Sunteți pe pagina 1din 9

525210

research-article2014
PENXXX10.1177/0148607114525210Journal of Parenteral and Enteral NutritionTappenden

Invited Review
Journal of Parenteral and Enteral
Nutrition
Intestinal Adaptation Following Resection Volume 38 Supplement 1
May 2014 23S­–31S
© 2014 American Society
for Parenteral and Enteral Nutrition
DOI: 10.1177/0148607114525210
jpen.sagepub.com
Kelly A. Tappenden, PhD, RD, FASPEN1 hosted at
online.sagepub.com

Abstract
Intestinal adaptation is a natural compensatory process that occurs following extensive intestinal resection, whereby structural and functional
changes in the intestine improve nutrient and fluid absorption in the remnant bowel. In animal studies, postresection structural adaptations
include bowel lengthening and thickening and increases in villus height and crypt depth. Functional changes include increased nutrient
transporter expression, accelerated crypt cell differentiation, and slowed transit time. In adult humans, data regarding adaptive changes are
sparse, and the mechanisms underlying intestinal adaptation remain to be fully elucidated. Several factors influence the degree of intestinal
adaptation that occurs post resection, including site and extent of resection, luminal stimulation with enteral nutrients, and intestinotrophic
factors. Two intestinotrophic growth factors, the glucagon-like peptide 2 analog teduglutide and recombinant growth hormone (somatropin),
are now approved for clinical use in patients with short bowel syndrome (SBS). Both agents enhance fluid absorption and decrease requirements
for parenteral nutrition (PN) and/or intravenous fluid. Intestinal adaptation has been thought to be limited to the first 1–2 years following
resection in humans. However, recent data suggest that a significant proportion of adult patients with SBS can achieve enteral autonomy,
even after many years of PN dependence, particularly with trophic stimulation. (JPEN J Parenter Enteral Nutr. 2014;38(suppl 1):23S-31S)

Keywords
short bowel syndrome; intestinal resection; intestinal adaptation; malabsorption

Intestinal adaptation is a natural compensatory process that differentiation into specialized mucosal cells (ie, enterocytes,
occurs following extensive intestinal resection, whereby struc- enteroendocrine cells, goblet cells, or Paneth cells).5 At the end
tural and functional changes in the intestine improve nutrient of their life cycles, the differentiated epithelial cells undergo
and fluid absorption in the remnant bowel.1,2 The degree of apoptosis.6 In animal studies, small bowel resection is fol-
intestinal adaptation that can be achieved is related to both the lowed by a rapid acceleration of crypt cell proliferation
extent of the resection and the anatomy of the remnant bowel. (Figure 1).7 As a result, crypt depth and villus height are
For example, adaptation is more pronounced in the ileum than increased.3,8 In contrast, attenuation of apoptosis does not
in the jejunum, which may account, in part, for the improved appear to be a mechanism for intestinal adaptation; instead, the
outcomes observed in patients who retain some ileal tissue rate may modestly increase as a consequence of enhanced
compared with patients with end-jejunostomy, even after con- crypt cell proliferation and a greater total number of entero-
trolling for the length of the remnant bowel.3,4 For adult cytes.9 Intestinal resection is also associated with local angio-
patients with short bowel syndrome (SBS), the degree of intes- genesis and resultant increases in tissue oxygenation and blood
tinal adaptation by the remnant bowel is one of the factors that flow (Figure 1).10-12 These changes support mucosal growth
determines whether permanent intestinal failure develops and and may also enhance absorptive function.
lifelong parenteral nutrition (PN) is necessary. This review
presents an overview of the structural and functional changes
that occur during intestinal adaptation following intestinal From the 1Department of Food Science and Human Nutrition, University
resection, factors that play a role in mediating these changes, of Illinois at Urbana-Champaign, Urbana, Illinois.
and different approaches to measuring intestinal adaptation in
Financial disclosure: The publication of the supplement in which
individual patients. This information is intended to provide cli- this article appears is supported by an educational grant from NPS
nicians with a better understanding of the multifaceted process Pharmaceuticals, Inc (Bedminster, New Jersey). K.A.T. has served as a
of intestinal adaptation and its importance in the successful consultant for NPS Pharmaceuticals, Inc. No financial compensation was
management of adult patients with SBS. provided to K.A.T. for the preparation of this work. Funding for medical
writing assistance was provided by NPS Pharmaceuticals, Inc.

Physiological Features of Intestinal Adaptation Received for publication December 11, 2013; accepted for publication
February 3, 2014.
Structural Changes Associated With Corresponding Author:
Intestinal Adaptation Kelly A. Tappenden, PhD, RD, FASPEN, Department of Food Science
and Human Nutrition, University of Illinois at Urbana-Champaign, 443
The epithelial mucosa of the intact small bowel undergoes con- Bevier Hall, 905 South Goodwin Avenue, Urbana, IL 61801, USA.
tinual self-renewal via crypt cell proliferation, migration, and Email: tappende@illinois.edu
Downloaded from pen.sagepub.com at RUTGERS UNIV on April 27, 2015
24S Journal of Parenteral and Enteral Nutrition 38(Suppl 1)

height was decreased for patients compared with control sub-


jects while they were maintained on PN but was increased
compared with control subjects 2 months after enteral feed-
ing resumed.20 Further, most animal studies involve resec-
tions of 75% or more of the intestinal tissue, but resections in
humans are rarely so extensive. In both human and animal
studies, the extent of resection correlates with the degree of
adaptation.15,18,21 Finally, mechanisms for adaptation may
differ between humans and other mammalian models.
Postresection adaptation clearly occurs in humans, as is dem-
onstrated by the majority of patients who progress from PN
Figure 1.  Structural and functional changes that occur in the dependence to enteral autonomy within the first several years
intestine during postresection adaptation. following surgery.22 Intestinal adaptation in humans may rely
more on functional rather than structural changes; however,
additional supporting clinical data are needed to draw defini-
In humans, gross changes in intestinal morphological fea-
tive conclusions.19
tures, including small bowel lengthening and dilation, have
been observed after resection or jejuno-ileal bypass.13,14
However, only limited evidence regarding modification of Functional Changes Associated With
intestinal microarchitecture post resection is available. Some
of the more frequently cited studies assessing structural intesti-
Intestinal Adaptation
nal adaptation in humans have been conducted in pediatric In addition to structural changes, functional adaptations that
patients. McDuffie et al15 evaluated intestinal morphological increase absorptive capacity also occur following resection
characteristics in 33 infants with neonatal necrotizing entero- (Figure 1). In animal studies, intestinal resection is associated
colitis both at the time of resection and also at ostomy reversal, with increased expression of transporter proteins and
which occurred at a mean of 74 days post surgery. In these exchangers involved in nutrient, electrolyte, and water
patients, villus height and crypt depth increased by 32% and absorption, including the sodium glucose cotransporter, Na+/
22%, respectively, at ostomy reversal compared with baseline H+ exchangers, and Na+/K+ adenosine triphosphatases.23-26
(P < .01 for both measures). Furthermore, the magnitude of the Importantly, these studies have documented an elevation of
change in villus height was significantly correlated with the transporter or exchanger expression that is not wholly a result
length of small bowel removed (r = 0.36; P < .05).15 of increased enterocyte mass. In addition, developing entero-
Few studies have been performed to date in adult patients, cytes express digestive enzymes and amino acid transporters
and these studies have been limited by small sample sizes and more rapidly following small bowel resection than in an
histological analyses performed at a single time point post intact bowel, suggesting an accelerated maturation pro-
resection. Doldi14 reported enterocyte hyperplasia and a 70%– cess.27,28 The net result of these changes in protein expression
75% increase in villus height in the small intestine of 13 and activity is to increase the digestive and absorptive capac-
patients at 2 years following jejuno-ileal bypass surgery com- ity of the remnant bowel.23
pared with controls; however, a statistical analysis was not per- Older studies in human adults documented enhanced
formed as part of this study. Joly et al16 documented a 35% absorption of glucose per unit length of intestine following
increase in crypt depth and a 22% increase in cell number/crypt resection.1,2 However, increased absorptive capacity observed
in the colon of 12 patients with jejuno-colonic anastomosis in these studies could be a result of localized hyperplasia, func-
compared with healthy controls (P < .005) at a mean of tional adaptation, or both. Data regarding potential mecha-
9.8 years following resection, but the histological status of the nisms for functional postresection intestinal adaptation in
small intestine was not evaluated. Several other studies showed humans are sparse. In a study by Ziegler et al,19 expression of
no changes in epithelial proliferation or crypt depth and villus PepT1, a transporter of dipeptides and tripeptides, was
height in the small intestine of patients with SBS compared increased in the colon, but not the small intestine, of patients
with healthy controls.17-19 with SBS compared with controls.19 In contrast, a later pro-
Several reasons may account for the limited evidence for spective case-control study failed to document a difference in
structural adaptation in human adults following resection. PepT1 mRNA or protein expression in the colon of 12 patients
Studies performed to date have included relatively few with SBS compared with 14 healthy volunteers.16 However,
patients and have examined single time points. Furthermore, this later study was performed a mean of 9.8 ± 5.7 years after
most patients are maintained on PN for at least some time resection, whereas in the earlier study, patients had been depen-
following resection; however, enteral nutrition is required for dent on PN for an average of 1.5–2.5 years. Thus, the rise in
maximal intestinal adaptation (see below). In a longitudinal colonic PepT1 expression may be an early event in the adapta-
study of 2 patients following small bowel resection, villus tion process.

Downloaded from pen.sagepub.com at RUTGERS UNIV on April 27, 2015


Tappenden 25S

Another mechanism for functional adaptation post resec- PN-induced mucosal atrophy reverses upon reintroduction of
tion is a deceleration of small bowel transit, which lengthens enteral nutrition.38,39 In contrast, luminal nutrients enhance
the contact time between luminal nutrients and the absorptive postresection adaptation.41,42 Further, increased nutrient com-
mucosa. In dogs, small intestinal transit time increased by 35% plexity is associated with greater adaptation, likely because of
and 29% following 50% and 75% resections, respectively, the greater digestive activity required for nutrient absorption.43
between 4 weeks and 12 weeks after surgery (P < .05 for each For example, rats and pigs fed a chow diet following intestinal
comparison).29 In patients who have undergone intestinal resection showed greater structural adaptation and digestive
resection, diarrhea decreases during the initial adaptive period enzyme activity than similar animals that received an elemen-
following surgery.30 Slowed gastrointestinal transit time, in tal diet.42,44
addition to other structural and functional changes, may con- Individual nutrients have been associated with intestinotro-
tribute to reduced diarrhea post resection. Indeed, compared phic effects following resection. Fats, in particular, appear to
with healthy controls, patients with an ileal resection and promote adaptation. Compared with standard chow, a high-fat
colon-in-continuity have higher plasma levels of peptide YY, a diet was associated with significantly increased bowel weight
hormone released by the distal intestine that acts to delay gas- and villus height at 14 days post resection in rats.45 In contrast,
tric emptying and intestinal transit.31 a low-fat diet reduced markers of intestinal adaptation com-
pared with a higher fat diet containing the same number of
calories.46 Specific fats may vary in their ability to stimulate
Key Factors in Intestinal Adaptation adaptation. Long-chain triglycerides are superior to medium-
Multiple factors can influence the extent of intestinal adapta- chain triglycerides in inducing hyperplasia of the intestinal epi-
tion that occurs post resection, including anatomic features, thelium in the postresection setting.47,48 Among the long-chain
route of nutrition delivery, and hormones and growth factors. triglycerides, however, the relative benefit of saturated versus
polyunsaturated fatty acids has not been resolved. In a study by
Anatomic features.  The site of intestinal resection influences Vanderhoof et al,49 resected rats receiving a diet high in men-
the extent of subsequent adaptation, with the ileum displaying haden oil, a polyunsaturated fatty acid, showed greater
greater adaptive potential than the jejunum.3,32,33 In humans, increases in mucosal weight and DNA and protein content
proximal resections are associated with decreased diarrhea and when compared with rats fed diets high in safflower oil, a less
steatorrhea relative to distal resections.34 The increased adap- highly unsaturated fatty acid, or beef tallow, a saturated fatty
tive capacity of the ileum relative to the jejunum may be attrib- acid. Similarly, diets high in docosahexaenoic acid and arachi-
utable to the fact that under normal conditions, the distal donic acid, 2 polyunsaturated fatty acids, stimulated greater
intestine is exposed to fewer luminal nutrients, which act as increases in mucosal mass than diets high in safflower oil or
potent stimuli for intestinal growth (see below). Indeed, in the hydrogenated coconut oil.50 In contrast, a study by Keelan et
unresected intestine, villus height gradually decreases along al51 demonstrated that resected rats receiving diets high in satu-
the proximal-distal axis.35 Furthermore, in transposition stud- rated fatty acids had significantly increased jejunal glucose
ies, an ileal segment that is moved proximally undergoes adap- transport compared with animals receiving diets high in poly-
tation, as is evident by significant increases in mucosal growth unsaturated fatty acids. This study did not evaluate the micro-
and glucose absorption, even in the absence of tissue loss.3,35 In architecture of the small bowel, but a more recent study by
contrast, villus size decreases in jejunal segments that have Sukhotnik et al52 showed that villus height and crypt depth
been transposed distally.35 increased significantly in resected rats receiving a diet high in
In addition to the site of resection, the extent of resection palmitic acid, a saturated fatty acid, compared with rats receiv-
affects the adaptive outcome. In both humans and animals, ing a diet with low palmitic acid content. However, the results
greater tissue loss is positively correlated with adaptation follow- of these animal studies may not directly translate to clinical
ing surgery.15,18,21 However, the ability of the gut to fully compen- applications. Patients with SBS and colon-in-continuity are
sate for tissue loss is limited. In a retrospective study of 268 adult generally not able to tolerate high-fat diets because of steator-
patients with SBS, remnant bowel length of <75 cm was signifi- rhea and reduced nutrient absorption.53
cantly associated with greater probability of permanent depen- Like long-chain triglycerides, short-chain fatty acids
dence on PN in a multivariate analysis.36 The correlation between (SCFAs) have been shown to enhance intestinal adaptation.
remnant bowel length and anatomy with patient outcomes is dis- Most SCFAs in the human diet are derived from carbohydrates
cussed more fully in an accompanying article.37 that undergo bacterial fermentation in the colon.54 In unre-
sected rats maintained on PN, supplementation with SCFAs
Role of enteral nutrients. Enteral stimulation is required to significantly mitigated PN-induced declines in mucosal weight
maintain the structure of the intestinal mucosa. The absence of and DNA and RNA content.55 Furthermore, SCFA supplemen-
enteral nutrition induces mucosal atrophy and decreases diges- tation of PN increases nutrient transporter expression and
tive enzyme and nutrient transporter activity in animals and nutrient absorption.56,57 Following experimental resection, PN
humans, even when adequate calories are provided via PN.38-40 supplemented with SCFAs enhances intestinal adaptation in

Downloaded from pen.sagepub.com at RUTGERS UNIV on April 27, 2015


26S Journal of Parenteral and Enteral Nutrition 38(Suppl 1)

mature rats and neonatal piglets.58,59 Although supporting data volume requirements than diet modification alone. However, 3
are sparse, patients with SBS and colon-in-continuity may also months following treatment discontinuation, the GH-induced
benefit from SCFAs. In a study of 6 patients with SBS, dietary PN/IV reductions were partially reversed and no longer signifi-
supplementation for 20 days with pectin, a starch that acts as a cantly different from baseline values.78
substrate for bacterial SCFA production, increased colonic lev- GH appears to be more effective as a part of a multimodal
els of SCFAs and resulted in a nonsignificant trend for therapy in combination with glutamine. Most, but not all, ani-
increased fluid absorption.60 mal studies evaluating GH with glutamine document improve-
The amino acid glutamine, rather than glucose, is the pri- ments in measures of intestinal adaptation.65,79-81 In humans,
mary energy source for enterocytes, a fact that has prompted GH plus glutamine is associated with greater and longer lasting
interest in exploring the role of glutamine in intestinal growth.61 reductions in PN/IV volume requirements and infusion fre-
In animal studies, the efficacy of glutamine depends on the quency than GH alone.78 Furthermore, in a subsequent study
route of administration. When used as a supplement to PN, glu- by Guo et al,82 10 adult patients with SBS showed significant
tamine counteracts PN-induced intestinal atrophy and improves increases in villus height, mucosal proliferation, and nitrogen
parameters of postresection adaptation.62-65 In contrast, gluta- absorption following 4 weeks of treatment with GH, gluta-
mine added to chow or an elemental diet has no effect on or mine, and enteral nutrition. A meta-analysis of 5 human studies
decreases measures of structural or functional intestinal adap- indicated that GH, with or without glutamine, improves energy
tation.66-69 It is likely that any modest effects of glutamine are absorption in patients with SBS.83 However, a subanalysis of
masked by the greater trophic stimulation provided by luminal data evaluating GH monotherapy showed no effect on energy
nutrition in these studies. No studies assessing the effect of absorption. Furthermore, in most studies, any benefits of GH
glutamine alone on intestinal adaptation have been conducted were reversed upon treatment discontinuation. The authors of
in humans with SBS, but glutamine in combination with the meta-analysis concluded that data regarding a potential
growth hormone (GH) has shown some efficacy (see below). benefit of GH for patients with SBS are inconclusive.83
Somatropin, a recombinant form of human GH, was approved
Hormones and growth factors. A variety of mediators have for use as a 4-week treatment regimen in patients with SBS in
been posited to act as intestinotrophic factors (Table 1). Two of 2003.84 However, cotreatment with glutamine is not a require-
these, recombinant human GH (somatropin) and the glucagon- ment for somatropin administration.
like peptide 2 (GLP-2) analog teduglutide, are now approved Further details regarding the pharmacological stimulation
for clinical use in adult patients with SBS. of adaptation and the clinical effects of teduglutide and soma-
tropin are provided in a companion paper.85
Growth hormone.  In the late 1970s, Taylor et al70 demon-
strated that hypophysectomy reduced measures of adaptation GLP-2.  GLP-2 is synthesized by enteroendocrine L cells
in resected rats. GH, a pituitary hormone that induces systemic of the distal small intestine and colon in response to nutrient
proliferative and anabolic effects, was quickly identified as stimulation.86 In preclinical models, plasma GLP-2 concentra-
a pituitary-derived factor with the potential to mediate intes- tions increase following resection, and postprandial GLP-2
tinal adaptation.71 However, subsequent preclinical studies levels correlate with the extent of intestinal adaptation.87,88 In
yielded mixed results. In some experiments, exogenous GH addition, PN supplemented with GLP-2 enhanced parameters
significantly increased small bowel length, mucosal height, of postresection adaptation compared with PN alone, including
jejunal villus height, and/or glutamine and leucine transport small bowel weight, small bowel surface area, villus height,
in resected animals.72-74 In contrast, other experiments docu- microvillus height, crypt cell proliferation, nutrient transporter
mented no effect of GH on postresection mucosal growth.74,75 expression, and nutrient absorption.89,90 Even in the absence of
Two small clinical studies have evaluated the effect of GH resection, exogenous GLP-2 augments small bowel weight and
alone on intestinal absorption in adult patients with SBS. The length, villus height, crypt cell proliferation, and jejunal pro-
first study included 10 patients and showed no improvement tein content.91 Expression of GLP-2 also increases in humans
in absorption of energy, nitrogen, or fluid following 8 weeks following intestinal resection, particularly if the colon is in
of treatment with 0.024 mg/kg/d GH, although body weight continuity.92 Furthermore, in two phase 3 studies and a long-
increased by a mean of 2.3 kg (P = .005).76 In contrast, the second term extension study, adult patients with SBS who were treated
study evaluating 12 patients with SBS who received GH (0.05 with the GLP-2 analog teduglutide showed increases in villus
mg/kg/d) for 3 weeks demonstrated significant enhancements height and decreases in PN/IV fluid requirements.93-95 Tedu-
in absorption of energy (P < .002), nitrogen (P < .04), and car- glutide is now approved for clinical use in adults with SBS
bohydrate (P < .04) as well as a mean increase in body weight who are dependent on PN.96
of 2.4 kg (P < .003).77 In a double-blind, placebo-controlled
study conducted in PN-dependent adult patients with SBS, 4 Other factors.  A number of other proteins have shown intes-
weeks of treatment with GH combined with an optimized diet tinotrophic effects in preclinical studies, including epidermal
resulted in greater reductions in PN and/or intravenous (IV) growth factor (EGF),97-101 insulin-like growth factor-1,102-104

Downloaded from pen.sagepub.com at RUTGERS UNIV on April 27, 2015


Tappenden 27S

Table 1.  Growth Factors Approved for Treatment of Short Bowel Syndrome in Adults.

Effects on Intestinal Adaptation: Effects on Intestinal Adaptation:


Agent Preclinical Studies Clinical Studies Stage of Clinical Development
GH and somatropin •• GH increased small bowel •• Somatropin reduced mean PN/ •• One phase 3, placebo-controlled
length, mucosal height, jejunal IV volume requirements and randomized clinical trial
villus height, and glutamine and infusion frequency in patients completed; no long-term
leucine transport in some, but not with SBS; PN/IV effects extension studies available.78
all, studies.72-75 partially reversed after treatment •• Somatropin is approved for clinical
discontinuation.78 use as a 4-week treatment regimen
in adult patients with SBS.84
GH with glutamine •• GH plus glutamine increased •• Somatropin plus glutamine •• One phase 3, placebo-controlled
mucosal thickness, villus height, reduced PN/IV volume randomized clinical trial
crypt depth, cell proliferation, requirements and infusion completed; no long-term
DNA content, and nutrient frequency in patients with SBS extension studies available.78
absorption in some, but not all, (superior results to somatropin •• Somatropin is approved for
studies.65,79,80 alone).78 clinical use as a 4-week treatment
•• GH plus glutamine increased regimen in adult patients with
villus height, mucosal SBS.84
proliferation, and nitrogen
absorption in patients with SBS.82
•• GH plus glutamine increased
energy absorption.83
•• Some patients with SBS
achieved independence from
PN/IV following treatment with
GH, glutamine, and optimized
diet.124,125
GLP-2 and teduglutide •• GLP-2 increased small bowel •• Teduglutide increased villus •• Two phase 3, placebo-controlled
weight, small bowel surface area, height in patients with SBS.93,126 randomized clinical trials and
villus height, microvillus height, •• Teduglutide increased plasma 2 long-term extension studies
crypt cell proliferation, nutrient citrulline levels in patients with completed.94,95,126,127
transporter expression, and SBS.95,126 •• Teduglutide is approved for
nutrient absorption in resected •• Teduglutide reduced mean PN/IV clinical use in adults with
rats.89,90 volume requirements in patients short bowel syndrome who are
•• GLP-2 increased small bowel with SBS and decreased number dependent on PN.96
weight and length, villus height, of infusion days for a majority of
crypt cell proliferation, and patients; effects on PN/IV were
jejunal protein content.91 partially reversed after treatment
discontinuation.94,95,126
•• A small number of patients
treated with teduglutide achieved
independence from PN/IV.94,126,127
GH, growth hormone; GLP-2, glucagon-like peptide 2; PN/IV, parenteral nutrition and/or intravenous fluid; SBS, short bowel syndrome.

and hepatocyte growth factor.105,106 Of these, only EGF has humans is relatively small. Reasons for this discrepancy
been tested clinically, in a single pilot study of 5 pediatric include the small size of the SBS population and the invasive
patients.107 Following 6 weeks of treatment with 100 mcg/kg/d nature of many of the procedures required to assess structural
EGF, patients demonstrated significant improvements in car- and functional adaptation. For example, histological analysis
bohydrate absorption and the percentage of calories obtained of the small intestine requires biopsy, which can be a particular
from enteral feeds (P < .05 for each measure, compared with hardship for patients who have already often endured multiple
baseline). Additional details regarding these and other poten- abdominal surgeries. Magnifying endoscopy may provide a
tial mediators of intestinal adaptation are discussed in a review means of evaluating the microarchitecture of the intestine in
by McMellen et al.71 the absence of biopsy, but further research on this method in
the SBS population is needed.82,108 For assessing functional
adaptation in patients with SBS, 72-hour metabolic balance
Assessing Intestinal Adaptation studies, with fecal and urinary analyses at baseline and after an
Compared with the wealth of animal studies investigating intervention, are the gold standard.109,110 However, this method
postresection intestinal adaptation, the number of studies in is complex, costly, and neither patient- nor clinician-friendly.

Downloaded from pen.sagepub.com at RUTGERS UNIV on April 27, 2015


28S Journal of Parenteral and Enteral Nutrition 38(Suppl 1)

Plasma citrulline, a nonessential amino acid produced by occurs early in the postoperative course, continued improve-
enterocytes, is considered to be a surrogate biomarker for ments in intestinal absorptive capacity are possible and do
enterocyte mass.111 In several studies, plasma citrulline levels occur for many years following resection. The timing of intes-
were strongly correlated with remnant small bowel length.112-116 tinal adaptation following resection may differ in pediatric
In patients with SBS, a postabsorptive plasma citrulline level patients, who experience growth of their remnant intestines as
of <20 µmol/L (ie, half the normal level in adult controls) was they age122,123; however, a full exploration of the topic is
significantly associated with permanent PN dependence when beyond the scope of this paper.
measured >2 years following the last abdominal surgery.112
However, whether citrulline levels accurately reflect the func-
tional absorptive capacity of the small intestine remains unre-
Conclusions
solved. Some studies have reported a correlation between A wealth of preclinical data has highlighted many of the mech-
plasma citrulline levels and nutrient absorption, whereas others anisms responsible for adaptation in nonhuman animals,
have not.112,114-117 Additional studies are required to determine including mucosal hyperplasia, angiogenesis, accelerated
whether plasma citrulline provides an accurate assessment of enterocyte differentiation, increased expression and activity of
postresection functional adaptation. nutrient transporters, and slowed intestinal transit. However,
Other parameters have been used as surrogates for postre- whether some or all of those processes contribute to intestinal
section adaptation in human studies, including weaning off adaptation in humans as well remains to be fully resolved.
PN, fluid composite effect (ie, increase in urine production, Newer, less invasive techniques for assessing intestinal micro-
plus decrease in PN and/or IV fluids, plus reduction in oral architecture and improved methods of evaluating intestinal
fluid intake),95 oral intake and stool weight/oral intake ratio,109 absorptive function may provide additional insights into the
and absorption of inert sugars.118 natural history of postresection adaptation in human patients.
Many, but not all, patients with SBS achieve enteral auton-
omy in the postresection period. Maximizing adaptation in the
Timing of Intestinal Adaptation in Human intestinal remnant by application of newer intestinotrophic
Adults therapies may enhance outcomes for these patients. Recent
Data related to the timing and duration of intestinal adaptation studies suggest that some patients can achieve PN indepen-
in human adults are sparse. According to general consensus, dence even many years following the initial resection. These
the majority of intestinal adaptation in adults occurs within the data argue against the long-held dogmatic assertion that adap-
first 2 years following resection.119 In a study of 50 patients tation occurs only in the first 12–24 months following resec-
with extensive bowel resection, there was no improvement in tion and provide additional hope for patients with SBS.
fecal weight or fecal excretion of fat, potassium, or nitrogen at
≥1 year (mean, 81 months) after surgery compared with Acknowledgments
<6 months after surgery.30 In the oft-cited report by Messing Medical writing assistance was provided by Heather Heerssen,
et al,22 95% of 64 patients with transient intestinal failure PhD, of Complete Healthcare Communications, Inc (Chadds Ford,
obtained enteral autonomy within the first 2 years post resec- Pennsylvania) under the direction of the author. K.A.T. is respon-
tion. In contrast, other studies suggest that significant adapta- sible for the content of the manuscript and had final approval of all
tion can occur beyond the 2-year mark in adults. Gouttebel et revisions.
al120 evaluated calcium absorption in 30 patients with SBS
between 1.5 and 120 months after intestinal resection. Patients
References
who were tested more than 2 years post surgery (mean, 1. Dowling RH, Booth CC. Functional compensation after small-bowel
resection in man: demonstration by direct measurement. Lancet.
52 months) absorbed significantly more calcium than patients
1966;2:146-147.
who were evaluated within 24 months of resection (mean, 2. Weinstein LD, Shoemaker CP, Hersh T, Wright HK. Enhanced
9 months).120 Furthermore, in a recent study that followed intestinal absorption after small bowel resection in man. Arch Surg.
268 patients with SBS at a single institution for a median of 1969;99:560-562.
4 years (range, 0.3–24 years), 26% were weaned off PN more 3. Dowling RH, Booth CC. Structural and functional changes following
small intestinal resection in the rat. Clin Sci. 1967;32:139-149.
than 2 years after the restoration of digestive continuity.36
4. Carbonnel F, Cosnes J, Chevret S, et al. The role of anatomic factors
Similarly, among 72 patients with SBS who achieved enteral in nutritional autonomy after extensive small bowel resection. JPEN J
autonomy during a long-term follow-up study, 47% had been Parenter Enteral Nutr. 1996;20:275-280.
PN dependent for 2–5 years and 18% had been PN dependent 5. Umar S. Intestinal stem cells. Curr Gastroenterol Rep. 2010;12:340-348.
for >5 years.121 Trophic stimulation may enhance this latent 6. Hall PA, Coates PJ, Ansari B, Hopwood D. Regulation of cell number in
the mammalian gastrointestinal tract: the importance of apoptosis. J Cell
adaptive response. In a 52-week study of patients with SBS, 3
Sci. 1994;107(pt 12):3569-3577.
of 25 patients receiving 0.05 mg/kg/d teduglutide were weaned 7. Loran MR, Crocker TT. Population dynamics of intestinal epithelia
off PN after 2–25 years of PN therapy.94 Taken together, these in the rat two months after partial resection of the ileum. J Cell Biol.
data suggest that although most intestinal adaptation in adults 1963;19:285-291.

Downloaded from pen.sagepub.com at RUTGERS UNIV on April 27, 2015


Tappenden 29S

8. Lauronen J, Pakarinen MP, Kuusanmaki P, et al. Intestinal adaptation after 30. Cosnes J, Carbonnel F, Beaugerie L, et al. Functional adaptation after
massive proximal small-bowel resection in the pig. Scand J Gastroenterol. extensive small bowel resection in humans. Eur J Gastroenterol Hepatol.
1998;33:152-158. 1994;6:197-202.
9. Helmrath MA, Erwin CR, Shin CE, Warner BW. Enterocyte apopto- 31. Nightingale JM, Kamm MA, van der Sijp JR, Ghatei MA, Bloom SR,
sis is increased following small bowel resection. J Gastrointest Surg. Lennard-Jones JE. Gastrointestinal hormones in short bowel syndrome:
1998;2:44-49. peptide YY may be the “colonic brake” to gastric emptying. Gut.
10. Martin CA, Perrone EE, Longshore SW, et al. Intestinal resection 1996;39:267-272.
induces angiogenesis within adapting intestinal villi. J Pediatr Surg. 32. Thompson JS, Quigley EM, Adrian TE. Factors affecting outcome fol-
2009;44:1077-1082; discussion 1083. lowing proximal and distal intestinal resection in the dog: an examination
11. Rowland KJ, Yao J, Wang L, et al. Immediate alterations in intestinal oxy- of the relative roles of mucosal adaptation, motility, luminal factors, and
gen saturation and blood flow after massive small bowel resection as mea- enteric peptides. Dig Dis Sci. 1999;44:63-74.
sured by photoacoustic microscopy. J Pediatr Surg. 2012;47:1143-1149. 33. Appleton GV, Bristol JB, Williamson RC. Proximal enterectomy provides
12. Ulrich-Baker MG, Hollwarth ME, Kvietys PR, Granger DN. Blood flow a stronger systemic stimulus to intestinal adaptation than distal enterec-
responses to small bowel resection. Am J Physiol. 1986;251:G815-G822. tomy. Gut. 1987;28(suppl):165-168.
13. Thompson JS, Langnas AN, Pinch LW, Kaufman S, Quigley EM, 34. Cosnes J, Gendre JP, Le Quintrec Y. Role of the ileocecal valve and site of
Vanderhoof JA. Surgical approach to short-bowel syndrome: experience intestinal resection in malabsorption after extensive small bowel resection.
in a population of 160 patients. Ann Surg. 1995;222:600-605; discussion Digestion. 1978;18:329-336.
605-607. 35. Altmann GG, Leblond CP. Factors influencing villus size in the small
14. Doldi SB. Intestinal adaptation following jejuno-ileal bypass. Clin Nutr. intestine of adult rats as revealed by transposition of intestinal segments.
1991;10:138-145. Am J Anat. 1970;127:15-36.
15. McDuffie LA, Bucher BT, Erwin CR, Wakeman D, White FV, Warner 36. Amiot A, Messing B, Corcos O, Panis Y, Joly F. Determinants of home
BW. Intestinal adaptation after small bowel resection in human infants. parenteral nutrition dependence and survival of 268 patients with non-
J Pediatr Surg. 2011;46:1045-1051. malignant short bowel syndrome. Clin Nutr. 2013;32:368-374.
16. Joly F, Mayeur C, Messing B, et al. Morphological adaptation with 37. Tappenden KA. Pathophysiology of short bowel syndrome: consider-
preserved proliferation/transporter content in the colon of patients ations of resected and residual anatomy. JPEN J Parenter Enteral Nutr.
with short bowel syndrome. Am J Physiol Gastrointest Liver Physiol. 2014;38(suppl 1):14S-22S.
2009;297:G116-G123. 38. Buchman AL, Moukarzel AA, Bhuta S, et al. Parenteral nutrition is asso-
17. O’Keefe SJ, Haymond MW, Bennet WM, Oswald B, Nelson DK, Shorter ciated with intestinal morphologic and functional changes in humans.
RG. Long-acting somatostatin analogue therapy and protein metabolism JPEN J Parenter Enteral Nutr. 1995;19:453-460.
in patients with jejunostomies. Gastroenterology. 1994;107:379-388. 39. Guedon C, Schmitz J, Lerebours E, et al. Decreased brush border hydrolase
18. Porus RL. Epithelial hyperplasia following massive small bowel resection activities without gross morphologic changes in human intestinal mucosa
in man. Gastroenterology. 1965;48:753-757. after prolonged total parenteral nutrition of adults. Gastroenterology.
19. Ziegler TR, Fernandez-Estivariz C, Gu LH, et al. Distribution of the H+/ 1986;90:373-378.
peptide transporter PepT1 in human intestine: up-regulated expression 40. Levine GM, Deren JJ, Steiger E, Zinno R. Role of oral intake in main-
in the colonic mucosa of patients with short-bowel syndrome. Am J Clin tenance of gut mass and disaccharide activity. Gastroenterology.
Nutr. 2002;75:922-930. 1974;67:975-982.
20. Pironi L, Paganelli GM, Miglioli M, et al. Morphologic and cytoprolif- 41. Feldman EJ, Dowling RH, McNaughton J, Peters TJ. Effects of oral ver-
erative patterns of duodenal mucosa in two patients after long-term total sus intravenous nutrition on intestinal adaptation after small bowel resec-
parenteral nutrition: changes with oral refeeding and relation to intestinal tion in the dog. Gastroenterology. 1976;70:712-719.
resection. JPEN J Parenter Enteral Nutr. 1994;18:351-354. 42. Ford WD, Boelhouwer RU, King WW, de Vries JE, Ross JS, Malt RA.
21. Juno RJ, Knott AW, Erwin CR, Warner BW. A serum factor(s) after small Total parenteral nutrition inhibits intestinal adaptive hyperplasia in young
bowel resection induces intestinal epithelial cell proliferation: effects of rats: reversal by feeding. Surgery. 1984;96:527-534.
timing, site, and extent of resection. J Pediatr Surg. 2003;38:868-874. 43. Weser E, Babbitt J, Hoban M, Vandeventer A. Intestinal adaptation: dif-
22. Messing B, Crenn P, Beau P, Boutron-Ruault MC, Rambaud JC, ferent growth responses to disaccharides compared with monosaccharides
Matuchansky C. Long-term survival and parenteral nutrition depen- in rat small bowel. Gastroenterology. 1986;91:1521-1527.
dence in adult patients with the short bowel syndrome. Gastroenterology. 44. Bines JE, Taylor RG, Justice F, et al. Influence of diet complexity on
1999;117:1043-1050. intestinal adaptation following massive small bowel resection in a pre-
23. Sigalet DL, Martin GR. Mechanisms underlying intestinal adaptation after clinical model. J Gastroenterol Hepatol. 2002;17:1170-1179.
massive intestinal resection in the rat. J Pediatr Surg. 1998;33:889-892. 45. Sukhotnik I, Mor-Vaknin N, Drongowski RA, Miselevich I, Coran AG,
24. Hines OJ, Bilchik AJ, Zinner MJ, et al. Adaptation of the Na+/glucose Harmon CM. Effect of dietary fat on early morphological intestinal adapta-
cotransporter following intestinal resection. J Surg Res. 1994;57:22-27. tion in a rat with short bowel syndrome. Pediatr Surg Int. 2004;20:419-424.
25. Hines OJ, Bilchik AJ, McFadden DW, et al. Up-regulation of Na+,K+ 46. Sukhotnik I, Shiloni E, Krausz MM, et al. Low-fat diet impairs postre-
adenosine triphosphatase after massive intestinal resection. Surgery. section intestinal adaptation in a rat model of short bowel syndrome.
1994;116:401-407; discussion 408. J Pediatr Surg. 2003;38:1182-1187.
26. Musch MW, Bookstein C, Rocha F, et al. Region-specific adaptation of 47. Vanderhoof JA, Grandjean CJ, Kaufman SS, Burkley KT, Antonson DL.
apical Na/H exchangers after extensive proximal small bowel resection. Effect of high percentage medium-chain triglyceride diet on mucosal
Am J Physiol Gastrointest Liver Physiol. 2002;283:G975-G985. adaptation following massive bowel resection in rats. JPEN J Parenter
27. Chaves M, Smith MW, Williamson RC. Increased activity of digestive Enteral Nutr. 1984;8:685-689.
enzymes in ileal enterocytes adapting to proximal small bowel resection. 48. Chen WJ, Yang CL, Lai HS, Chen KM. Effects of lipids on intestinal
Gut. 1987;28:981-987. adaptation following 60% resection in rats. J Surg Res. 1995;58:253-259.
28. Menge H, Sepulveda FV, Smith MW. Cellular adaptation of amino acid 49. Vanderhoof JA, Park JH, Herrington MK, Adrian TE. Effects of dietary
transport following intestinal resection in the rat. J Physiol. 1983;334: menhaden oil on mucosal adaptation after small bowel resection in rats.
213-223. Gastroenterology. 1994;106:94-99.
29. Quigley EM, Thompson JS. The motor response to intestinal resection: 50. Kollman KA, Lien EL, Vanderhoof JA. Dietary lipids influence intestinal
motor activity in the canine small intestine following distal resection. adaptation after massive bowel resection. J Pediatr Gastroenterol Nutr.
Gastroenterology. 1993;105:791-798. 1999;28:41-45.

Downloaded from pen.sagepub.com at RUTGERS UNIV on April 27, 2015


30S Journal of Parenteral and Enteral Nutrition 38(Suppl 1)

51. Keelan M, Cheeseman CI, Clandinin MT, Thomson AB. Intestinal mor- 71. McMellen ME, Wakeman D, Longshore SW, McDuffie LA, Warner BW.
phology and transport after ileal resection in rat is modified by dietary Growth factors: possible roles for clinical management of the short bowel
fatty acids. Clin Invest Med. 1996;19:63-70. syndrome. Semin Pediatr Surg. 2010;19:35-43.
52. Sukhotnik I, Hayari L, Bashenko Y, et al. Dietary palmitic acid modulates 72. Shulman DI, Hu CS, Duckett G, Lavallee-Grey M. Effects of short-term
intestinal re-growth after massive small bowel resection in a rat. Pediatr growth hormone therapy in rats undergoing 75% small intestinal resec-
Surg Int. 2008;24:1313-1321. tion. J Pediatr Gastroenterol Nutr. 1992;14:3-11.
53. Matarese LE. Nutrition and fluid optimization for patients with short 73. Benhamou PH, Canarelli JP, Leroy C, De Boissieu D, Dupont C.
bowel syndrome. JPEN J Parenter Enteral Nutr. 2013;37:161-170. Stimulation by recombinant human growth hormone of growth and
54. Macfarlane S, Macfarlane GT. Regulation of short-chain fatty acid pro- development of remaining bowel after subtotal ileojejunectomy in rats.
duction. Proc Nutr Soc. 2003;62:67-72. J Pediatr Gastroenterol Nutr. 1994;18:446-452.
55. Koruda MJ, Rolandelli RH, Bliss DZ, Hastings J, Rombeau JL, Settle RG. 74. Iannoli P, Miller JH, Ryan CK, Gu LH, Ziegler TR, Sax HC. Human
Parenteral nutrition supplemented with short-chain fatty acids: effect on growth hormone induces system B transport in short bowel syndrome.
the small-bowel mucosa in normal rats. Am J Clin Nutr. 1990;51:685-689. J Surg Res. 1997;69:150-158.
56. Tappenden KA, McBurney MI. Systemic short-chain fatty acids rapidly 75. Ljungmann K, Grofte T, Kissmeyer-Nielsen P, et al. GH decreases
alter gastrointestinal structure, function, and expression of early response hepatic amino acid degradation after small bowel resection in rats with-
genes. Dig Dis Sci. 1998;43:1526-1536. out enhancing bowel adaptation. Am J Physiol Gastrointest Liver Physiol.
57. Tappenden KA, Drozdowski LA, Thomson AB, McBurney MI. Short- 2000;279:G700-G706.
chain fatty acid-supplemented total parenteral nutrition alters intestinal 76. Ellegard L, Bosaeus I, Nordgren S, Bengtsson BA. Low-dose recombi-
structure, glucose transporter 2 (GLUT2) mRNA and protein, and proglu- nant human growth hormone increases body weight and lean body mass in
cagon mRNA abundance in normal rats. Am J Clin Nutr. 1998;68:118-125. patients with short bowel syndrome. Ann Surg. 1997;225:88-96.
58. Bartholome AL, Albin DM, Baker DH, Holst JJ, Tappenden KA. 77. Seguy D, Vahedi K, Kapel N, Souberbielle JC, Messing B. Low-dose
Supplementation of total parenteral nutrition with butyrate acutely increases growth hormone in adult home parenteral nutrition-dependent short
structural aspects of intestinal adaptation after an 80% jejunoileal resection bowel syndrome patients: a positive study. Gastroenterology. 2003;124:
in neonatal piglets. JPEN J Parenter Enteral Nutr. 2004;28:210-223. 293-302.
59. Tappenden KA, Thomson AB, Wild GE, McBurney MI. Short-chain fatty 78. Byrne TA, Wilmore DW, Iyer K, et al. Growth hormone, glutamine, and
acid-supplemented total parenteral nutrition enhances functional adapta- an optimal diet reduces parenteral nutrition in patients with short bowel
tion to intestinal resection in rats. Gastroenterology. 1997;112:792-802. syndrome: a prospective, randomized, placebo-controlled, double-blind
60. Atia A, Girard-Pipau F, Hebuterne X, et al. Macronutrient absorption char- clinical trial. Ann Surg. 2005;242:655-661.
acteristics in humans with short bowel syndrome and jejunocolonic anasto- 79. Zhou X, Li YX, Li N, Li JS. Glutamine enhances the gut-trophic effect of
mosis: starch is the most important carbohydrate substrate, although pectin growth hormone in rat after massive small bowel resection. J Surg Res.
supplementation may modestly enhance short chain fatty acid production 2001;99:47-52.
and fluid absorption. JPEN J Parenter Enteral Nutr. 2011;35:229-240. 80. Spadoni JM, Aguilar-Nascimento JE, Silva MH, Spadoni-Neto B, Costa
61. Windmueller HG, Spaeth AE. Identification of ketone bodies and gluta- PA, Alessio DM. Effects of the combined use of glutamine and growth
mine as the major respiratory fuels in vivo for postabsorptive rat small hormone in the intestinal adaptation after massive resection of the small
intestine. J Biol Chem. 1978;253:69-76. bowel in rats. Acta Cir Bras. 2005;20:382-389.
62. O’Dwyer ST, Smith RJ, Hwang TL, Wilmore DW. Maintenance of small 81. Vanderhoof JA, Kollman KA, Griffin S, Adrian TE. Growth hormone and
bowel mucosa with glutamine-enriched parenteral nutrition. JPEN J glutamine do not stimulate intestinal adaptation following massive small
Parenter Enteral Nutr. 1989;13:579-585. bowel resection in the rat. J Pediatr Gastroenterol Nutr. 1997;25:327-331.
63. Tamada H, Nezu R, Imamura I, et al. The dipeptide alanyl-glutamine pre- 82. Guo M, Li Y, Wang Z, Wu B, Wang J, Li J. Morphological adaptation in
vents intestinal mucosal atrophy in parenterally fed rats. JPEN J Parenter adult short bowel syndrome undergoing intestinal rehabilitation. J Invest
Enteral Nutr. 1992;16:110-116. Surg. 2013;26:1-5.
64. Tamada H, Nezu R, Matsuo Y, Imamura I, Takagi Y, Okada A. Alanyl 83. Wales PW, Nasr A, de Silva N, Yamada J. Human growth hormone and
glutamine-enriched total parenteral nutrition restores intestinal adaptation glutamine for patients with short bowel syndrome. Cochrane Database
after either proximal or distal massive resection in rats. JPEN J Parenter Syst Rev. 2010;(6):CD006321.
Enteral Nutr. 1993;17:236-242. 84. Zorbtive® (somatropin [rDNA origin]). Full prescribing information.
65. Gu Y, Wu ZH, Xie JX, Jin DY, Zhuo HC. Effects of growth hormone Rockland, MA: EMD Serono, Inc; 2003.
(rhGH) and glutamine supplemented parenteral nutrition on intestinal 85. Jeppesen PB. Pharmacologic options for intestinal rehabilitation in
adaptation in short bowel rats. Clin Nutr. 2001;20:159-166. patients with short bowel syndrome. JPEN J Parenter Enteral Nutr.
66. Neves Jde S, Aguilar-Nascimento JE, Gomes-da-Silva MH, et al. 2014;38(suppl 1):45S-52S.
Glutamine alone or combined with short-chain fatty acids fails to 86. Janssen P, Rotondo A, Mule F, Tack J. Review article: a comparison of
enhance gut adaptation after massive enterectomy in rats. Acta Cir Bras. glucagon-like peptides 1 and 2. Aliment Pharmacol Ther. 2013;37:18-36.
2006;21(suppl 4):2-7. 87. Dahly EM, Gillingham MB, Guo Z, et al. Role of luminal nutrients and
67. Michail S, Mohammadpour H, Park JH, Vanderhoof JA. Effect of glu- endogenous GLP-2 in intestinal adaptation to mid-small bowel resection.
tamine-supplemented elemental diet on mucosal adaptation following Am J Physiol Gastrointest Liver Physiol. 2003;284:G670-G682.
bowel resection in rats. J Pediatr Gastroenterol Nutr. 1995;21:394-398. 88. Ljungmann K, Hartmann B, Kissmeyer-Nielsen P, Flyvbjerg A, Holst
68. Vanderhoof JA, Blackwood DJ, Mohammadpour H, Park JH. Effects of JJ, Laurberg S. Time-dependent intestinal adaptation and GLP-2 altera-
oral supplementation of glutamine on small intestinal mucosal mass fol- tions after small bowel resection in rats. Am J Physiol Gastrointest Liver
lowing resection. J Am Coll Nutr. 1992;11:223-227. Physiol. 2001;281:G779-G785.
69. Yang H, Larsson J, Permert J, Braaf Y, Wiren M. No effect of bolus glu- 89. Martin GR, Wallace LE, Sigalet DL. Glucagon-like peptide-2 induces
tamine supplementation on the postresectional adaptation of small bowel intestinal adaptation in parenterally fed rats with short bowel syndrome.
mucosa in rats receiving chow ad libitum. Dig Surg. 2000;17:256-260. Am J Physiol Gastrointest Liver Physiol. 2004;286:G964-G972.
70. Taylor B, Murphy GM, Dowling RH. Pituitary hormones and the small 90. Sigalet DL, Bawazir O, Martin GR, et al. Glucagon-like peptide-2
bowel: effect of hypophysectomy on intestinal adaptation to small bowel induces a specific pattern of adaptation in remnant jejunum. Dig Dis Sci.
resection in the rat. Eur J Clin Invest. 1979;9:115-127. 2006;51:1557-1566.

Downloaded from pen.sagepub.com at RUTGERS UNIV on April 27, 2015


Tappenden 31S

91. Tsai CH, Hill M, Asa SL, Brubaker PL, Drucker DJ. Intestinal growth- 110. Jeppesen PB, Sanguinetti EL, Buchman A, et al. Teduglutide (ALX-
promoting properties of glucagon-like peptide-2 in mice. Am J Physiol 0600), a dipeptidyl peptidase IV resistant glucagon-like peptide 2 ana-
Endocrinol Metab. 1997;273:E77-E84. logue, improves intestinal function in short bowel syndrome patients. Gut.
92. Jeppesen PB, Hartmann B, Thulesen J, et al. Elevated plasma glucagon- 2005;54:1224-1231.
like peptide 1 and 2 concentrations in ileum resected short bowel patients 111. Lutgens L, Lambin P. Biomarkers for radiation-induced small bowel
with a preserved colon. Gut. 2000;47:370-376. epithelial damage: an emerging role for plasma citrulline. World J
93. Tappenden KA, Edelman J, Joelsson B. Teduglutide enhances structural Gastroenterol. 2007;13:3033-3042.
adaptation of the small intestinal mucosa in patients with short bowel syn- 112. Crenn P, Coudray-Lucas C, Thuillier F, Cynober L, Messing B.

drome. J Clin Gastroenterol. 2013;47:602-607. Postabsorptive plasma citrulline concentration is a marker of absorp-
94. O’Keefe SJ, Jeppesen PB, Gilroy R, Pertkiewicz M, Allard JP, Messing B. tive enterocyte mass and intestinal failure in humans. Gastroenterology.
Safety and efficacy of teduglutide after 52 weeks of treatment in patients 2000;119:1496-1505.
with short bowel syndrome-intestinal failure. Clin Gastroenterol Hepatol. 113. Santarpia L, Catanzano F, Ruoppolo M, et al. Citrulline blood levels as
2013;11:815-823. indicators of residual intestinal absorption in patients with short bowel
95. Jeppesen PB, Pertkiewicz M, Messing B, et al. Teduglutide reduces need syndrome. Ann Nutr Metab. 2008;53:137-142.
for parenteral support among patients with short bowel syndrome with 114. Luo M, Fernandez-Estivariz C, Manatunga AK, et al. Are plasma citrul-
intestinal failure. Gastroenterology. 2012;143:1473-1481. line and glutamine biomarkers of intestinal absorptive function in patients
96. Seidner DL, Schwartz LK, Winkler MF, Jeejeebhoy K, Boullata JI, with short bowel syndrome? JPEN J Parenter Enteral Nutr. 2007;31:1-7.
Tappenden KA. Increased intestinal absorption in the era of teduglutide 115. Papadia C, Sherwood RA, Kalantzis C, et al. Plasma citrulline concentra-
and its impact on management strategies in patients with short bowel tion: a reliable marker of small bowel absorptive capacity independent of
syndrome-associated intestinal failure. JPEN J Parenter Enteral Nutr. intestinal inflammation. Am J Gastroenterol. 2007;102:1474-1482.
2013;37:201-211. 116. Jianfeng G, Weiming Z, Ning L, et al. Serum citrulline is a simple quanti-
97. O’Loughlin E, Winter M, Shun A, Hardin JA, Gall DG. Structural and tative marker for small intestinal enterocytes mass and absorption function
functional adaptation following jejunal resection in rabbits: effect of epi- in short bowel patients. J Surg Res. 2005;127:177-182.
dermal growth factor. Gastroenterology. 1994;107:87-93. 117. Peters JH, Wierdsma NJ, Teerlink T, van Leeuwen PA, Mulder CJ, van
98. Shin CE, Helmrath MA, Falcone RA Jr, et al. Epidermal growth factor Bodegraven AA. Poor diagnostic accuracy of a single fasting plasma
augments adaptation following small bowel resection: optimal dosage, citrulline concentration to assess intestinal energy absorption capacity.
route, and timing of administration. J Surg Res. 1998;77:11-16. Am J Gastroenterol. 2007;102:2814-2819.
99. Erwin CR, Helmrath MA, Shin CE, Falcone RA Jr, Stern LE, Warner BW. 118. Sigalet DL, Martin GR, Poole A. Differential sugar absorption as a marker
Intestinal overexpression of EGF in transgenic mice enhances adaptation for adaptation in short bowel syndrome. J Pediatr Surg. 2000;35:661-664.
after small bowel resection. Am J Physiol. 1999;277:G533-G540. 119. Buchman AL, Scolapio J, Fryer J. AGA technical review on short

100. Chaet MS, Arya G, Ziegler MM, Warner BW. Epidermal growth fac- bowel syndrome and intestinal transplantation. Gastroenterology.
tor enhances intestinal adaptation after massive small bowel resection. 2003;124:1111-1134.
J Pediatr Surg. 1994;29:1035-1038; discussion 1038-1039. 120. Gouttebel MC, Saint Aubert B, Colette C, Astre C, Monnier LH, Joyeux
101. Helmrath MA, Shin CE, Fox JW, Erwin CR, Warner BW. Adaptation H. Intestinal adaptation in patients with short bowel syndrome: measure-
after small bowel resection is attenuated by sialoadenectomy: the role for ment by calcium absorption. Dig Dis Sci. 1989;34:709-715.
endogenous epidermal growth factor. Surgery. 1998;124:848-854. 121. Pironi L, Forbes A, Joly F, et al. Survival of patients identified as can-
102. Dahly EM, Guo Z, Ney DM. IGF-I augments resection-induced muco- didates for intestinal transplantation: a 3-year prospective follow-up.
sal hyperplasia by altering enterocyte kinetics. Am J Physiol Regul Integr Gastroenterology. 2008;135:61-71.
Comp Physiol. 2003;285:R800-R808. 122. Squires RH, Duggan C, Teitelbaum DH, et al. Natural history of pedi-
103. Vanderhoof JA, McCusker RH, Clark R, et al. Truncated and native insu- atric intestinal failure: initial report from the Pediatric Intestinal Failure
linlike growth factor I enhance mucosal adaptation after jejunoileal resec- Consortium. J Pediatr. 2012;161:723-728 e722.
tion. Gastroenterology. 1992;102:1949-1956. 123. Spencer AU, Neaga A, West B, et al. Pediatric short bowel syndrome:
104. Gillingham MB, Dahly EM, Carey HV, Clark MD, Kritsch KR, Ney redefining predictors of success. Ann Surg. 2005;242:403-409; discussion
DM. Differential jejunal and colonic adaptation due to resection and 409-412.
IGF-I in parenterally fed rats. Am J Physiol Gastrointest Liver Physiol. 124. Byrne TA, Persinger RL, Young LS, Ziegler TR, Wilmore DW. A new treat-
2000;278:G700-G709. ment for patients with short-bowel syndrome: growth hormone, glutamine,
105. Katz MS, Thatch KA, Schwartz MZ. Chronology of the effect of massive and a modified diet. Ann Surg. 1995;222:243-254; discussion 254-255.
small bowel resection and hepatocyte growth factor (HGF) on intestinal 125. Byrne TA, Cox S, Karimbakas M, et al. Bowel rehabilitation: an alterna-
adaptation. J Surg Res. 2011;171:399-403. tive to long-term parenteral nutrition and intestinal transplantation for some
106. Kato Y, Yu D, Schwartz MZ. Enhancement of intestinal adaptation by patients with short bowel syndrome. Transplant Proc. 2002;34:887-890.
hepatocyte growth factor. J Pediatr Surg. 1998;33:235-239. 126. Jeppesen PB, Gilroy R, Pertkiewicz M, Allard JP, Messing B, O’Keefe
107. Sigalet DL, Martin GR, Butzner JD, Buret A, Meddings JB. A pilot study SJ. Randomised placebo-controlled trial of teduglutide in reducing paren-
of the use of epidermal growth factor in pediatric short bowel syndrome. teral nutrition and/or intravenous fluid requirements in patients with short
J Pediatr Surg. 2005;40:763-768. bowel syndrome. Gut. 2011;60:902-914.
108. Guo M, Li Y, Wang Z, Wu B, Wang J, Li J. Magnifying endoscopy for 127. Schwartz LK, O’Keefe SJ, Jeppesen PB, Pertkiewicz M, Youssef N,
monitoring intestinal adaptation: a case report. Surg Laparosc Endosc Fujioka K. Long-term safety and efficacy of teduglutide for the treatment
Percutan Tech. 2011;21:e187-e189. of intestinal failure associated with short bowel syndrome: final results of
109. Crenn P, Morin MC, Joly F, Penven S, Thuillier F, Messing B. Net diges- the STEPS-2 study, a 2-year, multicenter, open-label clinical trial. Paper
tive absorption and adaptive hyperphagia in adult short bowel patients. presented at: American College of Gastroenterology Annual Scientific
Gut. 2004;53:1279-1286. Meeting; 2013; San Diego, CA.

Downloaded from pen.sagepub.com at RUTGERS UNIV on April 27, 2015

S-ar putea să vă placă și