Topic Highlights Open Access
Copyright ©2007 Baishideng Publishing Group Co., Limited. All rights reserved.
World J Gastroenterol. Jun 14, 2007; 13(22): 3033-3042
Published online Jun 14, 2007. doi: 10.3748/wjg.v13.i22.3033
Biomarkers for radiation-induced small bowel epithelial damage: An emerging role for plasma Citrulline
Ludy Lutgens, Philippe Lambin, Department of Radiation Oncology (Maastro), GROW Research Institute, University of Maastricht, Maastricht 6200 MD, The Netherlands
Author contributions: All authors contributed equally to the work.
Correspondence to: Dr. Ludy Lutgens, MD, PhD, Department of Radiation Oncology (Maastro), GROW Research Institute, University of Maastricht, Tanslaan 12, 6202 AZ Maastricht, The Netherlands. ludy.lutgens@maastro.nl
Telephone: +31-88-4455600 Fax: +31-88-4455773
Received: December 15, 2006
Revised: February 3, 2007
Accepted: March 15, 2007
Published online: June 14, 2007

Abstract

Reduction of cancer treatment-induced mucosal injury has been recognized as an important target for improving the therapeutic ratio as well as reducing the economic burden associated with these treatment related sequellae. Clinical studies addressing this issue are hampered by the fact that specific objective parameters, which enable monitoring of damage in routine clinical practice, are lacking. This review summarizes pros and cons of currently available endpoints for intestinal injury. The metabolic background and characteristics of plasma citrulline, a recently investigated biomarker specifically for small intestinal injury, are discussed in more detail.

Key Words: Biomarker, Citrulline, Small bowel, Radiation injury



INTRODUCTION

An increase in the use of multiple treatment modalities is characteristic for current developments in curative cancer treatment. Whereas this strategy has yielded superior treatment results in a variety of solid tumors, treatment related acute toxicity has increased as well[1-6]. Severe radiation induced intestinal injury occurring during a treatment course has a detrimental effect on treatment outcome in cancer patients due to necessary reductions in treatment intensity and/or treatment interruption. In addition, this acute type of epithelial gut damage has also been suggested as one of several mechanisms contributing to late treatment related sequellae[7]. Cancer treatments related epithelial gastrointestinal toxicity has also been recognized as a significant economic burden[8]. Hence, prevention and/or reduction of epithelial gut damage is expected to have a significant clinical and socio-economic impact. However, clinical studies addressing treatment induced gut damage are hampered by the fact that objective parameters, which enable monitoring of damage in routine clinical practice, are lacking. In case of radiation treatment for pelvic and/or abdominal cancers the small bowel is an important dose-limiting organ with regard to both early and late treatment related morbidity. The clonogenic crypt cell is a central target of intestinal epithelial radiation damage[9-12]. Radiation will result in an impairment or loss of cell production and eventually in the loss of functional cells, becoming manifest within days or weeks following single dose or fractionated radiation[9]. A wide diversity of functional disorders has been observed following ionizing radiation such as changes in trans-epithelial transport processes[13,14], gut barrier function[15], motility dysfunction[16,17], or the absorption of various nutrients such as carbohydrates, amino acids, proteins, vitamins and bile acid[18-26]. Some of these functional changes have been correlated with the epithelial cell mass available for absorption[23-26] suggesting a cellular basis in at least part of radiation induced functional disorders.

BIOMARKERS FOR EPITHELIAL INTESTINAL DAMAGE
Clinical symptoms

Clinical symptoms are most commonly used as a surrogate endpoint during and following treatment. Clinical symptoms of acute radiation enteritis include anorexia, nausea, vomiting, abdominal cramps and diarrhea. These symptoms may occur immediately following the start of treatment, although more usually, radiation sequellae become manifest during the 2nd or 3rd wk of fractionated treatment and lasting 2-6 wk following treatment. Whereas very early symptoms are attributable to altered intestinal motor activity, mucosal injury is the prominent feature underlying symptoms later on during the course of treatment, although altered intestinal motor activity is another contributing factor throughout the treatment course and following treatment[27-29]. Beside the fact that toxicity-grading systems are not used uniformly by investigators[30], they are being adjusted on a regular basis. More importantly however, clinical symptoms correlate poorly with objective parameters of gut damage such as altered morphology[28,31], sugar permeability tests[32-34] or treatment related parameters[30], illustrating the complexity of the pathophysiology of clinical symptoms related to cytotoxic treatment induced small bowel damage[35-37].

Because of the limitations related to the assessment of morphological endpoints in patients, investigators have used several surrogate endpoints for measuring small bowel dysfunction.

Mucosal transport and barrier function

Mucosal transport and barrier function is another frequently used item for measuring small bowel dysfunction. Surrogate endpoints are the assessment of gut barrier function through measuring absorption of test markers[38,39] or tests for nutrient malabsorption[20,40], bile acid or vitamin B12 absorption[19,22,41-43]. These function tests are qualitative tests mainly suited for diagnostic purposes. The endpoints used do not address damage to target cells. Consequently, they lack a dose response relationship. Although not as troublesome as taking mucosal biopsies, these tests are impractical for monitoring purposes during and following radiation treatment. Enterocyte transport has been used as a surrogate endpoint for epithelial cell mass. Overgaard et al[23] demonstrated a dose response relationship for jejunal glucose absorption in mice following single dose upper abdominal irradiation. A linear correlation was observed between jejunal glucose absorption and the absorptive surface. Kirichenko et al[26] used a nuclear scintigraphic technique to quantify active enterocyte transport in mice. At 3.5 d after single dose whole body irradiation (WBI) absorption of the isotope correlated significantly with a surrogate endpoint for the jejunal absorptive surface, i.e. the number of cells per villus. A strong correlation was observed between absorption and radiation dose at this time point. For the dose points used in these experiments, i.e. 4, 6, 8 and 12.5 Gy, no correlation was seen between jejunal crypt regeneration, radiation dose and absorption. The results of both experiments[23,26] indicate a cellular basis for the absorptive function and a correlation with the absorptive area. Both function tests were investigated for their applicability as a clinical assay for radiation-induced epithelial cell loss in the gut and indirectly for quantification of radiation damage to the target cell for epithelial small bowel damage. To date none of these assays have been introduced in clinical practice for routine use during and following fractionated treatment, mainly for practical reasons.

Diamine oxidase

Diamine oxidase (DAO), a cytoplasmic enzyme found in almost all organs is present in a particularly high concentration in the epithelial cells of the small intestine[44-46]. Following injury to intestinal epithelial cells DAO is released into the intestinal lumen and intercellular space where it is taken up by lymphatics and blood vessels[45]. Circulating DAO is rapidly cleared by the liver[47]. The plasma DAO activity has been suggested as a candidate marker for measuring ischemic small bowel injury[48-50]. Ely et al[51] demonstrated a radiation dose-dependent decline of ileal tissue and plasma DAO activity in rats. Nadir values were observed at 3 d after radiation. At this time point a linear dose-response relationship was demonstrated for plasma and tissue DAO activity at a dose range of 0-6 Gy and 2-8 Gy, respectively. DeBell et al[52] investigated the time course of tissue and plasma DAO activity changes following irradiation. They found that the decline of plasma DAO activity preceded the decline of jejunal tissue DAO activity. In addition, the calculated RBE values for both parameters were not the same. These data do not support a direct correlation between the changes of plasma DAO activity and intestinal tissue DAO activity as was in fact also the case for the observation made by Bounous et al[49] These authors observed a 7.5 and 1.4 -fold increase in serum DAO activity 24 and 30 h following the onset of symptoms in a patient with a lethal acute intestinal ischemia.

Fatty acid-binding proteins

Fatty acid-binding proteins (FABP) are small (15 kDa) cytoplasmic proteins. Intestinal-type FABP (I-FABP) and liver-type FABP (L-FABP) are produced in small intestinal enterocytes, mainly in the villi, not in the crypt[53,54]. I-FABP has been demonstrated to be a sensitive biomarker for intestinal disease associated with tissue necrosis. Upon small bowel enterocyte necrosis I-FABP and L-FABP are readily shed into the circulation[53]. In ischemic bowel disease a rapid increase in plasma and urinary I-FABP concentration is observed[55,56]. In contrast, I-FABP and L-FABP were not elevated in patients with intestinal disease not associated with a significant degree of tissue necrosis[57]. In transplant recipients histologic graft rejection was not preceded by increased levels of serum I-FABP[58]. Taken together, I-FABP and L-FABP seem to be sensitive biomarkers for ischemic bowel disease. However, its use for intestinal damage initially targeting clonogenic crypt cells, as in radiation induced intestinal damage[9-12] and transplant rejection[59], has been disappointing so far.

Calprotectin

Calprotectin is a protein abundant in neutrophils. The fecal concentration of calprotectin has been identified as a sensitive biomarker of intestinal inflammation[60]. In patients with Crohn’s disease the marker correlates with changes in intestinal permeability[61]. The test is highly sensitive. The marker was tested in a validated animal model for late intestinal radiation injury[62,63]. Fecal excretion of transferrin, the rodent analogue of calprotectin, the first 2 wk after treatment correlated with validated endpoints for acute and late intestinal radiation injury. Interestingly, the high sensitivity of the test allows treatment of a limited volume of small bowel. However, in contrast to these experimental conditions the marker does not allow discrimination of anatomical sites of intestinal injury due to a low specificity[64].

CITRULLINE: A BIOMARKER FOR VIABLE SMALL BOWEL ENTEROCYTES

While radiation-induced tissue damage is unlikely to be expressed or quantified by a single functional or morphological parameter[36], an assay measuring damage to relevant target cells involved in the initiation of tissue damage is of great importance for both experimental and clinical research. Ideally, such an assay must be tissue-specific, display a dose-response relationship and in case of the small intestinal epithelium also a volume-response relationship. In addition, the assay must be easily accessible in clinical practice and independent of experimental conditions such as concurrent medical conditions, medication and nutritional status. Citrulline is a candidate biomarker fulfilling most of these criteria. The assay assesses radiation-induced epithelial cell loss, an important initiating factor in the pathogenesis of acute and chronic intestinal radiation injury and one of several pathophysiological mechanisms underlying clinical symptoms. Citrulline is a nitrogen end product of small bowel enterocyte metabolism. Plasma citrulline has been identified as a biomarker for functional small bowel enterocyte mass under various clinical and experimental conditions. In addition to surgery[65-68], celiac and non-celiac diseases[69], viral enteritis[70] and acute cellular rejection following small bowel transplantation[70-75], cytotoxic treatments was identified as another event associated with decreased plasma citrulline level due to epithelial cell loss[30,76-79]. As a whole, plasma citrulline seems to be a quantitative parameter independent of the underlying cause for epithelial cell loss.

Small intestinal intermediary metabolism

The small bowel epithelium plays an important role in the intermediary metabolism of amino acids, particularly glutamine, citrulline and arginine[80,81] thereby conditioning the availability of dietary amino acids to extra-intestinal organs[82]. Intestinal dysfunction resulting from intestinal diseases or injuries affect intermediary and inter-organ metabolism[83-85]. Hence, any factor affecting the intestinal mucosal cell mass will have an impact on protein and amino acid metabolism[83,86-90]. Since the pioneering work of Windmueller and Spaeth during the 1970’s many research groups have demonstrated that amino acids are the major fuel for the small bowel epithelium, both under conditions of fasting and feeding[91-97]. Windmueller and Spaeth identified glutamine as the quantitatively most important arterial energy source[91,98-100] for the rat jejunum in fasted animals. Measurements in different species consistently demonstrated a concentration dependent high rate of intestinal glutamine extraction from the blood. Thus, 25%-33% of the total plasma glutamine is extracted by the small bowel in each single pass[98]. Glutamine is the most abundant amino acid in plasma and plays a key role in whole body protein and amino acid metabolism[98,101]. Organs may be classified as glutamine producers and as glutamine consumers[102]. Skeletal muscle is by far the most important producer and the small bowel the most important consumer. The gut epithelium has been identified as the predominant site of glutamine uptake and metabolism[98]. Of all epithelial cells, enterocytes are the cells mostly responsible for glutamine utilization[103,104]. The first step in enteral glutamine catabolism is the conversion to glutamate and ammonia by the mitochondrial enzyme glutaminase in a non-reversible reaction[105]. The intestinal uptake of glutamine from the blood varies with the availability of substrate in the lumen. However, the intestinal metabolism of plasma glutamine is sustained during competitive luminal substrate provision, even under conditions of luminal overloading with glutamate[91,98]. The gut epithelial cell has access to glutamine from the arterial blood supply and the gut lumen[98]. The metabolic fate of glutamine from both routes is nearly identical indicating a common metabolic pool[91]. Major glutamine carbon products are CO2 (55%-65%), lactate (8%-16%), citrate (2%-7%), citrulline (4%-6%), proline (5%-6%), alanine (0.5%-4%) and ornithine (0.5%-2%). Major glutamine nitrogen products are ammonia (23%-36%), alanine (33%-36%), citrulline (10%-34%) and proline (7%-10%).

An endproduct of glutamine metabolism

Citrulline was identified as an endproduct of nitrogen glutamine metabolism in the rat intestine accounting for 27.6% of metabolised glutamine[96,98,99]. Citrulline is an intermediate in the urea cycle[106,107], which is comprised of 5 enzymes, 2 being mitochondrial [(CPSI) and (OCT)] and 3 being cytosolic enzymes (arginino succinate synthethase (ASS), arginino succinate lyase (ASL) and arginase). Windmueller and Spaeth[80] did not detect any urea-cycle intermediate following luminal administration of citrulline to the intestinal mucosal cells and concluded that intestinal mucosal cells contain an incomplete urea cycle. However, others suggested a complete urea cycle in rodent enteroctyes[108,109]. Wu finally demonstrated urea synthesis in porcine enterocytes from ammonia, glutamine and arginine in a dose-dependent manner, thus providing the evidence that, in addition to periportal hepatocytes[106], small intestinal enterocytes[104,110] contain a complete urea cycle. Whereas an activity was observed of all urea cycle enzymes, the activity of OCT was by far the highest of all (i.e. a factor 10-20)[110]. In contrast to hepatocytes in which CPSI and ASS are considered the regulatory enzymes due to an exceedingly high arginase activity, in enterocytes the arginase activity seems to be the limiting factor for urea synthesis[110]. Given the high rate of glutamine/glutamate metabolism[91,93,96,111] and the relative abundant OCT activity[110] in small intestinal enterocytes, the majority of citrulline produced from glutamine/glutamate[80,104] will not be further metabolised in the urea cycle but instead released in the portal circulation. Thus only 5% of the glutamine-derived ammonia was converted to urea indicating the low capacity of urea synthesis from glutamine (or ammonia) in enterocytes[110]. Hence, although the small intestinal mucosa contains a metabolically significant urea cycle, the liver is without doubt the major organ for urea synthesis in mammals[106,112]. Furthermore, citrulline can be effectively regarded as an endproduct of glutamine/glutamate metabolism of intestinal enterocytes as suggested by Windmueller and Spaeth[80,98] and confirmed in many studies since then[66,69,83,86-88,90,104,111,113-115].

Pathways for citrulline synthesis

The synthesis of citrulline from glutamine involves 5 mitochondrial enzymes; phosphate-dependent glutaminase (PDG), pyrroline-5-carboxylate synthase (P5CS), ornithine aminotransferase (OAT), OCT and CPSI with P5CS being the key regulatory enzyme[104,116-118]. P5CS is unique to small intestinal enterocytes[116,119-121]. PDG converts glutamine to glutamate and ammonia. Glutamate is then converted to pyrroline-5 carboxylate by P5CS. Pyrroline-5-carboxylate is then converted to ornithine by OAT. Glutamine derived ammonia plus HCO3- are converted to carbamoyl phosphate by CPSI. Carbamoyl phosphate and ornithine are finally converted to citrulline by OCT. Pyrroline-5-carboxylate is a common precursor of both ornithine and proline. For a long time, glutamine and glutamate have been considered the only precursor for pyrroline-5-carboxylate. Wu et al[122] have demonstrated proline oxidase (PROox) activity in porcine enterocytes with the synthesis of citrulline and arginine from proline being another important pathway for citrulline synthesis. This pathway involves 4 mitochondrial enzymes, being PROox[123], OAT, OCT and PCSI. Proline is converted to pyrroline-5-carboxylate by PROox[124]. The subsequent metabolic steps are the same as for citrulline synthesis from glutamine, involving OAT, CPSI and OCT. As a consequence, glutamine-derived nitrogen intermediates such as glutamate and ammonia are necessary for the synthesis of citrulline from proline[122]. Based on the relative enzyme activities[104,125] PROox and CPSI are suggested key regulatory enzymes in citrulline synthesis from proline[122]. Small intestinal PROox activity is relatively high, i.e. 10- and 6-fold greater than the activity in the liver and the kidney of piglets, respectively[123]. Furthermore, the total cell mass of small intestine is relatively large compared to the liver and kidneys, respectively, i.e. 162% (liver) and 970% (kidneys) in 6 wk old pigs[126]. Hence, the small intestine may be a major site of proline degradation and subsequent synthesis of citrulline from proline[122]. In contrast to glutamine, the luminal proline derived from the diet is the most important source of proline for citrulline synthesis[98,122].

Metabolic fate of citrulline released into the portal vein

Under physiologic conditions there is no appreciable uptake of citrulline by the liver[80]. Labelled citrulline was supplied to the liver by a continuous portal infusion at a concentration 1.5 times the usual portal blood concentration. Less than 10% of the labelled citrulline had disappeared from the perfusate after about 40 passes clearly indicating that very little citrulline in the portal blood released by the intestine is metabolised by the liver[80]. Thus citrulline produced and released by the small intestine simply passes through the liver and reaches the systemic circulation. Subsequently the kidney is the major consumer of circulating citrulline extracting about 35% of arterial citrulline in each pass[80,86]. The relevance of this pathway is demonstrated quantitatively by the increase of the plasma citrulline level observed in patients with renal failure[127].

Source of circulating citrulline

It is now generally accepted that the small intestinal absorptive epithelial cell is the major source of circulating citrulline[104,122,128]. Windmueller and Spaeth investigated the existence of alternative sources of circulating citrulline[80]. Within 5 min after exclusion of either the intestine alone or all portal drained viscera from the circulation, plasma citrulline concentration fell by only 27% and 20%, respectively. Hence, more than 70% of the plasma citrulline concentration is sustained. Based on the high rate of citrulline uptake by the kidney accounting for 83% of the citrulline released by the small bowel, it can be estimated that clearance of citrulline from the plasma should be complete after 4.3 min in the rat in the absence of any input[80]. These findings indicate the existence of extra-splanchnic production sites and/or storage sites of citrulline. The cerebrospinal fluid[129] and skeletal muscle[130] are known sites with citrulline concentrations exceeding that of plasma but could not be identified as citrulline releasing sites[80]. Measurement of arteriovenous concentration differences across the hindquarter after complete removal of the portal drained viscera revealed a small net release of citrulline accounting for only 24% of citrulline uptake by the kidney under physiological conditions. Hence, whereas skeletal muscle may be considered a storage site for citrulline, it is not a substantial source for circulating citrulline under normal physiological circumstances[80]. The liver does not release citrulline unless provided with un-physiologically high doses of ammonium in conjunction with high concentrations of ornithine or proline in the perfusate, indicating that all the citrulline formed from ornithine or proline is converted to arginine[80]. This is probably due to the efficient metabolic channelling of citrulline to ASS and the high activity of typeIarginase in hepatocytes leading to a subsequent rapid hydrolysis of arginine into urea and ornithine[131]. Hence, despite the results observed with the organ exclusion experiments performed by Windmueller and Spaeth, no other site but the intestine has been identified so far that releases significant amounts of citrulline under physiological conditions. The role of the small intestine as the major source for circulating citrulline is demonstrated by experiments in which the plasma citrulline concen-tration is reduced by means of small bowel targeted interventions such as specific inhibitors of OAT[116] or OCT[132], yielding a similar decrease of plasma citrulline concentration as observed after small bowel resecti-on[83,86,113,114]. Furthermore, strong lines of evidence have been obtained since then through clinical observations which are in agreement with this concept[66,69,71,73,87,90,133,134]. Experimental and clinical data suggest a non-homogenous distribution of citrulline production. The distribution of P5CS activity in rats was 26%, 31%, 33% and 10% in the duodenum, upper jejunum, lower jejunum and ileum, respectively[120]. The release of citrulline measured as venous minus arterial concentration in patients admitted for elective gastrointestinal surgery was 30.4 ± 4.0 μmol/L and 8.4 ± 1.7 μmol/L for the jejunum and ileum, respectively[111].

Determinants of intestinal citrulline synthesis and plasma citrulline level

The activity of intestinal citrulline-synthesizing enzymes changes as a function of the feeding regimen, i.e. during the suckling and (post) weaning period. Weaning-induced changes in plasma cortisol levels are suggested to play a role in the difference observed between suckling and weaning animals, rather than developmental changes related to age[135-138]. Except for the interaction of metabolites with specific enzymes[137-141], substrate availability is another determinant of the citrulline production by enterocytes[104,122,140,142]. Several inborn errors[143-148] may give rise to specific changes in citrulline concentration. Enhanced NO synthase activity in patients with SLE has been associated with hypercitrullinemia[149]. Taken the central role of glutamine metabolism in the small intestinal citrulline synthesis[104,122], any metabolic condition substantially influencing intestinal glutamine metabolism is likely to have a major impact on citrulline synthesis as well. In this respect cumulative data indicate an important role for glucose metabolism[140,141,150]. A major determinant, however, under steady state conditions for the rate of citrulline released into the portal and subsequently the systemic circulation is the actual number of functional enterocytes[65-69,83,86,87,90,113,114,134,151]. This was further demonstrated by clinical data on small bowel trans-plantation[70-75,152]. In addition to a variation in citrulline synthesis, alterations in citrulline utilization will have an influence on the plasma citrulline concentration[80,119,127].

Plasma citrulline a surrogate endpoint for enterocyte mass

Effectively, citrulline can be regarded as an endproduct of glutamine and/or proline metabolism of intestinal enterocytes[80,98,104,110,122]. Several enzymes are involved in the synthesis of citrulline from glutamine and/or proline[104,110,122]. Whereas OAT[116,121] can be categorized as a ubiquitous enzyme, OCT, CPSI, PROox are highly polarized and P5CS is extremely polarized[119]. Thus high activities of OCT[153], CPSI[153] and PROox[121,123] are found in the small intestine and liver. P5CS activity is almost exclusively found in small intestinal enterocytes[116,120,121]. This unique enzymatic profile, the unique role of the small intestine in whole body glutamine metabolism[96,154] and the relatively high small bowel enterocyte cell mass[126] make the small bowel epithelium the most important source of circulating citrulline[128,131]. Taken together these data indicate a high specificity for circulating citrulline, i.e. small intestinal enterocytes. Thus under steady state conditions, citrulline can be considered a marker for the functional epithelial cell mass of the small bowel, a concept amply demonstrated in experimental and clinical studies[65-75,80,83,86-88,90,104,111,113-116,132-134,152]. Of notice, the lower plasma citrulline level observed in short bowel patients was sustained up to one year after treatment[66] emphasizing its strict dependence on the epithelial cell mass. As such, plasma citrulline concentration has been proposed as a biological marker for viable small bowel epithelium[65-67,69,71,73-75,152]. Crenn et al[69] have recently correlated plasma citrulline concentration with histologically graded villous atrophy in 42 patients with celiac and 10 patients with non-celiac villous atrophy disease. These authors identified a threshold value of 10 μmol/L (25% of the mean normal baseline value) to be predictive for severe and extensive villous atrophy and 20 μmol/L to be predictive for severe villous atrophy, whatever the extent. The plasma level of citrulline was thus indicative for the degree of villous atrophy. This finding is indicative for a possible volume effect. Crenn et al[69] demonstrated the use of plasma citrulline for monitoring treatment response in patients with celiac disease, indicating the simplicity of the marker in clinical practice. The accuracy of the assay has been assessed for various clinical settings. Crenn et al[66] measured plasma citrulline level in 57 patients with nonmalignant short bowel syndrome defined by a postduodenal remnant small bowel length of less than 200 cm. Minimal follow up was 2 years after definite digestive circuit modification. The threshold of plasma citrulline that best discriminated short bowel patients from controls was 30 μmol/L yielding a sensitivity, specificity, PPV and NPV of 77%, 75%, 76% and 77%, respectively. The best threshold of plasma citrulline for discrimination of transient from permanent intestinal failure was 20 μmol/L yielding a sensitivity, specificity, PPV and NPV of 92%, 90%, 95% and 85%, respectively. In a series of 52 patients with celiac and nonceliac villous atrophy Crenn et al[69] correlated plasma citrulline and mucosal atrophy assessed by endoscopic mucosal biopsies. The threshold of plasma citrulline for discrimination between nondestructive and destructive mucosal lesions (modified Marsh classification) was 20 μmol/L yielding a sensitivity, specificity, PPV and NPV of 95%, 90%, 88% and 96%, respectively. Gondolesi et al[75] measured plasma citrulline in 49 intestinal transplant recipients within 12 h before or after endoscopic biopsies taken according to a protocol (i.e. twice weekly for 6 wk, once weekly until 6 mo and monthly until 1 year postintestinal transplant). The sensitivity and specificity of the citrulline assay for diagnosing transplant rejection in adults was 80% and 58%, respectively.

PLASMA CITRULLINE: A SURROGATE ENDPOINT FOR RADIATION INDUCED EPITHELIAL CELL LOSS

Taken together, plasma citrulline is a candidate marker for measuring radiation-induced epithelial small bowel damage. The data indicate that this biomarker is tissue-specific, i.e. small intestinal epithelium. The biomarker corresponds with an important morpholocal endpoint, i.e. mucosal atrophy, and is easily accessible in clinical practice. Although experimental[120] and clinical data[111] suggest a non-homogenous distribution of citrulline production, a volume effect is suggested by the data provided by Crenn et al[66,69].

A decrease of intestinal absorptive function follo-wing irradiation has been correlated to the loss of functionally active enterocytes constituting the absorptive mucosal surface[23-26]. The correlation between radiation-induced epithelial cell loss and plasma citrulline level was demonstrated in mice by Lutgens et al[76] Following treatment with a single whole body irradiation (WBI) (dose range 0-14.9 Gy) blood and jejunal tissue were sampled for analysis. At 84 h and 4 d after WBI a dose response relationship was observed for plasma citrulline level. At this time point plasma citrulline correlated with mucosal surface, a surrogate endpoint for functional enterocyte mass. Plasma citrulline level decreased as a function of dose and time after WBI. Whereas the time effect was significant for all dose levels used, a significant dose-response relationship was observed only at d 4 after WBI. Remarkably, a rapid decline of plasma citrulline was observed at the first 2 d after WBI independent of the WBI doses used whereas recovery was more rapid for the lowest dose (i.e. 8 Gy) and incomplete during the observation period for the highest dose levels used (i.e. 11 and 12 Gy). This time and dose pattern is in agreement with the radiation effect on the hierarchically structured intestinal epithelium[155]. Interestingly, using the epithelial surface lining as a parameter did not yield significant changes except for the 4 d time point for the highest dose levels (i.e. 11 and 12 Gy) whereas for citrullinemia significant changes were observed for all dose levels used at the 4 d time point. Furthermore, plasma citrulline levels remained significantly decreased at the 11 d time point. For the dose range used in our experiments, mean values for mucosal surface lining ranged between 56% and 130% of control values, whereas for citrullinemia mean values ranged between 6% and 121% of control values. Thus citrullinemia seems to be more sensitive for detecting and monitoring small bowel radiation-induced epithelial cell loss than the representative morphologic endpoint used in these experiments. After WBI doses of 1-3 Gy no effect on citrullinemia could be demonstrated whereas this parameter was inversely proportional to WBI doses of 3-12 Gy. The threshold dose for the citrulline assay (about 3 Gy) is significantly lower as compared to the microcolony assay (about 8 Gy). Furthermore, in contrast to the microcolony assay the citrulline assay permits repeated measurements within the same animal. Therefore the citrulline assay and the microcolony assay are supplementary, both with regard to the dose range as with regard to their applicability.

The use of plasma citrulline as an assay for acute small bowel epithelial radiation injury was demonstrated by Lutgens et al[76] Amifostine was administered to mice as a radioprotective agent with a consistently found dose modification factor (DMF) of 1.6 using the microcolony assay as endpoint[156]. The DMF observed for citrulline (1.5) was in complete agreement with literature data. Vanclee et al[79] have used the citrulline assay to demonstrate a protective effect of keratinocyte growth factor on cytotoxic treatment induced intestinal injury.

The feasibility of plasma citrulline as a surrogate marker for radiation-induced small bowel injury was demonstrated by Lutgens et al[30] in a prospective clinical study in patients treated with fractionated radiotherapy for abdominal and/or pelvic cancer sites. A dose and volume effect was observed using dose volume histogram parameters and plasma citrulline levels as endpoints. Median nadir citrulline levels were observed during the 3rd wk of fractionated radiotherapy. This time course of plasma citrulline was further established in two clinical studies using archive material of patients treated with intensive myeloablative therapy[77,78]. Following conditioning treatment with high dose chemotherapy and fractionated WBI nadir plasma citrulline levels were observed around 7 d after hematopoietic stem cell transplant. Sensitivity and specificity of the citrulline assay were better compared to standard endpoints used for assessment of gut damage[77].

CONCLUSION

Radiation-induced small bowel damage is unlikely to be expressed or quantified by a single functional or morphological parameter. Several biomarkers are currently available differing with respect to kinetics, related target cells and pathophysiological processes involved and the convenience for clinical use. It is thus challenging to choose a (set of) biomarker(s) that is best suited to a specific experimental or clinical setting. Citrulline is a promising candidate biomarker. A dose-response relationship[76] and a correlation with epithelial cell mass[76,79] have been recently demonstrated in experimental studies. The time course of plasma citrulline following radiation[30,76,78] is in agreement with well known radiation effects on the hierarchically structured intestinal epithelium[155] and clinical observations of acute intestinal injury. The feasibility of the marker was demonstrated in a series of patients treated with fractionated radiotherapy for pelvic and/or abdominal cancers[30]. Unlike most other used endpoints, the citrulline assay can be applied to both experimental and clinical settings facilitating translational research. Also citrulline can be repeatedly measured enabling monitoring of treatment effects. Finally, the assay is simple to apply and relatively cheap. Like surgery[65-68], celiac and non-celiac disease[69] and acute cellular rejection following small bowel transplantation[70-73,75,152], ionizing irradiation has been demonstrated to be an additional event associated with reduced small bowel epithelial cell mass that can be monitored by plasma citrulline[30,76-79].

Footnotes

S- Editor Wang J L- Editor Alpini GD E- Editor Lu W

References
1.  Han SC, Kim DH, Higgins SA, Carcangiu ML, Kacinski BM. Chemoradiation as primary or adjuvant treatment for locally advanced carcinoma of the vulva. Int J Radiat Oncol Biol Phys. 2000;47:1235-1244.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 88]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
2.  Green JA, Kirwan JM, Tierney JF, Symonds P, Fresco L, Collingwood M, Williams CJ. Survival and recurrence after concomitant chemotherapy and radiotherapy for cancer of the uterine cervix: a systematic review and meta-analysis. Lancet. 2001;358:781-786.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 805]  [Cited by in F6Publishing: 839]  [Article Influence: 36.5]  [Reference Citation Analysis (0)]
3.  Påhlman L, Hohenberger W, Günther K, Fietkau R, Metzger U. Is radiochemotherapy necessary in the treatment of rectal cancer? Eur J Cancer. 1998;34:438-448.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 17]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
4.  Curran WJ. Evolving chemoradiation treatment strategies for locally advanced non-small-cell lung cancer. Oncology (Williston Park). 2003;17:7-14.  [PubMed]  [DOI]  [Cited in This Article: ]
5.  Stehman FB, Rose PG, Greer BE, Roy M, Plante M, Penalver M, Jhingran A, Eifel P, Montz F, Wharton JT. Innovations in the treatment of invasive cervical cancer. Cancer. 2003;98:2052-2063.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 22]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
6.  Thomas PR, Lindblad AS. Adjuvant postoperative radiotherapy and chemotherapy in rectal carcinoma: a review of the Gastrointestinal Tumor Study Group experience. Radiother Oncol. 1988;13:245-252.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 147]  [Cited by in F6Publishing: 141]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
7.  Richter KK, Langberg CW, Sung CC, Hauer-Jensen M. Increased transforming growth factor beta (TGF-beta) immunoreactivity is independently associated with chronic injury in both consequential and primary radiation enteropathy. Int J Radiat Oncol Biol Phys. 1997;39:187-195.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 55]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
8.  Elting LS, Cooksley C, Chambers M, Cantor SB, Manzullo E, Rubenstein EB. The burdens of cancer therapy. Clinical and economic outcomes of chemotherapy-induced mucositis. Cancer. 2003;98:1531-1539.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 417]  [Cited by in F6Publishing: 426]  [Article Influence: 20.3]  [Reference Citation Analysis (0)]
9.  Potten CS. Effects of radiation on murine gastrointestinal cell proliferation. Radiation and gut. Amsterdam: Elsevier Science B.V 1995; 61-84.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Potten CS, Booth C, Pritchard DM. The intestinal epithelial stem cell: the mucosal governor. Int J Exp Pathol. 1997;78:219-243.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 370]  [Cited by in F6Publishing: 345]  [Article Influence: 12.8]  [Reference Citation Analysis (0)]
11.  Osborne JW. Early and late radiation effects (external irradiation) on the gut. Radiation and gut. Amsterdam: Elsevier 1995; 145-209.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Paris F, Fuks Z, Kang A, Capodieci P, Juan G, Ehleiter D, Haimovitz-Friedman A, Cordon-Cardo C, Kolesnick R. Endothelial apoptosis as the primary lesion initiating intestinal radiation damage in mice. Science. 2001;293:293-297.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 966]  [Cited by in F6Publishing: 925]  [Article Influence: 40.2]  [Reference Citation Analysis (0)]
13.  Gunter-Smith PJ. Gamma radiation affects active electrolyte transport by rabbit ileum: basal Na and Cl transport. Am J Physiol. 1986;250:G540-G545.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Lebrun F, Benderitter M, Berroud A, Voisin P, Griffiths NM. Potential role of the membrane in the development of intestinal cellular damage after whole-body gamma irradiation of the rat. Can J Physiol Pharmacol. 2002;80:686-693.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 7]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
15.  Guzman-Stein G, Bonsack M, Liberty J, Delaney JP. Abdominal radiation causes bacterial translocation. J Surg Res. 1989;46:104-107.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 63]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
16.  Picard C, Wysocki J, Fioramonti J, Griffiths NM. Intestinal and colonic motor alterations associated with irradiation-induced diarrhoea in rats. Neurogastroenterol Motil. 2001;13:19-26.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 20]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
17.  Fraser R, Frisby C, Blackshaw LA, Schirmer M, Howarth G, Yeoh E. Small intestinal dysmotility following abdominal irradiation in the rat small intestine. Neurogastroenterol Motil. 1998;10:413-419.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 12]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
18.  Becciolini A, Balzi M, Potten CS. Radiation effects on proliferation and differentiation in the rat small intestine. Radiation and gut. 1 ed. Amsterdam: Elsevier Science B.V 1995; 85-143.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Yeoh E, Horowitz M, Russo A, Muecke T, Ahmad A, Robb T, Chatterton B. A retrospective study of the effects of pelvic irradiation for carcinoma of the cervix on gastrointestinal function. Int J Radiat Oncol Biol Phys. 1993;26:229-237.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 23]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
20.  Thomson AB, Cheeseman CI, Walker K. Effect of abdominal irradiation on the kinetic parameters of intestinal uptake of glucose, galactose, leucine, and gly-leucine in the rat. J Lab Clin Med. 1983;102:813-827.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Thomson AB, Cheeseman CI, Walker K. Effect of external abdominal irradiation on the dimensions and characteristics of the barriers to passive transport in the rat intestine. Lipids. 1984;19:405-418.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 8]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]
22.  Thomson AB, Cheeseman CI, Walker K. Intestinal uptake of bile acids: effect of external abdominal irradiation. Int J Radiat Oncol Biol Phys. 1984;10:671-685.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 14]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
23.  Overgaard J, Matsui M. Effect of radiation on glucose absorption in the mouse jejunum in vivo. Radiother Oncol. 1990;18:71-77.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 20]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
24.  Juby LD, Dixon MF, Axon AT. Abnormal intestinal permeability and jejunal morphometry. J Clin Pathol. 1987;40:714-718.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 29]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
25.  Gunter-Smith PJ. Gamma radiation affects active electrolyte transport by rabbit ileum. II. Correlation of alanine and theophylline response with morphology. Radiat Res. 1989;117:419-432.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 19]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
26.  Kirichenko AV, Mason KA, Straume M, Teates CD, Rich TA. Nuclear scintigraphic assessment of radiation-induced intestinal dysfunction. Radiat Res. 2000;153:164-172.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 8]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
27.  Erickson BA, Otterson MF, Moulder JE, Sarna SK. Altered motility causes the early gastrointestinal toxicity of irradiation. Int J Radiat Oncol Biol Phys. 1994;28:905-912.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 24]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
28.  Fraser R, Frisby C, Schirmer M, Blackshaw A, Langman J, Yeoh E, Rowland R, Horowitz M. Effects of fractionated abdominal irradiation on small intestinal motility--studies in a novel in vitro animal model. Acta Oncol. 1997;36:705-710.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 9]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
29.  Otterson MF, Sarna SK, Moulder JE. Effects of fractionated doses of ionizing radiation on small intestinal motor activity. Gastroenterology. 1988;95:1249-1257.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Lutgens LC, Deutz N, Granzier-Peeters M, Beets-Tan R, De Ruysscher D, Gueulette J, Cleutjens J, Berger M, Wouters B, von Meyenfeldt M. Plasma citrulline concentration: a surrogate end point for radiation-induced mucosal atrophy of the small bowel. A feasibility study in 23 patients. Int J Radiat Oncol Biol Phys. 2004;60:275-285.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 95]  [Cited by in F6Publishing: 100]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
31.  Keefe DM, Brealey J, Goland GJ, Cummins AG. Chemotherapy for cancer causes apoptosis that precedes hypoplasia in crypts of the small intestine in humans. Gut. 2000;47:632-637.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 269]  [Cited by in F6Publishing: 279]  [Article Influence: 11.6]  [Reference Citation Analysis (0)]
32.  Johansson JE, Ekman T. Gastro-intestinal toxicity related to bone marrow transplantation: disruption of the intestinal barrier precedes clinical findings. Bone Marrow Transplant. 1997;19:921-925.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 37]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
33.  Johansson JE, Ekman T. Gut mucosa barrier preservation by orally administered IgA-IgG to patients undergoing bone marrow transplantation: a randomised pilot study. Bone Marrow Transplant. 1999;24:35-39.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 10]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
34.  Blijlevens NM, van't Land B, Donnelly JP, M'Rabet L, de Pauw BE. Measuring mucosal damage induced by cytotoxic therapy. Support Care Cancer. 2004;12:227-233.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 39]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
35.  Thomson AB, Keelan M, Thiesen A, Clandinin MT, Ropeleski M, Wild GE. Small bowel review: diseases of the small intestine. Dig Dis Sci. 2001;46:2555-2566.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 33]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
36.  Griffiths NM. The example of gastrointestinal damage induced by ionising radiation: are there accessible markers? Cell Mol Biol (Noisy-le-grand). 2001;47:427-435.  [PubMed]  [DOI]  [Cited in This Article: ]
37.  MacNaughton WK. Review article: new insights into the pathogenesis of radiation-induced intestinal dysfunction. Aliment Pharmacol Ther. 2000;14:523-528.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 87]  [Cited by in F6Publishing: 95]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
38.  Travis S, Menzies I. Intestinal permeability: functional assessment and significance. Clin Sci (Lond). 1992;82:471-488.  [PubMed]  [DOI]  [Cited in This Article: ]
39.  Bjarnason I, MacPherson A, Hollander D. Intestinal permeability: an overview. Gastroenterology. 1995;108:1566-1581.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 627]  [Cited by in F6Publishing: 648]  [Article Influence: 22.3]  [Reference Citation Analysis (0)]
40.  Craig RM, Atkinson AJ. D-xylose testing: a review. Gastroenterology. 1988;95:223-231.  [PubMed]  [DOI]  [Cited in This Article: ]
41.  Yeoh EK, Horowitz M. Radiation enteritis. Surg Gynecol Obstet. 1987;165:373-379.  [PubMed]  [DOI]  [Cited in This Article: ]
42.  Yeoh E, Horowitz M, Russo A, Muecke T, Robb T, Chatterton B. The effects of abdominal irradiation for seminoma of the testis on gastrointestinal function. J Gastroenterol Hepatol. 1995;10:125-130.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 14]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
43.  Yeoh E, Horowitz M, Russo A, Muecke T, Robb T, Maddox A, Chatterton B. Effect of pelvic irradiation on gastrointestinal function: a prospective longitudinal study. Am J Med. 1993;95:397-406.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 116]  [Cited by in F6Publishing: 104]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
44.  Shaff RE, Beaven MA. Turnover and synthesis of diamine oxidase (DAO) in rat tissues. Studies with heparin and cycloheximide. Biochem Pharmacol. 1976;25:1057-1062.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 52]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
45.  Wollin A, Navert H, Bounous G. Effect of intestinal ischemia on diamine oxidase activity in rat intestinal tissue and blood. Gastroenterology. 1981;80:349-355.  [PubMed]  [DOI]  [Cited in This Article: ]
46.  Biegański T, Kusche J, Lorenz W, Hesterberg R, Stahlknecht CD, Feussner KD. Distribution and properties of human intestinal diamine oxidase and its relevance for the histamine catabolism. Biochim Biophys Acta. 1983;756:196-203.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 99]  [Cited by in F6Publishing: 103]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
47.  D'Agostino L, Ciacci C, Capuano G, Daniele B, D'Argenio G, Barone MV, Rodinò S, Budillon G, Mazzacca G. Metabolic fate of plasma diamine oxidase: evidence of isolated and perfused rat liver uptake. Digestion. 1986;34:243-250.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 20]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
48.  Bragg LE, Thompson JS, West WW. Intestinal diamine oxidase levels reflect ischemic injury. J Surg Res. 1991;50:228-233.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 34]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
49.  Bounous G, Echavé V, Vobecky SJ, Navert H, Wollin A. Acute necrosis of the intestinal mucosa with high serum levels of diamine oxidase. Dig Dis Sci. 1984;29:872-874.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 30]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
50.  Rose SG, Thompson JS, Spanta AD, Quigley EM. The effect of intestinal autotransplantation on serum diamine oxidase activity. J Surg Res. 1991;50:223-227.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 7]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]
51.  Ely MJ, Speicher JM, Catravas GN, Snyder SL. Radiation effects on diamine oxidase activities in intestine and plasma of the rat. Radiat Res. 1985;103:158-162.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 10]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
52.  DeBell RM, Ledney GD, Snyder SL. Quantification of gut injury with diamine oxidase activity: development of a fission neutron RBE and measurements with combined injury in mouse models. Radiat Res. 1987;112:508-516.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 7]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]
53.  Lieberman JM, Sacchettini J, Marks C, Marks WH. Human intestinal fatty acid binding protein: report of an assay with studies in normal volunteers and intestinal ischemia. Surgery. 1997;121:335-342.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 156]  [Cited by in F6Publishing: 167]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
54.  Ockner RK, Manning JA. Fatty acid-binding protein in small intestine. Identification, isolation, and evidence for its role in cellular fatty acid transport. J Clin Invest. 1974;54:326-338.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 344]  [Cited by in F6Publishing: 367]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
55.  Cronk DR, Houseworth TP, Cuadrado DG, Herbert GS, McNutt PM, Azarow KS. Intestinal fatty acid binding protein (I-FABP) for the detection of strangulated mechanical small bowel obstruction. Curr Surg. 2006;63:322-325.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 63]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
56.  Niewold TA, Meinen M, van der Meulen J. Plasma intestinal fatty acid binding protein (I-FABP) concentrations increase following intestinal ischemia in pigs. Res Vet Sci. 2004;77:89-91.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 50]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
57.  Pelsers MM, Namiot Z, Kisielewski W, Namiot A, Januszkiewicz M, Hermens WT, Glatz JF. Intestinal-type and liver-type fatty acid-binding protein in the intestine. Tissue distribution and clinical utility. Clin Biochem. 2003;36:529-535.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 223]  [Cited by in F6Publishing: 244]  [Article Influence: 12.2]  [Reference Citation Analysis (0)]
58.  Kaufman SS, Lyden ER, Marks WH, Lieberman J, Sudan DL, Fox IF, Shaw BW, Horslen SP, Langnas AN. Lack of utility of intestinal fatty acid binding protein levels in predicting intestinal allograft rejection. Transplantation. 2001;71:1058-1060.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 15]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
59.  Lee RG, Nakamura K, Tsamandas AC, Abu-Elmagd K, Furukawa H, Hutson WR, Reyes J, Tabasco-Minguillan JS, Todo S, Demetris AJ. Pathology of human intestinal transplantation. Gastroenterology. 1996;110:1820-1834.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 192]  [Cited by in F6Publishing: 191]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
60.  Konikoff MR, Denson LA. Role of fecal calprotectin as a biomarker of intestinal inflammation in inflammatory bowel disease. Inflamm Bowel Dis. 2006;12:524-534.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 290]  [Cited by in F6Publishing: 297]  [Article Influence: 16.5]  [Reference Citation Analysis (0)]
61.  Berstad A, Arslan G, Folvik G. Relationship between intestinal permeability and calprotectin concentration in gut lavage fluid. Scand J Gastroenterol. 2000;35:64-69.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 73]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
62.  Richter KK, Fagerhol MK, Carr JC, Winkler JM, Sung CC, Hauer-Jensen M. Association of granulocyte transmigration with structural and cellular parameters of injury in experimental radiation enteropathy. Radiat Oncol Investig. 1997;5:275-282.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
63.  Richter KK, Wang J, Fagerhol MK, Hauer-Jensen M. Radiation-induced granulocyte transmigration predicts development of delayed structural changes in rat intestine. Radiother Oncol. 2001;59:81-85.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 13]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
64.  Summerton CB, Longlands MG, Wiener K, Shreeve DR. Faecal calprotectin: a marker of inflammation throughout the intestinal tract. Eur J Gastroenterol Hepatol. 2002;14:841-845.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 123]  [Cited by in F6Publishing: 139]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
65.  Rhoads JM, Plunkett E, Galanko J, Lichtman S, Taylor L, Maynor A, Weiner T, Freeman K, Guarisco JL, Wu GY. Serum citrulline levels correlate with enteral tolerance and bowel length in infants with short bowel syndrome. J Pediatr. 2005;146:542-547.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 120]  [Cited by in F6Publishing: 121]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
66.  Crenn P, Coudray-Lucas C, Thuillier F, Cynober L, Messing B. Postabsorptive plasma citrulline concentration is a marker of absorptive enterocyte mass and intestinal failure in humans. Gastroenterology. 2000;119:1496-1505.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 365]  [Cited by in F6Publishing: 380]  [Article Influence: 15.8]  [Reference Citation Analysis (0)]
67.  Jianfeng G, Weiming Z, Ning L, Fangnan L, Li T, Nan L, Jieshou L. Serum citrulline is a simple quantitative marker for small intestinal enterocytes mass and absorption function in short bowel patients. J Surg Res. 2005;127:177-182.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 82]  [Cited by in F6Publishing: 70]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
68.  Chang RW, Javid PJ, Oh JT, Andreoli S, Kim HB, Fauza D, Jaksic T. Serial transverse enteroplasty enhances intestinal function in a model of short bowel syndrome. Ann Surg. 2006;243:223-228.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 76]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
69.  Crenn P, Vahedi K, Lavergne-Slove A, Cynober L, Matuchansky C, Messing B. Plasma citrulline: A marker of enterocyte mass in villous atrophy-associated small bowel disease. Gastroenterology. 2003;124:1210-1219.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 255]  [Cited by in F6Publishing: 256]  [Article Influence: 12.2]  [Reference Citation Analysis (0)]
70.  Gondolesi G, Fishbein T, Chehade M, Tschernia A, Magid M, Kaufman S, Raymond K, Sansaricq C, LeLeiko N. Serum citrulline is a potential marker for rejection of intestinal allografts. Transplant Proc. 2002;34:918-920.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 50]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
71.  Gondolesi GE, Kaufman SS, Sansaricq C, Magid MS, Raymond K, Iledan LP, Tao Y, Florman SS, LeLeiko NS, Fishbein TM. Defining normal plasma citrulline in intestinal transplant recipients. Am J Transplant. 2004;4:414-418.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 28]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
72.  Pappas PA, Saudubray JM, Tzakis AG, Rabier D, Carreno MR, Gomez-Marin O, Huijing F, Gelman B, Levi DM, Nery JR. Serum citrulline as a marker of acute cellular rejection for intestinal transplantation. Transplant Proc. 2002;34:915-917.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 41]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
73.  Pappas PA, G Tzakis A, Gaynor JJ, Carreno MR, Ruiz P, Huijing F, Kleiner G, Rabier D, Kato T, Levi DM. An analysis of the association between serum citrulline and acute rejection among 26 recipients of intestinal transplant. Am J Transplant. 2004;4:1124-1132.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 56]  [Cited by in F6Publishing: 56]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
74.  David AI, Gaynor JJ, Zis PP, Conanan L, Goldsmith L, Esquenazi V, Selvaggi G, Weppler D, Nishida S, Moon J. An association of lower serum citrulline levels within 30 days of acute rejection in patients following small intestine transplantation. Transplant Proc. 2006;38:1731-1732.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 17]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
75.  Gondolesi G, Ghirardo S, Raymond K, Hoppenhauer L, Surillo D, Rumbo C, Fishbein T, Sansaricq C, Sauter B. The value of plasma citrulline to predict mucosal injury in intestinal allografts. Am J Transplant. 2006;6:2786-2790.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 49]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
76.  Lutgens LC, Deutz NE, Gueulette J, Cleutjens JP, Berger MP, Wouters BG, von Meyenfeldt MF, Lambin P. Citrulline: a physiologic marker enabling quantitation and monitoring of epithelial radiation-induced small bowel damage. Int J Radiat Oncol Biol Phys. 2003;57:1067-1074.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 103]  [Cited by in F6Publishing: 91]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
77.  Lutgens LC, Blijlevens NM, Deutz NE, Donnelly JP, Lambin P, de Pauw BE. Monitoring myeloablative therapy-induced small bowel toxicity by serum citrulline concentration: a comparison with sugar permeability tests. Cancer. 2005;103:191-199.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 120]  [Cited by in F6Publishing: 122]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
78.  Blijlevens NM, Lutgens LC, Schattenberg AV, Donnelly JP. Citrulline: a potentially simple quantitative marker of intestinal epithelial damage following myeloablative therapy. Bone Marrow Transplant. 2004;34:193-196.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 90]  [Cited by in F6Publishing: 92]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
79.  Vanclée A, Lutgens LC, Oving EB, Deutz NE, Gijbels MJ, Schouten HC, Bos GM. Keratinocyte growth factor ameliorates acute graft-versus-host disease in a novel nonmyeloablative haploidentical transplantation model. Bone Marrow Transplant. 2005;36:907-915.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 30]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
80.  Windmueller HG, Spaeth AE. Source and fate of circulating citrulline. Am J Physiol. 1981;241:E473-E480.  [PubMed]  [DOI]  [Cited in This Article: ]
81.  Windmueller HG, Spaeth AE. Respiratory fuels and nitrogen metabolism in vivo in small intestine of fed rats. Quantitative importance of glutamine, glutamate, and aspartate. J Biol Chem. 1980;255:107-112.  [PubMed]  [DOI]  [Cited in This Article: ]
82.  Wu G. Intestinal mucosal amino acid catabolism. J Nutr. 1998;128:1249-1252.  [PubMed]  [DOI]  [Cited in This Article: ]
83.  Wakabayashi Y, Yamada E, Yoshida T, Takahashi N. Effect of intestinal resection and arginine-free diet on rat physiology. Am J Physiol. 1995;269:G313-G318.  [PubMed]  [DOI]  [Cited in This Article: ]
84.  Sido B, Hack V, Hochlehnert A, Lipps H, Herfarth C, Dröge W. Impairment of intestinal glutathione synthesis in patients with inflammatory bowel disease. Gut. 1998;42:485-492.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 165]  [Cited by in F6Publishing: 178]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
85.  Buchman AL, Scolapio J, Fryer J. AGA technical review on short bowel syndrome and intestinal transplantation. Gastroenterology. 2003;124:1111-1134.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 376]  [Cited by in F6Publishing: 300]  [Article Influence: 14.3]  [Reference Citation Analysis (0)]
86.  Dejong CH, Welters CF, Deutz NE, Heineman E, Soeters PB. Renal arginine metabolism in fasted rats with subacute short bowel syndrome. Clin Sci (Lond). 1998;95:409-418.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 11]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
87.  Osowska S, Moinard C, Neveux N, Loï C, Cynober L. Citrulline increases arginine pools and restores nitrogen balance after massive intestinal resection. Gut. 2004;53:1781-1786.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 122]  [Cited by in F6Publishing: 110]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
88.  Yamada E, Wakabayashi Y, Saito A, Yoda K, Tanaka Y, Miyazaki M. Hyperammonaemia caused by essential aminoacid supplements in patient with short bowel. Lancet. 1993;341:1542-1543.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 13]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
89.  Grazer RE, Sutton JM, Friedstrom S, McBarron FD. Hyperammonemic encephalopathy due to essential amino acid hyperalimentation. Arch Intern Med. 1984;144:2278-2279.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 17]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
90.  Pita AM, Wakabayashi Y, Fernandez-Bustos MA, Virgili N, Riudor E, Soler J, Farriol M. Plasma urea-cycle-related amino acids, ammonium levels, and urinary orotic acid excretion in short-bowel patients managed with an oral diet. Clin Nutr. 2003;22:93-98.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 15]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
91.  Windmueller HG, Spaeth AE. Intestinal metabolism of glutamine and glutamate from the lumen as compared to glutamine from blood. Arch Biochem Biophys. 1975;171:662-672.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 255]  [Cited by in F6Publishing: 261]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
92.  Windmueller HG, Spaeth AE. Metabolism of absorbed aspartate, asparagine, and arginine by rat small intestine in vivo. Arch Biochem Biophys. 1976;175:670-676.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 103]  [Cited by in F6Publishing: 109]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
93.  Battezzati A, Brillon DJ, Matthews DE. Oxidation of glutamic acid by the splanchnic bed in humans. Am J Physiol. 1995;269:E269-E276.  [PubMed]  [DOI]  [Cited in This Article: ]
94.  Reeds PJ, Burrin DG, Jahoor F, Wykes L, Henry J, Frazer EM. Enteral glutamate is almost completely metabolized in first pass by the gastrointestinal tract of infant pigs. Am J Physiol. 1996;270:E413-E418.  [PubMed]  [DOI]  [Cited in This Article: ]
95.  Stoll B, Burrin DG, Henry J, Yu H, Jahoor F, Reeds PJ. Substrate oxidation by the portal drained viscera of fed piglets. Am J Physiol. 1999;277:E168-E175.  [PubMed]  [DOI]  [Cited in This Article: ]
96.  Windmueller HG. Glutamine utilization by the small intestine. Adv Enzymol Relat Areas Mol Biol. 1982;53:201-237.  [PubMed]  [DOI]  [Cited in This Article: ]
97.  Wu G, Knabe DA, Yan W, Flynn NE. Glutamine and glucose metabolism in enterocytes of the neonatal pig. Am J Physiol. 1995;268:R334-R342.  [PubMed]  [DOI]  [Cited in This Article: ]
98.  Windmueller HG, Spaeth AE. Uptake and metabolism of plasma glutamine by the small intestine. J Biol Chem. 1974;249:5070-5079.  [PubMed]  [DOI]  [Cited in This Article: ]
99.  Windmueller HG, Spaeth AE. Identification of ketone bodies and glutamine as the major respiratory fuels in vivo for postabsorptive rat small intestine. J Biol Chem. 1978;253:69-76.  [PubMed]  [DOI]  [Cited in This Article: ]
100.  Windmueller HG, Spaeth AE. Vascular perfusion of rat small intestine: metabolic studies with isolated and in situ preparations. Fed Proc. 1977;36:177-181.  [PubMed]  [DOI]  [Cited in This Article: ]
101.  Alteheld B, Stehle P, Furst P.  Measurement of amino acid concentrations in biological fluids and tissues using reversed-phase HPLC-based methods. In: Cynober L, editor. Metabolic and therapeutic aspects of amino acids in clinical nutrition. 2nd ed. CRC Press 2004; 29-44.  [PubMed]  [DOI]  [Cited in This Article: ]
102.  Oehler R, Roth E. Regulative capacity of glutamine. Curr Opin Clin Nutr Metab Care. 2003;6:277-282.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 34]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
103.  Watford M, Lund P, Krebs HA. Isolation and metabolic characteristics of rat and chicken enterocytes. Biochem J. 1979;178:589-596.  [PubMed]  [DOI]  [Cited in This Article: ]
104.  Wu G, Knabe DA, Flynn NE. Synthesis of citrulline from glutamine in pig enterocytes. Biochem J. 1994;299:115-121.  [PubMed]  [DOI]  [Cited in This Article: ]
105.  Pinkus LM, Windmueller HG. Phosphate-dependent glutaminase of small intestine: localization and role in intestinal glutamine metabolism. Arch Biochem Biophys. 1977;182:506-517.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 133]  [Cited by in F6Publishing: 148]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
106.  Meijer AJ Ureagenesis and ammoniagenesis: an update. In: Cynober L, editor. Metabolic and therapeutic aspects of amino acids in clinical nutrition. 2nd ed. CRC Press 2004; 111-122.  [PubMed]  [DOI]  [Cited in This Article: ]
107.  Coomes MW. Amino acid metabolism. Textbook of Biochemistry with clinical correlations. 5th ed. New York: Wiley-Liss; 2002; 779-823.  [PubMed]  [DOI]  [Cited in This Article: ]
108.  Raijman L. Citrulline synthesis in rat tissues and liver content of carbamoyl phosphate and ornithine. Biochem J. 1974;138:225-232.  [PubMed]  [DOI]  [Cited in This Article: ]
109.  Hurwitz R, Kretchmer N. Development of arginine-synthesizing enzymes in mouse intestine. Am J Physiol. 1986;251:G103-G110.  [PubMed]  [DOI]  [Cited in This Article: ]
110.  Wu G. Urea synthesis in enterocytes of developing pigs. Biochem J. 1995;312:717-723.  [PubMed]  [DOI]  [Cited in This Article: ]
111.  van der Hulst RR, von Meyenfeldt MF, Deutz NE, Soeters PB. Glutamine extraction by the gut is reduced in depleted [corrected] patients with gastrointestinal cancer. Ann Surg. 1997;225:112-121.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 46]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
112.  Meijer AJ, Lamers WH, Chamuleau RA. Nitrogen metabolism and ornithine cycle function. Physiol Rev. 1990;70:701-748.  [PubMed]  [DOI]  [Cited in This Article: ]
113.  Wakabayashi Y, Yamada E, Yoshida T, Takahashi H. Arginine becomes an essential amino acid after massive resection of rat small intestine. J Biol Chem. 1994;269:32667-32671.  [PubMed]  [DOI]  [Cited in This Article: ]
114.  Chen K, Nezu R, Sando K, Haque SM, Iiboshi Y, Masunari A, Yoshida H, Kamata S, Takagi Y, Okada A. Influence of glutamine-supplemented parenteral nutrition on intestinal amino acid metabolism in rats after small bowel resection. Surg Today. 1996;26:618-623.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 25]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
115.  Déchelotte P, Darmaun D, Rongier M, Hecketsweiler B, Rigal O, Desjeux JF. Absorption and metabolic effects of enterally administered glutamine in humans. Am J Physiol. 1991;260:G677-G682.  [PubMed]  [DOI]  [Cited in This Article: ]
116.  Flynn NE, Wu G. An important role for endogenous synthesis of arginine in maintaining arginine homeostasis in neonatal pigs. Am J Physiol. 1996;271:R1149-R1155.  [PubMed]  [DOI]  [Cited in This Article: ]
117.  Wakabayashi Y, Henslee JG, Jones ME. Pyrroline-5-carboxylate synthesis from glutamate by rat intestinal mucosa. Subcellular localization and temperature stability. J Biol Chem. 1983;258:3873-3882.  [PubMed]  [DOI]  [Cited in This Article: ]
118.  Kramer JJ, Henslee JG, Wakabayashi Y, Jones ME. Delta 1-pyrroline-5-carboxylate synthase from rat intestinal mucosa. Methods Enzymol. 1985;113:113-120.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 7]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]
119.  Wakabayashi Y The glutamate crossway. In: Cynober LA, editor. Metabolic and therapeutic aspects of amino acids in clinical nutrition. 2nd ed. CRC Press 2004; 135-152.  [PubMed]  [DOI]  [Cited in This Article: ]
120.  Wakabayashi Y, Yamada E, Hasegawa T, Yamada R. Enzymological evidence for the indispensability of small intestine in the synthesis of arginine from glutamate. I. Pyrroline-5-carboxylate synthase. Arch Biochem Biophys. 1991;291:1-8.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in F6Publishing: 74]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
121.  Wu G, Davis PK, Flynn NE, Knabe DA, Davidson JT. Endogenous synthesis of arginine plays an important role in maintaining arginine homeostasis in postweaning growing pigs. J Nutr. 1997;127:2342-2349.  [PubMed]  [DOI]  [Cited in This Article: ]
122.  Wu G. Synthesis of citrulline and arginine from proline in enterocytes of postnatal pigs. Am J Physiol. 1997;272:G1382-G1390.  [PubMed]  [DOI]  [Cited in This Article: ]
123.  Samuels SE, Aarts HL, Ball RO. Effect of dietary proline on proline metabolism in the neonatal pig. J Nutr. 1989;119:1900-1906.  [PubMed]  [DOI]  [Cited in This Article: ]
124.  Adams E, Frank L. Metabolism of proline and the hydroxyprolines. Annu Rev Biochem. 1980;49:1005-1061.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 353]  [Cited by in F6Publishing: 322]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
125.  Wu G, Knabe DA. Arginine synthesis in enterocytes of neonatal pigs. Am J Physiol. 1995;269:R621-R629.  [PubMed]  [DOI]  [Cited in This Article: ]
126.  Schoknecht PA, Pond WG. Short-term ingestion of a high protein diet increases liver and kidney mass and protein accretion but not cellularity in young pigs. Proc Soc Exp Biol Med. 1993;203:251-254.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 10]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
127.  Levillain O, Parvy P, Hassler C. Amino acid handling in uremic rats: citrulline, a reliable marker of renal insufficiency and proximal tubular dysfunction. Metabolism. 1997;46:611-618.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 32]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
128.  Wu G, Morris SM. Arginine metabolism: nitric oxide and beyond. Biochem J. 1998;336:1-17.  [PubMed]  [DOI]  [Cited in This Article: ]
129.  Shih VE. Congenital hyperammonemic syndromes. Clin Perinatol. 1976;3:3-14.  [PubMed]  [DOI]  [Cited in This Article: ]
130.  Chan W, Wang M, Kopple JD, Swendseid ME. Citrulline levels and urea cycle enzymes in uremic rats. J Nutr. 1974;104:678-683.  [PubMed]  [DOI]  [Cited in This Article: ]
131.  Wu G, Morris SM, Jr .  Arginine metabolism in mammals. In: Cynober LA, editor. Metabolic and therapeutic aspects of amino acids in clinical nutrition. 2nd ed. CRC Press 2004; 153-167.  [PubMed]  [DOI]  [Cited in This Article: ]
132.  Hoogenraad N, Totino N, Elmer H, Wraight C, Alewood P, Johns RB. Inhibition of intestinal citrulline synthesis causes severe growth retardation in rats. Am J Physiol. 1985;249:G792-G799.  [PubMed]  [DOI]  [Cited in This Article: ]
133.  Castillo L, Sánchez M, Vogt J, Chapman TE, DeRojas-Walker TC, Tannenbaum SR, Ajami AM, Young VR. Plasma arginine, citrulline, and ornithine kinetics in adults, with observations on nitric oxide synthesis. Am J Physiol. 1995;268:E360-E367.  [PubMed]  [DOI]  [Cited in This Article: ]
134.  Crenn P, Coudray-Lucas C, Cynober L, Messing B. Post-absorptive plasma citrulline concentration: a marker of intestinal failure in humans. Transplant Proc. 1998;30:2528.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 19]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
135.  Dugan ME, Knabe DA, Wu G. The induction of citrulline synthesis from glutamine in enterocytes of weaned pigs is not due primarily to age or change in diet. J Nutr. 1995;125:2388-2393.  [PubMed]  [DOI]  [Cited in This Article: ]
136.  Wu G, Meininger CJ, Kelly K, Watford M, Morris SM. A cortisol surge mediates the enhanced expression of pig intestinal pyrroline-5-carboxylate synthase during weaning. J Nutr. 2000;130:1914-1919.  [PubMed]  [DOI]  [Cited in This Article: ]
137.  Flynn NE, Wu G. Enhanced metabolism of arginine and glutamine in enterocytes of cortisol-treated pigs. Am J Physiol. 1997;272:G474-G480.  [PubMed]  [DOI]  [Cited in This Article: ]
138.  Flynn NE, Wu G. Glucocorticoids play an important role in mediating the enhanced metabolism of arginine and glutamine in enterocytes of postweaning pigs. J Nutr. 1997;127:732-737.  [PubMed]  [DOI]  [Cited in This Article: ]
139.  Flynn NE, Meininger CJ, Kelly K, Ing NH, Morris SM, Wu G. Glucocorticoids mediate the enhanced expression of intestinal type II arginase and argininosuccinate lyase in postweaning pigs. J Nutr. 1999;129:799-803.  [PubMed]  [DOI]  [Cited in This Article: ]
140.  Wu G. An important role for pentose cycle in the synthesis of citrulline and proline from glutamine in porcine enterocytes. Arch Biochem Biophys. 1996;336:224-230.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 19]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
141.  Dillon EL, Knabe DA, Wu G. Lactate inhibits citrulline and arginine synthesis from proline in pig enterocytes. Am J Physiol. 1999;276:G1079-G1086.  [PubMed]  [DOI]  [Cited in This Article: ]
142.  Houdijk AP, van Leeuwen PA, Teerlink T, Flinkerbusch EL, Boermeester MA, Sauerwein HP, Wesdorp RI. Glutamine-enriched enteral diet increases renal arginine production. JPEN J Parenter Enteral Nutr. 1994;18:422-426.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 50]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
143.  Carpenter TO, Levy HL, Holtrop ME, Shih VE, Anast CS. Lysinuric protein intolerance presenting as childhood osteoporosis. Clinical and skeletal response to citrulline therapy. N Engl J Med. 1985;312:290-294.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 28]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
144.  Rajantie J, Simell O, Rapola J, Perheentupa J. Lysinuric protein intolerance: a two-year trial of dietary supplementation therapy with citrulline and lysine. J Pediatr. 1980;97:927-932.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 37]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
145.  Kawamoto S, Strong RW, Kerlin P, Lynch SV, Steadman C, Kobayashi K, Nakagawa S, Matsunami H, Akatsu T, Saheki T. Orthotopic liver transplantation for adult-onset type II citrullinaemia. Clin Transplant. 1997;11:453-458.  [PubMed]  [DOI]  [Cited in This Article: ]
146.  Fletcher JM, Couper R, Moore D, Coxon R, Dorney S. Liver transplantation for citrullinaemia improves intellectual function. J Inherit Metab Dis. 1999;22:581-586.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 15]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
147.  Rabier D, Diry C, Rotig A, Rustin P, Heron B, Bardet J, Parvy P, Ponsot G, Marsac C, Saudubray JM. Persistent hypocitrullinaemia as a marker for mtDNA NARP T 8993 G mutation? J Inherit Metab Dis. 1998;21:216-219.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 22]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
148.  Kobayashi K, Horiuchi M, Saheki T. Pancreatic secretory trypsin inhibitor as a diagnostic marker for adult-onset type II citrullinemia. Hepatology. 1997;25:1160-1165.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 58]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
149.  Wanchu A, Khullar M, Sud K, Sakhuja V, Thennarasu K, Sud A, Bambery P. Serum and Urine Nitrite and Citrulline Levels among Patients with Systemic Lupus Erythematosus: A Possible Addition to Activity Parameters? J Clin Rheumatol. 2001;7:10-15.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 9]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
150.  Mithieux G. New data and concepts on glutamine and glucose metabolism in the gut. Curr Opin Clin Nutr Metab Care. 2001;4:267-271.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 56]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
151.  Crenn P, Thuillier F, Rakatoambinina B, Rongier M, Darmaun D, Messing B. Duodenal vs. gastric administration of labeled leucine for the study of splanchnic metabolism in humans. J Appl Physiol (1985). 2000;89:573-580.  [PubMed]  [DOI]  [Cited in This Article: ]
152.  Pappas PA, Tzakis AG, Saudubray JM, Gaynor JJ, Carreno MR, Huijing F, Kleiner G, Rabier D, Kato T, Levi DM. Trends in serum citrulline and acute rejection among recipients of small bowel transplants. Transplant Proc. 2004;36:345-347.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 23]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
153.  Jones ME, Anderson AD, Anderson C, Hode S. Citrulline synthesis in rat tissues. Arch Biochem Biophys. 1961;95:499-507.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 139]  [Cited by in F6Publishing: 151]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
154.  Oehler R, Roth E.  Glutamine metabolism. In: Cynober LA, editor. Metabolic and therapeutic aspects of amino acids in clinical nutrition. 2nd ed. CRC Press 2004; 169-182.  [PubMed]  [DOI]  [Cited in This Article: ]
155.  Potten CS, Hendry JH.  Radiation and gut. Amsterdam: Elsevier Science B.V 1995; PMC1382624.  [PubMed]  [DOI]  [Cited in This Article: ]
156.  Hanson WR. Radiation protection of murine intestine by WR-2721, 16,16-dimethyl prostaglandin E2, and the combination of both agents. Radiat Res. 1987;111:361-373.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 45]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]