151
|
Liu JD, Lumpkins B, Mathis G, Williams SM, Fowler J. Evaluation of encapsulated sodium butyrate with varying releasing times on growth performance and necrotic enteritis mitigation in broilers. Poult Sci 2019; 98:3240-3245. [PMID: 30789214 DOI: 10.3382/ps/pez049] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 01/25/2019] [Indexed: 12/25/2022] Open
Abstract
This study was conducted to evaluate the effect of an encapsulated sodium butyrate (Na-B) with targeted releasing times on growth performance and mitigating the impact of necrotic enteritis in broilers. Two Na-B (30% of Na-B content) products, CMA (2 h releasing time) and CMP (3 to 4 h releasing time), were evaluated in a necrotic enteritis challenge model. The experiment consisted of 4 Na-B treatments (500 and 1,000 ppm of each product) plus 2 control (non-challenged and challenged). A total of 336 Cobb-Cobb male broilers were placed 8 birds per pen into 7 replicate battery cages. On day 14, birds from challenged treatments were orally gavaged with ∼5,000 oocysts of Eimeria maxima. On day 19, 20, and 21, the challenged birds received 1 mL of 108 cfu/mL Clostridium perfringens. Total pen body weight (BW) and feed weight were assessed on day 14, 21, and 28 for BW gain and mortality-adjusted feed conversion ratio (FCR). On day 21, 3 birds were randomly selected per pen and scored for intestinal lesions. The duodenum and jejunum tissue were collected on day 21 and 28 for quantifying intestinal histology. Results revealed that adding CMA at 500 ppm showed significantly higher cumulative BW gain (P < 0.05) compared to the challenged control from day 1 to 21. Adding CMA at 500 ppm also showed the equivalent cumulative FCR that was comparable to the non-challenged control on day 21. The CMA treatments and CMP at 1,000 ppm treatment showed equivalent BW gain compared to the non-challenge control after an additional 7 d post-challenge on day 28. Both products at 500 or 1,000 ppm had the significantly (P < 0.05) lower intestinal lesion scores compared to the challenged control. However, there was no difference in lesion scores among the Na-B treatments. The Na-B product targeted to release in the anterior intestinal tract shows the beneficial effects on BW gain and feed utilization efficiency in current challenge model. Adding encapsulated Na-B product has the potential to mitigate the impact of necrotic enteritis in broilers.
Collapse
Affiliation(s)
- J D Liu
- Department of Poultry Science, University of Georgia, Athens, GA 30602
| | - B Lumpkins
- Southern Poultry Research, Inc., 96 Roquemore Road, Athens, GA 30607
| | - G Mathis
- Southern Poultry Research, Inc., 96 Roquemore Road, Athens, GA 30607
| | - S M Williams
- Poultry Diagnostic and Research Center, College of Veterinary Medicine, University of Georgia, Athens, GA 30602
| | - J Fowler
- Department of Poultry Science, University of Georgia, Athens, GA 30602
| |
Collapse
|
152
|
Li K, Zhang L, Xue J, Yang X, Dong X, Sha L, Lei H, Zhang X, Zhu L, Wang Z, Li X, Wang H, Liu P, Dong Y, He L. Dietary inulin alleviates diverse stages of type 2 diabetes mellitus via anti-inflammation and modulating gut microbiota in db/db mice. Food Funct 2019; 10:1915-1927. [PMID: 30869673 DOI: 10.1039/c8fo02265h] [Citation(s) in RCA: 145] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Type 2 diabetes mellitus (T2DM) is closely correlated with chronic low-grade inflammation and gut dysbiosis. Prebiotic inulin (INU) is conducive to modulate gut dysbiosis. However, the impact of dietary inulin on the diverse stages of T2DM remains largely unknown. In the present study, according to the fasting blood glucose (FBG) and oral glucose tolerance tests (OGTT), mice were randomly divided into six groups (15 mice per group): pre-diabetic group (PDM group); inulin-treated pre-diabetic group (INU/PDM group); early diabetic group (EDM group); inulin-treated early diabetic group (INU/EDM group); diabetic group (DM group); inulin-treated diabetic group (INU/DM group). All animal experiments were approved by the Ethics Committee of the General Hospital of Ningxia Medical University (No. 2016-232). After 6 weeks of inulin intervention, the mice were euthanized and the associated indicators were investigated. Dietary inulin significantly reduced FBG, body weights (BWs), glycated hemoglobin (GHb), blood lipid, plasma lipopolysaccharide (LPS), interleukin (IL)-6, tumor necrosis factor (TNF)-α and IL-17A in the three inulin-treated groups compared to the untreated groups. But for IL-17A, there remained no significant difference between the PDM group and the INU/PDM group. Moreover, the anti-inflammatory IL-10 showed significant alteration in the INU/PDM and INU/EDM groups, but no significant alteration in the INU/DM group. Sequencing analysis of the gut microbiota showed an elevation in the relative abundance of Cyanobacteria and Bacteroides and a reduction in the relative abundance of Ruminiclostridium_6 in three inulin-treated different stages of T2DM groups, as well as a reduction in the relative abundance of Deferribacteres and Tenericutes in the INU/DM group. A reduction in the relative abundance of Mucispirillum was detected in the INU/PDM and INU/EDM groups. Correlation analysis revealed that Cyanobacteria and Bacteroides abundance were positively correlated with IL-10; Deferribacteres, Tenericutes, Mucispirillum and Ruminiclostridium_6 abundance were closely related to IL-6, TNF-α or IL-17A respectively. Additionally, Mucispirillum and Ruminiclostridium_6 abundance were positively correlated with LPS. Taken together, dietary inulin alleviated the diverse stages of T2DM via suppressing inflammation and modulating gut microbiota.
Collapse
Affiliation(s)
- Ke Li
- Clinical Medical College, Ningxia Medical University, Yinchuan 750004, Ningxia, China.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
153
|
Sakwinska O, Bosco N. Host Microbe Interactions in the Lactating Mammary Gland. Front Microbiol 2019; 10:1863. [PMID: 31456777 PMCID: PMC6701204 DOI: 10.3389/fmicb.2019.01863] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 07/29/2019] [Indexed: 12/13/2022] Open
Abstract
The bacteria present in human milk constitute the human milk microbiome (hMM). Both the older culture-based work and the more recent studies using molecular detection of bacterial DNA have reached similar conclusions: the hMM mostly consists of commensal staphylococci such as Staphylococcus epidermidis, and streptococci. The prevalence of other bacterial groups such lactobacilli varies widely, while the abundance and prevalence of bifidobacteria is generally low. Recently, the hMM became accepted as a part of a physiologically normal state with suggested potential health benefits. Most research on the hMM has focused on its composition and potential effect on the breastfed infant. A major role as a microbiome inoculum for the infant gut has been proposed, but remains to be clearly demonstrated. Herein, we also discuss the emerging connection between the hMM and mammary gland physiology and lactation. Similarities between the mammary gland and mucosal interfaces are considerable, and in particular mucosal-like immune attributes of mammary gland. The potential role of hMM-host interactions in the mammary gland in maternal health is explored with a primary focus on lactational mastitis.
Collapse
Affiliation(s)
- Olga Sakwinska
- Nestlé Research, Nestlé Institute of Health Sciences, Lausanne, Switzerland
| | - Nabil Bosco
- Nestlé Research, Nestlé Institute of Health Sciences, Lausanne, Switzerland
- Nestlé Research Singapore Hub, Singapore, Singapore
| |
Collapse
|
154
|
Iacob S, Iacob DG. Infectious Threats, the Intestinal Barrier, and Its Trojan Horse: Dysbiosis. Front Microbiol 2019; 10:1676. [PMID: 31447793 PMCID: PMC6692454 DOI: 10.3389/fmicb.2019.01676] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 07/08/2019] [Indexed: 02/06/2023] Open
Abstract
The ecosystem of the gut microbiota consists of diverse intestinal species with multiple metabolic and immunologic activities and it is closely connected with the intestinal epithelia and mucosal immune response, with which it builds a complex barrier against intestinal pathogenic bacteria. The microbiota ensures the integrity of the gut barrier through multiple mechanisms, either by releasing antibacterial molecules (bacteriocins) and anti-inflammatory short-chain fatty acids or by activating essential cell receptors for the immune response. Experimental studies have confirmed the role of the intestinal microbiota in the epigenetic modulation of the gut barrier through posttranslational histone modifications and regulatory mechanisms induced by epithelial miRNA in the epithelial lumen. Any quantitative or functional changes of the intestinal microbiota, referred to as dysbiosis, alter the immune response, decrease epithelial permeability and destabilize intestinal homeostasis. Consequently, the overgrowth of pathobionts (Staphylococcus, Pseudomonas, and Escherichia coli) favors intestinal translocations with Gram negative bacteria or their endotoxins and could trigger sepsis, septic shock, secondary peritonitis, or various intestinal infections. Intestinal infections also induce epithelial lesions and perpetuate the risk of bacterial translocation and dysbiosis through epithelial ischemia and pro-inflammatory cytokines. Furthermore, the decline of protective anaerobic bacteria (Bifidobacterium and Lactobacillus) and inadequate release of immune modulators (such as butyrate) affects the release of antimicrobial peptides, de-represses microbial virulence factors and alters the innate immune response. As a result, intestinal germs modulate liver pathology and represent a common etiology of infections in HIV immunosuppressed patients. Antibiotic and antiretroviral treatments also promote intestinal dysbiosis, followed by the selection of resistant germs which could later become a source of infections. The current article addresses the strong correlations between the intestinal barrier and the microbiota and discusses the role of dysbiosis in destabilizing the intestinal barrier and promoting infectious diseases.
Collapse
Affiliation(s)
- Simona Iacob
- Infectious Diseases Department, "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania.,National Institute of Infectious Diseases "Prof. Dr. Matei Balş", Bucharest, Romania
| | - Diana Gabriela Iacob
- Infectious Diseases Department, "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania
| |
Collapse
|
155
|
Collado MC, Vinderola G, Salminen S. Postbiotics: facts and open questions. A position paper on the need for a consensus definition. Benef Microbes 2019; 10:711-719. [PMID: 31965850 DOI: 10.3920/bm2019.0015] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The past definitions of probiotics and prebiotics have been reviewed and updated recently. According to these concepts, probiotics comprise live microorganisms that confer a health benefit on the host when administered in adequate amounts, whereas a prebiotic is a substrate that is selectively utilised by host microorganisms, conferring a health benefit. The words probiotics and prebiotics can be found on labels of many foods and supplements. Consumers have a growing awareness of these terms' meanings, and many countries are increasingly using them for regulation purposes. At the same time, there is increasing evidence on the health effects of non-viable microorganisms and the metabolites that they can produce by fermentation or by their action on food components. Different terms have been used in the literature to refer to these bioactive compounds, which do not fall under the known categories of probiotics, prebiotics or synbiotics. The tentative term postbiotics has been the most used one so far. However, no definition of the term has gained international consensus to date. This work aims to provide information on the facts and the open questions about the so-called postbiotics.
Collapse
Affiliation(s)
- M C Collado
- Institute of Agrochemistry and Food Technology-National Research Council (IATA-CSIC), Av. Agustin Escardino 7, 46980 Valencia, Spain.,Functional Foods Forum, Faculty of Medicine, University of Turku, Turun Yliopisto, Turku 20014, Finland
| | - G Vinderola
- Instituto de Lactología Industrial (INLAIN, UNL-CONICET), Facultad de Ingeniería Química, Universidad Nacional del Litoral, Santiago del Estero 2829, Santa Fe 3000, Argentina
| | - S Salminen
- Functional Foods Forum, Faculty of Medicine, University of Turku, Turun Yliopisto, Turku 20014, Finland
| |
Collapse
|
156
|
Pan LL, Niu W, Fang X, Liang W, Li H, Chen W, Zhang H, Bhatia M, Sun J. Clostridium butyricum Strains Suppress Experimental Acute Pancreatitis by Maintaining Intestinal Homeostasis. Mol Nutr Food Res 2019; 63:e1801419. [PMID: 31034143 DOI: 10.1002/mnfr.201801419] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 04/13/2019] [Indexed: 01/24/2023]
Abstract
SCOPE Acute pancreatitis (AP) is a common abdominal inflammatory disease. Disturbed gut homeostasis secondary to pancreatic inflammation aggravates the condition retroactively. The current study investigates potential beneficial effects of Clostridium butyricum (C. butyricum) strains on AP and underlying mechanisms. METHODS AND RESULTS C. butyricum strains MIYAIRI 588 (CBM588) and CGMCC0313.1 (CB0313.1) were supplemented to mice for three weeks before experimental AP or SAP induction. Both CBM588 and CB0313.1 protected against AP, as evidenced by reduced serum amylase and lipase levels, pancreatic edema, and myeloperoxidase activity. Amelioration of both experimental AP and SAP by CB0313.1 indicated a non-model-specific effect. Moreover, C. butyricum inhibited pancreatic neutrophil and dendritic cell infiltration, nucleotide-binding domain leucine-rich repeat-containing family, pyrin domain-containing 3 inflammasome activation, and pro-inflammatory pathways. Additionally in the gut, C. butyricum strains attenuated AP-associated intestinal inflammation and barrier dysfunction, accompanied with reduced pathogenic bacteria Escherichia coli and Enterococcus penetration into pancreas. Gut microbiome analyses further revealed that beneficial effects of C. butyricum on pancreatic-gut homeostasis were correlated with improved dysbiosis. In particular, relative abundance of Desulfovibrionaceae decreased, and Verrucomicrobiaceae Clostridiaceae and Lactobacillaceae increased. CONCLUSIONS For the first time, a protective effect of C. butyricum in AP by modulating intestinal homeostasis is demonstrated.
Collapse
Affiliation(s)
- Li-Long Pan
- School of Medicine, Jiangnan University, Wuxi, 214122, P. R. China
| | - Wenying Niu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China
| | - Xin Fang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China
| | - Wenjie Liang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China
| | - Hongli Li
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China
| | - Madhav Bhatia
- Inflammation Research Group, Department of Pathology, University of Otago, Christchurch, 8140, New Zealand
| | - Jia Sun
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China
| |
Collapse
|
157
|
Li W, Zhu Y, Li Y, Shu M, Wen Y, Gao X, Wan C. The gut microbiota of hand, foot and mouth disease patients demonstrates down-regulated butyrate-producing bacteria and up-regulated inflammation-inducing bacteria. Acta Paediatr 2019; 108:1133-1139. [PMID: 30427066 DOI: 10.1111/apa.14644] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 10/08/2018] [Accepted: 11/09/2018] [Indexed: 12/15/2022]
Abstract
AIM This study explored the gut microbiota of children with hand, foot and mouth disease (HFMD). METHODS We enrolled 15 cases with HFMD admitted to the West China Second Hospital, Sichuan University, China, from July to September 2016 at a median age of three years. The controls were 15 healthy children of a similar age who underwent routine health examinations at the hospital during the same period. Gut microbiota was analysed using high throughput 16S ribosomal deoxyribonucleic acid sequencing. RESULTS The gut microbiota in the HFMD patients was distinct from the controls. Compared with the controls, the composition of gut microbiota in the HFMD cases represented a reduction of two butyrate-producing bacteria, Ruminococcus (0.73 ± 1.28 versus 7.78 ± 20.01, p = 0.026) and Roseburia (0.67 ± 1.69 versus 1.61 ± 3.27, p = 0.024) and an up-regulation of Escherichia (5.26 ± 10.50 versus 1.59 ± 5.90,p < 0.01) and Enterococcus (4.12 ± 12.49 versus 0.12 ± 0.41, p = 0.015). CONCLUSION The dysbiosis of gut microbiota of the HFMD cases included a reduction of butyrate-producing bacteria and an up-regulation of inflammation-inducing bacteria. These may have impaired the intestinal biological mucosal barrier and host immune functions, promoting the invasion of the enterovirus.
Collapse
Affiliation(s)
- Weiran Li
- Department of Paediatrics West China Second Hospital Sichuan University Chengdu China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education Chengdu China
| | - Yu Zhu
- Department of Paediatrics West China Second Hospital Sichuan University Chengdu China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education Chengdu China
| | - Yiyuan Li
- Department of Paediatrics West China Second Hospital Sichuan University Chengdu China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education Chengdu China
| | - Min Shu
- Department of Paediatrics West China Second Hospital Sichuan University Chengdu China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education Chengdu China
| | - Yang Wen
- Department of Paediatrics West China Second Hospital Sichuan University Chengdu China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education Chengdu China
| | - Xiaolin Gao
- Department of Paediatrics West China Second Hospital Sichuan University Chengdu China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education Chengdu China
| | - Chaomin Wan
- Department of Paediatrics West China Second Hospital Sichuan University Chengdu China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education Chengdu China
| |
Collapse
|
158
|
|
159
|
Traisaeng S, Herr DR, Kao HJ, Chuang TH, Huang CM. A Derivative of Butyric Acid, the Fermentation Metabolite of Staphylococcus epidermidis, Inhibits the Growth of a Staphylococcus aureus Strain Isolated from Atopic Dermatitis Patients. Toxins (Basel) 2019; 11:toxins11060311. [PMID: 31159213 PMCID: PMC6628397 DOI: 10.3390/toxins11060311] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 05/24/2019] [Accepted: 05/24/2019] [Indexed: 12/20/2022] Open
Abstract
The microbiome is a rich source of metabolites for the development of novel drugs. Butyric acid, for example, is a short-chain fatty acid fermentation metabolite of the skin probiotic bacterium Staphylococcus epidermidis (S. epidermidis). Glycerol fermentation of S. epidermidis resulted in the production of butyric acid and effectively hindered the growth of a Staphylococcus aureus (S. aureus) strain isolated from skin lesions of patients with atopic dermatitis (AD) in vitro and in vivo. This approach, however, is unlikely to be therapeutically useful since butyric acid is malodorous and requires a high concentration in the mM range for growth suppression of AD S. aureus. A derivative of butyric acid, BA–NH–NH–BA, was synthesized by conjugation of two butyric acids to both ends of an –NH–O–NH– linker. BA–NH–NH–BA significantly lowered the concentration of butyric acid required to inhibit the growth of AD S. aureus. Like butyric acid, BA–NH–NH–BA functioned as a histone deacetylase (HDAC) inhibitor by inducing the acetylation of Histone H3 lysine 9 (AcH3K9) in human keratinocytes. Furthermore, BA–NH–NH–BA ameliorated AD S. aureus-induced production of pro-inflammatory interleukin (IL)-6 and remarkably reduced the colonization of AD S. aureus in mouse skin. These results describe a novel derivative of a skin microbiome fermentation metabolite that exhibits anti-inflammatory and S. aureus bactericidal activity.
Collapse
Affiliation(s)
- Supitchaya Traisaeng
- Department of Life Sciences, National Central University, Taoyuan 32001, Taiwan.
| | - Deron Raymond Herr
- Department of Pharmacology, National University of Singapore, Singapore 117600, Singapore.
| | - Hsin-Jou Kao
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan 32001, Taiwan.
| | - Tsung-Hsien Chuang
- Immunology Research Center, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan.
| | - Chun-Ming Huang
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan 32001, Taiwan.
- Department of Dermatology, University of California, San Diego 3525 John Hopkins Court, Rm276, San Diego, CA 92121, USA.
| |
Collapse
|
160
|
Prentice PM, Schoemaker MH, Vervoort J, Hettinga K, Lambers TT, van Tol EAF, Acerini CL, Olga L, Petry CJ, Hughes IA, Koulman A, Ong KK, Dunger DB. Human Milk Short-Chain Fatty Acid Composition is Associated with Adiposity Outcomes in Infants. J Nutr 2019; 149:716-722. [PMID: 31050748 DOI: 10.1093/jn/nxy320] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 10/25/2018] [Accepted: 12/18/2018] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Presumed benefits of human milk (HM) in avoiding rapid infancy weight gain and later obesity could relate to its nutrient composition. However, data on breast milk composition and its relation with growth are sparse. OBJECTIVE We investigated whether short-chain fatty acids (SCFAs), known to be present in HM and linked to energy metabolism, are associated with infancy anthropometrics. METHODS In a prospective birth cohort, HM hindmilk samples were collected from 619 lactating mothers at 4-8 wk postnatally [median (IQR) age: 33.9 (31.3-36.5) y, body mass index (BMI) (kg/m2): 22.8 (20.9-25.2)]. Their offspring, born at 40.1 (39.1-41.0) wk gestation with weight 3.56 (3.22-3.87) kg and 51% male, were assessed with measurement of weight, length, and skinfold thickness at ages 3, 12, and 24 mo, and transformed to age- and sex-adjusted z scores. HM SCFAs were measured by 1H-nuclear magnetic resonance spectroscopy (NMR) and GC-MS. Multivariable linear regression models were conducted to analyze the relations between NMR HM SCFAs and infancy growth parameters with adjustment for potential confounders. RESULTS NMR peaks for HM butyrate, acetate, and formic acid, but not propionate, were detected. Butyrate peaks were 17.8% higher in HM from exclusively breastfeeding mothers than mixed-feeding mothers (P = 0.003). HM butyrate peak values were negatively associated with changes in infant weight (standardized B = -0.10, P = 0.019) and BMI (B = -0.10, P = 0.018) between 3 and 12 mo, and negatively associated with BMI (B = -0.10, P = 0.018) and mean skinfold thickness (B = -0.10, P = 0.049) at age 12 mo. HM formic acid peak values showed a consistent negative association with infant BMI at all time points (B < = -0.10, P < = 0.014), whereas HM acetate was negatively associated with skinfold thickness at 3 mo (B = -0.10, P = 0.028) and 24 mo (B = -0.10, P = 0.036). CONCLUSIONS These results suggest that HM SCFAs play a beneficial role in weight gain and adiposity during infancy. Further knowledge of HM SCFA function may inform future strategies to support healthy growth.
Collapse
Affiliation(s)
- Philippa M Prentice
- Department of Paediatrics, University of Cambridge, Cambridge, United Kingdom
| | | | - Jacques Vervoort
- Department of Agrotechnology and Food Sciences, Wageningen University, the Netherlands
| | - Kasper Hettinga
- Department of Agrotechnology and Food Sciences, Wageningen University, the Netherlands
| | - Tim T Lambers
- Mead Johnson Pediatric Nutrition Institute, Nijmegen, the Netherlands
| | - Eric A F van Tol
- Mead Johnson Pediatric Nutrition Institute, Nijmegen, the Netherlands
| | - Carlo L Acerini
- Department of Paediatrics, University of Cambridge, Cambridge, United Kingdom
| | - Laurentya Olga
- Department of Paediatrics, University of Cambridge, Cambridge, United Kingdom
| | - Clive J Petry
- Department of Paediatrics, University of Cambridge, Cambridge, United Kingdom
| | - Ieuan A Hughes
- Department of Paediatrics, University of Cambridge, Cambridge, United Kingdom
| | - Albert Koulman
- MRC Epidemiology Unit, Wellcome Trust-MRC Institute of Metabolic Science, NIHR Cambridge Comprehensive Biomedical Research Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, United Kingdom
| | - Ken K Ong
- Department of Paediatrics, University of Cambridge, Cambridge, United Kingdom.,Wellcome Trust-MRC Institute of Metabolic Science, NIHR Cambridge Comprehensive Biomedical Research Centre, University of Cambridge, Cambridge, United Kingdom.,MRC Epidemiology Unit, Wellcome Trust-MRC Institute of Metabolic Science, NIHR Cambridge Comprehensive Biomedical Research Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, United Kingdom
| | - David B Dunger
- Department of Paediatrics, University of Cambridge, Cambridge, United Kingdom.,Wellcome Trust-MRC Institute of Metabolic Science, NIHR Cambridge Comprehensive Biomedical Research Centre, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
161
|
Mellon SH, Bersani FS, Lindqvist D, Hammamieh R, Donohue D, Dean K, Jett M, Yehuda R, Flory J, Reus VI, Bierer LM, Makotkine I, Abu Amara D, Henn Haase C, Coy M, Doyle FJ, Marmar C, Wolkowitz OM. Metabolomic analysis of male combat veterans with post traumatic stress disorder. PLoS One 2019; 14:e0213839. [PMID: 30883584 PMCID: PMC6422302 DOI: 10.1371/journal.pone.0213839] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 03/02/2019] [Indexed: 12/26/2022] Open
Abstract
Posttraumatic stress disorder (PTSD) is associated with impaired major domains of psychology and behavior. Individuals with PTSD also have increased co-morbidity with several serious medical conditions, including autoimmune diseases, cardiovascular disease, and diabetes, raising the possibility that systemic pathology associated with PTSD might be identified by metabolomic analysis of blood. We sought to identify metabolites that are altered in male combat veterans with PTSD. In this case-control study, we compared metabolomic profiles from age-matched male combat trauma-exposed veterans from the Iraq and Afghanistan conflicts with PTSD (n = 52) and without PTSD (n = 51) (‘Discovery group’). An additional group of 31 PTSD-positive and 31 PTSD-negative male combat-exposed veterans was used for validation of these findings (‘Test group’). Plasma metabolite profiles were measured in all subjects using ultrahigh performance liquid chromatography/tandem mass spectrometry and gas chromatography/mass spectrometry. We identified key differences between PTSD subjects and controls in pathways related to glycolysis and fatty acid uptake and metabolism in the initial ‘Discovery group’, consistent with mitochondrial alterations or dysfunction, which were also confirmed in the ‘Test group’. Other pathways related to urea cycle and amino acid metabolism were different between PTSD subjects and controls in the ‘Discovery’ but not in the smaller ‘Test’ group. These metabolic differences were not explained by comorbid major depression, body mass index, blood glucose, hemoglobin A1c, smoking, or use of analgesics, antidepressants, statins, or anti-inflammatories. These data show replicable, wide-ranging changes in the metabolic profile of combat-exposed males with PTSD, with a suggestion of mitochondrial alterations or dysfunction, that may contribute to the behavioral and somatic phenotypes associated with this disease.
Collapse
Affiliation(s)
- Synthia H. Mellon
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of California, San Francisco, CA, United States of America
- * E-mail:
| | - F. Saverio Bersani
- Department of Psychiatry and UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA, United States of America
| | - Daniel Lindqvist
- Department of Psychiatry and UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA, United States of America
| | - Rasha Hammamieh
- Integrative Systems Biology, US Army Medical Research and Materiel Command, USACEHR, Fort Detrick, Frederick, MD, United States of America
| | - Duncan Donohue
- Integrative Systems Biology, US Army Medical Research and Materiel Command, USACEHR, Fort Detrick, Frederick, MD, United States of America
| | - Kelsey Dean
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, United States of America
| | - Marti Jett
- Integrative Systems Biology, US Army Medical Research and Materiel Command, USACEHR, Fort Detrick, Frederick, MD, United States of America
| | - Rachel Yehuda
- Department of Psychiatry, James J. Peters VA Medical Center, Bronx, NY and Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - Janine Flory
- Department of Psychiatry, James J. Peters VA Medical Center, Bronx, NY and Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - Victor I. Reus
- Department of Psychiatry and UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA, United States of America
| | - Linda M. Bierer
- Department of Psychiatry, James J. Peters VA Medical Center, Bronx, NY and Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - Iouri Makotkine
- Department of Psychiatry, James J. Peters VA Medical Center, Bronx, NY and Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - Duna Abu Amara
- Department of Psychiatry, New York University Langone Medical School, New York, NY, United States of America
| | - Clare Henn Haase
- Department of Psychiatry, New York University Langone Medical School, New York, NY, United States of America
| | - Michelle Coy
- Department of Psychiatry and UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA, United States of America
| | - Francis J. Doyle
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, United States of America
| | - Charles Marmar
- Department of Psychiatry, New York University Langone Medical School, New York, NY, United States of America
- Stephen and Alexandra Cohen Veteran Center for Posttraumatic Stress and Traumatic Brain Injury, New York, NY, United States of America
| | - Owen M. Wolkowitz
- Department of Psychiatry and UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA, United States of America
| |
Collapse
|
162
|
Silva LG, Ferguson BS, Avila AS, Faciola AP. Sodium propionate and sodium butyrate effects on histone deacetylase (HDAC) activity, histone acetylation, and inflammatory gene expression in bovine mammary epithelial cells. J Anim Sci 2019; 96:5244-5252. [PMID: 30252114 DOI: 10.1093/jas/sky373] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 09/19/2018] [Indexed: 12/12/2022] Open
Abstract
Histone deacetylase (HDAC) inhibition attenuates inflammation in rodents and short-chain fatty acids (SCFAs) are effective HDAC inhibitors. Therefore, the objective of this study was to evaluate the role of the SCFAs sodium propionate (SP) and sodium butyrate (SB) as HDAC-dependent regulators of inflammatory gene expression in bovine mammary epithelial cells (MAC-Ts). We postulated that SP and SB would decrease inflammation in MAC-Ts by inhibiting HDAC activity and increasing histone H3 acetylation and consequently decreasing inflammatory gene expression. For this study, MAC-Ts stimulated with lipopolysaccharide (LPS) were used as a model for bovine mammary epithelial cell inflammation. MAC-Ts were cultured in a basal medium. Cell lysates were incubated with SP or SB (0 to 5 mM) for 2 h prior to HDAC substrates incubation for an additional 2 h and HDACs activity was determined. Next, cells were pretreated with SP or SB (0 to 3.0 mM) for 2 h prior to LPS (1 µg/mL) stimulation for an additional 2 h and assessed for histone H3 acetylation. Then, cells were pretreated with SP or SB (1 mM) for 24 h prior to LPS (1 µg/mL) stimulation for an additional 2 h and RNA was isolated for inflammatory gene expression evaluation by PCR array and gene validation was performed using quantitative real-time PCR. One-way ANOVA followed by Tukey post hoc analysis was conducted and statistical significance set at P < 0.05. SP and SB concentration-dependently and selectively inhibited class I HDAC activity, which differed between SCFAs, where SB inhibited (P < 0.05) HDACs 2, 3, and 8, while SP inhibited (P < 0.05) HDACs 2 and 8. Histone H3 acetylation was concentration-dependently increased by SCFAs and likewise the differential regulation of HDAC activity, SCFAs effected differently histone H3 acetylation, where SB increased (P < 0.05) H3K9/14, H3K18 and H3K27 acetylation, while SP increased (P < 0.05) H3K9/14 and H3K18 acetylation. However, SCFAs did not decrease (P > 0.05) overall inflammatory gene expression. Under our experimental conditions, findings suggest that in MAC-Ts, SCFAs regulate epigenetic markers on nucleosomal DNA in addition to regulation of inflammatory gene events independent of HDAC activity. Nevertheless, examination of SCFAs and/or HDACs inhibitors in bovine mammary gland is worth being further investigated to delineate the potential impact of HDAC inhibition and histones hyperacetylation on mammary gland tissue inflammation.
Collapse
Affiliation(s)
- Lorrayny G Silva
- Department of Animal Sciences, University of Florida, Gainesville, FL
| | - Bradley S Ferguson
- Department Agriculture, Nutrition, and Veterinary Sciences, University of Nevada, Reno, NV
| | - Andre S Avila
- Department of Animal Sciences, University of Florida, Gainesville, FL.,Agricultural Science Center, Universidade Estadual do Oeste do Parana, Marechal Candido Rondon, PR, Brazil
| | - Antonio P Faciola
- Department of Animal Sciences, University of Florida, Gainesville, FL
| |
Collapse
|
163
|
Mandaliya DK, Seshadri S. Short Chain Fatty Acids, pancreatic dysfunction and type 2 diabetes. Pancreatology 2019; 19:280-284. [PMID: 30713129 DOI: 10.1016/j.pan.2019.01.021] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 01/10/2019] [Accepted: 01/25/2019] [Indexed: 02/07/2023]
Abstract
The microbiota living in gut influence the immune response, metabolism, mood and behavior. The diet plays a pivotal role in maintaining healthy gut microbiota composition and its fermentation leads to production of Short Chain Fatty Acids (SCFAs) mainly acetate, propionate and butyrate. During pancreatic dysfunction, insulin mediated suppression of glucagon is impaired leading to uncontrolled glucose production by liver and state of hyperglycemia. Insulin and glucagon balance is as important as insulin sensitivity which is reduced during Type 2 Diabetes (T2D). Glucagon like peptide-1 (GLP1) produced by Intestinal epithelial cells regulates insulin and glucagon secretion directly via GLP1 receptor on pancreatic cells or via nervous system. But half-life period of GLP1 is very short i.e. about 2 min, after which it is cleaved and inactivated. SCFAs are well documented to induce GLP1 but its direct effect on pancreatic dysfunction has not been reported. This review opens a new avenue to study the role of SCFAs as treatment to pancreatic dysfunction and T2D.
Collapse
Affiliation(s)
| | - Sriram Seshadri
- Institute of Science, Nirma University, Ahmedabad, Gujarat, 382481, India.
| |
Collapse
|
164
|
Nijhuis L, Peeters JGC, Vastert SJ, van Loosdregt J. Restoring T Cell Tolerance, Exploring the Potential of Histone Deacetylase Inhibitors for the Treatment of Juvenile Idiopathic Arthritis. Front Immunol 2019; 10:151. [PMID: 30792714 PMCID: PMC6374297 DOI: 10.3389/fimmu.2019.00151] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 01/17/2019] [Indexed: 12/24/2022] Open
Abstract
Juvenile Idiopathic Arthritis (JIA) is characterized by a loss of immune tolerance. Here, the balance between the activity of effector T (Teff) cells and regulatory T (Treg) cells is disturbed resulting in chronic inflammation in the joints. Presently, therapeutic strategies are predominantly aimed at suppressing immune activation and pro-inflammatory effector mechanisms, ignoring the opportunity to also promote tolerance by boosting the regulatory side of the immune balance. Histone deacetylases (HDACs) can deacetylate both histone and non-histone proteins and have been demonstrated to modulate epigenetic regulation as well as cellular signaling in various cell types. Importantly, HDACs are potent regulators of both Teff cell and Treg cell function and can thus be regarded as attractive therapeutic targets in chronic inflammatory arthritis. HDAC inhibitors (HDACi) have proven therapeutic potential in the cancer field, and are presently being explored for their potential in the treatment of autoimmune diseases. Specific HDACi have already been demonstrated to reduce the secretion of pro-inflammatory cytokines by Teff cells, and promote Treg numbers and suppressive capacity in vitro and in vivo. In this review, we outline the role of the different classes of HDACs in both Teff cell and Treg cell function. Furthermore, we will review the effect of different HDACi on T cell tolerance and explore their potential as a therapeutic strategy for the treatment of oligoarticular and polyarticular JIA.
Collapse
Affiliation(s)
- Lotte Nijhuis
- Laboratory of Translational Immunology, Department of Pediatric Immunology & Rheumatology, University Medical Center Utrecht, University of Utrecht, Utrecht, Netherlands
| | - Janneke G C Peeters
- Laboratory of Translational Immunology, Department of Pediatric Immunology & Rheumatology, University Medical Center Utrecht, University of Utrecht, Utrecht, Netherlands
| | - Sebastiaan J Vastert
- Laboratory of Translational Immunology, Department of Pediatric Immunology & Rheumatology, University Medical Center Utrecht, University of Utrecht, Utrecht, Netherlands
| | - Jorg van Loosdregt
- Laboratory of Translational Immunology, Department of Pediatric Immunology & Rheumatology, University Medical Center Utrecht, University of Utrecht, Utrecht, Netherlands
| |
Collapse
|
165
|
Cornell K, Alam M, Lyden E, Wood L, LeVan TD, Nordgren TM, Bailey K, Hanson C. Saturated Fat Intake Is Associated with Lung Function in Individuals with Airflow Obstruction: Results from NHANES 2007⁻2012. Nutrients 2019; 11:nu11020317. [PMID: 30717299 PMCID: PMC6413158 DOI: 10.3390/nu11020317] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 01/28/2019] [Accepted: 01/29/2019] [Indexed: 01/14/2023] Open
Abstract
Nutritional status is a well-recognized prognostic indicator in chronic obstructive pulmonary disease (COPD); however, very little is known about the relationship between lung function and saturated fat intake. We used data from the cross-sectional National Health and Nutrition Examination Surveys (NHANES) to assess the relationship between saturated fatty acid (SFA) intake and lung function in the general US adult population. Adults in NHANES (2007⁻2012) with pre-bronchodilator spirometry measurements and dietary SFA intake were included. Primary outcomes were lung function including forced expiratory volume in one second (FEV₁), FEV₁, forced vital capacity (FVC), FEV₁/FVC ratio, percent predicted FEV₁ and percent predicted FVC. Multivariable regression models in the general population as well as those with spirometry-defined airflow obstruction were used to assess the relationship between lung function measurements and dietary SFA intake after adjustment for confounders. 11,180 eligible participants were included in this study. Univariate analysis revealed a statistically significant positive association between total SFA intake and lung function outcomes; however, these relationships were attenuated after adjustment for covariates. A secondary analysis of individuals with spirometry-defined airflow obstruction (FEV₁/FVC < 0.7) revealed that a lower intake of SFA was associated with reduced FEV1 (β = -126.4, p = 0.04 for quartile 1 vs. quartile 4), FVC (β = -165.8. p = 0.01 for quartile 1 vs. quartile 4), and percent predicted FVC (β = -3.3. p = 0.04 for quartile 1 vs. quartile 4), after adjustment for relevant confounders. No associations were observed for the FEV₁/FVC ratio and percent predicted FEV₁. It is possible that characteristics such as food source and fatty acid chain length may influence associations between saturated fatty acid intake and health outcomes.
Collapse
Affiliation(s)
- Kasey Cornell
- Internal Medicine Pulmonary, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| | - Morshed Alam
- College of Public Health, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| | - Elizabeth Lyden
- College of Public Health, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| | - Lisa Wood
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle NSW 2308, Australia.
| | - Tricia D LeVan
- Internal Medicine Pulmonary, University of Nebraska Medical Center, Omaha, NE 68198, USA.
- College of Public Health Epidemiology, University of Nebraska, Omaha, NE 68198, USA.
- Research Service, VA Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA.
| | - Tara M Nordgren
- Division of Biomedical Sciences, University of California Riverside, Riverside, CA 92521, USA.
| | - Kristina Bailey
- Internal Medicine Pulmonary, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| | - Corrine Hanson
- Medical Nutrition Education, College of Allied Health Professions, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| |
Collapse
|
166
|
Zhou JS, Guo P, Yu HB, Ji H, Lai ZW, Chen YA. Growth performance, lipid metabolism, and health status of grass carp (Ctenopharyngodon idella) fed three different forms of sodium butyrate. FISH PHYSIOLOGY AND BIOCHEMISTRY 2019; 45:287-298. [PMID: 30238219 DOI: 10.1007/s10695-018-0561-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 08/20/2018] [Indexed: 06/08/2023]
Abstract
Sodium butyrate (SB) can be coated with fatty acid matrix. In this study, the effects of three SB forms, being zero-lipid-coated (SB-A), half-lipid-coated (SB-B), and 2/3 lipid-coated (SB-C) (w/w), on growth, lipid metabolism, and health status of grass carp (Ctenopharyngodon idella) were investigated. The three forms of SB were added to a control diet to form three SB diets, Con., SB-A, SB-B, and SB-C, where the pure SB in each SB diet was kept at the same level (500 mg kg-1). A total of 216 C. idella (14.10 ± 0.60 g/fish) were allotted into four groups (triplicate per group) and fed the four diets respectively for 56 days, and then fish were sampled and determined. Fish growth was not affected by any of the three forms of SB. Viscerosomatic index, intraperitoneal fat index, and crude lipid of hepatopancreas and muscle were significantly decreased and villus height of intestine and mRNA expression of MyD88 and TLR22 in hepatopancreas were significantly improved in SB diets compared with control (p < 0.05), respectively. MiSeq sequencing of the V3-V4 region of bacterial 16S rRNA gene revealed that SB increased the relative abundances of intestinal healthy bacteria, Fusobacteria and Bacteroides, and the abundances of Cetobacterium decreased in the SB-C group. In conclusion, the present results showed that three forms of SB, without affecting the growth of fish, respectively decreased lipid accumulation and probably have a beneficial effect on health of C. idella.
Collapse
Affiliation(s)
- Ji Shu Zhou
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Pan Guo
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Hai Bo Yu
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Hong Ji
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China.
| | - Zhou Wen Lai
- New Austrian Biotechnology Co., Ltd., Xiamen, 361004, Fujian Province, China
| | - Yi An Chen
- New Austrian Biotechnology Co., Ltd., Xiamen, 361004, Fujian Province, China
| |
Collapse
|
167
|
Zhong X, Zhang Z, Wang S, Cao L, Zhou L, Sun A, Zhong Z, Nabben M. Microbial-Driven Butyrate Regulates Jejunal Homeostasis in Piglets During the Weaning Stage. Front Microbiol 2019; 9:3335. [PMID: 30713531 PMCID: PMC6345722 DOI: 10.3389/fmicb.2018.03335] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 12/24/2018] [Indexed: 02/05/2023] Open
Abstract
Microbe-derived butyrate plays an important role in the gut health of young mammals during the weaning stage. A greater understanding of how butyrate regulates intestinal development is necessary for overcoming post-weaning diarrheal diseases. We aimed to investigate whether jejunal microbial metabolite butyrate modulates the apoptosis/proliferation balance and immune response in piglets during the post-weaning period of the first 3 weeks of life. On the one hand, during the first week post-weaning, the relative abundances of the dominant bacterial families Erysipelotrichaceae (P < 0.01) and Lachnospiraceae (P < 0.01) were increased, which induced decreases in both butyrate production (P < 0.05) and its receptor (G-protein coupled receptor 43) expression (P < 0.01). The resulting intestinal inflammation (inferred from increased TNF-α and IFN-γ expression) contributed to the onset of cell apoptosis and the inhibition of cell-proliferation along the crypt-villus axis, which were followed by impaired jejunal morphology (i.e., increased crypt-depth) (P < 0.05) and intestinal dysfunction (i.e., decreased creatine kinase, and lactate dehydrogenase) (P < 0.05). On the other hand, during the second week post-weaning, the relative abundances of Lactobacillaceae (P < 0.01) and Ruminococcaceae (P < 0.05) were increased. The increases were accompanied by increased butyrate production (P < 0.05) and its receptor expression (P < 0.01), leading to the inhibition of cell apoptosis and the stimulation of cell proliferation via decreased pro-inflammatory cytokines and thereby the improvement of intestinal development and function. Herein, this study demonstrates that microbial-driven butyrate might be a key modulator in the maintenance of intestinal homeostasis after weaning. The findings suggest that strategies to promote butyrate production can maintain the apoptosis/proliferation balance via minimizing intestinal inflammation, and thereby improving post-weaning jejunal adaptation toward gut health.
Collapse
Affiliation(s)
- Xi Zhong
- Intensive Care Unit, West China Hospital, Sichuan University, Chengdu, China
| | - Zhongwei Zhang
- Intensive Care Unit, West China Hospital, Sichuan University, Chengdu, China
| | - Shujin Wang
- Department of Genetics and Cell Biology, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, Netherlands
| | - Lili Cao
- Medical School, Chengdu University, Chengdu, China
| | - Lin Zhou
- Shenzhen Premix Inve Nutrition, Co., Ltd., Shenzhen, China
| | - Aomin Sun
- Department of Genetics and Cell Biology, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, Netherlands
| | | | - Miranda Nabben
- Department of Genetics and Cell Biology, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
168
|
Wang J, Tian S, Yu H, Wang J, Zhu W. Response of Colonic Mucosa-Associated Microbiota Composition, Mucosal Immune Homeostasis, and Barrier Function to Early Life Galactooligosaccharides Intervention in Suckling Piglets. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:578-588. [PMID: 30562014 DOI: 10.1021/acs.jafc.8b05679] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Suckling piglets were used to investigate the response of colonic mucosa-associated microbiota composition, mucosal immune homeostasis, and barrier function to early life galactooligosaccharides (GOS) intervention. Ten milliliter 1 g/kg body weight GOS solutions and physiological saline solutions were fed to the newborn piglets in the GOS group and in the control (CON) group a week time, respectively. Six piglets from each group were euthanized on day 8 and day 21. GOS piglets had a higher abundance of short-chain fatty acids (SCFAs) producer such as Prevotella, Barnesiella, Parabacteroides, and Unclassified Porphyromonadaceae in colonic mucosa ( P < 0.05). In addition, the total SCFAs level in colonic digesta of GOS piglets increased on day 8 ( P < 0.05) and day 21 ( P = 0.064). Meanwhile, a higher SCFAs concentration in colon of the GOS piglets altered the gene expression of inflammatory cytokines (IL-8 and IL-10) and barrier proteins (ZO-1 and Claudin-1) through regulating the phosphorylation of the NFκB and AMPK signaling pathway. In summary, these results provide important insights and understandings to reveal the relationship between the mucosal microbiota colonization and intestinal function at the early life stage of piglets.
Collapse
Affiliation(s)
- Jue Wang
- National Center for International Research on Animal Gut Nutrition, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, Laboratory of Gastrointestinal Microbiology, Joint International Research Laboratory of Animal Health and Food Safety, College of Animal Science and Technology , Nanjing Agricultural University , Nanjing 210095 , China
| | - Shiyi Tian
- National Center for International Research on Animal Gut Nutrition, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, Laboratory of Gastrointestinal Microbiology, Joint International Research Laboratory of Animal Health and Food Safety, College of Animal Science and Technology , Nanjing Agricultural University , Nanjing 210095 , China
| | - Hu Yu
- National Center for International Research on Animal Gut Nutrition, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, Laboratory of Gastrointestinal Microbiology, Joint International Research Laboratory of Animal Health and Food Safety, College of Animal Science and Technology , Nanjing Agricultural University , Nanjing 210095 , China
| | - Jing Wang
- National Center for International Research on Animal Gut Nutrition, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, Laboratory of Gastrointestinal Microbiology, Joint International Research Laboratory of Animal Health and Food Safety, College of Animal Science and Technology , Nanjing Agricultural University , Nanjing 210095 , China
| | - Weiyun Zhu
- National Center for International Research on Animal Gut Nutrition, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, Laboratory of Gastrointestinal Microbiology, Joint International Research Laboratory of Animal Health and Food Safety, College of Animal Science and Technology , Nanjing Agricultural University , Nanjing 210095 , China
| |
Collapse
|
169
|
Fu X, Liu Z, Zhu C, Mou H, Kong Q. Nondigestible carbohydrates, butyrate, and butyrate-producing bacteria. Crit Rev Food Sci Nutr 2018; 59:S130-S152. [PMID: 30580556 DOI: 10.1080/10408398.2018.1542587] [Citation(s) in RCA: 324] [Impact Index Per Article: 46.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Nondigestible carbohydrates (NDCs) are fermentation substrates in the colon after escaping digestion in the upper gastrointestinal tract. Among NDCs, resistant starch is not hydrolyzed by pancreatic amylases but can be degraded by enzymes produced by large intestinal bacteria, including clostridia, bacteroides, and bifidobacteria. Nonstarch polysaccharides, such as pectin, guar gum, alginate, arabinoxylan, and inulin fructans, and nondigestible oligosaccharides and their derivatives, can also be fermented by beneficial bacteria in the large intestine. Butyrate is one of the most important metabolites produced through gastrointestinal microbial fermentation and functions as a major energy source for colonocytes by directly affecting the growth and differentiation of colonocytes. Moreover, butyrate has various physiological effects, including enhancement of intestinal barrier function and mucosal immunity. In this review, several representative NDCs are introduced, and their chemical components, structures, and physiological functions, including promotion of the proliferation of butyrate-producing bacteria and enhancement of butyrate production, are discussed. We also describe the strategies for achieving directional accumulation of colonic butyrate based on endogenous generation mechanisms.
Collapse
Affiliation(s)
- Xiaodan Fu
- a College of Food Science and Engineering , Ocean University of China , Qingdao , China
| | - Zhemin Liu
- a College of Food Science and Engineering , Ocean University of China , Qingdao , China
| | - Changliang Zhu
- a College of Food Science and Engineering , Ocean University of China , Qingdao , China
| | - Haijin Mou
- a College of Food Science and Engineering , Ocean University of China , Qingdao , China
| | - Qing Kong
- a College of Food Science and Engineering , Ocean University of China , Qingdao , China
| |
Collapse
|
170
|
Quagliariello V, Masarone M, Armenia E, Giudice A, Barbarisi M, Caraglia M, Barbarisi A, Persico M. Chitosan-coated liposomes loaded with butyric acid demonstrate anticancer and anti-inflammatory activity in human hepatoma HepG2 cells. Oncol Rep 2018; 41:1476-1486. [PMID: 30569138 PMCID: PMC6365699 DOI: 10.3892/or.2018.6932] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Accepted: 10/17/2018] [Indexed: 02/07/2023] Open
Abstract
Butyric acid (BA) has been reported to induce anticancer effects on hepatocellular carcinoma (HCC) cells both in vitro and in vivo. However, its delivery and release in cancer tissues must be optimized. On the basis of these requirements, we prepared liposomes coated with chitosan and uncoated liposomes and both types were loaded with BA through a thin-film hydration method. The liposomes coated or uncoated with chitosan had a mean hydrodynamic size of 83.5 and 110.3 nm, respectively, with a homogeneous size distribution of the particles. For evaluation of the biological effects of the nanoformulations, the hepatoblastoma (HB) HepG2 cell line was utilized. BA-loaded liposomes coated with chitosan showed a considerable higher cytotoxicity than both uncoated liposomes and free BA, with IC50 values, after 72 h of incubation, of 7.5, 2.5 and 1.6 mM, respectively. Treatment of HepG2 cells for 5 h with the BA-loaded liposomes coated with chitosan at 5 mM lowered the extent of the increase in IL-8, IL-6, TNF-α and TGF-β expression of approximately 64, 58, 85 and 73.8%, respectively, when compared to the untreated cells. The BA-loaded liposomes coated with chitosan had marked capacity to be internalized in human HB cells showing an increased cytotoxic activity when compared with free BA and important anti-inflammatory effects by inhibiting production of cytokines with a central role in liver cell survival.
Collapse
Affiliation(s)
- Vincenzo Quagliariello
- Division of Cardiology, Istituto Nazionale Tumori, IRCCS Fondazione G. Pascale, I-80131 Naples, Italy
| | - Mario Masarone
- Internal Medicine and Hepatology Division, Department of Medicine, University of Medicine of Salerno, Salerno, I-80123 Naples, Italy
| | - Emilia Armenia
- Department of Thoracic and Cardio-Respiratory Sciences, University of Campania 'Luigi Vanvitelli', I-80138 Naples, Italy
| | - Aldo Giudice
- Istituto Nazionale Tumori IRCCS Fondazione Pascale, I-80131 Napoli, Italy
| | - Manlio Barbarisi
- Department of Medical, Surgical, Neurological, Metabolic and Aging Sciences, University of Campania 'Luigi Vanvitelli', I-80138 Naples, Italy
| | - Michele Caraglia
- Department of Precision Medicine, University of Campania 'Luigi Vanvitelli', I-80138 Naples, Italy
| | - Alfonso Barbarisi
- Department of Thoracic and Cardio-Respiratory Sciences, University of Campania 'Luigi Vanvitelli', I-80138 Naples, Italy
| | - Marcello Persico
- Internal Medicine and Hepatology Division, Department of Medicine, University of Medicine of Salerno, Salerno, I-80123 Naples, Italy
| |
Collapse
|
171
|
Gut Microbiome Dysbiosis and Immunometabolism: New Frontiers for Treatment of Metabolic Diseases. Mediators Inflamm 2018; 2018:2037838. [PMID: 30622429 PMCID: PMC6304917 DOI: 10.1155/2018/2037838] [Citation(s) in RCA: 180] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 10/23/2018] [Indexed: 02/06/2023] Open
Abstract
Maintenance of healthy human metabolism depends on a symbiotic consortium among bacteria, archaea, viruses, fungi, and host eukaryotic cells throughout the human gastrointestinal tract. Microbial communities provide the enzymatic machinery and the metabolic pathways that contribute to food digestion, xenobiotic metabolism, and production of a variety of bioactive molecules. These include vitamins, amino acids, short-chain fatty acids (SCFAs), and metabolites, which are essential for the interconnected pathways of glycolysis, the tricarboxylic acid/Krebs cycle, oxidative phosphorylation (OXPHOS), and amino acid and fatty acid metabolism. Recent studies have been elucidating how nutrients that fuel the metabolic processes impact on the ways immune cells, in particular, macrophages, respond to different stimuli under physiological and pathological conditions and become activated and acquire a specialized function. The two major inflammatory phenotypes of macrophages are controlled through differential consumption of glucose, glutamine, and oxygen. M1 phenotype is triggered by polarization signal from bacterial lipopolysaccharide (LPS) and Th1 proinflammatory cytokines such as interferon-γ, TNF-α, and IL-1β, or both, whereas M2 phenotype is triggered by Th2 cytokines such as interleukin-4 and interleukin-13 as well as anti-inflammatory cytokines, IL-10 and TGFβ, or glucocorticoids. Glucose utilization and production of chemical mediators including ATP, reactive oxygen species (ROS), nitric oxide (NO), and NADPH support effector activities of M1 macrophages. Dysbiosis is an imbalance of commensal and pathogenic bacteria and the production of microbial antigens and metabolites. It is now known that the gut microbiota-derived products induce low-grade inflammatory activation of tissue-resident macrophages and contribute to metabolic and degenerative diseases, including diabetes, obesity, metabolic syndrome, and cancer. Here, we update the potential interplay of host gut microbiome dysbiosis and metabolic diseases. We also summarize on advances on fecal therapy, probiotics, prebiotics, symbiotics, and nutrients and small molecule inhibitors of metabolic pathway enzymes as prophylactic and therapeutic agents for metabolic diseases.
Collapse
|
172
|
Nath A, Molnár MA, Csighy A, Kőszegi K, Galambos I, Huszár KP, Koris A, Vatai G. Biological Activities of Lactose-Based Prebiotics and Symbiosis with Probiotics on Controlling Osteoporosis, Blood-Lipid and Glucose Levels. ACTA ACUST UNITED AC 2018; 54:medicina54060098. [PMID: 30513975 PMCID: PMC6306850 DOI: 10.3390/medicina54060098] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Accepted: 11/28/2018] [Indexed: 02/07/2023]
Abstract
Lactose-based prebiotics are synthesized by enzymatic- or microbial- biotransformation of lactose and have unique functional values. In this comprehensive review article, the biochemical mechanisms of controlling osteoporosis, blood-lipid, and glucose levels by lactose-based prebiotics and symbiosis with probiotics are reported along with the results of clinical investigations. Interaction between lactose-based prebiotics and probiotics reduces osteoporosis by (a) transforming insoluble inorganic salts to soluble and increasing their absorption to gut wall; (b) maintaining and protecting mineral absorption surface in the intestine; (c) increasing the expression of calcium-binding proteins in the gut wall; (d) remodeling osteoclasts and osteoblasts formation; (e) releasing bone modulating factors; and (f) degrading mineral complexing phytic acid. Lactose-based prebiotics with probiotics control lipid level in the bloodstream and tissue by (a) suppressing the expressions of lipogenic- genes and enzymes; (b) oxidizing fatty acids in muscle, liver, and adipose tissue; (c) binding cholesterol with cell membrane of probiotics and subsequent assimilation by probiotics; (d) enzymatic-transformations of bile acids; and (e) converting cholesterol to coprostanol and its defecation. Symbiosis of lactose-based prebiotics with probiotics affect plasma glucose level by (a) increasing the synthesis of gut hormones plasma peptide-YY, glucagon-like peptide-1 and glucagon-like peptide-2 from entero-endocrine L-cells; (b) altering glucose assimilation and metabolism; (c) suppressing systematic inflammation; (d) reducing oxidative stress; and (e) producing amino acids. Clinical investigations show that lactose-based prebiotic galacto-oligosaccharide improves mineral absorption and reduces hyperlipidemia. Another lactose-based prebiotic, lactulose, improves mineral absorption, and reduces hyperlipidemia and hyperglycemia. It is expected that this review article will be of benefit to food technologists and medical practitioners.
Collapse
Affiliation(s)
- Arijit Nath
- Department of Food Engineering, Faculty of Food Science, Szent István University, Ménesi st 44, HU-1118 Budapest, Hungary.
- Soós Ernő Water Technology Research Centre, Faculty of Engineering, University of Pannonia, Üllő út., H-3 Nagykanizsa, Hungary.
| | - Máté András Molnár
- Department of Food Engineering, Faculty of Food Science, Szent István University, Ménesi st 44, HU-1118 Budapest, Hungary.
| | - Attila Csighy
- Department of Food Engineering, Faculty of Food Science, Szent István University, Ménesi st 44, HU-1118 Budapest, Hungary.
| | - Kornélia Kőszegi
- Department of Food Engineering, Faculty of Food Science, Szent István University, Ménesi st 44, HU-1118 Budapest, Hungary.
| | - Ildikó Galambos
- Soós Ernő Water Technology Research Centre, Faculty of Engineering, University of Pannonia, Üllő út., H-3 Nagykanizsa, Hungary.
| | - Klára Pásztorné Huszár
- Department of Refrigeration and Livestock Product Technology, Faculty of Food Science, Szent István University, Ménesi st 43⁻45, HU-1118 Budapest, Hungary.
| | - András Koris
- Department of Food Engineering, Faculty of Food Science, Szent István University, Ménesi st 44, HU-1118 Budapest, Hungary.
| | - Gyula Vatai
- Department of Food Engineering, Faculty of Food Science, Szent István University, Ménesi st 44, HU-1118 Budapest, Hungary.
| |
Collapse
|
173
|
Impact of Diet-Modulated Butyrate Production on Intestinal Barrier Function and Inflammation. Nutrients 2018; 10:nu10101499. [PMID: 30322146 PMCID: PMC6213552 DOI: 10.3390/nu10101499] [Citation(s) in RCA: 350] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 10/01/2018] [Accepted: 10/11/2018] [Indexed: 02/07/2023] Open
Abstract
A major challenge in affluent societies is the increase in disorders related to gut and metabolic health. Chronic over nutrition by unhealthy foods high in energy, fat, and sugar, and low in dietary fibre is a key environmental factor responsible for this development, which may cause local and systemic inflammation. A low intake of dietary fibre is a limiting factor for maintaining a viable and diverse microbiota and production of short-chain fatty acids in the gut. A suppressed production of butyrate is crucial, as this short-chain fatty acid (SCFA) can play a key role not only in colonic health and function but also at the systemic level. At both sites, the mode of action is through mediation of signalling pathways involving nuclear NF-κB and inhibition of histone deacetylase. The intake and composition of dietary fibre modulate production of butyrate in the large intestine. While butyrate production is easily adjustable it is more variable how it influences gut barrier function and inflammatory markers in the gut and periphery. The effect of butyrate seems generally to be more consistent and positive on inflammatory markers related to the gut than on inflammatory markers in the peripheral tissue. This discrepancy may be explained by differences in butyrate concentrations in the gut compared with the much lower concentration at more remote sites.
Collapse
|
174
|
Kim YC, Anderson AJ. Rhizosphere pseudomonads as probiotics improving plant health. MOLECULAR PLANT PATHOLOGY 2018; 19:2349-2359. [PMID: 29676842 PMCID: PMC6638116 DOI: 10.1111/mpp.12693] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 04/08/2018] [Accepted: 04/18/2018] [Indexed: 05/25/2023]
Abstract
Many root-colonizing microbes are multifaceted in traits that improve plant health. Although isolates designated as biological control agents directly reduce pathogen growth, many exert additional beneficial features that parallel changes induced in animal and other hosts by health-promoting microbes termed probiotics. Both animal and plant probiotics cause direct antagonism of pathogens and induce systemic immunity in the host to pathogens and other stresses. They also alter host development and improve host nutrition. The probiotic root-colonizing pseudomonads are generalists in terms of plant hosts, soil habitats and the array of stress responses that are ameliorated in the plant. This article illustrates how the probiotic pseudomonads, nurtured by the carbon (C) and nitrogen (N) sources released by the plant in root exudates, form protective biofilms on the root surface and produce the metabolites or enzymes to boost plant health. The findings reveal the multifunctional nature of many of the microbial metabolites in the plant-probiotic interplay. The beneficial effects of probiotics on plant function can contribute to sustainable yield and quality in agricultural production.
Collapse
Affiliation(s)
- Young Cheol Kim
- Department of Applied Biology, College of Agriculture and Life SciencesChonnam National UniversityGwangju 61186South Korea
| | - Anne J. Anderson
- Department of Biological EngineeringUtah State UniversityLoganUT 84322‐4105USA
| |
Collapse
|
175
|
Regulation of the effector function of CD8 + T cells by gut microbiota-derived metabolite butyrate. Sci Rep 2018; 8:14430. [PMID: 30258117 PMCID: PMC6158259 DOI: 10.1038/s41598-018-32860-x] [Citation(s) in RCA: 204] [Impact Index Per Article: 29.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 09/17/2018] [Indexed: 12/15/2022] Open
Abstract
The gut microbiota produces metabolites such as short-chain fatty acids (SCFAs) that regulate the energy homeostasis and impact on immune cell function of the host. Recently, innovative approaches based on the oral administration of SCFAs have been discussed for therapeutic modification of inflammatory immune responses in autoimmune diseases. So far, most studies have investigated the SCFA-mediated effects on CD4+ T cells and antigen presenting cells. Here we show that butyrate and, to a lesser degree, propionate directly modulate the gene expression of CD8+ cytotoxic T lymphocytes (CTLs) and Tc17 cells. Increased IFN-γ and granzyme B expression by CTLs as well as the molecular switch of Tc17 cells towards the CTL phenotype was mediated by butyrate independently of its interaction with specific SCFA-receptors GPR41 and GPR43. Our results indicate that butyrate strongly inhibited histone-deacetylases (HDACs) in CD8+ T cells thereby affecting the gene expression of effector molecules. Accordingly, the pan-HDAC inhibitors trichostatin A (TSA) and sodium valproate exerted similar influence on CD8+ T cells. Furthermore, higher acetate concentrations were also able to increase IFN-γ production in CD8+ T lymphocytes by modulating cellular metabolism and mTOR activity. These findings might have significant implications in adoptive immunotherapy of cancers and in anti-viral immunity.
Collapse
|
176
|
Brugman S, Ikeda-Ohtsubo W, Braber S, Folkerts G, Pieterse CMJ, Bakker PAHM. A Comparative Review on Microbiota Manipulation: Lessons From Fish, Plants, Livestock, and Human Research. Front Nutr 2018; 5:80. [PMID: 30234124 PMCID: PMC6134018 DOI: 10.3389/fnut.2018.00080] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 08/17/2018] [Indexed: 12/12/2022] Open
Abstract
During recent years the impact of microbial communities on the health of their host (being plants, fish, and terrestrial animals including humans) has received increasing attention. The microbiota provides the host with nutrients, induces host immune development and metabolism, and protects the host against invading pathogens (1-6). Through millions of years of co-evolution bacteria and hosts have developed intimate relationships. Microbial colonization shapes the host immune system that in turn can shape the microbial composition (7-9). However, with the large scale use of antibiotics in agriculture and human medicine over the last decades an increase of diseases associated with so-called dysbiosis has emerged. Dysbiosis refers to either a disturbed microbial composition (outgrowth of possible pathogenic species) or a disturbed interaction between bacteria and the host (10). Instead of using more antibiotics to treat dysbiosis there is a need to develop alternative strategies to combat disturbed microbial control. To this end, we can learn from nature itself. For example, the plant root (or "rhizosphere") microbiome of sugar beet contains several bacterial species that suppress the fungal root pathogen Rhizoctonia solani, an economically important fungal pathogen of this crop (11). Likewise, commensal bacteria present on healthy human skin produce antimicrobial molecules that selectively kill skin pathogen Staphylococcus aureus. Interestingly, patients with atopic dermatitis (inflammation of the skin) lacked antimicrobial peptide secreting commensal skin bacteria (12). In this review, we will give an overview of microbial manipulation in fish, plants, and terrestrial animals including humans to uncover conserved mechanisms and learn how we might restore microbial balance increasing the resilience of the host species.
Collapse
Affiliation(s)
- Sylvia Brugman
- Cell Biology and Immunology Group, Animal Sciences Group, Wageningen University and Research, Wageningen, Netherlands
| | - Wakako Ikeda-Ohtsubo
- Food and Feed Immunology Group, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Saskia Braber
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Sciences, Utrecht University, Utrecht, Netherlands
| | - Gert Folkerts
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Sciences, Utrecht University, Utrecht, Netherlands
| | - Corné M. J. Pieterse
- Plant-Microbe Interactions, Department of Biology, Science4Life, Utrecht University, Utrecht, Netherlands
| | - Peter A. H. M. Bakker
- Plant-Microbe Interactions, Department of Biology, Science4Life, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
177
|
|
178
|
Zhu J, Liao M, Yao Z, Liang W, Li Q, Liu J, Yang H, Ji Y, Wei W, Tan A, Liang S, Chen Y, Lin H, Zhu X, Huang S, Tian J, Tang R, Wang Q, Mo Z. Breast cancer in postmenopausal women is associated with an altered gut metagenome. MICROBIOME 2018; 6:136. [PMID: 30081953 PMCID: PMC6080540 DOI: 10.1186/s40168-018-0515-3] [Citation(s) in RCA: 183] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Accepted: 07/10/2018] [Indexed: 05/12/2023]
Abstract
BACKGROUND Increasing evidence suggests that gut microbiota play a role in the pathogenesis of breast cancer. The composition and functional capacity of gut microbiota associated with breast cancer have not been studied systematically. METHODS We performed a comprehensive shotgun metagenomic analysis of 18 premenopausal breast cancer patients, 25 premenopausal healthy controls, 44 postmenopausal breast cancer patients, and 46 postmenopausal healthy controls. RESULTS Microbial diversity was higher in breast cancer patients than in controls. Relative species abundance in gut microbiota did not differ significantly between premenopausal breast cancer patients and premenopausal controls. In contrast, relative abundance of 45 species differed significantly between postmenopausal patients and postmenopausal controls: 38 species were enriched in postmenopausal patients, including Escherichia coli, Klebsiella sp_1_1_55, Prevotella amnii, Enterococcus gallinarum, Actinomyces sp. HPA0247, Shewanella putrefaciens, and Erwinia amylovora, and 7 species were less abundant in postmenopausal patients, including Eubacterium eligens and Lactobacillus vaginalis. Acinetobacter radioresistens and Enterococcus gallinarum were positively but weakly associated with expression of high-sensitivity C-reactive protein; Shewanella putrefaciens and Erwinia amylovora were positively but weakly associated with estradiol levels. Actinomyces sp. HPA0247 negatively but weakly correlated with CD3+CD8+ T cell numbers. Further characterization of metagenome functional capacity indicated that the gut metagenomes of postmenopausal breast cancer patients were enriched in genes encoding lipopolysaccharide biosynthesis, iron complex transport system, PTS system, secretion system, and beta-oxidation. CONCLUSION The composition and functions of the gut microbial community differ between postmenopausal breast cancer patients and healthy controls. The gut microbiota may regulate or respond to host immunity and metabolic balance. Thus, while cause and effect cannot be determined, there is a reproducible change in the microbiota of treatment-naive patients relative to matched controls.
Collapse
Affiliation(s)
- Jia Zhu
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
- Department of Breast Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, 330000, Jiangxi, China
| | - Ming Liao
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Ziting Yao
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Wenying Liang
- Clabee Genomics, Urban Garden Building, Bookstore Road, Luohu District, Shenzhen, 518000, Guangdong, China
| | - Qibin Li
- Clabee Genomics, Urban Garden Building, Bookstore Road, Luohu District, Shenzhen, 518000, Guangdong, China
| | - Jianlun Liu
- Department of Breast Surgery, The Affiliated Tumor Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Huawei Yang
- Department of Breast Surgery, The Affiliated Tumor Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Yinan Ji
- Department of Breast Surgery, The Affiliated Tumor Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Wei Wei
- Department of Breast Surgery, The Affiliated Tumor Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Aihua Tan
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
- Department of Chemotherapy, The Affiliated Tumor Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Siyuan Liang
- Department of Colorectal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Yang Chen
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Haisong Lin
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Xiujuan Zhu
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Shengzhu Huang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Jiarong Tian
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Ruiqiang Tang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Qiuyan Wang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China.
| | - Zengnan Mo
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China.
| |
Collapse
|
179
|
Lépine A, de Vos P. Synbiotic Effects of the Dietary Fiber Long-Chain Inulin and Probiotic Lactobacillus acidophilus W37 Can be Caused by Direct, Synergistic Stimulation of Immune Toll-Like Receptors and Dendritic Cells. Mol Nutr Food Res 2018; 62:e1800251. [PMID: 29902355 PMCID: PMC6099370 DOI: 10.1002/mnfr.201800251] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 05/02/2018] [Indexed: 01/24/2023]
Abstract
SCOPE Synbiotic effects of dietary fibers and lactobacilli are usually explained by synergistic modulation of gut microbiota. New insight, however, has demonstrated that both dietary fibers and lactobacilli can directly stimulate immune cells and benefit consumer immunity. Here, the synergistic effects of immune active long-chain inulin (lcITF) and Lactobacillus acidophilus W37 (LaW37) on dendritic cells (DCs) are investigated. METHODS AND RESULTS Effects of lcITF and LaW37 alone or combined were studied on Toll-like receptor (TLRs) signaling and cytokine secretion by DCs in the presence and absence of media of intestinal epithelial cell (IEC) exposed to the ingredients. Also, the effects of DC responses against Salmonella Typhimurium (STM) were investigated. Synergistic effects were observed on TLR2 and 3. Synergistic effects were not always pro-inflammatory. LaW37 was strongly pro-inflammatory, while cytokine responses were regulatory when combined with lcITF. Exposure of DCs to IECs medium changed the DCs' response, which revealed synergistic enhancing effects of lcITF/LaW37 on production of IL-6 and IL-8. DCs' response in the presence of STM and LaW37 were so strong that lcITF had no additional effect. CONCLUSION It is demonstrated that synbiotic effects of dietary fibers and bacteria are not limited to the effects on gut microbiota but can also occur by synergistically directly stimulating IECs and/or immune cells.
Collapse
Affiliation(s)
- Alexia Lépine
- Immunoendocrinology GroupDivision of Medical BiologyDepartment of Pathology and Medical BiologyUniversity Medical Center Groningen, University of GroningenHanzeplein 19700RBGroningenThe Netherlands,Food and Biobased ResearchWageningen University and ResearchBornse Weilanden 96708WGWageningenThe Netherlands
| | - Paul de Vos
- Immunoendocrinology GroupDivision of Medical BiologyDepartment of Pathology and Medical BiologyUniversity Medical Center Groningen, University of GroningenHanzeplein 19700RBGroningenThe Netherlands
| |
Collapse
|
180
|
Salem I, Ramser A, Isham N, Ghannoum MA. The Gut Microbiome as a Major Regulator of the Gut-Skin Axis. Front Microbiol 2018; 9:1459. [PMID: 30042740 PMCID: PMC6048199 DOI: 10.3389/fmicb.2018.01459] [Citation(s) in RCA: 359] [Impact Index Per Article: 51.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 06/12/2018] [Indexed: 12/12/2022] Open
Abstract
The adult intestine hosts a myriad of diverse bacterial species that reside mostly in the lower gut maintaining a symbiosis with the human habitat. In the current review, we describe the neoteric advancement in our comprehension of how the gut microbiota communicates with the skin as one of the main regulators in the gut-skin axis. We attempted to explore how this potential link affects skin differentiation and keratinization, its influence on modulating the cutaneous immune response in various diseases, and finally how to take advantage of this communication in the control of different skin conditions.
Collapse
Affiliation(s)
- Iman Salem
- Center for Medical Mycology, Department of Dermatology, Case Western Reserve University, Cleveland, OH, United States
| | - Amy Ramser
- Dermatology, University Hospitals Cleveland Medical Center, Cleveland, OH, United States
| | - Nancy Isham
- Center for Medical Mycology, Department of Dermatology, Case Western Reserve University, Cleveland, OH, United States
| | - Mahmoud A. Ghannoum
- Center for Medical Mycology, Department of Dermatology, Case Western Reserve University, Cleveland, OH, United States
- Dermatology, University Hospitals Cleveland Medical Center, Cleveland, OH, United States
| |
Collapse
|
181
|
Hanuš O, Samková E, Křížová L, Hasoňová L, Kala R. Role of Fatty Acids in Milk Fat and the Influence of Selected Factors on Their Variability-A Review. Molecules 2018; 23:E1636. [PMID: 29973572 PMCID: PMC6100482 DOI: 10.3390/molecules23071636] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 06/29/2018] [Accepted: 07/02/2018] [Indexed: 11/16/2022] Open
Abstract
Fatty acids (FAs) of milk fat are considered to be important nutritional components of the diets of a significant portion of the human population and substantially affect human health. With regard to dairy farming, the FA profile is also seen as an important factor in the technological quality of raw milk. In this sense, making targeted modifications to the FA profile has the potential to significantly contribute to the production of dairy products with higher added value. Thus, FAs also have economic importance. Current developments in analytical methods and their increasing efficiency enable the study of FA profiles not only for scientific purposes but also in terms of practical technological applications. It is important to study the sources of variability of FAs in milk, which include population genetics, type of farming, and targeted animal nutrition. It is equally important to study the health and technological impacts of FAs. This review summarizes current knowledge in the field regarding sources of FA variability, including the impact of factors such as: animal nutrition, seasonal feed changes, type of animal farming (conventional and organic), genetic parameters (influence of breed), animal individuality, lactation, and milk yield. Potential practical applications (to improve food technology and consumer health) of FA profile information are also reviewed.
Collapse
Affiliation(s)
- Oto Hanuš
- Dairy Research Institute Ltd., 16000 Prague, Czech Republic.
| | - Eva Samková
- Department of Food Biotechnologies and Agricultural Products´ Quality, Faculty of Agriculture, University of South Bohemia, 37005 České Budějovice, Czech Republic.
| | - Ludmila Křížová
- Department of Animal Nutrition, Faculty of Veterinary Hygiene and Ecology, University of Veterinary and Pharmaceutical Sciences Brno, 61242 Brno, Czech Republic.
| | - Lucie Hasoňová
- Department of Food Biotechnologies and Agricultural Products´ Quality, Faculty of Agriculture, University of South Bohemia, 37005 České Budějovice, Czech Republic.
| | - Robert Kala
- Department of Food Biotechnologies and Agricultural Products´ Quality, Faculty of Agriculture, University of South Bohemia, 37005 České Budějovice, Czech Republic.
| |
Collapse
|
182
|
Analysis of gut microbiota diversity and auxiliary diagnosis as a biomarker in patients with schizophrenia: A cross-sectional study. Schizophr Res 2018; 197:470-477. [PMID: 29352709 DOI: 10.1016/j.schres.2018.01.002] [Citation(s) in RCA: 218] [Impact Index Per Article: 31.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 12/26/2017] [Accepted: 01/01/2018] [Indexed: 12/20/2022]
Abstract
With the advent of sequencing technology, characterization of schizophrenia with underlying probing of gut microbiome can provide abundant clues for diagnosis and prognosis of schizophrenia. In this study, we first compared the difference of gut microbiota between schizophrenia patients and healthy controls by 16S rRNA sequencing. We further explored whether gut microbiota can be used as a biomarker to assist in the diagnosis of schizophrenia. We restricted inclusion criteria strictly to control confounding bias. Finally, we investigated differences in fecal microbiota between 64 schizophrenia patients and 53 healthy controls. At the phylum level, we found that the abundance of Proteobacteria in the schizophrenia patients was significantly increased. At the genus level, the relative abundance of Succinivibrio, Megasphaera, Collinsella, Clostridium, Klebsiella and Methanobrevibacter was significantly higher whereas the abundance of Blautia, Coprococcus, Roseburia was decreased compared to health controls. The receiver operating characteristic curve analysis demonstrated that 12 significant microbiota biomarkers were capable of being used as diagnostic factors for distinguishing the schizophrenia cohort from those in the control cohort (AUC = 0.837). We performed PICRUSt analysis and found that several metabolic pathways differed significantly between healthy controls and schizophrenia patients, including vitamin B6 and fatty acid. In conclusion, there are some difference of gut microbiota between schizophrenia patients and healthy controls and the insights from this study could be used to develop microbiota-based diagnosis for schizophrenia.
Collapse
|
183
|
Lee M, Lin W, Wang S, Lin L, Yu B, Lee T. Evaluation of potential antioxidant and anti-inflammatory effects of Antrodia cinnamomea powder and the underlying molecular mechanisms via Nrf2- and NF-κB-dominated pathways in broiler chickens. Poult Sci 2018; 97:2419-2434. [DOI: 10.3382/ps/pey076] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 03/29/2018] [Indexed: 12/20/2022] Open
|
184
|
Bedford A, Gong J. Implications of butyrate and its derivatives for gut health and animal production. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2018; 4:151-159. [PMID: 30140754 PMCID: PMC6104520 DOI: 10.1016/j.aninu.2017.08.010] [Citation(s) in RCA: 258] [Impact Index Per Article: 36.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 08/10/2017] [Indexed: 02/06/2023]
Abstract
Butyrate is produced by microbial fermentation in the large intestine of humans and animals. It serves as not only a primary nutrient that provides energy to colonocytes, but also a cellular mediator regulating multiple functions of gut cells and beyond, including gene expression, cell differentiation, gut tissue development, immune modulation, oxidative stress reduction, and diarrhea control. Although there are a large number of studies in human medicine using butyrate to treat intestinal disease, the importance of butyrate in maintaining gut health has also attracted significant research attention to its application for animal production, particularly as an alternative to in-feed antibiotics. Due to the difficulties of using butyrate in practice (i.e., offensive odor and absorption in the upper gut), different forms of butyrate, such as sodium butyrate and butyrate glycerides, have been developed and examined for their effects on gut health and growth performance across different species. Butyrate and its derivatives generally demonstrate positive effects on animal production, including enhancement of gut development, control of enteric pathogens, reduction of inflammation, improvement of growth performance (including carcass composition), and modulation of gut microbiota. These benefits are more evident in young animals, and variations in the results have been reported. The present article has critically reviewed recent findings in animal research on butyrate and its derivatives in regard to their effects and mechanisms behind and discussed the implications of these findings for improving animal gut health and production. In addition, significant findings of medical research in humans that are relevant to animal production have been cited.
Collapse
|
185
|
Li M, van Esch BCAM, Henricks PAJ, Folkerts G, Garssen J. The Anti-inflammatory Effects of Short Chain Fatty Acids on Lipopolysaccharide- or Tumor Necrosis Factor α-Stimulated Endothelial Cells via Activation of GPR41/43 and Inhibition of HDACs. Front Pharmacol 2018; 9:533. [PMID: 29875665 PMCID: PMC5974203 DOI: 10.3389/fphar.2018.00533] [Citation(s) in RCA: 209] [Impact Index Per Article: 29.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 05/02/2018] [Indexed: 12/22/2022] Open
Abstract
Background and Aim: Previously, we found that short chain fatty acids (SCFA) inhibit LPS or TNFα-induced endothelial inflammatory responses and excessive vascular cell adhesion molecule-1 (VCAM-1) expression, two important steps in the development of atherosclerosis. However, the mechanisms involved are still unclear. We hypothesized that the effects of SCFA are associated with activation of G-protein coupled receptor 41/43 (GPR41/43) and/or inhibition of histone deacetylases (HDACs). Methods: The expression and location of GPR41/43 and HDAC3 in human umbilical vein endothelial cells (HUVEC) were confirmed. HUVEC were pre-incubated with acetate, butyrate or propionate alone or in combination with GLPG0974 (GLPG, antagonist of GPR43) or β-hydroxybutyrate (SHB, antagonist of GPR41) and then exposed to LPS or TNFα. Interleukin (IL)-6 and IL-8 levels and VCAM-1 expression were measured. HDAC activity was measured after treatment with butyrate, propionate and trichostatin A (TSA, HDAC inhibitor). The peripheral blood mononuclear cell (PBMC) adhesive level was also determined after TSA treatment. Results: GPR41/43 were expressed on the membrane of HUVEC and HDAC3 was located in cytoplasm and nucleus. The GLPG and/or SHB treatments restored the inhibitory effects of acetate on IL-6 and IL-8 production and the inhibitory effects of butyrate or propionate on IL-6 production, but not on IL-8. In contrast, GLPG and/or SHB treatments did not affect the inhibitory effects of butyrate or propionate on TNFα-induced VCAM-1 expression. TSA showed similar effects on IL-8 production and VCAM-1 expression as butyrate and propionate. In addition, TSA significantly inhibited the adhesion of PBMC to an endothelial monolayer. Conclusion: Activation of GPR41/43 mediates the effects of acetate on IL-6 and IL-8 production and the effects of butyrate and propionate on IL-6 production. Furthermore, inhibition of HDACs mediates the effects of butyrate and propionate on IL-8 production, VCAM-1 expression, and PBMC adhesion to an endothelial monolayer. These data indicate the beneficial roles of SCFA in preventing vascular inflammation and relevant diseases by activation of GPR41/43 and inhibition of HDACs.
Collapse
Affiliation(s)
- Meng Li
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Betty C A M van Esch
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands.,Nutricia Research, Immunology, Utrecht, Netherlands
| | - Paul A J Henricks
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Gert Folkerts
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands.,Nutricia Research, Immunology, Utrecht, Netherlands
| |
Collapse
|
186
|
Celiberto LS, Graef FA, Healey GR, Bosman ES, Jacobson K, Sly LM, Vallance BA. Inflammatory bowel disease and immunonutrition: novel therapeutic approaches through modulation of diet and the gut microbiome. Immunology 2018; 155:36-52. [PMID: 29693729 DOI: 10.1111/imm.12939] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 03/28/2018] [Accepted: 04/18/2018] [Indexed: 12/11/2022] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory condition of the gastrointestinal tract, thought to at least in part reflect an aberrant immune response to gut bacteria. IBD is increasing in incidence, particularly in populations that have recently immigrated to western countries. This suggests that environmental factors are involved in its pathogenesis. We hypothesize that the increase in IBD rates might reflect the consumption of an unhealthy Western diet, containing excess calories and lacking in key nutritional factors, such as fibre and vitamin D. Several recent studies have determined that dietary factors can dramatically influence the activation of immune cells and the mediators they release through a process called immunonutrition. Moreover, dietary changes can profoundly affect the balance of beneficial versus pathogenic bacteria in the gut. This microbial imbalance can alter levels of microbiota-derived metabolites that in turn can influence innate and adaptive intestinal immune responses. If the diet-gut microbiome disease axis does indeed underpin much of the 'western' influence on the onset and progression of IBD, then tremendous opportunity exists for therapeutic changes in lifestyle, to modulate the gut microbiome and to correct immune imbalances in individuals with IBD. This review highlights four such therapeutic strategies - probiotics, prebiotics, vitamin D and caloric restriction - that have the potential to improve and add to current IBD treatment regimens.
Collapse
Affiliation(s)
- Larissa S Celiberto
- Department of Paediatrics, BC Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Franziska A Graef
- Department of Paediatrics, BC Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Genelle R Healey
- Department of Paediatrics, BC Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Else S Bosman
- Department of Paediatrics, BC Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Kevan Jacobson
- Department of Paediatrics, BC Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Laura M Sly
- Department of Paediatrics, BC Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Bruce A Vallance
- Department of Paediatrics, BC Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
187
|
Bortoluzzi C, Pedroso AA, Mallo JJ, Puyalto M, Kim WK, Applegate TJ. Sodium butyrate improved performance while modulating the cecal microbiota and regulating the expression of intestinal immune-related genes of broiler chickens. Poult Sci 2018; 96:3981-3993. [PMID: 29050425 DOI: 10.3382/ps/pex218] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 08/04/2017] [Indexed: 12/15/2022] Open
Abstract
This study evaluated the effect of sodium butyrate (SB) on performance, expression of immune-related genes in the cecal tonsils, and cecal microbiota of broiler chickens when dietary energy and amino acids concentrations were reduced. Day-old male Ross 708 broiler chicks were fed dietary treatments in a 3 × 2 factorial design (8 pens per treatment) with 3 dietary formulations (control diet; reduction of 2.3% of amino acids and 60 kcal/kg; and reduction of 4.6% of amino acids and 120 kcal/kg) with or without the inclusion of 0.1% of SB. Feed intake (FI), body weight gain (BW gain), and feed conversion ratio (FCR) were recorded until 28 d of age. From 14 to 28 d, there was an interaction of nutrient density by SB (P = 0.003) wherein BW gain of birds fed SB was impaired less by the energy/amino acids reduction than unsupplemented birds. A similar result was obtained from 1 to 28 d (P = 0.004). No interaction (P < 0.05) between nutrient density by SB was observed for FCR. Nutritional density of the diets and SB modified the structure, composition, and predicted function of the cecal microbiota. The nutritionally reduced diet altered the imputed function performed by the microbiota and the SB supplementation reduced these variations, keeping the microbial function similar to that observed in chickens fed a control diet. The frequency of bacterial species presenting the butyryl-CoA: acetate CoA-transferase gene increased in the microbiota of chickens fed a nutritionally reduced diet without SB supplementation, and was not changed by nutrient density of the diet when supplemented with SB (interaction; P = 0.01). SB modulated the expression of immune related genes in the cecal tonsils; wherein SB upregulated the expression of A20 in broilers fed control diets (P < 0.05) and increased IL-6 expression (P < 0.05). These results show that SB had positive effects on the productive performance of broilers fed nutritionally reduced diets, partially by modulating the cecal microbiota and exerting immune-modulatory effects.
Collapse
Affiliation(s)
- C Bortoluzzi
- Department of Animal Science, Purdue University, West Lafayette, IN, 47907.,Department of Poultry Science, University of Georgia, Athens, GA, 30602
| | | | | | | | - W K Kim
- Department of Poultry Science, University of Georgia, Athens, GA, 30602
| | - T J Applegate
- Department of Animal Science, Purdue University, West Lafayette, IN, 47907.,Department of Poultry Science, University of Georgia, Athens, GA, 30602
| |
Collapse
|
188
|
Bernard L, Bonnet M, Delavaud C, Delosière M, Ferlay A, Fougère H, Graulet B. Milk Fat Globule in Ruminant: Major and Minor Compounds, Nutritional Regulation and Differences Among Species. EUR J LIPID SCI TECH 2018. [DOI: 10.1002/ejlt.201700039] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Laurence Bernard
- Université Clermont Auvergne, INRA, VetAgro Sup, UMR Herbivores; F-63122 Saint-Genès-Champanelle France
| | - Muriel Bonnet
- Université Clermont Auvergne, INRA, VetAgro Sup, UMR Herbivores; F-63122 Saint-Genès-Champanelle France
| | - Carole Delavaud
- Université Clermont Auvergne, INRA, VetAgro Sup, UMR Herbivores; F-63122 Saint-Genès-Champanelle France
| | - Mylène Delosière
- Université Clermont Auvergne, INRA, VetAgro Sup, UMR Herbivores; F-63122 Saint-Genès-Champanelle France
| | - Anne Ferlay
- Université Clermont Auvergne, INRA, VetAgro Sup, UMR Herbivores; F-63122 Saint-Genès-Champanelle France
| | - Hélène Fougère
- Université Clermont Auvergne, INRA, VetAgro Sup, UMR Herbivores; F-63122 Saint-Genès-Champanelle France
| | - Benoît Graulet
- Université Clermont Auvergne, INRA, VetAgro Sup, UMR Herbivores; F-63122 Saint-Genès-Champanelle France
| |
Collapse
|
189
|
Khor BH, Narayanan SS, Sahathevan S, Gafor AHA, Daud ZAM, Khosla P, Sabatino A, Fiaccadori E, Chinna K, Karupaiah T. Efficacy of Nutritional Interventions on Inflammatory Markers in Haemodialysis Patients: A Systematic Review and Limited Meta-Analysis. Nutrients 2018; 10:nu10040397. [PMID: 29570616 PMCID: PMC5946182 DOI: 10.3390/nu10040397] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 03/20/2018] [Accepted: 03/21/2018] [Indexed: 12/31/2022] Open
Abstract
Low-grade chronic inflammation is prevalent in patients undergoing haemodialysis (HD) treatment and is linked to the development of premature atherosclerosis and mortality. The non-pharmacological approach to treat inflammation in HD patients through nutritional intervention is well cited. We aimed to assess the efficacy of different nutritional interventions at improving inflammatory outcomes in HD patients, based on markers such as C-reactive protein (CRP), interleukin-6 (IL-6), and tumour necrosis factor-α (TNF-α). We searched PubMed, Cochrane Library, and Embase for randomized controlled trials (RCT) published before June 2017. Inclusion criteria included RCTs on adult patients on maintenance HD treatment with duration of nutritional interventions for a minimum 4 weeks. Risk of bias was assessed using the Jadad score. In total, 46 RCTs experimenting different nutritional interventions were included in the review and categorized into polyphenols rich foods, omega-3 fatty acids, antioxidants, vitamin D, fibres, and probiotics. Meta-analyses indicated significant reduction in CRP levels by omega-3 fatty acids (Random model effect: -0.667 mg/L, p < 0.001) and vitamin E (fixed model effect: -0.257 mg/L, p = 0.005). Evidence for other groups of nutritional interventions was inconclusive. In conclusion, our meta-analysis provided evidence that omega-3 fatty acids and vitamin E could improve inflammatory outcomes in HD patients.
Collapse
Affiliation(s)
- Ban-Hock Khor
- Dietetics Program, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia.
| | | | - Sharmela Sahathevan
- Dietetics Program, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia.
| | - Abdul Halim Abdul Gafor
- Department of Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia.
| | - Zulfitri Azuan Mat Daud
- Department of Nutrition and Dietetics, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor 43400, Malaysia.
| | - Pramod Khosla
- Department of Nutrition & Food Sciences, College of Liberal Arts & Sciences, Wayne State University, Detroit, MI 48202, USA.
| | - Alice Sabatino
- Acute and Chronic Renal Failure Unit, Department of Clinical and Experimental Medicine, University of Parma, 43126 Parma, Italy.
| | - Enrico Fiaccadori
- Acute and Chronic Renal Failure Unit, Department of Clinical and Experimental Medicine, University of Parma, 43126 Parma, Italy.
| | - Karuthan Chinna
- Department of Social and Preventive Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia.
| | - Tilakavati Karupaiah
- Dietetics Program, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia.
- School of BioSciences, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya 47500, Malaysia.
| |
Collapse
|
190
|
Abstract
The gut microbiota comprises a complex community of microorganism species that resides in our gastrointestinal ecosystem and whose alterations influence not only various gut disorders but also central nervous system disorders such as Alzheimer's disease (AD). AD, the most common form of dementia, is a neurodegenerative disorder associated with impaired cognition and cerebral accumulation of amyloid-β peptides (Aβ). Most notably, the microbiota-gut-brain axis is a bidirectional communication system that is not fully understood, but includes neural, immune, endocrine, and metabolic pathways. Studies in germ-free animals and in animals exposed to pathogenic microbial infections, antibiotics, probiotics, or fecal microbiota transplantation suggest a role for the gut microbiota in host cognition or AD-related pathogenesis. The increased permeability of the gut and blood-brain barrier induced by microbiota dysbiosis may mediate or affect AD pathogenesis and other neurodegenerative disorders, especially those associated with aging. In addition, bacteria populating the gut microbiota can secrete large amounts of amyloids and lipopolysaccharides, which might contribute to the modulation of signaling pathways and the production of proinflammatory cytokines associated with the pathogenesis of AD. Moreover, imbalances in the gut microbiota can induce inflammation that is associated with the pathogenesis of obesity, type 2 diabetes mellitus, and AD. The purpose of this review is to summarize and discuss the current findings that may elucidate the role of the gut microbiota in the development of AD. Understanding the underlying mechanisms may provide new insights into novel therapeutic strategies for AD.
Collapse
Affiliation(s)
- Chunmei Jiang
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Institute of Neurology, Department of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Guangning Li
- Department of Neurology, Huadu District People's Hospital, Southern Medical University, Guangzhou, China
| | - Pengru Huang
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Institute of Neurology, Department of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Zhou Liu
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Institute of Neurology, Department of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Bin Zhao
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Institute of Neurology, Department of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
191
|
The effect of Clostridium butyricum on symptoms and fecal microbiota in diarrhea-dominant irritable bowel syndrome: a randomized, double-blind, placebo-controlled trial. Sci Rep 2018; 8:2964. [PMID: 29445178 PMCID: PMC5813237 DOI: 10.1038/s41598-018-21241-z] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 01/29/2018] [Indexed: 02/06/2023] Open
Abstract
Irritable bowel syndrome (IBS) is a common disorder in gastrointestinal system and impairs the quality of life of the patients. Clostridium butyricum (CB) is a probiotics that has been used in several gastrointestinal diseases. The efficacy of CB in treating IBS is still unknown. This prospective, multi-centre, randomized, double-blind, placebo-controlled trial aimed to assess the efficacy and safety of CB in treating diarrhea-predominant IBS (IBS-D) and analyze the fecal microbiota after treatment. Two hundred patients with IBS-D were recruited and were given CB or placebo for 4 weeks. End points included change from baseline in IBS symptoms, quality of life, stool consistency and frequency. Compared with placebo, CB is effective in improving the overall IBS-D symptoms (-62.12 ± 74.00 vs. -40.74 ± 63.67, P = 0.038) as well as quality of life (7.232 ± 14.06 vs. 3.159 ± 11.73, P = 0.032) and stool frequency (-1.602 ± 1.416 vs. -1.086 ± 1.644, P = 0.035). The responder rates are found higher in CB compared with the placebo (44.76% vs. 30.53%, P = 0.042). The change in fecal microbiota was analyzed and function pathways of CB in treating IBS-D were predicted. In conclusion, CB improves overall symptoms, quality of life and stool frequency in IBS-D patients and is considered to be used as a probiotics in treating IBS-D clinically.
Collapse
|
192
|
|
193
|
Stanley D, Moore RJ, Wong CHY. An insight into intestinal mucosal microbiota disruption after stroke. Sci Rep 2018; 8:568. [PMID: 29330443 PMCID: PMC5766598 DOI: 10.1038/s41598-017-18904-8] [Citation(s) in RCA: 126] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 12/19/2017] [Indexed: 12/12/2022] Open
Abstract
Recent work from our laboratory has provided evidence that indicates selective bacterial translocation from the host gut microbiota to peripheral tissues (i.e. lung) plays a key role in the development of post-stroke infections. Despite this, it is currently unknown whether mucosal bacteria that live on and interact closely with the host intestinal epithelium contribute in regulating bacterial translocation after stroke. Here, we found that the microbial communities within the mucosa of gastrointestinal tract (GIT) were significantly different between sham-operated and post-stroke mice at 24 h following surgery. The differences in microbiota composition were substantial in all sections of the GIT and were significant, even at the phylum level. The main characteristics of the stroke-induced shift in mucosal microbiota composition were an increased abundance of Akkermansia muciniphila and an excessive abundance of clostridial species. Furthermore, we analysed the predicted functional potential of the altered mucosal microbiota induced by stroke using PICRUSt and revealed significant increases in functions associated with infectious diseases, membrane transport and xenobiotic degradation. Our findings revealed stroke induces far-reaching and robust changes to the intestinal mucosal microbiota. A better understanding of the precise molecular events leading up to stroke-induced mucosal microbiota changes may represent novel therapy targets to improve patient outcomes.
Collapse
Affiliation(s)
- Dragana Stanley
- School of Health Medical and Applied Sciences, Central Queensland University, Bruce Highway, Rockhampton, Queensland, 4702, Australia
| | - Robert J Moore
- School of Science, RMIT University, Bundoora, Victoria, 3083, Australia.,Infection and Immunity Program, Biomedicine Discovery Institute, Department of Microbiology, Monash University, Clayton, Victoria, 3800, Australia
| | - Connie H Y Wong
- Centre for Inflammatory Diseases, Department of Medicine, School of Clinical Sciences, Monash University, Clayton, Victoria, 3168, Australia.
| |
Collapse
|
194
|
Liu Y, Upadhyaya B, Fardin-Kia AR, Juenemann RM, Dey M. Dietary resistant starch type 4-derived butyrate attenuates nuclear factor-kappa-B1 through modulation of histone H3 trimethylation at lysine 27. Food Funct 2018; 7:3772-3781. [PMID: 27713965 DOI: 10.1039/c6fo00856a] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Indigestible resistant starches (RS) are substrates for gut-microbial metabolism and have been shown to attenuate intestinal inflammation but the supporting evidence is inconsistent and lacks mechanistic explanation. We have recently reported dietary RS type 4 (RS4) induced improvements in immunometabolic functions in humans and a concomitant increase in butyrogenic gut-bacteria. Since inflammation is a key component in metabolic diseases, here we investigated the effects of RS4-derived butyrate on the epigenetic repression of pro-inflammatory genes in vivo and in vitro. RS4-fed mice, compared to the control-diet group, had higher cecal butyrate and increased tri-methylation of lysine 27 on histone 3 (H3K27me3) in the promoter of nuclear factor-kappa-B1 (NFκB1) in the colon tissue. The H3K27me3-enrichment inversely correlated with the concentration dependent down-regulation of NFκB1 in sodium butyrate treated human colon epithelial cells. Two additional inflammatory genes were attenuated by sodium butyrate, but were not linked with H3K27me3 changes. This exploratory study presents a new opportunity for studying underlying H3K27me3 and other methylation modifying mechanisms linked to RS4 biological activity.
Collapse
Affiliation(s)
- Yi Liu
- Department of Health and Nutritional Sciences, Box 2203, South Dakota State University, Brookings, SD 57007, USA.
| | - Bijaya Upadhyaya
- Department of Health and Nutritional Sciences, Box 2203, South Dakota State University, Brookings, SD 57007, USA.
| | - Ali Reza Fardin-Kia
- Office of Regulatory Science, Center for Food Safety and Applied Nutrition, HFS-717, US Food and Drug Administration, College Park, MD 20740, USA
| | - Robert M Juenemann
- Department of Health and Nutritional Sciences, Box 2203, South Dakota State University, Brookings, SD 57007, USA.
| | - Moul Dey
- Department of Health and Nutritional Sciences, Box 2203, South Dakota State University, Brookings, SD 57007, USA.
| |
Collapse
|
195
|
Abstract
The gastrointestinal (GI) tract is the residence of trillions of microorganisms that include bacteria, archaea, fungi and viruses. The collective genomes of whole microbial communities (microbiota) integrate the gut microbiome. Up to 100 genera and 1000 distinct bacterial species were identified in digestive tube niches. Gut microbiomes exert permanent pivotal functions by promoting food digestion, xenobiotic metabolism and regulation of innate and adaptive immunological processes. Proteins, peptides and metabolites released locally and at distant sites trigger many cell signalling and pathways. This intense crosstalk maintains the host-microbial homeostasis. Diet, age, diet, stress and diseases cause increases or decreases in relative abundance and diversity bacterial specie of GI and other body sites. Studies in animal models and humans have shown that a persistent imbalance of gut's microbial community, named dysbiosis, relates to inflammatory bowel diseases (IBD), irritable bowel syndrome (IBS), diabetes, obesity, cancer, cardiovascular and central nervous system disorders. Notably specific bacterial communities are promising clinical target to treat inflammatory and infectious diseases. In this context, intestinal microbiota transplantation (IMT) is one optional treatment for IBD, in particular to patients with recurrent Clostridium difficile-induced pseudo-membrane colitis. Here we discuss on recent discoveries linking whole gut microbiome dysbiosis to metabolic and inflammatory diseases and potential prophylactic and therapeutic applications of faecal and phage therapy, probiotic and prebiotic diets.
Collapse
Affiliation(s)
- José E Belizário
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.
| | - Joel Faintuch
- Department of Gastroenterology of Medical School, University of Sao Paulo, São Paulo, Brazil
| |
Collapse
|
196
|
Liu H, Wang J, He T, Becker S, Zhang G, Li D, Ma X. Butyrate: A Double-Edged Sword for Health? Adv Nutr 2018; 9:21-29. [PMID: 29438462 PMCID: PMC6333934 DOI: 10.1093/advances/nmx009] [Citation(s) in RCA: 686] [Impact Index Per Article: 98.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Accepted: 11/11/2017] [Indexed: 02/06/2023] Open
Abstract
Butyrate, a four-carbon short-chain fatty acid, is produced through microbial fermentation of dietary fibers in the lower intestinal tract. Endogenous butyrate production, delivery, and absorption by colonocytes have been well documented. Butyrate exerts its functions by acting as a histone deacetylase (HDAC) inhibitor or signaling through several G protein-coupled receptors (GPCRs). Recently, butyrate has received particular attention for its beneficial effects on intestinal homeostasis and energy metabolism. With anti-inflammatory properties, butyrate enhances intestinal barrier function and mucosal immunity. However, the role of butyrate in obesity remains controversial. Growing evidence has highlighted the impact of butyrate on the gut-brain axis. In this review, we summarize the present knowledge on the properties of butyrate, especially its potential effects and mechanisms involved in intestinal health and obesity.
Collapse
Affiliation(s)
- Hu Liu
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China
| | - Ji Wang
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China
| | - Ting He
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China
| | - Sage Becker
- Department of Animal Science, Oklahoma State University, Stillwater, OK; Departments of Internal Medicine and Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX
| | - Guolong Zhang
- Department of Animal Science, Oklahoma State University, Stillwater, OK; Departments of Internal Medicine and Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX
| | - Defa Li
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China
| | - Xi Ma
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China,Internal Medicine and Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX,Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX,Address correspondence to XM (e-mail: )
| |
Collapse
|
197
|
NAKASHIMA A, YAMADA K, IWATA O, SUGIMOTO R, ATSUJI K, OGAWA T, ISHIBASHI-OHGO N, SUZUKI K. β-Glucan in Foods and Its Physiological Functions. J Nutr Sci Vitaminol (Tokyo) 2018; 64:8-17. [DOI: 10.3177/jnsv.64.8] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
198
|
Kwon G, Lee J, Koh JH, Lim YH. Lifespan Extension of Caenorhabditis elegans by Butyricicoccus pullicaecorum and Megasphaera elsdenii with Probiotic Potential. Curr Microbiol 2017; 75:557-564. [PMID: 29222621 DOI: 10.1007/s00284-017-1416-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 12/06/2017] [Indexed: 11/27/2022]
Abstract
Butyricicoccus pullicaecorum and Megasphaera elsdenii inhabit the human intestine and have probiotic potential. The aim of this study was to evaluate the effects of B. pullicaecorum and M. elsdenii on the lifespan of Caenorhabditis elegans. They significantly (P < 0.05) extended the lifespan of C. elegans compared with Escherichia coli OP50, a standard food for the worm. Analysis of age-related biomarkers such as lipofuscin, body size, and locomotory activity showed that they retarded aging. They all failed to extend the lifespan of daf-12 or dbl-1 loss-of-function C. elegans mutants compared with E. coli OP50-fed worms. However, the increase in lifespan was observed in daf-16, jnk-1, pmk-1, and skn-1 mutants. Moreover, they increased the resistance of C. elegans to a human pathogen, Salmonella typhimurium. In conclusion, B. pullicaecorum and M. elsdenii extend the lifespan of C. elegans via the transforming growth factor-beta (TGF-β) pathway associated with anti-inflammatory processes in the innate immune system.
Collapse
Affiliation(s)
- Gayeung Kwon
- Department of Public Health Science (Brain Korea 21 PLUS Program), Graduate School, Korea University, Seoul, 136-701, Republic of Korea
| | - Jiyun Lee
- Department of Public Health Science (Brain Korea 21 PLUS Program), Graduate School, Korea University, Seoul, 136-701, Republic of Korea
| | - Jong-Ho Koh
- Department of Bio-Food Analysis and Processing, Bio-Campus Korea Polytechnic College, Nonsan, Chungnam, 32943, Republic of Korea
| | - Young-Hee Lim
- Department of Public Health Science (Brain Korea 21 PLUS Program), Graduate School, Korea University, Seoul, 136-701, Republic of Korea.
- School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul, 136-701, Republic of Korea.
- Department of Laboratory Medicine, Guro Hospital, Korea University, Seoul, 152-703, Republic of Korea.
| |
Collapse
|
199
|
Wu SB, Rodgers NJ, Cui G, Sun Y, Choct M. Dynamics of intestinal metabolites and morphology in response to necrotic enteritis challenge in broiler chickens. Avian Pathol 2017; 45:346-56. [PMID: 27245303 DOI: 10.1080/03079457.2016.1140896] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Despite the relatively small contribution to metabolizable energy that volatile fatty acids (VFAs) provide in chickens, these organic acids have been reported to play beneficial roles in the gastrointestinal tract (GIT) of birds, for example, inhibition of the growth of some pathogenic bacteria. However, information regarding the dynamics of these metabolites in the GIT of chickens is still scarce, especially under disease conditions such as necrotic enteritis (NE). Here, we investigated the dynamics of VFAs and lactic acid, and intestinal morphology in response to NE predisposing factors, that is, excessive dietary fishmeal and Eimeria inoculation, and causative agent Clostridium perfringens producing NetB toxin. The experiment was designed in a 2 × 2 × 2 factorial arrangement of treatments with or without: fishmeal feeding, Eimeria inoculation and C. perfringens challenge. The results showed that these factors significantly influenced composition and concentration of VFAs and lactic acids, pH and histomorphometry in one way or another. These changes may be important for the onset of NE or only the synergetic responses to micro environmental stress. Eimeria appeared to be more important than fishmeal in predisposing birds to NE, thus the application of Eimeria in NE challenge provides more consistent success in inducing the disease. The metabolic responses to various adverse factors such as excessive dietary fishmeal and Eimeria infection are complex. Thus, intensive efforts are required to better understand NE so as to achieve the control of the disease in the absence of antibiotics.
Collapse
Affiliation(s)
- Shu-Biao Wu
- a School of Environmental and Rural Science , University of New England , Armidale , NSW , Australia.,b Poultry Cooperative Research Centre , University of New England , Armidale , NSW , Australia
| | - Nicholas J Rodgers
- a School of Environmental and Rural Science , University of New England , Armidale , NSW , Australia
| | - Guimei Cui
- c Biotechnology Research Centre , Shanxi Academy of Agricultural Sciences , Taiyuan , People's Republic of China
| | - Yi Sun
- c Biotechnology Research Centre , Shanxi Academy of Agricultural Sciences , Taiyuan , People's Republic of China
| | - Mingan Choct
- a School of Environmental and Rural Science , University of New England , Armidale , NSW , Australia.,b Poultry Cooperative Research Centre , University of New England , Armidale , NSW , Australia
| |
Collapse
|
200
|
do Nascimento GE, Winnischofer SMB, Ramirez MI, Iacomini M, Cordeiro LMC. The influence of sweet pepper pectin structural characteristics on cytokine secretion by THP-1 macrophages. Food Res Int 2017; 102:588-594. [DOI: 10.1016/j.foodres.2017.09.037] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 08/30/2017] [Accepted: 09/11/2017] [Indexed: 10/18/2022]
|