Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Clin Colorectal Cancer ; 21(3): e196-e204, 2022 09.
Article in English | MEDLINE | ID: mdl-35668002

ABSTRACT

INDUCTION: chemotherapy (IC) followed by chemoradiation (CRT) is an attractive approach in high-risk locally advanced rectal cancer. Additionally, ASA has shown potential to improve outcomes alongside CRT in rectal cancer. The ICAR trial aimed to evaluate the safety and efficacy of IC followed by CRT with or without ASA on MRI tumor response. METHODS: Single-center, double-blind, randomized phase II trial to evaluate induction treatment with CAPOX, followed by capecitabine-based chemoradiotherapy with ASA (arm 1) or placebo (arm 2) in high-risk stage II-III rectal adenocarcinoma staged by MRI. The primary endpoint was MRI tumor regression grade (mrTRG). Secondary endpoints were pathological response, surgical outcomes, postoperative complications, treatment tolerance, DFS, and OS. RESULTS: Between January 2018 and August 2019, 27 patients were eligible, 25 (92.5%) completed IC, and 23 patients were randomly assigned (12 to ASA group; 11 to placebo group). In the ASA arm, 3 pts (25%) presented distant disease progression at restaging. Seven patients (30.4%) had cCR after neoadjuvant treatment. All 13 patients submitted to surgery after neoadjuvant treatment underwent R0 resections except for 1 patient with positive CRM, and 12 patients (92.3%) had sphincter preservation. After a median follow-up of 34.9 months, the 2-year DFS was 83.1% and 3-year OS was 81.5%. CONCLUSION: There was good compliance in both treatment arms and encouraging cCR rate. ASA during CRT was safe but failed to improve on MRI tumor response. The study was closed due to the absence of benefits.


Subject(s)
Induction Chemotherapy , Rectal Neoplasms , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Chemoradiotherapy/methods , Double-Blind Method , Humans , Induction Chemotherapy/methods , Neoadjuvant Therapy/methods , Neoplasm Staging , Rectal Neoplasms/pathology , Treatment Outcome
2.
J Pediatr Gastroenterol Nutr ; 66 Suppl 3: S52-S55, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29762379

ABSTRACT

BACKGROUND: A subset of children with functional gastrointestinal disorders (FGIDs), which includes functional dyspepsia, may have duodenal disaccharidase deficiencies. OBJECTIVES: To determine the frequency, demographics, and clinical characteristics associated with duodenal disaccharidase deficiencies in children with functional dyspepsia. METHODS: Children ages 4 to 18 years undergoing esophagogastroduodenoscopy (EGD) evaluation for dyspepsia were enrolled in either a retrospective (study 1) or prospective (study 2) evaluation. Those with histologic abnormalities were excluded. Duodenal biopsies were obtained for disaccharidase enzyme analysis. In the retrospective study, both demographic and clinical characteristics were obtained via chart review. In the prospective study, parents completed the Rome II Questionnaire on Gastrointestinal Symptoms before the EGD. RESULTS: One hundred and twenty-nine children (n = 101, study 1; n = 28, study 2) were included. Mean age was 11.2 ±â€Š3.8 (SD) years in study 1 and 10.6 ±â€Š3.2 years in study 2. Forty-eight (47.5%) of subjects in study 1 and 13 (46.4%) of subjects in study 2 had at least 1 disaccharidase deficiency identified. All of those with a disaccharidase deficiency in both studies had lactase deficiency with 8 (7.9%) and 5 (17.9%) of those in studies 1 and 2, respectively, having an additional disaccharidase deficiency. The second most common disaccharidase deficiency pattern was that of pan-disaccharidase deficiency (PDD) in both studies. In study 1 (where both race and ethnicity were captured), self-identified Hispanic (vs non-Hispanic, P < 0.05) and non-white (vs white, P < 0.01) children were more likely to have lactase deficiency. Age, sex, and type of gastrointestinal symptom were not associated with presence or absence of a disaccharidase deficiency. CONCLUSIONS: Approximately half of children with functional dyspepsia undergoing EGD were identified as having a disaccharidase deficiency (predominantly lactase deficiency). Race/ethnicity may be associated with the likelihood of identifying a disaccharidase deficiency. Other clinical characteristics were not able to distinguish those with versus without a disaccharidase deficiency.


Subject(s)
Disaccharidases/deficiency , Duodenum/enzymology , Dyspepsia/etiology , Intestinal Mucosa/enzymology , Malabsorption Syndromes/epidemiology , Adolescent , Child , Child, Preschool , Duodenum/pathology , Endoscopy, Digestive System , Female , Humans , Intestinal Mucosa/pathology , Malabsorption Syndromes/complications , Malabsorption Syndromes/diagnosis , Male , Prospective Studies , Retrospective Studies
3.
J Pediatr Gastroenterol Nutr ; 66 Suppl 3: S61-S64, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29762381

ABSTRACT

BACKGROUND AND HYPOTHESES: Human starch digestion is a multienzyme process involving 6 different enzymes: salivary and pancreatic α-amylase; sucrase and isomaltase (from sucrose-isomaltase [SI]), and maltase and glucoamylase (from maltase-glucoamylase [MGAM]). Together these enzymes cleave starch to smaller molecules ultimately resulting in the absorbable monosaccharide glucose. Approximately 80% of all mucosal maltase activity is accounted for by SI and the reminder by MGAM. Clinical studies suggest that starch may be poorly digested in those with congenital sucrase-isomaltase deficiency (CSID). Poor starch digestion occurs in individuals with CSID and can be documented using a noninvasive C-breath test (BT). METHODS: C-Labled starch was used as a test BT substrate in children with CSID. Sucrase deficiency was previously documented in study subjects by both duodenal biopsy enzyme assays and C-sucrose BT. Breath CO2 was quantitated at intervals before and after serial C-substrate loads (glucose followed 75 minutes later by starch). Variations in metabolism were normalized against C-glucose BT (coefficient of glucose absorption). Control subjects consisted of healthy family members and a group of children with functional abdominal pain with biopsy-proven sucrase sufficiency. RESULTS: Children with CSID had a significant reduction of C-starch digestion mirroring that of their duodenal sucrase and maltase activity and C-sucrase BT. CONCLUSIONS: In children with CSID, starch digestion may be impaired. In children with CSID, starch digestion correlates well with measures of sucrase activity.


Subject(s)
Breath Tests/methods , Carbohydrate Metabolism, Inborn Errors/diagnosis , Duodenum/enzymology , Starch/metabolism , Sucrase-Isomaltase Complex/deficiency , Adolescent , Carbon Isotopes/metabolism , Case-Control Studies , Child , Child, Preschool , Digestion/physiology , Female , Humans , Infant , Male , Sucrase-Isomaltase Complex/analysis
7.
J Pediatr Gastroenterol Nutr ; 48(4): 412-8, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19330928

ABSTRACT

BACKGROUND: Congenital sucrase-isomaltase deficiency (CSID) is characterized by absence or deficiency of the mucosal sucrase-isomaltase enzyme. Specific diagnosis requires upper gastrointestinal biopsy with evidence of low to absent sucrase enzyme activity and normal histology. The hydrogen breath test (BT) is useful, but is not specific for confirmation of CSID. We investigated a more specific 13C-sucrose labeled BT. OBJECTIVES: Determine whether CSID can be detected with the 13C-sucrose BT without duodenal biopsy sucrase assay, and if the 13C-sucrose BT can document restoration of sucrose digestion by CSID patients after oral supplementation with sacrosidase (Sucraid). METHODS: Ten CSID patients were diagnosed by low biopsy sucrase activity. Ten controls were children who underwent endoscopy and biopsy because of dyspepsia or chronic diarrhea with normal mucosal enzymes activity and histology. Uniformly labeled 13C-glucose and 13C-sucrose loads were orally administered. 13CO2 breath enrichments were assayed using an infrared spectrophotometer. In CSID patients, the 13C-sucrose load was repeated adding Sucraid. Sucrose digestion and oxidation were calculated as a mean percent coefficient of glucose oxidation averaged between 30 and 90 minutes. RESULTS: Classification of patients by 13C-sucrose BT percent coefficient of glucose oxidation agreed with biopsy sucrase activity. The breath test also documented the return to normal of sucrose digestion and oxidation after supplementation of CSID patients with Sucraid. CONCLUSIONS: 13C-sucrose BT is an accurate and specific noninvasive confirmatory test for CSID and for enzyme replacement management.


Subject(s)
Breath Tests/methods , Carbohydrate Metabolism, Inborn Errors/diagnosis , Carbon Isotopes , Sucrase-Isomaltase Complex/deficiency , Sucrose/metabolism , beta-Fructofuranosidase/therapeutic use , Adolescent , Biopsy , Carbohydrate Metabolism, Inborn Errors/drug therapy , Carbohydrate Metabolism, Inborn Errors/enzymology , Carbon Dioxide/metabolism , Child , Child, Preschool , Dietary Supplements , Female , Glucose/metabolism , Humans , Infant , Male , Sucrase/metabolism , Sucrase-Isomaltase Complex/genetics
8.
J Nutr ; 139(4): 684-90, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19193815

ABSTRACT

Starch is the major source of food glucose and its digestion requires small intestinal alpha-glucosidic activities provided by the 2 soluble amylases and 4 enzymes bound to the mucosal surface of enterocytes. Two of these mucosal activities are associated with sucrase-isomaltase complex, while another 2 are named maltase-glucoamylase (Mgam) in mice. Because the role of Mgam in alpha-glucogenic digestion of starch is not well understood, the Mgam gene was ablated in mice to determine its role in the digestion of diets with a high content of normal corn starch (CS) and resulting glucose homeostasis. Four days of unrestricted ingestion of CS increased intestinal alpha-glucosidic activities in wild-type (WT) mice but did not affect the activities of Mgam-null mice. The blood glucose responses to CS ingestion did not differ between null and WT mice; however, insulinemic responses elicited in WT mice by CS consumption were undetectable in null mice. Studies of the metabolic route followed by glucose derived from intestinal digestion of (13)C-labeled and amylase-predigested algal starch performed by gastric infusion showed that, in null mice, the capacity for starch digestion and its contribution to blood glucose was reduced by 40% compared with WT mice. The reduced alpha-glucogenesis of null mice was most probably compensated for by increased hepatic gluconeogenesis, maintaining prandial glucose concentration and total flux at levels comparable to those of WT mice. In conclusion, mucosal alpha-glucogenic activity of Mgam plays a crucial role in the regulation of prandial glucose homeostasis.


Subject(s)
Digestion , Glucose/metabolism , Homeostasis , Starch/metabolism , alpha-Glucosidases/metabolism , Animal Feed , Animals , Fasting , Genotype , Insulin/blood , Mice , Mice, Knockout , Mucous Membrane/enzymology , Sucrase/metabolism , alpha-Glucosidases/deficiency , alpha-Glucosidases/genetics
9.
J Nutr ; 138(4): 685-92, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18356321

ABSTRACT

The detailed mechanistic aspects for the final starch digestion process leading to effective alpha-glucogenesis by the 2 mucosal alpha-glucosidases, human sucrase-isomaltase complex (SI) and human maltase-glucoamylase (MGAM), are poorly understood. This is due to the structural complexity and vast variety of starches and their intermediate digestion products, the poorly understood enzyme-substrate interactions occurring during the digestive process, and the limited knowledge of the structure-function properties of SI and MGAM. Here we analyzed the basic catalytic properties of the N-terminal subunit of MGAM (ntMGAM) on the hydrolysis of glucan substrates and compared it with those of human native MGAM isolated by immunochemical methods. In relation to native MGAM, ntMGAM displayed slower activity against maltose to maltopentose (G5) series glucose oligomers, as well as maltodextrins and alpha-limit dextrins, and failed to show the strong substrate inhibitory "brake" effect caused by maltotriose, maltotetrose, and G5 on the native enzyme. In addition, the inhibitory constant for acarbose was 2 orders of magnitude higher for ntMGAM than for native MGAM, suggesting lower affinity and/or fewer binding configurations of the active site in the recombinant enzyme. The results strongly suggested that the C-terminal subunit of MGAM has a greater catalytic efficiency due to a higher affinity for glucan substrates and larger number of binding configurations to its active site. Our results show for the first time, to our knowledge, that the C-terminal subunit of MGAM is responsible for the MGAM peptide's "glucoamylase" activity and is the location of the substrate inhibitory brake. In contrast, the membrane-bound ntMGAM subunit contains the poorly inhibitable "maltase" activity of the internally duplicated enzyme.


Subject(s)
Glycoside Hydrolase Inhibitors , Protein Subunits/chemistry , Starch/metabolism , alpha-Glucosidases/chemistry , Acarbose , Catalysis , Dextrins/metabolism , Humans , Hydrogen-Ion Concentration , Oligosaccharides/metabolism , Polysaccharides/metabolism , Protein Subunits/metabolism , Recombinant Proteins , Sucrase-Isomaltase Complex/metabolism , alpha-Glucosidases/metabolism
10.
J Pediatr Gastroenterol Nutr ; 45(1): 32-43, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17592362

ABSTRACT

BACKGROUND: Starches are the major source of dietary glucose in weaned children and adults. However, small intestine alpha-glucogenesis by starch digestion is poorly understood due to substrate structural and chemical complexity, as well as the multiplicity of participating enzymes. Our objective was dissection of luminal and mucosal alpha-glucosidase activities participating in digestion of the soluble starch product maltodextrin (MDx). PATIENTS AND METHODS: Immunoprecipitated assays were performed on biopsy specimens and isolated enterocytes with MDx substrate. RESULTS: Mucosal sucrase-isomaltase (SI) and maltase-glucoamylase (MGAM) contributed 85% of total in vitro alpha-glucogenesis. Recombinant human pancreatic alpha-amylase alone contributed <15% of in vitro alpha-glucogenesis; however, alpha-amylase strongly amplified the mucosal alpha-glucogenic activities by preprocessing of starch to short glucose oligomer substrates. At low glucose oligomer concentrations, MGAM was 10 times more active than SI, but at higher concentrations it experienced substrate inhibition whereas SI was not affected. The in vitro results indicated that MGAM activity is inhibited by alpha-amylase digested starch product "brake" and contributes only 20% of mucosal alpha-glucogenic activity. SI contributes most of the alpha-glucogenic activity at higher oligomer substrate concentrations. CONCLUSIONS: MGAM primes and SI activity sustains and constrains prandial alpha-glucogenesis from starch oligomers at approximately 5% of the uninhibited rate. This coupled mucosal mechanism may contribute to highly efficient glucogenesis from low-starch diets and play a role in meeting the high requirement for glucose during children's brain maturation. The brake could play a constraining role on rates of glucose production from higher-starch diets consumed by an older population at risk for degenerative metabolic disorders.


Subject(s)
Enterocytes/metabolism , Glucan 1,4-alpha-Glucosidase/metabolism , Glucose/metabolism , Polysaccharides/metabolism , alpha-Glucosidases/metabolism , Animals , Biopsy , Child , Digestion , Duodenum/enzymology , Enterocytes/enzymology , Humans , Immunoprecipitation , Intestinal Mucosa/enzymology , Mice , Oligo-1,6-Glucosidase/metabolism , Starch/metabolism
11.
J Nutr ; 137(7): 1725-33, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17585022

ABSTRACT

Digestion of starch requires activities provided by 6 interactive small intestinal enzymes. Two of these are luminal endo-glucosidases named alpha-amylases. Four are exo-glucosidases bound to the luminal surface of enterocytes. These mucosal activities were identified as 4 different maltases. Two maltase activities were associated with sucrase-isomaltase. Two remaining maltases, lacking other identifying activities, were named maltase-glucoamylase. These 4 activities are better described as alpha-glucosidases because they digest all linear starch oligosaccharides to glucose. Because confusion persists about the relative roles of these 6 enzymes, we ablated maltase-glucoamylase gene expression by homologous recombination in Sv/129 mice. We assayed the alpha-glucogenic activities of the jejunal mucosa with and without added recombinant pancreatic alpha-amylase, using a range of food starch substrates. Compared with wild-type mucosa, null mucosa or alpha-amylase alone had little alpha-glucogenic activity. alpha-Amylase amplified wild-type and null mucosal alpha-glucogenesis. alpha-Amylase amplification was most potent against amylose and model resistant starches but was inactive against its final product limit-dextrin and its constituent glucosides. Both sucrase-isomaltase and maltase-glucoamylase were active with limit-dextrin substrate. These mucosal assays were corroborated by a 13C-limit-dextrin breath test. In conclusion, the global effect of maltase-glucoamylase ablation was a slowing of rates of mucosal alpha-glucogenesis. Maltase-glucoamylase determined rates of digestion of starch in normal mice and alpha-amylase served as an amplifier for mucosal starch digestion. Acarbose inhibition was most potent against maltase-glucoamylase activities of the wild-type mouse. The consortium of 6 interactive enzymes appears to be a mechanism for adaptation of alpha-glucogenesis to a wide range of food starches.


Subject(s)
Glucose/biosynthesis , Intestinal Mucosa/enzymology , Jejunum/enzymology , Starch/metabolism , alpha-Glucosidases/metabolism , Acarbose/metabolism , Acarbose/pharmacology , Animals , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/physiology , Isomaltose/metabolism , Maltose/metabolism , Mice , Mice, Knockout , alpha-Glucosidases/genetics
12.
Nutr Rev ; 65(2): 95-8, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17345962

ABSTRACT

A limited fraction of the human adult population retains intestinal lactase-phlorizin hydrolase (LPH) activity during adulthood, and this is called the lactase persistence phenotype. However, 95% of all adults have adult-type hypolactasia (ATH) and have difficulty digesting milk sugar. Rarely, some infants are born with an inability to digest lactase (congenital lactase deficiency or CLD) due to low levels of LPH activity, which results in severe clinical consequences if not properly diagnosed and treated by lactose avoidance. Recently, it has been shown that both recessive LPH deficiencies, CLD and ATH, are related to DNA variants affecting the lactase (LCT) gene, but they are mediated through very different molecular mechanisms. The LCT mutations resulting in childhood CLD lead to low LPH activity through nonsense-mediated LCT mRNA decay, whereas the critical nucleotide variants for the ATH phenotype represent distal enhancer polymorphisms, which regulate developmentally LCT transcript levels in intestinal cells.


Subject(s)
Chromosomes, Human, Pair 2 , Lactase/deficiency , Lactase/genetics , Lactose Intolerance/genetics , Polymorphism, Genetic , Chromosome Mapping , Genetic Markers , Haplotypes , Humans
13.
Clin Gastroenterol Hepatol ; 4(3): 276-87, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16527688

ABSTRACT

Sugars normally are absorbed in the small intestine. When carbohydrates are malabsorbed, the osmotic load produced by the high amount of low molecular weight sugars and partially digested starches in the small intestine can cause symptoms of intestinal distention, rapid peristalsis, and diarrhea. Colonic bacteria normally metabolize proximally malabsorbed dietary carbohydrate through fermentation to small fatty acids and gases (ie, hydrogen, methane, and carbon dioxide). When present in large amounts, the malabsorbed sugars and starches can be excreted in the stool. Sugar intolerance is the presence of abdominal symptoms related to the proximal or distal malabsorption of dietary carbohydrates. The symptoms consist of meal-related abdominal cramps and distention, increased flatulence, borborygmus, and diarrhea. Infants and young children with carbohydrate malabsorption show more intense symptoms than adults; the passage of undigested carbohydrates through the colon is more rapid and is associated with detectable carbohydrates in copious watery acid stools. Dehydration often follows feeding of the offending sugar. In this review we present the clinical and current molecular aspects of disaccharidase digestion.


Subject(s)
Digestion/genetics , Digestion/physiology , Disaccharidases/deficiency , Disaccharides/metabolism , Malabsorption Syndromes/genetics , Malabsorption Syndromes/metabolism , Disaccharidases/genetics , Humans , Mutation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...