1
|
Zuo H, Jiang W, Gao J, Ma Z, Li C, Peng Y, Jin J, Zhan X, Lv W, Liu X, Hu J, Zhang M, Jia Y, Xu Z, Tang J, Zheng R, Zuo B. SYISL Knockout Promotes Embryonic Muscle Development of Offspring by Modulating Maternal Gut Microbiota and Fetal Myogenic Cell Dynamics. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024:e2410953. [PMID: 39680624 DOI: 10.1002/advs.202410953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 12/02/2024] [Indexed: 12/18/2024]
Abstract
Embryonic muscle fiber formation determines post-birth muscle fiber totals. The previous research shows SYISL knockout significantly increases muscle fiber numbers and mass in mice, but the mechanism remains unclear. This study confirms that the SYISL gene, maternal gut microbiota, and their interaction significantly affect the number of muscle fibers in mouse embryos through distinct mechanisms, as SYISL knockout alters maternal gut microbiota composition and boosts butyrate levels in embryonic serum. Both fecal microbiota transplantation and butyrate feeding significantly increase muscle fiber numbers in offspring, with butyrate inhibiting histone deacetylases and increasing histone acetylation in embryonic muscle. Combined analysis of RNA-seq between wild-type and SYISL knockout mice with ChIP-seq for H3K9ac and H3K27ac reveals that SYISL and maternal microbiota interaction regulates myogenesis via the butyrate-HDAC-H3K9ac/H3K27ac pathway. Furthermore, scRNA-seq analysis shows that SYISL knockout alone significantly increases the number and proportion of myogenic cells and their dynamics, independently of regulating histone acetylation levels. Cell communication analysis suggests that this may be due to the downregulation of signaling pathways such as MSTN and TGFβ. Overall, multiple pathways are highlighted through which SYISL influences embryonic muscle development, offering valuable insights for treating muscle diseases and improving livestock production.
Collapse
Affiliation(s)
- Hao Zuo
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, Huazhong Agricultural University, Wuhan, 430070, China
- Hubei Hongshan Laboratory, Wuhan, 430070, China
| | - Wei Jiang
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, Huazhong Agricultural University, Wuhan, 430070, China
| | - Jianwei Gao
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, Huazhong Agricultural University, Wuhan, 430070, China
| | - Zhibo Ma
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, Huazhong Agricultural University, Wuhan, 430070, China
| | - Chen Li
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, Huazhong Agricultural University, Wuhan, 430070, China
| | - Yaxin Peng
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, Huazhong Agricultural University, Wuhan, 430070, China
| | - Jianjun Jin
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, Huazhong Agricultural University, Wuhan, 430070, China
| | - Xizhen Zhan
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, Huazhong Agricultural University, Wuhan, 430070, China
| | - Wei Lv
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, Huazhong Agricultural University, Wuhan, 430070, China
| | - Xiao Liu
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, Huazhong Agricultural University, Wuhan, 430070, China
| | - Jingjing Hu
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, Huazhong Agricultural University, Wuhan, 430070, China
| | - Mengdi Zhang
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, Huazhong Agricultural University, Wuhan, 430070, China
| | - Yiming Jia
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, Huazhong Agricultural University, Wuhan, 430070, China
| | - Zaiyan Xu
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, 430070, China
- Department of Basic Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Junming Tang
- Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medicine Science, Hubei University of Medicine, Shiyan, 442000, China
| | - Rong Zheng
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, Huazhong Agricultural University, Wuhan, 430070, China
| | - Bo Zuo
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, Huazhong Agricultural University, Wuhan, 430070, China
- Hubei Hongshan Laboratory, Wuhan, 430070, China
| |
Collapse
|
2
|
Zhang ZX, Peng J, Ding WW. Lipocalin-2 and intestinal diseases. World J Gastroenterol 2024; 30:4864-4879. [PMID: 39679305 PMCID: PMC11612708 DOI: 10.3748/wjg.v30.i46.4864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 09/25/2024] [Accepted: 11/04/2024] [Indexed: 11/21/2024] Open
Abstract
Dysfunction of the intestinal barrier is a prevalent phenomenon observed across a spectrum of diseases, encompassing conditions such as mesenteric artery dissection, inflammatory bowel disease, cirrhosis, and sepsis. In these pathological states, the integrity of the intestinal barrier, which normally serves to regulate the selective passage of substances between the gut lumen and the bloodstream, becomes compromised. This compromised barrier function can lead to a range of adverse consequences, including increased permeability to harmful substances, the translocation of bacteria and their products into systemic circulation, and heightened inflammatory responses within the gut and beyond. Understanding the mechanisms underlying intestinal barrier dysfunction in these diverse disease contexts is crucial for the development of targeted therapeutic interventions aimed at restoring barrier integrity and ameliorating disease progression. Lipocalin-2 (LCN2) expression is significantly upregulated during episodes of intestinal inflammation, making it a pivotal indicator for gauging the extent of such inflammatory processes. Notably, however, LCN2 derived from distinct cellular sources, whether intestinal epithelial cells or immune cells, exhibits notably divergent functional characteristics. Furthermore, the multifaceted nature of LCN2 is underscored by its varying roles across different diseases, sometimes even demonstrating contradictory effects.
Collapse
Affiliation(s)
- Zhong-Xu Zhang
- Department of Trauma and Acute Care Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, Jiangsu Province, China
| | - Jian Peng
- Department of Trauma and Acute Care Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, Jiangsu Province, China
| | - Wei-Wei Ding
- Department of Trauma and Acute Care Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, Jiangsu Province, China
| |
Collapse
|
3
|
Liu JQ, Zhou HB, Bai WF, Wang J, Li Q, Fan LY, Chang H, Shi SL. Assessment of progression of pulmonary fibrosis based on metabonomics and analysis of intestinal microbiota. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2024; 52:201-217. [PMID: 38488151 DOI: 10.1080/21691401.2024.2326616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 02/27/2024] [Indexed: 03/19/2024]
Abstract
The main purpose of this study was to explore the changes of biomarkers in different developmental stages of bleomycin-induced pulmonary fibrosis (PF) in rats via comprehensive pathophysiology, UPLC-QTOF/MS metabonomic technology, and 16S rRNA gene sequencing of intestinal microbiota. The rats were randomly divided into normal control and 1-, 2- and 4-week model group. The rat model of PF was established by one-time intratracheal instillation of bleomycin. The levels of inflammatory and fibrosis-related factors such as hydroxyproline (HYP), type III procollagen (COL-III), type IV collagen (COL-IV), hyaluronidase (HA), laminin (LN), interleukin (IL)-1β, IL-6, malondialdehyde (MDA) increased and superoxide dismutase (SOD) decreased as the PF cycle progressed. In the 1-, 2- and 4-week model group, 2, 19 and 18 potential metabolic biomarkers and 3, 16 and 12 potential microbial biomarkers were detected, respectively, which were significantly correlated. Glycerophospholipid metabolism pathway was observed to be an important pathway affecting PF at 1, 2 and 4 weeks; arginine and proline metabolism pathways significantly affected PF at 2 weeks. Linoleic acid metabolism pathway exhibited clear metabolic abnormalities at 2 and 4 weeks of PF, and alpha-linolenic acid metabolism pathway significantly affected PF at 4 weeks.
Collapse
Affiliation(s)
- Jia-Qi Liu
- Department of Pharmacy, Baotou Medical College, Baotou, PR China
| | - Hong-Bing Zhou
- Department of Pharmacy, Baotou Medical College, Baotou, PR China
- Institute of Bioactive Substance and Function of Mongolian Medicine and Chinese Materia Medica, Baotou Medical College, Baotou, PR China
| | - Wan-Fu Bai
- Department of Pharmacy, Baotou Medical College, Baotou, PR China
| | - Jia Wang
- Department of Pharmacy, Baotou Medical College, Baotou, PR China
| | - Qian Li
- Department of Pharmacy, Baotou Medical College, Baotou, PR China
| | - Li-Ya Fan
- Department of Pharmacy, Baotou Medical College, Baotou, PR China
| | - Hong Chang
- Department of Pharmacy, Baotou Medical College, Baotou, PR China
| | - Song-Li Shi
- Department of Pharmacy, Baotou Medical College, Baotou, PR China
- Institute of Bioactive Substance and Function of Mongolian Medicine and Chinese Materia Medica, Baotou Medical College, Baotou, PR China
| |
Collapse
|
4
|
Zhao L, Zheng J, Gu Y, Xu X, Yu J, Li J, Yang S, Chen B, Du J, Dong R. Quercetin intervention mitigates small intestinal damage and immunologic derangement induced by polystyrene nanoplastics: Insights from multi-omics analysis in mice. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 361:124862. [PMID: 39216663 DOI: 10.1016/j.envpol.2024.124862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 08/27/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024]
Abstract
Nanoplastics (NPs), which belong to emerging environmental pollutants, threaten environmental sustainability and human health. Despite recent studies have reported that NPs damage the gastrointestinal tract and immune homeostasis, the underlying mechanisms remain unclear. Polyphenols have been found to promote NPs excretion by interacting with intestinal flora (IF). However, the potential mechanisms and action targets of this are still poorly understood. To address these knowledge gaps, we investigated the impact of quercetin and three concentrations of polystyrene nanoplastics (PS-NPs) in mice using an integrated phenotypic and multi-omics analysis. Our findings demonstrated that PS-NPs accumulate within the intestine, resulting in impairments to intestinal tissue and barrier function, as well as disturbing the expression of immune-response small intestinal genes and composition of IF. Exposure to PS-NPs significantly elevate the level of intestinal IgG and CD20+ B cells, while inhibiting T cells activation. Furthermore, PS-NPs could induce systemic immune and serum insulin level disorders. Quercetin might mitigate PS-NPs-induced intestinal damage and immune disorders though reversing IF disorders, gene expression changes, and their interaction.
Collapse
Affiliation(s)
- Long Zhao
- Key Lab of Public Health Safety of the Ministry of Education, Institute of Nutrition, School of Public Health, Fudan University, Shanghai 200032, China
| | | | - Yiying Gu
- Key Lab of Public Health Safety of the Ministry of Education, Institute of Nutrition, School of Public Health, Fudan University, Shanghai 200032, China
| | - Xin Xu
- Key Lab of Public Health Safety of the Ministry of Education, Institute of Nutrition, School of Public Health, Fudan University, Shanghai 200032, China
| | - Jianguo Yu
- Zhongshan Community Health Care Center, Songjiang District, Shanghai 201613, China
| | - Jing Li
- Zhongshan Community Health Care Center, Songjiang District, Shanghai 201613, China
| | - Shuyu Yang
- Nutrilite Health Institute, Shanghai 200023, China
| | - Bo Chen
- Key Lab of Public Health Safety of the Ministry of Education, Institute of Nutrition, School of Public Health, Fudan University, Shanghai 200032, China
| | - Jun Du
- Nutrilite Health Institute, Shanghai 200023, China.
| | - Ruihua Dong
- Key Lab of Public Health Safety of the Ministry of Education, Institute of Nutrition, School of Public Health, Fudan University, Shanghai 200032, China.
| |
Collapse
|
5
|
Zhang L, Cai M, Zhang X, Wang S, Pang L, Chen X, Zheng C, Sun Y, Liang Y, Guo S, Wei F, Zhang Y. Integrated analysis of microbiome and host transcriptome unveils correlations between lung microbiota and host immunity in bronchoalveolar lavage fluid of pneumocystis pneumonia patients. Microbes Infect 2024; 26:105374. [PMID: 38849069 DOI: 10.1016/j.micinf.2024.105374] [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: 03/03/2024] [Revised: 05/29/2024] [Accepted: 06/02/2024] [Indexed: 06/09/2024]
Abstract
OBJECTIVE The lung microbiota of patients with pulmonary diseases is disrupted and impacts the immunity. The microbiological and immune landscape of the lungs in patients with pneumocystis pneumonia (PCP) remains poorly understood. METHODS Multi-omics analysis and machine learning were performed on bronchoalveolar lavage fluid to explore interaction between the lung microbiota and host immunity in PCP. Then we constructed a diagnostic model using differential genes with LASSO regression and validated by qPCR. The immune infiltration analysis was performed to explore the landscape of lung immunity in patients with PCP. RESULTS Patients with PCP showed a low alpha diversity of lung microbiota, accompanied by the elevated abundance of Firmicutes, and the differential expressed genes (DEGs) analysis displayed a downregulation of MAPK signaling. The MAPK10, TGFB1, and EFNA3 indicated a potential to predict PCP (AUC = 0.86). The lung immune landscape in PCP showed the lower levels of naïve CD4+ T cells and activated dendritic cells. The correlation analysis of the MAPK signaling pathway-related DEGs and the differential microorganisms at the level of phylum showed that the Firmicutes was negatively correlated with these DEGs. CONCLUSION We profiled the characteristics of lung microbiota and immune landscape in PCP, which may contribute to elucidating the mechanism of PCP.
Collapse
Affiliation(s)
- Ling Zhang
- Department of Respiratory and Critical Care Medicine, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China; Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
| | - Miaotian Cai
- Department of Respiratory and Critical Care Medicine, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
| | - Xin Zhang
- Department of Respiratory and Critical Care Medicine, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China; Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
| | - Sitong Wang
- Department of Respiratory and Critical Care Medicine, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
| | - Lijun Pang
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
| | - Xue Chen
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China; Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing Key Laboratory for HIV/AIDS Research, Beijing, 100069, China
| | - Caopei Zheng
- Department of Respiratory and Critical Care Medicine, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China; Laboratory for Clinical Medicine, Capital Medical University, China
| | - Yuqing Sun
- Department of Respiratory and Critical Care Medicine, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China; Laboratory for Clinical Medicine, Capital Medical University, China
| | - Ying Liang
- Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing Key Laboratory for HIV/AIDS Research, Beijing, 100069, China; Laboratory for Clinical Medicine, Capital Medical University, China
| | - Shan Guo
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
| | - Feili Wei
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China.
| | - Yulin Zhang
- Department of Respiratory and Critical Care Medicine, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China; Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing Key Laboratory for HIV/AIDS Research, Beijing, 100069, China; Laboratory for Clinical Medicine, Capital Medical University, China; Beijing Research Center for Respiratory Infectious Diseases, China.
| |
Collapse
|
6
|
Huang Q, Wen C, Gu S, Jie Y, Li G, Yan Y, Tian C, Wu G, Yang N. Synergy of gut microbiota and host genome in driving heterosis expression of chickens. J Genet Genomics 2024; 51:1121-1134. [PMID: 38950856 DOI: 10.1016/j.jgg.2024.06.011] [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: 04/09/2024] [Revised: 06/18/2024] [Accepted: 06/19/2024] [Indexed: 07/03/2024]
Abstract
Heterosis has been widely utilized in agricultural production. Despite over a century of extensive research, the underlying mechanisms of heterosis remain elusive. Most hypotheses and research have focused on the genetic basis of heterosis. However, the potential role of gut microbiota in heterosis has been largely ignored. Here, we carefully design a crossbreeding experiment with two distinct broiler breeds and conduct 16S rRNA amplicon and transcriptome sequencing to investigate the synergistic role of gut microbiota and host genes in driving heterosis. We find that the breast muscle weight of hybrids exhibits a high heterosis, 6.28% higher than the mid-parent value. A notable difference is observed in the composition and potential function of cecal microbiota between hybrids and their parents. Over 90% of differentially colonized microbiota and differentially expressed genes exhibit nonadditive patterns. Integrative analyses uncover associations between nonadditive genes and nonadditive microbiota, including a connection between the expression of cellular signaling pathways and metabolism-related genes and the abundance of Odoribacter, Oscillibacter, and Alistipes in hybrids. Moreover, higher abundances of these microbiota are related to better meat yield. In summary, these findings highlight the importance of gut microbiota in heterosis, serving as crucial factors that modulate heterosis expression in chickens.
Collapse
Affiliation(s)
- Qiang Huang
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center for Molecular Design Breeding, China Agricultural University, Beijing 100193, China; National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Chaoliang Wen
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center for Molecular Design Breeding, China Agricultural University, Beijing 100193, China; National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; Sanya Institute of China Agricultural University, Hainan 572025, China.
| | - Shuang Gu
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center for Molecular Design Breeding, China Agricultural University, Beijing 100193, China; National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Yuchen Jie
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center for Molecular Design Breeding, China Agricultural University, Beijing 100193, China; National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Guangqi Li
- Beijing Huadu Yukou Poultry Industry Co. Ltd., Beijing 101206, China
| | - Yiyuan Yan
- Beijing Huadu Yukou Poultry Industry Co. Ltd., Beijing 101206, China
| | - Chuanyao Tian
- Beijing Huadu Yukou Poultry Industry Co. Ltd., Beijing 101206, China
| | - Guiqin Wu
- Beijing Huadu Yukou Poultry Industry Co. Ltd., Beijing 101206, China
| | - Ning Yang
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center for Molecular Design Breeding, China Agricultural University, Beijing 100193, China; National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; Sanya Institute of China Agricultural University, Hainan 572025, China.
| |
Collapse
|
7
|
Zhao Y, Chen Z, Dong R, Liu Y, Zhang Y, Guo Y, Yu M, Li X, Wang J. Multiomics analysis reveals the potential mechanism of high-fat diet in dextran sulfate sodium-induced colitis mice model. Food Sci Nutr 2024; 12:8309-8323. [PMID: 39479684 PMCID: PMC11521715 DOI: 10.1002/fsn3.4426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 08/05/2024] [Accepted: 08/09/2024] [Indexed: 11/02/2024] Open
Abstract
A high-fat diet (HFD) is recognized as an important contributor to inflammatory bowel disease (IBD). However, the precise underlying mechanism of HFD on IBD remains elusive. This study aimed to investigate the potential mechanism by which HFD affects IBD using 16S rRNA-sequencing and RNA-seq technology. Results indicated that HFD-treated mice exhibited notable alternations in the structure and composition of the gut microbiota, with some of these alternations being associated with the pathogenesis of IBD. Analysis of the colon transcriptome revealed 11 hub genes and 7 hub pathways among control, DSS-induced colitis, and HFD + DSS-treated groups. Further analysis explores the relationship between the hub pathways and genes, as well as the hub genes and gut microbiota. Overall, the findings indicate that the impact of HFD on DSS-induced colitis may be linked to intestinal dysbiosis and specific genes such as Abca8b, Ace2, Apoa1, Apoa4, Apoc3, Aspa, Dpp4, Maob, Slc34a2, Slc7a9, and Trpm6. These results provide valuable insights for determining potential therapeutic targets for addressing HFD-induced IBD.
Collapse
Affiliation(s)
- Yuyang Zhao
- Department of GastroenterologyChina‐Japan Union Hospital of Jilin UniversityChangchunJilinChina
| | - Zhimin Chen
- Department of PharmacologyCollege of Basic Medical Sciences, Jilin UniversityChangchunJilinChina
| | - Ruiyi Dong
- College of Physical Education, Hunan Normal UniversityChangshaChina
| | - Yufan Liu
- Department of PharmacologyCollege of Basic Medical Sciences, Jilin UniversityChangchunJilinChina
| | - Yixin Zhang
- Department of PharmacologyCollege of Basic Medical Sciences, Jilin UniversityChangchunJilinChina
| | - Yan Guo
- Department of PharmacologyCollege of Basic Medical Sciences, Jilin UniversityChangchunJilinChina
| | - Meiyi Yu
- Department of PharmacologyCollege of Basic Medical Sciences, Jilin UniversityChangchunJilinChina
| | - Xiang Li
- Department of PharmacologyCollege of Basic Medical Sciences, Jilin UniversityChangchunJilinChina
| | - Jiangbin Wang
- Department of GastroenterologyChina‐Japan Union Hospital of Jilin UniversityChangchunJilinChina
| |
Collapse
|
8
|
Zhang Y, Shen W, Chen Z, He J, Feng L, Wang L, Chen S. Resistant starch reduces glycolysis by HK2 and suppresses high-fructose corn syrup-induced colon tumorigenesis. J Gastroenterol 2024; 59:905-920. [PMID: 39141107 PMCID: PMC11415400 DOI: 10.1007/s00535-024-02138-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 07/25/2024] [Indexed: 08/15/2024]
Abstract
BACKGROUND The intake of high-fructose corn syrup (HFCS) may increase the risk of colorectal cancer (CRC). This study aimed to explore the potential effects and mechanisms of resistant starch (RS) in HFCS-induced colon tumorigenesis. METHODS The azoxymethane/dextran sodium sulfate (AOM/DSS) and ApcMin/+ mice models were used to investigate the roles of HFCS and RS in CRC in vivo. An immunohistochemistry (IHC) staining analysis was used to detect the expression of proliferation-related proteins in tissues. 16S rRNA sequencing for microbial community, gas chromatography for short-chain fatty acids (SCFAs), and mass spectrometry analysis for glycolysis products in the intestines were performed. Furthermore, lactic acid assay kit was used to detect the glycolysis levels in vitro. RESULTS RS suppressed HFCS-induced colon tumorigenesis through reshaping the microbial community. Mechanistically, the alteration of the microbial community after RS supplement increased the levels of intestinal SCFAs, especially butyrate, leading to the suppression of glycolysis and CRC cell proliferation by downregulating HK2. CONCLUSIONS Our study identified RS as a candidate of protective factors in CRC and may provide a potential target for HFCS-related CRC treatment.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Weiyi Shen
- Department of Pathology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhehang Chen
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiamin He
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lijun Feng
- Department of Nutriology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lan Wang
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Shujie Chen
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
9
|
Marsh R, Santos CD, Yule A, Dellschaft NS, Hoad CL, Ng C, Major G, Smyth AR, Rivett D, van der Gast C. Impact of extended Elexacaftor/Tezacaftor/Ivacaftor therapy on the gut microbiome in cystic fibrosis. J Cyst Fibros 2024; 23:967-976. [PMID: 38749891 DOI: 10.1016/j.jcf.2024.05.002] [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: 01/22/2024] [Revised: 04/29/2024] [Accepted: 05/04/2024] [Indexed: 09/20/2024]
Abstract
BACKGROUND There is a paucity of knowledge on the longer-term effects of CF transmembrane conductance regulator (CFTR) modulator therapies upon the gut microbiome and associated outcomes. In a pilot study, we investigated longitudinal Elexacaftor/Tezacaftor/Ivacaftor (ETI) therapy on the gut microbiota, metabolomic functioning, and clinical outcomes in people with CF (pwCF). STUDY DESIGN Faecal samples from 20 pwCF were acquired before and then following 3, 6, and 17+ months of ETI therapy. Samples were subjected to microbiota sequencing and targeted metabolomics to profile and quantify short-chain fatty acid composition. Ten healthy matched controls were included for comparison. Clinical data, including markers of intestinal function were integrated to investigate relationships. RESULTS Extended ETI therapy increased core microbiota diversity and composition, which translated to gradual shifts in whole microbiota composition towards that observed in healthy controls. Despite becoming more similar over time, CF microbiota and functional metabolite compositions remained significantly different to healthy controls. Antibiotic treatment for pulmonary infection significantly explained a relatively large degree of variation within the whole microbiota and rarer satellite taxa. Clinical outcomes were not significantly different following ETI. CONCLUSIONS Whilst differences persisted, a positive trajectory towards the microbiota observed in healthy controls was found. We posit that progression was predominately impeded by pulmonary antibiotics administration. We recommend future studies use integrated omics approaches within a combination of long-term longitudinal patient studies and model experimental systems. This will deepen our understanding of the impacts of CFTR modulator therapy and respiratory antibiotic interventions upon the gut microbiome and gastrointestinal pathophysiology in CF.
Collapse
Affiliation(s)
- Ryan Marsh
- Department of Applied Sciences, Northumbria University, Newcastle, UK
| | | | - Alexander Yule
- School of Medicine, University of Nottingham, UK; NIHR Nottingham Biomedical Research Centre, UK
| | | | - Caroline L Hoad
- Sir Peter Mansfield Imaging Centre, University of Nottingham, UK
| | - Christabella Ng
- School of Medicine, University of Nottingham, UK; NIHR Nottingham Biomedical Research Centre, UK
| | - Giles Major
- School of Medicine, University of Nottingham, UK; Nestlé Institute of Health Sciences, Société des Produits Nestlé, Lausanne, Switzerland
| | - Alan R Smyth
- School of Medicine, University of Nottingham, UK; NIHR Nottingham Biomedical Research Centre, UK; School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, UK
| | - Damian Rivett
- Department of Natural Sciences, Manchester Metropolitan University, UK
| | - Christopher van der Gast
- Department of Applied Sciences, Northumbria University, Newcastle, UK; Department of Respiratory Medicine, Northern Care Alliance NHS Foundation Trust, Salford, UK.
| |
Collapse
|
10
|
Green N, Chan C, Ooi CY. The gastrointestinal microbiome, small bowel bacterial overgrowth, and microbiome modulators in cystic fibrosis. Pediatr Pulmonol 2024; 59 Suppl 1:S70-S80. [PMID: 39105345 DOI: 10.1002/ppul.26913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 01/12/2024] [Accepted: 01/31/2024] [Indexed: 08/07/2024]
Abstract
People with cystic fibrosis (pwCF) have an altered gastrointestinal microbiome. These individuals also demonstrate propensity toward developing small intestinal bacterial overgrowth (SIBO). The dysbiosis present has intestinal and extraintestinal implications, including potential links with the higher rates of gastrointestinal malignancies described in CF. Given these implications, there is growing interest in therapeutic options for microbiome modulation. Alternative therapies, including probiotics and prebiotics, and current CF transmembrane conductance regulator gene modulators are promising interventions for ameliorating gut microbiome dysfunction in pwCF. This article will characterize and discuss the current state of knowledge and expert opinions on gut dysbiosis and SIBO in the context of CF, before reviewing the current evidence supporting gut microbial modulating therapies in CF.
Collapse
Affiliation(s)
- Nicole Green
- Department of Pediatrics, Division of Gastroenterology and Hepatology, Seattle Children's Hospital, University of Washington, Seattle, Washington, USA
| | - Christopher Chan
- Discipline of Paediatrics & Child Health, Randwick Clinical Campus, School of Clinical Medicine, UNSW Medicine & Health, University of New South Wales, Sydney, New South Wales, Australia
| | - Chee Y Ooi
- Discipline of Paediatrics & Child Health, Randwick Clinical Campus, School of Clinical Medicine, UNSW Medicine & Health, University of New South Wales, Sydney, New South Wales, Australia
- Department of Gastroenterology, Sydney Children's Hospital, Randwick, New South Wales, Australia
| |
Collapse
|
11
|
Song X, Fu X, Niu S, Wang P, Qi J, Shi S, Chang H, Bai W. Exploring the effects of Saorilao-4 on the gut microbiota of pulmonary fibrosis model rats based on 16S rRNA sequencing. J Appl Microbiol 2024; 135:lxae178. [PMID: 39020259 DOI: 10.1093/jambio/lxae178] [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: 03/06/2024] [Revised: 06/16/2024] [Accepted: 07/17/2024] [Indexed: 07/19/2024]
Abstract
AIMS Pulmonary fibrosis (PF) is a progressive and incurable lung disease for which treatment options are limited. Here, we aimed to conduct an exploratory study on the effects of the Mongolian medicine Saorilao-4 (SRL) on the gut microbiota structure, species abundance, and diversity of a rat PF model as well as the mechanisms underlying such effects. METHODS AND RESULTS Rat fecal samples were analyzed using 16S rRNA sequencing technology. Bioinformatic and correlation analyses were performed on microbiota data to determine significant associations. SRL substantially attenuated the adverse effects exerted by PF on the structure and diversity of gut microbiota while regulating its alpha and beta diversities. Linear discriminant analysis effect size enabled the identification of 62 differentially abundant microbial taxa. Gut microbiota abundance analysis revealed that SRL significantly increased the relative abundance of bacterial phyla such as Firmicutes and Bacteroidetes. Moreover, SRL increased the proportion of beneficial bacteria, such as Lactobacillus and Bifidobacteriales, decreased the proportion of pathogenic bacteria, such as Rikenellaceae, and balanced the gut microbiota by regulating metabolic pathways. CONCLUSIONS SRL may attenuate PF by regulating gut microbiota. This exploratory study establishes the groundwork for investigating the metagenomics of PF.
Collapse
Affiliation(s)
- Xinni Song
- School of Pharmacy, Baotou Medical College, Baotou 014040, China
| | - Xinyue Fu
- School of Pharmacy, Baotou Medical College, Baotou 014040, China
| | - Shufang Niu
- School of Pharmacy, Baotou Medical College, Baotou 014040, China
| | - Peng Wang
- The Second Affiliated Hospital of Baotou Medical College, Baotou 014030, China
| | - Jun Qi
- The First Affiliated Hospital of Baotou Medical College, Baotou 014010, China
| | - Songli Shi
- School of Pharmacy, Baotou Medical College, Baotou 014040, China
| | - Hong Chang
- School of Pharmacy, Baotou Medical College, Baotou 014040, China
| | - Wanfu Bai
- School of Pharmacy, Baotou Medical College, Baotou 014040, China
| |
Collapse
|
12
|
Viteri-Echeverría J, Andrés A, Calvo-Lerma J, Heredia A, García-Hernández J, Asensio-Grau A. In vitro screening of the impact of dietary prebiotic components, probiotic strains, and their symbiotic combinations on colonic microbiota in children with cystic fibrosis. Food Funct 2024; 15:6512-6522. [PMID: 38804915 DOI: 10.1039/d4fo00325j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Children with Cystic Fibrosis (CF) are more likely to have intestinal dysbiosis due to recurrent antibiotic therapy and the conventional hypercaloric diet administered to them. This study aimed at evaluating the effect of isolated prebiotic components and probiotic strains, and their combinations as potential synbiotics, on the intestinal microbiota of CF children. A static in vitro colonic fermentation model was used by colonizing vials with faecal inoculum, a culture medium, and the substrates to be tested. Post treatment, aliquots were taken to determine ammonium, lactate, and short-chain fatty acids production and to profile the microbiota composition by 16s rRNA sequencing. At genus level, Escherichia-Shigella decreased (15.8%) with the treatment pectin + L. rhamnosus, followed by the beta-glucan + L. salivarius (15.5%). Inversely, the most increase in Bacteroides (44%) was obtained by the treatment with Pectin + L. reuteri. Lactate and acetic acid production was significantly increased with prebiotics and their combinations with L. rhamnosus and L. salivarius. In conclusion, the use of beta-glucan and pectin in combination with probiotic strains from the Lactobacillaceae family suggest potential to modulate dysbiosis and metabolic activity on CF colonic microbiota, encouraging further studies in animal studies or clinical settings to confirm the findings in vivo.
Collapse
Affiliation(s)
- Jazmín Viteri-Echeverría
- University Institute of Food Engineering (FoodUPV), Polytechnic University of Valencia, Camino de Vera s/n, 46022, València, Spain.
| | - Ana Andrés
- University Institute of Food Engineering (FoodUPV), Polytechnic University of Valencia, Camino de Vera s/n, 46022, València, Spain.
| | - Joaquim Calvo-Lerma
- Research Group in Innovative Technologies for Sustainable Food (ALISOST). University of Valencia, Avda. Vicent Andrés Estellés s/n, Burjassot, 46100, València, Spain.
| | - Ana Heredia
- University Institute of Food Engineering (FoodUPV), Polytechnic University of Valencia, Camino de Vera s/n, 46022, València, Spain.
| | - Jorge García-Hernández
- Advanced Food Microbiology Centre (CAMA), Polytechnic University of Valencia, Camino de Vera s/n, 46022 València, Spain.
| | - Andrea Asensio-Grau
- University Institute of Food Engineering (FoodUPV), Polytechnic University of Valencia, Camino de Vera s/n, 46022, València, Spain.
| |
Collapse
|
13
|
Tsugawa H, Ishihara T, Ogasa K, Iwanami S, Hori A, Takahashi M, Yamada Y, Satoh-Takayama N, Ohno H, Minoda A, Arita M. A lipidome landscape of aging in mice. NATURE AGING 2024; 4:709-726. [PMID: 38609525 DOI: 10.1038/s43587-024-00610-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 03/07/2024] [Indexed: 04/14/2024]
Abstract
Understanding the molecular mechanisms of aging is crucial for enhancing healthy longevity. We conducted untargeted lipidomics across 13 biological samples from mice at various life stages (2, 12, 19 and 24 months) to explore the potential link between aging and lipid metabolism, considering sex (male or female) and microbiome (specific pathogen-free or germ-free) dependencies. By analyzing 2,704 molecules from 109 lipid subclasses, we characterized common and tissue-specific lipidome alterations associated with aging. For example, the levels of bis(monoacylglycero)phosphate containing polyunsaturated fatty acids increased in various organs during aging, whereas the levels of other phospholipids containing saturated and monounsaturated fatty acids decreased. In addition, we discovered age-dependent sulfonolipid accumulation, absent in germ-free mice, correlating with Alistipes abundance determined by 16S ribosomal RNA gene amplicon sequencing. In the male kidney, glycolipids such as galactosylceramides, galabiosylceramides (Gal2Cer), trihexosylceramides (Hex3Cer), and mono- and digalactosyldiacylglycerols were detected, with two lipid classes-Gal2Cer and Hex3Cer-being significantly enriched in aged mice. Integrated analysis of the kidney transcriptome revealed uridine diphosphate galactosyltransferase 8A (UGT8a), alkylglycerone phosphate synthase and fatty acyl-coenzyme A reductase 1 as potential enzymes responsible for the male-specific glycolipid biosynthesis in vivo, which would be relevant to sex dependency in kidney diseases. Inhibiting UGT8 reduced the levels of these glycolipids and the expression of inflammatory cytokines in the kidney. Our study provides a valuable resource for clarifying potential links between lipid metabolism and aging.
Collapse
Affiliation(s)
- Hiroshi Tsugawa
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, Tokyo, Japan.
- Metabolome Informatics Research Team, RIKEN Center for Sustainable Resource Science, Yokohama, Japan.
- Molecular and Cellular Epigenetics Laboratory, Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan.
| | - Tomoaki Ishihara
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Department of Pharmacy, Nagasaki International University, Sasebo, Japan
| | - Kota Ogasa
- Division of Physiological Chemistry and Metabolism, Graduate School of Pharmaceutical Sciences, Keio University, Tokyo, Japan
| | - Seigo Iwanami
- Division of Physiological Chemistry and Metabolism, Graduate School of Pharmaceutical Sciences, Keio University, Tokyo, Japan
| | - Aya Hori
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Mikiko Takahashi
- Metabolome Informatics Research Team, RIKEN Center for Sustainable Resource Science, Yokohama, Japan
| | - Yutaka Yamada
- Metabolome Informatics Research Team, RIKEN Center for Sustainable Resource Science, Yokohama, Japan
| | - Naoko Satoh-Takayama
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Hiroshi Ohno
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Aki Minoda
- Laboratory for Cellular Epigenomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud University, Nijmegen, the Netherlands
| | - Makoto Arita
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.
- Molecular and Cellular Epigenetics Laboratory, Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan.
- Division of Physiological Chemistry and Metabolism, Graduate School of Pharmaceutical Sciences, Keio University, Tokyo, Japan.
- Human Biology-Microbiome-Quantum Research Center (WPI-Bio2Q), Keio University, Tokyo, Japan.
| |
Collapse
|
14
|
Cai L, Zhu H, Mou Q, Wong PY, Lan L, Ng CWK, Lei P, Cheung MK, Wang D, Wong EWY, Lau EHL, Yeung ZWC, Lai R, Meehan K, Fung S, Chan KCA, Lui VWY, Cheng ASL, Yu J, Chan PKS, Chan JYK, Chen Z. Integrative analysis reveals associations between oral microbiota dysbiosis and host genetic and epigenetic aberrations in oral cavity squamous cell carcinoma. NPJ Biofilms Microbiomes 2024; 10:39. [PMID: 38589501 PMCID: PMC11001959 DOI: 10.1038/s41522-024-00511-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: 04/09/2023] [Accepted: 03/27/2024] [Indexed: 04/10/2024] Open
Abstract
Dysbiosis of the human oral microbiota has been reported to be associated with oral cavity squamous cell carcinoma (OSCC) while the host-microbiota interactions with respect to the potential impact of pathogenic bacteria on host genomic and epigenomic abnormalities remain poorly studied. In this study, the mucosal bacterial community, host genome-wide transcriptome and DNA CpG methylation were simultaneously profiled in tumors and their adjacent normal tissues of OSCC patients. Significant enrichment in the relative abundance of seven bacteria species (Fusobacterium nucleatum, Treponema medium, Peptostreptococcus stomatis, Gemella morbillorum, Catonella morbi, Peptoanaerobacter yurli and Peptococcus simiae) were observed in OSCC tumor microenvironment. These tumor-enriched bacteria formed 254 positive correlations with 206 up-regulated host genes, mainly involving signaling pathways related to cell adhesion, migration and proliferation. Integrative analysis of bacteria-transcriptome and bacteria-methylation correlations identified at least 20 dysregulated host genes with inverted CpG methylation in their promoter regions associated with enrichment of bacterial pathogens, implying a potential of pathogenic bacteria to regulate gene expression, in part, through epigenetic alterations. An in vitro model further confirmed that Fusobacterium nucleatum might contribute to cellular invasion via crosstalk with E-cadherin/β-catenin signaling, TNFα/NF-κB pathway and extracellular matrix remodeling by up-regulating SNAI2 gene, a key transcription factor of epithelial-mesenchymal transition (EMT). Our work using multi-omics approaches explored complex host-microbiota interactions and provided important insights into genetic and functional basis in OSCC tumorigenesis, which may serve as a precursor for hypothesis-driven study to better understand the causational relationship of pathogenic bacteria in this deadly cancer.
Collapse
Affiliation(s)
- Liuyang Cai
- Department of Microbiology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Hengyan Zhu
- Department of Microbiology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Qianqian Mou
- Department of Microbiology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Po Yee Wong
- Department of Microbiology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Linlin Lan
- Department of Otorhinolaryngology, Head and Neck Surgery, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Cherrie W K Ng
- Department of Otorhinolaryngology, Head and Neck Surgery, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Pu Lei
- Department of Microbiology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Man Kit Cheung
- Department of Microbiology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Daijuanru Wang
- Department of Microbiology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Eddy W Y Wong
- Department of Otorhinolaryngology, Head and Neck Surgery, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Eric H L Lau
- Department of Otorhinolaryngology, Head and Neck Surgery, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Zenon W C Yeung
- Department of Otorhinolaryngology, Head and Neck Surgery, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Ronald Lai
- Department of Otorhinolaryngology, Head and Neck Surgery, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Katie Meehan
- Department of Otorhinolaryngology, Head and Neck Surgery, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Sherwood Fung
- Department of Chemical Pathology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Kwan Chee A Chan
- Department of Chemical Pathology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Vivian W Y Lui
- Georgia Cancer Center, Augusta, GA, 30912, USA
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Alfred S L Cheng
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jun Yu
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Paul K S Chan
- Department of Microbiology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jason Y K Chan
- Department of Otorhinolaryngology, Head and Neck Surgery, The Chinese University of Hong Kong, Hong Kong SAR, China.
| | - Zigui Chen
- Department of Microbiology, The Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
15
|
Guo J, Yang L. Regulation effect of the intestinal flora and intervention strategies targeting the intestinal flora in alleviation of pulmonary fibrosis development. BIOSCIENCE OF MICROBIOTA, FOOD AND HEALTH 2024; 43:293-299. [PMID: 39364128 PMCID: PMC11444866 DOI: 10.12938/bmfh.2023-100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 02/25/2024] [Indexed: 10/05/2024]
Abstract
Pulmonary fibrosis is an end-stage respiratory disease characterized by fibroblast proliferation and accumulation of extracellular matrix and collagen, which is accompanied by inflammatory damage. The disease is mainly based on pulmonary dysfunction and respiratory failure, the incidence of it is increasing year by year, and the current treatment methods for it are limited. In recent years, it has been found that gut microbes play a crucial role in the pathogenesis and development of pulmonary fibrosis. The microecological disturbance caused by changes in the composition of the intestinal flora can affect the course of pulmonary fibrosis. The regulatory network or information exchange system for gut-lung crosstalk is called the "gut-lung axis". This review focuses on the frontier research on entero-pulmonary regulation in pulmonary fibrosis and on intervention strategies for changing the gut microbiota to improve pulmonary fibrosis, including fecal microbiota transplantation, traditional Chinese medicine interventions, and supplementation with probiotics. In addition, the present problems in this field are also raised in order to provide strong theoretical and strategic support for the future exploration of regulatory mechanisms and therapeutic drug development. This paper reviews the interaction of the intestinal flora with pulmonary fibrosis, introduces the research progress for improving pulmonary fibrosis through interventions targeted at the intestinal flora, and provides new ideas for the treatment of pulmonary fibrosis.
Collapse
Affiliation(s)
- Jianquan Guo
- Key Laboratory of Coal Environmental Pathogenicity and Prevention (Shanxi Medical University), Ministry of Education, Taiyuan, Shanxi 030001, PR China
- School of Public Health, Shanxi Medical University, Taiyuan, Shanxi 030001, PR China
| | - Liyang Yang
- School of Basic Medical Sciences, Shanxi University of Chinese Medicine, Jinzhong, Shanxi 030619, PR China
| |
Collapse
|
16
|
Caley LR, Wood HM, Bottomley D, Fuentes Balaguer A, Wilkinson L, Dyson J, Young C, White H, Benton S, Brearley M, Quirke P, Peckham DG. The gut microbiota in adults with cystic fibrosis compared to colorectal cancer. J Cyst Fibros 2024; 23:262-268. [PMID: 38104000 DOI: 10.1016/j.jcf.2023.12.004] [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: 06/16/2023] [Revised: 12/06/2023] [Accepted: 12/06/2023] [Indexed: 12/19/2023]
Abstract
BACKGROUND Gut dysbiosis is implicated in colorectal cancer (CRC) pathogenesis. Cystic fibrosis (CF) is associated with both gut dysbiosis and increased CRC risk. We therefore compared the faecal microbiota from individuals with CF to CRC and screening samples. We also assessed changes in CRC-associated taxa before and after triple CF transmembrane conductance regulator (CFTR) modulator therapy. METHODS Bacterial DNA amplification comprising V4 16S rRNA analysis was conducted on 84 baseline and 53 matched follow-up stool samples from adults with CF. These data were compared to an existing cohort of 430 CRC and 491 control gFOBT samples from the NHS Bowel Cancer Screening Programme. Data were also compared to 26 previously identified CRC-associated taxa from a published meta-analysis. RESULTS Faecal CF samples had a lower alpha diversity and clustered distinctly from both CRC and control samples, with no clear clinical variables explaining the variation. Compared to controls, CF samples had an increased relative abundance in 6 of the 20 enriched CRC-associated taxa and depletion of 2 of the 6 taxa which have been reported as reduced in CRC. Commencing triple modulator therapy had subtle influence on the relative abundance of CRC-associated microbiota (n = 23 paired CF samples). CONCLUSIONS CF stool samples were clearly dysbiotic, clustering distinctly from both CRC and control samples. Several bacterial shifts in CF samples resembled those observed in CRC. Studies assessing the impact of dietary or other interventions and the longer-term use of CFTR modulators on reducing this potentially pro-oncogenic milieu are needed.
Collapse
Affiliation(s)
- L R Caley
- Leeds Institute of Medical Research, Clinical Sciences Building, St James's University Hospital, University of Leeds, LS9 7TF, UK
| | - H M Wood
- Pathology & Data Analytics, Leeds Institute of Medical Research, University of Leeds, Leeds, LS9 7TF, UK
| | - D Bottomley
- Pathology & Data Analytics, Leeds Institute of Medical Research, University of Leeds, Leeds, LS9 7TF, UK
| | - A Fuentes Balaguer
- Pathology & Data Analytics, Leeds Institute of Medical Research, University of Leeds, Leeds, LS9 7TF, UK
| | - L Wilkinson
- Pathology & Data Analytics, Leeds Institute of Medical Research, University of Leeds, Leeds, LS9 7TF, UK
| | - J Dyson
- Pathology & Data Analytics, Leeds Institute of Medical Research, University of Leeds, Leeds, LS9 7TF, UK
| | - C Young
- Pathology & Data Analytics, Leeds Institute of Medical Research, University of Leeds, Leeds, LS9 7TF, UK
| | - H White
- Leeds Beckett University, Nutrition, Health & Environment, Leeds, LS1 3HE UK
| | - S Benton
- NHS Bowel Cancer Screening South of England Hub, Royal Surrey NHS Foundation Trust, Guildford, GU2 7YS, UK
| | - M Brearley
- NHS Bowel Cancer Screening South of England Hub, Royal Surrey NHS Foundation Trust, Guildford, GU2 7YS, UK
| | - P Quirke
- Pathology & Data Analytics, Leeds Institute of Medical Research, University of Leeds, Leeds, LS9 7TF, UK
| | - D G Peckham
- Leeds Institute of Medical Research, Clinical Sciences Building, St James's University Hospital, University of Leeds, LS9 7TF, UK; Department of Respiratory Medicine, Leeds Teaching Hospitals NHS Trust, Leeds, LS9 7TF, UK.
| |
Collapse
|
17
|
Dong Y, He L, Zhu Z, Yang F, Ma Q, Zhang Y, Zhang X, Liu X. The mechanism of gut-lung axis in pulmonary fibrosis. Front Cell Infect Microbiol 2024; 14:1258246. [PMID: 38362497 PMCID: PMC10867257 DOI: 10.3389/fcimb.2024.1258246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 01/16/2024] [Indexed: 02/17/2024] Open
Abstract
Pulmonary fibrosis (PF) is a terminal change of a lung disease that is marked by damage to alveolar epithelial cells, abnormal proliferative transformation of fibroblasts, excessive deposition of extracellular matrix (ECM), and concomitant inflammatory damage. Its characteristics include short median survival, high mortality rate, and limited treatment effectiveness. More in-depth studies on the mechanisms of PF are needed to provide better treatment options. The idea of the gut-lung axis has emerged as a result of comprehensive investigations into the microbiome, metabolome, and immune system. This theory is based on the material basis of microorganisms and their metabolites, while the gut-lung circulatory system and the shared mucosal immune system act as the connectors that facilitate the interplay between the gastrointestinal and respiratory systems. The emergence of a new view of the gut-lung axis is complementary and cross-cutting to the study of the mechanisms involved in PF and provides new ideas for its treatment. This article reviews the mechanisms involved in PF, the gut-lung axis theory, and the correlation between the two. Exploring the gut-lung axis mechanism and treatments related to PF from the perspectives of microorganisms, microbial metabolites, and the immune system. The study of the gut-lung axis and PF is still in its early stages. This review systematically summarizes the mechanisms of PF related to the gut-lung axis, providing ideas for subsequent research and treatment of related mechanisms.
Collapse
Affiliation(s)
- Yawei Dong
- Key Laboratory of Gansu Provincial Prescription Mining and Innovative Translational Laboratory, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
- Gansu Provincial Traditional Chinese Medicine New Product Creation Engineering Laboratory, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Lanlan He
- Key Laboratory of Gansu Provincial Prescription Mining and Innovative Translational Laboratory, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
- Gansu Provincial Traditional Chinese Medicine New Product Creation Engineering Laboratory, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Zhongbo Zhu
- Key Laboratory of Gansu Provincial Prescription Mining and Innovative Translational Laboratory, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
- Gansu Provincial Traditional Chinese Medicine New Product Creation Engineering Laboratory, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Fan Yang
- Key Laboratory of Gansu Provincial Prescription Mining and Innovative Translational Laboratory, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
- Gansu Provincial Traditional Chinese Medicine New Product Creation Engineering Laboratory, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Quan Ma
- Key Laboratory of Gansu Provincial Prescription Mining and Innovative Translational Laboratory, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
- Gansu Provincial Traditional Chinese Medicine New Product Creation Engineering Laboratory, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
- Respiratory Medicine, Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Yanmei Zhang
- Key Laboratory of Gansu Provincial Prescription Mining and Innovative Translational Laboratory, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
- Gansu Provincial Traditional Chinese Medicine New Product Creation Engineering Laboratory, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Xuhui Zhang
- Key Laboratory of Gansu Provincial Prescription Mining and Innovative Translational Laboratory, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
- Gansu Provincial Traditional Chinese Medicine New Product Creation Engineering Laboratory, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
- Respiratory Medicine, Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Xiping Liu
- Key Laboratory of Gansu Provincial Prescription Mining and Innovative Translational Laboratory, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
- Gansu Provincial Traditional Chinese Medicine New Product Creation Engineering Laboratory, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| |
Collapse
|
18
|
Duckworth LA, Sutton KA, Shaikh N, Wang J, Hall-Moore C, Holtz LR, Tarr PI, Rubenstein RC. Quantification of Enteric Dysfunction in Cystic Fibrosis: Inter- and Intraindividual Variability. J Pediatr 2024; 265:113800. [PMID: 37866678 PMCID: PMC10869934 DOI: 10.1016/j.jpeds.2023.113800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 09/18/2023] [Accepted: 10/17/2023] [Indexed: 10/24/2023]
Abstract
OBJECTIVES To test the utility of various biomarkers as indicators of gut dysfunction in cystic fibrosis (CF) and determine whether intraindividual variations in these measures are repeatable over short intervals and whether interindividual variations correlate with clinical outcomes. STUDY DESIGN We performed a cross-sectional, limited longitudinal study of children with CF aged 1-21 years who provided blood and stool samples at 2 or 3 visits, 2 weeks and 3 months apart, which were assayed for markers of intestinal inflammation (fecal calprotectin [fCal], lipocalin-2 [fLcn2], neopterin), and permeability (plasma lipopolysaccharide [LPS] antibodies, LPS-binding protein) by enzyme immunoassays. Control specimens were obtained from children without CF who had undergone esophagogastroduodenoscopy and had no evidence of gut inflammation. RESULTS Twenty-six of 29 participants with CF completed the study. Sixty-nine stools (57 case/12 control) and 76 plasmas (60 case/16 control) were analyzed. LPS antibody had reliable intraindividual stability. fCal, fLcn2, and neopterin were significantly greater in CF than in control samples. fCal was negatively correlated with 3-month interval change (Δ) in weight-for-age z-score, body mass index/weight-for-length z-score, and forced expiratory volume in 1 second. fLcn2 was negatively correlated with FEV1 but not with anthropometrics. No marker correlated with Δbody mass index/weight-for-length z-score or ΔFEV1. CONCLUSIONS fLcn2 is elevated in people with CF and might predict worse interval pulmonary function. Expanded studies are warranted to test if fLcn2 correlates with changes in additional outcomes.
Collapse
Affiliation(s)
- Laura A Duckworth
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Washington University in St Louis, St Louis, MO.
| | - Kimberly A Sutton
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Washington University in St Louis, St Louis, MO
| | - Nurmohammad Shaikh
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Washington University in St Louis, St Louis, MO
| | - Jinli Wang
- Center for Biostatistics and Data Science, Washington University in St Louis, St Louis, MO
| | - Carla Hall-Moore
- Center for Biostatistics and Data Science, Washington University in St Louis, St Louis, MO
| | - Lori R Holtz
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Washington University in St Louis, St Louis, MO
| | - Phillip I Tarr
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Washington University in St Louis, St Louis, MO
| | - Ronald C Rubenstein
- Division of Allergy and Pulmonary Medicine, Department of Pediatrics, Washington University in St Louis, St Louis, MO
| |
Collapse
|
19
|
Wang HY, Liu LX, Chen XY, Zhang YD, Li WX, Li WW, Wang L, Mo XL, Wei H, Ji P, Xie P. Comprehensive analysis of the gut microbiome and post-translational modifications elucidates the route involved in microbiota-host interactions. Zool Res 2024; 45:95-107. [PMID: 38114436 PMCID: PMC10839661 DOI: 10.24272/j.issn.2095-8137.2023.008] [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: 04/06/2023] [Accepted: 09/08/2023] [Indexed: 12/21/2023] Open
Abstract
The gut microbiome interacts with the host to maintain body homeostasis, with gut microbial dysbiosis implicated in many diseases. However, the underlying mechanisms of gut microbe regulation of host behavior and brain functions remain unclear. This study aimed to elucidate the influence of gut microbiota on brain functions via post-translational modification mechanisms in the presence or absence of bacteria without any stimulation. We conducted succinylome analysis of hippocampal proteins in germ-free (GF) and specific pathogen-free (SPF) mice and metagenomic analysis of feces from SPF mice. These results were integrated with previously reported hippocampal acetylome and phosphorylome data from the same batch of mice. Subsequent bioinformatics analyses revealed 584 succinylation sites on 455 proteins, including 54 up-regulated succinylation sites on 91 proteins and 99 down-regulated sites on 51 proteins in the GF mice compared to the SPF mice. We constructed a panoramic map of gut microbiota-regulated succinylation, acetylation, and phosphorylation, and identified cross-talk and relative independence between the different types of post-translational modifications in modulating complicated intracellular pathways. Pearson correlation analysis indicated that 13 taxa, predominantly belonging to the Bacteroidetes phylum, were correlated with the biological functions of post-translational modifications. Positive correlations between these taxa and succinylation and negative correlations between these taxa and acetylation were identified in the modulation of intracellular pathways. This study highlights the hippocampal physiological changes induced by the absence of gut microbiota, and proteomic quantification of succinylation, phosphorylation, and acetylation, contributing to our understanding of the role of the gut microbiome in brain function and behavioral phenotypes.
Collapse
Affiliation(s)
- Hai-Yang Wang
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- College of Stomatology and Affiliated Stomatological Hospital of Chongqing Medical University, Chongqing 401147, China
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing 401147, China
| | - Lan-Xiang Liu
- Department of Neurology, Yongchuan Hospital of Chongqing Medical University, Chongqing 402160, China
| | - Xue-Yi Chen
- Department of Pathology, Faculty of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Yang-Dong Zhang
- Department of Neurology, First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Wen-Xia Li
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Wen-Wen Li
- Department of Pathology, Faculty of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Lian Wang
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Xiao-Long Mo
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Department of Neurology, First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Hong Wei
- Yu-Yue Pathology Scientific Research Center, Chongqing 401329, China. E-mail:
| | - Ping Ji
- College of Stomatology and Affiliated Stomatological Hospital of Chongqing Medical University, Chongqing 401147, China
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing 401147, China. E-mail:
| | - Peng Xie
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- College of Stomatology and Affiliated Stomatological Hospital of Chongqing Medical University, Chongqing 401147, China
- Department of Neurology, First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China. E-mail:
| |
Collapse
|
20
|
Cheng ZX, Wu YX, Jie ZJ, Li XJ, Zhang J. Genetic evidence on the causality between gut microbiota and various asthma phenotypes: a two-sample Mendelian randomization study. Front Cell Infect Microbiol 2024; 13:1270067. [PMID: 38274730 PMCID: PMC10808785 DOI: 10.3389/fcimb.2023.1270067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 12/26/2023] [Indexed: 01/27/2024] Open
Abstract
Introduction Asthma is a multifarious disease that manifests in various phenotypes. Among the various factors that contribute to the development of asthma, the gut microbiota has recently emerged as a compelling area of investigation. This study aims to investigate the causal relationships between gut microbiota and distinct asthma phenotypes. Methods The genome-wide association study (GWAS) summary statistics for 211 gut microbial taxa were used as study exposure. Five traits pertaining to various asthma phenotypes (asthma, allergic asthma, childhood asthma, suggestive for eosinophilic asthma and obesity-related asthma) were included as study outcome. We conducted Mendelian randomization (MR) analysis and sensitivity analysis for each bacterial taxa and asthma phenotypes. Result We discovered a total of 58 associations that exhibited evidence of causality. Out of these, 4 associations remained significant even after applying multiple correction. An increased risk of asthma was causally associated with higher abundance of genus Holdemanella (OR = 1.11; CI: 1.05-1.17; p = 0.027), genus Oxalobacter (OR = 1.09; CI: 1.04-1.15; p = 0.025) and genus Butyricimonas (OR = 1.14; CI: 1.06-1.22; p = 0.027). Order NB1n was causally linked with an increased risk of obesity-related asthma (OR = 1.17; CI: 1.07-1.29; p = 0.015). There was limited overlap among the taxa that exhibited potential causal relationships with distinct asthma phenotypes. Conclusion Our research has provided genetic evidence that establishes multiple causal relationships between the gut microbiota and distinct asthma phenotypes, supporting the role of the gut microbiota in various asthma phenotypes. It is possible that different taxa play a role in the development of distinct asthma phenotypes. The causal relationships identified in this study require further investigation.
Collapse
Affiliation(s)
- Zi-Xuan Cheng
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yi-Xing Wu
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhi-Jun Jie
- Department of Respiratory and Critical Care Medicine, the Fifth People’s Hospital of Shanghai, Fudan University, Shanghai, China
| | - Xing-Jing Li
- Department of Respiratory Medicine, Zhongshan Hospital Wusong Branch, Fudan University, Shanghai, China
| | - Jing Zhang
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
21
|
Mandarino Alves A, Lecchi C, Lopez S, Stornetta A, Mathai PP, Villalta PW, Ishii S, Balskus EP, Balbo S, Khoruts A. Dysfunctional mucus structure in cystic fibrosis increases vulnerability to colibactin-mediated DNA adducts in the colon mucosa. Gut Microbes 2024; 16:2387877. [PMID: 39133871 PMCID: PMC11321416 DOI: 10.1080/19490976.2024.2387877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 04/22/2024] [Accepted: 07/30/2024] [Indexed: 08/15/2024] Open
Abstract
Colibactin is a recently characterized pro-carcinogenic genotoxin produced by pks+ Escherichia coli. We hypothesized that cystic fibrosis (CF)-associated dysfunctional mucus structure increases the vulnerability of host mucosa to colibactin-induced DNA damage. In this pilot study, we tested healthy-appearing mucosal biopsy samples obtained during screening and surveillance colonoscopies of adult CF and non-CF patients for the presence of pks+ E. coli, and we investigated the possibility of detecting a novel colibactin-specific DNA adduct that has not been yet been demonstrated in humans. While CF patients had a lower incidence of pks+ E. coli carriage (~8% vs 29%, p = 0.0015), colibactin-induced DNA adduct formation was detected, but only in CF patients and only in those who were not taking CFTR modulator medications. Moreover, the only patient found to have colon cancer during this study had CF, harbored pks+ E. coli, and had colibactin-induced DNA adducts in the mucosal samples. Larger studies with longitudinal follow-up should be done to extend these initial results and further support the development of colibactin-derived DNA adducts to stratify patients and their risk.
Collapse
Affiliation(s)
- Amanda Mandarino Alves
- Department of Medicine, Division of Gastroenterology, Hepatology, and Nutrition, University of Minnesota, Minneapolis, MN, USA
| | - Chiara Lecchi
- Department of Environmental Health Sciences, School of Public Health, University of Minnesota, Minneapolis, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Sharon Lopez
- Department of Medicine, Division of Gastroenterology, Hepatology, and Nutrition, University of Minnesota, Minneapolis, MN, USA
| | - Alessia Stornetta
- Department of Environmental Health Sciences, School of Public Health, University of Minnesota, Minneapolis, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Prince P. Mathai
- Department of Medicine, Division of Gastroenterology, Hepatology, and Nutrition, University of Minnesota, Minneapolis, MN, USA
| | - Peter W. Villalta
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN, USA
| | - Satoshi Ishii
- Department of Soil, Water, and Climate, University of Minnesota, Minneapolis, MN, USA
- BioTechnology Institute, University of Minnesota, Minneapolis, MN, USA
| | - Emily P. Balskus
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA
| | - Silvia Balbo
- Department of Environmental Health Sciences, School of Public Health, University of Minnesota, Minneapolis, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Alexander Khoruts
- Department of Medicine, Division of Gastroenterology, Hepatology, and Nutrition, University of Minnesota, Minneapolis, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
- BioTechnology Institute, University of Minnesota, Minneapolis, MN, USA
- Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
22
|
Chetty A, Blekhman R. Multi-omic approaches for host-microbiome data integration. Gut Microbes 2024; 16:2297860. [PMID: 38166610 PMCID: PMC10766395 DOI: 10.1080/19490976.2023.2297860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 12/18/2023] [Indexed: 01/05/2024] Open
Abstract
The gut microbiome interacts with the host through complex networks that affect physiology and health outcomes. It is becoming clear that these interactions can be measured across many different omics layers, including the genome, transcriptome, epigenome, metabolome, and proteome, among others. Multi-omic studies of the microbiome can provide insight into the mechanisms underlying host-microbe interactions. As more omics layers are considered, increasingly sophisticated statistical methods are required to integrate them. In this review, we provide an overview of approaches currently used to characterize multi-omic interactions between host and microbiome data. While a large number of studies have generated a deeper understanding of host-microbiome interactions, there is still a need for standardization across approaches. Furthermore, microbiome studies would also benefit from the collection and curation of large, publicly available multi-omics datasets.
Collapse
Affiliation(s)
- Ashwin Chetty
- Committee on Genetics, Genomics and Systems Biology, The University of Chicago, Chicago, IL, USA
| | - Ran Blekhman
- Section of Genetic Medicine, Department of Medicine, The University of Chicago, Chicago, IL, USA
| |
Collapse
|
23
|
Joo MK, Lee JW, Woo JH, Kim HJ, Kim DH, Choi JH. Regulation of colonic neuropeptide Y expression by the gut microbiome in patients with ulcerative colitis and its association with anxiety- and depression-like behavior in mice. Gut Microbes 2024; 16:2319844. [PMID: 38404132 PMCID: PMC10900276 DOI: 10.1080/19490976.2024.2319844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 02/13/2024] [Indexed: 02/27/2024] Open
Abstract
Patients with inflammatory bowel disease (IBD), including ulcerative colitis (UC), show an increased incidence of anxiety and depression; however, the association between UC-associated psychiatric disorders and the gut microbiota is unclear. This study aimed to examine whether gut microbiota from patients with UC can alter colonic gene expression, leading to anxiety- and depression-like behavior in mice receiving fecal microbiota transplantation (FMT). RNA sequencing transcriptome analyses revealed a difference in colonic gene expression between mice receiving FMT from patients with UC (UC-FMT mice) and those receiving FMT from healthy controls (HC-FMT mice). Gene ontology analysis revealed the downregulation of neuropeptide signaling pathways, including neuropeptide Y (NPY) expression, in the colons of UC-FMT mice. The protein levels of NPY also decreased in the colon and plasma of UC-FMT mice compared to those in HC-FMT mice. The oral administration of Enterococcus mundtii (EM), a bacterium isolated from the feces of patients with UC, reduced NPY expression in the colons of mice and induced intestinal inflammation, anxiety, and depression-like behavior. Reduced NPY protein levels were also observed in the plasma and hippocampus of EM-treated mice. Intraperitoneal administration of NPY significantly alleviated anxiety- and depressive-like behaviors induced by EM in mice. Capsular polysaccharide in EM was associated with EM-induced NPY downregulation in the colon. Analysis of Gene Expression Omnibus datasets showed markedly reduced NPY expression in the inflamed colons of patients with UC compared with that in the colons of healthy controls. In summary, EM-induced reduction in the colonic expression of NPY may be associated with a decrease in hippocampal NPY and anxiety- and depression-like behavior in mice.
Collapse
Affiliation(s)
- Min-Kyung Joo
- Department of Biomedical and Pharmaceutical Sciences, Kyung Hee University, Seoul, Korea
- College of Pharmacy, Kyung Hee University, Seoul, Korea
| | - Jae-Won Lee
- Department of Biomedical and Pharmaceutical Sciences, Kyung Hee University, Seoul, Korea
- College of Pharmacy, Kyung Hee University, Seoul, Korea
| | - Jeong-Hwa Woo
- College of Pharmacy, Kyung Hee University, Seoul, Korea
| | - Hyo-Jong Kim
- Department of Internal Medicine, School of Medicine, Kyung Hee University, Seoul, Korea
| | - Dong-Hyun Kim
- Department of Biomedical and Pharmaceutical Sciences, Kyung Hee University, Seoul, Korea
- College of Pharmacy, Kyung Hee University, Seoul, Korea
| | - Jung-Hye Choi
- Department of Biomedical and Pharmaceutical Sciences, Kyung Hee University, Seoul, Korea
- College of Pharmacy, Kyung Hee University, Seoul, Korea
| |
Collapse
|
24
|
Shahila Ismail KI, Kumar CVS, Aneesha U, Syama PS, Sajini KP. Comparative analysis of gut bacteria of silkworm Bombyx mori L. on exposure to temperature through 16S rRNA high throughput metagenomic sequencing. J Invertebr Pathol 2023; 201:107992. [PMID: 37741505 DOI: 10.1016/j.jip.2023.107992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 09/13/2023] [Accepted: 09/20/2023] [Indexed: 09/25/2023]
Abstract
Global warming is one of the serious threats that adversely affects the development and reproduction of silkworms. The ideal temperature for silkworms to carryout normal life activities is 20-30 °C. Certain bivoltine silkworms that are raised in tropical regions are thermotolerant. But, prolonged exposure to high temperatures may be fatal. In the present study, fifth instar larvae of bivoltine silkworm were exposed to heat shock at 40 ± 2 °C for a short period of one hour per day to examine the changes in the gut microflora. The study used high throughput sequencing to evaluate the impact of intestinal microbes of silkworms in response to high temperature. The findings demonstrated that elevated temperature has a negative impact on the intestinal microbes of silkworm compared to the control which were reared under the optimum temperature (25 ± 3° C). Four hundred and fifty eight (458) species of microbes were reported in the control group whereas only 434 species were reported in the temperature exposed group. The digestive process of silkworms may also be impaired by heat shock due to their effect on digestive enzymes. So, the results indicated that heat shock has an impact on the intestinal microflora of silkworms that control the activity of associated digestive enzymes which affects the digestion and nutritional intake, eventually impacting the growth and development of silkworm larvae and cocoons produced. The morphometric parameters of silkworm larvae and cocoons also showed a considerable drop when exposed to heat shock.
Collapse
Affiliation(s)
- K I Shahila Ismail
- P.G. and Research Department of Zoology, Govt. Victoria College, Palakkad, India.
| | - C V Sreeranjit Kumar
- P.G. and Research Department of Zoology, Govt. Victoria College, Palakkad, India.
| | - U Aneesha
- P.G. and Research Department of Zoology, Govt. Victoria College, Palakkad, India
| | - P S Syama
- P.G. and Research Department of Zoology, Govt. Victoria College, Palakkad, India
| | - K P Sajini
- P.G. and Research Department of Zoology, Govt. Victoria College, Palakkad, India
| |
Collapse
|
25
|
Wen J, He JQ. The Causal Impact of the Gut Microbiota on Respiratory Tuberculosis Susceptibility. Infect Dis Ther 2023; 12:2535-2544. [PMID: 37815754 PMCID: PMC10651823 DOI: 10.1007/s40121-023-00880-4] [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: 08/08/2023] [Accepted: 09/22/2023] [Indexed: 10/11/2023] Open
Abstract
INTRODUCTION Recent cross-sectional research has demonstrated a substantial link between tuberculosis (TB) and gut microbiota. Nevertheless, the causal impact of the gut microbiota on TB susceptibility in humans remains unknown. METHODS The Mendelian randomization (MR) method was utilized for investigating the causality between them. The main method used for MR analysis was the inverse variance weighted (IVW) test, with the MR-Egger, weighted median, weighted mode, and simple median methods serving as supplements. And several sensitivity tests were carried out to validate the MR findings. RESULTS The IVW outcomes suggested that three bacterial traits exhibited associations with susceptibility to respiratory TB after Bonferroni correction, namely Lachnospiraceae UCG010 (odds ratio [OR] 1.73, 95% confidence interval [CI] 1.17-2.55, P = 0.005), Eubacterium (brachy group) (OR 1.33, 95% CI 1.07-1.65, P = 0.009), and Ruminococcaceae UCG005 (OR 0.71, 95% CI 0.52-0.98, P = 0.034). Sensitivity analyses demonstrated that horizontal pleiotropy and heterogeneity were absent, thereby guaranteeing the reliability of the results. CONCLUSION This research sheds light on the causal impact of gut microbiota on respiratory tuberculosis susceptibility, improving our knowledge of therapeutic strategies for managing TB.
Collapse
Affiliation(s)
- Jiayu Wen
- Department of Respiratory and Critical Care Medicine, The Second People's Hospital of Meishan City, 177 Longtan Avenue, Section 1, Huairen Street, Renshou County, Meishan, 620500, China
| | - Jian-Qing He
- Department of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University, No. 37, Guo Xue Alley, Chengdu, 610041, China.
| |
Collapse
|
26
|
Xiang Y, Zhang C, Wang J, Cheng Y, Wang L, Tong Y, Yan D. Identification of host gene-microbiome associations in colorectal cancer patients using mendelian randomization. J Transl Med 2023; 21:535. [PMID: 37563724 PMCID: PMC10416448 DOI: 10.1186/s12967-023-04335-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Accepted: 07/09/2023] [Indexed: 08/12/2023] Open
Abstract
BACKGROUND There are many studies indicating that alterations in the abundance of certain gut microbiota are associated with colorectal cancer (CRC). However, a causal relationship has not been identified due to confounding factors such as lifestyle, environmental, and possible reverse causal associations between the two. Furthermore, certain host gene mutations can also contribute to the development of CRC. However, the association between genes and gut microbes in patients with CRC has not been extensively studied. METHODS We conducted a two-sample Mendelian randomization (MR) study to reveal the causal relationship between gut microbiota and CRC. We obtained SNPs associated with gut microbiome abundance as instrumental variables (IVs) from a large-scale, multi-ethnic GWAS study, and extracted CRC-related datasets from an East Asian Population genetic consortia GWAS (AGWAS) study and FinnGen consortium, respectively. We analyzed a total of 166 bacterial features at four taxonomic levels, including order, family, genus, and species. The inverse-variance-weighted (IVW), weighted median, MR-Egger, and simple median methods were applied to the MR analysis, and the robustness of the results were tested using a series of sensitivity analyses. We extracted IVs of gut microbiota with direct causal association with CRC for SNP annotation to identify the genes in which these genetic variants were located to reveal the possible host gene-microbiome associations in CRC patients. RESULTS The findings from our MR analysis based on CRC-associated GWAS datasets from AGWAS revealed causal relationships between 6 bacterial taxa and CRC at a locus-wide significance level (P < 1 × 10-5). The IVW method found that family Porphyromonadaceae, genera Anaerotruncus, Intestinibacter, Slackia, and Ruminococcaceae UCG004, and species Eubacterium coprostanoligenes group were positively associated with CRC risk, which was generally consistent with the results of other complementary analyses. The results of a meta-analysis of the MR estimates from the AGWAS and the FinnGen datasets showed that family Porphyromonadaceae and genera Slackia, Anaerotruncus, and Intestinibacter replicated the same causal association. Sensitivity analysis of all causal associations did not indicate significant heterogeneity, horizontal pleiotropy, or reverse causal associations. We annotated the SNPs at a locus-wide significance level of the above intestinal flora and identified 24 host genes that may be related to pathogenic intestinal microflora in CRC patients. CONCLUSION This study supported the causal relationship of gut microbiota on CRC and revealed a possible correlation between genes and pathogenic microbiota in CRC. These findings suggested that the study of the gut microbiome and its further multi-omics analysis was important for the prevention and treatment of CRC.
Collapse
Affiliation(s)
- Yaoxian Xiang
- Department of Oncology, Beijing Luhe Hospital Affiliated to Capital Medical University, Beijing, 101149, China
| | - Chan Zhang
- Department of Oncology, Beijing Luhe Hospital Affiliated to Capital Medical University, Beijing, 101149, China
| | - Jing Wang
- Department of Oncology, Beijing Luhe Hospital Affiliated to Capital Medical University, Beijing, 101149, China
| | - Yurong Cheng
- Department of Oncology, Beijing Luhe Hospital Affiliated to Capital Medical University, Beijing, 101149, China
| | - Li Wang
- Department of Oncology, Beijing Luhe Hospital Affiliated to Capital Medical University, Beijing, 101149, China
| | - Yingying Tong
- Department of Oncology, Beijing Luhe Hospital Affiliated to Capital Medical University, Beijing, 101149, China.
| | - Dong Yan
- Department of Oncology, Beijing Luhe Hospital Affiliated to Capital Medical University, Beijing, 101149, China.
| |
Collapse
|
27
|
Woodall CA, Hammond A, Cleary D, Preston A, Muir P, Pascoe B, Sheppard SK, Hay AD. Oral and gut microbial biomarkers of susceptibility to respiratory tract infection in adults: A feasibility study. Heliyon 2023; 9:e18610. [PMID: 37593638 PMCID: PMC10432180 DOI: 10.1016/j.heliyon.2023.e18610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 07/10/2023] [Accepted: 07/24/2023] [Indexed: 08/19/2023] Open
Abstract
We conducted a feasibility cohort study which aimed to recruit and retain adults from the community to collect saliva (oral) and stool (gut) samples at three time points, at the start of the study (baseline), during a respiratory tract infection (RTI) and post-RTI. Community RTIs place a huge burden on health care services, and a non-invasive microbial diagnostic tool to predict the most vulnerable to respiratory infection would be ideal. To this aim, we analysed oral-gut baseline samples comparing those who reported RTI symptoms to those who remained healthy throughout the study for microbial biomarkers of respiratory susceptibility. Amplicon sequence variants (ASV) were identified by 16S sequence profiling to reveal oral-gut microbes. Reverse transcriptase-polymerase chain reaction (RT-PCR) was applied to target common respiratory microbes. Two general practices were recruited, and the participant recruitment rate was 1.3%. A total of 40 adult participants were retained, of which 19 acquired an RTI whereas 21 remained healthy. In healthy baseline oral and gut samples, ASVs from participants with RTI symptoms compared to those who remained healthy were similar with a high relative abundance of Streptococcus sp., and Blautia sp., respectively. Linear discriminant analysis effect size (LEfSe) revealed baseline oral microbes differed, indicating participants who suffered RTI symptoms had enhanced Streptococcus sobrinus and Megamonas sp., and depletion of Lactobacillus salivarius, Synergistetes, Verrucomicrobia and Dethiosulfovibrio. Furthermore, a random forest model ranked Streptococcus (4.13) as the highest mean decrease in accuracy (MDA) and RT-PCR showed a higher level of carriage of coagulase-negative Staphylococcus. Baseline core gut microbes were similar in both participant groups whereas LEfSe analysis revealed enhanced Veillonella, Rikenellaceae, Enhydobacteria, Eggerthella and Xanthomonsdales and depleted Desulfobulbus and Coprobacillus. Sutterella (4.73) had a high MDA value. Overall, we demonstrated the feasibility of recruiting and retaining adult participants from the community to provide multiple biological samples for microbial profiling. Our analyses identified potential oral-gut microbial biomarkers of respiratory infection susceptibility in otherwise healthy participants.
Collapse
Affiliation(s)
- Claire A. Woodall
- School of Cellular and Molecular Medicine, Bristol Medical School, University of Bristol, Bristol, UK
| | - Ashley Hammond
- Centre for Academic Primary Care, Population Health Science, Bristol Medical School, University of Bristol, Bristol, UK
| | - David Cleary
- Institute of Microbiology and Infection, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, UK
| | - Andrew Preston
- The Milner Centre for Evolution and Department of Biology and Biochemistry, University of Bath, Bath, UK
| | - Peter Muir
- Public Health England, Southwest Regional Laboratory, National Infection Service, Southmead Hospital, Bristol, UK
| | - Ben Pascoe
- Department of Biology, University of Oxford, Oxford, UK
| | | | - Alastair D. Hay
- Centre for Academic Primary Care, Population Health Science, Bristol Medical School, University of Bristol, Bristol, UK
| |
Collapse
|
28
|
Queen J, Shaikh F, Sears CL. Understanding the mechanisms and translational implications of the microbiome for cancer therapy innovation. NATURE CANCER 2023; 4:1083-1094. [PMID: 37525016 DOI: 10.1038/s43018-023-00602-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 06/21/2023] [Indexed: 08/02/2023]
Abstract
The intersection of the microbiota and cancer and the mechanisms that define these interactions are a fascinating, rapidly evolving area of cancer biology and therapeutics. Here we present recent insights into the mechanisms by which specific bacteria or their communities contribute to carcinogenesis and discuss the bidirectional interplay between microbiota and host gene or epigenome signaling. We conclude with comments on manipulation of the microbiota for the therapeutic benefit of patients with cancer.
Collapse
Affiliation(s)
- Jessica Queen
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Fyza Shaikh
- Cancer Immunology Program, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Cynthia L Sears
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Cancer Immunology Program, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Microbiology and Molecular Immunology, Bloomberg School of Public Health, Baltimore, MD, USA.
| |
Collapse
|
29
|
Lou S, Yang M, Li T, Zhao W, Cevasco H, Yang YT, Gerstein M. Constructing a full, multiple-layer interactome for SARS-CoV-2 in the context of lung disease: Linking the virus with human genes and microbes. PLoS Comput Biol 2023; 19:e1011222. [PMID: 37410793 PMCID: PMC10325097 DOI: 10.1371/journal.pcbi.1011222] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 05/28/2023] [Indexed: 07/08/2023] Open
Abstract
The COVID-19 pandemic caused by the SARS-CoV-2 virus has resulted in millions of deaths worldwide. The disease presents with various manifestations that can vary in severity and long-term outcomes. Previous efforts have contributed to the development of effective strategies for treatment and prevention by uncovering the mechanism of viral infection. We now know all the direct protein-protein interactions that occur during the lifecycle of SARS-CoV-2 infection, but it is critical to move beyond these known interactions to a comprehensive understanding of the "full interactome" of SARS-CoV-2 infection, which incorporates human microRNAs (miRNAs), additional human protein-coding genes, and exogenous microbes. Potentially, this will help in developing new drugs to treat COVID-19, differentiating the nuances of long COVID, and identifying histopathological signatures in SARS-CoV-2-infected organs. To construct the full interactome, we developed a statistical modeling approach called MLCrosstalk (multiple-layer crosstalk) based on latent Dirichlet allocation. MLCrosstalk integrates data from multiple sources, including microbes, human protein-coding genes, miRNAs, and human protein-protein interactions. It constructs "topics" that group SARS-CoV-2 with genes and microbes based on similar patterns of co-occurrence across patient samples. We use these topics to infer linkages between SARS-CoV-2 and protein-coding genes, miRNAs, and microbes. We then refine these initial linkages using network propagation to contextualize them within a larger framework of network and pathway structures. Using MLCrosstalk, we identified genes in the IL1-processing and VEGFA-VEGFR2 pathways that are linked to SARS-CoV-2. We also found that Rothia mucilaginosa and Prevotella melaninogenica are positively and negatively correlated with SARS-CoV-2 abundance, a finding corroborated by analysis of single-cell sequencing data.
Collapse
Affiliation(s)
- Shaoke Lou
- Program in Computational Biology & Bioinformatics, Yale University, New Haven, Connecticut, United States of America
- Department of Molecular Biophysics & Biochemistry, Yale University, New Haven, Connecticut, United States of America
| | - Mingjun Yang
- School of Electronic Engineering and Computer Science, Queen Mary University of London, Mile End Road, London, United Kingdom
| | - Tianxiao Li
- Program in Computational Biology & Bioinformatics, Yale University, New Haven, Connecticut, United States of America
- Department of Molecular Biophysics & Biochemistry, Yale University, New Haven, Connecticut, United States of America
| | - Weihao Zhao
- Program in Computational Biology & Bioinformatics, Yale University, New Haven, Connecticut, United States of America
- Department of Molecular Biophysics & Biochemistry, Yale University, New Haven, Connecticut, United States of America
| | - Hannah Cevasco
- Program in Computational Biology & Bioinformatics, Yale University, New Haven, Connecticut, United States of America
| | - Yucheng T. Yang
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
- MOE Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Shanghai, China
- MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Mark Gerstein
- Program in Computational Biology & Bioinformatics, Yale University, New Haven, Connecticut, United States of America
- Department of Molecular Biophysics & Biochemistry, Yale University, New Haven, Connecticut, United States of America
- Department of Computer Science, Yale University, New Haven, Connecticut, United States of America
- Department of Statistics & Data Science Yale University, New Haven, Connecticut, United States of America
- Department of Biomedical Informatics & Data Science, Yale University, New Haven, Connecticut, United States of America
| |
Collapse
|
30
|
Baldwin-Hunter BL, Rozenberg FD, Annavajhala MK, Park H, DiMango EA, Keating CL, Uhlemann AC, Abrams JA. The gut microbiome, short chain fatty acids, and related metabolites in cystic fibrosis patients with and without colonic adenomas. J Cyst Fibros 2023; 22:738-744. [PMID: 36717332 DOI: 10.1016/j.jcf.2023.01.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 12/02/2022] [Accepted: 01/23/2023] [Indexed: 01/29/2023]
Abstract
BACKGROUND Adults with cystic fibrosis (CF) are at increased risk for colon cancer. CF patients have reductions in intestinal bacteria that produce short chain fatty acids (SCFAs), although it is unclear whether this corresponds with intestinal SCFA levels and the presence of colonic neoplasia. The aim of this study was to compare gut microbiome and SCFA composition in patients with and without CF, and to assess associations with colonic adenomas. METHODS Colonic aspirates were obtained from adults with and without CF undergoing colon cancer screening or surveillance colonoscopy. Microbiome characterization was performed by 16S rRNA V3-V4 sequencing. Targeted profiling of SCFAs and related metabolites was performed by LC-MS. RESULTS 42 patients (21 CF, 21 control) were enrolled. CF patients had significantly reduced alpha diversity and decreased relative abundance of many SCFA-producing taxa. There were no significant differences in SCFA levels in CF patients, although there were reduced levels of branched chain fatty acids (BCFAs) and related metabolites. CF patients with adenomas, but not controls with adenomas, had significantly increased relative abundance of Bacteroides fragilis. CF microbiome composition was significantly associated with isovalerate concentration and the presence of adenomas. CONCLUSIONS CF patients have marked disturbances in the gut microbiome, and CF patients with adenomas had notably increased relative abundance of B. fragilis, a pathogen known to promote colon cancer. Reductions in BCFAs but not SCFAs were found in CF. Further studies are warranted to evaluate the role of B. fragilis as well the biological significance of reductions in BCFAs in CF.
Collapse
Affiliation(s)
| | - Felix D Rozenberg
- Microbiome and Pathogen Genomics Collaborative Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Medini K Annavajhala
- Microbiome and Pathogen Genomics Collaborative Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Heekuk Park
- Microbiome and Pathogen Genomics Collaborative Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Emily A DiMango
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA; Gunnar Esiason Adult Cystic Fibrosis and Lung Disease Program, Columbia University Irving Medical Center, New York, NY, USA
| | - Claire L Keating
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA; Gunnar Esiason Adult Cystic Fibrosis and Lung Disease Program, Columbia University Irving Medical Center, New York, NY, USA
| | - Anne-Catrin Uhlemann
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA; Microbiome and Pathogen Genomics Collaborative Center, Columbia University Irving Medical Center, New York, NY, USA; Digestive and Liver Disease Research Center, Columbia University Irving Medical Center, New York, NY, USA.
| | - Julian A Abrams
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA; Digestive and Liver Disease Research Center, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
31
|
Zhou X, Ji L, Ma Y, Tian G, Lv K, Yang J. Intratumoral Microbiota-Host Interactions Shape the Variability of Lung Adenocarcinoma and Lung Squamous Cell Carcinoma in Recurrence and Metastasis. Microbiol Spectr 2023; 11:e0373822. [PMID: 37074188 PMCID: PMC10269859 DOI: 10.1128/spectrum.03738-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 03/10/2023] [Indexed: 04/20/2023] Open
Abstract
Differences in tissue microbiota-host interaction between lung squamous cell carcinoma (LUSC) and lung adenocarcinoma (LUAD) about recurrence and metastasis have not been well studied. In this study, we performed bioinformatics analyses to identify the genes and tissue microbes significantly associated with recurrence or metastasis. All lung cancer patients were divided into the recurrence or metastasis (RM) group and the nonrecurrence and nonmetastasis (non-RM) group according to whether or not they had recurred or metastasized within 3 years after the initial surgery. Results showed that there were significant differences between LUAD and LUSC in gene expression and microbial abundance associated with recurrence and metastasis. Compared with non-RM, the bacterial community of RM had a lower richness in LUSC. In LUSC, host genes significantly correlated with tissue microbe, whereas host-tissue microbe interaction in LUAD was rare. Then, we established a novel multimodal machine learning model based on genes and microbes to predict the recurrence and metastasis risk of a LUSC patient, which achieves an area under the curve (AUC) of 0.81. In addition, the predicted risk score was significantly associated with the patient's survival. IMPORTANCE Our study elucidates significant differences in RM-associated host-microbe interactions between LUAD and LUSC. Besides, the microbes in tumor tissue could be used to predict the RM risk of LUSC, and the predicted risk score is associated with patients' survival.
Collapse
Affiliation(s)
- Xiangfeng Zhou
- Department of Mathematics, Ocean University of China, Qingdao, China
- Geneis Beijing Co., Ltd., Beijing, China
| | - Lei Ji
- Geneis Beijing Co., Ltd., Beijing, China
- Qingdao Geneis Institute of Big Data Mining and Precision Medicine, Qingdao, China
| | - Yanyu Ma
- Qingdao Geneis Institute of Big Data Mining and Precision Medicine, Qingdao, China
- Department of Mathematics, Zhejiang University of Science and Technology, Hangzhou, Zhejiang, China
| | - Geng Tian
- Geneis Beijing Co., Ltd., Beijing, China
- Qingdao Geneis Institute of Big Data Mining and Precision Medicine, Qingdao, China
| | - Kebo Lv
- Department of Mathematics, Ocean University of China, Qingdao, China
| | - Jialiang Yang
- Geneis Beijing Co., Ltd., Beijing, China
- Qingdao Geneis Institute of Big Data Mining and Precision Medicine, Qingdao, China
- Chifeng Municipal Hospital, Chifeng, Inner Mongolia, China
- Academician Workstation, Changsha Medical University, Changsha, China
| |
Collapse
|
32
|
Birch RJ, Peckham D, Wood HM, Quirke P, Konstant-Hambling R, Brownlee K, Cosgriff R, Consortium GER, Burr N, Downing A. The risk of colorectal cancer in individuals with mutations of the cystic fibrosis transmembrane conductance regulator (CFTR) gene: An English population-based study. J Cyst Fibros 2023; 22:499-504. [PMID: 36253274 DOI: 10.1016/j.jcf.2022.10.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 06/21/2022] [Accepted: 10/03/2022] [Indexed: 06/10/2023]
Abstract
BACKGROUND Studies have demonstrated a higher risk of developing colorectal cancer (CRC) in individuals with Cystic Fibrosis (CF), and also a potentially increased risk in carriers of cystic fibrosis transmembrane conductance regulator (CFTR) mutations. Life expectancy for those with CF is rising, increasing the number at risk of developing CRC. METHODS The incidence of CRC amongst individuals with CF was calculated using data from CORECT-R and linked UK CF Registry and Secondary User Services (SUS) data. Crude, age-specific and age-standardised rates were compared to those without CF. The presence of CFTR mutations in individuals with CRC was assessed using 100,000 Genomes Project data. FINDINGS The crude incidence rate of CRC in the CF population was 0.29 per 1,000 person-years (28 cases). The CF population were significantly younger than those without (median age at CRC diagnosis 52 years versus 73 years; p<0·01). When age-adjusted, there was a 5-fold increased CRC incidence amongst individuals with CF compared to those without (SIR 5.0 95%CI 3.2-6.9). When compared to other population studies the overall prevalence of CFTR mutations in the CRC population was significantly higher than expected (p<0·01). INTERPRETATION CF is linked to an increased risk of CRC. The incidence of CFTR mutations in the CRC population is higher than would be expected, suggesting an association between CFTR function and CRC risk. Further research is needed to develop effective screening strategies for these populations. FUNDING Cancer Research UK (grants C23434/A23706 & C10674/A27140).
Collapse
Affiliation(s)
- Rebecca J Birch
- Leeds Institute of Medical Research at St James's, University of Leeds; Leeds Institute for Data Analytics, University of Leeds.
| | - Daniel Peckham
- Leeds Institute of Medical Research at St James's, University of Leeds; Leeds Teaching Hospitals NHS Trust
| | - Henry M Wood
- Leeds Institute of Medical Research at St James's, University of Leeds
| | - Philip Quirke
- Leeds Institute of Medical Research at St James's, University of Leeds
| | | | | | | | | | | | - Amy Downing
- Leeds Institute of Medical Research at St James's, University of Leeds; Leeds Institute for Data Analytics, University of Leeds
| |
Collapse
|
33
|
Gong H, Wang T, Wu M, Chu Q, Lan H, Lang W, Zhu L, Song Y, Zhou Y, Wen Q, Yu J, Wang B, Zheng X. Maternal effects drive intestinal development beginning in the embryonic period on the basis of maternal immune and microbial transfer in chickens. MICROBIOME 2023; 11:41. [PMID: 36869365 PMCID: PMC9983169 DOI: 10.1186/s40168-023-01490-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 02/06/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND Nutrition drives immunity and health in animals, and maternal immunity benefits offspring. In our previous study, a nutritional intervention strategy was found to promote the immunity of hens, which subsequently improved immunity and growth in offspring chicks. Maternal effects clearly exist, but how are mothers' immune advantages transferred to their offspring, and how do they benefit them? RESULTS Here, we traced the beneficial effects back to the process of egg formation in the reproductive system, and we focused on the embryonic intestinal transcriptome and development, as well as on maternal microbial transfer in offspring. We found that maternal nutritional intervention benefits maternal immunity, egg hatching, and offspring growth. The results of protein and gene quantitative assays showed that the transfer of immune factors into egg whites and yolks depends on maternal levels. Histological observations indicated that the promotion of offspring intestinal development begins in the embryonic period. Microbiota analyses suggested that maternal microbes transfer to the embryonic gut from the magnum to the egg white. Transcriptome analyses revealed that offspring embryonic intestinal transcriptome shifts are related to development and immunity. Moreover, correlation analyses showed that the embryonic gut microbiota is correlated with the intestinal transcriptome and development. CONCLUSIONS This study suggests that maternal immunity positively influences offspring intestinal immunity establishment and intestinal development beginning in the embryonic period. Adaptive maternal effects might be accomplished via the transfer of relatively large amounts of maternal immune factors and by shaping of the reproductive system microbiota by strong maternal immunity. Moreover, reproductive system microbes may be useful resources for the promotion of animal health. Video Abstract.
Collapse
Affiliation(s)
- Haizhou Gong
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118 China
- Key Laboratory of Animal Production, Product Quality and Security (Jilin Agricultural University), Ministry of Education, Changchun, 130118 China
| | - Taiping Wang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118 China
| | - Min Wu
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118 China
| | - Qianran Chu
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118 China
| | - Hainan Lan
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118 China
| | - Wuying Lang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118 China
| | - Lingyu Zhu
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118 China
| | - Yang Song
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118 China
| | - Yujie Zhou
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118 China
| | - Qiongyi Wen
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118 China
| | - Jing Yu
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118 China
| | - Baolin Wang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118 China
| | - Xin Zheng
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118 China
- Key Laboratory of Animal Production, Product Quality and Security (Jilin Agricultural University), Ministry of Education, Changchun, 130118 China
| |
Collapse
|
34
|
Wang J, Cao Y, Hou W, Bi D, Yin F, Gao Y, Huang D, Li Y, Cao Z, Yan Y, Zhao J, Kong D, Lv X, Huang L, Zhong H, Wu C, Chen Q, Yang R, Wei Q, Qin H. Fecal microbiota transplantation improves VPA-induced ASD mice by modulating the serotonergic and glutamatergic synapse signaling pathways. Transl Psychiatry 2023; 13:17. [PMID: 36670104 PMCID: PMC9859809 DOI: 10.1038/s41398-023-02307-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 01/02/2023] [Accepted: 01/06/2023] [Indexed: 01/22/2023] Open
Abstract
Autism spectrum disorder (ASD) is a complex behavioral disorder diagnosed by social interaction difficulties, restricted verbal communication, and repetitive behaviors. Fecal microbiota transplantation (FMT) is a safe and efficient strategy to adjust gut microbiota dysbiosis and improve ASD-related behavioral symptoms, but its regulatory mechanism is unknown. The impact of the microbiota and its functions on ASD development is urgently being investigated to develop new therapeutic strategies for ASD. We reconstituted the gut microbiota of a valproic acid (VPA)-induced autism mouse model through FMT and found that ASD is in part driven by specific gut dysbiosis and metabolite changes that are involved in the signaling of serotonergic synapse and glutamatergic synapse pathways, which might be associated with behavioral changes. Further analysis of the microbiota showed a profound decrease in the genera Bacteroides and Odoribacter, both of which likely contributed to the regulation of serotonergic and glutamatergic synapse metabolism in mice. The engraftment of Turicibacter and Alistipes was also positively correlated with the improvement in behavior after FMT. Our results suggested that successful transfer of the gut microbiota from healthy donors to ASD mice was sufficient to improve ASD-related behaviors. Modulation of gut dysbiosis by FMT could be an effective approach to improve ASD-related behaviors in patients.
Collapse
Affiliation(s)
- Jifeng Wang
- grid.412538.90000 0004 0527 0050Department of Pathology, Shanghai Tenth People’s Hospital Affiliated to Tongji University, 200072 Shanghai, China
| | - Yuan Cao
- grid.412538.90000 0004 0527 0050Department of Pathology, Shanghai Tenth People’s Hospital Affiliated to Tongji University, 200072 Shanghai, China
| | - Weiliang Hou
- grid.412538.90000 0004 0527 0050Intestinal Microenvironment Treatment Center, Shanghai Tenth People’s Hospital Affiliated to Tongji University, 200072 Shanghai, China
| | - Dexi Bi
- grid.412538.90000 0004 0527 0050Department of Pathology, Shanghai Tenth People’s Hospital Affiliated to Tongji University, 200072 Shanghai, China
| | - Fang Yin
- grid.412538.90000 0004 0527 0050Intestinal Microenvironment Treatment Center, Shanghai Tenth People’s Hospital Affiliated to Tongji University, 200072 Shanghai, China
| | - Yaohui Gao
- grid.412538.90000 0004 0527 0050Department of Pathology, Shanghai Tenth People’s Hospital Affiliated to Tongji University, 200072 Shanghai, China
| | - Dengfeng Huang
- grid.412538.90000 0004 0527 0050Department of Pathology, Shanghai Tenth People’s Hospital Affiliated to Tongji University, 200072 Shanghai, China
| | - Yingying Li
- grid.412538.90000 0004 0527 0050Department of Pathology, Shanghai Tenth People’s Hospital Affiliated to Tongji University, 200072 Shanghai, China
| | - Zhan Cao
- grid.412538.90000 0004 0527 0050Intestinal Microenvironment Treatment Center, Shanghai Tenth People’s Hospital Affiliated to Tongji University, 200072 Shanghai, China
| | - Yinmei Yan
- grid.412538.90000 0004 0527 0050Department of Pediatrics, Shanghai Tenth People’s Hospital Affiliated to Tongji University, 200072 Shanghai, China
| | - Jianhua Zhao
- Shanghai Majorbio Bio-pharm Technology Co.,Ltd, 201210 Shanghai, China
| | - Dewu Kong
- Shanghai Majorbio Bio-pharm Technology Co.,Ltd, 201210 Shanghai, China
| | - Xiaoqiong Lv
- grid.412538.90000 0004 0527 0050Intestinal Microenvironment Treatment Center, Shanghai Tenth People’s Hospital Affiliated to Tongji University, 200072 Shanghai, China
| | - Linsheng Huang
- grid.412538.90000 0004 0527 0050Department of Pediatrics, Shanghai Tenth People’s Hospital Affiliated to Tongji University, 200072 Shanghai, China
| | - Hui Zhong
- grid.412538.90000 0004 0527 0050Department of Pediatrics, Shanghai Tenth People’s Hospital Affiliated to Tongji University, 200072 Shanghai, China
| | - Chunyan Wu
- grid.412538.90000 0004 0527 0050Intestinal Microenvironment Treatment Center, Shanghai Tenth People’s Hospital Affiliated to Tongji University, 200072 Shanghai, China
| | - Qiyi Chen
- grid.412538.90000 0004 0527 0050Intestinal Microenvironment Treatment Center, Shanghai Tenth People’s Hospital Affiliated to Tongji University, 200072 Shanghai, China
| | - Rong Yang
- Department of Pediatrics, Shanghai Tenth People's Hospital Affiliated to Tongji University, 200072, Shanghai, China.
| | - Qing Wei
- Department of Pathology, Shanghai Tenth People's Hospital Affiliated to Tongji University, 200072, Shanghai, China.
| | - Huanlong Qin
- Intestinal Microenvironment Treatment Center, Shanghai Tenth People's Hospital Affiliated to Tongji University, Department of Gastrointestinal Surgery, Shanghai Tenth People's Hospital Affiliated to Tongji University, 200072, Shanghai, China.
| |
Collapse
|
35
|
Gilbert B, Kaiko G, Smith S, Wark P. A systematic review of the colorectal microbiome in adult cystic fibrosis patients. Colorectal Dis 2023; 25:843-852. [PMID: 36598333 DOI: 10.1111/codi.16472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 11/28/2022] [Accepted: 12/17/2022] [Indexed: 01/05/2023]
Abstract
AIM Cystic fibrosis (CF) is a hereditary, life-limiting, multi-system condition that results in chronic respiratory infections, pancreatic insufficiency and intestinal inflammation. Evidence indicates that CF patients develop colorectal cancer (CRC) earlier and more often than the general population. Intestinal dysbiosis resulting from genetics and CF treatment is a contributing factor. This systematic review aims to evaluate the literature to compare the microbiome of adult CF patients to non-CF patients and to assess if these changes correspond with known CRC microbiome alterations. METHODS A systematic review across five databases was performed according to PRISMA guidelines. Studies focusing on adult CF patients using next generation sequencing and with appropriate non-CF controls were included. Two reviewers independently screened results and assessed study quality using the Newcastle-Ottawa scale. RESULTS The search generated 2757 results. 118 studies were retained after reviewing the title/abstract and full article review found five studies met the inclusion criteria. All studies consistently showed reduced microbial diversity in CF patients and unique clustering between CF and control cohorts. Thirty-four genera and 27 species were differently expressed between CF and controls. The CF cohort had a reduced number of short-chain fatty acid (SCFA) producing bacteria and a higher abundance of bacteria associated with CRC compared to controls. CONCLUSION There was substantial heterogeneity across all the studies with regard to methodologies and reporting. However, all studies consistently found CF patients had reduced microbial diversity, fewer SCFA producing bacteria and increased CRC-associated bacteria. Further prospective studies employing consistent multi-omics approaches are needed to improve our understanding of the CF gut microbiome and its involvement in early onset CRC. SIGNIFICANCE STATEMENT This is the first systematic review to assess adult CF colorectal microbiome changes. This study shows CF patients have reduced SCFA producing bacteria and increased CRC-associated bacteria compared to non-CF patients and may help to explain the increased risk of CRC in the CF cohort.
Collapse
Affiliation(s)
- Brent Gilbert
- University of Newcastle, School of Medicine, Newcastle, New South Wales, Australia.,John Hunter Hospital, Newcastle, New South Wales, Australia
| | - Gerard Kaiko
- University of Newcastle, School of Medicine, Newcastle, New South Wales, Australia.,Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Stephen Smith
- University of Newcastle, School of Medicine, Newcastle, New South Wales, Australia.,Hunter Medical Research Institute, Newcastle, New South Wales, Australia.,Calvary Mater Hospital, Newcastle, New South Wales, Australia
| | - Peter Wark
- University of Newcastle, School of Medicine, Newcastle, New South Wales, Australia.,John Hunter Hospital, Newcastle, New South Wales, Australia.,Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| |
Collapse
|
36
|
Caley LR, White H, de Goffau MC, Floto RA, Parkhill J, Marsland B, Peckham DG. Cystic Fibrosis-Related Gut Dysbiosis: A Systematic Review. Dig Dis Sci 2023; 68:1797-1814. [PMID: 36600119 DOI: 10.1007/s10620-022-07812-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 12/23/2022] [Indexed: 01/06/2023]
Abstract
BACKGROUND AND AIMS Cystic Fibrosis (CF) is associated with gut dysbiosis, local and systemic inflammation, and impaired immune function. Gut microbiota dysbiosis results from changes in the complex gut milieu in response to CF transmembrane conductance regulator (CFTR) dysfunction, pancreatic malabsorption, diet, medications, and environmental influences. In several diseases, alteration of the gut microbiota influences local and systemic inflammation and disease outcomes. We conducted a systematic review of the gut microbiota in CF and explored factors influencing dysbiosis. METHODS An electronic search of three databases was conducted in January 2019, and re-run in June 2021. Human, animal, and in vitro studies were included. The primary outcome was differences in the gut microbiota between people with CF (pwCF) and healthy controls. Secondary outcomes included the relationship between the gut microbiota and other factors, including diet, medication, inflammation, and pulmonary function in pwCF. RESULTS Thirty-eight studies were identified. The literature confirmed the presence of CF-related gut dysbiosis, characterized by reduced diversity and several taxonomic changes. There was a relative increase of bacteria associated with a pro-inflammatory response coupled with a reduction of those considered anti-inflammatory. However, studies linking gut dysbiosis to systemic and lung inflammation were limited. Causes of gut dysbiosis were multifactorial, and findings were variable. Data on the impact of CFTR modulators on the gut microbiota were limited. CONCLUSIONS CF-related gut dysbiosis is evident in pwCF. Whether this influences local and systemic disease and is amenable to interventions with diet and drugs, such as CFTR modulators, requires further investigation.
Collapse
Affiliation(s)
- L R Caley
- Leeds Institute of Medical Research, St James's University Hospital, Clinical Sciences Building, Leeds, LS9 7TF, UK
| | - H White
- Nutrition, Health & Environment, Leeds Beckett University, Leeds, UK
| | - M C de Goffau
- Wellcome Sanger Institute, Cambridge, UK.,Tytgat Institute for Liver and Intestinal Research, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - R A Floto
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, Cambridge, UK.,Cambridge Centre for Lung Infection, Royal Papworth Hospital, Cambridge, UK
| | - J Parkhill
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - B Marsland
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Australia
| | - D G Peckham
- Leeds Institute of Medical Research, St James's University Hospital, Clinical Sciences Building, Leeds, LS9 7TF, UK. .,Department of Respiratory Medicine, Leeds Teaching Hospitals NHS Trust, Leeds, UK.
| |
Collapse
|
37
|
Saralegui C, García-Durán C, Romeu E, Hernáez-Sánchez ML, Maruri A, Bastón-Paz N, Lamas A, Vicente S, Pérez-Ruiz E, Delgado I, Luna-Paredes C, Caballero JDD, Zamora J, Monteoliva L, Gil C, del Campo R. Statistical Evaluation of Metaproteomics and 16S rRNA Amplicon Sequencing Techniques for Study of Gut Microbiota Establishment in Infants with Cystic Fibrosis. Microbiol Spectr 2022; 10:e0146622. [PMID: 36255300 PMCID: PMC9784762 DOI: 10.1128/spectrum.01466-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 09/23/2022] [Indexed: 01/05/2023] Open
Abstract
Newborn screening for cystic fibrosis (CF) can identify affected but asymptomatic infants. The selection of omic technique for gut microbiota study is crucial due to both the small amount of feces available and the low microorganism load. Our aims were to compare the agreement between 16S rRNA amplicon sequencing and metaproteomics by a robust statistical analysis, including both presence and abundance of taxa, to describe the sequential establishment of the gut microbiota during the first year of life in a small size sample (8 infants and 28 fecal samples). The taxonomic assignations by the two techniques were similar, whereas certain discrepancies were observed in the abundance detection, mostly the lower predicted relative abundance of Bifidobacterium and the higher predicted relative abundance of certain Firmicutes and Proteobacteria by amplicon sequencing. During the first months of life, the CF gut microbiota is characterized by a significant enrichment of Ruminococcus gnavus, the expression of certain virulent bacterial traits, and the detection of human inflammation-related proteins. Metaproteomics provides information on composition and functionality, as well as data on host-microbiome interactions. Its strength is the identification and quantification of Actinobacteria and certain classes of Firmicutes, but alpha diversity indices are not comparable to those of amplicon sequencing. Both techniques detected an aberrant microbiota in our small cohort of infants with CF during their first year of life, dominated by the enrichment of R. gnavus within a human inflammatory environment. IMPORTANCE In recent years, some techniques have been incorporated for the study of microbial ecosystems, being 16S rRNA gene sequencing being the most widely used. Metaproteomics provides the advantage of identifying the interaction between microorganisms and human cells, but the available databases are less extensive as well as imprecise. Few studies compare the statistical differences between the two techniques to define the composition of an ecosystem. Our work shows that the two methods are comparable in terms of microorganism identification but provide different results in alpha diversity analysis. On the other hand, we have studied newborns with cystic fibrosis, for whom we have described the establishment of an intestinal ecosystem marked by the inflammatory response of the host and the enrichment of Ruminococcus gnavus.
Collapse
Affiliation(s)
- Claudia Saralegui
- Servicio de Microbiología, Hospital Universitario Ramón y Cajal and Instituto Ramón y Cajal de Investigación Sanitaria, Madrid, Spain
- CIBERINFEC, Madrid, Spain
- Unidad de Fibrosis Quística, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Carmen García-Durán
- Departamento de Microbiología y Parasitología, Universidad Complutense de Madrid and Instituto Ramón y Cajal de Investigación Sanitaria, Madrid, Spain
| | - Eduardo Romeu
- Unidad de Proteómica, Universidad Complutense de Madrid, Madrid, Spain
| | | | - Ainhize Maruri
- Servicio de Microbiología, Hospital Universitario Ramón y Cajal and Instituto Ramón y Cajal de Investigación Sanitaria, Madrid, Spain
- CIBERINFEC, Madrid, Spain
- Unidad de Fibrosis Quística, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Natalia Bastón-Paz
- Servicio de Microbiología, Hospital Universitario Ramón y Cajal and Instituto Ramón y Cajal de Investigación Sanitaria, Madrid, Spain
- CIBERINFEC, Madrid, Spain
- Unidad de Fibrosis Quística, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Adelaida Lamas
- Unidad de Fibrosis Quística, Hospital Universitario Ramón y Cajal, Madrid, Spain
- Servicio de Pediatría, Hospital Universitario Ramón y Cajal and Instituto Ramón y Cajal de Investigación Sanitaria, Madrid, Spain
| | - Saioa Vicente
- Unidad de Fibrosis Quística, Hospital Universitario Ramón y Cajal, Madrid, Spain
- Servicio de Pediatría, Hospital Universitario Ramón y Cajal and Instituto Ramón y Cajal de Investigación Sanitaria, Madrid, Spain
| | - Estela Pérez-Ruiz
- Unidad de Fibrosis Quística, Hospital Regional Universitario de Málaga, Málaga, Spain
| | - Isabel Delgado
- Unidad de Fibrosis Quística, Hospital Virgen del Rocío, Seville, Spain
| | - Carmen Luna-Paredes
- Sección de Neumología y Alergia Infantil, Unidad Multidisciplinar Fibrosis Quística, Hospital Doce de Octubre, Madrid, Spain
| | - Juan de Dios Caballero
- Servicio de Microbiología, Hospital Universitario Ramón y Cajal and Instituto Ramón y Cajal de Investigación Sanitaria, Madrid, Spain
- CIBERINFEC, Madrid, Spain
- Unidad de Fibrosis Quística, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Javier Zamora
- Unidad de Bioestadística, Hospital Universitario Ramón y Cajal and Instituto Ramón y Cajal de Investigación Sanitaria and CIBERESP, Madrid, Spain
| | - Lucía Monteoliva
- Departamento de Microbiología y Parasitología, Universidad Complutense de Madrid and Instituto Ramón y Cajal de Investigación Sanitaria, Madrid, Spain
- Unidad de Proteómica, Universidad Complutense de Madrid, Madrid, Spain
| | - Concepción Gil
- Departamento de Microbiología y Parasitología, Universidad Complutense de Madrid and Instituto Ramón y Cajal de Investigación Sanitaria, Madrid, Spain
- Unidad de Proteómica, Universidad Complutense de Madrid, Madrid, Spain
| | - Rosa del Campo
- Servicio de Microbiología, Hospital Universitario Ramón y Cajal and Instituto Ramón y Cajal de Investigación Sanitaria, Madrid, Spain
- CIBERINFEC, Madrid, Spain
- Unidad de Fibrosis Quística, Hospital Universitario Ramón y Cajal, Madrid, Spain
- Universidad Alfonso X El Sabio, Madrid, Spain
| |
Collapse
|
38
|
He D, Liu L, Zhang Z, Yang X, Jia Y, Wen Y, Cheng S, Meng P, Li C, Zhang H, Pan C, Zhang F. Association between gut microbiota and longevity: a genetic correlation and mendelian randomization study. BMC Microbiol 2022; 22:302. [PMID: 36510142 PMCID: PMC9746102 DOI: 10.1186/s12866-022-02703-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 11/11/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Longevity is one of the most complex phenotypes, and its genetic basis remains unclear. This study aimed to explore the genetic correlation and potential causal association between gut microbiota and longevity. RESULTS Linkage disequilibrium score (LDSC) regression analysis and a bi-directional two-sample Mendelian Randomization (MR) analysis were performed to analyze gut microbiota and longevity-related traits. LDSC analysis detected four candidate genetic correlations, including Veillonella (genetic correlation = 0.5578, P = 4.67 × 10- 2) and Roseburia (genetic correlation = 0.4491, P = 2.67 × 10- 2) for longevity, Collinsella (genetic correlation = 0.3144, P = 4.07 × 10- 2) for parental lifespan and Sporobacter (genetic correlation = 0.2092, P = 3.53 × 10- 2) for healthspan. Further MR analysis observed suggestive causation between Collinsella and parental longevity (father's age at death) (weighted median: b = 1.79 × 10- 3, P = 3.52 × 10- 2). Reverse MR analysis also detected several causal effects of longevity-related traits on gut microbiota, such as longevity and Sporobacter (IVW: b = 7.02 × 10- 1, P = 4.21 × 10- 25). Statistical insignificance of the heterogeneity test and pleiotropy test supported the validity of the MR study. CONCLUSION Our study found evidence that gut microbiota is causally associated with longevity, or vice versa, providing novel clues for understanding the roles of gut microbiota in aging development.
Collapse
Affiliation(s)
- Dan He
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, Xi'an Jiaotong University, 710061, Xi'an, China
- Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education of China, Xi'an Jiaotong University, Xi'an, China
| | - Li Liu
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, Xi'an Jiaotong University, 710061, Xi'an, China
- Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education of China, Xi'an Jiaotong University, Xi'an, China
| | - Zhen Zhang
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, Xi'an Jiaotong University, 710061, Xi'an, China
- Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education of China, Xi'an Jiaotong University, Xi'an, China
| | - Xuena Yang
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, Xi'an Jiaotong University, 710061, Xi'an, China
- Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education of China, Xi'an Jiaotong University, Xi'an, China
| | - Yumeng Jia
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, Xi'an Jiaotong University, 710061, Xi'an, China
- Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education of China, Xi'an Jiaotong University, Xi'an, China
| | - Yan Wen
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, Xi'an Jiaotong University, 710061, Xi'an, China
- Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education of China, Xi'an Jiaotong University, Xi'an, China
| | - Shiqiang Cheng
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, Xi'an Jiaotong University, 710061, Xi'an, China
- Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education of China, Xi'an Jiaotong University, Xi'an, China
| | - Peilin Meng
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, Xi'an Jiaotong University, 710061, Xi'an, China
- Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education of China, Xi'an Jiaotong University, Xi'an, China
| | - Chun'e Li
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, Xi'an Jiaotong University, 710061, Xi'an, China
- Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education of China, Xi'an Jiaotong University, Xi'an, China
| | - Huijie Zhang
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, Xi'an Jiaotong University, 710061, Xi'an, China
- Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education of China, Xi'an Jiaotong University, Xi'an, China
| | - Chuyu Pan
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, Xi'an Jiaotong University, 710061, Xi'an, China
- Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education of China, Xi'an Jiaotong University, Xi'an, China
| | - Feng Zhang
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, Xi'an Jiaotong University, 710061, Xi'an, China.
- Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education of China, Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
39
|
Liang X, Fu Y, Cao WT, Wang Z, Zhang K, Jiang Z, Jia X, Liu CY, Lin HR, Zhong H, Miao Z, Gou W, Shuai M, Huang Y, Chen S, Zhang B, Chen YM, Zheng JS. Gut microbiome, cognitive function and brain structure: a multi-omics integration analysis. Transl Neurodegener 2022; 11:49. [PMID: 36376937 PMCID: PMC9661756 DOI: 10.1186/s40035-022-00323-z] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 11/01/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Microbiome-gut-brain axis may be involved in the progression of age-related cognitive impairment and relevant brain structure changes, but evidence from large human cohorts is lacking. This study was aimed to investigate the associations of gut microbiome with cognitive impairment and brain structure based on multi-omics from three independent populations. METHODS We included 1430 participants from the Guangzhou Nutrition and Health Study (GNHS) with both gut microbiome and cognitive assessment data available as a discovery cohort, of whom 272 individuals provided fecal samples twice before cognitive assessment. We selected 208 individuals with baseline microbiome data for brain magnetic resonance imaging during the follow-up visit. Fecal 16S rRNA and shotgun metagenomic sequencing, targeted serum metabolomics, and cytokine measurements were performed in the GNHS. The validation analyses were conducted in an Alzheimer's disease case-control study (replication study 1, n = 90) and another community-based cohort (replication study 2, n = 1300) with cross-sectional dataset. RESULTS We found protective associations of specific gut microbial genera (Odoribacter, Butyricimonas, and Bacteroides) with cognitive impairment in both the discovery cohort and the replication study 1. Result of Bacteroides was further validated in the replication study 2. Odoribacter was positively associated with hippocampal volume (β, 0.16; 95% CI 0.06-0.26, P = 0.002), which might be mediated by acetic acids. Increased intra-individual alterations in gut microbial composition were found in participants with cognitive impairment. We also identified several serum metabolites and inflammation-associated metagenomic species and pathways linked to impaired cognition. CONCLUSIONS Our findings reveal that specific gut microbial features are closely associated with cognitive impairment and decreased hippocampal volume, which may play an important role in dementia development.
Collapse
Affiliation(s)
- Xinxiu Liang
- College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
- School of Life Sciences, Westlake University, 18 Shilongshan Rd, Cloud Town, Hangzhou, 310024, China
| | - Yuanqing Fu
- School of Life Sciences, Westlake University, 18 Shilongshan Rd, Cloud Town, Hangzhou, 310024, China
| | - Wen-Ting Cao
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Medical Statistics and Epidemiology, School of Public Health, Sun Yat-Sen University, Guangzhou, 510080, China
- School of Public Health, Hainan Medical University, Haikou, 571199, China
| | - Zhihong Wang
- Chinese Center for Disease Control and Prevention, National Institute for Nutrition and Health, Beijing, 100050, China
- Key Laboratory of Trace Element Nutrition, National Health Commission, Beijing, 100050, China
| | - Ke Zhang
- School of Life Sciences, Westlake University, 18 Shilongshan Rd, Cloud Town, Hangzhou, 310024, China
| | - Zengliang Jiang
- School of Life Sciences, Westlake University, 18 Shilongshan Rd, Cloud Town, Hangzhou, 310024, China
- Westlake Intelligent Biomarker Discovery Lab, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, China
| | - Xiaofang Jia
- Chinese Center for Disease Control and Prevention, National Institute for Nutrition and Health, Beijing, 100050, China
- Key Laboratory of Trace Element Nutrition, National Health Commission, Beijing, 100050, China
| | - Chun-Ying Liu
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Medical Statistics and Epidemiology, School of Public Health, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Hong-Rou Lin
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Medical Statistics and Epidemiology, School of Public Health, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Haili Zhong
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Medical Statistics and Epidemiology, School of Public Health, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Zelei Miao
- School of Life Sciences, Westlake University, 18 Shilongshan Rd, Cloud Town, Hangzhou, 310024, China
| | - Wanglong Gou
- School of Life Sciences, Westlake University, 18 Shilongshan Rd, Cloud Town, Hangzhou, 310024, China
| | - Menglei Shuai
- School of Life Sciences, Westlake University, 18 Shilongshan Rd, Cloud Town, Hangzhou, 310024, China
| | - Yujing Huang
- School of Life Sciences, Westlake University, 18 Shilongshan Rd, Cloud Town, Hangzhou, 310024, China
| | - Shengdi Chen
- Department of Neurology and Institute of Neurology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Bing Zhang
- Chinese Center for Disease Control and Prevention, National Institute for Nutrition and Health, Beijing, 100050, China.
- Key Laboratory of Trace Element Nutrition, National Health Commission, Beijing, 100050, China.
| | - Yu-Ming Chen
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Medical Statistics and Epidemiology, School of Public Health, Sun Yat-Sen University, Guangzhou, 510080, China.
| | - Ju-Sheng Zheng
- School of Life Sciences, Westlake University, 18 Shilongshan Rd, Cloud Town, Hangzhou, 310024, China.
- Westlake Intelligent Biomarker Discovery Lab, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, China.
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, 310024, China.
| |
Collapse
|
40
|
Khan FH, Bhat BA, Sheikh BA, Tariq L, Padmanabhan R, Verma JP, Shukla AC, Dowlati A, Abbas A. Microbiome dysbiosis and epigenetic modulations in lung cancer: From pathogenesis to therapy. Semin Cancer Biol 2022; 86:732-742. [PMID: 34273520 DOI: 10.1016/j.semcancer.2021.07.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 06/25/2021] [Accepted: 07/11/2021] [Indexed: 02/07/2023]
Abstract
The lung microbiome plays an essential role in maintaining healthy lung function, including host immune homeostasis. Lung microbial dysbiosis or disruption of the gut-lung axis can contribute to lung carcinogenesis by causing DNA damage, inducing genomic instability, or altering the host's susceptibility to carcinogenic insults. Thus far, most studies have reported the association of microbial composition in lung cancer. Mechanistic studies describing host-microbe interactions in promoting lung carcinogenesis are limited. Considering cancer as a multifaceted disease where epigenetic dysregulation plays a critical role, epigenetic modifying potentials of microbial metabolites and toxins and their roles in lung tumorigenesis are not well studied. The current review explains microbial dysbiosis and epigenetic aberrations in lung cancer and potential therapeutic opportunities.
Collapse
Affiliation(s)
- Faizan Haider Khan
- Discipline of Pathology, Lambe Institute for Translational Research, School of Medicine, National University of Ireland Galway, Galway, Ireland
| | | | | | - Lubna Tariq
- Department of Biotechnology, Baba Ghulam Shah Badshah University, Rajouri, India
| | - Roshan Padmanabhan
- Department of Medicine, Case Western Reserve University, and University Hospital, Cleveland, OH, 44106, USA
| | - Jay Prakash Verma
- Institute of Environment and Sustainable Development, Banaras Hindu University Varanasi, India
| | | | - Afshin Dowlati
- Division of Hematology and Oncology, Department of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA; University Hospitals Seidman Cancer Center, Cleveland, OH, 44106, USA; Developmental Therapeutics Program, Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH, 44116, USA
| | - Ata Abbas
- Division of Hematology and Oncology, Department of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA; Developmental Therapeutics Program, Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH, 44116, USA.
| |
Collapse
|
41
|
Diener C, Dai CL, Wilmanski T, Baloni P, Smith B, Rappaport N, Hood L, Magis AT, Gibbons SM. Genome-microbiome interplay provides insight into the determinants of the human blood metabolome. Nat Metab 2022; 4:1560-1572. [PMID: 36357685 PMCID: PMC9691620 DOI: 10.1038/s42255-022-00670-1] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 09/30/2022] [Indexed: 11/12/2022]
Abstract
Variation in the blood metabolome is intimately related to human health. However, few details are known about the interplay between genetics and the microbiome in explaining this variation on a metabolite-by-metabolite level. Here, we perform analyses of variance for each of 930 blood metabolites robustly detected across a cohort of 1,569 individuals with paired genomic and microbiome data while controlling for a number of relevant covariates. We find that 595 (64%) of these blood metabolites are significantly associated with either host genetics or the gut microbiome, with 69% of these associations driven solely by the microbiome, 15% driven solely by genetics and 16% under hybrid genome-microbiome control. Additionally, interaction effects, where a metabolite-microbe association is specific to a particular genetic background, are quite common, albeit with modest effect sizes. This knowledge will help to guide targeted interventions designed to alter the composition of the human blood metabolome.
Collapse
Affiliation(s)
| | | | | | | | - Brett Smith
- Institute for Systems Biology, Seattle, WA, USA
| | | | - Leroy Hood
- Institute for Systems Biology, Seattle, WA, USA
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | | | - Sean M Gibbons
- Institute for Systems Biology, Seattle, WA, USA.
- Department of Bioengineering, University of Washington, Seattle, WA, USA.
- eScience Institute, University of Washington, Seattle, WA, USA.
| |
Collapse
|
42
|
Zeng S, Wang S, Ross RP, Stanton C. The road not taken: host genetics in shaping intergenerational microbiomes. Trends Genet 2022; 38:1180-1192. [PMID: 35773025 DOI: 10.1016/j.tig.2022.05.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 05/31/2022] [Accepted: 05/31/2022] [Indexed: 02/09/2023]
Abstract
The early-life gut microbiome is linked to human phenotypes as an imbalanced microbiome of this period is implicated in diseases throughout life. Several determinants of early-life gut microbiome are explored, however, mechanisms of acquisition, colonization, and stability of early-life gut microbiome and their interindividual variability remain elusive. Host genetics play a vital role to shape the gut microbiome and interact with it to modulate individual phenotypes in human studies and animal models. Given the microbial linkage between host generations, we discuss the current state of roles of host genetics in forming intergenerational microbiomes associated with mothers, offspring, and those vertically transmitted, providing a basis for taking into account host genetics in future early-life microbiome research. We further expand our discussion to the bidirectional interactions between host gene expression and microbiome in human health.
Collapse
Affiliation(s)
- Shuqin Zeng
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, 610041, China; APC Microbiome Ireland, University College Cork, Cork, T12 YT20, Ireland; Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork, P61 C996, Ireland
| | - Shaopu Wang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, 610041, China; APC Microbiome Ireland, University College Cork, Cork, T12 YT20, Ireland; Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork, P61 C996, Ireland.
| | - R Paul Ross
- APC Microbiome Ireland, University College Cork, Cork, T12 YT20, Ireland
| | - Catherine Stanton
- APC Microbiome Ireland, University College Cork, Cork, T12 YT20, Ireland; Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork, P61 C996, Ireland
| |
Collapse
|
43
|
Cheung MK, Yue GGL, Lauw S, Li CSY, Yung MY, Ng SC, Yip HC, Kwan HS, Chiu PWY, Lau CBS. Alterations in gut microbiota of esophageal squamous cell carcinoma patients. J Gastroenterol Hepatol 2022; 37:1919-1927. [PMID: 35816164 DOI: 10.1111/jgh.15941] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 06/21/2022] [Accepted: 07/05/2022] [Indexed: 12/09/2022]
Abstract
BACKGROUND AND AIM Esophageal squamous cell carcinoma (ESCC) is the most common histological subtype of esophageal cancer worldwide. Patients with ESCC display an altered esophageal microbiota compared with healthy individuals; however, little is known about the gut microbiota in ESCC. METHODS Here, we characterized the fecal microbiota of 15 ESCC patients and 16 healthy control subjects using 16S rRNA gene sequencing. RESULTS After controlling for potential confounders, significant alterations in both taxonomic and functional composition of the gut microbiota in ESCC patients were observed. By contrast, alpha diversity of the gut microbiota did not significantly differ between the cases and controls. We observed an enrichment of potentially pro-inflammatory and/or carcinogenic bacteria, such as Butyricimonas, Veillonella, and Streptococcus, and a depletion of butyrate-producing and/or potentially anti-inflammatory bacteria, such as Butyricicoccus, Lachnospiraceae NK4A136 group, and Eubacterium eligens group, in the gut microbiota of ESCC patients. The log-ratios of Streptococcus to Butyricicoccus and Streptococcus to Lachnospiraceae NK4A136 group of the gut microbiota were identified as potential diagnostic biomarkers for ESCC, with an area under the receiver operating characteristic (ROC) curve (AUC) of 0.863 (95% confidence interval: 0.707-1.000) and 0.825 (0.673-0.977), respectively. The diagnostic performance of both microbial biomarkers was validated in another ESCC cohort. CONCLUSIONS This pilot study has revealed an altered gut microbiota in ESCC patients and has paved the way for large-scale prospective cohort studies to examine the causative relationship between ESCC and gut dysbiosis.
Collapse
Affiliation(s)
- Man Kit Cheung
- Department of Surgery, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| | - Grace Gar Lee Yue
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong.,State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Susana Lauw
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| | - Cindy Shin Yee Li
- Department of Surgery, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| | - Man Yee Yung
- Department of Surgery, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| | - Siew Chien Ng
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| | - Hon Chi Yip
- Department of Surgery, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| | - Hoi Shan Kwan
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| | - Philip Wai Yan Chiu
- Department of Surgery, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| | - Clara Bik San Lau
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong.,State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Shatin, Hong Kong
| |
Collapse
|
44
|
Sun X, Yuan Q, Du B, Jin X, Huang X, Li Q, Zhong Y, Pan Z, Xu S, Sima Y. Relationship between Changes in Intestinal Microorganisms and Effect of High Temperature on the Growth and Development of Bombyx mori Larvae. Int J Mol Sci 2022; 23:10289. [PMID: 36142203 PMCID: PMC9499401 DOI: 10.3390/ijms231810289] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/28/2022] [Accepted: 09/03/2022] [Indexed: 12/02/2022] Open
Abstract
Temperature is an important environmental factor affecting the growth and development of silkworm (Bombyx mori). To analyze the effect of intestinal microbes on silkworm in response to a high-temperature environment, this study used a combination of high throughput sequencing and biochemical assays to detect silkworm intestinal microbes treated with high temperature for 72 h. The results show that high temperature affects the intestinal microbes of silkworm and that there are sex differences, specifically, females were more sensitive. The changes in the metabolism and transport ability of silkworm intestinal tissues under high temperature are related to the intestinal microbes. High temperatures may affect the intestinal microbes of silkworms, regulating the activity of related digestive enzymes and substance transport in the intestine, thereby affecting the silkworm's digestion and absorption of nutrients, and ultimately affecting growth and development.
Collapse
Affiliation(s)
- Xiaoning Sun
- School of Biology and Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou 215123, China
| | - Qian Yuan
- School of Biology and Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou 215123, China
| | - Beibei Du
- School of Biology and Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou 215123, China
| | - Xinye Jin
- School of Biology and Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou 215123, China
| | - Xiyun Huang
- School of Biology and Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou 215123, China
| | - Qiuying Li
- School of Biology and Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou 215123, China
| | - Yueqiao Zhong
- School of Biology and Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou 215123, China
| | - Zhonghua Pan
- School of Biology and Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou 215123, China
- Institute of Agricultural Biotechnology & Ecology (IABE), Soochow University, Suzhou 215123, China
| | - Shiqing Xu
- School of Biology and Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou 215123, China
- Institute of Agricultural Biotechnology & Ecology (IABE), Soochow University, Suzhou 215123, China
| | - Yanghu Sima
- School of Biology and Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou 215123, China
- Institute of Agricultural Biotechnology & Ecology (IABE), Soochow University, Suzhou 215123, China
| |
Collapse
|
45
|
Wrigley-Carr HE, van Dorst JM, Ooi CY. Intestinal dysbiosis and inflammation in cystic fibrosis impacts gut and multi-organ axes. MEDICINE IN MICROECOLOGY 2022. [DOI: 10.1016/j.medmic.2022.100057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
46
|
Crosstalk between mucosal microbiota, host gene expression, and sociomedical factors in the progression of colorectal cancer. Sci Rep 2022; 12:13447. [PMID: 35927305 PMCID: PMC9352898 DOI: 10.1038/s41598-022-17823-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 08/01/2022] [Indexed: 11/24/2022] Open
Abstract
Various omics-based biomarkers related to the occurrence, progression, and prognosis of colorectal cancer (CRC) have been identified. In this study, we attempted to identify gut microbiome-based biomarkers and detect their association with host gene expression in the initiation and progression of CRC by integrating analysis of the gut mucosal metagenome, RNA sequencing, and sociomedical factors. We performed metagenome and RNA sequencing on colonic mucosa samples from 13 patients with advanced CRC (ACRC), 10 patients with high-risk adenoma (HRA), and 7 normal control (NC) individuals. All participants completed a questionnaire on sociomedical factors. The interaction and correlation between changes in the microbiome and gene expression were assessed using bioinformatic analysis. When comparing HRA and NC samples, which can be considered to represent the process of tumor initiation, 28 genes and five microbiome species were analyzed with correlation plots. When comparing ACRC and HRA samples, which can be considered to represent the progression of CRC, seven bacterial species and 21 genes were analyzed. When comparing ACRC and NC samples, 16 genes and five bacterial species were analyzed, and four correlation plots were generated. A network visualizing the relationship between bacterial and host gene expression in the initiation and progression of CRC indicated that Clostridium spiroforme and Tyzzerella nexilis were hub bacteria in the development and progression of CRC. Our study revealed the interactions of and correlation between the colonic mucosal microbiome and host gene expression to identify potential roles of the microbiome in the initiation and progression of CRC. Our results provide gut microbiome-based biomarkers that may be potential diagnostic markers and therapeutic targets in patients with CRC.
Collapse
|
47
|
Wang Y, Lei X, Lu C, Pan Y. Predicting Microbe-Disease Association Based on Multiple Similarities and LINE Algorithm. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2022; 19:2399-2408. [PMID: 34014827 DOI: 10.1109/tcbb.2021.3082183] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Numerous microbes have been found to have vital impacts on human health through affecting biological processes. Therefore, exploring potential associations between microbes and diseases will promote the understanding and diagnosis of diseases. In this study, we present a novel computational model, named MSLINE, to infer potential microbe-disease associations by integrating Multiple Similarities and Large-scale Information Network Embedding (LINE) based on known associations. Specifically, on the basis of known microbe-disease associations from the Human Microbe-Disease Association Database, we first increase the known associations by collecting proven associations from existing literatures. We then construct a microbe-disease heterogeneous network (MDHN) by integrating known associations and multiple similarities (including Gaussian interaction profile kernel similarity, microbe function similarity, disease semantic similarity and disease-symptom similarity). After that, we implement random walk and LINE algorithm on MDHN to learn its structure information. Finally, we score the microbe-disease associations according to the structure information for every nodes. In the Leave-one-out cross validation and 5-fold cross validation, MSLINE performs better compared to other existing methods. Moreover, case studies of different diseases proved that MSLINE could predict the potential microbe-disease associations efficiently.
Collapse
|
48
|
Wu Y, Meng H, Qiao B, Li N, Zhang Q, Jia W, Xing H, Li Y, Yuan J, Yang Z. Yifei Sanjie Formula Treats Chronic Obstructive Pulmonary Disease by Remodeling Pulmonary Microbiota. Front Med (Lausanne) 2022; 9:927607. [PMID: 35847812 PMCID: PMC9277004 DOI: 10.3389/fmed.2022.927607] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 06/10/2022] [Indexed: 11/13/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is one of the most common pulmonary diseases. Evidence suggests that dysbiosis of pulmonary microbiota leads to the COPD pathological process. Yifei Sanjie Formula (YS) is widely used to treat diseases in respiratory systems, yet little is known about its mechanisms. In the present study, we first established the fingerprint of YS as the background for UHPLC-QTOF-MS. Components were detected, including alkaloids, amino acid derivatives, phenylpropanoids, flavonoids, terpenoids, organic acids, phenols, and the like. The therapeutic effect of YS on COPD was evaluated, and the pulmonary function and ventilatory dysfunction (EF50, TV, and MV) were improved after the administration of YS. Further, the influx of lymphocytes was inhibited in pulmonary parenchyma, accompanied by down-regulation of inflammation cytokines via the NLRP3/caspase-1/IL-1β signaling pathway. The severity of pulmonary pathological damage was reversed. Disturbed pulmonary microbiota was discovered to involve an increased relative abundance of Ralstonia and Mycoplasma and a decreased relative abundance of Lactobacillus and Bacteroides in COPD animals. However, the subversive effect was shown. The abundance and diversity of pulmonary microflora were remodeled, especially increasing beneficial genua Lactobacillus and Bacteroides, as well as downregulating pathogenic genua Ralstonia and Mycoplasma in the YS group. Environmental factor correlation analysis showed that growing pulmonary microbiota was positively correlated with the inflammatory factor, referring to Ralstonia and Mycoplasma, as well as negatively correlated with the inflammatory factor, referring to Lactobacillus and Bacteroides. These results suggest that the effects of YS involved remodeling lung microbes and anti-inflammatory signal pathways, revealing that intervention microbiota and an anti-inflammatory may be a potential therapeutic strategy for COPD.
Collapse
Affiliation(s)
- Yueying Wu
- Yunnan Provincial Key Laboratory of Molecular Biology for Sinomedicine, Kunming, China
- First Clinical School of Medicine, Yunnan University of Chinese Medicine, Kunming, China
| | - Hui Meng
- Yunnan Provincial Key Laboratory of Molecular Biology for Sinomedicine, Kunming, China
| | - Bo Qiao
- Yunnan Provincial Key Laboratory of Molecular Biology for Sinomedicine, Kunming, China
| | - Ning Li
- Yunnan Provincial Key Laboratory of Molecular Biology for Sinomedicine, Kunming, China
- First Clinical School of Medicine, Yunnan University of Chinese Medicine, Kunming, China
| | - Qiang Zhang
- Basic Medical School, Shanghai University of Chinese Medicine, Shanghai, China
| | - Wenqing Jia
- Yunnan Provincial Key Laboratory of Molecular Biology for Sinomedicine, Kunming, China
| | - Haijing Xing
- Yunnan Provincial Key Laboratory of Molecular Biology for Sinomedicine, Kunming, China
| | - Yuqing Li
- Yunnan Provincial Key Laboratory of Molecular Biology for Sinomedicine, Kunming, China
| | - Jiali Yuan
- Yunnan Provincial Key Laboratory of Molecular Biology for Sinomedicine, Kunming, China
- Jiali Yuan
| | - Zhongshan Yang
- Yunnan Provincial Key Laboratory of Molecular Biology for Sinomedicine, Kunming, China
- *Correspondence: Zhongshan Yang
| |
Collapse
|
49
|
Priya S, Burns MB, Ward T, Mars RAT, Adamowicz B, Lock EF, Kashyap PC, Knights D, Blekhman R. Identification of shared and disease-specific host gene-microbiome associations across human diseases using multi-omic integration. Nat Microbiol 2022; 7:780-795. [PMID: 35577971 PMCID: PMC9159953 DOI: 10.1038/s41564-022-01121-z] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 04/06/2022] [Indexed: 12/19/2022]
Abstract
While gut microbiome and host gene regulation independently contribute to gastrointestinal disorders, it is unclear how the two may interact to influence host pathophysiology. Here we developed a machine learning-based framework to jointly analyse paired host transcriptomic (n = 208) and gut microbiome (n = 208) profiles from colonic mucosal samples of patients with colorectal cancer, inflammatory bowel disease and irritable bowel syndrome. We identified associations between gut microbes and host genes that depict shared as well as disease-specific patterns. We found that a common set of host genes and pathways implicated in gastrointestinal inflammation, gut barrier protection and energy metabolism are associated with disease-specific gut microbes. Additionally, we also found that mucosal gut microbes that have been implicated in all three diseases, such as Streptococcus, are associated with different host pathways in each disease, suggesting that similar microbes can affect host pathophysiology in a disease-specific manner through regulation of different host genes. Our framework can be applied to other diseases for the identification of host gene-microbiome associations that may influence disease outcomes.
Collapse
Affiliation(s)
- Sambhawa Priya
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN, USA
- Bioinformatics and Computational Biology, University of Minnesota, Minneapolis, MN, USA
| | - Michael B Burns
- Department of Biology, Loyola University Chicago, Chicago, IL, USA
| | - Tonya Ward
- BioTechnology Institute, College of Biological Sciences, University of Minnesota, Minneapolis, MN, USA
| | - Ruben A T Mars
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Beth Adamowicz
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN, USA
| | - Eric F Lock
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - Purna C Kashyap
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Dan Knights
- BioTechnology Institute, College of Biological Sciences, University of Minnesota, Minneapolis, MN, USA
- Department of Computer Science and Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Ran Blekhman
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN, USA.
- Department of Ecology, Evolution, and Behavior, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
50
|
Changing paradigms in the treatment of gastrointestinal complications of cystic fibrosis in the era of cystic fibrosis transmembrane conductance regulator modulators. Paediatr Respir Rev 2022; 42:9-16. [PMID: 33485777 DOI: 10.1016/j.prrv.2020.12.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 12/17/2020] [Indexed: 12/17/2022]
Abstract
Cystic fibrosis (CF) - although primarily a lung disease - also causes a variety of gastrointestinal manifestations which are important for diagnosis, prognosis and quality of life. All parts of the gastrointestinal tract can be affected by CF. Besides the well-known pancreatic insufficiency, gastroesophageal reflux disease, liver disease and diseases of the large intestine are important pathologies that impact on prognosis and also impair quality of life. Diagnosis and management of gastrointestinal manifestations will be discussed in this review. Since optimisation of CF therapy is associated with a significantly longer life-span of CF patients nowadays, also gastrointestinal malignancies, which are more common in CF than in the non-CF population need to be considered. Furthermore, novel evidence on the role of the gut microbiome in CF is emerging. The introduction of cystic fibrosis transmembrane conductance regulator (CFTR) protein modulators gives hope for symptom alleviation and even cure of gastrointestinal manifestations of CF.
Collapse
|