Você está na página 1de 7

European Journal of Internal Medicine 23 (2012) 499–505

Contents lists available at SciVerse ScienceDirect

European Journal of Internal Medicine


journal homepage: www.elsevier.com/locate/ejim

Review article

Diabetic gastrointestinal autonomic neuropathy: Current status and new


achievements for everyday clinical practice
A. Gatopoulou a,⁎, N. Papanas a, b, E. Maltezos a, b
a
Second Department of Internal Medicine, Democritus University of Thrace, Greece
b
Outpatient Clinic of Obesity, Diabetes and Metabolism of the Second Department of Internal Medicine, Democritus University of Thrace, Greece

a r t i c l e i n f o a b s t r a c t

Article history: Gastrointestinal symptoms occur frequently among patients with diabetes mellitus and are associated with
Received 13 February 2012 considerable morbidity. Diabetic gastrointestinal autonomic neuropathy represents a complex disorder with
Received in revised form 27 February 2012 multifactorial pathogenesis, which is still not well understood. It appears to involve a spectrum of metabolic
Accepted 1 March 2012
and cellular changes that affect gastrointestinal motor and sensory control. It may affect any organ in the
Available online 28 March 2012
digestive system. Clinical manifestations are often underestimated, and therefore autonomic neuropathy
Keywords:
should be suspected in all diabetic patients with unexplained gastrointestinal symptoms. Advances in technol-
Diabetes mellitus ogy have now enabled assessment of gastrointestinal motor function. Moreover, novel pharmacological
Diagnosis approaches, along with endoscopic and surgical treatment options, contribute to improved outcomes. This
Autonomic neuropathy review summarises the progress achieved in diabetic gastrointestinal autonomic neuropathy during the last
Gastrointestinal system years, focusing on clinical issues of practical importance to the everyday clinician.
Gastroparesis © 2012 European Federation of Internal Medicine. Published by Elsevier B.V. All rights reserved.
Treatment

1. Introduction motility. Symptoms appear to be more prevalent among women [6].


There are miscellaneous oesophageal motility disorders in diabetes,
Gastrointestinal symptoms occur more commonly in diabetic although these may be clinically silent, such as hypotensive lower
patients than in the general population and lead to a significant impair- oesophageal construction, decreased amplitude of constructions and
ment in the quality of life [1]. As high as 75% of patients with diabetes simultaneous prolonged aperistaltic constructions in the body of the
may experience post-prandial fullness with nausea, bloating, abdomi- oesophagus [7]. Usually, patients report heartburn, regurgitation
nal pain, diarrhoea and/or constipation [1]. Such complaints represent and dysphagia, suggesting acid reflux or pill-induced oesophageal
a major cause of morbidity and have a negative impact on healthcare erosions. Candida should be suspected in cases with odynophagia [7].
costs in diabetes [1,2]. However, they are often underestimated [2]. The most important manifestation of gastrointestinal autonomic
Gastrointestinal autonomic neuropathy may involve any organ in neuropathy is gastroparesis. This is a symptomatic chronic disorder of
the digestive system, such as the oesophagus, stomach, gallbladder, the stomach characterised by diminished gastric emptying after
pancreas, small and large intestine. Both abnormalities of gastric exclusion of obstruction [8–10]. It is usually diagnosed in patients
function (sensor and motor modality) and impairment of gastrointes- presenting with intense and prolonged symptoms of nausea, vomiting,
tinal hormonal secretion have already been described [3]. The under- early satiety and epigastric pain. Its prevalence has been estimated at
lying pathogenic mechanisms are still incompletely understood and 30–50% of patients with long-standing diabetes [9,10]. Both chronic
the best medical treatment remains to be defined [4,5]. and acute hyperglycaemia may reduce gastric emptying [9–11].
This review summarises the progress achieved in diabetic gastro- Patients with gastroparesis, generally, show delayed gastric emptying
intestinal autonomic neuropathy during the last years, focusing on (28%–65%), but accelerated gastric emptying may also be the
clinical issues of practical importance to the busy everyday clinician. case [9–11]. Poor post-prandial glucose control and post-prandial
upper abdominal symptoms usually occur in gastroparesis, both in
2. Epidemiology delayed and in accelerated gastric emptying [8]. Young females of child-
bearing age are mostly affected [12]. The reasons for this predilection
Gastrointestinal symptoms, malnutritional status and inadequate are unclear, but may be due, at least partly, to gender differences in
glycaemic control may be associated with impaired gastrointestinal neuronal nitric oxide synthase [nNOS] dimerisation [9].
Likewise, abnormalities in oesophageal motility are detected in al-
⁎ Corresponding author at: 2nd, Odysseos Rd, Alexandroupolis, Greece. Tel.: + 30
most 50% of diabetic patients with autonomic neuropathy [13]. Re-
2551074130. cent studies have confirmed that these perturbations tend to
E-mail address: gatop@otenet.gr (A. Gatopoulou). aggravate with long diabetes duration [14]. Gastro-oesophageal

0953-6205/$ – see front matter © 2012 European Federation of Internal Medicine. Published by Elsevier B.V. All rights reserved.
doi:10.1016/j.ejim.2012.03.001
500 A. Gatopoulou et al. / European Journal of Internal Medicine 23 (2012) 499–505

reflux disease is seen more frequently in diabetic subjects and is asso- typical gastrointestinal symptoms, as evidenced by the high preva-
ciated with neuropathy and poor glycaemic control [15]. lence of entirely asymptomatic or only poorly symptomatic severe
Furthermore, lithogenic bile composition and stasis of bile in the peptic ulcer disease in diabetes [8].
gallbladder may contribute to stone formation in diabetes, explaining Finally, it is now known that autonomic neuropathy may develop
the relatively higher frequency of choledocholithiasis. Impaired gall- early and is, indeed, demonstrable in some patients with pre-diabetes
bladder contraction may, possibly, be explained by a decreased sensi- [28], but there is little information on gastrointestinal neuropathy in
tivity of gallbladder smooth muscles to plasma cholecystokinin and this group.
by a reduction of cholecystokinin receptors on gallbladder wall [16].
Moreover, a mutation in the gene encoding the expression of chole- 3. Pathogenesis
cystokinin A (CCK-A) receptor on the gallbladder smooth muscle in
diabetic patients with gallstones may be the cause of decreased gall- The pathogenesis of gastrointestinal autonomic neuropathy is
bladder motility [17]. Insulin resistance per se has also been implicat- multifaceted [4]. The main pathogenic mechanisms of both upper
ed in gallbladder dysmotility, leading to gallstone formation or and lower gastrointestinal dysfunction include abnormalities of
acalculus cholecystitis [18]. motor function, visceral hypersensitivity, altered secretion of gastro-
Faecal incontinence, particularly nocturnal, is another unpleasant intestinal hormones, inflammatory state, autonomic dysfunction and
symptom. Overt steatorrhoea is noted only in a minority of patients genetic predisposition [5] (Table 1). All these are enhanced in the
[19]. Autonomic dysfunction has been held responsible for the im- presence of acute or chronic hyperglycaemia [5]. The latter exerts a
pairment of normal internal anal relaxation [19]. Interestingly, gly- particularly noxious effect on the interstitial Cajal cells, ultimately
caemic excursions may directly influence sphincter function. Indeed, leading to impaired gastric motility [29].
acute hyperglycaemia inhibits external anal sphincter function and Both type 1 and type 2 diabetes mellitus are affected [4,5]. Generally,
decreases rectal compliance, thereby, possibly, increasing the risk of patients with the former appear to have a relatively higher risk. Never-
faecal incontinence [20]. theless, the potential differences in the pathogenic mechanisms be-
Constipation is a further common complaint, affecting approxi- tween the two diabetes types are still unclear and most studies do not
mately 60% of patients with long-standing diabetes [21]. Severe distinguish between these two types [4,5]. Arguably, the relatively
constipation with megacolon or colonic intestinal pseudo-obstruction higher risk in type 1 diabetes may be attributed to the longer duration
may, albeit rarely, occur as well. Complications of severe constipation of hyperglycaemia, which, in turn, aggravates gastric emptying and
such as stercoral ulcer, perforation and overflow diarrhoea are infre- intestinal motility, but this is far from proven [4,9].
quently encountered [22]. Based on studies with radio-opaque Gastroparesis is usually considered the most significant manifesta-
markers to measure colonic segmental transit time, there is evidence tion of gastrointestinal autonomic neuropathy [5]. An examination of
for slow transit constipation in the diabetic population [23]. However, antral biopsies from patients with gastroparesis found that the total
no difference has been observed between subjects with and those number of nerve fibres was significantly reduced, in comparison to
without cardiovascular autonomic neuropathy [24]. non-diabetic controls [30]. In diabetic subjects, the myenteric plexus
Moreover, diarrhoea is a frequent symptom and may be reported was also more frequently infiltrated with lymphocytes [30]. In a further
by up to 20% of patients. It reflects perturbations in small bowel work studying gastric mucosal biopsies obtained during endoscopy,
transit, while bacterial overgrowth may also be a contributory factor reduced density and abnormal morphology of gastric mucosal nerves
[21]. It occurs at any time of the day, but it is most typically nocturnal. were detected in diabetic candidates for pancreas transplantation as
Characteristically, it is seen in patients with poorly controlled insulin- compared to control subjects [31].
dependent diabetes mellitus exhibiting peripheral and autonomic Ghrelin, a hormone secreted by gastric entero-endocrine cells, has
neuropathy [25]. been shown to enhance gastric emptying in diabetic gastroparesis,
Unexplained upper abdominal pain is another complaint that may suggesting that it may exert an additional contributory role [32].
occur in diabetic patients with neuropathy, sometimes along with an- Τhe nitric oxide (NO) produced by nNOS neurons (neuronal nitric
orexia and weight loss [26]. According to one hypothesis, diabetic oxide synthese neurons) is a major non-adrenergic, non-cholinergic
radiculopathy or neuropathy of thoracic nerve roots could be the inhibitory neurotransmitter, which mediates the smooth muscle
cause [26]. Of note, depression rather than glycaemic control has relaxation in the gastrointestinal tract and, hence, is important in
been linked with development of gastrointestinal symptoms in diabe- gut motility [33]. Loss of NO-mediated function in the stomach can
tes, suggesting that emotional status is relevant to [27]. Another work result in delayed gastric emptying or changes in gastric accommoda-
has shown that visceral neuropathy may reduce the perception of tion [34]. Loss of nNOS expression in diabetes may represent inhibition

Table 1
Summary of affected organs, common symptoms and pathogenic mechanisms.

Organ Abnormality Symptoms Pathogenesis⁎

Oesophagus Dysmotility, candidiasis Dysphagia, reflux odynophagia Multifactorial


Stomach Gastroparesis Nausea, vomiting, gastric outlet obstruction, bezoars • Abnormalities of gastrointestinal motor function
(i.e. ICC dysfunction, nerve density, loss of nNOS)
Small intestine Dysmotility, impaired fluid reabsorption Bacterial overgrowth, malabsorption diarrhoea • Visceral hypersensitivity
(central and peripheral mechanisms)
Large intestine Dysmotility, ischaemia Constipation, megacolon faecal incontinence, • Altered secretion of gastrointestinal hormones
ischaemic colitis, bowel infarction (i.e. ghrelin)
Gallbladder Dysmotility, gallstones, Biliary obstruction, sepsis • Autonomic dysfunction
cholelithiasis, choledocholithiasis • Oxidative stress (haem-1 oxygenase)
• Genetic predisposition
(Ano-1 expression, CCK-A receptor)
• Autoimmune (autoantibodies)
• Inflammation
• Growth factors (IGF-1)
• Hyperglycaemia

ICC: interstitial Cajal cells; CCK: cholecystokinin; IGF: insulin-like growth factor; nNOS: neuronal nitric oxide synthase.
⁎ Common pathogenic mechanisms of gastrointestinal autonomic neuropathy.
A. Gatopoulou et al. / European Journal of Internal Medicine 23 (2012) 499–505 501

of nNOS by advanced glycation end-products, which can bind to 4. Diagnosis


myenteric neurons and inhibit nNOS [9,34].
In the muscle layers, interstitial cells of Cajal (ICC) selectively Gastrointestinal autonomic neuropathy should be suspected in all
express Ano-1 [35]. The latter is a calcium-activated chloride channel diabetic patients with unexplained unpleasant gastrointestinal symp-
and participates in control mechanisms of smooth muscle contrac- toms. Its diagnosis is continuously being improved, as presented in
tion. Changes in Ano-1 expression may alter the electroplysiological Table 2.
properties of the channel [35]. One study has recently found impaired
splicing of Ano-1 gene in diabetic gastroparesis. This defect was 4.1. Gastroparesis
accountable for the diminution of chloride currents and the delay in
contractility, as compared with non-diabetic controls [36]. It was Clinical manifestations of diabetic gastrointestinal neuropathy
inferred that altered Ano-1 expression in ICC might be implicated in may overlap with symptoms occurring in organic disorders, notably
the pathogenesis of diabetic gastroparesis [36]. peptic ulcers, gastric or bowel obstruction, cancer and biliary disease.
Diabetes is characterised by high oxidative stress, which has recent- Therefore, diagnosis should be reserved for patients with symptoms
ly been recognised to exert a decisive role in the pathogenesis of and normal gastrointestinal imaging. Physical examination may be
gastrointestinal complications [37–39]. Recent data suggests that loss entirely normal, as well [51]. Nonetheless, autonomic neuropathy
of mechanisms normally conferring protection from increased oxida- might be suspected by an exaggerated postural drop in systolic and
tive stress, such as upregulation of macrophage haeme oxygenase-1 diastolic blood pressure and/or by a loss of normal sinus arrhythmia
(an important natural cytoprotective molecule) is of paramount im- on deep breathing [52]. Endoscopy and hepatobiliary ultrasound
portance for the development of gastroparesis [39,40]. One study need to be always performed to rule out other diagnoses. Proximal
demonstrated that inhibition of haeme oxygenase-1 activity in mice small bowel obstruction should be excluded by double-contrast bari-
caused a loss of receptor tyrosine kinase expression, thereby reducing um study or computed tomography [52].
gastric emptying and inducing diabetic gastroparesis [40]. A definitive diagnosis is based on objective measurements of
Recently, the role of autoimmune processes in the pathogenesis of gastric emptying [52]. Scintigraphy is the most common and widely
gastrointestinal neuromuscular disorders has attracted considerable available method for measuring gastric emptying and is considered
interest. It is now established that auto-antibodies play a pivotal the gold standard against which newer diagnostic tools are compared
role in the development of gastric dysmotility [9,41]. However, [9,53]. The standard technique involves scintigraphic determination
there is scant evidence supporting the autoimmune pathogenesis of of emptying of a solid meal. The American Neurogastroenterology
diabetic gastroenteropathies [42]. and Motility Society recommends the use of a 99 mTc (Technetium)
Diabetes leads to an imbalance between inhibitory and excitatory sulphur colloid-labelled egg, two slices of bread, strawberry jam and
enteric neuropeptide rations, which can directly cause altered gut water (255 Kcal). All medications that may affect gastric motility
motility. A further putative mechanism involves the interaction of should be stopped at least 48 h prior to the test. During the examina-
neuropeptides from enteric nervous system with immune cells acti- tion, blood glucose should be b15 mmol/l (270 mg/dl). The test is
vating pro-inflammatory cytokine production, ultimately resulting based on determining the percentage of food remaining in the stom-
in inflammation and neuronal loss. Thus, inflammation may cause ach during the examination: normal values are 37%–90% at 1 h, 30–
nerve damage leading to gastrointestinal motility disorders [42–44]. 60% at 2 h and 0–10% at 4 h [53,54].
Furthermore, there is strong evidence to support a role for various The gastric emptying breath test using 13C, a stable, non-
growth factors in the pathogenesis of gastrointestinal autonomic neu- radioactive isotope, has been validated in subjects with diabetes.
ropathy. Recent studies have demonstrated that myopathy may play After an overnight fast, the patient ingests a meal with 13C-octanoic
a role in diabetic gastroparesis. A reduction in insulin/insulin-growth acid, and breath samples are obtained before meal and every 15 min
factor 1 (IGF-I) signalling in diabetes causes ICC depletion, subsequent to 3 h after the meal and then every 30 min up to 5 h. The ratio
13
stem cell factor depletion and resultant smooth muscle atrophy CO2/ 12CO2 in the samples is measured by spectrometry. The result
[45,46]. In this context, insulin is of paramount importance. There is a is expressed as percentage of 13C recovery per hour (% dose/h) and
deficiency in insulin secretion and/or sensitivity, depending on type as a cumulative value over 5 h, providing a reliable measure of gastric
of diabetes. In both types, however, insulin-growth factor I (IGF-I) is emptying [9].
reduced, resulting in smooth muscle atrophy, which, in turn, contrib- A novel variation of the 13C-octanoic acid technique is the 13C-
utes to impaired gastrointestinal motility [24,35]. The administration Spiroulina platensis blue-green algae breath test. This is a promising
of growth factors, hormones and antioxidants has been extensively new method to evaluate gastric emptying non-invasively and without
studied. Impressively, IGF-1 may reverse experimental diabetic neu- radiation, but it is not yet approved by the Food and Drug Administra-
ropathy, providing further proof of concept for its pathogenic role [47]. tion (FDA) or the European Medicine Agency (EMEA) [55]. A recent
Moreover, impaired vagal activity has been implicated in the path- study has validated the test using breath samples at 45, 150, and
ogenesis of gastroparesis. Vagal nerve dysfunction, as manifested by a 180 min after digesting a shelf-stable 238 Kcal meal consisting of
reduced response to sham feeding-induced serum pancreatic poly-
peptide and ghrelin, is seen in patients with diabetic gastroparesis
Table 2
[48]. Both afferent and efferent innervation of the gastrointestinal Progress achieved in the diagnosis of gastrointestinal autonomic neuropathy.
tract is adversely affected in diabetes, more so in the presence of
gastroparesis [49]. Oesophageal function testing: impedance, Bravo pH capsule monitoring,
high-resolution manometry
In addition, visceral hypersensitivity is an important factor underly-
Standardisation of protocol for scintigraphy and 13C octanoic acid breath test
ing gastrointestinal discomfort [5]. In diabetic gastrointestinal autonom- Use of 13C Spiroulina platensis breath test: a promising newer
ic neuropathy, there is evidence of altered central processing of visceral non-invasive method
stimuli. Recently, one study has evaluated the brain processing of pain- Smart Pill: a wireless capsule to measure pressure pH, gut transit time
Magnetic Resonance Imaging: validation needed
ful visceral stimuli in type 1 diabetic patients with autonomic neuropa-
3-D Ultrasonography to measure gastric accommodation
thy and gastrointestinal symptoms in comparison to healthy volunteers Single photon emission computed tomography to measure gastric volume
[50]. Diabetic patients exhibited higher sensory thresholds, as well as a and accommodation
different pattern of electrical activity in many cerebral regions (anterior GCSI-DD (Gastroparesis Cardinal Symptom Index Daily Diary):
cingulate cortex, posterior insula and medial frontal gyrus), indicating a modified new questionnaire
Mucosal biopsies during endoscopy: the future diagnostic method?
altered central neuronal pain perception processing [50].
502 A. Gatopoulou et al. / European Journal of Internal Medicine 23 (2012) 499–505

freeze-dried egg mix, saltine crackers and 100 mg 13C-Spiroulina measured by a non-invasive, radio-opaque marker method. Tradi-
platensis. Sensitivity and specificity for the diagnosis of delayed tionally, barostat and manometry can be used to study anorectal
gastric emptying were 89% and 80%, respectively [56]. and sensory function [19,20,23].
The Smart Pill (Smart Pill Corporation, Buffalo, NY, USA) is a
further new diagnostic adjunct, which has received FDA approval. It 4.4. Gastrointestinal questionnaires
is a non-digestible capsule measuring gastric emptying time, as well
as transit time in the small and large intestine [55]. The change in Several questionnaires have proven useful to quantify gastrointesti-
pH between the distal stomach and the proximal small intestine nal symptoms. These include the Diabetes Bowel Symptom Question-
documents capsule movement along the gastrointestinal tract and, naire (DBSQ), a measure of gastrointestinal symptoms and glycaemic
therefore, provides a measure of the time from ingestion to arrival control in patients with diabetes [62]; the Gastroparesis Cardinal
in the small intestine [56]. This is usually associated with antral Symptom index (GCSI) consisting of nine commonly reported symp-
phasic contractions at the maximal frequency of the migrating toms [63]; and the new modified GCSI-DD (Gastroparesis Cardinal
motor complex [57]. The wireless capsule acquires data continuously Symptom index-daily diary). One advantage of the latter is that it
for up to 5 days, and this permits calculation of small bowel, colon may give expression to daily symptom variability [4,9].
and whole gut transit. An “event” button is used to denote sleep,
meal, occurrence of symptoms etc. [55]. 5. Treatment
New imaging modalities include magnetic resonance imaging
(MRI), single photon emission computed tomography imaging Management of gastrointestinal autonomic neuropathy is, gener-
(SPECT) and ultrasonography. At the moment, MRI has not been ally, suboptimal and needs to be improved. Therapeutic progress
validated in the same degree as scintigraphy. Patients are typically achieved is summarised in Table 3. Ideally, it should target both
examined over 120 min and both diameters of proximal and distal symptom relief and glycaemic control [4].
stomach, as well as the motor function, can be measured. It remains
a promising technology but is not widely validated [55,58]. SPECT
5.1. Nutritional status
can measure total gastric volume during fasting and during the first
30 min following a liquid nutrient meal of 300 Kcal and demonstrates
It is important to assess patients' nutritional status, especially in
the effects of disease on gastric volume and gastric accommodation
cases of gastroparesis and diarrhoea, and to recognise any dehydra-
[59]. Three-dimensional ultrasonography or, most recently, 3D
tion, weight loss, and electrolyte imbalance. In malnourished patients
reconstruction of images acquired by ordinary ultrasonography
with over 5% weight loss over 3 months, enteral feeding should be
assisted by magnetic scan-head tracking, can evaluate gastric func-
considered [9]. A satisfactory response to a trial of nasogastric or
tion non-invasively providing information of intragastric meal distri-
nasojejunal feeding is an indication to attempt direct feeding through
bution and gastric volume. But it is time consuming and harder in
a percutaneous endoscopically placed gastrostomy [PEG] or an endo-
obese patients [9,60].
scopically/surgically inserted jejunostomy [52]. Total parenteral
Andro-duodenal manometry and the barostat represent two
nutrition should be reserved only for patients who fail enteral feeding
further diagnostic improvements [52,55]. The former measures
[9]. Usually, nutritional consultation and dietary manipulation (low
contractions in distal stomach and duodenum and may distinguish
fat/fibre, small portions meals) manage to improve symptoms [64].
neuropathic from muscle disease. It measures phasic not tonic
contractions in the distal stomach and duodenum, but it is invasive
and, currently, not widely available [55]. The latter measures the 5.2. Gastroparesis
tone of hollow organs and estimates changes in the tone by changing
the volume of the air in an intra-gastric balloon. It is rarely performed Currently available pharmacological therapy for patients with
in clinical practice [52,55]. symptomatic gastroparesis mainly includes prokinetic drugs such as
Finally, the rhythm of the gastric pacemaker can be measured by metoclopramide, domperidone, erythromycin [65]. In patients with
cutaneous electrogastrography (EGG) [55]. This provides information oesophageal dysfunction and reflux disease, proton pump inhibitors
about the gastric myoelectric frequency and the amplitude of the ECG should be prescribed, as well [4]. Metoclopramide and domperidone
signal in the normal or abnormal frequency ranges. At the moment, are dopamine D2 receptors antagonists, which accelerate gastric
however, interpretation of results is rather controversial [55]. emptying and improve symptoms but have central nervous system
side effects [66]. Erythromycin is a motilin receptor agonist that also
4.2. Oesophageal dysfunction accelerates gastric emptying, but it is associated with rapid tachyphy-
laxis [67]. Antiemetics (phenothiazines and antihistamine agents)
Τhe assessment of oesophageal motility has relied on conventional have also been used. They relieve acute nausea but their efficacy for
manometry for many years. Recently, there have been two major chronic nausea is unclear [56]. One study has recently suggested
developments in this field: the wireless Bravo pH capsule, which that continuous subcutaneous insulin infusion therapy in patients
allows catheter-free monitoring; and impedance-pH measurement,
a catheter-based technique that enables detection of acidic and non- Table 3
acidic reflux. Two new procedures have also recently become avail- Therapeutic innovations for gastrointestinal autonomic neuropathy.
able to assess esophageal motility: high-resolution manometry,
Novel prokinetic agents: prucalopride, ATI-7505 and velusetrag
which uses many closely, spaced pressure sensors and provides spa-
Novel 5-HT-4 receptor agonists; acotiamide
tiotemporal plots of esophageal pressure changes; and impedance Azithromycin: an alternative to erythromycin
manometry, a test that directly measures bolus transit and provides Motilin agonist: GSK962040
conventional manometric data [61]. Novel ghrelin agonist: TZP-101
Iberocast: an herbal mixture
Enterra device with gastric electrical stimulation electrode
4.3. Diarrhoea/constipation endoscopically implanted
Two-channel gastric electrical stimulation
Similarly to upper gastrointestinal dysfunction, patients should Exenatide: a X glucagon-like peptide 1 (GLP-1) agonist to reduce accelerated
undergo endoscopic examinations, ultrasound, or computed tomog- gastric emptying
Stem cell-based therapies or cellular target agents such as haemin: future therapy?
raphy to exclude other diagnoses. Colonic transit time may be
A. Gatopoulou et al. / European Journal of Internal Medicine 23 (2012) 499–505 503

with diabetic gastroparesis improved glycaemic control and de- 5.5. Constipation
creased the number of hospitalisation days [68].
To further improve symptom relief, several potential novel proki- Management of constipation involves traditional laxatives and is
netic agents are now being developed: prucalopride, ATI-7505 and still symptomatic aiming at symptom relief [4]. Lubiprostone, a
velusetrag are novel types of 5-HT-4 receptor agonist; acotiamide is novel drug for constipation, has recently become available in the US.
a M1 and M2 muscarinic antagonist [5,56,69]. Azithromycin is anoth- It is approved for the treatment of chronic idiopathic constipation,
er macrolide antibiotic providing an alternative to erythromycin for and acts by activating CIC-2 chloride channels and, thereby, modify-
gastroprokinetic applications [5]. GSK962040 is a small-molecule ing intestinal transit time. This agent may prove useful for the future
motilin agonist nowadays being investigated for its potential efficacy treatment of constipation from gastrointestinal autonomic neuropa-
[70]. TZP-101 is a novel ghrelin agonist under development for the thy in diabetes [84].
treatment of gastroparesis and post-operative ileus [71]. Moreover,
iberocast, an herbal mixture, may have beneficial effects on functional 5.6. Pain management
dyspepsia and gastroparesis but the underlying mechanism of action
is unclear [72]. Botulinum injection of the pylorus at endoscopy failed Analgesia is often challenging. Medications such as tricyclic and tet-
to show significant symptomatic benefit [73]. racyclic antidepressants, gabapentin and pregabalin can be used, but
In refractory gastroparesis, gastric electrical stimulation can be opiates should be provided sparingly and only in refractory cases [9].
considered as rescue therapy. The Enterra device (Medtronic, Inc) Medications commonly used for chronic pain such as tramadol are
has been clinically available but its efficacy is controversial [74]. In usually effective, but there is currently limited experience with their
one recent study, a gastric electrical stimulation electrode was endo- use in gastroparesis [9,85].
scopically implanted in patients with gastroparesis, but further stud-
ies are needed to validate this approach [75]. A meta-analysis has 5.7. Emerging stem cell therapies
recently found that patients with diabetic gastroparesis seem to be
more responsive to gastric electrical stimulation both subjectively Stem cell-based therapies have been proposed to improve gastroin-
and objectively compared to patients with idiopathic or postsurgical testinal motility [86,87]. This approach is based on the ability of mesen-
gastroparesis [76]. Another study evaluated the effects of two- chymal stem cells to differentiate into several cell types including
channel gastric pacing in patients with gastroparesis and demonstrat- gastrointestinal smooth muscle cells. Future studies are now desirable
ed that two channel gastric pacing seem to be a safe treatment to ascertain whether this approach can restore normal digestive func-
approach, which is able to normalise and enhance slow wave activity, tion and reverse gastrointestinal complications in diabetic patients
as well as accelerating gastric emptying in patients with diabetic [86,87].
gastroparesis [77].
In extremely severe cases, gastrectomy may be performed. This 6. Conclusions
should be only attempted in highly selected patients after weighting
the risk of malnutrition and post-operative weight loss against antic- Gastrointestinal autonomic neuropathy represents a major factor of
ipated symptom relief [5]. morbidity in diabetic patients and has a profound negative impact on
In cases of accelerated emptying, dietary manoeuvres may be nec- the quality of life [1,4]. Importantly, this complication can easily go
essary [78]. These mainly include avoidance of consuming fluids unrecognised for a long time [1–8]. Thus, a high index of suspicion is
during and 30 min after meals and addition of fibre supplements. required in the case of persistent gastrointestinal symptoms, especially
Treatment with the GLP-1 agonist exenatide also seems to delay in patients with long-standing diabetes and other chronic complica-
gastric emptying and reduce post-prandial glycaemia [79]. tions [1–4,7]. Diagnosis will be confirmed by careful clinical examina-
tion, exclusion of other causes and use of specific tests, both
5.3. Oesophageal dysfunction established and emerging [1–4].
The pathogenesis of gastrointestinal autonomic neuropathy is
Conventional therapy with proton pump inhibitors is suggested to complex and multifactorial. An array of metabolic and cellular
manage oesophageal dysmotility especially associated with reflux. changes, along with hyperglycaemia, increased oxidative stress and,
Patients are also advised to drink fluids immediately after taking arguably, autoimmune and genetic factors appear to be involved
medications in order to avoid pill-induced oesophagitis [4]. [35]. Despite the great number of putative pathophysiological mech-
anisms, the exact contribution of each of these remains a matter of
5.4. Diarrhoea controversy [44].
New technologies and advances in diagnosis are being applied in
Given that bacterial overgrowth is found in almost 40% of diabetic diabetic subjects with gastrointestinal autonomic neuropathy
patients with diarrhoea [80], therapy should include intermittent (Table 1) [56]. For instance, the SmartPill is a new promising diagnos-
administration of selective antibiotics [81]. Rifaximin is the most tic tool. This is a wireless capsule that records luminal pH,
extensively studied antibiotic. This agent improves symptoms in temperature and pressure during transit through the gastrointestinal
33%–92% and eradicates bacterial overgrowth in up to 80% of patients tract [5]. Early diagnosis with gastric mucosal biopsies performed
[82]. In the event of severe diarrhoea, somatostatin analogues may be during endoscopy is another diagnostic innovation [31].
required [4,83]. In cases with diarrhoea, constipation and faecal in- New therapeutic strategies and emerging pharmacological agents
continence, it is mandatory to exclude other common causes, notably are being evaluated [5,9,56]. These mainly include muscarinic receptor
celiac or pancreatic disease [83]. Loperamide may prove helpful in antagonists, ghrelin receptors, motilin and 5-HT4 receptor agonists
faecal incontinence [83]. Alosentron, a selective 5-HT3 antagonist, [5,56]. Gastric electrical stimulation with endoscopically implanted
was approved in 2000 for the treatment of diarrhoea-predominant electrodes can be considered in refractory cases [75,76]. Τhe potential
irritable bowel syndrome in females with no experience in diabetic use of stem cell-based therapies or new drugs with cellular target
patients. It is thought to act by reducing inappropriate colonic motil- such as haemin may improve treatment in the future [35,86].
ity and colorectal hypersensitivity. A small incidence of reversible Meanwhile, optimal patient management requires multidisciplin-
ischaemic colitis led to an initial withdrawal of alosendron, while ary approach and should focus on the relief of gastrointestinal symp-
additional experience was more positive, allowing its re-introduction toms, as well as improvement in nutritional status and glycaemic
but limiting its use to sever cases [84]. control [4]. This goal should be pursued by the everyday clinician.
504 A. Gatopoulou et al. / European Journal of Internal Medicine 23 (2012) 499–505

Learning points [18] Nakeeb A, Comuzzie AG, Al Azzawi H, Sonnenberg GE, Kisselbach AH, Pitt HA.
Insulin resistance causes human gallbladder dysmotility. J Gastrointest Surg
2006;10:940–8.
• Gastrointestinal autonomic neuropathy represents a major factor of [19] Bharucha AE. Fecal incontinence. Gastroenterology 2003;124:1672–85.
morbidity in diabetic patients and should be suspected in all sub- [20] Russo A, Botten R, Kong MF, Chapman IM, Fraser RJ, Horowitz M, et al. Effects of
acute hyperglycaemia on anorectal motor and sensory function in diabetes melli-
jects with unexplained unpleasant gastrointestinal symptoms in tus. Diabet Med 2004;21:176–82.
everyday practice. [21] Phillips LK, Rayner CK, Jones KL, Horowitz M. An update on autonomic neuropa-
• The pathogenesis of gastrointestinal autonomic neuropathy is com- thy affecting the gastrointestinal tract. Curr Diab Rep 2006;6:417–23.
[22] Chandran M, Chu NV, Edelman SV. Gastrointestinal disturbances in diabetes. Curr
plex, multifactorial and remains largely unclear. Diab Rep 2003;3:43–8.
• New technologies and advances in diagnosis are being applied to [23] Rossol S. Constipation in patients with diabetes mellitus. MMW Forschr Med
improve diagnosis, such as the Smartpill, 3D-ultrasonography, 2007;149:39–42.
[24] Jung HK, Kim DY, Moon IH, Hong YS. Colonic transit time in diabetic patients—
MRI, SPECT, and 13C Spiroulina platensis breath test.
comparison with healthy subjects and the effect of autonomic neuropathy. Yonsei
• Emerging pharmacological agents and strategies are being evaluat- Med J 2003;44:265–72.
ed, as well. The most important ones include muscarinic receptor [25] Lysy J, Israeli E, Goldin E. The prevalence of chronic diarrhea among diabetic
agonists, ghrelin receptor agonists, motilin and 5-HT4 receptor ago- patients. Am J Gastroenterol 1999;94:2165–70.
[26] O'Connor RC, Andary MT, Russo RB, DeLano M. Thoracic radiculopathy. Phys Med
nists, and gastric electrical stimulation with endoscopically Rehabil Clin N Am 2002;13:623–44.
implanted electrodes for refractory cases. [27] Quan C, Talley NJ, Jones MP, Spies J, Horowitz M. Gain and loss of gastrointestinal
symptoms in diabetes mellitus: associations with psychiatric disease, glycemic
control, and autonomic neuropathy over 2 years of follow up. Am J Gastroentrol
Conflicts of interest 2008;103:2023–30.
[28] Papanas N, Vinik AI, Ziegler D. Neuropathy in prediabetes: does the clock start
This review was written independently. No company or institution ticking early? Nat Rev Endocrinol 2011;7:682–90.
[29] Coleski R, Hasler WL. Coupling and propagation of normal and dysrhythmic gas-
supported it financially. N Papanas has been an advisory board member tric slow waves during acute hyperglycaemia in healthy humans. Neurogastroen-
of Trigo Care International; has participated in sponsored studies by terol Motil 2009;21:492–9.
Novo Nordisk and Novartis; received honoraria as a speaker for Novo [30] Harberson J, Thomas RM, Harbison SP, Parkman HP. Gastric neuromuscular
pathology in gastroparesis: analysis of full thickness antral biopsies. Dig Dis Sci
Nordisk and Pfizer; and attended conferences sponsored by TrigoCare 2010;55:359–70.
International, Novo Nordisk, sanofi-aventis and Pfizer. E Maltezos has [31] Selim MM, Wendelschafer-Crabb G, Redmon JB, Khoruts A, Hodges JS, Koch K,
participated in sponsored studies by Novo Nordisk and Novartis; and et al. Gastric mucosal nerve density A biomarker for diabetic autonomic neurop-
athy? Neurology 2010;75:973–81.
attended conferences sponsored by Wyeth, Pfizer and Bayer.
[32] Murray C, Martin NM, Patterson M, Taylor SA, Ghatei MA, Kamm MA, et al. Ghrelin
enhances gastric emptying in diabetic gastroparesis: a double blind, placebo con-
References trolled, crossover study. Gut 2005;54:1693–8.
[33] Bult H, Boeckxstaens CE, Pelckmans PA, Jordaens FH, Van Maercke YM, Herman
[1] Bytzer P, Talley NJ, Leemon M, Young LJ, Jones MP, Horowitz M. Prevalence of gas- AG. Nitric oxide as an inhibitory non-adrenergic non-cholinergic neurotransmit-
trointestinal symptoms associated with diabetes mellitus: a population-based ter. Nature 1990;345:346–7.
survey of 15000 adults. Arch Intern Med 2001;161:1989–96. [34] Takahashi T. Pathophysiological significance of neuronal nitric oxide synthase in
[2] Wang YR, Fisher RS, Parkman HP. Gastroparesis-related hospitalizations in the the gastrointestinal tract. J Gastroenterol 2003;38:421–30.
United States: trends, characteristics and outcomes, 1995–2004. Am J Gastroen- [35] Ordog T, Hayashi Y, Gibbons J. Cellular pathogenesis of diabetic gastroenteropa-
terol 2008;103:313–22. thy. Minerva Gastroenterol Dietol 2009;55:315–43.
[3] Ziegler D. Diagnosis and treatment of diabetic autonomic neuropathy. Curr Diab [36] Mazzone A, Bernard CE, Strege PR, Beyder A, Galietta LJ, Pasricha PJ, et al. Altered
Rep 2001;1:216–27. expression of Ano variants in human diabetic gastroparesis. J Biol Chem 2011;15:
[4] Kempler P, Amarenco G, Freeman R, Frontoni S, Horowitz M, Stevens M, et al. Gas- 13393–403.
trointestinal autonomic neuropathy, erectile-, bladder- and sudomotor dysfunc- [37] Bhor VM, Raghuram N, Sivakami S. Oxidative damage and altered antioxidant en-
tion in patients with diabetes mellitus: clinical impact, assessment, diagnosis, zyme activities in the small intestine of streptozotocine-induced diabetic rats. Int
and management. Diabetes Metab Res Rev 2011 [Jul 11. Epub ahead of print]. J Biochem Cell Biol 2004;36:89–97.
[5] Tack J, Janssen P. Gastrointestinal motility. Curr Opin Gastroenterol 2010;26: [38] Samra YK, Wang M, Peat NJ, Smith NC, Shotton HR, Lincoln J. Selective susceptibil-
647–55. ity of different populations of sympathetic neurons to diabetic neuropathy in vivo
[6] Camilleri M, Bharucha E, Farrugia G. Epidemiology, mechanisms and management is reflected by increased vulnerability to oxidative stress in vitro. Neurosci Lett
of diabetic gastroparesis. Clin Gastroenterol Hepatol 2011;9:5–12. 2006;407:199–204.
[7] Verne GN, Snisky CA. Diabetes and the gastrointestinal tract. Gastroenterol Clin [39] Kashyap P, Farrugia G. Oxidative stress: key player in gastrointestinal complica-
North Am 1998;27:861–74. tions of diabetes. Neurogastroenterol Motil 2011;23:111–4.
[8] Boehme MW, Autschbach F, Ell C, Raeth U. Prevalence of silent gastric ulcer, ero- [40] Choi KM, Gibbons SJ, Nguyen TV, Stoltz GJ, Lurken MS, Ordog T, et al. Hemeoxy-
sions or severe acute gastritis in patients with type 2 diabetes mellitus-a genase-1 protects interstitial cellsof Cajal from oxidative stress and reverses
cross-sectional study. Hepatogastrenterology 2007;54:643–8. diabetic gastroparesis. Gastroenterology 2008;135:2055–64.
[9] Kashyap P, Farrugia G. Diabetic gastroparesis: what we have learned and hand to [41] Kashyap P, Farrugia G. Enteric autoantibodies and gut motility disorders. Gastro-
unlearn in the past 5 years. Gut 2010;59:1716–26. enterol Clin North Am 2008;37:397–410.
[10] Parkman HP, Hasler WL, Fisher RS. American Gastroenterological Association [42] Bour-Jordan H, Bluestone JA. Sensory neurons link the nervous system ans auto-
technical review on the diagnosis and treatment of gastroparesis. Gastroenterol- immune diabetes. Cell 2006;127:1097–9.
ogy 2004;127:1592–622. [43] Gutierrez-Canas I, Juarranz Y, Santiago B, Arranz A, Martinez C, Galindo M, et al. VIP
[11] Bharucha AE, Camillieri M, Forstrom LA, Zinsmeister AR. Relationship between down-regulates TLR4 expression and TLR4-Mediated chemokine production in
clinical features and gastric emptying disturbances in diabetes mellitus. Clin human rheumatoid synovial fibroblasts. Rheumatology (Oxford) 2006;45:527–32.
Endocrinol (Oxf) 2009;70:415–20. [44] Chandrasekharan B, Srinivasan S. Diabetes and the enteric nervous system.
[12] Jung HK, Choung RS, Locke 3rd GR, Schleck CD, Zinsmeister AR, Szarka LA, et al. Neurogastroentrol Motil 2007;19:951–60.
The incidence, the prevalence and the outcomes of patients with gastroparesis [45] Coulson FR, Jacoby DB, Fryer AD. Insulin regulates neuronal M2 muscarinic recep-
in Olmsted County, Minnesota from 1996 to 2006. Gastroenterology 2009;136: tor function in the ileum of diabetic rats. J Pharmacol Exp Ther 2004;308:760–6.
1225–33. [46] Horvath VJ, Vittal H, Lörincz A, Chen H, Almeida-Porada G, Redelman D, et al. Re-
[13] Horowitz M, Maddox AF, Wishart JM, Harding PE, Chatteron BE, Shearman DJ. duced stem cell factor links smooth myopathy and loss of interstitial cells of cajal
Relationship between esophageal transit and solid and liquid gastric emptying in murine diabetic gastroparesis. Gastroenterology 2006;130:759–70.
in diabetes mellitus. Eur J Nucl Med 1991;18:229–34. [47] Schmidt RE, Dorsey DA, Beaudet LN, Plurad SB, Parvin CA, Ohara S. Effect of IGF-I
[14] Kinekawa F, Kubo F, Matsuka K, Kobayashi M, Furuta Y, Okada H, et al. Esophageal and neurotrophin-e on gracile neuroaxonal dystrophy in diabetic and aging rats.
function worsens with long duration of diabetes. J Gastroenterol 2008;43:338–44. Brain Res 2000;876:88–94.
[15] Horikawa A, Ishii-Nozawa R, Ohguro M, Tagagi S, Ohtuji M, Yamada M, et al. Prev- [48] Gaddipati KV, Simonian HP, Kresge KM, Boden GH, Parkman HP. Abnormal ghre-
alence of GORD in type 2 diabetes and a comparison of clinical profiles between lin and pancreatic polyptetide responses in gastroparesis. Dig Dis Sci 2006;51:
diabetic patients with and without GORD. Diabet Med 2009;26:228–33. 1339–46.
[16] Paragliarulo M, Fornari F, Fraguell M, Zoli M, Giangregorio F, Grigolon A, et al. [49] Tougas G, Hunt RH, Fitzpatrick D, Upton AR. Evidence of impaired afferent vagal
Gallstone diseases and related risk factor in a large cohort of diabetic patients. function in patients with diabetes gastroparesis. Pacing Clin Electrophysiol
Dig Liver Dis 2004;36:130–4. 1992;15:1597–602.
[17] Ding X, Lu CY, Liu CA, Shi YJ. Correlation between gene expression of CCK-A recep- [50] Frokjaer JB, Egsgaard LL, Graversen C, Søfteland E, Dimcevski G, Blauenfeldt RA,
tor and emptying dysfunction of the gallbladder in patients with gallstones and et al. Gastrointestinal symptoms in type-1: is it all about brain plasticity? Eur J
diabetes mellitus. Hepatobilliary Panceat Dis Int 2005;4:295–8. Pain 2011;15:249–57.
A. Gatopoulou et al. / European Journal of Internal Medicine 23 (2012) 499–505 505

[51] Tack J, Talley NJ, Camilleri M, Holtmann G, Hu P, Malagelada JR, et al. Functional [69] Camilleri M, Kerstens R, Beyens G. A double-blind, placebo-controlled trial to
gastroduodenal disorders. Gastroenterology 2006;130:1466–79. evaluate the safety and tolerability of prucalopride oral solution in constipated
[52] Patric A, Epstein O. Review article: gastroparesis. Aliment Pharmacol Ther elderly patients living in a nursing facility. Gastroenterology 2009;136:240
2008;27:724–40. [Abstract].
[53] Abell TL, Camillieri M, Donohoe K, Hasler WL, Lin HC, Maurer AH, et al. Consensus [70] Sanger GJ, Westaway SM, Barnes AA, Macpherson DT, Muir AI, Jarvie EM, et al.
recommendations for gastric emptying scintigraphy: a joint report of the Ameri- GSK962040 a small molecule selective motilin agonist, effective as a stimulant
can Neurogastroenterology and Motility Society and the Society of Nuclear of human and rabbit gastrointestinal motility. Neurogastroenterol Motil
Medicine. Am J Gastroenterol 2008;103:753–63. 2009;21:657–64.
[54] Szarka LA, Camilleri M, Vella A, Burton D, Baxter K, Simonson J, et al. A stable [71] Ejskaer N, Dimcevski G, Wo J, Hellström PM, Gormsen LC, Sarosiek I, et al. Safety
isotope breath test with standard meal for abnormal gastric emptying of solids and efficacy of ghrelin agonist TZP-101, in relieving symptoms in patients with di-
in the clinic and in research. Clin Gastroenterol Hepatol 2008;6:635–43. abetic gastroparesis: a randomized, placebo-controlled study. Neurogastroenterol
[55] Szarka L, Camillieri M. Stomach dysfunction in diabetes mellitus: emerging tech- Motil 2010;22:1069-e281.
nology and pharmacology. J Diabetes Sci Technol 2010;4:180–9. [72] Braden B, Caspary W, Borner N, Vinson B, Schneider AR. Clinical effects of STW5
[56] Cassilly D, Kantor S, Knight LC, Maurer AH, Fisher RS, Semler J, et al. Gastric emp- [Iberocast] are not based on acceleration of gastric emptying in patients with
tying of a non-digestible solid: assessment with simultaneous Smart Pill pH and functional dyspepsia and gastroparesis. Neurogastroenterol Motil 2009;21:632–8.
pressure capsule, antroduodenal manometry, gastric emptying scintigraphy. Neu- [73] Bai Y, Xu MJ, Yang X, Xu C, Gao J, Zou DW, et al. A systematic review on intrapy-
rogastroenterol Motil 2008;20:311–9. loric botulinum toxin injection for gastroparesis. Digestion 2010;81:27–34.
[57] Kuo B, McCallum RW, Koch KL, Sitrin MD, Wo JM, Chey WD, et al. Comparison of [74] McCallum RW, Snape W, Brady F, Wa J, Parkman HP, Nowak T. Gastric electrical
gastric emptying of a nondigestible capsule to a radio-labelled meal in healthy stimulation with Enterra therapy improves symptoms from diabetic gastroparesis
and gastroparetic subjects. Aliment Pharmacol Ther 2008;27:186–96. in a prospective study. Clin Gastroenterol Hepatol 2010;8:947–54.
[58] Bilecen D, Scheffler K, Seifritz E, Bongartz G, Steinbrich W. Hydro-MRI for the [75] Daram SR, Tang SJ, Abell TL. Temporary gastric electrical stimulation for gastro-
visualization of gastri wall motility using RARE magnetic resonance imaging paresis: endoscopic placement of electrodes [ENDOstim]. Surg Endosc 2011;25:
sequences. Abdom Imaging 2000;25:30–4. 3444–5.
[59] Bouras EP, Delgado-Aros S, Camilleri M, Castillo EJ, Burton DD, Thomforde GM, [76] Chu H, Lin Z, Zhong L, McCallum RW, Hou X. A meta-analysis: the treatment of
et al. SPECT imaging of the stomach: comparison with barostat and effects of high-frequency gastric electrical stimulation for gastroparesis. J Gastroenterol
sex, age, body mass index and fundoplication Sigle photon emission computed to- Hepatol 2011 [Dec 1 Epub ahead of print].
mography. Gut 2002;51:781–6. [77] Lin Z, Sarosiek I, Forster J, Ross RA, Chen JD, McCallum RW. Two-channel gastric
[60] Liao D, Gregersen H, Hausken T, Gilja OH, Mundt M, Kassab G. Analysis of surface pacing in patients with diabetic gastroparesis. Neurogastroenterol Motil
geometry of the human stomach using real-time 3-D ultrasonography in vivo. 2011;23:912-e396.
Neurogastroenterol Motil 2004;16:315–24. [78] Bharucha AE, Camilleri M, Forstrom LA, Zinsmeister AR. Relationship between
[61] Wilson JA, Vela MF. New esophageal function testing (impedance, Bravo pH mon- clinical features and gastric emptying disturbances in diabetes mellitus. Clin
itoring and high-resolution manometry): clinical relevance. Curr Gastroenterol Endocrinol 2009;70:415–20.
Rep 2008;10:222–30. [79] Linnebjerg H, Park S, Kothare PA, Trautmann ME, Mace K, Fineman M, et al. Effect
[62] Quan C, Talley NJ, Cross S, Jones M, Hammer J, Giles N, et al. Development and of exenatide on gastric emptying and relationship to postpradial glycemia in type
validation of the Diabetes Bowel Symptom Questionnaire. Aliment Pharmacol 2 diabetes. Regul Pept 2008;151:123–9.
Ther 2003;17:1179–87. [80] Zietz B, Lock G, Straub RH, Braun B, Schölmerich J, Palitzsch KD. Small-bowel
[63] Revicki DA, Rentz AM, Dubois D, Kahrilas P, Stanghellini V, Talley NJ, et al. Devel- bacterial overgrowth in diabetic subject is associated with cardiovascular auto-
opment and validation of a patient-assessed gastroparesis symptom severity nomic neuropathy. Diabetes Care 2000;23:1200–1.
measure; the Gastroparesis Cardinal Symptom Index. Aliment Pharmacol Ther [81] Bures J, Cyrany J, Kohoutova D, Förstl M, Rejchrt S, Kvetina J, et al. Small intestinal
2003;18:141–50. bacterial overgrowth syndrome. World J Gastroenterol 2010;16:2978–90.
[64] Parkman HP, Yates KP, Hasler WL, Nguyan L, Pasricha PJ, Snape WJ, et al. Dietary [82] Pimentel M. Review of rifaximine as treatment for small intestinal bacterial over-
intake and nutritional deficiencies in patients with diabetic or idiopathic gastro- growth and irritable bowel syndrome. Expert Opin Investig Drugs 2009;18:349–58.
paresis. Gastroenterology 2011;142:486–98. [83] Samson M, Verhagen M. Intestinal function. In: Horowitz M, Samson M, editors.
[65] Tack J. Prokinetics and fundic relaxants in upper functional GI disorders. Curr Opin Gastrointestinal function in diabetes mellitus. Chichester: John Wiley and Sons;
Pharmacol 2008;8:690–6. 2004. p. 177–217.
[66] Patterson D, Abell T, Rothstein R, Koch K, Barnett J. A double-blind multicenter [84] Sanger G, Alpers D. Development of drugs for gastrointestinal motor disorders:
comparison of domperidone and metoclopramide in the treatment of diabetic translating science to clinical need. Neurogastroenterol Motil 2008;20:177–84.
patients with symptoms of gastroparesis. Am J Gastroenterol 1999;94:1230–4. [85] Camilleri M. Novel pharmacology: asimadoline, a kappa-opiod agonist and viscer-
[67] Dhir R, Richter JE. Erythromycin in the short- and long term control of dyspepsia al sensation. Neurogastroenterol Motil 2008;20:971–9.
symptoms in patients with gastroparesis. J Clin Gastroenterol 2004;38:237–42. [86] Tang DM, Fridernberg FK. Gastroparesis: approach, diagnostic evaluation and
[68] Sharma D, Morrison G, Joseph F, Purewal TS, Weston PJ. The role of continuous management. Dis Mon 2011;57:74–101.
subcutaneous insulin infusion therapy in patients with diabetic gastroparesis. [87] Volarevic V, Arsenijevic N, Lukic ML, Stojkovic M. Concise review Mesenchymal stem
Diabetologia 2011;54:2768–70. cell treatment of the complications of diabetes mellitus. Stem Cells 2011;29:5–10.

Você também pode gostar