Insighter Secures Fami-QS Certification

Insighter, one of the world’s leading manufacturers of natural growth promoters (NGPs) for animals, has been granted FAMI-QS certification for the production of Calcium Butyrate (Buta-ER), Benzoic Acid (Superstin), Colistizer, Tanrpo, Gutpro, Stab-CIM, etc., at its Nanxiong production site in Guangdong. The certification attests that Insighter has implemented and maintains a Feed Safety Management System including Good Manufacturing Practice (GMP) in compliance with the FAMI-QS code. The certification is further evidence Insighter’s commitment to developing quality and sustainable Solutions to Gut Problems.

FAMI-QS: the quality and safety system of reference for specialty feed ingredients and their mixtures

FAMI-QS is the internationally-recognized certification system for Specialty feed ingredients, the primary objective of which is to ensure compliance of certified companies with the EU’s most stringent feed regulations, and ultimately protect consumers’ health. Recognized by the European Commission, the stringent certification process covers the complete lifecycle of processed sensory additives, functional feed ingredients and premixtures  – from raw materials, processing, transportation and storage by the manufacturer to sale of the finished product. It also requires implementation of a procedure to handle customer complaints.

Certification is valid for three years and can be renewed on the basis of a new full audit or continual assessment. This proactive, systematic approach guarantees that manufacturers integrate best practices into their core activities.

“We are very proud to have completed the FAMI-QS certification process with flying colours,” said Dr. Peng Xianfeng, CEO. “As a trailblazer in NPGs, it is our responsibility to lead by example and this certification confirms Insighter’s commitment to ensuring that Quality, Health, Environmental and Safety concerns are an integral part of the way we conduct business.”

Besides FAMI-QS, Insigher’s Nanxiong Site had already secured several certifications for Quality, Safety and the Environment with, for example, the ISO 9001 (Quality Management System), ISO 22000 (Food Safety Management System) certifications. The FAMI-QS certification means that its site can now also be proud to have attained this internationally-valid recognition of their Feed Safety Management System.

饲料行业信息网对话福州饲料工业展报道广州英赛特彭险峰总经理

http://www.feedtrade.com.cn/news/review/2017-04-21/2028459.html

饲料行业信息网对话福州饲料工业展

http://news.feedtrade.com.cn/ 2017-04-21 11:18:21 中国饲料行业信息网

2017年4月18日,由中国饲料工业协会、全国畜牧总站主办的2017中国饲料工业展览会在“榕城”福州海峡国际会展中心隆重开幕。本届展会以“转型升级调结构,创新发展铸品牌”为主题,充分体现饲料工业连接种植养殖上下游、延长产业链的中枢和纽带功能,吸引了近20个国家和地区以及国内400多家饲料加工、饲料添加剂、饲料原料、饲料机械、畜禽养殖等企业参展,展会规模再创历史新高。

开幕式1_副本.jpg
开幕式

主持人:全国畜牧总站站长杨振海

出席嘉宾:农业部于康震副部长,原农业部副部长高鸿宾、中国工程院院士李德发、农业部畜牧业司孔亮副司长、中国饲料工业协会刘连贵副秘书长、福州市杭东副市长、金卫东、蔡辉益、陈少美等

开幕式.jpg

福建省2016年肉蛋奶总产量270万吨,生猪出栏1720万头,生猪、奶牛、肉鸡、蛋鸡规模化率均达到80%以上,全省饲料及添加剂企业364家,总产量883万吨,位居全国第11位。

开幕式后,农业部不副部长于康震一行对饲料工业展上福建天马、新希望六和、中粮、正邦等企业展会进行了视察。

于康震视察展位.jpg
于康震副部长一行视察福建天马、新希望六和、中粮、正邦、农信互联等

大企业.jpg
大型企业展位

 走进客户、对话企业:

……

八、广州英赛特:彭险峰总经理

DSC_9102_副本.jpg
DSC_9110_副本.jpg

1、 突破我们对于单宁酸是抗营养因子的传统认识,它还有除此之外的一些重要功能:

(1)肠道收敛控制腹泻(2)中和细菌肠毒素(3)抗肠道细菌及病毒(4)驱除蛔虫等肠道线虫(5)除臭(6)替代所有饲用抗生素,提高生产性能,促生长

单宁酸有两种产品:

强粘素:游离的单宁酸,主要用于禽料

保单:单宁的终极产品,是一种盐蛋白,主要用于猪料,适口性好

2、 苯甲酸:绿色新型肠道广谱抗菌剂

(1)完全无刺激性气味,不升华挥发(2)适口性好,无苯甲酸盐的苦涩味,不降低采食量(3)微丸制剂,不起拱,分散均匀、流散性好(4)食品级纯化工艺,原料纯度>99.95%

(5)广谱抗菌(6)肠溶制剂、肠道靶向释放,生物利用度<10%

……

Is “Enteric” Necessary for Butyrate? 丁酸盐所谓的“肠溶”是个伪命题!

The following article is from the internet and for your kind reference and comments:

外界对丁酸盐原粉存在两种误解:
1)丁酸盐如果没用脂肪包被的话过不了胃部!
2)由于后肠道是碱性环境,所以丁酸易被解离, 以阴离子形式存在, 而丁酸根阴离子是没有效果的,因无法被吸收,也无杀菌能力!
他们的理由是:
1)丁酸盐在胃部被溶解,而丁酸透过胃壁被吸收掉了。
2)有机酸在不同酸碱值下被解离的情形如下:

解离/非解离
有机酸种类 pKa pH 4 pH 6 pH 7
甲酸 3.75 1.8 : 1 178 : 1 1778 : 1
乳酸 3.83 1.5 : 1 148 : 1 1479 : 1
苯甲酸 4.19 0.6 : 1 65 : 1 646 : 1
丁酸 4.86 0.1 : 1 15 : 1 138 : 1
丙酸 4.88 0.1 : 1 13 : 1 132 : 1

酸性越强,丁酸越不易解离,大部分以完整丁酸形式存在 (解离/非解离 = 0.1 / 1)。
碱性越强,丁酸越易解离,大部分以丁酸根阴离子形式存在 (解离/非解离 = 138 / 1)。
而在后肠道中碱性较强 (pH > 7),丁酸根阴离子 / 丁酸比例会大于 138 / 1.  所以他们宣称丁酸盐到达不了后肠道。
事实如此吗?
事实1:
早在1990年Galfi and Bokori 添加0.17%丁酸盐原粉于日粮中,喂给从7 – 102公斤的猪只,回肠绒毛长度增长及盲肠隐窝深度加深。
事实2:
2006年Manzanilla等人指出添加0.3%丁酸盐原粉可增加空肠绒毛长度及加深隐窝深度。回肠隐窝深度也加深了。结肠的杯形细胞数目也增加了。
以上2个事实证明丁酸盐原粉是可过胃,也可到达前后肠道。
那丁酸盐原粉是如何过胃到达前肠道及后肠道呢?
A、丁酸盐在胃部解离释放出钙或钠阳离子及丁酸根阴离子。胃部是酸性环境富含氢离子(H+)。氢离子又与丁酸根阴离子结合成为完整的丁酸被运送到肠道。

B
1、丁酸以自动非离子扩散模式被吸收:
多年以来人们一直以为短链脂肪酸在结肠吸收是只以非离子扩散(non-ionic diffusion)的方式进入上皮细胞。
2、丁酸根阴离子以碳酸氢根离子转换的方式被吸收:
其实丁酸根阴离子还可在上皮细胞顶膜(apical membrane)及基底膜(basolateral membrane)透过HCO3 交换(碳酸氢根离子转换的方式) 进入上皮细胞, 达到丁酸吸收的目的,此过程在结肠膜内的空泡(vesicle)中被证实。
3、丁酸根阴离子也以其他短链脂肪酸浓度的增加的方式被吸收:
Charney等人于1998年在远端结肠中更发现, 纵然在碱性的环境下(pH 7.4),其他短链脂肪酸浓度的提高也有助丁酸根阴离子吸收,进入结肠上皮细胞内。并强调这种机制是不受酸碱值影响,也就是说短链脂肪酸浓度越高, 则丁酸根阴离子吸收度越高。

C、Smith, D. J. 等人于2012年(J. Agric. Food Chem. 60:3151-3157)以肉小鸡做试验,用碳14标定丁酸钙原粉中的丁酸,发现原粉丁酸钙在盲肠、结肠及泄殖腔释放的量在进食后2, 4,8及12小时分别为0.3%,0.2%,0.1% 及0.4%。 脂肪保护的丁酸钙在盲肠、 结肠及泄殖腔释放的量在进食后2, 4, 8及12小时分别为0.8%,0.4%, 0.2%及0.4%。
以上打破原粉到达不了后肠道的谣言!!

Butyrate: Short-chain fatty acids Absorption

Short-chain fatty acids and medium-chain fatty acids are primarily absorbed through the portal vein during lipid digestion,[6] while long-chain fatty acids are packed into chylomicrons and enter lymphatic capillaries, and enter the blood first at the subclavian vein.

https://en.wikipedia.org/wiki/Short-chain_fatty_acid

Kuksis, Arnis (2000). “Biochemistry of Glycerolipids and Formation of Chylomicrons”. In Christophe, Armand B.; DeVriese, Stephanie. Fat Digestion and Absorption. The American Oil Chemists Society. p. 163. ISBN 189399712X. Retrieved December 21, 2012.

Butyrate: Feeding the Gut and Beyond for Animal Health

James Pierce, MS, PhD; Nutriad Inc. Elgin, IL, USA – Maja Marien, DVM, PhD; Tim Goossens, PhD; Nutriad International NV Dendermonde, Belgium

Introduction

In North America, there has been a tremendous body of research in recent years to find the ideal product or program to replace or reduce the use of antibiotics in livestock and poultry production. This is driven by the legislation in the EU and the pressure the health-care system has placed on the use of sub-therapeutic levels of antibiotics for growth promotion. There have been many arguments made regarding the validity of such claims, but ultimately, the consumer will decide what they are willing to accept and pay for accordingly.

With the projected increase in the human population as well as the rise in income over the next 20 years, especially in BRIC countries, there will be a concomitant increase in demand for animal products. The demands to increase the safety and efficiency of food production has become a major focus globally. In recent years, many molecules and additives have been evaluated for their ability to improve animal health, growth, efficiency, and reproduction. The products evaluated have included: prebiotics, probiotics, botanical products, acids (inorganic, organic, short and medium chain fatty acids), chelated minerals, and enzymes.

In order to evaluate these compounds, one should aim at understanding as much as possible about their safety, efficacy, and mode of action. If we are primarily looking to replace antibiotic growth promoters, we should first evaluate the effect on digestion and gut health. The intestine is the largest organ in the body and thus involves a complex system in which several factors influence the final outcome. In the intestine, three major components (the mucosal barrier, the composition of the microbiota and the local immune system) provide defensive measures against different pathogens through permanent contact and communication with each other. Feed additives may interact with host cells (intestinal cells, immune cells), with the host’s microbiota, or with pathogens impairing the normal intestinal function.

Butyrate – Proposed modes of action

Butyrate is somewhat unique among its biological functions when compared with other short chain fatty acids (SCFA). The (SCFA) constitute a group of molecules that contain from one to seven carbon atoms and which exist as straight or branched-chain compounds: predominant SCFAs are acetic, propionic and butyric acid. Because SCFA are weak acids with a pK of ≤4.8 and the pH of the gastrointestinal tract (GIT) is nearly neutral, 90-99% of the SCFA are present in the GIT as anions rather than free acids.

Among the SCFA, butyric acid has received particular attention. Butyric acid is available in the salt form of Na, K, Mg or Ca. The advantage of salts over free acids is that they are generally odorless and easier to handle in the feed manufacturing process owing to their solid and less volatile form. For the purpose of the present article, the term ‘butyrate’ is used interchangeably for the acid, the salt and the anion forms.

Article-Butyrate-feeding the gut and beyond for animal health FIG1

Fig  1. Absorption of n-butyrate in the large intestine and subsequent metabolism. Butyrate transport with monocarboxylate transporters (MCT) are saturable and coupled with H+ transport. Butyrate is recognized by G-protein receptors (GPR41, GPR43). (Adapted from Guiloteau et al., 2010).

There are several potential fates of butyrate once consumed by the animal. Butyrate may function as a ligand for transmembrane receptors, as a modulator of gene activity, and as a direct energy source for cellular metabolism via B-oxidation. Several cell types, many of which associated with the digestive tract, have been described to be receptive to one or more of these biological effector functions, which explains the extensive range of biochemical effects that have been documented to be mediated by butyrate (Fig 1).

When butyrate is present in the blood stream or in the proximal parts of the intestinal tract, it induces the production of host defense peptides (Guilloteau et al., 2009). These peptides stimulate the development and repair of the intestinal tract through an increase in cell proliferation (Bartholome et al., 2004). Recently it has been shown that butyrate, when present in blood, stimulates a peptide that increases the absorption of glucose from the intestine. Indications that a similar mode of action can be expected in poultry, is shown by Hu and Guo (2007), who found an increased development of the villi when sodium-butyrate was added to the diet.

Butyrate also has been shown to stimulate several functions in the lower part of the intestinal tract. Studies have identified specific G-protein-coupled receptors, specifically GPR 41 and GPR 43, on gut epithelial cells in the epithelium of particularly the ileum, caeca and colon (Le Poul et al., 2003). When butyrate is attached to these receptors the production of several different peptides is stimulated (Cox et al., 2009, Tazoe et al., 2008). Some of these peptides have a positive effect on the development of the immune system and improve the functioning of the immune system in case of a health challenge (Cox et al., 2009). Other peptides have been shown to optimize gut motility, by reducing the rate of feed passage (Tazoe et al., 2008). In poultry, the emptying of the feed out of the gizzard into the small intestine is slowed down. Thus, it seems that butyrate is inducing a similar effect to passage rate as coarse particles such as oyster shell.

Indications that butyrate also stimulates the immune system in poultry were obtained by Leeson et al. (2005) in that, birds previously fed butyrate showed more ability to withstand against the stress of coccidial challenge at 21 d of age. Weber (2008) found when pigs where challenged with Escherichia coli lipopolysaccharide (LPS), sodium butyrate increased the magnitude of the cortisol response and increased skeletal muscle IL-6 mRNA expression, also indicating that dietary butyrate affects the response to inflammatory stimuli.

In summary, beneficial effects include, among others, stimulation of digestive enzyme production, enhanced development of intestinal villi, reduction of acute inflammatory responses, increased GIT retention time, inhibition of cancer cell growth and the secretion of host defense peptides (Guilloteau et al., 2012). Apart from effects in eukaryotic host cells, butyrate is also described to have an impact on the activity of prokaryotes residing in the animal’s GIT. For example, it has been shown to affect the colonization of Salmonella and Campylobacter and to influence the composition of the gut microbiota (see below).

Butyrate – Experimental results

Mucosal barrier

Dietary supplementation of butyrate has been shown to support enteric development and intestinal health of neonatal animals. At weaning, the small intestine of the piglet generally undergoes a decreased capacity of absorption that is associated with a marked reduction in villous height and crypt depth. These changes are accompanied with a decreased feed intake and poor growth (Piva et al., 2002; Pluske et al., 1996). Butyrate stimulates epithelial cell proliferation resulting in a larger absorptive surface, leading to improved feed utilization. Furthermore, butyrate in the weaner diet preserves villus length and thereby helps to maintain feed intake. Effect of butyrate on gut morphology is of great biological value to the weaning period when the weight of the small and large intestine increases three times faster than that of the whole body mass.

The following trial results (Ferket et al., 2010) were obtained in broilers, but do show the principle of the impact of butyrate supplementation on intestinal development and growth performance in young animals. Commercial broilers were randomly assigned to 32 floor pens containing 30 birds each and provided feed and water ad libitum until 49 days. Starter feed (pellet-crumbled) treatments consisting of 3 dietary supplementation levels of coated butyrate (0.0, 0.015, 0.03, and 0.06% butyrate) were subjected to 8 replicate pens per treatment from 1 – 14 days. Subsequently, all birds were fed common grower and finisher diets in pelleted form. Body weight (BW) and feed intake was determined at 7, 14, 21, 42, and 49 days and feed/gain (FCR) calculated. At 3, 8, and 14 days, 4 birds/treatment were sampled for gut histology evaluation. BW at 14 days increased linearly (p<0.01) as the level of butyrate increased (457 g vs. 470 g for 0 vs. 0.06% butyrate), but no effects on 1-14 days FCR were observed. Histomorphometic analysis correlated with early treatment effects on BW (see Fig 2A and 2B). The positive starter feed treatment effects was observed throughout the experiment, with 0.015% butyrate resulting in a 3% and 2% improvement in 42 days (p<0.02) and 49 days (p<0.10), respectively. A linear improvement in 1-42 days FCR by up to 3% was also observed as the level of butyrate increased in the starter feeds. Dietary supplementation of coated butyrate in starter feeds showed to have a lasting positive effect on broiler growth performance.

Article Butyrate feeding the gut-FIG2b               Article Butyrate feeding the gut-FIG2b

Fig. 2A Broiler chicks, 7 days, negative control                  Fig. 2B Broiler chicks, 7 days, 0.06% butyrate

 

Modulation of microbiota and impact on pathogens

Studies done by Galfi and coworkers (Galfi et al., 1991) have shown that butyrate increases the number of intestinal lactic acid and lactobacilli in butyrate-fed pigs, while decreasing the number of coliforms and E. coli. In his PhD-thesis Pérez Gutiérrez (2010) investigated microbial composition after including different additives in the feed of (weaning) pigs. The qPCR-data revealed an increase in lactobacilli in the butyrate group, while weaning piglets that received butyrate also had a more homogeneous microbial profile, which was regarded as a positive effect.

In studies of Van Immerseel and co-workers it was shown that butyrate, when present in the intestinal tract, was able to decrease Salmonella colonisation in broilers (Van Immerseel et al., 2005). The mode of action of this activity of butyrate seems at least partially mediated through modulation of gene expression. Butyrate specifically down-regulates Salmonella Pathogenicity Island 1 (SPI-1) gene expression, hereby preventing invasion of intestinal epithelial cells, one of the important steps of Salmonella pathogenesis in the bird (Gantois et al., 2006). The importance of an effective coating in order to get significant reduction in Salmonella colonisation in the ceca and internal organs in vivo was also shown in Fig 3. (Van Immerseel et al., 2005).

Article Butyrate feeding the gut-FIG3

Fig. 3: Percentage of animals with a certain amount of Salmonella in caecum (n=25 per group).

A trial done at the University of Bologna (Bosi et al., 2010) was done to look at the possible impact of butyrate (coated and uncoated) on E. coli K88 (ETEC) infection in piglets. Fifty-four piglets (prone to ETEC intestinal adhesion) weaned at 21-28 days were used (three control groups of six animals, three challenged groups of 12 animals, balanced for litter and weight). Experimental diets were obtained with the addition of free or fat coated sodium butyrate. Full factorial design of two factors (four diets x challenge (E. coli K88) – yes/no), so groups were: 1) control diet, unchallenged; 2) 2 kg/T uncoated butyrate, unchallenged; 3) 2 kg/T coated butyrate, unchallenged; 4) control diet, challenged; 5) 2 kg/T uncoated butyrate, challenged; 6) 2 kg/T coated butyrate, challenged. The challenge with ETEC increased mortality in the control group (15% mortality) and had an impact on the growth rate of the piglets. The piglets receiving the uncoated sodium butyrate showed a lower mortality rate (5%) and were to a lesser extent influenced by growth than the challenged control group. In the group offered the coated sodium butyrate in the diet, none of the piglets died and the growth rate was only marginally influenced.

 

Butyrate – More than just an active compound

The effects on immune development, gut motility as well as the Salmonella effect via gene expression are only possible when the butyrate arrives in the lower parts of the intestinal tract. It is generally accepted that unprotected butyrate is quickly absorbed in the proximal part of the intestinal tract. Therefore, in order to get butyrate in significant levels available in the lower part of the intestinal tract the butyrate needs to be protected to achieve a target release.

In recent research done at the University of Illinois (Stein et al, 2012) the importance of a good coating was again demonstrated. The researchers investigated the disappearance kinetics of different sources of butyrate in diets fed to weanling pigs. Weanling pigs (n = 24; 8.0 ± 0.5 kg BW) were randomly allotted to 3 dietary treatments (6 replicate pigs per dietary treatment): 1) a control diet, 2) the control diet + 4kg/T of an uncoated butyrate (50% product, UCB), 3) the control diet + 4kg/T of a coated butyrate (30% product, CB). The dietary treatments were provided to pigs daily for 7 d as 3 times the estimated energy requirement for maintenance. On the last day of the experiment, all pigs were euthanized to collect samples of contents in the stomach, duodenum, jejunum, ileum, cecum, and proximal and distal colon. Concentrations of dry matter (DM) and butyrate were analyzed in all samples. There was a similar pattern of the concentration of butyrate in the digestive tract, indicating that the concentration of butyrate was greater in the stomach than in the duodenum and jejunum, and gradually increased in the ileum. Weanling pigs fed the CB diet had greater (P < 0.05) concentrations of butyrate in the jejunum (190 % μg butyrate/g digesta DM) and ileum (188% μg butyrate/g digesta DM) than weanling pigs fed the control diet (µg butyrate/g digesta DM put as 100%). In the cecum and colon, endogenous production was more variable among piglets and therefore no significant differences could be demonstrated. In conclusion, the coated butyrate in spite of lower percentage of butyrate (30%), increased significantly the concentrations of butyrate in the lower intestinal contents of pigs. Supplementation of the more concentrated (50%) but less protected butyrate did not result in significant differences in the amount of butyrate in the intestinal tract compared with the control group.

Article Butyrate feeding the gut-FIG4

Fig. 4: Amount of butyrate in % compared to the unsupplemented control group.

Butyrate is a molecule of great interest to both human and veterinary medicine. Owing partly to the interest from human medicine, more and more profound knowledge on the mode of action has been obtained which allows for more sound advices to producers. In the current paper an attempt is made to summarize some of the newest developments on the research as well as to show experimental results in production animals; because, in the end the benefits to the efficiency of animal production are what matter. As shown above (Fig 4), butyrate has an effect on several levels (mucosal barrier, feed passage, microbiota, immune system, pathogens and others) and this combination of effects contribute to its general acceptance as helping for improved health as well as for improved performance.

References

Bartholome, A.L.,D.M. Albin, DH Baker, J.J. Holst, and K.A. Tappenden. 2004. Supplementation of total parental nutrition with butyrate increases structural aspects of intestinal adaptation after an 80% jejunoileal resection in neonatal piglets. JPEN. 28(4): 210-222.

Cox, M.A., J. Jackson, M. Stanton, A. Rojas-Trian, L. Bober, M. Laverty, X. Yang,  F. Zhu, J. Liu, S. Wang, F. Monsma, G. Vassileva, M. Maguire, E. Gustafson, M. Bayne, C. Chou, D. Lundell, and C. Jenh. 2009 Short-chain fatty acids act as anti-inflammatory mediators by regulating prostaglandin E2 and cytokines. China Natl. J. New Gastroenterol. 15(44): 5549-5557.

Galf, P., S. Neogradi. and T. Sakata. 1991. Effects of volatile fatty acids on the epithelial cell proliferation of digestive tract and its hormonal mediation. In: Tsuda, T., Y. Sasaki, and R. Kawashima editors, Physiological aspects of digestion and metabolism in ruminants, , Academic Press, Orlando, Florida, 49-59.

Gantois, I.,  R. Ducatelle, F. Pasmans, F. Haesebrouck, I. Hautefort, A. Thompson, J.C. Hinton, and F. Van Immerseel. 2006. Butyrate specifically down-regulates Salmonella pathogenicity island 1 gene expression. Appl Environ Microbiol. 72 (1): 946-949.

Guilloteau, P., R. Zabielski, J.C. David, J.W. Blum, J.A. Morisset, M. Biernat, J. Wolinski, D. Laubitz, and Y. Hamon. 2009. Sodium-butyrate as a growth promoter in milk replacer formula for young calves. J. Dairy Sci. 92: 1038-1049.

Guilloteau, P., L. Martin, V. Eeckhaut, R. Ducatelle, R. Zabielski., and F. Van Immerseel. 2012. From the gut to the peripheral tissues: the multiple effects of butyrate. Nutr Res Rev. 23(2):366-84.

Hu, Z. and Y. Guo. (2007). Effects of dietary sodium butyrate supplementation on the intestinal morphological structure, absorptive function and gut flora in chickens. Animal Feed Science and Technology, 132 (3): 240-249.

Leeson, S., Namkung, H., Antongiovanni, M. and Lee, E.H. (2005). Effect of butyric acid on the performance and carcass yield of broiler chickens. Poult Sci. 84: 1418-1422.

Le Poul, E., C. Loison,S. Struyf, J.Y. Springael., V. Lannoy, M. Decobecq, S. Brezillon, V. Dupriez, G. Vassart, J. Van Damme, M. Parmentier, and M. Detheux. 2003. Functional characterization of human receptors for short chain fatty acids and their role in polymorphonuclear cell activation. J Biol Chem. 278(28): 25481-25489.

Pérez Gutiérrez, O. 2010. Unraveling piglet gut microbiota dynamics in response to feed additives. Thesis submitted in fulfillment of the requirements for the degree of doctor at Wageningen University, The Netherlands.

Piva, A., M. Morlcchini, G. Casadie, P.P. Gatta, G. Biagi, and A. Prandini. 2002. Sodium butyrate improves growth performance of weaned piglets during the first period after weaning. Ital. J. Anim. Sci. 1: 35-41.

Pluske, J.R., M.J. Thompson, C.S. Atwood, P.H. Bird, I.H. Williams, and P.E. Hartmann. 1996. Maintenance of villus height and crypt depth, and enhancement of disaccharide digestion and monosaccharide absorption, in piglets fed on cows’ whole milk after weaning. Br J Nutr. 76 (3):409-22.

Scheppach W. and F. Weiler. 2004. The butyrate story: old wine in new bottles? Curr Opin Clin Nutr Metab Care.  7: 563–567.

Sossai, P. 2012. Butyric acid: what is the future for this old substance? Swiss Med Wkly. June 6;142:w13596. doi: 10.4414/smw.2012.13596.

Tazoe, H., Y. Otomo, L. Kaji, R. Tanaka, S.I. Karaki, and A. Kuwahara. 2008. Roles of short-chain fatty acids receptors, GPR41 and GPR43, on colonic functions. Acta Physiol Pol. 59 : 251-262.

Van Immerseel, F., F. Boyen, I. Gantois,L. Timbermont, L. Bohez, F. Pasmans, F. Haesebrouck, and R. Ducatelle 2005. Supplementation of coated butyric acid in the feed reduces colonization and shedding of Salmonella in poultry. Poult Sci. 84(12): 1851-1856.

Weber, T.E., and B.J. Kerr. 2008. Effect of sodium butyrate on growth performance and response to lipopolysaccharide in weanling pigs. J. Anim Sci. 86: 442-450.

Enteropathogenic Escherichia coli inhibits butyrate uptake in Caco-2 cells by altering the apical membrane MCT1 level

Alip Borthakur, Ravinder K. Gill, Kim Hodges, Krishnamurthy Ramaswamy, Gail Hecht, and Pradeep K. Dudeja from Section of Digestive Diseases and Nutrition, Department of Medicine, University of Illinois at Chicago and Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois

Enteropathogenic Escherichia coli (EPEC), a food-borne human pathogen, is responsible for infantile diarrhea, especially in developing countries. The pathophysiology of EPEC-induced diarrhea, however, is not completely understood. Our recent studies showed modulation of Na + /H + and Cl – /HCO 3 – exchange activities in Caco-2 cells in response to EPEC infection. We hypothesized that intestinal short-chain fatty acid absorption mediated by monocarboxylate transporter 1 (MCT1) might also be altered by EPEC infection. The aim of the current studies was to examine the effect of EPEC infection on butyrate uptake. Caco-2 cells were infected with wild-type EPEC, various mutant strains, or nonpathogenic E. coli HS4, and butyrate uptake was determined. EPEC, but not nonpathogenic E. coli, significantly decreased butyrate uptake. Infection of cells with strains harboring mutations in escN, which encodes a putative ATPase for the EPEC type III secretion system (TTSS), or in the espA, espB, or espD genes encoding structural components of the TTSS, had no effect on butyrate uptake, indicating the TTSS dependence. On the other hand, strains with mutations in the effector protein genes espF, espG, espH, and map inhibited butyrate uptake, similar to the wild-type EPEC. Surface expression of MCT1 decreased considerably after EPEC but not after nonpathogenic E. coli infection. In conclusion, our studies demonstrate inhibition of MCT1-mediated butyrate uptake in Caco-2 cells in response to EPEC infection. This inhibition was dependent on a functional TTSS and the structural proteins EspA, -B, and -D of the translocation apparatus.

【Keywords】  short-chain fatty acids ion transport diarrhea monocarboxylate transporter

 

BUTYRATE AND OTHER SHORT – CHAIN fatty acids (SCFAs) are produced by enteric bacterial fermentation of undigested carbohydrates and dietary fiber. SCFAs are avidly absorbed in the colon and serve as preferential fuel for colonic epithelial cells. Butyrate is the most important SCFA in colonocyte metabolism and is used preferentially over propionate and acetate. We have previously shown ( 12 ) that SCFA absorption across the epithelial membranes of the human ileum and colon involves an SCFA/HCO 3 – (OH – ) exchange mechanism. We and others also demonstrated the involvement of monocarboxylate transporter 1 (MCT1) in the luminal absorption of SCFAs in human intestinal epithelial cells ( 11, 28 ). Butyrate is known to stimulate water and NaCl absorption via activation of Na + /H + exchanger (NHE; see Refs. 6 and 25 ) and apical Cl – /HCO 3 – exchangers ( 24 ). The anti-inflammatory actions of butyrate are supported by both clinical and animal studies, implicating its role in suppressing mucosal inflammation ( 16 ). Also, butyrate-containing retention enemas have proven beneficial in the treatment of ulcerative and diversion colitis ( 6 ). Although decreased SCFA production resulting from impaired colonic fermentation has been shown to impair colonic absorption of sodium and water, little information is available on the factors that might influence efficient absorption of SCFAs by colonocytes. Particularly, no information is available on the modulation of SCFA uptake by pathogenic microorganisms that cause secretory and inflammatory diarrhea in humans.

 

Enteropathogenic Escherichia coli (EPEC) is an important noninvasive human enteric pathogen associated with diarrhea, particularly in infants. Its infection causes specific histopathological alterations of the intestinal enterocytes, called attaching and effacing (A/E) lesions, characterized by effacement of microvilli, close adherence of the bacteria to the host cell membrane, and recruitment of filamentous actin and other cytoskeletal proteins resulting in pedestal formation beneath the sites of attachment ( 32 ). The gene products required for producing A/E lesions are encoded by an 35-kb pathogenicity island in the bacterial chromosome, known as the Locus of Enterocyte Effacement (LEE). The LEE encodes a number of virulence genes, including the components of a type III secretion system (TTSS) that allows direct transfer of bacterial effector molecules into host cells ( 22 ). Several effectors, which are translocated into host cells via the TTSS, have been identified and characterized ( 21 ). These include Tir, EspF, EspG, EspH, and Map. The complete mechanism(s) of EPEC-induced diarrhea is not fully understood and appears to be multifactorial. One such factor may be impairment of ion and solute transport. Diarrhea associated with infection by enteric pathogens could result from either increased Cl – secretion, decreased NaCl absorption, or both. We have previously shown that Cl – /OH – exchange activity is inhibited in response to EPEC infection in Caco-2 cells ( 13 ). Also, EPEC infection increased the activity of NHE2, whereas the activity of NHE3, the predominant Na + -absorbing isoform, was inhibited ( 14 ). Based on the fact that EPEC-induced diarrhea is multifactorial, and that SCFAs maintain mucosal integrity and influence water and electrolyte absorption, it was of interest to determine if butyrate absorption by mucosal cells is affected by EPEC infection. We report here that butyrate uptake by intestinal epithelial Caco-2 cells is significantly decreased by EPEC infection. We further show that membrane expression of MCT1, the SCFA transporter in the intestinal epithelial cells, is also decreased in response to EPEC infection.

 

MATERIALS AND METHODS

 

Materials. sodium butyrate was obtained from NEN Life Science Products (Boston, MA). Caco-2 cells were obtained from ATCC (American Type Culture Collection, Manassas, VA). Sulfo-NHS-SS-biotin for biotinylation of cell surface proteins and streptavidin agarose were from Pierce (Rockford, IL). All other reagents were of at least reagent grade and were obtained from Sigma (St. Louis, MO) or Fisher Scientific (Pittsburgh, PA).

 

Cell culture. Caco-2 cells were grown at 37°C in an atmosphere of 5% CO 2. Cells were maintained in DMEM with 4.5 g/l glucose, 50 U/ml penicillin, 5 µg/ml streptomycin, 2 µg/ml gentamicin, and 20% FBS. Cells used for these studies were plated on 24-well plates at a density of 2 x 10 4 cells/well. Cells were used for bacterial infection and butyrate uptake on day 12-14 postplating and were grown overnight in a medium free of serum and antibiotic before infection.

 

Bacterial culture and infection of cells. The EPEC strains used in this study were as follows: wild-type EPEC strain E2348/69, CVD452 (E2348/69 escN :: Kanamycin; see Ref. 17 ), UMD864 (E2348/69 D48-759 espB1; see Ref. 7 ), UMD 870 (E2348/69 espD1 :: aph-3; Km; see Ref. 20 ), and E2348/69 espG orf3 ( 8 ) and the nonpathogenic isolate HS4 (obtained from J. B. Kaper’s laboratory). Strains were grown overnight in Milleva Luria Borth (LB) media. On the day of experiment, an aliquot of the overnight culture was inoculated in an appropriate volume of serum and antibiotic-free medium supplemented with 0.5% mannose. Bacteria were grown to midlog phase (optical density at 600 nm = 0.4). The culture was spun down and resuspended in the same volume of fresh media. Cell monolayers were then infected at a multiplicity of infection of 100. After infection for the desired time, media were removed, and cell monolayers were washed with PBS.

 

butyrate uptake was determined using cold butyrate concentrations ranging from 0.5 to 15.0 mM.

 

Biotinylation of cell surface proteins and Western blot. Biotinylation of cell surface proteins was carried out according to Akhter et al. ( 1 ) with minor modifications. Briefly, EPEC-infected or control cell monolayers were washed three times with PBS containing 0.1 mM CaCl 2 and 1 mM MgCl 2 at 4°C. The apical surface was then exposed to Sulfo-NHS-SS-biotin (Pierce) at a concentration of 1.5 mg/ml in borate buffer, pH 9.0, by incubation for 1 h at 4°C in horizontal motion. Cells were then quenched with PBS containing CaCl 2, MgCl 2, and 100 mM glycine for 20 min at 4°C. Cells were lysed in RIPA buffer (150 mM NaCl, 50 mM Tris·Cl, pH 7.4, 5 mM EDTA, 1% Triton X-100, 0.1% SDS, and protease inhibitor cocktail). After centrifugation, the supernatant was incubated overnight in streptavidin agarose and then washed three times with lysis buffer. The streptavidin agarose beads were spun down, and sample loading buffer for SDS-PAGE was added. Separated proteins were probed with anti-MCT1 antibody and visualized with enhanced chemiluminescence reagent.

 

Statistical analysis. Results are expressed as means ± SE. Each independent set of results represents data from at least six wells used in three to five separate experiments.

 

RESULTS

 

EPEC infection inhibits butyrate uptake in Caco-2 cells. To determine whether EPEC infection had any effect on butyrate uptake, Caco-2 cells were infected for 1 h, and butyrate uptake was determined as pH-driven butyrate uptake, as described in MATERIALS AND METHODS. Figure 1 shows that butyrate uptake was decreased significantly ( 60%) in EPEC-infected cells compared with uninfected controls. In contrast, infection with nonpathogenic E. coli had no significant effect.

 

Fig. 1. Enteropathogenic Escherichia coli (EPEC) infection inhibits butyrate uptake in Caco-2 cells. Caco-2 cells at 12-14 days postplating were serum starved overnight and then infected for 60 min with EPEC or nonpathogenic E. coli (HS4). butyrate uptake was subsequently measured as described in MATERIALS AND METHODS. Results represent means ± SE of 5 separate experiments performed in triplicate. * P < 0.05 compared with control.

 

Time course of EPEC inhibition of butyrate uptake. The time course of EPEC-mediated inhibition of butyrate uptake was determined by infecting Caco-2 monolayers with EPEC for 15, 30, 60, 90, and 120 min. As shown in Fig. 2, at earlier time points of infection (15 and 30 min), butyrate uptake was not significantly decreased, whereas 70% inhibition of butyrate uptake occurred by 60 min postinfection and persisted for at least 120 min. Therefore, all subsequent experiments were performed at 60 min postinfection.

 

Fig. 2. Time course of EPEC inhibition of butyrate uptake. Caco-2 monolayers were infected with EPEC for 15, 30, 60, 90, and 120 min, and butyrate uptake was measured. Results represent means ± SE of 3 independent experiments performed in triplicate.

 

EPEC infection decreases the maximal velocity of butyrate uptake without altering the Michaelis constant. The kinetics of butyrate uptake by EPEC-infected and uninfected Caco-2 cells were measured using increasing substrate concentrations in the range of 0.5-15 mM. As shown in Fig. 3, EPEC infection significantly decreased the activity of butyrate uptake at all concentrations examined. An analysis of the Lineweaver-Burke plot of this data showed that maximal velocity ( V max ) of butyrate uptake in EPEC-infected cells compared with uninfected controls was reduced significantly ( V max : in nmol·mg protein -1 ·5 min -1 : 15.9 ± 0.1 for control vs. 9.1 ± 0.1 in response to EPEC infection). In contrast, the apparent Michaelis constant ( K m ) did not change significantly (control 2.52 ± 0.04 mM vs. EPEC infected 2.71 ± 0.02 mM ).

 

Fig. 3. Effect of EPEC infection on the kinetics of butyrate uptake in Caco-2 cells. Caco-2 cells were infected with wild-type EPEC for 60 min, and butyrate uptake was determined in the presence of increasing concentrations (0.5-15 mM) of butyrate. The experiment was performed on 3 separate occasions of EPEC infection. A Michaelis-Menten plot from a representative experiment is shown.

 

TTSS is required to inhibit butyrate uptake. The role of various EPEC virulence proteins on the EPEC-induced decrease in butyrate uptake by Caco-2 cells was investigated using specific mutant strains. First, involvement of the TTSS was studied using an escN mutant strain. The product of this gene is the putative ATPase that drives type III secretion of various virulence proteins ( 9 ). As shown in Fig. 4, there was no inhibition of butyrate uptake by Caco-2 cells when infected with the escN mutant strain, in contrast to significant inhibition by wild-type EPEC. These results reveal that a functional TTSS is required for EPEC to inhibit butyrate uptake by Caco-2 cells.

 

Fig. 4. A functional type III secretion system (TTSS) is required for EPEC-mediated inhibition of butyrate uptake. Caco-2 cells were infected with EPEC or escN mutant strain for 60 min, and then butyrate uptake was measured. Results represent means ± SE of 3 independent experiments performed in triplicate. * P < 0.05 compared with control.

 

EPEC secreted proteins are involved in modulating butyrate uptake. We further analyzed the role of the TTSS in EPEC-modulated butyrate uptake by infecting cells with either espA, espB, or espD mutant strains. Each of these virulence genes encodes a structural component of the TTSS; therefore, mutation of any one of them renders TTSS ineffective ( 22 ). Figure 5 shows that infection of cells with these mutants had no effect on butyrate uptake compared with uninfected controls. Infection with wild-type EPEC, as expected, resulted in a marked decline in butyrate uptake. These results indicate that the structural components of the translocation apparatus themselves or the secreted effector molecules are required for the observed effect of EPEC on butyrate uptake.

 

Fig. 5. EPEC-secreted proteins EspA, EspB, and EspD are required for EPEC inhibition of butyrate uptake. Caco-2 cells were infected with EPEC or one of the mutant strains espA, espB, or espD for 60 min, and butyrate uptake was determined. Results represent means ± SE of 4 separate experiments performed in triplicate. * P < 0.05 compared with control.

 

Effector proteins EspF, EspG, EspH, and Map are not involved. EPEC infection results in the delivery of a number of effector proteins in the host cytosol. The role of these effector molecules in modulating butyrate uptake was also studied. Figure 6 shows that mutation of espF, – G, and – H, map, or of a double mutation of espG and its homolog orf3 had no impact on the inhibition of butyrate uptake caused by wild-type EPEC. These results suggest that these effector proteins are not involved in EPEC’s effect on butyrate uptake.

 

Fig. 6. EspF, EspG, EspH, and Map are not required for inhibition of butyrate uptake. Caco-2 cells were infected with EPEC, espF, espG, espH, map, or espG/orf3 double mutants for 60 min, and butyrate uptake was then measured. Results represent means ± SE of 3 separate experiments in triplicate. * P < 0.05 compared with control.

 

Membrane expression of MCT1 is decreased by EPEC infection. It has been previously shown by us and others ( 11, 28 ) that MCT1 is involved in the uptake of butyrate by Caco-2 cells. Therefore, to examine the effect of EPEC infection on MCT1 protein, cell surface proteins from control and infected cells were biotinylated and pulled down from the cell lysate by avidin, and separated proteins were probed with an anti-MCT1 antibody. As shown in Fig. 7 A, surface expression of MCT1 in infected cells was decreased considerably compared with uninfected controls or those infected with nonpathogenic E. coli (HS4), whereas levels of total cellular MCT1 remained constant in all groups. Densitometric scanning 50% decrease in the amount of surface MCT1 compared with total MCT1 in EPEC-infected cells ( Fig. 7 B ).

 

Fig. 7. A : surface expression of monocarboxylate transporter 1 (MCT1) is reduced in response to EPEC infection. Caco-2 cells were infected with EPEC or nonpathogenic E. coli ( HS4 ) for 60 min. Cell surface proteins were biotinylated as described in MATERIALS AND METHODS. Biotinylated proteins were pulled down from equal aliquots of cell lysates (protein contents normalized to 2.5 mg/ml) using equal amounts of streptavidin agarose. SDS-PAGE of the biotinylated proteins or the total proteins in cell lysates were probed with anti-MCT1 antibody. The blot is representative of 3 separate experiments. B : densitometric scanning of the relative band intensities and ratio of surface MCT1 to total MCT1. Values are means ± SE of 3 separate experiments. * P < 0.05 compared with control.

 

DISCUSSION

 

Absorption of SCFAs is important for colonocyte health and metabolism, epithelial integrity, as well as colonic fluid and electrolyte balance. Butyrate, a key SCFA, is known to have multiple regulatory roles in the mammalian colon, including stimulation of fluid and electrolyte absorption ( 24 ) by increasing electroneutral NaCl absorption ( 3 ) and inhibiting Cl – secretion ( 27 ). Recent studies ( 19 ) have also demonstrated that butyrate stimulates promoter activity and expression of the apical NHE3 in the human adenocarcinoma cell line Caco-2. Butyrate has also been implicated in suppressing mucosal inflammation ( 16 ). Decreased production or availability of butyrate has been shown to result in chronic inflammation and acute diarrhea ( 6 ). However, to date, the effects of enteric pathogens on the absorption of SCFAs have not been examined. EPEC is a human enteric pathogen infecting primarily infants and young children. The specific mechanisms by which EPEC causes early diarrhea in infected hosts, however, remain unclear. Diarrhea results from either increased secretion, impaired absorption, or both. EPEC infection has intact been shown to decrease secretagogue-induced Cl – secretion ( 15 ). We have also shown that EPEC infection increases NHE2 activity, whereas the activity of NHE3, the predominant Na + -absorbing isoform, is inhibited ( 14 ). Cl – /OH – exchange activity was also shown to be inhibited in Caco-2 cells infected with EPEC ( 13 ). We have also reported that EPEC infection induces inflammation ( 29 ) and disrupts the structure and barrier function of tight junctions after prolonged infections ( 26 ). These findings suggest that the mechanism(s) of EPEC-induced diarrhea are multifactorial and may also involve modulation of SCFA uptake. Caco-2 cell monolayers were selected to investigate this question, since previous studies from our laboratory ( 11 ) demonstrated that this cell line was a suitable in vitro model to study butyrate uptake. The data presented here suggest that EPEC infection of confluent differentiated Caco-2 monolayers results in a significant decrease in butyrate uptake. This effect was specific, since infection with nonpathogenic E. coli (HS4) had no effect on butyrate uptake. Time course studies of EPEC inhibition of butyrate uptake showed effective inhibition as early as 60 min postinfection that persisted until 120 min.

 

EPEC is noninvasive and does not produce toxins. Instead, it employs the TTSS to deliver virulence factors directly in the host cells ( 32 ). Several mutational studies have enabled identification of individual components of the TTSS. Requirement of an intact TTSS for EPEC to inhibit butyrate uptake is supported by the fact that mutation of the putative ATPase (EscN) blocked the inhibitory effect of EPEC infection. This mutant has also been shown to be ineffective in modulating Na + ( 14 ) and Cl – ( 10 ) uptake by Caco-2 cells. Mutant strains carrying mutations in espA, espB, or espD, all of which interfere with type III secretion, also failed to inhibit butyrate uptake in infected Caco-2 cells. EspA forms a filamentous organelle on the bacterial surface that is postulated to act as a channel for the type III system to deliver proteins to the host cell ( 9 ). EspB and EspD are translocated to the host cell membrane and together are thought to form the translocation pore. EspB also has been reported to be present in the cytoplasm; however, its function here is not known ( 9 ).

 

The EPEC-secreted effector molecules have previously been reported to alter host cell functions via different mechanisms. For example, EspF is known to disrupt tight junctions ( 23 ); EspG, and its homolog Orf3, both disrupt microtubules and produce subtle alterations in barrier functions ( 8, 30 ); EspH is known to alter pedestal morphology and filopodia formation ( 31 ), whereas Map has been reported to alter mitochondrial membrane potential ( 18 ). However, our current studies suggested that the secreted effector molecules EspF, EspG, EspH, and Map were not involved in mediating the observed decrease in butyrate uptake in response to EPEC infection, since these mutants behaved similar to wild-type EPEC in inhibiting butyrate uptake. Orf3, the espG homolog, is encoded in the EPEC genome in a locus distinct from the LEE. Because both effectors (EspG and Orf3) are known to induce disruption of microtubule networks beneath adherent bacteria ( 21 ), we used a double mutant of espG / orf3 to infect cells and observe the effects on butyrate uptake. There was again a significant inhibition of butyrate uptake, similar to that caused by wild-type EPEC, suggesting that these effector molecules are not involved in EPEC-mediated inhibition of butyrate uptake. However, the inability of nonpathogenic E. coli or type III secretion mutants to inhibit butyrate uptake seems to indicate that the observed effects are related to pathogenicity.

 

Our kinetic studies suggested that EPEC inhibited butyrate uptake via changes in the value of V max without significantly altering the apparent K m, indicating a decrease in the expression of the butyrate transporter on the plasma membrane. This was indeed found to be the case by performing biotinylation studies to quantitate the amount of surface MCT1, the butyrate transporter in Caco-2 cells. Although the surface expression of MCT1 was considerably lower in cells infected with EPEC compared with nonpathogenic E. coli, or uninfected control cells, total cellular MCT1 was the same in all the groups. These studies demonstrate the regulation of butyrate uptake either by retrieval of MCT1 from the apical plasma membrane or via reduced targeting to the apical membrane in response to EPEC infection. In this regard, previous studies of Buyse et. al. ( 5 ) also suggested that MCT1 is regulated by luminal leptin by altering translocation of MCT1 to the apical plasma membrane.

 

The effects of pathogenic organisms on fluid and electrolyte secretion by host intestinal epithelial cells have been well documented. Various enteric pathogens elicit a Cl – secretory response. The studies from our laboratory showed profound early effects of EPEC infection on intestinal epithelial absorption, an aspect that has not been studied previously. The observed effects of EPEC infection on butyrate uptake reported here and previously published results from our laboratory on Na + /H + and Cl – /OH – exchange activities strongly suggest that EPEC infection has profound effects on the host intestinal absorptive ion transport processes. We speculate that decreased butyrate availability caused by EPEC infection might compromise colonic epithelial integrity, resulting in inflammation of the epithelium, and also might contribute to EPEC-mediated diarrhea by inhibiting ion absorption in the colon. Our results demonstrate that EPEC requires a TTSS to inhibit butyrate uptake via regulation of MCT1. In lieu of the important role of SCFA in stimulating human colonic electrolyte absorption and being the key nutrient in the colon, our studies not only add to increased understanding of the mechanisms regulating human intestinal SCFA absorption but also suggest the potential role of inhibition of this key ion as an important contributory factor in the pathophysiology of EPEC-associated diarrhea.

 

GRANTS

 

These studies were supported by the Department of Veterans Affairs and the National Institute of Diabetes and Digestive and Kidney Diseases Grants DK-54016 (P. K. Dudeja), DK-68324 (P. K. Dudeja), DK-33349 (K. Ramaswamy), DK-67990 (K. Ramaswamy), and DK-50694 (G. Hecht).

【参考文献】
Akhter S, Kovbasnjuk O, Li X, Covet M, Noel J, Arpin M, Hubbard AL, and Donowitz M. Na + /H + exchanger 3 is in large complexes in the center of the apical surface of proximal tubule-derived OK cells. Am J Physiol Cell Physiol 283: C927-C940, 2002.

Alrefai WA, Tyagi S, Gill R, Saksena S, Hadjiagapiou C, Mansour F, Ramaswamy K, and Dudeja PK. Regulation of butyrate uptake in Caco2 cells by phorbol 12-myristate 13-acetate. Am J Physiol Gastrointest Liver Physiol 286: G197-G203, 2004.

Binder HJ and Mehta P. Short chain fatty acids stimulate Na and Cl absorption in vitro in the rat distal colon. Gastroenterology 96: 989-996, 1989.

Bradford M. A rapid and sensitive method for quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Anal Biochem 72: 248-254, 1976.

Buyse M, Sitaraman SV, Liu X, Bado A, and Merlin D. Luminal leptin enhances CD147/MCT1 mediated uptake of butyrate in the human intestinal cell line Caco2-BBE. J Biol Chem 277: 28182-28190, 2002.

Cook SI and Sellin JH. Review article: short chain fatty acids in health and disease. Pharmacol Ther 12: 499-507, 1998.

Donnenberg MS, Yu J, and Kaper JB. A second chromosomal gene necessary for intimate attachment of enteropathogenic E. coli to epithelial cells. J Bacteriol 175: 4670-4680, 1993.

Elliott SJ, Krejany EO, Mellies JL, Robins-Browne RM, Sasakawa C, and Kaper JB. EspG, a novel type III system-secreted protein from enteropathogenic E. coli with similarities to VirA of Shigella flexneri. Infect Immun 69: 4027-4033, 2001.

Gauthier A, Puente JL, and Finlay BB. Secretion of the enteropathogenic E. coli type III secretion system requires components of the type III apparatus for assembly and localization. Infect Immun 71: 3310-3319, 2003.

Gill Ravinder K, Hodges K, Borthakur A, Saksena S, Ramaswamy K, Hecht GA, and Dudeja PK. Role of enteropathogenic E. coli virulence genes in inhibition of apical Cl – /OH – exchange activity in Caco-2 cells (Abstract). Gastroenterology 126: A-95, 2004.

Hadjiagapiou C, Schmidt L, Dudeja PK, Lyden TJ, and Ramaswamy K. Mechanism(s) of butyrate uptake in Caco2 cells: Role of monocarboxylate transporter 1. Am J Physiol Gastrointest Liver Physiol 279: G775-G780, 2000.

Harig JM, Ng EK, Dudeja PK, Brasitus TA, and Ramaswamy K. Transport of n-butyrate into human colonic luminal membrane vesicles. Am J Physiol Gastrointest Liver Physiol 271: G415-G422, 1996.

Hecht G, Gill R, Saksena S, Tyagi S, Hodges K, Ramaswamy K, and Dudeja PK. Enteropathogenic E. coli inhibits Cl – /OH – exchange activity in Caco2 cells (Abstract). Gastroenterology 124: A482, 2003.

Hecht G, Hodges K, Gill R, Kear F, Tyagi S, Malakooti J, Ramaswamy K, and Dudeja PK. Differential regulation of Na + H + exchange isoform activities by enteropathogenic E. coli in human intestinal epithelial cells. Am J Physiol Gastrointest Liver Physiol 287: G370-G378, 2004.

Hecht G and Koutsouris A. Enteropathogenic E coli attenuates secretagogue-induced net intestinal ion transport but not Cl- secretion. Am J Physiol Gastrointest Liver Physiol 276: G781-G788, 1999.

Inan MS, Rasoulpour RJ, Yin L, Habbard AK, Rosenberg DW, and Giardena C. The luminal short chain fatty acid butyrate modulate NF-kappaB activity in a human colonic epithelial cell line. Gastroenterology 118: 724-734, 2000.

Jarvis KG, Giron JA, Jerse AE, McDaniel TK, Donnenberg MS, and Kaper JB. Enteropathogenic E. coli contains a putative type III secretion system necessary for the export of proteins involved in attaching and effacing lesion formation. Proc Natl Acad Sci USA 92: 7996-8000, 1995.

Kenny B and Jepson M. Targeting of an EPEC effector protein to host mitochondria. Cell Microbiol 2: 579-590, 2000.

Kiela PR, Hines ER, Collins JF, and Ghisan FK. Regulation of the rat NHE3 gene promoter by sodium butyrate. Am J Physiol Gastrointest Liver Physiol 281: G947-G956, 2001.

Lai LC, Wainwright LA, Stone KD, and Donnenberg MS. A third secreted protein that is encoded by the enteropathogenic E. coli pathogenicity island is required for transduction of signals and for attaching and effacing activities in host cells. Infect Immun 65: 2211-2217, 1997.

Matsuzawa T, Kuwae A, Yoshida S, sasakawa C, and Abe A. Enteropathogenic E. coli activates the RhoA signalling pathway via the stimulation of GEF-H1. EMBO J 23: 3570-3582, 2004.

McDaniel TK and Kaper JB. A cloned pathogenicity island from enteropathogenic E. coli confers the attaching and effacing phenotype on E coli k-12. Mol Microbiol 23: 399-407, 1997.

McNamara BP, Koutsouris A, O’conell CB, Nougayrede JP, Donnenberg MS, and Hecht G. Translocated EspF protein from enteropathogenic E. coli disrupts host intestinal barrier function. J Clin Invest 107:621-629, 2001.

Montrose MH and Kere J. Anion absorption in the intestine: anion transporters, short chain fatty acids, and role of the DRA gene product. Curr Top Membr Transp 50: 301-928, 2001.

Musch MW, Bookstein C, Xie Y, Sellin JH, and Chang EB. Short chain fatty acids increase intestinal Na absorption by induction of NHE3 in rat colon and human intestinal C2/bbe cells. Am J Physiol Gastrointest Liver Physiol 280: G687-G693, 2001.

Muza-Moons MM, Schneeberger EE, and Hecht GA. Enteropathogenic E. coli infection leads to appearance of aberrant tight junction strands in the lateral membrane of intestinal epithelial cells. Cell Microbiol 6: 783-793, 2004.

Resta-Lenert S, Trung F, Barrett KE, and Eckmann L. Inhibition of epithelial chloride secretion by butyrate: role of reduced adenylyl cyclase expression and activity. Am J Physiol Cell Physiol 281: C1837-C1849, 2001.

Ritzhaupt A, Wood IS, Ellis A, Hosie KB, and Shirazi-Beechey SP. Identification and characterization of a monocarboxylate transporter. J Physiol 513: 719-732, 1998.

Savkovic SV, Koutsouris A, and Hecht A. Activation of NF-kappaB in intestinal epithelial cells by enteropathogenic E. coli. Am J Physiol Cell Physiol 273: C1160-C1167, 1997.

Tomson FL, Viswanathan VK, Kanack KJ, Kanteti RP, Straub KV, Menet M, Kaper JB, and Hecht G. Enteropathogenic E. coli EspG disrupts microtubules and in conjunction with Orf3 enhances perturbation of the tight junction barrier. Mol Microbiol 56: 447-464, 2005.

Tu X, Nisan I, Yona C, Hanski E, and Rosenshine I. EspH, a new cytoskeleton-modulating effector of enterohaemorrhagic and enteropathogenic E. coli. Mol Microbiol 47: 595-606, 2003.

Vallance BA and Finlay BB. Exploitation of host cells by enteropathogenic E. coli. Proc Natl Acad Sci USA 97: 8799-8806, 2000.

Butyrate promotes the recovering of intestinal wound healing

http://www.feedadditive.com/docs/jas-90-Supplement_4-266.pdf

Article (PDF Available)inJournal of Animal Science 90(Supplement 4):266-268 · January 2013with198 Reads

X. Ma,*2 P. X. Fan,*L. S. Li,* S. Y. Qiao,* G. L. Zhang,† D. F. Li*
*State Key Lab of Animal Nutrition, China Agricultural University, Beijing, 100193, China; and †Department of Animal
Science, Oklahoma State University, Stillwater, Oklahoma, 74078

Abstract
Postweaning diarrhea is one of the most common causes of morbidity and mortality in weanling piglets. Feeding sodium butyrate to weanling piglets decreased the incidence of diarrhea, but the mechanism has not been fully elucidated. The present study was to evaluate the effect of sodium butyrate on diarrhea in relation to wound healing of intestinal barrier using IPEC-J2 cell model. Cultured cells were scratched to induce wound and then were treated with 4 mM sodium butyrate. The results showed that supplementation of the cells with sodium butyrate significantly promoted the process of wound healing, indicating the protective effects of butyrate on the intestinal mucosa. Butyrate treatment enhanced mRNA expression of the intestinal mucosal tight junction proteins occludin and zonula occluden protein-1 (P < 0.05), which suggested that the promotion of wound healing by butyrate is related to the maintenance of the function of the intestinal barrier. In addition, in the butyrate-treated group, intestinal total superoxide dismutase and glutathione peroxidase (P < 0.05), two of the main antioxidant enzymes, as well as glutathione (P < 0.05), one of the nonenzymatic antioxidant components, were enhanced whereas the malondialdehyde level, a marker of free radical mediated lipid peroxidation injury, was decreased (P < 0.05) compared with the control group. Collectively, these results indicate that dietary sodium butyrate might, at least partly, play an important role in recovering the intestinal tight junctions having a positive effect on maintaining the gut integrity.

Butyrate promotes the recovering of intestinal wound healing through its positive effect on the tight junctions (PDF Download Available).

 

 

Dietetic Supplementation with Butiric Acid. What is the Evidence?

http://www.feedadditive.com/docs/Dietetic-supplementation-with-butiric-acid.-what-is-the-evidence.pdf

Butyric acid is a short chain fatty acid with a central role in the metabolism and homeostasis of the digestive system, specially of the colon.

It is mainly provided to the body through the microbial fermentation of dietetic fiber, but also to a lesser extent through a few foods which content different chemical forms of butyric acid, although they are in very small quantities.

Due to the important positive butyric acid actions over the development of the intestinal epithelium, the balance of intestinal microbiota, the intestinal permeability and its noticeable anti-inflammatory effect, there have been many studies about the possible therapeutic usage of the supplementation with this short chain fatty acid in gastrointestinal pathologies, like:

  • Inflammatory bowel disease (Ulcerative Colitis and Crohn Disease)
  • Irritable bowel syndrome
  • Colon cancer
  • Constipation
  • Diarrhea 
  • Travelers diarrhea
  • Antibiotics associated diarrhea

Some of the main clinical trials and other possible therapeutic usages are detailed next:

216 patients with ulcerative colitis showed an incomplete response to standard mesalazine treatment. The treatment proposed included mesalazine, butyric acid and inuline, being effective in reducing disease activity with a marked improvement of symptoms and in the endoscopic appearance of mucosa.
Combined butyric acid/mesalazine treatment in ulcerative colitis with mild-moderate activity. Results of a multicentre pilot study. Minerva Gastroenterol Dietol. 2008 Sep;54(3):231-8. 

25 patients with ulcerative colitis completed this clinical trial in two groups, where one group treatment was mesalazine and the other was mesalazine plus butyric acid. The results of the present study indicate that oral butyrate is safe and well tolerated. These data also suggest that oral butyrate may improve the efficacy of oral mesalazine in active ulcerative colitis and prompt the need of a large scale investigation to confirm the present findings.
Combined oral sodium butyrate and mesalazine treatment compared to oral mesalazine alone in ulcerative colitis: randomized, double-blind, placebo-controlled pilot study. Dig Dis Sci. 2000 May;45(5):976-81. 

After 4 weeks there was a significant decrease of pain during defaecation in the microencapsulated sodium butyrate group versus the placebo group, which extended to improvement of urgency and bowel habit at 12 weeks of treatment in this trial of sixty-six patients with irritable bowel syndrome.
Microencapsulated sodium butyrate reduces the frequency of abdominal pain in patients with irritable bowel syndrome. Colorectal Dis. 2013 Feb;15(2):204-9. 

This article describes how butyric acid supplementation seems to be a promising therapy for irritable bowel syndrome. It is worth noting that no side effects were observed during treatment, which confirms the safety of its use in clinical practice.
Butyric acid in irritable bowel syndrome. Prz Gastroenterol. 2013;8(6):350-3. 

42 adult patients planning to travel to subtropical countries were enrolled in the study and randomized into a study group receiving butyric acid supplementation or placebo. In comparison to the control arm, the study arm noted significantly reduced occurrence of Travellers’ diarrhoea, being safe and may constitute a new method of travellers’ diarrhoea prevention.
Sodium butyrate and short chain fatty acids in prevention of travellers’ diarrhoea: a randomized prospective study. Travel Med Infect Dis. 2014 Mar-Apr;12(2):183-8. 

The present study demonstrated that simultaneous treatment with LGG and tributyrin prevents antibiotic-induced downregulation of genes and proteins involved with intestinal fluid and electrolyte homeostasis and intestinal barrier function in the intestinal tract.
Lactobacillus GG and tributyrin supplementation reduce antibiotic-induced intestinal injury. JPEN J Parenter Enteral Nutr. 2013 Nov;37(6):763-74.

This article presents the potential beneficial mechanisms of action of butyric acid in defecation disorders, which are primarily associated with reductions in pain during defecation and inflammation in the gut, among others.
Butyric acid in functional constipation. Prz Gastroenterol. 2013;8(5):295-8.  

This study shows the effects of tributyrin on growth, differentiation and vitamin D receptor expression in a human colon cancer cell line. Tributyrin was more potent in inhibiting growth and inducing cell differentiation than natural butyrate. The effect was further enhanced after addition of physiologic concentrations of dihydroxycholecalciferol.

This may provide a useful therapeutic approach in chemoprevention and treatment of colorectal cancer by the two nutrients occurring naturally in human diet.
Tributyrin, a stable and rapidly absorbed prodrug of butyric acid, enhances antiproliferative effects of dihydroxycholecalciferol in human colon cancer cells. J Nutr. 2001 Jun;131(6):1839-43.

The research about administration forms of butyric acid is aimed to assure that the ingredient reaches the final part of the intestine, and tributyrin seems to work as a prodrug of butyric acid, being a triglyceride that liberate butyric acid by the action of enzyme lipase.
Clinical and pharmacologic study of tributyrin: an oral butyrate prodrug.Cancer Chemother Pharmacol (2003) 51: 439.

Finally, note the article published in the World Journal of Gastroenterology: “Potential beneficial effects of butyrate in intestinal and extraintestinal diseases” in which they are reviewed and summarized the main trials and research on the wide range of clinical uses of butyric acid.
Potential beneficial effects of butyrate in intestinal and extraintestinal diseases. World J Gastroenterol. 2011 Mar 28; 17(12): 1519–1528.

 

David Manrique @ManriqueDVD 

María Eugenia González @EuNutricion

Farming Sector Needs to Encourage More Women into the Industry

Farming Sector Needs to Encourage More Women into the Industry

06 July 2016

UK – The farming sector needs to encourage more female participation in the industry and promote the significant role of women already working in agriculture.

Cabinet Secretary for Rural Economy and Connectivity, Fergus Ewing, kicked off proceedings by launching plans for Scottish Government research into women working in farming industries.

He said: “We have commissioned this research to identify the challenges and come up with policy initiatives to improve women’s participation in farming. In particular the contribution women make to agriculture in Scotland and the wider rural economy which is not always visible.”

Professor Sally Shortall, has been commissioned by the Scottish Government to carry out the research, assisted by colleagues at the James Hutton Institute.

“While we will focus on identifying the issues and producing actual policy recommendations to help improve the situation; the study is not anti-men or about positive discrimination,” Sally explained.

“It is about it is about improving the efficiency and economic viability of the farm and farming.”

Many of the other speakers echoed that message, that women in the sector were not interested in special treatment, merely for their skills and experience to be recognised and put to good use supporting the industry.

Gemma Thomson, Legal and Technical Policy Manager at NFU Scotland, said: “Promoting women in agriculture is not a new thing, but it is now building into a positive force. What we don’t want is positive discrimination however. That would be negative. But agriculture needs to be much better at promoting itself as a real career choice for females.”

SRUC Board Member and Vet Kate Richards noted that: “Women often feel they have to be better and give more to be taken seriously. That is an image that needs to change with women accepted as equals.”

Fergus Ewing also spoke of need to change perceptions. He said: “What we need to achieve is to change that image of the average Scottish farmer – being older and male.”

With over hundred women (and a few men) in the room, and seven inspirational female speakers, it is clear that that image is not necessarily a true representation of the industry. However, many of those at the event felt more needs to be done to encourage women into the sector, both those from a farming background, and those who might consider themselves ‘outsiders’.

One such ‘outsider’, SAOS’s Emma Patterson Taylor, said: “There are many advantages you can bring as an ‘outsider’ – you look at things from a different perspective and are not bound by the way it has always been done.”

SRUC’s Acting CEO and Principal, Janet Swadling, was also new to the sector when she joined SRUC twenty years ago. She concluded the event by saying: “I did not come from a farming background, but I am absolutely passionate about what I do, and am proud to have led the national college for the land-based sector for two and a half years.

“I think as an industry we all need to do more to promote the great work being done by women and men in the agricultural sector, and try and get more young women in particular studying and working in this field.”

Weekly Overview: How Will Brexit Affect the US Pork Market?

Weekly Overview: How Will Brexit Affect the US Pork Market?

05 July 2016

 

GLOBAL – US pig market analysts have been discussing the impact that the UK’s exit from the EU might have on the US’s pork market.

In the wake of brexit the US dollar has strengthened against the pound, making US products more expensive.

Steve Meyer and Len Steiner explained that when the dollar becomes stronger suddenly the price of US products goes up, making US pork less competitive relative to other exporting nations.

When the US dollar goes up the US market also becomes more attractive to ship to. The result is a shift in the relative flow of products into the US, ultimately impacting returns of US livestock producers.

Chris Hurt, Extension Economist at Purdue University, said the US only exports a small amount of pork to the EU. In 2015, only 0.2 per cent of US pork exports were destined for EU28 countries.

However, due to the strengthened the US dollar against the Euro, the 19 EU countries using the Euro are now at an immediate price advantage over US pork.

The longer-term economic implications of Brexit may be the most important and could reduce the rate of world economic growth. If Brexit does slow world income growth, that could be negative for global sales of pork, said Mr Hurt.

Following on with our Brexit coverage, the National Farmers Union has held a consultation on what a domestic UK farming policy should look like.

The agreed principles from NFU Council include ensuring the best access to markets in Europe, securing trade deals, preventing lower standard imports and creating an agricultural workers scheme.

In disease news this week, South Africa has reported three more outbreaks of African Swine Fever (ASF).

Reporting for ThePigSite, Glenneis Kriel spoke to the CEO of the South African Pork Producers Association, Simon Streicher, about the outbreaks.

Mr Streicher explained that the outbreaks have not had a big impact on commercial production, it is only affecting small-scale producers.

“Most commercial producers have sound biosecurity measures to prevent an outbreak. Small scale producers, on the other hand, cannot always afford these measurements, which ironically are aimed at protecting their investments,” Mr Streicher said.

ASF has also been reported this week on pig farms in Poland, Ukraine, Russiaand Lithuania.

There has also been another outbreak of Porcine Epidemic Diarrhoea (PED) this week in Huron County, Ontario, Canada.