University, Dali, 671000, Yunnan, China
| | - Jing Xia
- School of Basic Medicine, Dali University, Dali, 671000, Yunnan, China
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610000, Sichuan, China
| | - Huijie Jia
- School of Basic Medicine, Dali University, Dali, 671000, Yunnan, China
| | - Qianlong Dai
- School of Basic Medicine, Dali University, Dali, 671000, Yunnan, China
| | - Cui Chen
- School of Basic Medicine, Dali University, Dali, 671000, Yunnan, China
- Qujing Medical College, Qujing, 655011, Yunnan, China
| | - Fei He
- School of Basic Medicine, Dali University, Dali, 671000, Yunnan, China.
| | - Xiaobo Wang
- School of Basic Medicine, Dali University, Dali, 671000, Yunnan, China.
| | - Min Zhou
- School of Basic Medicine, Dali University, Dali, 671000, Yunnan, China.
| |
Collapse
|
9
|
Di Paola FJ, Alquati C, Conti G, Calafato G, Turroni S, D'Amico F, Ceccarelli C, Buttitta F, Bernardi A, Cuicchi D, Poggioli G, Turchetti D, Ferrari S, Cannizzaro R, Realdon S, Brigidi P, Ricciardiello L. Interplay between WNT/PI3K-mTOR axis and the microbiota in APC-driven colorectal carcinogenesis: data from a pilot study and possible implications for CRC prevention. J Transl Med 2024; 22:631. [PMID: 38970018 PMCID: PMC11227240 DOI: 10.1186/s12967-024-05305-5] [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: 03/07/2024] [Accepted: 05/16/2024] [Indexed: 07/07/2024] Open
Abstract
BACKGROUND Wnt/β-catenin signalling impairment accounts for 85% of colorectal cancers (CRCs), including sporadic and familial adenomatous polyposis (FAP) settings. An altered PI3K/mTOR pathway and gut microbiota also contribute to CRC carcinogenesis. We studied the interplay between the two pathways and the microbiota composition within each step of CRC carcinogenesis. METHODS Proteins and target genes of both pathways were analysed by RT-qPCR and IHC in tissues from healthy faecal immunochemical test positive (FIT+, n = 17), FAP (n = 17) and CRC (n = 15) subjects. CRC-related mutations were analysed through NGS and Sanger. Oral, faecal and mucosal microbiota was profiled by 16 S rRNA-sequencing. RESULTS We found simultaneous hyperactivation of Wnt/β-catenin and PI3K/mTOR pathways in FAP-lesions compared to CRCs. Wnt/β-catenin molecular markers positively correlated with Clostridium_sensu_stricto_1 and negatively with Bacteroides in FAP faecal microbiota. Alistipes, Lachnospiraceae, and Ruminococcaceae were enriched in FAP stools and adenomas, the latter also showing an overabundance of Lachnoclostridium, which positively correlated with cMYC. In impaired-mTOR-mutated CRC tissues, p-S6R correlated with Fusobacterium and Dialister, the latter also confirmed in the faecal-ecosystem. CONCLUSIONS Our study reveals an interplay between Wnt/β-catenin and PI3K/mTOR, whose derangement correlates with specific microbiota signatures in FAP and CRC patients, and identifies new potential biomarkers and targets to improve CRC prevention, early adenoma detection and treatment.
Collapse
Affiliation(s)
| | - Chiara Alquati
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
- Centre for Applied Biomedical Research (CRBA), University of Bologna, Bologna, Italy
| | - Gabriele Conti
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Giulia Calafato
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Silvia Turroni
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Federica D'Amico
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Claudio Ceccarelli
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | | | - Alice Bernardi
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Dajana Cuicchi
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Gilberto Poggioli
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Daniela Turchetti
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Simona Ferrari
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Renato Cannizzaro
- Oncological Gastroenterology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | - Stefano Realdon
- Oncological Gastroenterology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Patrizia Brigidi
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Luigi Ricciardiello
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy.
| |
Collapse
|
10
|
Kim YT, Mills DA. Exploring the gut microbiome: probiotics, prebiotics, synbiotics, and postbiotics as key players in human health and disease improvement. Food Sci Biotechnol 2024; 33:2065-2080. [PMID: 39130661 PMCID: PMC11315840 DOI: 10.1007/s10068-024-01620-1] [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: 04/10/2024] [Revised: 05/15/2024] [Accepted: 05/23/2024] [Indexed: 08/13/2024] Open
Abstract
The human gut microbiome accompanies us from birth, and it is developed and matured by diet, lifestyle, and environmental factors. During aging, the bacterial composition evolves in reciprocal communication with the host's physiological properties. Many diseases are closely related to the gut microbiome, which means the modulation of the gut microbiome can promote the disease targeting remote organs. This review explores the intricate interaction between the gut microbiome and other organs, and their improvement from disease by prebiotics, probiotics, synbiotics, and postbiotics. Each section of the review is supported by clinical trials that substantiate the benefits of modulation the gut microbiome through dietary intervention for improving primary health outcomes across various axes with the gut. In conclusion, the review underscores the significant potential of targeting the gut microbiome for therapeutic and preventative interventions in a wide range of diseases, calling for further research to fully unlock the microbiome's capabilities in enhancing human health.
Collapse
Affiliation(s)
- You-Tae Kim
- Department of Food Science and Technology, University of California-Davis, Davis, CA USA
| | - David A. Mills
- Department of Food Science and Technology, University of California-Davis, Davis, CA USA
| |
Collapse
|
11
|
Ryguła I, Pikiewicz W, Kaminiów K. Impact of Diet and Nutrition in Patients with Acne Vulgaris. Nutrients 2024; 16:1476. [PMID: 38794714 PMCID: PMC11124289 DOI: 10.3390/nu16101476] [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: 03/26/2024] [Revised: 05/08/2024] [Accepted: 05/12/2024] [Indexed: 05/26/2024] Open
Abstract
Acne vulgaris is a widespread a chronic inflammatory dermatosis that affects millions of people around the world, which has a significant influence on patients' standard of living. The progression of this dermatosis results in the appearance of inflammatory and non-inflammatory changes, and, in severe cases, disfiguring scars and hyperpigmentation. The aetiopathogenesis of acne is complex. It involves a complex interaction of many different factors, both endo- and exogenous in their effect on the hair and sebaceous unit. Genetic predisposition, hormones, the skin and gut microbiome, psychological stress, air pollutants, aggressive facial products, and certain medications are cited as factors influencing acne formation. The link between nutrition and acne is extensively debated for many years and is still relatively controversial. Diet is commonly recognised to have a direct relationship with certain biochemical markers and the transcription of genes related to sebaceous gland function, and the proliferation of bacteria and inflammation that encourage the progression of the disease. In this review, the authors take a closer look at the existing scientific reports on the involvement of nutrition in the development of acne vulgaris.
Collapse
Affiliation(s)
- Izabella Ryguła
- Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-752 Katowice, Poland;
| | - Wojciech Pikiewicz
- Collegium Medicum—Faculty of Medicine, WSB University, 41-300 Dabrowa Gornicza, Poland;
| | - Konrad Kaminiów
- Collegium Medicum—Faculty of Medicine, WSB University, 41-300 Dabrowa Gornicza, Poland;
| |
Collapse
|
12
|
Kim HJ, Kim YH. Exploring Acne Treatments: From Pathophysiological Mechanisms to Emerging Therapies. Int J Mol Sci 2024; 25:5302. [PMID: 38791344 PMCID: PMC11121268 DOI: 10.3390/ijms25105302] [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: 05/08/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
Acne vulgaris is a common dermatological condition that can present across different ages but predominantly affects adolescents and young adults. Characterized by various lesion types, the pathogenesis of acne is complex, involving genetic, hormonal, microbial, and inflammatory factors. This review comprehensively addresses current and emerging acne management strategies, emphasizing both topical and systemic treatments, procedural therapies, and dietary modifications. Key topical agents include retinoids, benzoyl peroxide, antibiotics, and other specialized compounds. Systemic options like antibiotics, hormonal therapies, and retinoids offer significant therapeutic benefits, particularly for moderate to severe cases. Procedural treatments such as laser devices, photodynamic therapy, chemical peels, and intralesional injections present viable alternatives for reducing acne symptoms and scarring. Emerging therapies focus on novel biologics, bacteriophages, probiotics, and peptides, providing promising future options. This review underscores the importance of personalized approaches to treatment due to the multifaceted nature of acne, highlighting the potential of innovative therapies for improving patient outcomes.
Collapse
Affiliation(s)
- Hyun Jee Kim
- Department of Dermatology, International St. Mary’s Hospital, College of Medicine, Catholic Kwandong University, Incheon 22711, Republic of Korea;
| | - Yeong Ho Kim
- Department of Dermatology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| |
Collapse
|
13
|
Rejeki PS, Pranoto A, Widiatmaja DM, Utami DM, Izzatunnisa N, Sugiharto, Lesmana R, Halim S. Combined Aerobic Exercise with Intermittent Fasting Is Effective for Reducing mTOR and Bcl-2 Levels in Obese Females. Sports (Basel) 2024; 12:116. [PMID: 38786985 PMCID: PMC11126026 DOI: 10.3390/sports12050116] [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: 03/06/2024] [Revised: 04/07/2024] [Accepted: 04/15/2024] [Indexed: 05/25/2024] Open
Abstract
The integration of combined aerobic exercise and intermittent fasting (IF) has emerged as a strategy for the prevention and management of obesity, including its associated health issues such as age-related metabolic diseases. This study aimed to examine the potential of combined aerobic exercise and IF as a preventative strategy against cellular senescence by targeting mTOR and Bcl-2 levels in obese females. A total of 30 obese women, aged 23.56 ± 1.83 years, body fat percentage (FAT) 45.21 ± 3.73% (very high category), BMI 30.09 ± 3.74 kg/m2 were recruited and participated in three different types of interventions: intermittent fasting (IF), exercise (EXG), and a combination of intermittent fasting and exercise (IFEXG). The intervention program was carried out 5x/week for 2 weeks. We examined mTOR and Bcl-2 levels using ELISA kits. Statistical analysis used the one-way ANOVA test and continued with Tukey's HSD post hoc test, with a significance level of 5%. The study results showed that a combination of aerobic exercise and IF significantly decreased mTOR levels (-1.26 ± 0.79 ng/mL) compared to the control group (-0.08 ± 1.33 ng/mL; p ≤ 0.05). However, combined aerobic exercise and IF did not affect Bcl-2 levels significantly (-0.07 ± 0.09 ng/mL) compared to the control group (0.01 ± 0.17 ng/mL, p ≥ 0.05). The IF-only group, exercise-only group, and combined group all showed a significant decrease in body weight and fat mass compared to the control group (p ≤ 0.05). However, the combined aerobic exercise and IF program had a significant effect in reducing the total percentage of body fat and fat mass compared to the IF-only group (p ≤ 0.05). Therefore, it was concluded that the combined intermittent fasting and exercise group (IFEXG) undertook the most effective intervention of the three in terms of preventing cellular senescence, as demonstrated by decreases in the mTOR level, body weight, and fat mass. However, the IFEXG did not present reduced Bcl-2 levels.
Collapse
Affiliation(s)
- Purwo Sri Rejeki
- Physiology Division, Department of Medical Physiology and Biochemistry, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, East Java, Indonesia
| | - Adi Pranoto
- Doctoral Program of Medical Science, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, East Java, Indonesia;
| | - Deandra Maharani Widiatmaja
- Medical Program, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, East Java, Indonesia; (D.M.W.); (D.M.U.); (N.I.)
| | - Dita Mega Utami
- Medical Program, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, East Java, Indonesia; (D.M.W.); (D.M.U.); (N.I.)
| | - Nabilah Izzatunnisa
- Medical Program, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, East Java, Indonesia; (D.M.W.); (D.M.U.); (N.I.)
| | - Sugiharto
- Department of Sport Science, Faculty of Sport Science, Universitas Negeri Malang, Malang 65145, East Java, Indonesia;
| | - Ronny Lesmana
- Physiology Division, Department of Biomedical Science, Faculty of Medicine, Universitas Padjajaran, Bandung 45363, West Java, Indonesia;
| | - Shariff Halim
- Faculty of Health Sciences, University Technology MARA (UiTM) Pulau Pinang, Bertam Campus, Kepala Batas 13200, Pulau Pinang, Malaysia;
| |
Collapse
|
14
|
He M, Wei W, Zhang Y, Xiang Z, Peng D, Kasimumali A, Rong S. Gut microbial metabolites SCFAs and chronic kidney disease. J Transl Med 2024; 22:172. [PMID: 38369469 PMCID: PMC10874542 DOI: 10.1186/s12967-024-04974-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 02/11/2024] [Indexed: 02/20/2024] Open
Abstract
The global incidence of Chronic Kidney Disease (CKD) is steadily escalating, with discernible linkage to the intricate terrain of intestinal microecology. The intestinal microbiota orchestrates a dynamic equilibrium in the organism, metabolizing dietary-derived compounds, a process which profoundly impacts human health. Among these compounds, short-chain fatty acids (SCFAs), which result from microbial metabolic processes, play a versatile role in influencing host energy homeostasis, immune function, and intermicrobial signaling, etc. SCFAs emerge as pivotal risk factors influencing CKD's development and prognosis. This paper review elucidates the impact of gut microbial metabolites, specifically SCFAs, on CKD, highlighting their role in modulating host inflammatory responses, oxidative stress, cellular autophagy, the immune milieu, and signaling cascades. An in-depth comprehension of the interplay between SCFAs and kidney disease pathogenesis may pave the way for their utilization as biomarkers for CKD progression and prognosis or as novel adjunctive therapeutic strategies.
Collapse
Affiliation(s)
- Meng He
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Wenqian Wei
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Yichen Zhang
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Zhouxia Xiang
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Dan Peng
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Ayijiaken Kasimumali
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Shu Rong
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China.
| |
Collapse
|
15
|
Tenchov R, Sasso JM, Wang X, Zhou QA. Antiaging Strategies and Remedies: A Landscape of Research Progress and Promise. ACS Chem Neurosci 2024; 15:408-446. [PMID: 38214973 PMCID: PMC10853939 DOI: 10.1021/acschemneuro.3c00532] [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: 08/12/2023] [Revised: 12/01/2023] [Accepted: 12/04/2023] [Indexed: 01/13/2024] Open
Abstract
Aging is typified by a gradual loss of physiological fitness and accumulation of cellular damage, leading to deteriorated functions and enhanced vulnerability to diseases. Antiaging research has a long history throughout civilization, with many efforts put forth to understand and prevent the effects of aging. Multiple strategies aiming to promote healthy aging and extend the lifespan have been developed including lifestyle adjustments, medical treatments, and social programs. A multitude of antiaging medicines and remedies have also been explored. Here, we use data from the CAS Content Collection to analyze the publication landscape of recent research related to antiaging strategies and treatments. We review the recent advances and delineate trends in research headway of antiaging knowledge and practice across time, geography, and development pipelines. We further assess the state-of-the-art antiaging approaches and explore their correlations with age-related diseases. The landscape of antiaging drugs has been outlined and explored. Well-recognized and novel, currently evaluated antiaging agents have also been summarized. Finally, we review clinical applications of antiaging products with their development pipelines. The objective of this review is to summarize current knowledge on preventive strategies and treatment remedies in the field of aging, to outline challenges and evaluate growth opportunities, in order to further efforts to solve the problems that remain.
Collapse
Affiliation(s)
- Rumiana Tenchov
- CAS, a Division of the American
Chemical Society, 2540 Olentangy River Road, Columbus, Ohio 43202, United States
| | - Janet M. Sasso
- CAS, a Division of the American
Chemical Society, 2540 Olentangy River Road, Columbus, Ohio 43202, United States
| | - Xinmei Wang
- CAS, a Division of the American
Chemical Society, 2540 Olentangy River Road, Columbus, Ohio 43202, United States
| | - Qiongqiong Angela Zhou
- CAS, a Division of the American
Chemical Society, 2540 Olentangy River Road, Columbus, Ohio 43202, United States
| |
Collapse
|
16
|
Ryguła I, Pikiewicz W, Grabarek BO, Wójcik M, Kaminiów K. The Role of the Gut Microbiome and Microbial Dysbiosis in Common Skin Diseases. Int J Mol Sci 2024; 25:1984. [PMID: 38396663 PMCID: PMC10889245 DOI: 10.3390/ijms25041984] [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: 01/05/2024] [Revised: 01/28/2024] [Accepted: 02/05/2024] [Indexed: 02/25/2024] Open
Abstract
Dermatoses are an increasingly common problem, particularly in developed countries. The causes of this phenomenon include genetic factors and environmental elements. More and more scientific reports suggest that the gut microbiome, more specifically its dysbiosis, also plays an important role in the induction and progression of diseases, including dermatological diseases. The gut microbiome is recognised as the largest endocrine organ, and has a key function in maintaining human homeostasis. In this review, the authors will take a close look at the link between the gut-skin axis and the pathogenesis of dermatoses such as atopic dermatitis, psoriasis, alopecia areata, and acne. The authors will also focus on the role of probiotics in remodelling the microbiome and the alleviation of dermatoses.
Collapse
Affiliation(s)
- Izabella Ryguła
- Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-752 Katowice, Poland;
| | - Wojciech Pikiewicz
- Collegium Medicum—Faculty of Medicine, WSB University, 41-300 Dabrowa Gornicza, Poland; (W.P.); (B.O.G.); (M.W.)
| | - Beniamin Oskar Grabarek
- Collegium Medicum—Faculty of Medicine, WSB University, 41-300 Dabrowa Gornicza, Poland; (W.P.); (B.O.G.); (M.W.)
| | - Michał Wójcik
- Collegium Medicum—Faculty of Medicine, WSB University, 41-300 Dabrowa Gornicza, Poland; (W.P.); (B.O.G.); (M.W.)
| | - Konrad Kaminiów
- Collegium Medicum—Faculty of Medicine, WSB University, 41-300 Dabrowa Gornicza, Poland; (W.P.); (B.O.G.); (M.W.)
| |
Collapse
|
17
|
Wu Y, Wang X, Wu W, Yang J. Mendelian randomization analysis reveals an independent causal relationship between four gut microbes and acne vulgaris. Front Microbiol 2024; 15:1326339. [PMID: 38371936 PMCID: PMC10869500 DOI: 10.3389/fmicb.2024.1326339] [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: 10/24/2023] [Accepted: 01/18/2024] [Indexed: 02/20/2024] Open
Abstract
Background Numerous studies have suggested a correlation between gut microbiota and acne vulgaris; however, no specific causal link has been explored. Materials and methods To investigate the possible causal relationship between gut microbiota and acne vulgaris, this study employed a large-scale genome-wide association study (GWAS) summary statistic. Initially, a two-sample Mendelian randomization (MR) analysis was utilized to identify the specific gut microflora responsible for acne vulgaris. We used the Inverse Variance Weighted (IVW) method as the main MR analysis method. Additionally, we assessed heterogeneity and horizontal pleiotropy, while also examining the potential influence of individual single-nucleotide polymorphisms (SNPs) on the analysis results. In order to eliminate gut microbiota with reverse causal associations, we conducted reverse MR analysis. Multivariate Mendelian randomization analysis (MVMR) was then employed to verify the independence of the causal associations. Finally, we performed SNP annotation on the instrumental variables of independent gut microbiota and acne vulgaris to determine the genes where these genetic variations are located. We also explored the biological functions of these genes through enrichment analysis. Result The IVW method of forward MR identified nine gut microbes with a causal relationship with acne vulgaris (p < 0.05). The findings from the sensitivity analysis demonstrate the absence of heterogeneity or horizontal pleiotropy, and leave-one-out analysis indicates that the results are not driven by a single SNP. Additionally, the Reverse MR analysis excluded two reverse-correlated pathogenic gut microbes. And then, MVMR was used to analyze seven gut microbes, and it was found that Cyanobacterium and Family XIII were risk factors for acne vulgaris, while Ruminococcus1 and Ruminiclostridium5 were protective factors for acne vulgaris. After conducting biological annotation, we identified six genes (PLA2G4A, FADS2, TIMP17, ADAMTS9, ZC3H3, and CPSF4L) that may be associated with the pathogenic gut microbiota of acne vulgaris patients. The enrichment analysis results indicate that PLA2G4A/FADS2 is associated with fatty acid metabolism pathways. Conclusion Our study found independent causal relationships between four gut microbes and acne vulgaris, and revealed a genetic association between acne vulgaris patients and gut microbiota. Consider preventing and treating acne vulgaris by interfering with the relative content of these four gut microbes.
Collapse
Affiliation(s)
- Yujia Wu
- School of Basic Medical Sciences, Dali University, Dali, China
| | - Xiaoyun Wang
- School of Basic Medical Sciences, Dali University, Dali, China
| | - Wenjuan Wu
- Department of Dermatology, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Jiankang Yang
- School of Basic Medical Sciences, Dali University, Dali, China
| |
Collapse
|
18
|
Sun J. Navigating the complex landscape of non-alcoholic fatty liver disease: From genomic associate to the AMPK/mTOR pathways. J Gastroenterol Hepatol 2023; 38:1863-1864. [PMID: 37931986 DOI: 10.1111/jgh.16407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 10/27/2023] [Indexed: 11/08/2023]
Affiliation(s)
- Jun Sun
- University of Illinois Chicago, Chicago, Illinois, USA
| |
Collapse
|
19
|
Marcondes-de-Castro IA, Reis-Barbosa PH, Marinho TS, Aguila MB, Mandarim-de-Lacerda CA. AMPK/mTOR pathway significance in healthy liver and non-alcoholic fatty liver disease and its progression. J Gastroenterol Hepatol 2023; 38:1868-1876. [PMID: 37438882 DOI: 10.1111/jgh.16272] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 06/06/2023] [Accepted: 06/14/2023] [Indexed: 07/14/2023]
Abstract
Obesity is related to several organs, but the liver is particularly affected. Adenosine monophosphate-activated protein kinase (AMPK) is a cellular energy sensor and regulator of liver lipid dysfunction and glucose metabolism. The mechanistic target of rapamycin (mTOR) is a protein kinase regulating cell growth, survival, metabolism, and immunity. Together, these pathways are involved in obesity, insulin resistance, non-alcoholic fatty liver disease (NAFLD) and its progression, and autophagy. During energy demand, liver kinase B (LKB) phosphorylation helps activate the AMPK/mTOR pathways. Likewise, the protein forkhead box O family (FOXO) negatively regulates adipogenesis by binding to the promoter sites of peroxisome proliferator-activated receptor-gamma coactivator 1-alpha, initiating adipogenesis. In addition, acetyl-CoA carboxylase, which regulates de novo lipogenesis, is linked to LKB and FOXO in developing NAFLD. The kinase complex, consisting of Unc-51-like autophagy-activating kinase 1 or 2 (ULK1, ULK2) by stimulating autophagy, and eliminating fat droplets in NAFLD, is regulated by mTORC1 and negatively regulated by AMPK that suppresses liver lipogenesis and increases fatty acid oxidation. Also, ULK1 is essential for initiating phagophore formation, establishing macrophagy, and generating autophagosomes. The selective breakdown of lipid droplets through macroautophagy, or macrolipophagy, occurs on a cellular energy level using free fatty acids. In addition, mTORC1 promotes lipogenesis by activating sterol regulatory element-binding protein. Finding new components and novel regulatory modes in signaling is significant for a better understanding of the AMPK/mTOR pathways, potentially facilitating the development of future diagnostic and therapeutic strategies for NAFLD and its progression to non-alcoholic steatohepatitis, cirrhosis, and hepatocellular carcinoma.
Collapse
Affiliation(s)
- Ilitch Aquino Marcondes-de-Castro
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Centre, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Pedro Henrique Reis-Barbosa
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Centre, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Thatiany Souza Marinho
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Centre, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Marcia Barbosa Aguila
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Centre, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Carlos Alberto Mandarim-de-Lacerda
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Centre, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| |
Collapse
|
20
|
Liu P, Li H, Xu H, Gong J, Jiang M, Xu Z, Shi J. Aggravated hepatic fibrosis induced by phenylalanine and tyrosine was ameliorated by chitooligosaccharides supplementation. iScience 2023; 26:107754. [PMID: 37731617 PMCID: PMC10507131 DOI: 10.1016/j.isci.2023.107754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 03/21/2023] [Accepted: 08/24/2023] [Indexed: 09/22/2023] Open
Abstract
Hepatic fibrosis is a classic pathological manifestation of metabolic chronic hepatopathy. The pathological process might either gradually deteriorate into cirrhosis and ultimately liver cancer with inappropriate nutrition supply, or be slowed down by several multifunctional nutrients, alternatively. Herein, we found diet with excessive phenylalanine (Phe) and tyrosine (Tyr) exacerbated hepatic fibrosis symptoms of liver dysfunction and gut microflora dysbiosis in mice. Chitooligosaccharides (COS) could ameliorate hepatic fibrosis with the regulation of amino acid metabolism by downregulating the mTORC1 pathway, especially that of Phe and Tyr, and also with the alleviation of the dysbiosis of gut microbiota, simultaneously. Conclusively, this work presents new insight into the role of Phe and Tyr in the pathologic process of hepatic fibrosis, while revealing the effectiveness and molecular mechanism of COS in improving hepatic fibrosis from the perspective of metabolites.
Collapse
Affiliation(s)
- Peng Liu
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, China
- Institute of Edible Fungi, Shanghai Academy of Agricultural Sciences, 1000 Jinqi Road, Shanghai 201403, China
| | - Heng Li
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, China
| | - Hongyu Xu
- National Engineering Research Center for Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi 214122, China
| | - Jinsong Gong
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, China
| | - Min Jiang
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, China
| | - Zhenghong Xu
- National Engineering Research Center for Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi 214122, China
| | - Jinsong Shi
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
21
|
Masenga SK, Povia JP, Lwiindi PC, Kirabo A. Recent Advances in Microbiota-Associated Metabolites in Heart Failure. Biomedicines 2023; 11:2313. [PMID: 37626809 PMCID: PMC10452327 DOI: 10.3390/biomedicines11082313] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 08/16/2023] [Accepted: 08/19/2023] [Indexed: 08/27/2023] Open
Abstract
Heart failure is a risk factor for adverse events such as sudden cardiac arrest, liver and kidney failure and death. The gut microbiota and its metabolites are directly linked to the pathogenesis of heart failure. As emerging studies have increased in the literature on the role of specific gut microbiota metabolites in heart failure development, this review highlights and summarizes the current evidence and underlying mechanisms associated with the pathogenesis of heart failure. We found that gut microbiota-derived metabolites such as short chain fatty acids, bile acids, branched-chain amino acids, tryptophan and indole derivatives as well as trimethylamine-derived metabolite, trimethylamine N-oxide, play critical roles in promoting heart failure through various mechanisms. Mainly, they modulate complex signaling pathways such as nuclear factor kappa-light-chain-enhancer of activated B cells, Bcl-2 interacting protein 3, NLR Family Pyrin Domain Containing inflammasome, and Protein kinase RNA-like endoplasmic reticulum kinase. We have also highlighted the beneficial role of other gut metabolites in heart failure and other cardiovascular and metabolic diseases.
Collapse
Affiliation(s)
- Sepiso K. Masenga
- HAND Research Group, School of Medicine and Health Sciences, Mulungushi University, Livingstone Campus, Livingstone 10101, Zambia; (J.P.P.); (P.C.L.)
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232-6602, USA
| | - Joreen P. Povia
- HAND Research Group, School of Medicine and Health Sciences, Mulungushi University, Livingstone Campus, Livingstone 10101, Zambia; (J.P.P.); (P.C.L.)
| | - Propheria C. Lwiindi
- HAND Research Group, School of Medicine and Health Sciences, Mulungushi University, Livingstone Campus, Livingstone 10101, Zambia; (J.P.P.); (P.C.L.)
| | - Annet Kirabo
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232-6602, USA
| |
Collapse
|
22
|
Gao Y, Tian T. mTOR Signaling Pathway and Gut Microbiota in Various Disorders: Mechanisms and Potential Drugs in Pharmacotherapy. Int J Mol Sci 2023; 24:11811. [PMID: 37511569 PMCID: PMC10380532 DOI: 10.3390/ijms241411811] [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: 06/12/2023] [Revised: 07/15/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
The mammalian or mechanistic target of rapamycin (mTOR) integrates multiple intracellular and extracellular upstream signals involved in the regulation of anabolic and catabolic processes in cells and plays a key regulatory role in cell growth and metabolism. The activation of the mTOR signaling pathway has been reported to be associated with a wide range of human diseases. A growing number of in vivo and in vitro studies have demonstrated that gut microbes and their complex metabolites can regulate host metabolic and immune responses through the mTOR pathway and result in disorders of host physiological functions. In this review, we summarize the regulatory mechanisms of gut microbes and mTOR in different diseases and discuss the crosstalk between gut microbes and their metabolites and mTOR in disorders in the gastrointestinal tract, liver, heart, and other organs. We also discuss the promising application of multiple potential drugs that can adjust the gut microbiota and mTOR signaling pathways. Despite the limited findings between gut microbes and mTOR, elucidating their relationship may provide new clues for the prevention and treatment of various diseases.
Collapse
Affiliation(s)
- Yuan Gao
- College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing 100044, China
| | - Tian Tian
- College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing 100044, China
| |
Collapse
|
23
|
Papa V, Schepis T, Coppola G, Chiappetta MF, Del Vecchio LE, Rozera T, Quero G, Gasbarrini A, Alfieri S, Papa A. The Role of Microbiota in Pancreatic Cancer. Cancers (Basel) 2023; 15:3143. [PMID: 37370753 DOI: 10.3390/cancers15123143] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 06/02/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
Pancreatic cancer (PC) has an unfavorable prognosis with few effective therapeutic options. This has led researchers to investigate the possible links between microbiota and PC. A disrupted gut microbiome can lead to chronic inflammation, which is involved in the pathogenesis of PC. In addition, some bacterial strains can produce carcinogens that promote the growth of cancer cells. Research has also focused on pancreatic and oral microbiota. Changes in these microbiota can contribute to the development and progression of PC. Furthermore, patients with periodontal disease have an increased risk of developing PC. The potential use of microbiota as a prognostic marker or to predict patients' responses to chemotherapy or immunotherapy is also being explored. Overall, the role of microbiota-including the gut, pancreatic, and oral microbiota-in PC is an active research area. Understanding these associations could lead to new diagnostic and therapeutic targets for this deadly disease.
Collapse
Affiliation(s)
- Valerio Papa
- Department of Translational Medicine and Surgery, School of Medicine, Catholic University, 00168 Rome, Italy
- Digestive Surgery Unit, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy
| | - Tommaso Schepis
- Center for Diagnosis and Treatment of Digestive Diseases, CEMAD, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy
| | - Gaetano Coppola
- Center for Diagnosis and Treatment of Digestive Diseases, CEMAD, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy
| | - Michele Francesco Chiappetta
- Center for Diagnosis and Treatment of Digestive Diseases, CEMAD, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy
| | - Livio Enrico Del Vecchio
- Center for Diagnosis and Treatment of Digestive Diseases, CEMAD, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy
| | - Tommaso Rozera
- Center for Diagnosis and Treatment of Digestive Diseases, CEMAD, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy
| | - Giuseppe Quero
- Department of Translational Medicine and Surgery, School of Medicine, Catholic University, 00168 Rome, Italy
- Digestive Surgery Unit, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy
| | - Antonio Gasbarrini
- Department of Translational Medicine and Surgery, School of Medicine, Catholic University, 00168 Rome, Italy
- Center for Diagnosis and Treatment of Digestive Diseases, CEMAD, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy
| | - Sergio Alfieri
- Department of Translational Medicine and Surgery, School of Medicine, Catholic University, 00168 Rome, Italy
- Digestive Surgery Unit, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy
| | - Alfredo Papa
- Department of Translational Medicine and Surgery, School of Medicine, Catholic University, 00168 Rome, Italy
- Center for Diagnosis and Treatment of Digestive Diseases, CEMAD, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy
| |
Collapse
|
24
|
Berard AR, Brubaker DK, Birse K, Lamont A, Mackelprang RD, Noël-Romas L, Perner M, Hou X, Irungu E, Mugo N, Knodel S, Muwonge TR, Katabira E, Hughes SM, Levy C, Calienes FL, Lauffenburger DA, Baeten JM, Celum C, Hladik F, Lingappa J, Burgener AD. Vaginal epithelial dysfunction is mediated by the microbiome, metabolome, and mTOR signaling. Cell Rep 2023; 42:112474. [PMID: 37149863 PMCID: PMC10242450 DOI: 10.1016/j.celrep.2023.112474] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 03/15/2023] [Accepted: 04/19/2023] [Indexed: 05/09/2023] Open
Abstract
Bacterial vaginosis (BV) is characterized by depletion of Lactobacillus and overgrowth of anaerobic and facultative bacteria, leading to increased mucosal inflammation, epithelial disruption, and poor reproductive health outcomes. However, the molecular mediators contributing to vaginal epithelial dysfunction are poorly understood. Here we utilize proteomic, transcriptomic, and metabolomic analyses to characterize biological features underlying BV in 405 African women and explore functional mechanisms in vitro. We identify five major vaginal microbiome groups: L. crispatus (21%), L. iners (18%), Lactobacillus (9%), Gardnerella (30%), and polymicrobial (22%). Using multi-omics we show that BV-associated epithelial disruption and mucosal inflammation link to the mammalian target of rapamycin (mTOR) pathway and associate with Gardnerella, M. mulieris, and specific metabolites including imidazole propionate. Experiments in vitro confirm that type strain G. vaginalis and M. mulieris supernatants and imidazole propionate directly affect epithelial barrier function and activation of mTOR pathways. These results find that the microbiome-mTOR axis is a central feature of epithelial dysfunction in BV.
Collapse
Affiliation(s)
- Alicia R Berard
- Department of Obstetrics & Gynecology, University of Manitoba, Winnipeg, MB R3E 3P5, Canada; Center for Global Health and Diseases, Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Douglas K Brubaker
- Weldon School of Biomedical Engineering and Regenstrief Center for Healthcare Engineering, Purdue University, West Lafayette, IN 47907, USA
| | - Kenzie Birse
- Department of Obstetrics & Gynecology, University of Manitoba, Winnipeg, MB R3E 3P5, Canada; Center for Global Health and Diseases, Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Alana Lamont
- Department of Obstetrics & Gynecology, University of Manitoba, Winnipeg, MB R3E 3P5, Canada
| | - Romel D Mackelprang
- Department of Global Health, University of Washington, Seattle, WA 98105, USA
| | - Laura Noël-Romas
- Department of Obstetrics & Gynecology, University of Manitoba, Winnipeg, MB R3E 3P5, Canada; Center for Global Health and Diseases, Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Michelle Perner
- Medical Microbiology and Infectious Disease University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Xuanlin Hou
- Department of Global Health, University of Washington, Seattle, WA 98105, USA
| | - Elizabeth Irungu
- Partners in Health Research and Development, Kenya Medical Research Institute, Mbagathi Road, Nairobi, Kenya
| | - Nelly Mugo
- Department of Global Health, University of Washington, Seattle, WA 98105, USA; Partners in Health Research and Development, Kenya Medical Research Institute, Mbagathi Road, Nairobi, Kenya
| | - Samantha Knodel
- Department of Obstetrics & Gynecology, University of Manitoba, Winnipeg, MB R3E 3P5, Canada; Center for Global Health and Diseases, Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Timothy R Muwonge
- Infectious Disease Institute, Makerere University, Makerere, Kampala, Uganda
| | - Elly Katabira
- Infectious Disease Institute, Makerere University, Makerere, Kampala, Uganda
| | - Sean M Hughes
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA 98195, USA
| | - Claire Levy
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA 98195, USA
| | | | | | - Jared M Baeten
- Department of Global Health, University of Washington, Seattle, WA 98105, USA; Department of Medicine, University of Washington, Seattle, WA 98195, USA; Department of Epidemiology, University of Washington, Seattle, WA 98195, USA; Gilead Sciences, Foster City, CA 94404, USA
| | - Connie Celum
- Department of Global Health, University of Washington, Seattle, WA 98105, USA; Department of Medicine, University of Washington, Seattle, WA 98195, USA; Department of Epidemiology, University of Washington, Seattle, WA 98195, USA
| | - Florian Hladik
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA 98195, USA; Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Department of Medicine, University of Washington, Seattle, WA 98195, USA
| | - Jairam Lingappa
- Department of Global Health, University of Washington, Seattle, WA 98105, USA; Department of Medicine, University of Washington, Seattle, WA 98195, USA; Department of Pediatrics, University of Washington, Seattle, WA 98195, USA
| | - Adam D Burgener
- Department of Obstetrics & Gynecology, University of Manitoba, Winnipeg, MB R3E 3P5, Canada; Center for Global Health and Diseases, Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA; Department of Medicine Solna, Karolinska Institutet, Framstegsgatan, 171 64 Solna, Sweden.
| |
Collapse
|
25
|
Liu XF, Shao JH, Liao YT, Wang LN, Jia Y, Dong PJ, Liu ZZ, He DD, Li C, Zhang X. Regulation of short-chain fatty acids in the immune system. Front Immunol 2023; 14:1186892. [PMID: 37215145 PMCID: PMC10196242 DOI: 10.3389/fimmu.2023.1186892] [Citation(s) in RCA: 60] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 04/24/2023] [Indexed: 05/24/2023] Open
Abstract
A growing body of research suggests that short-chain fatty acids (SCFAs), metabolites produced by intestinal symbiotic bacteria that ferment dietary fibers (DFs), play a crucial role in the health status of symbiotes. SCFAs act on a variety of cell types to regulate important biological processes, including host metabolism, intestinal function, and immune function. SCFAs also affect the function and fate of immune cells. This finding provides a new concept in immune metabolism and a better understanding of the regulatory role of SCFAs in the immune system, which impacts the prevention and treatment of disease. The mechanism by which SCFAs induce or regulate the immune response is becoming increasingly clear. This review summarizes the different mechanisms through which SCFAs act in cells. According to the latest research, the regulatory role of SCFAs in the innate immune system, including in NLRP3 inflammasomes, receptors of TLR family members, neutrophils, macrophages, natural killer cells, eosinophils, basophils and innate lymphocyte subsets, is emphasized. The regulatory role of SCFAs in the adaptive immune system, including in T-cell subsets, B cells, and plasma cells, is also highlighted. In addition, we discuss the role that SCFAs play in regulating allergic airway inflammation, colitis, and osteoporosis by influencing the immune system. These findings provide evidence for determining treatment options based on metabolic regulation.
Collapse
Affiliation(s)
- Xiao-feng Liu
- Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Wuxi, China
| | - Jia-hao Shao
- Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Wuxi, China
| | - Yi-Tao Liao
- Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Wuxi, China
| | - Li-Ning Wang
- School of Chinese Medicine, School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Yuan Jia
- Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Wuxi, China
| | - Peng-jun Dong
- Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Wuxi, China
| | - Zhi-zhong Liu
- Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Wuxi, China
| | - Dan-dan He
- Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Wuxi, China
| | - Chao Li
- Department of Spine, Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Wuxi, China
| | - Xian Zhang
- Department of Spine, Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Wuxi, China
| |
Collapse
|
26
|
Donati Zeppa S, Agostini D, Ferrini F, Gervasi M, Barbieri E, Bartolacci A, Piccoli G, Saltarelli R, Sestili P, Stocchi V. Interventions on Gut Microbiota for Healthy Aging. Cells 2022; 12:cells12010034. [PMID: 36611827 PMCID: PMC9818603 DOI: 10.3390/cells12010034] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/15/2022] [Accepted: 12/17/2022] [Indexed: 12/24/2022] Open
Abstract
In recent years, the improvement in health and social conditions has led to an increase in the average lifespan. Since aging is the most important risk factor for the majority of chronic human diseases, the development of therapies and intervention to stop, lessen or even reverse various age-related morbidities is an important target to ameliorate the quality of life of the elderly. The gut microbiota, that is, the complex ecosystem of microorganisms living in the gastrointestinal tract, plays an important role, not yet fully understood, in maintaining the host's health and homeostasis, influencing metabolic, oxidative and cognitive status; for this reason, it is also named "the forgotten endocrine organ" or "the second brain". On the other hand, the gut microbiota diversity and richness are affected by unmodifiable factors, such as aging and sex, and modifiable ones, such as diet, pharmacological therapies and lifestyle. In this review, we discuss the changes, mostly disadvantageous, for human health, induced by aging, in microbiota composition and the effects of dietary intervention, of supplementation with probiotics, prebiotics, synbiotics, psychobiotics and antioxidants and of physical exercise. The development of an integrated strategy to implement microbiota health will help in the goal of healthy aging.
Collapse
Affiliation(s)
- Sabrina Donati Zeppa
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Deborah Agostini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Fabio Ferrini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
- Correspondence: (F.F.); (M.G.)
| | - Marco Gervasi
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
- Correspondence: (F.F.); (M.G.)
| | - Elena Barbieri
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Alessia Bartolacci
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Giovanni Piccoli
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Roberta Saltarelli
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Piero Sestili
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Vilberto Stocchi
- Department of Human Science for Promotion of Quality of Life, Univerity San Raffaele, 00166 Rome, Italy
| |
Collapse
|
27
|
Cao Y, Han S, Lu H, Luo Y, Guo T, Wu Q, Luo F. Targeting mTOR Signaling by Dietary Polyphenols in Obesity Prevention. Nutrients 2022; 14:nu14235171. [PMID: 36501200 PMCID: PMC9735788 DOI: 10.3390/nu14235171] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 11/29/2022] [Accepted: 12/01/2022] [Indexed: 12/09/2022] Open
Abstract
Dietary polyphenols can be utilized to treat obesity and chronic disorders linked to it. Dietary polyphenols can inhibit pre-adipocyte proliferation, adipocyte differentiation, and triglyceride accumulation; meanwhile, polyphenols can also stimulate lipolysis and fatty acid β-oxidation, but the molecular mechanisms of anti-obesity are still unclear. The mechanistic target of rapamycin (mTOR) is a protein kinase that regulates cell growth, survival, metabolism, and immunity. mTOR signaling is also thought to play a key role in the development of metabolic diseases such as obesity. Recent studies showed that dietary polyphenols could target mTOR to reduce obesity. In this review, we systematically summarized the research progress of polyphenols in preventing obesity through the mTOR signaling pathway. Mechanistically, polyphenols can target multiple signaling pathways and gut microbiota to regulate the mTOR signaling pathway to exert anti-obesity effects. The main mechanisms include: modulating lipid metabolism, adipogenesis, inflammation, etc. Dietary polyphenols exerting an anti-obesity effect by targeting mTOR signaling will broaden our understanding of the anti-obesity mechanisms of polyphenols and provide valuable insights for researchers in this novel field.
Collapse
Affiliation(s)
- Yunyun Cao
- Hunan Provincial Key Laboratory of Grain-Oil Deep Process and Quality Control, Hunan Provincial Key Laboratory of Forestry Edible Resources Safety and Processing, Hunan Provincial Key Laboratory of Processed Food for Special Medical Purpose, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, China
| | - Shuai Han
- Hunan Provincial Key Laboratory of Grain-Oil Deep Process and Quality Control, Hunan Provincial Key Laboratory of Forestry Edible Resources Safety and Processing, Hunan Provincial Key Laboratory of Processed Food for Special Medical Purpose, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, China
| | - Han Lu
- Hunan Provincial Key Laboratory of Grain-Oil Deep Process and Quality Control, Hunan Provincial Key Laboratory of Forestry Edible Resources Safety and Processing, Hunan Provincial Key Laboratory of Processed Food for Special Medical Purpose, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, China
| | - Yi Luo
- Department of Clinic Medicine, Xiangya School of Medicine, Central South University, Changsha 410008, China
| | - Tianyi Guo
- Hunan Provincial Key Laboratory of Grain-Oil Deep Process and Quality Control, Hunan Provincial Key Laboratory of Forestry Edible Resources Safety and Processing, Hunan Provincial Key Laboratory of Processed Food for Special Medical Purpose, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, China
| | - Qi Wu
- Hunan Provincial Key Laboratory of Grain-Oil Deep Process and Quality Control, Hunan Provincial Key Laboratory of Forestry Edible Resources Safety and Processing, Hunan Provincial Key Laboratory of Processed Food for Special Medical Purpose, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, China
| | - Feijun Luo
- Hunan Provincial Key Laboratory of Grain-Oil Deep Process and Quality Control, Hunan Provincial Key Laboratory of Forestry Edible Resources Safety and Processing, Hunan Provincial Key Laboratory of Processed Food for Special Medical Purpose, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, China
- Correspondence:
| |
Collapse
|
28
|
The Impacts of Iron Overload and Ferroptosis on Intestinal Mucosal Homeostasis and Inflammation. Int J Mol Sci 2022; 23:ijms232214195. [PMID: 36430673 PMCID: PMC9697168 DOI: 10.3390/ijms232214195] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/12/2022] [Accepted: 11/15/2022] [Indexed: 11/18/2022] Open
Abstract
Intestinal homeostasis is maintained through the interplay of the intestinal mucosa, local and systemic immune factors, and the microbial content of the gut. Iron is a trace mineral in most organisms, including humans, which is essential for growth, systemic metabolism and immune response. Paradoxically, excessive iron intake and/or high iron status can be detrimental to iron metabolism in the intestine and lead to iron overload and ferroptosis-programmed cell death mediated by iron-dependent lipid peroxidation within cell membranes, which contributes to several intestinal diseases. In this review, we comprehensively review recent findings on the impacts of iron overload and ferroptosis on intestinal mucosal homeostasis and inflammation and then present the progress of iron overload and ferroptosis-targeting therapy in intestinal diseases. Understanding the involved mechanisms can provide a new understanding of intestinal disease pathogenesis and facilitate advanced preventive and therapeutic strategies for intestinal dysfunction and diseases.
Collapse
|
29
|
Xu L, Zhang Q, Dou X, Wang Y, Wang J, Zhou Y, Liu X, Li J. Fecal microbiota transplantation from young donor mice improves ovarian function in aged mice. J Genet Genomics 2022; 49:1042-1052. [PMID: 35654347 DOI: 10.1016/j.jgg.2022.05.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 05/18/2022] [Accepted: 05/19/2022] [Indexed: 12/29/2022]
Abstract
Advanced maternal age is characterized by declines in the quantity and quality of oocytes in the ovaries, and the aging process is accompanied by changes in gut microbiota composition. However, little is known about the relationship between gut microbiota and ovarian aging. By using fecal microbiota transplantation (FMT) to transplant material from young (5-week-old) into aged (42-week-old) mice, we find that the composition of gut microbiota in FMT-treated mice presents a "younger-like phenotype" and an increase of commensal bacteria, such as Bifidobacterium and Ruminococcaceae. Moreover, the FMT-treated mice show increased anti-inflammatory cytokine IL-4 and decreased pro-inflammatory cytokine IFN-γ. Fertility tests for assessing ovarian function reveal that the first litter size of female FMT-treated mice is significantly higher than that of the non-FMT group. Morphology analysis demonstrates a dramatic decrease in follicle atresia and apoptosis as well as an increase in cellular proliferation in the ovaries of the FMT-treated mice. Our results also show that FMT improves the immune microenvironment in aged ovaries, with decreased macrophages and macrophage-derived multinucleated giant cells (MNGCs). These results suggest that FMT from young donors could be a good choice for delaying ovarian aging.
Collapse
Affiliation(s)
- Li Xu
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Qiankun Zhang
- Department of Pathogen Biology-Microbiology Division, State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, China; Shanghai Personal Biotechnology, Shanghai 200231, China
| | - Xiaowei Dou
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, China; Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210003, China
| | - Yipeng Wang
- Department of Pathogen Biology-Microbiology Division, State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Jianwei Wang
- Department of Pathogen Biology-Microbiology Division, State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, China; Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, Jiangsu 215003, China
| | - Yong Zhou
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, China; Fertility Preservation Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong 510317, China
| | - Xingyin Liu
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, China; Department of Pathogen Biology-Microbiology Division, State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, China.
| | - Jing Li
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, China.
| |
Collapse
|
30
|
Tunbridge M, Perkins GB, Singer J, Salehi T, Ying T, Grubor-Bauk B, Barry S, Sim B, Hissaria P, Chadban SJ, Coates PT. Rapamycin and inulin for booster vaccine response stimulation (RIVASTIM)—rapamycin: study protocol for a randomised, controlled trial of immunosuppression modification with rapamycin to improve SARS-CoV-2 vaccine response in kidney transplant recipients. Trials 2022; 23:780. [PMID: 36109788 PMCID: PMC9477178 DOI: 10.1186/s13063-022-06634-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 08/04/2022] [Indexed: 11/10/2022] Open
Abstract
Kidney transplant recipients are at an increased risk of severe COVID-19-associated hospitalisation and death. Vaccination has been a key public health strategy to reduce disease severity and infectivity, but the effectiveness of COVID vaccines is markedly reduced in kidney transplant recipients. Urgent strategies to enhance vaccine efficacy are needed. Methods: RIVASTIM-rapamycin is a multicentre, randomised, controlled trial examining the effect of immunosuppression modification prior to a third dose of COVID-19 vaccine in kidney transplant recipients who have failed to develop protective immunity to a 2-dose COVID-19 vaccine schedule. Participants will be randomised 1:1 to either remain on standard of care immunosuppression with tacrolimus, mycophenolate, and prednisolone (control) or cease mycophenolate and commence sirolimus (intervention) for 4 weeks prior to and following vaccination. The primary outcome is the proportion of participants in each trial arm who develop protective serological neutralisation of live SARS-CoV-2 virus at 4–6 weeks following a third COVID-19 vaccination. Secondary outcomes include SARS-CoV-receptor binding domain IgG, vaccine-specific immune cell populations and responses, and the safety and tolerability of sirolimus switch. Discussion: Immunosuppression modification strategies may improve immunological vaccine response. We hypothesise that substituting the mTOR inhibitor sirolimus for mycophenolate in a triple drug regimen will enhance humoral and cell-mediated responses to COVID vaccination for kidney transplant recipients. Trial registration: Australia New Zealand Clinical Trials Registry ACTRN12621001412820. Registered on 20 October 2021; https://www.anzctr.org.au/Trial/Registration/TrialReview.aspx?id=382891&isReview=true
Collapse
|
31
|
He L, Yang FQ, Tang P, Gao TH, Yang CX, Tan L, Yue P, Hua YN, Liu SJ, Guo JL. Regulation of the intestinal flora: A potential mechanism of natural medicines in the treatment of type 2 diabetes mellitus. Biomed Pharmacother 2022; 151:113091. [PMID: 35576662 DOI: 10.1016/j.biopha.2022.113091] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 04/24/2022] [Accepted: 05/04/2022] [Indexed: 11/02/2022] Open
Abstract
Diabetes mellitus comprises a group of heterogeneous disorders, which are usually subdivided into type 1 diabetes mellitus (T1DM) and type 2 diabetes mellitus (T2DM). Both genetic and environmental factors have been implicated in the onset of diabetes. Type 1 diabetes primarily involves autoimmune insulin deficiency. In comparison, type 2 diabetes is contributed by the pathological state of insulin deficiency and insulin resistance. In recent years, significant differences were found in the abundance of microflora, intestinal barrier, and intestinal metabolites in diabetic subjects when compared to normal subjects. To further understand the relationship between diabetes mellitus and intestinal flora, this paper summarizes the interaction mechanism between diabetes mellitus and intestinal flora. Furthermore, the natural compounds found to treat diabetes through intestinal flora were classified and summarized. This review is expected to provide a valuable resource for the development of new diabetic drugs and the applications of natural compounds.
Collapse
Affiliation(s)
- Liying He
- Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Fang-Qing Yang
- Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Pan Tang
- Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Ting-Hui Gao
- Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Cai-Xia Yang
- Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Li Tan
- Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Pan Yue
- Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Ya-Nan Hua
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Si-Jing Liu
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Jin-Lin Guo
- Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|
32
|
Acne, Microbiome, and Probiotics: The Gut–Skin Axis. Microorganisms 2022; 10:microorganisms10071303. [PMID: 35889022 PMCID: PMC9318165 DOI: 10.3390/microorganisms10071303] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 06/20/2022] [Accepted: 06/25/2022] [Indexed: 11/17/2022] Open
Abstract
The objective of this narrative review was to check the influence of the human microbiota in the pathogenesis of acne and how the treatment with probiotics as adjuvant or alternative therapy affects the evolution of acne vulgaris. Acne is a chronic inflammatory skin disease involving the pilosebaceous units. The pathogenesis of acne is complex and multifactorial involving genetic, metabolic, and hormonal factors in which both skin and gut microbiota are implicated. Numerous studies have shown the bidirectionality between the intestinal microbiota and skin homeostasis, a communication mainly established by modifying the immune system. Increased data on the mechanisms of action regarding the relevance of Cutibacterium acnes, as well as the importance of the gut–skin axis, are becoming known. Diverse and varied in vitro studies have shown the potential beneficial effects of probiotics in this context. Clinical trials with both topical and oral probiotics are scarce, although they have shown positive results, especially with oral probiotics through the modulation of the intestinal microbiota, generating an anti-inflammatory response and restoring intestinal integrity, or through metabolic pathways involving insulin-like growth factor I (IGF-1). Given the aggressiveness of some standard acne treatments, probiotics should continue to be investigated as an alternative or adjuvant therapy.
Collapse
|
33
|
Özcan E, Lum GR, Hsiao EY. Interactions between the gut microbiome and ketogenic diet in refractory epilepsy. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2022; 167:217-249. [PMID: 36427956 DOI: 10.1016/bs.irn.2022.06.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Epilepsy is one of the most common neurological diseases globally, afflicting approximately 50 million people worldwide. While many antiepileptic drugs exist, an estimated one-third of individuals do not respond to available medications. The high fat, low carbohydrate ketogenic diet (KD) has been used to treat refractory epilepsy in cases when existing antiepileptic drugs fail. However, there are many variations of the KD, each of which varies greatly in its efficacy and side effects. Increasing evidence suggests that interactions between the KD and gut microbiome may modulate the effects of the diet on host physiology. Herein, we review existing evidence of microbiome differences in epileptic individuals compared to healthy controls. We highlight in particular both clinical and animal studies revealing effects of the KD on the composition and function of the microbiome, as well as proof-of-concept animal studies that implicate the microbiome in the antiseizure effects of the KD. We further synthesize findings suggesting that variations in clinical KD formulations may differentially influence host physiology and discuss the gut microbial interactions with specific dietary factors that may play a role. Overall, understanding interactions between the gut microbiota and specific nutritional components of clinical KDs could reveal foundational mechanisms that underlie the effectiveness, variability, and side effects of different KDs, with the potential to lead to precision nutritional and microbiome-based approaches to treat refractory epilepsy.
Collapse
Affiliation(s)
- Ezgi Özcan
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA, United States.
| | - Gregory R Lum
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Elaine Y Hsiao
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA, United States.
| |
Collapse
|
34
|
Li J, Li Y, Duan W, Zhao Z, Yang L, Wei W, Li J, Li Y, Yu Y, Dai B, Guo R. Shugan granule contributes to the improvement of depression-like behaviors in chronic restraint stress-stimulated rats by altering gut microbiota. CNS Neurosci Ther 2022; 28:1409-1424. [PMID: 35713215 PMCID: PMC9344086 DOI: 10.1111/cns.13881] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 05/16/2022] [Accepted: 05/24/2022] [Indexed: 12/14/2022] Open
Abstract
Aim The investigation aims to evaluate the potential effect of Shugan Granule (SGKL) on the gut, brain, and behaviors in rats exposed to chronic restraint stress (CRS). Methods The fecal microbiota and metabolite changes were studied in rats exposed to CRS and treated with SGKL (0.1 mg/kg/day). Depressive behaviors of these rats were determined through an open‐field experiment, forced swimming test, sucrose preference, and weighing. Moreover, LPS‐stimulated microglia and CRS‐stimulated rats were treated with SGKL to investigate the regulation between SGKL and the PI3K/Akt/pathway, which is inhibited by LY294002, a PI3K inhibitor. Results (i) SGKL improved the altered behaviors in CRS‐stimulated rats; (ii) SGKL ameliorated the CRS‐induced neuronal degeneration and tangled nerve fiber and also contributed to the recovery of intestinal barrier injury in these rats; (iii) SGKL inhibited the hippocampus elevations of TNF‐α, IL‐1β, and IL‐6 in response to CRS modeling; (iv) based on the principal coordinates analysis (PCoA), SGKL altered α‐diversity indices and shifted β‐diversity in CRS‐stimulated rats; (v) at the genus level, SGKL decreased the CRS‐enhanced abundance of Bacteroides; (vi) Butyricimonas and Candidatus Arthromitus were enriched in SGKL‐treated rats; (vii) altered gut microbiota and metabolites were correlated with behaviors, inflammation, and PI3K/Akt/mTOR pathway; (viii) SGKL increased the LPS‐decreased phosphorylation of the PI3K/Akt/mTOR pathway in microglia and inhibited the LPS‐induced microglial activation; (ix) PI3K/Akt/mTOR pathway inactivation reversed the SGKL effects in CRS rats. Conclusion SGKL targets the PI3K/Akt/mTOR pathway by altering gut microbiota and metabolites, which ameliorates altered behavior and inflammation in the hippocampus.
Collapse
Affiliation(s)
- Junnan Li
- Second Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Yannan Li
- Second Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Wenzhe Duan
- Second Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Zhonghui Zhao
- Second Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Lixuan Yang
- Second Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Wei Wei
- Second Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Jingchun Li
- Second Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Yang Li
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Yao Yu
- Beijing Changping Hospital of Integrated Chinese and Western Medicine, Beijing, China
| | - Baoan Dai
- Second Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Rongjuan Guo
- Department of Neurology, Dongfang Hospital Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
35
|
The increasing importance of the gut microbiome in acne vulgaris. Folia Microbiol (Praha) 2022; 67:825-835. [PMID: 35711021 DOI: 10.1007/s12223-022-00982-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 05/26/2022] [Indexed: 11/04/2022]
Abstract
Acne is a frequently presented dermatological condition brought about by an interplay among inflammation, increased sebum production, hyperkeratinisation, and predominantly Propionibacterium acnes (renamed as Cutibacterium acnes) proliferation, leading to debilitating psychological scars. However, it has been shown that it is the loss of microbial diversity in the skin and the imbalance among C. acnes phylotypes that brings about acne rather than the C. acnes species as a whole. Interestingly, recent evidence suggests that other microorganisms may be implicated, such as the fungi Malassezia and the bacteria Cutibacterium granulosum. A plethora of scientific evidence suggests that the gut microbiome is implicated in the overall health and physiology of the host; studies show that the gut microbiome of acne patients is distinct and depicts less microbial diversity compared to individuals without acne. Herein, using the key terms: acne, C. acnes, IGF-1, sebum, and gut microbiome, we carried out a review of the literature, using Google Scholar and PubMed, and discussed the role of the gut and skin microbiome in relation to acne, as a narrative review. The role of hormones, diet, sebum, and stress in relation to the gut microbiome was also investigated. Therapeutic implications and the use of pre-/postbiotics are also deliberated upon. In this light, future research should investigate the relationship between the gut microbiome and the agreed upon factors of acne pathology, potentially leading to the discovery of novel acne treatments with milder side effects.
Collapse
|
36
|
Gao J, Ma L, Yin J, Liu G, Ma J, Xia S, Gong S, Han Q, Li T, Chen Y, Yin Y. Camellia ( Camellia oleifera bel.) seed oil reprograms gut microbiota and alleviates lipid accumulation in high fat-fed mice through the mTOR pathway. Food Funct 2022; 13:4977-4992. [PMID: 35452062 DOI: 10.1039/d1fo04075h] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Camellia (Camellia oleifera bel.) seed oil (CO) is extensively used as an edible oil in China and Asian countries owing to its high nutritional and medicinal values. It has been shown that a high-fat diet enhances lipid accumulation and induces intestinal microbiota imbalance in mice. However, it is still to be learned whether CO prevents dyslipidemia through gut microbiota. Here, using 16S rRNA gene sequencing analysis of the gut microbiota, we found that oral CO relieved lipid accumulation and reversed gut microbiota dysbiosis. Compared to mice (C57BL/6J male mice) fed a high-fat diet, treatment with CO regulated the composition and functional profiling communities related to the lipid metabolism of gut microbiota. The abundances of Dubosiella, Lactobacillus, and Alistipes were markedly increased in CO supplementation mice. In addition, the colon levels of isobutyric acid, pentanoic acid, and isovaleric acid were similar between the control and CO supplementation mice. Besides, the results indicated that CO supplementation in mice alleviated lipid droplet accumulation in the hepatocytes and subcutaneous adipose tissue, although the liver index did not show a difference. Notably, CO supplementation for 6 weeks significantly reduced the levels of LDL, TC, and TG, while enhancing the level of HDL in serum and liver. Meanwhile, we also identified that CO supplementation suppressed the mammalian target of rapamycin (mTOR) signaling pathway in high fat-fed (HF-fed) mice. Taken together, our results suggest that CO improved dyslipidemia and alleviated lipid accumulation in HF-fed mice, the molecular mechanisms possibly associated with the reorganization of gut microbiota, in particular, Alistipes and Dubosiella, mediated the inhibition of the mTOR pathway.
Collapse
Affiliation(s)
- Jing Gao
- Research Institute of Oil Tea Camellia, Hunan Academy of Forestry, Shao shan South Road, No. 658, Changsha 410004, China. .,National Engineering Research Center for Oil Tea Camellia, Changsha 410004, China.,Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Chinese Academy of Sciences, Changsha, Hunan, China.
| | - Li Ma
- Research Institute of Oil Tea Camellia, Hunan Academy of Forestry, Shao shan South Road, No. 658, Changsha 410004, China. .,National Engineering Research Center for Oil Tea Camellia, Changsha 410004, China
| | - Jie Yin
- College of Animal Science and Technology, Hunan Co-Innovation Center of Animal Production Safety, Hunan Agricultural University, Changsha, China
| | - Gang Liu
- College of Animal Science and Technology, Hunan Co-Innovation Center of Animal Production Safety, Hunan Agricultural University, Changsha, China
| | - Jie Ma
- College of Animal Science and Technology, Hunan Co-Innovation Center of Animal Production Safety, Hunan Agricultural University, Changsha, China
| | - SiTing Xia
- College of Animal Science and Technology, Hunan Co-Innovation Center of Animal Production Safety, Hunan Agricultural University, Changsha, China
| | - SaiMing Gong
- College of Animal Science and Technology, Hunan Co-Innovation Center of Animal Production Safety, Hunan Agricultural University, Changsha, China
| | - Qi Han
- College of Animal Science and Technology, Hunan Co-Innovation Center of Animal Production Safety, Hunan Agricultural University, Changsha, China
| | - TieJun Li
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Chinese Academy of Sciences, Changsha, Hunan, China.
| | - YongZhong Chen
- Research Institute of Oil Tea Camellia, Hunan Academy of Forestry, Shao shan South Road, No. 658, Changsha 410004, China. .,National Engineering Research Center for Oil Tea Camellia, Changsha 410004, China
| | - YuLong Yin
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Chinese Academy of Sciences, Changsha, Hunan, China. .,College of Animal Science and Technology, Hunan Co-Innovation Center of Animal Production Safety, Hunan Agricultural University, Changsha, China
| |
Collapse
|
37
|
Cao LH, He HJ, Zhao YY, Wang ZZ, Jia XY, Srivastava K, Miao MS, Li XM. Food Allergy-Induced Autism-Like Behavior is Associated with Gut Microbiota and Brain mTOR Signaling. J Asthma Allergy 2022; 15:645-664. [PMID: 35603013 PMCID: PMC9122063 DOI: 10.2147/jaa.s348609] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 03/30/2022] [Indexed: 11/25/2022] Open
Abstract
Purpose Food allergy-induced autism-like behavior has been increasing for decades, but the causal drivers of this association are unclear. We sought to test the association of gut microbiota and mammalian/mechanistic target of rapamycin (mTOR) signaling with cow’s milk allergy (CMA)-induced autism pathogenesis. Methods Mice were sensitized intragastrically with whey protein containing cholera toxin before sensitization on intraperitoneal injection with whey-containing alum, followed by intragastric allergen challenge to induce experimental CMA. The food allergic immune responses, ASD-like behavioral tests and changes in the mTOR signaling pathway and gut microbial community structure were performed. Results CMA mice showed autism-like behavioral abnormalities and several distinct biomarkers. These include increased levels of 5-hydroxymethylcytosine (5-hmC) in the hypothalamus; c-Fos were predominantly located in the region of the lateral orbital prefrontal cortex (PFC), but not ventral; decreased serotonin 1A in amygdala and PFC. CMA mice exhibited a specific microbiota signature characterized by coordinate changes in the abundance of taxa of several bacterial genera, including the Lactobacillus. Interestingly, the changes were accompanied by promoted mTOR signaling in the brain of CMA mice. Conclusion We found that disease-associated microbiota and mTOR activation may thus play a pathogenic role in the intestinal, immunological, and psychiatric Autism Spectrum Disorder (ASD)-like symptoms seen in CAM associated autism. However, this is only a preliminary study, and their mechanisms require further investigation.
Collapse
Affiliation(s)
- Li-Hua Cao
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan Province, People’s Republic of China
| | - Hong-Juan He
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan Province, People’s Republic of China
| | - Yuan-Yuan Zhao
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan Province, People’s Republic of China
| | - Zhen-Zhen Wang
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan Province, People’s Republic of China
| | - Xing-Yuan Jia
- Department of Pharmacy, Henan Province Hospital of Traditional Chinese Medicine, Zhengzhou, 450046, Henan Province, People’s Republic of China
| | - Kamal Srivastava
- Department of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY, 10595, USA
- General Nutraceutical Technology, Elmsford, NY, 10523, USA
| | - Ming-San Miao
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan Province, People’s Republic of China
| | - Xiu-Min Li
- Department of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY, 10595, USA
- Department of Otolaryngology, New York Medical College, Valhalla, NY, 10595, USA
- Correspondence: Xiu-Min Li; Ming-San Miao, Tel +1 914-594-4197, Fax +1 371-65962546, Email ;
| |
Collapse
|
38
|
The Athlete Gut Microbiome and its Relevance to Health and Performance: A Review. Sports Med 2022; 52:119-128. [PMID: 36396898 PMCID: PMC9734205 DOI: 10.1007/s40279-022-01785-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/25/2022] [Indexed: 11/19/2022]
Abstract
The human gut microbiome is a complex ecosystem of microorganisms that play an important role in human health, influencing functions such as vitamin uptake, digestion and immunomodulation. While research of the gut microbiome has expanded considerably over the past decade, some areas such as the relationship between exercise and the microbiome remain relatively under investigated. Despite this, multiple studies have shown a potential bidirectional relationship between exercise and the gut microbiome, with some studies demonstrating the possibility of influencing this relationship. This, in turn, could provide a useful route to influence athletic performance via microbiome manipulation, a valuable prospect for many elite athletes and their teams. The evidence supporting the potential benefits of pursuing this route and associated future perspectives are discussed in this review.
Collapse
|
39
|
Mahmud MR, Akter S, Tamanna SK, Mazumder L, Esti IZ, Banerjee S, Akter S, Hasan MR, Acharjee M, Hossain MS, Pirttilä AM. Impact of gut microbiome on skin health: gut-skin axis observed through the lenses of therapeutics and skin diseases. Gut Microbes 2022; 14:2096995. [PMID: 35866234 PMCID: PMC9311318 DOI: 10.1080/19490976.2022.2096995] [Citation(s) in RCA: 166] [Impact Index Per Article: 55.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 06/10/2022] [Accepted: 06/27/2022] [Indexed: 02/08/2023] Open
Abstract
The human intestine hosts diverse microbial communities that play a significant role in maintaining gut-skin homeostasis. When the relationship between gut microbiome and the immune system is impaired, subsequent effects can be triggered on the skin, potentially promoting the development of skin diseases. The mechanisms through which the gut microbiome affects skin health are still unclear. Enhancing our understanding on the connection between skin and gut microbiome is needed to find novel ways to treat human skin disorders. In this review, we systematically evaluate current data regarding microbial ecology of healthy skin and gut, diet, pre- and probiotics, and antibiotics, on gut microbiome and their effects on skin health. We discuss potential mechanisms of the gut-skin axis and the link between the gut and skin-associated diseases, such as psoriasis, atopic dermatitis, acne vulgaris, rosacea, alopecia areata, and hidradenitis suppurativa. This review will increase our understanding of the impacts of gut microbiome on skin conditions to aid in finding new medications for skin-associated diseases.
Collapse
Affiliation(s)
- Md. Rayhan Mahmud
- Department of Production Animal Medicine, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Sharmin Akter
- Department of Microbiology, Jagannath University, Dhaka, Bangladesh
| | | | - Lincon Mazumder
- Department of Microbiology, Jagannath University, Dhaka, Bangladesh
| | - Israt Zahan Esti
- Department of Microbiology, Jagannath University, Dhaka, Bangladesh
| | | | - Sumona Akter
- Department of Microbiology, Jagannath University, Dhaka, Bangladesh
| | | | - Mrityunjoy Acharjee
- Department of Bioscience, Graduate School of Science and Technology, Shizuoka University, Shizuoka, Japan
| | | | | |
Collapse
|
40
|
Fat of the Gut: Epithelial Phospholipids in Inflammatory Bowel Diseases. Int J Mol Sci 2021; 22:ijms222111682. [PMID: 34769112 PMCID: PMC8584226 DOI: 10.3390/ijms222111682] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/20/2021] [Accepted: 10/27/2021] [Indexed: 12/15/2022] Open
Abstract
Inflammatory bowel diseases (IBD) comprise a distinct set of clinical symptoms resulting from chronic inflammation within the gastrointestinal (GI) tract. Despite the significant progress in understanding the etiology and development of treatment strategies, IBD remain incurable for thousands of patients. Metabolic deregulation is indicative of IBD, including substantial shifts in lipid metabolism. Recent data showed that changes in some phospholipids are very common in IBD patients. For instance, phosphatidylcholine (PC)/phosphatidylethanolamine (PE) and lysophosphatidylcholine (LPC)/PC ratios are associated with the severity of the inflammatory process. Composition of phospholipids also changes upon IBD towards an increase in arachidonic acid and a decrease in linoleic and a-linolenic acid levels. Moreover, an increase in certain phospholipid metabolites, such as lysophosphatidylcholine, sphingosine-1-phosphate and ceramide, can result in enhanced intestinal inflammation, malignancy, apoptosis or necroptosis. Because some phospholipids are associated with pathogenesis of IBD, they may provide a basis for new strategies to treat IBD. Current attempts are aimed at controlling phospholipid and fatty acid levels through the diet or via pharmacological manipulation of lipid metabolism.
Collapse
|
41
|
Morrow JD, Castaldi PJ, Chase RP, Yun JH, Lee S, Liu YY, Hersh CP. Peripheral blood microbial signatures in current and former smokers. Sci Rep 2021; 11:19875. [PMID: 34615932 PMCID: PMC8494912 DOI: 10.1038/s41598-021-99238-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 09/15/2021] [Indexed: 12/13/2022] Open
Abstract
The human microbiome has a role in the development of multiple diseases. Individual microbiome profiles are highly personalized, though many species are shared. Understanding the relationship between the human microbiome and disease may inform future individualized treatments. We hypothesize the blood microbiome signature may be a surrogate for some lung microbial characteristics. We sought associations between the blood microbiome signature and lung-relevant host factors. Based on reads not mapped to the human genome, we detected microbial nucleic acids through secondary use of peripheral blood RNA-sequencing from 2,590 current and former smokers with and without chronic obstructive pulmonary disease (COPD) from the COPDGene study. We used the Genome Analysis Toolkit (GATK) microbial pipeline PathSeq to infer microbial profiles. We tested associations between the inferred profiles and lung disease relevant phenotypes and examined links to host gene expression pathways. We replicated our analyses using a second independent set of blood RNA-seq data from 1,065 COPDGene study subjects and performed a meta-analysis across the two studies. The four phyla with highest abundance across all subjects were Proteobacteria, Actinobacteria, Firmicutes and Bacteroidetes. In our meta-analysis, we observed associations (q-value < 0.05) between Acinetobacter, Serratia, Streptococcus and Bacillus inferred abundances and Modified Medical Research Council (mMRC) dyspnea score. Current smoking status was associated (q < 0.05) with Acinetobacter, Serratia and Cutibacterium abundance. All 12 taxa investigated were associated with at least one white blood cell distribution variable. Abundance for nine of the 12 taxa was associated with sex, and seven of the 12 taxa were associated with race. Host-microbiome interaction analysis revealed clustering of genera associated with mMRC dyspnea score and smoking status, through shared links to several host pathways. This study is the first to identify a bacterial microbiome signature in the peripheral blood of current and former smokers. Understanding the relationships between systemic microbial signatures and lung-related phenotypes may inform novel interventions and aid understanding of the systemic effects of smoking.
Collapse
Affiliation(s)
- Jarrett D Morrow
- Channing Division of Network Medicine, Brigham and Women's Hospital, 181 Longwood Avenue, Boston, MA, 02115, USA.
| | - Peter J Castaldi
- Channing Division of Network Medicine, Brigham and Women's Hospital, 181 Longwood Avenue, Boston, MA, 02115, USA
| | - Robert P Chase
- Channing Division of Network Medicine, Brigham and Women's Hospital, 181 Longwood Avenue, Boston, MA, 02115, USA
| | - Jeong H Yun
- Channing Division of Network Medicine, Brigham and Women's Hospital, 181 Longwood Avenue, Boston, MA, 02115, USA
| | - Sool Lee
- Channing Division of Network Medicine, Brigham and Women's Hospital, 181 Longwood Avenue, Boston, MA, 02115, USA
| | - Yang-Yu Liu
- Channing Division of Network Medicine, Brigham and Women's Hospital, 181 Longwood Avenue, Boston, MA, 02115, USA
| | - Craig P Hersh
- Channing Division of Network Medicine, Brigham and Women's Hospital, 181 Longwood Avenue, Boston, MA, 02115, USA
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA
| |
Collapse
|
42
|
Icard P, Loi M, Wu Z, Ginguay A, Lincet H, Robin E, Coquerel A, Berzan D, Fournel L, Alifano M. Metabolic Strategies for Inhibiting Cancer Development. Adv Nutr 2021; 12:1461-1480. [PMID: 33530098 PMCID: PMC8321873 DOI: 10.1093/advances/nmaa174] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 08/14/2020] [Accepted: 12/14/2020] [Indexed: 12/11/2022] Open
Abstract
The tumor microenvironment is a complex mix of cancerous and noncancerous cells (especially immune cells and fibroblasts) with distinct metabolisms. These cells interact with each other and are influenced by the metabolic disorders of the host. In this review, we discuss how metabolic pathways that sustain biosynthesis in cancer cells could be targeted to increase the effectiveness of cancer therapies by limiting the nutrient uptake of the cell, inactivating metabolic enzymes (key regulatory ones or those linked to cell cycle progression), and inhibiting ATP production to induce cell death. Furthermore, we describe how the microenvironment could be targeted to activate the immune response by redirecting nutrients toward cytotoxic immune cells or inhibiting the release of waste products by cancer cells that stimulate immunosuppressive cells. We also examine metabolic disorders in the host that could be targeted to inhibit cancer development. To create future personalized therapies for targeting each cancer tumor, novel techniques must be developed, such as new tracers for positron emission tomography/computed tomography scan and immunohistochemical markers to characterize the metabolic phenotype of cancer cells and their microenvironment. Pending personalized strategies that specifically target all metabolic components of cancer development in a patient, simple metabolic interventions could be tested in clinical trials in combination with standard cancer therapies, such as short cycles of fasting or the administration of sodium citrate or weakly toxic compounds (such as curcumin, metformin, lipoic acid) that target autophagy and biosynthetic or signaling pathways.
Collapse
Affiliation(s)
- Philippe Icard
- Université Caen Normandie, Medical School, CHU de Caen, Caen, France
- Normandie Université, UNICAEN, INSERM U1086, Interdisciplinary Research Unit for Cancer Prevention and Treatment, Centre de Lutte Contre le Cancer Centre François Baclesse, Caen, France
- Service de Chirurgie Thoracique, Hôpital Cochin, Hôpitaux Universitaires Paris Centre, AP-HP, Paris-Descartes University, Paris, France
| | - Mauro Loi
- Radiotherapy Department, Humanitas Cancer Center, Rozzano, Milan, Italy
| | - Zherui Wu
- School of Medicine, Shenzhen University, Shenzhen, Guangdong, China
- INSERM UMR-S 1124, Cellular Homeostasis and Cancer, Paris-Descartes University, Paris, France
| | - Antonin Ginguay
- Service de Biochimie, Hôpital Cochin, Hôpitaux Universitaires Paris-Centre, AP-HP, Paris, France
- EA4466 Laboratoire de Biologie de la Nutrition, Faculté de Pharmacie de Paris, Université Paris-Descartes, Sorbonne Paris Cité, Paris, France
| | - Hubert Lincet
- INSERM U1052, CNRS UMR5286, Cancer Research Center of Lyon (CRCL), France
- ISPB, Faculté de Pharmacie, Université Lyon 1, Lyon, France
| | - Edouard Robin
- Service de Chirurgie Thoracique, Hôpital Cochin, Hôpitaux Universitaires Paris Centre, AP-HP, Paris-Descartes University, Paris, France
| | - Antoine Coquerel
- INSERM U1075, Comete “Mobilités: Attention, Orientation, Chronobiologie”, Université Caen, Caen, France
| | - Diana Berzan
- Service de Chirurgie Thoracique, Hôpital Cochin, Hôpitaux Universitaires Paris Centre, AP-HP, Paris-Descartes University, Paris, France
| | - Ludovic Fournel
- Service de Chirurgie Thoracique, Hôpital Cochin, Hôpitaux Universitaires Paris Centre, AP-HP, Paris-Descartes University, Paris, France
- INSERM UMR-S 1124, Cellular Homeostasis and Cancer, Paris-Descartes University, Paris, France
| | - Marco Alifano
- Service de Chirurgie Thoracique, Hôpital Cochin, Hôpitaux Universitaires Paris Centre, AP-HP, Paris-Descartes University, Paris, France
- INSERM U1138, Integrative Cancer Immunology, Paris, France
| |
Collapse
|
43
|
Wu X, Xiong F, Fang H, Zhang J, Chang M. Crosstalks between NOD1 and Histone H2A Contribute to Host Defense against Streptococcus agalactiae Infection in Zebrafish. Antibiotics (Basel) 2021; 10:antibiotics10070861. [PMID: 34356784 PMCID: PMC8300774 DOI: 10.3390/antibiotics10070861] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/08/2021] [Accepted: 07/10/2021] [Indexed: 11/30/2022] Open
Abstract
Correlation studies about NOD1 and histones have not been reported. In the present study, we report the functional correlation between NOD1 and the histone H2A variant in response to Streptococcus agalactiae infection. In zebrafish, NOD1 deficiency significantly promoted S. agalactiae proliferation and decreased larval survival. Transcriptome analysis revealed that the significantly enriched pathways in NOD1−/− adult zebrafish were mainly involved in immune and metabolism. Among 719 immunity-associated DEGs at 48 hpi, 74 DEGs regulated by NOD1 deficiency were histone variants. Weighted gene co-expression network analysis identified that H2A, H2B, and H3 had significant associations with NOD1 deficiency. Above all, S. agalactiae infection could induce the expression of intracellular histone H2A, as well as NOD1 colocalized with histone H2A, both in the cytoplasm and cell nucleus in the case of S. agalactiae infection. The overexpression of H2A variants such as zfH2A-6 protected against S. agalactiae infection and could improve cell survival in NOD1-deficient cells. Furthermore, NOD1 could interact with zfH2A-6 and cooperate with zfH2A-6 to inhibit the proliferation of S. agalactiae. NOD1 also showed a synergetic effect in inducing the expression of many antibacterial genes, especially antibacterial pattern recognition receptors PGRP2, PGRP5, and PGRP6. Collectively, these results firstly highlight the roles of NOD1 deficiency in the regulation of immune-related and metabolic pathways, and the correlation between zebrafish NOD1 and histone H2A variant in the defense against S. agalactiae infection.
Collapse
Affiliation(s)
- Xiaoman Wu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Key Laboratory of Aquaculture Disease Control, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; (X.W.); (F.X.); (H.F.); (J.Z.)
| | - Fan Xiong
- State Key Laboratory of Freshwater Ecology and Biotechnology, Key Laboratory of Aquaculture Disease Control, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; (X.W.); (F.X.); (H.F.); (J.Z.)
| | - Hong Fang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Key Laboratory of Aquaculture Disease Control, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; (X.W.); (F.X.); (H.F.); (J.Z.)
| | - Jie Zhang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Key Laboratory of Aquaculture Disease Control, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; (X.W.); (F.X.); (H.F.); (J.Z.)
| | - Mingxian Chang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Key Laboratory of Aquaculture Disease Control, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; (X.W.); (F.X.); (H.F.); (J.Z.)
- Innovation Academy for Seed Design, Chinese Academy of Sciences, Wuhan 430072, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Correspondence:
| |
Collapse
|
44
|
Beydoun S, Fardous AM, Saruna MM, Beydoun AG, Sorge JA, Ma H, Aoun G, Unnikrishnan A, Cabelof DC, Heydari AR. Succinylsulfathiazole modulates the mTOR signaling pathway in the liver of c57BL/6 mice via a folate independent mechanism. Exp Gerontol 2021; 150:111387. [PMID: 33957263 PMCID: PMC8165018 DOI: 10.1016/j.exger.2021.111387] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 04/25/2021] [Accepted: 04/28/2021] [Indexed: 01/27/2023]
Abstract
Researchers studying the effect of folate restriction on rodents have resorted to the use of the antibiotic succinylsulfathiazole (SST) in the folate depleted diet to induce a folate deficient status. SST has been used extensively in rodent studies since the 1940s. Its localized effect on the gut bacteria as well as its effectiveness in reducing folate producing species is well documented. The possible overlap between the pathways affected by folate depletion and SST could potentially produce a confounding variable in such studies. In our novel study, we analyzed the effect of SST on folate levels in c57Bl/6 male mice fed folate supplemented and deficient diets. We did not observe any significant difference on growth and weight gain at 21 weeks. SST did not significantly affect folate levels in the plasma, liver and colon tissues; however, it did alter energy metabolism and expression of key genes in the mTOR signaling pathway in the liver. This research sheds light on a possible confounding element when using SST to study folate depletion due to the potential overlap with multiple critical pathways such as mTOR. SUMMARY: The antibiotic succinylsulfathiazole (SST) is used to reduce folate producing bacteria in rodent folate depletion studies. SST can modulate critical energy and nutrient sensing pathways converging onto mTOR signaling, and potentially confounding cancer studies.
Collapse
Affiliation(s)
- Safa Beydoun
- Department of Nutrition and Food Science, Wayne State University, Detroit, MI 48202, USA; Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Ali M Fardous
- Department of Nutrition and Food Science, Wayne State University, Detroit, MI 48202, USA
| | - Michael M Saruna
- Department of Nutrition and Food Science, Wayne State University, Detroit, MI 48202, USA
| | - Ali G Beydoun
- Department of Nutrition and Food Science, Wayne State University, Detroit, MI 48202, USA
| | - Johnathan A Sorge
- Department of Nutrition and Food Science, Wayne State University, Detroit, MI 48202, USA
| | - Hongzhi Ma
- Department of Nutrition and Food Science, Wayne State University, Detroit, MI 48202, USA
| | - Ghada Aoun
- Department of Nutrition and Food Science, Wayne State University, Detroit, MI 48202, USA
| | - Archana Unnikrishnan
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health and Science Center, OK 73104, USA
| | - Diane C Cabelof
- Department of Nutrition and Food Science, Wayne State University, Detroit, MI 48202, USA; Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, MI 48202, USA
| | - Ahmad R Heydari
- Department of Nutrition and Food Science, Wayne State University, Detroit, MI 48202, USA; Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, MI 48202, USA.
| |
Collapse
|
45
|
Zhang YX, Qu SS, Zhang LH, Gu YY, Chen YH, Huang ZY, Liu MH, Zou W, Jiang J, Chen JQ, Wang YJ, Zhou FH. The Role of Ophiopogonin D in Atherosclerosis: Impact on Lipid Metabolism and Gut Microbiota. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2021; 49:1449-1471. [PMID: 34263719 DOI: 10.1142/s0192415x21500683] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Gut microbiota has been proven to play an important role in many metabolic diseases and cardiovascular disease, particularly atherosclerosis. Ophiopogonin D (OPD), one of the effective compounds in Ophiopogon japonicus, is considered beneficial to metabolic syndrome and cardiovascular diseases. In this study, we have illuminated the effect of OPD in ApoE knockout (ApoE[Formula: see text] mice on the development of atherosclerosis and gut microbiota. To investigate the potential ability of OPD to alleviate atherosclerosis, 24 eight-week-old male ApoE[Formula: see text] mice (C57BL/6 background) were fed a high-fat diet (HFD) for 12 weeks, and 8 male C57BL/6 mice were fed a normal diet, serving as the control group. ApoE[Formula: see text] mice were randomly divided into the model group, OPD group, and simvastatin group ([Formula: see text]= 8). After treatment for 12 consecutive weeks, the results showed that OPD treatment significantly decreased the plaque formation and levels of serum lipid compared with those in the model group. In addition, OPD improved oral glucose tolerance and insulin resistance as well as reducing hepatocyte steatosis. Further analysis revealed that OPD might attenuate atherosclerosis through inhibiting mTOR phosphorylation and the consequent lipid metabolism signaling pathways mediated by SREBP1 and SCD1 in vivo and in vitro. Furthermore, OPD treatment led to significant structural changes in gut microbiota and fecal metabolites in HFD-fed mice and reduced the relative abundance of Erysipelotrichaceae genera associated with cholesterol metabolism. Collectively, these findings illustrate that OPD could significantly protect against atherosclerosis, which might be associated with the moderation of lipid metabolism and alterations in gut microbiota composition and fecal metabolites.
Collapse
Affiliation(s)
- Ya-Xin Zhang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, P. R. China
| | - Shan-Shan Qu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, P. R. China
| | - Li-Hua Zhang
- Department of Gynaecology, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510310, P. R. China
| | - Yu-Yan Gu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, P. R. China
| | - Yi-Hao Chen
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, P. R. China
| | - Zhi-Yong Huang
- Department of Otolaryngology, The Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, P. R. China
| | - Meng-Hua Liu
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, P. R. China
| | - Wei Zou
- NHC Key Laboratory of Birth Defects Research, Prevention and Treatment Hunan Provincial Maternal and Child Health Care Hospital, Changsha 410008, P. R. China
| | - Jing Jiang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, P. R. China
| | - Jun-Qi Chen
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, P. R. China
| | - Yu-Jue Wang
- Department of Laboratory Animal Administration Center, Southern Medical University, Guangzhou 510515, P. R. China
| | - Feng-Hua Zhou
- The Fifth Affiliated Hospital, Southern Medical University, Guangzhou 510920, P. R. China
| |
Collapse
|
46
|
Zhu R, Lang T, Yan W, Zhu X, Huang X, Yin Q, Li Y. Gut Microbiota: Influence on Carcinogenesis and Modulation Strategies by Drug Delivery Systems to Improve Cancer Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2003542. [PMID: 34026439 PMCID: PMC8132165 DOI: 10.1002/advs.202003542] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 01/05/2021] [Indexed: 05/05/2023]
Abstract
Gut microbiota have close interactions with the host. It can affect cancer progression and the outcomes of cancer therapy, including chemotherapy, immunotherapy, and radiotherapy. Therefore, approaches toward the modulation of gut microbiota will enhance cancer prevention and treatment. Modern drug delivery systems (DDS) are emerging as rational and promising tools for microbiota intervention. These delivery systems have compensated for the obstacles associated with traditional treatments. In this review, the essential roles of gut microbiota in carcinogenesis, cancer progression, and various cancer therapies are first introduced. Next, advances in DDS that are aimed at enhancing the efficacy of cancer therapy by modulating or engineering gut microbiota are highlighted. Finally, the challenges and opportunities associated with the application of DDS targeting gut microbiota for cancer prevention and treatment are briefly discussed.
Collapse
Affiliation(s)
- Runqi Zhu
- State Key Laboratory of Drug Research and Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of Sciences501 Haike RoadShanghai201203China
- School of PharmacyUniversity of Chinese Academy of SciencesBeijing100049China
| | - Tianqun Lang
- State Key Laboratory of Drug Research and Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of Sciences501 Haike RoadShanghai201203China
- School of PharmacyUniversity of Chinese Academy of SciencesBeijing100049China
- Yantai Key Laboratory of Nanomedicine and Advanced PreparationsYantai Institute of Materia MedicaYantai264000China
| | - Wenlu Yan
- State Key Laboratory of Drug Research and Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of Sciences501 Haike RoadShanghai201203China
- School of PharmacyUniversity of Chinese Academy of SciencesBeijing100049China
| | - Xiao Zhu
- State Key Laboratory of Drug Research and Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of Sciences501 Haike RoadShanghai201203China
- School of PharmacyUniversity of Chinese Academy of SciencesBeijing100049China
| | - Xin Huang
- State Key Laboratory of Drug Research and Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of Sciences501 Haike RoadShanghai201203China
- School of PharmacyUniversity of Chinese Academy of SciencesBeijing100049China
| | - Qi Yin
- State Key Laboratory of Drug Research and Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of Sciences501 Haike RoadShanghai201203China
- School of PharmacyUniversity of Chinese Academy of SciencesBeijing100049China
- Yantai Key Laboratory of Nanomedicine and Advanced PreparationsYantai Institute of Materia MedicaYantai264000China
| | - Yaping Li
- State Key Laboratory of Drug Research and Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of Sciences501 Haike RoadShanghai201203China
- School of PharmacyUniversity of Chinese Academy of SciencesBeijing100049China
- Yantai Key Laboratory of Nanomedicine and Advanced PreparationsYantai Institute of Materia MedicaYantai264000China
- School of PharmacyYantai UniversityYantai264005China
| |
Collapse
|
47
|
Zhou H, Li G, Wang Y, Jiang R, Li Y, Wang H, Wang F, Ma H, Cao L. Microbial Metabolite Sodium Butyrate Attenuates Cartilage Degradation by Restoring Impaired Autophagy and Autophagic Flux in Osteoarthritis Development. Front Pharmacol 2021; 12:659597. [PMID: 33897442 PMCID: PMC8062861 DOI: 10.3389/fphar.2021.659597] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 03/01/2021] [Indexed: 12/16/2022] Open
Abstract
Osteoarthritis (OA) is a degenerative joint disease with multiple etiologies that affects individuals worldwide. No effective interventions are currently available to reverse the pathological process of OA. Sodium butyrate (NaB), a component of short-chain fatty acids (SCFAs), has multiple biological activities, including the attenuation of inflammation and anti-tumor activities in various diseases. However, whether the protective effects of NaB in OA are associated with the promotion of autophagy had not been investigated. Here, we explored the chondroprotective properties of NaB in an interleukin (IL)-1β-induced inflammatory chondrocyte model and an anterior cruciate ligament transection (ACLT) mouse model. Hematoxylin and eosin (HE), Safranin O, and immunohistochemical staining were performed to evaluate the effects of NaB treatment on articular cartilage. An optimal NaB dose for chondrocyte treatment was determined via cell counting kit-8 assays. Immunofluorescence and transmission electron microscopy were used to detect autophagy in chondrocytes. Flow cytometry was utilized to detect reactive oxygen species (ROS), cell cycle activity, and apoptosis in chondrocytes. Western blot and immunostaining were performed to evaluate the protein expression levels of relevant indicators. We found that the administration of NaB by oral gavage could attenuate cartilage degradation. In parallel, NaB treatment could enhance the activation of autophagy, increase autophagic flux, decrease extracellular matrix degradation, and reduce apoptosis by restraining inflammation, ROS production, and cell cycle arrest in IL-1β-treated chondrocytes. The protective effects of NaB could be partially abolished by the autophagy inhibitor 3-methyladenine (3-MA), which indicated that the protective effects of NaB against OA were partially governed by the enhancement of autophagy to restrain the formation of inflammatory mediators and ROS and regulate cell cycle progression and apoptosis in chondrocytes. In conclusion, NaB could attenuate OA progression by restoring impaired autophagy and autophagic flux via the phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR) pathway, both in vitro and in vivo, implying that NaB could represent a novel therapeutic approach for OA.
Collapse
Affiliation(s)
- Haikang Zhou
- Department of Orthopaedics, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Guoqing Li
- Department of Orthopaedics, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Yang Wang
- Department of Orthopaedics, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Rendong Jiang
- Department of Orthopaedics, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Yicheng Li
- Department of Orthopaedics, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Huhu Wang
- Department of Orthopaedics, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Fei Wang
- Xinjiang Uygur Autonomous Region Clinical Research Center for Orthopedic Diseases, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Hairong Ma
- Xinjiang Uygur Autonomous Region Clinical Research Center for Orthopedic Diseases, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Li Cao
- Department of Orthopaedics, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| |
Collapse
|
48
|
Wang Q, Sun Q, Wang J, Qiu X, Qi R, Huang J. Lactobacillus Plantarum 299v Changes miRNA Expression in the Intestines of Piglets and Leads to Downregulation of LITAF by Regulating ssc-miR-450a. Probiotics Antimicrob Proteins 2021; 13:1093-1105. [PMID: 33486700 DOI: 10.1007/s12602-021-09743-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/08/2021] [Indexed: 11/26/2022]
Abstract
Lactiplantibacillus plantarum subsp. plantarum 299v (L. plantarum 299v) is one of the most important probiotic strains in animal health, but the molecular mechanisms of how it exerts health benefits remain unclear. The purpose of this study was to explore the changes in miRNA expression profiles in the intestinal tissues of piglets by L. plantarum 299v and to explore its possible molecular regulatory mechanism in intestinal function. Neonatal piglets were orally administered L. plantarum 299v daily from 1 to 20 days old, and high-throughput sequencing was conducted to analyse the changes in miRNA expression in the jejunum and ileum. The results showed that 370 known porcine miRNAs were identified from eight libraries. Five miRNAs (ssc-miR-21-5p, -143-3p, -194b-5p, -192, and -126-3p) were highly expressed in the intestinal tissues. There were 15 differentially expressed miRNAs between the control group and the L. plantarum group, and only miR-450a was expressed differentially in both intestinal tissues. KEGG analysis revealed that the target genes of the 15 differentially expressed miRNAs were involved in 37 significantly enriched pathways (P < 0.01). Then, quantitative polymerase chain reaction confirmed that the miRNA expression was corresponded well with those from the sequencing. Luciferase reporter assays verified that lipopolysaccharide-induced TNF-α factor is a target of miR-450a. Our results also showed L. plantarum 299v could influence intestinal function by changing the levels of cytokines via miRNA expression. This is the first study to analyse differential expression miRNA profiles in intestinal tissue after L. plantarum 299v treatment and investigate the molecular regulatory mechanism of functional miRNA.
Collapse
Affiliation(s)
- Qi Wang
- Chongqing Academy of Animal Sciences, Rongchang, Chongqing, 402460, China
- Key Laboratory of Pig Industry Sciences, Ministry of Agriculture, Rongchang, Chongqing, 402460, China
| | - Qian Sun
- Chongqing Academy of Animal Sciences, Rongchang, Chongqing, 402460, China
| | - Jing Wang
- Chongqing Academy of Animal Sciences, Rongchang, Chongqing, 402460, China
- Key Laboratory of Pig Industry Sciences, Ministry of Agriculture, Rongchang, Chongqing, 402460, China
| | - Xiaoyu Qiu
- Chongqing Academy of Animal Sciences, Rongchang, Chongqing, 402460, China
| | - Renli Qi
- Chongqing Academy of Animal Sciences, Rongchang, Chongqing, 402460, China
- Key Laboratory of Pig Industry Sciences, Ministry of Agriculture, Rongchang, Chongqing, 402460, China
| | - Jinxiu Huang
- Chongqing Academy of Animal Sciences, Rongchang, Chongqing, 402460, China.
- Key Laboratory of Pig Industry Sciences, Ministry of Agriculture, Rongchang, Chongqing, 402460, China.
| |
Collapse
|
49
|
Choudhry H. The Microbiome and Its Implications in Cancer Immunotherapy. Molecules 2021; 26:E206. [PMID: 33401586 PMCID: PMC7795182 DOI: 10.3390/molecules26010206] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 12/22/2020] [Accepted: 12/30/2020] [Indexed: 02/06/2023] Open
Abstract
Cancer is responsible for ~18 million deaths globally each year, representing a major cause of death. Several types of therapy strategies such as radiotherapy, chemotherapy and more recently immunotherapy, have been implemented in treating various types of cancer. Microbes have recently been found to be both directly and indirectly involved in cancer progression and regulation, and studies have provided novel and clear insights into the microbiome-mediated emergence of cancers. Scientists around the globe are striving hard to identify and characterize these microbes and the underlying mechanisms by which they promote or suppress various kinds of cancer. Microbes may influence immunotherapy by blocking various cell cycle checkpoints and the production of certain metabolites. Hence, there is an urgent need to better understand the role of these microbes in the promotion and suppression of cancer. The identification of microbes may help in the development of future diagnostic tools to cure cancers possibly associated with the microbiome. This review mainly focuses on various microbes and their association with different types of cancer, responses to immunotherapeutic modulation, physiological responses, and prebiotic and postbiotic effects.
Collapse
Affiliation(s)
- Hani Choudhry
- Department of Biochemistry, Faculty of Sciences, Cancer and Mutagenesis Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
50
|
Pujari R, Banerjee G. Impact of prebiotics on immune response: from the bench to the clinic. Immunol Cell Biol 2020; 99:255-273. [PMID: 32996638 DOI: 10.1111/imcb.12409] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 08/31/2020] [Accepted: 09/29/2020] [Indexed: 12/23/2022]
Abstract
Several preclinical and clinical studies have shown the immunomodulatory role exerted by prebiotics in regulating the immune response. In this review, we describe the mechanistic and clinical studies that decipher the cell signaling pathways implicated in the process. Prebiotic fibers are conventionally known to serve as substrate for probiotic commensal bacteria that release of short-chain fatty acids in the intestinal tract along with several other metabolites. Subsequently, they then act on the local as well as the systemic immune cells and the gut-associated epithelial cells, primarily through G-protein-coupled receptor-mediated pathways. However, other pathways including histone deacetylase inhibition and inflammasome pathway have also been implicated in regulating the immunomodulatory effect. The prebiotics can also induce a microbiota-independent effect by directly acting on the gut-associated epithelial and innate immune cells through the Toll-like receptors. The cumulative effect results in the maintenance of the epithelial barrier integrity and modulation of innate immunity through secretion of pro- and anti-inflammatory cytokines, switches in macrophage polarization and function, neutrophil recruitment and migration, dendritic cell and regulatory T-cell differentiation. Extending these in vitro and ex vivo observations, some prebiotics have been well investigated, with successful human and animal trials demonstrating the association between gut microbes and immunity biomarkers leading to improvement in health endpoints across populations. This review discusses scientific insights into the association between prebiotics, innate immunity and gut microbiome from in vitro to human oral intervention.
Collapse
Affiliation(s)
- Radha Pujari
- Innovation Centre, Tata Chemicals Ltd, Pune, Maharashtra, India
| | - Gautam Banerjee
- Innovation Centre, Tata Chemicals Ltd, Pune, Maharashtra, India
| |
Collapse
|