You are on page 1of 9

Journal of Animal Science, 2020, Vol. 98, No.

8, 1–9

doi:10.1093/jas/skaa248
Advance Access publication August 6, 2020
Received: 4 June 2020 and Accepted: 30 July 2020
Board Invited Reviews

Board Invited Reviews


Ruminal acidosis, bacterial changes, and
lipopolysaccharides
Hugo F.  Monteiro and Antonio P. Faciola1
Department of Animal Sciences, University of Florida, Gainesville, FL 32611

Corresponding author: afaciola@ufl.edu


1

ORCiD numbers: 0000-0003-0935-6233 (A. P. Faciola).

Abstract
Acute and subacute ruminal acidosis (SARA) are common nutritional problems in both beef and dairy cattle. Therefore,
the objective of this review is to describe how ruminal Gram-negative bacteria could contribute to the pathogenesis
of ruminal acidoses, by releasing lipopolysaccharides (LPS; a component of their cell wall) in the ruminal fluid. When
cattle consume excessive amounts of highly fermentable carbohydrates without prior adaptation, normal fermentation
become disrupted. The fermentation of these carbohydrates quickly decreases ruminal pH due to the accumulation
of short-chain fatty acids and lactate in the rumen. As a consequence, ruminal epithelium may be damaged and
tissue function could be impaired, leading to a possible translocation of pathogenic substances from the rumen into
the bloodstream. Such changes in fermentation are followed by an increase in Gram-positive bacteria while Gram-
negative bacteria decrease. The lyses of Gram-negative bacteria during ruminal acidosis increase LPS concentration
in the ruminal fluid. Because LPS is a highly proinflammatory endotoxin in the circulatory system, past studies have
raised concerns regarding ruminal LPS contribution to the pathogenesis of ruminal acidosis. Although animals that
undergo these disorders do not always have an immune response, recent studies showed that different Gram-negative
bacteria have different LPS composition and toxicity, which may explain the differences in immune response. Given the
diversity of Gram-negative bacteria in the rumen, evaluating the changes in the bacterial community during ruminal
acidosis could be used as a way to identify which Gram-negative bacteria are associated with LPS release in the rumen.
By identifying and targeting ruminal bacteria with possible pathogenic LPS, nutritional strategies could be created to
overcome, or at least minimize, ruminal acidosis.

Key words:  Bacteroidetes, cattle, dysbiosis, endotoxin, Fibrobacteres, Gram-negative bacteria

  

Ruminal Acidosis in Cattle behavior), change the fermentation pattern toward nonfibrous
carbohydrates (NFC) fermentation (Owens et al., 1998; Nagaraja
Acute and subacute ruminal acidoses (SARA) have long been
and Titgemeyer, 2007; Miller-Cushon and DeVries, 2017). In the
described as a digestive disorders that affect beef and dairy cattle
rumen, these carbohydrates are quickly fermented into short-
(Owens et al., 1998; Nagaraja and Titgemeyer, 2007; Plaizier et al.,
chain fatty acids (SCFA) and lactate, and the accumulation of
2008). The transition from forage-based diets to concentrate-
these acids lowers ruminal pH to below normal fermentation
based diets, containing greater levels of rapidly fermentable
conditions (e.g., pH < 5.6), which can last for hours (Nagaraja
carbohydrates (e.g., transition to lactation or feedlot), or even
and Titgemeyer, 2007). This disruption negatively affects animal
the abrupt consumption of these carbohydrates (e.g., sorting

© The Author(s) 2020. Published by Oxford University Press on behalf of the American Society of Animal Science.
All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.

1
2 | Journal of Animal Science, 2020, Vol. 98, No. 8

Abbreviations investigating the origins of these endotoxins and the toxicity


EPM extracellular polymetric matrix of the LPS that could potentially affect the animal. Therefore,
EU endotoxin units the goal of this review is to describe the most current data on
GIT gastrointestinal tract bacterial changes during ruminal acidosis in order to elucidate
IL interleukin which bacterial groups could be contributing to the ruminal LPS
LAB lactic acid-producing bacteria increase. Understanding the origins of LPS could potentially
LAU lactate-utilizing bacteria indicate the toxicity of this endotoxin and the correlation of LPS
LPS lipopolysaccharides with ruminal acidosis.
NFC nonfibrous carbohydrates
PUFA polyunsaturated fatty acids
SARA subacute ruminal acidosis
Ruminal Lipopolysaccharide Origins
SCFA short-chain fatty acids
SFA saturated fatty acid Despite dietary, genetic, and environmental effects, different
TLR4 Toll-like receptor 4 ruminant species have been reported to have a core ruminal
microbiome composed of a mixture of Gram-positive and Gram-
negative bacteria (Henderson et  al., 2015). In cattle, the core
health and production; however, the pathogenesis of ruminal
ruminal microbiome has been reported to comprise 45% of all
acidosis is still not well understood.
ruminal bacteria, from which five out of six phyla are Gram-
A well-researched theory is that the increased acid load
negative bacteria (Xue et  al., 2018). During ruminal acidosis,
in the rumen may damage ruminal epithelium (Owens et  al.,
the bacterial community composition of the rumen changes,
1998; Nagaraja and Titgemeyer, 2007; Aschenbach et  al.,
and the concentration of Gram-positive bacteria increase, while
2019). According to Owens et  al. (1998), the animal undergoes
Gram-negative bacteria decrease (Andersen et al., 1994a; Li et al.,
a metabolic acidosis when the base excess in the blood is
2012b). The decrease in Gram-negative bacteria during acidosis is
decreased, due to ionized acid absorption by the rumen
followed by the accumulation of residues from bacterial lysis in
epithelium, and not corrected through blood pH homeostasis.
the ruminal fluid. As shown in Figure 1, Gram-negative bacteria
The accumulation of acids in the rumen also leads to an
contain LPS in their outer membrane and that is released during
increase in the ruminal osmotic pressure, which could cause
bacterial growth, division, and lysis; although bacterial lysis is
ruminal epithelium damage (Owens et  al., 1998; Nagaraja and
normally the main contributor to free LPS (Petsch and Anspach,
Titgemeyer, 2007; Aschenbach et  al., 2019). Tissue function
2000; Yaron et al., 2000), the diet associated with ruminal acidosis
becomes impaired when the ruminal epithelium is damaged
may increase the turnover rate of bacteria, and thus, intact cells
during ruminal acidosis, which increases the chances of
may also contribute to free LPS during ruminal acidosis as well
pathogenic substances translocating into the bloodstream
(Plaizier et  al., 2014). Thus, during ruminal acidosis, LPS from
(Plaizier et  al., 2012; Aschenbach et  al., 2019). Bacteria and
Gram-negative bacteria accumulates in the ruminal fluid and
bacterial endotoxins (lipopolysaccharides [LPS]) are believed to
may be toxic to the animal (Plaizier et al., 2012), such as the LPS
be able to translocate into the bloodstream in these situations,
from Escherichia coli that is known to evoke an immune response
potentially impacting animal health and production (Plaizier
when in the bloodstream (Nagaraja et  al., 1978b; Raetz et  al.,
et al., 2012, 2018; Aschenbach et al., 2019).
1991). Interestingly, the ruminal LPS has not been characterized
Although the diversity of the microbial population in
yet and its toxicity is not well understood. Therefore, in order to
the rumen is well noted, the literature is scarce of studies
understand the toxicity level of ruminal Gram-negative bacteria,

Figure 1.  Gram-negative bacterial cell-wall structure with emphasis in the lipopolysaccharide (LPS) presence in the outer membrane. Toxicity is mainly associated with
the lipid A composition. Courtesy of Yen-Ming Hsu, AB Biosciences, Inc. Boston, Massachusetts, USA.
Monteiro and Faciola  |  3

it is important to first understand how the pattern of ruminal upregulated in the rumen epithelium due to high ruminal LPS
LPS concentration is in animals under SARA and acute ruminal concentration, and that these could be one of the reasons for
acidosis, and how they are linked to the bacterial changes in animals experiencing rumenitis while with SARA.
both disorders. Along with an increase of LPS concentration, studies have
shown that animals with SARA have reduced richness and
Subacute ruminal acidosis diversity in both ruminal and fecal bacteria (Mao et  al., 2013;
SARA, while more prevalent in lactating dairy cows due to Plaizier et al., 2017a, 2017b), with some reporting an increase in
their high NFC intake in order to meet energy demands for E. coli in both the hindgut and feces (Plaizier et al., 2017b). Plaizier
lactation, it is also common in beef cattle consuming finishing et  al. (2017a) reported that among the bacterial phyla with
diets (Kleen et al., 2003; Aschenbach et al., 2011; Li et al., 2012a). more than 0.1% of relative abundance in the rumen, two Gram-
The excessive intake of NFC leads to an imbalance between the positive phyla (Firmicutes and Actinobacteria) increased with
production and clearance of ruminal SCFA. When the production SARA, while two Gram-negative phyla decreased (Cyanobacteria
of SCFA surpasses the rate of absorption or clearance through and Verrucomicrobia); the phylum Firmicutes had the greatest
the flow of ruminal content out of the rumen, the ruminal increase, from 22.4% in control cows to 35.7% in those with SARA.
pH drops and stays below the optimal threshold (below pH of However, one of the reasons for the increase in the phylum
5.6) for at least 3 h (Gozho et al., 2005; Penner et al., 2011). The Actinobacteria for animals with SARA was the increase in its
clinical signs are reduced feed intake and milk production, family Bifidobacteriaceae. In the rumen, the main genus of this
milk fat depression, diarrhea, and laminitis. Thus, except for family is Bifidobacterium, which is a major lactic acid producer in
laminitis, the symptoms of SARA are almost imperceptible on the rumen and is known to increase its abundance during SARA
a daily basis, which makes it a difficult problem to be identified and acute ruminal acidosis (Nagaraja and Titgemeyer, 2007;
compared with acute acidosis (Kleen et al., 2003; Plaizier et al., Plaizier et  al., 2017a). Besides that, the genus Bifidobacterium is
2008, 2012). also known to produce most of the bacteriocins against Gram-
The cause of these effects remains unclear; however, ruminal positive and Gram-negative bacteria, suppressing bacterial
LPS has been suggested to be one of the factors contributing to growth and increasing bacterial lysis in nonruminant studies
these effects (Nagaraja and Titgemeyer, 2007; Plaizier et al., 2012; (Martinez et al., 2013). The bacteriocin activity from Bifidobacteria
Aschenbach et al., 2019). Because of the dietary changes during has been reported to comprise a wide pH range (pH 2 to 10), but
the transition to SARA, the ruminal pH is later reduced, which most of them have enhanced activity at pH 4 to 7, which some
increases Gram-negative bacterial lysis. Besides that, substrates of those that target Gram-negative bacteria reported to have
for Gram-negative bacteria are reduced and the growth of some adequate activity at pH 4.8 to 5.5 and temperature 37 to 40  °C
Gram-negative bacteria may be impaired. Some research also (Martinez et  al., 2013), conditions similar to SARA. In Plaizier
suggests that Gram-positive bacteria produce bacteriocins et  al. (2017a), animals with SARA also had decreased bacterial
against a wide variety of bacteria, including many Gram- diversity in the feces, and all bacterial phyla that decreased
negative bacteria (Martinez et al., 2013), and that Gram-negative in the feces of those animals with SARA were Gram-negative
bacteria themselves produce microcins against other Gram- (Tenericutes, Proteobacteria, Cyanobacteria, Fibrobacteres,
negative bacteria (Sassone-Corsi et al., 2016). These substances Verrucomicrobia), while the phylum Actinobacteria of Gram-
could also increase the lysis of Gram-negative bacteria, given positive bacteria increased as in the rumen.
that SARA is marked by an increase in Gram-positive bacteria. Plaizier et  al. (2017a) also showed that that Firmicutes
Therefore, these dietary changes increase bacterial lysis in and Bacteroidetes together represented 76.9% and 94.4% of
animals under SARA, and the lysis of Gram-negative bacteria the ruminal and fecal microbial population, respectively. The
leads to the release of cell-wall components, most notably LPS. difference from the ruminal to fecal samples was mainly due to
Past studies that evaluated LPS in the rumen reported LPS Proteobacteria, which had a relative abundance of 13.1% in the
concentration in endotoxin units (EU), which 1 EU contains 100 rumen and only 0.67% in the feces. Most notably, E. coli, which
pg of LPS; thus, as one Gram-negative bacterium contains 10– LPS has been used as a model for inflammation studies, is one
15
g of LPS, around 105 bacteria are necessary to produce 1 EU of the main bacteria from Proteobacteria. Interestingly, these
(Raetz et al., 1991). In an induced SARA experiment, Gozho et al. diets with increased grain content in order to cause SARA also
(2007) reported that cows with SARA had increased free ruminal increase the passage rate of digesta out of the rumen and allow
LPS from 24,547 EU to 128,825 EU/mL compared with animals more fermentable substrates to reach the hindgut (Van Soest,
without SARA, although LPS was not detected in the blood. 1994). Increased grain fermentation in the hindgut may cause
In a similar study, Li et al. (2012b) reported an increase in free similar effects of ruminal acidosis, which increases epithelial
LPS in both ruminal (from 10,405 to 168,391 EU/mL) and cecal damage and may allow the entry of LPS to the circulation
digesta samples (16,508 to 118,522 EU/g of wet cecal digesta). (Gressley et al., 2011). The LPS concentration has been reported
Interestingly, some studies did report increases of LPS both to increase in the hindgut (from 16,508 to 118,522 EU/g of wet
in the rumen and in the blood, although the reasons are still digesta) when grain-based diets are fed to induce SARA (Li et al.,
unknown (Khafipour et  al., 2009; Zhao et  al., 2018). Khafipour 2012b). Thus, Proteobacteria could be an important source of
et al. (2009) showed that ruminal LPS increased from 28,184 to free LPS in the hindgut of animals experiencing SARA, as this is
107,152 EU/mL in a comparison of control dairy cows to those a major Gram-negative bacteria in the hindgut and it is almost
with induced SARA, and these animals also had an increased absent in the feces (Mao et al., 2015; Plaizier et al., 2017a).
plasma LPS from <0.05 to 0.52. Later, Zhao et al. (2018) reported Mccann et al. (2014) pointed out that changes in the ruminal
that when ruminal LPS increased from 30,768 to 130,589 EU/ bacterial population of cows right after parturition are also likely
mL between control dairy cows and those with SARA, and to be associated with SARA, as these animals also have a sudden
plasma LPS increased from <0.005 to 0.21 EU/mL, these animals intake of highly fermentable carbohydrates after calving. Similar
had an upregulated cascade of proinflammatory responses. to Plaizier et  al. (2017a), Wang et  al. (2012) reported that after
According to Zhao et  al. (2018), the expression and production calving, cows had an increase of the ruminal Gram-positive
of tumor necrosis factor-α, interleukin-1ß (IL-1ß), and IL-6 were phylum Firmicutes (55% at day −21 vs. 68% at day +21 after
4 | Journal of Animal Science, 2020, Vol. 98, No. 8

calving), and a decrease in the ruminal Gram-negative phylum thrive in these conditions and further reduce the ruminal pH
Bacteroidetes (14% at day −21 to 5% at day +21 after calving). (pH < 5.0; Wells et  al., 1997). Although some authors consider
The increase in Firmicutes was mainly due to an increase in acute ruminal acidosis when pH is below the threshold of 5.2
Anaerovibrio lipolytica, Streptococcus bovis, and Lactobacillus spp. (Nagaraja and Titgemeyer, 2007), whenever ruminal pH becomes
The last two bacteria are ruminal species responsible for the that low, it is because ruminal lactate concentration is greater
fermentation of starch and sugar mainly into lactate, thus than normal (>5.0  mM) and animals are undergoing acute
associated with SARA and acute ruminal acidosis (Nagaraja ruminal acidosis. Animals experiencing acute ruminal acidosis
and Titgemeyer, 2007). However, the decrease in Bacteroidetes have been reported to reach ruminal lactate concentrations from
was mainly related to a decrease in the family Rikenellaceae. 50 to 120  mM (Nagaraja et  al., 1998; Nagaraja and Titgemeyer,
A  similar decrease in Rikenellaceae was also observed in beef 2007). Clinical signs of acute ruminal acidosis include loss of
cattle transitioning from bermudagrass hay diets (crude protein appetite, ruminitis, laminitis, polioencephalomalacia, and death
[CP]  =  11.2%; neutral detergent fiber [NDF]  =  67%) to grazed (Nocek, 1997; Nagaraja et  al., 1998; Nagaraja and Titgemeyer,
winter wheat diets (CP  =  19.9%; NDF  =  44%), in an experiment 2007).
investigating the transition from low- to high-quality forages Unlike SARA, acute ruminal acidosis has been reported to
(Pitta et  al., 2010). This study also showed that the latter diet, depress SCFA concentration (SCFA < 100  mM), and this may
which was a more digestible diet (in vitro dry matter [DM] happen due to a few reasons: 1) ruminal pH in animals with acute
digestibility  =  57.2% vs. 80.3%), led to a decrease of the genus ruminal acidosis may impair fermentation by some bacterial
Rikenella, which is from Rikenellaceae family reported to communities until pH returns to normal conditions (Nagaraja
decrease after the transition to lactation. The decrease and lysis et  al., 1998); 2)  SCFA concentration may decrease because of
of these Gram-negative bacteria are another possible sources of the influx of liquid from the osmotic changes caused by acute
ruminal free LPS. acidosis (Owens et al., 1998); and 3) because the pKa of SCFA is
Recently, Dai et al. (2019) in a study evaluating LPS dosing into around 4.9, most of the SCFA in ruminal acidosis conditions will
dual-flow continuous culture fermenters in order to simulate be unprotonated, which should, in contrast, increase the SCFA
SARA conditions showed that LPS dosing decreased bacterial N absorption by the rumen epithelium (Bergman, 1990). Whether
compared with normal fermentation conditions, which could the major reason for lower ruminal SCFA concentration is
represent a decrease in microbial mass. Interestingly, this study because of depressed SCFA production, the ruminal influx of
showed that LPS dosing increased the relative abundance of some liquid diluting the SCFA concentration, or the increase in SCFA
bacteria related to starch digestion and pH reduction in ruminal absorption, Harmon et  al. (1985) reported that lactate isomers
fermentation (Anaeroplasma, Ruminobacter, Succiniclasticum, have different absorption rates by the ruminal epithelium.
Succinimonas, and Succinivibrio), despite those bacteria being The isomer l-lactate has considerably greater absorption than
Gram-negative. In a follow-up study, Dai et  al. (2020) reported d-lactate, and this makes the accumulation of d-lactate one of
that LPS may be used as a substrate for the growth of bacteria the possible causes of low ruminal pH (Harmon et al., 1985).
related to acidosis (S.  bovis, Succinivibrio dextrinosolvens, In acute ruminal acidosis, the association of Gram-negative
Lactobacillus ruminis, and Selenomonas ruminantium), and that bacterial LPS and acidosis was first reported by Mullenax et al.
when LPS is present in a medium containing mixed bacteria (1966) and Nagaraja et al. (1978b), which suggested that ruminal
from ruminal fermentation, bacterial genes associated with the bacteria contained an endotoxin similar to those found in
biosynthesis of LPS were also increased. E. coli and Salmonella typhosa. Mullenax et al. (1966) injected the
bacterial endotoxin extracted from the rumen in sheep and beef
Acute ruminal acidosis cattle and reported an animal toxicity similar to that of E. coli.
Acute ruminal acidosis has been reported to be more prevalent Nagaraja et  al. (1978b) showed that although similar to E.  coli,
in beef than dairy cattle, specifically in feedlot cattle consuming ruminal bacterial endotoxins had lesser toxicity when tested in
high-grain diets (Nagaraja et  al., 1998; Owens et  al., 1998; embryos, mice, rabbits, and Limulus lysate. The ruminal bacteria
Nagaraja and Titgemeyer, 2007). The cause of acute ruminal in the latter study was extracted from the rumen of cattle fed
acidosis is similar to SARA, however, at a greater level of highly hay or grains, containing both live and dead bacteria. The yield
fermentable carbohydrate intake (mainly starch). Usually, high- of total endotoxin was greater for hay-fed cattle, as these diets
producing dairy cows consume around 30% starch in their promote the growth of fibrolytic bacteria that are mostly Gram-
diet DM (60:40 forage to concentrate ratio), while beef cattle in negative. In a subsequent study (Nagaraja et al., 1978a), animals
feedlot diets may have up to double the starch concentration of under acute acidosis had an increased concentration of ruminal
dairy rations (10:90 forage to concentrate ratio). The transition free endotoxins by 18 times in 12  h. In that study, an in vitro
to these diets can cause the onset of acute ruminal acidosis, experiment also showed that the decrease in Gram-negative
as the ruminal microbial population of these animals are not bacteria was not associated with an increase in endotoxins, and
adapted to such high concentrations of starch (Owens et  al., thus, the release of endotoxins from intact cells seemed to have
1998; Nagaraja and Titgemeyer, 2007). increased during acidotic conditions. The authors suggested
Lactic acid-producing bacteria (LAB), specifically S.  bovis, that such an increase in free endotoxins may be due to other
rapidly ferment starch and glucose to produce lactate in the factors, such as low ruminal pH; however, the increase in free
rumen (Hungate, 1966; Nocek, 1997), reaching a doubling time endotoxins may be related as well to the large increase in Gram-
of just a few minutes (McAllister et  al., 1990). Lactate starts to positive bacteria during this period (e.g., bacteriocins).
accumulate because lactate-utilizing bacteria (LAU), particularly Similar to SARA, LPS concentration has been reported to
Megasphaera elsdenii and S.  ruminantium (predominant LAU), increase due to an increase in Gram-negative bacterial lysis
replicate slowly and take weeks to considerably increase their (Nagaraja and Titgemeyer, 2007). However, animals under
population in the rumen (Nocek, 1997; Russell and Rychlik, acute acidosis conditions have been reported to have free LPS
2001). Consequently, lactate accumulates quickly, thus dropping concentrations of up to 408 EU/mL of ruminal fluid (Andersen
ruminal pH. Because of a resistance to more acidic pH and due et al., 1994a), which is considerably greater compared with SARA.
to bacteriocin-mediated response to S.  bovis, Lactobacillus spp. Some studies also showed that the effects of acute ruminal
Monteiro and Faciola  |  5

acidosis may be more associated with the LPS toxicity to the decreased. The increase in Bacteroidetes is the opposite of what
animal than the LPS presence in the rumen itself (Dougherty it is expected in animals transitioning to high concentrate diets,
et  al., 1975; Aiumlamai et  al., 1992; Andersen et  al., 1994b), and because Bacteroidetes is the major Gram-negative bacteria
as these studies did not report an immune response as that in the rumen, further investigation is necessary. Interestingly,
observed by Mullenax et  al. (1966) and Nagaraja et  al. (1978b). two species decreased gradually in the study of Fernando et al.
The lack of LPS toxicity in some studies is another reason for (2010), Butyrivibrio fibrisolvens and Fibrobacter succinogenes. The
reviewing bacterial changes in these animals and thus possibly former is known as another major bacteriocin producers in the
tracking the origins of LPS during acute ruminal acidosis. rumen (Azevedo et al., 2015), while the latter is one of the main
Earlier research reported that increases in genera of bacteria Gram-negative cellulolytic bacteria in the rumen and contains
associated with lactate production in the rumen and possible LPS (Ransom-Jones et al., 2012).
association with acute acidosis (Nagaraja and Titgemeyer, 2007). Also using step-up protocols, Granja-Salcedo et  al. (2016)
These bacteria were Bifidobacterium, Butyrivibrio, Eubacterium, reported that the increase of grains in the diet (up to 80%
Lactobacillus, Mitsuokella, Prevotella, Ruminobacter, Selenomonas, concentrate) was correlated with a gradual decrease in the
Streptococcus, Succinimonas, and Succinivibrio. Besides the populations of Ruminococcus flavefaciens and Ruminococcus albus.
bacteriocins produced by Bifidobacterium spp. explained earlier These are other major cellulolytic bacteria that, although they
in the Subacute ruminal acidosis section, LAB bacteriocins are are Gram-positive, they have been reported to produce a series
some of the main bacteriocins characterized to date, as those of bacteriocins in the rumen (Morrison and Miron, 2000; Brulc
are of great importance for the food industry sector (Cotter et al., et al., 2011; Azevedo et al., 2015). Therefore, the combination of
2005; Perez et  al., 2014; Silva et  al., 2018). In the rumen, most slow increases of substrate for the production of organic acids,
of the characterized bacteriocins are produced by the phylum especially lactate, and the gradual removal of bacteria that can
Firmicutes, specifically Clostridiales and Lactobacillales, contribute to ruminal acute acidosis (through bacteriocins and
yet most of the LAB that increase in the rumen under acute LPS) may be another reason for the success of step-up diets.
ruminal acidosis have shown to produce bacteriocins (Russell However, because these protocols take weeks to be completed,
and Mantovani, 2002; Azevedo et al., 2015). The bacteriocins in they are not usually viable economically.
the rumen, although not completely understood, indicate that
the increase in LAB and Gram-positive bacteria during acute
ruminal acidosis may be associated with an increase in Gram-
negative bacterial lysis, as explained earlier.
Ruminal Lipopolysaccharide Toxicity
Past reviews on ruminal acidosis have emphasized the As described above, there is evidence that free LPS increases in
importance of animal adaptation to high concentrate diets in the rumen and hindgut of animals experiencing both SARA and
order to decrease the chances of acute ruminal acidosis (Owens acute ruminal acidosis. There is also evidence suggesting that
et  al., 1998; Nagaraja and Titgemeyer, 2007). This is a common Gram-negative bacterial lysis increases in the rumen ruminal
practice in the beef cattle industry, and it is known as a step-up acidosis (as indicated by increased LPS concentrations), despite
protocol, a method that allows LAU (mainly M.  elsdenii and some studies not finding a decrease in Gram-negative bacteria
S.  ruminantium) to sufficiently increase their population in the concentration. However, we could not find many studies
rumen in order to metabolize lactate produced from LAB. As that analyzed the LPS toxicity of individual ruminal bacteria,
explained earlier, LAU take longer to replicate in the rumen, and analyzed the toxicity of the bacteria that decrease during
thus, animals are fed diets with increasing levels of concentrate SARA and acute ruminal acidosis, or even that evaluated the
after being adapted to previous levels; these steps take a few reason why LPS concentration in the hindgut is also elevated
weeks to be completed. Although step-up protocols are costly, in animals experiencing such problems. As mentioned earlier,
they may eventually dilute the LPS effects from the microbial when ruminal bacterial LPS was extracted from mixed ruminal
changes, which happens either by reducing Gram-negative bacteria in both hay and grain-fed animals, ruminal LPS had less
bacterial lysis due to a less abrupt change of bacterial substrates toxicity than those of E. coli and S. typhi in a variety of models
or by the diluted effects of bacterial LPS toxicity. In any way, when (Nagaraja et  al., 1978b). We could find only one study that, in
step-up protocols are taken into account, besides allowing time fact, analyzed the toxicity of a single rumen bacterium and
for LAU to adequately replicate in the rumen, these procedures that was M. elsdenii (Nagaraja et al., 1979). However, the authors
may contribute to a decrease in the LPS effects as well. concluded that M. elsdenii LPS had low potency and would likely
Although the literature is scarce of studies evaluating not be the major problem of ruminal acidosis.
ruminal LPS changes in animals in step-up diets, some studies Thus, three possibilities are likely: 1)  total free ruminal
have evaluated bacterial changes during this period (Wells LPS, although less toxic than E.  coli and S.  typhosa LPS, has
et  al., 1997; Fernando et  al., 2010; Granja-Salcedo et  al., 2016). a concentration that overloads the ruminant detoxification
Wells et  al. (1997) reported that microbial changes are less system and triggers the effects of ruminal acidosis; a topic
expressive in animals gradually adapted to high concentrate reviewed by Aschenbach et  al. (2019); 2)  free LPS from specific
diets (up to 80% of cereal grain). According to the authors, bacterial groups are as toxic as that from E. coli and S. typhosa;
changes in ruminal pH were also not as pronounced as those however, such LPS have their toxic effects diluted into the
seen in animals with an abrupt transition to high concentrate pool of free ruminal LPS and decrease the toxicity of total free
diets; thus, as ruminal pH was not a major factor for microbial ruminal LPS; and 3) the difference in the LPS pattern in animals
changes in their study, the observed microbial changes in the transitioning to ruminal acidosis could be a signaling pathway
study of Wells et al. (1997) were reported to be mostly associated by the animal to detect gastrointestinal tract (GIT) changes.
with bacteriocins produced by Lactobacillus spp. To understand which of these possibilities is more likely
Fernando et  al. (2010), in a similar study, allowed animals to happen, it is important to understand how ruminal LPS
to have 1  wk of adaptation per diet, from 100% forage to up compares to other types of LPS, such as from E. coli. As explained
to 80% of concentrate in the diet. In their study, the phylum earlier, LPS is released in the GIT during bacterial lysis, growth,
Bacteroidetes gradually increased, while Fibrobacteres and division (Petsch and Anspach, 2000; Yaron et  al., 2000);
6 | Journal of Animal Science, 2020, Vol. 98, No. 8

however, bacterial lysis is the method that contributes the most immune response in ruminants: paracellular and transcellular
to free LPS concentration. The structure of LPS is composed of (Tomlinson and Blikslager, 2004; Mani et  al., 2012; Eckel and
three parts (Figure  1): a distal polysaccharide (O-antigen), a Ametaj, 2016). First, ruminal epithelial damage has been shown
core oligosaccharide, and the lipid A that attaches to the outer to be prevalent during ruminal acidosis; because of low pH and
membrane of Gram-negative bacteria (Raetz and Whitfield, high concentration of organic acids, a high osmotic pressure
2002; Matsuura, 2013). The distal polysaccharide has been promotes a barrier opening and endotoxins may translocate
reported to have up to 50 oligosaccharides composed of two to paracellularly into the epithelium (Aschenbach et  al., 2019).
eight sugar monomers, which may vary in a strain to species Undigested starch reaching the large intestine in these high-
level (Matsuura, 2013). This structure plays a major role in grain diets could also cause the development of lower gut
bacterial colonization, adhesion, and escape from predation acidosis, leading to more severe epithelium damage than that
(Moran, 2008). The variation of the core oligosaccharide only occurring in the rumen (Steele et  al., 2016), thus increasing
happens at a higher taxonomic level, and such variation is LPS paracellular translocation. The second pathway is the
not as common as the variation of the distal part (Raetz and transcellular translocation, which is mediated through TLR4
Whitfield, 2002). These two structures have been reported to be receptors explained before (Mani et  al., 2012). Of the total LPS
present in wild-type strains only and not in cultured strains transcytosis by the epithelial cells from the lumen, only 15%
(Raetz and Whitfield, 2002). Interestingly, these structures have reaches the basolateral side, with the remaining being either
been associated with bacterial resistance to antibiotics (Raetz recycled back to the lumen (38%) or packed into lysosomes (47%)
and Whitfield, 2002). Antibiotics have been used in ruminant (Beatty et al., 1999). However, chylomicrons have shown to have
nutrition, especially in feedlot beef cattle diets in which acute a high affinity to LPS, and thus, LPS can also enter the Golgi
acidosis is more prevalent (Russell and Strobel, 1989). Indeed, apparatus and be transported through the lymph and blood
concerns regarding bacterial resistance by Gram-negative (Ghoshal et al., 2009).
bacteria to antibiotics have been raised; although these were In a recent study evaluating which bacterial group could be
not associated directly with LPS at that time, the resistance was contributing to the pool of LPS in the human GIT, d′Hennezel
believed to be from the outer-membrane protection of these et  al. (2017) reported that the LPS from the Bacteroidales
bacteria (Russell and Strobel, 1989; Russell and Rychlik, 2001). are the most abundant in human feces (79% to 92.4% of the
Lastly, the lipid A  is the primary toxic structure of bacterial total LPS). Similar to Mullenax et al. (1966) and Nagaraja et al.
LPS, and it is present in high concentrations in the cell-wall (1978b), d′Hennezel et al. (2017) tested the toxicity of the total
of Gram-negative bacteria (around 106 residues/cell; Raetz and LPS from human feces against the LPS from E.  coli, however,
Whitfield, 2002). The lipid A from LPS is the portion that attaches in peripheral blood mononuclear cells (lymphocytes and
to macrophage and endothelial cell receptors (Toll-like receptor monocytes). The authors showed that when 10 LPS molecules
4 [TLR4]) to cause an immune response (Poltorak et  al., 1998; from mixed fecal samples were present in the media with 1
Hoshino et al., 1999; Aderem and Ulevitch, 2000). However, TLR4 E.  coli LPS, TLR4 signaling was not activated, which in some
receptors have also been reported to be present in the surface cases only 1 LPS from mixed fecal samples was able to stop
of human intestinal epithelial cells, and it is believed to play a the activity of 1 LPS from E.  coli. Later, in order to evaluate if
major role in the gut LPS signaling (Abreu, 2010). the LPS immunoinhibitory capacity could be an order-wide
Toxicity of LPS comes from the composition of lipid A (Netea feature, the authors tested the toxicity of LPS only from species
et al., 2002), as its acyl groups are correlated with TLR4 signaling, of the order Bacteroidales against E.  coli LPS in similar cells.
and has been demonstrated in Salmonella typhimurium to d′Hennezel et  al. (2017) showed that from the Bacteroidales
decrease its toxicity with decreased acylation when tested in LPS species tested, all were underacylated, which caused an
human macrophage cells (Matsuura et  al., 2012). These acyl inhibitory effect on TLR4 signaling (silent LPS), thus showing
groups may vary from 4 to 6 acyl chains, being 4 and 5 acyl groups that immunoinhibitory features of LPS may be conserved at
shown to be the LPS with less toxicity (immunoinhibitory/silent higher taxonomic levels.
LPS), and 6 the most likely to activate receptors and cause an Furthermore, d′Hennezel et  al. (2017) extracted the
immune response (E. coli LPS) (Netea et al., 2002; Matsuura, 2013). extracellular polymeric matrix (EPM; or extracellular polymeric
In a study evaluating the lipid A structure of bacterial LPS from substances) from 29 strains of the 4 main phyla in the human GIT
Yersinia, Yersinia enterocolitica, and Yersinia pseudotuberculosis, in order to evaluate its association with inflammation. The EPM
Rebeil et al. (2004) reported that acylation may vary depending is a matrix with active compounds surrounding the bacterial
on the temperature in which these bacteria are cultured. In their cell wall that helps in the colonization process through biofilm
study, Rebeil et al. (2004) found that 21 °C incubation temperature formation and intercellular aggregation (Rossi et  al., 2015).
promoted hexa-acylation of lipid A  and 37  °C promoted tetra- The authors reported that the bacterial EPM downregulated
acylation. Interestingly, the LPS used for comparisons with the inflammation through TLR4 and TLR2 by 4- to 20-fold in primary
ruminal LPS was E.  coli and S.  typhosa (Nagaraja et  al., 1978b), human immune cells, with the EPM of Bacteroidales reducing
which may have the most toxic LPS activities, suggesting inflammation the most; yet, EPM from Bacteroidales had one of
that in further evaluations: 1)  the lipid A  structure of ruminal the highest LPS contents of the bacterial EPM tested. This study
bacteria LPS should be characterized to understand its acylation was performed with fecal samples, which contain a less diverse
differences per bacterial group (mainly from those that decrease microbiome compared with rumen, thus providing a variety
during ruminal acidosis); 2)  silent ruminal LPS effects (tetra of LPS inferior to that of the rumen. However, these findings
and penta-acylated lipid A) should also be taken into account, from d′Hennezel et al. (2017) can potentially bring new context
as silent LPS does not trigger an immune response; and 3) the to ruminal acidosis research, as Bacteroidales comprise some
possibility of some ruminal bacteria having highly toxic lipid of the main genera found in the rumen, such as the Prevotella
A  LPS replacing those with silent LPS during ruminal acidosis spp. as explained previously (Henderson et al., 2015). Also, their
should also be considered. research sheds light on how future studies could analyze the
Regarding LPS translocation, there are two primary acylation of specific ruminal bacteria and test their toxicity in
pathways in which ruminal or caecal LPS could trigger an the models herein tested.
Monteiro and Faciola  |  7

Lastly, Mani et  al. (2013) using pigs as a model to absorption in the regulation of ruminal pH. J. Anim. Sci.
study intestinal endotoxin transport reported that n-3 89:1092–1107. doi:10.2527/jas.2010-3301
polyunsaturated fatty acids (PUFA) decreased LPS signaling Aschenbach,  J.  R., Q.  Zebeli, A.  K.  Patra, G.  Greco, S.  Amasheh,
possibly by also modulating TLR4, and that saturated fatty acid and G. B. Penner. 2019. Symposium Review: The importance
of the ruminal epithelial barrier for a healthy and productive
(SFA) supplementation increased LPS serum concentration.
cow. J. Dairy Sci. 102:1866–1882. doi:10.3168/jds.2018-15243
The mechanism by which fatty acids modulate TLR4 receptor is
Azevedo,  A.  C., C.  B.  Bento, J.  C.  Ruiz, M.  V.  Queiroz, and
directly associated with the lipid A composition of LPS. Nontoxic H.  C.  Mantovani. 2015. Distribution and genetic diversity of
LPS has been reported to have PUFA in its lipid A composition bacteriocin gene clusters in rumen microbial genomes. Appl.
and showed an antagonistic effect in mice when compared Environ. Microbiol. 81:7290–7304. doi:10.1128/AEM.01223-15
with LPS with lipid A  containing only SFA in its composition Beatty,  W.  L., S.  Méresse, P.  Gounon, J.  Davoust, J.  Mounier,
(Qureshi et  al., 1991). On the other hand, SFA (e.g., lauric and P.  J.  Sansonetti, and J.  P.  Gorvel. 1999. Trafficking of Shigella
myristic acids) are part of the lipid A  of LPS, and thus, SFA in lipopolysaccharide in polarized intestinal epithelial cells. J.
the lumen induce TLR4 activation, triggering LPS absorption (Lee Cell Biol. 145:689–698. doi:10.1083/jcb.145.4.689
et  al., 2004). Although cattle diets have low fatty acid content Bergman, E. N. 1990. Energy contributions of volatile fatty acids
from the gastrointestinal tract in various species. Physiol. Rev.
(<7% ether extract), the majority of the fatty acids that leave the
70:567–590. doi:10.1152/physrev.1990.70.2.567
rumen are SFA, which in high LPS conditions, such as in SARA
Brulc, J. M., C. J. Yeoman, M. K. Wilson, M. E. Berg Miller, P. Jeraldo,
and acute acidosis, this could be another possibility of triggering S. Jindou, N. Goldenfeld, H. J. Flint, R. Lamed, I. Borovok, et al.
LPS absorption and an immune response. 2011. Cellulosomics, a gene-centric approach to investigating
the intraspecific diversity and adaptation of Ruminococcus
flavefaciens within the rumen. PLoS One. 6:e25329. doi:10.1371/
Conclusion Remarks journal.pone.0025329
Cotter, P. D., C. Hill, and R. P. Ross. 2005. Bacteriocins: developing
In conclusion, although pH and lactate have been used as innate immunity for food. Nat. Rev. Microbiol. 3:777–788.
parameters for the pathogenesis for ruminal acidosis, there is doi:10.1038/nrmicro1273
evidence that LPS could greatly contribute to acidosis. The studies Dai,  X., E.  M.  Paula, A.  L.  J.  Lelis, L.  G.  Silva, V.  L.  N.  Brandao,
described here indicate that ruminal acidosis causes a dysbiosis H.  F.  Monteiro, P.  Fan, K.  C.  Jeong, A.  P.  Faciola, and
in the ruminal microbiome, and some Gram-negative bacteria S.  R.  Poulson. 2019. Effects of lipopolysaccharide dosing on
decrease their abundance in the rumen and lysis of these bacteria bacterial community composition and fermentation in a
increase, as indicated by the increase in free LPS. Greater LPS dual-flow continuous culture system. J. Dairy Sci. 102:334–350.
concentration during ruminal acidosis has also been reported in doi:10.3168/jds.2018-14807
Dai,  X., T.  J.  Hackmann, R.  R.  Lobo, A.  P.  Faciola. 2020. Applied
the hindgut, which increases the chances of evoking an immune
and Environmental Microbiology 86:e02193-19. doi:10.1128/
response. Other studies show that besides LPS increasing in the
AEM.02193-19
rumen during this time, increased LPS could also trigger the Dougherty, R. W., K. S. Coburn, H. M. Cook, and M. J. Allison. 1975.
growth of ruminal bacteria associated with acidosis. Therefore, Preliminary study of appearance of endotoxin in circulatiory
future studies should be performed to characterize ruminal system of sheep and cattle after induced grain engorgement.
LPS and its lipid A structure in order to accurately determine its Am. J.  Vet. Res. 36:831–832. Available from https://pubmed.
toxicity so that more strategies could be developed in order to ncbi.nlm.nih.gov/1147337/
overcome this major challenge in ruminant nutrition. Eckel,  E.  F., and B.  N.  Ametaj. 2016. Invited Review: Role of
bacterial endotoxins in the etiopathogenesis of periparturient
diseases of transition dairy cows. J. Dairy Sci. 99:5967–5990.
doi:10.3168/jds.2015-10727
Conflict of interest statement
Fernando,  S.  C., H.  T.  Purvis 2nd, F.  Z.  Najar, L.  O.  Sukharnikov,
The authors declare no real or perceived conflicts of interest. C. R. Krehbiel, T. G. Nagaraja, B. A. Roe, and U. Desilva. 2010.
Rumen microbial population dynamics during adaptation
to a high-grain diet. Appl. Environ. Microbiol. 76:7482–7490.
Literature Cited doi:10.1128/AEM.00388-10
Abreu,  M.  T. 2010. Toll-like receptor signalling in the intestinal Ghoshal,  S., J.  Witta, J.  Zhong, W.  de  Villiers, and E.  Eckhardt.
epithelium: how bacterial recognition shapes intestinal 2009. Chylomicrons promote intestinal absorption of
function. Nat. Rev. Immunol. 10:131–144. doi:10.1038/nri2707 lipopolysaccharides. J. Lipid Res. 50:90–97. doi:10.1194/jlr.
Aderem,  A., and R.  J.  Ulevitch. 2000. Toll-like receptors in the M800156-JLR200
induction of the innate immune response. Nature 406:782– Gozho,  G.  N., D.  O.  Krause, and J.  C.  Plaizier. 2007. Ruminal
787. doi:10.1038/35021228 lipopolysaccharide concentration and inflammatory response
Aiumlamai,  S., H.  Kindahl, G.  Fredriksson, L.  E.  Edqvist, during grain-induced subacute ruminal acidosis in dairy cows.
L. Kulander, and O. Eriksson. 1992. The role of endotoxins in J. Dairy Sci. 90:856–866. doi:10.3168/jds.S0022-0302(07)71569-2
induced ruminal acidosis in calves. Acta Vet. Scand. 33:117–127. Gozho,  G.  N., J.  C.  Plaizier, D.  O.  Krause, A.  D.  Kennedy, and
Available from https://pubmed.ncbi.nlm.nih.gov/1502995/ K.  M.  Wittenberg. 2005. Subacute ruminal acidosis induces
Andersen,  P.  H., B.  Bergelin, and K.  A.  Christensen. 1994a. ruminal lipopolysaccharide endotoxin release and triggers an
Effect of feeding regimen on concentration of free inflammatory response. J. Dairy Sci. 88:1399–1403. doi:10.3168/
endotoxin in ruminal fluid of cattle. J. Anim. Sci. 72:487–491. jds.S0022-0302(05)72807-1
doi:10.2527/1994.722487x Granja-Salcedo,  Y.  T., C.  S.  Ribeiro  Júnior, R.  B.  de  Jesus,
Andersen,  P.  H., M.  Hesselholt, and N.  Jarløv. 1994b. Endotoxin A.  S.  Gomez-Insuasti, A.  R.  Rivera, J.  D.  Messana,
and arachidonic acid metabolites in portal, hepatic and R.  C.  Canesin, and T.  T.  Berchielli. 2016. Effect of different
arterial blood of cattle with acute ruminal acidosis. Acta Vet. levels of concentrate on ruminal microorganisms and rumen
Scand. 35:223–234. Available from https://pubmed.ncbi.nlm. fermentation in Nellore steers. Arch. Anim. Nutr. 70:17–32. doi
nih.gov/7847191/ :10.1080/1745039X.2015.1117562
Aschenbach,  J.  R., G.  B.  Penner, F.  Stumpff, and G.  Gäbel. 2011. Gressley, T. F., M. B. Hall, and L. E. Armentano. 2011. Ruminant
Ruminant Nutrition Symposium: Role of fermentation acid Nutrition Symposium: Productivity, digestion, and health
8 | Journal of Animal Science, 2020, Vol. 98, No. 8

responses to hindgut acidosis in ruminants. J. Anim. Sci. Matsuura, M., K. Kawasaki, K. Kawahara, and M. Mitsuyama. 2012.
89:1120–1130. doi:10.2527/jas.2010-3460 Evasion of human innate immunity without antagonizing
Harmon,  D.  L., R.  A.  Britton, R.  L.  Prior, and R.  A.  Stock. 1985. TLR4 by mutant Salmonella enterica serovar Typhimurium
Net portal absorption of lactate and volatile fatty acids having penta-acylated lipid A. Innate Immun. 18:764–773.
in steers experiencing glucose-induced acidosis or fed a doi:10.1177/1753425912440599
70% concentrate diet ad libitum. J. Anim. Sci. 60:560–569. McAllister, T. A., K. J. Cheng, L. M. Rode, and C. W. Forsberg. 1990.
doi:10.2527/jas1985.602560x Digestion of barley, maize, and wheat by selected species
Henderson,  G., F.  Cox, S.  Ganesh, A.  Jonker, W.  Young, and of ruminal bacteria. Appl. Environ. Microbiol. 56:3146–3153.
P.  H.  Janssen; Global Rumen Census Collaborators. 2015. doi:10.1128/AEM.56.10.3146-3153.1990
Rumen microbial community composition varies with diet McCann,  J.  C., T.  A.  Wickersham, and J.  J.  Loor. 2014. High-
and host, but a core microbiome is found across a wide throughput methods redefine the rumen microbiome and its
geographical range. Sci. Rep. 5:14567. doi:10.1038/srep14567 relationship with nutrition and metabolism. Bioinform. Biol.
d′Hennezel, E., S. Abubucker, L. O. Murphy, and T. W. Cullen. 2017. Insights 8:109–125. doi:10.4137/BBI.S15389
Total lipopolysaccharide from the human gut microbiome Miller-Cushon, E. K., and T. J. DeVries. 2017. Feed sorting in dairy
silences toll-like receptor signaling. mSystems 2: e00046-17. cattle: causes, consequences, and management. J. Dairy Sci.
doi:10.1128/mSystems.00046-17 100:4172–4183. doi:10.3168/jds.2016-11983
Hoshino,  K., O.  Takeuchi, T.  Kawai, H.  Sanjo, T.  Ogawa, Moran, A. P. 2008. Relevance of fucosylation and Lewis antigen
Y.  Takeda, K.  Takeda, and S.  Akira. 1999. Cutting edge: Toll- expression in the bacterial gastroduodenal pathogen
like receptor 4 (TLR4)-deficient mice are hyporesponsive Helicobacter pylori. Carbohydr. Res. 343:1952–1965. doi:10.1016/j.
to lipopolysaccharide: evidence for TLR4 as the Lps gene carres.2007.12.012
product. J. Immunol. 162:3749–3752. Available from https:// Morrison,  M., and J.  Miron. 2000. Adhesion to cellulose
www.jimmunol.org/content/162/7/3749 by Ruminococcus albus: a combination of cellulosomes
Hungate,  R.  E. 1966. The rumen and its microbes. New York (NY): and Pil-proteins? FEMS Microbiol. Lett. 185:109–115.
Elsevier Science, Academic Press. doi:10.1111/j.1574-6968.2000.tb09047.x
Khafipour, E., D. O. Krause, and J. C. Plaizier. 2009. A grain-based Mullenax, C. H., R. F. Keeler, and M. J. Allison. 1966. Physiologic
subacute ruminal acidosis challenge causes translocation responses of ruminants to toxic factors extracted from
of lipopolysaccharide and triggers inflammation. J. Dairy Sci. rumen bacteria and rumen fluid. Am. J.  Vet. Res. 27:857–
92:1060–1070. doi:10.3168/jds.2008-1389 868. Available from https://www.cabdirect.org/cabdirect/
Kleen,  J.  L., G.  A.  Hooijer, J.  Rehage, and J.  P.  Noordhuizen. abstract/19671401936
2003. Subacute ruminal acidosis (SARA): a review. Nagaraja, T. G., E. E. Bartley, L. R. Fina, and H. D. Anthony. 1978a.
J. Vet. Med. A.  Physiol. Pathol. Clin. Med. 50:406–414. Relationship of rumen gram-negative bacteria and free
doi:10.1046/j.1439-0442.2003.00569.x endotoxin to lactic acidosis in cattle. J. Anim. Sci. 47:1329–
Lee, J. Y., L. Zhao, H. S. Youn, A. R. Weatherill, R. Tapping, L. Feng, 1337. doi:10.2527/jas1978.4761329x
W. H. Lee, K. A. Fitzgerald, and D. H. Hwang. 2004. Saturated Nagaraja,  T.  G., E.  E.  Bartley, L.  R.  Fina, H.  D.  Anthony, and
fatty acid activates but polyunsaturated fatty acid inhibits R.  M.  Bechtle. 1978b. Evidence of endotoxins in the rumen
Toll-like receptor 2 dimerized with toll-like receptor 6 or 1. J. bacteria of cattle fed hay or grain. J. Anim. Sci. 47:226–234.
Biol. Chem. 279:16971–16979. doi:10.1074/jbc.M312990200 doi:10.2527/jas1978.471226x
Li,  S., G.  N.  Gozho, N.  Gakhar, E.  Khafipour, D.  O.  Krause, and Nagaraja, T. G., L. R. Fina, B. A. Lassman, E. E. Bartley, H. D. Anthony,
J.  C.  Plaizier. 2012a. Evaluation of diagnostic measures for D.  A.  Sapienza, and B.  E.  Brent. 1979. Characterization of
subacute ruminal acidosis in dairy cows. Can. J.  Anim. Sci. endotoxin from the rumen bacterium Megasphaera elsdenii.
92:353–364. doi:10.4141/cjas2012-004 Am. J. Vet. Res. 40:35–39. Available from http://europepmc.org/
Li,  S., E.  Khafipour, D.  O.  Krause, A.  Kroeker, J.  C.  Rodriguez- article/MED/453682
Lecompte, G.  N.  Gozho, and J.  C.  Plaizier. 2012b. Effects of Nagaraja,  T.  G., M.  L.  Galyean, and N.  A.  Cole. 1998. Nutrition
subacute ruminal acidosis challenges on fermentation and and disease. Vet. Clin. North Am. Food Anim. Pract. 14:257–277.
endotoxins in the rumen and hindgut of dairy cows. J. Dairy doi:10.1016/s0749-0720(15)30253-x
Sci. 95:294–303. doi:10.3168/jds.2011-4447 Nagaraja,  T.  G., and E.  C.  Titgemeyer. 2007. Ruminal acidosis
Mani,  V., J.  H.  Hollis, and N.  K.  Gabler. 2013. Dietary oil in beef cattle: the current microbiological and nutritional
composition differentially modulates intestinal endotoxin outlook. J. Dairy Sci. 90:E17–E38. doi:10.3168/jds.2006-478
transport and postprandial endotoxemia. Nutr. Metab. (Lond). Netea,  M.  G., M.  van  Deuren, B.  J.  Kullberg, J.  M.  Cavaillon, and
10:6. doi:10.1186/1743-7075-10-6 J. W. Van der Meer. 2002. Does the shape of lipid A determine
Mani,  V., T.  E.  Weber, L.  H.  Baumgard, and N.  K.  Gabler. the interaction of LPS with Toll-like receptors? Trends Immunol.
2012. Growth and Development Symposium: Endotoxin, 23:135–139. doi:10.1016/s1471-4906(01)02169-x
inflammation, and intestinal function in livestock. J. Anim. Sci. Nocek,  J.  E. 1997. Bovine acidosis: implications on laminitis. J.
90:1452–1465. doi:10.2527/jas.2011-4627 Dairy Sci. 80:1005–1028. doi:10.3168/jds.S0022-0302(97)76026-0
Mao,  S., M.  Zhang, J.  Liu, and W.  Zhu. 2015. Characterising the Owens,  F.  N., D.  S.  Secrist, W.  J.  Hill, and D.  R.  Gill. 1998.
bacterial microbiota across the gastrointestinal tracts of Acidosis in cattle: a review. J. Anim. Sci. 76:275–286.
dairy cattle: membership and potential function. Sci. Rep. doi:10.2527/1998.761275x
5:16116. doi:10.1038/srep16116 Penner, G. B., M. A. Steele, J. R. Aschenbach, and B. W. McBride.
Mao, S. Y., R. Y. Zhang, D. S. Wang, and W. Y. Zhu. 2013. Impact 2011. Ruminant Nutrition Symposium: Molecular adaptation
of subacute ruminal acidosis (SARA) adaptation on rumen of ruminal epithelia to highly fermentable diets. J. Anim. Sci.
microbiota in dairy cattle using pyrosequencing. Anaerobe 89:1108–1119. doi:10.2527/jas.2010-3378
24:12–19. doi:10.1016/j.anaerobe.2013.08.003 Perez,  R.  H., T.  Zendo, and K.  Sonomoto. 2014. Novel
Martinez,  F.  A., E.  M.  Balciunas, A.  Converti, P.  D.  Cotter, and bacteriocins from lactic acid bacteria (LAB): various
R.  P.  de  Souza  Oliveira. 2013. Bacteriocin production by structures and applications. Microb. Cell Fact. 13(Suppl 1):S3.
Bifidobacterium spp. A  review. Biotechnol. Adv. 31:482–488. doi:10.1186/1475-2859-13-S1-S3
doi:10.1016/j.biotechadv.2013.01.010 Petsch,  D., and F.  B.  Anspach. 2000. Endotoxin removal from
Matsuura,  M. 2013. Structural modifications of bacterial protein solutions. J. Biotechnol. 76:97–119. doi:10.1016/
lipopolysaccharide that facilitate gram-negative bacteria s0168-1656(99)00185-6
evasion of host innate immunity. Front. Immunol. 4:109. Pitta, D. W., E. Pinchak, S. E. Dowd, J. Osterstock, V. Gontcharova,
doi:10.3389/fimmu.2013.00109 E.  Youn, K.  Dorton, I.  Yoon, B.  R.  Min, J.  D.  Fulford, et  al.
Monteiro and Faciola  |  9

2010. Rumen bacterial diversity dynamics associated with Rossi,  O., M.  T.  Khan, M.  Schwarzer, T.  Hudcovic, D.  Srutkova,
changing from bermudagrass hay to grazed winter wheat S. H. Duncan, E. H. Stolte, H. Kozakova, H. J. Flint, J. N. Samsom,
diets. Microb. Ecol. 59:511–522. doi:10.1007/s00248-009-9609-6 et  al. 2015. Faecalibacterium prausnitzii strain HTF-F and its
Plaizier,  J.  C., M.  Danesh  Mesgaran, H.  Derakhshani, H.  Golder, extracellular polymeric matrix attenuate clinical parameters
E. Khafipour, J. L. Kleen, I. Lean, J. Loor, G. Penner, and Q. Zebeli. in DSS-induced colitis. PLoS One. 10:e0123013. doi:10.1371/
2018. Review: Enhancing gastrointestinal health in dairy cows. journal.pone.0123013
Animal 12(s2):s399–s418. doi:10.1017/S1751731118001921 Russell,  J.  B., and H.  C.  Mantovani. 2002. The bacteriocins of
Plaizier, J. C., E. Khafipour, S. Li, G. N. Gozho, and D. O. Krause. ruminal bacteria and their potential as an alternative to
2012. Subacute ruminal acidosis (SARA), endotoxins and antibiotics. J. Mol. Microbiol. Biotechnol. 4:347–355. Available
health consequences. Anim. Feed Sci. Technol. 172:9–21. from https://pubmed.ncbi.nlm.nih.gov/12125815/
doi:10.1016/j.anifeedsci.2011.12.004 Russell,  J.  B., and J.  L.  Rychlik. 2001. Factors that alter rumen
Plaizier, J. C., D. O. Krause, G. N. Gozho, and B. W. McBride. 2008. microbial ecology. Science 292:1119–1122. doi:10.1126/
Subacute ruminal acidosis in dairy cows: the physiological science.1058830
causes, incidence and consequences. Vet. J. 176:21–31. Russell,  J.  B., and H.  J.  Strobel. 1989. Effect of ionophores on
doi:10.1016/j.tvjl.2007.12.016 ruminal fermentation. Appl. Environ. Microbiol. 55:1–6.
Plaizier, J. C., S. Li, A. M. Danscher, H. Derakshani, P. H. Andersen, doi:10.1128/AEM.55.1.1-6.1989
and E.  Khafipour. 2017a. Changes in microbiota in rumen Sassone-Corsi,  M., S.  P.  Nuccio, H.  Liu, D.  Hernandez, C.  T.  Vu,
digesta and feces due to a grain-based subacute ruminal A.  A.  Takahashi, R.  A.  Edwards, and M.  Raffatellu. 2016.
acidosis (SARA) challenge. Microb. Ecol. 74:485–495. Microcins mediate competition among Enterobacteriaceae
doi:10.1007/s00248-017-0940-z in the inflamed gut. Nature 540:280–283. doi:10.1038/
Plaizier, J. C., S. Li, M. Le Sciellour, B. L. Schurmann, P. Górka, and nature20557
G. B. Penner. 2014. Effects of duration of moderate increases Silva, C. C. G., S. P. M. Silva, and S. C. Ribeiro. 2018. Application of
in grain feeding on endotoxins in the digestive tract and bacteriocins and protective cultures in dairy food preservation.
acute phase proteins in peripheral blood of yearling calves. J. Front. Microbiol. 9:594. doi:10.3389/fmicb.2018.00594
Dairy Sci. 97:7076–7084. doi:10.3168/jds.2014-8162 Steele,  M.  A., G.  B.  Penner, F.  Chaucheyras-Durand, and
Plaizier, J. C., S. Li, H. M. Tun, and E. Khafipour. 2017b. Nutritional L. L. Guan. 2016. Development and physiology of the rumen
models of experimentally-induced subacute ruminal and the lower gut: targets for improving gut health. J. Dairy
acidosis (SARA) differ in their impact on rumen and hindgut Sci. 99:4955–4966. doi:10.3168/jds.2015-10351
bacterial communities in dairy cows. Front. Microbiol. 7:2128. Tomlinson, J. E., and A. T. Blikslager. 2004. Interactions between
doi:10.3389/fmicb.2016.02128 lipopolysaccharide and the intestinal epithelium. J. Am. Vet.
Poltorak,  A., X.  He, I.  Smirnova, M.  Y.  Liu, C.  Van  Huffel, X.  Du, Med. Assoc. 224:1446–1452. doi:10.2460/javma.2004.224.1446
D.  Birdwell, E.  Alejos, M.  Silva, C.  Galanos, et  al. 1998. Van  Soest,  P.  J. 1994. Nutritional ecology of the ruminant. 2nd ed.
Defective LPS signaling in C3H/HeJ and C57BL/10ScCr mice: Ithaca (NY): Cornell University Press.
mutations in Tlr4 gene. Science 282:2085–2088. doi:10.1126/ Wang, X., X. Li, C. Zhao, P. Hu, H. Chen, Z. Liu, G. Liu, and Z. Wang.
science.282.5396.2085 2012. Correlation between composition of the bacterial
Qureshi,  N., K.  Takayama, and R.  Kurtz. 1991. Diphosphoryl community and concentration of volatile fatty acids in the
lipid A  obtained from the nontoxic lipopolysaccharide rumen during the transition period and ketosis in dairy cows.
of Rhodopseudomonas sphaeroides is an endotoxin Appl. Environ. Microbiol. 78:2386–2392. doi:10.1128/AEM.07545-11
antagonist in mice. Infect. Immun. 59:441–444. doi:10.1128/ Wells,  J.  E., D.  O.  Krause, T.  R.  Callaway, and J.  B.  Russell. 1997.
IAI.59.1.441-444.1991 A bacteriocin-mediated antagonism by ruminal lactobacilli
Raetz, C. R., R. J. Ulevitch, S. D. Wright, C. H. Sibley, A. Ding, and against Streptococcus bovis. FEMS Microbiol. Ecol. 22:237–243.
C. F. Nathan. 1991. Gram-negative endotoxin: an extraordinary doi:10.1111/j.1574-6941.1997.tb00376.x
lipid with profound effects on eukaryotic signal transduction. Xue, M., H. Sun, X. Wu, L. L. Guan, and J. Liu. 2018. Assessment
FASEB J. 5:2652–2660. doi:10.1096/fasebj.5.12.1916089 of rumen microbiota from a large dairy cattle cohort
Raetz,  C.  R.  H., and C.  Whitfield. 2002. Lipopolysaccharide reveals the pan and core bacteriomes contributing to varied
endotoxins. Annu. Rev. Biochem. 71:635–700. doi:10.1146/ phenotypes. Appl. Environ. Microbiol. 84:e00970-18. doi:10.1128/
annurev.biochem.71.110601.135414 AEM.00970-18
Ransom-Jones, E., D. L. Jones, A. J. McCarthy, and J. E. McDonald. Yaron,  S., G.  L.  Kolling, L.  Simon, and K.  R.  Matthews. 2000.
2012. The Fibrobacteres: an important phylum of cellulose- Vesicle-mediated transfer of virulence genes from Escherichia
degrading bacteria. Microb. Ecol. 63:267–281. doi:10.1007/ coli O157:H7 to other enteric bacteria. Appl. Environ. Microbiol.
s00248-011-9998-1 66:4414–4420. doi:10.1128/aem.66.10.4414-4420.2000
Rebeil,  R., R.  K.  Ernst, B.  B.  Gowen, S.  I.  Miller, and Zhao, C., G. Liu, X. Li, Y. Guan, Y. Wang, X. Yuan, G. Sun, Z. Wang,
B.  J.  Hinnebusch. 2004. Variation in lipid A  structure in and X.  Li. 2018. Inflammatory mechanism of rumenitis in
the pathogenic Yersiniae. Mol. Microbiol. 52:1363–1373. dairy cows with subacute ruminal acidosis. BMC Vet. Res.
doi:10.1111/j.1365-2958.2004.04059.x 14:135. doi:10.1186/s12917-018-1463-7

You might also like