GB2344736A - Animal feed supplement comprising a phospholipid composition - Google Patents

Animal feed supplement comprising a phospholipid composition Download PDF

Info

Publication number
GB2344736A
GB2344736A GB9911191A GB9911191A GB2344736A GB 2344736 A GB2344736 A GB 2344736A GB 9911191 A GB9911191 A GB 9911191A GB 9911191 A GB9911191 A GB 9911191A GB 2344736 A GB2344736 A GB 2344736A
Authority
GB
United Kingdom
Prior art keywords
composition
lecithin
parts
phospholipid composition
phospholipid
Prior art date
Legal status (The legal status 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 status listed.)
Granted
Application number
GB9911191A
Other versions
GB2344736B (en
GB9911191D0 (en
Inventor
David John Garnett
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Lovesgrove Research Ltd
Original Assignee
Lovesgrove Research Ltd
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 Lovesgrove Research Ltd filed Critical Lovesgrove Research Ltd
Publication of GB9911191D0 publication Critical patent/GB9911191D0/en
Priority to EP99954205A priority Critical patent/EP1139783A1/en
Priority to CA002355301A priority patent/CA2355301A1/en
Priority to AU10623/00A priority patent/AU1062300A/en
Priority to MXPA01006207A priority patent/MXPA01006207A/en
Priority to PCT/GB1999/003743 priority patent/WO2000036929A1/en
Publication of GB2344736A publication Critical patent/GB2344736A/en
Priority to US09/868,271 priority patent/US6509055B1/en
Application granted granted Critical
Publication of GB2344736B publication Critical patent/GB2344736B/en
Anticipated expiration legal-status Critical
Expired - Lifetime legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23JPROTEIN COMPOSITIONS FOR FOODSTUFFS; WORKING-UP PROTEINS FOR FOODSTUFFS; PHOSPHATIDE COMPOSITIONS FOR FOODSTUFFS
    • A23J7/00Phosphatide compositions for foodstuffs, e.g. lecithin
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23KFODDER
    • A23K20/00Accessory food factors for animal feeding-stuffs
    • A23K20/10Organic substances
    • A23K20/158Fatty acids; Fats; Products containing oils or fats
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23KFODDER
    • A23K50/00Feeding-stuffs specially adapted for particular animals
    • A23K50/30Feeding-stuffs specially adapted for particular animals for swines

Landscapes

  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Polymers & Plastics (AREA)
  • Engineering & Computer Science (AREA)
  • Food Science & Technology (AREA)
  • Animal Husbandry (AREA)
  • Zoology (AREA)
  • Biochemistry (AREA)
  • Birds (AREA)
  • Fodder In General (AREA)

Abstract

A phospholipid composition comprises components in the ratio 18Ò1 parts phosphatidylcholine (PC), 14Ò1 parts phosphatidylethanolamine (PE), 4Ò1 parts lysophosphatidylcholine (LPC), 1Ò1 parts lysophosphatidylethanolamine (LPE) and 2Ò1 parts lysophosphatidylinositol (LPI). The composition may be used as a supplement for animal feed, such as standard creep diet or '486' type feed, and the composition may be provided with a carrier, such as silica talc, which may be included in an amount of 10-40% by weight. Alternatively, the composition may be provided in solution. The composition may be produced from crude soya lecithin (e.g. by enzyme action) or by mixing the synthetically derived pure components. The composition may comprise additional phospholipids such as phosphatidylinositol (PI), diphosphatidyl glycerol (DPG), phosphatidic acid (PA), lysophosphatidic acid (LPA) and/or acylphosphatidylethanolamine (APE). A preferred composition comprises the components (by Mol%), 25.63% PC, 19.27% PI, 7.52%LPC, 19.32% PE, 2.93% APE, 1.14% DPG, 3.95% LPE, 13.28% PA, 4.30%LPI and 2.66% LPA.

Description

Title: Specific Phospholipid Compositions for Inclusion in Animal Feeds.
DESCRIPTION This invention relates to specific phospholipid compositions for providing an improved animal feed, particularly compositions prepared from soya bean extracts.
Particular soya bean lecithins may be incorporated into animal feeds with beneficial effects on animal performance, especially growth rates. Soya bean lecithin is routinely used as an additive for animal diets, typically in amounts ranging from 0.1 to 3%, and is a permitted emulsifier for this use under EC Feed Regulations. Benefits provided by the lecithin incorporated in the animal feed include increased digestibility of fat and transport of lipids out of the liver. Lecithin may also provide a good source of choline. It is frequently included in compound animal feeds, milk replacers and aquatic diets.
Crude lecithin contains a variety of phospholipids and glycolipids as well as small amounts of phosphatic acid and neutral lipids. The most abundant phospholipids are phosphatidylcholine (PC), phosphatidylethanolamine (PE) and phosphatidylinositol (PI).
Phospholipases and lipases produced naturally by certain strains of bacteria, fungi and other organisms, or enzymes extracted from animal pancreas, can be used to achieve a conversion of the phospholipids to their deacylated derivatives known as lysolecithins, such as lysophosphatidylcholine (LPC), lysophosphatidylethanolamine (LPE) and lysophosphatidylinositol (LPI). These lysolecithins have a beneficial impact in animal growth when compared with lecithins (Schwarzer K and Adams C, Lipids 98 [1996] and UK Patent No. 2267033).
It has been known for some time that different lipid mixtures can have important and widely different effects on biological membranes. For example, the active lipid mixture AL721 is synthetically prepared from neutral glycerides, phosphatidylcholine and phosphatidylethanolamine in the ratio of 7: 2: 1 and has been used to inhibit attachment of viruses to cell surface receptor sites (Effects of Novel Compound (AL721) on HTLV-II Infectivity in vitro. Sarin P; Gallo R; Scheer D; Crews F; and Lippa A. New England Journal of Medicine 313 [1985] No. 20 1289-1290). This precise ratio of lipids appears to permeabilise the membrane and is associated with a decrease in membrane bound cholesterol (A Special Lipid Mixture for Membrane Fluidisation; Lyte M and Shinitzky M.
Biochem Biophys Acta 812 [1985] 133-138).
It is an obj ect of the present invention to provide new compositions for producing an improved animal feed which have beneficial effects on animal performance, especially growth rates.
A further object of the present invention is to provide new compositions which may be produced from crude lecithin, thereby providing compositions which are cheaper to produce and may be manufactured in bulk.
Accordingly, a first aspect of the present invention provides a novel phospholipid composition comprising the following components in the ratios given: Phosphatidylcholine 18 1 Phosphatidylethanolamine 1 Lysophosphatidylcholine 4 1 Lysophosphatidylethanolamine I + I Lysophosphatidylinositol 2 1 A second aspect of the present invention provides an animal feed supplement comprising a phospholipid composition having the following components in the ratios given: Phosphatidylcholine 18 1 Phosphatidylethanolamine 14 1 Lysophosphatidylcholine 4 1 Lysophosphatidylethanolamine 1 1 Lysophosphatidylinositol 2 1 Preferably, the ratio of components in the composition is as :- Phosphatidylcholine 18 Phosphatidylethanolamine 14 Lysophosphatidylcholine 4 Lysophosphatidylethanolamine 1 Lysophosphatidylinositol 2 The composition may include additional phospholipids, such as phosphatidylinositol (PI), acylphosphatidylethanolamine (APE), inositol (PI), diphosphatidyl glycerol (DPG), phosphatidic acid (PA) and lysophosphatic acid (LPA).
A preferred composition of the present invention has the following empirical formula: Phospholipid Weight% PC 9.69 PI 7.90 LPC 1.90 PE 6.88 APE 1.42 DPG 0.38 LPE 0.91 PG 0. 00 PA 4.47 LPI 1.20 PS 0.00 LPA 0.56 SPH 0.00 Others 0.00 The composition is preferably provided in an animal feed as a concentrate with a carrier, such as, silica talc, preferably being included in an amount of 10-40% by weight, more preferably 25%. Alternatively, the composition may be provided in solution.
The present invention will now be further illustrated by means of the following Examples in which Example 1 relates to an investigation into the uptake of the dye 3- [4, 5- Dimethylthiazol-2-yl]-2,5-diphenyltetrazoliumbromide (MTT) by a monolayer of Vero (monkey kidney) cells using cells treated with a composition of the present invention and cells treated with standard lecithin, Example 2 relates to an in vitro experiment to investigate the permeability of cell plasma membranes using cells which had been treated with a composition of the present invention and cells which had been treated with standard lecithin, by means of treating the respective cells with a media containing radioactive fatty acids and Example 3 relates to an in vivo experiment to determine the increase in daily liveweight gain in pigs fed with a feed supplement containing a composition of the present invention or standard lecithin, and with reference to the accompanying drawings in which: Figure 1 illustrates respectively the Spectra for Lecithin and for modified lecithin according to the present invention, using phosphorous nuclear magnetic resonance spectroscopy ; and Figure 2 is a graph illustrating the in vitro effects of lecithin and modified lecithin according to the present invention.
The present invention describes a narrow range of ratios and the empirical formula of the most biologically potent lecithins and their use in animal feed formulations.
Example 1 The rate of uptake of the dye 3- [4, 5-Dimethylthiazol-2-yl]-2, 5 diphenyltetrazoliumbromide (MTT) by a monolayer of Vero (monkey kidney) cells was measured. MTT is a tetrazolium salt which yields a yellow solution in water. When MTT is taken up by a living cell, dehydrogenase enzymes cleave the tetrazolium ring forming a water insoluble, purple, forrnazan within the cell. This formazan is solubilized in acidic isopropanol and its absorbance measured spectrophotometrically. The absorbance figure provides an indication of the rate of cellular absorbance, i. e., to determine whether the altered phospholipid ratio of the present invention does increase cellular absorption rates, indicated by more MTT being absorbe and metabolised.
Method Fifteen wells of 6-Well Linbro plates were seeded with Vero cells from a three day culture and incubated overnight under standard conditions in lml Dulbeccos Modified Eagles Media (DMEM) + 10% foetal calf serum to provide standard monolayers of cells.
Two ratios of phospholipids, shown in Table 2 below, named Lecithin ratio and Modified lecithin ratio were suspended in Hanks Balanced Salt Solution (HBSS) + 4% ethanol to yield a total phospholipid content of 3.9 mg ml' The Lecithin ratio is that found in unmodified soybean lecithin, the Modified lecithin ratio is that according to the present invention which may be obtained by various techniques such as mixing the synthetically derived pure components together or by enzymes acting on the crude lecithin.
Table 2.
Phospholipid Lecithin Ratio Modified I Species Ratio Pic 13-18 PE 11 14 ; LPC04 LPE 0 1 LPI 1 2 Triplicate wells of Vero cell monolayers were then treated with doses of the two ratios, shown in Table 2, for 90 minutes along with a negative control. This yielded a concentration per well of 0.78 or 0.39 mg mol-'total phospholipid. 1001 of a 5 mg ml' solution of MTT was then added to each well and incubated for 30 minutes. The cell monolayers were then washed twice with HBSS and any media removed from the wells. lml of acidified isopropanol was then added to each well and the plates swirled until all purple colouration was dissolved. This solution was then removed and measured spectrophotometrically at 570nm, with a background reading at 660nm.
Table 3.
Sample Name Ethanol (pi) HBSS (pl) Lecithin Ratio Modified lecithin Ratio (PI) Control 10 240 0 0 Lecithin 1 0 125 125 0 Lecithin 2 0 0 250 0 Modified 0 125 0 125 Lecithin1 Modified 0 0 0 250 Lecithin 2 The final absorbance readings were obtained by subtracting Abs660nm from Abs570nm and calculating the mean absorbance of the triplicate wells. The calculated figures are shown in Table 4 below.
Table 4.
Sample | Mean Absorbance Lecithin 1 0. 14933 Lecithin 2 0. 16266 Modified lecithin 1 0. 155 Modified lectihin 2 0. 1 Comparing the 250ut doses of Lecithin and Modified lecithin ratios of phospholipids there is an increase of 3. 7% in the level of metabolised MTT in the cells treated with the Modified lecithin ratio over that seen in the Lecithin ratio treated cells. A similar increase of 3.5% in the Modified lecithin treated cells is seen with the 125 l doses.
The negative control value shows the cells are not adversely effected by the treatment process. A far greater effect could be seen if the dispersion of the-phospholipids in the media could be improved, as quantities of the phospholipids were observed on the surface of the media during the assay. This suggests that the quantity of phospholipid acting on the cells in this assay is far below the stated maximum of 0.78 mg ml-'.
Thus, the ratio of different phospholipid species in Modified lecithin according to the present invention has the effect of enhancing cellular absorption over that seen in Lecithin.
Example 2 A simple in vitro test was performed to examine the biological properties of the enzyme treated product compared to the substrate lecithin. The enzyme treated product has a similar composition to the synthetic composition used in Example 1. The permeability of the cell plasma membrane to fatty acids was measured by treating cells with lecithin mixtures in media that contained radioactive fatty acids. C34 labeled stearic and palmitic acids were both used.
Method BHK 21 clone cells were cultured in the normal way using G-MEM media supplemented with 10% FBS, 10ml/L of 200mM L-Glutamine, 5% TSB and 10 ml/L anti biotic/antimycotic solution. All reagents were supplied by Sigma Chemicals, Dorset. The cell line was sourced from the European Collection of Animal Cell Cultures at the Centre for Applied Microbiology and Research at Porton Down.
The cells were cultured in 25cm2 flasks and all experiments were performed in triplicate. Cells in the wells were exposed to luCi ofC steahc and palmitic acid prepared in Hanks'Balanced Salt Solution from a stock 75uCi stock solution. They were co-exposed to varying concentrations of either lecithin or enzyme treated lecithin. These were prepared using an Ultra-Turrex blender to make up 1% aqueous emulsions. The cultures were left for 1 hour and then the media was removed by aspiration. The adhered cells were then washed twice in Hanks'solution and were then trypsinised using trypsin- EDTA solution. They were then pelleted by centrifuge at 1000 rpm for 5 minutes in preweighted Eppendorf tubes. The cell pellet was then lysed using distilled water and ultrasonic disrupter and left overnight. The Eppendorfs were then re-spun and 100p1 aliquots of the water removed for analysis in a scintillation counter. Sigmafluor was used as the scintillation liquid. Results were converted into DPM per mg cell solids after the Eppendorfs were re-weighed when dry. Figure 2 of the accompanying drawings shows the mean of three results for each experiment, revealing that the enzyme modified lecithin has a similar improvement in activity to the'synthetic'mixture used in Example 1.
Example 3 Six groups of pigs were grown in open pens containing 35 pigs in each. Standard creep diet and'486'type feed (BOCM-Pauls Ltd., England) was used which contained either lecithin or the modified lecithin of the present invention included at a level of 250g per ton of animal feed. The two types of lecithin were introduced to the feed during production as 25% concentrate in silica talc carrier. Summary results are shown in Table 5 below.
Table 5
Overall Total Overall Totai Lecithin-IModified Lecithin Pig Days 4879 I 5055 Weight Gain (Kg) 1657 1821 Total Feed (Kg) 2725.4 2878.5 DLG (g) 339. 6 360. i FCR 6451. 581 These results translate to an increase in daily liveweight gain in the group fed with the modified lecithin of+6. 1%.
The modified lecithin of the present invention has unusual biological properties not found in the substrate material or individual components. The specific compositions as defined herein are recommended for the improvement of growth rates of livestock.
Example 1 used the claimed composition formed by adding together synthetically derived pure components whereas Examples 2 and 3 used lecithins which had been modified by enzymes available in the art to produce the claimed composition. The precise empirical formula of the preferred composition of the present invention is given below in Table 6.
Table 6
Phospholipid Integral Molecular Mol% Weight % Weight PC 83.95 770 25.63 9. 69 PI 63. 14 835 19. 27 7 90 LPC24. 625157. 521. 90 PE 63. 29 725 19. 32 6. 88 APE 9. 59 990 2. 93 1. 42 DPG 3. 73 682. 5 1. 14 0. 38 LPE 12. 93 470 3 95 1 0. 91 PG 0 758 0. 00 0. 00 PA 43. 5 685 13. 28 4. 47 LPI 14. 1 570 4. 30 1. 20 PS 0 772 0. 00 0. 00 LPA 8. 73 430 2. 66 0. 56 SPH 0 770 0. 00 0. 00 Others 0 770 0. 00 0. 00 Standard 100 326.29 Totals 100.00 35. 31 Sample weight (mg) 235.2 Standard weight (mg) 11. 5 The increase in quantity of lysophosphatidylcholine, lysophosphatidylethanolamine and lysophosphatidylinositol in specified ratios improves the value of the lecithin as a feed supplement by increasing the rate of cellular absorption in the gut.
It is postulated that activity in the membrane results in changes to the fluidity of the membrane that facilitates passive flux of nutrients into the cell. The prior art composition AL721 was purported to effect the fluidity of cell membranes by modifying cholesterol level in the membrane and thereby the dynamics of the membrane (Intervention in Aging the Development and Application of Active Lipid. Shinitzky, M; Lyte. M; Heron. D and Samuel. D Basic Research & Pre-clinical Screening [1983] NY 175-186). Although this may be one mode of action of the modified lecithin mixture of the present invention it is thought that a more important mode of action is the substitution of the mixture into the membrane thereby directly increasing the motility of the individual phospholipids within the matrix. This may be due to the smaller space occupied by the tail groups of the monoacyl moieties and the concurrent disturbance of the bonds which serve normally to reinforce the matrix.
The digestive tracks of mammals and fish are composed of surfaces comprising tissue perfused with blood for the purposes of absorption of nutrients and these tissues are made up of individual cells which have plasma membranes which may be affected by these modified lecithins. Thus, when diets containing small amounts of the modified lecithin are fed to animals in experiments the feed is better absorbed into the bloodstream from the digestive system and the field trial results support this mechanistic hypothesis.

Claims (6)

  1. CLAIMS 1. A phospholipid composition comprising the following components in the ratios given: Phosphatidylcholine 18 1 Phosphatidylethanolamine 14 1 Lysophosphatidylcholine i 1 Lysophosphatidylethanolamine 1 < Lysophosphatidylinositol +
  2. 2. A phospholipid composition as claimed in claim 1, wherein the ratio of components is: Phosphatidylcholine 18 Phosphatidylethanolamine 4 Lysophosphatidylcholine 4 Lysophosphatidylethanolamine 1 Lysophosphatidylinositol 2
  3. 3. A phospholipid composition as claimed in claim 1 or claim 2 further comprising additional phospholipids selected from the group consisting of phosphatidyl inositol, diphosphatidyl glycerol, phosphatidic acid, lysophosphatidic acid and acylphosphatidylethanolamine.
  4. 4. A phospholipid composition as claimed in claim 3 having the empirical formula given below: Phospholipid Weight% PC 9.69 PI 7.90 LPC 1.90 PE 6.88 APE 1.42 DPG 0.38 LPE 0.91 PG 0. 00 PA 4.47 LPI 1.20 PS 0.00 LPA 0.56 SPH 0.00 Others 0.00
  5. 5. Use of a phospholipid composition as claimed in any one of the preceding claims as a supplement for animal feeds.
  6. 6. An animal feed supplement comprising a phospholipid composition as claimed in any one of claims 1 to 4 and a carrier therefor.
GB9911191A 1998-12-18 1999-05-14 Specific phospholipid compositions for inclusion in animal feeds Expired - Lifetime GB2344736B (en)

Priority Applications (6)

Application Number Priority Date Filing Date Title
PCT/GB1999/003743 WO2000036929A1 (en) 1998-12-18 1999-11-10 An animal feed supplement comprising specific phospholipid compositions
CA002355301A CA2355301A1 (en) 1998-12-18 1999-11-10 An animal feed supplement comprising specific phospholipid compositions
AU10623/00A AU1062300A (en) 1998-12-18 1999-11-10 An animal feed supplement comprising specific phospholipid compositions
MXPA01006207A MXPA01006207A (en) 1998-12-18 1999-11-10 An animal feed supplement comprising specific phospholipid compositions.
EP99954205A EP1139783A1 (en) 1998-12-18 1999-11-10 An animal feed supplement comprising specific phospholipid compositions
US09/868,271 US6509055B1 (en) 1998-12-18 2000-06-29 Animal feed supplement comprising specific phospholipid compositions

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
GBGB9828013.4A GB9828013D0 (en) 1998-12-18 1998-12-18 Specific phospholipid-lipid mixtures for inclusion in animal feed

Publications (3)

Publication Number Publication Date
GB9911191D0 GB9911191D0 (en) 1999-07-14
GB2344736A true GB2344736A (en) 2000-06-21
GB2344736B GB2344736B (en) 2002-11-06

Family

ID=10844573

Family Applications (2)

Application Number Title Priority Date Filing Date
GBGB9828013.4A Ceased GB9828013D0 (en) 1998-12-18 1998-12-18 Specific phospholipid-lipid mixtures for inclusion in animal feed
GB9911191A Expired - Lifetime GB2344736B (en) 1998-12-18 1999-05-14 Specific phospholipid compositions for inclusion in animal feeds

Family Applications Before (1)

Application Number Title Priority Date Filing Date
GBGB9828013.4A Ceased GB9828013D0 (en) 1998-12-18 1998-12-18 Specific phospholipid-lipid mixtures for inclusion in animal feed

Country Status (1)

Country Link
GB (2) GB9828013D0 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8603568B2 (en) 2010-01-15 2013-12-10 Kemin Industries, Inc. Hydrolyzed lecithin product to improve digestibility
CN108601373A (en) * 2016-03-02 2018-09-28 百思威中间体公司 Animal feed containing specific glycolipid

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4275089A (en) * 1977-05-12 1981-06-23 Kopas George A Purification of vegetable oil
EP0354442A1 (en) * 1988-08-09 1990-02-14 A. Nattermann & Cie. GmbH Phospholipid-containing composition, a process for its preparation and its use as an excipient for pharmaceutical substances
EP0372327A2 (en) * 1988-12-08 1990-06-13 N.V. Vandemoortele International Process for fractionating phosphatide mixtures
GB2267033A (en) * 1992-03-07 1993-11-24 David Garnett Animal feed containing a phospholipid component
WO1995004467A1 (en) * 1993-08-06 1995-02-16 A/S Biotec-Mackzymal Animal feeds comprising yeast glucan

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE3445949A1 (en) * 1984-12-17 1986-06-19 A. Nattermann & Cie GmbH, 5000 Köln METHOD FOR INSULATING A COMPONENT PHOSPHOLIPID-FREE PHOSPHATIDYLCHOLINE

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4275089A (en) * 1977-05-12 1981-06-23 Kopas George A Purification of vegetable oil
EP0354442A1 (en) * 1988-08-09 1990-02-14 A. Nattermann & Cie. GmbH Phospholipid-containing composition, a process for its preparation and its use as an excipient for pharmaceutical substances
EP0372327A2 (en) * 1988-12-08 1990-06-13 N.V. Vandemoortele International Process for fractionating phosphatide mixtures
GB2267033A (en) * 1992-03-07 1993-11-24 David Garnett Animal feed containing a phospholipid component
WO1995004467A1 (en) * 1993-08-06 1995-02-16 A/S Biotec-Mackzymal Animal feeds comprising yeast glucan

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8603568B2 (en) 2010-01-15 2013-12-10 Kemin Industries, Inc. Hydrolyzed lecithin product to improve digestibility
US9173419B2 (en) 2010-01-15 2015-11-03 Kemin Industries, Inc. Method of improving animal feeds using hydrolyzed lecithins
CN108601373A (en) * 2016-03-02 2018-09-28 百思威中间体公司 Animal feed containing specific glycolipid

Also Published As

Publication number Publication date
GB9828013D0 (en) 1999-02-10
GB2344736B (en) 2002-11-06
GB9911191D0 (en) 1999-07-14

Similar Documents

Publication Publication Date Title
McCaman et al. Intermediary Metabolism of Phospholipids in Brain Tissue: III. PHOSPHOCHOLINE-GLYCERIDE TRANSFERASE
Borchers Proteolytic activity of rumen fluid in vitro
CN1199694C (en) Application of composite fat used as stabilizing additive in medicine of digestive enzyme mixture
Kodama et al. Edifenphos, inhibitor of phosphatidylcholine biosynthesis in Pyricularia oryzae
US6017530A (en) Phospholipases in animal feed
Schulthess et al. Absorption of monoacylglycerols by small intestinal brush border membrane
US6509055B1 (en) Animal feed supplement comprising specific phospholipid compositions
Magee et al. Effect of chlorpromazine and azacyclonol on the labelling of phosphatides in brain slices
Żyla et al. In‐vitro and in‐vivo dephosphorylation of rapeseed meal by means of phytate‐degrading enzymes derived from Aspergillus niger
Sawada et al. Effect of lipid on protoheme ferro-lyase
Kiessling et al. Biochemical changes in rat tissues after prolonged alcohol consumption
Jett et al. Selective cytotoxicity of tumor cells induced by liposomes containing plant phosphatidylinositol
Franson et al. Phospholipase A activity of highly enriched preparations of cardiac sarcolemma from hamster and dog
White et al. Phosphatidic acid and phosphatidylinositol metabolism in Schizosaccharomyceas pombe
Hadley et al. Identification of N-(2-propenal) ethanolamine as a urinary metabolite of malondialdehyde
GB2344736A (en) Animal feed supplement comprising a phospholipid composition
Van der Steen et al. Lipid dependence of glycophorin-induced transbilayer movement of lysophosphatidylcholine in large unilamellar vesicles
Hamor et al. Quantitative composition of the fertilized ovum and constituent parts in the Atlantic salmon Salmo salar L.
Udenfriend et al. fi-glucuronidase (Boyland, Wallace & Williams
Dlomede et al. Modulation of ATPase activity by cholesterol and synthetic ether lipids in leukemic cells
US4849137A (en) Process for producing lysophospholipids containing substantially no lysophospholipids except LPC
Melnick et al. Effect of phthlate esters on energy coupling and succinate oxidation in rat liver mitochondria
Demisch et al. Increased concentrations of phosphatidylinositol (PI) and decreased esterification of arachidonic acid into phospholipids in platelets from patients with schizoaffective disorders or atypic phasic psychoses
Oshino et al. Mechanism of action of atpenin B on Raji cells
Hossack et al. Stability of the plasma membrane in Saccharomyces cerevisiae enriched with phosphatidylcholine or phosphatidylethanolamine

Legal Events

Date Code Title Description
732E Amendments to the register in respect of changes of name or changes affecting rights (sect. 32/1977)
PE20 Patent expired after termination of 20 years

Expiry date: 20190513