US20080044487A1 - Anti-inflammatory properties of marine lipid compositions - Google Patents

Anti-inflammatory properties of marine lipid compositions Download PDF

Info

Publication number
US20080044487A1
US20080044487A1 US11/800,229 US80022907A US2008044487A1 US 20080044487 A1 US20080044487 A1 US 20080044487A1 US 80022907 A US80022907 A US 80022907A US 2008044487 A1 US2008044487 A1 US 2008044487A1
Authority
US
United States
Prior art keywords
composition
omega
fatty acid
predicted
phospholipids
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.)
Abandoned
Application number
US11/800,229
Inventor
Inge Bruheim
Mikko Griinari
Sebastiano Banni
Per Christian Sæbo
Erik Fuglseth
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.)
Aker Biomarine AS
Original Assignee
Natural ASA
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 Natural ASA filed Critical Natural ASA
Priority to US11/800,229 priority Critical patent/US20080044487A1/en
Assigned to AKER BIOMARINE ASA reassignment AKER BIOMARINE ASA CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: NATURAL ASA
Assigned to AKER BIOMARINE ASA reassignment AKER BIOMARINE ASA CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: NATURAL ASA
Publication of US20080044487A1 publication Critical patent/US20080044487A1/en
Priority to US12/849,950 priority patent/US10117882B2/en
Priority to US16/180,564 priority patent/US10525068B2/en
Priority to US16/734,850 priority patent/US11400105B2/en
Priority to US17/874,677 priority patent/US20230047556A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/683Diesters of a phosphorus acid with two hydroxy compounds, e.g. phosphatidylinositols
    • A61K31/685Diesters of a phosphorus acid with two hydroxy compounds, e.g. phosphatidylinositols one of the hydroxy compounds having nitrogen atoms, e.g. phosphatidylserine, lecithin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/02Nutrients, e.g. vitamins, minerals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/06Phosphorus compounds without P—C bonds
    • C07F9/08Esters of oxyacids of phosphorus
    • C07F9/09Esters of phosphoric acids
    • C07F9/10Phosphatides, e.g. lecithin

Definitions

  • the present invention relates to novel marine lipid compositions comprising combinations of omega-3 fatty acid rich functional phospholipids and omega-3 fatty acid rich triglycerides.
  • food supplements, functional food, drugs and feed products comprising such compositions are provided along with methods of their use.
  • Marine lipids such as omega-3 rich triglycerides and omega-3 rich phospholipids can be isolated from a number of different natural sources such as fish, crustaceans, plankton, seals, whales as well as algae using extraction technologies. In addition, they can be prepared industrially using chemical or bio-catalytical methods such as enzyme catalyzed transesterification of crude soy lecithin with fish oil fatty acids [1].
  • omega-3 fatty acids are well known and the use as an anti-inflammatory agent has been described both for triglycerides and phospholipids [2-3].
  • omega-3 fatty acids are famous for their anti-inflammatory properties, and it has been shown that omega-3 fatty acids alleviate the symptoms of a series of autoimmune, atherosclerotic and inflammatory diseases including inflammatory bowel diseases and rheumatoid arthritis [4-6]. Suppression of inflammation has been proposed as one of the strategies to slow down the progress of these diseases.
  • this invention discloses the effect on marine lipid compositions on the concentration of markers of inflammation such as TNF- ⁇ and other cytokines such as interleukin-1 ⁇ and interleukin 6.
  • AA arachidonic acid
  • prostaglandins prostaglandins, thromboxanes and leukotrienes
  • this invention discloses the reduction of AA level and the improvement in the EPA/AA ratio in different lipid pools in tissues such as in the phospholipids isolated from adipose tissue, heart, testicles, plasma, brain and liver.
  • the invention provides a composition comprising a triglyceride and a phospholipids in a ratio ranging from 1:10 to 10:1; said phospholipids having the following structure: wherein R1 is OH or a fatty acid, R2 is OH or a fatty acid, and R3 is a mixture of H, choline, ethanolamine, inositol and serine, said phospholipid having at least 1% of DHA/EPA, said phospholipids have a concentration of OH in the range of 25-50%.
  • the invention provides a marine lipid composition characterized by providing higher uptake of omega-3 fatty acids into plasma as compared to administration of purified triglycerides, phospholipids, or natural marine phospholipids.
  • the invention provides a composition characterized by efficiently improving the AA/EPA ratio in plasma phospholipids as compared to administration of purified triglycerides, phospholipids, or natural marine phospholipids.
  • the invention is a marine lipid composition characterized by efficiently increasing the concentration of omega-3 fatty acids in tissues as compared to administration of purified triglycerides, phospholipids, or natural marine phospholipids.
  • the composition is characterized by reducing the concentration of biomarkers of inflammation as compared to administration of purified triglycerides, phospholipids, or natural marine phospholipids.
  • the marine lipid composition is formulated into an animal feed, a food product, a food supplement and a drug.
  • the present invention provides a composition comprising phospholipids having the following structure: wherein R1 is OH or a fatty acid, R2 is OH or a fatty acid, and R3 is a mixture of H, choline, ethanolamine, inositol and serine, said phospholipid having at least 1% of DHA/EPA at positions R1 and/or R2 and from about 20-50% of OH at positions R1 and/or R2.
  • the composition is acylated in a range from about 55% to about 85%.
  • the omega-3 fatty acids are selected from the group consisting of EPA, DHA, DPA and ⁇ -linolenic acid (ALA).
  • the composition is substantially free of organic solvents and volatile organic compounds such as short chain fatty acids, short chain aldehydes and short chain ketones.
  • the composition has at least 5% of a combination of EPA and DHA esterified.
  • the composition has at least 10% of a combination of EPA and DHA esterified.
  • the composition has at least 20% of a combination of EPA and DHA esterified.
  • the composition has at least 30% of a combination of EPA and DHA esterified.
  • said composition contains from about 5%, 10%, 20% and 30% EPA/DHA attached to position 1 and/ or position 2.
  • the composition has a ratio of EPA/DHA ranging from 1:1 to 4:1. In some embodiments, the composition has a ratio of EPA/DHA ranging from 2:1 to 4:1. In some embodiments, the composition is acylated in a range from 60% to 80%. In some embodiments, the composition is acylated in a range from 50% to 75%.
  • the composition further comprises a lipid carrier in a ratio of from 1:10 to 10:1 to said phospholipids.
  • the lipid carrier and said phospholipids are in a ratio of from about 5:1 to 1:5.
  • the composition comprises from about 20% to about 90% of said phospholipid composition and from about 10% to about 50% of said lipid carrier.
  • the present invention is not limited to any particular lipid carrier.
  • the lipid carrier is selected from the group consisting of a triglyceride, a diglyceride, an ethyl ester, and a methyl ester and combinations thereof.
  • the composition provides higher uptake of omega-3 fatty acids into plasma as compared to natural marine phospholipids when administered to subjects. In some embodiments, the composition improves the AA/EPA ratio in plasma phospholipids when administered to subjects as compared to natural marine phospholipids. In some embodiments, the composition increases the concentration of omega-3 fatty acids in tissues when administered to subjects as compared to natural marine phospholipids. In some embodiments, the composition reduces the concentration of biomarkers of inflammation when administered to subjects as compared to natural marine phospholipids. In some embodiments, the present invention provides a food product comprising the foregoing compositions. In some embodiments, the present invention provides an animal feed comprising the foregoing compositions. In some embodiments, the present invention provides a food supplement comprising the foregoing compositions. In some embodiments, the present invention provides a pharmaceutical composition comprising the foregoing compositions.
  • the present invention provides methods of preparing a bioavailable omega-3 fatty acid composition comprising: a) providing a purified phospholipid composition comprising omega-3 fatty acid residues and a purified triglyceride composition comprising omega-3 fatty acid residues; b) combining said phospholipid composition and said triglyceride composition to form a bioavailable omega-3 fatty acid composition.
  • the bioavailable phospholipid composition is one of the compositions described above.
  • the methods further comprise the step of encapsulating said bioavailable omega-3 fatty acid composition.
  • the bioavailable omega-3 fatty acid composition has increased bioavailability as compared to purified triglycerides or phospholipids comprising omega-3 fatty acid residues.
  • the methods further comprise the step of packaging the bioavailable omega-3 fatty acid composition for use in functional foods.
  • the methods further comprise the step of assaying the bioavailable omega-3 fatty acid composition for bioavailability.
  • the methods further comprise administering the bioavailable omega-3 fatty acid composition to a patient.
  • the present invention provides a food product, animal feed, food supplement or pharmaceutical composition made by the foregoing process.
  • the present invention provides methods for reducing symptoms of cognitive dysfunction in a child comprising administering an effective amount of a marine phospholipid composition, wherein said symptoms are selected from the group consisting of ability to complete task, ability to stay on task, ability to follow instructions, ability to complete assignments, psychomotor function, long term memory, short term memory, ability to make a decision, ability to follow through on decision, ability to self-sustain attention, ability to engage in conversations, sensitivity to surroundings, ability to plan, ability to carry out plan, ability to listen, interruptions in social situations, temper tantrums, level/frequency of frustration, level/frequency restlessness, frequency/level fidgeting, ability to exhibit delayed gratification, aggressiveness, demanding behavior/frequency of demanding behavior, sleep patterns, restive sleep, interrupted sleep, awakening behavior, disruptive behavior, ability to exhibit control in social situations, ability to extrapolate information and ability to integrate information.
  • the child exhibits one or more symptoms of Attention Deficit Hyperactivity Disorder (ADHD), is suspected of having ADHD, or has been diagnosed with ADHD.
  • ADHD Attention Deficit Hyperactivity Disorder
  • autistic spectrum disorder is suspected of having autistic spectrum disorder, or has been diagnosed with autistic spectrum disorder.
  • the present invention provides methods of increasing cognitive performance in an aging mammal comprising administering an effective amount of a marine phospholipid composition.
  • the cognitive performance is selected from the group consisting of memory loss, forgetfulness, short-term memory loss, aphasia, disorientation, disinhibition, and behavioral changes.
  • the mammal is a human.
  • the mammal is a pet selected from the group consisting of cats and dogs.
  • the mammal has symptoms of age-associated memory impairment or decline.
  • the marine phospholipid composition comprises phospholipids having the following structure: wherein R1 is OH or a fatty acid, R2 is OH or a fatty acid, and R3 is a mixture of H, choline, ethanolamine, inositol and serine, said phospholipid having at least 1% of omega-3 fatty acid moieties at positions R1 and/or R2.
  • the phospholipid composition comprises from about 20-50% of OH at positions R1 and/or R2.
  • the phospholipid composition further comprises a lipid carrier.
  • the phospholipid composition is prepared from natural marine phospholipids isolated from a marine organism. In some embodiments, the phospholipid composition is enzymatically prepared by reacting lecithin with DHA and EPA in the presence of an enzyme. In some embodiments, the lecithin is soybean or egg lecithin. In some embodiments, the omega-3 fatty acid moieties are selected from the group of EPA and DHA and combination thereof. In some embodiments, the effective amount of said phospholipid composition comprises from about 300 to about 1000 mg omega-3 fatty acids. In some embodiments, the phospholipid composition is administered orally. In some embodiments, the phospholipid composition is provided in a gel capsule or pill.
  • the present invention provides methods of treating a subject by administration of a marine phospholipid composition comprising administering a marine phospholipid composition to said subject under conditions such that a desired condition is improved, wherein said conditions is selected from the group consisting of fertility, physical endurance, sports performance, muscle soreness, inflammation, auto-immune stimulation, metabolic syndrome, obesity and type II diabetes.
  • the subject is a human.
  • the subject is a companion animal.
  • the present invention is not limited to any particular marine phospholipid composition.
  • the marine phospholipid composition comprises phospholipids having the following structure: wherein R1 is OH or a fatty acid, R2 is OH or a fatty acid, and R3 is a mixture of H, choline, ethanolamine, inositol and serine, said phospholipid having at least 1% of omega-3 fatty acid moieties at positions R1 and/or R2.
  • the phospholipid composition comprises from about 20-50% of OH at positions R1 and/or R2.
  • the phospholipid composition is prepared from natural marine phospholipids isolated from a marine organism.
  • the composition further comprises a lipid carrier.
  • the phospholipid composition is enzymatically prepared by reacting lecithin with DHA and EPA in the presence of an enzyme.
  • the-lecithin is soybean or egg lecithin.
  • the omega-3 fatty acid moieties are selected from the group of EPA and DHA and combination thereof.
  • the effective amount of said phospholipid composition comprises from about 300 to about 1000 mg omega-3 fatty acids.
  • the phospholipid composition is administered orally.
  • the phospholipid composition is provided in a gel capsule or pill.
  • the human is a male.
  • the present invention provides methods for prophylactically treating a subject by administration of a marine phospholipid composition
  • a marine phospholipid composition comprising administering a marine phospholipid composition to a subject under conditions such that an undesirable condition is prevented, wherein said undesirable condition is selected from the group consisting of weight gain, infertility, obesity, metabolic syndrome, diabetes type II, mortality in subjects with a high risk of sudden cardiac death, and induction of sustained ventricular tachycardia.
  • the subject is at risk for developing a condition selected from the group consisting of weight gain, obesity, metabolic syndrome, diabetes type II, mortality in subjects with a high risk of sudden cardiac death, and induction of sustained ventricular tachycardia.
  • the subject is a human. In some embodiments, the subject is a companion animal.
  • the present invention is not limited to any particular marine phospholipid composition.
  • the marine phospholipid composition comprises phospholipids having the following structure: wherein R1 is OH or a fatty acid, R2 is OH or a fatty acid, and R3 is a mixture of H, choline, ethanolamine, inositol and serine, said phospholipid having at least 1% of omega-3 fatty acid moieties at positions R1 and/or R2.
  • the phospholipid composition comprises from about 20-50% of OH at positions R1 and/or R2.
  • the phospholipid composition is prepared from natural marine phospholipids isolated from a marine organism.
  • the composition further comprises a lipid carrier.
  • the phospholipid composition is enzymatically prepared by reacting lecithin with DHA and EPA in the presence of an enzyme.
  • the lecithin is soybean or egg lecithin.
  • the omega-3 fatty acid moieties are selected from the group of EPA and DHA and combination thereof.
  • the effective amount of said phospholipid composition comprises from about 300 to about 1000 mg omega-3 fatty acids.
  • the phospholipid composition is administered orally.
  • the phospholipid composition is provided in a gel capsule or pill.
  • FIG. 1 The total amount of EPA consumed during the four-week rat trial (mean ⁇ SE).
  • FIG. 5 Schematic drawing of experimental set-up.
  • FIG. 6 EPA levels in plasma as a function of hours after one bolus intake of a marine phospholipid composition.
  • FIG. 7 EPA levels in plasma as a function of hours after one bolus intake of a marine phospholipid composition.
  • FIG. 8 ARA levels in plasma as a function of hours after one bolus intake of a marine phospholipid composition.
  • phospholipid refers to an organic compound having the following general structure: wherein R1 is a fatty acid residue or —OH, R2 is a fatty acid residue or —OH, and R3 is a —H or a nitrogen containing compound such as choline (HOCH 2 CH 2 N + (CH 3 ) 3 OH ⁇ ), ethanolamine (HOCH 2 CH 2 NH 2 ), inositol or serine. R1 and R2 cannot simultaneously be OH.
  • R3 is an —OH
  • the compound is a diacylglycerophosphate
  • R3 is a nitrogen-containing compound
  • the compound is a phosphatide such as lecithin, cephalin, phosphatidyl serine or plasmalogen.
  • the R1 site is herein referred to as position 1 of the phospholipid
  • the R2 site is herein referred to as position 2 of the phospholipid
  • the R3 site is herein referred to as position 3 of the phospholipid.
  • omega-3 fatty acid refers to polyunsaturated fatty acids that have the final double bond in the hydrocarbon chain between the third and fourth carbon atoms from the methyl end of the molecule.
  • Non-limiting examples of omega-3 fatty acids include, 5,8,11,14,17-eicosapentaenoic acid (EPA), 4,7,10,13,16,19-docosahexanoic acid (DHA) and 7,10,13,16,19-docosapentanoic acid (DPA).
  • bioavailability refers to the degree and rate at which a substance (as a drug) is absorbed into a living system or is made available at the site of physiological activity.
  • the term “functional food” refers to a food product to which a biologically active supplement has been added.
  • fish oil refers to any oil obtained from a marine source e.g. tuna oil, seal oil and algae oil.
  • lipase refers to any enzyme capable of hydrolyzing fatty acid esters
  • the term “food supplement” refers to a food product formulated as a dietary or nutritional supplement to be used as part of a diet.
  • acylation means fatty acids attached to the phospholipid. 100% acylation means that there are no lyso- or glycerol-phospholipids.
  • This invention discloses that the uptake/absorption of omega-3 fatty acids attached to phospholipids are dependent on the level of LPL and GPL.
  • the level of LPL should be in the range of 15-45% and the level of GPL should be 0%.
  • this invention discloses that the pure PC transesterified with EPA/DHA have a different effect on gene expression in the liver than 40% PC transesterified with EPA/DHA. It is disclosed that the two compositions regulated around 40 genes differently.
  • the invention discloses that the EPA/DHA ratio is important. The treatment containing a EPA/DHA ratio of 2:1 regulated key enzymes involved in the inflammatory response (NF- ⁇ B) in a positive way, the treatment containing a EPA/DHA ratio of 1:1 did not.
  • the present invention describes novel marine lipid compositions comprising an omega-3 containing phospholipid and a triacylglyceride (TG) in a ratio from about 1:10 to 10:1.
  • the ratio is in the range of from about 3:1 to 1:3, more preferably the ratio is in the range of about 1:2 to 2:1.
  • the TG is a fish oil such as tuna oil, herring oil, menhaden oil, cod liver oil and algae oil.
  • this invention is not limited to omega-3 containing oils as other TG sources are contemplated such as vegetable oils.
  • the phospholipids in the composition have the following structure: wherein R1 is OH or a fatty acid, R2 is OH or a fatty acid, and R3 is a mixture of H, choline, ethanolamine, inositol and serine. Attached to position 1 or position 2 are least 1% omega-3 fatty acids, preferably at least 5%, more preferably at least 10% omega-3 fatty acids, up to about 15%, 20%, 30%, 40%, 50%, or 60% omega-3 fatty acids.
  • the omega-3 fatty acids can be EPA, DHA, DPA or C18:3 (n-3), most preferably the omega-3 fatty acids are EPA and DHA.
  • the phospholipid composition preferably contains OH in position 1 or position 2 in a range of 25% to 50% in order to maximize absorption in-vivo.
  • the present invention provides bioavailable and bioefficient omega-3 fatty acids.
  • This invention shows that the novel marine lipid composition disclosed above enhances the uptake of the omega-3 fatty acid in vivo and incorporates omega-3 fatty acids more efficiently into tissues of adult rats than pure fish oil does.
  • An embodiment of the invention is to use the marine lipid composition for efficient increase of omega-3 fatty acids in the liver, brain, adipose tissue, plasma, testicles and heart.
  • this invention also discloses that the marine lipid compositions efficiently reduced the concentration of the pro-inflammatory precursor AA in total lipids and in phospholipids in tissues.
  • the concentration of AA in the different lipid pools in the liver, brain, adipose tissue, plasma, testicles and heart can be more efficiently reduced than using fish oil.
  • the composition can be used to improve the EPA/AA ratio, which is a bio-marker of silent inflammation.
  • the invention also discloses that the incorporation of the omega-3 fatty acids into monocytes is also more efficient using the claimed marine lipid composition as opposed to the fish oil.
  • Yet another embodiment of the invention is to use the marine lipid composition to reduce chronic and acute inflammation in humans and in animals. Acute inflammation is mediated by granulocytes or polymorphonuclear leukocytes, while chronic inflammation is mediated by mononuclear cells such as monocytes.
  • Monocytes protect against blood-borne pathogens and moves quickly to sites of infection in the tissues, secreting large amounts of pro-inflammatory prostaglandins. Furthermore, low grade chronic inflammation may be the underlying cause of many life-style related diseases such as obesity, arthritis, diabetes type II, metabolic syndrome, Alzheimer's disease, osteoarthritis, inflammatory bowel disease, allergy and asthma [14].
  • the marine lipid composition can be used to treat and prevent diseases linked to chronic inflammation.
  • This invention discloses that the inflammatory response of monocytes harvested from animals in lower in animals treated with the marine lipid composition compared to fish oil.
  • the concentrations of the pro-inflammatory cytokines such as interleukin-1 ⁇ , interleukin-6 as well as tumor necrosis factor ⁇ (TNF- ⁇ ) were reduced for the group fed the marine lipid composition compared to fish oil.
  • These cytokines are important markers of real inflammation as for examples I1-1 ⁇ induces fever.
  • I1-6 also induces fever in addition to being linked to the acute phase response.
  • TNF- ⁇ is involved in systemic inflammation as well and is released by white blood cells in the case of damage. It has a range of different biological effects such as increasing insulin resistance, stimulating the acute phase response in the liver and affecting the hypothalamus causing appetite suppression and fever.
  • This invention also discloses that the fatty acid composition of the brain and adipose tissue phospholipids changes after in take of omega-3 fatty acids for 30 days.
  • a significant reduction of the arachidonic acids can be found in the phospholipids in the brain and adipose tissue for the rats given either the EPA- or DHA-rich PL diets (PL 1 and PL 2, respectively). This may affect the inflammatory response in this tissue and thereby have a great impact on cognitive diseases/conditions such as Parkinson's or and Alzheimer's where the inflammatory component is fundamental for the progression of the disease.
  • This invention also discloses that the reduction of ARA is present also in the sn-2 position of the phospholipids in the brain.
  • adipose tissue In adipose tissue, accumulation of EPA and DHA in both total lipids (table 3) and PLs (table 8) is substantial when omega-3 supplements were fed and negligible when the control diet was fed. The increase was more pronounced in total lipids, which mainly consists of triglycerides (99% of fat cell lipid content).
  • This invention demonstrates that omega-3 phospholipids can increase the accumulation of EPA/DHA into adipose tissue. This is important as the adipose tissue can function as a reservoir for these fatty acids.
  • Arachidonic acid concentration in total lipids was higher in omega-3 supplemented animals, showing probably an increase of lipoprotein lipase activity, in agreement with the ability of omega 3 in decreasing plasma TAGs concentration.
  • arachidonic acid levels in adipose tissue PLs were significantly lower in omega-3 supplemented animals than the levels in controls. Peculiar enough, the PL-EPA diet was the most efficient in decreasing arachidonic acid.
  • the invention discloses that the reduction of ARA is also observed in the sn-2 position of the phospholipids of the omega-3 supplemented animals. This is very important as the pro-inflammatory eicosanoids are produced from ARA, which are catalytically hydrolyzed from position 2 on the phospholipid by the action of phospholipase A2. The phospholipase A2 is released after stimuli at the cell wall, it then moves to the nuclear membrane where the hydrolysis of the phospholipid takes place. The reduction of ARA in position 2 on the phospholipids may affect the inflammatory response in this tissue, which may have practical application in different pathologies of the adipose tissue and in its physiological activity of accumulation and release of fatty acids.
  • Fatty acid data from brain are well in line with the data from adipose tissue. Also in this tissue, we found a significant decrease of arachidonic acid in PLs, but surprisingly, only with PL-EPA and PL-DHA (table 7). On the other hand, DHA levels in both total lipids and PLs were not influenced by the omega-3 diets, while there was a small but significant increase in EPA levels. Lack of increase in DHA levels is likely to be attributable to the fact that the rats in this study were adults and pass the stage in development where they incorporate DHA in the brain (mainly PE). On the other hand, EPA being present at low concentration has more margin to increase.
  • liver as expected, we found for all omega-3 groups a significant increase of EPA and DHA and decrease of arachidonic acid. No great differences were expected between total lipids and PLs because about 80% of liver total lipids are PLs (table 4, 9 and 14).
  • Heart total lipids and PLs showed a strong increase of EPA and DHA with a concomitant decrease of arachidonic acid when omega-3 supplements were fed.
  • the strong decrease in the omega-6/omega-3 ratio in heart lipids is important considering the possible impact on the anti-inflammatory potential.
  • Observed change in heart tissue fatty acids increase of fatty acids with 6 or 5 double bonds
  • This change was most striking in the PL-DHA group where the increase of DHA was significantly higher than the increase in the TG-oil and PL-EPA groups.
  • the fluidity of myocardium cell membrane seems to play an important role in controlling arrhythmia. Ventricular arrhythmia, is one of the main causes of sudden cardiac death. Furthermore, atrial fibrillation is another pathological state with a high incidence and important health consequences.
  • Testicular long chain PUFAs are of special interest because there is a high rate of production of prostaglandins from the omega-6 PUFA (arachidonic acid mainly) into the semen or seminal fluid.
  • High rate of prostaglandin production does not indicate an active inflammatory process but a stimulus for the uterus smooth muscle to favor male fertility.
  • An omega-3 induced decrease of arachidonic acid as observed in other tissue could be detrimental to the male fertility if it occurred also in testis.
  • testicular tissue has also a high level of DPA (22:5 omega-6), which may serve as a reservoir for arachidonic acid.
  • Arachidonic acid could be formed according to the need, through the retroconversion mechanism in the peroxisomes.
  • Another embodiment of the invention is to formulate the marine lipid compositions into a feed product for the purpose of reducing low-grade chronic inflammation in animals. It can also be formulated into a food product and given to humans for the same purpose. Furthermore, it can be formulated as a functional food product, as a drug or as food supplement.
  • the compositions of this invention are contained in acceptable excipients and/or carriers for oral consumption.
  • the actual form of the carrier, and thus, the compositions itself, is not critical.
  • the carrier may be a liquid, gel, gelcap, capsule, powder, solid tablet (coated or non-coated), tea, or the like.
  • the composition is preferably in the form of a tablet or capsule and most preferably in the form of a hard gelatin capsule.
  • Suitable excipient and/or carriers include maltodextrin, calcium carbonate, dicalcium phosphate, tricalcium phosphate, microcrystalline cellulose, dextrose, rice flour, magnesium stearate, stearic acid, croscarmellose sodium, sodium starch glycolate, crospovidone, sucrose, vegetable gums, lactose, methylcellulose, povidone, carboxymethylcellulose, corn starch, and the like (including mixtures thereof).
  • Preferred carriers include calcium carbonate, magnesium stearate, maltodextrin, and mixtures thereof.
  • the various ingredients and the excipient and/or carrier are mixed and formed into the desired form using conventional techniques.
  • the tablet or capsule of the present invention may be coated with an enteric coating that dissolves at a pH of about 6.0 to 7.0.
  • a suitable enteric coating that dissolves in the small intestine but not in the stomach is cellulose acetate phthalate. Further details on techniques for formulation for and administration may be found in the latest edition of Remington's Pharmaceutical Sciences (Maack Publishing Co., Easton, Pa.).
  • the composition contains no traces of organic solvents which is an important property regarding the safety of consuming such compounds.
  • Phospholipids prepared using chemical or enzymatic methods in the presence of organic solvents may contain residual solvents that may be a health hazard.
  • VOC are often co-extracted when marine phospholipids are extracted, such VOC's may contribute to the smell taste of the phospholipids.
  • the supplement is provided as a powder or liquid suitable for adding by the consumer to a food or beverage.
  • the dietary supplement can be administered to an individual in the form of a powder, for instance to be used by mixing into a beverage, or by stirring into a semi-solid food such as a pudding, topping, sauce, puree, cooked cereal, or salad dressing, for instance, or by otherwise adding to a food.
  • compositions of the present invention may also be formulated with a number of other compounds. These compounds and substances add to the palatability or sensory perception of the particles (e.g., flavorings and colorings) or improve the nutritional value of the particles (e.g., minerals, vitamins, phytonutrients, antioxidants, etc.).
  • the dietary supplement may comprise one or more inert ingredients, especially if it is desirable to limit the number of calories added to the diet by the dietary supplement.
  • the dietary supplement of the present invention may also contain optional ingredients including, for example, herbs, vitamins, minerals, enhancers, colorants, sweeteners, flavorants, inert ingredients, and the like.
  • the dietary supplement of the present invention may contain one or more of the following: ascorbates (ascorbic acid, mineral ascorbate salts, rose hips, acerola, and the like), dehydroepiandosterone (DHEA), Fo-Ti or Ho Shu Wu (herb common to traditional Asian treatments), Cat's Claw ( ancient herbal ingredient), green tea (polyphenols), inositol, kelp, dulse, bioflavinoids, maltodextrin, nettles, niacin, niacinamide, rosemary, selenium, silica (silicon dioxide, silica gel, horsetail, shavegrass, and the like), spirulina, zinc, and the like.
  • Such optional ingredients may be either naturally occurring or concentrated forms.
  • the dietary supplements further comprise vitamins and minerals including, but not limited to, calcium phosphate or acetate, tribasic; potassium phosphate, dibasic; magnesium sulfate or oxide; salt (sodium chloride); potassium chloride or acetate; ascorbic acid; ferric orthophosphate; niacinamide; zinc sulfate or oxide; calcium pantothenate; copper gluconate; riboflavin; beta-carotene; pyridoxine hydrochloride; thiamin mononitrate; folic acid; biotin; chromium chloride or picolonate; potassium iodide; sodium selenate; sodium molybdate; phylloquinone; vitamin D 3 ; cyanocobalamin; sodium selenite; copper sulfate; vitamin A; vitamin C; inositol; potassium iodide.
  • vitamins and minerals including, but not limited to, calcium phosphate or acetate, tribasic; potassium
  • compositions comprise at least one food flavoring such as acetaldehyde(ethanal), acetoin(acetyl methylcarbinol), anethole(parapropenyl anisole), benzaldehyde(benzoic aldehyde), N-butyric acid (butanoic acid), d- or l-carvone(carvol), cinnamaldehyde(cinnamic aldehyde), citral(2,6-dimethyloctadien-2,6-al-8, gera-nial, neral), decanal(N-decylaldehyde, capraldehyde, capric aldehyde, caprinaldehyde, aldehyde C-10), ethyl acetate, ethyl butyrate, 3-methyl-3-phenyl glycidic acid ethyl ester(ethyl-methyl-phenyl-glycid)
  • compositions comprise at least one synthetic or natural food coloring (e.g., annatto extract, astaxanthin, beet powder, ultramarine blue, canthaxanthin, caramel, carotenal, beta carotene, carmine, toasted cottonseed flour, ferrous gluconate, ferrous lactate, grape color extract, grape skin extract, iron oxide, fruit juice, vegetable juice, dried algae meal, tagetes meal, carrot oil, corn endosperm oil, paprika, paprika oleoresin, riboflavin, saffron, tumeric, tumeric and oleoresin).
  • synthetic or natural food coloring e.g., annatto extract, astaxanthin, beet powder, ultramarine blue, canthaxanthin, caramel, carotenal, beta carotene, carmine, toasted cottonseed flour, ferrous gluconate, ferrous lactate, grape color extract, grape skin extract, iron oxide, fruit juice, vegetable juice, dried algae
  • compositions comprise at least one phytonutrient (e.g., soy isoflavonoids, oligomeric proanthcyanidins, indol-3-carbinol, sulforaphone, fibrous ligands, plant phytosterols, ferulic acid, anthocyanocides, triterpenes, omega 3/6 fatty acids, conjugated fatty acids such as conjugated linoleic acid and conjugated linolenic acid, polyacetylene, quinones, terpenes, cathechins, gallates, and quercitin).
  • phytonutrient e.g., soy isoflavonoids, oligomeric proanthcyanidins, indol-3-carbinol, sulforaphone, fibrous ligands, plant phytosterols, ferulic acid, anthocyanocides, triterpenes, omega 3/6 fatty acids, conjugated fatty acids such as conjugated linole
  • Sources of plant phytonutrients include, but are not limited to, soy lecithin, soy isoflavones, brown rice germ, royal jelly, bee propolis, acerola berry juice powder, Japanese green tea, grape seed extract, grape skin extract, carrot juice, bilberry, flaxseed meal, bee pollen, ginkgo biloba, primrose (evening primrose oil), red clover, burdock root, dandelion, parsley, rose hips, milk thistle, ginger, Siberian ginseng, rosemary, curcumin, garlic, lycopene, grapefruit seed extract, spinach, and broccoli.
  • the compositions comprise at least one vitamin (e.g., vitamin A, thiamin (B1), riboflavin (B2), pyridoxine (B6), cyanocobalamin (B12), biotin, ascorbic acid (vitamin C), retinoic acid (vitamin D), vitamin E, folic acid and other folates, vitamin K, niacin, and pantothenic acid).
  • the particles comprise at least one mineral (e.g., sodium, potassium, magnesium, calcium, phosphorus, chlorine, iron, zinc, manganese, flourine, copper, molybdenum, chromium, selenium, and iodine).
  • a dosage of a plurality of particles includes vitamins or minerals in the range of the recommended daily allowance (RDA) as specified by the United States Department of Agriculture.
  • the particles comprise an amino acid supplement formula in which at least one amino acid is included (e.g., 1-carnitine or tryptophan).
  • Thermomyces Lanuginosus lipase a variety of lipases is contemplated, including, but not limited to, Thermomyces Lanuginosus lipase, Rhizomucor miehei lipase, Candida Antarctica lipase, Pseudomonas fluorescence lipase, and Mucor javanicus lipase.
  • free fatty acids are added such as EPA and DHA, however any omega-3 fatty acid is contemplated.
  • a strong vacuum is applied to the reaction vessel for 72 hours.
  • the reaction length can be varied in order to obtain a composition with the desired amount of phospholipids and lyso-phospholipids. By extending the reaction time beyond 72 hours, a product comprising more than 65% phospholipids can be obtained.
  • a lipid carrier is added to the reaction mixture in order to reduce the viscosity of the solution.
  • the added amount of triglycerides can be 10%, 20%, 30%, 40% or more, it depends on the requested viscosity of the final product.
  • the lipid carrier can be a fish oil such as tuna oil, menhaden oil and herring oil, or any triglyceride, diglyceride, ethyl- or methylester of a fatty acid.
  • the product is subjected to a molecular distillation and the free fatty acids are removed, resulting in a final product comprising of phospholipids (lyso-phospholipids and phospholipids) and triglycerides in a ratio of preferably 2:1.
  • This invention further discloses a process for the enzymatic transesterification/esterification of phospholipids with fatty acids alkyl esters or free fatty acids in an evacuated vessel (B).
  • a reduced pressure is applied to the vessel B (0.001-30 mbar) and water vapor (moisture) is allowed to enter the reaction mixture through a tube from a second vessel (A) ( FIG. 5 for schematic drawing of the experimental setup).
  • the water in vessel A is heated to 25-30° C.
  • the rate of reaction could either be increased of the lipase dosage could be reduced.
  • the reuse of the enzymes was improved.
  • a novel marine phospholipid composition was prepared characterized by being acylated in the range of 55%-85%, having at least 5% EPA and/or DHA esterified, having a EPA/DHA ratio of at least 1.
  • the present invention utilizes a phospholipid, preferably a phosphatide such as lecithin.
  • a phospholipid preferably a phosphatide such as lecithin.
  • the present invention is not limited to the use of any particular phospholipid. Indeed, the use of a variety of phospholipids is contemplated.
  • the phospholipid is a phosphatidic or lysophosphatidic acid.
  • the phospholipid is a mixture of phosphatides such as phosphatidylcholine, phosphatidylethnolamine, phosphatidylserine and phosphatidylinositol.
  • the present invention is not limited to the use of any particular source of phospholipids.
  • the phospholipids are from soybeans, while in other embodiments, the phospholipids are from eggs.
  • the phospholipids utilized are commercially available, such as Alcolec 40P® from American Lecithin Company Inc (Oxford, Conn., USA).
  • Alcolec 40P® from American Lecithin Company Inc (Oxford, Conn., USA).
  • the present invention is not limited to the use of any particular enzyme. Indeed, the use of a variety of enzymes is contemplated, including, but not limited to Thermomyces Lanuginosus lipase, Rhizomucor miehei lipase, Candida Antarctica lipase, Pseudomonas fluorescence lipase, and Mucor javanicus lipase.
  • This invention is not limited to any particular fatty acid alkyl ester either. This includes, but not limited to: decanoic acid (10:0), undecanoic acid (11:0), 10-undecanoic acid (11:1), lauric acid (12:0), cis-5-dodecanoic acid (12:1), tridecanoic acid (13:0), myristic acid (14:0), myristoleic acid (cis-9-tetradecenoic acid, 14:1), pentadecanoic acid (15:0), palmitic acid (16:0), palmitoleic acid (cis-9-hexadecenoic acid, 16:1), heptadecanoic acid (17:1), stearic acid (18:0), elaidic acid (trans-9-octadecenoic acid, 18:1), oleic acid (cis-9-octadecanoic acid, 18:1), nonadecanoic acid (19:0), eicosanoi
  • the synthetic marine phospholipid compositions of the present invention are substantially free of volatile organic compounds and are therefore much more suitable as a food supplement for humans and animals. Accordingly, in preferred compositions, the present invention provides synthetic marine phospholipids compositions having high or increased palatability, wherein the high or increased palatability is due to low levels of organic solvents and/or volatile organic compounds. In preferred embodiments, palatability is assayed by feeding the composition to a panel of subjects, preferably human. In more preferred embodiments, the phospholipids compositions have high or increased palatability as compared to naturally extracted marine phospholipids. In other preferred embodiments, the synthetic marine phospholipids compositions of the present invention are safe for oral administration.
  • the difference in bioavailability and bioefficacy between the marine lipid composition of the present invention and a fish oil were investigated in a rat experiment.
  • the rat feed was prepared using AIN-93 except that soybean oil was removed from the feed.
  • the pelleted AIN-93 diet was ground and the marine lipid compositions (PL 1 and PL 2) as well as fish oil (TG oil) and control were added to this ground feed.
  • the marine lipid compositions were prepared using enzymatic (lipase) catalyzed transesterification of soy lecithin with fish oil fatty acids according to the method described in Example 4, followed by the addition of a triglyceride carrier and short path distillation.
  • Sprague Dawley rats Thirty six newly weaned male Sprague Dawley rats (start weight 168 ⁇ 11 g) were used in the experiment.
  • the rats were initially given low-essential oil rat feed, containing 20 g of sunflower oil and 10 g of flaxseed oil per kg of feed, for one week.
  • modified AIN-93 diet powder without the test oil was given to rats ad libitum until the start of the experiment. Feeding of rats was stopped 12 hours before the sampling, 30 days after the start of feeding.
  • Each rat was individually anesthetized with carbon dioxide, weighed and euthanized with cervical dislocation. Next, blood was sampled and centrifuged to separate plasma and blood cells.
  • the total fatty acid profile for the lipids in the brains (table 2), adipose tissue (table 3), liver (table 4), testicles (table 5) and heart (table 6) were isolated from the rats in example 1.
  • the PL 1 composition increases the DHA content in brain and adipose tissue more than the TG composition.
  • the PL 1 composition increase the EPA content in the adipose tissue more than the TG composition. It is to be observed that the PL1 composition increases the EPA/DHA content in the phospholipids and in the total lipids of the different tissues as well as reduces the AA/EPA ratio more than the TG oil composition.
  • TABLE 2 Fatty acid profile of the total lipids isolated from the rat brain (mmol/g lipids).
  • a marine phospholipid composition containing 8.4% EPA and 1.2% DHA was prepared using a crude soybean lecithin as a starting material according to [19].
  • a marine oil was added to the phospholipid mixture (30% w/w) so that the total level of EPA was 21.9% and for DHA 9.4%.
  • soy lecithin and lyso-phospholipids prepared according to [20] were added to the mixture in variable amounts so that a range of PC/LPC/GPC ratios could be obtained (Table 17).
  • all the treatments (MPL1-MPL5) contained exactly the same amount EPA and DHA.
  • Lipid compositions were consumed as a single bolus by adult Sprague-Dawley rats and the appearance of EPA/DHA in blood was measured at different time points from 1 to 12 hours after ingestion. The concentration of EPA/DHA was determined using GC-FID and reported as area percentage.
  • FIGS. 6 and 7 show that composition MPL2 and MPL3 results in the highest concentration of EPA/DHA in plasma after uptake. Comparing the surface area under each curve it is clear that MPL2 and MPL3 demonstrates a higher bioavailability of EPA/DHA than the other composition MPL1, MPL4 and MPL5.
  • Marine phospholipids were prepared using either 40% soy PC (American Lecithin Company Inc, Oxford, Conn., USA) (MPL1) or 96% pure soy PC (Phospholipid GmbH, GmbH, Germany) (MPL2) according to a method described by others [4]. Fatty acid content and the level of bi-products are shown in table 18.
  • the MPL treatments consisted of a mixture of phospholipids, lyso-phospholipids and glycerol-phospholipids. Looking only at the PC/LPC/GPC relationship, it was 64/33/2 and 42/40/18 for MPL1 and MPL2, respectively. Finally, all three treatments were emulsified into skimmed milk.
  • Sprague-Dawley rats were fed the milk emulsions for 1 week. Each rat was placed in its own cage to ensure that they got an even amount of test substance and the milk was consumed by the rat pups ad libitum. After 1 week the experiment was terminated and the rats were decapitated. The animals were kept without food for 24 hours before sampling. Entire livers were collected and frozen immediately using liquid nitrogen (stored at ⁇ 65° C.). Total RNA was isolated from the liver samples according to the Quiagen Rnaesy Midi Kit Protocol. The RNA samples were quantified and quality measured by NanoDrop and Bioanalyzer.
  • FDR False Discovery Rate
  • MPL1 and MPL2 are biologically different compounds due to the fact that over 40 genes were differentially expressed (table 19).
  • TABLE 19 List of genes differentially expressed (DE) by MPL1 versus MPL2. The list is sorted according to increasing p-values.
  • SLR Estimated signal log-ratio ( ⁇ 0: down regulated gene, >0: up regulated gene).
  • Fold change Estimated fold change corresponding to the parameter ( ⁇ 1: down regulated gene, >1: up regulated gene).
  • Affy fold change Estimated fold change using the Affymetrix definition ( ⁇ 1: down regulated gene, >1: up regulated gene)
  • MPL2 regulates 401 genes versus the control (table 20).
  • a number of genes listed are involved maintenance of the cell, in transcription and protein synthesis as well as signaling pathways. Others are involved in regulation of metabolism and the inflammatory response such as Tnf receptor-associated factor 6 (Traf6_predicted) (fold change of 0.53), guanine nucleotide binding protein alpha inhibiting 2 (Gnai2) (fold change of 0.6, gamma-butyrobetaine hydroxylase (Bbox1) (fold change of 1.32), monoglyceride lipase (Mg11) (fold change 0.52), nuclear NF-kappaB activating protein (fold change 0.65) and CCAAT/enhancer binding protein (C/EBP) (fold change of 0.66).
  • Tnf receptor-associated factor 6 Traf6_predicted
  • Gnai2 guanine nucleotide binding protein alpha inhibiting 2
  • Bbox1 gamma-butyrobetaine hydroxylase
  • fatty acid ethyl ester consisting of 10% EPA and 50% DHA (FAEE 10-50), obtained from Napro Pharma (Brattvaag, Norway) and 15 g TL-IM obtained from Novozymes (Bagsvaerd, Denmark) were mixed in an evacuated round bottomed glass flask for 15 minutes. Next, N 2 was released into the glass flask and the mixture was heated to 65° C. 20 g Alcolec 40P® from American Lecithin Company Inc (Oxford, Conn., USA) was then added to the reaction mixture.
  • Alcolec 40P® is a crude soybean phospholipid product containing 40% PC, 26% phosphatidylethanolamine, 11% phosphatidylinositol, 1% phosphatidylserine, 13% phytoglycolipids as well as 14% other phosphatides (w,w).
  • the glass flask was evacuated (20-30 mbar).
  • a second vessel containing water (30° C.) was connected to the reaction vessel through a plastic tube ( FIG. 1 ). The reduced pressure allowed moisture from the headspace of the second vessel to be added through the reaction mixture continuously. In order to obtain the final product the enzymes were removed by filtration.
  • a triglyceride carrier was added to the product, followed by removal of the residual free fatty acids and/or esters by short path distillation.
  • the isolated PC+LPC fraction was then dried under nitrogen prior to derivatization; finally the fatty acid profile was determined by analyzing the derivatives using GC-FID.
  • the relationship between PC, LPC and GPC was determined using HPLC with the method above, except that the UV detector was replaced by an evaporative light scattering detection (ELSD).
  • ELSD evaporative light scattering detection
  • the enzymes from example 7 were isolated by filtration and the possibility of reuse was determined in the following experiment. 30 g FAEE (10-50), 10 g Alcolec and 15 g used enzymes (equivalent to 7.5 g enzyme because the used enzymes had absorbed product from the first reaction). The reaction was performed at 65° C. and stirred at 200 rpm using a shaker incubator. The transesterified phospholipids were analyzed as in the previous example and the results are shown in table 21 below.

Abstract

Novel marine lipid compositions comprising triglycerides and omega-3 rich phospholipids are described. The compositions are characterized by providing highly bioavailable omega-3, increased tissue incorporation of omega-3 and reduced concentration of pro-inflammatory cytokines.

Description

  • This application claims the benefit of U.S. Provisional Applications 60/798,026, 60/798,027, and 60/798,030, all filed May 5, 2006, and U.S. Provisional Application 60/872,096, filed Dec. 1, 2006, each of which is incorporated herein by reference in their entirety.
  • FIELD OF THE INVENTION
  • The present invention relates to novel marine lipid compositions comprising combinations of omega-3 fatty acid rich functional phospholipids and omega-3 fatty acid rich triglycerides. In addition, food supplements, functional food, drugs and feed products comprising such compositions are provided along with methods of their use.
  • BACKGROUND OF THE INVENTION
  • Marine lipids such as omega-3 rich triglycerides and omega-3 rich phospholipids can be isolated from a number of different natural sources such as fish, crustaceans, plankton, seals, whales as well as algae using extraction technologies. In addition, they can be prepared industrially using chemical or bio-catalytical methods such as enzyme catalyzed transesterification of crude soy lecithin with fish oil fatty acids [1].
  • The anti-inflammatory properties of omega-3 fatty acids are well known and the use as an anti-inflammatory agent has been described both for triglycerides and phospholipids [2-3]. Actually, omega-3 fatty acids are famous for their anti-inflammatory properties, and it has been shown that omega-3 fatty acids alleviate the symptoms of a series of autoimmune, atherosclerotic and inflammatory diseases including inflammatory bowel diseases and rheumatoid arthritis [4-6]. Suppression of inflammation has been proposed as one of the strategies to slow down the progress of these diseases. Hence, this invention discloses the effect on marine lipid compositions on the concentration of markers of inflammation such as TNF-α and other cytokines such as interleukin-1β and interleukin 6. In addition, since arachidonic acid (AA) is the predominant precursor of the eicosanoid mediators of inflammatory responses (prostaglandins, thromboxanes and leukotrienes), this invention discloses the reduction of AA level and the improvement in the EPA/AA ratio in different lipid pools in tissues such as in the phospholipids isolated from adipose tissue, heart, testicles, plasma, brain and liver.
  • The bioavailability of EPA and DHA from fish oil triglycerides have been reported to be high in healthy adults. However, for certain conditions i.e. pathological conditions such as extrahepatic cholestasis and for pre-term infants the absorption can be low. For example it was shown that the absorption of DHA from egg lecithin in pre-term infants was 90% compared to 80% from triglycerides [7]. Absorption of long chain PUFA (AA and DHA) is less (75% and 62%, respectively) than the absorption of C18 PUFA (94%) in pre-term infants [8]. The difference between C18 PUFA and long chain PUFA absorption is likely to become less apparent in older children and adults. Sala-Vila et al [9-10] investigated the bioavailabilities of DHA-PL and DHA-TG in full term infants and found no differences based on plasma lipid enrichments. Valenzuela et al. [11] supplemented female rats with different forms of DHA including egg yolk PL and single cell algae TG. They found also no difference in absorption of DHA from PL and TG based on plasma lipid enrichments. However, the tissue and milk fat levels were higher in PL-DHA compared to the TG-DHA supplemented rats. These data indicate that although there were no differences in the bioavailability, efficacy with respect to tissue enrichment was higher for PL-DHA compared to TG-DHA. Furthermore, the relative absorption of EPA and DHA ethyl esters (4 g/d) compared to oleic acid calculated from peak concentrations was 94 and 100%, respectively. Estimates of relative absorption based on the area under the concentration curve indicated a relative absorption of 91% for EPA and 93% for DHA [12]. Bioavailability of C18:1, C18:2 and C18:3 in adult humans are close to 100% (note 94% in preterm infants). Thus the bioavailability of EPA and DHA delivered in different forms is, according to previous, work likely to be over 90%.
  • SUMMARY OF THE INVENTION
  • In some embodiments, the invention provides a composition comprising a triglyceride and a phospholipids in a ratio ranging from 1:10 to 10:1; said phospholipids having the following structure:
    Figure US20080044487A1-20080221-C00001

    wherein R1 is OH or a fatty acid, R2 is OH or a fatty acid, and R3 is a mixture of H, choline, ethanolamine, inositol and serine, said phospholipid having at least 1% of DHA/EPA, said phospholipids have a concentration of OH in the range of 25-50%. In further embodiments, the invention provides a marine lipid composition characterized by providing higher uptake of omega-3 fatty acids into plasma as compared to administration of purified triglycerides, phospholipids, or natural marine phospholipids. In further embodiments, the invention provides a composition characterized by efficiently improving the AA/EPA ratio in plasma phospholipids as compared to administration of purified triglycerides, phospholipids, or natural marine phospholipids. In still other embodiments, the invention is a marine lipid composition characterized by efficiently increasing the concentration of omega-3 fatty acids in tissues as compared to administration of purified triglycerides, phospholipids, or natural marine phospholipids. In still further embodiments, the invention the composition is characterized by reducing the concentration of biomarkers of inflammation as compared to administration of purified triglycerides, phospholipids, or natural marine phospholipids. In other embodiments of the invention, the marine lipid composition is formulated into an animal feed, a food product, a food supplement and a drug.
  • In some embodiments, the present invention provides a composition comprising phospholipids having the following structure:
    Figure US20080044487A1-20080221-C00002

    wherein R1 is OH or a fatty acid, R2 is OH or a fatty acid, and R3 is a mixture of H, choline, ethanolamine, inositol and serine, said phospholipid having at least 1% of DHA/EPA at positions R1 and/or R2 and from about 20-50% of OH at positions R1 and/or R2. In some embodiments, the composition is acylated in a range from about 55% to about 85%. In some embodiments, the omega-3 fatty acids are selected from the group consisting of EPA, DHA, DPA and α-linolenic acid (ALA). In some embodiments, the composition is substantially free of organic solvents and volatile organic compounds such as short chain fatty acids, short chain aldehydes and short chain ketones. In some embodiments, the composition has at least 5% of a combination of EPA and DHA esterified. In some embodiments, the composition has at least 10% of a combination of EPA and DHA esterified. In some embodiments, the composition has at least 20% of a combination of EPA and DHA esterified. In some embodiments, the composition has at least 30% of a combination of EPA and DHA esterified. In yet other embodiments, said composition contains from about 5%, 10%, 20% and 30% EPA/DHA attached to position 1 and/ or position 2. In some embodiments, the composition has a ratio of EPA/DHA ranging from 1:1 to 4:1. In some embodiments, the composition has a ratio of EPA/DHA ranging from 2:1 to 4:1. In some embodiments, the composition is acylated in a range from 60% to 80%. In some embodiments, the composition is acylated in a range from 50% to 75%.
  • In some embodiments, the composition further comprises a lipid carrier in a ratio of from 1:10 to 10:1 to said phospholipids. In some embodiments, the lipid carrier and said phospholipids are in a ratio of from about 5:1 to 1:5. In some embodiments, the composition comprises from about 20% to about 90% of said phospholipid composition and from about 10% to about 50% of said lipid carrier. The present invention is not limited to any particular lipid carrier. In some embodiments, the lipid carrier is selected from the group consisting of a triglyceride, a diglyceride, an ethyl ester, and a methyl ester and combinations thereof. In some embodiments, the composition provides higher uptake of omega-3 fatty acids into plasma as compared to natural marine phospholipids when administered to subjects. In some embodiments, the composition improves the AA/EPA ratio in plasma phospholipids when administered to subjects as compared to natural marine phospholipids. In some embodiments, the composition increases the concentration of omega-3 fatty acids in tissues when administered to subjects as compared to natural marine phospholipids. In some embodiments, the composition reduces the concentration of biomarkers of inflammation when administered to subjects as compared to natural marine phospholipids. In some embodiments, the present invention provides a food product comprising the foregoing compositions. In some embodiments, the present invention provides an animal feed comprising the foregoing compositions. In some embodiments, the present invention provides a food supplement comprising the foregoing compositions. In some embodiments, the present invention provides a pharmaceutical composition comprising the foregoing compositions.
  • In some embodiments, the present invention provides methods of preparing a bioavailable omega-3 fatty acid composition comprising: a) providing a purified phospholipid composition comprising omega-3 fatty acid residues and a purified triglyceride composition comprising omega-3 fatty acid residues; b) combining said phospholipid composition and said triglyceride composition to form a bioavailable omega-3 fatty acid composition. In some embodiments, the bioavailable phospholipid composition is one of the compositions described above. In some embodiments, the methods further comprise the step of encapsulating said bioavailable omega-3 fatty acid composition. In some embodiments, the bioavailable omega-3 fatty acid composition has increased bioavailability as compared to purified triglycerides or phospholipids comprising omega-3 fatty acid residues. In some embodiments, the methods further comprise the step of packaging the bioavailable omega-3 fatty acid composition for use in functional foods. In some embodiments, the methods further comprise the step of assaying the bioavailable omega-3 fatty acid composition for bioavailability. In some embodiments, the methods further comprise administering the bioavailable omega-3 fatty acid composition to a patient. In some embodiments, the present invention provides a food product, animal feed, food supplement or pharmaceutical composition made by the foregoing process.
  • In some embodiments, the present invention provides methods for reducing symptoms of cognitive dysfunction in a child comprising administering an effective amount of a marine phospholipid composition, wherein said symptoms are selected from the group consisting of ability to complete task, ability to stay on task, ability to follow instructions, ability to complete assignments, psychomotor function, long term memory, short term memory, ability to make a decision, ability to follow through on decision, ability to self-sustain attention, ability to engage in conversations, sensitivity to surroundings, ability to plan, ability to carry out plan, ability to listen, interruptions in social situations, temper tantrums, level/frequency of frustration, level/frequency restlessness, frequency/level fidgeting, ability to exhibit delayed gratification, aggressiveness, demanding behavior/frequency of demanding behavior, sleep patterns, restive sleep, interrupted sleep, awakening behavior, disruptive behavior, ability to exhibit control in social situations, ability to extrapolate information and ability to integrate information. In some embodiments, the child exhibits one or more symptoms of Attention Deficit Hyperactivity Disorder (ADHD), is suspected of having ADHD, or has been diagnosed with ADHD. In some embodiments, the child exhibits one or more symptoms of autistic spectrum disorder, is suspected of having autistic spectrum disorder, or has been diagnosed with autistic spectrum disorder. In further embodiments, the present invention provides methods of increasing cognitive performance in an aging mammal comprising administering an effective amount of a marine phospholipid composition. In some embodiments, the cognitive performance is selected from the group consisting of memory loss, forgetfulness, short-term memory loss, aphasia, disorientation, disinhibition, and behavioral changes. In some embodiments, the mammal is a human. In some embodiments, the mammal is a pet selected from the group consisting of cats and dogs. In some embodiments, the mammal has symptoms of age-associated memory impairment or decline.
  • The foregoing methods are not limited to the use of any particular marine phospholipid composition. In some embodiments, the marine phospholipid composition comprises phospholipids having the following structure:
    Figure US20080044487A1-20080221-C00003

    wherein R1 is OH or a fatty acid, R2 is OH or a fatty acid, and R3 is a mixture of H, choline, ethanolamine, inositol and serine, said phospholipid having at least 1% of omega-3 fatty acid moieties at positions R1 and/or R2. In some embodiments, the phospholipid composition comprises from about 20-50% of OH at positions R1 and/or R2. In some embodiments, the phospholipid composition further comprises a lipid carrier. In some embodiments, the phospholipid composition is prepared from natural marine phospholipids isolated from a marine organism. In some embodiments, the phospholipid composition is enzymatically prepared by reacting lecithin with DHA and EPA in the presence of an enzyme. In some embodiments, the lecithin is soybean or egg lecithin. In some embodiments, the omega-3 fatty acid moieties are selected from the group of EPA and DHA and combination thereof. In some embodiments, the effective amount of said phospholipid composition comprises from about 300 to about 1000 mg omega-3 fatty acids. In some embodiments, the phospholipid composition is administered orally. In some embodiments, the phospholipid composition is provided in a gel capsule or pill.
  • In some embodiments, the present invention provides methods of treating a subject by administration of a marine phospholipid composition comprising administering a marine phospholipid composition to said subject under conditions such that a desired condition is improved, wherein said conditions is selected from the group consisting of fertility, physical endurance, sports performance, muscle soreness, inflammation, auto-immune stimulation, metabolic syndrome, obesity and type II diabetes. In some embodiments, the subject is a human. In some embodiments, the subject is a companion animal. The present invention is not limited to any particular marine phospholipid composition. In some embodiments, the marine phospholipid composition comprises phospholipids having the following structure:
    Figure US20080044487A1-20080221-C00004

    wherein R1 is OH or a fatty acid, R2 is OH or a fatty acid, and R3 is a mixture of H, choline, ethanolamine, inositol and serine, said phospholipid having at least 1% of omega-3 fatty acid moieties at positions R1 and/or R2. In some embodiments, the phospholipid composition comprises from about 20-50% of OH at positions R1 and/or R2. In some embodiments, the phospholipid composition is prepared from natural marine phospholipids isolated from a marine organism. In some embodiments, the composition further comprises a lipid carrier. In some embodiments, the phospholipid composition is enzymatically prepared by reacting lecithin with DHA and EPA in the presence of an enzyme. In some embodiments, the-lecithin is soybean or egg lecithin. In some embodiments, the omega-3 fatty acid moieties are selected from the group of EPA and DHA and combination thereof. In some embodiments, the effective amount of said phospholipid composition comprises from about 300 to about 1000 mg omega-3 fatty acids. In some embodiments, the phospholipid composition is administered orally. In some embodiments, the phospholipid composition is provided in a gel capsule or pill. In some embodiments, the human is a male.
  • In some embodiments, the present invention provides methods for prophylactically treating a subject by administration of a marine phospholipid composition comprising administering a marine phospholipid composition to a subject under conditions such that an undesirable condition is prevented, wherein said undesirable condition is selected from the group consisting of weight gain, infertility, obesity, metabolic syndrome, diabetes type II, mortality in subjects with a high risk of sudden cardiac death, and induction of sustained ventricular tachycardia. In some embodiments, the subject is at risk for developing a condition selected from the group consisting of weight gain, obesity, metabolic syndrome, diabetes type II, mortality in subjects with a high risk of sudden cardiac death, and induction of sustained ventricular tachycardia.
  • In some embodiments, the subject is a human. In some embodiments, the subject is a companion animal. The present invention is not limited to any particular marine phospholipid composition. In some embodiments, the marine phospholipid composition comprises phospholipids having the following structure:
    Figure US20080044487A1-20080221-C00005

    wherein R1 is OH or a fatty acid, R2 is OH or a fatty acid, and R3 is a mixture of H, choline, ethanolamine, inositol and serine, said phospholipid having at least 1% of omega-3 fatty acid moieties at positions R1 and/or R2. In some embodiments, the phospholipid composition comprises from about 20-50% of OH at positions R1 and/or R2. In some embodiments, the phospholipid composition is prepared from natural marine phospholipids isolated from a marine organism. In some embodiments, the composition further comprises a lipid carrier. In some embodiments, the phospholipid composition is enzymatically prepared by reacting lecithin with DHA and EPA in the presence of an enzyme. In some embodiments, the lecithin is soybean or egg lecithin. In some embodiments, the omega-3 fatty acid moieties are selected from the group of EPA and DHA and combination thereof. In some embodiments, the effective amount of said phospholipid composition comprises from about 300 to about 1000 mg omega-3 fatty acids. In some embodiments, the phospholipid composition is administered orally. In some embodiments, the phospholipid composition is provided in a gel capsule or pill.
  • DESCRIPTION OF THE FIGURES
  • FIG. 1. The total amount of EPA consumed during the four-week rat trial (mean±SE).
  • FIG. 2. Relative EPA (20:5) content of plasma (mean±SE; n=6).
  • FIG. 3. Relative 20:5 content of red blood cells (mean±SE; n=5-6).
  • FIG. 4. Relative 20:5 content of monocytes (mean±SE; n=5-6).
  • FIG. 5. Schematic drawing of experimental set-up.
  • FIG. 6. EPA levels in plasma as a function of hours after one bolus intake of a marine phospholipid composition.
  • FIG. 7. EPA levels in plasma as a function of hours after one bolus intake of a marine phospholipid composition.
  • FIG. 8. ARA levels in plasma as a function of hours after one bolus intake of a marine phospholipid composition.
  • DEFINITIONS
  • As used herein, “phospholipid” refers to an organic compound having the following general structure:
    Figure US20080044487A1-20080221-C00006

    wherein R1 is a fatty acid residue or —OH, R2 is a fatty acid residue or —OH, and R3 is a —H or a nitrogen containing compound such as choline (HOCH2CH2N+(CH3)3OH), ethanolamine (HOCH2CH2NH2), inositol or serine. R1 and R2 cannot simultaneously be OH. When R3 is an —OH, the compound is a diacylglycerophosphate, while when R3 is a nitrogen-containing compound, the compound is a phosphatide such as lecithin, cephalin, phosphatidyl serine or plasmalogen.
  • The R1 site is herein referred to as position 1 of the phospholipid, the R2 site is herein referred to as position 2 of the phospholipid, and the R3 site is herein referred to as position 3 of the phospholipid.
  • As used herein, the term omega-3 fatty acid refers to polyunsaturated fatty acids that have the final double bond in the hydrocarbon chain between the third and fourth carbon atoms from the methyl end of the molecule. Non-limiting examples of omega-3 fatty acids include, 5,8,11,14,17-eicosapentaenoic acid (EPA), 4,7,10,13,16,19-docosahexanoic acid (DHA) and 7,10,13,16,19-docosapentanoic acid (DPA).
  • As used herein, the term “bioavailability” refers to the degree and rate at which a substance (as a drug) is absorbed into a living system or is made available at the site of physiological activity.
  • As used herein, the term “functional food” refers to a food product to which a biologically active supplement has been added.
  • As used herein, the term “fish oil” refers to any oil obtained from a marine source e.g. tuna oil, seal oil and algae oil.
  • As used herein, the term “lipase” refers to any enzyme capable of hydrolyzing fatty acid esters
  • As used herein, the term “food supplement” refers to a food product formulated as a dietary or nutritional supplement to be used as part of a diet.
  • As used herein, the term “acylation” means fatty acids attached to the phospholipid. 100% acylation means that there are no lyso- or glycerol-phospholipids.
  • DETAILED DESCRIPTION OF THE INVENTION
  • This invention discloses that the uptake/absorption of omega-3 fatty acids attached to phospholipids are dependent on the level of LPL and GPL. Preferably, in order to ensure maximum uptake the level of LPL should be in the range of 15-45% and the level of GPL should be 0%. Furthermore, this invention discloses that the pure PC transesterified with EPA/DHA have a different effect on gene expression in the liver than 40% PC transesterified with EPA/DHA. It is disclosed that the two compositions regulated around 40 genes differently. Furthermore, the invention discloses that the EPA/DHA ratio is important. The treatment containing a EPA/DHA ratio of 2:1 regulated key enzymes involved in the inflammatory response (NF-κB) in a positive way, the treatment containing a EPA/DHA ratio of 1:1 did not.
  • The present invention describes novel marine lipid compositions comprising an omega-3 containing phospholipid and a triacylglyceride (TG) in a ratio from about 1:10 to 10:1. Preferably the ratio is in the range of from about 3:1 to 1:3, more preferably the ratio is in the range of about 1:2 to 2:1. Preferably, the TG is a fish oil such as tuna oil, herring oil, menhaden oil, cod liver oil and algae oil. However, this invention is not limited to omega-3 containing oils as other TG sources are contemplated such as vegetable oils. The phospholipids in the composition have the following structure:
    Figure US20080044487A1-20080221-C00007

    wherein R1 is OH or a fatty acid, R2 is OH or a fatty acid, and R3 is a mixture of H, choline, ethanolamine, inositol and serine. Attached to position 1 or position 2 are least 1% omega-3 fatty acids, preferably at least 5%, more preferably at least 10% omega-3 fatty acids, up to about 15%, 20%, 30%, 40%, 50%, or 60% omega-3 fatty acids. The omega-3 fatty acids can be EPA, DHA, DPA or C18:3 (n-3), most preferably the omega-3 fatty acids are EPA and DHA. The phospholipid composition preferably contains OH in position 1 or position 2 in a range of 25% to 50% in order to maximize absorption in-vivo.
  • In some embodiments, the present invention provides bioavailable and bioefficient omega-3 fatty acids. This invention shows that the novel marine lipid composition disclosed above enhances the uptake of the omega-3 fatty acid in vivo and incorporates omega-3 fatty acids more efficiently into tissues of adult rats than pure fish oil does. An embodiment of the invention is to use the marine lipid composition for efficient increase of omega-3 fatty acids in the liver, brain, adipose tissue, plasma, testicles and heart. Furthermore, this invention also discloses that the marine lipid compositions efficiently reduced the concentration of the pro-inflammatory precursor AA in total lipids and in phospholipids in tissues. It is disclosed that the concentration of AA in the different lipid pools in the liver, brain, adipose tissue, plasma, testicles and heart can be more efficiently reduced than using fish oil. Hence, the composition can be used to improve the EPA/AA ratio, which is a bio-marker of silent inflammation. The invention also discloses that the incorporation of the omega-3 fatty acids into monocytes is also more efficient using the claimed marine lipid composition as opposed to the fish oil. Yet another embodiment of the invention is to use the marine lipid composition to reduce chronic and acute inflammation in humans and in animals. Acute inflammation is mediated by granulocytes or polymorphonuclear leukocytes, while chronic inflammation is mediated by mononuclear cells such as monocytes. Monocytes protect against blood-borne pathogens and moves quickly to sites of infection in the tissues, secreting large amounts of pro-inflammatory prostaglandins. Furthermore, low grade chronic inflammation may be the underlying cause of many life-style related diseases such as obesity, arthritis, diabetes type II, metabolic syndrome, Alzheimer's disease, osteoarthritis, inflammatory bowel disease, allergy and asthma [14]. Hence, the marine lipid composition can be used to treat and prevent diseases linked to chronic inflammation. This invention discloses that the inflammatory response of monocytes harvested from animals in lower in animals treated with the marine lipid composition compared to fish oil. The concentrations of the pro-inflammatory cytokines such as interleukin-1β, interleukin-6 as well as tumor necrosis factor α (TNF-α) were reduced for the group fed the marine lipid composition compared to fish oil. These cytokines are important markers of real inflammation as for examples I1-1β induces fever. I1-6 also induces fever in addition to being linked to the acute phase response. TNF-α is involved in systemic inflammation as well and is released by white blood cells in the case of damage. It has a range of different biological effects such as increasing insulin resistance, stimulating the acute phase response in the liver and affecting the hypothalamus causing appetite suppression and fever.
  • This invention also discloses that the fatty acid composition of the brain and adipose tissue phospholipids changes after in take of omega-3 fatty acids for 30 days. A significant reduction of the arachidonic acids can be found in the phospholipids in the brain and adipose tissue for the rats given either the EPA- or DHA-rich PL diets (PL 1 and PL 2, respectively). This may affect the inflammatory response in this tissue and thereby have a great impact on cognitive diseases/conditions such as Parkinson's or and Alzheimer's where the inflammatory component is fundamental for the progression of the disease. This invention also discloses that the reduction of ARA is present also in the sn-2 position of the phospholipids in the brain. This is very important as the pro-inflammatory eicosanoids are produced from ARA, which are catalytically hydrolyzed from position 2 on the phospholipid by the action of phospholipase A2. The phospholipase A2 is released after stimuli at the cell wall, it then moves to the nuclear membrane where the hydrolysis of the phospholipid takes place.
  • In adipose tissue, accumulation of EPA and DHA in both total lipids (table 3) and PLs (table 8) is substantial when omega-3 supplements were fed and negligible when the control diet was fed. The increase was more pronounced in total lipids, which mainly consists of triglycerides (99% of fat cell lipid content). This invention demonstrates that omega-3 phospholipids can increase the accumulation of EPA/DHA into adipose tissue. This is important as the adipose tissue can function as a reservoir for these fatty acids. Arachidonic acid concentration in total lipids was higher in omega-3 supplemented animals, showing probably an increase of lipoprotein lipase activity, in agreement with the ability of omega 3 in decreasing plasma TAGs concentration. On the other hand, arachidonic acid levels in adipose tissue PLs were significantly lower in omega-3 supplemented animals than the levels in controls. Peculiar enough, the PL-EPA diet was the most efficient in decreasing arachidonic acid. In addition, the invention discloses that the reduction of ARA is also observed in the sn-2 position of the phospholipids of the omega-3 supplemented animals. This is very important as the pro-inflammatory eicosanoids are produced from ARA, which are catalytically hydrolyzed from position 2 on the phospholipid by the action of phospholipase A2. The phospholipase A2 is released after stimuli at the cell wall, it then moves to the nuclear membrane where the hydrolysis of the phospholipid takes place. The reduction of ARA in position 2 on the phospholipids may affect the inflammatory response in this tissue, which may have practical application in different pathologies of the adipose tissue and in its physiological activity of accumulation and release of fatty acids.
  • Fatty acid data from brain are well in line with the data from adipose tissue. Also in this tissue, we found a significant decrease of arachidonic acid in PLs, but surprisingly, only with PL-EPA and PL-DHA (table 7). On the other hand, DHA levels in both total lipids and PLs were not influenced by the omega-3 diets, while there was a small but significant increase in EPA levels. Lack of increase in DHA levels is likely to be attributable to the fact that the rats in this study were adults and pass the stage in development where they incorporate DHA in the brain (mainly PE). On the other hand, EPA being present at low concentration has more margin to increase. Furthermore, this may affect the inflammatory response in this tissue, which may have a great impact in such diseases as Parkinson's and Alzheimer's where the inflammatory component is fundamental for the progression of the disease. Positional distribution of arachidonic acid show that the ARA content is reduced for the EPA-PL groups, as stated before this is very important as it influences the pro-inflammatory eicosanoid production.
  • In liver, as expected, we found for all omega-3 groups a significant increase of EPA and DHA and decrease of arachidonic acid. No great differences were expected between total lipids and PLs because about 80% of liver total lipids are PLs (table 4, 9 and 14).
  • Heart total lipids and PLs (table 6 and table 11, respectively) showed a strong increase of EPA and DHA with a concomitant decrease of arachidonic acid when omega-3 supplements were fed. The strong decrease in the omega-6/omega-3 ratio in heart lipids is important considering the possible impact on the anti-inflammatory potential. Observed change in heart tissue fatty acids (increase of fatty acids with 6 or 5 double bonds) also suggests a possible increase in membrane fluidity. This change was most striking in the PL-DHA group where the increase of DHA was significantly higher than the increase in the TG-oil and PL-EPA groups. The fluidity of myocardium cell membrane seems to play an important role in controlling arrhythmia. Ventricular arrhythmia, is one of the main causes of sudden cardiac death. Furthermore, atrial fibrillation is another pathological state with a high incidence and important health consequences.
  • Testicular long chain PUFAs are of special interest because there is a high rate of production of prostaglandins from the omega-6 PUFA (arachidonic acid mainly) into the semen or seminal fluid. High rate of prostaglandin production does not indicate an active inflammatory process but a stimulus for the uterus smooth muscle to favor male fertility. An omega-3 induced decrease of arachidonic acid as observed in other tissue could be detrimental to the male fertility if it occurred also in testis. Furthermore, testicular tissue has also a high level of DPA (22:5 omega-6), which may serve as a reservoir for arachidonic acid. Arachidonic acid could be formed according to the need, through the retroconversion mechanism in the peroxisomes. A similar mechanism may take place with DHA to form EPA in other tissues. Our data (table 5) show an increase of EPA and DHA and a small decrease of arachidonic acid in the total lipids fraction when omega-3 fatty acids are fed. However, there is no change in arachidonic acid levels in PL when TG-oil and PL-EPA are fed and interestingly a significant increase in the PL-DHA group. Furthermore, DPA n-6 concentration in total lipids was not influenced by omega-3 supplementation but there was a significant increase in DPA in the PL-EPA group (table 5). Overall, these data seem to indicate that the diets with omega-3 did not change the arachidonic and DPA n-6 concentrations in a way that would predict negative effects on male fertility. In contrast, increase in arachidonic acid content of testicular PLs (table 10) when PL-DHA was fed and increase in DPA when PL-EPA was fed could be interpreted to be positively associated with male fertility.
  • Another embodiment of the invention is to formulate the marine lipid compositions into a feed product for the purpose of reducing low-grade chronic inflammation in animals. It can also be formulated into a food product and given to humans for the same purpose. Furthermore, it can be formulated as a functional food product, as a drug or as food supplement.
  • In some embodiments, the compositions of this invention are contained in acceptable excipients and/or carriers for oral consumption. The actual form of the carrier, and thus, the compositions itself, is not critical. The carrier may be a liquid, gel, gelcap, capsule, powder, solid tablet (coated or non-coated), tea, or the like. The composition is preferably in the form of a tablet or capsule and most preferably in the form of a hard gelatin capsule. Suitable excipient and/or carriers include maltodextrin, calcium carbonate, dicalcium phosphate, tricalcium phosphate, microcrystalline cellulose, dextrose, rice flour, magnesium stearate, stearic acid, croscarmellose sodium, sodium starch glycolate, crospovidone, sucrose, vegetable gums, lactose, methylcellulose, povidone, carboxymethylcellulose, corn starch, and the like (including mixtures thereof). Preferred carriers include calcium carbonate, magnesium stearate, maltodextrin, and mixtures thereof. The various ingredients and the excipient and/or carrier are mixed and formed into the desired form using conventional techniques. The tablet or capsule of the present invention may be coated with an enteric coating that dissolves at a pH of about 6.0 to 7.0. A suitable enteric coating that dissolves in the small intestine but not in the stomach is cellulose acetate phthalate. Further details on techniques for formulation for and administration may be found in the latest edition of Remington's Pharmaceutical Sciences (Maack Publishing Co., Easton, Pa.).
  • In other embodiments, the composition contains no traces of organic solvents which is an important property regarding the safety of consuming such compounds. Phospholipids prepared using chemical or enzymatic methods in the presence of organic solvents may contain residual solvents that may be a health hazard. VOC are often co-extracted when marine phospholipids are extracted, such VOC's may contribute to the smell taste of the phospholipids.
  • In other embodiments, the supplement is provided as a powder or liquid suitable for adding by the consumer to a food or beverage. For example, in some embodiments, the dietary supplement can be administered to an individual in the form of a powder, for instance to be used by mixing into a beverage, or by stirring into a semi-solid food such as a pudding, topping, sauce, puree, cooked cereal, or salad dressing, for instance, or by otherwise adding to a food.
  • The compositions of the present invention may also be formulated with a number of other compounds. These compounds and substances add to the palatability or sensory perception of the particles (e.g., flavorings and colorings) or improve the nutritional value of the particles (e.g., minerals, vitamins, phytonutrients, antioxidants, etc.).
  • The dietary supplement may comprise one or more inert ingredients, especially if it is desirable to limit the number of calories added to the diet by the dietary supplement. For example, the dietary supplement of the present invention may also contain optional ingredients including, for example, herbs, vitamins, minerals, enhancers, colorants, sweeteners, flavorants, inert ingredients, and the like. For example, the dietary supplement of the present invention may contain one or more of the following: ascorbates (ascorbic acid, mineral ascorbate salts, rose hips, acerola, and the like), dehydroepiandosterone (DHEA), Fo-Ti or Ho Shu Wu (herb common to traditional Asian treatments), Cat's Claw (ancient herbal ingredient), green tea (polyphenols), inositol, kelp, dulse, bioflavinoids, maltodextrin, nettles, niacin, niacinamide, rosemary, selenium, silica (silicon dioxide, silica gel, horsetail, shavegrass, and the like), spirulina, zinc, and the like. Such optional ingredients may be either naturally occurring or concentrated forms.
  • In some embodiments, the dietary supplements further comprise vitamins and minerals including, but not limited to, calcium phosphate or acetate, tribasic; potassium phosphate, dibasic; magnesium sulfate or oxide; salt (sodium chloride); potassium chloride or acetate; ascorbic acid; ferric orthophosphate; niacinamide; zinc sulfate or oxide; calcium pantothenate; copper gluconate; riboflavin; beta-carotene; pyridoxine hydrochloride; thiamin mononitrate; folic acid; biotin; chromium chloride or picolonate; potassium iodide; sodium selenate; sodium molybdate; phylloquinone; vitamin D3; cyanocobalamin; sodium selenite; copper sulfate; vitamin A; vitamin C; inositol; potassium iodide. Suitable dosages for vitamins and minerals may be obtained, for example, by consulting the U.S. RDA guidelines.
  • In further embodiments, the compositions comprise at least one food flavoring such as acetaldehyde(ethanal), acetoin(acetyl methylcarbinol), anethole(parapropenyl anisole), benzaldehyde(benzoic aldehyde), N-butyric acid (butanoic acid), d- or l-carvone(carvol), cinnamaldehyde(cinnamic aldehyde), citral(2,6-dimethyloctadien-2,6-al-8, gera-nial, neral), decanal(N-decylaldehyde, capraldehyde, capric aldehyde, caprinaldehyde, aldehyde C-10), ethyl acetate, ethyl butyrate, 3-methyl-3-phenyl glycidic acid ethyl ester(ethyl-methyl-phenyl-glycidate, strawberry aldehyde, C-16 aldehyde), ethyl vanillin, geraniol(3,7-dimethyl-2,6 and 3,6-octadien-1-ol), geranyl acetate (geraniol acetate), limonene (d-, l-, and dl-), linalool (linalol, 3,7-dimethyl-1,6-octadien-3-ol), linalyl acetate(bergamol), methyl anthranilate(methyl-2-aminobenzoate), piperonal(3,4-methylenedioxy-benzaldehyde, heliotropin), vanillin, alfalfa (Medicago sativa L.), allspice (Pimenta officinalis), ambrette seed (Hibiscus abelmoschus), angelic (Angelica archangelica), Angostura (Galipea officinalis), anise (Pimpinella anisum), star anise (Illicium verum), balm (Melissa officinalis), basil (Ocimum basilicum), bay (Laurus nobilis), calendula (Calendula officinalis), (Anthemis nobilis), capsicum (Capsicum frutescens), caraway (Carum carvi), cardamom (Elettaria cardamomum), cassia, (Cinnamomum cassia), cayenne pepper (Capsicum frutescens), Celery seed (Apium graveolens), chervil (Anthriscus cerefolium), chives (Allium schoenoprasum), coriander (Coriandrum sativum), cumin (Cuminum cyminum), elder flowers (Sambucus canadensis), fennel (Foeniculum vulgare), fenugreek (Trigonella foenum-graecum), ginger (Zingiber officinale), horehound (Marrubium vulgare), horseradish (Armoracia lapathifolia), hyssop (Hyssopus officinalis), lavender (Lavandula officinalis), mace (Myristica fragrans), maroram (Majorana hortensis), mustard (Brassica nigra, Brassica juncea, Brassica hirta), nutmeg (Myristica fragrans), paprika (Capsicum annuum), black pepper (Piper nigrum), peppermint (Mentha piperita), poppy seed (Papayer somniferum), rosemary (Rosmarinus officinalis), saffron (Crocus sativus), sage (Salvia officinalis), savory (Satureia hortensis, Satureia montana), sesame (Sesamum indicum), spearmint (Mentha spicata), tarragon (Artemisia dracunculus), thyme (Thymus vulgaris, Thymus serpyllum), turmeric (Curcuma longa), vanilla (Vanilla planifolia), zedoary (Curcuma zedoaria), sucrose, glucose, saccharin, sorbitol, mannitol, aspartame. Other suitable flavoring are disclosed in such references as Remington's Pharmaceutical Sciences, 18th Edition, Mack Publishing, p. 1288-1300 (1990), and Furia and Pellanca, Fenaroli's Handbook of Flavor Ingredients, The Chemical Rubber Company, Cleveland, Ohio, (1971), known to those skilled in the art.
  • In other embodiments, the compositions comprise at least one synthetic or natural food coloring (e.g., annatto extract, astaxanthin, beet powder, ultramarine blue, canthaxanthin, caramel, carotenal, beta carotene, carmine, toasted cottonseed flour, ferrous gluconate, ferrous lactate, grape color extract, grape skin extract, iron oxide, fruit juice, vegetable juice, dried algae meal, tagetes meal, carrot oil, corn endosperm oil, paprika, paprika oleoresin, riboflavin, saffron, tumeric, tumeric and oleoresin).
  • In still further embodiments, the compositions comprise at least one phytonutrient (e.g., soy isoflavonoids, oligomeric proanthcyanidins, indol-3-carbinol, sulforaphone, fibrous ligands, plant phytosterols, ferulic acid, anthocyanocides, triterpenes, omega 3/6 fatty acids, conjugated fatty acids such as conjugated linoleic acid and conjugated linolenic acid, polyacetylene, quinones, terpenes, cathechins, gallates, and quercitin). Sources of plant phytonutrients include, but are not limited to, soy lecithin, soy isoflavones, brown rice germ, royal jelly, bee propolis, acerola berry juice powder, Japanese green tea, grape seed extract, grape skin extract, carrot juice, bilberry, flaxseed meal, bee pollen, ginkgo biloba, primrose (evening primrose oil), red clover, burdock root, dandelion, parsley, rose hips, milk thistle, ginger, Siberian ginseng, rosemary, curcumin, garlic, lycopene, grapefruit seed extract, spinach, and broccoli.
  • In still other embodiments, the compositions comprise at least one vitamin (e.g., vitamin A, thiamin (B1), riboflavin (B2), pyridoxine (B6), cyanocobalamin (B12), biotin, ascorbic acid (vitamin C), retinoic acid (vitamin D), vitamin E, folic acid and other folates, vitamin K, niacin, and pantothenic acid). In some embodiments, the particles comprise at least one mineral (e.g., sodium, potassium, magnesium, calcium, phosphorus, chlorine, iron, zinc, manganese, flourine, copper, molybdenum, chromium, selenium, and iodine). In some particularly preferred embodiments, a dosage of a plurality of particles includes vitamins or minerals in the range of the recommended daily allowance (RDA) as specified by the United States Department of Agriculture. In still other embodiments, the particles comprise an amino acid supplement formula in which at least one amino acid is included (e.g., 1-carnitine or tryptophan).
  • Transesterification of phosphatidylcholine (PC) under solvent free conditions has been performed by Haraldsson et al in 1999 [15], with the results of high incorporation of EPA/DHA and with the following hydrolysis profile PC/LPC/GPC=39/44/17. Extensive hydrolysis and by-product formation is generally considered a problem with transesterification reactions, resulting in low product yields. This invention discloses a process for transesterification of crude soybean lecithin (mixture of PC, PE and PI). In the first step, the lecithin is hydrolyzed using a lipase in the presence of water (pH=8). The use of a variety of lipases is contemplated, including, but not limited to, Thermomyces Lanuginosus lipase, Rhizomucor miehei lipase, Candida Antarctica lipase, Pseudomonas fluorescence lipase, and Mucor javanicus lipase. The first step takes around 24 hours and results in a product comprising predominantly of lyso-phospholipids and glycerophospholipids such as PC/LPC/GPC=0/15/85. In the second step, free fatty acids are added such as EPA and DHA, however any omega-3 fatty acid is contemplated. Next a strong vacuum is applied to the reaction vessel for 72 hours. However, the reaction length can be varied in order to obtain a composition with the desired amount of phospholipids and lyso-phospholipids. By extending the reaction time beyond 72 hours, a product comprising more than 65% phospholipids can be obtained. Next, a lipid carrier is added to the reaction mixture in order to reduce the viscosity of the solution. The added amount of triglycerides can be 10%, 20%, 30%, 40% or more, it depends on the requested viscosity of the final product. The lipid carrier can be a fish oil such as tuna oil, menhaden oil and herring oil, or any triglyceride, diglyceride, ethyl- or methylester of a fatty acid. In the final step, the product is subjected to a molecular distillation and the free fatty acids are removed, resulting in a final product comprising of phospholipids (lyso-phospholipids and phospholipids) and triglycerides in a ratio of preferably 2:1.
  • This invention further discloses a process for the enzymatic transesterification/esterification of phospholipids with fatty acids alkyl esters or free fatty acids in an evacuated vessel (B). A reduced pressure is applied to the vessel B (0.001-30 mbar) and water vapor (moisture) is allowed to enter the reaction mixture through a tube from a second vessel (A) (FIG. 5 for schematic drawing of the experimental setup). The water in vessel A is heated to 25-30° C. By adding moisture to an evacuated reaction vessel the rate of reaction could either be increased of the lipase dosage could be reduced. In addition, the reuse of the enzymes was improved. Finally, a novel marine phospholipid composition was prepared characterized by being acylated in the range of 55%-85%, having at least 5% EPA and/or DHA esterified, having a EPA/DHA ratio of at least 1.
  • Accordingly, in preferred embodiments, the present invention utilizes a phospholipid, preferably a phosphatide such as lecithin. The present invention is not limited to the use of any particular phospholipid. Indeed, the use of a variety of phospholipids is contemplated. In some embodiments, the phospholipid is a phosphatidic or lysophosphatidic acid. In more preferred embodiments, the phospholipid is a mixture of phosphatides such as phosphatidylcholine, phosphatidylethnolamine, phosphatidylserine and phosphatidylinositol. The present invention is not limited to the use of any particular source of phospholipids. In some embodiments, the phospholipids are from soybeans, while in other embodiments, the phospholipids are from eggs. In particularly preferred embodiments, the phospholipids utilized are commercially available, such as Alcolec 40P® from American Lecithin Company Inc (Oxford, Conn., USA). The present invention is not limited to the use of any particular enzyme. Indeed, the use of a variety of enzymes is contemplated, including, but not limited to Thermomyces Lanuginosus lipase, Rhizomucor miehei lipase, Candida Antarctica lipase, Pseudomonas fluorescence lipase, and Mucor javanicus lipase. This invention is not limited to any particular fatty acid alkyl ester either. This includes, but not limited to: decanoic acid (10:0), undecanoic acid (11:0), 10-undecanoic acid (11:1), lauric acid (12:0), cis-5-dodecanoic acid (12:1), tridecanoic acid (13:0), myristic acid (14:0), myristoleic acid (cis-9-tetradecenoic acid, 14:1), pentadecanoic acid (15:0), palmitic acid (16:0), palmitoleic acid (cis-9-hexadecenoic acid, 16:1), heptadecanoic acid (17:1), stearic acid (18:0), elaidic acid (trans-9-octadecenoic acid, 18:1), oleic acid (cis-9-octadecanoic acid, 18:1), nonadecanoic acid (19:0), eicosanoic acid (20:0), cis-11-eicosenoic acid (20:1), 11,14-eicosadienoic acid (20:2), heneicosanoic acid (21:0), docosanoic acid (22:0), erucic acid (cis-13-docosenoic acid, 22:1), tricosanoic acid (23:0), tetracosanoic acid (24:0), nervonic acid (24:1), pentacosanoic acid (25:0), hexacosanoic acid (26:0), heptacosanoic acid (27:0), octacosanoic acid (28:0), nonacosanoic acid (29:0), triacosanoic acid (30:0), vaccenic acid (t-11-octadecenoic acid, 18:1), tariric acid (octadec-6-ynoic acid, 18:1), and ricinoleic acid (12-hydroxyoctadec-cis-9-enoic acid, 18:1) and ω3, ω6, and ω9 fatty acyl residues such as 9,12,15-octadecatrienoic acid (α-linolenic acid) [18:3, ω3]; 6,9,12,15-octadecatetraenoic acid (stearidonic acid) [18:4, ω3]; 11,14,17-eicosatrienoic acid (dihomo-α-linolenic acid) [20:3, ω3]; 8,11,14,17-eicosatetraenoic acid [20:4, ω3], 5,8,11,14,17-eicosapentaenoic acid [20:5, ω3]; 7,10,13,16,19-docosapentaenoic acid [22:5, ω3]; 4,7,10,13,16,19-docosahexaenoic acid [22:6, ω3]; 9,12-octadecadienoic acid (linoleic acid) [18:2, ω6]; 6,9,12-octadecatrienoic acid (γ-linolenic acid) [18:3, ω6]; 8,11,14-eicosatrienoic acid (dihomo-γ-linolenic acid) [20:3 ω6]; 5,8,11,14-eicosatetraenoic acid (arachidonic acid) [20:4, ω6]; 7,10,13,16-docosatetraenoic acid [22:4, ω6]; 4,7,10,13,16-docosapentaenoic acid [22:5, ω6]; 6,9-octadecadienoic acid [18:2, ω9]; 8,1 1-eicosadienoic acid [20:2, ω9]; 5,8,1 1-eicosatrienoic acid (Mead acid) [20:3, ω9]; t10,c12 octadecadienoic acid; c10,t12 octadecadienoic acid; c9,t11 octadecadienoic acid; and t9,c11 octadecadienoic acid. Moreover, acyl residues may be conjugated, hydroxylated, epoxidated or hydroxyepoxidated acyl residues.
  • Marine phospholipids extracted from marine sources have a characteristic smell and taste of rancid fish. The GC profile of the volatiles confirms the presence of these degradation products, such as short chain aldehydes and carboxylic acids. In preferred embodiments, the synthetic marine phospholipid compositions of the present invention are substantially free of volatile organic compounds and are therefore much more suitable as a food supplement for humans and animals. Accordingly, in preferred compositions, the present invention provides synthetic marine phospholipids compositions having high or increased palatability, wherein the high or increased palatability is due to low levels of organic solvents and/or volatile organic compounds. In preferred embodiments, palatability is assayed by feeding the composition to a panel of subjects, preferably human. In more preferred embodiments, the phospholipids compositions have high or increased palatability as compared to naturally extracted marine phospholipids. In other preferred embodiments, the synthetic marine phospholipids compositions of the present invention are safe for oral administration.
  • Experimental
  • EXAMPLE 1
  • The difference in bioavailability and bioefficacy between the marine lipid composition of the present invention and a fish oil were investigated in a rat experiment. The rat feed was prepared using AIN-93 except that soybean oil was removed from the feed. The pelleted AIN-93 diet was ground and the marine lipid compositions (PL 1 and PL 2) as well as fish oil (TG oil) and control were added to this ground feed. The marine lipid compositions were prepared using enzymatic (lipase) catalyzed transesterification of soy lecithin with fish oil fatty acids according to the method described in Example 4, followed by the addition of a triglyceride carrier and short path distillation. The concentration of EPA, DHA and 18:3 n-3 in the different diets can be seen in the table below (table 1).
    TABLE 1
    Amount of different fatty acid in the final feed products
    g/100 g g/100 g g/100 g SUM g/100 g
    EPA DHA 18:3n3 EPA + DHA + 18:3n3
    Control T4 0 0 0.26 0.26
    TG Oil T1 0.61 0.39 0.24 1.23
    PL 1 T2 0.61 0.35 0.26 1.22
    PL 2 T3 0.24 0.73 0.26 1.23
  • Thirty six newly weaned male Sprague Dawley rats (start weight 168±11 g) were used in the experiment. The rats were initially given low-essential oil rat feed, containing 20 g of sunflower oil and 10 g of flaxseed oil per kg of feed, for one week. After the first week, modified AIN-93 diet powder without the test oil was given to rats ad libitum until the start of the experiment. Feeding of rats was stopped 12 hours before the sampling, 30 days after the start of feeding. Each rat was individually anesthetized with carbon dioxide, weighed and euthanized with cervical dislocation. Next, blood was sampled and centrifuged to separate plasma and blood cells. Then abdominal skin was removed and 70 ml of sterile Hepes-Hanks was injected into the peritoneal cavity to collect intraperitoneal lymphocytes. The abdomen was gently massaged for about 3 minutes after which the buffer solution was drained and centrifuged in Falcon tubes (200×g, 10 min) to collect the cells. The cells were resuspended into 1 ml of freezing fluid (10% DMSO, 90% fetal bovine serum) in 1.5 ml Eppendorf tubes. These tubes were then frozen to dry ice temperature for one hour by immersing the tubes in isopropanol placed on dry ice. This enabled a slower freezing rate than by putting the cells directly on dry ice. In the laboratory, the cells were stored overnight at −80° C. and then stored in liquid nitrogen. The derivatization of the lipids in order to perform gas chromatographic (GC) analysis was carried accordingly to [16]. The run conditions for the GC were according to [17]. The growth of rats did not differ between the feeding groups (data not shown). The intake of feed, and the intake fatty acids thereof, was monitored by keeping the rats in metabolic cages which allows the measurement of eaten and uneaten portion of feed. The PL 1 test group consumed somewhat less EPA than the TG oil group, whereas the PL 2 and the control group consumed much less EPA than both the PL 1 and the TG oil groups (FIG. 1). The amount of EPA in plasma varied between the groups and the results are shown FIG. 2. Even though the estimated intake of EPA was higher in the TG Oil group than the PL 1 group, the area % of EPA measured in plasma for PL1 was higher than for TG oil. Indicating a higher bioavailability of EPA from of PL 1 than from the TG oil. Furthermore, this was also observed in the FA profile of the red blood cells and the monocytes (FIG. 3 and FIG. 4, respectively). Demonstrating that the PL 1 composition was more efficient in enriching these cells with omega-3 than the TG oil group, hence being more bioefficient than TG.
  • EXAMPLE 2
  • The total fatty acid profile for the lipids in the brains (table 2), adipose tissue (table 3), liver (table 4), testicles (table 5) and heart (table 6) were isolated from the rats in example 1. The PL 1 composition increases the DHA content in brain and adipose tissue more than the TG composition. The PL 1 composition increase the EPA content in the adipose tissue more than the TG composition. It is to be observed that the PL1 composition increases the EPA/DHA content in the phospholipids and in the total lipids of the different tissues as well as reduces the AA/EPA ratio more than the TG oil composition.
    TABLE 2
    Fatty acid profile of the total lipids isolated from the rat brain (mmol/g lipids).
    18:1 18:2 18:3 n3 20:3 ARA EPA 22:4 22:5 n6 22:5 n3 DHA
    TG-oil 370 12 0.35 5.7 153.7 2.2 68.5 5.6 5.0 191.4
    PL-1 399 12 0.33 7.2 160.7 2.2 70.2 7.2 5.7 203.3
    PL-2 345 13 0.33 5.7 141.6 1.7 60.8 5.8 3.4 190.3
    Control 363 12 0.30 5.2 174.9 0.1 81.4 7.6 1.7 189.6
  • TABLE 3
    Fatty acid profile of the total lipids isolated from adipose tissue (mmol/g lipids).
    18:1 18:2 18:3 n3 20:3 ARA EPA 22:4 22:5 n6 22:5 n3 DHA
    TG-oil 350 306 40.2 1.4 6.3 21.7 6.3 0.9 11.3 27.5
    PL-1 500 236 58.2 1.9 7.0 24.0 7.0 1.0 7.2 30.1
    PL-2 420 536 45.7 2.4 6.8 12.8 6.8 2.7 104.0 50.9
    Control 419 113 28.2 0.8 3.8 0.3 3.8 0.6 26.1 0.8
  • TABLE 4
    Fatty acid profile of the total lipids in liver (mmol/g lipids).
    18:1 18:2 18:3 n3 20:3 ARA EPA 22:4 22:5 n6 22:5 n3 DHA
    TG-oil 304.2 514.1 31.9 13.2 278.9 147.8 2.5 2.9 49.9 272.8
    PL-1 264.3 501.6 28.1 12.8 317.3 120.2 2.4 2.6 49.8 259.0
    PL-2 226.6 462.7 19.9 12.3 289.0 80.4 3.0 5.5 30.4 263.1
    Control 359.2 493.7 21.7 8.9 480.9 9.2 12.0 4.5 14.7 145.8
  • TABLE 5
    Fatty acid profile of the total lipids in testicles.
    nmoles
    FA/mg n3 n6 20:3
    lipids 18:4 20:5 18:3 18:3 22:6 16:1 20:4 18:2 22:5 20:3 n9 22:4 18:1
    TG-oil 3.2 17.0 11.7 1.8 56.6 47.6 280.1 195.1 6.9 285.6 3.7 56.2 326.8
    PL-1 0.6 10.8 9.2 1.7 52.4 35.6 294.9 210.1 6.1 294.2 3.4 61.3 348.3
    PL-2 2.4 6.1 1.7 2.0 56.0 0.0 310.8 156.5 2.4 345.3 6.2 59.5 332.5
    Control 1.8 1.1 1.6 2.0 27.2 0.0 335.5 162.2 1.3 319.1 5.8 78.7 330.1
  • TABLE 6
    Fatty acid profile of the total lipids in heart.
    nmoles
    FA/mg n3 n6 20:3
    lipids 18:4 20:5 18:3 18:3 22:6 16:1 20:4 18:2 22:5 20:3 n9 22:4 18:1
    TG-oil 2.0 44.7 20.2 0.9 314.0 49.7 279.6 787.9 4.0 6.8 3.0 288.9
    PL-1 1.5 45.0 25.6 0.8 314.4 44.6 300.4 789.0 4.4 7.2 3.3 276.6
    PL-2 0.2 29.1 9.9 0.4 372.1 30.1 291.0 690.3 9.0 6.3 1.5 4.4 237.0
    Control 0.0 2.9 7.2 0.7 209.9 495.8 756.2 14.7 6.2 2.4 30.3 289.5
  • EXAMPLE 3
  • The fatty acid profile of the phospholipids isolated from the brain (table 7), adipose tissue (table 8), liver (table 9), testicles (table 10) and heart (Table 11) in the rats from example 1 were determined.
    TABLE 7
    Fatty acid profile of the phospholipids isolated from the brain (mmol/g lipids).
    18:1 18:2 18:3 n3 20:3 ARA EPA 22:4 22:5 n6 22:5 n3 DHA
    TG-oil 260.8 9.9 0.2 4.5 147.17 1.9 72.8 5.6 5.3 196.8
    PL-1 289.9 10.9 0.1 4.2 115.07 1.4 54.7 4.3 4.5 170.1
    PL-2 232.6 2.1 0.6 4.2 122.38 1.2 57.4 4.4 0.7 175.9
    Control 288.8 4.4 3.5 181.60 0.1 86.8 7.3 0.5 213.1
  • TABLE 8
    Fatty acid profile of the phospholipids isolated from
    the adipose tissue (mmol/g lipids).
    18:1 18:2 18:3 n3 20:3 ARA EPA 22:4 22:5 n6 22:5 n3 DHA
    TG-oil 0.7 0.9 0.04 0.04 0.45 0.10 0.0 0.0 0.0 0.17
    PL-1 0.6 0.5 0.04 0.02 0.24 0.06 0.0 0.0 0.0 0.09
    PL-2 0.5 0.6 0.03 0.04 0.37 0.05 0.0 0.0 0.0 0.13
    Control 0.5 0.4 0.04 0.03 0.57 0.01 0.0 0.0 0.0 0.04
  • TABLE 9
    Fatty acid profile of the phospholipids isolated from the liver (mmol/g lipids).
    18:1 18:2 18:3 n3 20:3 ARA EPA 22:4 22:5 n6 22:5 n3 DHA
    TG-oil 84.2 165.4 4.4 11.5 256.9 61.6 1.7 1.69 20.5 186.5
    PL-1 107.2 227.6 4.6 11.4 287.3 67.9 2.0 1.62 29.1 207.7
    PL-2 117.0 347.7 6.6 12.3 314.7 51.7 2.7 5.12 26.0 247.7
    Control 86.7 204.4 1.6 7.6 393.6 2.8 8.0 3.41 11.0 125.9
  • TABLE 10
    Fatty acid profile of the phospholipids isolated from the testicles
    nmoles FA/mg
    lipids 20:5 22:6 20:4 18:2 22:5 n3 22:5 n6 22:4 18:1
    TG-oil 3.18 23.00 173.10 55.30 1.55 234.24 20.50 118.11
    PL-1 3.71 31.41 227.44 82.56 2.05 317.90 28.27 143.41
    PL-2 3.339 46.79 335.87 127.50 2.04 265.36 45.48 155.71
    Control 0.430 14.08 204.27 49.39 0.36 161.20 35.70 118.18
  • TABLE 11
    Fatty acid profile of the phospholipids isolated from the heart.
    nmoles 20:3
    FA/mg lipids 20:5 n3 18:3 22:6 20:4 18:2 22:5 20:3 n9 22:4 18:1
    TG-oil 35.0 6.4 286.3 240.3 877.5 4.2 5.1 3.2 119.6
    PL-1 27.5 6.0 261.6 205.1 754.6 3.4 4.2 2.6 96.3
    PL-2 20.8 4.1 331.1 227.6 570.3 7.9 5.0 1.2 4.1 115.7
    Control 1.6 3.1 187.1 348.8 505.9 13.7 3.2 1.9 22.0 128.6
  • TABLE 12
    Fatty acid profile of the sn-2 position on the phospholipids isolated from
    the adipose tissue
    sn-2 EPA n3 18:3 DHA ARA 18:2
    TG-oil 0.06 0.03 0.09 0.24 0.57
    PL-1 0.09 0.04 0.12 0.29 0.70
    PL-2 0.04 0.11 0.35 0.60
    Control 0.00 0.04 0.45 0.49
  • TABLE 13
    Fatty acid profile of the sn-2 position on the phospholipids isolated
    from the brain
    sn-2 EPA DHA ARA 18:2 22:5 20:3 22:4 18:1
    TG-oil 1.6 164.7 123.6 9.8 5.0 3.9 72.8 197.4
    PL-1 1.3 146.3 96.5 11.6 3.7 4.1 51.8 260.2
    PL-2 1.2 173.5 120.3 3.2 4.4 4.2 57.4 223.4
    Control 0.1 203.0 171.6 4.4 7.1 3.2 81.1 258.4
  • TABLE 14
    Fatty acid profile of the sn-2 position of the phospholipids in the liver.
    n3 n6 20:3
    sn-2 18:4 20:5 18:3 18:3 22:6 16:1 20:4 18:2 22:5n3 22:5 20:3 n9 22:4 18:1 20:2
    TG-oil 0.5 50.2 2.3 0.3 145.3 6.9 188.6 149.5 18.6 0.9 6.4 0.5 0.9 45.1 2.7
    PL-1 0.4 60.1 2.9 0.4 176.7 0.0 235.6 193.0 24.3 1.2 8.3 0.8 1.6 77.9 2.5
    PL-2 0.1 45.8 4.2 0.5 212.4 22.2 264.5 343.7 25.6 3.7 9.6 0.6 2.0 87.0 2.5
    Control 0.0 2.5 1.3 0.4 108.6 31.7 330.9 204.4 11.0 2.7 6.0 1.2 6.4 62.6 3.7
  • TABLE 15
    Fatty acid profile of the sn-2 position of the phospholipids in the testis.
    sn-2 20:5 22:6 20:4 18:2 22:5 22:4 18:1
    TG-oil 2.9 21.8 167.6 60.7 224.4 20.5 109.3
    PL-1 3.4 28.4 210.7 77.2 290.6 28.2 120.3
    PL-2 2.5 41.6 304.7 110.4 244.1 45.4 137.8
    Control 0.4 13.6 202.1 50.9 157.2 35.6 113.7
  • TABLE 16
    Fatty acid profile of the sn-2 position of the phospholipids in the heart
    sn-2 20:5 n3 18:3 22:6 20:4 18:2 22:5 20:3 20:3 n9 22:4 18:1
    TG-oil 28.6 5.3 215.4 164.9 741.7 2.9 3.2 3.2 68.2
    PL-1 22.2 4.3 193.4 131.5 601.8 2.1 2.2 2.5 52.5
    PL-2 17.6 3.4 256.7 160.4 519.5 5.7 3.2 1.2 4.1 70.0
    Control 1.4 1.9 163.7 283.9 467.0 8.4 2.9 1.9 21.9 96.0
  • EXAMPLE 4
  • 50 g of soy lecithin from American Lecithin Company Inc (Oxford, Conn., USA), 40 g of TL-IM lipase from Novozymes (Bagsvaerd, Denmark) and 5 g of water (adjusted to pH=8 using NaOH) were mixed in a reaction vessel at 50° C. for 24 hours. Next, 10 g of free fatty acids containing 10% EPA and 50% DHA from Napro Pharma (Brattvaag, Norway) was added, followed by application of vacuum to the reaction vessel. After 72 hours the reaction was terminated and the phospholipid mixture was analyzed using HPLC and GC. The results showed that the relationship between PC/LPC/GPC was 65/35/0, and that the content of EPA and DHA was around 10% and 12%, respectively. Next, 20 g of sardine oil was added to the reaction mixture which comprised of 18% EPA and 12% DHA (relative GC peak area), followed by molecular distillation. The final product contained around 70% acetone insolubles, around 30% triglycerides and traces of free fatty acids.
  • EXAMPLE 5
  • A marine phospholipid composition containing 8.4% EPA and 1.2% DHA was prepared using a crude soybean lecithin as a starting material according to [19]. A marine oil was added to the phospholipid mixture (30% w/w) so that the total level of EPA was 21.9% and for DHA 9.4%. Furthermore, soy lecithin and lyso-phospholipids prepared according to [20] were added to the mixture in variable amounts so that a range of PC/LPC/GPC ratios could be obtained (Table 17). By using this method, all the treatments (MPL1-MPL5) contained exactly the same amount EPA and DHA. Lipid compositions were consumed as a single bolus by adult Sprague-Dawley rats and the appearance of EPA/DHA in blood was measured at different time points from 1 to 12 hours after ingestion. The concentration of EPA/DHA was determined using GC-FID and reported as area percentage. FIGS. 6 and 7 show that composition MPL2 and MPL3 results in the highest concentration of EPA/DHA in plasma after uptake. Comparing the surface area under each curve it is clear that MPL2 and MPL3 demonstrates a higher bioavailability of EPA/DHA than the other composition MPL1, MPL4 and MPL5.
    TABLE 17
    Hydrolysis profile of the compositions tested
    Treatment MPL1 MPL2 MPL3 MPL4 MPL5
    PC/LPC/GPC 85/15/0 70/30/0 55/45/0 40/60/0 47/37/16
  • EXAMPLE 6
  • Marine phospholipids were prepared using either 40% soy PC (American Lecithin Company Inc, Oxford, Conn., USA) (MPL1) or 96% pure soy PC (Phospholipid GmbH, Köln, Germany) (MPL2) according to a method described by others [4]. Fatty acid content and the level of bi-products are shown in table 18. The MPL treatments consisted of a mixture of phospholipids, lyso-phospholipids and glycerol-phospholipids. Looking only at the PC/LPC/GPC relationship, it was 64/33/2 and 42/40/18 for MPL1 and MPL2, respectively. Finally, all three treatments were emulsified into skimmed milk.
    TABLE 18
    Composition of the phospholipids used in example 2
    PC/LPC/ 18:2
    Composition GPC (n − 6) 18:3 (n − 3) EPA DHA
    MPL1 64/33/2  129 mg/g 9 mg/g 51 mg/g 171 mg/g
    MPL2 42/40/18 124 mg/g 9 mg/g 96 mg/g  96 mg/g
  • 18 newly weaned Sprague-Dawley rats were fed the milk emulsions for 1 week. Each rat was placed in its own cage to ensure that they got an even amount of test substance and the milk was consumed by the rat pups ad libitum. After 1 week the experiment was terminated and the rats were decapitated. The animals were kept without food for 24 hours before sampling. Entire livers were collected and frozen immediately using liquid nitrogen (stored at −65° C.). Total RNA was isolated from the liver samples according to the Quiagen Rnaesy Midi Kit Protocol. The RNA samples were quantified and quality measured by NanoDrop and Bioanalyzer. The isolated RNA was hybridized onto a gene chip RAE230 2.0 from Affymetrix (Santa Clara, Calif., USA). The expression level of each gene was measured using an Affymetrix GeneChip 3000 7G scanner. The results were suitable for all chips except 2 and they were excluded from the trial. Using statistical tools a list of genes expressed differentially between MPL1 versus MPL2 (Table 3) was obtained. The results are based on (log) probe set summary expression measures, computed by RMA, and linear models are fitted using Empirical Bayes methods for borrowing strength across genes (using the Limma package in R). The p-value are adjusted for multiple testing using the Benjamini-Hochberg-method, controlling the False Discovery Rate (FDR), where FDR=the proportion of null-hypotheses of no DE that are falsely rejected.
  • It was observed that MPL1 and MPL2 are biologically different compounds due to the fact that over 40 genes were differentially expressed (table 19).
    TABLE 19
    List of genes differentially expressed (DE) by MPL1 versus MPL2. The list is
    sorted according to increasing p-values. SLR: Estimated signal log-ratio (<0: down regulated
    gene, >0: up regulated gene). Fold change: Estimated fold change corresponding to the
    parameter (<1: down regulated gene, >1: up regulated gene). Affy fold change: Estimated fold
    change using the Affymetrix definition (<−1: down regulated gene, >1: up regulated gene) df:
    Degrees of freedom (= number of arrays − number of estimated parameters).
    Fold Affy
    Gene-name t p-value FDR SLR change Fold.change df
    Wiskott-Aldrich syndrome-like 8.35 0.00000 0.00261 1.02 2.02 2.02 14
    Similar to osteoclast inhibitory lectin 7.40 0.00000 0.00392 1.49 2.80 2.80 14
    neuron-glia-CAM-related cell adhesion −7.14 0.00000 0.00392 −0.55 0.68 −1.46 14
    molecule
    dehydrodolichyl diphosphate synthase 7.09 0.00000 0.00392 0.57 1.49 1.49 14
    (predicted)
    casein kinase II, alpha 1 polypeptide 7.05 0.00000 0.00392 1.47 2.77 2.77 14
    similar to cisplatin resistance-associated 7.03 0.00000 0.00392 0.94 1.92 1.92 14
    overexpressed protein (predicted)
    similar to Retinoblastoma-binding protein 2 7.01 0.00000 0.00392 0.67 1.59 1.59 14
    (RBBP-2)
    serine/threonine kinase 25 (STE20 homolog, 6.90 0.00000 0.00392 0.43 1.35 1.35 14
    yeast)
    Delangin (predicted) 6.89 0.00000 0.00392 0.69 1.61 1.61 14
    similar to Hypothetical protein MGC30714 −6.80 0.00000 0.00401 −0.40 0.76 −1.32 14
    myeloid/lymphoid or mixed-lineage leukemia 5 6.77 0.00000 0.00401 0.68 1.60 1.60 14
    (trithorax homolog, Drosophila) (predicted)
    Radixin 6.76 0.00000 0.00401 0.74 1.67 1.67 14
    similar to myocyte enhancer factor 2C 6.70 0.00000 0.00420 0.60 1.52 1.52 14
    WD repeat and FYVE domain containing 1 6.64 0.00000 0.00443 0.80 1.74 1.74 14
    (predicted)
    similar to Retinoblastoma-binding protein 2 6.55 0.00000 0.00509 0.87 1.82 1.82 14
    (RBBP-2)
    zinc and ring finger 1 (predicted) −6.50 0.00000 0.00528 −0.51 0.70 −1.42 14
    pumilio 1 (Drosophila) (predicted) 6.45 0.00000 0.00535 0.59 1.51 1.51 14
    leukocyte receptor cluster (LRC) member 8 6.44 0.00000 0.00535 1.06 2.08 2.08 14
    (predicted)
    Similar to collapsin response mediator protein- −6.40 0.00000 0.00542 −0.52 0.70 −1.43 14
    2A
    SWI/SNF related, matrix associated, actin 6.37 0.00000 0.00542 0.74 1.67 1.67 14
    dependent regulator of chromatin, subfamily a,
    member 4
    B-cell CLL/lymphoma 7C (predicted) 6.37 0.00000 0.00542 0.51 1.42 1.42 14
    synaptic vesicle glycoprotein 2b 6.33 0.00000 0.00568 0.97 1.96 1.96 14
    leptin receptor overlapping transcript −6.31 0.00000 0.00570 −0.58 0.67 −1.50 14
    Transcribed locus 6.22 0.00001 0.00658 0.44 1.36 1.36 14
    similar to mKIAA1321 protein 6.20 0.00001 0.00658 0.99 1.99 1.99 14
    retinol binding protein 2, cellular 6.18 0.00001 0.00660 0.59 1.51 1.51 14
    similar to Safb2 protein 6.16 0.00001 0.00660 0.62 1.53 1.53 14
    similar to Zbtb20 protein 6.15 0.00001 0.00660 0.49 1.40 1.40 14
    phosphofructokinase, liver, B-type 6.13 0.00001 0.00665 0.67 1.60 1.60 14
    Transcription elongation factor B (SIII), 6.10 0.00001 0.00668 0.43 1.35 1.35 14
    polypeptide 3
    Echinoderm microtubule associated protein like 6.10 0.00001 0.00668 1.00 2.00 2.00 14
    4 (predicted)
    DNA topoisomerase I, mitochondrial 6.08 0.00001 0.00678 0.61 1.53 1.53 14
    ectonucleoside triphosphate diphosphohydrolase 5 5.96 0.00001 0.00849 0.74 1.67 1.67 14
    nuclear factor I/X 5.94 0.00001 0.00859 0.65 1.57 1.57 14
    WW domain binding protein 4 5.90 0.00001 0.00914 0.58 1.50 1.50 14
    Acetyl-coenzyme A acetyltransferase 1 5.84 0.00001 0.00967 0.69 1.61 1.61 14
    similar to THO complex 2 5.84 0.00001 0.00967 0.74 1.67 1.67 14
  • MPL2 regulates 401 genes versus the control (table 20). A number of genes listed are involved maintenance of the cell, in transcription and protein synthesis as well as signaling pathways. Others are involved in regulation of metabolism and the inflammatory response such as Tnf receptor-associated factor 6 (Traf6_predicted) (fold change of 0.53), guanine nucleotide binding protein alpha inhibiting 2 (Gnai2) (fold change of 0.6, gamma-butyrobetaine hydroxylase (Bbox1) (fold change of 1.32), monoglyceride lipase (Mg11) (fold change 0.52), nuclear NF-kappaB activating protein (fold change 0.65) and CCAAT/enhancer binding protein (C/EBP) (fold change of 0.66). The data listed in table 4 show that a omega-3 rich phospholipid with an EPA/DHA ratio of 2:1 behaves differently compared to placebo. A phospholipid composition with an EPA/DHA ratio o 1:1 did not show any difference versus placebo on gene expression. Consequently, the EPA/DHA ratio is important and should preferably be 2:1.
    TABLE 20
    List of genes differentially expressed (DE) by MPL2 versus control. SLR:
    Estimated signal log-ratio (<0: down regulated gene, >0: up regulated gene). Fold change:
    Estimated fold change corresponding to the parameter (<1: down regulated gene, >1: up
    regulated gene). Affy fold change: Estimated fold change using the Affymetrix definition (<−1: down
    regulated gene, >1: up regulated gene) df: Degrees of freedom (= number of arrays −
    number of estimated parameters)
    Fold Affy
    Gene-ID Gene name t p-value FDR SLR change Fold.change df
    1367588_a_at ribosomal protein L13A −5.22 0.00005 0.00568 −0.44 0.74 −1.36 14
    1367844_at guanine nucleotide binding −5.47 0.00003 0.00417 −0.54 0.69 −1.46 14
    protein, alpha inhibiting 2
    1367958_at abl-interactor 1 −6.42 0.00000 0.00119 −0.69 0.62 −1.62 14
    1367971_at protein tyrosine phosphatase −6.01 0.00001 0.00190 −0.36 0.78 −1.28 14
    4a2
    1368057_at ATP-binding cassette, sub- −5.06 0.00007 0.00688 −0.54 0.69 −1.45 14
    family D (ALD), member 3
    1368405_at v-ral simian leukemia viral −4.86 0.00011 0.00913 −0.44 0.74 −1.36 14
    oncogene homolog A (ras
    related)
    1368646_at mitogen-activated protein 4.98 0.00008 0.00772 0.70 1.62 1.62 14
    kinase 9
    1368649_at dyskeratosis congenita 1, −6.73 0.00000 0.00080 −0.53 0.69 −1.44 14
    dyskerin
    1368662_at ring finger protein 39 −7.01 0.00000 0.00053 −0.61 0.66 −1.52 14
    1368703_at enigma homolog −5.38 0.00003 0.00450 −0.76 0.59 −1.70 14
    1368824_at caldesmon 1 −7.18 0.00000 0.00043 −1.00 0.50 −2.00 14
    1368841_at transcription factor 4 −4.94 0.00009 0.00828 −0.38 0.77 −1.31 14
    1368867_at GERp95 −7.83 0.00000 0.00019 −0.85 0.56 −1.80 14
    1369094_a_at protein tyrosine phosphatase, −7.22 0.00000 0.00042 −0.97 0.51 −1.96 14
    receptor type, D
    1369127_a_at prostaglandin F receptor 4.85 0.00011 0.00921 0.45 1.37 1.37 14
    1369174_at SMAD, mothers against DPP −5.19 0.00005 0.00581 −0.38 0.77 −1.30 14
    homolog 1 (Drosophila)
    1369227_at Choroidermia 5.04 0.00007 0.00718 0.47 1.39 1.39 14
    1369249_at progressive ankylosis homolog 5.36 0.00004 0.00467 0.48 1.39 1.39 14
    (mouse)
    1369501_at zinc finger protein 260 5.17 0.00005 0.00595 0.41 1.33 1.33 14
    1369517_at pleckstrin homology, Sec7 and 4.93 0.00009 0.00829 0.48 1.40 1.40 14
    coiled/coil domains 1
    1369546_at butyrobetaine (gamma), 2- 4.96 0.00009 0.00811 0.40 1.32 1.32 14
    oxoglutarate dioxygenase 1
    (gamma-butyrobetaine
    hydroxylase)
    1369628_at synaptic vesicle glycoprotein −7.20 0.00000 0.00042 −1.11 0.46 −2.15 14
    2b
    1369689_at N-ethylmaleimide sensitive 6.22 0.00001 0.00155 0.66 1.58 1.58 14
    fusion protein
    1369736_at epithelial membrane protein 1 5.74 0.00002 0.00286 0.62 1.54 1.54 14
    1369775_at nuclear ubiquitous casein −7.56 0.00000 0.00027 −0.79 0.58 −1.73 14
    kinase and cyclin-dependent
    kinase substrate
    1370184_at cofilin 1 −6.07 0.00001 0.00178 −0.38 0.77 −1.30 14
    1370260_at adducin 3 (gamma) −5.50 0.00003 0.00399 −0.76 0.59 −1.70 14
    1370328_at Dickkopf homolog 3 (Xenopus 4.80 0.00012 0.00964 0.59 1.51 1.51 14
    laevis)
    1370717_at AP1 gamma subunit binding 6.00 0.00001 0.00192 0.58 1.50 1.50 14
    protein 1
    1370831_at monoglyceride lipase −5.47 0.00003 0.00414 −0.94 0.52 −1.92 14
    1370901_at similar to hypothetical protein −4.83 0.00012 0.00948 −0.34 0.79 −1.27 14
    MGC36831 (predicted)
    1370946_at nuclear factor I/X −10.64 0.00000 0.00002 −1.17 0.45 −2.25 14
    1370949_at myristoylated alanine rich −7.58 0.00000 0.00026 −1.17 0.44 −2.26 14
    protein kinase C substrate
    1370993_at laminin, gamma 1 6.13 0.00001 0.00171 0.63 1.54 1.54 14
    1371034_at one cut domain, family −5.65 0.00002 0.00327 −1.77 0.29 −3.40 14
    member 1
    1371059_at protein kinase, cAMP- 5.24 0.00005 0.00556 0.48 1.40 1.40 14
    dependent, regulatory, type 2,
    alpha
    1371345_at methyl-CpG binding domain −5.32 0.00004 0.00491 −0.34 0.79 −1.27 14
    protein 3 (predicted)
    1371361_at similar to tensin −7.21 0.00000 0.00042 −0.60 0.66 −1.51 14
    1371394_x_at similar to Ab2-143 −5.11 0.00006 0.00645 −0.63 0.64 −1.55 14
    1371397_at nitric oxide synthase −5.53 0.00002 0.00383 −0.34 0.79 −1.26 14
    interacting protein (predicted)
    1371428_at −5.76 0.00001 0.00276 −0.37 0.77 −1.29 14
    1371430_at dystroglycan 1 −5.46 0.00003 0.00417 −0.62 0.65 −1.53 14
    1371432_at −4.95 0.00009 0.00811 −0.36 0.78 −1.28 14
    1371452_at bone marrow stromal cell- −5.05 0.00007 0.00705 −0.46 0.73 −1.37 14
    derived ubiquitin-like protein
    1371573_at ribosomal protein L36a −5.90 0.00001 0.00221 −0.40 0.76 −1.32 14
    (predicted)
    1371589_at Ubiquitin-Like 5 Protein −5.28 0.00004 0.00518 −0.57 0.68 −1.48 14
    1371590_s_at Ubiquitin-Like 5 Protein −4.94 0.00009 0.00829 −0.39 0.76 −1.31 14
    1371779_at sorting nexin 6 (predicted) 5.64 0.00002 0.00329 0.63 1.55 1.55 14
    1371826_at Transcribed locus −5.58 0.00002 0.00359 −0.48 0.72 −1.39 14
    1371896_at growth arrest and DNA- −6.02 0.00001 0.00189 −0.43 0.74 −1.35 14
    damage-inducible, gamma
    interacting protein 1 (predicted)
    1371918_at CD99 −5.35 0.00004 0.00476 −0.37 0.77 −1.29 14
    1372057_at CDNA clone MGC: 124976 −6.12 0.00001 0.00173 −0.38 0.77 −1.30 14
    IMAGE: 7110947
    1372137_at biogenesis of lysosome-related −6.03 0.00001 0.00187 −0.41 0.75 −1.32 14
    organelles complex-1, subunit
    1 (predicted)
    1372142_at arsA arsenite transporter, ATP- −4.93 0.00009 0.00829 −0.37 0.77 −1.30 14
    binding, homolog 1 (bacterial)
    (predicted)
    1372236_at Similar to Caspase recruitment −4.90 0.00010 0.00871 −0.36 0.78 −1.29 14
    domain protein 4
    1372469_at Transcribed locus −4.84 0.00011 0.00945 −0.36 0.78 −1.28 14
    1372697_at mitochondrial ribosomal −5.70 0.00002 0.00299 −0.58 0.67 −1.49 14
    protein S15
    1373031_at tripartite motif protein 8 −5.13 0.00006 0.00628 −0.44 0.74 −1.36 14
    (predicted)
    1373105_at interleukin 1 receptor-like 1 −5.01 0.00008 0.00742 −0.37 0.77 −1.30 14
    ligand (predicted)
    1373135_at similar to hypothetical protein −5.30 0.00004 0.00503 −0.55 0.68 −1.46 14
    MGC2744
    1373206_at similar to FAD104 (predicted) 6.73 0.00000 0.00080 0.64 1.56 1.56 14
    1373303_at similar to mKIAA3013 protein −5.28 0.00004 0.00514 −0.48 0.72 −1.39 14
    1373347_at DMT1-associated protein −6.18 0.00001 0.00162 −0.73 0.60 −1.66 14
    1373378_at ATP/GTP binding protein 1 5.39 0.00003 0.00449 0.51 1.42 1.42 14
    (predicted)
    1373804_at Forkhead box P1 (predicted) −5.28 0.00004 0.00518 −0.59 0.66 −1.51 14
    1373885_at chromobox homolog 5 −5.94 0.00001 0.00208 −1.04 0.48 −2.06 14
    (Drosophila HP1a) (predicted)
    1374002_at −6.78 0.00000 0.00074 −0.86 0.55 −1.82 14
    1374283_at fetal Alzheimer antigen −7.44 0.00000 0.00032 −0.74 0.60 −1.67 14
    (predicted)
    1374425_at transducin-like enhancer of −4.91 0.00010 0.00849 −0.40 0.76 −1.32 14
    split 1, homolog of Drosophila
    E(spl) (predicted)
    1374509_at Similar to RIKEN cDNA −5.62 0.00002 0.00337 −0.47 0.72 −1.39 14
    1110018O08
    1374511_at 5.60 0.00002 0.00345 0.55 1.47 1.47 14
    1374657_at Transcribed locus −4.88 0.00010 0.00890 −0.34 0.79 −1.27 14
    1374733_at symplekin (predicted) −5.04 0.00007 0.00716 −0.36 0.78 −1.28 14
    1374772_at similar to Chromosome 13 5.18 0.00005 0.00581 0.46 1.38 1.38 14
    open reading frame 21
    1374837_at B-cell CLL/lymphoma 7C −8.92 0.00000 0.00006 −0.71 0.61 −1.63 14
    (predicted)
    1374851_at similar to RIKEN cDNA −4.89 0.00010 0.00879 −0.39 0.76 −1.31 14
    2810405O22 (predicted)
    1374852_at hypothetical LOC362592 −5.20 0.00005 0.00579 −0.37 0.78 −1.29 14
    1375214_at UDP-N-acetyl-alpha-D- −5.31 0.00004 0.00500 −0.58 0.67 −1.50 14
    galactosamine:polypeptide N-
    acetylgalactosaminyltransferase
    2 (predicted)
    1375335_at heat shock 90 kDa protein 1, −5.26 0.00004 0.00538 −0.55 0.68 −1.46 14
    beta
    1375396_at pumilio 1 (Drosophila) −10.05 0.00000 0.00003 −0.92 0.53 −1.89 14
    (predicted)
    1375421_a_at praja 2, RING-H2 motif −6.51 0.00000 0.00102 −0.60 0.66 −1.52 14
    containing
    1375453_at −12.32 0.00000 0.00000 −1.02 0.49 −2.02 14
    1375469_at SWI/SNF related, matrix −7.97 0.00000 0.00017 −0.93 0.53 −1.90 14
    associated, actin dependent
    regulator of chromatin,
    subfamily a, member 4
    1375533_at vestigial like 4 (Drosophila) −5.30 0.00004 0.00505 −0.61 0.66 −1.52 14
    (predicted)
    1375548_at similar to RIKEN cDNA −5.64 0.00002 0.00328 −0.58 0.67 −1.50 14
    4732418C07 (predicted)
    1375621_at −7.05 0.00000 0.00051 −0.96 0.51 −1.95 14
    1375632_at similar to 60S ribosomal −4.85 0.00011 0.00921 −0.29 0.82 −1.22 14
    protein L38
    1375650_at bromodomain containing 4 −6.64 0.00000 0.00088 −0.48 0.71 −1.40 14
    (predicted)
    1375658_at Transcribed locus −5.00 0.00008 0.00756 −0.44 0.74 −1.35 14
    1375696_at interferon (alpha and beta) 4.81 0.00012 0.00958 0.59 1.51 1.51 14
    receptor 1 (predicted)
    1375703_at myeloid/lymphoid or mixed- −10.20 0.00000 0.00003 −1.02 0.49 −2.03 14
    lineage leukemia 5 (trithorax
    homolog, Drosophila)
    (predicted)
    1375706_at −5.01 0.00008 0.00743 −0.49 0.71 −1.40 14
    1375763_at similar to 2700008B19Rik −7.08 0.00000 0.00050 −0.54 0.69 −1.45 14
    protein
    1375958_at −5.13 0.00006 0.00628 −0.65 0.64 −1.57 14
    1376059_at 5.33 0.00004 0.00483 0.35 1.28 1.28 14
    1376256_at WD repeat and FYVE domain −9.16 0.00000 0.00005 −1.10 0.47 −2.15 14
    containing 1 (predicted)
    1376299_at similar to Retinoblastoma- −9.22 0.00000 0.00005 −0.89 0.54 −1.85 14
    binding protein 2 (RBBP-2)
    1376450_at transmembrane protein 5 −6.26 0.00001 0.00147 −0.55 0.68 −1.46 14
    (predicted)
    1376523_at At rich interactive domain 4A −5.53 0.00002 0.00383 −0.77 0.59 −1.70 14
    (Rbp1 like) (predicted)
    1376524_at hypothetical protein Dd25 −6.69 0.00000 0.00082 −0.66 0.63 −1.58 14
    1376532_at similar to FAD104 (predicted) 6.06 0.00001 0.00178 0.56 1.47 1.47 14
    1376728_at Transcribed locus −4.80 0.00012 0.00966 −0.35 0.78 −1.27 14
    1376917_at zinc finger protein 292 −5.21 0.00005 0.00571 −0.66 0.63 −1.58 14
    1376982_at Transcribed locus −5.49 0.00003 0.00405 −0.45 0.73 −1.37 14
    1377105_at −6.97 0.00000 0.00056 −0.89 0.54 −1.85 14
    1377302_a_at methylmalonic aciduria −5.10 0.00006 0.00660 −0.52 0.70 −1.43 14
    (cobalamin deficiency) type A
    (predicted)
    1377524_at similar to CG18661-PA −5.36 0.00003 0.00465 −0.43 0.74 −1.35 14
    (predicted)
    1377663_at ras homolog gene family, −5.00 0.00008 0.00756 −0.87 0.55 −1.82 14
    member E
    1377683_at similar to hypothetical protein −6.63 0.00000 0.00088 −0.56 0.68 −1.47 14
    FLJ13045 (predicted)
    1377728_at LOC499567 −5.45 0.00003 0.00419 −1.03 0.49 −2.04 14
    1377766_at Transcribed locus 4.80 0.00012 0.00964 0.37 1.29 1.29 14
    1377899_at similar to RIKEN cDNA −4.99 0.00008 0.00760 −0.46 0.73 −1.38 14
    2810025M15 (predicted)
    1377906_at DEAH (Asp-Glu-Ala-His) box −4.82 0.00012 0.00950 −0.73 0.60 −1.66 14
    polypeptide 36 (predicted)
    1377914_at serine/arginine repetitive −6.41 0.00000 0.00120 −0.98 0.51 −1.97 14
    matrix 1 (predicted)
    1378155_at similar to KIAA1096 protein −5.68 0.00002 0.00313 −0.89 0.54 −1.86 14
    1378163_at Transcribed locus −4.86 0.00011 0.00913 −0.78 0.58 −1.71 14
    1378170_at Transcribed locus −5.00 0.00008 0.00756 −0.92 0.53 −1.90 14
    1378194_a_at rap2 interacting protein x −4.82 0.00012 0.00950 −0.72 0.61 −1.65 14
    1378361_at chromodomain helicase DNA −7.32 0.00000 0.00039 −0.73 0.60 −1.66 14
    binding protein 7 (predicted)
    1378453_at −4.84 0.00011 0.00938 −0.74 0.60 −1.66 14
    1378504_at Insulin-like growth factor I −5.41 0.00003 0.00440 −0.96 0.51 −1.95 14
    mRNA, 3′ end of mRNA
    1378786_at Transcribed locus, weakly 4.89 0.00010 0.00879 0.33 1.25 1.25 14
    similar to NP_780607.2
    hypothetical protein
    LOC10905 [Mus musculus]
    1379062_at similar to Expressed sequence −6.60 0.00000 0.00090 −1.08 0.47 −2.12 14
    AU019823
    1379073_at Similar to RIKEN cDNA −5.51 0.00003 0.00394 −0.49 0.71 −1.40 14
    2310067G05
    1379101_at DEAH (Asp-Glu-Ala-His) box −5.55 0.00002 0.00375 −0.87 0.55 −1.82 14
    polypeptide 36 (predicted)
    1379112_At At rich interactive domain 4A −5.70 0.00002 0.00299 −0.44 0.74 −1.35 14
    (Rbp1 like) (predicted)
    1379232_at TBC1D12: TBC1 domain −6.98 0.00000 0.00056 −1.40 0.38 −2.63 14
    family, member 12 (predicted)
    1379330_s_at CDNA clone IMAGE: 7316839 −4.80 0.00012 0.00967 −0.36 0.78 −1.28 14
    1379332_at Transcribed locus, strongly −4.88 0.00010 0.00886 −0.61 0.66 −1.52 14
    similar to XP_417265.1
    PREDICTED: similar to F-
    box-WD40 repeat protein 6
    [Gallus gallus]
    1379399_at similar to cDNA sequence −5.37 0.00003 0.00459 −0.42 0.75 −1.34 14
    BC016188 (predicted)
    1379457_at neural precursor cell expressed, −5.39 0.00003 0.00449 −0.56 0.68 −1.48 14
    developmentally down-
    regulated gene 1 (predicted)
    1379469_at similar to transducin (beta)-like −6.23 0.00001 0.00153 −0.91 0.53 −1.88 14
    1 X-linked
    1379485_at eukaryotic translation initiation −7.08 0.00000 0.00050 −1.68 0.31 −3.21 14
    factor 3, subunit 10 (theta)
    (predicted)
    1379571_at plakophilin 4 (predicted) −5.42 0.00000 0.00436 −0.74 0.60 −1.67 14
    1379578_at similar to Zbtb20 protein −8.89 0.00000 0.00006 −0.71 0.61 −1.63 14
    1379662_a_at SNF related kinase 4.93 0.00009 0.00829 0.36 1.29 1.29 14
    1379715_at similar to CG9346-PA −4.93 0.00009 0.00829 −0.71 0.61 −1.63 14
    (predicted)
    1379826_at similar to hypothetical protein −5.95 0.00001 0.00208 −0.62 0.65 −1.54 14
    MGC31967
    1380008_at similar to Neurofilament triplet −5.11 0.00006 0.00645 −0.60 0.66 −1.52 14
    H protein (20 kDa
    neurofilament protein)
    (Neurofilament heavy
    polypeptide) (NF-H)
    (predicted)
    1380060_at DNA topoisomerase I, −5.23 0.00005 0.00566 −0.53 0.69 −1.44 14
    mitochondrial
    1380062_at membrane protein, −6.88 0.00000 0.00065 −0.75 0.59 −1.68 14
    palmitoylated 6 (MAGUK p55
    subfamily member 6)
    (predicted)
    1380166_at Similar to hypothetical protein 5.63 0.00002 0.00333 0.34 1.27 1.27 14
    FLJ12056
    1380371_at delangin (predicted) −9.37 0.00000 0.00005 −0.94 0.52 −1.91 14
    1380446_at myeloid/lymphoid or mixed- −5.00 0.00008 0.00756 −0.62 0.65 −1.54 14
    lineage leukemia (trithorax
    homolog, Drosophila);
    translocated to, 10 (predicted)
    1380503_at hypothetical LOC305452 −6.07 0.00001 0.00178 −0.62 0.65 −1.53 14
    (predicted)
    1380728_at Similar to collapsin response 6.09 0.00001 0.00178 0.49 1.41 1.41 14
    mediator protein-2A
    1381469_a_at PERQ amino acid rich, with −5.49 0.00003 0.00405 −0.51 0.70 −1.43 14
    GYF domain 1 (predicted)
    1381525_at −4.82 0.00012 0.00952 −0.41 0.75 −1.33 14
    1381542_at UBX domain containing 2 −6.15 0.00001 0.00171 −0.83 0.56 −1.78 14
    (predicted)
    1381548_at golgi phosphoprotein 4 −5.81 0.00001 0.00256 −0.69 0.62 −1.61 14
    (predicted)
    1381567_at hypothetical LOC294390 4.97 0.00008 0.00800 0.36 1.29 1.29 14
    (predicted)
    1381764_s_at ring finger protein 126 −5.54 0.00002 0.00382 −0.51 0.70 −1.42 14
    (predicted)
    1381809_at ankyrin repeat domain 11 −5.94 0.00001 0.00209 −1.11 0.46 −2.17 14
    (predicted)
    1381829_at −6.27 0.00000 0.00145 −1.07 0.48 −2.10 14
    1381878_at ubinuclein 1 (predicted) −5.82 0.00001 0.00252 −1.18 0.44 −2.26 14
    1381958_at similar to mKIAA0259 protein −6.90 0.00000 0.00062 −1.27 0.41 −2.42 14
    1382000_at 4.82 0.00012 0.00950 0.41 1.33 1.33 14
    1382009_at Transcribed locus −5.39 0.00003 0.00449 −0.69 0.62 −1.62 14
    1382027_at LOC498010 −6.28 0.00000 0.00144 −0.76 0.59 −1.70 14
    1382056_at similar to splicing factor p54 −8.13 0.00000 0.00016 −0.97 0.51 −1.96 14
    1382109_at nuclear NF-kappaB activating −5.83 0.00001 0.00250 −0.62 0.65 −1.53 14
    protein
    1382155_at 6.37 0.00000 0.00126 0.58 1.50 1.50 14
    1382193_at Transcribed locus −6.07 0.00001 0.00178 −1.42 0.37 −2.67 14
    1382306_at Ariadne ubiquitin-conjugating 6.59 0.00000 0.00090 0.59 1.50 1.50 14
    enzyme E2 binding protein
    homolog 1 (Drosophila)
    (predicted)
    1382307_at protein phosphatase 1, −4.79 0.00013 0.00976 −0.47 0.72 −1.39 14
    regulatory (inhibitor) subunit
    12A
    1382358_at Similar to SRY (sex −5.34 0.00004 0.00482 −0.65 0.64 −1.57 14
    determining region Y)-box 5
    isoform a
    1382372_at Aryl hydrocarbon receptor −5.07 0.00007 0.00680 −0.74 0.60 −1.67 14
    1382430_at similar to KIAA1585 protein −5.62 0.00002 0.00338 −0.58 0.67 −1.50 14
    (predicted)
    1382434_at ectonucleoside triphosphate −5.89 0.00001 0.00229 −0.73 0.60 −1.66 14
    diphosphohydrolase 5
    1382466_at similar to RIKEN cDNA −5.43 0.00003 0.00433 −0.98 0.51 −1.97 14
    6530403A03 (predicted)
    1382551_at similar to Intersectin 2 (SH3 −6.72 0.00000 0.00080 −1.41 0.38 −2.67 14
    domain-containing protein 1B)
    (SH3P18) (SH3P18-like
    WASP associated protein)
    1382558_at transcription factor 3 −6.13 0.00001 0.00171 −0.62 0.65 −1.54 14
    (predicted)
    1382573_at Transcribed locus 5.08 0.00007 0.00677 0.38 1.30 1.30 14
    1382584_at similar to mKIAA1321 protein −7.22 0.00000 0.00042 −1.15 0.45 −2.22 14
    1382620_at ankyrin repeat domain 11 −9.69 0.00000 0.00003 −0.95 0.52 −1.93 14
    (predicted)
    1382797_at similar to 1500019C06Rik −5.02 0.00008 0.00742 −0.47 0.72 −1.39 14
    protein
    1382813_at similar to RIKEN cDNA −5.36 0.00004 0.00467 −0.46 0.73 −1.38 14
    4930444A02 (predicted)
    1382862_at Transcribed locus −6.23 0.00001 0.00153 −1.16 0.45 −2.23 14
    1382904_at similar to hypothetical protein −9.04 0.00000 0.00005 −0.85 0.56 −1.80 14
    DKFZp434K1421 (predicted)
    1382935_at similar to Hypothetical protein −6.54 0.00000 0.00097 −0.64 0.64 −1.56 14
    KIAA0141
    1382939_at translocated promoter region −5.18 0.00005 0.00581 −1.13 0.46 −2.19 14
    (predicted)
    1382957_at similar to cisplatin resistance- −8.04 0.00000 0.00016 −1.08 0.47 −2.11 14
    associated overexpressed
    protein (predicted)
    1382960_at Transcribed locus −5.95 0.00001 0.00208 −0.77 0.59 −1.70 14
    1382972_at Transcribed locus, strongly 5.17 0.00005 0.00595 0.37 1.29 1.29 14
    similar to XP_226713.2
    PREDICTED: similar to Src-
    associated protein SAW
    [Rattus norvegicus]
    1383008_at SMC4 structural maintenance −5.19 0.00005 0.00581 −0.98 0.51 −1.97 14
    of chromosomes 4-like 1
    (yeast) (predicted)
    1383040_a_at −5.46 0.00003 0.00419 −0.47 0.72 −1.38 14
    1383052_a_at −6.54 0.00000 0.00097 −0.62 0.65 −1.53 14
    1383054_at −7.86 0.00000 0.00019 −0.76 0.59 −1.70 14
    1383060_at G kinase anchoring protein 1 −5.82 0.00001 0.00255 −0.44 0.74 −1.35 14
    (predicted)
    1383085_at Similar to Sh3bgrl protein −5.20 0.00005 0.00576 −0.86 0.55 −1.81 14
    1383179_at Similar to hypothetical protein −5.10 0.00006 0.00660 −0.75 0.60 −1.68 14
    HSPC129 (predicted)
    1383184_at zinc and ring finger 1 5.03 0.00007 0.00731 0.39 1.31 1.31 14
    (predicated)
    1383334_at Transcribed locus −5.37 0.00003 0.00461 −0.46 0.73 −1.37 14
    1383455_at glutamyl-prolyl-tRNA −6.80 0.00000 0.00072 −0.72 0.61 −1.65 14
    synthetase (predicted)
    1383535_at ankyrin repeat and SOCS box- 6.24 0.00001 0.00152 0.38 1.30 1.30 14
    containing protein 8 (predicted)
    1383615_a_at similar to HECT domain −6.06 0.00001 0.00178 −1.08 0.47 −2.11 14
    containing 1
    1383687_at −5.40 0.00003 0.00441 −0.43 0.74 −1.34 14
    1383776_at Transcribed locus 6.41 0.00000 0.00120 0.62 1.54 1.54 14
    1383786_at Transcribed locus −5.13 0.00006 0.00628 −0.50 0.71 −1.41 14
    1383825_at radixin −9.20 0.00000 0.00005 −1.01 0.50 −2.01 14
    1383827_at tousled-like kinase 1 −6.09 0.00001 0.00178 −1.25 0.42 −2.37 14
    (predicted)
    1384125_at myeloid/lymphoid or mixed- −5.97 0.00001 0.00202 −0.51 0.70 −1.42 14
    lineage leukemia 5 (trithorax
    homolog, Drosophila)
    (predicted)
    1384131_at ADP-ribosylation factor-like 6 6.10 0.00001 0.00176 0.70 1.63 1.63 14
    interacting protein 2 (predicted)
    1384146_at Similar to CD69 antigen (p60, −5.22 0.00005 0.00568 −1.35 0.39 −2.55 14
    early T-cell activation antigen)
    1384154_at WW domain binding protein 4 −5.33 0.00004 0.00483 −0.52 0.70 −1.43 14
    1384260_at Transcribed locus −6.36 0.00000 0.00126 −0.66 0.63 −1.58 14
    1384263_at ATP-binding cassette, sub- −7.27 0.00000 0.00040 −0.72 0.61 −1.64 14
    family A (ABC1), member 13
    (predicted)
    similar to hypothetical protein
    MGC33214 (predicted)
    1384339_s_at casein kinase II, alpha 1 −8.81 0.00000 0.00006 −1.83 0.28 −3.56 14
    polypeptide
    1384376_at similar to FLJ14281 protein −5.42 0.00003 0.00436 −0.65 0.64 −1.57 14
    1384394_at −7.21 0.00000 0.00042 −0.61 0.65 −1.53 14
    1384609_a_at similar to RIKEN cDNA −6.61 0.00000 0.00090 −0.91 0.53 −1.87 14
    B230380D07 (predicted)
    1384766_a_at similar to PHD finger protein −5.18 0.00005 −0.00581 −0.70 0.61 −1.63 14
    14 isoform 1
    1384791_at UDP-GlcNAc:betaGal beta- −5.08 0.00006 0.00671 −0.75 0.60 −1.68 14
    1,3-N-
    acetylglucosaminyltransferase
    1 (predicted)
    1384792_at formin binding protein 3 −6.70 0.00000 0.00082 −0.97 0.51 −1.96 14
    (predicted)
    1384857_at A kinase (PRKA) anchor −5.62 0.00002 0.00338 −1.05 0.48 −2.07 14
    protein (yotiao) 9
    1385006_at alpha thalassemia/mental −4.94 0.00009 0.00829 −0.45 0.73 −1.37 14
    retardation syndrome X-linked
    homolog (human)
    1385038_at similar to hedgehog-interacting −9.94 0.00000 0.00003 −0.80 0.57 −1.75 14
    protein
    1385076_at −5.78 0.00001 0.00271 −0.57 0.68 −1.48 14
    1385077_at similar to golgi-specific −7.83 0.00000 0.00019 −1.05 0.48 −2.07 14
    brefeldin A-resistance guanine
    nucleotide exchange factor 1
    (predicted)
    1385101_a_at Unknown (protein for −5.53 0.00002 0.00383 −0.97 0.51 −1.96 14
    MGC: 73017)
    1385108_at Transcribed locus −4.83 0.00011 0.00946 −1.27 0.42 −2.40 14
    1385240_at WD repeat domain 33 −4.81 0.00012 0.00958 −0.93 0.52 −1.91 14
    (predicted)
    1385320_at similar to Pdz-containing −5.26 0.00004 0.00530 −0.47 0.72 −1.38 14
    protein
    1385407_at TCDD-inducible poly(ADP- −5.46 0.00003 0.00417 −1.34 0.39 −2.54 14
    ribose) polymerase (predicted)
    1385408_at similar to mKIAA0518 protein −5.86 0.00001 0.00236 −1.31 0.40 −2.49 14
    1385689_at Transcribed locus −4.83 0.00011 0.00948 −0.68 0.62 −1.60 14
    1385852_at CREB binding protein −4.85 0.00011 0.00927 −0.53 0.69 −1.44 14
    hypothetical gene supported by
    NM_133381
    1385931_at hook homolog 3 −7.30 0.00000 0.00040 −1.66 0.32 −3.16 14
    1385999_at YME1-like 1 (S. cerevisiae) −4.80 0.00012 0.00964 −0.63 0.64 −1.55 14
    1386191_a_at Transcribed locus 5.22 0.00005 0.00568 0.46 1.37 1.37 14
    1386641_at Transcribed locus −5.41 0.00003 0.00440 −0.97 0.51 −1.95 14
    1386793_at similar to zinc finger protein 61 −5.28 0.00004 0.00514 −0.59 0.66 −1.51 14
    1387087_at CCAAT/enhancer binding −5.43 0.00003 0.00435 −0.61 0.66 −1.52 14
    protein (C/EBP), beta
    1387306_a_at early growth response 2 4.82 0.00012 0.00950 0.33 1.26 1.26 14
    1387365_at nuclear receptor subfamily 1, −4.86 0.00011 0.00913 −0.35 0.78 −1.27 14
    group H, member 3
    1387415_a_at syntaxin binding protein 5 4.86 0.00011 0.00913 0.40 1.32 1.32 14
    (tomosyn)
    1387458_at ring finger protein 4 7.05 0.00000 0.00051 0.75 1.69 1.69 14
    1387664_at ATPase, H+ transporting, V1 5.55 0.00002 0.00375 0.46 1.38 1.38 14
    subunit B, isoform 2
    1387757_at liver regeneration p-53 related 5.19 0.00005 0.00581 0.51 1.42 1.42 14
    protein
    1387760_a_at one cut domain, family −6.12 0.00001 0.00171 −1.58 0.34 −2.98 14
    member 1
    1387789_at v-ets erythroblastosis virus E26 −6.61 0.00000 0.00090 −0.58 0.67 −1.50 14
    oncogene like (avian)
    1387915_at Ratsg2 −4.79 0.00013 0.00976 −0.33 0.79 −1.26 14
    1387947_at v-maf musculoaponeurotic −5.08 0.00007 0.00674 −0.80 0.57 −1.74 14
    fibrosarcoma oncogene family,
    protein B (avian)
    1388022_a_at dynamin 1-like 4.95 0.00009 0.00811 0.43 1.35 1.35 14
    1388059_a_at solute carrier family 11 5.75 0.00001 0.00280 0.43 1.35 1.35 14
    (proton-coupled divalent metal
    ion transporters), member 2
    1388089_a_at ring finger protein 4 5.73 0.00002 0.00289 0.50 1.41 1.41 14
    1388157_at myristoylated alanine rich −5.76 0.00001 0.00276 −0.51 0.70 −1.43 14
    protein kinase C substrate
    1388196_at NCK-associated protein 1 5.31 0.00004 0.00500 0.48 1.40 1.40 14
    1388251_at protein kinase C, lambda 5.21 0.00005 0.00571 0.55 1.47 1.47 14
    1388313_at ribosomal protein s25 −4.83 0.00011 0.00946 −0.63 0.65 −1.54 14
    1388353_at proliferation-associated 2G4, −6.35 0.00000 0.00127 −0.67 0.63 −1.59 14
    38 kDa
    1388388_at Protein phosphatase 2, −5.32 0.00004 0.00487 −0.41 0.75 −1.33 14
    regulatory subunit B (B56),
    delta isoform (predicted)
    1388396_at serine/threonine kinase 25 −5.49 0.00003 0.00404 −0.34 0.79 −1.27 14
    (STE20 homolog, yeast)
    1388503_at similar to CREBBP/EP300 −6.06 0.00001 0.00178 −0.40 0.76 −1.32 14
    inhibitory protein 1
    1388714_at elongation factor RNA −5.88 0.00001 0.00229 −0.46 0.73 −1.37 14
    polymerase II (predicted)
    1388735_at Similar to keratin associated 4.84 0.00011 0.00945 0.50 1.41 1.41 14
    protein 10-6
    1388752_at BCL2-associated transcription −4.81 0.00012 0.00958 −0.40 0.76 −1.32 14
    factor 1 (predicted)
    1388849_at Protease, serine, 25 (predicted) −5.97 0.00001 0.00202 −0.50 0.71 −1.41 14
    1388888_at Transcribed locus 5.13 0.00006 0.00632 0.40 1.32 1.32 14
    1389268_at similar to DNA polymerase −5.21 0.00005 0.00571 −0.37 0.77 −1.29 14
    lambda
    1389307_at similar to Amyloid beta (A4) −4.91 0.00010 0.00854 −0.49 0.71 −1.40 14
    precursor-like protein 1
    1389419_at Transcribed locus −6.54 0.00000 0.00097 −1.30 0.40 −2.47 14
    1389432_at pre-B-cell leukemia −4.93 0.00009 0.00829 −0.55 0.69 −1.46 14
    transcription factor 2
    1389444_at Transcribed locus −6.84 0.00000 0.00068 −1.06 0.48 −2.09 14
    1389806_at Transcribed locus −7.63 0.00000 0.00026 −0.54 0.69 −1.46 14
    1389868_at similar to RCK −6.34 0.00000 0.00128 −1.47 0.36 −2.76 14
    1389963_at P55 mRNA for p55 protein −5.54 0.00002 0.00378 −0.44 0.74 −1.35 14
    1389986_at LOC499304 −5.39 0.00003 0.00449 −2.57 0.17 −5.93 14
    1389989_at alpha thalassemia/mental −4.93 0.00009 0.00829 −0.54 0.69 −1.45 14
    retardation syndrome X-linked
    homolog (human)
    1389998_at Nuclear receptor subfamily 2, −6.03 0.00001 0.00187 −0.69 0.62 −1.61 14
    group F, member 2
    1390048_at serine/arginine repetitive −5.81 0.00001 0.00256 −1.04 0.49 −2.05 14
    matrix 2 (predicted)
    1390120_a_at ring finger protein 1 −5.70 0.00002 0.00299 −0.36 0.78 −1.28 14
    1390121_at GLIS family zinc finger 2 4.81 0.00012 0.00958 0.43 1.34 1.34 14
    (predicted)
    1390227_at CDNA clone IMAGE: 7300848 −5.91 0.00001 0.00219 −1.03 0.49 −2.04 14
    1390360_a_at similar to Safb2 protein −4.79 0.00013 0.00976 −0.48 0.72 −1.39 14
    1390410_at Transcribed locus −4.79 0.00012 0.00973 −0.49 0.71 −1.40 14
    1390436_at Autophagy 7-like (S. cerevisiae) −7.88 0.00000 0.00019 −1.46 0.36 −2.76 14
    (predicted)
    1390448_at similar to 1110065L07Rik 5.08 0.00007 0.00674 0.32 1.25 1.25 14
    protein (predicted)
    1390454_at 4-nitrophenylphosphatase −5.47 0.00003 0.00415 −0.41 0.75 −1.33 14
    domain and non-neuronal
    SNAP25-like protein homolog
    1 (C. elegans) (predicted)
    1390576_at Transcribed locus −5.10 0.00006 0.00660 −0.67 0.63 −1.59 14
    1390660_at T-box 2 (predicted) 5.01 0.00008 0.00752 0.40 1.32 1.32 14
    1390706_at spectrin beta 2 −5.55 0.00002 0.00376 −0.71 0.61 −1.64 14
    1390739_at similar to zinc finger protein −5.51 0.00003 0.00395 −0.52 0.70 −1.43 14
    609
    similar to zinc finger protein
    609
    1390777_at sterol-C5-desaturase (fungal −6.59 0.00000 0.00090 −0.70 0.61 −1.63 14
    ERG3, delta-5-desaturase)
    homolog (S. cerevisae)
    1390779_at Similar to phosphoseryl-tRNA −4.86 0.00011 0.00913 −0.64 0.64 −1.56 14
    kinase
    1390813_at Similar to RNA-binding −5.19 0.00005 0.00581 −0.62 0.65 −1.54 14
    protein Musashi2-S
    1390884_a_at UDP-GlcNAc: betaGal beta- 4.87 0.00010 0.00904 0.49 1.40 1.40 14
    1,3-N-
    acetylglucosaminyltransferase
    7 (predicted)
    1391021_at similar to KIAA1749 protein −7.55 0.00000 0.00027 −0.74 0.60 −1.67 14
    (predicted)
    1391170_at similar to mKIAA1757 protein −9.21 0.00000 0.00005 −2.01 0.25 −4.04 14
    (predicted)
    1391222_at similar to Nedd4 binding −5.91 0.00001 0.00219 −0.81 0.57 −1.76 14
    protein 1 (predicted)
    1391297_at REST corepressor 1 (predicted) −5.17 0.00005 0.00595 −0.92 0.53 −1.89 14
    1391578_at −8.48 0.00000 0.00009 −1.11 0.46 −2.15 14
    1391584_at Transcribed locus 6.04 0.00001 0.00185 0.45 1.37 1.37 14
    1391625_at Wiskott-Aldrich syndrome-like −10.52 0.00000 0.00002 −1.28 0.41 −2.43 14
    (human)
    1391669_at protein tyrosine phosphatase, −6.21 0.00001 0.00156 −0.82 0.56 −1.77 14
    receptor type, B (predicted)
    1391689_at similar to Retinoblastoma- −9.06 0.00000 0.00005 −1.20 0.44 −2.30 14
    binding protein 2 (RBBP-2)
    1391701_at MYST histone −5.18 0.00005 0.00581 −0.98 0.51 −1.97 14
    acetyltransferase (monocytic
    leukemia) 3 (predicted)
    1391743_at ELAV (embryonic lethal, −4.80 0.00012 0.00964 −1.36 0.39 −2.58 14
    abnormal vision, Drosophila)-
    like 1 (Hu antigen R)
    (predicted)
    1391830_at copine VIII (predicted) −5.22 0.00005 0.00568 −1.08 0.47 −2.12 14
    1391838_at ankyrin repeat domain 11 −7.81 0.00000 0.00019 −1.15 0.45 −2.23 14
    (predicted)
    1391848_at RNA binding motif protein 27 −7.02 0.00000 0.00053 −0.76 0.59 −1.70 14
    (predicted)
    1391968_at Similar to expressed sequence −4.89 0.00010 0.00883 −0.69 0.62 −1.62 14
    AA415817
    1392000_at Similar to PHD finger protein 5.01 0.00008 0.00742 0.45 1.37 1.37 14
    14 isoform 1
    1392061_at minichromosome maintenance 5.34 0.00004 0.00482 0.54 1.46 1.46 14
    deficient 10 (S. cerevisiae)
    (predicted)
    1392269_at transcriptional regulator, −6.23 0.00001 0.00153 −1.13 0.46 −2.19 14
    SIN3A (yeast) (predicted)
    1392277_at −7.29 0.00000 0.00040 −0.48 0.72 −1.40 14
    1392322_at GTPase, IMAP family member 7 −4.83 0.00012 0.00948 −0.29 0.82 −1.22 14
    1392472_at similar to myocyte enhancer −9.77 0.00000 0.00003 −0.88 0.54 −1.84 14
    factor 2C
    1392552_at similar to transcription −6.15 0.00001 0.00169 −0.96 0.51 −1.95 14
    repressor p66 (predicted)
    1392564_at myeloid/lymphoid or mixed- −6.13 0.00001 0.00171 −0.57 0.68 −1.48 14
    lineage leukemia 5 (trithorax
    homolog, Drosophila)
    (predicted)
    1392629_a_at similar to MADP-1 protein −4.93 0.00009 0.00829 −0.82 0.57 −1.77 14
    (predicted)
    1392738_at similar to KIAA1096 protein −5.88 0.00001 0.00231 −0.75 0.59 −1.68 14
    1392825_at LOC499256 −5.20 0.00005 0.00580 −0.93 0.53 −1.90 14
    1392864_at Rho GTPase activating protein −8.05 0.00000 0.00016 −1.37 0.39 −2.58 14
    5 (predicted)
    1392932_at leukocyte receptor cluster −4.81 0.00012 0.00958 −0.79 0.58 −1.73 14
    (LRC) member 8 (predicted)
    1392936_at similar to RNA binding motif −4.82 0.00012 0.00950 −0.88 0.54 −1.85 14
    protein 25
    1392984_at copine III (predicted) −7.83 0.00000 0.00019 −0.95 0.52 −1.93 14
    1393151_at 5.03 0.00007 0.00726 0.65 1.57 1.57 14
    1393226_at Transcribed locus −4.94 0.00009 0.00828 −0.73 0.60 −1.66 14
    1393290_at similar to myocyte enhancer −5.65 0.00002 0.00327 −0.50 0.71 −1.42 14
    factor 2C
    1393322_at TAF15 RNA polymerase II, −6.18 0.00001 0.00162 −1.00 0.50 −2.00 14
    TATA box binding protein
    (TBP)-associated factor
    (predicted)
    1393378_at −5.72 0.00002 0.00293 −0.52 0.70 −1.43 14
    1393443_a_at similar to CGI-112 protein −5.33 0.00004 0.00483 −0.47 0.72 −1.39 14
    (predicted)
    1393505_x_at similar to RIKEN cDNA −7.60 0.00000 0.00026 −0.69 0.62 −1.61 14
    B230380D07 (predicted)
    1393511_at similar to galactose-3-O- 5.10 0.00006 0.00655 0.41 1.33 1.33 14
    sulfotransferase 4
    1393560_at −4.91 0.00010 0.00852 −0.51 0.70 −1.42 14
    1393576_at Transcribed locus −4.82 0.00012 0.00950 −0.62 0.65 −1.54 14
    1393593_at similar to KIAA0597 protein 5.43 0.00003 0.00435 0.57 1.48 1.48 14
    1393639_at myosin X (predicted) −4.95 0.00009 0.00811 −0.59 0.67 −1.50 14
    1393790_at HRAS-like suppressor 5.44 0.00003 0.00432 0.44 1.35 1.35 14
    (predicted)
    1393798_at alpha thalassemia/mental −5.00 0.00008 0.00757 −0.84 0.56 −1.79 14
    retardation syndrome X-linked
    homolog (human)
    1393804_at similar to hypothetical protein −6.79 0.00000 0.00073 −0.85 0.56 −1.80 14
    FLJ22490 (predicted)
    1393809_at Tnf receptor-associated factor 6 −8.48 0.00000 0.00009 −0.90 0.53 −1.87 14
    (predicted)
    1393811_at similar to putative repair and −6.08 0.00001 0.00178 −0.79 0.58 −1.73 14
    recombination helicase
    RAD26L
    1393910_at similar to Fam13a1 protein −4.85 0.00011 0.00921 −0.81 0.57 −1.75 14
    (predicted)
    1393981_at similar to KIAA0423 −5.24 0.00005 0.00556 −0.57 0.68 −1.48 14
    (predicted)
    1394003_at similar to DNA polymerase −5.59 0.00002 0.00349 −0.59 0.67 −1.50 14
    epsilon p17 subunit (DNA
    polymerase epsilon subunit 3)
    (Chromatin accessibility
    complex 17) (HuCHRAC17)
    (CHRAC-17)
    1394220_at Similar to hypothetical protein 5.46 0.00003 0.00417 0.43 1.34 1.34 14
    (predicted)
    1394243_at similar to spermine synthase −6.11 0.00001 0.00175 −0.60 0.66 −1.51 14
    1394436_at sperm associated antigen 9 −6.60 0.00000 0.00090 −0.91 0.53 −1.88 14
    (predicted)
    1394497_at similar to TCF7L2 protein −8.03 0.00000 0.00016 −1.06 0.48 −2.08 14
    1394594_at Transcribed locus 5.09 0.00006 0.00671 0.42 1.34 1.34 14
    1394715_at Dicer1, Dcr-1 homolog 5.14 0.00006 0.00627 0.54 1.46 1.46 14
    (Drosophila) (predicted)
    1394740_at 5.41 0.00003 0.00440 0.52 1.43 1.43 14
    1394742_at Transcribed locus −5.73 0.00002 0.00289 −0.98 0.51 −1.98 14
    1394746_at hect (homologous to the E6-AP −7.32 0.00000 0.00039 −0.94 0.52 −1.91 14
    (UBE3A) carboxyl terminus)
    domain and RCC1 (CHC1)-like
    domain (RLD) 1 (predicted)
    1394814_at translocated promoter region −6.13 0.00001 0.00171 −0.63 0.64 −1.55 14
    (predicted)
    1394849_at Transcribed locus −5.22 0.00005 0.00569 −1.61 0.33 −3.05 14
    1394865_at Transmembrane protein 7 −7.85 0.00000 0.00019 −0.92 0.53 −1.90 14
    (predicted)
    1394965_at enthoprotin 5.30 0.00004 0.00503 0.40 1.32 1.32 14
    1394969_at Transcribed locus 5.40 0.00003 0.00441 0.39 1.31 1.31 14
    1394985_at early endosome antigen 1 −7.60 0.00000 0.00026 −1.00 0.50 −2.00 14
    (predicted)
    1395211_s_at supervillin (predicted) −8.74 0.00000 0.00007 −0.98 0.51 −1.97 14
    1395237_at eukaryotic translation initiation −8.31 0.00000 0.00012 −0.87 0.55 −1.83 14
    factor 5B
    1395264_at similar to Rap1-interacting −6.85 0.00000 0.00067 −0.95 0.52 −1.93 14
    factor 1
    1395331_at similar to hypothetical protein 4.84 0.00011 0.00945 0.31 1.24 1.24 14
    CL25084 (predicted)
    1395338_at leucine-rich PPR-motif 5.24 0.00005 0.00555 0.75 1.68 1.68 14
    containing (predicted)
    1395516_at similar to hypothetical protein −4.89 0.00010 0.00883 −0.59 0.66 −1.51 14
    FLJ10154 (predicted)
    1395565_at COP9 signalosome subunit 4 5.55 0.00002 0.00376 0.40 1.32 1.32 14
    1395610_at similar to Hypothetical protein 5.66 0.00002 0.00325 0.33 1.26 1.26 14
    MGC30714
    1395616_at similar to Ab2-008 (predicted) −5.03 0.00007 0.00729 −0.50 0.71 −1.42 14
    1395625_at Transcribed locus −6.03 0.00001 0.00187 −0.76 0.59 −1.70 14
    1395739_at similar to RIKEN cDNA 5.05 0.00007 0.00698 0.54 1.46 1.46 14
    C920006C10 (predicted)
    1395814_at Transcribed locus −5.09 0.00006 0.00663 −0.78 0.58 −1.71 14
    1395976_at similar to phosphoinositol 4- −6.37 0.00000 0.00126 −0.57 0.67 −1.49 14
    phosphate adaptor protein-2
    1395981_at helicase, ATP binding 1 −5.76 0.00001 0.00276 −0.62 0.65 −1.54 14
    (predicted)
    1396036_at Ral GEF with PH domain and −6.67 0.00000 0.00084 −1.04 0.49 −2.06 14
    SH3 binding motif 2
    (predicted)
    1396063_at DEK oncogene (DNA binding) −4.82 0.00012 0.00952 −0.63 0.65 −1.55 14
    1396100_at similar to RIKEN cDNA −5.15 0.00006 0.00610 −0.56 0.68 −1.47 14
    2010009L17 (predicted)
    1396170_at WW domain binding protein 4 −7.78 0.00000 0.00020 −0.77 0.59 −1.71 14
    1396187_at Hypothetical protein 5.14 0.00006 0.00622 0.51 1.43 1.43 14
    LOC606294
    1396202_at Transcribed locus 4.97 0.00008 0.00795 0.52 1.44 1.44 14
    1396403_at −9.07 0.00000 0.00005 −1.01 0.50 −2.02 14
    1396803_at similar to THO complex 2 −7.09 0.00000 0.00050 −0.90 0.54 −1.86 14
    1397203_at PRP4 pre-mRNA processing −6.18 0.00001 0.00162 −0.67 0.63 −1.59 14
    factor 4 homolog B (yeast)
    (predicted)
    1397234_at G patch domain containing 1 −5.65 0.00002 0.00326 −0.49 0.71 −1.40 14
    (predicted)
    1397367_at A disintegrin and 5.05 0.00007 0.00698 0.47 1.38 1.38 14
    metalloprotease domain 23
    (predicted)
    1397508_at similar to RIKEN cDNA −5.08 0.00006 0.00671 −0.62 0.65 −1.54 14
    2310005B10
    1397552_at echinoderm microtubule −8.47 0.00000 0.00009 −1.39 0.38 −2.62 14
    associated protein like 4
    (predicted)
    1397627_at diaphanous homolog 1 −5.07 0.00007 0.00680 −0.52 0.70 −1.43 14
    (Drosophila) (predicted)
    1397647_at solute carrier family 25 5.51 0.00003 0.00395 0.62 1.54 1.54 14
    (mitochondrial carrier;
    ornithine transporter) member
    15 (predicted)
    1397669_at Chemokine (C—C motif) 5.78 0.00001 0.00271 0.51 1.43 1.43 14
    receptor 6 (predicted)
    1397674_at eukaryotic translation initiation −6.44 0.00000 0.00116 −0.76 0.59 −1.69 14
    factor 3, subunit 8, 110 kDa
    (predicted)
    1397676_at Similar to osteoclast inhibitory −6.68 0.00000 0.00084 −1.34 0.39 −2.54 14
    lectin
    1397758_at Similar to choline −4.83 0.00011 0.00946 −0.38 0.77 −1.30 14
    phosphotransferase 1;
    cholinephosphotransferase 1
    alpha;
    cholinephosphotransferase 1
    1397959_at similar to RIKEN cDNA −6.39 0.00000 0.00123 −1.14 0.45 −2.20 14
    D130059P03 gene (predicted)
    1398311_a_at kinase D-interacting substance 5.14 0.00006 0.00627 0.44 1.36 1.36 14
    220
    1398351_at Ubiquitin specific protease 7 −5.60 0.00002 0.00349 −0.42 0.75 −1.34 14
    (herpes virus-associated)
    (predicted)
    1398420_at Similar to E3 ubiquitin ligase −5.33 0.00004 0.00483 −0.94 0.52 −1.92 14
    SMURF2 (predicted)
    1398436_at ubiquitin specific protease 42 −6.36 0.00000 0.00126 −0.76 0.59 −1.69 14
    (predicted)
    1398486_at CDNA clone MGC: 93990 −8.09 0.00000 0.00016 −1.53 0.35 −2.89 14
    IMAGE: 7115381
    1398522_at similar to Ab2-034 (predicted) −4.92 0.00009 0.00832 −0.51 0.70 −1.42 14
    1398553_at similar to CGI-100-like protein −6.91 0.00000 0.00062 −1.68 0.31 −3.20 14
    1398834_at mitogen activated protein −4.94 0.00009 0.00828 −0.32 0.80 −1.25 14
    kinase kinase 2
    1398926_at prefoldin 1 (predicted) −5.95 0.00001 0.00208 −0.48 0.72 −1.40 14
    1398963_at TAF10 RNA polymerase II, −5.42 0.00003 0.00436 −0.41 0.75 −1.33 14
    TATA box binding protein
    (TBP)-associated factor
    (predicted)
    1399099_at heterogeneous nuclear −4.94 0.00009 0.00829 −0.54 0.69 −1.46 14
    ribonucleoprotein U-like 1
    (predicted)
    1399140_at Transcribed locus −5.16 0.00005 0.00597 −0.49 0.71 −1.40 14
    AFFX-BioB- Biotin synthase −4.89 0.00010 0.00879 −0.64 0.64 −1.56 14
    M_at biotin synthesis, sulfur
    insertion?
    AFFX- dethiobiotin synthetase −4.92 0.00009 0.00834 −0.70 0.62 −1.62 14
    BioDn-5_at
    AFFX-r2-Ec- dethiobiotin synthetase −5.41 0.00003 0.00440 −0.51 0.70 −1.43 14
    bioD-5_at
  • EXAMPLE 7
  • 60 g fatty acid ethyl ester consisting of 10% EPA and 50% DHA (FAEE 10-50), obtained from Napro Pharma (Brattvaag, Norway) and 15 g TL-IM obtained from Novozymes (Bagsvaerd, Denmark) were mixed in an evacuated round bottomed glass flask for 15 minutes. Next, N2 was released into the glass flask and the mixture was heated to 65° C. 20 g Alcolec 40P® from American Lecithin Company Inc (Oxford, Conn., USA) was then added to the reaction mixture. Alcolec 40P® is a crude soybean phospholipid product containing 40% PC, 26% phosphatidylethanolamine, 11% phosphatidylinositol, 1% phosphatidylserine, 13% phytoglycolipids as well as 14% other phosphatides (w,w). Next, the glass flask was evacuated (20-30 mbar). Finally, a second vessel containing water (30° C.), was connected to the reaction vessel through a plastic tube (FIG. 1). The reduced pressure allowed moisture from the headspace of the second vessel to be added through the reaction mixture continuously. In order to obtain the final product the enzymes were removed by filtration. Finally, a triglyceride carrier was added to the product, followed by removal of the residual free fatty acids and/or esters by short path distillation. In order to analyze the product, the sample was fractionated by HPLC-UV (λ=207 nm) with a silica column and methanol-water (92:8, v/v) as mobile phase. The isolated PC+LPC fraction was then dried under nitrogen prior to derivatization; finally the fatty acid profile was determined by analyzing the derivatives using GC-FID. Furthermore, the relationship between PC, LPC and GPC was determined using HPLC with the method above, except that the UV detector was replaced by an evaporative light scattering detection (ELSD). Integrated ELSD peak areas were reported for PC/LPC/GPC (total 100%); however for simplicity other PL species were not analyzed. The results obtained for example 7 is shown in table 20 below.
    TABLE 20
    Results obtained after transesterification using vacuum and water addition
    Reaction time PC/LPC/GPC* EPA/DHA** Acid value
    1 day 65/31/4 3.6/2.6 43
    2 days 52/45/3 5.3/4.7 55
    5 days 78/22/0 5.2/4.6 65
    6 days 72/26/2 6.0/5.3 75

    *ELSD peak area (total 100%). Only peaks relating to PC, LPC and GPC are integrated.

    **EPA/DHA attached to PC + LPC
  • EXAMPLE 8
  • The enzymes from example 7 were isolated by filtration and the possibility of reuse was determined in the following experiment. 30 g FAEE (10-50), 10 g Alcolec and 15 g used enzymes (equivalent to 7.5 g enzyme because the used enzymes had absorbed product from the first reaction). The reaction was performed at 65° C. and stirred at 200 rpm using a shaker incubator. The transesterified phospholipids were analyzed as in the previous example and the results are shown in table 21 below.
    TABLE 21
    Results obtained with reused enzymes using incubator shaker
    Reaction time PC/LPC/GPC* EPA/DHA** Acid value
    1 day 94/5/1 0.4/0.5 55
    2 days 87/11/2 0.7/0.7 76
    5 days 68/26/6 1.0/0.8 85

    *ELSD peak area (total 100%). Only peaks relating to PC, LPC and GPC are integrated.

    **EPA/DHA attached to PC + LPC
  • EXAMPLE 9
  • The same conditions as in example 1 were used, except that the amount of lipase was 10 g. The results are shown in table 22.
    TABLE 22
    Results obtained with reduced lipase dosage after transesterification
    using vacuum and water addition
    Reaction time PC/LPC/GPC* EPA/DHA** Acid value
    1 day 90/10/0 0.9/0.7 29
    2 days 74/24/2 2.1/1.5 87
    3 days 49/27/24 4.6/4.4 102
    6 days 25/32/43 6.7/6.9 115

    *ELSD peak area (total 100%). Only peaks relating to PC, LPC and GPC are integrated.

    **EPA/DHA attached to PC + LPC
  • EXAMPLE 10
  • The enzymes from example 7 were isolated by filtration and the possibility of reuse was determined in the following experiment. 30 g FAEE (10-50), 10 g Alcolec and 15 g used enzymes (equivalent to 7.5 g enzyme because the used enzymes had absorbed product from the first reaction). The reaction was performed using the same conditions as in example 3. See table 23 below for results.
    TABLE 23
    Reuse of enzymes from example 7
    using vapor addition into evacuated reaction vessel.
    Reaction time PC/LPC/GPC* EPA/DHA** Acid value
    1 day 79/17/4 1.0/0.9 55
    2 days 59/31/10 2.8/2.6 76
    3 days 52/34/14 3.8/3.5 85
    6 days 37/43/20 5.6/5.7 95

    *ELSD peak area (total 100%). Only peaks relating to PC, LPC and GPC are integrated.

    **EPA/DHA attached the fraction consisting of PC + LPC
  • EXAMPLE 11
  • The same conditions as in example 7 are applied, except that the pressure in the reaction vessel is 1 mbar. The results obtained are similar to the results in Table 23, except that the hydrolysis and the acid values are reduced. After 6 days the relationship between PC species is 80/10/0 and the acids value is 40. The incorporation of EPA/DHA is the same.
  • EXAMPLE 12
  • The safety of omega-3 rich phospholipids prepared in the presence of chloroform and omega-7 rich phospholipids prepared under solvent free conditions is to be examined by feeding pregnant rats for 1 week. It is to be found that the treatment containing omega-3 rich phospholipds with traces of chloroform will result in damage to the developing fetus than the treatment containing essentially no traces of organic solvents.
  • Having now fully described this invention, it will be appreciated by those skilled in the art that the same can be performed within a wide range of equivalent parameters, concentrations, and conditions without departing from the spirit and scope of the invention and without undue experimentation.
  • While this invention has been described in connection with specific embodiments thereof, it will be understood that it is capable of further modifications. This application is intended to cover any variations, uses, or adaptations of the invention following, in general, the principles of the invention and including such departures from the present disclosure as come within known or customary practice within the art to which the invention pertains and as may be applied to the essential features hereinbefore set forth.
  • REFERENCES
  • [1] WO 2006054183
  • [2] P. C. Calder. Prostaglandins, Leukotrienes and Essential Fatty Acids 2006; 75; 197-202.
  • [3] U.S. Pat. No. 5,434,183
  • [4] Alexander J W. Nutrition 14 (1998) 627.
  • [5] Belluzi A, Boschi S, Brignola C, Munarini A, Cariani G and Miglio F. Am J Clin Nutr 71 (2000) 339.
  • [6] Kremer J M. Am J Clin Nutr 71 (2000) 249
  • [7] V P Carnielli, G. Verlato, F. Pederzini, I. Luijendijk, A. Boerlage, D. Pedrotti and P. Sauer Am J Clin Nutr 1998;67; 97-103.
  • [8] M. Moya, E. Cortes, M. Juste, J. G. De Dios and A Vera Eur. J. Clin. Nutr. 2001; 55; 755-762.
  • [9] A. Sala-Vila, A. I. Castello, C. Campoy, M. Rivero, M. Rodriguez-Palermoe and M. C. Lopez-Sabater. J. Nutr. 2004; 134; 868-873.
  • [10] A. Sala-Vila, C. Campoy, A. I. Castellote, F. J. Garrido, M. Rivero, M. Rodriguez-Palmero and M. C. Lopez-Sabater. Prostaglandins, Leukotrienes and Essential Fatty Acids; 2006; 74; 143-148.
  • [11] A. Valenzuela, S. Nieto, J. Sanhueza, M. J. Nunez and C. Ferrer. Annals of Nutrition & Metabolism; 2005; 49; 325-332.
  • [12] J. B. Hansen, S: Grimsgaard, H. Nilsen, A. Nordoy and K. H. Bonaa. Lipids; 1998; 33; 131-138.
  • [13] U.S. provisional application entitled “Functional Phospholipid Compositions” with Ser. No. 60/798,027 filed May 5, 2006.
  • [14] P. C Calder, Philip C. Am. J. Clin. Nutr; 2006; 83; 1505S-1519S.
  • [15] G. G. Haraldsson, A. Thorarensen, JAOCS 75 (1999) 1143-1149.
  • [16] G. Lepage and C. C. Roy; J. Lipid Res; 1986; 27; 114-120.
  • [17] T. Moriguchi, S-Y Lim, R. Greiner, W. Lefkowitz, J. Loewke, J. Hoshiba and N. Salem. J. Lipid Res; 2004;. 45; 1437-1445.
  • [18] H. Salman, M. Bergman, H Bessler, S Alexandrova, B. Beilin, M. Djaldetti. Acta Phys Scand; 2000; 168, 431-436.
  • [19] Haraldsson G G and Thorarensen A, JAOCS 75 (1999) 1143.
  • [20] Samey D B, Fregapane G and Vulfson E N. JAOCS 71 (1994) 93.

Claims (26)

1. A composition comprising phospholipids having the following structure:
Figure US20080044487A1-20080221-C00008
wherein R1 is OH or a fatty acid, R2 is OH or a fatty acid, and R3 is a mixture of H, choline, ethanolamine, inositol and serine, said phospholipid having at least 1% of DHA/EPA at positions R1 and/or R2 and from about 20-50% of OH at positions R1 and/or R2.
2. The composition as claimed in claim 1, wherein said composition is acylated in a range from about 55% to about 85%.
3. The composition as claimed in claim 2, wherein said composition has a ratio of EPA/DHA ranging from 1:1 to 4:1.
4. The composition of claim 2, wherein said composition having a ratio of EPA/DHA ranging from 2:1 to 4:1.
5. The composition of claim 1, wherein said composition further comprises a lipid carrier in a ratio of from 1:10 to 10:1 to said phospholipids.
6. The composition of claim 5, wherein said lipid carrier is selected from the group consisting of a triglyceride, a diglyceride, an ethyl ester, and a methyl ester and combinations thereof.
7. The composition in claim 1, wherein said composition provides higher uptake of omega-3 fatty acids into plasma as compared to natural marine phospholipids.
8. The composition in claim 1, wherein said composition improves the AA/EPA ratio in plasma phospholipids as compared to natural marine phospholipids.
9. The composition in claim 1, wherein said composition increases the concentration of omega-3 fatty acids in tissues as compared to natural marine phospholipids.
10. The composition in claim 1, wherein said composition reduces the concentration of biomarkers of inflammation as compared to natural marine phospholipids.
11. A food product comprising the composition in claim 1.
12. An animal feed comprising the composition in claim 1.
13. A food supplement comprising the composition in claim 1.
14. A pharmaceutical comprising the composition in claim 1.
15. The composition of claim 5, wherein said lipid carrier and said phospholipids are in a ratio of from about 5:1 to 1:5.
16. The composition of claim 5, wherein said composition comprises from about 20% to about 90% of said phospholipid composition and from about 10% to about 50% of said lipid carrier.
17. A method of preparing a bioavailable omega-3 fatty acid composition comprising:
a) providing a purified phospholipid composition comprising omega-3 fatty acid residues and a purified triglyceride composition comprising omega-3 fatty acid residues;
b) combining said phospholipid composition and said triglyceride composition to form a bioavailable omega-3 fatty acid composition.
18. The method of claim 17, further comprising the step of encapsulating said bioavailable omega-3 fatty acid composition.
19. The method of claim 17, wherein said bioavailable omega-3 fatty acid composition has increased bioavailability as compared to purified triglycerides or phospholipids comprising omega-3 fatty acid residues.
20. The method of claim 17, further comprising the step of packaging the bioavailable omega-3 fatty acid composition for use in functional foods.
21. The method of claim 17, further comprising the step of assaying the bioavailable omega-3 fatty acid composition for bioavailability.
22. The method of claim 17, further comprising administering the bioavailable omega-3 fatty acid composition to a patient.
23. A food product comprising a bioavailable omega-3 fatty acid composition made the process of claim 17.
24. An animal feed comprising a bioavailable omega-3 fatty acid composition made the process of claim 17.
25. A food supplement comprising a bioavailable omega-3 fatty acid composition made the process of claim 17.
26. A pharmaceutical comprising a bioavailable omega-3 fatty acid composition made the process of claim 17.
US11/800,229 2006-05-05 2007-05-04 Anti-inflammatory properties of marine lipid compositions Abandoned US20080044487A1 (en)

Priority Applications (5)

Application Number Priority Date Filing Date Title
US11/800,229 US20080044487A1 (en) 2006-05-05 2007-05-04 Anti-inflammatory properties of marine lipid compositions
US12/849,950 US10117882B2 (en) 2006-05-05 2010-08-04 Anti-inflammatory properties of marine lipid compositions
US16/180,564 US10525068B2 (en) 2006-05-05 2018-11-05 Anti-inflammatory properties of marine lipid compositions
US16/734,850 US11400105B2 (en) 2006-05-05 2020-01-06 Anti-inflammatory properties of marine lipid compositions
US17/874,677 US20230047556A1 (en) 2006-05-05 2022-07-27 Anti-inflammatory properties of marine lipid compositions

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US79802606P 2006-05-05 2006-05-05
US79803006P 2006-05-05 2006-05-05
US79802706P 2006-05-05 2006-05-05
US87209606P 2006-12-01 2006-12-01
US11/800,229 US20080044487A1 (en) 2006-05-05 2007-05-04 Anti-inflammatory properties of marine lipid compositions

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US12/849,950 Continuation US10117882B2 (en) 2006-05-05 2010-08-04 Anti-inflammatory properties of marine lipid compositions

Publications (1)

Publication Number Publication Date
US20080044487A1 true US20080044487A1 (en) 2008-02-21

Family

ID=39101658

Family Applications (5)

Application Number Title Priority Date Filing Date
US11/800,229 Abandoned US20080044487A1 (en) 2006-05-05 2007-05-04 Anti-inflammatory properties of marine lipid compositions
US12/849,950 Active 2028-05-13 US10117882B2 (en) 2006-05-05 2010-08-04 Anti-inflammatory properties of marine lipid compositions
US16/180,564 Active US10525068B2 (en) 2006-05-05 2018-11-05 Anti-inflammatory properties of marine lipid compositions
US16/734,850 Active 2027-11-15 US11400105B2 (en) 2006-05-05 2020-01-06 Anti-inflammatory properties of marine lipid compositions
US17/874,677 Pending US20230047556A1 (en) 2006-05-05 2022-07-27 Anti-inflammatory properties of marine lipid compositions

Family Applications After (4)

Application Number Title Priority Date Filing Date
US12/849,950 Active 2028-05-13 US10117882B2 (en) 2006-05-05 2010-08-04 Anti-inflammatory properties of marine lipid compositions
US16/180,564 Active US10525068B2 (en) 2006-05-05 2018-11-05 Anti-inflammatory properties of marine lipid compositions
US16/734,850 Active 2027-11-15 US11400105B2 (en) 2006-05-05 2020-01-06 Anti-inflammatory properties of marine lipid compositions
US17/874,677 Pending US20230047556A1 (en) 2006-05-05 2022-07-27 Anti-inflammatory properties of marine lipid compositions

Country Status (1)

Country Link
US (5) US20080044487A1 (en)

Cited By (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100010088A1 (en) * 2007-11-01 2010-01-14 Wake Forest University School Of Medicine Compositions and Methods for Prevention and Treatment of Mammalian Diseases
WO2010077358A1 (en) * 2009-01-02 2010-07-08 Nestec S.A. Methods for increasing endogenous plasmalogen levels
WO2011139010A1 (en) * 2010-05-03 2011-11-10 연세대학교 산학협력단 Composition for the prevention or treatment of obesity, dyslipidaemia, fatty liver or insulin resistance syndrome, comprising piperonal as an active ingredient
US8168611B1 (en) 2011-09-29 2012-05-01 Chemo S.A. France Compositions, kits and methods for nutrition supplementation
US8183227B1 (en) 2011-07-07 2012-05-22 Chemo S. A. France Compositions, kits and methods for nutrition supplementation
WO2012095749A1 (en) * 2011-01-14 2012-07-19 Pharma Marine As Removal of monoglycerides from fatty acid concentrates
US8343753B2 (en) 2007-11-01 2013-01-01 Wake Forest University School Of Medicine Compositions, methods, and kits for polyunsaturated fatty acids from microalgae
CN103355254A (en) * 2013-06-21 2013-10-23 四川大学华西医院 Preparation method of rhesus metabolism syndrome model
EP2934505A4 (en) * 2012-12-24 2016-11-30 Qualitas Health Ltd Eicosapentaenoic acid (epa) formulations
US9629820B2 (en) 2012-12-24 2017-04-25 Qualitas Health, Ltd. Eicosapentaenoic acid (EPA) formulations
US10123986B2 (en) 2012-12-24 2018-11-13 Qualitas Health, Ltd. Eicosapentaenoic acid (EPA) formulations
CN110538185A (en) * 2019-10-09 2019-12-06 广东海洋大学深圳研究院 Application of sea cucumber phospholipid in preparation of medicine for preventing and treating inflammation
US10653725B2 (en) 2015-08-17 2020-05-19 Aker Biomarine Antarctic As Methods for improving health of farmed fish
JP2021502819A (en) * 2017-11-17 2021-02-04 ヒルズ・ペット・ニュートリシャン・インコーポレーテッド Compositions containing omega-3 polyunsaturated and medium chain fatty acids
US11065267B2 (en) 2017-12-21 2021-07-20 Aker Biomarine Antarctic As Lysophosphatidylcholine compositions
EP3958973A4 (en) * 2019-04-26 2023-06-07 Cytosolve, Inc. Compositions for improving brain health and memory

Families Citing this family (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080044487A1 (en) * 2006-05-05 2008-02-21 Natural Asa Anti-inflammatory properties of marine lipid compositions
WO2013033618A1 (en) 2011-09-02 2013-03-07 Arctic Nutrition As Lipid compositions with high dha content
US9549937B2 (en) 2013-12-05 2017-01-24 Burvia, LLC. Composition containing phospholipid-DHA and folate
US9233114B2 (en) 2013-12-05 2016-01-12 Buriva, LLC Dietary supplement containing phospholipid-DHA derived from eggs
US9216199B2 (en) 2013-12-05 2015-12-22 Buriva, LLC Nutritional supplement containing phospholipid-DHA derived from eggs
US9610302B2 (en) 2013-12-05 2017-04-04 Buriva, LLC. Composition containing phospholipid-DHA and B vitamins
CN110564786B (en) * 2019-08-02 2022-06-03 大渔华创(广州)海洋生物科技有限公司 EPA/DHA type lysophospholipid composition and preparation method thereof
WO2023079332A1 (en) * 2021-11-02 2023-05-11 MOSSAVI, Seid Mohamad Hosein Composition for burns, bed sores and diabetic foot ulcers

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050130937A1 (en) * 2003-10-22 2005-06-16 Enzymotec Ltd. Lipids containing omega-3 and omega-6 fatty acids

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SE9101642D0 (en) 1991-05-30 1991-05-30 Kabi Pharmacia Ab phospholipids
ID16442A (en) 1996-01-22 1997-10-02 Dow Chemical Co WEIGHT POLYMER ETHYLENE ULTRA LOW MOLECULES
DK1417211T3 (en) 2001-07-27 2007-10-08 Neptune Technologies & Bioress Natural marine phospholipids containing flavonoids and polyunsaturated phospholipids and their use
AU2005305610A1 (en) 2004-11-17 2006-05-26 Natural Asa Enzymatically synthesized marine phospholipids
US20080044487A1 (en) * 2006-05-05 2008-02-21 Natural Asa Anti-inflammatory properties of marine lipid compositions

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050130937A1 (en) * 2003-10-22 2005-06-16 Enzymotec Ltd. Lipids containing omega-3 and omega-6 fatty acids

Cited By (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8343753B2 (en) 2007-11-01 2013-01-01 Wake Forest University School Of Medicine Compositions, methods, and kits for polyunsaturated fatty acids from microalgae
US20100010088A1 (en) * 2007-11-01 2010-01-14 Wake Forest University School Of Medicine Compositions and Methods for Prevention and Treatment of Mammalian Diseases
WO2010077358A1 (en) * 2009-01-02 2010-07-08 Nestec S.A. Methods for increasing endogenous plasmalogen levels
US9884041B2 (en) * 2010-05-03 2018-02-06 Yonsei University Technology Holdings, Inc. Composition for the prevention or treatment of obesity, dyslipidaemia, fatty liver or insulin resistance syndrome, comprising piperonal as an active ingredient
WO2011139010A1 (en) * 2010-05-03 2011-11-10 연세대학교 산학협력단 Composition for the prevention or treatment of obesity, dyslipidaemia, fatty liver or insulin resistance syndrome, comprising piperonal as an active ingredient
US20130116315A1 (en) * 2010-05-03 2013-05-09 Tae Sun Park Composition for the prevention or treatment of obesity, dyslipidaemia, fatty liver or insulin resistance syndrome, comprising piperonal as an active ingredient
WO2012095749A1 (en) * 2011-01-14 2012-07-19 Pharma Marine As Removal of monoglycerides from fatty acid concentrates
US8183227B1 (en) 2011-07-07 2012-05-22 Chemo S. A. France Compositions, kits and methods for nutrition supplementation
US8168611B1 (en) 2011-09-29 2012-05-01 Chemo S.A. France Compositions, kits and methods for nutrition supplementation
US8545896B2 (en) 2011-09-29 2013-10-01 Chemo S. A. France Compositions, kits and methods for nutrition supplementation
US10123986B2 (en) 2012-12-24 2018-11-13 Qualitas Health, Ltd. Eicosapentaenoic acid (EPA) formulations
EP2934505A4 (en) * 2012-12-24 2016-11-30 Qualitas Health Ltd Eicosapentaenoic acid (epa) formulations
US9629820B2 (en) 2012-12-24 2017-04-25 Qualitas Health, Ltd. Eicosapentaenoic acid (EPA) formulations
US10039734B2 (en) 2012-12-24 2018-08-07 Qualitas Health, Ltd. Eicosapentaenoic acid (EPA) formulations
WO2014202023A1 (en) * 2013-06-21 2014-12-24 四川大学华西医院 Method for preparing rhesus macaque metabolic syndrome model
CN103355254A (en) * 2013-06-21 2013-10-23 四川大学华西医院 Preparation method of rhesus metabolism syndrome model
US10653725B2 (en) 2015-08-17 2020-05-19 Aker Biomarine Antarctic As Methods for improving health of farmed fish
JP2021502819A (en) * 2017-11-17 2021-02-04 ヒルズ・ペット・ニュートリシャン・インコーポレーテッド Compositions containing omega-3 polyunsaturated and medium chain fatty acids
JP7030194B2 (en) 2017-11-17 2022-03-04 ヒルズ・ペット・ニュートリシャン・インコーポレーテッド Compositions containing omega-3 polyunsaturated and medium chain fatty acids
US11065267B2 (en) 2017-12-21 2021-07-20 Aker Biomarine Antarctic As Lysophosphatidylcholine compositions
EP3958973A4 (en) * 2019-04-26 2023-06-07 Cytosolve, Inc. Compositions for improving brain health and memory
CN110538185A (en) * 2019-10-09 2019-12-06 广东海洋大学深圳研究院 Application of sea cucumber phospholipid in preparation of medicine for preventing and treating inflammation

Also Published As

Publication number Publication date
US20100316680A1 (en) 2010-12-16
US11400105B2 (en) 2022-08-02
US20190070203A1 (en) 2019-03-07
US20200179416A1 (en) 2020-06-11
US10525068B2 (en) 2020-01-07
US20230047556A1 (en) 2023-02-16
US10117882B2 (en) 2018-11-06

Similar Documents

Publication Publication Date Title
US11400105B2 (en) Anti-inflammatory properties of marine lipid compositions
Paul et al. Plasmalogens: A potential therapeutic target for neurodegenerative and cardiometabolic disease
US11865143B2 (en) Bioeffective krill oil compositions
WO2008149177A2 (en) Marine lipid compositions and uses thereof
IL176054A (en) Oil or fat compositions containing phospholipids and long-chain polyunsaturated fatty acid supplying compound and food using the same
US20080058286A1 (en) Novel applications of omega-3 rich phospholipids
Wang et al. Digestion, absorption, and metabolism characteristics of EPA-enriched phosphoethanolamine plasmalogens based on gastrointestinal functions in healthy mice
AU2014256345C1 (en) Bio effective krill oil compositions
US20080070870A1 (en) Use of omega-3 rich phospholipids in the area of cognitive function
Zhang Omega-3 phospholipids
AU2012101335A4 (en) Processes and products thereof

Legal Events

Date Code Title Description
AS Assignment

Owner name: AKER BIOMARINE ASA, NORWAY

Free format text: CHANGE OF NAME;ASSIGNOR:NATURAL ASA;REEL/FRAME:019805/0418

Effective date: 20070707

Owner name: AKER BIOMARINE ASA, NORWAY

Free format text: CHANGE OF NAME;ASSIGNOR:NATURAL ASA;REEL/FRAME:019805/0274

Effective date: 20070707

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION