Explore
Communities in English
Advertise on Engormix

Towards the control of necrotic enteritis in broiler chickens with in-feed antibiotics phasing-out worldwide

Published: July 27, 2017
By: Shawkat A. M'Sadeq 1, Shubiao Wu 1, Robert A. Swick 1, Mingan Choct 1,2. / 1 School of Environmental and Rural Science, Armidale 2351, Australia; 2 Poultry Cooperative Research Centre, Armidale 2351, Australia.
Summary

Poultry production has undergone a substantial increase compared to the livestock industries since 1970. However, the industry worldwide is now facing challenges with the removal of in-feed antibiotics completely or gradually, as the once well-controlled poultry diseases have re-emerged to cause tremendous loss of production. Necrotic enteritis (NE) is one of the most important diseases which costs the industry over two billion dollars annually. In this paper, we review the progress on the etiology of NE and its control through dietary modifications, pre- and probiotics, short chain fatty acids, and vaccination. The other likely measures resulted in the most advances in the toxin characterization are also discussed. Vaccine strategies may have greater potential for the control of NE mainly due to clearer etiology of NE having been elucidated in recent years with the identification of necrotic enteritis toxin B-like (NetB) toxin. Therefore, the use of alternatives to in-feed antibiotics with a better understanding of the relationship between nutrition and NE, and limiting exposure to infectious agents through biosecurity and vaccination, might be a tool to reduce the incidence of NE and to improve gut health in the absence of in-feed antibiotics. More importantly, the combinations of different measures may achieve greater protection of birds against the disease. Among all the alternatives investigated, prebiotics, organic acids and vaccination have shown improved gastrointestinal health and thus, have potential for the control of NE.

 
Introduction
Poultry production has undergone a substantial increase compared to other animal food-producing industries since 1970 (Yegani and Korver, 2008). Improvements in housing, genetic selection for growth rate, and advances in feed formulation achieved by matching nutrient requirements of the birds and nutrient contents of the feedstuffs, have resulted in higher meat yield, improved feed conversion and lower mortality rates (Choct et al., 1999; Cooper and Songer, 2009). As growth rate and feed conversion ratio improve, the birds' nutrition and health care are becoming more demanding (Choct et al., 1999). The nutritional and health status of poultry are interlinked with gut health which includes immune system, gut microbial balance and macro and micro-structural integrity of the gut. The health of the gastrointestinal tract (GIT) affects digestion, absorption and metabolism of nutrients, disease resistance and immune response (Kelly and Conway, 2001; Yegani and Korver, 2008). The disturbances of these processes can result in enteric diseases (Dekich, 1998). This makes it important to pay attention to gastrointestinal health; usually any slight change is mostly accompanied by disruption of gut health and thus overall performance.
Enteric diseases are one of the most important problems in the poultry industry because of high economic losses due to decreased weight gain, increased mortality rates, worse feed conversion ratio, greater medication costs, and increased risk of contamination of poultry products for human consumption (Timbermont et al., 2011). Several pathogens including viruses, bacteria, macroparasites and other infectious and non-infectious agents are reported as possible causes of enteric diseases either alone or in synergy (Reynolds, 2003). Many conditions have been associated with gastrointestinal problems such as diarrhea, wet droppings, dysbacteriosis, intestinal colibacillosis, malabsorption syndrome, coccidiosis and necrotic enteritis (NE). Enteric disorders are frequently associated with an overgrowth of Clostridium perfringens. Infections with this bacterium in poultry can cause NE, necrotic dermatitis, cholangiohepatitis, as well as gizzard erosion (Hafez, 2011). NE is the most common clostridial enteric disease in poultry, which typically occurs in broiler chickens (Cooper et al., 2013). NE is characterized by necrosis and inflammation of the GIT with a significant decline in growth performance and, in clinical cases, a massive increase in flock mortality. The total cost of NE outbreaks globally is estimated to be over $2 billion annually (Van der Sluis, 2000).
Antibiotics have been used as an effective tool to improve animal performance, by selectively modifying the gut microflora, decreasing bacterial fermentation, reducing thickness of the intestinal wall and suppressing bacterial catabolism. All these are important to improve health, nutrient availability and growth performance (Carlson and Fangman, 2000). Hence, dietary antimicrobials not only improve poultry growth and feed conversion efficiency, but also control enteric disease outbreak (Kim et al., 2011). The use of antibiotics in feed and for treating animals is second only to the medical use (Dahiya et al., 2006). It has been estimated that 11.15 million kg of antibiotics are used in animal feed in the USA alone each year (Union of Concerned Scientists, 2001) and 4.7 million kg or 35% of all antibiotics administered in Europe in 1999 were used in animal feed (European Commission Directorate General XXIV Directorate B, 1999). Hence, antibiotics have come under increasing scrutiny by government regulators, scientists and consumers because of the emergence of antibiotic-resistant “superbugs”. European countries have now prohibited the use of in-feed antibiotics in poultry feed (Van Immerseel et al., 2004). Without the use of in-feed antibiotics, the Animal Health Institute of America has estimated that the USA will require an additional 12 million pigs, 23 million cattle and 452 million chickens to reach the levels of production attained by the current practices (Dahiya et al., 2006).
With a ban of in-feed antibiotics in European countries, the incidence of NE has increased on the broiler farms of these countries (Casewell et al., 2003; Hofacre et al., 2003). At the same time, the focus on alternative strategies has increased to secure animal health and thus the efficiency of livestock production. These alternative strategies include modulation of gut microflora, augmentation of immune response and pathogen reduction through management, vaccination, nutritional strategies and feed additives.
 
2. Necrotic enteritis in broiler chickens
2.1. Epidemiology
NE has a high mortality rate with severe economic losses. The disease has been reported in many countries, including the United Kingdom (Parish, 1961), Australia (Nairn and Bamford, 1967), Canada (Helmboldt and Bryant, 1971) and France (Casewell et al., 2003). The primary causative agent of NE is C. perfringens and the source of C. perfringens is ultimately the chickens themselves (Cooper and Songer, 2009). Outbreaks of NE in poultry, past and present, have been associated with C. perfringens contamination of the chickens' feed (Eleazer and Harrell, 1976; Hofacre et al., 1986; Nairn and Bamford, 1967). Occurrences of NE are also affected by season (Kaldhusdal and Skjerve, 1996), dietary restriction (Olkowski et al., 2006), bedding on high fiber litter (Branton et al., 1997) and management-related stress (Craven, 2000).
NE usually occurs in broiler chicks at 2e6 weeks of age (Cooper and Songer, 2010). Generally, NE is not typically known as a seasonal disease, although the occurrences of NE between different latitudes appear to contradict this. Kaldhusdal and Skjerve (1996) suggested that univariate regression analysis in south-eastern Norway indicated that NE occurred more often during the months OctobereMarch than during the months AprileSeptember, whilst in Canada it mostly appeared in JulyeOctober (Long, 1973). In the United Kingdom, the peak incidence of NE is during winter with a lower incidence during the warmer seasons (Hermans and Morgan, 2007).
 
2.1.1. C. perfringens as a causative agent of NE
C. perfringens is a Gram-positive, spore forming anaerobic bacterium, able to produce several enzymes and toxins responsible for NE symptoms and lesions (Van Immerseel et al., 2004). C. perfringens can be found in poultry litter, feces, soil, dust and in healthy bird intestinal contents (Dahiya et al., 2005). It is expected that small numbers of C. perfringens are resident, transiently or permanently, in the GIT of most bird species (Cooper and Songer, 2009). When poultry meat is analyzed for C. perfringens, in some cases up to 84% of meat samples are positive (Craven et al., 2001b). It is reported that colonization or contamination of poultry by C. perfringens occurs during the early life of the animal, and can commence in the hatchery environment (Van Immerseel et al., 2004). C. perfringens is found in eggshell, paper pads and chicken dander in the hatchery (Craven et al., 2001a). Craven et al. (2003) indicated that the C. perfringens contamination found in broiler carcasses can begin in the breeder hen, and be transmitted through the hatchery and growing area. Free-living birds such as crows have high counts of C. perfringens in their intestinal droppings, which indicate that wild birds also suffer from NE (Asaoka et al., 2004). Craven et al. (2001b) found that swabs taken from poultry farms showed an incidence of C. perfringens from a variety of sources, including live flies, walls, dirt outside the entrance, fans, floor, nipple-drinker drip-cups, water pipes, litter material, chick delivery-tray liners and boots of farm staff before chicks were placed. Even feed samples taken after 2 weeks following bird placement had an incidence of C. perfringens. This indicates that different sources and strains of C. perfringens can colonize in the birds and produce NE. Additionally, NE has been reported in a variety of wild birds including ducks (Chakrabarty et al., 1983), wild geese (Wobeser and Rainnie, 1987), Laysan albatross (Work et al., 1998), mute swan (Duff et al., 2011), bobwhite quail (Shivaprasad et al., 2008) and wild crows (Asaoka et al., 2004; McOrist and Reece, 1992). With the gradual move from caged or housed chicken raising to free range worldwide, occurrence of NE in free range flocks becomes a major concern as the domestic chickens are prone to be infected through wild birds. This poses eminent challenge in the management and care of free range chickens which may be less than when they are raised in housed conditions.
 
2.1.2. Toxins to produce NE
C. perfringens is able to produce several types of toxins, while individual strains only produce a subset of these toxins (Van Immerseel et al., 2008). C. perfringens strains (A, B, C, D and E) are classified according to the production of four major extracellular toxins alpha (a), beta (b), epsilon (3) and iota (i), while various strains can also produce other toxins, including b 2 toxin, perfringolysin O [q-toxin], collagenase [k-toxin], enterotoxin, theta toxin etc. (Petit et al., 1999). Type A produces a-toxin, type B produces a, b and 3 toxins, type C produces a and b toxins, type D produces a and 3 toxins and type E produces a and i toxins (Brynestad and Granum, 2002). In poultry, NE has been reported being caused by type A and to a lesser extent by type C (Engström et al., 2003).
Alpha-toxin has been reported to be the main virulence factor in poultry NE (Van Immerseel et al., 2008) with phospholipase C and  sphingomyelinase activity (Flores-Díaz and Alape-Girón, 2003). Although this toxin was believed to be the main virulent factor in the pathogenesis of NE for long time, the importance of a-toxin in the pathogenesis of NE has been questionable. Early studies on the factors inducing NE showed that intro-duodenal infusions of both large volumes of C. perfringens broth culture and crude toxins (Al- Sheikhly and Truscott, 1977) into chickens, resulted in typical NE lesions. From these studies the authors concluded that a-toxin was the main toxin produced by C. perfringens, and then it was considered the most virulent factor in the pathogenesis of NE. Fukata et al. (1988) found that 21 out of 56 germ-free chickens, inoculated with either purified a-toxin or a supernatant of broth cultures of C. perfringens died after inoculation, whereas no bird died after receiving a culture supernatant neutralized by antialpha- toxin serum. Later on, Hofshagen and Stenwig (1992) found a significantly higher amount of a-toxin in isolates from birds with NE compared with isolates from birds without NE. In a more recent study, Cooper and Songer (2009) suggested that immunization with a-toxin gave substantial protection against NE and Rehman et al. (2009) concluded that a-toxin can damage the intestinal mucosal barrier. Contradicting these studies, an in vitro study (Gholamiandekhordi et al., 2006) demonstrated no difference in atoxin production between C. perfringens isolated from healthy flocks and those isolated from NE outbreaks flocks. The most convincing evidence that a-toxin of C. perfringens was not a major factor in producing NE in chickens came from a study using an atoxin negative mutant of C. perfringens generated from a virulent strain isolated from NE infected birds. This study demonstrated that the constructed mutant negating a-toxin gene induced the same type of NE lesion as that of the wild type strain (Keyburn et al., 2006). Other studies also argue against the role of a-toxin in NE. Furthermore, histological analysis of tissue in early stages of NE lesion development is inconsistent with the a-toxin phospholipase C or sphingomyelinase activities (Olkowski et al., 2008).
Recently, a new pore-forming toxin named necrotic enteritis toxin B-like (NetB) was discovered by Keyburn et al. (2008) as a virulence factor for the development of NE. NetB toxin showed limited amino acid similarity with pore-forming b-toxin (38% identity) and the a-toxin of Staphylococcus aureus (31% identity). This toxin was identified from C. perfringens type A strain isolated from infected chickens. Both recombinant and native NetB showed cytotoxic activities against the chicken leghorn male hepatoma cell line and the mechanism of action appeared to include the formation of a hydrophilic pore in the cell membrane with a functional diameter of 1.6e1.8 nm. A complemented NetB mutant was able to cause necrotic lesions in the gut of experimentally infected broilers. Additional evidence for the role of NetB in disease comes from the observation that chickens suffering from NE carry the NetB gene and produce highly conserved NetB toxin (Keyburn et al., 2010). This has been confirmed by surveys in North America, where all C. perfringens isolates from birds infected with NE carried the NetB gene, while only a small percentage of isolates from healthy birds carry the gene (Chalmers et al., 2008; Martin and Smyth, 2009). Keyburn et al. (2010) reported that NetB positive C. perfringens strain isolates from diseased birds were able to produce NetB in vitro and therefore only strains producing NetB were able to induce disease. This was supported by the report of Smyth and Martin (2010) where all NetB positive isolates induced NE in challenged birds, whereas none of the NetB negative isolates produced disease. This suggests that the presence of NetB is a critical factor in the development of NE.
 
2.2. Symptoms of necrotic enteritis in poultry
2.2.1. Clinical signs
The clinical form of NE is associated with signs such as ruffled feathers, relative immobility, depression, anorexia, diarrhea and decreased appetite (Al-Sheikhly and Al-Saieg, 1980; Al-Sheikhly and Truscott, 1977; Helmboldt and Bryant, 1971). Wet litter is also sometimes an early indicator of the disease (Riddell and Kong, 1992). Birds displaying clinical signs generally die within a few hours, with mortality rates up to 1% per day (Helmboldt and Bryant, 1971). In the acute form of NE, characterized by a sudden increase in flock mortality, birds usually die without premonitory signs (Kocher and Choct, 2008).
Diarrhea and associated wet litter were noted in the wood shaving litter of birds suffering from NE (Kalshusdal and Hofshagen, 1992). Elwinger and Tegl€of (1991) found a direct correlation between poor litter condition and sticky droppings. In field surveys conducted globally (Van der Sluis, 2000) and in the United Kingdom (Hermans and Morgan, 2003), researchers associated NE with diarrhea or wet litter. Williams (2005) noted that in the United Kingdom, any sudden increase in litter moisture of poultry farms is associated with NE and antibiotic therapy is initiated. Helmboldt and Bryant (1971) suggested that acute NE may be associated with diarrhea or wet litter (Nairn and Bamford, 1967).
 
2.2.2. Subclinical signs
In subclinical forms of NE there is no peak of mortality and no clinical signs are present (Timbermont et al., 2011). Subclinical forms of NE are usually associated with reduced feed intake and weight gain and increased feed conversion ratio (Kalshusdal and Hofshagen, 1992; Lovland and Kaldhusdal, 2001). They are also associated with hepatitis and cholangiohepatitis (Van Immerseel et al., 2004). During subclinical infection, bacteria can reach the portal blood stream and bile duct. Colonization of high numbers of C. perfringens in hepatic tissue result in cholangiohepatitis (Timbermont et al., 2011). Diseased livers are greatly enlarged and have a pale reticular pattern with white or red foci and histopathological lesions characterized by bile duct hyperplasia, cholangitis, fibrinoid necrosis and sometimes focal granulomatous inflammation (Løvland and Kaldhusdal,1999). Onderka et al. (1990) reported that inoculation of birds with C. perfringens produced cholangiohepatitis, enlarged liver, tan-colored liver with red and white foci and edematous gall bladder. During meat inspection at a slaughter house, infected livers were found, without any clinical signs in the flock (Timbermont et al., 2011). Although clinical forms of NE may cause high levels of mortality, the subclinical form of NE is more economically important than the clinical form because it may persist in the flock without any clinical manifestation. Thus, untreated birds suffering from subclinical NE can cause huge economic loses in the poultry production industry (Dahiya et al., 2006).
Gross lesions are usually restricted to the duodenum, jejunum and ileum (Timbermont et al., 2011) and also can occur in the caeca (Van Immerseel et al., 2004). The small intestine of infected birds is friable, dilated, hyperemic, thin walled and filled with gas, and the mucosal surfaces are covered by tan-orange pseudo-membranes and occasional hemorrhages (Broussard et al., 1986; Olkowski et al., 2006). Microscopic examination shows a strong inflammatory reaction to C. perfringens in the early stages of NE. The lamina propria is infiltrated and hyperemic with several inflammatory cells, especially at the interface of the basal domain of enterocytes and lamina propria. These areas are edematous, permitting the extensive disorder of the structural integrity between the enterocytes and the lamina propria (Olkowski et al., 2006). Histopathological examination of later stage NE lesions shows extensive villous necrosis (Broussard et al., 1986), coagulation of necrosis of the mucosa in all segments of the small and large intestine (Olkowski et al., 2006) and a clear line of demarcation between necrotic and viable tissue. An accumulation of heterophilic granulocytes at the junction is seen (Al-Sheikhly and Al-Saieg, 1980; Long, 1973). Hemorrhage or patchy congestion is present over all the lamina mucosa, particularly in the vicinity of the crypts. The crypts are usually misshapen and inflated by pink mucus and necrotic cellular debris (Olkowski et al., 2006).
 
2.3. Predisposing factors for necrotic enteritis
Although C. perfringens is the primary causative agent of NE in poultry, other contributory factors that alter the microenvironment of GIT and create a favorable environment for C. perfringens overgrowth are essential to produce both the clinical and subclinical types of NE. Factors that predispose the bird to NE include co-infection of other parasites most evidently coccidiosis, stress and immunosuppression, and nutrition.
 
2.3.1. Coccidiosis
Coccidiosis is an enteric disease caused in the fowl by numerous Eimeria species. Some species (Eimeria praecox, Eimeria mitis or Eimeria acervulina) produce less severe clinical coccidiosis than others (Eimeria brunetti, Eimeria maxima, Eimeria necatrix, and Eimeria tenella) (Williams, 2005). The intestinal damage caused by coccidia is an essential predisposing factor for NE (Al-Sheikhly and Al-Saieg, 1980; Rodgers et al., 2015; Williams, 2005), allowing C. perfringens overgrowth and production of toxins (Van Immerseel et al., 2008). Intestinal damage during Eimeria infection will result in leakage of plasma proteins into the lumen of the intestinal tract, which is a rich nutrient substrate and favorable for C. perfringens proliferation and toxin production (Van Immerseel et al., 2004). Collier et al. (2008) suggested that coccidial infection induces mucogenesis as a result of a host mucogenic response, providing a growth advantage for C. perfringens.
For these reasons, several studies have used Eimeria spp. In conjunction with C. perfringens to induce NE experimentally. E. maxima, E. acervulina and E. necatrix are known to be the most suitable species to induce NE (Al-Sheikhly and Al-Saieg, 1980; Hofacre et al., 1998; Van Immerseel et al., 2004; Williams, 2005). Eimeria vaccine has also been used to enhance C. perfringens to induce NE (Gholamiandehkordi et al., 2007; Timbermont et al., 2009). The time and dose of administration of the virulent Eimeria or coccidial vaccine are important for inducing experimental NE. The time should not be more than 4e5 days before C. perfringens challenge (Williams et al., 2003; Williams, 2005). The doses of Eimeria administration needed for experimentally inducing NE are different according to the models which have been used. For example, Wu et al. (2014) used a suspension of 5000 sporulated oocysts each of E. maxima and E. acervulina, and 2500 oocysts of E. brunetti; Williams et al. (2003) used 30,000 sporulated oocysts of E. maxima and Gholamiandehkordi et al. (2007) suggested using attenuated coccidial vaccines at 10-times the recommended vaccination doses.
 
2.3.2. Stress and immunosuppression
Any stressful condition in broiler chickens could predispose them to NE, because it could change the intestinal environment in such away that the risk of induction of NE is raised (McDevitt et al., 2006). For example, alteration in feeding regime (moving from a starter diet to a grower diet) in young chicken causes such stress in the GIT. Increases in stocking density are also frequently associated with NE (McDevitt et al., 2006; Tsiouris et al., 2015). Immunosuppressive agents such as infectious bursal disease, Marek's disease, chicken anemia virus and Gumboro disease have been suggested as causing an increase in the severity of NE (Lee et al., 2011; McReynolds et al., 2007; Timbermont et al., 2011; Williams et al., 2003). Indeed, in several studies, infectious bursal disease vaccine has been used as a predisposing factor to experimentally induce NE (Gholamiandehkordi et al., 2007; McReynolds et al., 2004, 2007; Timbermont et al., 2009). This has been done by inoculation with usual doses of infectious bursal disease vaccine (intermediate class) (Gholamiandehkordi et al., 2007; Timbermont et al., 2009) or at 10- times the recommended vaccination dose rate with infectious bursal disease vaccine (intermediate plus) (Nikpiran et al., 2008).
 
2.3.3. Nutritional factors
The key risk factor for the development of NE is alteration of the GIT environment which creates a favorable environment for C. perfringens growth. Diet is now widely recognized as having a strong impact on the incidence of NE in broiler chickens (Annett et al., 2002; Drew et al., 2004). Evidence arising from several studies has shown that there is a relationship between cereal type used in the diet, dietary protein levels and anti-nutritional factors and the incidence of NE (Kalshusdal and Hofshagen,1992; McDevitt et al., 2006; Riddell and Kong, 1992). It is widely believed that a diet with high levels of indigestible water soluble non-starch polysaccharides (NSPs) strongly influences the incidence of NE in broilers. Numerous studies have revealed that a diet comprising cereals such as barley, rye and wheat increase the digesta viscosity and enhance the development of NE as these grains contain high amounts of NSPs such as arabinoxylans or b glucans (Annett et al., 2002; Dahiya et al., 2006; Kalshusdal and Hofshagen, 1992; McDevitt et al., 2006). The NSPs and certain types of starch in the cereals are not digestible by the enzymes present in the bird's GIT (Iji and Tivey, 1998; Juskiewicz et al., 2004). They act as substrates for the gut microflora and provide an opportunity for these organisms, including pathogenic bacteria, to proliferate (Apajalahti et al., 2004; Choct et al., 1996; Iji and Tivey, 1998; McDevitt et al., 2006). As higher NSPs in diets lead to increased digesta viscosity, prolonged transit time and decreased nutrient digestibility (Choct et al., 1999), this may lead to the increased proliferation of C. perfringens, predisposing birds to NE (Annett et al., 2002). The NSPs are also hydrophilic, which encourages birds to drink more water in order to maintain homeostasis. The increased water intake also increases water excretion, affecting litter quality and thus allowing the pathogenic bacteria to proliferate (McDevitt et al., 2006). Furthermore, some NSPs interact with epithelial protein and glycoproteins, increasing mucin secretion from the tissues (Kleessen et al., 2003). This provides an opportunity to pathogenic microorganisms to adhere to the mucin and proliferate (McDevitt et al., 2006).
The level of crude protein, protein source, and the amino acid content of a diet have been thought to be associated with the incidence of NE. Poultry diets with high protein content or those rich in animal protein such as meat and bone meal or fish meal predispose birds to NE (Kaldhusdal and Skjerve, 1996; Williams et al., 2003; Williams, 2005; Wu et al., 2010, 2014). Diets that have a high protein concentration or imbalanced amino acid profiles reduce the digestibility of these compounds in the upper part of digestive system (McDevitt et al., 2006). Thus the indigestible protein in the lower part of GIT acts as substrate for the gut microflora (Timbermont et al., 2011). The fermentation of protein produces unfavorable outcomes such as phenols, thiols, amines, ammonia, indoles and increases the pH of the lower part of the GIT, which encourages the proliferation of pathogenic bacteria such as Clostridium sp. (Juskiewicz et al., 2004; Lan et al., 2005). Another possible explanation of the association between fish or meat meal and NE could be related to their higher zinc, glycine and methionine concentrations. Fish meal is relatively high in zinc (NRC, 1984), glycine and methionine (Dahiya et al., 2007). In an in vitro study, Baba et al. (1992) proposed that dietary zinc increased the production of a-toxin and protected the a-toxin destruction by trypsin. Glycine is an amino acid that stimulates the growth of C. perfringens (Dahiya et al., 2007) and is positively correlated to the C. perfringens population in the intestine (Wilkie et al., 2005).
Other anti-nutritional factors other than NSPs such as lectins, trypsin inhibitors and tannins may also predispose birds to NE. Lectins from wheat and soybeans are proteins and glycoproteins, which interact vigorously with epithelial tissues (Pusztai and Bardocz, 1996) and cause damage, change in microflora population and immune response in animals (Lan et al., 2005; Pusztai and Bardocz, 1996). Lectins alter bacterial attachment to the GIT and change the extent and rates of bacterial growth (Giovannini et al., 1996; McDevitt et al., 2006). Trypsin inhibitors, found in soybean meal, reduce the digestibility of protein and thus increase nitrogen concentration in the lower GIT, which provides suitable conditions for the growth of proteolytic bacteria such as C. perfringens (Clarke and Wiseman, 2005; McDevitt et al., 2006). Tannins are present in many dietary ingredients such as rapeseed meals and beans, and can interact strongly with protein, leading to tissue damage that may predispose birds to NE (McDevitt et al., 2006; Robins and Brooker, 2005). There are also other compounds such as mycotoxins, glucosinolates, alkaloids and polyphenols which interact with bacteria, altering bacterial proliferation (McDevitt et al., 2006) and may play a role in predisposing birds to NE.
The dietary factors play important roles in predisposing the birds to NE infection. However, diets having higher NSPs or containing high protein level may lead to more frequent NE occurrence in broiler chickens. Whereas other nutritional factors such as lectins, trypsin inhibitors and tannins may cause birds to be infected with NE, they are less significant. Attention may need to be paid to reduce levels of NSPs and protein especially animal protein in the diets to minimize the possibility for the flocks to succumb to NE disease.
 
2.3.4. Physical form of diet
The physical form of poultry diets may affect the physiological and morphological characteristics of the GIT (Engberg et al., 2004). Finely ground feed allows C. perfringens to growfaster than coarsely ground feed, which can lead to occurrence of NE in the field (Engberg et al., 2002). Branton et al. (1987) observed that use of a coarsely ground wheat diet decreased mortality from NE to 18.1%, whereas a finely ground wheat diet resulted in mortality of 28.9%. This may be related to that coarsely ground mash stimulates gastric function, including secretion of hydrochloric acid, and simultaneously increases the retention time of feed in the proventriculus and gizzard (Engberg et al., 2002).
 
Measures to control necrotic enteritis post antibiotic era
Antibiotics have played a major role in the suppression of clinical NE (Williams, 2005) and extensive reviews have been performed in the past. However, public concern over the use of in-feed antibiotics and the emergence of antibiotic-resistant bacteria has led many countries to ban the use of dietary antimicrobials. To be able to cope with the removal of antibiotics from animal feed, many studies have been conducted to investigate the possible alternatives to be used to control the diseases in animal production. Herein, we will discuss the development of antibiotics alternatives and their utilization in broiler chicken production. As a result of consumer pressure to reduce in-feed antibiotics in animal feed, Sweden was the first country to ban the use of antimicrobials for growth-promoting purposes (Kocher and Choct, 2008). In 1995, Denmark banned the use of avoparcin and virginiamycin in animal feed (Casewell et al., 2003). In 1997, the European Union restricted the use of some antibiotics in animal feed and followed this by a general ban on the use of all antibiotics in all animal feed in 2006 (Kocher and Choct, 2008). After the banning of sub-therapeutic antibiotics in animal feed, the incidence of NE in poultry farms increased in many European Union countries (Van Immerseel et al., 2004). For example, the incidence of NE in France was 4% in 1995 and increased to 12.4% in 1999 (Drouin, 1999). Similarly, in the USA, who also stopped using in-feed antibiotics, the incidence of NE and other diseases like gangrenous dermatitis, botulism and cholangiohepatitis has increased (Shane, 2004). Thus, the question has now become ‘how to control this disease in the absence of antibiotics’. To investigate a method for controlling NE, the factors that predispose birds to develop NE must be better understood and alternative dietary supplements and management strategies to control NE must be developed.
Based on the literature, there are three basic strategies used to control NE. These strategies are: amplification of immune response, pathogen reduction, and dietary modification and/or use of feed additives (Dahiya et al., 2006). This paper will review numerous strategies for controlling NE, including dietary modification, the use of feed enzymes, probiotics, prebiotics and organic acids, and vaccination strategy will be discussed in the following sections.
 
3.1. Dietary modifications and feed enzymes
Modification of diet and the addition of enzymes cannot provide total protection against NE, but may reduce the risk of NE by improving digestion. For example, dietary cereals such as barley, rye and wheat contain high amounts of arabinoxylans or b glucans, mannans, cellulose, lignin and ingredients, which cannot be digested by poultry. These non-digestible feed constituents increase the digesta viscosity and encourage the development of NE (Dahiya et al., 2006; McDevitt et al., 2006). Also, poultry diets with high protein content, or those rich in animal protein such as meat and bone meal or fish meal predispose birds to NE (Kaldhusdal and Skjerve,1996; Williams et al., 2003; Williams, 2005;Wu et al., 2010, 2014). Thus, reducing indigestible carbohydrates and proteins that predispose birds to NE may decrease the risk of NE outbreak in poultry farms.
It has been demonstrated that inclusion of exogenous enzymes to wheat, barley, oat or rye based diets can significantly decrease digesta viscosity in the small intestine (Bedford and Classen, 1992; Choct et al., 1999). Choct et al. (1999) reported that the addition of xylanase to wheat base diets decreased digesta viscosity and fermentation, increased nutrient digestion and digesta passage, and reduced the amount of nutrients available to the microflora, which, in turn, may reduce the bacterial population in the small intestine. Sinlae and Choct (2000) also demonstrated that dietary supplementation of xylanase reduced numbers of C. perfringens. Jackson et al. (2003) proposed that b mannanase would reduce the severity of challenge by Eimeria spp. and C. perfringens in broiler chickens, and thus reduce NE lesion scores in the intestine. However, supplementation of pentosanase in a wheat based diet did not have any beneficial effect on the susceptibility to NE in broiler chickens. Despite the contradictory results from published studies on the effect of feed enzymes on various bacterial populations, including C. perfringens in the broiler intestine, the supplementation of exogenous enzymes alone cannot provide complete protection against NE (Elwinger and Tegl€of, 1991; Riddell and Kong, 1992). Enzymes have no direct effect on C. perfringens, but only change the intestinal environment (Kocher and Choct, 2008). Therefore, further research is needed to investigate the effect of feed enzymes on the incidence of NE, so as to clarify the roles of the enzymes in the possible control of the disease.
 
3.2. Probiotics
Probiotics have been defined as a “live microbial feed supplements, which beneficially affect the host animal by improving its microbial balance” (Fuller, 1990). The characteristic of ideal probiotics are that they must be from host origin, resist to gastric acids and bile, persist in the intestinal tract, adhere to epithelium or mucus, produce inhibitory compounds, alter immune response and modulate the microflora activity (Patterson and Burkholder, 2003). The modes of action of probiotics include stimulating the immune system, maintaining gut microflora by competitive exclusion, altering metabolism through increased digestive enzyme activity, decreasing bacterial enzyme activity and ammonia production, and neutralizing enterotoxins (Collins and Gibson, 1999; Patterson andBurkholder, 2003; Simmering and Blaut, 2001; Walker and Duffy, 1998). The mechanisms of competitive exclusion of pathogens include competitive use of nutrients and mucosal binding sites, or production of short-chain fatty acids (SCFAs), low pH and bacteriocins, which are bactericidal or bacteriostatic for pathogenic bacteria (Ohland and MacNaughton, 2010).
A number of studies have reported the potential of undefined (normal gut flora) or defined (characterized bacterial strain) probiotics on the colonization of pathogenic bacteria. Mead (2000) reported that normal gut flora preparations showed efficacy against food borne pathogens such as Escherichia coli, Yersinia enterocolitica, Clostridium botulinum, C. perfringens, Salmonella, and Campylobacter jejuni. Morishita et al. (1997) showed that Lactobacillus acidophilus and Streptococcus faecium reduced the colonization of C. jejuni in the jejunum by 27%. A number of studies have been conducted to evaluate the effects of probiotics on NE in chickens. It has been reported that commercial probiotics reduce gross intestinal lesions from NE and improve feed efficiency (Hofacre et al., 1998). Craven et al. (1999) proposed that feeding normal gut flora to broiler chickens would reduce the C. perfringens colonization and decrease the incidence of NE. Hofacre et al. (2003) showed that mortality due to NE was reduced from 60 to 30% when birds were treated with lactic acid bacteria. La Ragione and Woodward (2003) found that colonization and determination of C. perfringens was suppressed when 1 day old and 20 day old birds were inoculated with 109 spores of Bacillus subtilis strain, then challenged 24 h later with 105 CFU of C. perfringens, colonization and determination of C. perfringens was suppressed. Jayaraman et al. (2013) found that dietary supplementation of B. subtilis in experimentally challenged broiler chickens with Eimeria and C. perfringens reduced FCR, intestinal lesion scores, and C. perfringens counts, and increased villus height to crypt depth ratios compared with an infected control without such supplementation.
 
3.3. Prebiotics
Prebiotics are generally defined as indigestible feed ingredients that selectively stimulate the growth or activity of beneficial bacteria that are already resident in the GIT (Gibson and Roberfroid, 1995). The potential effects of prebiotics on animals were recognized in the 1980s (Hajati and Rezaei, 2010). For a feed ingredient to be classified as a prebiotic, it must be neither digested nor absorbed in the upper part of digestive system, and it must be a selective substrate for one or a limited number of useful bacteria in the GIT, stimulate the bacteria to grow, be metabolically activated, be able to alter the intestinal microflora toward a healthier composition, and be palatable as a food ingredient (Collins and Gibson, 1999; Hajati and Rezaei, 2010). Among those, carbohydrates are commonly seen in the literature as prebiotics. For example, lactose, a disaccharide, has been used as a prebiotic in chickens (Hajati and Rezaei, 2010). We list here several other predominantly used prebiotics, i.e., mannooligosaccharides (MOS), fructo-oligosaccharides (FOS), xylo-oligosaccharides, glycol-oligosaccharides, galactooligosaccharides, gluco-oligosaccharides, lactitol, lactulose, maltooligosaccharides, and trans-galacto-oligosaccharides (Collins and Gibson, 1999; Gibson and Roberfroid, 1995; Patterson et al., 1997). In general, prebiotics can prevent the colonization of bacterial pathogens in the GIT (Bengmark, 2001), lower the gut pH through SCFA production (Gibson and Wang, 1994), and stimulate the immune system (Monsan and Paul, 1995). The following sections will detail the effectiveness of some prebiotics on the control of NE.
MOS is extracted from the yeast cell wall of Saccharomyces cerevisiae yeast (Hofacre et al., 2003). The yeast cell wall consists of protein, glucans, and mannan (Klis et al., 2002). MOS consists of mannoproteins, chitin, b (1,3) glucan, and b (1,6) glucan (Kollár et al., 1997). The exact mode of action of MOS is unclear, but bglucans and mannans are the units with primary functions on bacteria indirectly. The mannans act as a receptor for type-1 fimbriae which are used by some harmful bacteria such as E. coli and Salmonella to attach to the gastrointestinal wall (Oyofo et al., 1989). b-glucans act as microbial recognition receptors of the innate immune system (Gantner et al., 2003) and both mannans and b-glucans structures stimulate the immune system (Spring et al., 2000). The effects of MOS supplementation on the broiler immune system, gut microflora and gut morphology have been well documented. For instance, Shanmugasundaram and Selvaraj (2012) reported that dietary prebiotics (killed whole yeast cells) increased interleukin-10 mRNA by 9 fold in comparison to the control. Shanmugasundaram et al. (2013) also proposed that 0.2% of killed whole yeast cell prebiotics increased body weight gain, macrophage nitric oxide production and caecal tonsil interleukin-1 mRNA amounts in broilers challenged with coccidia. Ghosh et al. (2011) reported that antibody titer against Newcastle disease was significantly higher in birds fed a yeast cell wall preparation. In the same study, birds were orally infected with Salmonella pullorum at 45 d of age. After the oral infection, Salmonella counts were lower in digesta in birds fed the yeast cell wall preparation. This was supported by a number of studies, where dietary MOS were effective in reducing Salmonella infection (Fernandez et al., 2000; Spring et al., 2000). It has also been reported that the mannan oligosaccharides improved intestinal morphology, such as increasing villi height and altering mucosal architecture (Iji et al., 2001), and decreasing the crypt depth of the mucosa of the small intestine (Yang et al., 2009).
Some studies have been conducted on the effects of prebiotics on C. perfringens in reducing the severity of NE. Data from Sims et al. (2004) showed that 6 week old turkeys in a MOS treatment group had significantly fewer C. perfringens in their large intestines compared with control birds. This finding is supported by Kim et al. (2011) who reported that dietary supplementation with MOS decreased C. perfringens populations in broilers' small intestines. Dietary MOS also reduced the abundance of C. perfringens in the broiler caeca at 21 d of age (Yang et al., 2008). During NE infection, Mohamed and Hafez (2011) observed that dietary MOS reduced mortality by 12% compared with the positive control group. In the same study, dietary MOS supplementation showed a reduction in the severity of lesion scores to 0.57 in contrast to 1.8 in the control group. Also, the results obtained in our group showed that yeast cell wall extract was effective in reducing lesions and improving livability compared with the challenged control (M'Sadeq et al., 2015). In contrast, Hofacre et al. (2003) suggested neither addition of MOS nor FOS to the broilers diet had significant effect on intestinal lesion scores and mortality caused by NE. Although some controversial results have been observed, prebiotics such as MOS did show its effectiveness to control diseases including NE. Thus, prebiotics may be potentially effective antibiotic alternatives and it is worthwhile to further explore its utilization in the control of NE in broilers.
 
3.4. Organic acids
Organic acids have been used for decades in feed, to protect feed from microbial and fungal damage. These acids are added to foods as preservative agents and can also be used to control microbial contamination (Kumet al., 2010). In fact, the organic acids that have antimicrobial activity are simple monocarboxylic acids such as formic, acetic, propionic and butyric acids, or are carboxylic acids bearing a hydroxyl group on the alpha carbon such as lactic, malic, and tartaric acids. It has been noted that salts of some of these acids have performance benefits (Dibner and Buttin, 2002). Some short chain carboxylic organic acids, such as sorbic and fumaric acids containing double bonds, also have antifungal activity (Dibner and Buttin, 2002). Several of these organic acids can be produced in small quantities as a result of the fermentation activity of the GIT (Dorsa, 1997) where anaerobic microflora are predominant (Dixon and Hamilton, 1981).
The effects of organic acids on broiler chickens have been well documented. Hassan et al. (2010) proposed that the inclusion of organic acids to broiler diets enhanced growth and feed utilization and feed conversion ratio. Acidification with different organic acids has been shown to reduce pathogenic bacteria toxicity through controlling pathogenic colonization on the gastrointestinal wall, and by inhibiting damage to epithelial cells (Langhout, 2000). The basic principal mode of action of organic acids is that nondissociated organic acids can diffuse through lipophilic bacteria and mold membranes, and can disrupt the enzymatic reaction and the transport system of the bacteria (Cherrington et al., 1991). As described by Lambert and Stratford (1999), after penetration of organic acids into bacterial cytoplasm the non-ionized organic acids decompose to H (Hþ) ions and (A¯) ions. By the time of the decline of the pH inside the bacteria, a specific mechanism (Hþ- ATPase pump) will act to return the intracellular levels to normal pH. This process requires energy, which will lead to reduced energy accessibility for the proliferation of cells and eventually stop the bacterial growth or even kill it. Izat et al. (1990) observed that dietary supplementation of buffered propionic acid significantly reduced the total number of E. coli in the intestinal tract. Thompson and Hinton (1997) found that organic acids are bactericidal for Salmonella serotype Enteritidis PT4. Chaveerach et al. (2004) demonstrated that organic acids in the drinking water of young chicks could have a potential effect on Campylobacter infection. Czerwinski et al. (2012) found a negative correlation between Enterobacteriaceae numbers and the concentration of undissociated propionate, acetate and butyrate in the caeca. Dietary addition of organic acids can also improve the digestibility of protein and amino acids (Afsharmanesh and Pourreza, 2005), calcium, phosphorus, magnesium and zinc (Garcia et al., 2007). Also, it has been reported that organic acid supplementation can improve gastrointestinal villi height and cell proliferation (Adil et al., 2010). Other activities of organic acids associated with acidification include the increase of pancreatic secretions, microbial phytase activity, and the improvements of digestive enzyme activity (Dibner and Buttin, 2002).
Recently, microencapsulated organic acids have been used in poultry diets. The rationale behind using coated organic acids is that non-protected organic acids are digested and absorbed in the upper part of digestive tract, while coating organic acids prevents dissociation in the upper part of the digestive tract and directs their bioactivity towards the lower GIT. The applications of microencapsulated organic acids on animals are limited. Smulikowska et al. (2009) suggested that there is no growth promoting response to coated sodium butyrate or its salt in birds raised under optimized conditions. This finding has been supported by a number of other researchers (Czerwinski et al., 2012; Smulikowska et al., 2009; Zhang et al., 2011). Jerzsele et al. (2011) suggested that protected sodium butyrate had no beneficial effects in birds challenged with C. perfringens, but a combination of sodium butyrate with essential oils protected with vegetable fat increased body weight gain, villus height and decreased gross lesion scores compared with a control. In conclusion, organic acids have not shown effective protection of birds from pathogenic C. perfringens infection to alleviate the NE symptoms. However, further studies are required to investigation why such additives are not so far effective against NE as they are protective against E. coli (Izat et al., 1990) or Campylobacter (Chaveerach et al., 2004) in broiler chickens.
 
3.5. Vaccination
Vaccination has been an effective way to prevent humans and animals from many infectious diseases. It can enhance specific immunity of the organisms to viral and bacterial diseases. Vaccines have also been successfully applied to control numerous clostridial diseases in livestock animals (Walker, 1992). Therefore, vaccination against NE disease is proposed to provide an alternative treatment for NE in poultry especially when more convincing evidences was revealed that the toxin NetB is responsible for the disease (Keyburn et al., 2008, 2013).
Vaccination strategies have been put forward for the control of NE mainly in broiler chickens. These strategies were extensively reviewed recently by Mot et al. (2014). Prior to the discovery of  NetB, the earlier vaccination focused on toxins that may not be associated to NE by and large, for example, a-toxin. Thus, the vaccines developed only had limited success in controlling NE (Cooper et al., 2009; Hoang et al., 2008; Zekarias et al., 2008), while the partial protective effect of a-toxin based vaccine may be due to the association of a-toxin protein with cell membrane that can have immune interaction to perform such protection (Keyburn et al., 2013). On the other hand, vaccination against coccidiosis has been also used to protect birds from NE occurrence (Jackson et al., 2003; Tsiouris et al., 2013; Williams et al., 2003). As Eimeria infection is widely recognized as one of the most important NE predisposing factors in broilers, it seems reasonable that the reduced risk of coccidiosis in birds would alleviate NE outbreak in the flock.
The most important step forward to developing vaccines to immunize the birds against NE occurred following the discovery of NetB toxin (Keyburn et al., 2008). A recombinant NetB C. perfringens (rNetB) was constructed and attenuated as a vaccine by (Keyburn et al., 2013). The birds immunized with rNetB were significantly protected against NE challenged with a mild dose of virulent bacteria, while the effectiveness of the vaccination was not so when a more robust challenge was performed. Alternatively, when the birds were immunized with a combination of rNetB, bacterin and cell free toxoid, significant protection against moderate and severe challengewas observed. It was suggested that in vitro levels of NetB produced by virulent C. perfringens isolates were too low to produce strong immune response in the birds and thus the combined vaccination of birds with rNetB and other cellular or cell-free antigens may be necessary. Jang et al. (2012) compared four C. perfringens recombinant proteins as vaccine candidates using the Montanide™ ISA 71 VG adjuvant in an experimental model of NE.
When the broilers were immunized with purified clostridial recombinant NetB toxin, pyruvate: ferredoxin oxidoreductase (PFO), a-toxin, or elongation factor-Tu, significantly reduced gut lesions were observed. Furthermore, birds immunized with NetB toxin exhibited significantly increased body weight gains and greater NetB toxin antibody titers. The authors suggested that vaccination with NetB toxin or PFO, in combination with ISA 71 VG enhances protective immunity against NE in broiler chickens. Fernandes da Costa et al. (2013) employed similar strategy but used only a formaldehyde NetB toxoid or NetB genetic toxoid (W262A) without attenuated NetB containing C. perfringens strains or recombinants. The immunization of poultry with these toxoids resulted in the induction of antibody responses against NetB and provided partial protection against the disease. Furthermore, Mot et al. (2013) used crude supernatant containing active toxin or formaldehyde-inactivated supernatant (toxoid) of a NetB positive C. perfringens strain and administered vaccination on 1, 3 and 12 days post hatch. It was found that double vaccination at day 3 and day 12 resulted in a significant decrease in the number of chickens with NE lesions although single vaccination with crude supernatant at day 3 also produced significant protection. However, the efficacy of vaccination using toxoid was lower compared with crude supernatant, and vaccination of 1-day-old chickens with crude supernatant or toxoid did not induce protection, away supposed to be  practical in the broiler production. To be more practical for the industry, Keyburn et al. (2013) performed maternal immunization in hens injected subcutaneously with genetically toxoided rNetB(S254L) alone, C. perfringens type A toxoid and toxoid combined with rNetB (S254L). They observed strong serum immunoglobulin Y response to NetB immunized with rNetB (S254L) formulations with anti-NetB antibodies transferred to the eggs and then onto the hatched progeny. It was confirmed that birds derived from hens immunized with rNetB (S254L) combined with toxoid and challenged with a homologous strain (EHE-NE18) had significantly lower levels of disease in a subclinical form compared with birds from adjuvant only vaccinated hens. They demonstrated that maternal immunization with a NetB-enhanced toxoid vaccine is promising for the control of NE in broiler chickens.
Although the protection of birds against NE was achieved by vaccination using NetB or other antigen related vaccines or in combination, more practical protocols and effective vaccines have to be extensively examined to achieve full protection of birds against the disease. The vaccines targeting against NetB antigen and possibly other undiscovered toxins responsible for NE should be explored through a practical vaccination regime for industry application. Undoubtedly, other beneficial combinations of different vaccines, such as non-inactivated supernatants, formalininactivated crude toxoids, immunogenic proteins and modified toxins including those that indirectly protect against NE are also important aspects to be investigated so as to achieve desired immunization levels suitable in a commercial application
 
3.6. Other potential measures in controlling NE
Recent progress on the characterization of NetB toxin has suggested other possibilities to control NE. NetB has been characterized as a pore-forming toxin with a heptameric structure (Savva et al., 2013; Yan et al., 2013) where the polymerization of the proteins leads to the toxicity of the toxin to the epithelial cells of chicken intestine. It has been recognized that cholesterol plays a key role in the ability of NetB to oligomerize and form functional pores (Savva et al., 2013). This may provide some hints on the development of measures to reduce toxicity of NetB and thus to control NE in birds. Furthermore, the detailed information on the residues important for binding and toxicity will also facilitate vaccine development against NE.
Apart from the toxin NetB which directly produce toxicity to the intestinal tissues, a bacteriocin from NetB positive C. perfringens characterized by Timbermont et al. (2014), perfrin, was considered important in the pathogenesis of NE in broilers. This bacteriocin inhibits growth of other C. perfringens strains and leads to extensive and selective growth of C. perfringens secreting toxins that cause gut lesions. Consequently, it is reasonable to speculate that mechanisms preventing the production of such bacteriocin in the gut would be able to protect birds from infection of NE at least to some extent.
Selenium regulates major physiologic pathways of humans and animals as an essential micronutrient. It can enhance the immune and antioxidant systems. Recently, seleniumwas used for the in ovo injection to broiler eggs at 18 d of embryo age and the birds were later challenged with NE, and the protection effect was observed to increase the levels of antibody against NetB and a-toxin (Lee et al., 2014b). Again, the same group injected selenium in ovo that was incorporated into hydrolyzed soybean protein (B-Taxim [BT]) and observed similar protection of challenged birds against NE (Lee et al., 2014a). Accordingly, it seems promising that selenium administered in single or incorporated in hydrolyzed soybean protein shows certain degree of protection thus worth to pursue further for industrial implementation.
 
Conclusions
Many factors associated with infectious agents and diet can negatively affect the chicken gut health which may affect the health status and production performance of birds. Phasing out of in-feed antibiotics from poultry feed in Europe and recent removal or reduction of these compounds in the other parts of the world, either by government regulation or voluntarily, in countries including Australia and USA, is a challenge to the poultry industry. Studies from Denmark and Sweden have confirmed that the key problem of in-feed antibiotic withdrawal from poultry diets is the control of NE. Currently, much extensive multidisciplinary research has been done to alleviate the problems associated with antibiotic withdrawal from poultry diets, but to date no single preventive therapy has been established to substitute in-feed antibiotics.
Several nutraceutical supplements including probiotics, prebiotics, essential oils, organic acids, enzymes and vaccines have been used to reduce the incidence of NE. However, no product has been as effective as antibiotics in terms of controlling NE and registered for commercial applications. Recent investigations have shown that the use of some antibiotic alternatives in poultry diet, for instance, prebiotics, organic acids and vaccines have improved gastrointestinal health, integrity and reduced the intestinal colonization of pathogenic bacteria including NE. In particular, the vaccine strategies may have greater potential for the control of NE mainly due to clearer etiology having been elucidated in recent years with the identification of NetB toxin. Therefore, using alternatives to in-feed antibiotics with a better understanding of the relationship between nutrition and NE, and limiting exposure to infectious agents through biosecurity and vaccination, might be a tool to reduce the incidence of NE and improve gut health in the absence of in-feed antibiotics. More importantly, the combinations of different measures may achieve greater protection of birds against the disease.
 
Acknowledgment
Ministry of Higher Education of Kurdistan region of Iraq is acknowledged for its financial sponsor of Shawkat M'Sadeq for his PhD study at the University of New England, Australia.
 
This article was originally published in Animal Nutrition, Volume 1, Issue 1, March 2015, Pages 1-11. https://doi.org/10.1016/j.aninu.2015.02.004. This is an Open Access article under the CC BYNC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
 
References
Adil S, Banday T, Bhat GA, Mir MS, Rehman M. Effect of dietary supplementation of organic acids on performance, intestinal histomorphology, and serum biochemistry of broiler chicken. Vet Med Int 2010;2010:479e85.
Afsharmanesh M, Pourreza J. Effect of calcium, citric acid, ascorbic acid vitamin D3 on the efficacy of microbial phytase in broiler starters fed wheat-based diets on performance, bone mineralization and ileal digestibility. Int J Poult Sci 2005;4: 418e24.
Al-Sheikhly F, Al-Saieg A. Role of coccidia in the occurrence of necrotic enteritis of chickens. Avian Dis 1980;24:324e33.
Al-Sheikhly F, Truscott RB. The pathology of necrotic enteritis of chickens following infusion of crude toxins of Clostridium perfringens into the duodenum. Avian Dis 1977;21:230e40.
Annett CB, Viste JR, Chirino-Trejo M, Classen HL, Middleton DM, Simko E. Necrotic enteritis: effect of barley, wheat and corn diets on proliferation of Clostridium perfringens type A. Avian Pathol 2002;31:598e601.
Apajalahti J, Kettunen A, Graham H. Characteristics of the gastrointestinal microbial communities, with special reference to the chicken.Worlds Poult Sci J 2004;60: 223e32.
Asaoka Y, Yanai T, Hirayama H, Une Y, Saito E, Sakai H, et al. Fatal necrotic enteritis associated with Clostridium perfringens in wild crows (Corvus macrorhynchos). Avian Pathol 2004;33:19e24.
Baba E, Fuller AL, Gilbert JM, Thayer SG, McDougald LR. Effects of Eimeria brunetti infection and dietary zinc on experimental induction of necrotic enteritis in broiler chickens. Avian Dis 1992;36:59e62.
Bedford MR, Classen HL. Reduction of intestinal viscosity through manipulation of dietary rye and pentosanase concentration is effected through changes in the carbohydrate composition of the intestinal aqueous phase and results in improved growth rate and food conversion efficiency of broiler chicks. J Nutr 1992;122:560e9.
Bengmark S. Pre-, pro-and synbiotics. Curr Opin Clin Nutr Metab Care 2001;4:571.
Branton SL, Lott BD, Deaton JW, Maslin WR, Austin FW, Pote LM, et al. The effect of added complex carbohydrates or added dietary fiber on necrotic enteritis lesions in broiler chickens. Poult Sci 1997;76:24e8.
Branton SL, Reece FN, Hagler WM. Influence of a wheat diet on mortality of broiler chickens associated with necrotic enteritis. Poult Sci 1987;66:1326e30.
Broussard CT, Hofacre CL, Page RK, Fletcher OJ. Necrotic enteritis in cage-reared commercial layer pullets. Avian Dis 1986;30:617e9.
Brynestad S, Granum PE. Clostridium perfringens and foodborne infections. Int J Food Microbiol 2002;74:195e202.
Carlson MS, Fangman TJ. Swine antibiotics and feed additives: food safety considerations. Columbia, MO: University of Missouri Extension; 2000G2353.
Casewell M, Friis C, Marco E, McMullin P, Phillips I. The European ban on growth-promoting antibiotics and emerging consequences for human and animal health. J Antimicrob Chemother 2003;52:159e61.
Chakrabarty A, Boro B, Mukit A, Datta B, Baruah M. An epornitic of enteritis in brahminy ducks (Todorna ferruginea) due to enterotoxigenic strains of Clostridium perfringens, type C. J Zoo Anim Med 1983:24e7.
Chalmers G, Martin SW, Hunter DB, Prescott JF, Weber LJ, Boerlin P. Genetic diversity of Clostridium perfringens isolated from healthy broiler chickens at a commercial farm. Vet Microbiol 2008;127:116e27.
Chaveerach P, Keuzenkamp DA, Lipman LJA, Van Knapen F. Effect of organic acids in drinking water for young broilers on Campylobacter infection, volatile fatty acid production, gut microflora and histological cell changes. Poult Sci 2004;83: 330e4.
Cherrington CA, Hinton M, Mead GC, Chopra I. Organic acids: chemistry, antibacterial activity and practical applications. Adv Microb Physiol 1991;32:87e108.
Choct M, Hughes RJ, Bedford MR. Effects of a xylanase on individual bird variation, starch digestion throughout the intestine, and ileal and caecal volatile fatty acid production in chickens fed wheat. Br Poult Sci 1999;40:419e22.
Choct M, Hughes RJ, Wang J, Bedford MR, Morgan AJ, Annison G. Increased small intestinal fermentation is partly responsible for the anti-nutritive activity of non-starch polysaccharides in chickens. Br Poult Sci 1996;37:609e21.
Clarke E, Wiseman J. Effects of variability in trypsin inhibitor content of soya bean meals on true and apparent ileal digestibility of amino acids and pancreas size in broiler chicks. Anim Feed Sci Technol 2005;121:125e38.
Collier CT, Hofacre CL, Payne AM, Anderson DB, Kaiser P, Mackie RI, et al. Coccidiainduced mucogenesis promotes the onset of necrotic enteritis by supporting Clostridium perfringens growth. Vet Immunol Immunopathol 2008;122:104e15
Collins MD, Gibson GR. Probiotics, prebiotics, and synbiotics: approaches for modulating the microbial ecology of the gut. Am J Clin Nutr 1999;69:1052e7.
Cooper KK, Songer JG. Necrotic enteritis in chickens: a paradigm of enteric infection by Clostridium perfringens type A. Anaerobe 2009;15:55e60.
Cooper KK, Songer JG. Virulence of Clostridium perfringens in an experimental model of poultry necrotic enteritis. Vet Microbiol 2010;142:323e8.
Cooper KK, Songer JG, Uzal FA. Diagnosing clostridial enteric disease in poultry. J Vet Diagn Invest 2013:1e14.
Cooper KK, Trinh HT, Songer JG. Immunization with recombinant alpha toxin partially protects broiler chicks against experimental challenge with Clostridium perfringens. Vet Microbiol 2009;133:92e7.
Craven SE. Colonization of the intestinal tract by Clostridium perfringens and fecal shedding in diet-stressed and unstressed broiler chickens. Poult Sci 2000;79: 843e9.
Craven SE, Cox NA, Bailey JS, Cosby DE. Incidence and tracking of Clostridium perfringens through an integrated broiler chicken operation. Avian Dis 2003;47: 707e11.
Craven SE, Cox NA, Stern NJ, Mauldin JM. Prevalence of Clostridium perfringens in commercial broiler hatcheries. Avian Dis 2001a;45:1050e3.
Craven SE, Stern NJ, Bailey JS, Cox NA. Incidence of Clostridium perfringens in broiler chickens and their environment during production and processing. Avian Dis 2001b;45:887e96.
Craven SE, Stern NJ, Cox NA, Bailey JS, Berrang M. Cecal carriage of Clostridium perfringens in broiler chickens given Mucosal Starter Culture™. Avian Dis 1999;43:484e90.
Czerwinski J, Højberg O, Smulikowska S, Engberg RM, Mieczkowska A. Effects of sodium butyrate and salinomycin upon intestinal microbiota, mucosal morphology and performance of broiler chickens. Arch Anim Nutr 2012;66:102e16.
Dahiya JP, Hoehler D, Van Kessel AG, Drew MD. Dietary encapsulated glycine influences Clostridium perfringens and Lactobacilli growth in the gastrointestinaltract of broiler chickens. J Nutr 2007;137:1408e14.
Dahiya JP, Hoehler D, Wilkie D, Van Kessel A, Drew M. Dietary glycine concentration affects intestinal Clostridium perfringens and lactobacilli populations in broiler chickens. Poult Sci 2005;84:1875e85.
Dahiya JP, Wilkie DC, Van Kessel AG, Drew MD. Potential strategies for controlling necrotic enteritis in broiler chickens in post-antibiotic era. Anim Feed Sci  Technol 2006;129:60e88.
Dekich MA. Broiler industry strategies for control of respiratory and enteric diseases. Poult Sci 1998;77:1176e80.
Dibner JJ, Buttin P. Use of organic acids as a model to study the impact of gut microflora on nutrition and metabolism. J Appl Poult Res 2002;11:453e63.
Dixon RC, Hamilton PATB. Effect of feed ingredients on the antifungal activity of propionic acid. Poult Sci 1981;60:2407e11.
Dorsa WJ. New and established carcass decontamination procedures commonly used in the beef-processing industry. J Food Prot 1997;60:1146e51.
Drew MD, Syed NA, Goldade BG, Laarveld B, Van Kessel AG. Effects of dietary protein source and level on intestinal populations of Clostridium perfringens in broiler chickens. Poult Sci 2004;83:414e20.
Drouin P. Retour en force de l'enterite necrotique. Filieres Avicoles. 1999. p. 78e9.
Duff J, Holliman A, Hateley G, Irvine R, Wight A, Williamson S. Water bird mortality associated with freezing winter weather and disease. Vet Rec 2011;168:593.
Eleazer TH, Harrell JS. Clostridium perfringens infection in turkey poults. Avian Dis 1976;20:774e6.
Elwinger K, Tegl€of B. Performance of broiler chickens as influenced by a dietary enzyme complex with and without antibiotic supplementation. Arch Gefluegelkd Ger Fr 1991;55:69e73.
Engberg RM, Hedemann MS, Jensen BB. The influence of grinding and pelleting of feed on the microbial composition and activity in the digestive tract of broiler chickens. Br Poult Sci 2002;43:569e79.
Engberg RM, Hedemann MS, Steenfeldt S, Jensen BB. Influence of whole wheat and xylanase on broiler performance and microbial composition and activity in the digestive tract. Poult Sci 2004;83:925e38
Engstr€om BE, Fermer C, Lindberg A, Saarinen E, Båverud V, Gunnarsson A. Molecular typing of isolates of Clostridium perfringens from healthy and diseased poultry. Vet Microbiol 2003;94:225e35.
European Commission Directorate General XXIV Directorate B. Opinion of the Scientific Steering Committee on Antimicrobial Resistance. http://ec.europa.eu/ food/fs/sc/ssc/out50_en.pdf; 1999 [online].
Fernandes da Costa SP, Mot D, Bokori-Brown M, Savva CG, Basak AK, Van Immerseel F, et al. Protection against avian necrotic enteritis after immunisation with NetB genetic or formaldehyde toxoids. Vaccine 2013;31:4003e8
Fernandez F, Hinton M, Van Gils B. Evaluation of the effect of mannanoligosaccharides on the competitive exclusion of Salmonella Enteritidis colonization in broiler chicks. Avian Pathol 2000;29:575e81.
Flores-Díaz M, Alape-Girón A. Role of Clostridium perfringens phospholipase C in the pathogenesis of gas gangrene. Toxicon 2003;42:979e86.
Fukata T, Hadate Y, Baba E, Uemura T, Arakawa A. Influence of Clostridium perfringens and its toxin in germ-free chickens. Res Vet Sci 1988;44:68e70.
Fuller R. Probiotics in human medicine. Gut 1990;32:439e42.
Gantner BN, Simmons RM, Canavera SJ, Akira S, Underhill DM. Collaborative induction of inflammatory responses by dectin-1 and Toll-like receptor 2. J Exp Med 2003;197:1107e17.
Garcia V, Catalá-Gregori P, Hernandez F, Megias MD, Madrid J. Effect of formic acid and plant extracts on growth, nutrient digestibility, intestine mucosa morphology, and meat yield of broilers. J Appl Poult Res 2007;16:555e62.
Gholamiandehkordi AR, Immerseel FV, Timbermont L, Lanckriet A, Broeck WVD, Pedersen K, et al. Quantification of gut lesions in a subclinical necrotic enteritis model. Avian Pathol 2007;36:375e82.
Gholamiandekhordi AR, Ducatelle R, Heyndrickx M, Haesebrouck F, Van Immerseel F. Molecular and phenotypical characterization of Clostridium perfringens isolates from poultry flocks with different disease status. Vet Microbiol 2006;113:143e52.
Ghosh TK, Haldar S, Bedford MR, Muthusami N, Samanta I. Assessment of yeast cell wall as replacements for antibiotic growth promoters in broiler diets: effects on performance, intestinal histo-morphology and humoral immune responses. J Anim Physiol Anim Nutr 2011;96:275e84.
Gibson GR, Roberfroid MB. Dietary modulation of the human colonic microbiota: introducing the concept of prebiotics. J Nutr 1995;125:1401e12.
Gibson GR, Wang X. Bifidogenic properties of different types of fructo-oligosaccharides. Food Microbiol 1994;11:491e8.
Giovannini C, Mancini E, De Vincenzi M. Inhibition of the cellular metabolism of Caco-2 cells by prolamin peptides from cereals toxic for coeliacs. Toxicol In Vitro 1996;10:533e8.
Hafez HM. Enteric diseases of poultry with special attention to Clostridium perfringens. Pak Vet J 2011;31:175e84.
Hajati H, Rezaei M. The application of prebiotics in poultry production. Int J Poult Sci 2010;9:298e304.
Hassan HMA, Mohamed MA, Youssef AW, Hassan ER. Effect of using organic acids to substitute antibiotic growth promoters on performance and intestinal microflora of broilers. Asian-Australas J Anim Sci 2010;23:1348e53.
Helmboldt CF, Bryant ES. The pathology of necrotic enteritis in domestic fowl. Avian Dis 1971;15:775e80.
Hermans PG, Morgan KL. 57. The epidemiology of necrotic enteritis in broiler chickens. Res Vet Sci 2003;74:19.
Hermans PG, Morgan KL. Prevalence and associated risk factors of necrotic enteritis on broiler farms in the United Kingdom; a cross-sectional survey. Avian Pathol 2007;36:43e51.
Hoang TH, Hong HA, Clark GC, Titball RW, Cutting SM. Recombinant Bacillus subtilis expressing the Clostridium perfringens alpha toxoid is a candidate orally delivered vaccine against necrotic enteritis. Infect Immun 2008;76:5257e65.
Hofacre CL, Beacorn T, Collett S, Mathis G. Using competitive exclusion, mannanoligosaccharide and other intestinal products to control necrotic enteritis. J Appl Poult Res 2003;12:60e4.
Hofacre CL, French JD, Page RK, Fletcher OJ. Subcutaneous clostridial infection in broilers. Avian Dis 1986;30:620e2.
Hofacre CL, Froyman R, George B, Goodwin MA, Brown J. Use of aviguard, virginiamycin, or bacitracin MD against Clostridium perfringens-associated necrotizing enteritis. J Appl Poult Res 1998;7:412e8.
Hofshagen M, Stenwig H. Toxin production by Clostridium perfringens isolated from broiler chickens and capercaillies (Tetrao urogallus) with and without necrotizing enteritis. Avian Dis 1992;36:837e43.
Iji PA, Saki AA, Tivey DR. Intestinal structure and function of broiler chickens on diets supplemented with a mannan oligosaccharide. J Sci Food Agric 2001;81: 1186e92.
Iji PA, Tivey DR. Natural and synthetic oligosaccharides in broiler chicken diets. Worlds Poult Sci J 1998;54:129e43.
Izat AL, Tidwell NM, Thomas RA, Reiber MA, Adams MH, Colberg M, et al. Effects of a buffered propionic acid in diets on the performance of broiler chickens and on microflora of the intestine and carcass. Poult Sci 1990;69:818e26.
Jackson ME, Anderson DM, Hsiao HY, Mathis GF, Fodge DW. Beneficial effect of bmannanase feed enzyme on performance of chicks challenged with Eimeria sp. and Clostridium perfringens. Avian Dis 2003;47:759e63.
Jang SI, Lillehoj HS, Lee S-H, Lee KW, Lillehoj EP, Hong YH, et al. Vaccination with Clostridium perfringens recombinant proteins in combination with Montanide™ ISA 71 VG adjuvant increases protection against experimental necrotic enteritis in commercial broiler chickens. Vaccine 2012;30:5401e6.
Jayaraman S, Thangavel G, Kurian H, Mani R, Mukkalil R, Chirakkal H. Bacillus subtilis PB6 improves intestinal health of broiler chickens challenged with Clostridium perfringens-induced necrotic enteritis. Poult Sci 2013;92:370e4.
Jerzsele A, Szeker K, Csizinszky R, Gere E, Jakab C, Mallo JJ, et al. Efficacy of protected sodium butyrate, a protected blend of essential oils, their combination, and Bacillus amyloliquefaciens spore suspension against artificially induced necrotic enteritis in broilers. Poult Sci 2011;91:837e43.
Juskiewicz J, Zdunczyk Z, Jankowski J. Selected parameters of gastrointestinal tract metabolism of turkeys fed diets with flavomycin and different inulin content. Worlds Poult Sci J 2004;60:177e85.
Kaldhusdal M, Skjerve E. Association between cereal contents in the diet and incidence of necrotic enteritis in broiler chickens in Norway. Prev Vet Med 1996;28:1e16.
Kalshusdal M, Hofshagen M. Barley inclusion and avoparcin supplementation in broiler diets. 2. Clinical, pathological, and bacteriological findings in a mild form of necrotic enteritis. Poult Sci 1992;71:1145e53.
Kelly D, Conway S. Genomics at work: the global gene response to enteric bacteria. Gut 2001;49:612e3.
Keyburn AL, Boyce JD, Vaz P, Bannam TL, Ford ME, Parker D, et al. NetB, a new toxin that is associated with avian necrotic enteritis caused by Clostridium perfringens. PLoS Pathog 2008;4:e26.
Keyburn AL, Portela RW, Sproat K, Ford ME, Bannam TL, Yan X, et al. Vaccination with recombinant NetB toxin partially protects broiler chickens from necrotic enteritis. Vet Res 2013;44:54.
Keyburn AL, Sheedy SA, Ford ME, Williamson MM, Awad MM, Rood JI, et al. Alphatoxin of Clostridium perfringens is not an essential virulence factor in necrotic enteritis in chickens. Infect Immun 2006;74:6496e500.
Keyburn AL, Yan X-X, Bannam TL, Van Immerseel F, Rood JI, Moore RJ. Association between avian necrotic enteritis and Clostridium perfringens strains expressing NetB toxin. Vet Res 2010;41:1e8.
Kim GB, Seo YM, Kim CH, Paik IK. Effect of dietary prebiotic supplementation on the performance, intestinal microflora, and immune response of broilers. Poult Sci 2011;90:75e82.
Kleessen B, Hartmann L, Blaut M. Fructans in the diet cause alterations of intestinal mucosal architecture, released mucins and mucosa-associated bifidobacteria in gnotobiotic rats. Br J Nutr 2003;89:597e606.
Klis FM, Mol P, Hellingwerf K, Brul S. Dynamics of cell wall structure in Saccharomyces cerevisiae. FEMS Microbiol Rev 2002;26:239e56.
Kocher A, Choct M. Improving broiler chicken performance. http://www.rirdc.gov. au/fullreports/index.html; 2008.
Kollár R, Reinhold BB, Petráková E, Yeh HJC, Ashwell G, Drgonová J, et al. Architecture of the yeast cell wall. J Biol Chem 1997;272:17762e75.
Kum S, Eren U, Onol AG, Sandikci M. Effects of dietary organic acid supplementation on the intestinal mucosa in broilers. Rev Med Vet 2010;10:463e8
La Ragione RM, Woodward MJ. Competitive exclusion by Bacillus subtilis spores of Salmonella enterica serotype Enteritidis and Clostridium perfringens in young chickens. Vet Microbiol 2003;94:245e56.
Lambert RJ, Stratford M. Weak acid preservatives: modelling microbial inhibition and response. J Appl Microbiol 1999;86:157e64.
Lan Y, Verstegen MWA, Tamminga S, Williams BA. The role of the commensal gut microbial community in broiler chickens. Worlds Poult Sci J 2005;61:95e104.
Langhout P. New additives for broiler chickens. World Poult 2000;16:22e7.
Lee KW, Lillehoj HS, Jeong W, Jeoung HY, An DJ. Avian necrotic enteritis: experimental models, host immunity, pathogenesis, risk factors, and vaccine development. Poult Sci 2011;90:1381e90.
Lee SH, Lillehoj HS, Jang SI, Jeong M, Kim DK, Xu S, et al. Immune and anti-oxidant effects of in ovo selenium proteinate on post-hatch experimental avian necrotic enteritis. Vet Parasitol 2014a;206:115e22.
Lee SH, Lillehoj HS, Jang SI, Jeong MS, Xu SZ, Kim JB, et al. Effects of in ovo injection with selenium on immune and antioxidant responses during experimental necrotic enteritis in broiler chickens. Poult Sci 2014b;93:1113e21.
Long JR. Necrotic enteritis in broiler chickens. I. A review of the literature and the prevalence of the disease in Ontario. Can J Comp Med 1973;37:302e8.
Lovland A, Kaldhusdal M. Severely impaired production performance in broiler flocks with high incidence of Clostridium perfringens-associated hepatitis. Avian Pathol 2001;30:73e81.
Løvland A, Kaldhusdal M. Liver lesions seen at slaughter as an indicator of necrotic  enteritis in broiler flocks. FEMS Immunol Med Microbiol 1999;24:345e51. 
M'Sadeq SA, Wu SB, Choct M, Forder R, Swick RA. Use of yeast cell wall extract as a tool to reduce the impact of necrotic enteritis in broilers. Poult Sci 2015 [in press].
Martin TG, Smyth JA. Prevalence of netB among some clinical isolates of Clostridium perfringens from animals in the United States. Vet Microbiol 2009;136:202e5.
McDevitt RM, Brooker JD, Acamovic T, Sparks NHC. Necrotic enteritis; a continuing challenge for the poultry industry. Worlds Poult Sci J 2006;62:221e47.
McOrist S, Reece RL. Clostridial enteritis in free living lorikeets (Trichoglossus spp.). Avian Pathol 1992;21:503e7.
McReynolds JL, Byrd JA, Anderson RC, Moore RW, Edrington TS, Genovese KJ, et al. Evaluation of immunosuppressants and dietary mechanisms in an experimental disease model for necrotic enteritis. Poult Sci 2004;83:1948e52.
McReynolds JL, Byrd JA, Genovese KJ, Poole TL, Duke SE, Farnell MB, et al. Dietary lactose and its effect on the disease condition of necrotic enteritis. Poult Sci 2007;86:1656e61.
Mead G. Prospects for ‘competitive exclusion’ treatment to control salmonellas and other foodborne pathogens in poultry. Vet J 2000;159:111e23.
Mohamed MA, Hafez MSA. The effect of a mannanoligosaccharides (Biomos®) on necrotic enteritis infection in broiler chickens. Int J Poult Sci 2011;10:685e90.
Monsan PF, Paul F. Oligosaccharide feed additives. Biotechnology in animal feeds and animal feeding. 1995. p. 233e45. New York, USA.
Morishita TY, Aye PP, Harr BS, Cobb CW, Clifford JR. Evaluation of an avian-specific probiotic to reduce the colonization and shedding of Campylobacter jejuni in broilers. Avian Dis 1997;41:850e5.
Mot D, Timbermont L, Delezie E, Haesebrouck F, Ducatelle R, Van Immerseel F. Dayof- hatch vaccination is not protective against necrotic enteritis in broiler chickens. Avian Pathol 2013;42:179e84.
Mot D, Timbermont L, Haesebrouck F, Ducatelle R, Van Immerseel F. Progress and problems in vaccination against necrotic enteritis in broiler chickens. Avian Pathol 2014;43:290e300.
Nairn ME, Bamford VW. Necrotic enteritis of broiler chickens in western Australia. Aust Vet J 1967;43:49e54.
Nikpiran H, Shojadoost B, Peighambari SM. Experimental induction of necrotic enteritis in broiler chickens by Clostridium perfringens isolates from the outbreaks in Iran. J Vet Res 2008;63:127e32
NRC. Nutrient requirement of poultry. 9th revised ed. Washington, D.C.: National Academy Press; 1984.
Ohland CL, MacNaughton WK. Probiotic bacteria and intestinal epithelial barrier function. Am J Physiol Gastrointest Liver Physiol 2010;298:G807e19.
Olkowski AA, Wojnarowicz C, Chirino-Trejo M, Drew MD. Responses of broiler chickens orally challenged with Clostridium perfringens isolated from field cases of necrotic enteritis. Res Vet Sci 2006;81:99e108.
Olkowski AA, Wojnarowicz C, Chirino-Trejo M, Laarveld B, Sawicki G. Sub-clinical necrotic enteritis in broiler chickens: novel etiological consideration based on ultra-structural and molecular changes in the intestinal tissue. Res Vet Sci 2008;85:543e53.
Onderka DK, Langevin CC, Hanson JA. Fibrosing cholehepatitis in broiler chickens induced by bile duct ligations or inoculation of Clostridium perfringens. Can J Vet Res 1990;54:285e90.
Oyofo BA, DeLoach JR, Corrier DE, Norman JO, Ziprin RL, Mollenhauer HH. Effect of carbohydrates on Salmonella typhimurium colonization in broiler chickens. Avian Dis 1989;33:531e4.
Parish WE. Necrotic enteritis in the fowl (Gallus gallus domesticus) histopathology of the disease and isolation of a strain of Clostridium welchii. J Comp Pathol 1961;71:377e93.
Patterson J, Orban J, Sutton A, Richards G. Selective enrichment of bifidobacteria in the intestinal tract of broilers by thermally produced kestoses and effect on broiler performance. Poult Sci 1997;76:497e500.
Patterson JA, Burkholder KM. Application of prebiotics and probiotics in poultry production. Poult Sci 2003;82:627e31.
Petit L, Gibert M, Popoff MR. Clostridium perfringens: toxinotype and genotype. Trends Microbiol 1999;7:104e10.
Pusztai A, Bardocz S. Biological effects of plant lectins on the gastrointestinal tract: metabolic consequences and applications. Trends Glycosci Glycotechnol 1996;8:149e66.
Rehman H, Ijaz A, Specht A, Dill D, Hellweg P, M€anner K, et al. In vitro effects of alpha toxin from Clostridium perfringens on the electrophysiological parameters of jejunal tissues from laying hens preincubated with inulin and N-acetyl-Lcysteine. Poult Sci 2009;88:199e204.
Reynolds DL. Multi-causal enteric diseases. In: Saif YM, editor. Diseases of poultry. 11th ed. Ames, IA: Blackwell; 2003. p. 1169e70
Riddell C, Kong X-M. The influence of diet on necrotic enteritis in broiler chickens. Avian Dis 1992;36:499e503.
Robins C, Brooker JD. The effects of Acacia aneura feeding on abomasal and intestinal structure and function in sheep. Anim Feed Sci Technol 2005;121:205e15.
Rodgers N, Swick RA, Geier MS, Moore R, Choct M, Wu S-B. A multifactorial analysis of the extent to which Eimeria and fishmeal predispose broiler chickens to necrotic enteritis. Avian Dis 2015;59:38e45.
Savva CG, da Costa SPF, Bokori-Brown M, Naylor CE, Cole AR, Moss DS, et al. Molecular architecture and functional analysis of NetB, a pore-forming toxin from Clostridium perfringens. J Biol Chem 2013;288:3512e22.
Shane S. Update on the poultry disease situation in the USA-significant disease outbreaks in recent years. Overall, the health status is good but there is no room for complacency. Poult Int 2004;43:10e5.
Shanmugasundaram R, Selvaraj RK. Effect of killed whole yeast cell prebiotic supplementation on broiler performance and intestinal immune cell parameters. Poult Sci 2012;91:107e11.
Shanmugasundaram R, Sifri M, Selvaraj RK. Effect of yeast cell product (CitriStim) supplementation on broiler performance and intestinal immune cell parameters during an experimental coccidial infection. Poult Sci 2013;92:358e63.
Shivaprasad H, Uzal F, Kokka R, Fisher DJ, McClane BA, Glenn Songer A. Ulcerative enteritis-like disease associated with Clostridium perfringens type A in bobwhite quail (Colinus virginianus). Avian Dis 2008;52:635e40.
Simmering R, Blaut M. Pro-and prebiotics e the tasty guardian angels. Appl Microbiol Biotechnol 2001;55:19e28.
Sims MD, Dawson KA, Newman KE, Spring P, Hoogell DM. Effects of dietary mannan oligosaccharide, bacitracin methylene disalicylate, or both on the live performance and intestinal microbiology of turkeys. Poult Sci 2004;83: 1148e54.
Sinlae M, Choct M. Xylanase supplementation affects the caecal microflora of broilers. In: Proceeding of the Australian poultry science symposium, vol. 12; 2000. p. 209. Sydney.
Smulikowska S, Czerwinski J, Mieczkowska A, Jankowiak J. The effect of fat-coated organic acid salts and a feed enzyme on growth performance, nutrient utilization, microflora activity, and morphology of the small intestine in broiler chickens. J Anim Feed Sci 2009;18:478e89.
Smyth JA, Martin TG. Disease producing capability of netB positive isolates of C. perfringens recovered from normal chickens and a cow, and netB positive and negative isolates from chickens with necrotic enteritis. Vet Microbiol 2010;146: 76e84.
Spring P, Wenk C, Dawson K, Newman K. The effects of dietary mannaoligosaccharides on cecal parameters and the concentrations of enteric bacteria in the ceca of Salmonella-challenged broiler chicks. Poult Sci 2000;79: 205e11.
Thompson JL, Hinton M. Antibacterial activity of formic and propionic acids in the diet of hens on Salmonellas in the crop. Br Poult Sci 1997;38:59e65.
Timbermont L, De Smet L, Van Nieuwerburgh F, Parreira VR, Van Driessche G, Haesebrouck F, et al. Perfrin, a novel bacteriocin associated with netB positive Clostridium perfringens strains from broilers with necrotic enteritis. Vet Res 2014;45:1e10.
Timbermont L, Haesebrouck F, Ducatelle R, Van Immerseel F. Necrotic enteritis in broilers: an updated review on the pathogenesis. Avian Pathol 2011;40:341e7.
Timbermont L, Lanckriet A, Gholamiandehkordi AR, Pasmans F, Martel A, Haesebrouck F, et al. Origin of Clostridium perfringens isolates determines the ability to induce necrotic enteritis in broilers. Comp Immunol Microbiol Infect Dis 2009;32:503e12.
Tsiouris V, Georgopoulou I, Batzios C, Pappaioannou N, Diakou A, Petridou E, et al. The role of an attenuated anticoccidial vaccine on the intestinal ecosystem and on the pathogenesis of experimental necrotic enteritis in broiler chickens. Avian Pathol 2013;42:163e70.
Tsiouris V, Georgopoulou I, Batzios C, Pappaioannou N, Ducatelle R, Fortomaris P. High stocking density as a predisposing factor for necrotic enteritis in broiler chicks. Avian Pathol 2015;44:59e66.
Union of Concerned Scientists. Estimates of antimicrobial abuse in livestock. http:// www.ucsusa.org/food_and_agriculture/our-failing-food-system/industrialagriculture/ hogging-it-estimates-of.html; 2001 .
Van der Sluis W. Clostridial enteritis is an often underestimated problem. World Poult 2000;16:42e3.
Van Immerseel F, De Buck J, Pasmans F, Huyghebaert G, Haesebrouck F, Ducatelle R. Clostridium perfringens in poultry: an emerging threat for animal and public health. Avian Pathol 2004;33:537e49.
Van Immerseel F, Rood JI, Moore RJ, Titball RW. Rethinking our understanding of the pathogenesis of necrotic enteritis in chickens. Trends Microbiol 2008;17:32e6.
Walker PD. Bacterial vaccines: old and new, veterinary and medical. Vaccine 1992;10:977e90.
Walker WA, Duffy LC. Diet and bacterial colonization: role of probiotics and prebiotics. J Nutr Biochem 1998;9:668e75.
Wilkie DC, Van Kessel AG,White LJ, Laarveld B, Drew MD. Dietary amino acids affect intestinal Clostridium perfringens populations in broiler chickens. Can J Anim Sci 2005;85:185e93.
Williams R, Marshall R, La Ragione R, Catchpole J. A new method for the experimental production of necrotic enteritis and its use for studies on the relationships between necrotic enteritis, coccidiosis and anticoccidial vaccination of chickens. Parasitol Res 2003;90:19e26.
Williams RB. Intercurrent coccidiosis and necrotic enteritis of chickens: rational, integrated disease management by maintenance of gut integrity. Avian Pathol 2005;34:159e80.
Wobeser G, Rainnie D. Epizootic necrotic enteritis in wild geese. J Wildl Dis 1987;23:376e85.
Work T, Smith M, Duncan R. Necrotizing enteritis as a cause of mortality in Laysan albatross, Diomedea immutabilis, chicks on Midway Atoll, Hawaii. Avian Dis 1998:1e5.
Wu SB, Rodgers N, Choct M. Optimized necrotic enteritis model producing clinical and subclinical infection of Clostridium perfringens in broiler chickens. Avian Dis 2010;54:1058e65.
Wu SB, Stanley D, Rodgers N, Swick RA, Moore RJ. Two necrotic enteritis predisposing factors, dietary fishmeal and Eimeria infection, induce large changes in the caecal microbiota of broiler chickens. Vet Microbiol 2014;169:188e97.
Yan X-X, Porter CJ, Hardy SP, Steer D, Smith AI, Quinsey NS, et al. Structural and functional analysis of the pore-forming toxin NetB from Clostridium perfringens. MBio 2013;4. e00019-00013.
Yang Y, Iji P, Choct M. Dietary modulation of gut microflora in broiler chickens: a review of the role of six kinds of alternatives to in-feed antibiotics.Worlds Poult Sci J 2009;65:97e114.
Yang Y, Iji P, Kocher A, Mikkelsen L, Choct M. Effects of dietary mannanoligosaccharide on growth performance, nutrient digestibility and gut development of broilers given different cereal-based diets. J Anim Physiol Anim Nutr 2008;92:650e9.
Yegani M, Korver DR. Factors affecting intestinal health in poultry. Poult Sci 2008;87:2052e63.
Zekarias B, Mo H, Curtiss R. Recombinant attenuated Salmonella enterica serovar Typhimurium expressing the carboxy-terminal domain of alpha toxin from Clostridium perfringens induces protective responses against necrotic enteritis in chickens. Clin Vaccine Immunol 2008;15:805e16.
Zhang WH, Jiang Y, Zhu QF, Gao F, Dai SF, Chen J, et al. Sodium butyrate maintains growth performance by regulating the immune response in broiler chickens. Br Poult Sci 2011;52:292e301.
Related topics
Authors:
Shubiao Wu
University of New England
Follow
Robert Swick
University of New England
Follow
Mingan Choct
University of New England
Follow
Join to be able to comment.
Once you join Engormix, you will be able to participate in all content and forums.
* Required information
Would you like to discuss another topic? Create a new post to engage with experts in the community.
Create a post
Forest Lin
ADM Aditivos
30 de agosto de 2017
nice article! it's headache for NE prevention worldwide.
Join Engormix and be part of the largest agribusiness social network in the world.
LoginRegister