Explore
Communities in English
Advertise on Engormix

Poultry nutrient- Managing the gut

Managing the Gut to Maximize Nutrient Utilization

Published: November 10, 2010
By: Stephen R. Collett (The University of Georgia)
Introduction

The impact of gastrointestinal disease ranges from erosive to catastrophic, and considerable time, effort and financial resources are devoted to limiting the risk and consequence of disease challenge. Intense breeding and selection programmes have provided birds with the genetic potential to perform in the face of disease challenge, while good biosecurity has reduced the risk and consequence of infectious disease. At the production level, capital investment in poultry housing and equipment, and best operating practices, have helped to limit environmental stress.

Ironically, relatively little effort has been directed at managing the ecology of the gastrointestinal tract, the very site of nutrient digestion and absorption. This neglect is in part due to ignorance, but perhaps more importantly, it stems from the success of in-feed antibiotics and anticoccidials in preventing enteric disease. In the absence of these in-feed antimicrobials the paradigm of gut health management has to change. Working with the forces of nature to control the ecology of the digestive tract is the only alternative. Although intestinal ecology is very complex, and, difficult to understand, the position is changing rapidly. Advanced molecular techniques are being used to characterise details of the gut flora at the cellular level, in the hopes that novel strategies to preserve gut health are revealed.

Managing Gut Health - form, function and flora

In the high volume low margin poultry industry the economic impact of low grade (subclinical) disease is commonly rate-limiting. Health and disease are, however, not mutually exclusive. Disease only measurably impacts performance once the aggregate of homeostatic stressors exceed the bird's coping mechanisms. The negative impact of any challenge is thus directly proportional to the prevailing level of stress. While low grade stress or sub-clinical disease may go unnoticed, it is frequently sufficient to preclude birds from achieving genetic potential. These subtle changes in flock health are signalled by a decline in flock uniformity, because the degree of stress experienced by each bird in a flock varies; flock uniformity is rapidly reduced and skewed by disease challenge (Collett, 2003; Klasing, 1998). Since flock uniformity is correlated with economic return, it serves as a very useful, sensitive and early indicator of changes in flock health.

Health tracking systems provide real time information which allows early detection and response to disease-challenge. There are, however, no clearly defined objective, visual indicators of low grade gastro-intestinal disease, that are closely correlated with bird productivity. Early detection, appraisal and response to overt disease remains an art. A thorough understanding of the complex ecology and epidemiology of disease in intensive production systems is essential. In order to predict the impact of gastrointestinal disease on performance, necropsy findings must be evaluated in conjunction with all other available information and not viewed as absolute. Each component of the digestive tract must be evaluated relative to form, function and microbial content.

The importance of early gut development cannot be over emphasized. The efficiency of digestion and absorption is directly proportional to the surface area and integrity of the intestinal epithelial lining. The epithelium, with its network of microfibers and intercellular tight junctions, make up the intestinal cytoskeleton which separates the host from the complex microenvironment of the enteron. A series of villi and microvilli adorn the epithelial surface, increasing the surface area by approximately 600 times. This intricate design allows the ebb and flow of copious amounts of water and the absorption of nutrients to occur on a continuous basis, while at the same time, preventing the microbial inhabitants and their toxic metabolites from gaining access to the body. The complex but harmonious relationship between the gut microbiota and the host, is crucial to normal function (Collier et al., 2003; Neish, 2002). Any breakdown in this highly evolved relationship, stimulates a protective response by the host, which involves a cascade of events causing inflammation and gastrointestinal disease.

The efficiency of nutrient assimilation hinges on the early establishment and maintenance of a favourable gut lumen environment. Colonization of the gut with pioneer bacteria species, that are able to modulate expression of genes in the gut epithelia to create favourable conditions for establishment of a stable and beneficial climax flora, should be the starting point of any gut health management program (Guarner and Malagelada, 2003; Hooper and Gordon, 2001).

Steps to control gut health should start at the parent flock level, since the first organisms to gain access to the hatchling gut originate from the parent. Vertical transmission of gut inhabitants (from parent to offspring) can be transovarial (inside the egg) or as a result of shell contamination. In the artificially clean hatchery environment, even low doses of beneficial bacteria can significantly improve resistance to pathogen colonization and artificial seeding of the gut at an early age has been shown to be beneficial (Chaing and Hseih, 1995; Edens et al., 1997; England et al., 1996; Fernandez et al., 2002; Fukata et al., 1991; Hofacre et al., 2003; Hofacre et al., 1998; Hofacre, 2003; La Ragione et al., 2004; Mohan et al., 1996; Owings et al., 1990).

The first organisms to colonize the gut determine the composition of the climax flora by creating the microenvironment necessary for complex microbial community architectural development (Hooper and Gordon, 2001). In addition to seeding the gut with the correct pioneer species, it is crucial to enhance their ability to proliferate, compete and colonize, so as to avoid pathogen proliferation. As colonization proceeds, organisms attach to one another and the epithelium, by a series of fibrils to form a tightly adherent mat over the gut surface (Giron et al., 2002). Pathogens are thereby precluded access to the epithelial surface and their ability to colonize is compromised by a process of competitive exclusion (Nurmi and Rantala, 1973). Microbe attachment to host cell docking sites on the intestinal epithelium is dependent on surface molecule structure and is the pivotal first step in the colonisation of the gut (Giron et al., 2002; Sharon and Lis, 1993; Stavric et al., 1987) . Since several gut pathogens recognise and attach to specific gut epithelia glycoproteins, products that mimic these docking sites are also useful in preventing attachment and reducing the risk of pathogen colonization (Finucane et al., 1999; Giron et al., 2002; Spring, 1996).

In-feed antibiotics have for the past 50 years been used to enhance feed efficiency by subtly changing the composition of the gut flora (Anderson et al., 2000; Collier et al., 2003; Lu et al., 2003; Rosen, 1996; Rosen, 1995). Weak organic acids can also be used to change gut flora community structure. As weak proton donors, they are able to escape inactivation in the proventriculus and gizzard, while their presence in the small intestine modifies microbial community composition by providing a competitive advantage to the acid tolerant organisms (Davidson, 2001; Ricke, 2003). Such manipulation of the microbiota has both short and long term implications.

Poultry production although cyclical in nature, should from a gut flora point of view, be viewed as a continuous system. Members of the gut microbial community surviving in litter residue are carried over from one cycle to the next, thus serving as the "seed stock" for the gut flora of the next placement (Lilijebjelke et al., 2003). In view of this, it is surprising that although there are literally thousands of growth-promotant trials demonstrating their efficacy, the literature is devoid of data showing the long-term effect of such programs. While in-feed antibiotics can alter the gut flora within a couple of weeks, it takes several grow-out cycles to change the house (litter) flora (Avellaneda et al., 2003; Idris et al., 2003; Lilijebjelke et al., 2003; Schildknect et al., 2003a,2003b). This is by no means a new concept, both rotation and shuttle programs have been used for decades to avoid the lack of response to in-feed antibiotics following their persistent use.
The realization that even minor changes in the microbial community composition can affect long term productivity (through incremental displacement and replacement of the house flora) has highlighted the significance of microbial community management. Attention to detail is more critical than ever. Mycotoxins, previously recognised for their host toxicity, also have strong antimicrobial properties and may, for example, affect long term productivity by altering the gut flora (Kubena et al., 2001; Swamy et al., 2002a; Swamy et al., 2002b). In such instances, mycotoxin binders may be useful in helping to preserve the stability of the microbiota, even when mycotoxin levels are low (Kubena et al., 2001; Swamy et al., 2002a; Swamy et al., 2002b).

The efficiency of nutrient assimilation is highly correlated with the functional surface area of the intestinal lining (Sklan, 2001). Inflammatory response at the gut interface has a direct impact on feed assimilation (intake, digestion and absorption) by compromising functional surface area. Irrespective of the initiating cause, disease process begins with an inflammatory response and it is the extent of this inflammatory response that determines its performance effect. An appropriate immune response limits the consequence of challenge, while an inadequate or excessive response depresses assimilation efficiency and productivity (Klasing, 1998; Klasing et al., 1987).

Once initiated, inflammation stimulates acute-phase protein synthesis in the liver which causes several behavioural, hormonal and metabolic perturbations, the ramifications of which range from negligible to dramatic (Klasing, 1998; Klasing and Johnstone, 1991). Local inflammatory response causes tissue damage, while systemic or fever response causes feed intake to decline, protein turnover to accelerate and a rapid transition into negative nitrogen balance (Collier et al., 2003; Klasing, 1998; Klasing et al., 1987). With systemic challenge, most (70%) of the negative impact on growth rate and feed efficiency is attributed to reduced feed intake, while the inefficiencies of catabolism and nutrient absorption account for the rest (30%) (Collier et al., 2003; Klasing, 1998; Klasing and Barnes, 1988; Klasing et al., 1987; Klipper et al., 2000). Low level antigen stimulation at the gut/ingesta interface, seldom results in systemic/fever response and is in fact essential for cellular homeostasis (Petrof et al., 2004; Sansonetti and Di Santo, 2007). Excessive pro-inflammatory mediator release can, however, damage host tissue, thereby causing localized inflammatory disease and reduced feed efficiency (Klasing, 1998; Klasing and Barnes, 1988; Klasing et al., 1987; Klipper et al., 2000,, 2001).

Antigen induced inflammation of the gut lining stimulates mucus secretion, increased paracellular permeability, and accelerated feed passage (peristalsis) (Collier et al., 2003; Cooper, 1984). The cascade of events that follows, is self perpetuating since increased permeability enhances toxin and antigen penetration which stimulates inflammation, and the resulting increase in mucus production attracts mucolytic species such as Clostridium perfringens, which produce tissue damaging cytotoxins (Collier et al., 2008; Collier et al., 2003).

The nature and extent of the inflammatory response is influenced by several nutritional factors. Dietary polyunsaturated fatty acids (PUFA) for example, indirectly determine the type of immune response that follows cell damage. Lipids incorporated into the cell membrane during growth and development are subsequently used as the substrate for immune system, communication molecule synthesis (Klasing, 1998; Korver and Klasing, 1995; Korver and Klasing, 1997). Cereal grains are high in linoleic acid (n-6 PUFA precursor for arachidonic acid) which favours the production of prostaglandins, leukotreins and thromboxanes, while fish oil is high in n-3 PUFA, which favours production of interleukin-1 and prostaglandin-E (Fritsche et al., 1991; Korver and Klasing, 1997).

Maternal antibodies provide crucial support to the maturing immune system of the hatchling, but early gastrointestinal defence is primarily dependent on the innate component of the immune system. During the first 3 days post hatch, the chick immune system learns to tolerate innocuous gut antigens. Gut barrier function matures by day 3 and oral exposure to ‘new' antigens after this is more likely to lead to an inappropriate immune response and negatively impact feed efficiency (Geyra et al., 2001; Klipper et al., 2001,, 2004; Uni et al., 2000; Uni et al., 1999). It may be advantageous to ensure that pre-starter rations contain the feed ingredients (antigens) necessary to ensure that the mature immune system is tolerant of these feed antigens after closure of gut barrier function.

Maternally derived antibodies also play a crucial role in guiding the development of the hatchling immune system. There is a fine line between tolerance and response. Maternal antibodies to hazardous/pathogenic antigens block development of tolerance thus enabling the developing immune system to differentiate potentially hazardous antigens from innocuous feed or commensal antigens (Klipper et al., 2004). Gut health management should start at the parent flock level (Fernandez et al., 2002).

The practice of maximising disease resistance is too costly for the modern poultry industry (Klasing, 1998). Immune stimulation needs careful evaluation since the cost of an exaggerated or inappropriate immune response is counterproductive in terms of feed efficiency (Collier et al., 2003; Grimble, 2001; Klasing, 1998; Klasing et al., 1987). Feed is the single largest input cost and as feed margin has declined, the cost driven poultry industry has shifted emphasis from disease resistance to disease resilience (Klasing, 1998).

While an excessive or inappropriate immune response erodes productivity by adversely affecting the feed conversion, an inadequate immune response usually results in increased flock mortality. Specific infectious diseases, nutritional deficiencies, toxicities and stress are all factors that can cause immune suppression and result in an inadequate immune response (Ferket et al., 1999; Ferket and Qureshi, 1992; Qureshi et al., 1998; Siegel, 1994; Surai, 2002; Swamy et al., 2002a; Swamy et al., 2002b; Sword et al., 1991).

It is possible to maintain productivity in the face of disease challenge by modulating the immune response. Stimulation of the protective antibody response enhances disease resistance and suppression of the acute phase/fever response preserves feed intake and bird performance (Collett, 2003; Collett and Dawson, 2001; Ferket et al., 2002; Klasing, 1998; Klasing et al., 1987; Parks et al., 2001; Savage et al., 1996). Both endogenous and exogenous anti-inflammatory agents help to maintain gut health by modulating the immune response so as to preserve the integrity of the gut lining and reduce the fever response (Choct et al., 2004; Ferket et al., 2002; Grimble, 2001; Kelly et al., 2004; Klasing, 1998; Korver et al., 1998; Korver et al., 1997; Parks et al., 2001; Surai, 2002; Sword et al., 1991).

Years of breeding and selection for growth rate have increased broiler appetite, daily feed intake, and feed passage rate. Interestingly, retention times for the small intestine have remained fairly constant while those for the proventriculus and gizzard have declined (Denbow, 2000; Washburn, 1991). This is probably because feed processing at the feed mill reduces the need for particle size reduction in the gizzard, while nutrient utilization is proportional to small intestine retention time. Protein digestion is, however, potentially compromised by short proventriculus/gizzard retention, since the conversion pepsinogen to pepsin is pH dependant (Denbow, 2000; Klipper et al., 2004; Sklan et al., 1975).

To adequately manage gut health the nutritionist should consider ingredient blend, in addition to nutrient specification, since ingredient characteristics such as viscosity, particle size, digestibility (starch), and lipid or protein content affect passage time (Bedford, 1996; Classen, 1996; Refstie et al., 1999; Sell et al., 1983; Sibbald, 1979; Weurding et al., 2001). Certain undesirable ingredient characteristics such as viscosity and non-digestible nutrient content can be countered with concomitant enzyme and/or osmolyte usage (Bedford, 2000; Cronje, 2007; Kettunen et al., 2001). Apart from the direct feed efficiency implication of reduced digestion and absorption, the through flow of undigested nutrients impacts downstream gut ecology (Gidenne, 1997). Undigested protein is strongly inflammatory and provides a competitive advantage to the proteolytic organisms, like Clostridium perfringens, in the caecae (Klipper et al., 2001,, 2004; Lillehoj and Trout, 1996). Potentially toxic compounds such as ammonia, amines, phenols and indoles, are generated by the proteolytic and ureolytic activity of the caecal flora on undigested nutrients that make their way through to the caecal pouches. These toxic compounds affect flora ecology (Gidenne, 1997) . The effect of this is likely more pronounced with soluble nutrients because liquids pass through the digestive tract 15% faster than solids (Klipper et al., 2004; Sklan et al., 1975; Sklan and Hurwitz, 1980).

The organisms of the lower gastrointestinal tract are normally kept in check by intense competition for a limited source of nutrients (Zinser and Kolter, 2004). Any factor that reduces digestion efficiency in the upper gastrointestinal tract or increases nitrogen turnover, could potentially alter caecal ecology. Urine (uric acid) and feed (undigested protein) nitrogen are used by caecal flora to synthesise microbial protein (Bjornhag and Sperber, 1977) and volatile fatty acids (VFA) formed during uric acid degradation, have antibacterial activity (Annison et al., 1968; Cherrington et al., 1990; Cherrington et al., 1991; Davidson, 2001; Sudo and Duke, 1980). Since caecal ecology is affected by protein through-flow, exogenous protein enzymes can be used to help stabilize caecal flora communities. The amount of protein nitrogen reaching the caeca can be further reduced if nutrient credit allocated permits a reduction in dietary protein (Schang and Azacona, 2003).

Conclusion

Strategies to improve gut health in commercial operations need to be cost effective, sustainable, farm specific and holistic. Intervention / product selection needs to be science based but practical and each intervention must address the specific objective for its inclusion. It is possible to enhance gut health and improve productivity by seeding of the hatchling gut with favorable flora, early modification of the gut environment to promote climax flora development, pathogen exclusion (competitive and selective), immune modulation, and ingredient/nutrient management.

References

Anderson, D., McCracken, V., Aminov, R., Simpson, J., Mackie, R., Verstegen, M. and Gaskins, H. (2000) Gut microbiology and growth-promoting antibiotics in swine. Nutritional Abstracts and Reviews Series B: Livestock Feeds and Feeding, 70, 101-108.
Annison, E. F., Hill, K. J. and Kenworthy, R. (1968) Volatile fatty acids in the digestive tract of the fowl. Br J Nutr, 22, 207-16.
Avellaneda, G., Lu, J., Liu, T., Lee, M., Holfacre, C. and Maurer, J. (2003) The Impact Of Growth-Promoting Antibiotics On Total Poultry Microbiota As Well As Enterococcus Population Present On Poultry Carcass. Congress of the World Veterinary Poultry Association July Program and Abstracts. Poultry Disease Research Center, University of Georgia.
Bedford, M. (1996) Interaction between ingested feed and the digestive system in poultry. Journal of Applied Poultry Research, 5, 86-95.
Bedford, M. (2000) Removal of antibiotic growth promoters from poultry diets: implications and strategies to minimise subsequent problems. World's Poultry Science Journal. Volume Number., 56.
Bjornhag, D. and Sperber, I. (1977) Transport of various food components through the digestive tract of turkeys, geese and guinea fowl. Swedish Journal of Agricultural Science, 7, 57-66.
Chaing, S. and Hseih, W. (1995) Effect of direct-fed microorganisms on broiler growth performanceand litter ammonia level. Asian Australian Journal of Animal Science, 1995, 159-162.
Cherrington, C. A., Hinton, M. and Chopra, I. (1990) Effect of short-chain organic acids on macromolecular synthesis in Escherichia coli. J Appl Bacteriol, 68, 69-74.
Cherrington, C. A., Hinton, M., Mead, G. C. and Chopra, I. (1991) Organic acids: chemistry, antibacterial activity and practical applications. Adv Microb Physiol, 32, 87-108.
Choct, M., Naylor, A. J. and Reinke, N. (2004) Selenium supplementation affects broiler growth performance, meat yield and feather coverage. Br Poult Sci, 45, 677-83.
Classen, H. L. (1996) Cereal grain starch and exogenous enzymes in poultry diets. Animal Feed Science Technology, 62, 21-27.
Collett, S. (2003) The Value of Understanding Total Productivity. . In Lyons, T. and Jacques, K. (eds.) Nutritional Biotechnology In The Feed and Food Industries Proceedings of Alltech's 20th Annual Symposium. Nicholasville, KY, Nottingham University Press, pp. 85-91.
Collett, S. and Dawson, K. (2001) Alternatives to subtherapeutic antibiotics: What are the options? How effective are they?2nd International Poultry Broiler Nutritionist's Conference. Poultry beyond 2005. Sheraton Rotorua, New Zealand.
Collier, C. T., Hofacre, C. L., Payne, A. M., Anderson, D. B., Kaiser, P., Mackie, R. I. and Gaskins, H. R. (2008) Coccidia-induced mucogenesis promotes the onset of necrotic enteritis by supporting Clostridium perfringens growth. Vet Immunol Immunopathol, 122, 104-15.
Collier, C. T., van der Klis, J. D., Deplancke, B., Anderson, D. B. and Gaskins, H. R. (2003) Effects of tylosin on bacterial mucolysis, Clostridium perfringens colonization, and intestinal barrier function in a chick model of necrotic enteritis. Antimicrobial Agents Chemotherapy, 47, 3311-7.
Cooper, B. T. (1984) Small intestinal permeability in clinical practice. J Clin Gastroenterol, 6, 499-501.
Cronje, P. (2007) Gut Health, Osmoregulation and resilience to heat stress in poultry. 19th Annual Australian Poultry Science Symposium. Sydney, New South Wales, pp. 9-13.
Davidson, P. (2001) Chemical preservatives and natural antimicrobial compounds. In
Doyle, M., Beuchat, L. and Montville, T. (eds.) Food Microbiology - Fundamentals and Frontiers. 2nd edn. Washington, DC, American Society for Microbiology, pp. 593-627.
Denbow, D. (2000) Gastrointestinal Anatomy and Physiology. In Wihittow, G. (ed.) Sturkies Avian Physiology. 5th edn. New York, Academic Press, pp. 299-325.
Edens, F. W., Parkhurst, C. R., Casas, I. A. and Dobrogosz, W. J. (1997) Principles of ex ovo competitive exclusion and in ovo administration of Lactobacillus reuteri. Poult Sci, 76, 179-96.
England, J., Watkins, S., Saleh, E., Waldroup, P., Casas, I. and Burnham, D. (1996) Effects of Lactobacillus reuteri on live performance and intestinal development of male turkeys. Journal of Applied Poultry Research, 5, 311-324.
Ferket, P., Parks, C. and Grimes, J. (2002) Benefits of dietary antibiotic and mannanoligosaccharide supplementation for poultry. Multi State Poultry Meeting. Ohio.
Ferket, P. R., Quershi, M. A. and Edens, F. W. (1999) Trace minerals in immunity and stress in poultry. 60th Minnesota Nutrition Conference and Zinpro Technical Symposium. Bloomington, Minnesota., University of Minnesota Extension Service, pp. 21-37.
Ferket, P. R. and Qureshi, M. A. (1992) Performance and immunity of heat-stressed broilers fed vitamin- and electrolyte-supplemented drinking water. Poultry Science, 71, 88-97.
Fernandez, F., Hinton, M. and Van Gils, B. (2002) Dietary mannan-oligosaccharides and their effect on chicken caecal microflora in relation to Salmonella Enteritidis colonization. Avian Pathology, 31, 49-58.
Finucane, M., Dawson, K. A., Spring, P. and Newman, K. E. (1999) Effects of Mannanoligosaccharide and BMD on Gut Microflora of Turkey Poults. Poultry Sci., 78 (Suppl. 1), 77.
Fritsche, K. L., Cassity, N. A. and Huang, S. C. (1991) Effect of dietary fat source on antibody production and lymphocyte proliferation in chickens. Poult Sci, 70, 611-7.
Fukata, T., Hadate, Y., Baba, E. and Arakawa, A. (1991) Influence of bacteria on Clostridium perfringens infections in young chickens. Avian Dis, 35, 224-7.
Geyra, A., Uni, Z. and Sklan, D. (2001) Enterocyte dynamics and mucosal development in the posthatch chick. Poult Sci, 80, 776-82.
Gidenne, T. (1997) Caeco-colic digestion in the growing rabbit: impact of nutritional factors and related disturbances. Livestock Production Science, 51.
Giron, J. A., Torres, A. G., Freer, E. and Kaper, J. B. (2002) The flagella of enteropathogenic Escherichia coli mediate adherence to epithelial cells. Mol Microbiol, 44, 361-79.
Grimble, R. F. (2001) Nutritional modulation of immune function. Proceedings of Nutrition Society, 60, 389-97.
Guarner, F. and Malagelada, J. R. (2003) Gut flora in health and disease. Lancet, 361, 512-9.
Hofacre, C., Beacorn, T., Collett, S. and Mathis, G. (2003) Using competitive Exclusion, Mannan-Oligosaccharide and other Intestinal Products to Control Necrotic Enteritis. J.Appl.Poultry Res., 12, 60 - 64.
Hofacre, C. L., Froyman, R., Gautrias, B., George, B., Goodwin, M. A. and Brown, J. (1998) Use of Aviguard and other intestinal bioproducts in experimental Clostridium perfringens-associated necrotizing enteritis in broiler chickens. Avian Dis, 42, 579-84.
Hofacre, C. L. L., M.D., and Maurer, J.J. (2003) Enhancing Microflora and its Effects of Clostridium. 2003 37th National Meeting on Poultry Health A Processing. October 9-11th, Ocean City Maryland., 106-108.
Hooper, L. V. and Gordon, J. I. (2001) Commensal host-bacterial relationships in the gut. Science, 292, 1115-8.
Idris, U., Lu, J. I., Lee, M., Sanchez, S., Hofacre, C. and Maurer, J. (2003) Factors Affecting Epidemiology Of Antibiotic-Resistant Campylobacter Jejuni And Campylobarcter Coli. Program and Abstracts, Congress of the World Veterinary Poultry Association.
Kelly, D., Campbell, J. I., King, T. P., Grant, G., Jansson, E. A., Coutts, A. G., Pettersson, S. and Conway, S. (2004) Commensal anaerobic gut bacteria attenuate inflammation by regulating nuclear-cytoplasmic shuttling of PPAR-gamma and RelA. Nat Immunol, 5, 104-12.
Kettunen, H., Peuranen, S. and Tiihonen, K. (2001) Betaine aids in the osmoregulation of duodenal epithelium of broiler chicks, and affects the movement of water across the small intestinal epithelium in vitro. Comp Biochem Physiol A Mol Integr Physiol, 129, 595-603.
Klasing, K. C. (1998) Nutritional modulation of resistance to infectious diseases. Poult Sci, 77, 1119-25.
Klasing, K. C. and Barnes, D. M. (1988) Decreased amino acid requirements of growing chicks due to immunologic stress. Journal of Nutrition, 118, 1158-64.
Klasing, K. C. and Johnstone, B. J. (1991) Monokines in growth and development. Poultry Science, 70, 1781-9.
Klasing, K. C., Laurin, D. E., Peng, R. K. and Fry, D. M. (1987) Immunologically mediated growth depression in chicks: influence of feed intake, corticosterone and interleukin-1. Journal of Nutrition, 117, 1629-37.
Klipper, E., Sklan, D. and Friedman, A. (2000) Immune responses of chickens to dietary protein antigens. I. Induction of systemic and intestinal immune responses following oral administration of soluble proteins in the absence of adjuvant. Veterinary Immunology and Immunopathology, 74, 209-23.
Klipper, E., Sklan, D. and Friedman, A. (2001) Response, tolerance and ignorance following oral exposure to a single dietary protein antigen in Gallus domesticus. Vaccine, 19, 2890-7.
Klipper, E., Sklan, D. and Friedman, A. (2004) Maternal antibodies block induction of oral tolerance in newly hatched chicks. Vaccine, 22, 493-502.
Korver, D. and Klasing, K. (1995) n-3 polyunsaturated fatty acids improve growth rate of broiler chickens and decrease interleukin-1 production. Poultry Science, 74:, 15.
Korver, D. R. and Klasing, K. C. (1997) Dietary fish oil alters specific and inflammatory immune responses in chicks. J Nutr, 127, 2039-46.
Korver, D. R., Roura, E. and Klasing, K. C. (1998) Effect of dietary energy level and oil source on broiler performance and response to an inflammatory challenge. Poult Sci, 77, 1217-27.
Korver, D. R., Wakenell, P. and Klasing, K. C. (1997) Dietary fish oil or lofrin, a 5- lipoxygenase inhibitor, decrease the growth-suppressing effects of coccidiosis in broiler chicks. Poult Sci, 76, 1355-63.
Kubena, L. F., Bailey, R. H., Byrd, J. A., Young, C. R., Corrier, D. E., Stanker, L. H. and Rottinghaust, G. E. (2001) Cecal volatile fatty acids and broiler chick susceptibility to Salmonella typhimurium colonization as affected by aflatoxins and T-2 toxin. Poult Sci, 80, 411-7.
La Ragione, R. M., Narbad, A., Gasson, M. J. and Woodward, M. J. (2004) In vivo characterization of Lactobacillus johnsonii FI9785 for use as a defined competitive exclusion agent against bacterial pathogens in poultry. Lett Appl Microbiol, 38, 197-205.
Lilijebjelke, K., Hofacre, C., Liu Tongrui and Maurer, J. (2003) Molecular Epidemiology of Salmonella on Poultry Farms In NE Georgia. Program and Abstracts, Congress of the World Veterinary Poultry Association.
Lillehoj, H. S. and Trout, J. M. (1996) Avian gut-associated lymphoid tissues and intestinal immune responses to Eimeria parasites. Clin Microbiol Rev, 9, 349-60.
Lu, J., Idris, U., Harmon, B., Hofacre, C., Maurer, J. J. and Lee, M. D. (2003) Diversity and succession of the intestinal bacterial community of the maturing broiler chicken. Appl Environ Microbiol, 69, 6816-24.
Mohan, B., Kadirvel, R., Natarajan, A. and M, B. (1996) Effect of probiotic supplementation on growth, nitrogen utilization and serum cholesterol in broilers. British Poultry Science, 37.
Neish, A. S. (2002) The gut microflora and intestinal epithelial cells: a continuing dialogue. Microbes Infect, 4, 309-17.
Nurmi, E. and Rantala, M. (1973) New aspects of Salmonella infection in broiler production. Nature, 241, 210-1.
Owings, W., Reynolds, D., Hasiak, R. and Ferket, P. (1990) Influence of dietary supplementation with Streptococcus faecium M-74 on broiler body weigt, feed conversio, carcass characteristics and intestinal microbial colonization. Poultry Science, 69, 1257-1264.
Parks, C. W., Grimes, J. L., Ferket, P. R. and Fairchild, A. S. (2001) The effect of mannanoligosaccharides, bambermycins, and virginiamycin on performance of large white male market turkeys. Poult Sci, 80, 718-23.
Petrof, E. O., Ciancio, M. J. and Chang, E. B. (2004) Role and regulation of intestinal epithelial heat shock proteins in health and disease. Chin J Dig Dis, 5, 45-50.
Qureshi, M. A., Hussain, I. and Heggen, C. L. (1998) Understanding immunology in disease development and control. Poultry Science, 77, 1126-9.
Refstie, S., Svihus, B., Shearer, K. and Storebakken, T. (1999) Nutrient digestibility in Atlantic salmon and broiler chickens related to viscosity and non-starch polysaccharide content in different soyabean products. Animal Feed Science Technology, 79, 331-345.
Ricke, S. C. (2003) Perspectives on the use of organic acids and short chain fatty acids as antimicrobials. Poult Sci, 82, 632-9.
Rosen, G., Vol. II, p141. (1996) Proceedings of the World's Poultry Science Society. New Delhi., pp. 141.
Rosen, G. D. (1995) Antibacterials in poultry and pig nutrition. In Biotechnology in the Animal Feeds and Animal Feeding Ed by R.J. Wallace and A. Chesson. VCH Verlagsgesellschaft mbH D-69461 Weinheim. 8, 143-172.
Sansonetti, P. J. and Di Santo, J. P. (2007) Debugging how bacteria manipulate the immune response. Immunity, 26, 149-61.
Savage, T., Cotter, P. and Zakrzewska, E. (1996) The effect of feeding a mannanoligosaccharide on immunoglobulin plasma IgA and bile IgA of Wrolstad MW male turkeys. Poultry Sci., 75(Suppl. 1), 143.
Schang, M. J. and Azacona, J. O. (2003) Natural enzyme applications to optimize animal performance. In Lyons, T. P. and Jacques, K. A. (eds.) Nutritional Biotechnology In The Feed and Food Industries Proceedings of Alltech's 20th Annual Symposium. Nicholasville, KY, Nottingham University Press.
Schildknect, E., Rakebrand, L., Jensen, L. and Skinner (2003a) Changes In Anticoccidial Sensitivity Profiles Of Coccidia From Broiler Chickens Raised On Built-Up Litter For Eight Production Cycles Following A Coccidiosis Challenge. International Poultry Scientific Forum Abstracts. pp. 10.
Schildknect, E., Rakebrand, L., Jensen, L. and Skinner (2003b) Changes In Live Performance Of Broiler Chickens Raised On Built-Up Litter For Eight Production Cycles Following A Coccidiosis. International Poultry Scientific Forum Abstracts. Atlanta, Georgia.
Sell, J., Eastwood, J. and Mateos, G. (1983) Influence of supplemental fat on metabolizable energy and ingesta transit time in laying hens. Nutrition Rep. Intern., 28, 487-495.
Sharon, N. and Lis, H. (1993) Carbohydrates in cell recognition. Sci Am, 268, 82-9.
Sibbald, I. R. (1979) Passage of feed through the adult rooster. Poult Sci, 58, 446-59.
Siegel, H. (1994) Stress and Immunity. Proceedings of the 9th European Poultry Conference. Glasgow UK, pp. 22-125.
Sklan, D. (2001) Development of the digestive tract of poultry. World's Poultry Science Journal, 57.
Sklan, D., Dubrov, D., Eisner, U. and Hurwitz, S. (1975) 51Cr-EDTA, 91Y and 141Ce as nonabsorbed reference substances in the gastrointestinal tract of the chicken. J Nutr, 105, 1549-52.
Sklan, D. and Hurwitz, S. (1980) Protein digestion and absorption in young chicks and turkeys. J Nutr, 110, 139-44.
Spring, P. (1996) Effects of mannanoligosaccharide on different caecal parameters and on the attachment of enteric pathogens in poultry. Zurich, Swiss Fed. Inst Technology.
Stavric, S., Gleeson, T., Blanchfield, B. and Pivnick, H. (1987) Effect of environmental temperature on the susceptibility of young chickens to
Salmonella typhimurium. Australian Veterinary Journal, 55, 413.
Sudo, S. and Duke, G. (1980) Kinetics of absorption of volatile fatty acids from the ceca of domestic turkeys. Comp. Biochem. Physiol. 67, 231-237.
Surai, P. (2002) Selenium in poultry nutrition: a new look at an old element. 1 Antioxidant properties, deficiency and toxicity. World's Poultry Science Journal, 58B, 333-347.
Swamy, H. V., Smith, T. K., Cotter, P. F., Boermans, H. J. and Sefton, A. E. (2002a) Effects of feeding blends of grains naturally contaminated with Fusarium mycotoxins on production and metabolism in broilers. Poult Sci, 81, 966-75.
Swamy, H. V., Smith, T. K., MacDonald, E. J., Boermans, H. J. and Squires, E. J. (2002b) Effects of feeding a blend of grains naturally contaminated with Fusarium mycotoxins on swine performance, brain regional neurochemistry, and serum chemistry and the efficacy of a polymeric glucomannan mycotoxin adsorbent. J Anim Sci, 80, 3257-67.
Sword, J. T., Pope, A. L. and Hoekstra, W. G. (1991) Endotoxin and lipid peroxidation in vivo in selenium- and vitamin E-deficient and -adequate rats. Journal of Nutrition, 121, 251-7.
Uni, Z., Geyra, A., Ben-Hur, H. and Sklan, D. (2000) Small intestinal development in the young chick: crypt formation and enterocyte proliferation and migration. Br Poult Sci, 41, 544-51.
Uni, Z., Noy, Y. and Sklan, D. (1999) Posthatch development of small intestinal function in the poult. Poult Sci, 78, 215-22.
Washburn, K. W. (1991) Efficiency of feed utilization and rate of feed passage through the digestive system. Poult Sci, 70, 447-52.
Weurding, R. E., Veldman, A., Veen, W. A., van der Aar, P. J. and Verstegen, M. W. (2001) Starch digestion rate in the small intestine of broiler chickens differs among feedstuffs. J Nutr, 131, 2329-35.
Zinser, E. R. and Kolter, R. (2004) Escherichia coli evolution during stationary phase. Research Microbiology, 155, 328-36.

This presentation was given at the 9º Seminario de Actualización Avícola de AMEVEA, Entre Ríos, Argentina in September 2010. Engormix thank the author and the organizing committee for the contribution.
Related topics:
Authors:
Dr. Steve Collett
University of Georgia
Recommend
Comment
Share
Amir Attar
Javaneh khorasan
12 de enero de 2011
thanks to author because i think the gut is related to profitability in poultry farm and managing of it by nutrition is so important
Recommend
Reply
Alloui Nadir
13 de noviembre de 2010
Very good review article
Recommend
Reply
Richard Fan
14 de junio de 2013

yes, Intestinal Environment is very import for the animals, it is usually been destroyed by the use of antibiotics, in China, we use probiotics to reduce the use of antibiotics, so that the intestinal environment will be repaired, feed conversion ratio be improved and more profit will be made. Thanks.

Recommend
Reply
Dr. Manoranjan Sharma
14 de marzo de 2011
The article is a informative and useful one to understand more on the gut health management and the parameters while formulating feed.
Recommend
Reply
Dr. Zaib Ur Rehman
17 de enero de 2011
Very good article on, the different factors affecting the intestinal health of broilers. Author also discusses genetic parameters affecting the intestinal micro flora of broiler. This article will help the nutritionist during feed formulation
Recommend
Reply
Dr G Naresh Kumar
29 de diciembre de 2010
wonderful, indeed i would like to thank the author as he explained the gut related factors in detailed .
Recommend
Reply
Waseem Alshible
14 de diciembre de 2010
Thank you For your good Article
Recommend
Reply
Hafiz Imran Shakoor
24 de noviembre de 2010
Very good review on gut heath management. Good gut heath is main target, whether it is achieved through growth promoters (AGP, non AGPs, correct ingredient selection or through modulating gut micro flora.
Recommend
Reply
Dr Jaydip Mulik
13 de noviembre de 2010
I really salute the author as he has explained the gut health related paremeters/factors in depth. Author has elaborately covered all the factors influencing the Gut health.I hereby requesting the Author to update us on the different effective tools to maintain the gut healthy in commercial chicks in deep.
Recommend
Reply
Arshaq Ramzee
11 de noviembre de 2010
Authos has dicussed some very important points which are esstentail for good digestion especially the genetic aspect of the present broiler breeds that has resulted in some minor changes in digestive tract. Another important point is undigestible protein and its role in gut health. In fact a great article with some good points to help a nutritionists to formulate a good feed.
Recommend
Reply
Profile picture
Would you like to discuss another topic? Create a new post to engage with experts in the community.
Join Engormix and be part of the largest agribusiness social network in the world.