Download as pdf or txt
Download as pdf or txt
You are on page 1of 7

ORIGINAL ARTICLE: HEPATOLOGY

AND

NUTRITION

Gluten-free Sourdough Wheat Baked Goods Appear Safe for Young Celiac Patients: A Pilot Study

Raffaella Di Cagno, yMaria Barbato, yChiara Di Camillo, Carlo Giuseppe Rizzello, Maria De Angelis, zGiammaria Giuliani, Massimo De Vincenzi, Marco Gobbetti, and ySalvatore Cucchiara

ABSTRACT
Objectives: This study was aimed at showing the safety, for young patients with celiac disease (CD), of sweet baked goods made of wheat our, which was rendered gluten-free during sourdough fermentation. Methods and Results: As shown by R5 antibody-based sandwich and competitive enzyme-linked immunosorbent assay (ELISA), selected lactobacilli and fungal proteases, routinely used in bakeries, degraded gluten to <10 ppm during sourdough fermentation. The resulting our was mainly a mixture of water-/salt-soluble low-size peptides and free amino acids. Gliadin and glutenin fractions extracted from the pepsintrypsin (PT) digest of the fermented wheat our induced the expression of interferon (IFN)-g at the level comparable with the negative control. After fermentation, the wheat our was spray dried and used for making sweet baked goods. Eight patients with CD in remission were enrolled for the clinical challenge, and they daily consumed 200 g of sweet baked goods equivalent to 10 g of native gluten. Hematology, serology (total serum IgA, IgG and IgA antigluten, endomysial and tissue transglutaminase IgA antibodies), and intestinal permeability analyses were carried out over time. One patient interrupted the trial after 15 days and another after 30 days only due to difculties in the compliance of the daily consumption. All of the other patients showed normal values of hematology, serology, and intestinal permeability during 60 days of challenge. Conclusions: This study showed that a wheat ourfermented product, having gluten completely degraded, is not toxic for patients with CD. Nevertheless, these foods should not be recommended for patients with celiac disease until a formal trial has been done. Key Words: celiac disease, lactic acid bacteria, sourdough-baked goods

(JPGN 2010;51: 777783) eliac disease (CD) (gluten-sensitive enteropathy) is a chronic gastrointestinal tract disorder in which ingestion of gluten, the protein network present in food products made from wheat, rye, and barley, and their cross-related varieties, leads to damage of the small intestinal mucosa by an autoimmune mechanism in
Received February 19, 2010; accepted July 13, 2010. From the Department of Plant Protection and Applied Microbiology, University of Bari, Bari, the yDipartimento di Pediatria, La Sapienza Universita di Roma, Rome, the zGiuliani S.p.A., Milan and the Istituto ` Superiore di Sanita, Laboratorio di Metabolismo e Biochimica Patolo` gica, Rome, Italy. Address correspondence and reprint requests to Carlo G. Rizzello, Department of Plant Protection and Applied Microbiology, University of Bari, 70126 Bari, Italy (e-mail: rizzello@agr.uniba.it). The authors report no conicts of interest. Copyright # 2010 by European Society for Pediatric Gastroenterology, Hepatology, and Nutrition and North American Society for Pediatric Gastroenterology, Hepatology, and Nutrition DOI: 10.1097/MPG.0b013e3181f22ba4

genetically susceptible individuals (1). The gluten network is formed while mixing flour and water by interactions between gliadins and glutenins. Gluten is an unusual protein because it is consumed in relatively large amounts, is partially resistant to luminal digestion by gastric, pancreatic, and brush border enzymes in the human small intestine, and when absorbed, it is susceptible to posttranslational modification (deamidation) by mucosal transglutaminase (1). Proline mainly imparts resistance to digestion because many proteases are unable to cleave peptide bonds located at N- and C-termini of proline (2,3). The variability of the gluten network is extremely large; public databases such as GenBank include more than 200 entries for genes encoding gluten proteins in Triticum aestivum. Epidemiology of CD is increasing ($1% of Europeans and North Americans) (1), as well as the prevalence of CD in elderly people (4). During the last decades, cereal food technology changed extensively to modify the dietary habits of entire populations, previously nave to massive gluten exposure. Cereal baked goods are manufactured by fast processes in which long-time fermentation by sourdough, a cocktail of acidifying and proteolytic lactic acid bacteria, has been almost completely replaced by the use of chemical and/or bakers yeast leavening agents. Under these technological conditions, cereal components (eg, proteins) are not degraded during manufacture (5). The gluten-free diet (GFD) is effective and safe, and at present, it is the only available treatment for CD. Nevertheless, development and strict adherence to GFD are also complicated by formidable economical, cultural, and distribution difficulties in developing countries (6). Despite the proven benefits of the GFD, it may be exceedingly difficult to completely avoid gluten-containing foods, and effective adherence to the GFD is estimated to be only 45% to 80% (7). Beyond other therapeutic options, oral supplementation with microbial oligopeptidases is proposed as an alternative to the GFD (813). In the last decade, several studies (1419) were carried out in the authors laboratory aimed at showing the capacity of proteolytic enzymes, mainly peptidases, of selected sourdough lactobacilli to degrade gluten during food processing. More recently, it was shown that selected sourdough lactobacilli, in combination with fungal proteases, decreased the residual concentration of gluten (T aestivum and Triticum durum flours) to <10 ppm during sourdough fermentation (13,19). This study was aimed at showing the safety, for young patients with CD, of sweet baked goods made of wheat flour that were rendered gluten-free during sourdough fermentation. Preliminarily, the hydrolyzed wheat flour was characterized through immunological and ex vivo analyses. Furthermore, sweet baked goods, containing the equivalent of $10 g of native gluten, were administered daily to 8 patients with CD for 60 days following a proof-of-concept open study. Hematology, serology, and intestinal permeability analyses were carried out.

JPGN

Volume 51, Number 6, December 2010

777

Copyright 2010 by ESPGHAN and NASPGHAN. Unauthorized reproduction of this article is prohibited.

Di Cagno et al

JPGN

Volume 51, Number 6, December 2010

MATERIALS AND METHODS Microorganisms and Enzymes


Lactobacillus sanfranciscensis 7A, LS3, LS10, LS19, LS23, LS38, and LS47, Lactobacillus alimentarius 15 mol/L, Lactobacillus brevis 14G, and Lactobacillus hilgardii 51B were selected based on their peptidase activities, with particular reference to degradation of Pro-rich peptides (14,20). Strains were propagated for 24 hours at 308C in modified MRS broth (Oxoid, Basingstoke, UK), with the addition of fresh yeast extract (5%, v/v) and 28 mmol/L maltose at a final pH of 5.6 (modified MRS). When used for sourdough fermentation, cells of lactobacilli were cultivated until the late exponential phase of growth was reached ($12 hours). Fungal proteases from Aspergillus oryzae (500,000 hemoglobin units on the tyrosine basis per gram) and Aspergillus niger (3000 spectrophotometric acid protease units/g), routinely used as improvers in the bakery industry, were purchased from BIO-CAT Inc. (Troy, VA).

Proteins were selectively extracted from flour according to the method of Weiss et al (21). Two-dimensional electrophoresis of proteins extracted from various fractions was carried out with the immobiline-polyacrylamide system (14). Gels were silver stained, and spot intensities were normalized as reported by Bini et al (22). Multidimensional HPLC (MDLC) coupled with electrospray ionization (ESI)-ion trap mass spectrometry (MS) was used to detect epitopes (13). The protein concentration of the flours used for making baked goods was determined by the Bradford method (23). The organic nitrogen concentration was determined using the Kjeldahl method. Free amino acids were analyzed by a series 30 Amino Acid Analyzer (Biochrom Ltd, Cambridge Science Park, UK).

Ex Vivo Assays
Gliadins and glutenins, extracted from fermented and nonfermented wheat flour, were subjected to sequential pepsin-trypsin (PT) hydrolysis to simulate the in vivo digestion (18). After digestion, PT digests were heated at 1008C for 30 minutes to inactivate enzymes and freeze-dried for further analysis. Duodenal biopsies were obtained from 10 CD patients on a GFD (age range 8 to 17 years). All of the patients with CD expressed the HLA-DQ2 phenotype. CD was diagnosed according to European Society for Paediatric Gastroenterology, Hepatology, and Nutrition (ESPGHAN) criteria (24). Immediately after excision, all of the biopsies were placed into RPMI-1640 (GibcoInvitrogen, Paisley, UK) ice-chilled culture medium, which is used largely for cultivating mammalian cells and/or for handling and transport of biopsies (3060 minutes) (13,25). Within 30 minutes, duodenal biopsies were cultured for 4 hours using the organ culture method originally described by Browing and Trier (26). Biopsies from each patient with CD were cultured with PT digest (5 mg/mL) of gliadin/glutenins from fermented or nonfermented wheat flour or with medium alone (negative control). Tissue samples were taken and kept in RNAlater (Qiagen GmbH, Hilden, Germany) to preserve RNA. Total RNA was extracted from tissue using the Rneasy Mini Kit (Qiagen GmbH) according to the manufacturers instructions. Concentration of mRNA was estimated by ultraviolet absorbance at 260 nm. Aliquots of total RNA (500 ng) were reverse transcribed using random hexamers and the TaqMan Reverse Transcription Reagents (Applied Biosystems, Monza, Italy) with 3.125 U/mL of MultiScribe Reverse Transcriptase in a final volume of 50 mL. cDNA samples were stored at 208C. Quantitative real-time polymerase chain reaction (RT-PCR) for interferon (IFN)-g gene was carried out in 96-well plates on the ABI Prism 7500HT Fast Sequence Detection System (Applied Biosystems). TaqMan Gene Expression Assay for IFN-g gene and Pre-Developed TaqMan Assay for glyceraldehyde-3-phosphate dehydrogenase (GAPDH, endogenous control) gene (Applied Biosystems) with the provided primers (Appled Biosystems) were used. The assays were carried out using 20 mix of PCR primers and TaqMan Minor Groove Binder 6-FAM dye labeled probes with a nonfluorescent quencher at the 30 end of the probe as described by De Angelis et al (13). Six serial dilutions (200.1 ng/mL) of IFN-g cDNA were used as a template for each primer/probe set. The standard curve was generated by plotting the threshold cycle (CT) values against the log of the amount of cDNA. The average value of the target gene was normalized using the GAPDH gene. A normal duodenal biopsy was used as calibrator in all of the experiments. IFN-g proteins secreted in the supernatant were quantified by ELISA in 96-well, round-bottomed plates (Tema Ricerca, Milan, Italy), according to the manufacturers recommendations (13). www.jpgn.org

Hydrolysis of Gluten During Sourdough Fermentation and Making Sweet Baked Goods
The main characteristics of the wheat (T aestivum cv Appulo) flour used were as follows: moisture, 12.8%; protein, 10.3% of dry matter (d.m.); fat, 1.8% of d.m.; ash, 0.6% of d.m.; and total carbohydrates, 74.5% of d.m. Wheat flour and tap water (ratio 1:4), containing ca 1 109 CFU/g of lactobacilli cells and 200 ppm of both fungal proteases, were used for sourdough fermentation at 378C for 48 hours under stirring conditions ($200 rpm). After fermentation, wheat sourdough was subjected to spray drying to remove water, and the resulting hydrolyzed flour was further milled and used. The spray drying method allows to obtain dry powder from liquid or semiliquid matrices by rapidly drying with a hot gas, and it is commonly applied to thermally sensitive materials (eg, foods, pharmaceuticals). The following formula was used for making sweet baked goods at the pilot plant of the Department of Plant Protection and Applied Microbiology: fermented wheat flour (50%, w/w), sucrose (11.3%, w/w), eggs (8.8%, w/w), pasteurized yolk (1.2%, w/w), egg whites (13%, w/w), butter (7.3%, w/w), milk cream (7.3%, w/w), sodium bicarbonate (1%, w/w), vanilla bean, and lemon peel as flavoring agents. The dough was whipped with a continuous highspeed bakery mixer (dough mixing time, 5 minutes) (Chopin and Co, Boulogne, Sur-Seine, France), and sweet baked goods (weight of $100 g) were baked at 1608C for 40 minutes (Combo 3, Zucchelli, Verona, Italy).

Immunological Analyses and Characterization of Wheat Flour


Immunological analyses were carried out by using R5 antibody-based sandwich and competitive enzyme-linked immunosorbent assays (ELISAs) and R5 antibody-based Western blotting. The R5 monoclonal antibody and the horseradish peroxidase-conjugated R5 antibody were used for gluten analysis. The R5-based sandwich ELISA was carried out with the Transia plate detection kit following the instructions of the manufacturer (Diffchamb, Vastra, Frolunda, Sweden). The R5-based competitive ELISA was carried out at the gluten unit of the Centro National de Biotecnologia (Madrid, Spain). For R5-based Western blot analysis, after 1dimensional SDS-PAGE, proteins were electrotransferred onto polyvinylidene difluoride membranes. The membranes were incubated directly with R5-horseradish peroxidase antibody, and the blots were developed by immunodetection with ECL Western blotting analysis system (Amersham Pharmacia, Uppsala, Sweden).

778

Copyright 2010 by ESPGHAN and NASPGHAN. Unauthorized reproduction of this article is prohibited.

JPGN

Volume 51, Number 6, December 2010

Gluten-free Baked Goods Safe for Celiac Patients

FIGURE 1. Serology analyses of celiac disease patients at 0 (t0), 30 (t1), and 60 (t2) days of the clinical challenge. Total serum IgA (A); IgA anti-gluten antibodies (IgA-AGA) (B); IgG anti-gluten antibodies (IgG-AGA) (C); and tissue-transglutaminase (IgA-tTG) antibodies (D). Aggregate data are shown in box plots. The center line of the box represents the median (&); the top and bottom of the box represent the 75th and 25th percentile of the data, respectively. The top and bottom of the bars represent the 5th and 95th percentile of the data, respectively. Dotted lines represent the cutoff values provided by the manufacturers.

Protocol of the Clinical Challenge


Eight patients with CD (age median 13, range 817 years) were admitted to the study. Because 2 patients interrupted the clinical challenge (see below), the age median of individuals who followed the trial was 11 years. CD was diagnosed according to ESPGHAN (24). At recruitment all of the patients with CD showed normal values for total serum IgA (0.343.48 g/L) (Fig. 1A). All of the patients were on a GFD for at least 3 years and did not report any other associated diseases. The protocol for recruitment of patients with CD and the clinical challenge were approved by the University Hospital Umberto I, Sapienza University of Rome, Italy. The parents of patients qualified for the study gave their informed consent. All of the enrolled patients with CD were asked to maintain a strict GFD during challenge; the only permitted gluten-containing dietary products were those provided in the study. During the first assessment, volunteers underwent clinical and nutritional evaluation. All patients with CD consumed daily $200 g of sweet baked goods, corresponding to 100 g of processed wheat flour, which www.jpgn.org

contained $10 g of hydrolyzed gluten. The challenge lasted 60 days.

Hematology Analyses
Blood samples were collected as venous blood in evacuated tubes containing K3EDTA (Becton-Dickinson, Rutherford, NJ) and were analyzed within 4 hours. The following parameters were considered: WBC, white blood cell; RBC, red blood cell; HGB, hemoglobin; HCT, hematocrit; MCV, mean corpuscular volume; MCH, mean corpuscular hemoglobin; MCHC, mean corpuscular hemoglobin concentration; RDW-SD, RBC distribution width standard deviation; PLT, platelet count; MPV, mean platelet volume; PDW, platelet distribution width; PCT, plateletcrit; NEUT, neutrophils; LYMPH, lymphocytes; MONO, monocytes; EO, eosinophils; BASO, basophils; LUC, large unstained cells; VES, red blood cell sedimentation rate; PCR, C-reactive protein; glucose, creatinine; total proteins; albumins; a-amylase; lipases; AST/GOT, aspartate aminotransferase/glutamic-oxaloacetic transaminase; ALT/GPT;

779

Copyright 2010 by ESPGHAN and NASPGHAN. Unauthorized reproduction of this article is prohibited.

Di Cagno et al alanine aminotransferase/glutamic-pyruvic transaminase; g-GT, g-glutamyl-transferase; total bilirubin; direct bilirubin; ALP, alkaline phosphatase; total cholesterol; HDL cholesterol, cholesterol high-density lipoprotein; LDL cholesterol, cholesterol lowdensity lipoprotein; triglycerides; ferritin; TSH, thyroid-stimulating hormone; FT4, free thyroxine; FT3, free triiodothyronine; antiTPO, anti-thyroid peroxidase antibodies; and anti-TG, antithyroglobulin antibodies.

JPGN

Volume 51, Number 6, December 2010

TABLE 1. Organic nitrogen concentration (expressed as percentage of total organic nitrogen concentration) of the water-/salt-soluble, gliadin, and glutenin protein fractions extracted from nonfermented and fermented wheat our Organic nitrogen concentration, % Protein fraction Nonfermented wheat our 16.1 0.1 36.9 0.1 47.0 0.2 Fermented wheat our 87.5 0.1 Not detected 12.5 0.2

Serology Analyses
Serum samples were kept frozen at 308C until the assays were carried out. Quantitative determination of total serum IgA was carried out using the UniCAP System (Pharmacia & Upjohn Diagnostics, Uppsala, Sweden). IgG and IgA AGA (anti-gluten antibodies) were determined by commercially available ELISA kits (Eurospital, Trieste, Italy). The cutoff values provided by the manufacturer were 15.0 and 50.0 UA/mL for IgA and IgG AGA, respectively (Fig. 1B and C). Endomysial antibodies (EMA) were determined by an indirect immunofluorescence assay on sections of monkey esophagus (Eurospital). Tissue transglutaminase (tTG) IgA antibodies were measured by commercially available ELISA kits (Eurospital). The cutoff value provided by the manufacturer was 16.0 UA/mL (Fig. 1D).

Water/salt soluble Gliadin Glutenin

Intestinal Permeability
Enrolled patients with CD were hospitalized at the University Hospital Umberto I, La Sapienza University of Rome, Italy, and followed a lactulose- and mannitol-free diet 24 hours before intestinal permeability assay. After an overnight fast, patients with CD voided a urine sample and 1 mL of chlorhexidine 20% (wt/vol) was added to inhibit bacterial growth. Further more, they drank 120 mL of deionized water containing 5.0 g of lactulose and 1.0 g of mannitol. One hour after the test was started, patients were encouraged to drink up to 150 mL of tap water to enhance diuresis. Urine was collected during the next 6 hours following the solution administration. Also in this case, 1 mL of chlorhexidine 20% (wt/vol) was added to samples. The total volume of collected urine was measured, and a 10-mL aliquot was stored at 208C until analysis. Carbohydrates were determined by HPLC according to the method previously described by Lostia et al (27,28). The fractional excretion of lactulose was calculated from the ratio milligrams of lactulose excreted/milligrams of lactulose assumed. The milligrams of lactulose excreted were obtained from milligrams per liter lactulose per liters of urine. The same occurred with mannitol. The values of lactulose and mannitol from samples before administration were subtracted from those of samples collected for 6 hours. Results were expressed as the ratio of the fractional excretion of lactulose to the fractional excretion of mannitol (L/M ratio) (27,28). As reported for healthy children or patients with CD under strict GFD (pediatric age) (2729), values of L/M ratio in the range 0.00 to 0.03 were considered to be normal.

not shown). Two-dimensional electrophoresis of the gliadin fraction did not reveal the presence of protein spots in the fermented wheat flour (data not shown). Only a few spots were detectable for the glutenin fraction (data not shown). No gliadin/glutenin epitopes were found at the lowest detectable concentration (20 ppm) by nano-ESI-MS/MS analysis (data not shown). The lowest detectable concentration was estimated by injecting different concentrations (1100 ppm) of the synthetic epitope QLQPFPQPQLPY together with the fermented wheat flour (19). After fermentation, wheat flour was subjected to spray drying. Compared to nonfermented wheat flour (Table 1), the water-/salt-soluble fraction increased from 16.1% to 87.5% of the total protein fractions (total organic nitrogen content of 1.80% that approximately corresponded [N 5.70] to the protein concentration of 10.26%). This fraction consisted of low-molecular-mass peptides and free amino acids mainly derived from the degradation of all wheat proteins. Indeed, no organic nitrogen was detectable in the gliadin fraction of fermented wheat flour, and the level of glutenin fraction decreased from 47% to 12.5%. The concentration of total free amino acids confirmed these differences ($1032 32 versus 15,320 102 mg/kg for nonfermented and fermented wheat flour, respectively) (P < 0.01).

Cytokine Expression in Duodenal Biopsies of Patients With CD


Gliadins and glutenins were extracted from fermented and nonfermented wheat flour, subjected to PT hydrolysis, pooled, and used to treat duodenal biopsies from patients with CD. Compared to the negative control, the exposure of biopsies to PT digest from nonfermented wheat flour resulted in a significant (P < 0.05) increase of the production of IFN-g mRNA (Fig. 2A and B). The release of IFN-g mRNA by biopsies treated with the PT digest of fermented wheat flour did not significantly (P > 0.05) differ from that of the negative control (medium alone) (Fig. 2A and C). Increased levels of IFN-g protein were found in the culture supernatant of all biopsies. A good agreement was found between proteins and level of mRNA. The highest synthesis of IFN-g proteins was found in biopsies treated with PT digest from nonfermented wheat flour (data not shown).

RESULTS Hydrolysis of Gluten During Sourdough Fermentation


The native concentration of gluten of the wheat (T aestivum cv Appulo) flour was $103,127 123 ppm. After sourdough fermentation by selected sourdough lactobacilli and fungal proteases, the concentration of residual gluten was <10 ppm. This was shown by sandwich and competitive ELISA analyses. The absence of gluten was confirmed by R5 antibody-based Western blotting (data

Clinical Challenge
Eight patients with CD underwent a clinical challenge daily consuming 200 g of sweet baked goods, which contained the equivalent of $10 g of native gluten. At recruitment all of the patients had normal values of hematology and serology parameters. www.jpgn.org

780

Copyright 2010 by ESPGHAN and NASPGHAN. Unauthorized reproduction of this article is prohibited.

JPGN

Volume 51, Number 6, December 2010

Gluten-free Baked Goods Safe for Celiac Patients challenge. All hematology parameters were within the normal values at recruitment. For 60 days, none of the 42 hematology parameters (see Materials and Methods) varied with respect to the initial values. At recruitment all of the patients with CD showed normal values for total serum IgA (0.343.48 g/L) (Fig. 1A). IgG- and IgAAGA and IgA-tTG antibodies of all of the patients with CD were within normal values at all times (Fig. 1BD). The data distribution was highly homogeneous; no outliers were found for any serology parameters over time. In particular, the values for IgA-AGA and IgA-tTG antibodies were <6.0 and 8.0 UA/mL, respectively. Response for EMA antibodies was always negative for all of the patients at all times. When levels of gluten (0.24.3 g), markedly lower than those of this study, were administered daily to numerous children with CD for 60 days, abnormal values of IgG- and IgAAGA, IgA-tTG, and EMA antibodies were found in 90% of individuals (10,3032). The intestinal permeability was determined at recruitment and during clinical challenge up to 60 days. Daily consumption of 200 g of sweet baked goods made with fermented wheat flour (corresponding to $10 g of native gluten) did not cause variation of the intestinal permeability of all of the patients with CD for all times (Fig. 3). All L/M ratios were <0.03. The median value decreased over time. Several reports (33,34) showed abnormal values of intestinal permeability in patients with CD already after a single intake of gluten ($3050 g).

DISCUSSION
Bacterial-derived endopeptidases are recommended as a supplement in the diet to abolish the antigenicity of the 33-mer and other Pro-rich putative immunotoxic epitopes (11,19,35). Problems with the stability of endopeptidases in an acidic gastric environment and efficient mixing with gluten are frequently associated with the oral supplementation (36). Besides, administration of peptidases with the meal did actually enhance the immunoresponse, by exposing more rapidly the small intestinal mucosa to immunopeptides from fragmented gluten proteins (36). The use of microbial peptidases during food processing is not associated with these problems and guarantees the easiest and noninvasive tool to eliminate, in a dose-dependent manner, the toxicity of gliadins and glutenins from wheat (37). This study showed that the combination of fungal proteases, routinely used as bakery improvers, and lactobacilli peptidases completely degrades gluten (<10 ppm) during sourdough fermentation of wheat flour. Fungal proteases are indispensable to generate polypeptides of intermediate dimensions (eg, 440 amino acids) from native proteins that are suddenly transported inside the lactobacilli cells to be hydrolyzed through the complex peptidase system of lactobacilli (19). A large number of intracellular peptidases (eg, PepN, PepO, PEP, PepX, PepT, PepV, PepQ, PepR) are needed to hydrolyze the 33-mer or other synthetic immunogenic polypeptides to free amino acids (13). After hydrolysis, the wheat flour was spray dried and used for making sweet baked goods. Bread, pasta, or sweet baked goods may be manufactured with the wheat flour rendered gluten-free. From a technological point of view, the use of this spray-dried flour has the same technical problems as other naturally gluten-free raw materials (13,16,19). On the contrary, the use of the wheat flour rendered gluten-free has indubitably economical, nutritional, social, and sensory advantages compared with the naturally gluten-free counterpart used in the GFD. As shown by sensory analysis, the properties of sourdough baked goods manufactured with wheat flour rendered gluten-free were almost comparable to those of the full-gluten counterpart; these goods were highly superior to GF baked goods (19).

FIGURE 2. Effect on the expression of mRNA interferon gamma (IFN-g) of the medium alone (RPMI, negative control) (A); PT (pepsin-trypsin) digests of gliadins and glutenins from nonfermented (B) and fermented wheat our (C). GAPDH, glyceraldehyde-3-phosphate dehydrogenase (endogenous control). CD-P1 to10, celiac disease patients. Due to difficulties in the compliance of the daily consumption, 1 patient interrupted the trial after 15 days and another after 30 days of challenge. The other 6 patients reached the endpoint of 60 days. None of the 8 patients with CD had clinical complaints during the www.jpgn.org

781

Copyright 2010 by ESPGHAN and NASPGHAN. Unauthorized reproduction of this article is prohibited.

Di Cagno et al

JPGN

Volume 51, Number 6, December 2010

FIGURE 3. Intestinal permeability of celiac disease patients at 0 (t0), 30 (t1), and 60 (t2) days of the clinical challenge. Aggregate data are shown in box plots. The center line of the box represents the median (&); the top and bottom of the box represent the 75th and 25th percentile of the data, respectively. The top and bottom of the bars represent the 5th and 95th percentile of the data, respectively. The dotted line represents the reference cutoff value for healthy subjects (2729).

mucosa, and especially the appearance of abnormal values of celiac antibodies. All of the patients with CD enrolled in this study showed normal values of hematology and serology during 60 days of clinical challenge. The measurement of the intestinal permeability following an oral dose administration of usually nonabsorbable versus absorbable sugars is the most widely accepted method for monitoring the small intestinal mucosal (tight junctions) leakiness, as in Crohn disease and CD (10,28,41). Even after a single-dose gluten challenge, remarkable abnormal values of the L/M ratio were found in patients with CD under GFD (33,34). All of the patients with CD who daily consumed 200 g of sweet baked goods made with fermented wheat flour showed values of L/M ratio that were $0.03. Overall, this value is considered to be the threshold for normal intestinal permeability in clinical practice (28,29,33). The findings of this study provide the rationale for exploring tools that could reduce the toxicity of gluten for patients with CD beyond the standard GFD. This is the first time that a wheat flour derived product has been shown to be nontoxic after administration for 60 days to patients with CD; nevertheless, these foods should not be recommended for patients with CD until a formal trial has been done. There is no doubt that a period of 60 days, although repeatedly shown to be sufficient to evaluate gluten toxicity in the majority of patients, may not be long enough to evaluate toxicity in all patients with CD who occasionally may show different sensitivity to gluten. Prolonged trials must be planned to state the safety of the baked goods manufactured by applying this promising and rediscovered biotechnology.

After hydrolysis, some glutenin polypeptides persisted in the wheat-fermented flour. Glutenins may contain sequences that activate T cells from the small intestine and result in the secretion of large amounts of IFN-g (12). Based on these findings, the diagnostic value of R5 antibody to detect toxic epitopes may be in part limited. Ex vivo assays based on organ culture of treated intestinal biopsies from 10 patients with CD were used to confirm the absence of toxicity (38). As expected, the PT digest from nonfermented wheat flour caused a marked increase in IFN-g. IFN-g coordinates many aspects of the innate and adaptive immune responses, and it is the principal cytokine produced by a-b CD4 reactive T cells upon gluten activation (12,39). The PT digests of fermented wheat flour induced IFN-g as the negative control. Based on the above results, a clinical challenge was started daily administering 200 g of sweet baked goods made with wheat flour rendered gluten-free (corresponding to $10 g of native gluten). Standardized gluten challenges based on the daily intake of 10 to 20 g were frequently considered, independently of the body weight of enrolled patients with CD (32). The daily intake of 10 to 20 g of gluten was shown to shorten the time of challenge and minimize discomfort (32). Eight patients with CD in remission were enrolled, and hematology, serology, and intestinal permeability analyses were carried out. Even though intestinal biopsy showing the typical picture of flat mucosa is still considered the criterion standard for CD diagnosis, the importance of serology is growing, thanks to reliable tests that were shown to be highly predictive for this condition (37). All of the most complementary serology analyses were carried out in this study. For ethical reasons, no positive controls (patients with CD fed with native gluten) were included in this proof-of-concept open study. Nevertheless, several reports showed that pathological titers of celiac antibodies were detectable within 2 to 7 weeks after reintroduction of gluten in patients with CD (10,30,31,40). Laurin et al (32) demonstrated that levels of gluten (1.7 g/day) caused the deterioration of the intestinal

REFERENCES
1. Tye-Din J, Anderson R. Immunopathogenesis of celiac disease. Curr Gastroenterol Rep 2008;10:45865. 2. Hausch F, Shan L, Santiago NA, et al. Intestinal digestive resistance of immunodominant gliadin peptides. Am J Physiol Gastrointest Liver Physiol 2002;283:9961003. 3. Shan L, Molberg O, Parrot I, et al. Structural basis for gluten intolerance in coeliac sprue. Science 2002;297:22759. 4. Vilppula A, Kaukinen K, Luostarinen L, et al. Increasing prevalence and high incidence of celiac disease in elderly people: a population-based study. BMC Gastroenterol 2009;9:49. 5. Gobbetti M. The sourdough microora: interactions between lactic acid bacteria and yeasts. Trends Food Sci Technol 1998;9:26774. 6. Fasano A, Araya M, Bhatnagar S, et al. Federation of International Societies of Pediatric Gastroenterology, Hepatology, and Nutrition Consensus report on celiac disease. J Pediatr Gastroenterol Nutr 2008;47:2149. 7. Lefer DA, Edwards-George J, Dennis M, et al. Factors that inuence adherence to a gluten-free diet in adults with celiac disease. Dig Dis Sci 2008;53:157381. 8. Catassi C, Fabiani E, Iacono G, et al. A prospective, double-blind, placebo-controlled trial to establish a safe gluten threshold for patients with celiac disease. Am J Clin Nutr 2007;85:1606. 9. Pyle GG, Paaso B, Anderson BE, et al. Effect of pretreatment of food gluten with prolyl endopeptidase on gluten-induced malabsorption in celiac spue. Clin Gastroenterol Hepatol 2005;3:68794. 10. Troncone R, Caputo M, Micillo M, et al. Immunologic and intestinal permeability tests as predictors of relapse during gluten challenge in childhood coeliac disease. Scand J Gastroenterol 1994;29:1447. 11. Mitea C, Havenaar R, Drijfhout JW, et al. Efcient degradation of gluten by a prolyl endoprotease in a gastrointestinal model: implications for coeliac disease. Gut 2008;57:2532. 12. Stepniak D, Spaenij-Dekking L, Mitea C, et al. Highly efcient gluten degradation with a newly identied prolyl endoprotease: implications for celiac disease. Am J Physiol Gastrointest Liver Physiol 2006;291: G6219. 13. De Angelis M, Cassone A, Rizzello CG, et al. Sourdough lactobacilli and fungal proteases detoxied durum wheat gluten: mechanism of epitopes hydrolysis. Appl Environ Microbiol 2010;76:50818.

782

www.jpgn.org

Copyright 2010 by ESPGHAN and NASPGHAN. Unauthorized reproduction of this article is prohibited.

JPGN

Volume 51, Number 6, December 2010

Gluten-free Baked Goods Safe for Celiac Patients

14. Di Cagno R, De Angelis M, Lavermicocca P, et al. Proteolysis by sourdough lactic acid bacteria: effects on wheat our protein fractions and gliadin peptides involved in human cereal intolerance. Appl Environ Microbiol 2002;68:62333. 15. Di Cagno R, De Angelis M, Auricchio S, et al. Sourdough bread made from wheat and nontoxic ours and started with selected lactobacilli is tolerated in celiac sprue patients. Appl Environ Microbiol 2004;70: 108896. 16. Di Cagno R, De Angelis M, Alfonsi G, et al. Pasta made from durum wheat semolina fermented with selected lactobacilli as a tool for a potential decrease of the gluten intolerance. J Agric Food Chem 2005; 53:4393402. 17. De Angelis M, Coda R, Silano M, et al. Fermentation by selected sourdough lactic acid bacteria to decrease the intolerance to rye and barley ours. J Cereal Sci 2006;43:30114. 18. De Angelis M, Rizzello CG, Fasano A, et al. VSL#3 probiotic preparation has the capacity to hydrolyze gliadin polypeptides responsible for Celiac Sprue. Biochim Biophys Acta 2006;1762:8093. 19. Rizzello CG, De Angelis M, Di Cagno R, et al. Highly efcient gluten degradation by lactobacilli and fungal proteases during food processing: new perspectives for celiac disease. Appl Environ Microbiol 2007;73: 4499507. 20. Dewar DH, Amato M, Ellis HJ, et al. The toxicity of high molecular weight glutenin subunits of wheat to patients with coeliac disease. Eur J Gastroenterol Hepatol 2006;18:48391. 21. Weiss W, Volgelmeier C, Gorg A. Electrophoretic characterization of wheat grain allergens from different cultivars involved in bakersasthma. Electrophoresis 1993;14:80516. 22. Bini L, Magi B, Marzocchi B, et al. Protein expression proles in human breast ductal carcinoma and histologically normal tissue. Electrophoresis 1997;18:283241. 23. Bradford MM. A rapid and sensitive method for the quantication of microgram quantities of protein utilizing the principle of protein-dye binding. Anal Biochem 1976;72:24854. 24. European Society of Paediatric Gastroenterology and Nutrition. Revised criteria for diagnosis of coeliac disease. Report of Working Group of European Society of Paediatric Gastroenterology and Nutrition. Arch Dis Child 1990;65:90911. 25. Berstad AE, Brandtzaeg P. Expression of cell membrane complement regulatory glycoproteins along the normal and diseased human gastrointestinal tract. Gut 1998;42:5229. 26. Browning TH, Trier JS. Organ culture of mucosal biopsies of small intestine. J Clin Invest 1969;48:142332.

27. Caffarelli C, Cavagni G, Menzies IS, et al. Elimination diet and intestinal permeability in atopic eczema: a preliminary study. Clin Exp Allergy 1993;23:2831. 28. Lostia AM, Lionetto L, Principessa L, et al. A liquid chromatography/ mass spectrometry method for the evaluation of intestinal permeability. Clin Biochem 2008;41:88792. 29. Marsilio R, DAntiga L, Cancan L, et al. Simultaneous HPLC determination with light- scattering detection of lactulose and mannitol in studies of intestinal permeability in pediatrics. Clin Chem 1998;44: 168591. 30. Dinari G, Rosenbach Y, Marcus H, et al. IgA antigliadin antibodies in childhood celiac disease. Isr Med Sci 1988;24:28690. 31. Valleta AE, Trevisol D, Mastella G. IgA antigliadin antibodies in the monitoring of gluten challenge in celiac disease. J Pediatr Gastroenterol Nutr 1990;10:16973. 32. Laurin P, Wolving M, Falth Magnusson K. Even small amounts of gluten cause relapse in children with celiac disease. J Pediatr Gastroenterol Nutr 2002;34:2630. 33. Greco L, DAdamo G, Truscelli A, et al. Intestinal permeability after single dose gluten challenge in coeliac disease. Arch Dis Child 1991; 66:8702. 34. Hamilton I, Cobden I, Rothwell J, et al. Intestinal permeability in coeliac disease: the response to gluten withdrawal and single-dose gluten challenge. Gut 1982;23:20210. 35. Gass J, Bethune MT, Siegel M, et al. Combination enzyme therapy for gastric digestion of dietary gluten in patients with celiac sprue. Gastroenterology 2007;133:47280. 36. Shan L, Marti T, Sollid LM, et al. Comparative biochemical analysis of three bacterial prolyl endopeptidases: implications for celiac sprue. Biochem J 2004;383:3118. 37. Volta U, Granito A, Fiorini E. Usefulness of antibodies to deamidated gliadin peptides in celiac disease diagnosis and follow-up. Dig Dis Sci 2008;53:15828. 38. Kilmartin C, Lynch S, Abuzakouk M, et al. Avenin fails to induce a Th1 response in coeliac tissue following in vitro culture. Gut 2003;52:4752. 39. Monteleone IG, Del Vecchio Blanco G, Vavassori P, et al. Regulation of the T helper cell type 1 transcription factor T-bet in coeliac disease mucosa. Gut 2004;53:10905. 40. Fotoulaki M, Nousia-Arvanitakis S, Augoustidou-Savvopoulou P, et al. Clinical application of immunological markers as monitoring tests in celiac disease. Dig Dis Sci 1999;44:21338. 41. Schuppan D, Junker Y, Barisani D. Celiac disease: from pathogenesis to novel therapies. Gastroenterology 2009;137:19123.

www.jpgn.org

783

Copyright 2010 by ESPGHAN and NASPGHAN. Unauthorized reproduction of this article is prohibited.

You might also like