101
|
McGrattan A, Stewart CJ, Cassidy A, Woodside JV, McEvoy CT. Diet Patterns, the Gut Microbiome, and Alzheimer's Disease. J Alzheimers Dis 2022; 88:933-941. [PMID: 35634849 DOI: 10.3233/jad-220205] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Given the complex bidirectional communication system that exists between the gut microbiome and the brain, there is growing interest in the gut microbiome as a novel and potentially modifiable risk factor for Alzheimer's disease (AD). Gut dysbiosis has been implicated in the pathogenesis and progression of AD by initiating and prolonging neuroinflammatory processes. The metabolites of gut microbiota appear to be critical in the mechanism of the gut-brain axis. Gut microbiota metabolites, such as trimethylamine-n-oxide, lipopolysaccharide, and short chain fatty acids, are suggested to mediate systemic inflammation and intracerebral amyloidosis via endothelial dysfunction. Emerging data suggest that the fungal microbiota (mycobiome) may also influence AD pathology. Importantly, 60% of variation in the gut microbiome is attributable to diet, therefore modulating the gut microbiome through dietary means could be an effective approach to reduce AD risk. Given that people do not eat isolated nutrients and instead consume a diverse range of foods and combinations of nutrients that are likely to be interactive, studying the effects of whole diets provides the opportunity to account for the interactions between different nutrients. Thus, dietary patterns may be more predictive of real-life effect on gut microbiome and AD risk than foods or nutrients in isolation. Accumulating evidence from experimental and animal studies also show potential effects of gut microbiome on AD pathogenesis. However, data from human dietary interventions are lacking. Well-designed intervention studies are needed in diverse populations to determine the influence of diet on gut microbiome and inform the development of effective dietary strategies for prevention of AD.
Collapse
Affiliation(s)
- Andrea McGrattan
- School of Biomedical, Nutritional and Sport Sciences, Newcastle University, UK
| | | | - Aedín Cassidy
- Institute For Global Food Security, Queen's University Belfast, UK
| | - Jayne V Woodside
- Institute For Global Food Security, Queen's University Belfast, UK
| | - Claire T McEvoy
- Institute For Global Food Security, Queen's University Belfast, UK.,Global Brain Health Institute, University of California San Francisco, USA and Trinity College Dublin, Ireland
| |
Collapse
|
102
|
Bouyahya A, Omari NE, EL Hachlafi N, Jemly ME, Hakkour M, Balahbib A, El Menyiy N, Bakrim S, Naceiri Mrabti H, Khouchlaa A, Mahomoodally MF, Catauro M, Montesano D, Zengin G. Chemical Compounds of Berry-Derived Polyphenols and Their Effects on Gut Microbiota, Inflammation, and Cancer. Molecules 2022; 27:3286. [PMID: 35630763 PMCID: PMC9146061 DOI: 10.3390/molecules27103286] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/27/2022] [Accepted: 05/08/2022] [Indexed: 12/15/2022] Open
Abstract
Berry-derived polyphenols are bioactive compounds synthesized and secreted by several berry fruits. These polyphenols feature a diversity of chemical compounds, including phenolic acids and flavonoids. Here, we report the beneficial health effects of berry-derived polyphenols and their therapeutical application on gut-microbiota-related diseases, including inflammation and cancer. Pharmacokinetic investigations have confirmed the absorption, availability, and metabolism of berry-derived polyphenols. In vitro and in vivo tests, as well as clinical trials, showed that berry-derived polyphenols can positively modulate the gut microbiota, inhibiting inflammation and cancer development. Indeed, these compounds inhibit the growth of pathogenic bacteria and also promote beneficial bacteria. Moreover, berry-derived polyphenols exhibit therapeutic effects against different gut-microbiota-related disorders such as inflammation, cancer, and metabolic disorders. Moreover, these polyphenols can manage the inflammation via various mechanisms, in particular the inhibition of the transcriptional factor Nf-κB. Berry-derived polyphenols have also shown remarkable effects on different types of cancer, including colorectal, breast, esophageal, and prostate cancer. Moreover, certain metabolic disorders such as diabetes and atherosclerosis were also managed by berry-derived polyphenols through different mechanisms. These data showed that polyphenols from berries are a promising source of bioactive compounds capable of modulating the intestinal microbiota, and therefore managing cancer and associated metabolic diseases. However, further investigations should be carried out to determine the mechanisms of action of berry-derived polyphenol bioactive compounds to validate their safety and examinate their clinical uses.
Collapse
Affiliation(s)
- Abdelhakim Bouyahya
- Laboratory of Human Pathologies Biology, Department of Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat 10106, Morocco
| | - Nasreddine El Omari
- Laboratory of Histology, Embryology, and Cytogenetic, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Rabat 10100, Morocco;
| | - Naoufal EL Hachlafi
- Microbial Biotechnology and Bioactive Molecules Laboratory, Sciences and Technologies Faculty, Sidi Mohmed Ben Abdellah University, Imouzzer Road Fez, Fez 30003, Morocco;
| | - Meryem El Jemly
- Faculty of Pharmacy, University Mohammed VI for Health Science, Casablanca 82403, Morocco;
| | - Maryam Hakkour
- Laboratory of Biodiversity, Ecology, and Genome, Faculty of Sciences, Mohammed V University in Rabat, Rabat 10106, Morocco; (M.H.); (A.B.)
| | - Abdelaali Balahbib
- Laboratory of Biodiversity, Ecology, and Genome, Faculty of Sciences, Mohammed V University in Rabat, Rabat 10106, Morocco; (M.H.); (A.B.)
| | - Naoual El Menyiy
- Laboratory of Pharmacology, National Agency of Medicinal and Aromatic Plants, Taounate 34025, Morocco;
| | - Saad Bakrim
- Molecular Engineering, Valorization and Environment Team, Polydisciplinary Faculty of Taroudant, Ibn Zohr University, Agadir 80000, Morocco;
| | - Hanae Naceiri Mrabti
- Laboratory of Pharmacology and Toxicology, Bio Pharmaceutical and Toxicological Analysis Research Team, Faculty of Medicine and Pharmacy, Mohammed V University, Rabat 10000, Morocco;
| | - Aya Khouchlaa
- Laboratory of Biochemistry, National Agency of Medicinal and Aromatic Plants, Taounate 34025, Morocco;
| | - Mohamad Fawzi Mahomoodally
- Department of Health Sciences, Faculty of Medicine and Health Sciences, University of Mauritius, Reduit 80837, Mauritius;
| | - Michelina Catauro
- Department of Engineering, University of Campania “Luigi Vanvitelli”, Via Roma 29, 81031 Aversa, Italy
| | - Domenico Montesano
- Department of Pharmacy, University of Naples Federico II, Via D. Montesano 49, 80131 Naples, Italy;
| | - Gokhan Zengin
- Physiology and Biochemistry Research Laboratory, Department of Biology, Science Faculty, Selcuk University, 42130 Konya, Turkey
| |
Collapse
|
103
|
Abstract
Inflammatory bowel diseases (IBD), namely, Crohn's disease (CD) and ulcerative colitis (UC), are lifelong and incurable chronic inflammatory diseases affecting 6.8 million people worldwide. By 2030, the prevalence of IBD is estimated to reach 1% of the population in Western countries, and thus there is an urgent need to develop effective therapies to reduce the burden of this disease. Microbiome dysbiosis is at the heart of the IBD pathophysiology, and current research and development efforts for IBD treatments have been focused on gut microbiome regulation. Diet can shape the intestinal microbiome. Diet is also preferred over medication, is safe, and has been proven to be an effective strategy for the management of IBD. Therefore, although often overlooked, dietary interventions targeting the microbiome represent ideal treatments for IBD. Here, I summarize the latest research on diet as a treatment for IBD from infancy to adulthood, compile evidence of the mechanisms of action behind diet as treatment, and, lastly, provide insights into future research focusing on culturally tailored diets for ethnic minority groups with increased incidence of IBD yet underrepresented in nutrition research.
Collapse
Affiliation(s)
- Ana Maldonado-Contreras
- University of Massachusetts Chan Medical School, Department of Microbiology and Physiological Systems, Program of Microbiome Dynamics, Worcester, Massachusetts, USA
| |
Collapse
|
104
|
Environmental Factors and the Risk of Developing Type 1 Diabetes-Old Disease and New Data. BIOLOGY 2022; 11:biology11040608. [PMID: 35453807 PMCID: PMC9027552 DOI: 10.3390/biology11040608] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 04/10/2022] [Accepted: 04/14/2022] [Indexed: 12/16/2022]
Abstract
Simple Summary Despite many studies, the risk factors of type 1 diabetes (T1DM) in children and adolescents are still not fully understood and remain a big challenge. Therefore, an extensive online search for scientific research on factors related to diabetes has been performed for the identification of new factors of unexplained etiology. A better understanding of the role of viral, bacterial, and yeast-like fungi infections related to the risk of T1DM in children and adolescents and the identification of new risk factors, especially those spread by the droplet route, is of great importance for people and families with diabetes. Abstract The incidence of type 1 diabetes (T1D) is increasing worldwide. The onset of T1D usually occurs in childhood and is caused by the selective destruction of insulin-producing pancreatic islet cells (β-cells) by autoreactive T cells, leading to insulin deficiency. Despite advanced research and enormous progress in medicine, the causes of T1D are still not fully understood. Therefore, an extensive online search for scientific research on environmental factors associated with diabetes and the identification of new factors of unexplained etiology has been carried out using the PubMed, Cochrane, and Embase databases. The search results were limited to the past 11 years of research and discovered 143 manuscripts published between 2011 and 2022. Additionally, 21 manuscripts from between 2000 and 2010 and 3 manuscripts from 1974 to 2000 were referenced for historical reference as the first studies showcasing a certain phenomenon or mechanism. More and more scientists are inclined to believe that environmental factors are responsible for the increased incidence of diabetes. Research results show that higher T1D incidence is associated with vitamin D deficiency, a colder climate, and pollution of the environment, as well as the influence of viral, bacterial, and yeast-like fungi infections. The key viral infections affecting the risk of developing T1DM are rubella virus, mumps virus, Coxsackie virus, cytomegalovirus, and enterovirus. Since 2020, i.e., from the beginning of the COVID-19 pandemic, more and more studies have been looking for a link between Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) and diabetes development. A better understanding of the role of viral, bacterial, and yeast-like fungi infections related to the risk of T1DM in children and adolescents and the identification of new risk factors, especially those spread by the droplet route, is of great importance for people and families with diabetes.
Collapse
|
105
|
Kheirandish P, Petri RM, Sener-Aydemir A, Schwartz-Zimmermann HE, Berthiller F, Zebeli Q, Pacífico C. Characterization of Microbial Intolerances and Ruminal Dysbiosis Towards Different Dietary Carbohydrate Sources Using an in vitro Model. J Appl Microbiol 2022; 133:458-476. [PMID: 35396778 PMCID: PMC9545568 DOI: 10.1111/jam.15573] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 03/15/2022] [Accepted: 04/01/2022] [Indexed: 11/30/2022]
Abstract
AIM This study aimed to characterize the critical points for determining the development of dysbiosis associated with feed intolerances and ruminal acidosis. METHODS AND RESULTS A metabologenomics approach was used to characterize dynamic microbial and metabolomics shifts using the rumen simulation technique (RUSITEC) by feeding native cornstarch (ST), chemically-modified cornstarch (CMS), or sucrose (SU). SU and CMS elicited the most drastic changes as rapidly as 4 h after feeding. This was accompanied by a swift accumulation of D-lactate, and the decline of benzoic and malonic acid. A consistent increase in Bifidobacterium and Lactobacillus as well as a decrease in fibrolytic bacteria was observed for both CMS and ST after 24 h, indicating intolerances within the fiber degrading populations. However, an increase in Lactobacillus was already evident in SU after 8 h. An inverse relationship between Fibrobacter and Bifidobacterium was observed in ST. In fact, Fibrobacter was positively correlated with several short-chain fatty acids (SCFA), while Lactobacillus was positively correlated with lactic acid, hexoses, hexose-phosphates, pentose phosphate pathway (PENTOSE-P-PWY) and heterolactic fermentation (P122-PWY). CONCLUSIONS The feeding of sucrose and modified starches, followed by native cornstarch, had a strong disruptive effect in the ruminal microbial community. Feed intolerances were shown to develop at different rates based on the availability of glucose for ruminal microorganisms. SIGNIFICANCE OF THE STUDY These results can be used to establish patterns of early dysbiosis (biomarkers) and develop strategies for preventing undesirable shifts in the ruminal microbial ecosystem.
Collapse
Affiliation(s)
- Parisa Kheirandish
- Christian Doppler Laboratory for Innovative Gut Health Concepts of Livestock, Institute of Animal Nutrition and Functional Plant Compounds, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine, Vienna, Austria
| | - Renee Maxine Petri
- Agriculture and Agri-Food Canada, Sherbrooke Research and Development Centre, Sherbrooke, Canada
| | - Arife Sener-Aydemir
- Christian Doppler Laboratory for Innovative Gut Health Concepts of Livestock, Institute of Animal Nutrition and Functional Plant Compounds, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine, Vienna, Austria
| | - Heidi Elisabeth Schwartz-Zimmermann
- Christian Doppler Laboratory for Innovative Gut Health Concepts of Livestock, Institute of Bioanalytics and Agro-Metabolomics, Department of Agrobiotechnology (IFA-Tulln), University of Natural Resources and Life Sciences, Vienna, Austria
| | - Franz Berthiller
- Christian Doppler Laboratory for Innovative Gut Health Concepts of Livestock, Institute of Bioanalytics and Agro-Metabolomics, Department of Agrobiotechnology (IFA-Tulln), University of Natural Resources and Life Sciences, Vienna, Austria
| | - Qendrim Zebeli
- Christian Doppler Laboratory for Innovative Gut Health Concepts of Livestock, Institute of Animal Nutrition and Functional Plant Compounds, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine, Vienna, Austria
| | - Cátia Pacífico
- Christian Doppler Laboratory for Innovative Gut Health Concepts of Livestock, Institute of Animal Nutrition and Functional Plant Compounds, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine, Vienna, Austria
| |
Collapse
|
106
|
Hu C, Patil Y, Gong D, Yu T, Li J, Wu L, Liu X, Yu Z, Ma X, Yong Y, Chen J, Gooneratne R, Ju X. Heat Stress-Induced Dysbiosis of Porcine Colon Microbiota Plays a Role in Intestinal Damage: A Fecal Microbiota Profile. Front Vet Sci 2022; 9:686902. [PMID: 35300220 PMCID: PMC8921775 DOI: 10.3389/fvets.2022.686902] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 01/24/2022] [Indexed: 12/30/2022] Open
Abstract
The pathological mechanisms of gastrointestinal disorders, including inflammatory bowel disease (IBD), in pigs are poorly understood. We report the induction of intestinal inflammation in heat-stressed (HS) pigs, fecal microbiota transplantation from pigs to mice, and explain the role of microorganisms in IBD. 24 adult pigs were subjected to HS (34 ± 1 °C; 75–85% relative humidity for 24h) while 24 control pigs (CP) were kept at 25 ± 3°C and the same humidity. Pigs were sacrificed on days 1, 7, 14, 21. Colonic content microbiome analyses were conducted. Pseudo-germ-free mice were fed by gavage with fecal microbiota from HS-pigs and CP to induce pig-like responses in mice. From 7 d, HS-pigs exhibited fever and diarrhea, and significantly lower colonic mucosal thickness, crypt depth/width, and goblet cell number. Compared with each control group, the concentration of cortisol in the peripheral blood of HS pigs gradually increased, significantly so on days 7, 14, and 21 (P < 0.01). While the concentration of LPS in HS pigs' peripheral blood was significantly higher on days 7, 14 (P < 0.01), and 21 (P < 0.05) compared with that of the control group. The colonic microbiome composition of HS-pigs was different to that of CP. By day 14, opportunistic pathogens (e.g., Campylobacterales) had increased in HS-pigs. The composition of the colonic microbiome in mice administered feces from HS-pigs was different from those receiving CP feces. Bacteroides were significantly diminished, Akkermansia were significantly increased, and intestinal damage and goblet cell numbers were higher in mice that received HS-pig feces. Moreover, we verified the relevance of differences in the microbiota of the colon among treatments. Heat stress promotes changes in gut microbiome composition, which can affect the colonic microbial structure of mice through fecal microbiota transplantation; the molecular mechanisms require further investigation. This study enhanced our understanding of stress-induced inflammation in the colon and the increase in diarrhea in mammals subjected to prolonged HS. Our results provide useful information for preventing or ameliorating deficits in pig production caused by prolonged exposure to high temperatures.
Collapse
Affiliation(s)
- Canying Hu
- Department of Animal Science, Agricultural College, Guangdong Ocean University, Zhanjiang, China
- Shenzhen Institute of Guangdong Ocean University, Shenzhen, China
| | - Yadnyavalkya Patil
- Department of Wine, Food and Molecular Biosciences, Faculty of Agriculture and Life Sciences, Lincoln University, Lincoln, New Zealand
| | - Dongliang Gong
- Department of Animal Science, Agricultural College, Guangdong Ocean University, Zhanjiang, China
| | - Tianyue Yu
- Department of Animal Science, Agricultural College, Guangdong Ocean University, Zhanjiang, China
| | - Junyu Li
- Department of Veterinary Medicine, College of Agriculture, Guangdong Ocean University, Zhanjiang, China
| | - Lianyun Wu
- Department of Animal Science, Agricultural College, Guangdong Ocean University, Zhanjiang, China
| | - Xiaoxi Liu
- Department of Animal Science, Agricultural College, Guangdong Ocean University, Zhanjiang, China
| | - Zhichao Yu
- Department of Animal Science, Agricultural College, Guangdong Ocean University, Zhanjiang, China
| | - Xinbing Ma
- Department of Animal Science, Agricultural College, Guangdong Ocean University, Zhanjiang, China
| | - Yanhong Yong
- Department of Veterinary Medicine, College of Agriculture, Guangdong Ocean University, Zhanjiang, China
| | - Jinjun Chen
- Department of Animal Science, Agricultural College, Guangdong Ocean University, Zhanjiang, China
| | - Ravi Gooneratne
- Department of Wine, Food and Molecular Biosciences, Faculty of Agriculture and Life Sciences, Lincoln University, Lincoln, New Zealand
| | - Xianghong Ju
- Department of Animal Science, Agricultural College, Guangdong Ocean University, Zhanjiang, China
- Shenzhen Institute of Guangdong Ocean University, Shenzhen, China
- Department of Veterinary Medicine, College of Agriculture, Guangdong Ocean University, Zhanjiang, China
- *Correspondence: Xianghong Ju
| |
Collapse
|
107
|
Abstract
BACKGROUND Obesity develops due to an imbalance in energy homeostasis, wherein energy intake exceeds energy expenditure. Accumulating evidence shows that manipulations of dietary protein and their component amino acids affect the energy balance, resulting in changes in fat mass and body weight. Amino acids are not only the building blocks of proteins but also serve as signals regulating multiple biological pathways. SCOPE OF REVIEW We present the currently available evidence regarding the effects of dietary alterations of a single essential amino acid (EAA) on energy balance and relevant signaling mechanisms at both central and peripheral levels. We summarize the association between EAAs and obesity in humans and the clinical use of modifying the dietary EAA composition for therapeutic intervention in obesity. Finally, similar mechanisms underlying diets varying in protein levels and diets altered of a single EAA are described. The current review would expand our understanding of the contribution of protein and amino acids to energy balance control, thus helping discover novel therapeutic approaches for obesity and related diseases. MAJOR CONCLUSIONS Changes in circulating EAA levels, particularly increased branched-chain amino acids (BCAAs), have been reported in obese human and animal models. Alterations in dietary EAA intake result in improvements in fat and weight loss in rodents, and each has its distinct mechanism. For example, leucine deprivation increases energy expenditure, reduces food intake and fat mass, primarily through regulation of the general control nonderepressible 2 (GCN2) and mammalian target of rapamycin (mTOR) signaling. Methionine restriction by 80% decreases fat mass and body weight while developing hyperphagia, primarily through fibroblast growth factor 21 (FGF-21) signaling. Some effects of diets with different protein levels on energy homeostasis are mediated by similar mechanisms. However, reports on the effects and underlying mechanisms of dietary EAA imbalances on human body weight are few, and more investigations are needed in future.
Collapse
Affiliation(s)
- Fei Xiao
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Innovation Center for Intervention of Chronic Disease and Promotion of Health, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, China
| | - Feifan Guo
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Innovation Center for Intervention of Chronic Disease and Promotion of Health, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, China; Shanghai Jiao Tong University Affiliated Sixth People's Hospital, China.
| |
Collapse
|
108
|
Foolchand A, Ghazi T, Chuturgoon AA. Malnutrition and Dietary Habits Alter the Immune System Which May Consequently Influence SARS-CoV-2 Virulence: A Review. Int J Mol Sci 2022; 23:2654. [PMID: 35269795 PMCID: PMC8910702 DOI: 10.3390/ijms23052654] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/22/2022] [Accepted: 02/23/2022] [Indexed: 02/06/2023] Open
Abstract
COVID-19, resulting from the SARS-CoV-2 virus, is a major pandemic that the world is fighting. SARS-CoV-2 primarily causes lung infection by attaching to the ACE2 receptor on the alveolar epithelial cells. However, the ACE2 receptor is also present in intestinal epithelial cells, suggesting a link between nutrition, virulence and clinical outcomes of COVID-19. Respiratory viral infections perturb the gut microbiota. The gut microbiota is shaped by our diet; therefore, a healthy gut is important for optimal metabolism, immunology and protection of the host. Malnutrition causes diverse changes in the immune system by repressing immune responses and enhancing viral vulnerability. Thus, improving gut health with a high-quality, nutrient-filled diet will improve immunity against infections and diseases. This review emphasizes the significance of dietary choices and its subsequent effects on the immune system, which may potentially impact SARS-CoV-2 vulnerability.
Collapse
Affiliation(s)
| | | | - Anil A. Chuturgoon
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, College of Health Sciences, Howard College Campus, University of Kwa-Zulu Natal, Durban 4041, South Africa; (A.F.); (T.G.)
| |
Collapse
|
109
|
Wu H, Norton V, Cui K, Zhu B, Bhattacharjee S, Lu YW, Wang B, Shan D, Wong S, Dong Y, Chan SL, Cowan D, Xu J, Bielenberg DR, Zhou C, Chen H. Diabetes and Its Cardiovascular Complications: Comprehensive Network and Systematic Analyses. Front Cardiovasc Med 2022; 9:841928. [PMID: 35252405 PMCID: PMC8891533 DOI: 10.3389/fcvm.2022.841928] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 01/18/2022] [Indexed: 12/12/2022] Open
Abstract
Diabetes mellitus is a worldwide health problem that usually comes with severe complications. There is no cure for diabetes yet and the threat of these complications is what keeps researchers investigating mechanisms and treatments for diabetes mellitus. Due to advancements in genomics, epigenomics, proteomics, and single-cell multiomics research, considerable progress has been made toward understanding the mechanisms of diabetes mellitus. In addition, investigation of the association between diabetes and other physiological systems revealed potentially novel pathways and targets involved in the initiation and progress of diabetes. This review focuses on current advancements in studying the mechanisms of diabetes by using genomic, epigenomic, proteomic, and single-cell multiomic analysis methods. It will also focus on recent findings pertaining to the relationship between diabetes and other biological processes, and new findings on the contribution of diabetes to several pathological conditions.
Collapse
Affiliation(s)
- Hao Wu
- Department of Surgery, Vascular Biology Program, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States
| | - Vikram Norton
- Department of Surgery, Vascular Biology Program, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States
| | - Kui Cui
- Department of Surgery, Vascular Biology Program, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States
| | - Bo Zhu
- Department of Surgery, Vascular Biology Program, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States
| | - Sudarshan Bhattacharjee
- Department of Surgery, Vascular Biology Program, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States
| | - Yao Wei Lu
- Department of Surgery, Vascular Biology Program, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States
| | - Beibei Wang
- Department of Surgery, Vascular Biology Program, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States
| | - Dan Shan
- Department of Surgery, Vascular Biology Program, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States
| | - Scott Wong
- Department of Surgery, Vascular Biology Program, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States
| | - Yunzhou Dong
- Department of Surgery, Vascular Biology Program, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States
| | - Siu-Lung Chan
- Department of Surgery, Vascular Biology Program, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States
| | - Douglas Cowan
- Department of Surgery, Vascular Biology Program, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States
| | - Jian Xu
- Department of Medicine, Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma, OK, United States
| | - Diane R Bielenberg
- Department of Surgery, Vascular Biology Program, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States
| | - Changcheng Zhou
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, United States
| | - Hong Chen
- Department of Surgery, Vascular Biology Program, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States
| |
Collapse
|
110
|
Chronic Inflammation as the Underlying Mechanism of the Development of Lung Diseases in Psoriasis: A Systematic Review. Int J Mol Sci 2022; 23:ijms23031767. [PMID: 35163689 PMCID: PMC8836589 DOI: 10.3390/ijms23031767] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 01/30/2022] [Accepted: 02/02/2022] [Indexed: 01/04/2023] Open
Abstract
Psoriasis is a systemic inflammatory disease caused by dysfunctional interactions between the innate and adaptive immune responses. The systemic inflammation in psoriasis may be associated with the development of comorbidities, including lung diseases. In this review, we aimed to provide a summary of the evidence regarding the prevalence of lung diseases in patients with psoriasis and the potential underlying mechanisms. Twenty-three articles published between March 2010 and June 2021 were selected from 195 initially identified records. The findings are discussed in terms of the prevalence of asthma, chronic obstructive pulmonary disease, interstitial lung disease, obstructive sleep apnea, pulmonary hypertension, and sarcoidosis in psoriasis. A higher prevalence of lung diseases in psoriasis has been confirmed in asthma, chronic obstructive pulmonary disease, obstructive sleep apnea, and pulmonary hypertension. These conditions are important as they are previously unrecognized causes of morbidity and mortality in psoriasis. The development of lung diseases in patients with psoriasis can be explained by several mechanisms, including common risk factors, shared immune and molecular characteristics associated with chronic inflammation, as well as other mechanisms. Understanding the prevalence of lung diseases in psoriasis and their underlying mechanisms can help implement appropriate preventative and therapeutic strategies to address respiratory diseases in patients with psoriasis.
Collapse
|
111
|
Vetuschi A, Battista N, Pompili S, Cappariello A, Prete R, Taticchi A, Selvaggini R, Latella G G, Corsetti A, Sferra R. The antiinflammatory and antifibrotic effect of olive phenols and Lactiplantibacillus plantarum IMC513 in dextran sodium sulfate-induced chronic colitis. Nutrition 2022; 94:111511. [PMID: 34813981 DOI: 10.1016/j.nut.2021.111511] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 09/17/2021] [Accepted: 10/03/2021] [Indexed: 12/19/2022]
Abstract
OBJECTIVES After a chronic intestinal injury, several intestinal cells switch their phenotype to activated myofibroblasts, which in turn release an abnormal amount of extracellular matrix proteins, leading to the onset of the fibrotic process. To date, no resolutive pharmacological treatments are available, and the identification of new therapeutic approaches represents a crucial goal to achieve. The onset, maintenance, and progression of inflammatory bowel disease are related to abnormal intestinal immune responses to environmental factors, including diet and intestinal microflora components. This study aimed to evaluate the potential antiinflammatory and antifibrotic effect of a biologically debittered olive cream and its probiotic oral administration in an experimental model of dextran sodium sulfate (DSS)-induced chronic colitis. METHODS Chronic colitis was induced in mice by three cycles of oral administration of 2.5% DSS (5 d of DSS followed by 7 d of tap water). Mice were randomly divided into five groups: 10 control mice fed with standard diet (SD), 20 mice receiving SD and DSS (SD+DSS), 20 mice receiving an enriched diet (ED) with olive cream and DSS (ED+DSS), 20 mice receiving SD plus probiotics (PB; Lactiplantibacillus plantarum IMC513) and DSS (SD+PB+DSS), and 20 mice receiving ED plus PB and DSS (ED+ PB+DSS). Clinical features and large bowel macroscopic, histologic, and immunohistochemical findings were evaluated. RESULTS The simultaneous administration of ED and PB induced a significant reduction in macroscopic and microscopic colitis scores compared with the other DSS-treated groups. In addition, ED and PB led to a significant decrease in the expression of inflammatory cytokines and profibrotic molecules. CONCLUSIONS The concomitant oral administration of a diet enriched with biologically debittered olive cream and a specific probiotic strain (Lactiplantibacillus plantarum IMC513) can exert synergistic antiinflammatory and antifibrotic action in DSS-induced chronic colitis. Further studies are needed to define the cellular and molecular mechanisms modulated by olive cream compounds and by Lactiplantibacillus plantarum IMC513.
Collapse
Affiliation(s)
- Antonella Vetuschi
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Natalia Battista
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - Simona Pompili
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy.
| | - Alfredo Cappariello
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Roberta Prete
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - Agnese Taticchi
- Department of Agricultural, Food and Environmental Sciences, University of Perugia, Perugia, Italy
| | - Roberto Selvaggini
- Department of Agricultural, Food and Environmental Sciences, University of Perugia, Perugia, Italy
| | - Giovanni Latella G
- Department of Life, Health and Environmental Sciences-Gastroenterology, Hepatology and Nutrition Division, University of L'Aquila, L'Aquila, Italy
| | - Aldo Corsetti
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - Roberta Sferra
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| |
Collapse
|
112
|
Little M, Dutta M, Li H, Matson A, Shi X, Mascarinas G, Molla B, Weigel K, Gu H, Mani S, Cui JY. Understanding the physiological functions of the host xenobiotic-sensing nuclear receptors PXR and CAR on the gut microbiome using genetically modified mice. Acta Pharm Sin B 2022; 12:801-820. [PMID: 35256948 PMCID: PMC8897037 DOI: 10.1016/j.apsb.2021.07.022] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 05/29/2021] [Accepted: 07/09/2021] [Indexed: 12/12/2022] Open
Abstract
Pharmacological activation of the xenobiotic-sensing nuclear receptors pregnane X receptor (PXR) and constitutive androstane receptor (CAR) is well-known to increase drug metabolism and reduce inflammation. Little is known regarding their physiological functions on the gut microbiome. In this study, we discovered bivalent hormetic functions of PXR/CAR modulating the richness of the gut microbiome using genetically engineered mice. The absence of PXR or CAR increased microbial richness, and absence of both receptors synergistically increased microbial richness. PXR and CAR deficiency increased the pro-inflammatory bacteria Helicobacteraceae and Helicobacter. Deficiency in both PXR and CAR increased the relative abundance of Lactobacillus, which has bile salt hydrolase activity, corresponding to decreased primary taurine-conjugated bile acids (BAs) in feces, which may lead to higher internal burden of taurine and unconjugated BAs, both of which are linked to inflammation, oxidative stress, and cytotoxicity. The basal effect of PXR/CAR on the gut microbiome was distinct from pharmacological and toxicological activation of these receptors. Common PXR/CAR-targeted bacteria were identified, the majority of which were suppressed by these receptors. hPXR-TG mice had a distinct microbial profile as compared to wild-type mice. This study is the first to unveil the basal functions of PXR and CAR on the gut microbiome.
Collapse
Key Words
- BA, bile acid
- BSH, bile salt hydrolase
- Bile acids
- CA, cholic acid
- CAR
- CAR, constitutive androstane receptor
- CDCA, chenodeoxycholic acid
- CITCO, 6-(4-chlorophenyl)imidazo[2,1-b][1,3]thiazole-5-carbaldehyde O-(3,4-dichlorobenzyl)oxime
- CV, conventional
- CYP, cytochrome P450
- DCA, deoxycholic acid
- EGF, epidermal growth factor
- Feces
- GF, germ free
- GLP-1, glucagon-like peptide-1
- GM-CSF, granulocyte-macrophage colony-stimulating factor
- Gut microbiome
- HDCA, hyodeoxycholic acid
- IBD, inflammatory bowel disease
- IFNγ, interferon-gamma
- IL, interleukin
- IS, internal standards
- Inflammation
- LCA, lithocholic acid
- LC–MS/MS, liquid chromatography–tandem mass spectrometry
- MCA, muricholic acid
- MCP-1, monocyte chemoattractant protein-1
- Mice
- NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells
- NSAID, non-steroidal anti-inflammatory drug
- Nuclear receptor
- OH, hydroxylated
- OTUs, operational taxonomy units
- PA, indole-3 propionic acid
- PBDEs, polybrominated diphenyl ethers
- PCBs, polychlorinated biphenyls
- PCoA, Principle Coordinate Analysis
- PXR
- PXR, pregnane X receptor
- PiCRUSt, Phylogenetic Investigation of Communities by Reconstruction of Observed States
- QIIME, Quantitative Insights Into Microbial Ecology
- SCFAs, short-chain fatty acids
- SNP, single-nucleotide polymorphism
- SPF, specific-pathogen-free
- T, wild type
- T-, taurine conjugated
- TCPOBOP, 1,4-bis-[2-(3,5-dichloropyridyloxy)]benzene, 3,3′,5,5′-Tetrachloro-1,4-bis(pyridyloxy)benzene
- TGR-5, Takeda G-protein-coupled receptor 5
- TLR4, toll-like receptor 4
- TNF, tumor necrosis factor
- UDCA, ursodeoxycholic acid
- YAP, yes-associated protein
- hPXR-TG, humanized PXR transgenic
Collapse
Affiliation(s)
- Mallory Little
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA 98105, USA
| | - Moumita Dutta
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA 98105, USA
| | - Hao Li
- Department of Medicine, Molecular Pharmacology and Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Adam Matson
- University of Connecticut, Hartford, CT 06106, USA
| | - Xiaojian Shi
- Arizona Metabolomics Laboratory, College of Health Solutions, Arizona State University, Phoenix, AZ 85004, USA
| | - Gabby Mascarinas
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA 98105, USA
| | - Bruk Molla
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA 98105, USA
| | - Kris Weigel
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA 98105, USA
| | - Haiwei Gu
- Arizona Metabolomics Laboratory, College of Health Solutions, Arizona State University, Phoenix, AZ 85004, USA
| | - Sridhar Mani
- Department of Medicine, Molecular Pharmacology and Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Julia Yue Cui
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA 98105, USA
| |
Collapse
|
113
|
Goessler KF, Gualano B, Nonino CB, Bonfá E, Nicoletti CF. Lifestyle Interventions and Weight Management in Systemic Lupus Erythematosus Patients: A Systematic Literature Review and Metanalysis. J Lifestyle Med 2022; 12:37-46. [PMID: 35300036 PMCID: PMC8918379 DOI: 10.15280/jlm.2022.12.1.37] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 01/05/2022] [Indexed: 11/22/2022] Open
Abstract
Background We aimed to identify and describe different types of lifestyle interventions primarily or secondarily focused on weight loss in SLE patients. Methods A systematic search of controlled trials published until June 2021 that assigned adults patients after dietary or exercise intervention resulted in 248 studies initially screened. Results Six studies with seven interventions (3 dietary and 4 exercise training programs) fulfilled the eligibility criteria and were included in the meta-analysis with a median of age 35.8 (31.3 to 49.0 years); median of BMI 26.6 (25.2 to 33.6 kg/m2). After six to twelve weeks of diet or exercise program, no differences were observed in body weight [-1.539 (-4.482 to 1.405) kg (CI 95%), p = 0.306]. Also, a subgroup analysis also revelated no body weight difference following dietary intervention [-3.561 (-9.604 to 2.481) kg (CI 95%), p = 0.248] or exercise intervention [-0.910 (-4.279 to 2.460) kg (CI 95%), p = 0.597]. Conclusion The results showed that different protocols of exercise intervention or diets were not effective to reduce body weight in patients with SLE. However, only one of the selected trials had a specific study design and protocol focusing on weight loss management.
Collapse
Affiliation(s)
- Karla F. Goessler
- Applied Physiology and Nutrition Research Group, School of Physical Education and Sport, Faculdade de Medicina FMUSP, Sao Paulo, Brazil
| | - Bruno Gualano
- Applied Physiology and Nutrition Research Group, School of Physical Education and Sport, Faculdade de Medicina FMUSP, Sao Paulo, Brazil
- Rheumatology Division, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Sao Paulo, Brazil
| | - Carla B. Nonino
- Department of Health Science, Ribeirão Preto School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Eloisa Bonfá
- Rheumatology Division, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Sao Paulo, Brazil
| | - Carolina Ferreira Nicoletti
- Applied Physiology and Nutrition Research Group, School of Physical Education and Sport, Faculdade de Medicina FMUSP, Sao Paulo, Brazil
- Rheumatology Division, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Sao Paulo, Brazil
| |
Collapse
|
114
|
Changes in Metabolic Regulation and the Microbiota Composition after Supplementation with Different Fatty Acids in db/db Mice. INTERNATIONAL JOURNAL OF FOOD SCIENCE 2022; 2022:3336941. [PMID: 35036426 PMCID: PMC8759926 DOI: 10.1155/2022/3336941] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Accepted: 12/15/2021] [Indexed: 12/20/2022]
Abstract
Introduction The effects of fatty acids on health vary and depend on the type, amount, and route of consumption. EPA and DHA have a defined role in health, unlike coconut oil. Objective The aim was to investigate the changes in metabolic regulation and the composition of the culture-dependent microbiota after supplementation with different fatty acids in db/db mice. Material and Methods. We were using 32 8-week-old db/db mice, supplemented for eight weeks with EPA/DHA derived from microalgae as well as coconut oil. The lipid, hormonal profiles, and composition of the culture-dependent microbiota and the phylogenetic analysis based on the 16S rRNA gene sequencing were determined for identification of the intestinal microbiota. Results Enriched diet with EPA/DHA reduced TNF-α, C-peptide, insulin resistance, resistin, and the plasma atherogenic index, but increased TC, LDL-c, VLDL-c, and TG without changes in HDL-c. Coconut oil raised the HDL-c, GIP, and TNF-α, with TG, insulin resistance, adiponectin, and C-peptide reduced. Conclusion The most abundant microbial populations were Firmicutes and the least Proteobacteria. EPA/DHA derived from microalgae contributes to improving the systemic inflammatory status, but depressed the diversity of the small intestine microbiota. Coconut oil only decreased the C-peptide, raising TNF-α, with an unfavorable hormonal and lipid profile.
Collapse
|
115
|
Effects of Dietary Enterococcus faecalis YFI-G720 on the Growth, Immunity, Serum Biochemical, Intestinal Morphology, Intestinal Microbiota, and Disease Resistance of Crucian Carp (Carassius auratus). FISHES 2022. [DOI: 10.3390/fishes7010018] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Diseases of crucian carp (Carassius auratus) are closely related to intestinal parameters. Enterococcus faecalis has strong colonization ability in the intestinal tract, and produces natural antibiotics, bacteriocin, and other bacteriostatic substances, which can effectively inhibit some pathogenic bacteria and improve the intestinal microenvironment. This study aimed to assess the effects of E. faecalis YFI-G720 which was isolated from the intestinal of crucian carp on the growth, immunity, intestinal health, and disease resistance of crucian carp. Fish (48.16 ± 0.55 g) were fed four diets, commercial diet or diet containing E. faecalis at 105 CFU/g (EF1), 106 CFU/g (EF2), or 107 CFU/g (EF3) for 28 days. The results showed that supplementation of E. faecalis significantly improved the weight gain ratio (WGR) and the specific growth rate (SGR) compared with control group (p < 0.05). Intestinal mucosal epithelial cells in EF2 were intact and normal, but there was obvious vacuolation in CG. Compared with CG, serum C3 and IgM in EF2 were significantly increased at the end of the experiment (p < 0.05), and serum alkaline phosphatase was significantly higher in all experimental groups (p < 0.05). Among studied immune-related genes, expression was detected by qPCR, C3, IgM, and IL-1βwere upregulated in all experimental groups to varying degrees from 14 days, with highest expression in EF2 at 28 days. Intestinal microbiota structure analyzed through high-throughput sequencing, and the results showed that the relative abundance of Aeromonas and Acinetobacter decreased while Cetobacterium increased in all experimental groups, with the greatest changes in EF2. Challenge tests showed that fish fed E. faecalis were more resistant to Aeromonas veronii (p < 0.05). In conclusion, dietary E. faecalis YFI-G720 at 106 CFU/g can improve the health status, immune parameters, intestinal microbiota composition, and disease resistance of crucian carp.
Collapse
|
116
|
Malo J, Alam MJ, Islam S, Mottalib MA, Rocki MMH, Barmon G, Tinni SA, Barman SK, Sarker T, Khan MNI, Kaliannan K, Hasanat MA, Rahman S, Pathan MF, Khan AKA, Malo MS. Intestinal alkaline phosphatase deficiency increases the risk of diabetes. BMJ Open Diabetes Res Care 2022; 10:10/1/e002643. [PMID: 35082135 PMCID: PMC8796214 DOI: 10.1136/bmjdrc-2021-002643] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 12/06/2021] [Indexed: 11/24/2022] Open
Abstract
INTRODUCTION Our previous case-control study demonstrated that a high level of intestinal alkaline phosphatase (IAP), an endotoxin-detoxifying anti-inflammatory enzyme secreted by villus-associated enterocytes and excreted with stool, plays a protective role against type 2 diabetes mellitus (T2DM) irrespective of obesity. In the current study, we investigated the long-term effect of IAP deficiency (IAPD) on the pathogenesis of T2DM. RESEARCH DESIGN AND METHODS A healthy cohort of participants without diabetes (30-60 years old), comprising 188 without IAPD (IAP level: ≥65 U/g stool) and 386 with IAPD (IAP level: <65 U/g stool), were followed up for 5 years. We measured stool IAP (STAP) and fasting plasma glucose, and calculated the risk ratio (RR) using log-binomial regression model. RESULTS T2DM incidence rates were 8.0%, 11.7%, 25.6%, and 33.3% in participants with 'persistent no IAPD' (IAP level: always ≥65 U/g stool), 'remittent IAPD' (IAP level: increased from <65 U/g stool to ≥65 U/g stool), 'persistent IAPD' (IAP level: always <65 U/g stool), and 'incident IAPD' (IAP level: decreased from ≥65 U/g stool to <65 U/g stool), respectively. Compared with 'persistent no IAPD' the risk of developing T2DM with 'incident IAPD' was 270% higher (RR: 3.69 (95% CI 1.76 to 7.71), χ2 p<0.001). With 'persistent IAPD' the risk was 230% higher (RR: 3.27 (95% CI 1.64 to 6.50), p<0.001). 'Remittent IAPD' showed insignificant risk (RR: 2.24 (95% CI 0.99 to 5.11), p=0.0541). Sensitivity analyses of persistent IAP levels revealed that, compared with participants of the highest persistent IAP pentile (always >115 U/g stool), the rate of increase of fasting glycemia was double and the risk of developing T2DM was 1280% higher (RR: 13.80 (95% CI 1.87 to 101.3), p=0.0099) in participants of the lowest persistent IAP pentile (always <15 U/g stool). A diabetes pathogenesis model is presented. CONCLUSIONS IAPD increases the risk of developing T2DM, and regular STAP tests would predict individual vulnerability to T2DM. Oral IAP supplementation might prevent T2DM.
Collapse
Affiliation(s)
| | - Md Jahangir Alam
- Department of Statistics, University of Rajshahi, Rajshahi, Bangladesh
| | - Salequl Islam
- Department of Microbiology, Jahangirnagar University, Savar, Bangladesh
| | - Md Abdul Mottalib
- Department of Biochemistry and Molecular Biology, BIRDEM, Dhaka, Bangladesh
| | | | - Ginok Barmon
- Diabetic Association of Bangladesh, Dhaka, Bangladesh
| | | | | | - Tapas Sarker
- Diabetic Association of Bangladesh, Dhaka, Bangladesh
| | | | - Kanakaraju Kaliannan
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Muhammad Abul Hasanat
- Department of Endocrinology, Bangabandhu Sheikh Mujib Medical University, Dhaka, Bangladesh
| | - Salimur Rahman
- Department of Hepatology, Bangabandhu Sheikh Mujib Medical University, Dhaka, Bangladesh
| | | | - A K Azad Khan
- Diabetic Association of Bangladesh, Dhaka, Bangladesh
| | - Madhu S Malo
- Diabetic Association of Bangladesh, Dhaka, Bangladesh
- Department of Biochemistry and Molecular Biology, BIRDEM, Dhaka, Bangladesh
- Centre for Global Health Research, Diabetic Association of Bangladesh, Dhaka, Bangladesh
| |
Collapse
|
117
|
LIU X, TAN F, CUI M, LI D, YAO P. Effects of red meat diet on gut microbiota in mice. FOOD SCIENCE AND TECHNOLOGY 2022. [DOI: 10.1590/fst.28321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Xiaoyan LIU
- Xinjiang Medical University, P.R. China; Xinjiang Medical University, P.R. China
| | - Fang TAN
- Xinjiang Medical University, P.R. China
| | - Min CUI
- Xinjiang Medical University, P.R. China
| | - Danping LI
- Xinjiang Medical University, P.R. China; Xinjiang Medical University, P.R. China
| | - Ping YAO
- Xinjiang Medical University, P.R. China
| |
Collapse
|
118
|
Khushboo, Dubey KK. Microbial metabolites beneficial in regulation of obesity. CURRENT DEVELOPMENTS IN BIOTECHNOLOGY AND BIOENGINEERING 2022:355-375. [DOI: 10.1016/b978-0-12-823506-5.00006-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
119
|
Kandel Gambarte PC, Wolansky MJ. The gut microbiota as a biomarker for realistic exposures to pesticides: A critical consideration. Neurotoxicol Teratol 2022; 91:107074. [DOI: 10.1016/j.ntt.2022.107074] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 12/24/2021] [Accepted: 01/10/2022] [Indexed: 10/19/2022]
|
120
|
Sianipar IR, Sestramita S, Pradnjaparamita T, Yunir E, Harbuwono DS, Soewondo P, Tahapary DL. The role of Intestinal-Fatty Acid Binding Proteins and Chitinase-3-Like Protein 1 across the spectrum of dysglycemia. Diabetes Metab Syndr 2022; 16:102366. [PMID: 34942410 DOI: 10.1016/j.dsx.2021.102366] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 12/07/2021] [Accepted: 12/09/2021] [Indexed: 11/15/2022]
Abstract
BACKGROUND AND AIMS Recent studies underlie the importance of intestinal permeability and chronic inflammation in the pathogenesis of T2DM. Our study compared the concentrations of FABP2 and YKL40 as markers of intestinal permeability and inflammation among normoglycemia, prediabetes and T2DM. METHODS We recruited 122 participants (45 normoglycemic, 26 prediabetes, and 51 T2DM) of whom we measured the fasting serum levels of FABP2 and YKL-40 using ELISA method. RESULTS The levels of FABP2 were significantly higher in the T2DM group [2.890 (1.880-4.070)] in comparison to both prediabetes [2.025 (1.145-2.343), p = 0.0085] and normoglycemia group [1.72 (1.250-2.645), p = 0.011]. The levels of YKL-40 were also significantly higher in the T2DM group [68.70 (44.61-166.6)] in comparison to both prediabetes [28.85 (20.64-41.53), p < 0.0001] and normoglycemia group [28.64 (19.25-43.87), p < 0.001]. CONCLUSIONS Our study observed that the levels of FABP2 and YKL-40 were highest in the T2DM group supporting the available evidences on the role of intestinal permeability disruption and chronic low-grade inflammation in the pathogenesis of T2DM.
Collapse
Affiliation(s)
- Imelda R Sianipar
- Department of Medical Physiology, Faculty of Medicine, Universitas Indonesia, Indonesia.
| | - Sestramita Sestramita
- Graduate Student of Master Program in Biomedical Science, Faculty of Medicine, Universitas Indonesia, Indonesia
| | - Tika Pradnjaparamita
- Metabolic, Cardiovascular and Aging Research Cluster, The Indonesian Medical Education and Research Institute, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Em Yunir
- Metabolic, Cardiovascular and Aging Research Cluster, The Indonesian Medical Education and Research Institute, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia; Division of Endocrinology, Metabolism and Diabetes, Department of Internal Medicine, dr. Cipto Mangunkusumo National General Hospital/Faculty of Medicine Universitas Indonesia, Indonesia
| | - Dante S Harbuwono
- Metabolic, Cardiovascular and Aging Research Cluster, The Indonesian Medical Education and Research Institute, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia; Division of Endocrinology, Metabolism and Diabetes, Department of Internal Medicine, dr. Cipto Mangunkusumo National General Hospital/Faculty of Medicine Universitas Indonesia, Indonesia
| | - Pradana Soewondo
- Metabolic, Cardiovascular and Aging Research Cluster, The Indonesian Medical Education and Research Institute, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia; Division of Endocrinology, Metabolism and Diabetes, Department of Internal Medicine, dr. Cipto Mangunkusumo National General Hospital/Faculty of Medicine Universitas Indonesia, Indonesia.
| | - Dicky L Tahapary
- Metabolic, Cardiovascular and Aging Research Cluster, The Indonesian Medical Education and Research Institute, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia; Division of Endocrinology, Metabolism and Diabetes, Department of Internal Medicine, dr. Cipto Mangunkusumo National General Hospital/Faculty of Medicine Universitas Indonesia, Indonesia.
| |
Collapse
|
121
|
Portincasa P, Bonfrate L, Khalil M, Angelis MD, Calabrese FM, D’Amato M, Wang DQH, Di Ciaula A. Intestinal Barrier and Permeability in Health, Obesity and NAFLD. Biomedicines 2021; 10:83. [PMID: 35052763 PMCID: PMC8773010 DOI: 10.3390/biomedicines10010083] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/20/2021] [Accepted: 12/28/2021] [Indexed: 02/07/2023] Open
Abstract
The largest surface of the human body exposed to the external environment is the gut. At this level, the intestinal barrier includes luminal microbes, the mucin layer, gastrointestinal motility and secretion, enterocytes, immune cells, gut vascular barrier, and liver barrier. A healthy intestinal barrier is characterized by the selective permeability of nutrients, metabolites, water, and bacterial products, and processes are governed by cellular, neural, immune, and hormonal factors. Disrupted gut permeability (leaky gut syndrome) can represent a predisposing or aggravating condition in obesity and the metabolically associated liver steatosis (nonalcoholic fatty liver disease, NAFLD). In what follows, we describe the morphological-functional features of the intestinal barrier, the role of major modifiers of the intestinal barrier, and discuss the recent evidence pointing to the key role of intestinal permeability in obesity/NAFLD.
Collapse
Affiliation(s)
- Piero Portincasa
- Clinica Medica “A. Murri”, Department of Biomedical Sciences & Human Oncology, University of Bari Medical School, 70124 Bari, Italy; (L.B.); (M.K.); (A.D.C.)
| | - Leonilde Bonfrate
- Clinica Medica “A. Murri”, Department of Biomedical Sciences & Human Oncology, University of Bari Medical School, 70124 Bari, Italy; (L.B.); (M.K.); (A.D.C.)
| | - Mohamad Khalil
- Clinica Medica “A. Murri”, Department of Biomedical Sciences & Human Oncology, University of Bari Medical School, 70124 Bari, Italy; (L.B.); (M.K.); (A.D.C.)
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, Via Amendola 165/a, 70126 Bari, Italy; (M.D.A.); (F.M.C.)
| | - Maria De Angelis
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, Via Amendola 165/a, 70126 Bari, Italy; (M.D.A.); (F.M.C.)
| | - Francesco Maria Calabrese
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, Via Amendola 165/a, 70126 Bari, Italy; (M.D.A.); (F.M.C.)
| | - Mauro D’Amato
- Gastrointestinal Genetics Lab, CIC bioGUNE-BRTA, 48160 Derio, Spain;
- Ikerbasque, Basque Foundation for Science, 48009 Bilbao, Spain
| | - David Q.-H. Wang
- Department of Medicine and Genetics, Division of Gastroenterology and Liver Diseases, Marion Bessin Liver Research Center, Einstein-Mount Sinai Diabetes Research Center, Albert Einstein College of Medicine, New York, NY 10461, USA;
| | - Agostino Di Ciaula
- Clinica Medica “A. Murri”, Department of Biomedical Sciences & Human Oncology, University of Bari Medical School, 70124 Bari, Italy; (L.B.); (M.K.); (A.D.C.)
| |
Collapse
|
122
|
Ahmed I, Yusuf K, Roy BC, Stubbs J, Anant S, Attard TM, Sampath V, Umar S. Dietary Interventions Ameliorate Infectious Colitis by Restoring the Microbiome and Promoting Stem Cell Proliferation in Mice. Int J Mol Sci 2021; 23:339. [PMID: 35008767 PMCID: PMC8745185 DOI: 10.3390/ijms23010339] [Citation(s) in RCA: 8] [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: 11/09/2021] [Revised: 12/21/2021] [Accepted: 12/25/2021] [Indexed: 12/15/2022] Open
Abstract
Decreases in short-chain-fatty-acids (SCFAs) are linked to inflammatory bowel disease (IBD). Yet, the mechanisms through which SCFAs promote wound healing, orchestrated by intestinal stem cells, are poorly understood. We discovered that, in mice with Citrobacter rodentium (CR)-induced infectious colitis, treatment with Pectin and Tributyrin diets reduced the severity of colitis by restoring Firmicutes and Bacteroidetes and by increasing mucus production. RNA-seq in young adult mouse colon (YAMC) cells identified higher expression of Lgr4, Lgr6, DCLK1, Muc2, and SIGGIR after Butyrate treatment. Lineage tracing in CR-infected Lgr5-EGFP-IRES-CreERT2/ROSA26-LacZ (Lgr5-R) mice also revealed an expansion of LacZ-labeled Lgr5(+) stem cells in the colons of both Pectin and Tributyrin-treated mice compared to control. Interestingly, gut microbiota was required for Pectin but not Tributyrin-induced Lgr5(+) stem cell expansion. YAMC cells treated with sodium butyrate exhibited increased Lgr5 promoter reporter activity due to direct Butyrate binding with Lgr5 at -4.0 Kcal/mol, leading to thermal stabilization. Upon ChIP-seq, H3K4me3 increased near Lgr5 transcription start site that contained the consensus binding motif for a transcriptional activator of Lgr5 (SPIB). Thus, a multitude of effects on gut microbiome, differential gene expression, and/or expansion of Lgr5(+) stem cells seem to underlie amelioration of colitis following dietary intervention.
Collapse
Affiliation(s)
- Ishfaq Ahmed
- Department of Surgery, University of Kansas Medical Center, Kansas City, KS 66160, USA; (I.A.); (K.Y.); (B.C.R.)
| | - Kafayat Yusuf
- Department of Surgery, University of Kansas Medical Center, Kansas City, KS 66160, USA; (I.A.); (K.Y.); (B.C.R.)
| | - Badal C. Roy
- Department of Surgery, University of Kansas Medical Center, Kansas City, KS 66160, USA; (I.A.); (K.Y.); (B.C.R.)
| | - Jason Stubbs
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA;
| | - Shrikant Anant
- Cancer Biology Department, University of Kansas Medical Center, Kansas City, KS 66160, USA;
| | - Thomas M. Attard
- Department of Pediatrics and Gastroenterology, Children’s Mercy Hospital, Kansas City, KS 66160, USA; (T.M.A.); (V.S.)
| | - Venkatesh Sampath
- Department of Pediatrics and Gastroenterology, Children’s Mercy Hospital, Kansas City, KS 66160, USA; (T.M.A.); (V.S.)
| | - Shahid Umar
- Department of Surgery, University of Kansas Medical Center, Kansas City, KS 66160, USA; (I.A.); (K.Y.); (B.C.R.)
| |
Collapse
|
123
|
Intestinal microbiota and their metabolic contribution to type 2 diabetes and obesity. J Diabetes Metab Disord 2021; 20:1855-1870. [PMID: 34900829 PMCID: PMC8630233 DOI: 10.1007/s40200-021-00858-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 07/14/2021] [Indexed: 02/06/2023]
Abstract
Obesity and type 2 diabetes mellitus (T2DM) are common, chronic metabolic disorders with associated significant long-term health problems at global epidemic levels. It is recognised that gut microbiota play a central role in maintaining host homeostasis and through technological advances in both animal and human models it is becoming clear that gut microbiota are heavily involved in key pathophysiological roles in the aetiology and progression of both conditions. This review will focus on current knowledge regarding microbiota interactions with short chain fatty acids, the host inflammatory response, signaling pathways, integrity of the intestinal barrier, the interaction of the gut-brain axis and the subsequent impact on the metabolic health of the host.
Collapse
|
124
|
Sayers B, Wijeyesekera A, Gibson G. Exploring the potential of prebiotic and polyphenol-based dietary interventions for the alleviation of cognitive and gastrointestinal perturbations associated with military specific stressors. J Funct Foods 2021. [DOI: 10.1016/j.jff.2021.104753] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
|
125
|
Evaluation of Microbiome Alterations Following Consumption of BIOHM, a Novel Probiotic. Curr Issues Mol Biol 2021; 43:2135-2146. [PMID: 34940122 PMCID: PMC8928933 DOI: 10.3390/cimb43030148] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/21/2021] [Accepted: 11/24/2021] [Indexed: 12/11/2022] Open
Abstract
Gastrointestinal microbiome dysbiosis may result in harmful effects on the host, including those caused by inflammatory bowel diseases (IBD). The novel probiotic BIOHM, consisting of Bifidobacterium breve, Saccharomyces boulardii, Lactobacillus acidophilus, L. rhamnosus, and amylase, was developed to rebalance the bacterial–fungal gut microbiome, with the goal of reducing inflammation and maintaining a healthy gut population. To test the effect of BIOHM on human subjects, we enrolled a cohort of 49 volunteers in collaboration with the Fermentation Festival group (Santa Barbara, CA, USA). The profiles of gut bacterial and fungal communities were assessed via stool samples collected at baseline and following 4 weeks of once-a-day BIOHM consumption. Mycobiome analysis following probiotic consumption revealed an increase in Ascomycota levels in enrolled individuals and a reduction in Zygomycota levels (p value < 0.01). No statistically significant difference in Basidiomycota was detected between pre- and post-BIOHM samples and control abundance profiles (p > 0.05). BIOHM consumption led to a significant reduction in the abundance of Candida genus in tested subjects (p value < 0.013), while the abundance of C. albicans also trended lower than before BIOHM use, albeit not reaching statistical significance. A reduction in the abundance of Firmicutes at the phylum level was observed following BIOHM use, which approached levels reported for control individuals reported in the Human Microbiome Project data. The preliminary results from this clinical study suggest that BIOHM is capable of significantly rebalancing the bacteriome and mycobiome in the gut of healthy individuals, suggesting that further trials examining the utility of the BIOHM probiotic in individuals with gastrointestinal symptoms, where dysbiosis is considered a source driving pathogenesis, are warranted.
Collapse
|
126
|
Emodin Improves Intestinal Health and Immunity through Modulation of Gut Microbiota in Mice Infected by Pathogenic Escherichia coli O 1. Animals (Basel) 2021; 11:ani11113314. [PMID: 34828045 PMCID: PMC8614316 DOI: 10.3390/ani11113314] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 11/12/2021] [Accepted: 11/15/2021] [Indexed: 02/07/2023] Open
Abstract
The effect of emodin on the intestinal mucosal barrier of a mouse E. coli O1-induced diarrhea model was observed. Following successful establishment of a diarrhea model, the mice were treated with drugs for seven days. Intestinal lesions and the shape and the number of goblet cells were assessed via hematoxylin-eosin and periodic-acid-Schiff staining, while changes in inflammatory factors, ultrastructure of the small intestine, expression of MUC-2, and changes in the intestinal microbiota were analyzed via RT-PCR, electron microscopy, immunofluorescence, and 16S rRNA sequencing. Examination showed that emodin ameliorated pathological damage to the intestines of diarrheic mice. RT-PCR indicated that emodin reduced TNF-α, IL-β, IL-6, MPO, and COX-2 mRNA levels in duodenal tissues and increased the levels of sIgA and MUC-2 and the number of goblet cells. Microbiome analysis revealed that Escherichia coli O1 reduced bacterial richness and altered the distribution pattern of bacterial communities at the phylum and order levels in cecum contents. Notably, pathogenic Clostridiales and Enterobacteriales were significantly increased in diarrheic mice. However, emodin reversed the trend. Thus, emodin protected against intestinal damage induced by E. coli O1 and improved intestinal mucosal barrier function in mice by increasing the abundance of beneficial intestinal microbiota and inhibiting the abundance of harmful bacteria, thereby alleviating diarrhea.
Collapse
|
127
|
Yang J, Qiu X, Zhou M, Wang D. Crocetin attenuating Urinary tract Infection and adherence of uropathogenic E. coli in NRK-52E cells via an inflammatory pathway. J Food Biochem 2021; 45:e13998. [PMID: 34792197 DOI: 10.1111/jfbc.13998] [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: 06/12/2021] [Revised: 09/17/2021] [Accepted: 09/19/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND AND OBJECTIVE Urinary tract infections (UTI) are commonly treated with broad-spectrum antibiotics, but treatment has limitation due to causes of nephrotoxicity in uroepithelial cells. Recently, the researcher focuses their research on alternative therapy for the treatment of UTI. This study evaluated the anti-infectious effect of crocetin against adherence of pathogenic [2-14 C]-acetate labeled Escherichia coli (MTCC-729) to rat proximal renal tubular cells (NRK-52E cells) and explores the possible mechanism of action. MATERIALS AND METHODS In vitro cytotoxicity and radio acetate labeled tests were performed on NRK-52E cells. The rats were divided into five different groups as follows: normal control (NC), disease control (DC), and various doses of crocetin (1.25, 2.5, and 5 mg/kg) treated group rats. White blood cells in blood, urine, and bacterial colony counts were estimated at regular intervals. Pro-inflammatory cytokines, such as interleukin-6 (IL-6), monocyte chemoattractant protein-1 (MCP-1), interleukin-10 (IL-10), and interleukin-8 (IL-8), were also estimated. In the current study, we estimated the mRNA expression of toll-like receptor-4 (TLR-4) and toll-like receptor-2 (TLR-2) in the renal and bladder tissues. RESULTS Crocetin significantly (p < .05) inhibited the adherence of E. Coli in NRK-52E cells. Crocetin suppresses the lipid peroxidation (LPO) 42% in cells treated with H2 O2 cells without crocetin. The white blood cells (WBC) count in blood and urine were augmented and crocetin treatment significantly (p < .05) reduced the WBC in urine and blood. The pro-inflammatory cytokines, such as IL-6, MCP-1, IL-10, and IL-8, significantly (p < .05) increased in the DC group and crocetin significantly (p < .05) reduced the pro-inflammatory cytokines. Dose-dependent treatment of crocetin significantly reduced the mRNA expression of TLR2 and TLR4 in the renal and bladder tissues. CONCLUSION Crocetin considerably reduced the bacterial adherence to NRK-52E cells, attenuated the H2 O2 induced toxicity in NRK-52E cells and also improved the renal tubular function, and reduced the inflammatory response via altering the inflammatory and antioxidant markers. PRACTICAL APPLICATION As we all know that urinary tract infection is the most common disease worldwide. In this study, we scrutinized the protective effect of crocetin against urinary tract infection. Crocetin treatment considerably reduced the zone of inhibition and improved radioactivity. Crocetin significantly reduced the levels of cytokines and inflammatory mediators. Crocetin can be used as a protective drug in the treatment of urinary tract infections.
Collapse
Affiliation(s)
- Jian Yang
- Department of Nephrology, Baoji Hospital of traditional Chinese Medicine, Baoji, China
| | - Xin Qiu
- Department of Nephrology, Baoji Hospital of traditional Chinese Medicine, Baoji, China
| | - Meilan Zhou
- Department of Nephrology, Xijing Hospital, The First Affiliated Hospital of Fourth Military Medical University, Xi'an, China
| | - Di Wang
- Department of Nephrology, Xijing Hospital, The First Affiliated Hospital of Fourth Military Medical University, Xi'an, China
| |
Collapse
|
128
|
Higashimura Y, Hirabayashi M, Nishikawa H, Inoue R, Nagai E, Matsumoto K, Enomoto T, Mizushima K, Takagi T, Naito Y. Dietary intake of yacon roots ( Smallanthus sonchifolius) affects gut microbiota and fecal mucin and prevents intestinal inflammation in mice. J Clin Biochem Nutr 2021; 69:272-279. [PMID: 34857989 PMCID: PMC8611369 DOI: 10.3164/jcbn.20-203] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Accepted: 02/28/2021] [Indexed: 12/23/2022] Open
Abstract
Consumption of yacon (Smallanthus sonchifolius) is associated with beneficial effects such as prevention of metabolic diseases. Yacon root is known to contain various bioactive components including indigestible carbohydrates, but the alteration of intestinal environment after treatment with yacon has not been fully investigated. This study investigated yacon-containing diet effects on the intestinal environment in mice, including microbial composition, short-chain fatty acid levels, and mucus content. After mice were administered yacon-containing diet for 4 weeks, 16S rRNA gene sequencing analyses revealed their fecal microbiota profiles. Organic acid concentrations in cecal contents were measured using an HPLC system. Compared to the control group, yacon-containing diet-received mice had significantly higher the concentrations of succinic acid, lactic acid, acetic acid, and propionic acid. The fecal mucin content was also higher in yacon-containing diet-received mice. Results of 16S rRNA gene sequencing analyses showed that the relative abundances of 27 taxa differed significantly in yacon-containing diet-received mice. Furthermore, results show effects of yacon administration on intestinal inflammation using 2,4,6-trinitrobenzene sulfonic acid induced colitis model in mice. Increased colonic damage and myeloperoxidase activity after 2,4,6-trinitrobenzene sulfonic acid treatment were suppressed in yacon-containing diet-received mice. Results suggest that oral intake of yacon root modulates the intestinal environment, thereby inhibiting intestinal inflammation.
Collapse
Affiliation(s)
- Yasuki Higashimura
- Department of Food Science, Faculty of Bioresources and Environmental Sciences, Ishikawa Prefectural University, Nonoichi, Ishikawa 921-8836, Japan
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Misaki Hirabayashi
- Department of Food Science, Faculty of Bioresources and Environmental Sciences, Ishikawa Prefectural University, Nonoichi, Ishikawa 921-8836, Japan
| | - Hitomi Nishikawa
- Department of Food Science, Faculty of Bioresources and Environmental Sciences, Ishikawa Prefectural University, Nonoichi, Ishikawa 921-8836, Japan
| | - Ryo Inoue
- Department of Applied Biological Sciences, Faculty of Agriculture, Setsunan University, Hirakata, Osaka 573-0101, Japan
| | - Emiko Nagai
- Department of Food Science, Faculty of Bioresources and Environmental Sciences, Ishikawa Prefectural University, Nonoichi, Ishikawa 921-8836, Japan
| | - Kenji Matsumoto
- Department of Food Science, Faculty of Bioresources and Environmental Sciences, Ishikawa Prefectural University, Nonoichi, Ishikawa 921-8836, Japan
| | - Toshiki Enomoto
- Department of Food Science, Faculty of Bioresources and Environmental Sciences, Ishikawa Prefectural University, Nonoichi, Ishikawa 921-8836, Japan
| | - Katsura Mizushima
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Tomohisa Takagi
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Yuji Naito
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| |
Collapse
|
129
|
Metagenomic Sequencing Reveals that High-Grain Feeding Alters the Composition and Metabolism of Cecal Microbiota and Induces Cecal Mucosal Injury in Sheep. mSystems 2021; 6:e0091521. [PMID: 34609166 PMCID: PMC8547435 DOI: 10.1128/msystems.00915-21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
The cecum serves as an additional fermentation site for ruminants, but it lacks buffering capacity and has a relatively simple epithelial structure compared to the rumen. The role of high-grain (HG) diets in manipulating the rumen microbiome has been well elucidated, yet the microbial response to such diets in the cecum and the subsequent microbe-host interactions remain largely unexplored. Here, we describe the modification of the cecal microbiome and host epithelial gene expression based on data from 20 sheep grouped to feed an HG diet for 7, 14, and 28 days. Our data indicate that the alteration of cecal microbial fermentation was manifested by a decrease in luminal pH and an increase in acetate and butyrate concentrations following the diet change to HG. We further demonstrate that the alteration of the microbiome was driven by microbes that are likely acetate producers (e.g., Blautia spp. and Akkermansia spp.) and butyrate producers (e.g., Anaerostipes spp. and Roseburia spp.). Moreover, the core microbiota in the cecal microbiome was predominantly maintained after HG diet feeding, while the specific populations of the cecal microbiomes adaptively varied at the species and genomic levels time dependently. Association analysis suggests that the perturbations of the cecal microbiome under the HG diet were closely linked to the variations in the two key enzymes that catalyze the conversion of pyruvate to acetyl-CoA and urease enzymes that hydrolyze urea into ammonia, alongside mucosal inflammatory responses. Overall, our findings here provide novel insights into understanding microbiome-host interactions in the hindgut of ruminants. IMPORTANCE High-grain (HG) diets are known to alter the rumen microbiome. However, the responses of the hindgut microbiota and its epithelial function need further investigation in ruminants. Using 20 sheep as the experimental model, we found that the microbial fermentation pattern of the cecum changed after switching to the HG diet. The taxa of the acetate and butyrate producers increased with the feeding time. Moreover, enzymes engaged in carbon and nitrogen metabolisms of the cecal microbiome are altered. The expression of epithelial genes related to volatile fatty acid (VFA) absorption and metabolism, cytokines, and tight junction proteins, alongside light microscopy visualization of epithelial tissue, suggested that the HG diet may induce cecal mucosal inflammatory responses. Our findings reveal cecal microbial and metabolic perturbations in response to HG diets in sheep and provide a new reference for the research on hindgut microbial homeostasis and host health in ruminants.
Collapse
|
130
|
Effects of Feeding a Hypoallergenic Diet with a Nutraceutical on Fecal Dysbiosis Index and Clinical Manifestations of Canine Atopic Dermatitis. Animals (Basel) 2021; 11:ani11102985. [PMID: 34680004 PMCID: PMC8532801 DOI: 10.3390/ani11102985] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/11/2021] [Accepted: 10/14/2021] [Indexed: 12/25/2022] Open
Abstract
Simple Summary The prevalence of canine atopic dermatitis in developed countries has been growing constantly over the last few decades. Genetic predisposition represents only part of the problem, and environmental factors are believed to be an important boost to the rapid rise in atopic dogs. Although a complete understanding of the gut–skin axis has not yet been achieved, a growing number of studies demonstrate a close relationship between gastrointestinal imbalance and skin diseases. The aim of this study was to assess the effect of a nutraceutical product on the Dysbiotic Index and skin lesions of atopic dogs. Abstract Background: an imbalance of the intestinal microbiota can cause health problems in the gastrointestinal tract and in other organs. Canine Atopic Dermatitis (CAD) is a genetically predisposed, inflammatory and pruritic allergic skin disease with multifactorial etiology and multimodal treatment. The aim of this study was to assess the effect of a nutraceutical product on Dysbiotic Index (DI) and the skin lesions of atopic dogs. Methods: a nutraceutical product was administered to 32 dogs with CAD. The product was associated with a standardized hypoallergenic diet for 60 days; the dietary regimen continued for 120 days, while ongoing therapies remained unchanged. Values of Visual Analogic Scale (VAS), Canine Atopic Dermatitis Lesional Index (CADLI) and DI were evaluated on day 0, 60, 120. Results: all the 32 dogs showed a statistically significant decrease (p < 0.001) to V60 of VAS and CADLI, which persisted and increased to V120 when diet alone was continued. The decrease in the DI value was also statistically significant (p < 0.001). Conclusion: the intake of nutraceutical associated with diet resulted in a decrease in the index of intestinal dysbiosis, with an improvement in the subjective severity of cutaneous lesions.
Collapse
|
131
|
Panneerselvam J, Madka V, Rai R, Morris KT, Houchen CW, Chandrakesan P, Rao CV. Inflammatory Mediators and Gut Microbial Toxins Drive Colon Tumorigenesis by IL-23 Dependent Mechanism. Cancers (Basel) 2021; 13:5159. [PMID: 34680308 PMCID: PMC8533859 DOI: 10.3390/cancers13205159] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 10/03/2021] [Accepted: 10/08/2021] [Indexed: 01/01/2023] Open
Abstract
Obesity-associated chronic inflammation predisposes colon cancer risk development. Interleukin-23 (IL-23) is a potential inflammatory mediator linking obesity to chronic colonic inflammation, altered gut microbiome, and colon carcinogenesis. We aimed to elucidate the role of pro-inflammatory eicosanoids and gut bacterial toxins in priming dendritic cells and macrophages for IL-23 secretion to promote colon tumor progression. To investigate the association of IL-23 with obesity and colon tumorigenesis, we utilized TCGA data set and colonic tumors from humans and preclinical models. To understand IL-23 production by inflammatory mediators and gut microbial toxins, we performed several in vitro mechanistic studies to mimic the tumor microenvironment. Colonic tumors were utilized to perform the ex vivo experiments. Our findings showed that IL-23 is elevated in obese individuals, colonic tumors and correlated with reduced disease-free survival. In vitro studies showed that IL-23 treatment increased the colon tumor cell self-renewal, migration, and invasion while disrupting epithelial barrier permeability. Co-culture experiments of educated dendritic cells/macrophages with colon cancer cells significantly increased the tumor aggression by increasing the secretory levels of IL-23, and these observations are further supported by ex vivo rat colonic tumor organotypic experiments. Our results demonstrate gut microbe toxins and eicosanoids facilitate IL-23 production, which plays an important role in obesity-associated colonic tumor progression. This newly identified nexus represents a potential target for the prevention and treatment of obesity-associated colon cancer.
Collapse
Affiliation(s)
- Janani Panneerselvam
- Center for Cancer Prevention and Drug Development, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (J.P.); (V.M.)
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (R.R.); (C.W.H.); (P.C.)
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Venkateshwar Madka
- Center for Cancer Prevention and Drug Development, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (J.P.); (V.M.)
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (R.R.); (C.W.H.); (P.C.)
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Rajani Rai
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (R.R.); (C.W.H.); (P.C.)
| | - Katherine T. Morris
- Department of Surgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
| | - Courtney W. Houchen
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (R.R.); (C.W.H.); (P.C.)
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Parthasarathy Chandrakesan
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (R.R.); (C.W.H.); (P.C.)
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Chinthalapally V. Rao
- Center for Cancer Prevention and Drug Development, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (J.P.); (V.M.)
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (R.R.); (C.W.H.); (P.C.)
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- VA Medical Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
132
|
Nowland TL, Kirkwood RN, Pluske JR. Review: Can early-life establishment of the piglet intestinal microbiota influence production outcomes? Animal 2021; 16 Suppl 2:100368. [PMID: 34649827 DOI: 10.1016/j.animal.2021.100368] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 08/07/2021] [Accepted: 08/27/2021] [Indexed: 12/21/2022] Open
Abstract
The gastrointestinal tract microbiota is involved in the development and function of many body processes. Studies demonstrate that early-life microbial colonisation is the most important time for shaping intestinal and immune development, with perturbations to the microbiota during this time having long-lasting negative implications for the host. Piglets face many early-life events that shape the acquisition and development of their intestinal microbiota. The pork industry has a unique advantage in that the producer has a degree of control over what piglets are exposed to, providing conditions that allow for optimum piglet growth and development. An influx of publications within this area has occurred in recent times and with this, interest surrounding its application in pork production has increased. However, it can be difficult to distinguish which research is of most relevance to industry in terms of delivering repeatable and reliable production outcomes. In this review, we describe the literature surrounding research within pigs, predominantly during the preweaning period that has either provided solutions to industry problems or is generating information targeted at addressing relevant industry issues, with the focus being on studies demonstrating causation where possible. This review will provide a basis for the development of new studies targeted at understanding how to better support initial intestinal microbiota colonisation in order to improve piglet health and survival.
Collapse
Affiliation(s)
- T L Nowland
- Livestock Sciences, South Australian Research and Development Institute, PPPI Building, University of Adelaide, Roseworthy, SA 5371, Australia.
| | - R N Kirkwood
- School of Animal and Veterinary Sciences, The University of Adelaide, Roseworthy, SA 5371, Australia
| | - J R Pluske
- Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, VIC 3010, Australia
| |
Collapse
|
133
|
Shiba T, Komatsu K, Sudo T, Sawafuji R, Saso A, Ueda S, Watanabe T, Nemoto T, Kano C, Nagai T, Ohsugi Y, Katagiri S, Takeuchi Y, Kobayashi H, Iwata T. Comparison of Periodontal Bacteria of Edo and Modern Periods Using Novel Diagnostic Approach for Periodontitis With Micro-CT. Front Cell Infect Microbiol 2021; 11:723821. [PMID: 34616690 PMCID: PMC8488429 DOI: 10.3389/fcimb.2021.723821] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 08/17/2021] [Indexed: 11/07/2022] Open
Abstract
Ancient dental calculus, formed from dental plaque, is a rich source of ancient DNA and can provide information regarding the food and oral microbiology at that time. Genomic analysis of dental calculus from Neanderthals has revealed the difference in bacterial composition of oral microbiome between Neanderthals and modern humans. There are few reports investigating whether the pathogenic bacteria of periodontitis, a polymicrobial disease induced in response to the accumulation of dental plaque, were different between ancient and modern humans. This study aimed to compare the bacterial composition of the oral microbiome in ancient and modern human samples and to investigate whether lifestyle differences depending on the era have altered the bacterial composition of the oral microbiome and the causative bacteria of periodontitis. Additionally, we introduce a novel diagnostic approach for periodontitis in ancient skeletons using micro-computed tomography. Ancient 16S rDNA sequences were obtained from 12 samples at the Unko-in site (18th-19th century) of the Edo era (1603–1867), a characteristic period in Japan when immigrants were not accepted. Furthermore, modern 16S rDNA data from 53 samples were obtained from a database to compare the modern and ancient microbiome. The microbial co-occurrence network was analyzed based on 16S rDNA read abundance. Eubacterium species, Mollicutes species, and Treponema socranskii were the core species in the Edo co-occurrence network. The co-occurrence relationship between Actinomyces oricola and Eggerthella lenta appeared to have played a key role in causing periodontitis in the Edo era. However, Porphyromonas gingivalis, Fusobacterium nucleatum subsp. vincentii, and Prevotella pleuritidis were the core and highly abundant species in the co-occurrence network of modern samples. These results suggest the possibility of differences in the pathogens causing periodontitis during different eras in history.
Collapse
Affiliation(s)
- Takahiko Shiba
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Keiji Komatsu
- Department of Lifetime Oral Health Care Sciences, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Takeaki Sudo
- Institute of Education, Tokyo Medical and Dental University, Tokyo, Japan
| | - Rikai Sawafuji
- Department of Evolutionary Studies of Biosystems, The Graduate University for Advanced Studies (SOKENDAI), Kanagawa, Japan
| | - Aiko Saso
- Department of Physical Therapy, Faculty of Rehabilitation, Niigata University of Health and Welfare, Niigata, Japan
| | - Shintaroh Ueda
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan.,Department of Legal Medicine, Toho University School of Medicine, Tokyo, Japan
| | - Takayasu Watanabe
- Department of Chemistry, Nihon University School of Dentistry, Tokyo, Japan
| | - Takashi Nemoto
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Chihiro Kano
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Takahiko Nagai
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yujin Ohsugi
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Sayaka Katagiri
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yasuo Takeuchi
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hiroaki Kobayashi
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Takanori Iwata
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
134
|
Childs CE, Munblit D, Ulfman L, Gómez-Gallego C, Lehtoranta L, Recker T, Salminen S, Tiemessen M, Collado MC. Potential Biomarkers, Risk Factors and their Associations with IgE-mediated Food Allergy in Early Life: A Narrative Review. Adv Nutr 2021; 13:S2161-8313(22)00081-3. [PMID: 34596662 PMCID: PMC8970818 DOI: 10.1093/advances/nmab122] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Food allergy affects the quality of life of millions of people worldwide and presents a significant psychological and financial burden for both national and international public health. In the past few decades, the prevalence of allergic disease has been on the rise worldwide. Identified risk factors for food allergy include family history, mode of delivery, variations in infant feeding practices, prior diagnosis of other atopic diseases such as eczema, and social economic status. Identifying reliable biomarkers which predict the risk of developing food allergy in early life would be valuable in both preventing morbidity and mortality and by making current interventions available at the earliest opportunity. There is also the potential to identify new therapeutic targets. This narrative review provides details on the genetic, epigenetic, dietary and microbiome influences upon the development of food allergy and synthesizes the currently available data indicating potential biomarkers. While there is a large body of research evidence available within each field of potential risk factors, there are very limited number of studies which span multiple methodological fields, for example including immunology, microbiome, genetic/epigenetic factors and dietary assessment. We recommend that further collaborative research with detailed cohort phenotyping is required to identify biomarkers, and whether these vary between at-risk populations and the wider population. The low incidence of oral food challenge confirmed food allergy in the general population, and the complexities of designing nutritional intervention studies will provide challenges for researchers to address in generating high quality, reliable and reproducible research findings. STATEMENT OF SIGNIFICANCE Food allergy affects the quality of life of millions of people worldwide and presents a significant psychological and financial burden for both national and international public health. Identifying reliable biomarkers which predict the risk of developing food allergy would be valuable in both preventing morbidity and mortality and by making current interventions available at the earliest opportunity. This review provides details on the genetic, epigenetic, dietary and microbiome influences upon the development of food allergy. This helps in identifying reliable biomarkers to predict the risk of developing food allergy, which could be valuable in both preventing morbidity and mortality and by making interventions available at the earliest opportunity.
Collapse
Affiliation(s)
- Caroline E Childs
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, United Kingdom,Institute for Life Sciences, University of Southampton, Southampton, United Kingdom
| | - Daniel Munblit
- Imperial College London, London, United Kingdom,Department of Paediatrics and Paediatric Infectious Diseases, Institute of Child’s Health, Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia,Inflammation, Repair and Development Section, National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, United Kingdom
| | | | - Carlos Gómez-Gallego
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | | | | | | | | | | |
Collapse
|
135
|
Bornbusch SL, Harris RL, Grebe NM, Roche K, Dimac-Stohl K, Drea CM. Antibiotics and fecal transfaunation differentially affect microbiota recovery, associations, and antibiotic resistance in lemur guts. Anim Microbiome 2021; 3:65. [PMID: 34598739 PMCID: PMC8485508 DOI: 10.1186/s42523-021-00126-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 09/19/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Antibiotics alter the diversity, structure, and dynamics of host-associated microbial consortia, including via development of antibiotic resistance; however, patterns of recovery from microbial imbalances and methods to mitigate associated negative effects remain poorly understood, particularly outside of human-clinical and model-rodent studies that focus on outcome over process. To improve conceptual understanding of host-microbe symbiosis in more naturalistic contexts, we applied an ecological framework to a non-traditional, strepsirrhine primate model via long-term, multi-faceted study of microbial community structure before, during, and following two experimental manipulations. Specifically, we administered a broad-spectrum antibiotic, either alone or with subsequent fecal transfaunation, to healthy, male ring-tailed lemurs (Lemur catta), then used 16S rRNA and shotgun metagenomic sequencing to longitudinally track the diversity, composition, associations, and resistomes of their gut microbiota both within and across baseline, treatment, and recovery phases. RESULTS Antibiotic treatment resulted in a drastic decline in microbial diversity and a dramatic alteration in community composition. Whereas microbial diversity recovered rapidly regardless of experimental group, patterns of microbial community composition reflected long-term instability following treatment with antibiotics alone, a pattern that was attenuated by fecal transfaunation. Covariation analysis revealed that certain taxa dominated bacterial associations, representing potential keystone species in lemur gut microbiota. Antibiotic resistance genes, which were universally present, including in lemurs that had never been administered antibiotics, varied across individuals and treatment groups. CONCLUSIONS Long-term, integrated study post antibiotic-induced microbial imbalance revealed differential, metric-dependent evidence of recovery, with beneficial effects of fecal transfaunation on recovering community composition, and potentially negative consequences to lemur resistomes. Beyond providing new perspectives on the dynamics that govern host-associated communities, particularly in the Anthropocene era, our holistic study in an endangered species is a first step in addressing the recent, interdisciplinary calls for greater integration of microbiome science into animal care and conservation.
Collapse
Affiliation(s)
| | - Rachel L. Harris
- Department of Evolutionary Anthropology, Duke University, Durham, USA
| | - Nicholas M. Grebe
- Department of Evolutionary Anthropology, Duke University, Durham, USA
| | - Kimberly Roche
- Program in Computational Biology & Bioinformatics, Duke University, Durham, USA
| | | | - Christine M. Drea
- Department of Evolutionary Anthropology, Duke University, Durham, USA
| |
Collapse
|
136
|
Harkins P, Burke E, Swales C, Silman A. 'All disease begins in the gut'-the role of the intestinal microbiome in ankylosing spondylitis. Rheumatol Adv Pract 2021; 5:rkab063. [PMID: 34557624 PMCID: PMC8452999 DOI: 10.1093/rap/rkab063] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 07/26/2021] [Indexed: 12/21/2022] Open
Abstract
Ankylosing spondylitis is a chronic, debilitating arthritis with a predilection for the axial skeleton. It has a strong genetic predisposition, but the precise pathogenetic mechanisms involved in its development have not yet been fully elucidated. This has implications both for early diagnosis and for effective management. Recently, alterations in the intestinal microbiome have been implicated in disease pathogenesis. In this review, we summarize studies assessing the intestinal microbiome in AS pathogenesis, in addition to synthesizing the literature exploring the postulated mechanisms by which it exerts it pathogenic potential. Finally, we review studies analysing manipulation of the microbiome as a potential therapeutic avenue in AS management.
Collapse
Affiliation(s)
- Patricia Harkins
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Eoghan Burke
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Catherine Swales
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Alan Silman
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| |
Collapse
|
137
|
Glycosaminoglycan biosynthesis pathway in host genome is associated with Helicobacter pylori infection. Sci Rep 2021; 11:18235. [PMID: 34521966 PMCID: PMC8440747 DOI: 10.1038/s41598-021-97790-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 08/31/2021] [Indexed: 02/08/2023] Open
Abstract
Helicobacter pylori is a causative pathogen of many gastric and extra-gastric diseases. It has infected about half of the global population. There were no genome-wide association studies (GWAS) for H. pylori infection conducted in Chinese population, who carried different and relatively homogenous strain of H. pylori. In this work, we performed SNP (single nucleotide polymorphism)-based, gene-based and pathway-based genome-wide association analyses to investigate the genetic basis of host susceptibility to H. pylori infection in 480 Chinese individuals. We also profiled the composition and function of the gut microbiota between H. pylori infection cases and controls. We found several genes and pathways associated with H. pylori infection (P < 0.05), replicated one previously reported SNP rs10004195 in TLR1 gene region (P = 0.02). We also found that glycosaminoglycan biosynthesis related pathway was associated with both onset and progression of H. pylori infection. In the gut microbiome association study, we identified 2 species, 3 genera and several pathways had differential abundance between H. pylori infected cases and controls. This paper is the first GWAS for H. pylori infection in Chinese population, and we combined the genetic and microbial data to comprehensively discuss the basis of host susceptibility to H. pylori infection.
Collapse
|
138
|
Zhu B, Chan SL, Li J, Li K, Wu H, Cui K, Chen H. Non-alcoholic Steatohepatitis Pathogenesis, Diagnosis, and Treatment. Front Cardiovasc Med 2021; 8:742382. [PMID: 34557535 PMCID: PMC8452937 DOI: 10.3389/fcvm.2021.742382] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 08/13/2021] [Indexed: 12/12/2022] Open
Abstract
There has been a rise in the prevalence of non-alcohol fatty liver disease (NAFLD) due to the popularity of western diets and sedentary lifestyles. One quarter of NAFLD patients is diagnosed with non-alcoholic steatohepatitis (NASH), with histological evidence not only of fat accumulation in hepatocytes but also of liver cell injury and death due to long-term inflammation. Severe NASH patients have increased risks of cirrhosis and liver cancer. In this review, we discuss the pathogenesis and current methods of diagnosis for NASH, and current status of drug development for this life-threatening liver disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Hong Chen
- Department of Surgery, Vascular Biology Program, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States
| |
Collapse
|
139
|
Misiakiewicz-Has K, Maciejewska-Markiewicz D, Rzeszotek S, Pilutin A, Kolasa A, Szumilas P, Stachowska E, Wiszniewska B. The Obscure Effect of Tribulus terrestris Saponins Plus Inulin on Liver Morphology, Liver Fatty Acids, Plasma Glucose, and Lipid Profile in SD Rats with and without Induced Type 2 Diabetes Mellitus. Int J Mol Sci 2021; 22:ijms22168680. [PMID: 34445384 PMCID: PMC8395419 DOI: 10.3390/ijms22168680] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/01/2021] [Accepted: 08/09/2021] [Indexed: 01/08/2023] Open
Abstract
Diabetes is a predictor of nonalcoholic fatty liver disease (NAFLD). There are data suggesting that Tribulus terrestris (TT) saponins act as antidiabetic agents and protect against NAFLD. The effect of saponins may be increased by fermentable fibers such as inulin. The aim of the present study was to investigate the influence of TT saponins and TT saponins plus inulin on the plasma lipid profile and liver fatty acids of rats with induced diabetes mellitus type 2 (T2DM). The study was performed on 36 male Sprague–Dawley rats divided into two main groups: control and diabetic. Animals of the diabetic (DM) group were fed a high-fat diet and injected with streptozotocin (low doses). Animals of the control group (nDM) were on a regular diet and were injected with buffer. After the injections, the animals were split into subgroups: three non-diabetic (nDM): (i) control (c-C); (ii) saponin-treated rats (C-Sap); (iii) rats treated with saponins + inulin (C-Sap + IN), and three diabetic subgroups (DM): (iv) control (c-DM); (v) saponin-treated rats (DM-Sap); (vi) rats treated with saponins + inulin (DM-Sap + IN). Liver fatty acids were extracted and analyzed by gas chromatography, and plasma glucose and lipids were measured. The study showed significant changes in liver morphology, liver fatty acids, plasma lipid profile, and plasma glucose. In summary, supplementation with TT saponins or saponins with inulin for one month decreased the level of steatosis in rats with induced type 2 diabetes. Moreover, there were favorable effects on the plasma lipid profile in the rats. However, additional supplementation with inulin had a negative effect on liver morphology (with a microvesicular type of steatosis) in the non-diabetes group. Moreover, supplementation with inulin had a negative effect on plasma glucose in both diabetic and non-diabetic rats. These data show that a diet enriched with fermentable fibers reveals different effects in different organisms, and not all sources and forms of fiber are beneficial to health.
Collapse
Affiliation(s)
- Kamila Misiakiewicz-Has
- Department of Histology and Embryology, Pomeranian Medical University in Szczecin, 70-111 Szczecin, Poland; (S.R.); (A.P.); (A.K.); (B.W.)
- Correspondence:
| | - Dominika Maciejewska-Markiewicz
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University in Szczecin, 70-204 Szczecin, Poland; (D.M.-M.); (E.S.)
| | - Sylwia Rzeszotek
- Department of Histology and Embryology, Pomeranian Medical University in Szczecin, 70-111 Szczecin, Poland; (S.R.); (A.P.); (A.K.); (B.W.)
| | - Anna Pilutin
- Department of Histology and Embryology, Pomeranian Medical University in Szczecin, 70-111 Szczecin, Poland; (S.R.); (A.P.); (A.K.); (B.W.)
| | - Agnieszka Kolasa
- Department of Histology and Embryology, Pomeranian Medical University in Szczecin, 70-111 Szczecin, Poland; (S.R.); (A.P.); (A.K.); (B.W.)
| | - Paweł Szumilas
- Department of Social Medicine and Public Health, Pomeranian Medical University in Szczecin, 48 Żołnierska Str., 71-210 Szczecin, Poland;
| | - Ewa Stachowska
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University in Szczecin, 70-204 Szczecin, Poland; (D.M.-M.); (E.S.)
| | - Barbara Wiszniewska
- Department of Histology and Embryology, Pomeranian Medical University in Szczecin, 70-111 Szczecin, Poland; (S.R.); (A.P.); (A.K.); (B.W.)
| |
Collapse
|
140
|
Lacourt-Ventura MY, Vilanova-Cuevas B, Rivera-Rodríguez D, Rosario-Acevedo R, Miranda C, Maldonado-Martínez G, Maysonet J, Vargas D, Ruiz Y, Hunter-Mellado R, Cubano LA, Dharmawardhane S, Lampe JW, Baerga-Ortiz A, Godoy-Vitorino F, Martínez-Montemayor MM. Soy and Frequent Dairy Consumption with Subsequent Equol Production Reveals Decreased Gut Health in a Cohort of Healthy Puerto Rican Women. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18168254. [PMID: 34444002 PMCID: PMC8391519 DOI: 10.3390/ijerph18168254] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/23/2021] [Accepted: 07/26/2021] [Indexed: 11/16/2022]
Abstract
The U.S. Hispanic female population has one of the highest breast cancer (BC) incidence and mortality rates, while BC is the leading cause of cancer death in Puerto Rican women. Certain foods may predispose to carcinogenesis. Our previous studies indicate that consuming combined soy isoflavones (genistein, daidzein, and glycitein) promotes tumor metastasis possibly through increased protein synthesis activated by equol, a secondary dietary metabolite. Equol is a bacterial metabolite produced in about 20-60% of the population that harbor and exhibit specific gut microbiota capable of producing it from daidzein. The aim of the current study was to investigate the prevalence of equol production in Puerto Rican women and identify the equol producing microbiota in this understudied population. Herein, we conducted a cross-sectional characterization of equol production in a clinically based sample of eighty healthy 25-50 year old Puerto Rican women. Urine samples were collected and evaluated by GCMS for the presence of soy isoflavones and metabolites to determine the ratio of equol producers to equol non-producers. Furthermore, fecal samples were collected for gut microbiota characterization on a subset of women using next generation sequencing (NGS). We report that 25% of the participants were classified as equol producers. Importantly, the gut microbiota from equol non-producers demonstrated a higher diversity. Our results suggest that healthy women with soy and high dairy consumption with subsequent equol production may result in gut dysbiosis by having reduced quantities (diversity) of healthy bacterial biomarkers, which might be associated to increased diseased outcomes (e.g., cancer, and other diseases).
Collapse
Affiliation(s)
- Mercedes Y. Lacourt-Ventura
- Department of Biochemistry, School of Medicine, Universidad Central del Caribe, Bayamón 00956, Puerto Rico; (M.Y.L.-V.); (R.R.-A.); (D.V.); (L.A.C.)
| | - Brayan Vilanova-Cuevas
- Department of Microbiology and Medical Zoology, Medical Sciences Campus, University of Puerto Rico, San Juan 00921, Puerto Rico; (B.V.-C.); (F.G.-V.)
| | | | - Raysa Rosario-Acevedo
- Department of Biochemistry, School of Medicine, Universidad Central del Caribe, Bayamón 00956, Puerto Rico; (M.Y.L.-V.); (R.R.-A.); (D.V.); (L.A.C.)
| | - Christine Miranda
- Retrovirus Research Center, Internal Medicine Department, School of Medicine, Universidad Central del Caribe, Bayamón 00956, Puerto Rico; (C.M.); (G.M.-M.); (J.M.); (Y.R.); (R.H.-M.)
| | - Gerónimo Maldonado-Martínez
- Retrovirus Research Center, Internal Medicine Department, School of Medicine, Universidad Central del Caribe, Bayamón 00956, Puerto Rico; (C.M.); (G.M.-M.); (J.M.); (Y.R.); (R.H.-M.)
| | - Johanna Maysonet
- Retrovirus Research Center, Internal Medicine Department, School of Medicine, Universidad Central del Caribe, Bayamón 00956, Puerto Rico; (C.M.); (G.M.-M.); (J.M.); (Y.R.); (R.H.-M.)
- Hematology and Oncology Group, HIMA-San Pablo Bayamón Hospital, Bayamón 00961, Puerto Rico
| | - Darlene Vargas
- Department of Biochemistry, School of Medicine, Universidad Central del Caribe, Bayamón 00956, Puerto Rico; (M.Y.L.-V.); (R.R.-A.); (D.V.); (L.A.C.)
| | - Yelitza Ruiz
- Retrovirus Research Center, Internal Medicine Department, School of Medicine, Universidad Central del Caribe, Bayamón 00956, Puerto Rico; (C.M.); (G.M.-M.); (J.M.); (Y.R.); (R.H.-M.)
- Hematology and Oncology Group, HIMA-San Pablo Bayamón Hospital, Bayamón 00961, Puerto Rico
| | - Robert Hunter-Mellado
- Retrovirus Research Center, Internal Medicine Department, School of Medicine, Universidad Central del Caribe, Bayamón 00956, Puerto Rico; (C.M.); (G.M.-M.); (J.M.); (Y.R.); (R.H.-M.)
- Hematology and Oncology Group, HIMA-San Pablo Bayamón Hospital, Bayamón 00961, Puerto Rico
| | - Luis A. Cubano
- Department of Biochemistry, School of Medicine, Universidad Central del Caribe, Bayamón 00956, Puerto Rico; (M.Y.L.-V.); (R.R.-A.); (D.V.); (L.A.C.)
| | - Suranganie Dharmawardhane
- Department of Biochemistry, Medical Sciences Campus, University of Puerto Rico, San Juan 00921, Puerto Rico; (S.D.); (A.B.-O.)
| | - Johanna W. Lampe
- Fred Hutchinson Cancer Research Center, Division of Public Health Sciences, Seattle, WA 98109, USA;
| | - Abel Baerga-Ortiz
- Department of Biochemistry, Medical Sciences Campus, University of Puerto Rico, San Juan 00921, Puerto Rico; (S.D.); (A.B.-O.)
| | - Filipa Godoy-Vitorino
- Department of Microbiology and Medical Zoology, Medical Sciences Campus, University of Puerto Rico, San Juan 00921, Puerto Rico; (B.V.-C.); (F.G.-V.)
| | - Michelle M. Martínez-Montemayor
- Department of Biochemistry, School of Medicine, Universidad Central del Caribe, Bayamón 00956, Puerto Rico; (M.Y.L.-V.); (R.R.-A.); (D.V.); (L.A.C.)
- Correspondence: ; Tel.: +1-787-798-3001 (ext. 2152)
| |
Collapse
|
141
|
Chalifour B, Li J. Characterization of the gut microbiome in wild rocky mountainsnails (Oreohelix strigosa). Anim Microbiome 2021; 3:49. [PMID: 34274024 PMCID: PMC8285866 DOI: 10.1186/s42523-021-00111-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 07/04/2021] [Indexed: 11/30/2022] Open
Abstract
Background The Rocky Mountainsnail (Oreohelix strigosa) is a terrestrial gastropod of ecological importance in the Rocky Mountains of western United States and Canada. Across the animal kingdom, including in gastropods, gut microbiomes have profound effects on the health of the host. Current knowledge regarding snail gut microbiomes, particularly throughout various life history stages, is limited. Understanding snail gut microbiome composition and dynamics can provide an initial step toward better conservation and management of this species. Results In this study, we employed 16S rRNA gene amplicon sequencing to examine gut bacteria communities in wild-caught O. strigosa populations from the Front Range of Colorado. These included three treatment groups: (1) adult and (2) fetal snails, as well as (3) sub-populations of adult snails that were starved prior to ethanol fixation. Overall, O. strigosa harbors a high diversity of bacteria. We sequenced the V4 region of the 16S rRNA gene on an Illumina MiSeq and obtained 2,714,330 total reads. We identified a total of 7056 unique operational taxonomic units (OTUs) belonging to 36 phyla. The core gut microbiome of four unique OTUs accounts for roughly half of all sequencing reads returned and may aid the snails’ digestive processes. Significant differences in microbial composition, as well as richness, evenness, and Shannon Indices were found across the three treatment groups. Conclusions Comparisons of gut microbiomes in O. strigosa adult, fetal, and starved samples provide evidence that the host internal environments influence bacterial community compositions, and that bacteria may be transmitted vertically from parent to offspring. This work provides the first comprehensive report on the structure and membership of bacterial populations in the gastropod family Oreohelicidae and reveals similarities and differences across varying life history metrics. Strong differentiation between these life history metrics demonstrates the need for wider sampling for studies of dynamics of the snail gut microbiome. Supplementary Information The online version contains supplementary material available at 10.1186/s42523-021-00111-6.
Collapse
Affiliation(s)
- Bridget Chalifour
- Department of Ecology and Evolutionary Biology, University of Colorado Boulder, 1900 Pleasant Street, 334 UCB, Boulder, CO, 80309, USA.
| | - Jingchun Li
- Department of Ecology and Evolutionary Biology, University of Colorado Boulder, 1900 Pleasant Street, 334 UCB, Boulder, CO, 80309, USA.,Museum of Natural History, University of Colorado Boulder, 265 UCB, Boulder, CO, 80309, USA
| |
Collapse
|
142
|
Nutrición y enfermedad inflamatoria intestinal: posibles mecanismos en la incidencia y manejo. REVISTA MÉDICA CLÍNICA LAS CONDES 2021. [DOI: 10.1016/j.rmclc.2021.01.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
143
|
Lee KS, Jeong YJ, Lee MS. Escherichia coli Shiga Toxins and Gut Microbiota Interactions. Toxins (Basel) 2021; 13:toxins13060416. [PMID: 34208170 PMCID: PMC8230793 DOI: 10.3390/toxins13060416] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 06/03/2021] [Accepted: 06/08/2021] [Indexed: 12/19/2022] Open
Abstract
Escherichia coli (EHEC) and Shigella dysenteriae serotype 1 are enterohemorrhagic bacteria that induce hemorrhagic colitis. This, in turn, may result in potentially lethal complications, such as hemolytic uremic syndrome (HUS), which is characterized by thrombocytopenia, acute renal failure, and neurological abnormalities. Both species of bacteria produce Shiga toxins (Stxs), a phage-encoded exotoxin inhibiting protein synthesis in host cells that are primarily responsible for bacterial virulence. Although most studies have focused on the pathogenic roles of Stxs as harmful substances capable of inducing cell death and as proinflammatory factors that sensitize the host target organs to damage, less is known about the interface between the commensalism of bacterial communities and the pathogenicity of the toxins. The gut contains more species of bacteria than any other organ, providing pathogenic bacteria that colonize the gut with a greater number of opportunities to encounter other bacterial species. Notably, the presence in the intestines of pathogenic EHEC producing Stxs associated with severe illness may have compounding effects on the diversity of the indigenous bacteria and bacterial communities in the gut. The present review focuses on studies describing the roles of Stxs in the complex interactions between pathogenic Shiga toxin-producing E. coli, the resident microbiome, and host tissues. The determination of these interactions may provide insights into the unresolved issues regarding these pathogens.
Collapse
Affiliation(s)
- Kyung-Soo Lee
- Environmental Diseases Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Daejeon 34141, Korea;
- Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), 127 Gajeong-ro, Yuseong-gu, Daejeon 34113, Korea
| | - Yu-Jin Jeong
- Environmental Diseases Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Daejeon 34141, Korea;
- Correspondence: (Y.-J.J.); (M.-S.L.)
| | - Moo-Seung Lee
- Environmental Diseases Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Daejeon 34141, Korea;
- Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), 127 Gajeong-ro, Yuseong-gu, Daejeon 34113, Korea
- Correspondence: (Y.-J.J.); (M.-S.L.)
| |
Collapse
|
144
|
Ferreira RLU, Sena-Evangelista KCM, de Azevedo EP, Pinheiro FI, Cobucci RN, Pedrosa LFC. Selenium in Human Health and Gut Microflora: Bioavailability of Selenocompounds and Relationship With Diseases. Front Nutr 2021; 8:685317. [PMID: 34150830 PMCID: PMC8211732 DOI: 10.3389/fnut.2021.685317] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 05/11/2021] [Indexed: 12/12/2022] Open
Abstract
This review covers current knowledge of selenium in the dietary intake, its bioavailability, metabolism, functions, biomarkers, supplementation and toxicity, as well as its relationship with diseases and gut microbiota specifically on the symbiotic relationship between gut microflora and selenium status. Selenium is essential for the maintenance of the immune system, conversion of thyroid hormones, protection against the harmful action of heavy metals and xenobiotics as well as for the reduction of the risk of chronic diseases. Selenium is able to balance the microbial flora avoiding health damage associated with dysbiosis. Experimental studies have shown that inorganic and organic selenocompounds are metabolized to selenomethionine and incorporated by bacteria from the gut microflora, therefore highlighting their role in improving the bioavailability of selenocompounds. Dietary selenium can affect the gut microbial colonization, which in turn influences the host's selenium status and expression of selenoproteoma. Selenium deficiency may result in a phenotype of gut microbiota that is more susceptible to cancer, thyroid dysfunctions, inflammatory bowel disease, and cardiovascular disorders. Although the host and gut microbiota benefit each other from their symbiotic relationship, they may become competitors if the supply of micronutrients is limited. Intestinal bacteria can remove selenium from the host resulting in two to three times lower levels of host's selenoproteins under selenium-limiting conditions. There are still gaps in whether these consequences are unfavorable to humans and animals or whether the daily intake of selenium is also adapted to meet the needs of the bacteria.
Collapse
Affiliation(s)
| | - Karine Cavalcanti Maurício Sena-Evangelista
- Postgraduate Program in Nutrition, Federal University of Rio Grande do Norte, Natal, Brazil.,Department of Nutrition, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Eduardo Pereira de Azevedo
- Graduate Program of Biotechnology, Laureate International Universities - Universidade Potiguar, Natal, Brazil
| | - Francisco Irochima Pinheiro
- Graduate Program of Biotechnology, Laureate International Universities - Universidade Potiguar, Natal, Brazil.,Medical School, Laureate International Universities - Universidade Potiguar, Natal, Brazil
| | - Ricardo Ney Cobucci
- Graduate Program of Biotechnology, Laureate International Universities - Universidade Potiguar, Natal, Brazil.,Medical School, Laureate International Universities - Universidade Potiguar, Natal, Brazil
| | - Lucia Fatima Campos Pedrosa
- Postgraduate Program in Nutrition, Federal University of Rio Grande do Norte, Natal, Brazil.,Department of Nutrition, Federal University of Rio Grande do Norte, Natal, Brazil
| |
Collapse
|
145
|
Peng F, Zhang H, He X, Song Z. Effects of Ursolic Acid on Intestinal Health and Gut Bacteria Antibiotic Resistance in Mice. Front Physiol 2021; 12:650190. [PMID: 34122127 PMCID: PMC8195277 DOI: 10.3389/fphys.2021.650190] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 05/05/2021] [Indexed: 01/04/2023] Open
Abstract
Ursolic acid (UA), a natural pentacyclic triterpenoid, has been widely reported to exert anti-oxidant and anti-inflammatory properties. However, the effects of UA on the intestinal homeostasis and gut microbiota were rarely explored. The aim of the present study was to investigate the effects of UA on intestinal health and gut microflora antibiotic-resistance in antibiotic-exposed mice. Kunming mice (n = 80) were randomly allocated into three groups and fed with one of the following diets, respectively: Cont group (n = 20), the basal diet; UA group (n = 20), the basal diet supplemented with 150 mg/kg UA; Tet group (n = 40), the basal diet supplemented with 659 mg/kg chlortetracycline. After 14 days, 10 mice in each group were euthanatized and the remaining 30 mice in the Tet group were randomly allocated into three sub-groups (n = 10 per group) as follows: the Tet group which were kept feeding a Tet diet for 14 days; the Natural Restoration (NatR) group which received a basal diet for 14 days; and the UA therapy (UaT) group which fed a basal diet supplemented with 150 mg/kg UA for 14 days. Throughout the experiment, the weight and the food intake of each mouse were recorded once weekly. Serum LPS and diamine oxidase (DAO), jejunal morphology, jejunal tight junction proteins and nutrient transporters, colonic inflammatory cytokines, gut microbiota and its antibiotic resistance gene (ARG) were examined at euthanasia. The results showed that UA treatment significantly increased average daily food intake (ADFI) of mice. Notably, UA increased the jejunal villi height, decreased the jejunal crypt depth and promoted the expression of jejunum nutrient transporters. UaT group had higher villi height, lower crypt depth and higher nutrient transporter mRNA expression in jejunum than NatR group. Besides, UA decreased serum DAO content, upregulated mRNA expression of ZO-1, claudin-1 and occludin and downregulated TNF-α and IL-6. The mRNA abundances of ZO-1, claudin-1 and occludin and TNF-α and IL-6 in UaT group were, respectively upregulated and downregulated than NatR group. Furthermore, an analysis of 16S rDNA sequences demonstrated that UA increased the abundance of beneficial bacteria in the gut. And the results of ARG test showed that UA downregulated the expression of antibiotic-induced resistance genes. The UaT group inhibited the increase of harmful bacteria abundance and suppressed the mRNA abundances of ARG compared to the NatR group. In conclusion, considering the positive effects of UA on the growth performance and intestinal mucosal barrier, we anticipate that these findings could be a stepping stone for developing UA as a novel substitute of antibiotics.
Collapse
Affiliation(s)
- Fang Peng
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China.,Ministry of Education Engineering Research Center of Feed Safety and Efficient Use, Changsha, China
| | - Haihan Zhang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China.,Ministry of Education Engineering Research Center of Feed Safety and Efficient Use, Changsha, China
| | - Xi He
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China.,Ministry of Education Engineering Research Center of Feed Safety and Efficient Use, Changsha, China
| | - Zehe Song
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China.,Ministry of Education Engineering Research Center of Feed Safety and Efficient Use, Changsha, China
| |
Collapse
|
146
|
Ren X, Wang L, Chen Z, Hou D, Xue Y, Diao X, Shen Q. Foxtail Millet Improves Blood Glucose Metabolism in Diabetic Rats through PI3K/AKT and NF-κB Signaling Pathways Mediated by Gut Microbiota. Nutrients 2021; 13:nu13061837. [PMID: 34072141 PMCID: PMC8228963 DOI: 10.3390/nu13061837] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/22/2021] [Accepted: 05/25/2021] [Indexed: 12/11/2022] Open
Abstract
Foxtail millet (FM) is receiving ongoing increased attention due to its beneficial health effects, including the hypoglycemic effect. However, the underlying mechanisms of the hypoglycemic effect have been underexplored. In the present study, the hypoglycemic effect of FM supplementation was confirmed again in high-fat diet and streptozotocin-induced diabetic rats with significantly decreased fasting glucose (FG), glycated serum protein, and areas under the glucose tolerance test (p < 0.05). We employed 16S rRNA and liver RNA sequencing technologies to identify the target gut microbes and signaling pathways involved in the hypoglycemic effect of FM supplementation. The results showed that FM supplementation significantly increased the relative abundance of Lactobacillus and Ruminococcus_2, which were significantly negatively correlated with FG and 2-h glucose. FM supplementation significantly reversed the trends of gene expression in diabetic rats. Specifically, FM supplementation inhibited gluconeogenesis, stimulated glycolysis, and restored fatty acid synthesis through activation of the PI3K/AKT signaling pathway. FM also reduced inflammation through inhibition of the NF-κB signaling pathway. Spearman’s correlation analysis indicated a complicated set of interdependencies among the gut microbiota, signaling pathways, and metabolic parameters. Collectively, the above results suggest that the hypoglycemic effect of FM was at least partially mediated by the increased relative abundance of Lactobacillus, activation of the PI3K/AKT signaling pathway, and inhibition of the NF-κB signaling pathway.
Collapse
Affiliation(s)
- Xin Ren
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Engineering and Technology Research Center of Food Additives, Beijing Technology and Business University, Beijing 100048, China; (X.R.); (L.W.)
- Key Laboratory of Plant Protein and Grain Processing, National Engineering and Technology Research Center for Fruits and Vegetables, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (D.H.); (Y.X.)
| | - Linxuan Wang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Engineering and Technology Research Center of Food Additives, Beijing Technology and Business University, Beijing 100048, China; (X.R.); (L.W.)
| | - Zenglong Chen
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China;
| | - Dianzhi Hou
- Key Laboratory of Plant Protein and Grain Processing, National Engineering and Technology Research Center for Fruits and Vegetables, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (D.H.); (Y.X.)
| | - Yong Xue
- Key Laboratory of Plant Protein and Grain Processing, National Engineering and Technology Research Center for Fruits and Vegetables, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (D.H.); (Y.X.)
| | - Xianmin Diao
- Center for Crop Germplasm Resources, Institute of Crop Science, Chinese Academy of Agricultural Sciences, Beijing 100081, China;
| | - Qun Shen
- Key Laboratory of Plant Protein and Grain Processing, National Engineering and Technology Research Center for Fruits and Vegetables, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (D.H.); (Y.X.)
- Correspondence: ; Tel.: +86-10-62737524
| |
Collapse
|
147
|
Sitkin SI, Avalueva EB, Oreshko LS, Khavkin AI. Intestinal microbiota and dysbiosis in celiac disease. ROSSIYSKIY VESTNIK PERINATOLOGII I PEDIATRII (RUSSIAN BULLETIN OF PERINATOLOGY AND PEDIATRICS) 2021; 66:116-122. [DOI: 10.21508/1027-4065-2021-66-2-116-122] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Affiliation(s)
- S. I. Sitkin
- Mechnikov North-Western State Medical University; Almazov National Medical Research Centre; State Research Institute of Highly Pure Biopreparations
| | | | | | - A. I. Khavkin
- Veltischev Research and Clinical Institute for Pediatrics at the Pirogov Russian National Research Medical University
| |
Collapse
|
148
|
Zhang L, Guo T, Zhan N, Sun T, Shan A. Effects of the antimicrobial peptide WK3 on diarrhea, growth performance and intestinal health of weaned piglets challenged with enterotoxigenic Escherichia coli K88. Food Nutr Res 2021; 65:3448. [PMID: 34262420 PMCID: PMC8254467 DOI: 10.29219/fnr.v65.3448] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 12/01/2020] [Accepted: 12/16/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Antibiotics are very effective for treating diarrhea in weaned pigs, but the global prohibition of antibiotics makes it urgent to find an alternative to antibiotics. OBJECTIVE An experiment was conducted to determine the antimicrobial activity of a linear trpzip-like β-hairpin antimicrobial peptide WK3 in vivo and to assess its effects on growth performance and intestinal health. DESIGN Thirty-two piglets were weaned at 21 days and housed in individual metabolic cages, which were randomly divided into four groups and were maintained on a corn-soybean meal-based basal diet. Group 1 included a blank group. Groups 2, 3, and 4 were orally infected by feeding with Enterotoxigenic Escherichia coli (ETEC) K88, which was followed by saline treatment (group 2), enrofloxacin injection at a dose of 2.5 mg/kg (group 3), and WK3 injection at a dose of 2 mg/kg (group 4). The experiment lasted for 6 days, and feed and water were provided ad libitum. RESULTS Both WK3 and enrofloxacin effectively attenuated diarrhea and improved growth performance of piglets. Compared with the control group, WK3 significantly improved the villus height in the ileum (P < 0.05) but did not affect the villus height in the duodenum or jejunum. Additionally, we did not observe any obvious difference in crypt depth or villus height/crypt depth among the duodenum, jejunum and ileum (P > 0.05). WK3 also reduced the numbers of Enterococcus spp (P < 0.01) in the cecal contents, and the number of Enterobacterium spp tended to decrease (0.05 < P < 0.1). Moreover, the jejunal mucosa of the WK3 group exhibited lower interleukin-1α (IL-1a; P < 0.01), toll-like receptors-4 (TLR-4; P < 0.05), and myeloid differentiation primary response 88 (MyD88; P < 0.01) messenger ribonucleic acid (mRNA) expression levels. The jejunum of the WK3 group also exhibited an increased antioxidant capacity, reduced concentration of malondialdehyde (MDA; P < 0.05), and enhanced superoxide dismutase (SOD) activity (P < 0.05). CONCLUSIONS WK3 has the potential to replace antibiotics as a new generation feed additive.
Collapse
Affiliation(s)
| | | | - Na Zhan
- Institute of Animal Nutrition, Northeast Agricultural University, Xiangfang District, Harbin, People’s Republic of China
| | - Taotao Sun
- Institute of Animal Nutrition, Northeast Agricultural University, Xiangfang District, Harbin, People’s Republic of China
| | - Anshan Shan
- Institute of Animal Nutrition, Northeast Agricultural University, Xiangfang District, Harbin, People’s Republic of China
| |
Collapse
|
149
|
Arrona Cardoza P, Spillane MB, Morales Marroquin E. Alzheimer's disease and gut microbiota: does trimethylamine N-oxide (TMAO) play a role? Nutr Rev 2021; 80:271-281. [PMID: 33942080 DOI: 10.1093/nutrit/nuab022] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease that affects memory and cognitive function. Clinical evidence has put into question our current understanding of AD development, propelling researchers to look into further avenues. Gut microbiota has emerged as a potential player in AD pathophysiology. Lifestyle factors, such as diet, can have negative effects on the gut microbiota and thus host health. A Western-type diet has been highlighted as a risk factor for both gut microbiota alteration as well as AD development. The gut-derived trimethylamine N-oxide (TMAO) has been previously implied in the development of cardiovascular diseases with recent evidence suggesting a plausible role of TMAO in AD development. Therefore, the main goal of the present review is to provide the reader with potential mechanisms of action through which consumption of a Western-type diet could increase AD risk, by acting through microbiota-produced TMAO. Although a link between TMAO and AD is far from definitive, this review will serve as a call for research into this new area of research.
Collapse
Affiliation(s)
- Pablo Arrona Cardoza
- P. Arrona Cardoza is with the Tecnológico de Monterrey, School of Medicine and Health Science, Monterrey, Nuevo Leon, Mexico. M.B Spillane is with the H.C. Drew School of Health and Human Performance, McNeese State University, Lake Charles, Louisiana, USA. E. Morales Marroquin is with the School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas, USA. E. Morales Marroquin is with the Center for Pediatric Population Health, UTHealth School of Public Health and Children's Health System of Texas, Dallas, Texas, USA
| | - Micheil B Spillane
- P. Arrona Cardoza is with the Tecnológico de Monterrey, School of Medicine and Health Science, Monterrey, Nuevo Leon, Mexico. M.B Spillane is with the H.C. Drew School of Health and Human Performance, McNeese State University, Lake Charles, Louisiana, USA. E. Morales Marroquin is with the School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas, USA. E. Morales Marroquin is with the Center for Pediatric Population Health, UTHealth School of Public Health and Children's Health System of Texas, Dallas, Texas, USA
| | - Elisa Morales Marroquin
- P. Arrona Cardoza is with the Tecnológico de Monterrey, School of Medicine and Health Science, Monterrey, Nuevo Leon, Mexico. M.B Spillane is with the H.C. Drew School of Health and Human Performance, McNeese State University, Lake Charles, Louisiana, USA. E. Morales Marroquin is with the School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas, USA. E. Morales Marroquin is with the Center for Pediatric Population Health, UTHealth School of Public Health and Children's Health System of Texas, Dallas, Texas, USA
| |
Collapse
|
150
|
Cortez APB, Fisberg M, de Morais MB. Intestinal permeability and small intestine bacterial overgrowth in excess weight adolescents. Pediatr Obes 2021; 16:e12741. [PMID: 33089672 DOI: 10.1111/ijpo.12741] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 08/21/2020] [Accepted: 09/28/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Increased intestinal permeability may be associated with certain disorders, such as obesity and small intestine bacterial overgrowth (SIBO). OBJECTIVE This study aimed to investigate intestinal permeability and SIBO in excess weight adolescents. METHODS This cross-sectional study included 67 adolescents with excess weight and 66 normal weight adolescents. Excess weight was defined as a body mass index for age (BMI/age) > +1 SD, which includes having excess body weight and obesity. SIBO was diagnosed by a breath test after the ingestion of lactulose according to the production of hydrogen and methane. Zonulin (haptoglobin) was considered an indicator of intestinal permeability. RESULTS Adolescents with excess weight had a higher height/age Z-score (median [25th; 75th percentile]: +0.6 [-0.4; +1.0]) than those in the normal weight group (-0.1 [-0.6; +0.7]; P = .014). Zonulin (mg/mL) in the excess weight (2.3 [1.5; 3.8]) adolescents was higher than that in the normal weight (1.6 [1.0; 2.2]) adolescents (P < .001). SIBO was diagnosed in 23.3% (31/133) of the adolescents. The adolescents with SIBO had a lower (P < .05) BMI/age (+0.6 [-0.6; +1.9]) and height/age (-0.3 [-0.7; +0.3]) than the adolescents without SIBO (+1.3 [+0.1; +2.6] and +0.2 [-0.5; +1.0], respectively). No association was found between zonulin and SIBO. CONCLUSION Excess weight is associated with increased intestinal permeability. No relationship was found between SIBO and intestinal permeability; however, SIBO was related to lower BMI and height for age Z-scores.
Collapse
Affiliation(s)
- Ana Paula Bidutte Cortez
- Post-Graduate Program in Nutrition, Federal University of São Paulo (Universidade Federal de São Paulo - UNIFESP), São Paulo, Brazil
| | - Mauro Fisberg
- Division of Pediatric Gastroenterology, Federal University of São Paulo (Universidade Federal de São Paulo - UNIFESP), São Paulo, Brazil.,Nutrition and Feeding Difficulties Center-Pensi Institute-Sabará Children's Hospital (Hospital Infantil Sabará), São Paulo, Brazil
| | - Mauro Batista de Morais
- Division of Pediatric Gastroenterology, Federal University of São Paulo (Universidade Federal de São Paulo - UNIFESP), São Paulo, Brazil
| |
Collapse
|