Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
1.
FASEB J ; 34(3): 3983-3995, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31957074

RESUMO

BACKGROUND AND AIMS: Intestinal adaptation in short bowel syndrome (SBS) includes morphologic processes and functional mechanisms. This study investigated whether digestive enzyme expression in the duodenum and colon is upregulated in SBS patients. METHOD: Sucrase-isomaltase (SI), lactase-phlorizin hydrolase (LPH), and neutral Aminopeptidase N (ApN) were analyzed in duodenal and colonic biopsies from nine SBS patients in a late stage of adaptation as well as healthy and disease controls by immunoelectron microscopy (IEM), Western blots, and enzyme activities. Furthermore, proliferation rates and intestinal microbiota were analyzed in the mucosal specimen. RESULTS: We found significantly increased amounts of SI, LPH, and ApN in colonocytes in most SBS patients with large variation and strongest effect for SI and ApN. Digestive enzyme expression was only partially elevated in duodenal enterocytes due to a low proliferation level measured by Ki-67 staining. Microbiome analysis revealed high amounts of Lactobacillus resp. low amounts of Proteobacteria in SBS patients with preservation of colon and ileocecal valve. Colonic expression was associated with a better clinical course in single cases. CONCLUSION: In SBS patients disaccharidases and peptidases can be upregulated in the colon. Stimulation of this colonic intestinalization process by drugs, nutrients, and pre- or probiotics might offer better therapeutic approaches.


Assuntos
Intestino Grosso/enzimologia , Síndrome do Intestino Curto/enzimologia , Aminopeptidases/metabolismo , Western Blotting , Dissacaridases/metabolismo , Feminino , Humanos , Lactase-Florizina Hidrolase/metabolismo , Lactobacillus/fisiologia , Masculino , Microscopia Imunoeletrônica , Peptídeo Hidrolases/metabolismo , Proteobactérias/fisiologia , Complexo Sacarase-Isomaltase/metabolismo
2.
Curr Opin Gastroenterol ; 36(2): 101-109, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31990709

RESUMO

PURPOSE OF REVIEW: Disaccharidase testing, as applied to the evaluation of gastrointestinal disturbances is available but it is not routinely considered in the diagnostic work-up. The purpose of this review was to determine if disaccharidase testing is clinically useful and to consider how the results could alter patient management. RECENT FINDINGS: Indicate that carbohydrate maldigestion could contribute functional bowel disorders and negatively impact the fecal microbiome. Diagnostic techniques include enzyme activity assays performed on random endoscopically obtained small intestinal biopsies, immunohistochemistry, stable isotope tracer and nonenriched substrate load breath testing, and genetic testing for mutations. More than 40 sucrase--isomaltase gene variants coding for defective or reduced enzymatic activity have been reported and deficiency conditions are more common than previously thought. SUMMARY: The rationale for disaccharidase activity testing relates to a need to fully assess unexplained recurrent abdominal discomfort and associated symptoms. All disaccharidases share the same basic mechanism of mucosal expression and deficiency has far reaching consequences. Testing for disaccharidase expression appears to have an important role in symptom evaluation, but there are accuracy and logistical issues that should be considered. It is likely that specific recommendations for patient management, dietary modification, and enzyme supplementation would come from better testing methods.


Assuntos
Dissacaridases/análise , Gastroenteropatias/diagnóstico , Dissacaridases/deficiência , Dissacaridases/metabolismo , Fermentação , Gastroenteropatias/metabolismo , Gastroenteropatias/fisiopatologia , Microbioma Gastrointestinal/fisiologia , Humanos , Síndromes de Malabsorção/diagnóstico , Síndromes de Malabsorção/metabolismo , Síndromes de Malabsorção/fisiopatologia
3.
BMC Gastroenterol ; 20(1): 331, 2020 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-33036568

RESUMO

BACKGROUND: Disaccharides such as lactose and sucrose are sugars commonly found in human diet. They are broken down by mucosal disaccharidases in the duodenum. Previous small studies found no associations between gastrointestinal (GI) symptoms and combined low disaccharidase activity. We aim to explore the associations of low activity of disaccharidase and combinations of low activity of different disaccharidases with general GI symptom presentations in a large cohort of pediatric patients. METHODS: We examined a cohort (0-21 yrs.) who have undergone esophagogastroduodenoscopy and received disaccharidase activity assay from duodenal biopsy in the time period 2010 to 2012. Disaccharidase assays tested for activity of lactase, sucrase, maltase, and palatinase. GI symptoms were grouped into four categories, abdominal pain, diarrhea, weight loss, and gastroesophageal reflux. RESULTS: Of the 347 subjects, we found an association between low lactase activity and abdominal pain (OR = 1.78; 95% CI = 1.07-2.97; p < 0.05). Subjects with a lactase/sucrase ratio < 0.2 were found to be associated with abdominal pain (OR = 2.25; 95% CI = 1.25-4.04; p < 0.05), Subjects with low pandisaccharidase may be correlated with abdominal pain and have a unique frequency of GI symptoms due to low frequency of diarrhea and weight loss, but they were not statistically significant. CONCLUSIONS: Low activities of certain disaccharidase combinations may be associated with GI symptoms in subjects; a prospective study may be needed to investigate further.


Assuntos
Dissacaridases , Lactase , Criança , Duodeno , Humanos , Estudos Prospectivos , Estudos Retrospectivos
4.
FASEB J ; 32(7): 3903-3911, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29465310

RESUMO

Dietary starch is finally converted to glucose for absorption by the small intestine mucosal α-glucosidases (sucrase-isomaltase [SI] and maltase-glucoamylase), and control of this process has health implications. Here, the molecular mechanisms were analyzed associated with starch-triggered maturation and transport of SI. Biosynthetic pulse-chase in Caco-2 cells revealed that the high MW SI species (265 kDa) induced by maltose (an α-amylase starch digestion product) had a higher rate of early trafficking and maturation compared with a glucose-induced SI (245 kDa). The maltose-induced SI was found to have higher affinity to lipid rafts, which are associated with enhanced targeting to the apical membrane and higher activity. Accordingly, in situ maltose-hydrolyzing action was enhanced in the maltose-treated cells. Thus, starch digestion products at the luminal surface of small intestinal enterocytes are sensed and accelerate the intracellular processing of SI to enhance starch digestion capacity in the intestinal lumen.-Chegeni, M., Amiri, M., Nichols, B. L., Naim, H. Y., Hamaker, B. R. Dietary starch breakdown product sensing mobilizes and apically activates α-glucosidases in small intestinal enterocytes.


Assuntos
Enterócitos/metabolismo , Amido/metabolismo , alfa-Glucosidases/metabolismo , Células CACO-2 , Humanos , Intestino Delgado/citologia , Maltose/metabolismo , Microdomínios da Membrana/metabolismo , Transdução de Sinais
5.
Proc Natl Acad Sci U S A ; 113(21): 6035-40, 2016 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-27162343

RESUMO

The primary sweet sensor in mammalian taste cells for sugars and noncaloric sweeteners is the heteromeric combination of type 1 taste receptors 2 and 3 (T1R2+T1R3, encoded by Tas1r2 and Tas1r3 genes). However, in the absence of T1R2+T1R3 (e.g., in Tas1r3 KO mice), animals still respond to sugars, arguing for the presence of T1R-independent detection mechanism(s). Our previous findings that several glucose transporters (GLUTs), sodium glucose cotransporter 1 (SGLT1), and the ATP-gated K(+) (KATP) metabolic sensor are preferentially expressed in the same taste cells with T1R3 provides a potential explanation for the T1R-independent detection of sugars: sweet-responsive taste cells that respond to sugars and sweeteners may contain a T1R-dependent (T1R2+T1R3) sweet-sensing pathway for detecting sugars and noncaloric sweeteners, as well as a T1R-independent (GLUTs, SGLT1, KATP) pathway for detecting monosaccharides. However, the T1R-independent pathway would not explain responses to disaccharide and oligomeric sugars, such as sucrose, maltose, and maltotriose, which are not substrates for GLUTs or SGLT1. Using RT-PCR, quantitative PCR, in situ hybridization, and immunohistochemistry, we found that taste cells express multiple α-glycosidases (e.g., amylase and neutral α glucosidase C) and so-called intestinal "brush border" disaccharide-hydrolyzing enzymes (e.g., maltase-glucoamylase and sucrase-isomaltase). Treating the tongue with inhibitors of disaccharidases specifically decreased gustatory nerve responses to disaccharides, but not to monosaccharides or noncaloric sweeteners, indicating that lingual disaccharidases are functional. These taste cell-expressed enzymes may locally break down dietary disaccharides and starch hydrolysis products into monosaccharides that could serve as substrates for the T1R-independent sugar sensing pathways.


Assuntos
Dissacarídeos/farmacologia , Regulação Enzimológica da Expressão Gênica/fisiologia , Papilas Gustativas/enzimologia , Paladar/fisiologia , alfa-Glucosidases/biossíntese , Animais , Proteínas Facilitadoras de Transporte de Glucose/genética , Proteínas Facilitadoras de Transporte de Glucose/metabolismo , Camundongos , Camundongos Transgênicos , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Transportador 1 de Glucose-Sódio/genética , Transportador 1 de Glucose-Sódio/metabolismo , alfa-Glucosidases/genética
6.
J Pediatr Gastroenterol Nutr ; 66 Suppl 3: S24-S29, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29762372

RESUMO

The mucosal maltase enzymes are characterized by an activity that produces glucose from linear glucose polymers, assayed with the disaccharide maltose. The related enzyme isomaltase produces glucose from branched glucose polymers, assayed with palatinose. Maltase and isomaltase activities are part of the 4 disaccharidases assayed from clinical duodenal biopsy homogenates. The reported maltase activities are more difficult to interpret than lactase or sucrase activities because both the sucrase-isomaltase and maltase-glucoamylase proteins have overlapping maltase activities. The early work of Dahlqvist identified 4 maltase activities from human small intestinal mucosa. On one peptide, sucrase (maltase Ib) and isomaltase (maltase Ia) activities shared maltase activities but identified the enzymes as sucrase-isomaltase. On the other peptide, no distinguishing characteristics of the 2 maltase activities (maltases II and III) were detected and the activities identified as maltase-glucoamylase. The nutritional/clinical importance of small intestinal maltase and isomaltase activities are due to their crucial role in the digestion of food starches to absorbable free glucose. This review focuses on the interpretation of biopsy maltase activities in the context of reported lactase, sucrase, maltase, and palatinase biopsy assay activity patterns. We present a classification of mucosal maltase deficiencies and novel primary maltase deficiency (Ib, II, III) and provide a clarification of the role of maltase activity assayed from clinically obtained duodenal biopsies, as a path toward future clinical and molecular genomic investigations.


Assuntos
Mucosa Intestinal/enzimologia , alfa-Glucosidases/deficiência , Animais , Digestão/fisiologia , Humanos , Mucosa Intestinal/metabolismo , Mutação , alfa-Glucosidases/análise , alfa-Glucosidases/metabolismo
7.
J Pediatr Gastroenterol Nutr ; 66 Suppl 3: S52-S55, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29762379

RESUMO

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.


Assuntos
Dissacaridases/deficiência , Duodeno/enzimologia , Dispepsia/etiologia , Mucosa Intestinal/enzimologia , Síndromes de Malabsorção/epidemiologia , Adolescente , Criança , Pré-Escolar , Duodeno/patologia , Endoscopia do Sistema Digestório , Feminino , Humanos , Mucosa Intestinal/patologia , Síndromes de Malabsorção/complicações , Síndromes de Malabsorção/diagnóstico , Masculino , Estudos Prospectivos , Estudos Retrospectivos
8.
J Pediatr Gastroenterol Nutr ; 66 Suppl 3: S61-S64, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29762381

RESUMO

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.


Assuntos
Testes Respiratórios/métodos , Erros Inatos do Metabolismo dos Carboidratos/diagnóstico , Duodeno/enzimologia , Amido/metabolismo , Complexo Sacarase-Isomaltase/deficiência , Adolescente , Isótopos de Carbono/metabolismo , Estudos de Casos e Controles , Criança , Pré-Escolar , Digestão/fisiologia , Feminino , Humanos , Lactente , Masculino , Complexo Sacarase-Isomaltase/análise
9.
J Pediatr Gastroenterol Nutr ; 65(2): e35-e42, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28267073

RESUMO

BACKGROUND AND OBJECTIVE: Although named because of its sucrose hydrolytic activity, this mucosal enzyme plays a leading role in starch digestion because of its maltase and glucoamylase activities. Sucrase-deficient mutant shrews, Suncus murinus, were used as a model to investigate starch digestion in patients with congenital sucrase-isomaltase deficiency.Starch digestion is much more complex than sucrose digestion. Six enzyme activities, 2 α-amylases (Amy), and 4 mucosal α-glucosidases (maltases), including maltase-glucoamylase (Mgam) and sucrase-isomaltase (Si) subunit activities, are needed to digest starch to absorbable free glucose. Amy breaks down insoluble starch to soluble dextrins; mucosal Mgam and Si can either directly digest starch to glucose or convert the post-α-amylolytic dextrins to glucose. Starch digestion is reduced because of sucrase deficiency and oral glucoamylase enzyme supplement can correct the starch maldigestion. The aim of the present study was to measure glucogenesis in suc/suc shrews after feeding of starch and improvement of glucogenesis by oral glucoamylase supplements. METHODS: Sucrase mutant (suc/suc) and heterozygous (+/suc) shrews were fed with C-enriched starch diets. Glucogenesis derived from starch was measured as blood C-glucose enrichment and oral recombinant C-terminal Mgam glucoamylase (M20) was supplemented to improve starch digestion. RESULTS: After feedings, suc/suc and +/suc shrews had different starch digestions as shown by blood glucose enrichment and the suc/suc had lower total glucose concentrations. Oral supplements of glucoamylase increased suc/suc total blood glucose and quantitative starch digestion to glucose. CONCLUSIONS: Sucrase deficiency, in this model of congenital sucrase-isomaltase deficiency, reduces blood glucose response to starch feeding. Supplementing the diet with oral recombinant glucoamylase significantly improved starch digestion in the sucrase-deficient shrew.


Assuntos
Erros Inatos do Metabolismo dos Carboidratos/tratamento farmacológico , Suplementos Nutricionais , Digestão/fisiologia , Fármacos Gastrointestinais/uso terapêutico , Glucana 1,4-alfa-Glucosidase/uso terapêutico , Amido/metabolismo , Complexo Sacarase-Isomaltase/deficiência , Sacarase/deficiência , Administração Oral , Animais , Animais Geneticamente Modificados , Biomarcadores/metabolismo , Glicemia/metabolismo , Erros Inatos do Metabolismo dos Carboidratos/metabolismo , Masculino , Distribuição Aleatória , Musaranhos , Complexo Sacarase-Isomaltase/metabolismo , Resultado do Tratamento
10.
Proc Natl Acad Sci U S A ; 110(15): E1380-9, 2013 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-23520048

RESUMO

Efficient wound healing is required to maintain the integrity of the intestinal epithelial barrier because of its constant exposure to a large variety of environmental stresses. This process implies a partial cell depolarization and the acquisition of a motile phenotype that involves rearrangements of the actin cytoskeleton. Here we address how polarized enterocytes harboring actin-rich apical microvilli undergo extensive cell remodeling to drive injury repair. Using live imaging technologies, we demonstrate that enterocytes in vitro and in vivo rapidly depolarize their microvilli at the wound edge. Through its F-actin-severing activity, the microvillar actin-binding protein villin drives both apical microvilli disassembly in vitro and in vivo and promotes lamellipodial extension. Photoactivation experiments indicate that microvillar actin is mobilized at the lamellipodium, allowing optimal migration. Finally, efficient repair of colonic mechanical injuries requires villin severing of F-actin, emphasizing the importance of villin function in intestinal homeostasis. Thus, villin severs F-actin to ensure microvillus depolarization and enterocyte remodeling upon injury. This work highlights the importance of specialized apical pole disassembly for the repolarization of epithelial cells initiating migration.


Assuntos
Actinas/química , Enterócitos/citologia , Proteínas dos Microfilamentos/fisiologia , Actinas/metabolismo , Animais , Apoptose , Diferenciação Celular , Linhagem Celular , Movimento Celular , Proliferação de Células , Endoscopia , Enterócitos/metabolismo , Feminino , Mucosa Intestinal/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas dos Microfilamentos/metabolismo , Microvilosidades/metabolismo , Fenótipo , Suínos , Cicatrização
11.
J Clin Biochem Nutr ; 54(1): 55-60, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24426192

RESUMO

Using the small intestine enterocyte Caco-2 cell model, sucrase-isomaltase (SI, the mucosal α-glucosidase complex) expression and modification were examined relative to exposure to different mono- and disaccharide glycemic carbohydrates. Caco-2/TC7 cells were grown on porous supports to post-confluence for complete differentiation, and dietary carbohydrate molecules of glucose, sucrose (disaccharide of glucose and fructose), maltose (disaccharide of two glucoses α-1,4 linked), and isomaltose (disaccharide of two glucoses α-1,6 linked) were used to treat the cells. qRT-PCR results showed that all the carbohydrate molecules induced the expression of the SI gene, though maltose (and isomaltose) showed an incremental increase in mRNA levels over time that glucose did not. Western blot analysis of the SI protein revealed that only maltose treatment induced a higher molecular weight band (Mw ~245 kDa), also at higher expression level, suggesting post-translational processing of SI, and more importantly a sensing of maltose. Further work is warranted regarding this putative sensing response as a potential control point for starch digestion and glucose generation in the small intestine.

12.
J Biol Chem ; 287(44): 36917-21, 2012 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-22988246

RESUMO

The quality of starch digestion, related to the rate and extent of release of dietary glucose, is associated with glycemia-related problems such as diabetes and other metabolic syndrome conditions. Here, we found that the rate of glucose generation from starch is unexpectedly associated with mucosal α-glucosidases and not just α-amylase. This understanding could lead to a new approach to regulate the glycemic response and glucose-related physiologic responses in the human body. There are six digestive enzymes for starch: salivary and pancreatic α-amylases and four mucosal α-glucosidases, including N- and C-terminal subunits of both maltase-glucoamylase and sucrase-isomaltase. Only the mucosal α-glucosidases provide the final hydrolytic activities to produce substantial free glucose. We report here the unique and shared roles of the individual α-glucosidases for α-glucans persisting after starch is extensively hydrolyzed by α-amylase (to produce α-limit dextrins (α-LDx)). All four α-glucosidases share digestion of linear regions of α-LDx, and three can hydrolyze branched fractions. The α-LDx, which were derived from different maize cultivars, were not all equally digested, revealing that the starch source influences glucose generation at the mucosal α-glucosidase level. We further discovered a fraction of α-LDx that was resistant to the extensive digestion by the mucosal α-glucosidases. Our study further challenges the conventional view that α-amylase is the only rate-determining enzyme involved in starch digestion and better defines the roles of individual and collective mucosal α-glucosidases. Strategies to control the rate of glucogenesis at the mucosal level could lead to regulation of the glycemic response and improved glucose management in the human body.


Assuntos
Metabolismo dos Carboidratos , Dextrinas/química , Glucose/química , Mucosa/enzimologia , alfa-Glucosidases/química , Animais , Humanos , Hidrólise , Cinética , Camundongos , Peso Molecular , Subunidades Proteicas/química , Amido/química , Zea mays/química , alfa-Amilases/química
13.
J Biol Chem ; 287(38): 31929-38, 2012 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-22851177

RESUMO

Starch digestion involves the breakdown by α-amylase to small linear and branched malto-oligosaccharides, which are in turn hydrolyzed to glucose by the mucosal α-glucosidases, maltase-glucoamylase (MGAM) and sucrase-isomaltase (SI). MGAM and SI are anchored to the small intestinal brush-border epithelial cells, and each contains a catalytic N- and C-terminal subunit. All four subunits have α-1,4-exohydrolytic glucosidase activity, and the SI N-terminal subunit has an additional exo-debranching activity on the α-1,6-linkage. Inhibition of α-amylase and/or α-glucosidases is a strategy for treatment of type 2 diabetes. We illustrate here the concept of "toggling": differential inhibition of subunits to examine more refined control of glucogenesis of the α-amylolyzed starch malto-oligosaccharides with the aim of slow glucose delivery. Recombinant MGAM and SI subunits were individually assayed with α-amylolyzed waxy corn starch, consisting mainly of maltose, maltotriose, and branched α-limit dextrins, as substrate in the presence of four different inhibitors: acarbose and three sulfonium ion compounds. The IC(50) values show that the four α-glucosidase subunits could be differentially inhibited. The results support the prospect of controlling starch digestion rates to induce slow glucose release through the toggling of activities of the mucosal α-glucosidases by selective enzyme inhibition. This approach could also be used to probe associated metabolic diseases.


Assuntos
Glucose/metabolismo , Mucosa/enzimologia , alfa-Glucosidases/metabolismo , Animais , Diabetes Mellitus/metabolismo , Drosophila melanogaster , Glicosídeo Hidrolases/química , Glicosilação , Humanos , Hidrólise , Concentração Inibidora 50 , Mucosa Intestinal/metabolismo , Cinética , Camundongos , Modelos Químicos , Obesidade/metabolismo , Estrutura Terciária de Proteína , Proteínas Recombinantes/química
15.
J Pediatr Gastroenterol Nutr ; 57(6): 704-12, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23838818

RESUMO

OBJECTIVES: Six enzyme activities are needed to digest starch to absorbable free glucose; 2 luminal α-amylases (AMY) and 4 mucosal maltase-glucoamylase (MGAM) and sucrase-isomaltase (SI) subunit activities are involved in the digestion. The AMY activities break down starch to soluble oligomeric dextrins; mucosal MGAM and SI can either directly digest starch to glucose or convert the post-α-amylolytic dextrins to glucose. We hypothesized that MGAM, with higher maltase than SI, drives digestion on ad limitum intakes and SI, with lower activity but more abundant amount, constrains ad libitum starch digestion. METHODS: Mgam null and wild-type (WT) mice were fed with starch diets ad libitum and ad limitum. Fractional glucogenesis (fGG) derived from starch was measured and fractional gluconeogenesis and glycogenolysis were calculated. Carbohydrates in small intestine were determined. RESULTS: After ad libitum meals, null and WT had similar increases of blood glucose concentration. At low intakes, null mice had less (f)GG (P = 0.02) than WT mice, demonstrating the role of Mgam activity in ad limitum feeding; null mice did not reduce fGG responses to ad libitum intakes demonstrating the dominant role of SI activity during full feeding. Although fGG was rising after feeding, fractional gluconeogenesis fell, especially for null mice. CONCLUSIONS: The fGNG (endogenous glucogenesis) in null mice complemented the fGG (exogenous glucogenesis) to conserve prandial blood glucose concentrations. The hypotheses that Mgam contributes a high-efficiency activity on ad limitum intakes and SI dominates on ad libitum starch digestion were confirmed.


Assuntos
Carboidratos da Dieta/metabolismo , Digestão , Gluconeogênese , Glucose/metabolismo , Amido/metabolismo , Complexo Sacarase-Isomaltase/metabolismo , alfa-Glucosidases/metabolismo , Animais , Glicemia/metabolismo , Digestão/genética , Mucosa Intestinal/enzimologia , Mucosa Intestinal/metabolismo , Intestino Delgado/enzimologia , Intestino Delgado/metabolismo , Camundongos , Camundongos Knockout , Mutação , Período Pós-Prandial , alfa-Glucosidases/genética
16.
Bioorg Med Chem ; 19(13): 3929-34, 2011 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-21669536

RESUMO

Inhibition of intestinal α-glucosidases and pancreatic α-amylases is an approach to controlling blood glucose and serum insulin levels in individuals with Type II diabetes. The two human intestinal glucosidases are maltase-glucoamylase and sucrase-isomaltase. Each incorporates two family 31 glycoside hydrolases responsible for the final step of starch hydrolysis. Here we compare the inhibition profiles of the individual N- and C-terminal catalytic subunits of both glucosidases by clinical glucosidase inhibitors, acarbose and miglitol, and newly discovered glucosidase inhibitors from an Ayurvedic remedy used for the treatment of Type II diabetes. We show that features of the compounds introduce selectivity towards the subunits. Together with structural data, the results enhance the understanding of the role of each catalytic subunit in starch digestion, helping to guide the development of new compounds with subunit specific antidiabetic activity. The results may also have relevance to other metabolic diseases such as obesity and cardiovascular disease.


Assuntos
Amido/metabolismo , Complexo Sacarase-Isomaltase/metabolismo , alfa-Glucosidases/metabolismo , 1-Desoxinojirimicina/análogos & derivados , 1-Desoxinojirimicina/química , 1-Desoxinojirimicina/farmacologia , Acarbose/química , Acarbose/farmacologia , Domínio Catalítico , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Inibidores de Glicosídeo Hidrolases , Cinética , Monossacarídeos/química , Compostos de Selênio/química , Compostos de Selênio/farmacologia , Complexo Sacarase-Isomaltase/antagonistas & inibidores , Álcoois Açúcares/química , Álcoois Açúcares/farmacologia , Sulfatos/química , Sulfatos/farmacologia
17.
JPGN Rep ; 2(1): e027, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37206939

RESUMO

Human breath is an easily, noninvasively obtained substance. It offers insight into metabolism and is used to diagnose disaccharide malabsorption, infection, small bowel bacterial over growth, and transit times. Herein, we discuss the readily available clinical breath tests, how they function, how they are administered and interpreted and some pitfalls in their use.

18.
Nutrition ; 78: 110857, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32599415

RESUMO

OBJECTIVES: Maltase-glucoamylase (Mgam) and sucrase-isomaltase (Si) are mucosal α-glucosidases required for the digestion of starch to glucose. We hypothesized that a dietary approach to reduce Mgam and Si activities can reduce glucose generation and absorption, and improve glucose control. METHODS: Rice starch was entrapped in alginate microspheres to moderate in vitro digestion properties. Three groups of 8-wk old mice (n = 8) were conditioned for 7 d with low 13C-starch-based materials differing in digestion rates (fast, slow, and slower), and then given a digestible 13C-labeled cornstarch test feeding to determine its digestion to glucose. RESULTS: Conditioning of the small intestine with the slowly digestible starches for 7 d reduced jejunal α-glucosidase and sucrase activities, as well as glucose absorption for the slowly digestible starch slower group (P < 0.01). A correlative relationship was found between glucose absorption from a cornstarch test feeding given at d 7 and jejunal α-glucosidase and sucrase activities (R2 = 0.64; 0.67). However, total prandial glucose levels during the 2-h feeding period did not differ. CONCLUSIONS: Decreased glucogenesis from a digestible starch feeding was found in mice conditioned on slowly digestible starch diets, suggesting that a dietary approach incorporating slowly digestible starches may change α-glucosidase activities to moderate glucose absorption rate.


Assuntos
Digestão , alfa-Glucosidases , Animais , Dieta , Glucose , Camundongos , Amido
19.
J Nutr ; 139(1): 188-91, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19056811

RESUMO

The Children's Nutrition Research Center (CNRC) is a unique cooperative venture among Baylor College of Medicine, Texas Children's Hospital, and the USDA/Agricultural Research Service. The CNRC is dedicated to defining the nutrient needs of children, from conception through adolescence, and the needs of pregnant women and nursing mothers. Scientific data from the Center enable healthcare providers and policy advisors to make dietary recommendations that improve the health of today's children and that of generations to come. CNRC research has already impacted feeding guidelines for normal U.S. children and all children of the world.


Assuntos
Ciências da Nutrição Infantil/história , United States Department of Agriculture/história , Pesquisa Biomédica/história , Pesquisa Biomédica/legislação & jurisprudência , Criança , Ciências da Nutrição Infantil/legislação & jurisprudência , História do Século XX , Hospitais/história , Humanos , Faculdades de Medicina/história , Texas , Estados Unidos
20.
J Nutr ; 139(4): 684-90, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19193815

RESUMO

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.


Assuntos
Digestão , Glucose/metabolismo , Homeostase , Amido/metabolismo , alfa-Glucosidases/metabolismo , Ração Animal , Animais , Jejum , Genótipo , Insulina/sangue , Camundongos , Camundongos Knockout , Mucosa/enzimologia , Sacarase/metabolismo , alfa-Glucosidases/deficiência , alfa-Glucosidases/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA