1
|
Chen N, Yao P, Farid MS, Zhang T, Luo Y, Zhao C. Effect of bioactive compounds in processed Camellia sinensis tea on the intestinal barrier. Food Res Int 2025; 199:115383. [PMID: 39658174 DOI: 10.1016/j.foodres.2024.115383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 10/18/2024] [Accepted: 11/14/2024] [Indexed: 12/12/2024]
Abstract
The human intestinal tract plays a pivotal role in safeguarding the body against noxious substances and microbial pathogens by functioning as a barrier. This barrier function is achieved through the combined action of physical, chemical, microbial, and immune components. Tea (Camellia sinensis) is the most widely consumed beverage in the world, and it is consumed and appreciated in a multitude of regions across the globe. Tea can be classified into various categories, including green, white, yellow, oolong, black, and dark teas, based on the specific processing methods employed. In recent times, there has been a notable surge in scientific investigation into the various types of tea. The recent surge in research on tea can be attributed to the plethora of bioactive compounds it contains, including polyphenols, polysaccharides, pigments, and theanine. The processing of different teas affects the active ingredients to varying degrees, resulting in a range of chemical reactions and the formation of different types and quantities of ingredients. The bioactive compounds present in tea are of great importance for the maintenance of the integrity of the intestinal barrier, operating through a variety of mechanisms. This literature review synthesizes scientific studies on the impact of the primary bioactive compounds and different processing methods of tea on the intestinal barrier function. This review places particular emphasis on the exploration of the barrier repair and regulatory effects of these compounds, including the mitigation of damage to different barriers following intestinal diseases. Specifically, the active ingredients in tea can alleviate damage to physical barriers and chemical barriers by regulating barrier protein expression. At the same time, they can also maintain the stability of immune and biological barriers by regulating the expression of inflammatory factors and the metabolism of intestinal flora. This investigation can establish a strong theoretical foundation for the future development of innovative tea products.
Collapse
Affiliation(s)
- Nan Chen
- College of Food Science and Engineering, Jilin University, Changchun 130062, China.
| | - Peng Yao
- College of Food Science and Engineering, Jilin University, Changchun 130062, China.
| | | | - Tiehua Zhang
- College of Food Science and Engineering, Jilin University, Changchun 130062, China.
| | - Yangchao Luo
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT 06269, United States.
| | - Changhui Zhao
- College of Food Science and Engineering, Jilin University, Changchun 130062, China.
| |
Collapse
|
2
|
Hernandez J, Rodriguez JB, Trak-Fellermeier MA, Galvan R, Macchi A, Martinez-Motta P, Palacios C. Suboptimal vitamin D status and overweight/obesity are associated with gut integrity and inflammation in minority children and adolescents: A cross-sectional analysis from the MetA-bone trial. Nutr Res 2025; 133:13-21. [PMID: 39662375 DOI: 10.1016/j.nutres.2024.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 11/18/2024] [Accepted: 11/18/2024] [Indexed: 12/13/2024]
Abstract
Preserving gut integrity is essential to preventing the development of chronic diseases. Several factors are associated with gut integrity and inflammation in adults. However, there is limited evidence in healthy children. This study evaluated the factors associated with gut integrity and inflammation in healthy children participating in the MetA-Bone trial. We hypothesized that age, sex, race/ethnicity, diet, vitamin D, and body composition will be associated with gut integrity and inflammation. Socio-demographic variables were collected with a questionnaire. Measures included markers of gut integrity (Intestinal Fatty Acid Binding Protein; I-FABP), and inflammation (IL-17 and calprotectin) determined by ELISA in 24-h urine and serum; serum 25(OH)D concentration (commercial lab), BMI percentile, and diet (24-h recalls). Analyses included descriptive statistics, chi-square, and adjusted logistic regressions. Participants (n=138) median age was 12.4 (11.1-13.3), 53.6% were male, 9.4% were Black/African American, and 71.1% were Hispanic/Latino. Children with suboptimal vitamin D were 3.35 times more likely to present gut integrity damage (elevated I-FABP) than those with optimal status (P = .005). Overweight/obesity and fructose intake were associated with inflammation (elevated calprotectin) (P < .05). Those with lower gut integrity damage had lower odds of having higher inflammation (P = .021). Other factors were not associated with inflammation. Suboptimal vitamin D status, overweight/obesity and inflammation may compromise the gut integrity in healthy children, suggesting an impairment on the intestinal barrier repair system. More research with a longitudinal design is needed to gain a deeper understanding of the role of additional factors linked to gut integrity and inflammation in healthy children.
Collapse
Affiliation(s)
- Jacqueline Hernandez
- Department of Dietetics and Nutrition, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL, USA.
| | - Jose Bastida Rodriguez
- Department of Dietetics and Nutrition, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL, USA
| | - Maria Angelica Trak-Fellermeier
- Department of Dietetics and Nutrition, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL, USA
| | - Rodolfo Galvan
- Department of Dietetics and Nutrition, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL, USA
| | - Alison Macchi
- Department of Dietetics and Nutrition, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL, USA
| | - Preciosa Martinez-Motta
- Department of Dietetics and Nutrition, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL, USA
| | - Cristina Palacios
- Department of Dietetics and Nutrition, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL, USA
| |
Collapse
|
3
|
Furukawa N, Kobayashi M, Ito M, Matsui H, Ohashi K, Murohara T, Takeda JI, Ueyama J, Hirayama M, Ohno K. Soy protein β-conglycinin ameliorates pressure overload-induced heart failure by increasing short-chain fatty acid (SCFA)-producing gut microbiota and intestinal SCFAs. Clin Nutr 2024; 43:124-137. [PMID: 39447394 DOI: 10.1016/j.clnu.2024.09.045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 09/26/2024] [Accepted: 09/27/2024] [Indexed: 10/26/2024]
Abstract
BACKGROUND AND AIMS Soybeans and their ingredients have antioxidant and anti-inflammatory effects on cardiovascular diseases. β-Conglycinin (β-CG), a major constituent of soy proteins, is protective against obesity, hypertension, and chronic kidney disease, but its effects on heart failure remain to be elucidated. We tested the effects of β-CG on left ventricular (LV) remodeling in pressure overload-induced heart failure. METHODS A transverse aortic constriction (TAC)-induced pressure overload was applied to the heart in 7-week-old C57BL6 male mice that were treated with β-CG, GlcNAc, or sodium propionate. Gut microbiota was analyzed by 16S rRNA sequencing. Fecal short-chain fatty acids (SCFAs) were quantified by GC-MS. The effects of oral antibiotics were examined in β-CG-fed mice. RESULTS β-CG ameliorated impaired cardiac contractions, cardiac hypertrophy, and myocardial fibrosis in TAC-operated mice. As β-CG is a highly glycosylated protein, we examined the effects of GlcNAc. GlcNAc had similar but less efficient effects on LV remodeling compared to β-CG. β-CG increased three major SCFA-producing intestinal bacteria, as well as fecal concentrations of SCFAs, in sham- and TAC-operated mice. Oral administration of antibiotics nullified the effects of β-CG in TAC-operated mice by markedly reducing SCFA-producing intestinal bacteria and fecal SCFAs. In contrast, oral administration of sodium propionate, one of SCFAs, ameliorated LV remodeling in TAC-operated mice to a similar extent as β-CG. CONCLUSIONS β-CG was protective against TAC-induced LV remodeling, which was likely to be mediated by increased SCFA-producing gut microbiota and increased intestinal SCFAs. Modified β-CG and/or derivatives arising from β-CG are expected to be developed as prophylactic and/or therapeutic agents to ameliorate devastating symptoms in heart failure.
Collapse
Affiliation(s)
- Nozomi Furukawa
- Department of Pathophysiological Laboratory Sciences, Nagoya University Graduate School of Medicine, Nagoya, Japan; Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| | - Miku Kobayashi
- Department of Pathophysiological Laboratory Sciences, Nagoya University Graduate School of Medicine, Nagoya, Japan; Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Mikako Ito
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroki Matsui
- Department of Laboratory Sciences, Gunma University Graduate School of Health Sciences, Maebashi, Japan
| | - Koji Ohashi
- Department of Molecular Medicine and Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Toyoaki Murohara
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Jun-Ichi Takeda
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan; Center for One Medicine Innovative Translational Research (COMIT), Institute for Advanced Study, Gifu University, Gifu, Japan
| | - Jun Ueyama
- Department of Pathophysiological Laboratory Sciences, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masaaki Hirayama
- Department of Pathophysiological Laboratory Sciences, Nagoya University Graduate School of Medicine, Nagoya, Japan; Department of Occupational Therapy, Chubu University College of Life and Health Sciences, Kasugai, Japan
| | - Kinji Ohno
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan; Graduate School of Nutritional Sciences, Nagoya University of Arts and Sciences, Nisshin, Japan
| |
Collapse
|
4
|
Yan H, Hu Y, Liang J, He K, Kuang X, Liu Q, Zhao L, Yang S. Yinchenhao Decoction mitigates intestinal impairment induced by high carbohydrate diet in largemouth bass (Micropterus salmoides): insights from inflammation, apoptosis, oxidative stress, tight junctions, and microbiota homeostasis. FISH PHYSIOLOGY AND BIOCHEMISTRY 2024; 50:2207-2223. [PMID: 39066864 DOI: 10.1007/s10695-024-01388-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 07/22/2024] [Indexed: 07/30/2024]
Abstract
As a major source of energy, carbohydrates have a protein-saving effect. However, excessive consumption of carbohydrates can lead to the disruption of the intestinal barrier in fish, especially for carnivorous fish. Therefore, traditional Chinese medicine component Yinchenhao Decoction (YD), was used to detect the effect on intestinal barriers and microbial community equilibrium for largemouth bass in current research. In this research, a series of NC (normal carbohydrate diet) and HC (high carbohydrate diet) with graded YD treatments during 10 weeks feeding trial. Results suggested that 2% and 4% YD treatments significantly reduced gut inflammation and mucosal loss caused by HC. Compared with NC, HC significantly decreased the relative expression of intestinal tight junction-related genes (zo1, claudin1, claudin7, and occludin). However, with the application of YD, the expression of tight junction-related genes (zo1, claudin1, and claudin7) increased significantly (p < 0.05). Likewise, administration of YD significantly reduced elevated plasma diamine oxidase (DAO) activity caused by HC (p < 0.05). Additionally, YD significantly downregulated the mRNA expression of endoplasmic reticulum stress (ERS)-related genes (grp78, atf6, chopα, ire1, xbp1, and eifα) and pro-apoptosis genes (casp3, casp8, and bax) (p < 0.05), while upregulating the anti-apoptosis gene bcl2 (p < 0.05). Moreover, YD significantly increased the mRNA expression of antioxidant genes and the enzyme activities of CAT and GPX, while decreased MDA concentration significantly (p < 0.05). Whereas, YD markedly decreased the expression of pro-inflammatory genes (il1β, tnfα, il8, and nf-κB) and the immune enzymes activity (ACP and AKP) (p < 0.05) by up-regulating the expression of anti-inflammatory genes (ikb and il10). Notably, YD modulated the largemouth bass intestinal microbial community, enhanced the diversity and increased the abundance of probiotic microorganisms in the intestinal microbiota. In summary, YD supplementation in HC alleviated inflammation, apoptosis, oxidative stress, tight-junction injury, and microbiota disequilibrium in the intestine, which suggested that YD could be a valuable functional additive in aquaculture.
Collapse
Affiliation(s)
- Haoxiao Yan
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Yifan Hu
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Ji Liang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Kuo He
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Xu Kuang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Qiao Liu
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Liulan Zhao
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Song Yang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China.
| |
Collapse
|
5
|
Mansour H, Slika H, Nasser SA, Pintus G, Khachab M, Sahebkar A, Eid AH. Flavonoids, gut microbiota and cardiovascular disease: Dynamics and interplay. Pharmacol Res 2024; 209:107452. [PMID: 39383791 DOI: 10.1016/j.phrs.2024.107452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 09/11/2024] [Accepted: 10/04/2024] [Indexed: 10/11/2024]
Abstract
Cardiovascular disease (CVD) remains the leading cause of global morbidity and mortality. Extensive efforts have been invested to explicate mechanisms implicated in the onset and progression of CVD. Besides the usual suspects as risk factors (obesity, diabetes, and others), the gut microbiome has emerged as a prominent and essential factor in the pathogenesis of CVD. With its endocrine-like effects, the microbiome modulates many physiologic processes. As such, it is not surprising that dysbiosis-by generating metabolites, inciting inflammation, and altering secondary bile acid signaling- could predispose to or aggravate CVD. Nevertheless, various natural and synthetic compounds have been shown to modulate the microbiome. Prime among these molecules are flavonoids, which are natural polyphenols mainly present in fruits and vegetables. Accumulating evidence supports the potential of flavonoids in attenuating the development of CVD. The ascribed mechanisms of these compounds appear to involve mitigation of inflammation, alteration of the microbiome composition, enhancement of barrier integrity, induction of reverse cholesterol transport, and activation of farnesoid X receptor signaling. In this review, we critically appraise the methods by which the gut microbiome, despite being essential to the human body, predisposes to CVD. Moreover, we dissect the mechanisms and pathways underlying the cardioprotective effects of flavonoids.
Collapse
Affiliation(s)
- Hadi Mansour
- Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Hasan Slika
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Gianfranco Pintus
- Department of Biomedical Sciences, University of Sassari, Sassari 07100, Italy
| | - Maha Khachab
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Beirut, Lebanon
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali H Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar.
| |
Collapse
|
6
|
Santangelo C, Scazzocchio B, Varì R, Ajmone-Cat MA, Tammaro A, Tait S, Masciola I, Tassinari R, Vincentini O, Di Benedetto R, Berry A, Cirulli F, Maranghi F, De Simone R, D’Archivio M. Insights into the Sex-Related Effects of Dietary Polyphenols and Metabolic Disruptors on Inflammatory and (Neuro) Endocrine Pathways in Obesity: The HEAL Project. Nutrients 2024; 16:3595. [PMID: 39519428 PMCID: PMC11547382 DOI: 10.3390/nu16213595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 09/27/2024] [Accepted: 10/11/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND this study was performed under the umbrella of the Health Extended Alliance for Innovative Therapies, Advanced Lab Research, and Integrated Approaches of Precision Medicine (HEAL ITALIA) partnership and funded under the National Recovery and Resilience Plan, Mission 4 Component 2 Investment 1.3, and by the European Union. OBJECTIVES the overall objective of the HEAL project is to identify innovative and effective therapeutic approaches to reduce disease burden. The present research falls within Spoke 7: Prevention Strategies: Integrated and gender medicine approaches for prevention strategies based on environmental, lifestyle, and clinical biometric data. Obesity represents a primary risk factor worldwide for the onset of numerous life-threatening diseases, including metabolic, cardiovascular, and neurological diseases. Environmental and gender-related factors influence obesity development. However, the molecular mechanisms underlying the effects of those agents on different organs of the human body are not fully understood yet. METHODS here, we present a study protocol aimed at shedding light on (i) the complex interplays among adipose tissue, brain and gut in obesity, and (ii) the effects of specific dietary components and environmental metabolism-disrupting compounds on those interactions. To this purpose, we combined ex vivo, in vitro, and in vivo approaches to gain additional knowledge about the molecular mechanisms underlying connections between organs. CONCLUSIONS the data provided by this study will contribute to defining new targets for therapeutic and/or preventive interventions, thereby allowing more personalized approaches to nutrition.
Collapse
Affiliation(s)
- Carmela Santangelo
- Gender-Specific Prevention and Health Unit, Centre for Gender-Specific Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (C.S.); (R.V.); (A.T.); (S.T.); (I.M.); (R.T.); (F.M.); (M.D.)
| | - Beatrice Scazzocchio
- Gender-Specific Prevention and Health Unit, Centre for Gender-Specific Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (C.S.); (R.V.); (A.T.); (S.T.); (I.M.); (R.T.); (F.M.); (M.D.)
| | - Rosaria Varì
- Gender-Specific Prevention and Health Unit, Centre for Gender-Specific Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (C.S.); (R.V.); (A.T.); (S.T.); (I.M.); (R.T.); (F.M.); (M.D.)
| | | | - Alessia Tammaro
- Gender-Specific Prevention and Health Unit, Centre for Gender-Specific Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (C.S.); (R.V.); (A.T.); (S.T.); (I.M.); (R.T.); (F.M.); (M.D.)
- Department of Biomedicine and Prevention, Tor Vergata University of Rome, 00133 Rome, Italy
| | - Sabrina Tait
- Gender-Specific Prevention and Health Unit, Centre for Gender-Specific Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (C.S.); (R.V.); (A.T.); (S.T.); (I.M.); (R.T.); (F.M.); (M.D.)
| | - Irene Masciola
- Gender-Specific Prevention and Health Unit, Centre for Gender-Specific Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (C.S.); (R.V.); (A.T.); (S.T.); (I.M.); (R.T.); (F.M.); (M.D.)
| | - Roberta Tassinari
- Gender-Specific Prevention and Health Unit, Centre for Gender-Specific Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (C.S.); (R.V.); (A.T.); (S.T.); (I.M.); (R.T.); (F.M.); (M.D.)
| | - Olimpia Vincentini
- Human Nutrition and Health Unit, Department of Food Safety, Nutrition, and Veterinary Public Health, Istituto Superiore di Sanità, 00161 Rome, Italy; (O.V.); (R.D.B.)
| | - Rita Di Benedetto
- Human Nutrition and Health Unit, Department of Food Safety, Nutrition, and Veterinary Public Health, Istituto Superiore di Sanità, 00161 Rome, Italy; (O.V.); (R.D.B.)
| | - Alessandra Berry
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.B.); (F.C.)
| | - Francesca Cirulli
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.B.); (F.C.)
| | - Francesca Maranghi
- Gender-Specific Prevention and Health Unit, Centre for Gender-Specific Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (C.S.); (R.V.); (A.T.); (S.T.); (I.M.); (R.T.); (F.M.); (M.D.)
| | - Roberta De Simone
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Massimo D’Archivio
- Gender-Specific Prevention and Health Unit, Centre for Gender-Specific Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (C.S.); (R.V.); (A.T.); (S.T.); (I.M.); (R.T.); (F.M.); (M.D.)
| |
Collapse
|
7
|
Marangelo C, Vernocchi P, Del Chierico F, Scanu M, Marsiglia R, Petrolo E, Fucà E, Guerrera S, Valeri G, Vicari S, Putignani L. Stratification of Gut Microbiota Profiling Based on Autism Neuropsychological Assessments. Microorganisms 2024; 12:2041. [PMID: 39458350 PMCID: PMC11510388 DOI: 10.3390/microorganisms12102041] [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: 09/13/2024] [Revised: 10/04/2024] [Accepted: 10/07/2024] [Indexed: 10/28/2024] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder. Investigations of gut microbiota (GM) play an important role in deciphering disease severity and symptoms. Overall, we stratified 70 ASD patients by neuropsychological assessment, based on Calibrated Severity Scores (CSSs) of the Autism Diagnostic Observation Schedule-Second edition (ADOS-2), Child Behavior Checklist (CBCL) and intelligent quotient/developmental quotient (IQ/DQ) parameters. Hence, metataxonomy and PICRUSt-based KEGG predictions of fecal GM were assessed for each clinical subset. Here, 60% of ASD patients showed mild to moderate autism, while the remaining 40% showed severe symptoms; 23% showed no clinical symptoms, 21% had a risk of behavior problems and 56% had clinical symptoms based on the CBCL, which assesses internalizing problems; further, 52% had no clinical symptoms, 21% showed risk, and 26% had clinical symptoms classified by CBCL externalizing problems. Considering the total CBCL index, 34% showed no clinical symptoms, 13% showed risk, and 52% had clinical symptoms. Here, 70% of ASD patients showed cognitive impairment/developmental delay (CI/DD). The GM of ASDs with severe autism was characterized by an increase in Veillonella, a decrease in Monoglobus pectinilyticus and a higher microbial dysbiosis index (MDI) when compared to mild-moderate ASDs. Patients at risk for behavior problems and showing clinical symptoms were characterized by a GM with an increase of Clostridium, Eggerthella, Blautia, Intestinibacter, Coprococcus, Ruminococcus, Onthenecus and Bariatricus, respectively. Peptidoglycan biosynthesis and biofilm formation KEGGs characterized patients with clinical symptoms, while potential microbiota-activated PPAR-γ-signaling was seen in CI/DD patients. This evidence derived from GM profiling may be used to further improve ASD understanding, leasing to a better comprehension of the neurological phenotype.
Collapse
Affiliation(s)
- Chiara Marangelo
- Research Unit of Microbiome, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (C.M.); (P.V.); (F.D.C.); (M.S.); (R.M.)
| | - Pamela Vernocchi
- Research Unit of Microbiome, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (C.M.); (P.V.); (F.D.C.); (M.S.); (R.M.)
| | - Federica Del Chierico
- Research Unit of Microbiome, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (C.M.); (P.V.); (F.D.C.); (M.S.); (R.M.)
| | - Matteo Scanu
- Research Unit of Microbiome, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (C.M.); (P.V.); (F.D.C.); (M.S.); (R.M.)
| | - Riccardo Marsiglia
- Research Unit of Microbiome, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (C.M.); (P.V.); (F.D.C.); (M.S.); (R.M.)
| | - Emanuela Petrolo
- Child and Adolescent Neuropsychiatry Unit, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (E.P.); (E.F.); (S.G.); (G.V.); or (S.V.)
| | - Elisa Fucà
- Child and Adolescent Neuropsychiatry Unit, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (E.P.); (E.F.); (S.G.); (G.V.); or (S.V.)
| | - Silvia Guerrera
- Child and Adolescent Neuropsychiatry Unit, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (E.P.); (E.F.); (S.G.); (G.V.); or (S.V.)
| | - Giovanni Valeri
- Child and Adolescent Neuropsychiatry Unit, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (E.P.); (E.F.); (S.G.); (G.V.); or (S.V.)
| | - Stefano Vicari
- Child and Adolescent Neuropsychiatry Unit, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (E.P.); (E.F.); (S.G.); (G.V.); or (S.V.)
- Life Sciences and Public Health Department, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Lorenza Putignani
- Unit of Microbiomics and Research Unit of Human Microbiome, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy
| |
Collapse
|
8
|
Ozdemir VF, Yanar M. Effects of propolis extract administration on immune parameters, faecal consistency scores, and growth performance of Holstein-Friesian calves. Trop Anim Health Prod 2024; 56:259. [PMID: 39292350 DOI: 10.1007/s11250-024-04128-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 09/11/2024] [Indexed: 09/19/2024]
Abstract
The purpose of the study was to investigate the effect of Ethanolic Extract of Propolis (EEP) administration on immune parameters, faecal consistency scores, growth performance, and feed efficiency of Holstein Friesian calves. A total of 24 calves were divided into two different groups, control (n = 12) and EEP (n = 12). Both groups consisted of 6 male and 6 female calves. The calves were fed milk amounting to 10% of their birth weight each day until they reached 60 days of age. Additionally, they were given starter feed and dry hay once a day. Calves assigned to the EEP group received 4 ml of EEP daily. Use of EEP increased (P < 0.05) the serum IgG and IgM levels at 2 months of age compared to the control group. EEP also showed efficacy (P < 0.01) in reducing faecal consistency in calves throughout the study. The levels of IL-1β, IL-6, TNF-α and NF-κB expression in calves treated with EEP were lower (P < 0.05) throughout the EEP application period. On the other hand, IGF-1 mRNA transcript levels were (P < 0.01) higher in EEP group calves than in the control group. Furthermore, EEP-fed calves consumed less dry matter for 1 kg of live weight gain during the weaning-4 months (P < 0.01) and birth-4 months (P < 0.05) periods. These results indicate that EEP supplementation, through its immunostimulatory effects, plays a crucial role in the control of neonatal calf diarrhoea. Growth and development as well as IGF-1, which stimulates growth in almost all somatic cells, was also significantly increased by EEP supplementation. The combined effect of the rich bioactive compounds found in EEP appears to have a significant impact on health and well-being, resulting in improved early life performance in dairy calves.
Collapse
Affiliation(s)
- Veysel Fatih Ozdemir
- Department of Animal Science, College of Agriculture, Atatürk University, 25240, Erzurum, Türkiye.
| | - Mete Yanar
- Department of Animal Science, College of Agriculture, Atatürk University, 25240, Erzurum, Türkiye
| |
Collapse
|
9
|
AlMarzooqi SK, Almarzooqi F, Sadida HQ, Jerobin J, Ahmed I, Abou-Samra AB, Fakhro KA, Dhawan P, Bhat AA, Al-Shabeeb Akil AS. Deciphering the complex interplay of obesity, epithelial barrier dysfunction, and tight junction remodeling: Unraveling potential therapeutic avenues. Obes Rev 2024; 25:e13766. [PMID: 38745386 DOI: 10.1111/obr.13766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 03/11/2024] [Accepted: 04/17/2024] [Indexed: 05/16/2024]
Abstract
Obesity stands as a formidable global health challenge, predisposing individuals to a plethora of chronic illnesses such as cardiovascular disease, diabetes, and cancer. A confluence of genetic polymorphisms, suboptimal dietary choices, and sedentary lifestyles significantly contribute to the elevated incidence of obesity. This multifaceted health issue profoundly disrupts homeostatic equilibrium at both organismal and cellular levels, with marked alterations in gut permeability as a salient consequence. The intricate mechanisms underlying these alterations have yet to be fully elucidated. Still, evidence suggests that heightened inflammatory cytokine levels and the remodeling of tight junction (TJ) proteins, particularly claudins, play a pivotal role in the manifestation of epithelial barrier dysfunction in obesity. Strategic targeting of proteins implicated in these pathways and metabolites such as short-chain fatty acids presents a promising intervention for restoring barrier functionality among individuals with obesity. Nonetheless, recognizing the heterogeneity among affected individuals is paramount; personalized medical interventions or dietary regimens tailored to specific genetic backgrounds and allergy profiles may prove indispensable. This comprehensive review delves into the nexus of obesity, tight junction remodeling, and barrier dysfunction, offering a critical appraisal of potential therapeutic interventions.
Collapse
Affiliation(s)
- Sara K AlMarzooqi
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Research Program, Sidra Medicine, Doha, Qatar
| | - Fajr Almarzooqi
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Research Program, Sidra Medicine, Doha, Qatar
| | - Hana Q Sadida
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Research Program, Sidra Medicine, Doha, Qatar
| | - Jayakumar Jerobin
- Qatar Metabolic Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Ikhlak Ahmed
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Research Program, Sidra Medicine, Doha, Qatar
| | - Abdul-Badi Abou-Samra
- Qatar Metabolic Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Khalid A Fakhro
- Department of Human Genetics, Sidra Medicine, Doha, Qatar
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
- Department of Genetic Medicine, Weill Cornell Medicine, Doha, Qatar
| | - Punita Dhawan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Ajaz A Bhat
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Research Program, Sidra Medicine, Doha, Qatar
| | - Ammira S Al-Shabeeb Akil
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Research Program, Sidra Medicine, Doha, Qatar
| |
Collapse
|
10
|
Zhao X, Guo J, Wang Y, Yi X. High-tannin food enhances spatial memory and scatter-hoarding in rodents via the microbiota-gut-brain axis. MICROBIOME 2024; 12:140. [PMID: 39075602 DOI: 10.1186/s40168-024-01849-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Accepted: 05/29/2024] [Indexed: 07/31/2024]
Abstract
BACKGROUND The mutually beneficial coevolutionary relationships between rodents and plant seeds have been a theme of research in plant-animal relationships. Seed tannins are important secondary metabolites of plants that regulate the food-hoarding behavior of rodents; however, the underlying molecular mechanisms are not yet clear. In this study, we investigated whether and how seed tannins improve spatial memory and regulate the hoarding behavior of Tamias sibiricus by altering their gut microbiota. RESULTS We showed that acorn tannins not only improved spatial memory but also enhanced scatter-hoarding in T. sibiricus. Changes in the composition and function of the gut microbiota in response to tannins from acorns are closely related to these improvements. Metabonomic analyses revealed the role of gut isovaleric acid and isobutyric acid as well as serum L-tryptophan in mediating the spatial memory of T. sibiricus via the gut microbiota. The hippocampal proteome provides further evidence that the microbiota-gut-brain axis regulates spatial memory and scatter-hoarding in animals. Our study is likely the first to report that plant secondary metabolites improve hippocampal function and spatial memory and ultimately modulate food-hoarding behavior via the microbiota-gut-brain axis. CONCLUSION Our findings may have resolved the long-standing puzzle about the hidden role of plant secondary metabolites in manipulating food-hoarding behavior in rodents via the microbiota-gut-brain axis. Our study is important for better understanding the mutualistic coevolution between plants and animals. Video Abstract.
Collapse
Affiliation(s)
- Xiangyu Zhao
- School of Life Sciences, Qufu Normal University, Qufu, 273165, China
| | - Jiawei Guo
- School of Life Sciences, Qufu Normal University, Qufu, 273165, China
- Present address: Huxi Middle School, Dongchangfu District, Liaocheng, 252000, China
| | - Yiming Wang
- School of Life Sciences, Qufu Normal University, Qufu, 273165, China
| | - Xianfeng Yi
- School of Life Sciences, Qufu Normal University, Qufu, 273165, China.
| |
Collapse
|
11
|
Ahmad A, Mahmood N, Raza MA, Mushtaq Z, Saeed F, Afzaal M, Hussain M, Amjad HW, Al-Awadi HM. Gut microbiota and their derivatives in the progression of colorectal cancer: Mechanisms of action, genome and epigenome contributions. Heliyon 2024; 10:e29495. [PMID: 38655310 PMCID: PMC11035079 DOI: 10.1016/j.heliyon.2024.e29495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 04/08/2024] [Accepted: 04/09/2024] [Indexed: 04/26/2024] Open
Abstract
Gut microbiota interacts with host epithelial cells and regulates many physiological functions such as genetics, epigenetics, metabolism of nutrients, and immune functions. Dietary factors may also be involved in the etiology of colorectal cancer (CRC), especially when an unhealthy diet is consumed with excess calorie intake and bad practices like smoking or consuming a great deal of alcohol. Bacteria including Fusobacterium nucleatum, Enterotoxigenic Bacteroides fragilis (ETBF), and Escherichia coli (E. coli) actively participate in the carcinogenesis of CRC. Gastrointestinal tract with chronic inflammation and immunocompromised patients are at high risk for CRC progression. Further, the gut microbiota is also involved in Geno-toxicity by producing toxins like colibactin and cytolethal distending toxin (CDT) which cause damage to double-stranded DNA. Specific microRNAs can act as either tumor suppressors or oncogenes depending on the cellular environment in which they are expressed. The current review mainly highlights the role of gut microbiota in CRC, the mechanisms of several factors in carcinogenesis, and the role of particular microbes in colorectal neoplasia.
Collapse
Affiliation(s)
- Awais Ahmad
- Department of Food Science, Government College University Faisalabad, Faisalabad, Pakistan
| | - Nasir Mahmood
- Department of Zoology, University of Central Punjab Bahawalpur, Bahawalpur, Pakistan
| | - Muhammad Ahtisham Raza
- Department of Food Science, Government College University Faisalabad, Faisalabad, Pakistan
| | - Zarina Mushtaq
- Department of Food Science, Government College University Faisalabad, Faisalabad, Pakistan
| | - Farhan Saeed
- Department of Food Science, Government College University Faisalabad, Faisalabad, Pakistan
| | - Muhammad Afzaal
- Department of Food Science, Government College University Faisalabad, Faisalabad, Pakistan
| | - Muzzamal Hussain
- Department of Food Science, Government College University Faisalabad, Faisalabad, Pakistan
| | - Hafiz Wasiqe Amjad
- International Medical School, Jinggangshan University, Ji'an, Jiangxi, China
| | | |
Collapse
|
12
|
Cheng X, Zhu Y, Huang J, Li Y, Jiang X, Yang Q. A neutral polysaccharide from Persicaria hydropiper (L.) Spach ameliorates lipopolysaccharide-induced intestinal barrier injury via regulating the gut microbiota and modulating AKT/PI3K/mTOR and MAPK signaling pathways. JOURNAL OF ETHNOPHARMACOLOGY 2024; 320:117403. [PMID: 37952732 DOI: 10.1016/j.jep.2023.117403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 10/13/2023] [Accepted: 11/07/2023] [Indexed: 11/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Persicaria hydropiper (L.) Spach, a herb that is prevalent across Asia and Europe, finds utility as both a culinary ingredient and medicinal herb. In China, P. hydropiper decoction is commonly employed to alleviate dysentery, gastroenteritis, and diarrhea symptoms. AIM OF THE STUDY To assess the effects of a neutral polysaccharide from P. hydropiper (PHP) on the intestinal barrier (IB) injury induced by lipopolysaccharide (LPS) in mice, and elucidate the molecular mechanisms involved. MATERIALS AND METHODS PHP was extracted from dried P. hydropiper herb using hot water extraction, followed by ethanol precipitation. The extract underwent successive isolation and purification steps involving anion-exchange and gel filtration chromatography. The primary structure of PHP was determined using Fourier-transformed infrared spectroscopy, ion chromatography, gas chromatography-mass spectrometry (GC-MS), and nuclear magnetic resonance (NMR) spectroscopy. Male BALB/c mice were randomly assigned to control (CON), model (MOD), berberine hydrochloride (BBR), and PHP (20, 40 and 80 mg/kg) groups. Histopathological changes in jejunal tissues were assessed through hematoxylin and eosin (HE) staining. The expression levels of proteins and genes involved in AKT/PI3K/mTOR and MAPK signaling pathways were evaluated using qRT-PCR and Western blotting, respectively. The composition and abundance of the gut microbiota in mice were analyzed using high-throughput 16S rRNA gene sequencing. Additionally, the concentrations of short-chain fatty acids (SCFAs) were determined using GC-MS. RESULTS The main components of PHP included arabinose, galactose, and glucose (molar ratio = 1.00:5.52:11.39). The backbone of PHP consisted of →4)-Glcp-(1→, →4,6)-Glcp-(1→, →4)-Galp-(1→, →4,6)-Galp-(1→. The branched chains primarily consisted of 5)-Araf-(1→ residues, which were attached to the backbone through →6)-Glcp-(1→ and →6)-Galp-(1→ at the 6-position. Histological analysis demonstrated that PHP exhibited a mitigating effect on intestinal damage induced by LPS. PHP could markedly reduce the mRNA levels of PI3K, AKT, mTOR, p70 S6K, Ras, Raf1, MEK1/2, p38, ERK1/2, and JNK, while downregulating the protein levels of p-mTOR, p-PI3K, p-AKT, p-p38, p-ERK, and p-JNK. PHP also modulated the diversities and abundances of the gut microbiota, resulting in an increase in the abundances of Lactobacillaceae, Anaerovoracaceae, Lachnospiraceae, Eggerthellaceae, and Desulfovibrionaceae and a decrease in the abundances of Muribaculaceae, Prevotellaceae, and Rikenellaceae. Additionally, PHP significantly increased the content of various SCFAs. CONCLUSION PHP emerges as a pivotal factor in the repair of IB injury by virtue of its ability to regulate the gut microbiota, elevate SCFA levels, and inhibit the MAPK and AKT/PI3K/mTOR pathways. It is worth noting that the therapeutic effect of high-dose PHP was remarkably significant, surpassing even the positive control of berberine hydrochloride.
Collapse
Affiliation(s)
- Xuanxuan Cheng
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, China; Key Laboratory of State Administration of Traditional Chinese Medicine for Production & Development of Cantonese Medicinal Materials, Guangzhou, China; Comprehensive Experimental Station of National Industrial Technology System for Chinese Materia Medica, Guangzhou, China; Guangdong Engineering Research Center of Good Agricultural Practice & Comprehensive Development for Cantonese Medicinal Materials, Guangzhou, China.
| | - Yuehua Zhu
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, China; Key Laboratory of State Administration of Traditional Chinese Medicine for Production & Development of Cantonese Medicinal Materials, Guangzhou, China; Comprehensive Experimental Station of National Industrial Technology System for Chinese Materia Medica, Guangzhou, China; Guangdong Engineering Research Center of Good Agricultural Practice & Comprehensive Development for Cantonese Medicinal Materials, Guangzhou, China.
| | - Jiahuan Huang
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, China; Key Laboratory of State Administration of Traditional Chinese Medicine for Production & Development of Cantonese Medicinal Materials, Guangzhou, China; Comprehensive Experimental Station of National Industrial Technology System for Chinese Materia Medica, Guangzhou, China; Guangdong Engineering Research Center of Good Agricultural Practice & Comprehensive Development for Cantonese Medicinal Materials, Guangzhou, China.
| | - Yufei Li
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, China; Key Laboratory of State Administration of Traditional Chinese Medicine for Production & Development of Cantonese Medicinal Materials, Guangzhou, China; Comprehensive Experimental Station of National Industrial Technology System for Chinese Materia Medica, Guangzhou, China; Guangdong Engineering Research Center of Good Agricultural Practice & Comprehensive Development for Cantonese Medicinal Materials, Guangzhou, China.
| | - Xiaolin Jiang
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, China; Key Laboratory of State Administration of Traditional Chinese Medicine for Production & Development of Cantonese Medicinal Materials, Guangzhou, China; Comprehensive Experimental Station of National Industrial Technology System for Chinese Materia Medica, Guangzhou, China; Guangdong Engineering Research Center of Good Agricultural Practice & Comprehensive Development for Cantonese Medicinal Materials, Guangzhou, China.
| | - Quan Yang
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, China; Key Laboratory of State Administration of Traditional Chinese Medicine for Production & Development of Cantonese Medicinal Materials, Guangzhou, China; Comprehensive Experimental Station of National Industrial Technology System for Chinese Materia Medica, Guangzhou, China; Guangdong Engineering Research Center of Good Agricultural Practice & Comprehensive Development for Cantonese Medicinal Materials, Guangzhou, China.
| |
Collapse
|
13
|
Pretorius L, Smith C. Green rooibos (Aspalathus linearis) promotes gut health: insight into mechanisms. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117379. [PMID: 37923252 DOI: 10.1016/j.jep.2023.117379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 10/20/2023] [Accepted: 10/31/2023] [Indexed: 11/07/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Paralleling the increasing incidence of gastrointestinal disorders world-wide, therapeutic investigations of nutraceuticals to promote gastrointestinal health are gaining popularity. Although anecdotally well-known for its gut health promoting potential, sparse scientific evidence supports this action of Aspalathus linearis (Burm.f.) R. Dahlgren - or rooibos - at the gastrointestinal epithelial level. AIM OF THE STUDY Traditionally, rooibos is considered to exert antispasmodic, anti-inflammatory, and anti-nociceptive effects in the gut. However, the direct effect on intestinal epithelium is unknown. Thus, to assess the validity of anecdotal claims, two larval zebrafish models were utilized to evaluate effects of rooibos on intestinal health. MATERIALS AND METHODS Firstly, a larval zebrafish model of gastrointestinal inflammation (2-day TNBS-exposure) was employed. Co-administration of 6α-methylprednisolone served as an internal treatment control. Assessments included live imaging techniques and post-mortem immunofluorescent staining of epithelial tight junction proteins. In addition, whole body H2O2 and prostaglandin E2 assays were performed. Secondly, a gastrointestinal motility assay was performed, with known pro- and anti-kinetic mediators to assess the effect of rooibos to alter functional outcome in vivo. RESULTS Aqueous and ethanol extracts of green rooibos rescued TNBS-induced reductions in neutral red stained length of larval mid-intestines. Subsequent experiments confirmed the rescue capacity of the aqueous green rooibos extract regarding whole body oxidative and inflammatory status. Concerning tight junction proteins, only the aqueous green rooibos extract - and not prednisolone - normalized both zona occludens-1 and occludin expression levels when compared the TNBS group. In terms of gastrointestinal motility, the aqueous green rooibos extract significantly reduced the extent of gut motility dysregulation achieved by kinetic modulators. CONCLUSIONS Data indicates the potential of a 2 mg/ml aqueous extract of green rooibos to improve gastrointestinal integrity and functionality in vivo, suggesting beneficial effects of rooibos may already occur at the level of the gut. This provides some evidence to support indigenous knowledge.
Collapse
Affiliation(s)
- Lesha Pretorius
- Experimental Medicine, Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, 7500, South Africa.
| | - Carine Smith
- Experimental Medicine, Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, 7500, South Africa.
| |
Collapse
|
14
|
de Jesus LB, Frota AF, de Araújo FM, de Jesus RLC, Costa MDFD, de Vasconcelos DFSA, Gois MB, Baccan GC, da Silva VDA, Costa SL. Effect of the Flavonoid Rutin on the Modulation of the Myenteric Plexuses in an Experimental Model of Parkinson's Disease. Int J Mol Sci 2024; 25:1037. [PMID: 38256111 PMCID: PMC10815896 DOI: 10.3390/ijms25021037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 01/08/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Recent discoveries have shown that enteric glial cells play an important role in different neurodegenerative disorders, such as Parkinson's disease (PD), which is characterized by motor dysfunctions caused by the progressive loss of dopaminergic neurons in the substance nigra pars compacta and non-motor symptoms including gastrointestinal dysfunction. In this study, we investigated the modulatory effects of the flavonoid rutin on the behavior and myenteric plexuses in a PD animal model and the response of enteric glia. Adult male Wistar rats were submitted to stereotaxic injection with 6-hydroxydopamine or saline, and they were untreated or treated with rutin (10 mg/kg) for 14 days. The ileum was collected to analyze tissue reactivity and immunohistochemistry for neurons (HuC/HuD) and enteric glial cells (S100β) in the myenteric plexuses. Behavioral tests demonstrated that treatment with rutin improved the motor capacity of parkinsonian animals and improved intestinal transit without interfering with the cell population; rutin treatment modulated the reactivity of the ileal musculature through muscarinic activation, reducing relaxation through the signaling pathway of nitric oxide donors, and increased the longitudinal contractility of the colon musculature in parkinsonian animals. Rutin revealed modulatory activities on the myenteric plexus, bringing relevant answers regarding the effect of the flavonoid in this system and the potential application of PD adjuvant treatment.
Collapse
Affiliation(s)
- Livia Bacelar de Jesus
- Laboratory of Neurochemistry and Cellular Biology, Federal University of Bahia, Salvador 40170-110, BA, Brazil; (L.B.d.J.); (A.F.F.); (F.M.d.A.); (M.d.F.D.C.)
| | - Annyta Fernandes Frota
- Laboratory of Neurochemistry and Cellular Biology, Federal University of Bahia, Salvador 40170-110, BA, Brazil; (L.B.d.J.); (A.F.F.); (F.M.d.A.); (M.d.F.D.C.)
| | - Fillipe Mendes de Araújo
- Laboratory of Neurochemistry and Cellular Biology, Federal University of Bahia, Salvador 40170-110, BA, Brazil; (L.B.d.J.); (A.F.F.); (F.M.d.A.); (M.d.F.D.C.)
| | - Rafael Leonne Cruz de Jesus
- Cardiovascular Physiology and Pharmacology Laboratory, Federal University of Bahia, Salvador 40170-110, BA, Brazil; (R.L.C.d.J.); (D.F.S.A.d.V.)
| | - Maria de Fátima Dias Costa
- Laboratory of Neurochemistry and Cellular Biology, Federal University of Bahia, Salvador 40170-110, BA, Brazil; (L.B.d.J.); (A.F.F.); (F.M.d.A.); (M.d.F.D.C.)
- National Institute for Translational Neurosciences (INCT/CNPq INNT), Rio de Janeiro 21941-902, RJ, Brazil
| | | | - Marcelo Biondaro Gois
- Faculty of Health Sciences, Federal University of Rondonópolis, Rondonópolis 78736-900, MT, Brazil;
| | - Gyselle Chrystina Baccan
- Laboratory of Neuroendocrine-Immunology, Federal University of Bahia, Salvador 40170-110, BA, Brazil;
| | - Victor Diogenes Amaral da Silva
- Laboratory of Neurochemistry and Cellular Biology, Federal University of Bahia, Salvador 40170-110, BA, Brazil; (L.B.d.J.); (A.F.F.); (F.M.d.A.); (M.d.F.D.C.)
| | - Silvia Lima Costa
- Laboratory of Neurochemistry and Cellular Biology, Federal University of Bahia, Salvador 40170-110, BA, Brazil; (L.B.d.J.); (A.F.F.); (F.M.d.A.); (M.d.F.D.C.)
- National Institute for Translational Neurosciences (INCT/CNPq INNT), Rio de Janeiro 21941-902, RJ, Brazil
| |
Collapse
|
15
|
Schytz Andersen-Civil AI, Arora P, Zhu L, Myhill LJ, Büdeyri Gökgöz N, Castro-Mejia JL, Leppä MM, Hansen LH, Lessard-Lord J, Salminen JP, Thamsborg SM, Sandris Nielsen D, Desjardins Y, Williams AR. Gut microbiota-mediated polyphenol metabolism is restrained by parasitic whipworm infection and associated with altered immune function in mice. Gut Microbes 2024; 16:2370917. [PMID: 38944838 PMCID: PMC11216105 DOI: 10.1080/19490976.2024.2370917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 06/17/2024] [Indexed: 07/02/2024] Open
Abstract
Polyphenols are phytochemicals commonly found in plant-based diets which have demonstrated immunomodulatory and anti-inflammatory properties. However, the interplay between polyphenols and pathogens at mucosal barrier surfaces has not yet been elucidated in detail. Here, we show that proanthocyanidin (PAC) polyphenols interact with gut parasites to influence immune function and gut microbial-derived metabolites in mice. PAC intake inhibited mastocytosis during infection with the small intestinal roundworm Heligmosomoides polygyrus, and altered the host tissue transcriptome at the site of infection with the large intestinal whipworm Trichuris muris, with a notable enhancement of type-1 inflammatory and interferon-driven gene pathways. In the absence of infection, PAC intake promoted the expansion of Turicibacter within the gut microbiota, increased fecal short chain fatty acids, and enriched phenolic metabolites such as phenyl-γ-valerolactones in the cecum. However, these putatively beneficial effects were reduced in PAC-fed mice infected with T. muris, suggesting concomitant parasite infection can attenuate gut microbial-mediated PAC catabolism. Collectively, our results suggest an inter-relationship between a phytonutrient and infection, whereby PAC may augment parasite-induced inflammation (most prominently with the cecum dwelling T. muris), and infection may abrogate the beneficial effects of health-promoting phytochemicals.
Collapse
Affiliation(s)
| | - Pankaj Arora
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Ling Zhu
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Laura J. Myhill
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | | | | | - Milla M. Leppä
- Natural Chemistry Research Group, Department of Chemistry, University of Turku, Turku, Finland
| | - Lars H. Hansen
- Department of Plant and Environmental Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Jacob Lessard-Lord
- Institute of Nutrition and Functional Foods (INAF), Laval University, Québec, QC, Canada
| | - Juha-Pekka Salminen
- Natural Chemistry Research Group, Department of Chemistry, University of Turku, Turku, Finland
| | - Stig M. Thamsborg
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | | | - Yves Desjardins
- Institute of Nutrition and Functional Foods (INAF), Laval University, Québec, QC, Canada
| | - Andrew R. Williams
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| |
Collapse
|
16
|
Kanner J. Food Polyphenols as Preventive Medicine. Antioxidants (Basel) 2023; 12:2103. [PMID: 38136222 PMCID: PMC10740609 DOI: 10.3390/antiox12122103] [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: 10/29/2023] [Revised: 12/04/2023] [Accepted: 12/04/2023] [Indexed: 12/24/2023] Open
Abstract
Reactive oxygen species (ROS) are the initiators in foods and in the stomach of oxidized dietary lipids, proteins, and lipid-oxidation end-products (ALEs), inducing in humans the development of several chronic diseases and cancer. Epidemiological, human clinical and animal studies supported the role of dietary polyphenols and derivatives in prevention of development of such chronic diseases. There is much evidence that polyphenols/derivatives at the right timing and concentration, which is critical, acts mostly in the aerobic stomach and generally in the gastrointestinal tract as reducing agents, scavengers of free radicals, trappers of reactive carbonyls, modulators of enzyme activity, generators of beneficial gut microbiota and effectors of cellular signaling. In the blood system, at low concentration, they act as generators of electrophiles and low concentration of H2O2, acting mostly as cellular signaling, activating the PI3K/Akt-mediated Nrf2/eNOS pathways and inhibiting the inflammatory transcription factor NF-κB, inducing the cells, organs and organism for eustress, adaptation and surviving.
Collapse
Affiliation(s)
- Joseph Kanner
- Department of Food Science, ARO, Volcani Center, Bet-Dagan 7505101, Israel; or
- Institute of Biochemistry, Food Science and Nutrtion, Faculty of Agriculture Food and Environment, The Hebrew University of Jerusalem, Rehovot 9190501, Israel
| |
Collapse
|
17
|
Gao F, He Q, Wu S, Zhang K, Xu Z, Kang J, Quan F. Catalpol ameliorates LPS-induced inflammatory response by activating AMPK/mTOR signaling pathway in rat intestinal epithelial cells. Eur J Pharmacol 2023; 960:176125. [PMID: 37890606 DOI: 10.1016/j.ejphar.2023.176125] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 10/01/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023]
Abstract
Intestinal inflammation is a common clinical intestinal disease. Catalpol, a natural iridoid compound, has been shown to have anti-inflammatory, anti-oxidant and anti-apoptotic functions, but the mechanism of its protection against intestinal inflammation is still unclear. This study investigated the protective effect and potential mechanism of catalpol on the lipopolysaccharide (LPS)-induced inflammatory response of intestinal epithelial cell-6 (IEC-6). The results showed that catalpol could inhibit LPS-induced inflammatory response by dose-dependently reducing the release of inflammatory factors, such as tumor necrosis (TNF)-α, interleukin (IL)-1β and IL-6, and inhibiting the nuclear factor kappa-B (NF-κB) signaling pathway. Catalpol ameliorated cellular oxidative stress by reducing reactive oxygen species (ROS) and malondialdehyde (MDA) levels and increasing superoxide dismutase (SOD) and glutathione peroxidase (GSH-PX) expression. Meanwhile, catalpol also inhibited cell apoptosis, decreased the expression of B-cell lymphoma 2 (Bcl-2) - associated X (Bax), caspase 3 and caspase 9, and increased the expression of Bcl-2. This study found that catalpol activates AMP-activated protein kinase (AMPK) signaling pathway and inhibit mammalian target of rapamycin (mTOR) phosphorylationthe. In a further study, after inhibiting AMPK with dorsomorphin, the anti-inflammatory effects of catalpol were significantly reduced. Therefore, catalpol ameliorates LPS-induced inflammatory response by activating AMPK/mTOR signaling pathway in IEC-6 cells.
Collapse
Affiliation(s)
- Feng Gao
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, China
| | - Qifu He
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, China
| | - Shenghui Wu
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, China
| | - Kang Zhang
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, China
| | - Zhiming Xu
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, China
| | - Jian Kang
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, China
| | - Fusheng Quan
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, China.
| |
Collapse
|
18
|
Song S, Shon J, Yang WR, Kang HB, Kim KH, Park JY, Lee S, Baik SY, Lee KR, Park YJ. Short-Term Effects of Weight-Loss Meal Replacement Programs with Various Macronutrient Distributions on Gut Microbiome and Metabolic Parameters: A Pilot Study. Nutrients 2023; 15:4744. [PMID: 38004139 PMCID: PMC10675061 DOI: 10.3390/nu15224744] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 10/21/2023] [Accepted: 10/31/2023] [Indexed: 11/26/2023] Open
Abstract
It has emerged the gut microbiome is crucially linked to metabolic health and obesity. Macronutrient distribution has been discussed as a key parameter in weight-loss programs, but little is known about its impact on the gut microbiome. We investigated the effects of weight-loss meal replacement programs with different macronutrient ratios on the gut microbiota and metabolic parameters in subjects with overweight and obesity. Three low-calorie meal replacement programs with different ratios of carbohydrates, proteins, and lipids were designed: a balanced diet (Group B, 60:15:30), a high-lipid-low-carbohydrate diet (Group F, 35:20:55), and a protein-enriched diet (Group P, 40:25:35). Sixty overweight or obese participants were provided with the meals twice daily for 3 weeks. In all groups, diet intervention resulted in reduced body weight and BMI. The relative abundance of Bacteroidetes and Firmicutes phyla decreased and increased, respectively, which increased the Firmicutes/Bacteroidetes (F/B) ratio in all subjects, particularly in Groups B and P. Alpha- and beta-diversity were augmented at the phylum level in Group P. In conclusion, short-term interventions with weight-loss meal replacement programs increased butyrate-producing bacteria and the F/B ratio. Moreover, the protein-enriched diet significantly increased alpha- and beta-diversity compared to the balanced diet and the high-lipid-low-carbohydrate diet.
Collapse
Affiliation(s)
- Seungmin Song
- Graduate Program in System Health Science and Engineering, Ewha Womans University, Seoul 03760, Republic of Korea
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Jinyoung Shon
- Graduate Program in System Health Science and Engineering, Ewha Womans University, Seoul 03760, Republic of Korea
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Woo-ri Yang
- Hyundai Greenfood Greating Laboratory, Yongin-si 16827, Republic of Korea
| | - Han-Bit Kang
- Hyundai Greenfood Greating Laboratory, Yongin-si 16827, Republic of Korea
| | - Keun-Ha Kim
- Hyundai Greenfood Greating Laboratory, Yongin-si 16827, Republic of Korea
| | - Ju-Yeon Park
- Hyundai Greenfood Greating Laboratory, Yongin-si 16827, Republic of Korea
| | - Sanghoo Lee
- SCL Healthcare Inc., Yongin-si 16954, Republic of Korea
| | - Sae Yun Baik
- Hanaro Medical Foundation, Seoul 03159, Republic of Korea
| | - Kyoung-Ryul Lee
- SCL Healthcare Inc., Yongin-si 16954, Republic of Korea
- Hanaro Medical Foundation, Seoul 03159, Republic of Korea
| | - Yoon Jung Park
- Graduate Program in System Health Science and Engineering, Ewha Womans University, Seoul 03760, Republic of Korea
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul 03760, Republic of Korea
| |
Collapse
|
19
|
Liu C, Zeng H, Jiang R, Wang K, Ouyang J, Wen S, Peng L, Xu H, Huang J, Liu Z. Effects of Mulberry Leaf Fu Tea on the Intestines and Intestinal Flora of Goto-Kakizaki Type 2 Diabetic Rats. Foods 2023; 12:4006. [PMID: 37959125 PMCID: PMC10648540 DOI: 10.3390/foods12214006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 10/29/2023] [Accepted: 10/30/2023] [Indexed: 11/15/2023] Open
Abstract
Type 2 diabetes mellitus is a disease caused by hyperglycemia, an imbalance in the intestinal flora and disruption of the endocrine system. At present, it is primarily controlled through drug treatment and an improved diet. Mulberry leaf and fu brick tea were considered to have excellent hypoglycemic effects. This study used mulberry leaves and fu brick tea as raw materials to develop a dietary regulator that can assist in the prevention and alleviation of diabetes. The experiment used the Goto-Kakizaki (GK) rat model to investigate the hypoglycemic effect of mulberry leaf fu tea (MFT) and its influence on the intestinal flora of diabetic rats through methods including ELISA, tissue section observation and 16S RNA microbial sequencing. The results showed that, compared with the GK group, the intervention of mulberry leaf fu tea significantly reduced the activities of α-glucosidase (p < 0.05) and α-amylase (p < 0.05) in the duodenum of GK diabetic rats. The height of the duodenal villi was significantly reduced (p < 0.001), leading to decreased intestinal sugar absorption. At the same time, MFT alleviates the imbalance of intestinal flora caused by high blood sugar, promotes the growth of beneficial bacteria (Lactobacillus, Bifidobacterium, etc.), and inhibits the reproduction of harmful bacteria (Blautia, Klebsiella, Helicobacter, Alistipes, etc.). MFT helps reduce the secretion of toxic substances (lipopolysaccharide, p < 0.001), decreases oxidative stress and inflammation, mitigates organ damage, and improves symptoms of diabetes. Finally, the random blood glucose value of GK rats dropped from 22.79 mmol/L to 14.06 mmol/L. In summary, mulberry leaf fu tea can lower sugar absorption in diabetic rats, reduce the body's oxidative stress and inflammatory response, regulate intestinal flora, and reduce blood sugar levels in GK rats. It is hinted that mulberry leaf fu tea could be used as a functional drink to help prevent the occurrence of diabetes.
Collapse
Affiliation(s)
- Changwei Liu
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha 410128, China
| | - Hongzhe Zeng
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha 410128, China
| | - Ronggang Jiang
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha 410128, China
| | - Kuofei Wang
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha 410128, China
| | - Jian Ouyang
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha 410128, China
| | - Shuai Wen
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha 410128, China
| | - Liyuan Peng
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha 410128, China
| | - Hao Xu
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha 410128, China
| | - Jianan Huang
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha 410128, China
- National Research Center of Engineering and Technology for Utilization of Botanical Functional Ingredients, Changsha 410128, China
- Co-Innovation Center of Education Ministry for Utilization of Botanical Functional Ingredients, Changsha 410128, China
- Key Laboratory for Evaluation and Utilization of Gene Resources of Horticultural Crops, Ministry of Agriculture and Rural Affairs of China, Hunan Agricultural University, Changsha 410128, China
| | - Zhonghua Liu
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha 410128, China
- National Research Center of Engineering and Technology for Utilization of Botanical Functional Ingredients, Changsha 410128, China
- Co-Innovation Center of Education Ministry for Utilization of Botanical Functional Ingredients, Changsha 410128, China
- Key Laboratory for Evaluation and Utilization of Gene Resources of Horticultural Crops, Ministry of Agriculture and Rural Affairs of China, Hunan Agricultural University, Changsha 410128, China
| |
Collapse
|
20
|
Wu B, Cox AD, Chang H, Kennett M, Rosa C, Chopra S, Li S, Reddivari L. Maize near-isogenic lines with enhanced flavonoids alleviated dextran sodium sulfate-induced murine colitis via modulation of the gut microbiota. Food Funct 2023; 14:9606-9616. [PMID: 37814601 DOI: 10.1039/d3fo02953k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/11/2023]
Abstract
The rising incidence of inflammatory bowel disease (IBD) has necessitated the search for safe and effective novel therapeutic strategies. Dietary flavonoids exhibited antioxidant, antiproliferative, and anticarcinogenic activities in several model systems with proven abilities to reduce inflammation and oxidative stress, thus they could be promising therapeutic agents for IBD prevention/treatment. However, understanding the role of a specific class of compounds in foods that promote health is difficult because of the chemically complex food matrices. This study aimed to utilize four maize near-isogenic lines to determine the anti-colitis effects of specific classes of flavonoids, anthocyanins and/or phlobaphenes, in a whole-food matrix. Results showed that the intake of anthocyanin and phlobaphene-enriched maize diets effectively alleviated dextran sodium sulfate (DSS)-induced colitis in mice via reducing the intestinal permeability and restoring the barrier function. Anthocyanin diets were more effective in maintaining the crypt structure and muc2 protein levels and reducing inflammation. Bacterial communities of mice consuming diets enriched with anthocyanins and phlobaphenes were more similar to the healthy control compared to the DSS control group, suggesting the role of flavonoids in modulating the gut microbiota to retrieve intestinal homeostasis. Microbiota depletion rendered these compounds ineffective against colitis. Lower serum concentrations of several phenolic acids were detected in the microbiota-depleted mice, indicating that gut microbiota plays a role in flavonoid metabolism and bioavailability.
Collapse
Affiliation(s)
- Binning Wu
- Department of Food Science, Purdue University, West Lafayette, Indiana, USA.
- Department of Plant Science, The Pennsylvania State University, University Park, Pennsylvania, USA
- Interdisciplinary Graduate Program in Plant Biology, The Pennsylvania State University, University Park, PA, USA
| | - Abigail D Cox
- Department of Comparative Pathobiology, Purdue University, West Lafayette, Indiana, USA
| | - Haotian Chang
- Department of Food Science, Purdue University, West Lafayette, Indiana, USA.
| | - Mary Kennett
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, Pennsylvania, USA
| | - Cristina Rosa
- Interdisciplinary Graduate Program in Plant Biology, The Pennsylvania State University, University Park, PA, USA
- Department of Plant Pathology and Environmental Microbiology, The Pennsylvania State University, University Park, Pennsylvania, USA
| | - Surinder Chopra
- Department of Plant Science, The Pennsylvania State University, University Park, Pennsylvania, USA
- Interdisciplinary Graduate Program in Plant Biology, The Pennsylvania State University, University Park, PA, USA
| | - Shiyu Li
- Department of Food Science, Purdue University, West Lafayette, Indiana, USA.
| | - Lavanya Reddivari
- Department of Food Science, Purdue University, West Lafayette, Indiana, USA.
| |
Collapse
|
21
|
Wilson SMG, Peach JT, Fausset H, Miller ZT, Walk ST, Yeoman CJ, Bothner B, Miles MP. Metabolic impact of polyphenol-rich aronia fruit juice mediated by inflammation status of gut microbiome donors in humanized mouse model. Front Nutr 2023; 10:1244692. [PMID: 37727634 PMCID: PMC10505616 DOI: 10.3389/fnut.2023.1244692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 08/16/2023] [Indexed: 09/21/2023] Open
Abstract
Background The Aronia melanocarpa fruit is emerging as a health food owing to its high polyphenolic content and associated antioxidant activity. Antioxidant-rich foods, such as Aronia fruit, may counter inflammatory stimuli and positively modulate the gut microbiome. However, a comprehensive study characterizing the impact of Aronia fruit supplementation has not been completed. Therefore, we completed analyses measuring the metabolic, microbial, and inflammatory effects of a diet supplemented with Aronia fruit juice. Method Humanized mice were generated by colonizing gnotobiotic mice with microbiomes from human donors presenting disparate inflammation levels. Blood and fecal samples were collected throughout the course of an 8-week dietary intervention with either Aronia juice or a carbohydrate-matched beverage alone (2 weeks) or in combination with a high-fat diet to induce inflammation (6 weeks). Samples were analyzed using 16S rRNA gene sequencing (stool) and liquid chromatography-mass spectrometry (serum). Results We demonstrated transfer of microbiome composition and diversity and metabolic characteristics from humans with low and high inflammation levels to second-generation humanized mice. Aronia supplementation provided robust protection against high-fat diet induced metabolic and microbiome changes that were dependent in part on microbiome donor. Aronia induced increases in bacteria of the Eggerthellaceae genus (7-fold) which aligns with its known ability to metabolize (poly)phenols and in phosphatidylcholine metabolites which are consistent with improved gut barrier function. The gut microbiome from a low inflammation phenotype donor provided protection against high-fat diet induced loss of microbiome β-diversity and global metabolomic shifts compared to that from the high inflammation donor. Conclusion These metabolic changes elucidate pathway-specific drivers of reduced inflammation stemming from both Aronia and the gut microbiota.
Collapse
Affiliation(s)
- Stephanie M. G. Wilson
- Department of Food Systems, Nutrition, and Kinesiology, Montana State University, Bozeman, MT, United States
| | - Jesse T. Peach
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, MT, United States
| | - Hunter Fausset
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, MT, United States
| | - Zachary T. Miller
- Department of Research Centers, Montana State University, Bozeman, MT, United States
| | - Seth T. Walk
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT, United States
| | - Carl J. Yeoman
- Department of Animal and Range Sciences, Montana State University, Bozeman, MT, United States
| | - Brian Bothner
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, MT, United States
| | - Mary P. Miles
- Department of Food Systems, Nutrition, and Kinesiology, Montana State University, Bozeman, MT, United States
| |
Collapse
|
22
|
Mishima MDV, Martino HSD, Kolba N, Agarwal N, Jackson C, da Silva BP, Grancieri M, de Assis A, de São José VPB, Tako E. Chia Phenolic Extract Appear to Improve Small Intestinal Functionality, Morphology, Bacterial Populations, and Inflammation Biomarkers In Vivo ( Gallus gallus). Nutrients 2023; 15:3643. [PMID: 37630833 PMCID: PMC10458096 DOI: 10.3390/nu15163643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/07/2023] [Accepted: 08/17/2023] [Indexed: 08/27/2023] Open
Abstract
Phenolic compounds can act as a substrate for colonic resident microbiota. Once the metabolites are absorbed and distributed throughout the body, they can have diverse effects on the gut. The objective of this study was to evaluate the effects of the intra-amniotic administration of a chia phenolic extract on intestinal inflammation, intestinal barrier, brush border membrane functionality, intestinal microbiota, and morphology in vivo (Gallus gallus model). Cornish-cross fertile broiler eggs, at 17 days of embryonic incubation, were separated into groups as follows: non-injected (NI; this group did not receive an injection); 18 MΩ H2O (H2O; injected with ultrapure water), and 10 mg/mL (1%) chia phenolic extract (CPE; injected with phenolic extract diluted in ultrapure water). Immediately after hatch (21 days), chickens were euthanized and their small intestine, cecum, and cecum content were collected and analyzed. The chia phenolic extract reduced the tumor necrosis factor-alpha (TNF-α) and increased the sucrose isomaltase (SI) gene expression, reduced the Bifidobacterium and E. coli populations, reduced the Paneth cell diameter, increased depth crypt, and maintained villus height compared to the non-injected control group. Chia phenolic extract may be a promising beneficial compound for improving intestinal health, demonstrating positive changes in intestinal inflammation, functionality, microbiota, and morphology.
Collapse
Affiliation(s)
- Marcella Duarte Villas Mishima
- Department of Food Science, Stocking Hall, Cornell University, Ithaca, NY 14853, USA; (M.D.V.M.); (N.K.); (N.A.); (C.J.)
| | - Hércia Stampini Duarte Martino
- Department of Nutrition and Health, Federal University of Viçosa, Viçosa 36570-900, MG, Brazil; (H.S.D.M.); (B.P.d.S.); (M.G.); (A.d.A.); (V.P.B.d.S.J.)
| | - Nikolai Kolba
- Department of Food Science, Stocking Hall, Cornell University, Ithaca, NY 14853, USA; (M.D.V.M.); (N.K.); (N.A.); (C.J.)
| | - Nikita Agarwal
- Department of Food Science, Stocking Hall, Cornell University, Ithaca, NY 14853, USA; (M.D.V.M.); (N.K.); (N.A.); (C.J.)
| | - Cydney Jackson
- Department of Food Science, Stocking Hall, Cornell University, Ithaca, NY 14853, USA; (M.D.V.M.); (N.K.); (N.A.); (C.J.)
| | - Bárbara Pereira da Silva
- Department of Nutrition and Health, Federal University of Viçosa, Viçosa 36570-900, MG, Brazil; (H.S.D.M.); (B.P.d.S.); (M.G.); (A.d.A.); (V.P.B.d.S.J.)
| | - Mariana Grancieri
- Department of Nutrition and Health, Federal University of Viçosa, Viçosa 36570-900, MG, Brazil; (H.S.D.M.); (B.P.d.S.); (M.G.); (A.d.A.); (V.P.B.d.S.J.)
| | - Andressa de Assis
- Department of Nutrition and Health, Federal University of Viçosa, Viçosa 36570-900, MG, Brazil; (H.S.D.M.); (B.P.d.S.); (M.G.); (A.d.A.); (V.P.B.d.S.J.)
| | | | - Elad Tako
- Department of Food Science, Stocking Hall, Cornell University, Ithaca, NY 14853, USA; (M.D.V.M.); (N.K.); (N.A.); (C.J.)
| |
Collapse
|
23
|
Lela L, Russo D, De Biasio F, Gorgoglione D, Ostuni A, Ponticelli M, Milella L. Solanum aethiopicum L. from the Basilicata Region Prevents Lipid Absorption, Fat Accumulation, Oxidative Stress, and Inflammation in OA-Treated HepG2 and Caco-2 Cell Lines. PLANTS (BASEL, SWITZERLAND) 2023; 12:2859. [PMID: 37571013 PMCID: PMC10421219 DOI: 10.3390/plants12152859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/28/2023] [Accepted: 07/31/2023] [Indexed: 08/13/2023]
Abstract
Obesity is widely associated with intestine barrier impairment, nonalcoholic fatty liver disease (NAFLD) outbreaks, oxidative stress, and inflammation. In a previous investigation, the Solanum aethiopicum L. growing in Basilicata Region has demonstrated to have antioxidant activity; hence this investigation was aimed to evaluate for the first time the antilipidemic and anti-inflammatory activity of the Lucanian S. aethiopicum L. peel extract in vitro on OA-treated HepG2 and Caco-2 cell lines. It was shown that the extract could reduce lipogenesis by down-regulating SREBP-1c and HMGCR expression and fatty acid β-oxidation by up-regulating PPARα, CPT1A, and UCP2 expression. In addition, the S. aethiopicum L. peel extract might also improve oxidative stress by reducing endoplasmic reticulum stress and regulating the Nrf2 and Nf-κB molecular pathways. Altogether, these results demonstrated for the first time the possible application of the Lucanian S. aethiopicum peel extract for preventing obesity and managing NAFLD.
Collapse
Affiliation(s)
- Ludovica Lela
- Department of Science, University of Basilicata, V.le Ateneo Lucano 10, 85100 Potenza, Italy; (L.L.); (D.R.); (A.O.)
| | - Daniela Russo
- Department of Science, University of Basilicata, V.le Ateneo Lucano 10, 85100 Potenza, Italy; (L.L.); (D.R.); (A.O.)
- Spinoff Bioactiplant s.r.l., Via dell’Ateneo Lucano 10, 85100 Potenza, Italy
| | | | | | - Angela Ostuni
- Department of Science, University of Basilicata, V.le Ateneo Lucano 10, 85100 Potenza, Italy; (L.L.); (D.R.); (A.O.)
| | - Maria Ponticelli
- Department of Science, University of Basilicata, V.le Ateneo Lucano 10, 85100 Potenza, Italy; (L.L.); (D.R.); (A.O.)
| | - Luigi Milella
- Department of Science, University of Basilicata, V.le Ateneo Lucano 10, 85100 Potenza, Italy; (L.L.); (D.R.); (A.O.)
| |
Collapse
|
24
|
Subramaniam S, Kamath S, Ariaee A, Prestidge C, Joyce P. The impact of common pharmaceutical excipients on the gut microbiota. Expert Opin Drug Deliv 2023; 20:1297-1314. [PMID: 37307224 DOI: 10.1080/17425247.2023.2223937] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 06/07/2023] [Indexed: 06/14/2023]
Abstract
INTRODUCTION Increasing attention is being afforded to understanding the bidirectional relationships that exist between oral medications and the gut microbiota, in an attempt to optimize pharmacokinetic performance and mitigate unwanted side effects. While a wealth of research has investigated the direct impact of active pharmaceutical ingredients (APIs) on the gut microbiota, the interactions between inactive pharmaceutical ingredients (i.e. excipients) and the gut microbiota are commonly overlooked, despite excipients typically representing over 90% of the final dosage form. AREAS COVERED Known excipient-gut microbiota interactions for various classes of inactive pharmaceutical ingredients, including solubilizing agents, binders, fillers, sweeteners, and color additives, are reviewed in detail. EXPERT OPINION Clear evidence indicates that orally administered pharmaceutical excipients directly interact with gut microbes and can either positively or negatively impact gut microbiota diversity and composition. However, these relationships and mechanisms are commonly overlooked during drug formulation, despite the potential for excipient-microbiota interactions to alter drug pharmacokinetics and interfere with host metabolic health. The insights derived from this review will inform pharmaceutical scientists with the necessary design considerations for mitigating potential adverse pharmacomicrobiomic interactions when formulating oral dosage forms, ultimately providing clear avenues for improving therapeutic safety and efficacy.
Collapse
Affiliation(s)
- Santhni Subramaniam
- UniSA Clinical & Health Sciences, University of South Australia, Adelaide, Australia
| | - Srinivas Kamath
- UniSA Clinical & Health Sciences, University of South Australia, Adelaide, Australia
| | - Amin Ariaee
- UniSA Clinical & Health Sciences, University of South Australia, Adelaide, Australia
| | - Clive Prestidge
- UniSA Clinical & Health Sciences, University of South Australia, Adelaide, Australia
| | - Paul Joyce
- UniSA Clinical & Health Sciences, University of South Australia, Adelaide, Australia
| |
Collapse
|
25
|
Kaur H, Kaur G, Ali SA. IL-33's role in the gut immune system: A comprehensive review of its crosstalk and regulation. Life Sci 2023; 327:121868. [PMID: 37330043 DOI: 10.1016/j.lfs.2023.121868] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 06/02/2023] [Accepted: 06/14/2023] [Indexed: 06/19/2023]
Abstract
The intestinal tract is the largest immune organ in the human body, comprising a complex network of immune cells and epithelial cells that perform a variety of functions such as nutrient absorption, digestion, and waste excretion. Maintenance of homeostasis and effective responses to injury in the colonic epithelium are crucial for maintaining homeostasis between these two cell types. The onset and perpetuation of gut inflammation, characterizing inflammatory bowel diseases (IBD), are triggered by constitutive dysregulation of cytokine production. IL-33 is a newly characterized cytokine that has emerged as a critical modulator of inflammatory disorders. IL-33 is constitutively expressed in the nuclei of different cell types such as endothelial, epithelial, and fibroblast-like cells. Upon tissue damage or pathogen encounter, IL-33 is released as an alarmin and signals through a heterodimer receptor that consists of serum Stimulation-2 (ST2) and IL-1 receptor accessory protein (IL-1RAcP). IL-33 has the ability to induce Th2 cytokine production and enhance both Th1 and Th2, as well as Th17 immune responses. Exogenous administration of IL-33 in mice caused pathological changes in most mucosal tissues such as the lung and the gastrointestinal (GI) tract associated with increased production of type 2 cytokines and chemokines. In vivo and in vitro, primary studies have exhibited that IL-33 can activate Th2 cells, mast cells, or basophils to produce type 2 cytokines such as IL-4, IL-5, and IL-13. Moreover, several novel cell populations, collectively referred to as "type 2 innate lymphoid cells," were identified as being IL-33 responsive and are thought to be important for initiating type 2 immunity. Nevertheless, the underlying mechanisms by which IL-33 promotes type 2 immunity in the GI tract remain to be fully understood. Recently, it has been discovered that IL-33 plays important roles in regulatory immune responses. Highly suppressive ST2 + FoxP3+ Tregs subsets regulated by IL-33 were identified in several tissues, including lymphoid organs, gut, lung, and adipose tissues. This review aims to comprehensively summarize the current knowledge on IL-33's role in the gut immune system, its crosstalk, and regulation. The article will provide insights into the potential applications of IL-33-based therapies in the treatment of gut inflammatory disorders.
Collapse
Affiliation(s)
- Harpreet Kaur
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Gurjeet Kaur
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, NSW 2052, Australia; Mark Wainwright Analytical Centre, Bioanalytical Mass Spectrometry Facility, University of New South Wales, Sydney, NSW 2052, Australia
| | - Syed Azmal Ali
- Division Proteomics of Stem Cells and Cancer, German Cancer Research Center, 69120 Heidelberg, Germany.
| |
Collapse
|
26
|
Miguéns-Gómez A, Sierra-Cruz M, Blay MT, Rodríguez-Gallego E, Beltrán-Debón R, Terra X, Pinent M, Ardévol A. GSPE Pre-Treatment Exerts Long-Lasting Preventive Effects against Aging-Induced Changes in the Colonic Enterohormone Profile of Female Rats. Int J Mol Sci 2023; 24:ijms24097807. [PMID: 37175514 PMCID: PMC10177949 DOI: 10.3390/ijms24097807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 04/22/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023] Open
Abstract
The impact that healthy aging can have on society has raised great interest in understanding aging mechanisms. However, the effects this biological process may have on the gastrointestinal tract (GIT) have not yet been fully described. Results in relation to changes observed in the enteroendocrine system along the GIT are controversial. Grape seed proanthocyanidin extracts (GSPE) have been shown to protect against several pathologies associated with aging. Based on previous results, we hypothesized that a GSPE pre-treatment could prevent the aging processes that affect the enteroendocrine system. To test this hypothesis, we treated 21-month-old female rats with GSPE for 10 days. Eleven weeks after the treatment, we analyzed the effects of GSPE by comparing these aged animals with young animals. Aging induced a greater endocrine response to stimulation in the upper GIT segments (cholecystokinin (CCK) and glucagon-like peptide-1 (GLP-1)), a decrease in the mRNA abundance of GLP-1, peptide YY (PYY) and chromogranin A (ChgA) in the colon, and an increase in colonic butyrate. GSPE-treated rats were protected against a decrease in enterohormone expression in the colon. This effect is not directly related to the abundance of microbiome or short-chain fatty acids (SCFA) at this location. GSPE may therefore be effective in preventing a decrease in the colonic abundance of enterohormone expression induced by aging.
Collapse
Affiliation(s)
- Alba Miguéns-Gómez
- MoBioFood Research Group, Departament de Bioquímica i Biotecnologia, Universitat Rovira i Virgili, c/Marcel·lí Domingo n°1, 43007 Tarragona, Spain
| | - Marta Sierra-Cruz
- MoBioFood Research Group, Departament de Bioquímica i Biotecnologia, Universitat Rovira i Virgili, c/Marcel·lí Domingo n°1, 43007 Tarragona, Spain
| | - M Teresa Blay
- MoBioFood Research Group, Departament de Bioquímica i Biotecnologia, Universitat Rovira i Virgili, c/Marcel·lí Domingo n°1, 43007 Tarragona, Spain
- Institut d'Investigació Sanitària Pere Virgili (IISPV), 43005 Tarragona, Spain
| | - Esther Rodríguez-Gallego
- MoBioFood Research Group, Departament de Bioquímica i Biotecnologia, Universitat Rovira i Virgili, c/Marcel·lí Domingo n°1, 43007 Tarragona, Spain
- Institut d'Investigació Sanitària Pere Virgili (IISPV), 43005 Tarragona, Spain
| | - Raúl Beltrán-Debón
- MoBioFood Research Group, Departament de Bioquímica i Biotecnologia, Universitat Rovira i Virgili, c/Marcel·lí Domingo n°1, 43007 Tarragona, Spain
- Institut d'Investigació Sanitària Pere Virgili (IISPV), 43005 Tarragona, Spain
| | - Ximena Terra
- MoBioFood Research Group, Departament de Bioquímica i Biotecnologia, Universitat Rovira i Virgili, c/Marcel·lí Domingo n°1, 43007 Tarragona, Spain
- Institut d'Investigació Sanitària Pere Virgili (IISPV), 43005 Tarragona, Spain
| | - Montserrat Pinent
- MoBioFood Research Group, Departament de Bioquímica i Biotecnologia, Universitat Rovira i Virgili, c/Marcel·lí Domingo n°1, 43007 Tarragona, Spain
- Institut d'Investigació Sanitària Pere Virgili (IISPV), 43005 Tarragona, Spain
| | - Anna Ardévol
- MoBioFood Research Group, Departament de Bioquímica i Biotecnologia, Universitat Rovira i Virgili, c/Marcel·lí Domingo n°1, 43007 Tarragona, Spain
- Institut d'Investigació Sanitària Pere Virgili (IISPV), 43005 Tarragona, Spain
| |
Collapse
|
27
|
Wang H, Chen L, Yang B, Du J, Chen L, Li Y, Guo F. Structures, Sources, Identification/Quantification Methods, Health Benefits, Bioaccessibility, and Products of Isorhamnetin Glycosides as Phytonutrients. Nutrients 2023; 15:nu15081947. [PMID: 37111165 PMCID: PMC10143801 DOI: 10.3390/nu15081947] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/12/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023] Open
Abstract
In recent years, people have tended to consume phytonutrients and nutrients in their daily diets. Isorhamnetin glycosides (IGs) are an essential class of flavonoids derived from dietary and medicinal plants such as Opuntia ficus-indica, Hippophae rhamnoides, and Ginkgo biloba. This review summarizes the structures, sources, quantitative and qualitative analysis technologies, health benefits, bioaccessibility, and marketed products of IGs. Routine and innovative assay methods, such as IR, TLC, NMR, UV, MS, HPLC, UPLC, and HSCCC, have been widely used for the characterization and quantification of IGs. All of the therapeutic effects of IGs discovered to date are collected and discussed in this study, with an emphasis on the relevant mechanisms of their health-promoting effects. IGs exhibit diverse biological activities against cancer, diabetes, hepatic diseases, obesity, and thrombosis. They exert therapeutic effects through multiple networks of underlying molecular signaling pathways. Owing to these benefits, IGs could be utilized to make foods and functional foods. IGs exhibit higher bioaccessibility and plasma concentrations and longer average residence time in blood than aglycones. Overall, IGs as phytonutrients are very promising and have excellent application potential.
Collapse
Affiliation(s)
- Hong Wang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Lijia Chen
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Binrui Yang
- Nutrition Science, Amway (Shanghai) Innovation & Science Co., Ltd., Shanghai 201203, China
| | - Jun Du
- Nutrition Science, Amway (Shanghai) Innovation & Science Co., Ltd., Shanghai 201203, China
| | - Liang Chen
- Nutrition Science, Amway (Shanghai) Innovation & Science Co., Ltd., Shanghai 201203, China
| | - Yiming Li
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Fujiang Guo
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
28
|
Wu J, Zhang D, Zhao M, Zheng X. Gut Microbiota Dysbiosis and Increased NLRP3 Levels in Patients with Pregnancy-Induced Hypertension. Curr Microbiol 2023; 80:168. [PMID: 37024673 PMCID: PMC10079714 DOI: 10.1007/s00284-023-03252-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 03/01/2023] [Indexed: 04/08/2023]
Abstract
Pregnancy-induced hypertension (PIH) is one of the most common diseases, causing high maternal morbidity and mortality. However, the correlation of gut microbiota in PIH has not been reported. Our aim was to characterize the intestinal microbiota of patients with PIH compared with healthy people. We analyzed and compared the gut microbiota communities in the feces of 28 PIH patients with pregnancy(not pre-pregnancy) body mass index (including height and weight)-matched healthy controls using 16S rRNA gene sequencing and then investigate the relationships among gut microbiota, cytokines, and PIH. Compared with the healthy group, microbial α diversity was lower in the PIH group, but not statistically significant different. At the phylum level, Firmicutes, Bacteroidetes, Proteobacteria, and Actinobacteria exhibited obvious differences between the PIH and control groups. LEfSe analysis found 33 differentially abundant taxa between the two groups. The production of pro-inflammatory cytokines in PIH serum or placenta tissues was higher than that of the control group. In addition to alterations in gut microbiota composition, we also found that the Bac_Prevotellaceae, Pre_Prevotella bacteria were positively correlated with NLRP3 level, but negatively correlated with Bac_Bacteroidaceae, Bac_Bacteroides. PIH patients had gut microbiota dysbiosis and increased NLRP3 levels, which will lead to a better understanding of the relationship between the gut microbiota and PIH.
Collapse
Affiliation(s)
- Jingjing Wu
- Department of Obstetrics, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Dongmei Zhang
- Department of Obstetrics, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China.
| | - Meijing Zhao
- Department of Obstetrics, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Xiaowei Zheng
- Clinical Laboratory, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| |
Collapse
|
29
|
Liang L, Saunders C, Sanossian N. Food, gut barrier dysfunction, and related diseases: A new target for future individualized disease prevention and management. Food Sci Nutr 2023; 11:1671-1704. [PMID: 37051344 PMCID: PMC10084985 DOI: 10.1002/fsn3.3229] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 12/20/2022] [Accepted: 12/28/2022] [Indexed: 03/09/2023] Open
Abstract
Dysfunction of gut barrier is known as "leaky gut" or increased intestinal permeability. Numerous recent scientific evidences showed the association between gut dysfunction and multiple gastrointestinal tract (GI) and non-GI diseases. Research also demonstrated that food plays a crucial role to cause or remedy gut dysfunction related to diseases. We reviewed recent articles from electronic databases, mainly PubMed. The data were based on animal models, cell models, and human research in vivo and in vitro models. In this comprehensive review, our aim focused on the relationship between dietary factors, intestinal permeability dysfunction, and related diseases. This review synthesizes currently available literature and is discussed in three parts: (a) the mechanism of gut barrier and function, (b) food and dietary supplements that may promote gut health, and food or medication that may alter gut function, and (c) a table that organizes the synthesized information by general mechanisms for diseases related to leaky gut/intestinal permeability and associated dietary influences. With future research, dietary intervention could be a new target for individualized disease prevention and management.
Collapse
Affiliation(s)
- Linda Liang
- University of Southern CaliforniaLos AngelesCaliforniaUSA
| | | | - Nerses Sanossian
- Department of NeurologyMedical School of Southern CaliforniaLos AngelesCaliforniaUSA
| |
Collapse
|
30
|
Popiolek-Kalisz J, Glibowski P. Apple Peel Supplementation Potential in Metabolic Syndrome Prevention. Life (Basel) 2023; 13:life13030753. [PMID: 36983908 PMCID: PMC10056680 DOI: 10.3390/life13030753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/06/2023] [Accepted: 03/08/2023] [Indexed: 03/12/2023] Open
Abstract
(1) Introduction: Apples are a source of bioactive substances, e.g., anthocyanidins and flavonols, and dietary fiber. Their highest concentrations are observed in the skin. Metabolic syndrome (MetS) is a set of conditions originally associated with obesity. Excessive adipose tissue accompanying obesity leads to chronic inflammation and metabolic disorders, which result in the development of dyslipidemia, elevated blood pressure, and glucose levels. Thus, supplementation of apple peels, a source of antioxidant substances and fiber, could potentially be a method supporting the prevention of MetS. This paper summarizes the results of available research on the potential impact of apple peel supplementation on the components of MetS. (2) Results: The results from in vitro and animal model studies indicate a positive effect of apple peel supplementation on lipid profile, glucose levels, and blood pressure regulation mediators. Only one human study was performed, and it showed that the consumption of apple peels had an effect on endothelial function but not on other clinical parameters. At the moment, there are no results from observations on large groups of people available. (3) Conclusions: The results of in vitro and animal-model studies indicate the potential of apple peel supplementation in MetS prevention, but it has not been clinically confirmed in human studies. Conducting large human studies could allow a definite clarification of the role of apple peel supplementation in MetS prevention.
Collapse
Affiliation(s)
- Joanna Popiolek-Kalisz
- Clinical Dietetics Unit, Department of Bioanalytics, Medical University of Lublin, 20-093 Lublin, Poland
- Department of Biotechnology, Microbiology and Human Nutrition, University of Life Sciences in Lublin, 20-704 Lublin, Poland
- Department of Cardiology, Cardinal Wyszynski Hospital in Lublin, 20-718 Lublin, Poland
- Correspondence:
| | - Paweł Glibowski
- Department of Biotechnology, Microbiology and Human Nutrition, University of Life Sciences in Lublin, 20-704 Lublin, Poland
| |
Collapse
|
31
|
Enichen E, Harvey C, Demmig-Adams B. COVID-19 Spotlights Connections between Disease and Multiple Lifestyle Factors. Am J Lifestyle Med 2023; 17:231-257. [PMID: 36883129 PMCID: PMC9445631 DOI: 10.1177/15598276221123005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The SARS-CoV-2 virus (severe acute respiratory syndrome coronavirus 2), and the disease it causes (COVID-19), have had a profound impact on global human society and threaten to continue to have such an impact with newly emerging variants. Because of the widespread effects of SARS-CoV-2, understanding how lifestyle choices impact the severity of disease is imperative. This review summarizes evidence for an involvement of chronic, non-resolving inflammation, gut microbiome disruption (dysbiosis with loss of beneficial microorganisms), and impaired viral defenses, all of which are associated with an imbalanced lifestyle, in severe disease manifestations and post-acute sequelae of SARS-CoV-2 (PASC). Humans' physiological propensity for uncontrolled inflammation and severe COVID-19 are briefly contrasted with bats' low propensity for inflammation and their resistance to viral disease. This insight is used to identify positive lifestyle factors with the potential to act in synergy for restoring balance to the immune response and gut microbiome, and thereby protect individuals against severe COVID-19 and PASC. It is proposed that clinicians should consider recommending lifestyle factors, such as stress management, balanced nutrition and physical activity, as preventative measures against severe viral disease and PASC.
Collapse
Affiliation(s)
- Elizabeth Enichen
- Department of Ecology and Evolutionary Biology, University of Colorado, Boulder, CO, USA (EE, CH, BDA)
| | - Caitlyn Harvey
- Department of Ecology and Evolutionary Biology, University of Colorado, Boulder, CO, USA (EE, CH, BDA)
| | - Barbara Demmig-Adams
- Department of Ecology and Evolutionary Biology, University of Colorado, Boulder, CO, USA (EE, CH, BDA)
| |
Collapse
|
32
|
Liu Z, Wang T, Zhu Y, Zhao H, Zhou Z, Wu Q. Improvements in Gut Microbiota Dysbiosis in Aged Mice Transplanted with Adipose-Derived Stem Cells. Stem Cells Dev 2023; 32:185-196. [PMID: 36680756 DOI: 10.1089/scd.2022.0257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Adipose-derived stem cells (ASCs), as a cell therapy with considerable therapeutic potential, have received increasing attention in tissue repair, endocrine regulation, immune regulation, and aging and obesity research. Gut microbiota are present in all organisms and play important roles in the development of aging and obesity. Dysbiosis activates inflammatory pathways that may contribute to the development of aging and obesity. We used C57BL/6 J mice of different ages to carry out the experiment. Young mice were used as donors for ASC. Feces from the three groups were collected for 16sRNA sequencing to analyze the species composition of intestinal microorganisms, and then, predicted metabolic pathways by PICRUSt2 using 16s rRNA gene sequences. Immune cell levels in abdominal adipose tissue were assessed by flow cytometry. The content of IL-6, IL-1β, TNF-α, and lipopolysaccharides in serum was measured by ELISA kit. Our 16sRNA sequencing data showed restoration of gut microbiota diversity and an increase in beneficial flora (Akkermansia, Lactobacillus, Prevotella) 7 days after ASC transplantation. In addition, the inflammatory environment improved in older transplanted mice.
Collapse
Affiliation(s)
- Zebiao Liu
- Guangxi Universities Key Laboratory of Stem Cell and Biopharmaceutical Technology, Research Center for Biomedical Sciences, School of Life Sciences, Guangxi Normal University, Guilin, China
| | - Tao Wang
- Guangxi Universities Key Laboratory of Stem Cell and Biopharmaceutical Technology, Research Center for Biomedical Sciences, School of Life Sciences, Guangxi Normal University, Guilin, China
| | - Yu Zhu
- Guangxi Universities Key Laboratory of Stem Cell and Biopharmaceutical Technology, Research Center for Biomedical Sciences, School of Life Sciences, Guangxi Normal University, Guilin, China
| | - Hongxia Zhao
- Guangxi Universities Key Laboratory of Stem Cell and Biopharmaceutical Technology, Research Center for Biomedical Sciences, School of Life Sciences, Guangxi Normal University, Guilin, China
| | - Zuping Zhou
- Guangxi Universities Key Laboratory of Stem Cell and Biopharmaceutical Technology, Research Center for Biomedical Sciences, School of Life Sciences, Guangxi Normal University, Guilin, China
| | - Qiong Wu
- Guangxi Universities Key Laboratory of Stem Cell and Biopharmaceutical Technology, Research Center for Biomedical Sciences, School of Life Sciences, Guangxi Normal University, Guilin, China
| |
Collapse
|
33
|
Zhou J, Yue J, Yao Y, Hou P, Zhang T, Zhang Q, Yi L, Mi M. Dihydromyricetin Protects Intestinal Barrier Integrity by Promoting IL-22 Expression in ILC3s through the AMPK/SIRT3/STAT3 Signaling Pathway. Nutrients 2023; 15:nu15020355. [PMID: 36678226 PMCID: PMC9861697 DOI: 10.3390/nu15020355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/06/2023] [Accepted: 01/08/2023] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Previous studies indicate that dihydromyricetin (DHM) could alleviate intestinal inflammation and improve intestinal barrier integrity, yet the underlying mechanism remains obscure. METHODS C57BL/6 male mice were fed with a control diet, high-fat diet (HFD), or HFD + DHM diet for 12 weeks. The intestinal permeability and expression of intestinal tight junction (TJ) protein were detected to evaluate the effects of DHM on intestinal barrier integrity. The interleukin 22 (IL-22) production of group 3 innate lymphoid cells (ILC3s) in small intestine lamina propria was tested to clarify the effects of DHM on ILC3s. In addition, an MNK3 cell line, which expresses the same transcription factors and cytokines as ILC3, was used to investigate the molecular mechanism under DHM-induced IL-22 expression. RESULTS DHM effectively protected HFD-fed mice against intestinal barrier destruction by promoting ILC3 activation and IL-22 secretion, and IL-22 expression increased the expression levels of TJ molecules to protect intestinal barrier integrity. Moreover, DHM increased activation of the AMPK/SIRT3/STAT3 pathway, which in turn promoted IL-22 expression in MNK3 cells. CONCLUSIONS DHM improved IL-22 production in ILC3 cells to alleviate HFD-induced intestinal barrier destruction via the AMPK/SIRT3/STAT3 pathway.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Long Yi
- Correspondence: (L.Y.); or mantian (M.M.); Tel./Fax: +86-2368771549 (M.M.)
| | - Mantian Mi
- Correspondence: (L.Y.); or mantian (M.M.); Tel./Fax: +86-2368771549 (M.M.)
| |
Collapse
|
34
|
Xiong HH, Lin SY, Chen LL, Ouyang KH, Wang WJ. The Interaction between Flavonoids and Intestinal Microbes: A Review. Foods 2023; 12:foods12020320. [PMID: 36673411 PMCID: PMC9857828 DOI: 10.3390/foods12020320] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 12/27/2022] [Accepted: 01/04/2023] [Indexed: 01/11/2023] Open
Abstract
In recent years, research on the interaction between flavonoids and intestinal microbes have prompted a rash of food science, nutriology and biomedicine, complying with future research trends. The gut microbiota plays an essential role in the maintenance of intestinal homeostasis and human health, but once the intestinal flora dysregulation occurs, it may contribute to various diseases. Flavonoids have shown a variety of physiological activities, and are metabolized or biotransformed by gut microbiota, thereby producing new metabolites that promote human health by modulating the composition and structure of intestinal flora. Herein, this review demonstrates the key notion of flavonoids as well as intestinal microbiota and dysbiosis, aiming to provide a comprehensive understanding about how flavonoids regulate the diseases by gut microbiota. Emphasis is placed on the microbiota-flavonoid bidirectional interaction that affects the metabolic fate of flavonoids and their metabolites, thereby influencing their metabolic mechanism, biotransformation, bioavailability and bioactivity. Potentially by focusing on the abundance and diversity of gut microbiota as well as their metabolites such as bile acids, we discuss the influence mechanism of flavonoids on intestinal microbiota by protecting the intestinal barrier function and immune system. Additionally, the microbiota-flavonoid bidirectional interaction plays a crucial role in regulating various diseases. We explain the underlying regulation mechanism of several typical diseases including gastrointestinal diseases, obesity, diabetes and cancer, aiming to provide a theoretical basis and guideline for the promotion of gastrointestinal health as well as the treatment of diseases.
Collapse
Affiliation(s)
- Hui-Hui Xiong
- College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang 330045, China
| | - Su-Yun Lin
- College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang 330045, China
| | - Ling-Li Chen
- College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang 330045, China
| | - Ke-Hui Ouyang
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang 330045, China
| | - Wen-Jun Wang
- College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang 330045, China
- Correspondence: ; Tel.: +86-791-83813655
| |
Collapse
|
35
|
Li L, Yang HY, Ma Y, Liang XH, Xu M, Zhang J, Huang ZX, Meng LH, Zhou J, Xian J, Suo YJ, Huang S, Cai JW, Meng BH, Zhao ZY, Lu JL, Xu Y, Wang TG, Li M, Chen YH, Wang WQ, Bi YF, Ning G, Shen FX, Hu RY, Chen G, Chen L, Chen LL, Deng HC, Gao ZN, Huo YN, Li Q, Liu C, Mu YM, Qin GJ, Shi LX, Su Q, Wan Q, Wang GX, Wang SY, Wang YM, Wu SL, Xu YP, Yan L, Yang T, Ye Z, Yu XF, Zhang YF, Zhao JJ, Zeng TS, Tang XL, Qin YF, Luo ZJ. Whole fresh fruit intake and risk of incident diabetes in different glycemic stages: a nationwide prospective cohort investigation. Eur J Nutr 2023; 62:771-782. [PMID: 36261730 PMCID: PMC9941276 DOI: 10.1007/s00394-022-02998-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 08/31/2022] [Indexed: 11/04/2022]
Abstract
PURPOSE Fruit intake is beneficial to several chronic diseases, but controversial in diabetes. We aimed to investigate prospectively the associations of whole fresh fruit intake with risk of incident type 2 diabetes (T2D) in subjects with different glucose regulation capacities. METHODS The present study included 79,922 non-diabetic participants aged ≥ 40 years from an ongoing nationwide prospective cohort in China. Baseline fruit intake information was collected by a validated food frequency questionnaire. Plasma HbA1c, fasting and 2 h post-loading glucose levels were measured at both baseline and follow-up examinations. Cox proportional hazards models were used to calculate hazard ratio (HR) and 95% confidence intervals (CI) for incident diabetes among participants with normal glucose tolerance (NGT) and prediabetes, after adjusted for multiple confounders. Restricted cubic spline analysis was applied for dose-response relation. RESULTS During a median 3.8-year follow-up, 5886 (7.36%) participants developed diabetes. Overall, we identified a linear and dose-dependent inverse association between dietary whole fresh fruit intake and risk of incident T2D. Each 100 g/d higher fruit intake was associated with 2.8% lower risk of diabetes (HR 0.972, 95%CI [0.949-0.996], P = 0.0217), majorly benefiting NGT subjects with 15.2% lower risk (HR 0.848, 95%CI [0.766-0.940], P = 0.0017), while not significant in prediabetes (HR 0.981, 95%CI 0.957-4.005, P = 0.1268). Similarly, the inverse association was present in normoglycemia individuals with a 48.6% lower risk of diabetes when consuming fruits > 7 times/week comparing to those < 1 time/week (HR 0.514, 95% CI [0.368-0.948]), but not in prediabetes (HR 0.883, 95% CI [0.762-1.023]). CONCLUSION These findings suggest that higher frequency and amount of fresh fruit intake may protect against incident T2D, especially in NGT, but not in prediabetes, highlighting the dietary recommendation of higher fresh fruit consumption to prevent T2D in normoglycemia population.
Collapse
Affiliation(s)
- Li Li
- grid.412594.f0000 0004 1757 2961Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, No. 6 of Shuangyong Road, Nanning, 530021 Guangxi China
| | - Hai-Yan Yang
- grid.412594.f0000 0004 1757 2961Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, No. 6 of Shuangyong Road, Nanning, 530021 Guangxi China
| | - Yan Ma
- grid.412594.f0000 0004 1757 2961Department of Ultrasonography, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xing-Huan Liang
- grid.412594.f0000 0004 1757 2961Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, No. 6 of Shuangyong Road, Nanning, 530021 Guangxi China
| | - Min Xu
- grid.16821.3c0000 0004 0368 8293Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China ,Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, China
| | - Jie Zhang
- grid.412594.f0000 0004 1757 2961Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, No. 6 of Shuangyong Road, Nanning, 530021 Guangxi China
| | - Zhen-Xing Huang
- grid.412594.f0000 0004 1757 2961Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, No. 6 of Shuangyong Road, Nanning, 530021 Guangxi China
| | - Li-Heng Meng
- grid.412594.f0000 0004 1757 2961Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, No. 6 of Shuangyong Road, Nanning, 530021 Guangxi China
| | - Jia Zhou
- grid.412594.f0000 0004 1757 2961Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, No. 6 of Shuangyong Road, Nanning, 530021 Guangxi China
| | - Jing Xian
- grid.412594.f0000 0004 1757 2961Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, No. 6 of Shuangyong Road, Nanning, 530021 Guangxi China
| | - Ying-Jun Suo
- grid.412594.f0000 0004 1757 2961Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, No. 6 of Shuangyong Road, Nanning, 530021 Guangxi China
| | - Song Huang
- grid.412594.f0000 0004 1757 2961Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, No. 6 of Shuangyong Road, Nanning, 530021 Guangxi China
| | - Jin-Wei Cai
- grid.412594.f0000 0004 1757 2961Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, No. 6 of Shuangyong Road, Nanning, 530021 Guangxi China
| | - Bi-Hui Meng
- grid.412594.f0000 0004 1757 2961Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, No. 6 of Shuangyong Road, Nanning, 530021 Guangxi China
| | - Zhi-Yun Zhao
- grid.16821.3c0000 0004 0368 8293Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China ,Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, China
| | - Jie-Li Lu
- grid.16821.3c0000 0004 0368 8293Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China ,Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, China
| | - Yu Xu
- grid.16821.3c0000 0004 0368 8293Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China ,Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, China
| | - Tian-Ge Wang
- grid.16821.3c0000 0004 0368 8293Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China ,Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, China
| | - Mian Li
- grid.16821.3c0000 0004 0368 8293Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China ,Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, China
| | - Yu-Hong Chen
- grid.16821.3c0000 0004 0368 8293Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China ,Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, China
| | - Wei-Qing Wang
- grid.16821.3c0000 0004 0368 8293Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China ,Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, China
| | - Yu-Fang Bi
- grid.16821.3c0000 0004 0368 8293Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China ,Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, China
| | - Guang Ning
- grid.16821.3c0000 0004 0368 8293Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China ,Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, China
| | - Fei-Xia Shen
- grid.414906.e0000 0004 1808 0918The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Ru-Ying Hu
- grid.433871.aZhejiang Provincial Center for Disease Control and Prevention, Zhejiang, China
| | - Gang Chen
- grid.256112.30000 0004 1797 9307Fujian Provincial Hospital, Fujian Medical University, Fuzhou, China
| | - Li Chen
- grid.452402.50000 0004 1808 3430Qilu Hospital of Shandong University, Jinan, China
| | - Lu-Lu Chen
- grid.33199.310000 0004 0368 7223Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hua-Cong Deng
- grid.452206.70000 0004 1758 417XThe First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zheng-Nan Gao
- grid.452337.40000 0004 0644 5246Dalian Municipal Central Hospital, Dalian, China
| | - Ya-Nan Huo
- grid.415002.20000 0004 1757 8108Jiangxi Provincial People’s Hospital Affiliated to Nanchang University, Nanchang, China
| | - Qiang Li
- grid.412463.60000 0004 1762 6325The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chao Liu
- grid.412676.00000 0004 1799 0784Jiangsu Province Hospital on Integration of Chinese and Western Medicine, Nanjing, China
| | - Yi-Ming Mu
- grid.414252.40000 0004 1761 8894Chinese People’s Liberation Army General Hospital, Beijing, China
| | - Gui-Jun Qin
- grid.412633.10000 0004 1799 0733The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Li-Xin Shi
- grid.452244.1Affiliated Hospital of Guiyang Medical College, Guiyang, China
| | - Qing Su
- grid.412987.10000 0004 0630 1330Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qin Wan
- grid.488387.8The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Gui-Xia Wang
- grid.430605.40000 0004 1758 4110The First Hospital of Jilin University, Changchun, China
| | - Shuang-Yuan Wang
- grid.16821.3c0000 0004 0368 8293Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China ,Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, China
| | - You-Min Wang
- grid.412679.f0000 0004 1771 3402The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Sheng-Li Wu
- Karamay Municipal People’s Hospital, Xinjiang, China
| | - Yi-Ping Xu
- grid.16821.3c0000 0004 0368 8293Clinical Trials Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li Yan
- grid.12981.330000 0001 2360 039XSun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Tao Yang
- grid.412676.00000 0004 1799 0784The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhen Ye
- grid.433871.aZhejiang Provincial Center for Disease Control and Prevention, Zhejiang, China
| | - Xue-Feng Yu
- grid.33199.310000 0004 0368 7223Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yin-Fei Zhang
- grid.459667.fCentral Hospital of Shanghai Jiading District, Shanghai, China
| | - Jia-Jun Zhao
- grid.460018.b0000 0004 1769 9639Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Tian-Shu Zeng
- grid.33199.310000 0004 0368 7223Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xu-Lei Tang
- grid.412643.60000 0004 1757 2902The First Hospital of Lanzhou University, Lanzhou, China
| | - Ying-Fen Qin
- Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, No. 6 of Shuangyong Road, Nanning, 530021, Guangxi, China.
| | - Zuo-Jie Luo
- Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, No. 6 of Shuangyong Road, Nanning, 530021, Guangxi, China.
| | | |
Collapse
|
36
|
Chen S, Liu H, Zhang J, Zhou B, Zhuang S, He X, Wang T, Wang C. Effects of different levels of rutin on growth performance, immunity, intestinal barrier and antioxidant capacity of broilers. ITALIAN JOURNAL OF ANIMAL SCIENCE 2022. [DOI: 10.1080/1828051x.2022.2116732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Affiliation(s)
- Shun Chen
- College of Animal Science and Technology, National Experimental Teaching Demonstration Centre of Animal Science, Nanjing Agricultural University, Nanjing, China
| | - Huijuan Liu
- College of Animal Science and Technology, National Experimental Teaching Demonstration Centre of Animal Science, Nanjing Agricultural University, Nanjing, China
| | - Jiaqi Zhang
- College of Animal Science and Technology, National Experimental Teaching Demonstration Centre of Animal Science, Nanjing Agricultural University, Nanjing, China
| | - Binbin Zhou
- College of Animal Science and Technology, National Experimental Teaching Demonstration Centre of Animal Science, Nanjing Agricultural University, Nanjing, China
| | - Su Zhuang
- College of Animal Science and Technology, National Experimental Teaching Demonstration Centre of Animal Science, Nanjing Agricultural University, Nanjing, China
| | - Xiaofang He
- Co-Innovation Center for School of Animal Science and Food Engineering, Jinling Institute of Technology, Nanjing, China
| | - Tian Wang
- College of Animal Science and Technology, National Experimental Teaching Demonstration Centre of Animal Science, Nanjing Agricultural University, Nanjing, China
| | - Chao Wang
- College of Animal Science and Technology, National Experimental Teaching Demonstration Centre of Animal Science, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
37
|
Su L, Zeng Y, Li G, Chen J, Chen X. Quercetin improves high-fat diet-induced obesity by modulating gut microbiota and metabolites in C57BL/6J mice. Phytother Res 2022; 36:4558-4572. [PMID: 35906097 DOI: 10.1002/ptr.7575] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 07/09/2022] [Accepted: 07/12/2022] [Indexed: 12/13/2022]
Abstract
High-fat diet-induced obesity is characterized by low-grade inflammation, which has been linked to gut microbiota dysbiosis. We hypothesized that quercetin supplementation would alter gut microbiota and reduce inflammation in obese mice. Male C57BL/6J mice, 4 weeks of age, were divided into 3 groups, including a low-fat diet group, a high-fat diet (HFD) group, and a high-fat diet plus quercetin (HFD+Q) group. The mice in HFD+Q group were given 50 mg per kg BW quercetin by gavage for 20 weeks. The body weight, fat accumulation, gut barrier function, glucose tolerance, and adipose tissue inflammation were determined in mice. 16 s rRNA amplicon sequence and non-targeted metabolomics analysis were used to explore the alteration of gut microbiota and metabolites. We found that quercetin significantly alleviated HFD-induced obesity, improved glucose tolerance, recovered gut barrier function, and reduced adipose tissue inflammation. Moreover, quercetin ameliorated HFD-induced gut microbiota disorder by regulating the abundance of gut microbiota, such as Adlercreutzia, Allobaculum, Coprococcus_1, Lactococcus, and Akkermansia. Quercetin influenced the production of metabolites that were linked to alterations in obesity-related inflammation and oxidative stress, such as Glycerophospho-N-palmitoyl ethanolamine, sanguisorbic acid dilactone, O-Phospho-L-serine, and P-benzoquinone. Our results demonstrate that the anti-obesity effects of quercetin may be mediated through regulation in gut microbiota and metabolites.
Collapse
Affiliation(s)
- Lijie Su
- Department of Nutrition and Food Hygiene, School of Public Health, Guangzhou Medical University, Guangzhou, China
| | - Yupeng Zeng
- Department of Nutrition and Food Hygiene, School of Public Health, Guangzhou Medical University, Guangzhou, China
| | - Guokun Li
- Department of Nutrition and Food Hygiene, School of Public Health, Guangzhou Medical University, Guangzhou, China
| | - Jing Chen
- Sino-French Hoffmann Institute, School of Basic Medical Science, Guangzhou Medical University, Guangzhou, China
| | - Xiaoyi Chen
- Department of Nutrition and Food Hygiene, School of Public Health, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
38
|
Anti-Inflammatory and Antibacterial Effects and Mode of Action of Greek Arbutus, Chestnut, and Fir Honey in Mouse Models of Inflammation and Sepsis. Microorganisms 2022; 10:microorganisms10122374. [PMID: 36557628 PMCID: PMC9784341 DOI: 10.3390/microorganisms10122374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/23/2022] [Accepted: 11/24/2022] [Indexed: 12/03/2022] Open
Abstract
Background: Honey has been shown to possess anti-inflammatory and bactericidal properties that may be useful for the prevention and treatment of infections as well as of acute and chronic inflammatory diseases. The antimicrobial potency of honey could be attributed to its physicochemical characteristics combined with the presence of certain compounds, such as hydrogen peroxide and polyphenols. Honey's bacteriostatic or bactericidal capacity varies depending on its composition and the bacterial type of each infection. Nevertheless, not all honey samples possess anti-inflammatory or antibacterial properties and their mechanism of action has not been clearly elucidated. Objectives: We therefore investigated the anti-inflammatory properties of three different honey samples that derived from different geographical areas of Greece and different botanical origins, namely, arbutus, chestnut, and fir; they were compared to manuka honey, previously known for its anti-inflammatory and antibacterial activity. Materials and Methods: To test the anti-inflammatory activity of the different samples, we utilized the in vivo model of LPS-driven inflammation, which induces septic shock without the presence of pathogens. To evaluate the antibacterial action of the same honey preparations, we utilized the cecal-slurry-induced peritonitis model in mice. Since acute inflammation and sepsis reduce the biotransformation capacity of the liver, the expression of key enzymes in the process was also measured. Results: The administration of all Greek honey samples to LPS-stimulated mice revealed a potent anti-inflammatory activity by suppressing the TNFα serum levels and the expression of TNFα and iNOS in the liver at levels comparable to those of the manuka honey, but they had no effect on IL-6 or IL-1β. It was shown that the LPS-induced suppression of CYP1A1 in the liver was reversed by Epirus and Crete fir honey, while, correspondingly, the suppression of CYP2B10 in the liver was reversed by Evros chestnut and Epirus fir honey. The effect of the same honey samples in polymicrobial peritonitis in mice was also evaluated. Even though no effect was observed on the disease severity or peritoneal bacterial load, the bacterial load in the liver was reduced in mice treated with Evros chestnut, Epiros fir, and Crete fir, while the bacterial load in the lungs was reduced in Epirus arbutus, Crete fir, and manuka honey-treated mice. Conclusion: Our findings suggest that these specific Greek honey samples possess distinct anti-inflammatory and antibacterial properties, as evidenced by the reduced production of pro-inflammatory mediators and the impaired translocation of bacteria to tissues in septic mice. Their mode of action was comparable or more potent to those of manuka honey.
Collapse
|
39
|
Yuvaraj S, Sasikumar S, Puhari SSM, Ramprasath T, Baskaran N, Vasudevan V, Selvam GS. Chrysin reduces hypercholesterolemia-mediated atherosclerosis through modulating oxidative stress, microflora, and apoptosis in experimental rats. J Food Biochem 2022; 46:e14349. [PMID: 35892244 DOI: 10.1111/jfbc.14349] [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: 02/28/2022] [Revised: 07/04/2022] [Accepted: 07/11/2022] [Indexed: 12/29/2022]
Abstract
Chrysin (Chy) is known for various biological proprieties such as inhibitory effects on inflammation, cancer, oxidative stress, aging, and atherosclerosis. However, the hypolipidemic activity of Chy and its mechanistic action remains unclear in cardiovascular diseases (CVD). In this study, we focused on the hypolipidemic proprieties of Chy in hypercholesterolemia-induced atherosclerosis. Male Wistar rats (150-220 g) were divided into four groups as follows: Group I control was fed with standard laboratory chow. Rats in Group II were fed a high-fat diet (HFD) for 60 days. After 60 days of HFD, Group III rats received Chy (100 mg/kg body weight); Group IV rats received Atorvastatin (Atv; 10 mg/kg body weight) for 30 days. Biochemical studies showed Chy, Atv treatment decreased the activities of liver marker enzymes and the levels of Reactive Oxygen Species (ROS) and lipid profile. Gene expression analysis on nuclear factor erythroid 2-related factor 2 (Nrf2) and its regulated genes were significantly reduced in the intestine and increased in the aorta by Chy and Atv. Gut microbial species such as Bacteroidetes, Lactobacillus, Enterococcus, and Clostridium leptum copy numbers were significantly increased by Chy and Atv treatment. In addition, Chy and Atv modulated the expression of inflammatory genes including TLR4, TNFα, NLRP3, and IL-17 in the aorta and intestine compared with hypercholesterolemic control rats. Chy and Atv effectively increased the caspase-3 mRNA expression in the intestine, but these decreased in the aorta. The present study concludes that by reducing oxidative stress and increasing gut microbial colonization, Chy may provide an effective therapeutic approach for the prevention of hypercholesterolemia-mediated atherosclerosis. PRACTICAL APPLICATIONS: Our study focused on a therapeutic model representing the clinical presentation of atherosclerosis in humans. Statins are commonly used in the treatment of cardiovascular complications, patients with hypercholesterolemia face difficulties in the continuation of statin therapy. The reason for statin discontinuation has been associated with toxicological effects. It is necessary to investigate the potentiality of the natural compound as an alternative medicine to statin with fewer side effects. The main theme of our study is to compare the therapeutic potential of Chy and Atv. Chy is a natural bioflavonoid that could be considered as an alternative medicinal compound to statins and to avoid toxicity problems associated with statins. Chy is a bioflavonoid present in Passiflora caerulea (blue passion flower), Oroxylum indicum (Indian trumpet flower), Pelargonium crispum, propolis, and honey. Consuming Chy-rich foods will reduce hypercholesterolemia-mediated cardiovascular complications. Overall, the present studies provided a key to developing bioactive compounds-based foods for CVD patients.
Collapse
Affiliation(s)
- Subramani Yuvaraj
- Molecular Cardiology Unit, Department of Biochemistry, Centre for Excellence in Genomic Sciences, School of Biological Sciences, Madurai Kamaraj University, Madurai, Tamil Nadu, India
| | - Sundrasen Sasikumar
- Molecular Cardiology Unit, Department of Biochemistry, Centre for Excellence in Genomic Sciences, School of Biological Sciences, Madurai Kamaraj University, Madurai, Tamil Nadu, India
| | - Shanavas Syed Mohamed Puhari
- Molecular Cardiology Unit, Department of Biochemistry, Centre for Excellence in Genomic Sciences, School of Biological Sciences, Madurai Kamaraj University, Madurai, Tamil Nadu, India
| | - Tharmarajan Ramprasath
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, Georgia, USA
| | - Nagarethinam Baskaran
- National Institute of Food Technology, Entrepreneurship and Management - Thanjavur (NIFTEM-T), Ministry of Food Processing Industries, Thanjavur, Tamil Nadu, India
| | - Varadaraj Vasudevan
- Molecular Cardiology Unit, Department of Biochemistry, Centre for Excellence in Genomic Sciences, School of Biological Sciences, Madurai Kamaraj University, Madurai, Tamil Nadu, India
| | - Govindan Sadasivam Selvam
- Molecular Cardiology Unit, Department of Biochemistry, Centre for Excellence in Genomic Sciences, School of Biological Sciences, Madurai Kamaraj University, Madurai, Tamil Nadu, India
| |
Collapse
|
40
|
Pham VH, Abbas W, Huang J, Guo F, Zhang K, Kong L, Zhen W, Guo Y, Wang Z. Dietary coated essential oil and organic acid mixture supplementation improves health of broilers infected with avian pathogenic Escherichia coli. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2022; 12:245-262. [PMID: 36712401 PMCID: PMC9868345 DOI: 10.1016/j.aninu.2022.09.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 09/26/2022] [Accepted: 09/28/2022] [Indexed: 01/19/2023]
Abstract
Colibacillosis caused by avian pathogenic Escherichia coli (APEC) is a very prevalent disease in poultry farms in China. The exploration of effective non-antibiotic substances is of great significance for the control of APEC infections. This experiment evaluated the efficacy of coated essential oil and organic acid (EOA) supplementation to prevent E. coli O78 infection in broiler chickens. A total of 288 one-day-old male broiler chicks were randomly distributed into 4 groups with 6 replicates per group. Chickens were fed a diet either supplemented with EOA (500 mg/kg feed) or not, and either uninfected or infected with E. coli O78 intratracheally. Results showed that E. coli O78 infection reduced body weight gain, increased mortality and the ratio of feed to gain along with cecal and liver E. coli load, damaged gut mucosa, induced local and systemic inflammation, and altered cecal microbial composition, diversity and function (P < 0.05). Supplemental EOA improved feed conversion efficiency, lowered gross lesion scores and cecal E. coli population, enhanced intestinal goblet cells and serum IgG concentration, and tended to decrease serum IL-12 production (P < 0.05). Essential oil and organic acid addition downregulated IFN-γ mRNA, tended to decrease mucin-2 mRNA levels while upregulating IL-10 mRNA, and tended to increase ZO-1 gene expression in the jejuna of infected birds at 7 d after E. coli O78 challenge (P < 0.05). The 16S rRNA gene sequencing indicated that both EOA addition and E. coli O78 challenge altered the diversity and composition of the cecal microbiota community. Furthermore, infected birds fed EOA showed decreased Bacteroidetes and genus Lactobacillus abundance compared with the infected control. LEfSe analysis showed that Firmicutes, Ruminococcaceae, Clostridiales, Clostridia, Lactobacillus, Lactobacilaceae, and cc-115 were enriched in the non-infected but EOA-treated group (P < 0.05). Collectively, dietary EOA supplementation could mildly alleviate E. coli-induced gut injury and inflammation.
Collapse
Affiliation(s)
- Van Hieu Pham
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China,Faculty of Animal Science and Veterinary Medicine, Thai Nguyen University Agriculture and Forestry, Thai Nguyen, Viet Nam
| | - Waseem Abbas
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Jinyu Huang
- Menon Animal Nutrition Technology Co. Ltd., Shanghai, 201807, China
| | - Fangshen Guo
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Kaichen Zhang
- Tengzhou Heyi Food Co. Ltd., Zaozhuang, 277000, China
| | - Linhua Kong
- Tengzhou Heyi Food Co. Ltd., Zaozhuang, 277000, China
| | - Wenrui Zhen
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Yuming Guo
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Zhong Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China,Corresponding author.
| |
Collapse
|
41
|
Volovat SR, Augustin I, Zob D, Boboc D, Amurariti F, Volovat C, Stefanescu C, Stolniceanu CR, Ciocoiu M, Dumitras EA, Danciu M, Apostol DGC, Drug V, Shurbaji SA, Coca LG, Leon F, Iftene A, Herghelegiu PC. Use of Personalized Biomarkers in Metastatic Colorectal Cancer and the Impact of AI. Cancers (Basel) 2022; 14:4834. [PMID: 36230757 PMCID: PMC9562853 DOI: 10.3390/cancers14194834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 09/18/2022] [Accepted: 09/29/2022] [Indexed: 12/09/2022] Open
Abstract
Colorectal cancer is a major cause of cancer-related death worldwide and is correlated with genetic and epigenetic alterations in the colonic epithelium. Genetic changes play a major role in the pathophysiology of colorectal cancer through the development of gene mutations, but recent research has shown an important role for epigenetic alterations. In this review, we try to describe the current knowledge about epigenetic alterations, including DNA methylation and histone modifications, as well as the role of non-coding RNAs as epigenetic regulators and the prognostic and predictive biomarkers in metastatic colorectal disease that can allow increases in the effectiveness of treatments. Additionally, the intestinal microbiota's composition can be an important biomarker for the response to strategies based on the immunotherapy of CRC. The identification of biomarkers in mCRC can be enhanced by developing artificial intelligence programs. We present the actual models that implement AI technology as a bridge connecting ncRNAs with tumors and conducted some experiments to improve the quality of the model used as well as the speed of the model that provides answers to users. In order to carry out this task, we implemented six algorithms: the naive Bayes classifier, the random forest classifier, the decision tree classifier, gradient boosted trees, logistic regression and SVM.
Collapse
Affiliation(s)
- Simona-Ruxandra Volovat
- Department of Medical Oncology-Radiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Str., 700115 Iasi, Romania
| | - Iolanda Augustin
- Department of Medical Oncology, AI.Trestioreanu Institute of Oncology, 022328 Bucharest, Romania
| | - Daniela Zob
- Department of Medical Oncology, AI.Trestioreanu Institute of Oncology, 022328 Bucharest, Romania
| | - Diana Boboc
- Department of Medical Oncology-Radiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Str., 700115 Iasi, Romania
| | - Florin Amurariti
- Department of Medical Oncology-Radiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Str., 700115 Iasi, Romania
| | - Constantin Volovat
- Department of Medical Oncology, “Euroclinic” Center of Oncology, 2 Vasile Conta Str., 700106 Iasi, Romania
| | - Cipriana Stefanescu
- Department of Biophysics and Medical Physics-Nuclear Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Str., 700115 Iasi, Romania
| | - Cati Raluca Stolniceanu
- Department of Biophysics and Medical Physics-Nuclear Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Str., 700115 Iasi, Romania
| | - Manuela Ciocoiu
- Department of Pathophysiology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Eduard Alexandru Dumitras
- Department of Pathophysiology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- Department of Anesthesiology and Intensive Care, Regional Institute of Oncology, 700115 Iasi, Romania
| | - Mihai Danciu
- Pathology Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | | | - Vasile Drug
- Department of Gastroenterology, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Str., 700115 Iasi, Romania
- Gastroenterology Clinic, Institute of Gastroenterology and Hepatology, ‘St. Spiridon’ Clinical Hospital, 700115 Iasi, Romania
| | - Sinziana Al Shurbaji
- Gastroenterology Clinic, Institute of Gastroenterology and Hepatology, ‘St. Spiridon’ Clinical Hospital, 700115 Iasi, Romania
| | - Lucia-Georgiana Coca
- Faculty of Computer Science, Alexandru Ioan Cuza University, 700115 Iasi, Romania
| | - Florin Leon
- Faculty of Automatic Control and Computer Engineering, Gheorghe Asachi Technical University, 700115 Iasi, Romania
| | - Adrian Iftene
- Faculty of Computer Science, Alexandru Ioan Cuza University, 700115 Iasi, Romania
| | - Paul-Corneliu Herghelegiu
- Faculty of Automatic Control and Computer Engineering, Gheorghe Asachi Technical University, 700115 Iasi, Romania
| |
Collapse
|
42
|
Miguéns-Gómez A, Sierra-Cruz M, Pérez-Vendrell AM, Rodríguez-Gallego E, Beltrán-Debón R, Terra X, Ardévol A, Pinent M. Differential effects of a cafeteria diet and GSPE preventive treatments on the enterohormone secretions of aged vs. young female rats. Food Funct 2022; 13:10491-10500. [PMID: 36148543 DOI: 10.1039/d2fo02111k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Grape seed derived procyanidins (GSPE) have been shown to effectively prevent intestinal disarrangements induced by a cafeteria diet in young rats. However, little is known about the effects of procyanidins and cafeteria diet on enterohormone secretion in aged rats, as the ageing processes modify these effects. To study these effects in aged rats, we subjected 21-month-old and young 2-month-old female rats to two sub-chronic preventive GSPE treatments. After three months of cafeteria diet administration, we analysed the basal and stimulated secretion and mRNA expression of CCK, PYY and GLP-1, caecal SCFA and intestinal sizes. We found that the effects of a cafeteria diet on the basal duodenal CCK secretion are age dependent. GLP-1 in the ileum was not modified regardless of the rat's age, and GSPE preventive effects differed in the two age groups. GSPE pre-treatment reduced GLP-1, PYY and ChgA in mRNA in aged ileum tissue, while the cafeteria diet increased these in aged colon. The GSPE treatments only modified low-abundance SCFAs. The cafeteria diet in aged rats increases the caecum size differently from that in young rats and GSPE pre-treatment prevents this increase. Therefore, ageing modifies nutrient sensing, and the cafeteria diet acts mainly on the duodenum and colon, while procyanidins have a larger effect on the ileum.
Collapse
Affiliation(s)
- Alba Miguéns-Gómez
- MoBioFood Research Group, Universitat Rovira i Virgili, Departament de Bioquímica i Biotecnologia, c/Marcel·lí Domingo n°1, 43007 Tarragona, Spain.
| | - Marta Sierra-Cruz
- MoBioFood Research Group, Universitat Rovira i Virgili, Departament de Bioquímica i Biotecnologia, c/Marcel·lí Domingo n°1, 43007 Tarragona, Spain.
| | - Anna Maria Pérez-Vendrell
- Monogastric Nutrition, Centre Mas de Bover, IRTA, Ctra. Reus-El Morell Km 3.8, 43120 Constantí, Spain
| | - Esther Rodríguez-Gallego
- MoBioFood Research Group, Universitat Rovira i Virgili, Departament de Bioquímica i Biotecnologia, c/Marcel·lí Domingo n°1, 43007 Tarragona, Spain.
| | - Raúl Beltrán-Debón
- MoBioFood Research Group, Universitat Rovira i Virgili, Departament de Bioquímica i Biotecnologia, c/Marcel·lí Domingo n°1, 43007 Tarragona, Spain.
| | - Ximena Terra
- MoBioFood Research Group, Universitat Rovira i Virgili, Departament de Bioquímica i Biotecnologia, c/Marcel·lí Domingo n°1, 43007 Tarragona, Spain.
| | - Anna Ardévol
- MoBioFood Research Group, Universitat Rovira i Virgili, Departament de Bioquímica i Biotecnologia, c/Marcel·lí Domingo n°1, 43007 Tarragona, Spain.
| | - Montserrat Pinent
- MoBioFood Research Group, Universitat Rovira i Virgili, Departament de Bioquímica i Biotecnologia, c/Marcel·lí Domingo n°1, 43007 Tarragona, Spain.
| |
Collapse
|
43
|
Aspalathus linearis (Rooibos) and Agmatine May Act Synergistically to Beneficially Modulate Intestinal Tight Junction Integrity and Inflammatory Profile. Pharmaceuticals (Basel) 2022; 15:ph15091097. [PMID: 36145318 PMCID: PMC9501288 DOI: 10.3390/ph15091097] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/17/2022] [Accepted: 08/28/2022] [Indexed: 11/25/2022] Open
Abstract
In order to promote gastrointestinal health, significant increases in the prevalence of gastrointestinal disorders should be paralleled by similar surges in therapeutics research. Nutraceutical interventions may play a significant role in patient management. The current study aimed to determine the potential of Aspalathus linearis (rooibos) to prevent gastrointestinal dysregulation resulting from high-dose trace-amine (TA) exposure. Considering the substantial female bias in functional gastrointestinal disorders, and the suggested phytoestrogenicity of rooibos, the study design allowed for a comparison between the effects of an ethanol extract of green rooibos and 17β-estradiol (E2). High levels of ρ-tyramine (TYR) and agmatine (AGM), but not β-phenethylamine (PEA) or tryptamine (TRP), resulted in prostaglandin E2 (PGE2) hypersecretion, increased tight-junction protein (TJP; occludin and ZO-1) secretion and (dissimilarly) disrupted the TJP cellular distribution profile. Modulating benefits of rooibos and E2 were TA-specific. Rooibos pre-treatment generally reduced IL-8 secretion across all TA conditions and prevented PGE2 hypersecretion after exposure to both TYR and AGM, but was only able to normalise TJP levels and the distribution profile in AGM-exposed cells. In contrast, E2 pre-treatment prevented only TYR-associated PGE2 hypersecretion and TJP dysregulation. Together, the data suggest that the antioxidant and anti-inflammatory effects of rooibos, rather than phytoestrogenicity, affect benefits illustrated for rooibos.
Collapse
|
44
|
Zhu Y, Wei YL, Karras I, Cai PJ, Xiao YH, Jia CL, Qian XL, Zhu SY, Zheng LJ, Hu X, Sun AD. Modulation of the gut microbiota and lipidomic profiles by black chokeberry ( Aronia melanocarpa L.) polyphenols via the glycerophospholipid metabolism signaling pathway. Front Nutr 2022; 9:913729. [PMID: 35990329 PMCID: PMC9387202 DOI: 10.3389/fnut.2022.913729] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 06/17/2022] [Indexed: 11/13/2022] Open
Abstract
Black chokeberry (Aronia melanocarpa L.) is rich in polyphenols with various physiological and pharmacological activities. However, the relationship between the modulation effect of black chokeberry polyphenols on obesity and the alteration of lipid metabolism is not clearly understood. This study aimed to investigate the beneficial effects of the black chokeberry polyphenols (BCPs) treatment on the structure of gut microbiota, lipid metabolism, and associated mechanisms in high-fat diet (HFD)-induced obese rats. Here, we found that a high-fat diet promoted body weight gain and lipid accumulation in rats, while oral BCPs supplementation reduced body weight, liver, and white adipose tissue weight and alleviated dyslipidemia and hepatic steatosis in HFD-induced obese rats. In addition, BCPs supplementation prevented gut microbiota dysbiosis by increasing the relative abundance of Bacteroides, Prevotella, Romboutsia, and Akkermansia and decreasing the relative abundance of Desulfovibrio and Clostridium. Furthermore, 64 lipids were identified as potential lipid biomarkers through lipidomics analysis after BCPs supplementation, especially PE (16:0/22:6), PE (18:0/22:6), PC (20:3/19:0), LysoPE (24:0), LysoPE (24:1), and LysoPC (20:0). Moreover, our studies provided new evidence that composition of gut microbiota was closely related to the alteration of lipid profiles after BCPs supplementation. Additionally, BCPs treatment could ameliorate the disorder of lipid metabolism by regulating the mRNA and protein expression of genes related to the glycerophospholipid metabolism signaling pathway in HFD-induced obese rats. The mRNA and protein expression of PPARα, CPT1α, EPT1, and LCAT were significantly altered after BCPs treatment. In conclusion, the results of this study indicated that BCPs treatment alleviated HFD-induced obesity by modulating the composition and function of gut microbiota and improving the lipid metabolism disorder via the glycerophospholipid metabolism signaling pathway.
Collapse
Affiliation(s)
- Yue Zhu
- College of Biological Sciences and Technology, Beijing Forestry University, Beijing, China.,Beijing Key Laboratory of Forest Food Processing and Safety, Beijing Forestry University, Beijing, China
| | - Yu-Long Wei
- College of Biological Sciences and Technology, Beijing Forestry University, Beijing, China.,Beijing Key Laboratory of Forest Food Processing and Safety, Beijing Forestry University, Beijing, China
| | - Ioanna Karras
- College of Agricultural, Consumer and Environmental Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Peng-Ju Cai
- College of Biological Sciences and Technology, Beijing Forestry University, Beijing, China.,Beijing Key Laboratory of Forest Food Processing and Safety, Beijing Forestry University, Beijing, China
| | - Yu-Hang Xiao
- College of Biological Sciences and Technology, Beijing Forestry University, Beijing, China
| | - Cheng-Li Jia
- Beijing Key Laboratory of Forest Food Processing and Safety, Beijing Forestry University, Beijing, China
| | - Xiao-Lin Qian
- College of Biological Sciences and Technology, Beijing Forestry University, Beijing, China.,Beijing Key Laboratory of Forest Food Processing and Safety, Beijing Forestry University, Beijing, China
| | - Shi-Yu Zhu
- College of Biological Sciences and Technology, Beijing Forestry University, Beijing, China.,Beijing Key Laboratory of Forest Food Processing and Safety, Beijing Forestry University, Beijing, China
| | - Lu-Jie Zheng
- College of Biological Sciences and Technology, Beijing Forestry University, Beijing, China.,Beijing Key Laboratory of Forest Food Processing and Safety, Beijing Forestry University, Beijing, China
| | - Xin Hu
- College of Biological Sciences and Technology, Beijing Forestry University, Beijing, China.,Beijing Key Laboratory of Forest Food Processing and Safety, Beijing Forestry University, Beijing, China
| | - Ai-Dong Sun
- College of Biological Sciences and Technology, Beijing Forestry University, Beijing, China.,Beijing Key Laboratory of Forest Food Processing and Safety, Beijing Forestry University, Beijing, China
| |
Collapse
|
45
|
Sharma P, Silva C, Pfreundschuh S, Ye H, Sampath H. Metabolic protection by the dietary flavonoid 7,8-dihydroxyflavone requires an intact gut microbiome. Front Nutr 2022; 9:987956. [PMID: 36061902 PMCID: PMC9428675 DOI: 10.3389/fnut.2022.987956] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 07/29/2022] [Indexed: 11/18/2022] Open
Abstract
Background 7,8-dihydroxyflavone (DHF) is a naturally occurring flavonoid found in Godmania, Tridax, and Primula species that confers protection against high-fat diet (HFD) induced metabolic pathologies selectively in female mice. We have previously reported that this metabolic protection is associated with early and stable remodeling of the intestinal microbiome, evident in female but not male DHF-supplemented mice. Early changes in the gut microbiome in female DHF-fed mice were highly predictive of subsequent metabolic protection, suggesting a causative association between the gut microbiome and the metabolic effects of DHF. Objective To investigate a causal association between the gut microbiome and the metabolic effects of DHF using a model of antibiotic-induced gut microbiome ablation. Materials and methods Age-matched male and female C57Bl6/J mice were given ad libitum access to HFD and drinking water containing vehicle or DHF for 12 weeks. For antibiotic (Abx) treatment, female mice were given drinking water containing a cocktail of antibiotics for 2 weeks prior to HFD feeding and throughout the feeding period. Metabolic phenotyping consisted of longitudinal assessments of body weights, body composition, food, and water intake, as well as measurement of energy expenditure, glucose tolerance, and plasma and hepatic lipids. Protein markers mediating the cellular effects of DHF were assessed in brown adipose tissue (BAT) and skeletal muscle. Results Metabolic protection conferred by DHF in female HFD-fed mice was only apparent in the presence of an intact gut microbiome. Abx-treated mice were not protected from HFD-induced obesity by DHF administration. Further, tissue activation of the tropomyosin-related kinase receptor B (TrkB) receptor, which has been attributed to the biological activity of DHF, was lost upon gut microbiome ablation, indicating a requirement for microbial “activation” of DHF for its systemic effects. In addition, we report for the first time that DHF supplementation significantly activates TrkB in BAT of female, but not male, mice uncovering a novel target tissue of DHF. DHF supplementation also increased uncoupling protein 1 (UCP1) and AMP-activated protein kinase (AMPK) protein in BAT, consistent with protection from diet-induced obesity. Conclusion These results establish for the first time a requirement for the gut microbiome in mediating the metabolic effects of DHF in female mice and uncover a novel target tissue that may mediate these sexually-dimorphic protective effects.
Collapse
Affiliation(s)
- Priyanka Sharma
- Department of Nutritional Sciences, Rutgers University, New Brunswick, NJ, United States
- Rutgers Center for Lipid Research, New Jersey Institute for Food, Nutrition, and Health, Rutgers University, New Brunswick, NJ, United States
- Center for Microbiome, Nutrition, and Health, New Jersey Institute for Food, Nutrition, and Health, Rutgers University, New Brunswick, NJ, United States
| | - Camila Silva
- Department of Biotechnology, Rutgers University, New Brunswick, NJ, United States
| | - Sarah Pfreundschuh
- Department of Nutritional Sciences, Rutgers University, New Brunswick, NJ, United States
| | - Hong Ye
- Department of Nutritional Sciences, Rutgers University, New Brunswick, NJ, United States
| | - Harini Sampath
- Department of Nutritional Sciences, Rutgers University, New Brunswick, NJ, United States
- Rutgers Center for Lipid Research, New Jersey Institute for Food, Nutrition, and Health, Rutgers University, New Brunswick, NJ, United States
- Center for Microbiome, Nutrition, and Health, New Jersey Institute for Food, Nutrition, and Health, Rutgers University, New Brunswick, NJ, United States
- *Correspondence: Harini Sampath,
| |
Collapse
|
46
|
Cheng Y, Tang S, Wu T, Pan S, Xu X. Lactobacillus casei-fermented blueberry pomace ameliorates colonic barrier function in high fat diet mice through MAPK-NF-κB-MLCK signaling pathway. J Funct Foods 2022. [DOI: 10.1016/j.jff.2022.105139] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
47
|
Xu F, Zhu Y, Lu M, Qin L, Zhao D, Ren T. Effects of Hydroxy-Alpha-Sanshool on Intestinal Metabolism in Insulin-Resistant Mice. Foods 2022; 11:foods11142040. [PMID: 35885283 PMCID: PMC9322383 DOI: 10.3390/foods11142040] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 07/07/2022] [Accepted: 07/08/2022] [Indexed: 11/16/2022] Open
Abstract
To explore the hydroxy-alpha-sanshool (HAS) effects on the intestinal metabolites of insulin-resistant mice, the blank group (BG), model group (MG), and HAS dose group (DG) were designed. The insulin resistance (IR) model was induced through streptozotocin (STZ) combined with a high-fat and high-sugar diet. Based on the availability of the model, the HAS dose was given by gavage for 28 days. The determination of cecum and key serum indexes was made, including the contents of insulin (INS), triglycerides (TG), total cholesterol (TC), glycosylated serum protein (GSP), and glycosylated hemoglobin (GHb). The changes in gut microbiota and metabolites in cecal contents were detected by 16S rRNA gene amplicon sequencing and UPLC/HRMS technology, respectively. The results that the levels of GSP, GHb, TG, and TC were significantly increased; this was not the case for INS; or for the changes in the gut microbiota and metabolites in MG. However, the intervention of HAS effectively reversed these changes, for instance, it decreased levels of GSP, GHb, TG, TC, and alterations of metabolite composition for linoleic acid and tyrosine metabolism and recovered trends of declining species diversity and richness of the gut microbiota in MG. It was indicated that HAS alleviated IR by regulating the gut microbiota and metabolites and affecting lipid and amino acid metabolism pathways.
Collapse
Affiliation(s)
- Fangyan Xu
- College of Brewing and Food Engineering, Guizhou University, Guiyang 550025, China; (F.X.); (M.L.); (L.Q.); (D.Z.)
| | - Yuping Zhu
- School of Basic Medicine, Guizhou Medical University, Guiyang 550025, China;
| | - Mintao Lu
- College of Brewing and Food Engineering, Guizhou University, Guiyang 550025, China; (F.X.); (M.L.); (L.Q.); (D.Z.)
| | - Likang Qin
- College of Brewing and Food Engineering, Guizhou University, Guiyang 550025, China; (F.X.); (M.L.); (L.Q.); (D.Z.)
| | - Degang Zhao
- College of Brewing and Food Engineering, Guizhou University, Guiyang 550025, China; (F.X.); (M.L.); (L.Q.); (D.Z.)
- Guiyang Station for DUS Testing Center of New Plant Varieties of the Ministry of Agriculture and Rural Affairs of the People’s Republic of China in Guizhou Academy of Agricultural Sciences, Guiyang 550006, China
| | - Tingyuan Ren
- College of Brewing and Food Engineering, Guizhou University, Guiyang 550025, China; (F.X.); (M.L.); (L.Q.); (D.Z.)
- Guiyang Station for DUS Testing Center of New Plant Varieties of the Ministry of Agriculture and Rural Affairs of the People’s Republic of China in Guizhou Academy of Agricultural Sciences, Guiyang 550006, China
- Correspondence:
| |
Collapse
|
48
|
Pharmacological Effects of Polyphenol Phytochemicals on the Intestinal Inflammation via Targeting TLR4/NF-κB Signaling Pathway. Int J Mol Sci 2022; 23:ijms23136939. [PMID: 35805952 PMCID: PMC9266441 DOI: 10.3390/ijms23136939] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/20/2022] [Accepted: 06/21/2022] [Indexed: 02/05/2023] Open
Abstract
TLR4/NF-κB is a key inflammatory signaling transduction pathway, closely involved in cell differentiation, proliferation, apoptosis, and pro-inflammatory response. Toll like receptor 4 (TLR4), the first mammalian TLR to be characterized, is the innate immune receptor that plays a key role in inflammatory signal transductions. Nuclear factor kappa B (NF-κB), the TLR4 downstream, is the key to accounting for the expression of multiple genes involved in inflammatory responses, such as pro-inflammatory cytokines. Inflammatory bowel disease (IBD) in humans is a chronic inflammatory disease with high incidence and prevalence worldwide. Targeting the TLR4/NF-κB signaling pathway might be an effective strategy to alleviate intestinal inflammation. Polyphenol phytochemicals have shown noticeable alleviative effects by acting on the TLR4/NF-κB signaling pathway in intestinal inflammation. This review summarizes the pharmacological effects of more than 20 kinds of polyphenols on intestinal inflammation via targeting the TLR4/NF-κB signaling pathway. We expected that polyphenol phytochemicals targeting the TLR4/NF-κB signaling pathway might be an effective approach to treat IBD in future clinical research applications.
Collapse
|
49
|
Yan X, Zhai Y, Zhou W, Qiao Y, Guan L, Liu H, Jiang J, Peng L. Intestinal Flora Mediates Antiobesity Effect of Rutin in High-Fat-Diet Mice. Mol Nutr Food Res 2022; 66:e2100948. [PMID: 35616308 DOI: 10.1002/mnfr.202100948] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 04/12/2022] [Indexed: 11/09/2022]
Abstract
SCOPE Intestinal flora plays a critical role in the development of . Rutin is a natural flavonoid with potential prebiotic effects on regulating the intestinal flora composition that is beneficial for host health. Therefore, this study hypothesizes that rutin supplementation has beneficial effects on high-fat-diet (HFD)-induced obesity and metabolic disorder through the modulation of intestinal flora in mice. METHODS AND RESULTS The obesity-alleviating property of rutin using 6-week-old C57BL/6J male mice fed on HFD with or without rutin supplementation for 16 weeks is investigated. Rutin supplementation effectively reduces body-weight gain, insulin resistance, and acted favorably on the intestinal barrier, thereby reducing endotoxemia and systemic inflammation. Sequencing of 16S rRNA genes from fecal samples indicate that rutin exerted modulatory effects on HFD-induced intestinal flora disorders (e.g., rutin decreased Firmicutes abundance and increased Bacteroidetes and Verrucomicrobia abundance). Antibiotic treatment and fecal microbiota transplantation further demonstrate that the salutary effects of rutin on obesity control are strongly dependent on the intestinal flora. CONCLUSION Rutin can be considered as a prebiotic agent for improving intestinal flora disorders and obesity-associated metabolic perturbations in obese individuals.
Collapse
Affiliation(s)
- Xu Yan
- College of Life Sciences, Hebei University, Baoding, Hebei, 071002, China.,Beijing Key Laboratory for Immune-Mediated Inflammatory Diseases, Institute of Medical Science, China-Japan Friendship Hospital, No. 2 Yinghua East Street, Chaoyang District, Beijing, 100029, China
| | - Yuanyuan Zhai
- College of Life Sciences, Hebei University, Baoding, Hebei, 071002, China.,Beijing Key Laboratory for Immune-Mediated Inflammatory Diseases, Institute of Medical Science, China-Japan Friendship Hospital, No. 2 Yinghua East Street, Chaoyang District, Beijing, 100029, China
| | - Wenling Zhou
- College of Life Sciences, Hebei University, Baoding, Hebei, 071002, China.,Beijing Key Laboratory for Immune-Mediated Inflammatory Diseases, Institute of Medical Science, China-Japan Friendship Hospital, No. 2 Yinghua East Street, Chaoyang District, Beijing, 100029, China
| | - Yuan Qiao
- Beijing Key Laboratory for Immune-Mediated Inflammatory Diseases, Institute of Medical Science, China-Japan Friendship Hospital, No. 2 Yinghua East Street, Chaoyang District, Beijing, 100029, China
| | - Lingling Guan
- Beijing Key Laboratory for Immune-Mediated Inflammatory Diseases, Institute of Medical Science, China-Japan Friendship Hospital, No. 2 Yinghua East Street, Chaoyang District, Beijing, 100029, China
| | - Hao Liu
- Beijing Key Laboratory for Immune-Mediated Inflammatory Diseases, Institute of Medical Science, China-Japan Friendship Hospital, No. 2 Yinghua East Street, Chaoyang District, Beijing, 100029, China
| | - Jizhi Jiang
- College of Life Sciences, Hebei University, Baoding, Hebei, 071002, China
| | - Liang Peng
- Beijing Key Laboratory for Immune-Mediated Inflammatory Diseases, Institute of Medical Science, China-Japan Friendship Hospital, No. 2 Yinghua East Street, Chaoyang District, Beijing, 100029, China
| |
Collapse
|
50
|
Sun L, Guo L, Xu G, Li Z, Appiah MO, Yang L, Lu W. Quercetin Reduces Inflammation and Protects Gut Microbiota in Broilers. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27103269. [PMID: 35630745 PMCID: PMC9147699 DOI: 10.3390/molecules27103269] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/18/2022] [Accepted: 05/18/2022] [Indexed: 12/12/2022]
Abstract
The aim of this study was to investigate the effects of quercetin on inflammatory response and intestinal microflora in broiler chicken jejuna. A total of 120 broiler chickens were allocated into 3 groups: saline-challenged broilers fed a basal diet (CTR group), lipopolysaccharide (LPS)-challenged broilers fed a basal diet (L group) and LPS-challenged broilers fed a basal diet supplemented with 200 mg/kg quercetin (LQ group). Our results showed that LPS significantly increased expression of tumor necrosis factor (TNF)-α, interleukin (IL)-1β, IL-6, IL-8, interferon (IFN)-γ, toll-like receptor (TLR)-4, Bax, Caspase-3 and diamine oxidase activity (DAO), and decreased expression of zona occludens-1 (ZO-1), Occludin and Bcl-2 in the jejunum, while dietary quercetin prevented the adverse effects of LPS injection. LPS injection significantly decreased the number of Actinobacteria, Armatimonadetes and Fibrobacteriae at the phylum level when compared to the CTR group. Additionally, at genus level, compared with the CTR group, the abundance of Halomonas, Micromonospora, Nitriliruptor, Peptococcus, Rubellimicrobium, Rubrobacter and Slaclda in L group was significantly decreased, while dietary quercetin restored the numbers of these bacteria. In conclusion, our results demonstrated that dietary quercetin could alleviate inflammatory responses of broiler chickens accompanied by modulating jejunum microflora.
Collapse
|