WO2014020209A1 - Use of milk protein hydrolysates as gastrointestinal protectors - Google Patents

Use of milk protein hydrolysates as gastrointestinal protectors Download PDF

Info

Publication number
WO2014020209A1
WO2014020209A1 PCT/ES2013/070550 ES2013070550W WO2014020209A1 WO 2014020209 A1 WO2014020209 A1 WO 2014020209A1 ES 2013070550 W ES2013070550 W ES 2013070550W WO 2014020209 A1 WO2014020209 A1 WO 2014020209A1
Authority
WO
WIPO (PCT)
Prior art keywords
seq
hydrolyzate
peptides
milk
casein
Prior art date
Application number
PCT/ES2013/070550
Other languages
Spanish (es)
French (fr)
Inventor
Mª Isidra RECIO SÁNCHEZ
Mª Angeles Beatriz MIRALLES BURAGLIA
Daniel MARTÍNEZ MAQUEDA
Blanca HERNÁNDEZ LEDESMA
Sonia Cristina DE PASCUAL-TERESA FERNÁNDEZ
Lourdes Amigo Garrido
Mª del Rocío GIRÓN MORENO
Carlos GOICOECHEA GARCÍA
Original Assignee
Consejo Superior De Investigaciones Científicas (Csic)
Universidad Rey Juan Carlos
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Consejo Superior De Investigaciones Científicas (Csic), Universidad Rey Juan Carlos filed Critical Consejo Superior De Investigaciones Científicas (Csic)
Publication of WO2014020209A1 publication Critical patent/WO2014020209A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/01Hydrolysed proteins; Derivatives thereof
    • A61K38/012Hydrolysed proteins; Derivatives thereof from animals
    • A61K38/018Hydrolysed proteins; Derivatives thereof from animals from milk
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/1703Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • A61K38/1709Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4717Plasma globulins, lactoglobulin
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4732Casein

Definitions

  • the present invention is within biology and medicine and can be applied to the food and pharmaceutical industry.
  • the invention relates to the use of peptides and / or enzymatic hydrolysates of milk proteins to improve the protection of the intestinal tract by stimulating mucin secretion, and to the compositions containing said peptides.
  • the present invention also collects new peptide sequences with opioid activity.
  • the intestine is the organ responsible for the absorption of nutrients, it acts as a barrier, signal recognition and synthesis of bioactive compounds. These functions are regulated by hormones and cytokines, but it is known that certain components of the diet can also play a fundamental role in modulating these functions.
  • Epithelial cells Intestinals which are forming a monolayer that covers the entire surface of the gastrointestinal tract, are especially important in this modulation because they interact directly with the intestinal content, including food, its digested and bacterial flora. Therefore, the food, in addition to a source of nutrients, can act as a "modulator" of different physiological functions.
  • the organ where this concept can be applied directly is the gastrointestinal tract because it is in contact with a large number of substances that we ingest. Modulation of intestinal functions through diet has recently been described as essential to improve human health (Shimizu and Son, 2007 Curr. Pharm. Design 13: 885-895).
  • a key function of the intestinal epithelium is to act as a physical barrier between the external and internal environment.
  • the intestinal surface is covered by a mucous layer that plays an important physiological role that includes the lubrication of the cell surface, the defense against colonization by pathogenic bacteria and the protection against intestinal proteases.
  • the main structural components of the mucus are the mucins, high molecular weight glycoproteins produced by the goblet cells of the epithelial surface.
  • the epithelium of the intestine both thin and thick, contains goblet cells that produce mucins.
  • Mucin 2 is the predominant gel-forming mucin in the healthy intestine of human, rat and mouse.
  • the present invention provides new peptide sequences with stimulating activity. of the secretion of intestinal mucins in human intestinal cells. This biological activity would not have been previously described for these peptides, so it implies a new use of them.
  • their production is described by enzymatic hydrolysis of low-cost food proteins (whey proteins, caseinates, etc.), resulting in hydrolysates containing the new peptides in relevant concentrations and therefore with protective activity at the intestinal level.
  • the present invention provides hydrolysates of whey and casein proteins that contain a series of peptides that positively affect the secretory activity of mucins, stimulating the formation of the intestinal mucosa and that affect the expression of the MUC5AC gene in intestinal secretory epithelial cells of mucins (HT29-MTX). New peptides derived from caseins capable of inducing mucin secretion and presenting opioid activity in guinea pig ileum are also described.
  • a first aspect of the invention relates to the use of at least one isolated bioactive peptide that: a) possesses mucin secretion stimulating activity and / or opioid activity; b) is present in an enzymatic hydrolyzate of at least one milk protein selected from a whey protein hydrolyzate (preferably a whey protein hydrolyzate with trypsin) or a milk casein hydrolyzate (preferably a casein hydrolyzate of pepsin milk); c) its structure comprises a sequence with at least two aromatic amino acids, where the first aromatic amino acid is a tyrosine located in the first or second position with respect to the N-terminal end of said sequence and the second aromatic amino acid is a phenylalanine or a tyrosine located in third or fourth position with respect to the first tyrosine (which means that between the first tyrosine residue and the second aromatic amino acid there is one or two separation amino acids); and where said structure is selected from SEQ ID No: 1, SEQ ID
  • the formation of said intestinal mucosa is due to a greater mucin secretion that is induced by the above isolated peptides.
  • mucin formation is due to increased expression of the MUC5AC gene in secretory human goblet cells (HT29-MTX), when exposed to at least one of the isolated bioactive peptides. This stimulation of the formation of the intestinal mucosa by the bioactive peptides above, is useful to promote gastrointestinal protection.
  • Table 1 shows the list of the sequences of the bioactive peptides SEQ ID No: 1 to SEQ ID No: 12 contemplated within the scope of the present invention.
  • enzymatic hydrolyzate refers to the combination of a protein substrate that serves as a starting material and proteolytic enzymes in a suitable ratio and under specific conditions of pH and hydrolysis time that gives rise to peptides with the sequence desired.
  • the starting material of the present invention would be any suitable substrate comprising one or more proteins or peptides, of animal, plant, or microorganism origin, containing the amino acid sequence of the bioactive peptides of interest (SEQ ID No 1, SEQ ID No. 2, SEQ ID No. 3, SEQ ID No. 4, SEQ ID No. 5, SEQ ID No. 6, SEQ ID No. 7, SEQ ID No. 8, SEQ ID No. 9, SEQ ID No. 10, SEQ ID No. 11 and SEQ ID No. 12, table 1), preferably milk, casein or whey protein concentrates.
  • suitable substrate comprising one or more proteins or peptides, of animal, plant, or microorganism origin, containing the amino acid sequence of the bioactive peptides of interest (SEQ ID No 1, SEQ ID No. 2, SEQ ID No. 3, SEQ ID No. 4, SEQ ID No. 5, SEQ ID No. 6, SEQ ID No. 7, SEQ ID No. 8, SEQ ID No. 9, SEQ ID No. 10, SEQ ID No. 11 and S
  • any preparation containing the starting proteins could be used: ⁇ -lactoglobulin, ⁇ -casein, or 3 ⁇ 4i-casein of different species, or peptides or fragments thereof of any size, alone or mixed with other proteins.
  • ⁇ -lactoglobulin ⁇ -casein, or 3 ⁇ 4i-casein of different species
  • peptides or fragments thereof of any size, alone or mixed with other proteins.
  • whole casein, caseinates and milk in its different forms of presentation fermented milk products, hydrolysed milk proteins, milk by-products, dairy products for animal feed, etc.
  • Said starting material is dissolved or dispersed, at an appropriate concentration, in water or in a buffer solution, at a pH suitable for the action of the proteolytic enzyme.
  • proteolytic enzyme capable of breaking the protein present in the starting material and providing the peptides of interest may be used, but preferably pepsin at pH 2.0-3.0 or trypsin at pH 7.5-8.5.
  • proteolytic microorganisms that carried out fermentation of the substrate and hydrolysis of the protein could also be used.
  • the hydrolysis conditions pH, temperature, enzyme-substrate ratio, reaction interruption etc., are optimized in order to select the most active hydrolysates.
  • the bioactive peptides are obtained using casein hydrolyzed with pepsin at pH 2.5, in an enzyme / substrate ratio 4/100 (w / w) and performing hydrolysis at 37 ° C, for a period of time between 10 minutes and 24 hours, but preferably for less than 6 hours.
  • the peptides are obtained by hydrolyzing a whey protein concentrate with trypsin at pH 8.0, in a 5/100 enzyme / substrate ratio (w / w) and performing hydrolysis at 37 ° C, over a period of time. of time between 10 minutes and 24 hours, but preferably for less than 4 hours.
  • the pharmaceutical composition refers to the mixture of an active ingredient, or mixture of active ingredients, with suitable excipients (lactose, maltodextrin, etc.) to give oral administration forms, preferably tablets or capsules.
  • suitable excipients lactose, maltodextrin, etc.
  • the active principle is understood as the individual peptides, the mixture thereof or the enzymatic hydrolysates that contain them.
  • the doses of the peptides of interest may be between 1 to 10 mg / day, but preferably they will be administered at doses of 5 to 7 mg / day.
  • functional ingredient refers to a food ingredient that in addition to exercising a Beneficial effect on the individual who consumes it for its nutritional value, satisfactorily demonstrates improving one or more functions in the body.
  • the function in the organism of the functional ingredient is referred both to the improvement of a physiological function, and to the decrease in the risk of suffering from a disease.
  • the functional ingredient may contain the individual peptides, the mixture thereof or the enzymatic hydrolysates that contain them, in addition to different excipients in order to improve their organoleptic or technological characteristics (lactose, fructose, starch, maltodextrins, etc.).
  • This functional ingredient could be incorporated into different foods compatible with a healthy diet such as yogurts, juices, infant formulas, cereal bars, dairy preparations and desserts, powdered food preparations, etc.
  • the pharmaceutical composition or functional ingredient above are useful for stimulating the formation of the intestinal mucosa.
  • the isolated bioactive peptide for the manufacture of the pharmaceutical composition or functional ingredient comprises at least one peptide selected from at least one of the group consisting of: SEQ ID No: 1, SEQ ID No: 2 and a combination from both.
  • Said peptides SEQ ID No: 1 and / or SEQ ID No: 2 are present in whey protein hydrolysates (preferably in whey protein hydrolysates with trypsin) and increase the secretory activity of mucins in producing intestinal epithelial cells. of mucins, and therefore, stimulate the formation of the intestinal mucosa.
  • whey protein enzyme hydrolyzate comprising at least one of the individual peptides SEQ ID No: 1, SEQ ID No: 2, or a combination thereof, is equally valid for the preparation of the pharmaceutical composition or functional ingredient.
  • bioactive peptides identified in Table 2 SEQ ID No: 1 and SEQ ID No: 2
  • SEQ ID No: 2 upon knowing their sequence, can be obtained by chemical and / or enzymatic synthesis, hydrolysis of proteins of different species (cow , sheep, goat, etc.), hydrolysis of recombinant human proteins or by recombinant methods of heterologous production.
  • the isolated bioactive peptide for the manufacture of the pharmaceutical composition or functional ingredient comprises at least one peptide selected from at least one of the group consisting of: SEQ ID No: 3, SEQ ID No: 4, SEQ ID No: 5, SEQ ID No: 6, SEQ ID No: 7, SEQ ID No: 8, SEQ ID No: 9, SEQ ID No: 10, SEQ ID No: 11, SEQ ID No: 12 and any combination of the same.
  • Peptides SEQ ID No: 3, SEQ ID No: 4, SEQ ID No: 5, SEQ ID No: 6, SEQ ID No: 7, SEQ ID No: 8, SEQ ID No: 9, SEQ ID No: 10, SEQ ID No: 11 and / or SEQ ID No: 12 are present in casein hydrolysates (preferably in casein hydrolysates of milk with pepsin) and increase the secretory activity of mucins in intestinal mucin-producing epithelial cells, thus stimulating the formation of the intestinal mucosa.
  • the available technology allows the bioactive peptides identified in Table 3 (SEQ ID No: 3, SEQ ID No: 4, SEQ ID No: 5, SEQ ID No: 6, SEQ ID No: 7, SEQ ID No: 8, SEQ ID No: 9, SEQ ID No: 10, SEQ ID No: 11, SEQ ID No: 12), upon knowing its sequence, can be obtained by chemical and / or enzymatic synthesis, hydrolysis of proteins of different species (cow, sheep , goat, etc.), hydrolysis of recombinant human proteins or by recombinant methods of heterologous production.
  • the isolated bioactive peptide for the manufacture of the pharmaceutical composition or functional ingredient comprises at least one peptide selected from at least one of the group consisting of: SEQ ID No: 3, SEQ ID No: 4, SEQ ID No: 5 and any combination thereof.
  • the peptides SEQ ID No: 3, SEQ ID No: 4 and / or SEQ ID No: 5 are present in a hydrolyzate of the milk / 3-casein fraction (preferably in milk / 3-casein hydrolysates with pepsin) Therefore, for the preparation of the pharmaceutical composition or functional ingredient, as the invention is understood, it is possible to use at least one of the peptides defined by SEQ ID No: 3, SEQ ID No: 4, SEQ ID No: 5 , or any combination of said peptides, or an enzymatic hydrolyzate of the milk / 3-casein fraction comprising at least one of said peptides.
  • the isolated bioactive peptide for the manufacture of the pharmaceutical composition or functional ingredient comprises at least one peptide selected from at least one of the group consisting of: SEQ ID No: 6, SEQ ID No: 7, SEQ ID No: 8, SEQ ID No: 9, SEQ ID No: 10, SEQ ID No: 11, SEQ ID No: 12 and any combination thereof.
  • Peptides SEQ ID No: 6, SEQ ID No: 7, SEQ ID No: 8, SEQ ID No: 9, SEQ ID No: 10, SEQ ID No: 11 and / or SEQ ID No: 12 are present in hydrolysates of the 3 ⁇ 4i-casein fraction of milk (preferably in sl -casein hydrolysates of milk with pepsin), and as mentioned above increase the secretory activity of mucins in intestinal epithelial cells producing mucins.
  • any of the peptides SEQ ID No: 8, SEQ ID No: 9, SEQ ID No: 10, SEQ ID No: 11, SEQ ID No: 12 or a combination thereof in addition to stimulating secretion activity of mucins, present opioid activity in guinea pig ileum preparations (Table 4).
  • the present invention also contemplates the protection of an isolated bioactive peptide that: a) possesses mucin secretion stimulating activity and / or opioid activity; b) is present in an enzymatic hydrolyzate of at least one milk protein selected from a whey protein hydrolyzate (preferably a whey protein hydrolyzate with trypsin) or a milk casein hydrolyzate (preferably a casein hydrolyzate of pepsin milk); c) its structure comprises a sequence with at least two aromatic amino acids, where the first aromatic amino acid is a tyrosine located in the first or second position with respect to the N-terminal end of said sequence and the second aromatic amino acid is a phenylalanine or a tyrosine located in third
  • the use of the first aspect of the invention can also be protected as a method of treatment to stimulate the formation of the intestinal mucosa and / or favor gastrointestinal protection characterized in that it comprises administering to an individual a therapeutically effective amount of a peptide.
  • isolated bioactive that: a) has mucin secretion stimulating activity and / or opioid activity; b) is present in an enzymatic hydrolyzate of at least one milk protein selected from a whey protein hydrolyzate (preferably a whey protein hydrolyzate with trypsin) or a milk casein hydrolyzate (preferably a milk casein hydrolyzate with pepsin); c) its structure comprises a sequence with at least two aromatic amino acids, where the first aromatic amino acid is a tyrosine located in the first or second position with respect to the N-terminal end of said sequence and the second aromatic amino acid is a phenylalanine or a tyrosine located in third or fourth position with respect to the first tyrosine; and where said structure is selected from SEQ ID No: 1, SEQ ID No: 2, SEQ ID No: 3, SEQ ID No: 4, SEQ ID No: 6, SEQ ID No: 7, SEQ ID No: 8, SEQ ID No: 9, SEQ ID No: 10, S
  • the term "therapeutically effective amount” refers to the amount of the peptides of the invention calculated to produce the desired effect. Specifically, it refers to the amount of peptide, or mixture of peptides, or of the hydrolysates that contain the peptides, which is capable of inducing the secretion of mucins at the gastrointestinal level and thus contributing to the protection thereof.
  • the doses of the peptides of interest may be between 1 to 10 mg / day, but preferably they will be administered at doses of 5 to 7 mg / day.
  • the doses of the hydrolysates containing them may be between 0.5-5 g / day, but preferably they will be administered at doses of 1-2 g / day.
  • Figure 1 Effect of peptides SEQ ID No: 5 (A), SEQ ID No: 6 (B), SEQ ID No: 8 (C) at a concentration of 0.1 mM on mucin secretion in HT29- cells MTX determined by enzyme conjugated lectin assay (ELLA). The data are expressed as a percentage of mucin secretion versus control (untreated cells). Each point represents the mean + standard error of the average of three biological replicates in triplicate. Significant differences obtained through two-way ANOVA applying the Bonferroni test. *** P ⁇ 0.001; * P ⁇ 0.05. Figure 2.
  • FIG. 3 Effect of peptides SEQ ID No: 2 (A), SEQ ID No: 6 (B) and SEQ ID No: 8 (C) in concentrations of 0.5, 0.1 and 0.05 mM on the MUC5AC mRNA level determined by quantitative PCR (RT-qPCR) between 2 and 24 hours of exposure.
  • the data are expressed as relative expression level of MUC5AC versus the control (untreated cells), using cyclophilin as the reference gene.
  • Each point represents the mean + the standard error of the average of three biological replicates in triplicate.
  • the ⁇ -casomorphine 7 bovine f (60-66) of the ⁇ -casein A2 of YPFPGPI sequence was purchased from Bachem (Bubendorf, Switzerland) and was used as a positive control.
  • whey protein hydrolyzate In the preparation of whey protein hydrolyzate, a whey protein concentrate (Friesland Foods Domo, Zwolle, The Netherlands) was dissolved in 5% w / v water and heated at 90 ° C for 10 minutes. Hydrolysis was carried out by applying food grade porcine trypsin (Biocatalysts, Nantgarw, Wales, UK) with an enzyme-substrate ratio of 5% w / w, for 3 hours at 37 ° C and pH 8.0. The reaction was terminated by inactivation of the enzyme by heat treatment at 95 ° C for 15 minutes.
  • porcine trypsin Biocatalysts, Nantgarw, Wales, UK
  • Casein hydrolyzate was obtained from commercial casein (Promilk 85, Arras Cedex, France) at 6% w / v in water with a final pH of 2.5. Hydrolysis was carried out with food grade pepsin (Biocatalysts) in an enzyme-substrate ratio of 2% w / w, being added twice (initial moment and three hours of hydrolysis). It was incubated at 40 ° C for 6 hours with final inactivation of the enzyme by heating at 80-85 ° C for 15 minutes. The peptides contained in the hydrolysates were identified by HPLC-MS / MS. a.2) Analysis via RP-HPLC coupled online to tandem mass spectrometry (RP-HPLC-MS / MS)
  • Esquire-LC equipment (Bruker Daltonik GMBH, Bremen, Germany) was used.
  • the HPLC equipment (1100 series) consisted of a quaternary pump, an automatic injector, an eluent degassing system and a variable wavelength ultraviolet detector (Agilent Technologies, Waldbronn, Germany) coupled in line to a mass spectrometer of ionic trap Esquire 3000 (Bruker Daltonik).
  • the column was a Hi-Pore C18 column (250 x 4.6 mm ID, 5 particle size) (Bio-Rad Laboratories, Richmond, CA, USA).
  • Solvent A was a mixture of water and trifluoroacetic acid (1000: 0.37) and solvent B a mixture of acetonitrile and trifluoroacetic acid (1000: 0.27). 50 ⁇ of prepared sample was injected at a concentration of 4.5 mg / ml. A flow of 0.8 ml / min was used, with a linear gradient from 0% to 50% of solvent B in 60 minutes. The eluent was monitored at 214 nm and the sample flow to the mass spectrometer nebulizer was 275 ⁇ / min.
  • Electrospray ionization used N2 as a nebulizer agent (8 L / min), fogging pressure 60 psi, drying temperature 350 ° C and a capillary voltage of 4 KV.
  • the mass spectra were acquired in the range of 100-2500 m / z and a fragmentation ramp between 0.3 and 2 V was used.
  • the Esquire ControlTM 5.0 program (Bruker Daltonik) was used as the data acquisition system and for To identify the sequence of the different peptides, the Data AnalysisTM 3.0, Sequence EditorTM 2.1 and BiotoolsTM 2.1 software were all used by Bruker Daltonik. a.3) Cell cultures
  • the HT29-MTX cell line derived from human colon adenocarcinoma was used, with mucin secretion capacity donated by Dr. Thécla Lesuffleur (INSERM UMR S 938, Paris, France).
  • Cells were grown in Dulbecco-modified Eagle medium (DMEM) supplemented with 10% inactivated bovine fetal serum and 10 mL / L of penicillin-streptomycin at 37 ° C, in a humidified atmosphere with 5% C0 2 . Weekly passes were performed using 0.05% trypsin / EDTA (all reagents were from Gibco, Paisley, UK).
  • the cells were seeded in 12-well plates with a density of 5> ⁇ 10 5 cells per well (Nunc, Roskilde, Denmark). The cell line was used between passes 12 and 22. The experiments were performed 21 days after confluence. From 24 hours before the test, we worked with culture medium without serum or antibiotic to eliminate the interference of proteins or hormones. On the day of the test, the medium was removed, the cell monolayer was washed twice with phosphate buffered saline (PBS) and the medium was added with the peptides in concentration 0.05, 0.1 and 0.5 mM, incubating at 37 ° C for times between 30 minutes and 24 hours. In the control wells only the culture medium was added.
  • PBS phosphate buffered saline
  • the quantitative determination of total mucins was performed by an ELLA assay based on the use of wheat germ agglutinin, which has shown a strong reactivity against specific sugars present in the mucins secreted by goblet cells.
  • the total glycoprotein content in the samples studied was obtained by interpolation in a standard curve constructed from commercial porcine gastric mucin (Sigma, St Louis, CA, USA).
  • the upholstery of the 96-well polystyrene plate was performed with the samples from the calibration line and with the samples to be tested dissolved in sodium carbonate buffer (0.5 M, pH 9.6), leaving to incubate overnight at 4 ° C
  • the blockade was performed with bovine serum albumin (BSA) (Sigma) at 2% for 1 hour at 37 ° C.
  • BSA bovine serum albumin
  • Biotinylated wheat germ agglutinin Vector Laboratories, Peterborough, UK
  • PBS-Tween-BSA (1: 1000
  • RNA copy number was determined by quantitative PCR with real-time fluorescence measurement, using SYBR Green as a fluorophore in Lightcycler 480 (Roche, Mannhein, Germany) with 384 well plates.
  • the cDNA was obtained from 375 ng of RNA by using High Capacity cDNA Reverse Transcription Kit (Applied Biosystems) according to the manufacturer's instructions.
  • MUC5AC access code NCBI AJ001402
  • target gene oligos 2870-2899 / 3109-3091 were used
  • cyclophilin access code NCBI Y00052
  • oligos 280-340 / 445- were used 421 (Zoghbi et al., 2006 Am. J.
  • Each cDNA sample was studied in triplicate.
  • Each reaction tube contained 5 ⁇ of 2 ⁇ SYBR Green real-time PCR Master Mix (Applied Biosystems), 0.25 ⁇ to 10 ⁇ of each of the specific oligos, 0.27 ⁇ of cDNA and 4.23 of H 2 0.
  • the amplification was started at 95 ° C for 5 minutes and 45 cycles at 95 ° C for 10 seconds, 60 ° C for 10 seconds and 72 ° C for 10 seconds. Controls were used to confirm the absence of oligo dimers and to verify that there was no DNA contamination.
  • FL-PM guinea pig ileum
  • the in vitro organ bath technique was used, in which the equipment used is made up of 10-ml glass cups that were filled with 5 ml of Krebs solution (NaCl 118, KC1 4.75; CaCl 2 2.54; KH 2 P0 4 1.19; MgS0 4 1.2; NaHC0 3 25; 11 mM glucose), which is continuously bubbled with carbon dioxide (95% 0 2 and 5% C0 2 ) and maintained at a temperature of 37 ° C.
  • the preparation is then given a baseline tension of 1 g, allowed to rest for 15 minutes for stabilization and electrically stimulated continuously.
  • the stimulation conditions were constant: simple square pulses of 0.3 Hz frequency, 2 ms duration and supramaximal voltage.
  • the contractile activity of Preparations were recorded using a data recording and analysis system coupled to an isometric force transducer.
  • concentration-response curves were performed, by adding to the bath of organs of cumulative increasing doses of the peptides, spaced in 10 min (considered as the interval of time required for the peptides to exert their effect).
  • the concentrations evaluated were: 6, 1 x 1CT 8 , 1.8 x 1CT 7 , 5.5 x 1CT 7 , 1.6 x 1CT 6 and 5 x 1CT 6 M.
  • a single dose of the opioid antagonist naloxone (10 ⁇ 6 M) was added to check if their effect reversal took place and to confirm that the inhibitory effect was mediated by selective interaction with opioid receptors.
  • results are expressed as% contraction inhibition, considering as 100% the average amplitude of the last 5 contractions before the addition of the first dose of the curve.
  • Each preparation was used to carry out a single concentration-response curve.
  • Example 1 Effect of different synthetic food peptides on the secretory activity of mucins in HT29-MTX cells.
  • Table 5 shows the maximum mucin secretion values in HT29-MTX cells after the addition of the peptides of SEQ ID No: 1, SEQ ID No: 2, SEQ ID No: 3, SEQ ID No: 4, SEQ ID No: 5, SEQ ID No: 6, SEQ ID No: 7 and SEQ ID No: 8 at the concentration of 0.1 mM.
  • Table 5 shows these values for six other peptides previously described as opioids. Eleven of the 14 food peptides tested resulted in a significant increase (P ⁇ 0.001) of mucin secretory activity in HT29-MTX cells. The maximum secretion values ranged from 191 to 587% with respect to the control (untreated cells). It should be noted that SEQ ID No: 2 and SEQ ID No: 5 produced greater increases in mucin secretion than ⁇ -casomorphine 7.
  • Neocasomorphine / 3 B -casein f (114-
  • Figure 1 shows the effect during the exposure time of the peptides of SEQ ID No: 5, SEQ ID No: 6 and SEQ ID No: 8. at a 0.1 mM concentration on the secretion of mucins in HT29-MTX cells .
  • SEQ ID No: 5 showed increased mucin secretion values at 30 minutes and 2 hours followed by a significant increase at 4 hours and 8 hours (Fig 1A).
  • SEQ ID No: 6 and SEQ ID No: 8 showed absolute levels lower than those presented by the previously described but significant peptides at 2 and 4 hours, respectively (Fig IB and 1C).
  • Example 2 Effect of different milk protein hydrolysates derived from whey proteins and caseins on the expression of the MUC5AC gene of HT29-MTX cells.
  • Example 3 Effect of different milk peptides derived from whey proteins and caseins on the expression of the MUC5AC gene of HT29-MTX cells.
  • Concentrations between 0.05 and 0.5 mM of the peptides of SEQ ID No: 2, SEQ ID No: 6 and SEQ ID No: 8 were added to the cell incubation medium for times between 2 and 24 hours.
  • Figure 3 shows the results of quantitative PCR at concentrations of 0.05 mM, 0.1 mM and 0.5 mM. Treatment of cells with SEQ ID No: 2 caused a significant increase
  • Example 4 Opioid activity of food peptides by guinea pig ileum assay.
  • Figure 4 shows the opioid agonist activity of peptides SEQ ID No: 9 and SEQ ID No: 10 determined in vitro by assay in the FL-PM preparation of guinea pig ileum.
  • the peptides induced an inhibition of electrically induced contractions that ranged between 20 and 30% in the case of SEQ ID No: 9 (Fig 4A) and between 25 and 35% in the case of SEQ ID No: 10 (Fig. 4B), in the concentration 5.5 10 ⁇ 7 M.
  • the effect of all the peptides was reversed with the administration of naloxone (10 ⁇ 6 M).
  • naloxone (10 ⁇ 6 M).
  • morphine resulted in a maximum inhibition of 75% while / 3-casomorphine 7 produced a maximum inhibition of 20%.

Abstract

The present invention relates to the use of an isolated bioactive peptide which has stimulating activity for the secretion of mucins and/or opioid activity, which is included in an enzymatic hydrolysate of at least one milk protein selected from a whey protein hydrolysate or a milk casein hydrolysate, in which the structure thereof includes a sequence having at least two aromatic amino acids, wherein the first aromatic amino acid is a tyrosine in the first or second position relative to the N-terminal end of said sequence and the second aromatic amino acid is a phenylalanine or a tyrosine in the third or fourth position relative to the first tyrosine; in order to stimulate the formation of intestinal mucus and/or to promote gastrointestinal protection.

Description

USO DE HIDROLIZADOS DE PROTEINAS LACTEAS COMO PROTECTORES A NIVEL  USE OF HYDROLYZES OF MILK PROTEINS AS LEVEL PROTECTORS
GASTROINTESTINAL  GASTROINTESTINAL
DESCRIPCION DESCRIPTION
Sector de la técnica Technical sector
La presente invención se encuentra dentro de la biología y la medicina pudiendo aplicarse a la industria alimentaria y farmacéutica. La invención se refiere al uso de péptidos y/o hidrolizados enzimáticos de proteínas lácteas para mejorar la protección del tracto intestinal mediante la estimulación de la secreción de mucinas, y a las composiciones que contienen dichos péptidos. Además, la presente invención también recoge nuevas secuencias peptídicas con actividad opioide . The present invention is within biology and medicine and can be applied to the food and pharmaceutical industry. The invention relates to the use of peptides and / or enzymatic hydrolysates of milk proteins to improve the protection of the intestinal tract by stimulating mucin secretion, and to the compositions containing said peptides. In addition, the present invention also collects new peptide sequences with opioid activity.
Estado de la técnica State of the art
Durante los últimos años, muchas de las investigaciones sobre la fracción proteica de los alimentos se han enfocado al estudio de los péptidos biológicamente activos presentes en la secuencia de las mismas . Estos péptidos se encuentran en estado inactivo cuando forman parte de la proteína precursora pero pueden liberarse mediante hidrólisis enzimática, bien durante la digestión gastrointestinal o durante el procesado de los alimentos. Una vez liberados, los péptidos pueden actuar como compuestos biológicamente activos en el organismo. Así, por ejemplo, se ha demostrado que pueden ejercer actividad opiode, antihipertensiva o antimicrobiana; y cada vez existe más evidencia de que pueden tener actividad en el sistema inmune o mejorar la biodisponibilidad de minerales . During the last years, many of the investigations on the protein fraction of foods have focused on the study of biologically active peptides present in the sequence thereof. These peptides are in an inactive state when they are part of the precursor protein but can be released by enzymatic hydrolysis, either during gastrointestinal digestion or during food processing. Once released, the peptides can act as biologically active compounds in the body. Thus, for example, it has been shown that they can exert opiode, antihypertensive or antimicrobial activity; and there is increasing evidence that they may have activity in the immune system or improve the bioavailability of minerals.
El intestino es el órgano responsable de la absorción de nutrientes, ejerce funciones de barrera, de reconocimiento de señales y de síntesis de compuestos bioactivos. Estas funciones están reguladas por hormonas y citoquinas, pero se sabe que ciertos componentes de la dieta también pueden jugar un papel fundamental en la modulación de dichas funciones. Las células epiteliales intestinales, que se encuentran formando una monocapa que cubre toda la superficie del tracto gastrointestinal, son especialmente importantes en esta modulación porque interaccionan directamente con el contenido intestinal, incluyendo alimentos, sus digeridos y la flora bacteriana. Por lo tanto, el alimento, además de una fuente de nutrientes, puede actuar como un "modulador" de distintas funciones fisiológicas. El órgano donde este concepto puede aplicarse directamente es el tracto gastrointestinal porque está en contacto con una gran cantidad de sustancias que ingerimos. La modulación de las funciones intestinales mediante la dieta ha sido recientemente calificada como esencial para mejorar la salud humana (Shimizu and Son, 2007 Curr. Pharm. Design 13: 885-895) . The intestine is the organ responsible for the absorption of nutrients, it acts as a barrier, signal recognition and synthesis of bioactive compounds. These functions are regulated by hormones and cytokines, but it is known that certain components of the diet can also play a fundamental role in modulating these functions. Epithelial cells Intestinals, which are forming a monolayer that covers the entire surface of the gastrointestinal tract, are especially important in this modulation because they interact directly with the intestinal content, including food, its digested and bacterial flora. Therefore, the food, in addition to a source of nutrients, can act as a "modulator" of different physiological functions. The organ where this concept can be applied directly is the gastrointestinal tract because it is in contact with a large number of substances that we ingest. Modulation of intestinal functions through diet has recently been described as essential to improve human health (Shimizu and Son, 2007 Curr. Pharm. Design 13: 885-895).
Una función clave del epitelio intestinal es actuar como barrera física entre el medio externo y el interno. La superficie intestinal está cubierta por una capa mucosa que juega un importante papel fisiológico que incluye la lubricación de la superficie celular, la defensa frente a la colonización por bacterias patógenas y la protección frente a las proteasas intestinales. Los principales componentes estructurales del mucus son las mucinas, glicoproteínas de alto peso molecular producidas por las células caliciformes de la superficie epitelial. El epitelio del intestino, tanto delgado como grueso, contiene células caliciformes productoras de mucinas. La mucina 2 es la mucina formadora de gel predominante en el intestino sano de humano, rata y ratón. Cualquier situación experimental o patológica que cambie directa o indirectamente la cantidad de mucina, su estructura o el contenido acuoso o iónico va a provocar una alteración de la barrera mucosa. La úlcera gástrica o la colitis ulcerosa son ejemplos de las consecuencias de un fallo en la expresión y/o composición del mucus gástrico o intestinal, respectivamente. A key function of the intestinal epithelium is to act as a physical barrier between the external and internal environment. The intestinal surface is covered by a mucous layer that plays an important physiological role that includes the lubrication of the cell surface, the defense against colonization by pathogenic bacteria and the protection against intestinal proteases. The main structural components of the mucus are the mucins, high molecular weight glycoproteins produced by the goblet cells of the epithelial surface. The epithelium of the intestine, both thin and thick, contains goblet cells that produce mucins. Mucin 2 is the predominant gel-forming mucin in the healthy intestine of human, rat and mouse. Any experimental or pathological situation that changes directly or indirectly the amount of mucin, its structure or the aqueous or ionic content will cause an alteration of the mucosal barrier. Gastric ulcer or ulcerative colitis are examples of the consequences of a failure in the expression and / or composition of the gastric or intestinal mucus, respectively.
Estudios recientes han demostrado que algunos componentes de la dieta pueden tener una influencia positiva sobre las características del mucus intestinal, produciendo un aumento en la secreción o en la expresión de mucinas o incluso aumentando el número de células caliciformes intestinales, aunque el modo de acción de cada compuesto puede diferir (Barceló et al., 2000 Gut 46:218-224). Por ejemplo, la fibra dietética parece favorecer la capacidad total de secreción de mucinas en el lumen del intestino delgado, aunque el efecto estimulador en la secreción es dependiente tanto de la cantidad como de la calidad de la fibra dietética ingerida (Tanabe et al, 2005 J. Nutr. 135: 2431-2437) y puede deberse a un incremento en el número de células caliciformesRecent studies have shown that some components of the diet can have a positive influence on the characteristics of the intestinal mucus, producing an increase in the secretion or expression of mucins or even increasing the number of intestinal goblet cells, although the mode of The action of each compound may differ (Barceló et al., 2000 Gut 46: 218-224). For example, dietary fiber seems to favor the total mucin secretion capacity in the lumen of the small intestine, although the stimulating effect on secretion is dependent on both the quantity and quality of the ingested dietary fiber (Tanabe et al, 2005 J. Nutr. 135: 2431-2437) and may be due to an increase in the number of goblet cells
(Ito et al., 2009 J. Nutr. 139: 1640-1647). Respecto a los genes codificadores de las mucinas (MUC) , se ha demostrado que concentraciones bajas de butirato estimulan la expresión de MUC2(Ito et al., 2009 J. Nutr. 139: 1640-1647). Regarding the genes encoding the mucins (MUC), it has been shown that low concentrations of butyrate stimulate the expression of MUC2
(Gaudier et al., 2004 Am. J. Physiol. Gastrointest . Líver Physiol. 278: G1168-G1174) mientras que elevadas concentraciones de este ácido graso de cadena corta reprimen la expresión, lo que puede hacer disminuir la función intestinal de barrera (Burger Van Paasen et al., 2009 Biochem. J. 420: 211-219). (Gaudier et al., 2004 Am. J. Physiol. Gastrointest. Líver Physiol. 278: G1168-G1174) while high concentrations of this short-chain fatty acid suppress expression, which can decrease the intestinal function of the barrier ( Burger Van Paasen et al., 2009 Biochem. J. 420: 211-219).
En el campo de las proteínas alimentarias se está investigando el papel de las proteínas y péptidos en la regulación de distintas funciones a nivel intestinal. En particular, Trompette et al., 2003 ( J. Nutr. 133: 3499-3503) han comprobado que algunos péptidos lácteos, concretamente el péptido YPFPGPI, /3-casomorfina 7, que corresponde al fragmento (60-66) de la /3-caseína A2 bovina, induce la producción de mucina en yeyuno de rata, mediante la activación del sistema nervioso entérico y la interacción con receptores opioides. También se comprobó que un hidrolizado de a-lactoalbúmina inducía la liberación de mucinas mientras que ni la caseína ni una mezcla de aminoácidos producían este efecto. Puesto que esta actividad secretora desaparecía en presencia de naloxona, un antagonista opioide, se planteó que la secreción se producía por un mecanismo nervioso implicando la activación de receptores opioides tipo μ (Claustre et al., 2002 Am. J. Physiol. Gastrointest. Liver Physiol. 283: G521-G528; Trompette et al., 2003 J. Nutr. 133: 3499- 3503). La presencia de estos receptores en células intestinales sugiere la posibilidad de que las /3-casomorfinas , que se producen en el lumen intestinal durante la digestión, pueden controlar la producción de mucinas mediante la acción directa sobre las células caliciformes intestinales. Posteriormente, se ha demostrado que el péptido YPFPGPI, /3-casomorfina 7, induce la secreción de la mucina 5AC en células caliciformes humanas de colon (HT29-MTX) (Zoghbi et al., 2006 Am. J. Physiol. Gastrointest. Liver Physiol. 290: G1105- G1113; Plaisancié et al. 2010 Int Patent Appl. WO2010/130956) . Sin embargo, no todos los péptidos opioides tienen actividad sobre la producción de mucinas intestinales . La administración luminal en yeyuno de rata de los péptidos /3-casomorfina-6 bovina (YPFPGP) , β- casomorfina 4 (YPFP) y /3-casomorfina 4-NH2 (YPFF-NH2), aunque poseen afinidad por receptores opioides (Teschemacher et al., 1997 Biopolymers 43:99-117; Meisel and FitzGerald 2000 Br. J. Nutr. 84:S27-S31), no inducen la secreción de mucinas (Trompette et al., 2003 J. Nutr. 133: 3499-3503). In the field of food proteins, the role of proteins and peptides in the regulation of different functions at the intestinal level is being investigated. In particular, Trompette et al., 2003 (J. Nutr. 133: 3499-3503) have found that some dairy peptides, specifically the YPFPGPI peptide, / 3-casomorphine 7, which corresponds to fragment (60-66) of the / Bovine 3-casein A2, induces mucin production in rat jejunum, by activating the enteric nervous system and interacting with opioid receptors. It was also found that an a-lactoalbumin hydrolyzate induced mucin release while neither casein nor a mixture of amino acids produced this effect. Since this secretory activity disappeared in the presence of naloxone, an opioid antagonist, it was suggested that secretion was produced by a nervous mechanism involving the activation of μ type opioid receptors (Claustre et al., 2002 Am. J. Physiol. Gastrointestinal. Liver Physiol. 283: G521-G528; Trompette et al., 2003 J. Nutr. 133: 3499-3503). The presence of these receptors in intestinal cells suggests the possibility that / 3-casomorphins, which occur in the intestinal lumen during digestion, can control the mucin production through direct action on intestinal goblet cells. Subsequently, it has been shown that the YPFPGPI peptide, / 3-casomorphine 7, induces the secretion of mucin 5AC in human colon goblet cells (HT29-MTX) (Zoghbi et al., 2006 Am. J. Physiol. Gastrointest. Liver Physiol. 290: G1105-G1113; Plaisancié et al. 2010 Int Patent Appl. WO2010 / 130956). However, not all opioid peptides have activity on the production of intestinal mucins. The luminal administration in rat jejunum of bovine peptides / 3-casomorphine-6 (YPFPGP), β-casomorphine 4 (YPFP) and / 3-casomorphine 4-NH 2 (YPFF-NH 2 ), although they have an affinity for opioid receptors (Teschemacher et al., 1997 Biopolymers 43: 99-117; Meisel and FitzGerald 2000 Br. J. Nutr. 84: S27-S31), do not induce mucin secretion (Trompette et al., 2003 J. Nutr. 133: 3499-3503).
Por otro lado, con el fin de establecer una asociación entre la producción de mucinas y su biosintesis, se han realizado estudios de expresión de los genes que codifican para mucinas. En células intestinales de rata (DHE) se ha demostrado que la exposición al péptido YPFPGPI, /3-casomorfina 7, provoca un aumento en la expresión de los principales genes implicados en la producción de mucinas de rata (rMuc) , rMuc2 y rMuc3. En células humanas HT29-MTX, este péptido también da lugar a un aumento en la expresión del gen codificador de la principal mucina secretada, MUC5AC (Zoghbi et al., 2006 Am. J. Physiol. Gastrointest. Liver Physiol. 290: G1105- G1113) . En un estudio in vivo, se ha observado un aumento en la expresión de los genes rMuc3 y rMuc4 en ratas a las que se les administró una dieta rica en caseínas hidrolizadas comparándola con una dieta rica en aminoácidos y una dieta exenta de proteínas (Han et al., 2008 J. Agrie. Food Chem. 56: 5572-5576). On the other hand, in order to establish an association between mucin production and its biosynthesis, expression studies of the genes coding for mucins have been carried out. In rat intestinal cells (DHE) it has been shown that exposure to the YPFPGPI peptide, / 3-casomorphine 7, causes an increase in the expression of the main genes involved in the production of rat mucins (rMuc), rMuc2 and rMuc3. In human HT29-MTX cells, this peptide also results in an increase in the expression of the gene encoding the main secreted mucin, MUC5AC (Zoghbi et al., 2006 Am. J. Physiol. Gastrointestinal. Liver Physiol. 290: G1105- G1113). In an in vivo study, an increase in the expression of the rMuc3 and rMuc4 genes has been observed in rats given a diet rich in hydrolyzed caseins compared to a diet rich in amino acids and a protein-free diet (Han et al., 2008 J. Agrie. Food Chem. 56: 5572-5576).
Sin embargo, aunque se conoce que los hidrolizados que contienen β- casomorfina 7 tienen actividad secretora de mucinas, no se han identificado otras secuencias peptídicas que estén presentes en hidrolizados de caseínas lácteas y que sean también responsables de inducir secreción de mucinas. Por tanto, la presente invención proporciona nuevas secuencias peptídicas con actividad estimulante de la secreción de mucinas intestinales en células intestinales humanas . Esta actividad biológica no habría sido previamente descrita para estos péptidos, por lo que supone un nuevo uso de los mismos. Además, se describe la producción de las mismas mediante hidrólisis enzimática de proteínas alimentarias de bajo costo (proteínas suero, caseinatos, etc.), dando lugar a hidrolizados que contienen los nuevos péptidos en concentraciones relevantes y por tanto con actividad protectora a nivel intestinal. However, although it is known that hydrolysates containing β-casomorphine 7 have mucin secretory activity, other peptide sequences that are present in milk casein hydrolysates and that are also responsible for inducing mucin secretion have not been identified. Therefore, the present invention provides new peptide sequences with stimulating activity. of the secretion of intestinal mucins in human intestinal cells. This biological activity would not have been previously described for these peptides, so it implies a new use of them. In addition, their production is described by enzymatic hydrolysis of low-cost food proteins (whey proteins, caseinates, etc.), resulting in hydrolysates containing the new peptides in relevant concentrations and therefore with protective activity at the intestinal level.
Descripción de la Invención Description of the Invention
La presente invención proporciona hidrolizados de proteínas de suero y caseína que contienen una serie de péptidos que afectan positivamente a la actividad secretora de mucinas, estimulando la formación de la mucosa intestinal y que afectan a la expresión del gen MUC5AC en células epiteliales intestinales secretoras de mucinas (HT29-MTX). También se describen nuevos péptidos derivados de las caseínas capaces de inducir la secreción de mucinas y que presentan actividad opioide en íleon de cobayo. The present invention provides hydrolysates of whey and casein proteins that contain a series of peptides that positively affect the secretory activity of mucins, stimulating the formation of the intestinal mucosa and that affect the expression of the MUC5AC gene in intestinal secretory epithelial cells of mucins (HT29-MTX). New peptides derived from caseins capable of inducing mucin secretion and presenting opioid activity in guinea pig ileum are also described.
Un primer aspecto de la invención se refiere al uso de al menos un péptido bioactivo aislado que: a) posee actividad estimulante de secreción de mucinas y/o actividad opioide; b) está presente en un hidrolizado enzimático de al menos una proteína de la leche seleccionado entre un hidrolizado de proteínas de suero (preferentemente un hidrolizado de proteínas de suero con tripsina) o un hidrolizado de caseína de la leche (preferentemente un hidrolizado de caseína de la leche con pepsina) ; c) su estructura comprende una secuencia con al menos dos aminoácidos aromáticos, donde el primer aminoácido aromático es una tirosina situada en primera o segunda posición respecto al extremo N-terminal de dicha secuencia y el segundo aminoácido aromático es una fenilalanina o una tirosina situada en tercera o cuarta posición con respecto a la primera tirosina (lo que quiere decir que entre el primer residuo de tirosina y el segundo aminoácido aromático hay uno o dos aminoácidos de separación) ; y donde dicha estructura se selecciona entre SEQ ID No: 1, SEQ ID No: 2, SEQ ID No: 3, SEQ ID No: 4, SEQ ID No: 5, SEQ ID No: 6, SEQ ID No: 7, SEQ ID No: 8, SEQ ID No: 9, SEQ ID No: 10, SEQ ID No: 11 y SEQ ID No: 12; para la fabricación de una composición farmacéutica o un ingrediente funcional para estimular la formación de la mucosa intestinal. La formación de dicha mucosa intestinal se debe a una mayor secreción de mucinas que se induce por los péptidos aislados anteriores. De hecho, se ha comprobado que la formación de mucinas se debe al incremento de la expresión del gen MUC5AC en células caliciformes humanas secretoras (HT29-MTX), cuando se exponen a al menos uno de los péptidos bioactivos aislados . Esta estimulación de la formación de la mucosa intestinal por parte de los péptidos bioactivos anteriores, resulta de utilidad para favorecer la protección gastrointestinal. A first aspect of the invention relates to the use of at least one isolated bioactive peptide that: a) possesses mucin secretion stimulating activity and / or opioid activity; b) is present in an enzymatic hydrolyzate of at least one milk protein selected from a whey protein hydrolyzate (preferably a whey protein hydrolyzate with trypsin) or a milk casein hydrolyzate (preferably a casein hydrolyzate of pepsin milk); c) its structure comprises a sequence with at least two aromatic amino acids, where the first aromatic amino acid is a tyrosine located in the first or second position with respect to the N-terminal end of said sequence and the second aromatic amino acid is a phenylalanine or a tyrosine located in third or fourth position with respect to the first tyrosine (which means that between the first tyrosine residue and the second aromatic amino acid there is one or two separation amino acids); and where said structure is selected from SEQ ID No: 1, SEQ ID No: 2, SEQ ID No: 3, SEQ ID No: 4, SEQ ID No: 6, SEQ ID No: 7, SEQ ID No: 8, SEQ ID No: 9, SEQ ID No: 10, SEQ ID No: 11 and SEQ ID No: 12; for the manufacture of a pharmaceutical composition or a functional ingredient to stimulate the formation of the intestinal mucosa. The formation of said intestinal mucosa is due to a greater mucin secretion that is induced by the above isolated peptides. In fact, it has been proven that mucin formation is due to increased expression of the MUC5AC gene in secretory human goblet cells (HT29-MTX), when exposed to at least one of the isolated bioactive peptides. This stimulation of the formation of the intestinal mucosa by the bioactive peptides above, is useful to promote gastrointestinal protection.
En la Tabla 1 se muestra la lista de las secuencias de los péptidos bioactivos SEQ ID No: 1 a SEQ ID No: 12 contemplados en el ámbito de la presente invención. Table 1 shows the list of the sequences of the bioactive peptides SEQ ID No: 1 to SEQ ID No: 12 contemplated within the scope of the present invention.
Tabla 1 . Table 1 .
YLLF SEQ ID No: 1 Secuencia de aminoácidos del fragmento 102-105 de la /3-lactoglobulina bovina YLLF SEQ ID No: 1 Amino acid sequence of fragment 102-105 of bovine / 3-lactoglobulin
YLLF-NH2 SEQ ID No: 2 Secuencia de aminoácidos del fragmento 102-105 amidado de la /3-lactoglobulina bovina YLLF-NH 2 SEQ ID No: 2 Amino acid sequence of the aminated 102-105 fragment of the bovine / 3-lactoglobulin
YPSF-NH2 SEQ ID No: 3 Secuencia de aminoácidos del fragmento 41-44 amidado de la /3-caseina humana YPSF-NH 2 SEQ ID No: 3 Amino acid sequence of the amidated fragment 41-44 of the human / 3-casein
YPFV-NH2 SEQ ID No: 4 Secuencia de aminoácidos del fragmento 51-54 amidado de la /3-caseina humana YPFV-NH 2 SEQ ID No: 4 Amino acid sequence of the amidated fragment 51-54 of the human / 3-casein
YPFVE SEQ ID No: 5 Secuencia de aminoácidos del fragmento 51-55 de la /3-caseina humana  YPFVE SEQ ID No: 5 Amino acid sequence of fragment 51-55 of the human / 3-casein
RYLGY SEQ ID No: 6 Secuencia de aminoácidos del fragmento 90-94 de la Qsl-caseína bovina YLGY SEQ ID No: 7 Secuencia de aminoácidos del fragmento 91-94 de la Qsl-caseína bovina RYLGY SEQ ID No: 6 Amino acid sequence of fragment 90-94 of the Q sl- bovine casein YLGY SEQ ID No: 7 Amino acid sequence of fragment 91-94 of the Q sl- bovine casein
AYFYPEL SEQ ID No: 8 Secuencia de aminoácidos del fragmento 143-149 de la Qsl-caseína bovina AYFYPEL SEQ ID No: 8 Amino acid sequence of fragment 143-149 of Q sl- bovine casein
YFYPEL SEQ ID No: 9 Secuencia de aminoácidos del fragmento 144-149 de la Qsl-caseína bovina YFYPEL SEQ ID No: 9 Amino acid sequence of fragment 144-149 of the Q sl- bovine casein
YFYPE SEQ ID No: 10 Secuencia de aminoácidos del fragmento 144-148 de la Qsi-caseína bovina YFYPE SEQ ID No: 10 Amino acid sequence of fragment 144-148 of the bovine Q s i-casein
YFYP SEQ ID No: 11 Secuencia de aminoácidos del fragmento 144-147 de la Qsl-caseína bovina YFYP SEQ ID No: 11 Amino acid sequence of fragment 144-147 of Q sl- bovine casein
YFY SEQ ID No: 12 Secuencia de aminoácidos del fragmento 144-146 de la Qsl-caseína bovina YFY SEQ ID No: 12 Amino acid sequence of fragment 144-146 of the Q sl- bovine casein
En el ámbito de la presente invención, hidrolizado enzimático se refiere a la combinación de un sustrato proteico que sirve de material de partida y enzimas proteoliticas en una relación adecuada y en condiciones especificas de pH y tiempo de hidrólisis que da lugar a péptidos con la secuencia deseada. Within the scope of the present invention, enzymatic hydrolyzate refers to the combination of a protein substrate that serves as a starting material and proteolytic enzymes in a suitable ratio and under specific conditions of pH and hydrolysis time that gives rise to peptides with the sequence desired.
El material de partida de la presente invención seria cualquier sustrato apropiado que comprendiese una o más proteínas o péptidos, de origen animal, vegetal, o procedentes de microorganismos, que contengan la secuencia de aminoácidos de los péptidos bioactivos de interés (SEQ ID No 1, SEQ ID No 2, SEQ ID No 3, SEQ ID No 4, SEQ ID No 5, SEQ ID No 6, SEQ ID No 7, SEQ ID No 8, SEQ ID No 9, SEQ ID No 10, SEQ ID No 11 y SEQ. ID. N° 12, tabla 1), preferiblemente leche, caseína o concentrados de proteínas de suero. Dado que todos ellos son secuencias lácteas, resulta obvio que podría usarse cualquier preparado que contenga las proteínas de partida: β-lactoglobulina, β-caseína, o ¾i-caseína de diferentes especies, o péptidos o fragmentos de las mismas de cualquier tamaño, solos o mezclados con otras proteínas. Por ejemplo: caseína entera, caseinatos y leche en sus diferentes formas de presentación, productos lácteos fermentados, hidrolizados de proteínas lácteas, subproductos lácteos, derivados lácteos para alimentación animal, etc. Dicho material de partida se disuelve o dispersa, a una concentración apropiada, en agua o en una disolución tampón, a un pH adecuado para la actuación de la enzima proteolitica . Puede emplearse cualquier enzima proteolitica capaz de romper la proteina presente en el material de partida y proporcionar los péptidos de interés, pero preferiblemente pepsina a pH 2,0-3,0 o tripsina a pH 7,5-8,5. También podrían emplearse microorganismos proteolíticos que llevasen a cabo una fermentación del sustrato y la hidrólisis de la proteína. The starting material of the present invention would be any suitable substrate comprising one or more proteins or peptides, of animal, plant, or microorganism origin, containing the amino acid sequence of the bioactive peptides of interest (SEQ ID No 1, SEQ ID No. 2, SEQ ID No. 3, SEQ ID No. 4, SEQ ID No. 5, SEQ ID No. 6, SEQ ID No. 7, SEQ ID No. 8, SEQ ID No. 9, SEQ ID No. 10, SEQ ID No. 11 and SEQ ID No. 12, table 1), preferably milk, casein or whey protein concentrates. Since they are all milk sequences, it is obvious that any preparation containing the starting proteins could be used: β-lactoglobulin, β-casein, or ¾i-casein of different species, or peptides or fragments thereof of any size, alone or mixed with other proteins. For example: whole casein, caseinates and milk in its different forms of presentation, fermented milk products, hydrolysed milk proteins, milk by-products, dairy products for animal feed, etc. Said starting material is dissolved or dispersed, at an appropriate concentration, in water or in a buffer solution, at a pH suitable for the action of the proteolytic enzyme. Any proteolytic enzyme capable of breaking the protein present in the starting material and providing the peptides of interest may be used, but preferably pepsin at pH 2.0-3.0 or trypsin at pH 7.5-8.5. Proteolytic microorganisms that carried out fermentation of the substrate and hydrolysis of the protein could also be used.
Las condiciones de hidrólisis: pH, temperatura, relación enzima- sustrato, interrupción de la reacción etc., se optimizan con el fin de seleccionar los hidrolizados más activos. En una realización particular, se obtienen los péptidos bioactivos empleando caseína hidrolizada con pepsina a pH 2,5, en una relación enzima/sustrato 4/100 (p/p) y realizando la hidrólisis a 37°C, durante un periodo de tiempo comprendido entre 10 minutos y 24 horas, pero, preferiblemente durante un tiempo inferior a 6 horas. En otra realización particular, los péptidos se obtienen hidrolizando un concentrado de proteínas de suero con tripsina a pH 8,0, en una relación enzima/sustrato 5/100 (p/p) y realizando la hidrólisis a 37°C, durante un periodo de tiempo comprendido entre 10 minutos y 24 horas, pero, preferiblemente durante un tiempo inferior a 4 horas . The hydrolysis conditions: pH, temperature, enzyme-substrate ratio, reaction interruption etc., are optimized in order to select the most active hydrolysates. In a particular embodiment, the bioactive peptides are obtained using casein hydrolyzed with pepsin at pH 2.5, in an enzyme / substrate ratio 4/100 (w / w) and performing hydrolysis at 37 ° C, for a period of time between 10 minutes and 24 hours, but preferably for less than 6 hours. In another particular embodiment, the peptides are obtained by hydrolyzing a whey protein concentrate with trypsin at pH 8.0, in a 5/100 enzyme / substrate ratio (w / w) and performing hydrolysis at 37 ° C, over a period of time. of time between 10 minutes and 24 hours, but preferably for less than 4 hours.
En el ámbito de la presente invención, la composición farmacéutica se refiere a la mezcla de un principio activo, o mezcla de principios activos, con los excipientes adecuados (lactosa, maltodextrina, etc.) para dar formas orales de administración, preferentemente comprimidos o cápsulas . Se entiende por principio activo los péptidos individuales, la mezcla de los mismos o los hidrolizados enzimáticos que los contienen. Las dosis de los péptidos de interés pueden estar comprendidas entre 1 a 10 mg/día, pero preferiblemente se administrarán a dosis de 5 a 7 mg/día. Within the scope of the present invention, the pharmaceutical composition refers to the mixture of an active ingredient, or mixture of active ingredients, with suitable excipients (lactose, maltodextrin, etc.) to give oral administration forms, preferably tablets or capsules. . The active principle is understood as the individual peptides, the mixture thereof or the enzymatic hydrolysates that contain them. The doses of the peptides of interest may be between 1 to 10 mg / day, but preferably they will be administered at doses of 5 to 7 mg / day.
En el ámbito de la presente invención, ingrediente funcional se refiere a un ingrediente alimentario que además de ejercer un efecto beneficioso en el individuo que lo consume por su valor nutritivo, demuestra satisfactoriamente mejorar una o más funciones en el organismo. La función en el organismo del ingrediente funcional viene referida tanto a la mejora de una función fisiológica, como a la disminución del riesgo a padecer una enfermedad. El ingrediente funcional puede contener los péptidos individuales, la mezcla de los mismos o los hidrolizados enzimáticos que los contienen, además de distintos excipientes con el fin de mejorar sus características organolépticas o tecnológicas (lactosa, fructosa, almidón, maltodextrinas , etc.). Este ingrediente funcional podría incorporarse a distintos alimentos compatibles con una dieta saludable como yogures, zumos, fórmulas infantiles, barritas de cereales, preparados y postres lácteos, preparados alimenticios en polvo, etc. Within the scope of the present invention, functional ingredient refers to a food ingredient that in addition to exercising a Beneficial effect on the individual who consumes it for its nutritional value, satisfactorily demonstrates improving one or more functions in the body. The function in the organism of the functional ingredient is referred both to the improvement of a physiological function, and to the decrease in the risk of suffering from a disease. The functional ingredient may contain the individual peptides, the mixture thereof or the enzymatic hydrolysates that contain them, in addition to different excipients in order to improve their organoleptic or technological characteristics (lactose, fructose, starch, maltodextrins, etc.). This functional ingredient could be incorporated into different foods compatible with a healthy diet such as yogurts, juices, infant formulas, cereal bars, dairy preparations and desserts, powdered food preparations, etc.
La composición farmacéutica o el ingrediente funcional anteriores son de utilidad para estimular la formación de la mucosa intestinal . The pharmaceutical composition or functional ingredient above are useful for stimulating the formation of the intestinal mucosa.
En una realización preferida, el péptido bioactivo aislado para la fabricación de la composición farmacéutica o del ingrediente funcional comprende al menos un péptido seleccionado entre al menos uno del grupo que consiste en: SEQ ID No: 1, SEQ ID No: 2 y una combinación de ambos. Dichos péptidos SEQ ID No: 1 y/o SEQ ID No: 2 (Tabla 2) están presentes en hidrolizados de proteínas de suero (preferentemente en hidrolizados de proteínas de suero con tripsina) e incrementan la actividad secretora de mucinas en células epiteliales intestinales productoras de mucinas, y por tanto, estimulan la formación de la mucosa intestinal. Debe entenderse que el uso de un hidrolizado enzimático de proteínas de suero, que comprenda al menos uno de los péptidos individuales SEQ ID No: 1, SEQ ID No: 2, o una combinación de los mismos, es igualmente válido para la elaboración de la composición farmacéutica o del ingrediente funcional. In a preferred embodiment, the isolated bioactive peptide for the manufacture of the pharmaceutical composition or functional ingredient comprises at least one peptide selected from at least one of the group consisting of: SEQ ID No: 1, SEQ ID No: 2 and a combination from both. Said peptides SEQ ID No: 1 and / or SEQ ID No: 2 (Table 2) are present in whey protein hydrolysates (preferably in whey protein hydrolysates with trypsin) and increase the secretory activity of mucins in producing intestinal epithelial cells. of mucins, and therefore, stimulate the formation of the intestinal mucosa. It should be understood that the use of a whey protein enzyme hydrolyzate, comprising at least one of the individual peptides SEQ ID No: 1, SEQ ID No: 2, or a combination thereof, is equally valid for the preparation of the pharmaceutical composition or functional ingredient.
Tabla 2. Secuencias de péptidos lácteos con actividad secretora de mucinas derivados de proteínas de suero . YLLF SEQ ID No: 1 Table 2. Sequences of dairy peptides with secretory activity of mucins derived from whey proteins. YLLF SEQ ID No: 1
YLLF-NH2 SEQ ID No: 2 YLLF-NH 2 SEQ ID No: 2
La tecnología disponible permite que los péptidos bioactivos identificados en la Tabla 2 (SEQ ID No: 1 y SEQ ID No: 2), al conocerse su secuencia, puedan obtenerse por síntesis química y/o enzimática, hidrólisis de proteínas de diferentes especies (vaca, oveja, cabra, etc.), hidrólisis de proteínas humanas recombinantes o por métodos recombinantes de producción heterologa. The available technology allows the bioactive peptides identified in Table 2 (SEQ ID No: 1 and SEQ ID No: 2), upon knowing their sequence, can be obtained by chemical and / or enzymatic synthesis, hydrolysis of proteins of different species (cow , sheep, goat, etc.), hydrolysis of recombinant human proteins or by recombinant methods of heterologous production.
En otra realización preferida, el péptido bioactivo aislado para la fabricación de la composición farmacéutica o del ingrediente funcional comprende al menos un péptido seleccionado entre al menos uno del grupo que consiste en: SEQ ID No: 3, SEQ ID No: 4, SEQ ID No: 5, SEQ ID No: 6, SEQ ID No: 7, SEQ ID No: 8, SEQ ID No: 9, SEQ ID No: 10, SEQ ID No: 11, SEQ ID No: 12 y cualquier combinación de los mismos. Los péptidos SEQ ID No : 3, SEQ ID No: 4, SEQ ID No : 5, SEQ ID No: 6, SEQ ID No: 7, SEQ ID No: 8, SEQ ID No: 9, SEQ ID No: 10, SEQ ID No: 11 y/o SEQ ID No: 12 (Tabla 3) están presentes en hidrolizados de caseínas (preferentemente en hidrolizados de caseína de la leche con pepsina) e incrementan la actividad secretora de mucinas en células epiteliales intestinales productoras de mucinas, estimulando por tanto la formación de la mucosa intestinal. In another preferred embodiment, the isolated bioactive peptide for the manufacture of the pharmaceutical composition or functional ingredient comprises at least one peptide selected from at least one of the group consisting of: SEQ ID No: 3, SEQ ID No: 4, SEQ ID No: 5, SEQ ID No: 6, SEQ ID No: 7, SEQ ID No: 8, SEQ ID No: 9, SEQ ID No: 10, SEQ ID No: 11, SEQ ID No: 12 and any combination of the same. Peptides SEQ ID No: 3, SEQ ID No: 4, SEQ ID No: 5, SEQ ID No: 6, SEQ ID No: 7, SEQ ID No: 8, SEQ ID No: 9, SEQ ID No: 10, SEQ ID No: 11 and / or SEQ ID No: 12 (Table 3) are present in casein hydrolysates (preferably in casein hydrolysates of milk with pepsin) and increase the secretory activity of mucins in intestinal mucin-producing epithelial cells, thus stimulating the formation of the intestinal mucosa.
Tabla 3. Secuencias de los péptidos lácteos con actividad secretora de mucinas derivados de caseínas . Table 3. Sequences of dairy peptides with secretory activity of casein-derived mucins.
YPSF-NH2 SEQ ID No: 3 YPSF-NH2 SEQ ID No: 3
YPFV-NH2 SEQ ID No: 4  YPFV-NH2 SEQ ID No: 4
YPFVE SEQ ID No: 5  YPFVE SEQ ID No: 5
RYLGY SEQ ID No: 6  RYLGY SEQ ID No: 6
YLGY SEQ ID No: 7  YLGY SEQ ID No: 7
AYFYPEL SEQ ID No: 8  AYFYPEL SEQ ID No: 8
YFYPEL SEQ ID No: 9  YFYPEL SEQ ID No: 9
YFYPE SEQ ID No: 10  YFYPE SEQ ID No: 10
YFYP SEQ ID No: 11 YFY SEQ ID No: 12 YFYP SEQ ID No: 11 YFY SEQ ID No: 12
La tecnología disponible permite que los péptidos bioactivos identificados en la Tabla 3 (SEQ ID No: 3, SEQ ID No: 4, SEQ ID No: 5, SEQ ID No: 6, SEQ ID No: 7, SEQ ID No: 8, SEQ ID No: 9, SEQ ID No: 10, SEQ ID No: 11, SEQ ID No: 12), al conocerse su secuencia, puedan obtenerse por síntesis química y/o enzimática, hidrólisis de proteínas de diferentes especies (vaca, oveja, cabra, etc.) , hidrólisis de proteínas humanas recombinantes o por métodos recombinantes de producción heterologa. The available technology allows the bioactive peptides identified in Table 3 (SEQ ID No: 3, SEQ ID No: 4, SEQ ID No: 5, SEQ ID No: 6, SEQ ID No: 7, SEQ ID No: 8, SEQ ID No: 9, SEQ ID No: 10, SEQ ID No: 11, SEQ ID No: 12), upon knowing its sequence, can be obtained by chemical and / or enzymatic synthesis, hydrolysis of proteins of different species (cow, sheep , goat, etc.), hydrolysis of recombinant human proteins or by recombinant methods of heterologous production.
En otra realización preferida, el péptido bioactivo aislado para la fabricación de la composición farmacéutica o del ingrediente funcional comprende al menos un péptido seleccionado entre al menos uno del grupo que consiste en: SEQ ID No: 3, SEQ ID No: 4, SEQ ID No: 5 y cualquier combinación de los mismos. Los péptidos SEQ ID No: 3, SEQ ID No: 4 y/o SEQ ID No: 5 están presentes en un hidrolizado de la fracción de /3-caseína de la leche (preferentemente en hidrolizados de /3-caseína de la leche con pepsina) . Por lo tanto, para la elaboración de la composición farmacéutica o del ingrediente funcional, según se entiende la invención, es posible utilizar al menos uno de los péptidos definidos por SEQ ID No: 3, SEQ ID No: 4, SEQ ID No: 5, o una combinación cualquiera de dichos péptidos, o un hidrolizado enzimático de la fracción de /3-caseína de la leche que comprenda al menos uno de dichos péptidos. In another preferred embodiment, the isolated bioactive peptide for the manufacture of the pharmaceutical composition or functional ingredient comprises at least one peptide selected from at least one of the group consisting of: SEQ ID No: 3, SEQ ID No: 4, SEQ ID No: 5 and any combination thereof. The peptides SEQ ID No: 3, SEQ ID No: 4 and / or SEQ ID No: 5 are present in a hydrolyzate of the milk / 3-casein fraction (preferably in milk / 3-casein hydrolysates with pepsin) Therefore, for the preparation of the pharmaceutical composition or functional ingredient, as the invention is understood, it is possible to use at least one of the peptides defined by SEQ ID No: 3, SEQ ID No: 4, SEQ ID No: 5 , or any combination of said peptides, or an enzymatic hydrolyzate of the milk / 3-casein fraction comprising at least one of said peptides.
En otra realización preferida, el péptido bioactivo aislado para la fabricación de la composición farmacéutica o del ingrediente funcional comprende al menos un péptido seleccionado entre al menos uno del grupo que consiste en: SEQ ID No: 6, SEQ ID No: 7, SEQ ID No: 8, SEQ ID No: 9, SEQ ID No: 10, SEQ ID No: 11, SEQ ID No: 12 y cualquier combinación de los mismos. Los péptidos SEQ ID No: 6, SEQ ID No: 7, SEQ ID No: 8, SEQ ID No: 9, SEQ ID No: 10, SEQ ID No: 11 y/o SEQ ID No: 12 están presentes en hidrolizados de la fracción de ¾i-caseína de la leche (preferentemente en hidrolizados de sl- caseína de la leche con pepsina) , y como se mencionó anteriormente incrementan la actividad secretora de mucinas en células epiteliales intestinales productoras de mucinas. Además, cualquiera de los péptidos SEQ ID No: 8, SEQ ID No: 9, SEQ ID No: 10, SEQ ID No: 11, SEQ ID No: 12 o una combinación de los mismos, además de actividad estimulante de la secreción de mucinas, presentan actividad opioide en preparaciones de íleon de cobayo (Tabla 4) . En consecuencia y al igual que en los casos anteriores, para la elaboración de la composición farmacéutica o del ingrediente funcional según la invención, es posible utilizar al menos uno de los péptidos individuales definidos por SEQ ID No: 6, SEQ ID No: 7, SEQ ID No: 8, SEQ ID No: 9, SEQ ID No: 10, SEQ ID No: 11, SEQ ID No: 12, o cualquier combinación de dichos péptidos, o un hidrolizado enzimático de la fracción de ¾i-caseína de la leche que comprenda al menos uno de dichos péptidos . Al poseer estas secuencias actividad opioide podrían ejercer distintas funciones fisiológicas relacionadas con la interacción con receptores opioides a nivel intestinal como el retardo del tránsito intestinal, actividades relacionadas con el sistema inmune, etc. In another preferred embodiment, the isolated bioactive peptide for the manufacture of the pharmaceutical composition or functional ingredient comprises at least one peptide selected from at least one of the group consisting of: SEQ ID No: 6, SEQ ID No: 7, SEQ ID No: 8, SEQ ID No: 9, SEQ ID No: 10, SEQ ID No: 11, SEQ ID No: 12 and any combination thereof. Peptides SEQ ID No: 6, SEQ ID No: 7, SEQ ID No: 8, SEQ ID No: 9, SEQ ID No: 10, SEQ ID No: 11 and / or SEQ ID No: 12 are present in hydrolysates of the ¾i-casein fraction of milk (preferably in sl -casein hydrolysates of milk with pepsin), and as mentioned above increase the secretory activity of mucins in intestinal epithelial cells producing mucins. In addition, any of the peptides SEQ ID No: 8, SEQ ID No: 9, SEQ ID No: 10, SEQ ID No: 11, SEQ ID No: 12 or a combination thereof, in addition to stimulating secretion activity of mucins, present opioid activity in guinea pig ileum preparations (Table 4). Accordingly and as in the previous cases, for the preparation of the pharmaceutical composition or the functional ingredient according to the invention, it is possible to use at least one of the individual peptides defined by SEQ ID No: 6, SEQ ID No: 7, SEQ ID No: 8, SEQ ID No: 9, SEQ ID No: 10, SEQ ID No: 11, SEQ ID No: 12, or any combination of said peptides, or an enzymatic hydrolyzate of the ¾i-casein fraction of the milk comprising at least one of said peptides. By possessing these opioid activity sequences, they could exercise different physiological functions related to the interaction with opioid receptors at the intestinal level such as delayed intestinal transit, activities related to the immune system, etc.
Tabla 4. Secuencias de los péptidos lácteos derivados de caseínas con actividad opioide . Table 4. Sequences of milk peptides derived from caseins with opioid activity.
Figure imgf000013_0001
Figure imgf000013_0001
Debe entenderse que con el uso de al menos uno de los péptidos bioactivos aislados anteriores, para la fabricación de una composición farmacéutica o un ingrediente funcional para estimular la formación de la mucosa intestinal (y en consecuencia mejorar la protección gastrointestinal), como se describió anteriormente, la presente invención también contempla la protección de un péptido bioactivo aislado que: a) posee actividad estimulante de secreción de mucinas y/o actividad opioide; b) está presente en un hidrolizado enzimático de al menos una proteina de la leche seleccionado entre un hidrolizado de proteínas de suero (preferentemente un hidrolizado de proteínas de suero con tripsina) o un hidrolizado de caseína de la leche (preferentemente un hidrolizado de caseína de la leche con pepsina) ; c) su estructura comprende una secuencia con al menos dos aminoácidos aromáticos, donde el primer aminoácido aromático es una tirosina situada en primera o segunda posición respecto al extremo N-terminal de dicha secuencia y el segundo aminoácido aromático es una fenilalanina o una tirosina situada en tercera o cuarta posición con respecto a la primera tirosina; y donde dicha estructura se selecciona entre SEQ ID No: 1, SEQ ID No: 2, SEQ ID No: 3, SEQ ID No: 4, SEQ ID No: 5, SEQ ID No: 6, SEQ ID No: 7, SEQ ID No: 8, SEQ ID No: 9, SEQ ID No: 10, SEQ ID No: 11 y SEQ ID No: 12; para estimular la formación de la mucosa intestinal y/o favorecer la protección gastrointestinal, y por tanto para su uso como estimulante de la formación de la mucosa intestinal y/o como protector gastrointestinal. No obstante, es posible que para su aplicación, preferentemente dicho péptido se encuentre formando parte como un componente de una composición farmacéutica o de un ingrediente funcional, como los definidos anteriormente. It should be understood that with the use of at least one of the above isolated bioactive peptides, for the manufacture of a pharmaceutical composition or a functional ingredient to stimulate the formation of the intestinal mucosa (and consequently improve gastrointestinal protection), as described above. , the present invention also contemplates the protection of an isolated bioactive peptide that: a) possesses mucin secretion stimulating activity and / or opioid activity; b) is present in an enzymatic hydrolyzate of at least one milk protein selected from a whey protein hydrolyzate (preferably a whey protein hydrolyzate with trypsin) or a milk casein hydrolyzate (preferably a casein hydrolyzate of pepsin milk); c) its structure comprises a sequence with at least two aromatic amino acids, where the first aromatic amino acid is a tyrosine located in the first or second position with respect to the N-terminal end of said sequence and the second aromatic amino acid is a phenylalanine or a tyrosine located in third or fourth position with respect to the first tyrosine; and where said structure is selected from SEQ ID No: 1, SEQ ID No: 2, SEQ ID No: 3, SEQ ID No: 4, SEQ ID No: 6, SEQ ID No: 7, SEQ ID No: 8, SEQ ID No: 9, SEQ ID No: 10, SEQ ID No: 11 and SEQ ID No: 12; to stimulate the formation of the intestinal mucosa and / or promote gastrointestinal protection, and therefore for its use as a stimulant of the formation of the intestinal mucosa and / or as a gastrointestinal protector. However, it is possible that for its application, said peptide is preferably being part as a component of a pharmaceutical composition or a functional ingredient, as defined above.
Del mismo modo, el uso del primer aspecto de la invención puede ser igualmente protegido como un método de tratamiento para estimular la formación de la mucosa intestinal y/o favorecer la protección gastrointestinal caracterizado porque comprende administrar a un individuo una cantidad terapéuticamente eficaz de un péptido bioactivo aislado que: a) posee actividad estimulante de secreción de mucinas y/o actividad opioide; b) está presente en un hidrolizado enzimático de al menos una proteína de la leche seleccionado entre un hidrolizado de proteínas de suero (preferentemente un hidrolizado de proteínas de suero con tripsina) o un hidrolizado de caseína de la leche (preferentemente un hidrolizado de caseína de la leche con pepsina) ; c) su estructura comprende una secuencia con al menos dos aminoácidos aromáticos, donde el primer aminoácido aromático es una tirosina situada en primera o segunda posición respecto al extremo N-terminal de dicha secuencia y el segundo aminoácido aromático es una fenilalanina o una tirosina situada en tercera o cuarta posición con respecto a la primera tirosina; y donde dicha estructura se selecciona entre SEQ ID No: 1, SEQ ID No: 2, SEQ ID No: 3, SEQ ID No: 4, SEQ ID No: 5, SEQ ID No: 6, SEQ ID No: 7, SEQ ID No: 8, SEQ ID No: 9, SEQ ID No: 10, SEQ ID No: 11 y SEQ ID No: Similarly, the use of the first aspect of the invention can also be protected as a method of treatment to stimulate the formation of the intestinal mucosa and / or favor gastrointestinal protection characterized in that it comprises administering to an individual a therapeutically effective amount of a peptide. isolated bioactive that: a) has mucin secretion stimulating activity and / or opioid activity; b) is present in an enzymatic hydrolyzate of at least one milk protein selected from a whey protein hydrolyzate (preferably a whey protein hydrolyzate with trypsin) or a milk casein hydrolyzate (preferably a milk casein hydrolyzate with pepsin); c) its structure comprises a sequence with at least two aromatic amino acids, where the first aromatic amino acid is a tyrosine located in the first or second position with respect to the N-terminal end of said sequence and the second aromatic amino acid is a phenylalanine or a tyrosine located in third or fourth position with respect to the first tyrosine; and where said structure is selected from SEQ ID No: 1, SEQ ID No: 2, SEQ ID No: 3, SEQ ID No: 4, SEQ ID No: 6, SEQ ID No: 7, SEQ ID No: 8, SEQ ID No: 9, SEQ ID No: 10, SEQ ID No: 11 and SEQ ID No:
Según la presente invención, el término cantidad terapéuticamente eficaz se refiere a la cantidad de los péptidos de la invención calculada para producir el efecto deseado. Concretamente, viene referido a la cantidad de péptido, o mezcla de péptidos, o de los hidrolizados que contienen los péptidos, que es capaz de inducir la secreción de mucinas a nivel gastrointestinal y así contribuir a la protección del mismo. Las dosis de los péptidos de interés pueden estar comprendidos entre 1 a 10 mg/día, pero preferiblemente se administrarán a dosis de 5 a 7 mg/día. Las dosis de los hidrolizados que los contienen pueden estar comprendidos entre 0.5- 5 g/día, pero preferiblemente se administrarán a dosis de 1-2 g/día . According to the present invention, the term "therapeutically effective amount" refers to the amount of the peptides of the invention calculated to produce the desired effect. Specifically, it refers to the amount of peptide, or mixture of peptides, or of the hydrolysates that contain the peptides, which is capable of inducing the secretion of mucins at the gastrointestinal level and thus contributing to the protection thereof. The doses of the peptides of interest may be between 1 to 10 mg / day, but preferably they will be administered at doses of 5 to 7 mg / day. The doses of the hydrolysates containing them may be between 0.5-5 g / day, but preferably they will be administered at doses of 1-2 g / day.
Explicación detallada de las figuras Detailed explanation of the figures
Figura 1. Efecto de los péptidos SEQ ID No: 5 (A), SEQ ID No : 6 (B) , SEQ ID No: 8 (C) en una concentración de 0,1 mM sobre la secreción de mucinas en células HT29-MTX determinado mediante ensayo de lectina conjugado a enzimas (ELLA) . Los datos están expresados como porcentaje de secreción de mucinas frente a control (células sin tratar) . Cada punto representa la media + error estándar de la media de tres réplicas biológicas por triplicado. Diferencias significativas obtenidas mediante ANOVA de dos vías aplicando el test de Bonferroni . ***P<0,001; *P<0,05. Figura 2. Efecto de los hidrolizados obtenidos de proteínas de suero (A), y de caseínas (B) en concentraciones de 0,1 y 1,0 % sobre el nivel de RNAm de MUC5AC determinado mediante PCR cuantitativa (RT-qPCR) entre 2 y 24 horas de exposición. Los datos están expresados como nivel de expresión relativa de MUC5AC frente al control (células sin tratar) , empleando ciclofilina como gen de referencia. Cada punto representa la media + el error estándar de la media de tres réplicas biológicas por triplicado. Diferencias significativas obtenidas mediante ANOVA de dos vías aplicando el test de Bonferroni . *P<0,05. Figure 1. Effect of peptides SEQ ID No: 5 (A), SEQ ID No: 6 (B), SEQ ID No: 8 (C) at a concentration of 0.1 mM on mucin secretion in HT29- cells MTX determined by enzyme conjugated lectin assay (ELLA). The data are expressed as a percentage of mucin secretion versus control (untreated cells). Each point represents the mean + standard error of the average of three biological replicates in triplicate. Significant differences obtained through two-way ANOVA applying the Bonferroni test. *** P <0.001; * P <0.05. Figure 2. Effect of hydrolysates obtained from whey proteins (A), and caseins (B) in concentrations of 0.1 and 1.0% on the level of MUC5AC mRNA determined by quantitative PCR (RT-qPCR) between 2 and 24 hours of exposure. The data are expressed as relative expression level of MUC5AC versus the control (untreated cells), using cyclophilin as the reference gene. Each point represents the mean + the standard error of the average of three biological replicates in triplicate. Significant differences obtained through two-way ANOVA applying the Bonferroni test. * P <0.05.
Figura 3. Efecto de los péptidos SEQ ID No: 2 (A), SEQ ID No: 6 (B) y SEQ ID No : 8 (C) en concentraciones de 0,5, 0,1 y 0,05 mM sobre el nivel de RNAm de MUC5AC determinado mediante PCR cuantitativa (RT-qPCR) entre 2 y 24 horas de exposición. Los datos están expresados como nivel de expresión relativa de MUC5AC frente al control (células sin tratar), empleando ciclofilina como gen de referencia. Cada punto representa la media + el error estándar de la media de tres réplicas biológicas por triplicado. Diferencias significativas obtenidas mediante ANOVA de dos vías aplicando el test de Bonferroni. **P<0,01; *P<0,05. Figure 3. Effect of peptides SEQ ID No: 2 (A), SEQ ID No: 6 (B) and SEQ ID No: 8 (C) in concentrations of 0.5, 0.1 and 0.05 mM on the MUC5AC mRNA level determined by quantitative PCR (RT-qPCR) between 2 and 24 hours of exposure. The data are expressed as relative expression level of MUC5AC versus the control (untreated cells), using cyclophilin as the reference gene. Each point represents the mean + the standard error of the average of three biological replicates in triplicate. Significant differences obtained through two-way ANOVA applying the Bonferroni test. ** P <0.01; * P <0.05.
Figura 4. Efecto de los péptidos SEQ ID No: 9 (A) y SEQ ID No: 10Figure 4. Effect of peptides SEQ ID No: 9 (A) and SEQ ID No: 10
(B) en la preparación fibra longitudinal-plexo mientérico (FL-PM) de íleon de cobayo. Cada punto representa la media del porcentaje de inhibición de la respuesta contráctil inducida eléctricamente de 3-4 réplicas + error estándar de la media en al menos 3 animales diferentes por la administración acumulativa de los péptidos . (B) in the preparation longitudinal fiber-myenteric plexus (FL-PM) of guinea pig ileum. Each point represents the average percentage inhibition of the electrically induced contractile response of 3-4 replicates + standard error of the mean in at least 3 different animals by the cumulative administration of the peptides.
Ejemplos de realización de la invención Examples of embodiment of the invention
A continuación se ilustrará la invención mediante unos ensayos realizados por los inventores, que ponen de manifiesto la especificidad y efectividad de los hidrolizados de de las proteínas de suero y caseínas y de los péptidos que contienen sobre la secreción de mucinas y la expresión del gen MUC5AC en células HT29- MTX. Además, se ilustra la actividad opioide de algunos de ellos en ensayos realizados en preparaciones de íleon de cobayo. a) MATERIALES Y MÉTODOS a.l) Péptidos e hidrolizados The invention will now be illustrated by tests carried out by the inventors, which show the specificity and effectiveness of the hydrolysates of whey proteins and caseins and the peptides they contain on mucin secretion and the expression of the MUC5AC gene in HT29-MTX cells. In addition, the opioid activity of some of them is illustrated in trials conducted in guinea pig ileum preparations. a) MATERIALS AND METHODS al) Peptides and hydrolysates
La β-casomorfina 7 bovina f (60-66) de la β-caseina A2 de secuencia YPFPGPI se adquirió de Bachem (Bubendorf, Suiza) y se utilizó como control positivo. Los péptidos SEQ ID No: 1, SEQ ID No: 2, SEQ ID No: 3, SEQ ID No: 4, SEQ ID No: 5, SEQ ID No: 6, SEQ ID No: 7, SEQ ID No: 8, SEQ ID No: 9, SEQ ID No: 10, SEQ ID No: 11 y SEQ ID No: 12, y los péptidos oí-lactorfina humana y bovina f (50-53) de la OÍ- lactoalbúmina de secuencia YGLF-NH2, la forma desaminada de la OÍ - lactorfina bovina f (50-53) de la α-lactoalbúmina de secuencia YGLF, la morficeptina bovina, f (60-63) de la β-caseina A2 de secuencia YPFP-NH2, la neocasomorfina bovina f (114-119) de la β-caseina de secuencia YPVEPF y la exorfina de α-caseína bovina, f (91-96) de secuencia YLGYLE, se obtuvieron mediante síntesis en fase sólida (FMOC) en un sintetizador 433A (Applied Biosystems, Warrington, UK) . La pureza de todos estos péptidos sintéticos fue superior al 90% verificándose mediante cromatografía líquida en fase inversa acoplada a espectrometría de masas en tándem (HPLC-MS/MS) . The β-casomorphine 7 bovine f (60-66) of the β-casein A2 of YPFPGPI sequence was purchased from Bachem (Bubendorf, Switzerland) and was used as a positive control. Peptides SEQ ID No: 1, SEQ ID No: 2, SEQ ID No: 3, SEQ ID No: 4, SEQ ID No: 5, SEQ ID No: 6, SEQ ID No: 7, SEQ ID No: 8, SEQ ID No: 9, SEQ ID No: 10, SEQ ID No: 11 and SEQ ID No: 12, and human and bovine oi-lactorphine peptides f (50-53) of the YGLF-NH 2 sequence OIL-lactalbumin peptides , the deaminated form of the OI - bovine lactorphine f (50-53) of the YGLF sequence α-lactalbumin, bovine morphiceptin, f (60-63) of the YPFP-NH 2 sequence β-casein A2, the neocasomorphine bovine f (114-119) of the β-casein of YPVEPF sequence and the bovine α-casein exorphine, f (91-96) of YLGYLE sequence, were obtained by solid phase synthesis (FMOC) in a 433A synthesizer (Applied Biosystems, Warrington, UK). The purity of all these synthetic peptides was greater than 90%, being verified by reverse phase liquid chromatography coupled to tandem mass spectrometry (HPLC-MS / MS).
En la preparación del hidrolizado de proteínas de suero, un concentrado de proteínas de suero (Friesland Foods Domo, Zwolle, Holanda) se disolvió en agua al 5% p/v y se calentó a 90°C durante 10 minutos. La hidrólisis se desarrolló aplicando tripsina porcina de grado alimentario (Biocatalysts , Nantgarw, Wales, UK) con una relación enzima-sustrato del 5% p/p, durante 3 horas a 37 °C y pH 8,0. La reacción finalizó mediante inactivación de la enzima por tratamiento térmico a 95 °C durante 15 minutos. In the preparation of whey protein hydrolyzate, a whey protein concentrate (Friesland Foods Domo, Zwolle, The Netherlands) was dissolved in 5% w / v water and heated at 90 ° C for 10 minutes. Hydrolysis was carried out by applying food grade porcine trypsin (Biocatalysts, Nantgarw, Wales, UK) with an enzyme-substrate ratio of 5% w / w, for 3 hours at 37 ° C and pH 8.0. The reaction was terminated by inactivation of the enzyme by heat treatment at 95 ° C for 15 minutes.
El hidrolizado de caseínas se obtuvo a partir de caseína comercial (Promilk 85, Arras Cedex, Francia) al 6% p/v en agua con un pH final de 2,5. La hidrólisis se llevó a cabo con pepsina de grado alimentario (Biocatalysts) en una relación enzima-sustrato del 2% p/p, añadiéndose dos veces (momento inicial y tres horas de hidrólisis) . Se incubó a 40 °C durante 6 horas con inactivación final de la enzima por calentamiento a 80-85°C durante 15 minutos. La identificación de los péptidos contenidos en los hidrolizados se realizó mediante HPLC-MS/MS. a.2) Análisis mediante RP-HPLC acoplado on-line a espectrometría de masas en tándem (RP-HPLC-MS/MS ) Casein hydrolyzate was obtained from commercial casein (Promilk 85, Arras Cedex, France) at 6% w / v in water with a final pH of 2.5. Hydrolysis was carried out with food grade pepsin (Biocatalysts) in an enzyme-substrate ratio of 2% w / w, being added twice (initial moment and three hours of hydrolysis). It was incubated at 40 ° C for 6 hours with final inactivation of the enzyme by heating at 80-85 ° C for 15 minutes. The peptides contained in the hydrolysates were identified by HPLC-MS / MS. a.2) Analysis via RP-HPLC coupled online to tandem mass spectrometry (RP-HPLC-MS / MS)
Se empleó un equipo Esquire-LC (Bruker Daltonik GMBH, Bremen, Alemania) . El equipo de HPLC (serie 1100) estaba formado por una bomba cuaternaria, un inyector automático, un sistema de desgasificación de eluyentes y un detector ultravioleta de longitud de onda variable (Agilent Technologies, Waldbronn, Alemania) acoplado en línea a un espectrómetro de masas de trampa iónica Esquire 3000 (Bruker Daltonik) . La columna fue una columna Hi-Pore C18 (250 x 4,6 mm d.i., 5 \im de tamaño de partícula) (Bio-Rad Laboratories, Richmond, CA, USA) . El disolvente A fue una mezcla de agua y ácido trifluoroacético (1000:0,37) y el disolvente B una mezcla de acetonitrilo y ácido trifluoroacético (1000:0,27) . Se inyectaron 50 μΐ de muestra preparada a una concentración de 4,5 mg/ml. Se empleó un flujo de 0,8 ml/min, con un gradiente lineal del 0% al 50% de disolvente B en 60 minutos. El eluyente se monitorizó a 214 nm y el flujo de la muestra hacia el nebulizador del espectrómetro de masas fue de 275 μΐ/min. La ionización por electrospray (ESI) utilizó N2 como agente nebulizador (8 L/min) , presión de nebulización 60 psi, temperatura de secado 350 °C y un voltaje del capilar de 4 KV. Los espectros de masa se adquirieron en el rango de 100-2500 m/ z y se empleó un rampa de fragmentación comprendida entre 0,3 y 2 V. Como sistema de adquisición de datos se utilizó el programa Esquire ControlTM 5.0 (Bruker Daltonik) y para identificar la secuencia de los diferentes péptidos se utilizaron los programas informáticos Data AnalysisTM 3.0, Sequence EditorTM 2.1 y BiotoolsTM 2.1 todos ellos de Bruker Daltonik. a.3) Cultivos celulares Esquire-LC equipment (Bruker Daltonik GMBH, Bremen, Germany) was used. The HPLC equipment (1100 series) consisted of a quaternary pump, an automatic injector, an eluent degassing system and a variable wavelength ultraviolet detector (Agilent Technologies, Waldbronn, Germany) coupled in line to a mass spectrometer of ionic trap Esquire 3000 (Bruker Daltonik). The column was a Hi-Pore C18 column (250 x 4.6 mm ID, 5 particle size) (Bio-Rad Laboratories, Richmond, CA, USA). Solvent A was a mixture of water and trifluoroacetic acid (1000: 0.37) and solvent B a mixture of acetonitrile and trifluoroacetic acid (1000: 0.27). 50 μΐ of prepared sample was injected at a concentration of 4.5 mg / ml. A flow of 0.8 ml / min was used, with a linear gradient from 0% to 50% of solvent B in 60 minutes. The eluent was monitored at 214 nm and the sample flow to the mass spectrometer nebulizer was 275 μΐ / min. Electrospray ionization (ESI) used N2 as a nebulizer agent (8 L / min), fogging pressure 60 psi, drying temperature 350 ° C and a capillary voltage of 4 KV. The mass spectra were acquired in the range of 100-2500 m / z and a fragmentation ramp between 0.3 and 2 V was used. The Esquire ControlTM 5.0 program (Bruker Daltonik) was used as the data acquisition system and for To identify the sequence of the different peptides, the Data AnalysisTM 3.0, Sequence EditorTM 2.1 and BiotoolsTM 2.1 software were all used by Bruker Daltonik. a.3) Cell cultures
Se utilizó la línea celular HT29-MTX derivada de adenocarcinoma de colon humano, con capacidad de secreción de mucinas donada por la Dra. Thécla Lesuffleur (INSERM UMR S 938, París, Francia) . Las células se cultivaron en medio Eagle modificado por Dulbecco (DMEM) suplementado con 10% de suero fetal bovino inactivado y 10 mL/L de penicilina-estreptomicina a 37 °C, en atmósfera humidificada con 5% de C02. Los pases semanales se realizaron empleando tripsina/EDTA 0,05% (todos los reactivos eran de Gibco, Paisley, UK) . Las células se sembraron en placas de 12 pocilios con una densidad de 5><105 células por pocilio (Nunc, Roskilde, Dinamarca) . La linea celular se utilizó entre los pases 12 y 22. Los experimentos se realizaron 21 días después de confluencia. Desde 24 horas antes del ensayo se trabajó con medio de cultivo sin suero ni antibiótico para eliminar la interferencia de proteínas u hormonas. El día del ensayo, se retiró el medio, se lavó dos veces la monocapa celular con buffer fosfato salino (PBS) y se adicionó el medio con los péptidos en concentración 0,05, 0,1 y 0,5 mM, incubándose a 37 °C durante tiempos comprendidos entre 30 minutos y 24 horas. En los pocilios control se añadió únicamente el medio de cultivo. A los tiempos seleccionados, se recogieron los sobrenadantes que se mantuvieron congelados a -70 °C hasta su análisis. La extracción y posterior purificación del ARN celular se llevó a cabo con el kit Nucleospin® RNA II (Macherey-Nagel , Düren, Alemania) . a.4) Ensayo de Lectina Conjugada a Enzima (Enzyme-Linked Lectin Assay; ELLA) . The HT29-MTX cell line derived from human colon adenocarcinoma was used, with mucin secretion capacity donated by Dr. Thécla Lesuffleur (INSERM UMR S 938, Paris, France). Cells were grown in Dulbecco-modified Eagle medium (DMEM) supplemented with 10% inactivated bovine fetal serum and 10 mL / L of penicillin-streptomycin at 37 ° C, in a humidified atmosphere with 5% C0 2 . Weekly passes were performed using 0.05% trypsin / EDTA (all reagents were from Gibco, Paisley, UK). The cells were seeded in 12-well plates with a density of 5><10 5 cells per well (Nunc, Roskilde, Denmark). The cell line was used between passes 12 and 22. The experiments were performed 21 days after confluence. From 24 hours before the test, we worked with culture medium without serum or antibiotic to eliminate the interference of proteins or hormones. On the day of the test, the medium was removed, the cell monolayer was washed twice with phosphate buffered saline (PBS) and the medium was added with the peptides in concentration 0.05, 0.1 and 0.5 mM, incubating at 37 ° C for times between 30 minutes and 24 hours. In the control wells only the culture medium was added. At the selected times, supernatants were collected that were kept frozen at -70 ° C until analysis. Extraction and subsequent purification of cellular RNA was carried out with the Nucleospin® RNA II kit (Macherey-Nagel, Düren, Germany). a.4) Enzyme Conjugated Lectin Assay (Enzyme-Linked Lectin Assay; ELLA).
La determinación cuantitativa de mucinas totales se realizó mediante un ensayo ELLA basado en el empleo de aglutinina de germen de trigo, que ha mostrado una fuerte reactividad frente a azúcares específicos presentes en las mucinas secretadas por las células caliciformes. El contenido total de glicoproteínas en las muestras estudiadas se obtuvo mediante interpolación en una curva estándar construida a partir de mucina gástrica porcina comercial (Sigma, St Louis, CA, USA) . El tapizado de la placa de poliestireno de 96 pocilios se realizó con las muestras de la recta de calibrado y con las muestras a ensayar disueltas en buffer carbonato sódico (0,5 M, pH 9,6), dejando en incubación toda la noche a 4 °C. Se realizó el bloqueo con seroalbúmina bovina (BSA) (Sigma) al 2% durante 1 hora a 37 °C. Se añadió aglutinina de germen de trigo biotinilada (Vector Laboratories, Peterborough, UK) en PBS-Tween-BSA (1:1000), y las muestras se incubaron 1 hora a 37 °C. Posteriormente, para amplificar la señal se añadió el complejo avidina-peroxidasa (Vector laboratories ) en PBS-Tween-BSA (1:50000) . La determinación colorimétrica se llevó a cabo mediante lectura de la absorbancia a 492 nm en un lector de placas Multiskan Ascent (Labsystems, Barcelona, España) utilizando una disolución de o-fenilendiamina dihidrocloruro (OPD) (Dako, Glostrup, Dinamarca) . Los datos de cada muestra ensayada se expresaron como porcentaje de secreción de glicoproteina frente a su muestra control en el mismo tiempo y ensayo celular. a.5) Ensayo de PCR cuantitativa The quantitative determination of total mucins was performed by an ELLA assay based on the use of wheat germ agglutinin, which has shown a strong reactivity against specific sugars present in the mucins secreted by goblet cells. The total glycoprotein content in the samples studied was obtained by interpolation in a standard curve constructed from commercial porcine gastric mucin (Sigma, St Louis, CA, USA). The upholstery of the 96-well polystyrene plate was performed with the samples from the calibration line and with the samples to be tested dissolved in sodium carbonate buffer (0.5 M, pH 9.6), leaving to incubate overnight at 4 ° C The blockade was performed with bovine serum albumin (BSA) (Sigma) at 2% for 1 hour at 37 ° C. Biotinylated wheat germ agglutinin (Vector Laboratories, Peterborough, UK) was added in PBS-Tween-BSA (1: 1000), and the samples were incubated 1 hour at 37 ° C. Subsequently, to amplify the signal, the avidin-peroxidase complex (Vector laboratories) in PBS-Tween-BSA (1: 50,000) was added. Colorimetric determination was carried out by reading the absorbance at 492 nm in a Multiskan Ascent plate reader (Labsystems, Barcelona, Spain) using a solution of o-phenylenediamine dihydrochloride (OPD) (Dako, Glostrup, Denmark). The data of each sample tested was expressed as a percentage of glycoprotein secretion against its control sample at the same time and cell assay. a.5) Quantitative PCR assay
La determinación del número de copias de ARN se llevó a cabo mediante PCR cuantitativa con medida de la fluorescencia en tiempo real, empleando SYBR Green como fluoróforo en el equipo Lightcycler 480 (Roche, Mannhein, Alemania) con placas de 384 pocilios. Se obtuvo el ADNc a partir de 375 ng de ARN mediante el uso de High Capacity cDNA Reverse Transcription Kit (Applied Biosystems) de acuerdo con las instrucciones del fabricante. Para MUC5AC (código de acceso NCBI AJ001402), gen diana, se utilizaron los oligos 2870- 2889/3109-3091, mientras que para el gen de referencia ciclofilina (código de acceso NCBI Y00052) se utilizaron los oligos 280- 340/445-421 (Zoghbi et al., 2006 Am. J. Physiol. Gastrointest. Líver Physiol . 290: 1105-1113) . Se estudió cada muestra de ADNc por triplicado. Cada tubo de reacción contenia 5 μΐ de 2χ SYBR Green real-time PCR Master Mix (Applied Biosystems), 0,25 μΐ a 10 μΜ de cada uno de los oligos específicos, 0,27 μΐ de ADNc y 4,23 de H20. La amplificación se inició a 95 °C durante 5 minutos y 45 ciclos a 95 °C durante 10 segundos, 60 °C durante 10 segundos y 72 °C durante 10 segundos. Se utilizaron controles para confirmar la ausencia de dímeros de oligos y para verificar que no había contaminación por ADN. Mediante el análisis de las curvas de fusión y electroforesis en gel de agarosa al 2% se comprobó que todos los ensayos amplificaron un único producto. Para calcular la eficiencia (E=10-l/pendiente) se determinó una curva con los números de ciclos (Ct) obtenidos de la amplificación de cantidades conocidas de ADNc. Los valores de expresión relativa se calcularon utilizando el método del umbral critico (Δ ACt) . Todos los experimentos se realizaron por triplicado partiendo de tres réplicas biológicas. a.6) Análisis estadístico The RNA copy number was determined by quantitative PCR with real-time fluorescence measurement, using SYBR Green as a fluorophore in Lightcycler 480 (Roche, Mannhein, Germany) with 384 well plates. The cDNA was obtained from 375 ng of RNA by using High Capacity cDNA Reverse Transcription Kit (Applied Biosystems) according to the manufacturer's instructions. For MUC5AC (access code NCBI AJ001402), target gene, oligos 2870-2899 / 3109-3091 were used, while for the reference gene cyclophilin (access code NCBI Y00052) oligos 280-340 / 445- were used 421 (Zoghbi et al., 2006 Am. J. Physiol. Gastrointest. Líver Physiol. 290: 1105-1113). Each cDNA sample was studied in triplicate. Each reaction tube contained 5 μΐ of 2 χ SYBR Green real-time PCR Master Mix (Applied Biosystems), 0.25 μΐ to 10 μΜ of each of the specific oligos, 0.27 μΐ of cDNA and 4.23 of H 2 0. The amplification was started at 95 ° C for 5 minutes and 45 cycles at 95 ° C for 10 seconds, 60 ° C for 10 seconds and 72 ° C for 10 seconds. Controls were used to confirm the absence of oligo dimers and to verify that there was no DNA contamination. By analyzing the melting and electrophoresis curves in 2% agarose gel it was found that all the trials amplified a single product. To calculate the efficiency (E = 10-l / slope) a curve was determined with the number of cycles (Ct) obtained from the amplification of known amounts of cDNA. Relative expression values were calculated using the critical threshold method (Δ ACt). All experiments were performed in triplicate based on three biological replicas. a.6) Statistical analysis
Los datos se analizaron mediante ANOVA de dos vías, utilizando el software GraphPad Prism, seguido del test de Bonferroni para las comparaciones individuales . Las diferencias entre medias y controles se consideraron significativas con valores de P<0,05(*), P<0,01(**) o P<0, 001 (***) . a.7) Valoración in vitro de la actividad opioide de los péptidos en la preparación fibra longitudinal-plexo mientérico (FL-PM) de íleon de cobayo Data were analyzed using two-way ANOVA, using GraphPad Prism software, followed by the Bonferroni test for individual comparisons. The differences between means and controls were considered significant with values of P <0.05 (*), P <0.01 (**) or P <0.001 (***). a.7) In vitro evaluation of the opioid activity of the peptides in the longitudinal fiber-myenteric plexus (FL-PM) preparation of guinea pig ileum
Se ha estudiado la posible naturaleza opioide de los péptidos mediante la valoración de su capacidad para inhibir las contracciones inducidas eléctricamente de la preparación fibra longitudinal-plexo mientérico de íleon de cobayo (FL-PM) . Para este estudio se han utilizado cobayos albino hembra procedentes de Harían Ibérica (Castelar, España) , con un peso comprendido entre 250 - 300 g. La preparación FL-PM se obtuvo a partir de dichos segmentos intestinales, según la modificación de la técnica descrita por Ambache (1954 J. Physiol. Lond. 125: 53-55) para aislar fibras longitudinales de íleon de conejo. Se utilizó la técnica in vitro de baño de órganos, en la que el equipo utilizado está formado por copas de vidrio con capacidad para 10 mi que se llenaron con 5 mi de solución Krebs (NaCl 118, KC1 4.75; CaCl2 2.54; KH2P04 1.19; MgS04 1.2; NaHC03 25; glucosa 11 mM) , que se burbujea continuamente con gas carbógeno (95% 02 y 5% C02) y se mantiene a la temperatura de 37 °C. A continuación se le confiere a la preparación una tensión basal de 1 g, se dejan transcurrir 15 min de reposo para su estabilización y se estimulan eléctricamente de forma continuada. Las condiciones de estimulación fueron constantes: pulsos simples cuadrados de 0,3 Hz de frecuencia, 2 ms de duración y voltaje supramaximal . La actividad contráctil de las preparaciones se registró mediante un sistema de registro de datos y análisis acoplado a un transductor de fuerza isométrica. The possible opioid nature of the peptides has been studied by assessing their ability to inhibit electrically induced contractions of the longitudinal-myenteric fiber preparation of guinea pig ileum (FL-PM). For this study, female albino guinea pigs from Harían Ibérica (Castelar, Spain), weighing between 250 - 300 g, have been used. The FL-PM preparation was obtained from said intestinal segments, according to the modification of the technique described by Ambache (1954 J. Physiol. Lond. 125: 53-55) to isolate longitudinal fibers of rabbit ileum. The in vitro organ bath technique was used, in which the equipment used is made up of 10-ml glass cups that were filled with 5 ml of Krebs solution (NaCl 118, KC1 4.75; CaCl 2 2.54; KH 2 P0 4 1.19; MgS0 4 1.2; NaHC0 3 25; 11 mM glucose), which is continuously bubbled with carbon dioxide (95% 0 2 and 5% C0 2 ) and maintained at a temperature of 37 ° C. The preparation is then given a baseline tension of 1 g, allowed to rest for 15 minutes for stabilization and electrically stimulated continuously. The stimulation conditions were constant: simple square pulses of 0.3 Hz frequency, 2 ms duration and supramaximal voltage. The contractile activity of Preparations were recorded using a data recording and analysis system coupled to an isometric force transducer.
Una vez que, tras el comienzo de la estimulación eléctrica, las preparaciones presentaron una respuesta contráctil estable, se realizaron curvas concentración-respuesta, por adición al baño de órganos de dosis crecientes acumulativas de los péptidos, espaciadas en 10 min (considerado como el intervalo de tiempo requerido para que los péptidos ejerzan su efecto) . Las concentraciones evaluadas fueron: 6, 1 x 1CT8, 1,8 x 1CT7, 5,5 x 1CT7, 1,6 x 1CT6 y 5 x 1CT6 M. Tras la administración de la última dosis y tras los 10 min de espera, se añadió una única dosis del antagonista opioide naloxona (10~6 M) para comprobar si tenia lugar la reversión del efecto de los mismos y corroborar que el efecto inhibitorio estaba mediado por la interacción selectiva con receptores opioides. Con el fin de comparar el efecto de los nuevos péptidos con el de un agonista opioide conocido y con un péptido alimentario con actividad opioide descrita, de forma paralela a la evaluación de los nuevos péptidos, también se realizaron curvas de morfina y de β-casomorfina 7 siguiendo el mismo protocolo y se han utilizado como curvas patrón de referencia. Once, after the beginning of the electrical stimulation, the preparations presented a stable contractile response, concentration-response curves were performed, by adding to the bath of organs of cumulative increasing doses of the peptides, spaced in 10 min (considered as the interval of time required for the peptides to exert their effect). The concentrations evaluated were: 6, 1 x 1CT 8 , 1.8 x 1CT 7 , 5.5 x 1CT 7 , 1.6 x 1CT 6 and 5 x 1CT 6 M. After administration of the last dose and after 10 min waiting, a single dose of the opioid antagonist naloxone (10 ~ 6 M) was added to check if their effect reversal took place and to confirm that the inhibitory effect was mediated by selective interaction with opioid receptors. In order to compare the effect of the new peptides with that of a known opioid agonist and with a food peptide with described opioid activity, in parallel to the evaluation of the new peptides, morphine and β-casomorphine curves were also performed 7 following the same protocol and have been used as reference standard curves.
Los resultados se expresan como % de inhibición de la contracción, considerando como 100% la amplitud media de las 5 últimas contracciones antes de la adición de la primera dosis de la curva. Cada preparación se utilizó para llevar a cabo una única curva concentración-respuesta . The results are expressed as% contraction inhibition, considering as 100% the average amplitude of the last 5 contractions before the addition of the first dose of the curve. Each preparation was used to carry out a single concentration-response curve.
Ejemplo 1. Efecto de diferentes péptidos sintéticos alimentarios en la actividad secretora de mucinas en células HT29-MTX. Example 1. Effect of different synthetic food peptides on the secretory activity of mucins in HT29-MTX cells.
La Tabla 5 presenta los valores de máxima secreción de mucina en las células HT29-MTX tras la adición de los péptidos de SEQ ID No: 1, SEQ ID No: 2, SEQ ID No: 3, SEQ ID No: 4, SEQ ID No: 5, SEQ ID No: 6, SEQ ID No: 7 y SEQ ID No: 8 a la concentración de 0,1 mM. Además, la Tabla 5 muestra estos valores para otros seis péptidos previamente descritos como opioides. Once de los 14 péptidos alimentarios ensayados dieron lugar a un incremento significativo (P<0,001) de la actividad secretora de mucinas en las células HT29- MTX. Los valores máximos de secreción oscilaron entre 191 y 587% respecto al control (células sin tratar) . Cabe destacar que la SEQ ID No: 2 y la SEQ ID No: 5 produjeron mayores aumentos en la secreción de mucinas que la β-casomorfina 7. Table 5 shows the maximum mucin secretion values in HT29-MTX cells after the addition of the peptides of SEQ ID No: 1, SEQ ID No: 2, SEQ ID No: 3, SEQ ID No: 4, SEQ ID No: 5, SEQ ID No: 6, SEQ ID No: 7 and SEQ ID No: 8 at the concentration of 0.1 mM. In addition, Table 5 shows these values for six other peptides previously described as opioids. Eleven of the 14 food peptides tested resulted in a significant increase (P <0.001) of mucin secretory activity in HT29-MTX cells. The maximum secretion values ranged from 191 to 587% with respect to the control (untreated cells). It should be noted that SEQ ID No: 2 and SEQ ID No: 5 produced greater increases in mucin secretion than β-casomorphine 7.
Tabla 5. Secreción máxima de mucina respecto al control (células HT29-MTX no tratadas) por efecto de la adición de diferentes péptidos sintéticos alimentarios en concentración 0,1 mM. Datos obtenidos de tres replicados biológicos por triplicado. Diferencias significati as entre valores medios y controles (células no tratadas) mediante ANOVA de dos vías (Test de Bonferroni) . Table 5. Maximum mucin secretion with respect to the control (untreated HT29-MTX cells) due to the addition of different synthetic food peptides in 0.1 mM concentration. Data obtained from three biological replicates in triplicate. Significant differences between mean and control values (untreated cells) using two-way ANOVA (Bonferroni test).
Mucina Mucina
Péptido  Peptide
secretada secreted
Proteina o Protein or
o  or
Secuencia Denominación P alimentaria Control  Sequence Designation P food Control
SEQ ID Des-NH2-/3-lactorfina /3B-lactoglobulina SEQ ID Des-NH 2 - / 3-lactorphine / 3 B- lactoglobulin
163 <0, , 05 No: 1 bovina f (102-105)  163 <0,, 05 No: 1 bovine f (102-105)
SEQ ID /3B-lactoglobulina SEQ ID / 3 B -lactoglobulin
/3-lactorfina bovina 452 <o, 001 No: 2 f (102-105)  / 3-bovine lactorphine 452 <o, 001 No: 2 f (102-105)
SEQ ID /3H-caseina f (41-SEQ ID / 3 H -casein f (41-
/3-casorfina humana 215 <o, 001 No: 3 44) / 3-human casorphine 215 <, 001 No: 3 44)
SEQ ID /3H-caseina f (51-SEQ ID / 3 H -casein f (51-
Valmuceptina 259 <o, 001 No: 4 54) Valmuceptin 259 <, 001 No: 4 54)
SEQ ID /3-casomorfina 5 /3H-caseina f (51-SEQ ID / 3-casomorphine 5/3 H -casein f (51-
339 <o, 001 No: 5 humana 55) 339 <, 001 No: 5 human 55)
SEQ ID ¾j ,-) -caseína  SEQ ID ¾j, -) -casein
- 191 <o, 001 No: 6 f (90-94)  - 191 <or, 001 No: 6 f (90-94)
SEQ ID ¾j ,-) -caseína  SEQ ID ¾j, -) -casein
- 226 <o, 001 No: 7 f (91-94)  - 226 <o, 001 No: 7 f (91-94)
SEQ ID asiB_caseína SEQ ID a s iB _ casein
- 220 <o, 001 No: 8 f (143-149)  - 220 <o, 001 No: 8 f (143-149)
/3-casomorfina 7 /3B-caseína A2 / 3-casomorphine 7/3 B- casein A2
YPFPGPI 282 <o, 001 bovina f (60-66)  YPFPGPI 282 <, 001 bovine f (60-66)
α-lactorfina humana aH/B-lactalbúmina human α-lactorphine to H / B -lactalbumin
YGLF '-NH2 587 <o, 001 y bovina f (50-53) Des-NH2- Q-lactorfina aH/B-lactalbúmina YGLF '-NH 2 587 <o, 001 and bovine f (50-53) Des-NH 2 - Q-lactorphine to H / B -lactalbumin
YGLF 209 <0,001 humana y bovina f (50-53)  YGLF 209 <0.001 human and bovine f (50-53)
/3B-caseína A2 / 3 B- casein A2
YPFP-NH? MMoorrffiicceeppttiinnaa bboovviinnaa 130 ns f (60-63)  YPFP-NH? MMoorrffiicceeppttiinnaa bboovviinnaa 130 ns f (60-63)
Neocasomorfina /3B-caseína f (114-Neocasomorphine / 3 B -casein f (114-
YPVEPF 120 ns bovina 119) YPVEPF 120 ns bovine 119)
α-caseína exorfina o¾-caseína f (91- α-casein exorphine o¾-casein f (91-
YLGYLE 104 ns YLGYLE 104 ns
2-7 bovina 96)  2-7 bovine 96)
La Figura 1 muestra el efecto durante el tiempo de exposición de los péptidos de SEQ ID No: 5, SEQ ID No: 6 y SEQ ID No: 8. a una concentración 0,1 mM sobre la secrección de mucinas en células HT29-MTX. La SEQ ID No: 5 mostró valores incrementados de secreción de mucinas a los 30 minutos y 2 horas seguido de un incremento significativo a las 4 horas y a las 8 horas (Fig 1A) . Las SEQ ID No: 6 y SEQ ID No: 8 mostraron niveles absolutos inferiores a los que habían presentado los péptidos anteriormente descritos pero significativos a las 2 y 4 horas, respectivamente (Fig IB y 1C) . Figure 1 shows the effect during the exposure time of the peptides of SEQ ID No: 5, SEQ ID No: 6 and SEQ ID No: 8. at a 0.1 mM concentration on the secretion of mucins in HT29-MTX cells . SEQ ID No: 5 showed increased mucin secretion values at 30 minutes and 2 hours followed by a significant increase at 4 hours and 8 hours (Fig 1A). SEQ ID No: 6 and SEQ ID No: 8 showed absolute levels lower than those presented by the previously described but significant peptides at 2 and 4 hours, respectively (Fig IB and 1C).
Estos resultados muestran que no todas la secuencias opioides inducen la secreción de mucinas. Así, por ejemplo, los péptidos morficeptina bovina, neocasomorfina bovina y α-caseína exorfina 2-7 bovina, que se habían descrito previamente como péptidos agonistas opioides (Teschemacher 2003 Curr. Pharm. Design 9: 1331-1344), no estimularon la producción de mucinas en células epiteliales intestinales. Las secuencias YGLF-NH2 e YGLF, son capaces de favorecer la secreción de mucinas. Por otra parte, se describe por primera vez que los péptidos SEQ ID No: 1, SEQ ID No: 2, SEQ ID No: 3, SEQ ID No: 4 y SEQ ID No: 5, SEQ ID No: 6, SEQ ID No: 7 y SEQ ID No: 8 son capaces de favorecer la secreción de mucinas promoviendo de esta forma una actividad protectora a nivel intestinal. These results show that not all opioid sequences induce mucin secretion. Thus, for example, bovine morphiceptin, bovine neocasomorphine and bovine α-casein 2-7 bovine peptides, which had previously been described as opioid agonist peptides (Teschemacher 2003 Curr. Pharm. Design 9: 1331-1344), did not stimulate production of mucins in intestinal epithelial cells. The YGLF-NH2 and YGLF sequences are capable of promoting mucin secretion. On the other hand, it is described for the first time that the peptides SEQ ID No: 1, SEQ ID No: 2, SEQ ID No: 3, SEQ ID No: 4 and SEQ ID No: 5, SEQ ID No: 6, SEQ ID No: 7 and SEQ ID No: 8 are capable of promoting the secretion of mucins, thus promoting a protective activity at the intestinal level.
Ejemplo 2. Efecto de diferentes hidrolizados de proteínas lácteas derivados de las proteínas de suero y de las caseínas en la expresión del gen MUC5AC de las células HT29-MTX. Example 2. Effect of different milk protein hydrolysates derived from whey proteins and caseins on the expression of the MUC5AC gene of HT29-MTX cells.
Con el objetivo de evaluar si los hidrolizados de las proteínas de suero y de las caseínas afectaban a la expresión del gen MUC5AC se realizaron ensayos de estimulación de las células HT29-MTX después de 2, 4, 8 y 24 horas de incubación con concentraciones de los hidrolizados de 0,1% y 1%. Se observó que el hidrolizado de proteínas de suero daba lugar a un incremento de copias de MUC5AC entre las 4 y las 8 horas. La expresión basal cuando se añadió la concentración de 1% a las 4 horas fue 1,53 veces el valor basal (Figura 2A) . El hidrolizado de caseínas dio lugar a un incremento de copias de MUC5AC a entre las 4 y las 8 horas. La expresión basal cuando se añadió la concentración de 1% a las 4 horas fue 1,86 veces el valor basal (P<0,5) (Figura 2B) . In order to assess whether whey protein and casein hydrolysates affected the expression of the MUC5AC gene, performed stimulation tests of HT29-MTX cells after 2, 4, 8 and 24 hours of incubation with hydrolyzate concentrations of 0.1% and 1%. It was observed that whey protein hydrolyzate resulted in an increase in copies of MUC5AC between 4 and 8 hours. Baseline expression when the concentration of 1% was added at 4 hours was 1.53 times the baseline value (Figure 2A). Casein hydrolyzate resulted in an increase in copies of MUC5AC between 4 and 8 hours. The baseline expression when the 1% concentration was added at 4 hours was 1.86 times the baseline value (P <0.5) (Figure 2B).
Estos resultados indican que tanto los hidrolizados de proteínas de suero como los hidrolizados de caseínas ensayados producen en las células caliciformes humanas secretoras de mucinas una sobreexpresión del gen MUC5AC . These results indicate that both whey protein hydrolysates and casein hydrolysates tested produce overexpression of the MUC5AC gene in human goblet secreting cells.
Ejemplo 3. Efecto de diferentes péptidos lácteos derivados de las proteínas de suero y de las caseínas en la expresión del gen MUC5AC de células HT29-MTX. Example 3. Effect of different milk peptides derived from whey proteins and caseins on the expression of the MUC5AC gene of HT29-MTX cells.
Concentraciones comprendidas entre 0,05 y 0,5 mM de los péptidos de SEQ ID No: 2, SEQ ID No: 6 y SEQ ID No: 8 se añadieron al medio de incubación de las células para tiempos comprendidos entre 2 y 24 horas . Concentrations between 0.05 and 0.5 mM of the peptides of SEQ ID No: 2, SEQ ID No: 6 and SEQ ID No: 8 were added to the cell incubation medium for times between 2 and 24 hours.
La Figura 3 muestra los resultados de PCR cuantitativa en a unas concentraciones de 0,05 mM, 0,1 mM y 0,5 mM. El tratamiento de las células con la SEQ ID No : 2 provocó un aumento significativoFigure 3 shows the results of quantitative PCR at concentrations of 0.05 mM, 0.1 mM and 0.5 mM. Treatment of cells with SEQ ID No: 2 caused a significant increase
(P<0,05) del número de copias del gen MUC5AC a las 4 horas de exposición a la concentración de 0,5 mM (2,1 veces la expresión basal) (Fig. 3A) . El tratamiento de las células con SEQ ID No: 6 dio lugar a un aumento del número de copias de mRNA a las 4 horas(P <0.05) of the number of copies of the MUC5AC gene at 4 hours of exposure to the concentration of 0.5 mM (2.1 times the baseline expression) (Fig. 3A). Treatment of cells with SEQ ID No: 6 resulted in an increase in the number of mRNA copies at 4 hours
(entre 1,5 y 1,7 veces la expresión basal), aunque debido a la variabilidad encontrada, este incremento no fue estadísticamente significativo (Fig. 3B) . El tratamiento de las células con la SEQ ID No: 8 provocó un aumento significativo (P<0,05) (1,9 veces la expresión basal) del número de copias del gen MUC5AC a las 4 horas de exposición a la concentración de 0,1 mM (Fig. 3C) . Los tiempos en los que se detecta sobreexpresion coinciden con el tiempo al que las células tratadas con este péptido habían mostrado una mayor secreción de mucinas . (between 1.5 and 1.7 times the baseline expression), although due to the variability found, this increase was not statistically significant (Fig. 3B). Treatment of the cells with SEQ ID No: 8 caused a significant increase (P <0.05) (1.9 times the baseline expression) of the number of copies of the MUC5AC gene at 4 hours of exposure to the concentration of 0 , 1 mM (Fig. 3C). The times in which overexpression is detected coincide with the time at which the cells treated with this peptide had shown a greater mucin secretion.
Ejemplo 4. Actividad opioide de péptidos alimentarios mediante ensayo en íleon de cobayo . Example 4. Opioid activity of food peptides by guinea pig ileum assay.
La Figura 4 presenta la actividad agonista opioide de los péptidos SEQ ID No: 9 y SEQ ID No: 10 determinada in vítro mediante ensayo en la preparación FL-PM de íleon de cobayo. Los péptidos indujeron una inhibición de las contracciones inducidas eléctricamente que osciló entre 20 y 30% en el caso de la SEQ ID No: 9 (Fig 4A) y entre 25 y 35% en el caso de la SEQ ID No: 10 (Fig. 4B) , en la concentración 5,5 10~7 M. El efecto de todos los péptidos revirtió con la administración de naloxona (10~6 M) . En curvas similares la morfina dio lugar a una inhibición máxima de un 75% mientras que la /3-casomorfina 7 produjo una inhibición máxima de un 20%. Figure 4 shows the opioid agonist activity of peptides SEQ ID No: 9 and SEQ ID No: 10 determined in vitro by assay in the FL-PM preparation of guinea pig ileum. The peptides induced an inhibition of electrically induced contractions that ranged between 20 and 30% in the case of SEQ ID No: 9 (Fig 4A) and between 25 and 35% in the case of SEQ ID No: 10 (Fig. 4B), in the concentration 5.5 10 ~ 7 M. The effect of all the peptides was reversed with the administration of naloxone (10 ~ 6 M). In similar curves morphine resulted in a maximum inhibition of 75% while / 3-casomorphine 7 produced a maximum inhibition of 20%.
Estos resultados indican que las secuencias ensayadas, cuya actividad opioide no había sido previamente descrita, producen una inhibición de la contracción en la preparación FL-PM de íleon de cobayo mediada por receptores opioides superior a la de la β- casomorfina 7. These results indicate that the sequences tested, whose opioid activity had not been previously described, produce an inhibition of the contraction in the FL-PM preparation of guinea pig ileum mediated by opioid receptors greater than that of β-casomorphine 7.

Claims

REIVINDICACIONES
1. Uso de al menos un péptido bioactivo aislado que: a) posee actividad estimulante de secreción de mucinas y/o actividad opioide; b) está presente en un hidrolizado enzimático de al menos una proteina de la leche seleccionado entre un hidrolizado de proteínas de suero o un hidrolizado de caseína de la leche; c) su estructura comprende una secuencia con al menos dos aminoácidos aromáticos, donde el primer aminoácido aromático es una tirosina situada en primera o segunda posición respecto al extremo N-terminal de dicha secuencia y el segundo aminoácido aromático es una fenilalanina o una tirosina situada en tercera o cuarta posición con respecto a la primera tirosina; y donde dicha estructura se selecciona entre SEQ ID No: 1, SEQ ID No: 2, SEQ ID No: 3, SEQ ID No: 4, SEQ ID No: 5, SEQ ID No: 6, SEQ ID No: 7, SEQ ID No: 8, SEQ ID No: 9, SEQ ID No: 10, SEQ ID No: 11 y SEQ ID No: 12; para la fabricación de una composición farmacéutica o un ingrediente funcional para estimular la formación de la mucosa intestinal . 1. Use of at least one isolated bioactive peptide that: a) possesses mucin secretion stimulating activity and / or opioid activity; b) is present in an enzymatic hydrolyzate of at least one milk protein selected from a whey protein hydrolyzate or a milk casein hydrolyzate; c) its structure comprises a sequence with at least two aromatic amino acids, where the first aromatic amino acid is a tyrosine located in the first or second position with respect to the N-terminal end of said sequence and the second aromatic amino acid is a phenylalanine or a tyrosine located in third or fourth position with respect to the first tyrosine; and where said structure is selected from SEQ ID No: 1, SEQ ID No: 2, SEQ ID No: 3, SEQ ID No: 4, SEQ ID No: 6, SEQ ID No: 7, SEQ ID No: 8, SEQ ID No: 9, SEQ ID No: 10, SEQ ID No: 11 and SEQ ID No: 12; for the manufacture of a pharmaceutical composition or a functional ingredient to stimulate the formation of the intestinal mucosa.
2. Uso según la reivindicación 1, donde la composición farmacéutica o el ingrediente funcional es para favorecer la protección gastrointestinal . 2. Use according to claim 1, wherein the pharmaceutical composition or functional ingredient is to promote gastrointestinal protection.
3. Uso según una de las reivindicaciones 1 ó 2, donde dicho péptido aislado está presente en un hidrolizado de proteínas de suero, posee actividad estimulante de la secreción de mucinas y comprende una estructura seleccionada del grupo que consiste en: SEQ ID No: 1, SEQ ID No: 2 y una combinación de ambas. 3. Use according to one of claims 1 or 2, wherein said isolated peptide is present in a whey protein hydrolyzate, has mucin secretion stimulating activity and comprises a structure selected from the group consisting of: SEQ ID No: 1 , SEQ ID No: 2 and a combination of both.
4. Uso según una de las reivindicaciones 1 ó 2, donde dicho péptido aislado está presente en un hidrolizado de caseína de la leche, posee actividad estimulante de la secreción de mucinas y comprende una estructura seleccionada del grupo que consiste en: SEQ ID No: 3, SEQ ID No: 4, SEQ ID No: 5, SEQ ID No: 6, SEQ ID No: 7, SEQ ID No: 8, SEQ ID No: 9, SEQ ID No: 10, SEQ ID No: 11, SEQ ID No: 12 y cualquier combinación de las mismas . 4. Use according to one of claims 1 or 2, wherein said isolated peptide is present in a milk casein hydrolyzate, It has a mucin secretion stimulating activity and comprises a structure selected from the group consisting of: SEQ ID No: 3, SEQ ID No: 4, SEQ ID No: 5, SEQ ID No: 6, SEQ ID No: 7, SEQ ID No: 8, SEQ ID No: 9, SEQ ID No: 10, SEQ ID No: 11, SEQ ID No: 12 and any combination thereof.
5. Uso según una de las reivindicaciones 1 ó 2, donde dicho péptido aislado está presente en un hidrolizado de la fracción de /3-caseina de la leche, posee actividad estimulante de la secreción de mucinas y comprende una estructura seleccionada del grupo que consiste en: SEQ ID No: 3, SEQ ID No: 4, SEQ ID No: 5 y una combinación de las mismas . 5. Use according to one of claims 1 or 2, wherein said isolated peptide is present in a hydrolyzate of the milk / 3-casein fraction, has mucin secretion stimulating activity and comprises a structure selected from the group consisting of in: SEQ ID No: 3, SEQ ID No: 4, SEQ ID No: 5 and a combination thereof.
6. Uso según una de las reivindicaciones 1 ó 2, donde dicho péptido aislado está presente en un hidrolizado de la fracción de sl- caseina de la leche, posee actividad estimulante de la secreción de mucinas y comprende una estructura seleccionada del grupo que consiste en: SEQ ID No: 6, SEQ ID No: 7, SEQ ID No: 8, SEQ ID No: 9, SEQ ID No: 10, SEQ ID No: 11, SEQ ID No: 12 y una combinación de las mismas . 6. Use according to one of claims 1 or 2, wherein said isolated peptide is present in a hydrolyzate of the sl -casein fraction of milk, has mucin secretion stimulating activity and comprises a structure selected from the group consisting of : SEQ ID No: 6, SEQ ID No: 7, SEQ ID No: 8, SEQ ID No: 9, SEQ ID No: 10, SEQ ID No: 11, SEQ ID No: 12 and a combination thereof.
7. Uso según la reivindicación 6 donde, cuando el péptido comprende una estructura seleccionada del grupo que consiste en: SEQ ID No:7. Use according to claim 6 wherein, when the peptide comprises a structure selected from the group consisting of: SEQ ID No:
8. SEQ ID No: 9, SEQ ID No: 10, SEQ ID No: 11, SEQ ID No: 12 y una combinación de las mismas; dicho péptido, además de actividad estimulante de la secreción de mucinas, presenta actividad opioide . 8. SEQ ID No: 9, SEQ ID No: 10, SEQ ID No: 11, SEQ ID No: 12 and a combination thereof; said peptide, in addition to mucin secretion stimulating activity, has opioid activity.
8. Péptido bioactivo aislado que: a) posee actividad estimulante de secreción de mucinas y/o actividad opioide; b) está presente en un hidrolizado enzimático de al menos una proteina de la leche seleccionado entre un hidrolizado de proteínas de suero o un hidrolizado de caseína de la leche; c) su estructura comprende una secuencia con al menos dos aminoácidos aromáticos, donde el primer aminoácido aromático es una tirosina situada en primera o segunda posición respecto al extremo N-terminal de dicha secuencia y el segundo aminoácido aromático es una fenilalanina o una tirosina situada en tercera o cuarta posición con respecto a la primera tirosina; y donde dicha estructura se selecciona entre SEQ ID No: 1, SEQ ID No: 2, SEQ ID No: 3, SEQ ID No: 4, SEQ ID No: 5, SEQ ID No: 6, SEQ ID No: 7, SEQ ID No: 8, SEQ ID No: 9, SEQ ID No: 10, SEQ ID No: 11 y SEQ ID No: 12; para estimular la formación de la mucosa intestinal y/o favorecer la protección gastrointestinal. 8. Isolated bioactive peptide that: a) has mucin secretion stimulating activity and / or opioid activity; b) is present in an enzymatic hydrolyzate of at least one milk protein selected from a whey protein hydrolyzate or a milk casein hydrolyzate; c) its structure comprises a sequence with at least two aromatic amino acids, where the first aromatic amino acid is a tyrosine located in first or second position with respect to the end N-terminal of said sequence and the second aromatic amino acid is a phenylalanine or a tyrosine located in third or fourth position with respect to the first tyrosine; and where said structure is selected from SEQ ID No: 1, SEQ ID No: 2, SEQ ID No: 3, SEQ ID No: 4, SEQ ID No: 6, SEQ ID No: 7, SEQ ID No: 8, SEQ ID No: 9, SEQ ID No: 10, SEQ ID No: 11 and SEQ ID No: 12; to stimulate the formation of the intestinal mucosa and / or promote gastrointestinal protection.
9. Un método de tratamiento para estimular la formación de la mucosa intestinal y/o favorecer la protección gastrointestinal caracterizado porque comprende administrar a un individuo una cantidad terapéuticamente eficaz de un péptido bioactivo aislado que : a) posee actividad estimulante de secreción de mucinas y/o actividad opioide; b) está presente en un hidrolizado enzimático de al menos una proteina de la leche seleccionado entre un hidrolizado de proteínas de suero o un hidrolizado de caseína de la leche; c) su estructura comprende una secuencia con al menos dos aminoácidos aromáticos, donde el primer aminoácido aromático es una tirosina situada en primera o segunda posición respecto al extremo N-terminal de dicha secuencia y el segundo aminoácido aromático es una fenilalanina o una tirosina situada en tercera o cuarta posición con respecto a la primera tirosina; y donde dicha estructura se selecciona entre SEQ ID No: 1, SEQ ID No: 2, SEQ ID No: 3, SEQ ID No: 4, SEQ ID No: 5, SEQ ID No: 6, SEQ ID No: 7, SEQ ID No: 8, SEQ ID No: 9, SEQ ID No: 10, SEQ ID No: 11 y SEQ ID No: 9. A method of treatment to stimulate the formation of the intestinal mucosa and / or promote gastrointestinal protection characterized in that it comprises administering to an individual a therapeutically effective amount of an isolated bioactive peptide that: a) possesses mucin secretion stimulating activity and / or opioid activity; b) is present in an enzymatic hydrolyzate of at least one milk protein selected from a whey protein hydrolyzate or a milk casein hydrolyzate; c) its structure comprises a sequence with at least two aromatic amino acids, where the first aromatic amino acid is a tyrosine located in the first or second position with respect to the N-terminal end of said sequence and the second aromatic amino acid is a phenylalanine or a tyrosine located in third or fourth position with respect to the first tyrosine; and where said structure is selected from SEQ ID No: 1, SEQ ID No: 2, SEQ ID No: 3, SEQ ID No: 4, SEQ ID No: 6, SEQ ID No: 7, SEQ ID No: 8, SEQ ID No: 9, SEQ ID No: 10, SEQ ID No: 11 and SEQ ID No:
PCT/ES2013/070550 2012-07-30 2013-07-26 Use of milk protein hydrolysates as gastrointestinal protectors WO2014020209A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
ES201231227 2012-07-30
ESP201231227 2012-07-30

Publications (1)

Publication Number Publication Date
WO2014020209A1 true WO2014020209A1 (en) 2014-02-06

Family

ID=50027312

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/ES2013/070550 WO2014020209A1 (en) 2012-07-30 2013-07-26 Use of milk protein hydrolysates as gastrointestinal protectors

Country Status (1)

Country Link
WO (1) WO2014020209A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3595698A4 (en) * 2017-03-16 2020-12-23 Microsintesis Inc. Propiotic molecules for reducing pathogen virulence
GB2608587A (en) * 2021-06-25 2023-01-11 Mjn Us Holdings Llc Use of extensively hydrolysed protein

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1986002079A1 (en) * 1984-09-28 1986-04-10 Victor Brantl Pharmacologically active peptide
JPS61271300A (en) * 1985-05-28 1986-12-01 Ajinomoto Co Inc Novel peptide
CN101525376A (en) * 2009-04-17 2009-09-09 天津商业大学 Opioid peptide for source of Alphas1-casein and preparation method thereof
JP2010132574A (en) * 2008-12-02 2010-06-17 Meiji Milk Prod Co Ltd Antioxidant
WO2010125192A1 (en) * 2009-04-30 2010-11-04 Consejo Superior De Investigaciones Cientificas Use of a casein-derived peptide and compositions thereof as antihypertensive

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1986002079A1 (en) * 1984-09-28 1986-04-10 Victor Brantl Pharmacologically active peptide
JPS61271300A (en) * 1985-05-28 1986-12-01 Ajinomoto Co Inc Novel peptide
JP2010132574A (en) * 2008-12-02 2010-06-17 Meiji Milk Prod Co Ltd Antioxidant
CN101525376A (en) * 2009-04-17 2009-09-09 天津商业大学 Opioid peptide for source of Alphas1-casein and preparation method thereof
WO2010125192A1 (en) * 2009-04-30 2010-11-04 Consejo Superior De Investigaciones Cientificas Use of a casein-derived peptide and compositions thereof as antihypertensive

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
MARTINEZ-MAQUEDA, D. ET AL.: "Food derived peptides stimulate mucin secretion and gene expression in intestinal cells", JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY, vol. 60, no. 35, 5 September 2012 (2012-09-05), pages 8600 - 8605 *
RECIO, I. ET AL.: "Food derived peptides stimulate mucin secretion and gene expression in intestinal cells", ANNALS OF NUTRITION AND METABOLISM., vol. 58, no. SUPPL., 2011, pages 117 *
ROUFIK, S. ET AL.: "In vitro digestibility of bioactive peptides derived from bovine (beta-lactoglobulin", INTERNATIONAL DAIRY JOURNAL., vol. 16, no. 4, 1 April 2006 (2006-04-01), pages 294 - 302 *
TROMPETTE, A. ET AL.: "Milk bioactive peptides and (beta-casomorphines induce mucus release in rat jejunum", THE JOURNAL OF NUTRITION, vol. 133, no. 11, 1 November 2003 (2003-11-01), pages 3499 - 3503 *

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3595698A4 (en) * 2017-03-16 2020-12-23 Microsintesis Inc. Propiotic molecules for reducing pathogen virulence
US11912788B2 (en) 2017-03-16 2024-02-27 Microsintesis Inc. Probiotic molecules for reducing pathogen virulence
GB2608587A (en) * 2021-06-25 2023-01-11 Mjn Us Holdings Llc Use of extensively hydrolysed protein

Similar Documents

Publication Publication Date Title
ES2343499B1 (en) MICROORGANISMS TO IMPROVE THE STATE OF HEALTH OF INDIVIDUALS WITH DISORDERS RELATED TO THE INTAKE OF GLUTEN.
ES2871117T3 (en) Procedures to modulate the gut microbiota
ES2808939T3 (en) Pharmaceutical and food compositions based on Hafnia alvei to induce satiety and prolong satiety
ES2402014B1 (en) PEPTIDE SECRETED BY LACTOBACILLUS PLANTARUM WITH IMMUNOMODULATORY FUNCTION
ES2915100T3 (en) Microbial lysozyme for use in the treatment of irritable bowel syndrome or inflammatory bowel disease
ES2887086T3 (en) Procedure to produce egg yolk with high af-16 content
BR112020014676A2 (en) COMBINATION THERAPY FOR CANCER TREATMENT OR PREVENTION
Ceelen et al. Modulation of connexin signaling by bacterial pathogens and their toxins
US20180153959A1 (en) Pharmaceutical and food compositions for inducing satiation and prolonging satiety in subjects in need thereof
WO2014020209A1 (en) Use of milk protein hydrolysates as gastrointestinal protectors
ES2885830T3 (en) Compositions for the inhibition of Giardia lamblia
ES2321602T3 (en) COMPOSITION TO PROMOTE THE PROLIFERATION OF LACTOBACILLUS CASEI SUBSP.CASEI.
ES2329316B1 (en) OBTAINING AND ANTIHIPERTENSIVE PROPERTIES OF PEPTIDES DERIVED FROM EGG CLEAR PROTEINS.
EP3439684B1 (en) Hafnia alvei based pharmaceutical and food compositions for inducing satiation and prolonging satiety
US20200131495A1 (en) Proteins derived from clpb and uses thereof
Tanabe et al. Identification of a peptide in enzymatic hydrolyzate of cheese that inhibits ovalbumin permeation in Caco-2 cells
US11369645B2 (en) Hafnia alvei impact on regulation of appetite and metabolic syndrome aspects
KR20190139910A (en) Brassicaea extract and its uses
US20090068174A1 (en) Therapeutic alkaline protease compositions and use in facilitating the transport of agents across the gastrointestinal mucosal lining
EP4295843A1 (en) Compositions comprising amino acids for use in the prevention and/or treatment of intestinal diseases
Boscaini Novel insights into the mechanisms underlying the anti-obesity
Boscaini Novel insights into the mechanisms underlying the anti-obesity effects of whey protein
KR101648330B1 (en) Composition containing collagen hydrolyzate having enhanced proliferation of osteoblast
JP2020015757A (en) Pharmaceutical composition and food composition for inducing satiation and extending satiety in object in need thereof
ES2354784A1 (en) Enzymatic procedure for the production of bioactive peptides. (Machine-translation by Google Translate, not legally binding)

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 13825783

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 13825783

Country of ref document: EP

Kind code of ref document: A1