US20040220090A1 - Advanced glycation end-product intermediaries and post-amadori inhibition - Google Patents

Advanced glycation end-product intermediaries and post-amadori inhibition Download PDF

Info

Publication number
US20040220090A1
US20040220090A1 US10/834,419 US83441904A US2004220090A1 US 20040220090 A1 US20040220090 A1 US 20040220090A1 US 83441904 A US83441904 A US 83441904A US 2004220090 A1 US2004220090 A1 US 2004220090A1
Authority
US
United States
Prior art keywords
ribose
age
amadori
post
formation
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
US10/834,419
Inventor
Billy Hudson
Parvin Todd
Raja Khalifah
Aaron Booth
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.)
University of Kansas Medical Center
Original Assignee
University of Kansas Medical Center
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
Priority claimed from US08/711,555 external-priority patent/US5985857A/en
Application filed by University of Kansas Medical Center filed Critical University of Kansas Medical Center
Priority to US10/834,419 priority Critical patent/US20040220090A1/en
Publication of US20040220090A1 publication Critical patent/US20040220090A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6803General methods of protein analysis not limited to specific proteins or families of proteins
    • G01N33/6842Proteomic analysis of subsets of protein mixtures with reduced complexity, e.g. membrane proteins, phosphoproteins, organelle proteins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/04Nitro compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4415Pyridoxine, i.e. Vitamin B6
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/675Phosphorus compounds having nitrogen as a ring hetero atom, e.g. pyridoxal phosphate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/05Dipeptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/06Tripeptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/07Tetrapeptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/06Dipeptides
    • C07K5/06008Dipeptides with the first amino acid being neutral
    • C07K5/06017Dipeptides with the first amino acid being neutral and aliphatic
    • C07K5/06034Dipeptides with the first amino acid being neutral and aliphatic the side chain containing 2 to 4 carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/06Dipeptides
    • C07K5/06008Dipeptides with the first amino acid being neutral
    • C07K5/06017Dipeptides with the first amino acid being neutral and aliphatic
    • C07K5/06034Dipeptides with the first amino acid being neutral and aliphatic the side chain containing 2 to 4 carbon atoms
    • C07K5/06052Val-amino acid
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/08Tripeptides
    • C07K5/0802Tripeptides with the first amino acid being neutral
    • C07K5/0812Tripeptides with the first amino acid being neutral and aromatic or cycloaliphatic
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2400/00Assays, e.g. immunoassays or enzyme assays, involving carbohydrates
    • G01N2400/02Assays, e.g. immunoassays or enzyme assays, involving carbohydrates involving antibodies to sugar part of glycoproteins
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value
    • G01N2500/04Screening involving studying the effect of compounds C directly on molecule A (e.g. C are potential ligands for a receptor A, or potential substrates for an enzyme A)

Definitions

  • the instant invention is in the field of Advanced Glycation End-products (AGEs), their formation, detection, identification, inhibition, and inhibitors thereof.
  • AGEs Advanced Glycation End-products
  • Nonenzymatic glycation by glucose and other reducing sugars is an important post-translational modification of proteins that has been increasingly implicated in diverse pathologies. Irreversible nonenzymatic glycation and crosslinking through a slow, glucose-induced process may mediate many of the complications associated with diabetes. Chronic hyperglycemia associated with diabetes can cause chronic tissue damage which can lead to complications such as retinopathy, nephropathy, and atherosclerotic disease. (Cohen and Ziyadeh, 1996, J. Amer. Soc. Nephrol. 7:183-190).
  • Glycated proteins have also been shown to be toxic, antigenic, and capable of triggering cellular injury responses after uptake by specific cellular receptors (see for example, Vlassara, Bucala & Striker, 1994, Lab. Invest.
  • the instant disclosure teaches previously unknown, and unpredicted mechanism of formation of post-Amadori advanced glycation end products (Maillard products; AGEs) and methods for identifying and characterizing effective inhibitors of post-Amadori AGE formation.
  • the instant disclosure demonstrates the unique isolation and kinetic characterization of a reactive protein intermediate competent in forming post-Amadori AGEs, and for the first time teaching methods which allow for the specific elucidation and rapid quantitative kinetic study of “late” stages of the protein glycation reaction.
  • Glycation reactions are known to be initiated by reversible Schiff-base (aldimine or ketimine) addition reactions with lysine side-chain ⁇ -amino and terminal ⁇ -amino groups, followed by essentially irreversible Amadori rearrangements to yield ketoamine products e.g. 1-amino-1-deoxy-ketoses from the reaction of aldoses (Baynes et al., 1989, in The Maillard Reaction in Aging, Diabetes, and Nutrition , ed. Monnier and Baynes, (Alan R. Liss, New York, pp 43-67).
  • sugars initially react in their open-chain (not the predominant pyranose and furanose structures) aldehydo or keto forms with lysine side chain ⁇ -amino and terminal ⁇ -amino groups through reversible Schiff base condensation (Scheme I).
  • the resulting aldimine or ketimine products then undergo Amadori rearrangements to give ketoamine Amadori products, i.e. 1-amino-1-deoxy-ketoses from the reaction of aldoses (Means & Chang, 1982, Diabetes 31, Suppl. 3:1-4; Harding, 1985, Adv. Protein Chem. 37:248-334).
  • FFI 2-(2-furoyl)-4(5)-(2-furanyl)-1H-imidazole
  • Inhibition of AGE formation has utility in the areas of, for example, food spoilage, animal protein aging, and personal hygiene such as combating the browning of teeth.
  • Some notable, though quantitatively minor, advanced glycation end-products are pentosidine and N ⁇ -carboxymethyllysine (Sell and Monnier, 1989, J. Biol. Chem. 264:21597-21602; Ahmed et al., 1986, J. Biol. Chem. 261:4889-4894).
  • Inhibitory compounds that are biodegradeble and/or naturally metabolized are more desirable for use as therapeutics than highly reactive compounds which may have toxic side effects, such as aminoguanidine.
  • AGE post-Amadori advanced glycation end-product
  • This stable product is a presumed sugar saturated Amadori/Schiff base product produced by the further reaction of the early stage protein/sugar Amadori product with more sugar.
  • this post-Amadori/Schiff base intermediary has been generated by the reaction of target protein with ribose sugar.
  • the instant invention provides for a method of generating stable protein-sugar AGE formation intermediary precursors via a novel method of high sugar inhibition.
  • the sugar used is ribose.
  • the instant invention provides for a method for identifying an effective inhibitor of the formation of late Maillard products comprising: generating stable protein-sugar post-Amadori advanced glycation end-product intermediates by incubating a protein with sugar at a sufficient concentration and for sufficient length of time to generate stable post-Amadori AGE intermediates; contacting said stable protein-sugar post-Amadori advanced glycation end-product intermediates with an inhibitor candidate; identifying effective inhibition by monitoring the formation of post-Amadori AGEs after release of the stable protein-sugar post-Amadori advanced glycation end-product intermediates from sugar induced equilibrium.
  • Appropriate sugars include, and are not limited to ribose, lyxose, xylose, arabinose, glucose, fructose, maltose, lactose, mannose, fructose and galactose.
  • the sugar used is ribose.
  • the instant invention teaches that an effective inhibitor of post-Amadori AGE formation via “late” reactions can be identified and characterized by the ability to inhibit the formation of post-Amadori AGE endproducts in an assay comprising; generating stable protein-sugar post-Amadori advanced glycation end-product intermediates by incubating a protein with sugar at a sufficient concentration and for sufficient length of time to generate stable post-Amadori AGE intermediates; contacting said stable protein-sugar post-Amadori advanced glycation end-product intermediates with an inhibitor candidate; identifying effective inhibition by monitoring the formation of post-Amadori AGEs after release of the stable protein-sugar post-Amadori advanced glycation end-product intermediates from sugar induced equilibrium.
  • the assay uses ribose.
  • the methods of the instant invention allow for the rapid screening of candidate post-Amadori AGE formation inhibitors for effectiveness, greatly reducing the cost and amount of work required for the development of effective small molecule inhibitors of post-Amadori AGE formation.
  • effective inhibitors of post-Amadori “late” reactions of AGE formation include derivatives of vitamin B 6 and vitamin B 1 , in the preferred embodiment the specific species being pyridoxamine and thiamine pyrophosphate.
  • the instant invention teaches new methods for rapidly inducing diabetes like pathologies in rats comprising administering ribose to the subject animal. Further provided for is the use of identified inhibitors pyridoxamine and thiamine pyrophosphate in vivo to inhibit post-Amadori AGE induced pathologies.
  • FIG. 1 is a series of graphs depicting the effect of vitamin B 6 derivatives on AGE formation in bovine serum albumin (BSA).
  • FIG. 1A Pyridoxamine (PM);
  • FIG. 1B pyridoxal phosphate (PLP);
  • FIG. 1C pyridoxal (PL);
  • FIG. 1D pyridoxine (PN).
  • FIG. 2 is a series of graphs depicting the effect of vitamin B 1 derivatives and aminoguanidine (AG) on AGE formation in bovine serum albumin.
  • FIG. 2A Thiamine pyrophosphate (TPP);
  • FIG. 2B thiamine monophosphate (TP);
  • FIG. 2D aminoguanidine (AG).
  • FIG. 3 is a series of graphs depicting the effect of vitamin B 6 derivatives on AGE formation in human methemoglobin (Hb).
  • FIG. 3A Pyridoxamine (PM);
  • FIG. 3B pyridoxal phosphate (PLP);
  • FIG. 3C pyridoxal (PL);
  • FIG. 3D pyridoxine
  • FIG. 4 is a series of graphs depicting the effect of vitamin B 1 derivatives and aminoguanidine (AG) on AGE formation in human methemoglobin.
  • FIG. 2A Thiamine pyrophosphate (TPP);
  • FIG. 2B thiamine monophosphate (TP);
  • FIG. 2D aminoguanidine (AG).
  • FIG. 5 is a bar graph comparison of the inhibition of the glycation of ribonuclease A by thiamine pyrophosphate (TPP), pyridoxamine (PM) and aminoguanidine (AG).
  • TPP thiamine pyrophosphate
  • PM pyridoxamine
  • AG aminoguanidine
  • FIG. 6A is a graph of the kinetics of glycation of RNase A (10 mg/mL) by ribose as monitored by ELISA.
  • FIG. 6B is a graph showing the dependence of reciprocal half-times on ribose concentration at pH 7.5.
  • FIG. 7 are two graphs showing a comparison of uninterrupted and interrupted glycation of RNase by glucose ( 7 B) and ribose ( 7 A), as detected by ELISA.
  • FIG. 8 are two graphs showing kinetics of pentosidine fluorescence (arbitrary units) increase during uninterrupted and interrupted ribose glycation of RNase.
  • FIG. 8A Uninterrupted glycation in the presence of 0.05 M ribose.
  • FIG. 8B Interrupted glycation after 8 and 24 hours of incubation.
  • FIG. 9 is a graph which shows the kinetics of reactive intermediate buildup.
  • FIG. 10 are graphs of Post-Amadori inhibition of AGE formation by ribose.
  • FIG. 10A graphs data where aliquots were diluted into inhibitor containing buffers at time 0.
  • FIG. 10B graphs data where samples were interrupted at 24 h, and then diluted into inhibitor containing buffers.
  • FIG. 11 is a graph showing dependence of the initial rate of formation of antigenic AGE on pH following interruption of glycation.
  • FIG. 12 are two graphs showing the effect of pH jump on ELISA detected AGE formation after interrupted glycation. Interrupted samples left 12 days at 37° C. in pH 5.0 buffer produced substantial AGEs (33%; FIG. 12 B) when pH was changed to 7.5, as compared to the normal control sample not exposed to low pH (FIG. 12 A)
  • FIG. 13 is a series of graphs depicting the effect of vitamin B 6 derivatives on AGE formation during uninterrupted glycation of ribonuclease A (RNase A) by ribose.
  • FIG. 13A Pyridoxamine (PM);
  • FIG. 13B pyridoxal-5′-phosphate (PLP);
  • FIG. 13C pyridoxal (PL);
  • FIG. 13D pyridoxine
  • FIG. 14 is a series of graphs depicting the effect of vitamin B 1 derivatives and aminoguanidine (AG) on AGE formation during uninterrupted glycation of ribonuclease A (RNase A) by ribose.
  • FIG. 14A Thiamine pyrophosphate (TPP);
  • FIG. 14B thiamine monophosphate (TP);
  • FIG. 14D aminoguanidine (AG).
  • FIG. 15 is a series of graphs depicting the effect of vitamin B 6 derivatives on AGE formation during uninterrupted glycation of bovine serum albumin (BSA) by ribose.
  • FIG. 15A Pyridoxamine (PM);
  • FIG. 15B pyridoxal-5′-phosphate (PLP);
  • FIG. 15C pyridoxal (PL);
  • FIG. 15D pyridoxine (PN).
  • FIG. 16 is a series of graphs depicting the effect of vitamin B 1 derivatives and aminoguanidine (AG) on AGE formation during uninterrupted glycation of bovine serum albumin (BSA) by ribose.
  • FIG. 16A Thiamine pyrophosphate (TPP);
  • FIG. 16B thiamine monophosphate (TP);
  • FIG. 16D aminoguanidine (AG).
  • FIG. 17 is a series of graphs depicting the effect of vitamin B 6 derivatives on AGE formation during uninterrupted glycation of human methemoglobin (Hb) by ribose.
  • FIG. 17A Pyridoxamine (PM);
  • FIG. 17B pyridoxal-5′-phosphate (PLP);
  • FIG. 17C pyridoxal (PL);
  • FIG. 17D pyridoxine (PN).
  • FIG. 18 is a series of graphs depicting the effect of vitamin B 6 derivatives on post-Amadori AGE formation after interrupted glycation by ribose.
  • FIG. 18D RNase and pyridoxamine (PM)
  • FIG. 19 are graphs depicting the effect of thiamine pyrophosphate on post-Amadori AGE formation after interrupted glycation by ribose.
  • FIG. 19A RNase
  • FIG. 19B BSA.
  • FIG. 20 are graphs depicting the effect of aminoguanidine on post-Amadori AGE formation after interrupted glycation by ribose.
  • FIG. 20A RNase, FIG. 20B BSA.
  • FIG. 21 is a graph depicting the effect of N ⁇ -acetyl-L-lysine on post-Amadori AGE formation after interrupted glycation by ribose.
  • FIG. 22 are bar graphs showing a comparison of post-Amadori inhibition of AGE formation by thiamine pyrophosphate (TPP), pyridoxamine (PM) and aminoguanidine (AG) after interrupted glycation of RNase (FIG. 22A) and BSA (FIG. 22B) by ribose.
  • TPP thiamine pyrophosphate
  • PM pyridoxamine
  • AG aminoguanidine
  • FIG. 23 is a bar graph showing the effects of Ribose treatment in vivo alone on rat tail-cuff blood pressure. Treatment was with 0.05 M, 0.36 M, and 1 M Ribose (R) injected for 1, 2 or 8 Days (D).
  • FIG. 24 is a bar graph showing the effects of Ribose treatment in vivo alone on rat creatinine clearance (Clearance per 100 g Body Weight). Treatment was with 0.05 M, 0.30 M, and 1 M Ribose (R) injected for 1, 2 or 8 Days (D).
  • FIG. 25 is a bar graph showing the effects of Ribose treatment in vivo alone on rat Albuminuria (Albumin effusion rate). Treatment was with 0.30 M, and 1 M Ribose (R) injected for 1, 2 or 8 Days (D).
  • FIG. 26 is a bar graph showing the effects of inhibitor treatment in vivo, with or without ribose, on rat tail-cuff blood pressure.
  • Treatment groups were: 25 mg/100 g body weight aminoguanidine (AG); 25 or 250 mg/100 g body weight Pyridoxamine (P); 250 mg/100 g body weight Thiamine pyrophosphate (T), or with 1 M Ribose (R).
  • FIG. 27 is a bar graph showing the effects of inhibitor treatment in vivo, with or without ribose, on rat creatinine clearance (Clearance per 100 g body weight).
  • Treatment groups were: 25 mg/100 g body weight aminoguanidine (AG); 25 or 250 mg/100 g body weight Pyridoxamine (P); 250 mg/100 g body weight Thiamine pyrophosphate (T), or with 1 M Ribose (R).
  • FIG. 28 is a bar graph showing the effects of inhibitor treatment in vivo without ribose, and ribose alone on rat Albuminuria (Albumin effusion rate).
  • Treatment groups were: 25 mg/100 g body weight aminoguanidine (AG); 250 mg/100 g body weight Pyridoxamine (P); 250 mg/100 g body weight Thiamine pyrophosphate (T), or treatment with 1 M Ribose (R) for 8 days (D). Control group had no treatment.
  • FIG. 29 is a bar graph showing the effects of inhibitor treatment in vivo, with 1 M ribose , on rat Albuminuria (Albumin effusion rate).
  • Treatment groups were: 25 mg/100 g body weight aminoguanidine (AG); 25 and 250 mg/100 g body weight Pyridoxamine (P); 250 mg/100 g body weight Thiamine pyrophosphate (T), or treatment with 1 M Ribose (R) for 8 days (D) alone. Control group had no treatment.
  • FIG. 30A depicts Scheme 1 showing a diagram of AGE formation from protein.
  • FIG. 30B depicts Scheme 2, a chemical structure of aminoguanidine.
  • FIG. 30C depicts Scheme 3, chemical structures for thiamine, thiamine-5′-phosphate, and thiamine pyrophosphate.
  • FIG. 30D depicts Scheme 4, chemical structures of pyridoxine, pyridoxamine, pyridoxal-5′-phosphate, and pyridoxal.
  • FIG. 30E depicts Scheme 5, kinetics representation of AGE formation.
  • FIG. 30F depicts Scheme 6, kinetics representation of AGE formation and intermediate formation.
  • FIG. 31 shows a 125 MHz C-13 NMR Resonance spectrum of Riobonuclease Amadori Intermediate prepared by 24 HR reaction with 99% [2-C13]Ribose.
  • Alloxan induced diabetic Lewis rats have been used as a model for protein aging to demonstrate the in vivo effectiveness of inhibitors of AGE formation. The correlation being demonstrated is between inhibition of late diabetes related pathology and effective inhibition of AGE formation (Brownlee, Cerami, and Vlassara, 1988, New Eng. J. Med. 318(20):1315-1321). Streptozotocin induction of diabetes in Lewis rats, New Zealand White rabbits with induced diabetes, and genetically diabetic BB/Worcester rats have also been utilized, as described in, for example, U.S. Pat. No. 5,334,617 (incorporated by reference). A major problem with these model systems is the long time period required to demonstrate AGE related injury, and thus to test compounds for AGE inhibition.
  • An important tool for studying AGE formation is the use of polyclonal and monoclonal antibodies that are specific for AGEs elicited by the reaction of several sugars with a variety of target proteins.
  • the antibodies are screened for resultant specificity for AGEs that is independent of the nature of the protein component of the AGE (Nakayama et al., 1989, Biochem. Biophys. Res. Comm. 162: 740-745; Nakayama et al., 1991, J. Immunol. Methods 140: 119-125; Horiuchi et al., 1991, J. Biol. Chem. 266: 7329-7332; Araki et al., 1992, J. Biol. Chem. 267: 10211-10214; Makita et al., 1992, J. Biol. Chem. 267: 5133-5138).
  • Such antibodies have been used to monitor AGE formation in vivo and in vitro.
  • thiamine is practically devoid of pharmacodynamic actions when given in usual therapeutic doses; and even large doses were not known to have any effects.
  • Thiamine pyrophosphate is the physiologically active form of thiamine, and it functions mainly in carbohydrate metabolism as a coenzyme in the decarboxylation of ⁇ -keto acids.
  • Tablets of thiamine hydrochloride are available in amounts ranging from 5 to 500 mg each.
  • Thiamine hydrochloride injection solutions are available which contain 100 to 200 mg/ml.
  • intravenous doses of as high as 100 mg/L of parenteral fluid are commonly used, with the typical dose of 50 to 100 mg being administered.
  • GI absorption of thiamine is believed to be limited to 8 to 15 mg per day, but may be exceed by oral administration in divided doses with food.
  • the instant invention has found, as shown by in vitro testing, that administration of thiamine pyrophosphate at levels above what is normally found in the human body or administered for dietary therapy, is an effective inhibitor of post-Amadori antigenic AGE formation, and that this inhibition is more complete than that possible by the administration of aminoguanidine.
  • Vitamin B 6 is typically available in the form of pyridoxine hydrochloride in over-the-counter preparations available from many sources.
  • Beach pharmaceuticals Beelith Tablets contain 25 mg of pyridoxine hydrochloride that is equivalent to 20 mg of B 6
  • other preparations include Marlyn Heath Care Marlyn Formula 50 which contain 1 mg of pyridoxine HCl and Marlyn Formula 50 Mega Forte which contains 6 mg of pyridoxine HCl, Wyeth-Ayerst Stuart Prenatal® tablets which contain 2.6 mg pyridoxine HCl, J&J-Merck Corp.
  • pyridoxine is a primary alcohol
  • pyridoxal is the corresponding aldehyde
  • pyridoxamine contains an aminomethyl group at this position.
  • pyridoxine is a primary alcohol
  • pyridoxal is the corresponding aldehyde
  • pyridoxamine contains an aminomethyl group at this position.
  • Each of these three forms can be utilized by mammals after conversion by the liver into pyridoxal-5′-phosphate, the active form of the vitamin. It has long been believed that these three forms are equivalent in biological properties, and have been treated as equivalent forms of vitamin B 6 by the art.
  • the Council on Pharmacy and Chemistry has assigned the name pyridoxine to the vitamin.
  • the most active antimetabolite to pyridoxine is 4-deoxypyridoxine, for which the antimetabolite activity has been attributed to the formation in vivo of 4-deoxypyridoxine-5-phosphate, a competitive inhibitor of several pyridoxal phosphate-dependent enzymes.
  • the pharmacological actions of pyridoxine are limited, as it elicits no outstanding pharmacodynamic actions after either oral or intravenous administration, and it has low acute toxicity, being water soluble. It has been suggested that neurotoxicity may develop after prolonged ingestion of as little as 200 mg of pyridoxine per day.
  • pyridoxine phosphate is involved in several metabolic transformations of amino acids including decarboxylation, transamination, and racemization, as well as in enzymatic steps in the metabolism of sulfur-containing and hydroxy-amino acids.
  • pyridoxal phosphate is aminated to pyridoxamine phosphate by the donor amino acid, and the bound pyridoxamine phosphate is then deaminated to pyridoxal phosphate by the acceptor ⁇ -keto acid.
  • vitamin B complex is known to be a necessary dietary supplement involved in specific breakdown of amino acids.
  • the instant invention has discovered that effective dosages of the metabolically transitory pyridoxal amine form of vitamin B 6 (pyridoxamine), at levels above what is normally found in the human body, is an effective inhibitor of post-Amadori antigenic AGE formation, and that this inhibition may be more complete than that possible by the administration of aminoguanidine.
  • B 6 vitamers especially pyridoxal phosphate (PLP) have been previously proposed to act as “competitive inhibitors” of early glycation, since as aldehydes they themselves can form Schiff bases adducts with protein amino groups (Khatami et al., 1988, Life Sciences 43:1725-1731) and thus limit the amount of amines available for glucose attachment.
  • PBP pyridoxal phosphate
  • effectiveness in limiting initial sugar attachment is not a predictor of inhibition of the conversion of any Amadori products formed to AGEs.
  • the instant invention describes inhibitors of “late” glycation reactions as indicated by their effects on the in vitro formation of antigenic AGEs (Booth et al., 1996, Biochem. Biophys. Res. Com. 220:113-119).
  • Bovine pancreatic ribonuclease A (RNase) was chromatographically pure, aggregate-free grade from Worthington Biochemicals.
  • Bovine Serum albumin (BSA; fraction V, fatty-acid free), human methemoglobin (Hb), D-glucose, pyridoxine, pyridoxal, pyridoxal 5′phosphate, pyridoxamine, thiamine, thiamine monophosphate, thiamine pyrophosphate, and goat alkaline phosphatase-conjugated anti-rabbit IgG were all from Sigma Chemicals.
  • Aminoguanidine hydrochloride was purchased from Aldrich Chemicals.
  • Bovine serum albumin, ribonuclease A, and human hemoglobin were incubated with glucose at 37° C. in 0.4 M sodium phosphate buffer of pH 7.5 containing 0.02% sodium azide.
  • the protein, glucose (at 1.0 M), and prospective inhibitors were introduced into the incubation mixture simultaneously. Solutions were kept in the dark in capped tubes. Aliquots were taken and immediately frozen until analyzed by ELISA at the conclusion of the reaction. The incubations were for 3 weeks (Hb) or 6 weeks (RNase, BSA).
  • Immunogen preparation followed earlier protocols (Nakayama et al., 1989, Biochem. Biophys. Res. Comm. 162:740-745; Horiuchi et al., 1991, J. Biol. Chem. 266:7329-7332; Makita et al., 1992, J. Biol. Chem. 267:5133-5138). Briefly, immunogen was prepared by glycation of BSA (R479 antibodies) or RNase (R618 antibodies) at 1.6 g protein in 15 ml for 60-90 days using 1.5 M glucose in 0.4 M sodium phosphate buffer of pH 7.5 containing 0.05% sodium azide at pH 7.4 and 37° C.
  • BSA R479 antibodies
  • RNase R618 antibodies
  • New Zealand white rabbit males of 8-12 weeks were immunized by subcutaneous administration of a 1 ml solution containing 1 mg/ml of glycated protein in Freund's adjuvant.
  • the primary injection used the complete adjuvant and three boosters were made at three week intervals with Freund's incomplete adjuvant. Rabbits were bled three weeks after the last booster.
  • the serum was collected by centrifugation of clotted whole blood.
  • the antibodies are AGE-specific, being unreactive with either native proteins (except for the carrier) or with Amadori intermediates.
  • the polyclonal anti-AGE antibodies have proven to be a sensitive and valuable analytical tool for the study of “late” AGE formation in vitro and in vivo.
  • CmL carboxymethyl lysine
  • Rabbit anti-AGE antibodies were diluted at a titer of about 1:350 in incubation buffer, and incubated for 1 h at 37° C., followed by washing.
  • antibodies R479 used to detect glycated RNase were raised against glycated BSA
  • antibodies R618 used to detect glycated BSA and glycated Hb were raised against glycated RNase.
  • An alkaline phosphatase-conjugated antibody to rabbit IgG was then added as the secondary antibody at a titer of 1:2000 or 1:2500 (depending on lot) and incubated for 1 h at 37° C., followed by washing.
  • the p-nitrophenylphosphate substrate solution was then added (200 ul/well) to the plates, with the absorbance of the released p-nitrophenolate being monitored at 410 nm with a Dynatech MR 4000 microplate reader.
  • FIGS. 1 A-D are graphs which show the effect of vitamin B 6 derivatives on post-Amadori AGE formation in bovine serum albumin glycated with glucose.
  • BSA (10 mg/ml) was incubated with 10 M glucose in the presence and absence of the various indicated derivative in 0.4 M sodium phosphate buffer of pH 7.5 at 37° C. for 6 weeks. Aliquots were assayed by ELISA using R618 anti-AGE antibodies. Concentrations of the inhibitors were 3, 15 and 50 mM.
  • Inhibitors used in FIGS. ( 1 A) Pyridoxamine (PM); ( 1 B) pyridoxal phosphate (PLP); ( 1 C) pyridoxal (PL); ( 1 D) pyridoxine (PN).
  • FIG. 1 (control curves) demonstrates that reaction of BSA with 1.0 M glucose is slow and incomplete after 40 days, even at the high sugar concentration used to accelerate the reaction.
  • the simultaneous inclusion of different concentrations of various B 6 vitamers markedly affects the formation of post-Amadori antigenic AGEs.
  • FIG. 1A-D Pyridoxamine and pyridoxal phosphate strongly suppressed post-Amadori antigenic AGE formation at even the lowest concentrations tested, while pyridoxal was effective above 15 mM. Pyridoxine was slightly effective at the highest concentrations tested.
  • FIGS. 2 A-D are graphs which show the effect of vitamin B 1 derivatives and aminoguanidine (AG) on post-Amadori AGE formation in bovine serum albumin.
  • BSA (10 mg/ml) was incubated with 1.0 M glucose in the presence and absence of the various indicated derivative in 0.4 M sodium phosphate buffer of pH 7.5 at 37° C. for 6 weeks. Aliquots were assayed by ELISA using R618 anti-AGE antibodies. Concentrations of the inhibitors were 3, 15 and 50 mM.
  • Inhibitors used in FIGS. ( 2 A) Thiamine pyrophosphate (TPP); ( 2 B) thiamine monophosphate (TP); ( 2 C) thiamine (T); ( 2 D) aminoguanidine (AG).
  • FIGS. 3 A-D are graphs which show the effect of vitamin B 6 derivatives on post-Amadori AGE formation in human methemoglobin.
  • Hb (1 mg/ml) was incubated with 1.0 M glucose in the presence and absence of the various indicated derivative in 0.4 M sodium phosphate buffer of pH 7.5 at 37° C. for 3 weeks. Aliquots were assayed by ELISA using R618 anti-AGE antibodies. Concentrations of the inhibitors were 0.5, 3, 15 and 50 mM.
  • Inhibitors used in FIGS. ( 3 A) Pyridoxamine (PM); ( 3 B) pyridoxal phosphate (PLP); ( 3 C) pyridoxal (PL); ( 3 D) pyridoxine (PN).
  • Hb of a diabetic patient contains a component that binds to anti-AGE antibodies, and it was proposed that this glycated Hb (termed Hb-AGE, not to be confused with Hb Alc ) could be useful in measuring long-term exposure to glucose.
  • the in vitro incubation of Hb with glucose produces antigenic AGEs at an apparently faster rate than observed with BSA.
  • B 6 and B 1 FIG. 4A-C vitamers exhibited the same inhibition trends in Hb, with pyridoxamine and thiamine pyrophosphate being the most effective inhibitors in each of their respective families.
  • aminoguanidine only inhibited the rate of post-Amadori AGE formation, and not the final levels of post-Amadori AGE formed (FIG. 4D).
  • FIGS. 4 A-D are graphs which show the effect of vitamin B 1 derivatives and aminoguanidine (AG) on post-Amadori AGE formation in human methemoglobin.
  • Hb (1 mg/ml) was incubated with 1.0 M glucose in the presence and absence of the various indicated derivative in 0.4 M sodium phosphate buffer of pH 7.5 at 37° C. for 3 weeks. Aliquots were assayed by ELISA using R618 anti-AGE antibodies. Concentrations of the inhibitors were 0.5, 3, 15 and 50 mM.
  • Inhibitors used in FIGS. ( 4 A) Thiamine pyrophosphate (TPP); ( 4 B) thiamine monophosphate (TP); ( 4 C) thiamine (T); ( 4 D) aminoguanidine (AG).
  • FIG. 5 is a bar graph which shows a comparison of the inhibition of the glycation of ribonuclease A by thiamine pyrophosphate (TPP), pyridoxamine (PM) and aminoguanidine (AG).
  • TPP thiamine pyrophosphate
  • PM pyridoxamine
  • AG aminoguanidine
  • RNase (1 mg/ml) was incubated with 1.0 M glucose (glc) in the presence and absence of the various indicated derivative in 0.4 M sodium phosphate buffer of pH 7.5 at 37° C. for 6 weeks. Aliquots were assayed by ELISA using R479 anti-AGE antibodies. The indicated percent inhibition was computed from ELISA readings in the absence and presence of the inhibitors at the 6 week time point. Concentrations of the inhibitors were 0.5, 3, 15 and 50 mM.
  • Such assays do not yield information on inhibition of post-Amadori conversion steps to “late” post-Amadori AGE products, since such steps lead to no change in the amount of labeled sugar that is attached to the proteins.
  • Other “glycation” assays have relied on the sugar-induced increase of non-specific protein fluorescence, but this can also be induced by dicarbonyl oxidative fragments of free sugar, such as glycoaldehyde or glyoxal (Hunt et al., 1988, Biochem. 256:205-212), independently of Amadori product formation.
  • pyridoxamine is a candidate amine potentially capable of forming a Schiff-base linkage with the carbonyls of open-chain sugars, with dicarbonyl fragments, with Amadori products, or with post-Amadori intermediates.
  • the mechanism of inhibition of B 1 compounds is not obvious. All the forms contain an amino functionality, so that the marked efficiency of only the pyrophosphate form suggests an important requirement for a strong negative charge.
  • Pentosidine production in RNase was quantitated by HPLC (Sell & Monnier, 1989, J. Biol. Chem. 264:21597-21602; Odetti et al., 1992, Diabetes 41:153-159).
  • Ribose-modified protein samples were hydrolyzed in 6 N HCl for 18 h at 100° C. and then dried in a Speed Vac. The samples were then redissolved, and aliquots were taken into 0.1% trifluoroacetic acid and analyzed by HPLC on a Shimadzu system using a Vydac C-18 column equilibrated with 0.1% TFA.
  • Glycation is much slower with glucose than with ribose (note the difference in time scales between FIG. 7A and FIG. 7B). However, unlike the case with ribose, interruption after 3 days of glycation by 1.0 M glucose produced negligible buildup of precursor to ELISA-reactive AGE antigens (FIG. 7B, dashed curve).
  • FIG. 9 shows such an experiment where the kinetics of intermediate buildup are measured for RNase A in the presence of 0.5 M ribose (solid symbols and curve). For comparison, the amount of AGE present before ribose removal at each interruption point is also shown (open symbols and dashed lines). As expected (cf. FIG. 7A), little AGE is formed prior to removal (or dilution) of ribose, so that ELISA readings after the 5 day secondary incubation periods are mostly a measure of AGE formed after ribose removal. The results in FIG. 9 show that the rate of buildup of intermediate in 0.5 M ribose is exponential and very fast, with a half-time of about 3.3 h. This is about 3-fold more rapid than the observed rate of conversion of the intermediate to antigenic AGEs after interruption (open symbols and dashed curve FIG. 7A).
  • the 24 h ribated sample was simply diluted 100-fold into solutions containing varying final concentrations of ribose ranging from 0.005 M to 0.505 M (FIG. 10A).
  • the rate and extent of AGE formation are clearly seen to be diminished by increasing ribose concentrations.
  • the kinetics appear exponential and do not show the induction period that occurs with uninterrupted glycation (FIGS. 6A and 7A) in high ribose concentrations.
  • FIG. 10B A second experiment (FIG. 10B) was also conducted in which the 24 h interrupted sample was extensively dialyzed in the cold to release free and reversibly bound ribose as well as any inhibitory products that may have formed during the 24 h incubation with ribose. Following this, aliquots were diluted 100-fold into varying concentrations of freshly made ribose, and the formation of antigenic AGE products was monitored as above. There results were nearly identical to the experiment of FIG. 10A where the dialysis step was omitted. In both cases, the rate and extent of AGE formation were diminished by increasing concentrations of ribose, and the kinetics appeared exponential with no induction period.
  • FIG. 20A demonstrates that aminoguanidine has modest effects on blocking the formation of antigenic AGEs in RNase under these conditions, with little inhibition below 50 mM. Approximately 50% inhibition is achieved only at or above 100-250 mM. Note again that in these experiments, the protein was exposed to aminoguanidine only after interruption and removal of free and reversibly bound ribose. Comparable results were also obtained with the interrupted glycation of BSA (FIG. 20B).
  • the data may imply the participation of a second amino group, such as from a neighboring lysine, in the formation of dominant antigenic AGEs.
  • a second amino group such as from a neighboring lysine
  • the interrupted glycation method allowed examination of aminoguanidine efficacy on only post-Amadori steps of AGE formation. Equally important, it allowed studies in the absence of free sugar or dicarbonyl-reactive fragments from free sugar (Wolff & Dean, 1987, Biochem. J. 245:243-250; Wells-Knecht et al., 1995, Biochemistry 34:-3702-3709) that can combine with aminoguanidine.
  • the results of FIG. 20 demonstrate that aminoguanidine has, at best, only a modest effect on post-Amadori AGE formation reactions, achieving 50% inhibition at concentrations above 100-250 mM.
  • aminoguanidine thus predominantly arises either from inhibiting early steps of glycation (Schiff base formation) or from scavenging highly reactive dicarbonyls generated during glycation. Contrary to the original claims, it does not appear to inhibit AGE formation by complexing the Amadori intermediate.
  • interrupted glycation is not limited for kinetic studies. Interrupted glycation can simplify structural studies of glycated proteins and identifying unknown AGEs using techniques such as 13 C NMR that has been used to detect Amadori-adducts of RNase (Neglia et al., 1983, J. Biol. Chem. 258:14279-14283; 1985, J. Biol. Chem. 260:5406-5410). The combined use of structural and kinetic approaches should also be of special interest.
  • candidate AGEs such as the recently proposed (carboxymethyl)lysine (Reddy et al., 1995, Biochemistry 34:10872-10878; cf. Makita et al., 1992, J. Biol. Chem. 267:5133-5138) should display the same kinetics of formation from the reactive intermediate that we have observed.
  • the availability of the interrupted kinetics approach will also help to determine the importance of the Amadori pathway to the formation of this AGE.
  • monitoring of the interrupted glycation reaction by techniques such as 13 C NMR should identify resonances of other candidate antigenic AGEs as being those displaying similar kinetics of appearance.
  • Table I lists the 13 C NMR peaks of the Amadori intermediate of RNase prepared by reaction with C-2 enriched ribose.
  • the downfield peak near 205 ppm is probably due to the carbonyl of the Amadori product.
  • the ability to remove excess free and Schiff base sugars through interrupted glycation will considerably simplify isolation, identification, and structural characterization.
  • Ribonuclease A was reacted for 24 hr with 0.5 M ribose 99% enriched at C-2, following which excess and Schiff base bound ribose was removed by extensive dialysis in the cold. The sample was then warmed back to 37° C. immediately before taking a 2 hr NMR scan. The signals from RNase reacted with natural abundance ribose under identical conditions were then subtracted from the NMR spectrum. Thus all peaks shown are due to enriched C-13 that originated at the C-2 position. Some of the peaks arise from degradation products of the intermediate, and these can be identified by the increase in the peak intensity over time.
  • FIG. 31 shows the NMR spectrum obtained.
  • the interrupted glycation method for following post-Amadori kinetics of AGE formation allows for the rapid quantitative study of “late” stages of the glycation reaction. Importantly, this method allows for inhibition studies that are free of pathways of AGE formation which arise from glycoxidative products of free sugar or Schiff base (Namiki pathway) as illustrated in Scheme I. Thus the interrupted glycation method allows for the rapid and unique identification and characterization of inhibitors of “late” stages of glycation which lead to antigenic AGE formation.
  • pyridoxamine and thiamine pyrophosphate are unique inhibitors of the post-Amadori pathway of AGE formation. Importantly, it was found that efficacy of inhibition of overall glycation of protein, in the presence of high concentrations of sugar, is not predictive of the ability to inhibit the post-Amadori steps of AGE formation where free sugar is removed.
  • pyridoxamine, thiamine pyrophosphate and aminoguanidine are potent inhibitors of AGE formation in the overall glycation of protein by glucose, aminoguanidine differs from the other two in that it is not an effective inhibitor of post-Amadori AGE formation.
  • Aminoguanidine markedly slows the initial rate of AGE formation by ribose under uninterrupted conditions, but has no effect on the final levels of antigenic AGEs produced. Examination of different proteins (RNase, BSA and hemoglobin), confirmed that the inhibition results are generally non-specific as to the protein used, even though there are individual variations in the rates of AGE formation and inhibition.
  • Bovine serum albumin, ribonuclease A, and human hemoglobin were incubated with ribose at 37° C. in 0.4 M sodium phosphate buffer of pH 7.5 containing 0.02% sodium azide.
  • the protein (10 mg/ml or 1 mg/ml), 0.05 M ribose, and prospective inhibitors (at 0.5, 3, 15 and 50 mM) were introduced into the incubation mixture simultaneously. Solutions were kept in the dark in capped tubes. Aliquots were taken and immediately frozen until analyzed by ELISA at the conclusion of the reaction. The incubations were for 3 weeks (Hb) or 6 weeks (RNase, BSA). Glycation reactions were monitored for constant pH throughout the duration of the experiments.
  • Glycation was first carried out by incubating protein (10 mg/ml) with 0.5 M ribose at 37° C. in 0.4 M phosphate buffer at pH 7.5 containing 0.2% sodium azide for 24 h in the absence of inhibitors. Glycation was then interrupted to remove excess and reversibly bound (Schiff base) sugar by extensive dialysis against frequent cold buffer changes at 4° C. The glycated intermediate samples containing maximal amount of Amadori product and little AGE (depending on protein) were then quickly warmed to 37° C. without re-addition of ribose.
  • Kinetics data was routinely fit to mono- or bi-exponential functions using non-linear least squares methods utilizing either SCIENTIST 2.0 (MicroMath, Inc.) or ORIGIN (Microcal, Inc.) software that permit user-defined functions and control of parameters to iterate on. Standard deviations of the parameters of the fitted functions (initial and final ordinate values and rate constants) were returned as measures of the precision of the fits. Apparent half-times for bi-exponential kinetics fits were determined with the “solve” function of MathCad software (MathSoft, Inc.).
  • FIG. 18 shows the effects of pyridoxamine (FIG. 18A), pyridoxal phosphate (FIG.
  • FIG. 20 shows the results of testing aminoguanidine for inhibition of post-Amadori AGE formation kinetics with both BSA and RNase. At 50 mM, inhibition was about 20% in the case of BSA (FIG. 20B), and less than 15% with RNase (FIG. 20A). The possibility of inhibition by simple amino-containing functionalities was also tested using N + -acetyl-L-lysine (FIG. 21), which contains only a free N ⁇ -amino group. N ⁇ -acetyl-L-lysine at up to 50 mM failed to exhibit any significant inhibition of AGE formation.
  • aminoguanidine is an apparently potent inhibitor of many manifestations of nonenzymatic glycation (Brownlee et al., 1986; Brownlee, 1992, 1994, 1995).
  • the inhibitory effects of aminoguanidine on various phenomena that are induced by reducing sugars are widely considered as proof of the involvement of glycation in many such phenomena.
  • Aminoguanidine has recently entered into a second round of Phase III clinical trials (as pimagedine) for ameliorating the complications of diabetes thought to be caused by glycation of connective tissue proteins due to high levels of sugar.
  • FIG. 22 are bar graphs which depict summarized comparative data of percent inhibition at defined time points using various concentrations of inhibitor.
  • FIG. 22A graphs the data for inhibition after interrupted glycation of RNase AGE formation in ribose.
  • FIG. 22B graphs the data for inhibition after interrupted glycation of BSA AGE formation by ribose.
  • Hyperglycemia can be rapidly induced (within one or two days) in rats by administration of streptozocin (aka. streptozotocin, STZ) or alloxan. This has become a common model for diabetes melitus. However, these rats manifest nephropathy only after many months of hyperglycemia, and usually just prior to death from end-stage renal disease (ESRD). It is believed that this pathology is caused by the irreversible glucose chemical modification of long-lived proteins such as collagen of the basement membrane. STZ-diabetic rats show albuminuria very late after induction of hyperglycemia, at about 40 weeks usually only just prior to death.
  • Rats were then kept for 4 days with no further ribose administration, at which time they were sacrificed and the following physiological measurements were determined: (i) initial and final body weight; (ii) final stage kidney weight; (iii) initial and final tail-cuff blood pressure; (iv) creatinine clearance per 100 g body weight.
  • Tail-cuff blood pressure increased from 66 ⁇ 4 to 83 ⁇ 3 to rats treated with low ribose (1 ⁇ 50 mM), and from 66 ⁇ 4 to 106 ⁇ 5 for rats treated with high ribose (2 ⁇ 300 mM). These results are shown in the bar graph of FIG. 23.
  • Groups of rats (3-6) were intraperitoneally given 0.3 M “low ribose dose” (LR) or 1.0 M “high ribose dose” (HR) by twice-daily injections for either (i) 1 day, (ii) a “short-term” (S) of 4 days, or (iii) a “long-term” (L) of 8 days. Additionally, these concentrations of ribose were included in drinking water.
  • LR low ribose dose
  • HR high ribose dose
  • ribose plus low dose (“LP” ) of pyridoxamine 25 mg/kg body weight injected as 0.5 ml with 9 cc ribose
  • ribose plus high dose (“HT”) of thiamine pyrophosphate 250 mg/kg body weight injected as 0.5 ml with 9 cc ribose
  • Intervention compounds were pre-administered for one day prior to introducing them with ribose.
  • diabetic nephropathy Persistent hyperglycemia in diabetes mellitus leads to diabetic nephropathy in perhaps one third of human patients. Clinically, diabetic nephropathy is defined by the presence of:
  • Renal function depends on blood flow (not measured) and the glomerular clearance, which can be measured by either inulin clearance (not measured) or creatinine clearance. Glomerular permeability is measured by albumin filtration rate, but this parameter is quite variable. It is also a log-distribution function: a hundred-fold increase in albumin excretion represents only a two-fold decrease in filtration capacity.
  • ribose appears to very rapidly induce manifestations of diabetic nephropathy, as reflected in hypertension, creatinine clearance and albuminuria, even though the latter is not large.
  • hyperglycemia is rapidly established in 1-2 days, but clinical manifestations of diabetic nephropathy arise very late, perhaps as much as 40 weeks for albuminuria.
  • albuminuria is highly variable from day to day and from animal to animal, although unlike humans, most STZ rats do eventually develop nephropathy.
  • pyridoxamine at 25 mg/kg body weight appears to effectively prevent two of the three manifestations usually attributed to diabetes, namely the impairment of creatinine clearance and albumin filtration. It did so as effectively as aminoguanidine.
  • the effectiveness of thiamine pyrophosphate was not manifest, but it should be emphasized that this may be due to its use at elevated concentrations of 250 mg/kg body weight. Pyridoxamine would have appeared much less effective if only the results at 250 mg/kg body weight are considered.
  • a typical adult human being of average size weighs between 66-77 Kg.
  • diabetic patients may tend to be overweight and can be over 112 Kg.
  • a range of doses for administration of pyridoxamine or thiamine pyrophosphate that is predicted to be effective for inhibiting post-Amadori AGE formation and thus inhibiting related pathologies would fall in the range of 1 mg/100 g body weight to 200 mg/100 g body weight.
  • the appropriate range when co-administered with aminoguanidine will be similar. Calculated for an average adult of 75 Kg, the range (at 10 mg/1 Kg body weight) can be approximately 750 mg to upwards of 150 g (at 2 g/1 Kg body weight). This will naturally vary according to the particular patient.

Abstract

The instant invention provides compositions and methods for modeling post-Amadori AGE formation and the identification and characterization of effective inhibitors of post-Amadori AGE formation, and such identified inhibitor compositions.

Description

  • This application claims priority to U.S. Provisional Application for Patent Ser. No. 60/003,268, filed Sep. 12, 1995, the contents of which are hereby incorporated in the entirety.[0001]
  • STATEMENT OF GOVERNMENT RIGHTS
  • [0002] Some of the work disclosed has been supported in part by NIH Grant DK 43507, therefore, the United States Government may have certain rights in the invention.
  • BACKGROUND OF THE INVENTION
  • The instant invention is in the field of Advanced Glycation End-products (AGEs), their formation, detection, identification, inhibition, and inhibitors thereof. [0003]
  • Protein Aging and Advanced Glycosylation End-Products [0004]
  • Nonenzymatic glycation by glucose and other reducing sugars is an important post-translational modification of proteins that has been increasingly implicated in diverse pathologies. Irreversible nonenzymatic glycation and crosslinking through a slow, glucose-induced process may mediate many of the complications associated with diabetes. Chronic hyperglycemia associated with diabetes can cause chronic tissue damage which can lead to complications such as retinopathy, nephropathy, and atherosclerotic disease. (Cohen and Ziyadeh, 1996, [0005] J. Amer. Soc. Nephrol. 7:183-190). It has been shown that the resulting chronic tissue damage associated with long-term diabetes mellitus arise in part from in situ immune complex formation by accumulated immunoglobulins and/or antigens bound to long-lived structural proteins that have undergone Advanced Glycosylation End-product (AGE) formation, via non-enzymatic glycosylation (Brownlee et al., 1983, J. Exp. Med. 158:1739-1744). The primary protein target is thought to be extra-cellular matrix associated collagen. Nonenzymatic glycation of proteins, lipids, and nucleic acids may play an important role in the natural processes of aging. Recently protein glycation has been associated with β-amyloid deposits and formation of neurofibrillary tangles in Alzheimer disease, and possibly other neurodegenerative diseases involving amyloidosis (Colaco and Harrington, 1994, NeuroReport 5: 859-861). Glycated proteins have also been shown to be toxic, antigenic, and capable of triggering cellular injury responses after uptake by specific cellular receptors (see for example, Vlassara, Bucala & Striker, 1994, Lab. Invest. 70:138-151; Vlassara et al., 1994, PNAS(USA) 91:11704-11708; Daniels & Hauser, 1992, Diabetes 41:1415-1421; Brownlee, 1994, Diabetes 43:836-841; Cohen et al., 1994, Kidney Int. 45:1673-1679; Brett et al., 1993, Am. J. Path. 143:1699-1712; and Yan et al., 1994, PNAS(USA) 91:7787-7791).
  • The appearance of brown pigments during the cooking of food is a universally recognized phenomenon, the chemistry of which was first described by Maillard in 1912, and which has subsequently led to research into the concept of protein aging. It is known that stored and heat-treated foods undergo nonenzymatic browning that is characterized by crosslinked proteins which decreases their bioavailibility. It was found that this Maillard reaction occurred in vivo as well, when it was found that a glucose was attached via an Amadori rearrangement to the amino-terminal of the α-chain of hemoglobin. [0006]
  • The instant disclosure teaches previously unknown, and unpredicted mechanism of formation of post-Amadori advanced glycation end products (Maillard products; AGEs) and methods for identifying and characterizing effective inhibitors of post-Amadori AGE formation. The instant disclosure demonstrates the unique isolation and kinetic characterization of a reactive protein intermediate competent in forming post-Amadori AGEs, and for the first time teaching methods which allow for the specific elucidation and rapid quantitative kinetic study of “late” stages of the protein glycation reaction. [0007]
  • In contrast to such “late” AGE formation, the “early” steps of the glycation reaction have been relatively well characterized and identified for several proteins (Harding, 1985, [0008] Adv. Protein Chem. 37:248-334; Monnier & Baynes eds., 1989, The Maillard Reaction in Aging, Diabetes, and Nutrition (Alan R. Liss, New York); Finot et al., 1990, eds. The Maillard Reaction in Food Processing, Human Nutrition and Physiology (Birkhauser Verlag, Basel)). Glycation reactions are known to be initiated by reversible Schiff-base (aldimine or ketimine) addition reactions with lysine side-chain ε-amino and terminal α-amino groups, followed by essentially irreversible Amadori rearrangements to yield ketoamine products e.g. 1-amino-1-deoxy-ketoses from the reaction of aldoses (Baynes et al., 1989, in The Maillard Reaction in Aging, Diabetes, and Nutrition, ed. Monnier and Baynes, (Alan R. Liss, New York, pp 43-67). Typically, sugars initially react in their open-chain (not the predominant pyranose and furanose structures) aldehydo or keto forms with lysine side chain ε-amino and terminal α-amino groups through reversible Schiff base condensation (Scheme I). The resulting aldimine or ketimine products then undergo Amadori rearrangements to give ketoamine Amadori products, i.e. 1-amino-1-deoxy-ketoses from the reaction of aldoses (Means & Chang, 1982, Diabetes 31, Suppl. 3:1-4; Harding, 1985, Adv. Protein Chem. 37:248-334). These Amadori products then undergo, over a period of weeks and months, slow and irreversible Maillard “browning” reactions, forming fluorescent and other products via rearrangement, dehydration, oxidative fragmentation, and cross-linking reactions. These post-Amadori reactions, (slow Maillard “browning” reactions), lead to poorly characterized Advanced Glycation End-products (AGEs).
  • As with Amadori and other glycation intermediaries, free glucose itself can undergo oxidative reactions that lead to the production of peroxide and highly reactive fragments like the dicarbonyls glyoxal and glycoaldehyde. Thus the elucidation of the mechanism of formation of a variety of AGEs has been extremely complex since most in vitro studies have been carried out at extremely high sugar concentrations. [0009]
  • In contrast to the relatively well characterized formation of these “early” products, there has been a clear lack of understanding of the mechanisms of forming the “late” Maillard products produced in post-Amadori reactions, because of their heterogeneity, long reaction times, and complexity. The lack of detailed information about the chemistry of the “late” Maillard reaction stimulated research to identify fluorescent AGE chromophores derived from the reaction of glucose with amino groups of polypeptides. One such chromophore, 2-(2-furoyl)-4(5)-(2-furanyl)-1H-imidazole (FFI) was identified after nonenzymatic browning of bovine serum albumin and polylysine with glucose, and postulated to be representative of the chromophore present in the intact polypeptides. (Pongor et al., 1984, [0010] PNAS(USA) 81:2684-2688). Later studies established FFI to be an artifact formed during acid hydrolysis for analysis.
  • A series of U.S. Patents have issued in the area of inhibition of protein glycosylation and cross-linking of protein sugar amines based upon the premise that the mechanism of such glycosylation and cross-linking occurs via saturated glycosylation and subsequent cross-linking of protein sugar amines via a single basic, and repeating reaction. These patents include U.S. Pat. Nos. 4,665,192; 5,017,696; 4,758,853; 4,908,446; 4,983,604; 5,140,048; 5,130,337; 5,262,152; 5,130,324; 5,272,165; 5,221,683; 5,258,381; 5,106,877; 5,128,360; 5,100,919; 5,254,593; 5,137,916; 5,272,176; 5,175,192; 5,218,001; 5,238,963; 5,358,960; 5,318,982; and 5,334,617. (All U.S. Patents cited are hereby incorporated by reference in their entirety). [0011]
  • The focus of these U.S. Patents, are a method for inhibition of AGE formation focused on the carbonyl moiety of the early glycosylation Amadori product, and in particular the most effective inhibition demonstrated teaches the use of exogenously administered aminoguanidine. The effectiveness of aminoguanidine as an inhibitor of AGE formation is currently being tested in clinical trials. [0012]
  • Inhibition of AGE formation has utility in the areas of, for example, food spoilage, animal protein aging, and personal hygiene such as combating the browning of teeth. Some notable, though quantitatively minor, advanced glycation end-products are pentosidine and N[0013] π-carboxymethyllysine (Sell and Monnier, 1989, J. Biol. Chem. 264:21597-21602; Ahmed et al., 1986, J. Biol. Chem. 261:4889-4894).
  • The Amadori intermediary product and subsequent post-Amadori AGE formation, as taught by the instant invention, is not fully inhibited by reaction with aminoguanidine. Thus, the formation of post-Amadori AGEs as taught by the instant disclosure occurs via an important and unique reaction pathway that has not been previously shown, or even previously been possible to demonstrate in isolation. It is a highly desirable goal to have an efficient and effective method for identifying and characterizing effective post-Amadori AGE inhibitors of this “late” reaction. By providing efficient screening methods and model systems, combinatorial chemistry can be employed to screen candidate compounds effectively, and thereby greatly reducing time, cost, and effort in the eventual validation of inhibitor compounds. It would be very useful to have in vivo methods for modeling and studying the effects of post-Amadori AGE formation which would then allow for the efficient characterization of effective inhibitors. [0014]
  • Inhibitory compounds that are biodegradeble and/or naturally metabolized are more desirable for use as therapeutics than highly reactive compounds which may have toxic side effects, such as aminoguanidine. [0015]
  • SUMMARY OF THE INVENTION
  • In accordance with the present invention, a stable post-Amadori advanced glycation end-product (AGE) precursor has been identified which can then be used to rapidly complete the post-Amadori conversion into post-Amadori AGEs. This stable product is a presumed sugar saturated Amadori/Schiff base product produced by the further reaction of the early stage protein/sugar Amadori product with more sugar. In a preferred embodiment, this post-Amadori/Schiff base intermediary has been generated by the reaction of target protein with ribose sugar. [0016]
  • The instant invention provides for a method of generating stable protein-sugar AGE formation intermediary precursors via a novel method of high sugar inhibition. In a preferred embodiment the sugar used is ribose. [0017]
  • The instant invention provides for a method for identifying an effective inhibitor of the formation of late Maillard products comprising: generating stable protein-sugar post-Amadori advanced glycation end-product intermediates by incubating a protein with sugar at a sufficient concentration and for sufficient length of time to generate stable post-Amadori AGE intermediates; contacting said stable protein-sugar post-Amadori advanced glycation end-product intermediates with an inhibitor candidate; identifying effective inhibition by monitoring the formation of post-Amadori AGEs after release of the stable protein-sugar post-Amadori advanced glycation end-product intermediates from sugar induced equilibrium. Appropriate sugars include, and are not limited to ribose, lyxose, xylose, arabinose, glucose, fructose, maltose, lactose, mannose, fructose and galactose. In a preferred embodiment the sugar used is ribose. [0018]
  • The instant invention teaches that an effective inhibitor of post-Amadori AGE formation via “late” reactions can be identified and characterized by the ability to inhibit the formation of post-Amadori AGE endproducts in an assay comprising; generating stable protein-sugar post-Amadori advanced glycation end-product intermediates by incubating a protein with sugar at a sufficient concentration and for sufficient length of time to generate stable post-Amadori AGE intermediates; contacting said stable protein-sugar post-Amadori advanced glycation end-product intermediates with an inhibitor candidate; identifying effective inhibition by monitoring the formation of post-Amadori AGEs after release of the stable protein-sugar post-Amadori advanced glycation end-product intermediates from sugar induced equilibrium. In a preferred embodiment the assay uses ribose. [0019]
  • Thus the methods of the instant invention allow for the rapid screening of candidate post-Amadori AGE formation inhibitors for effectiveness, greatly reducing the cost and amount of work required for the development of effective small molecule inhibitors of post-Amadori AGE formation. The instant invention teaches that effective inhibitors of post-Amadori “late” reactions of AGE formation include derivatives of vitamin B[0020] 6 and vitamin B1, in the preferred embodiment the specific species being pyridoxamine and thiamine pyrophosphate.
  • The instant invention teaches new methods for rapidly inducing diabetes like pathologies in rats comprising administering ribose to the subject animal. Further provided for is the use of identified inhibitors pyridoxamine and thiamine pyrophosphate in vivo to inhibit post-Amadori AGE induced pathologies.[0021]
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 is a series of graphs depicting the effect of vitamin B[0022] 6 derivatives on AGE formation in bovine serum albumin (BSA). FIG. 1A Pyridoxamine (PM); FIG. 1B pyridoxal phosphate (PLP); FIG. 1C pyridoxal (PL); FIG. 1D pyridoxine (PN).
  • FIG. 2 is a series of graphs depicting the effect of vitamin B[0023] 1 derivatives and aminoguanidine (AG) on AGE formation in bovine serum albumin. FIG. 2A Thiamine pyrophosphate (TPP); FIG. 2B thiamine monophosphate (TP); FIG. 2C thiamine (T); FIG. 2D aminoguanidine (AG).
  • FIG. 3 is a series of graphs depicting the effect of vitamin B[0024] 6 derivatives on AGE formation in human methemoglobin (Hb). FIG. 3A Pyridoxamine (PM); FIG. 3B pyridoxal phosphate (PLP); FIG. 3C pyridoxal (PL); FIG. 3D pyridoxine (PN).
  • FIG. 4 is a series of graphs depicting the effect of vitamin B[0025] 1 derivatives and aminoguanidine (AG) on AGE formation in human methemoglobin. FIG. 2A Thiamine pyrophosphate (TPP); FIG. 2B thiamine monophosphate (TP); FIG. 2C thiamine (T); FIG. 2D aminoguanidine (AG).
  • FIG. 5 is a bar graph comparison of the inhibition of the glycation of ribonuclease A by thiamine pyrophosphate (TPP), pyridoxamine (PM) and aminoguanidine (AG). [0026]
  • FIG. 6A is a graph of the kinetics of glycation of RNase A (10 mg/mL) by ribose as monitored by ELISA. FIG. 6B is a graph showing the dependence of reciprocal half-times on ribose concentration at pH 7.5. [0027]
  • FIG. 7 are two graphs showing a comparison of uninterrupted and interrupted glycation of RNase by glucose ([0028] 7B) and ribose (7A), as detected by ELISA.
  • FIG. 8 are two graphs showing kinetics of pentosidine fluorescence (arbitrary units) increase during uninterrupted and interrupted ribose glycation of RNase. FIG. 8A Uninterrupted glycation in the presence of 0.05 M ribose. FIG. 8B Interrupted glycation after 8 and 24 hours of incubation. [0029]
  • FIG. 9 is a graph which shows the kinetics of reactive intermediate buildup. [0030]
  • FIG. 10 are graphs of Post-Amadori inhibition of AGE formation by ribose. FIG. 10A graphs data where aliquots were diluted into inhibitor containing buffers at [0031] time 0. FIG. 10B graphs data where samples were interrupted at 24 h, and then diluted into inhibitor containing buffers.
  • FIG. 11 is a graph showing dependence of the initial rate of formation of antigenic AGE on pH following interruption of glycation. [0032]
  • FIG. 12 are two graphs showing the effect of pH jump on ELISA detected AGE formation after interrupted glycation. Interrupted samples left 12 days at 37° C. in pH 5.0 buffer produced substantial AGEs (33%; FIG. 12 B) when pH was changed to 7.5, as compared to the normal control sample not exposed to low pH (FIG. 12 A) [0033]
  • FIG. 13 is a series of graphs depicting the effect of vitamin B[0034] 6 derivatives on AGE formation during uninterrupted glycation of ribonuclease A (RNase A) by ribose. FIG. 13A Pyridoxamine (PM); FIG. 13B pyridoxal-5′-phosphate (PLP); FIG. 13C pyridoxal (PL); FIG. 13D pyridoxine (PN).
  • FIG. 14 is a series of graphs depicting the effect of vitamin B[0035] 1 derivatives and aminoguanidine (AG) on AGE formation during uninterrupted glycation of ribonuclease A (RNase A) by ribose. FIG. 14A Thiamine pyrophosphate (TPP); FIG. 14B thiamine monophosphate (TP); FIG. 14C thiamine (T); FIG. 14D aminoguanidine (AG).
  • FIG. 15 is a series of graphs depicting the effect of vitamin B[0036] 6 derivatives on AGE formation during uninterrupted glycation of bovine serum albumin (BSA) by ribose. FIG. 15A Pyridoxamine (PM); FIG. 15B pyridoxal-5′-phosphate (PLP); FIG. 15C pyridoxal (PL); FIG. 15D pyridoxine (PN).
  • FIG. 16 is a series of graphs depicting the effect of vitamin B[0037] 1 derivatives and aminoguanidine (AG) on AGE formation during uninterrupted glycation of bovine serum albumin (BSA) by ribose. FIG. 16A Thiamine pyrophosphate (TPP); FIG. 16B thiamine monophosphate (TP); FIG. 16C thiamine (T); FIG. 16D aminoguanidine (AG).
  • FIG. 17 is a series of graphs depicting the effect of vitamin B[0038] 6 derivatives on AGE formation during uninterrupted glycation of human methemoglobin (Hb) by ribose. FIG. 17A Pyridoxamine (PM); FIG. 17B pyridoxal-5′-phosphate (PLP); FIG. 17C pyridoxal (PL); FIG. 17D pyridoxine (PN).
  • FIG. 18 is a series of graphs depicting the effect of vitamin B[0039] 6 derivatives on post-Amadori AGE formation after interrupted glycation by ribose. FIG. 18A BSA and Pyridoxamine (PM); FIG. 18B BSA and pyridoxal-5′-phosphate (PLP); FIG. 18C BSA and pyridoxal (PL); FIG. 18D RNase and pyridoxamine (PM)
  • FIG. 19 are graphs depicting the effect of thiamine pyrophosphate on post-Amadori AGE formation after interrupted glycation by ribose. FIG. 19A RNase, FIG. 19B BSA. [0040]
  • FIG. 20 are graphs depicting the effect of aminoguanidine on post-Amadori AGE formation after interrupted glycation by ribose. FIG. 20A RNase, FIG. 20B BSA. [0041]
  • FIG. 21 is a graph depicting the effect of N[0042] α-acetyl-L-lysine on post-Amadori AGE formation after interrupted glycation by ribose.
  • FIG. 22 are bar graphs showing a comparison of post-Amadori inhibition of AGE formation by thiamine pyrophosphate (TPP), pyridoxamine (PM) and aminoguanidine (AG) after interrupted glycation of RNase (FIG. 22A) and BSA (FIG. 22B) by ribose. [0043]
  • FIG. 23 is a bar graph showing the effects of Ribose treatment in vivo alone on rat tail-cuff blood pressure. Treatment was with 0.05 M, 0.36 M, and 1 M Ribose (R) injected for 1, 2 or 8 Days (D). [0044]
  • FIG. 24 is a bar graph showing the effects of Ribose treatment in vivo alone on rat creatinine clearance (Clearance per 100 g Body Weight). Treatment was with 0.05 M, 0.30 M, and 1 M Ribose (R) injected for 1, 2 or 8 Days (D). [0045]
  • FIG. 25 is a bar graph showing the effects of Ribose treatment in vivo alone on rat Albuminuria (Albumin effusion rate). Treatment was with 0.30 M, and 1 M Ribose (R) injected for 1, 2 or 8 Days (D). [0046]
  • FIG. 26 is a bar graph showing the effects of inhibitor treatment in vivo, with or without ribose, on rat tail-cuff blood pressure. Treatment groups were: 25 mg/100 g body weight aminoguanidine (AG); 25 or 250 mg/100 g body weight Pyridoxamine (P); 250 mg/100 g body weight Thiamine pyrophosphate (T), or with 1 M Ribose (R). [0047]
  • FIG. 27 is a bar graph showing the effects of inhibitor treatment in vivo, with or without ribose, on rat creatinine clearance (Clearance per 100 g body weight). Treatment groups were: 25 mg/100 g body weight aminoguanidine (AG); 25 or 250 mg/100 g body weight Pyridoxamine (P); 250 mg/100 g body weight Thiamine pyrophosphate (T), or with 1 M Ribose (R). [0048]
  • FIG. 28 is a bar graph showing the effects of inhibitor treatment in vivo without ribose, and ribose alone on rat Albuminuria (Albumin effusion rate). Treatment groups were: 25 mg/100 g body weight aminoguanidine (AG); 250 mg/100 g body weight Pyridoxamine (P); 250 mg/100 g body weight Thiamine pyrophosphate (T), or treatment with 1 M Ribose (R) for 8 days (D). Control group had no treatment. [0049]
  • FIG. 29 is a bar graph showing the effects of inhibitor treatment in vivo, with 1 M ribose , on rat Albuminuria (Albumin effusion rate). Treatment groups were: 25 mg/100 g body weight aminoguanidine (AG); 25 and 250 mg/100 g body weight Pyridoxamine (P); 250 mg/100 g body weight Thiamine pyrophosphate (T), or treatment with 1 M Ribose (R) for 8 days (D) alone. Control group had no treatment. [0050]
  • FIG. 30A depicts [0051] Scheme 1 showing a diagram of AGE formation from protein. FIG. 30B depicts Scheme 2, a chemical structure of aminoguanidine. FIG. 30C depicts Scheme 3, chemical structures for thiamine, thiamine-5′-phosphate, and thiamine pyrophosphate. FIG. 30D depicts Scheme 4, chemical structures of pyridoxine, pyridoxamine, pyridoxal-5′-phosphate, and pyridoxal. FIG. 30E depicts Scheme 5, kinetics representation of AGE formation. FIG. 30F depicts Scheme 6, kinetics representation of AGE formation and intermediate formation.
  • FIG. 31 shows a 125 MHz C-13 NMR Resonance spectrum of Riobonuclease Amadori Intermediate prepared by 24 HR reaction with 99% [2-C13]Ribose.[0052]
  • DETAILED DESCRIPTION
  • Animal Models for Protein Aging [0053]
  • Alloxan induced diabetic Lewis rats have been used as a model for protein aging to demonstrate the in vivo effectiveness of inhibitors of AGE formation. The correlation being demonstrated is between inhibition of late diabetes related pathology and effective inhibition of AGE formation (Brownlee, Cerami, and Vlassara, 1988, [0054] New Eng. J. Med. 318(20):1315-1321). Streptozotocin induction of diabetes in Lewis rats, New Zealand White rabbits with induced diabetes, and genetically diabetic BB/Worcester rats have also been utilized, as described in, for example, U.S. Pat. No. 5,334,617 (incorporated by reference). A major problem with these model systems is the long time period required to demonstrate AGE related injury, and thus to test compounds for AGE inhibition. For example, 16 weeks of treatment was required for the rat studies described in U.S. Patent 5,334,617, and 12 weeks for the rabbit studies. Thus it would be highly desirable and useful to have a model system for AGE related diabetic pathology that will manifest in a shorter time period, allowing for more efficient and expeditious determination of AGE related injury and the effectiveness of inhibitors of post-Amadori AGE formation.
  • Antibodies to AGEs [0055]
  • An important tool for studying AGE formation is the use of polyclonal and monoclonal antibodies that are specific for AGEs elicited by the reaction of several sugars with a variety of target proteins. The antibodies are screened for resultant specificity for AGEs that is independent of the nature of the protein component of the AGE (Nakayama et al., 1989, [0056] Biochem. Biophys. Res. Comm. 162: 740-745; Nakayama et al., 1991, J. Immunol. Methods 140: 119-125; Horiuchi et al., 1991, J. Biol. Chem. 266: 7329-7332; Araki et al., 1992, J. Biol. Chem. 267: 10211-10214; Makita et al., 1992, J. Biol. Chem. 267: 5133-5138). Such antibodies have been used to monitor AGE formation in vivo and in vitro.
  • Thiamine—Vitamin B[0057] 1
  • The first member of the Vitamin B complex to be identified, thiamine is practically devoid of pharmacodynamic actions when given in usual therapeutic doses; and even large doses were not known to have any effects. Thiamine pyrophosphate is the physiologically active form of thiamine, and it functions mainly in carbohydrate metabolism as a coenzyme in the decarboxylation of α-keto acids. Tablets of thiamine hydrochloride are available in amounts ranging from 5 to 500 mg each. Thiamine hydrochloride injection solutions are available which contain 100 to 200 mg/ml. [0058]
  • For treating thiamine deficiency, intravenous doses of as high as 100 mg/L of parenteral fluid are commonly used, with the typical dose of 50 to 100 mg being administered. GI absorption of thiamine is believed to be limited to 8 to 15 mg per day, but may be exceed by oral administration in divided doses with food. [0059]
  • Repeated administration of glucose may precipitate thiamine deficiency in under nourished patients, and this has been noted during the correction of hyperglycemia. [0060]
  • Surprisingly, the instant invention has found, as shown by in vitro testing, that administration of thiamine pyrophosphate at levels above what is normally found in the human body or administered for dietary therapy, is an effective inhibitor of post-Amadori antigenic AGE formation, and that this inhibition is more complete than that possible by the administration of aminoguanidine. [0061]
  • Pyridoxine—Vitamin B[0062] 6
  • Vitamin B[0063] 6 is typically available in the form of pyridoxine hydrochloride in over-the-counter preparations available from many sources. For example Beach pharmaceuticals Beelith Tablets contain 25 mg of pyridoxine hydrochloride that is equivalent to 20 mg of B6, other preparations include Marlyn Heath Care Marlyn Formula 50 which contain 1 mg of pyridoxine HCl and Marlyn Formula 50 Mega Forte which contains 6 mg of pyridoxine HCl, Wyeth-Ayerst Stuart Prenatal® tablets which contain 2.6 mg pyridoxine HCl, J&J-Merck Corp. Stuart Formula® tablets contain 2 mg of pyridoxine HCl, and the CIBA Consumer Sunkist Children's chewable multivitamins which contain 1.05 mg of pyridoxine HCl, 150% of the U.S. RDA for children 2 to 4 years of age, and 53% of the U.S. RDA for children over 4 years of age and adults. (Physician's Desk Reference for nonprescription drugs, 14th edition (Medical Economics Data Production Co., Montvale, N.J., 1993).
  • There are three related forms of pyridoxine, which differ in the nature of the substitution on the carbon atom in [0064] position 4 of the pyridine nucleus: pyridoxine is a primary alcohol, pyridoxal is the corresponding aldehyde, and pyridoxamine contains an aminomethyl group at this position. Each of these three forms can be utilized by mammals after conversion by the liver into pyridoxal-5′-phosphate, the active form of the vitamin. It has long been believed that these three forms are equivalent in biological properties, and have been treated as equivalent forms of vitamin B6 by the art. The Council on Pharmacy and Chemistry has assigned the name pyridoxine to the vitamin.
  • The most active antimetabolite to pyridoxine is 4-deoxypyridoxine, for which the antimetabolite activity has been attributed to the formation in vivo of 4-deoxypyridoxine-5-phosphate, a competitive inhibitor of several pyridoxal phosphate-dependent enzymes. The pharmacological actions of pyridoxine are limited, as it elicits no outstanding pharmacodynamic actions after either oral or intravenous administration, and it has low acute toxicity, being water soluble. It has been suggested that neurotoxicity may develop after prolonged ingestion of as little as 200 mg of pyridoxine per day. Physiologically, as a coenzyme, pyridoxine phosphate is involved in several metabolic transformations of amino acids including decarboxylation, transamination, and racemization, as well as in enzymatic steps in the metabolism of sulfur-containing and hydroxy-amino acids. In the case of transamination, pyridoxal phosphate is aminated to pyridoxamine phosphate by the donor amino acid, and the bound pyridoxamine phosphate is then deaminated to pyridoxal phosphate by the acceptor α-keto acid. Thus vitamin B complex is known to be a necessary dietary supplement involved in specific breakdown of amino acids. For a general review of the vitamin B complex see [0065] The Pharmacological Basis of Therapeutics, 8th edition, ed. Gilman, Rall, Nies, and Taylor (Pergamon Press, New York, 1990, pp. 1293-4; pp. 1523-1540).
  • Surprisingly, the instant invention has discovered that effective dosages of the metabolically transitory pyridoxal amine form of vitamin B[0066] 6 (pyridoxamine), at levels above what is normally found in the human body, is an effective inhibitor of post-Amadori antigenic AGE formation, and that this inhibition may be more complete than that possible by the administration of aminoguanidine.
  • Formation of Stable Amadori/Schifff Base Intermediary [0067]
  • The typical study of the reaction of a protein with glucose to form AGEs has been by ELISA using antibodies directed towards antigenic AGEs, and the detection of the production of an acid-stable fluorescent AGE, pentosidine, by HPLC following acid hydrolysis. Glycation of target proteins (i.e. BSA or RNase A) with glucose and ribose were compared by monitoring ELISA reactivity of polyclonal rabbit anti-Glucose-AGE-RNase and anti-Glucose-AGE-BSA antibodies. The antigen was generated by reacting 1 M glucose with RNase for 60 days and BSA for 90 days. The antibodies (R618 and R479) were screened and showed reactivity with only AGEs and not the protein, except for the carrier immunogen BSA. [0068]
  • EXAMPLE 1
  • Thiamine Pyrophosphate and Pyridoxamine Inhibit the Formation of Antigenic Advanced Glycation End-Products from Glucose: Comparison with Aminoguanidine [0069]
  • Some B[0070] 6 vitamers, especially pyridoxal phosphate (PLP), have been previously proposed to act as “competitive inhibitors” of early glycation, since as aldehydes they themselves can form Schiff bases adducts with protein amino groups (Khatami et al., 1988, Life Sciences 43:1725-1731) and thus limit the amount of amines available for glucose attachment. However, effectiveness in limiting initial sugar attachment is not a predictor of inhibition of the conversion of any Amadori products formed to AGEs. The instant invention describes inhibitors of “late” glycation reactions as indicated by their effects on the in vitro formation of antigenic AGEs (Booth et al., 1996, Biochem. Biophys. Res. Com. 220:113-119).
  • Chemicals [0071]
  • Bovine pancreatic ribonuclease A (RNase) was chromatographically pure, aggregate-free grade from Worthington Biochemicals. Bovine Serum albumin (BSA; fraction V, fatty-acid free), human methemoglobin (Hb), D-glucose, pyridoxine, pyridoxal, [0072] pyridoxal 5′phosphate, pyridoxamine, thiamine, thiamine monophosphate, thiamine pyrophosphate, and goat alkaline phosphatase-conjugated anti-rabbit IgG were all from Sigma Chemicals. Aminoguanidine hydrochloride was purchased from Aldrich Chemicals.
  • Uninterrupted Glycation with Glucose [0073]
  • Bovine serum albumin, ribonuclease A, and human hemoglobin were incubated with glucose at 37° C. in 0.4 M sodium phosphate buffer of pH 7.5 containing 0.02% sodium azide. The protein, glucose (at 1.0 M), and prospective inhibitors (at 0.5, 3, 15 and 50 mM) were introduced into the incubation mixture simultaneously. Solutions were kept in the dark in capped tubes. Aliquots were taken and immediately frozen until analyzed by ELISA at the conclusion of the reaction. The incubations were for 3 weeks (Hb) or 6 weeks (RNase, BSA). [0074]
  • Preparation of Polyclonal Antibodies to AGE Proteins [0075]
  • Immunogen preparation followed earlier protocols (Nakayama et al., 1989, [0076] Biochem. Biophys. Res. Comm. 162:740-745; Horiuchi et al., 1991, J. Biol. Chem. 266:7329-7332; Makita et al., 1992, J. Biol. Chem. 267:5133-5138). Briefly, immunogen was prepared by glycation of BSA (R479 antibodies) or RNase (R618 antibodies) at 1.6 g protein in 15 ml for 60-90 days using 1.5 M glucose in 0.4 M sodium phosphate buffer of pH 7.5 containing 0.05% sodium azide at pH 7.4 and 37° C. New Zealand white rabbit males of 8-12 weeks were immunized by subcutaneous administration of a 1 ml solution containing 1 mg/ml of glycated protein in Freund's adjuvant. The primary injection used the complete adjuvant and three boosters were made at three week intervals with Freund's incomplete adjuvant. Rabbits were bled three weeks after the last booster. The serum was collected by centrifugation of clotted whole blood. The antibodies are AGE-specific, being unreactive with either native proteins (except for the carrier) or with Amadori intermediates. The polyclonal anti-AGE antibodies have proven to be a sensitive and valuable analytical tool for the study of “late” AGE formation in vitro and in vivo. The nature of the dominant antigenic AGE epitope or hapten remains in doubt, although recently it has been proposed that the protein glycoxidation product carboxymethyl lysine (CmL) may be a dominant antigen of some antibodies (Reddy et al., 1995, Biochem. 34:10872-10878). Earlier studies have failed to reveal ELISA reactivity with model CmL compounds (Makita et al., 1992, J. Biol. Chem. 267:5133-5138).
  • ELISA Detection of AGE Products [0077]
  • The general method of Engvall (1981, [0078] Methods Enzymol. 70:-419-439) was used to perform the ELISA. Typically, glycated protein samples were diluted to approximately 1.5 ug/ml in 0.1 M sodium carbonate buffer of pH 9.5 to 9.7. The protein was coated overnight at room temperature onto 96-well polystyrene plates by pippetting 200 ul of the protein solution in each well (0.3 ug/well). After coating, the protein was washed from the wells with a saline solution containing 0.05% Tween-20. The wells were then blocked with 200 ul of 1% casein in carbonate buffer for 2 h at 37° C. followed by washing. Rabbit anti-AGE antibodies were diluted at a titer of about 1:350 in incubation buffer, and incubated for 1 h at 37° C., followed by washing. In order to minimize background readings, antibodies R479 used to detect glycated RNase were raised against glycated BSA, and antibodies R618 used to detect glycated BSA and glycated Hb were raised against glycated RNase. An alkaline phosphatase-conjugated antibody to rabbit IgG was then added as the secondary antibody at a titer of 1:2000 or 1:2500 (depending on lot) and incubated for 1 h at 37° C., followed by washing. The p-nitrophenylphosphate substrate solution was then added (200 ul/well) to the plates, with the absorbance of the released p-nitrophenolate being monitored at 410 nm with a Dynatech MR 4000 microplate reader.
  • Controls containing unmodified protein were routinely included, and their readings were subtracted, the corrections usually being negligible. The validity of the use of the ELISA method in quantitatively studying the kinetics of AGE formation depends on the linearity of the assay (Kemeny & Challacombe, 1988, [0079] ELISA and Other Solid Phase Immunoassays, John Wiley & Sons, Chichester, U.K.). Control experiments were carried out, for example, demonstrating that the linear range for RNase is below a coating concentration of about 0.2-0.3 mg/well.
  • Results [0080]
  • FIG. 1 A-D are graphs which show the effect of vitamin B[0081] 6 derivatives on post-Amadori AGE formation in bovine serum albumin glycated with glucose. BSA (10 mg/ml) was incubated with 10 M glucose in the presence and absence of the various indicated derivative in 0.4 M sodium phosphate buffer of pH 7.5 at 37° C. for 6 weeks. Aliquots were assayed by ELISA using R618 anti-AGE antibodies. Concentrations of the inhibitors were 3, 15 and 50 mM. Inhibitors used in FIGS. (1A) Pyridoxamine (PM); (1B) pyridoxal phosphate (PLP); (1C) pyridoxal (PL); (1D) pyridoxine (PN).
  • FIG. 1 (control curves) demonstrates that reaction of BSA with 1.0 M glucose is slow and incomplete after 40 days, even at the high sugar concentration used to accelerate the reaction. The simultaneous inclusion of different concentrations of various B[0082] 6 vitamers markedly affects the formation of post-Amadori antigenic AGEs. (FIG. 1A-D) Pyridoxamine and pyridoxal phosphate strongly suppressed post-Amadori antigenic AGE formation at even the lowest concentrations tested, while pyridoxal was effective above 15 mM. Pyridoxine was slightly effective at the highest concentrations tested.
  • FIG. 2 A-D are graphs which show the effect of vitamin B[0083] 1 derivatives and aminoguanidine (AG) on post-Amadori AGE formation in bovine serum albumin. BSA (10 mg/ml) was incubated with 1.0 M glucose in the presence and absence of the various indicated derivative in 0.4 M sodium phosphate buffer of pH 7.5 at 37° C. for 6 weeks. Aliquots were assayed by ELISA using R618 anti-AGE antibodies. Concentrations of the inhibitors were 3, 15 and 50 mM. Inhibitors used in FIGS. (2A) Thiamine pyrophosphate (TPP); (2B) thiamine monophosphate (TP); (2C) thiamine (T); (2D) aminoguanidine (AG).
  • Of the various B[0084] 1 vitamers similarly tested (FIG. 2A-D), thiamine pyrophosphate was effective at all concentrations tested (FIG. 2C), whereas thiamine and thiamine monophosphate were not inhibitory. Most significantly it is remarkable to note the decrease in the final levels of AGEs formed observed with thiamine pyrophosphate, pyridoxal phosphate and pyridoxamine. Aminoguanidine (FIG. 2D) produced some inhibition of post-Amadori AGE formation in BSA, but less so than the above compounds. Similar studies were carried out with human methemaglobin and bovine ribonuclease A.
  • FIG. 3 A-D are graphs which show the effect of vitamin B[0085] 6 derivatives on post-Amadori AGE formation in human methemoglobin. Hb (1 mg/ml) was incubated with 1.0 M glucose in the presence and absence of the various indicated derivative in 0.4 M sodium phosphate buffer of pH 7.5 at 37° C. for 3 weeks. Aliquots were assayed by ELISA using R618 anti-AGE antibodies. Concentrations of the inhibitors were 0.5, 3, 15 and 50 mM. Inhibitors used in FIGS. (3A) Pyridoxamine (PM); (3B) pyridoxal phosphate (PLP); (3C) pyridoxal (PL); (3D) pyridoxine (PN).
  • It had been previously reported that Hb of a diabetic patient contains a component that binds to anti-AGE antibodies, and it was proposed that this glycated Hb (termed Hb-AGE, not to be confused with Hb[0086] Alc) could be useful in measuring long-term exposure to glucose. The in vitro incubation of Hb with glucose produces antigenic AGEs at an apparently faster rate than observed with BSA. Nevertheless, the different B6 (FIG. 3A-D) and B1 (FIG. 4A-C) vitamers exhibited the same inhibition trends in Hb, with pyridoxamine and thiamine pyrophosphate being the most effective inhibitors in each of their respective families. Significantly, in Hb, aminoguanidine only inhibited the rate of post-Amadori AGE formation, and not the final levels of post-Amadori AGE formed (FIG. 4D).
  • With RNase the rate of post-Amadori antigenic AGE formation by glucose was intermediate between that of Hb and BSA, but the extent of inhibition within each vitamer series was maintained. Again pyridoxamine and thiamine pyrophosphate were more effective that aminoguanidine (FIG. 5). [0087]
  • FIG. 4 A-D are graphs which show the effect of vitamin B[0088] 1 derivatives and aminoguanidine (AG) on post-Amadori AGE formation in human methemoglobin. Hb (1 mg/ml) was incubated with 1.0 M glucose in the presence and absence of the various indicated derivative in 0.4 M sodium phosphate buffer of pH 7.5 at 37° C. for 3 weeks. Aliquots were assayed by ELISA using R618 anti-AGE antibodies. Concentrations of the inhibitors were 0.5, 3, 15 and 50 mM. Inhibitors used in FIGS. (4A) Thiamine pyrophosphate (TPP); (4B) thiamine monophosphate (TP); (4C) thiamine (T); (4D) aminoguanidine (AG).
  • FIG. 5 is a bar graph which shows a comparison of the inhibition of the glycation of ribonuclease A by thiamine pyrophosphate (TPP), pyridoxamine (PM) and aminoguanidine (AG). RNase (1 mg/ml) was incubated with 1.0 M glucose (glc) in the presence and absence of the various indicated derivative in 0.4 M sodium phosphate buffer of pH 7.5 at 37° C. for 6 weeks. Aliquots were assayed by ELISA using R479 anti-AGE antibodies. The indicated percent inhibition was computed from ELISA readings in the absence and presence of the inhibitors at the 6 week time point. Concentrations of the inhibitors were 0.5, 3, 15 and 50 mM. [0089]
  • Discussion [0090]
  • These results demonstrate that certain derivatives of B[0091] 1 and B6 vitamins are capable of inhibiting “late” post-Amadori AGE formation. Some of these vitamers successfully inhibited the final levels of post-Amadori AGE produced, in contrast to aminoguanidine, suggesting that they have greater interactions with Amadori or post-Amadori precursors to antigenic AGEs. The Amadori and post-Amadori intermediates represent a crucial juncture where the “classical” pathway of nonenzymatic glycation begins to become essentially irreversible (Scheme I). In earlier inhibition studies “glycation” was usually measured either as Schiff base formed (after reduction with labeled cyanoborohydride) or as Amadori product formed (after acid precipitation using labeled sugar). Such assays do not yield information on inhibition of post-Amadori conversion steps to “late” post-Amadori AGE products, since such steps lead to no change in the amount of labeled sugar that is attached to the proteins. Other “glycation” assays have relied on the sugar-induced increase of non-specific protein fluorescence, but this can also be induced by dicarbonyl oxidative fragments of free sugar, such as glycoaldehyde or glyoxal (Hunt et al., 1988, Biochem. 256:205-212), independently of Amadori product formation.
  • In the case of pyridoxal (PL) and pyridoxal phosphate (PLP), the data support the simple mechanism of inhibition involving competitive Schiff-base condensation of these aldehydes with protein amino groups at glycation sites. Due to internal hemiacetal formation in pyridoxal but not pyridoxal phosphate, stronger inhibition of post-Amadori AGE formation by PLP is expected by this competitive mechanism. This indeed is observed in the data (FIG. 1B, 1C, FIG. 3B, 3C). The inhibition by pyridoxamine is necessarily different, as pyridoxamine lacks an aldehyde group. However, pyridoxamine is a candidate amine potentially capable of forming a Schiff-base linkage with the carbonyls of open-chain sugars, with dicarbonyl fragments, with Amadori products, or with post-Amadori intermediates. The mechanism of inhibition of B[0092] 1 compounds is not obvious. All the forms contain an amino functionality, so that the marked efficiency of only the pyrophosphate form suggests an important requirement for a strong negative charge.
  • A significant unexpected observation is that the extent of inhibition by aminoguanidine, and some of the other compounds, is considerably less at late stages of the reaction, than during the early initial phase. This suggests a different mechanism of action than that of pyridoxamine and thiamine pyrophosphate, suggesting that the therapeutic potential of these compounds will be enhanced by co-administration with aminoguanidine. [0093]
  • EXAMPLE 2
  • Kinetics of Non-enzymatic Glycation: Paradoxical Inhibition by Ribose and Facile Isolation of Protein Intermediate for Rapid Post-Amadori AGE Formation [0094]
  • While high concentrations of glucose are used to cause the non-enzymatic glycation of proteins, paradoxically, it was found that ribose at high concentrations is inhibitory to post-Amadori AGE formation in ribonuclease by acting on the post-Amadori “late” stages of the glycation reaction. This unexpectedly inhibitory effect suggests that the “early” reactive intermediates, presumably Amadori products, can be accumulated with little formation of “late” post-Amadori AGE products (AGEs; Maillard products). Investigation into this phenomenon has demonstrated: (1) ability to define conditions for the kinetic isolation of Amadori (or post-Amadori) glycated intermediate(s); (2) the ability study the fast kinetics of buildup of such an intermediate; (3) the ability to study the surprisingly rapid kinetics of conversion of such intermediates to AGE products in the absence of free or reversibly bound sugar; (4) the ability to use these intermediates to study and characterize inhibition of post-Amadori steps of AGE formation thus providing a novel system to investigate the mechanism of reaction and the efficacy of potential agents that could block AGE formation; and (5) with this knowledge it is also further possible to use [0095] 13C NMR to study the reactive intermediates and monitor their conversion to various candidate AGEs (Khalifah et al., 1996, Biochemistry 35(15):4645-4654).
  • Chemicals and Materials [0096]
  • As in Example 1 Above. [0097]
  • Preparation of Polyclonal Antibodies to AGEs [0098]
  • As in Example 1 Above. [0099]
  • ELISA Detection of AGE [0100]
  • Products As in Example 1 above. [0101]
  • Amino Acid Analysis [0102]
  • Amino acid analyses were carried out at the Biotechnology Support Facility of the Kansas University Medical Center. Analyses were performed after hydrolysis of glycated protein (reduced with sodium cyanoborohydride) with 6 N HCl at 110° C. for 18-24 h. Phenyl isothiocyanate was used for derivatization, and PTH derivatives were analyzed by reverse-phase HPLC on an Applied Biosystems amino acid analyzer (420A derivatizer, 130A separation system, 920A data analysis system). [0103]
  • Pentosidine Reverse-Phase HPLC Analysis [0104]
  • Pentosidine production in RNase was quantitated by HPLC (Sell & Monnier, 1989, [0105] J. Biol. Chem. 264:21597-21602; Odetti et al., 1992, Diabetes 41:153-159). Ribose-modified protein samples were hydrolyzed in 6 N HCl for 18 h at 100° C. and then dried in a Speed Vac. The samples were then redissolved, and aliquots were taken into 0.1% trifluoroacetic acid and analyzed by HPLC on a Shimadzu system using a Vydac C-18 column equilibrated with 0.1% TFA. A gradient of 0-6% acetonitrile (0.1% in TFA) was run in 30 min at a flow rate of about 1 ml/min. Pentosidine was detected by 335 nm excitation/385 nm emission fluorescence, and its elution time was determined by running a synthesized standard. Due to the extremely small levels of pentosidine expected (Grandhee & Monnier, 1991, J. Biol. Chem. 266:11649-11653; Dyer et al., 1991, J. Biol. Chem. 266:11654-11660), no attempt was made to quantitate the absolute concentrations. Only relative concentrations were determined from peak areas.
  • Glycation Modifications [0106]
  • Modification with ribose or glucose was generally done at 37° C. in 0.4 M phosphate buffer of pH 7.5 containing 0.02% sodium azide. The high buffer concentration was always used with 0.5 M ribose modifications. The solutions were kept in capped tubes and opened only to remove timed aliquots that were immediately frozen for later carrying out the various analyses. “Interrupted glycation” experiments were carried out by first incubating protein with the ribose at 37° C. for 8 or 24 h, followed by immediate and extensive dialysis against frequent cold buffer changes at 4° C. The samples were then reincubated by quickly warming to 37° C. in the absence of external ribose. Aliquots were taken and frozen at various intervals for later analysis. Due to the low molecular weight of RNase, protein concentrations were remeasured after dialysis even when low molecular weight cut-off dialysis tubing was used. An alternative procedure was also devised (see below) in which interruption was achieved by simple 100-fold dilution from reaction mixtures containing 0.5 M ribose. Protein concentrations were estimated from UV spectra. The difference in molar extinction between the peak (278 nm) and trough (250 nm) was used for RNase concentration determinations in order to compensate for the general increase in UV absorbance that accompanies glycation. Time-dependent UV-difference spectral studies were carried out to characterize the glycation contributions of the UV spectrum. [0107]
  • Data Analysis and Numerical Simulations of Kinetics [0108]
  • Kinetic data were routinely fit to monoexponential or biexponential functions using nonlinear least-squares methods. The kinetic mechanisms of Schemes 5-6 have been examined by numerical simulations of the differential equations of the reaction. Both simulations and fitting to observed kinetics data were carried out with the SCIENTIST 2.0 software package (Micromath, Inc.). Determination of apparent half-times (FIG. 6B) from kinetic data fit to two-exponential functions (FIG. 6A) was carried out with the “solve” function of MathCAD 4.0 software (MathSoft, Inc.). [0109]
  • Results [0110]
  • Comparison of Glycation by Glucose and Ribose [0111]
  • The reaction of RNase A with ribose and glucose has been followed primarily with ELISA assays, using R479 rabbit AGE-specific antibodies developed against glucose-modified BSA. To a lesser extent, the production of pentosidine, the only known acid-stable fluorescent AGE, was quantiated by HPLC following acid hydrolysis. Preliminary studies using 0.05 M ribose at 37° C. showed that the rate of antigenic AGE formation appears to be modestly increased (roughly 2-3 fold as measured by the apparent half-time) as the pH is increased from 5.0 to 7.5, with an apparent small induction period at the beginning of the kinetics in all cases. The glycation of RNase with 0.05_M ribose at pH 7.5 (half-time near 6.5 days) appears to be almost an order of magnitude faster than that of glycation with 1.0 M glucose (half-time in excess of 30 days; see FIG. 7B, solid line). The latter kinetics also displayed a small induction period but incomplete leveling off even after 60 days, making it difficult to estimate a true half-time. [0112]
  • When the dependence of the kinetics on ribose concentration was examined at pH 7.5, a most unexpected result was obtained. The rate of reaction initially increased with increasing ribose concentration, but at concentrations above 0.15 M the rate of reaction leveled off and then significantly decreased (FIG. 6A). A plot of the dependence of the reciprocal half-time on the concentration of ribose (FIG. 6B) shows that high ribose concentrations are paradoxically inhibitory to post-Amadori antigenic AGE formation. This unusual but consistent effect was found to be independent of changes in the concentration of either buffer (2-fold) or RNase (10-fold), and it was not changed by affinity purification of the R479 antibody on a column of immobilized AGE-RNase. It is also not due to effects of ribose on the ELISA assay itself. The measured inhibitory effect by ribose on post-Amadori AGE formation is not likely due to ribose interference with antibody recognition of the AGE antigenic sites on protein in the ELISA assay. Prior to the first contact with the primary anti-AGE antibody on the ELISA plates, glycated protein has been diluted over 1000-fold, washed extensively with Tween-20 after adsorption, and blocked with a 1% casein coating followed by further washing with Tween-20. [0113]
  • Kinetics of Formation of Post-Amadori Antigenic AGEs by “Interrupted Glycation”[0114]
  • In view of the small induction period seen, an attempt was made to determine whether there was some accumulation during the reaction, of an early precursor such as an Amadori intermediate, capable of producing the ELISA-detectable post-Amadori antigenic AGEs. RNase was glycated at pH 7.5 and 37° C. with a high ribose concentration of 0.5 M, and the reaction was interrupted after 24 h by immediate cooling to 4° C. and dialysis against several changes of cold buffer over a period of 24 h to remove free and reversibly bound (Schiff base) ribose. Such a ribose-free sample was then rapidly warmed to 37° C. without re-adding any ribose, and was sampled for post-Amadori AGE formation over several days. The AGE antigen production of this 24 h “interrupted glycation” sample is shown by the dashed line and open triangles in FIG. 7A, the time spent in the cold dialysis is not included. An uninterrupted control (solid line and filled circles) is also shown for comparison. Dramatically different kinetics of post-Amadori antigenic AGE formation are evident in the two samples. The kinetics of AGE antigen production of the ribose-free interrupted sample now show (1) monoexponential kinetics with no induction period, (2) a greatly enhanced rate of antigenic AGE formation, with remarkable half-times of the order of 10 h, and (3) production of levels of antigen comparable to those seen in long incubations in the continued presence of ribose (see FIG. 6A). Equally significant, the data also demonstrate that negligible AGE antigen was formed during the cold dialysis period, as shown by the small difference between the open triangle and filled circle points at [0115] time 1 day in FIG. 7A. Very little, if any, AGE was formed by the “interruption” procedure itself. These observations show that a fully competent isolatable intermediate or precursor to antigenic AGE has been generated during the 24 h contact with ribose prior to the removal of the free and reversibly bound sugar.
  • Samples interrupted after only 8 h produced a final amount of AGE antigen that was about 72% of the 24 h interrupted sample. Samples of RNase glycated with only 0.05 M ribose and interrupted at 8 h by cold dialysis and reincubation at 37° C. revealed less than 5% production of ELISA-reactive antigen after 9 days. Interruption at 24 h, however, produced a rapid rise of ELISA antigen (similar to FIG. 7A) to a level roughly 50% of that produced in the uninterrupted presence of 0.05 M ribose. [0116]
  • The same general interruption effects were also seen with other proteins (BSA and Hemoglobin). Except for a somewhat different absolute value of the rate constants, and the amount of antigenic AGEs formed during the 24 h 0.5 M ribose incubation, the same dramatic increase in the rate of AGE antigen formation was observed after removal of 0.5 M ribose. [0117]
  • Glycation is much slower with glucose than with ribose (note the difference in time scales between FIG. 7A and FIG. 7B). However, unlike the case with ribose, interruption after 3 days of glycation by 1.0 M glucose produced negligible buildup of precursor to ELISA-reactive AGE antigens (FIG. 7B, dashed curve). [0118]
  • Kinetics of Pentosidine Formation [0119]
  • The samples subjected to ELISA testing were also assayed for the production of pentosidine, an acid-stable AGE. The content of pentosidine was measured for the same RNase samples analyzed for antibody reactivity by ELISA. Glycation by ribose in 0.4 M phosphate buffer at pH 7.5 produced pentosidine in RNase A that was quantitated by fluroescence after acid hydrolysis. FIG. 8A shows that under uninterrupted conditions, 0.05 M ribose produces a progressive increase in pentosidine. However, when glycation is carried out under “interrupted” conditions using 0.5 M ribose, a dramatic increase in the rate of pentosidine formation is seen immediately after removal of excess ribose (FIG. 8B), which is similar to, but slightly more rapid than, the kinetics of the appearance of antigenic AGEs (FIG. 7A). A greater amount of pentosidine was also produced with 24 h interruption as compared with 8 h. Reproducible differences between the kinetics of formation of pentosidine and antigenic AGEs can also be noted. A significant amount of pentosidine is formed during the 24 h incubation and also during the cold dialysis, resulting in a jump of the dashed vertical line in FIG. 8B. Our observations thus demonstrate that a pentosidine precursor accumulates during ribose glycation that can rapidly produce pentosidine after ribose removal (cf. Odetti et al., 1992, [0120] Diabetes 41:153-159).
  • Rate of Buildup of the Reactive Intermediate(s) [0121]
  • The “interrupted glycation” experiments described above demonstrate that a precursor or precursors to both post-Amadori antigenic AGEs and pentosidine can be accumulated during glycation with ribose. The kinetics of formation of this intermediate can be independently followed and quantitated by a variation of the experiments described above. The amount of intermediate generated in RNase at different contact times with ribose can be assayed by the maximal extent to which it can produce antigenic AGE after interruption. At variable times after initiating glycation, the free and reversibly-bound ribose is removed by dialysis in the cold or by rapid dilution (see below). Sufficient time (5 days, which represents several half-lives according to FIG. 7A) is then allowed after warming to 37° C. for maximal development of post-Amadori antigenic AGEs. The [0122] ELISA readings 5 days after each interruption point, representing maximal AGE development, would then be proportional to the intermediate concentration present at the time of interruption.
  • FIG. 9 shows such an experiment where the kinetics of intermediate buildup are measured for RNase A in the presence of 0.5 M ribose (solid symbols and curve). For comparison, the amount of AGE present before ribose removal at each interruption point is also shown (open symbols and dashed lines). As expected (cf. FIG. 7A), little AGE is formed prior to removal (or dilution) of ribose, so that ELISA readings after the 5 day secondary incubation periods are mostly a measure of AGE formed after ribose removal. The results in FIG. 9 show that the rate of buildup of intermediate in 0.5 M ribose is exponential and very fast, with a half-time of about 3.3 h. This is about 3-fold more rapid than the observed rate of conversion of the intermediate to antigenic AGEs after interruption (open symbols and dashed curve FIG. 7A). [0123]
  • In these experiments the removal of ribose at each interruption time was achieved by 100-fold dilution, and not by dialysis. Simple dilution reduced the concentration of ribose from 0.05 M to 0.005 M. It was independently determined (FIG. 6A) that little AGE is produced in this time scale with the residual 5 mM ribose. This dilution approach was primarily dictated by the need for quantitative point-to-point accuracy. Such accuracy would not have been achieved by the dialysis procedure that would be carried out independently for each sample at each interruption point. Our results show that dilution was equivalent to dialysis. [0124]
  • A separate control experiment (see FIG. 10 below) demonstrated that the instantaneous 100-fold dilution gave nearly identical results to the dialysis procedure. These control experiments demonstrate that the reversible ribose-protein binding (Schiff base) equilibrium is quite rapid on this time scale. This is consistent with data of Bunn and Higgins (1981, [0125] Science 213: 222-224) that indicated that the half-time of Schiff base formation with 0.5 M ribose should be on the order of a few minutes. The 100-fold rapid dilution method to reduce ribose is a valid method where quantitative accuracy is essential and cannot be achieved by multiple dialysis of many samples.
  • Direct Inhibition of Post-Amadori AGE Formation from the Intermediate by Ribose and Glucose [0126]
  • The increase in the rate of AGE formation after interruption and sugar dilution suggests, but does not prove, that high concentrations of ribose are inhibiting the reaction at or beyond the first “stable” intermediate, presumably the Amadori derivative (boxed in Scheme I). A test of this was then carried out by studying the effect of directly adding ribose, on the post-Amadori reaction. RNase was first incubated for 24 h in 0.5 M ribose in order to prepare the intermediate. Two protocols were then carried out to measure possible inhibition of the post-Amadori formation of antigenic AGEs by different concentrations of ribose. In the first experiment, the 24 h ribated sample was simply diluted 100-fold into solutions containing varying final concentrations of ribose ranging from 0.005 M to 0.505 M (FIG. 10A). The rate and extent of AGE formation are clearly seen to be diminished by increasing ribose concentrations. Significantly, up to the highest concentration of 0.5 M ribose, the kinetics appear exponential and do not show the induction period that occurs with uninterrupted glycation (FIGS. 6A and 7A) in high ribose concentrations. [0127]
  • A second experiment (FIG. 10B) was also conducted in which the 24 h interrupted sample was extensively dialyzed in the cold to release free and reversibly bound ribose as well as any inhibitory products that may have formed during the 24 h incubation with ribose. Following this, aliquots were diluted 100-fold into varying concentrations of freshly made ribose, and the formation of antigenic AGE products was monitored as above. There results were nearly identical to the experiment of FIG. 10A where the dialysis step was omitted. In both cases, the rate and extent of AGE formation were diminished by increasing concentrations of ribose, and the kinetics appeared exponential with no induction period. [0128]
  • The question of whether glucose or other sugars can also inhibit the formation of AGEs from the reactive intermediate obtained by interrupted glycation in 0.5 M ribose was also investigated. The effects of glucose at concentrations of 1.0-2.0 M were tested (data not shown). Glucose was clearly not as inhibitory as ribose. When the 24 h ribose interrupted sample was diluted 100-fold into these glucose solutions, the amount of antigenic AGE formed was diminished by about 30%, but there was little decrease in the apparent rate constant. Again, the kinetics appeared exponential. [0129]
  • Effect of pH on Post-Amadori Kinetics of AGE Formation [0130]
  • The interrupted glycation method was used to investigate the pH dependence of the post-Amadori kinetics of AGE formation from the reactive intermediate. In these experiments, RNase A was first reacted for 24 h with 0.5 M ribose at pH 7.5 to generate the reactive intermediate. The kinetics of the decay of the intermediate to AGEs were then measured by ELISA. FIG. 11 shows that an extremely wide pH range of 5.0-9.5 was achievable when the kinetics were measured by initial rates. A remarkable bell-shaped dependence was observed, showing that the kinetics of antigenic AGEs formation are decreased at both acidic and alkaline pH ranges, with an optimum near [0131] pH 8.
  • A single “pH jump” experiment was also carried out on the pH 5.0 sample studied above which had the slowest rate of antigenic AGE formation. After 12 days at 37° C. in pH 5.0 buffer, the pH was adjusted quickly to 7.5, and antigenic AGE formation was monitored by ELISA. Within experimental error, the sample showed identical kinetics (same first order rate constant) of AGE formation to interrupted glycation samples that had been studied directly at pH 7.5 (FIG. 12). In this experiment, the relative amounts of antigenic AGE could not be directly compared on the same ELISA plate, but the pH-jumped sample appeared to have formed substantial though somehow diminished levels of antigenic AGEs. These results demonstrate that intermediate can be prepared free of AGE and stored at pH for later studies of conversion to AGEs. [0132]
  • Inhibition of Post-Amadori AGE Formation by Aminoguanidine [0133]
  • The efficacy of aminoguanidine was tested by this interrupted glycation method, i.e., by testing its effect on post-Amadori formation of antigenic AGEs after removal of excess and reversibly bound ribose. FIG. 20A demonstrates that aminoguanidine has modest effects on blocking the formation of antigenic AGEs in RNase under these conditions, with little inhibition below 50 mM. Approximately 50% inhibition is achieved only at or above 100-250 mM. Note again that in these experiments, the protein was exposed to aminoguanidine only after interruption and removal of free and reversibly bound ribose. Comparable results were also obtained with the interrupted glycation of BSA (FIG. 20B). [0134]
  • Amino Acid Analysis of Interrupted Glycation Samples [0135]
  • Amino acid analysis was carried out on RNase after 24 h contact with 0.5 M ribose (undialyzed), after extensive dialysis of the 24 h glycated sample, and after 5 days of incubation of the latter sample at 37° C. These determinations were made after sodium cyanoborohydride reduction, which reduces Schiff base present on lysines or the terminal amino group. All three samples, normalized to alanine (12 residues), showed the same residual lysine content (4.0±0.5 out of the original 10 in RNase). This indicates that after 24 h contact with 0.5 M ribose, most of the formed Schiff base adducts had been converted to Amadori or subsequent products. No arginine or histidine residues were lost by modification. [0136]
  • Discussion [0137]
  • The use of rapidly reacting ribose and the discovery of its reversible inhibition of post-Amadori steps have permitted the dissection and determination of the kinetics of different steps of protein glycation in RNase. Most previous kinetic studies of protein “glycation” have actually been restricted to the “early” steps of Schiff base formation and subsequent Amadori rearrangement. Some kinetic studies have been carried out starting with synthesized fructosylamines, i.e. small model Amadori compounds of glucose (Smith and Thornalley, 1992, [0138] Eur. J. Biochem. 210:729-739, and references cited therein), but such studies, with few exceptions, have hitherto not been possible with proteins. One notable exception is the demonstration by Monnier (Odetti et al., 1992, supra) that BSA partially glycated with ribose can rapidly produce pentosidine after ribose removal. The greater reactivity of ribose has also proven a distinct advantage in quantitatively defining the time course of AGE formation. It is noted that glucose and ribose are both capable of producing similar AGE products, such as pentosidine (Grandhee & Monnier, 1991, supra; Dyer et al. 1991, supra), and some 13C NMR model compound work has been done with ADP-ribose (Cervantes-Laurean et al., 1993, Biochemistry 32:1528-1534). The present work shows that antigenic AGE products of ribose fully cross-react with anti-AGE antibodies directed against glucose-modified proteins, suggesting that ribose and glucose produce similar antigenic AGEs. The primary kinetic differences observed between these two sugars are probably due to relative differences in the rate constants of steps leading to post-Amadori AGE formation, rather than in the mechanism.
  • The results presented reveal a marked and paradoxical inhibition of overall AGE formation by high concentrations of ribose (FIG. 6) that has not been anticipated by earlier studies. This inhibition is rapidly reversible in the sense that it is removed by dialysis of initially modified protein (FIG. 7A) or by simple 100-fold dilution (as used in FIG. 11). The experiments of FIG. 10 demonstrate that it is not due to the accumulation of dialyzable inhibitory intermediates during the initial glycation, since dialysis of 24 h modified protein followed by addition of different concentrations of fresh ribose induces the same inhibition. The data of FIG. 10A,B show that the inhibition occurs by reversible and rapid interaction of ribose with protein intermediate containing reactive Amadori products. The inhibition is unlikely to apply to the early step of formation of Amadori product due to the rapid rate of formation of the presumed Amadori intermediate that was determined in the experiment of FIG. 9. The identification of the reactive intermediate as an Amadori product is well supported by the amino acid analysis carried out (after sodium cyanoborohydrate reduction) before and after dialysis at the 24 h interruption point. The unchanged residual lysine content indicates that any dischageable Schiff bases have already been irreversibly converted (presumably by Amadori rearrangement) by the 24 h time. [0139]
  • The secondary ribose suppression of “late” but not “early” glycation steps significantly enhances the accumulation of a fully-competent reactive Amadori intermediate containing little AGE. Its isolation by the interruption procedure is of importance for kinetic and structural studies, since it allows one to make studies in the absence of free or Schiff base bound sugar and their attendant reactions and complications. For example, the post-Amadori conversion rates to antigenic AGE and pentosidine AGE products have been measured (FIG. 7A, open symbols, and FIG. 8B), and demonstrated to be much faster (t ½˜10 h) than reflected in the overall kinetics under uninterrupted conditions (FIG. 6A and FIG. 8A). The rapid formation of pentosidine that was measured appears consistent with an earlier interrupted ribose experiment on BSA by Odetti et al. (1992, supra). Since ribose and derivatives such as ADP-ribose are normal metabolites, the very high rates of AGE formation seen here suggest that they should be considered more seriously as sources of potential glycation in various cellular compartments (Cervantes-Laurean et al., 1993, supra), even though their concentrations are well below those of the less reactive glucose. [0140]
  • Another new application of the isolation of intermediate is in studying the pH dependence of this complex reaction. The unusual bell-shaped pH profile seen for the post-Amadori AGE formation (FIG. 11) is in striking contrast to the mild pH dependence of the overall reaction. The latter kinetics reflect a composite effect of pH on all steps in the reaction, including Schiff base and Amadori product formation, each of which may have a unique pH dependence. This complexity is especially well illustrated by studies of hemoglobin glycation (Lowery et al., 1985, [0141] J. Biol. Chem. 260:11611-11618). The bell-shaped pH profile suggests, but does not prove, the involvement of two ionizing groups. If true, the data may imply the participation of a second amino group, such as from a neighboring lysine, in the formation of dominant antigenic AGEs. The observed pH profile and the pH-jump observations described suggest that a useful route to isolating and maintaining the reactive intermediate would be by the rapid lowering of the pH to near 5.0 after 24 h interruption.
  • The kinetic studies provide new insights into the mechanisms of action of aminoguanidine (guanylhydrazine), an AGE inhibitor proposed by Cerami and co-workers to combine with Amadori intermediates (Brownlee et al., 1986, supra). This proposed pharmacological agent is now in Phase III clinical trials for possible therapeutic effects in treating diabetes (Vlassara et al., 1994, supra) . However, its mechanism of AGE inhibition is likely to be quite complex, since it is multifunctional. As a nucelophilic hydrazine, it can reversibly add to active carbonyls, including aldehydo carbonyls of open-chain glucose and ribose (Khatami et al., 1988, [0142] Life Sci. 43:1725-1731; Hirsch et al., 1995, Carbohyd. Res. 267:17-25), as well as keto carbonyls of Amadori compounds. It is also a guanidinium compound that can scavange highly reactive dicarbonyl glycation intermediates such as glyoxal and glucosones (Chen & Cerami, 1993, J. Carbohyd. Chem. 12:731-742; Hirsch et al., 1992, Carbohyd. Res. 232:125-130; Ou & Wolff, 1993, Biochem. Pharmacol. 46:1139-1144). The interrupted glycation method allowed examination of aminoguanidine efficacy on only post-Amadori steps of AGE formation. Equally important, it allowed studies in the absence of free sugar or dicarbonyl-reactive fragments from free sugar (Wolff & Dean, 1987, Biochem. J. 245:243-250; Wells-Knecht et al., 1995, Biochemistry 34:-3702-3709) that can combine with aminoguanidine. The results of FIG. 20 demonstrate that aminoguanidine has, at best, only a modest effect on post-Amadori AGE formation reactions, achieving 50% inhibition at concentrations above 100-250 mM. The efficacy of aminoguanidine thus predominantly arises either from inhibiting early steps of glycation (Schiff base formation) or from scavenging highly reactive dicarbonyls generated during glycation. Contrary to the original claims, it does not appear to inhibit AGE formation by complexing the Amadori intermediate.
  • The use of interrupted glycation is not limited for kinetic studies. Interrupted glycation can simplify structural studies of glycated proteins and identifying unknown AGEs using techniques such as [0143] 13C NMR that has been used to detect Amadori-adducts of RNase (Neglia et al., 1983, J. Biol. Chem. 258:14279-14283; 1985, J. Biol. Chem. 260:5406-5410). The combined use of structural and kinetic approaches should also be of special interest. For example, although the identity of the dominant antigenic AGEs reacting with the polyclonal antibodies remains uncertain, candidate AGEs, such as the recently proposed (carboxymethyl)lysine (Reddy et al., 1995, Biochemistry 34:10872-10878; cf. Makita et al., 1992, J. Biol. Chem. 267:5133-5138) should display the same kinetics of formation from the reactive intermediate that we have observed. The availability of the interrupted kinetics approach will also help to determine the importance of the Amadori pathway to the formation of this AGE. Similarly, monitoring of the interrupted glycation reaction by techniques such as 13C NMR should identify resonances of other candidate antigenic AGEs as being those displaying similar kinetics of appearance. Table I lists the 13C NMR peaks of the Amadori intermediate of RNase prepared by reaction with C-2 enriched ribose. The downfield peak near 205 ppm is probably due to the carbonyl of the Amadori product. In all cases, the ability to remove excess free and Schiff base sugars through interrupted glycation will considerably simplify isolation, identification, and structural characterization.
  • Table I lists the peaks that were assigned to the Post-Amadori Intermediate due to their invariant or decreasing intensity with time. Peak positions are listed in ppm downfield from TMS. [0144]
    TABLE I
    125 MHz C-13 NMR Resonances of Ribonuclease Amadori
    Intermediate Prepared by 24 HR Reaction with 99% [2-C13]Ribose
    216.5 ppm 108.5 ppm
    211.7 105.9
    208 103.9
    103
    172  95.8
    165
    163.9  73.65
    162.1  70.2
     69.7
  • Ribonuclease A was reacted for 24 hr with 0.5 M ribose 99% enriched at C-2, following which excess and Schiff base bound ribose was removed by extensive dialysis in the cold. The sample was then warmed back to 37° C. immediately before taking a 2 hr NMR scan. The signals from RNase reacted with natural abundance ribose under identical conditions were then subtracted from the NMR spectrum. Thus all peaks shown are due to enriched C-13 that originated at the C-2 position. Some of the peaks arise from degradation products of the intermediate, and these can be identified by the increase in the peak intensity over time. FIG. 31 shows the NMR spectrum obtained. [0145]
  • EXAMPLE 3
  • In Vitro Inhibition of the Formation of Antigenic Advanced Glycation End-Products (AGES) by Derivatives of Vitamins B[0146] 1 and B6 and Aminoguanidine. Inhibition of Post-Amadori Kinetics Differs from that of Overall Glycation
  • The interrupted glycation method for following post-Amadori kinetics of AGE formation allows for the rapid quantitative study of “late” stages of the glycation reaction. Importantly, this method allows for inhibition studies that are free of pathways of AGE formation which arise from glycoxidative products of free sugar or Schiff base (Namiki pathway) as illustrated in Scheme I. Thus the interrupted glycation method allows for the rapid and unique identification and characterization of inhibitors of “late” stages of glycation which lead to antigenic AGE formation. [0147]
  • Among the vitamin B[0148] 1 and B6 derivatives examined, pyridoxamine and thiamine pyrophosphate are unique inhibitors of the post-Amadori pathway of AGE formation. Importantly, it was found that efficacy of inhibition of overall glycation of protein, in the presence of high concentrations of sugar, is not predictive of the ability to inhibit the post-Amadori steps of AGE formation where free sugar is removed. Thus while pyridoxamine, thiamine pyrophosphate and aminoguanidine are potent inhibitors of AGE formation in the overall glycation of protein by glucose, aminoguanidine differs from the other two in that it is not an effective inhibitor of post-Amadori AGE formation. Aminoguanidine markedly slows the initial rate of AGE formation by ribose under uninterrupted conditions, but has no effect on the final levels of antigenic AGEs produced. Examination of different proteins (RNase, BSA and hemoglobin), confirmed that the inhibition results are generally non-specific as to the protein used, even though there are individual variations in the rates of AGE formation and inhibition.
  • Chemicals and Materials [0149]
  • As in Example 1 above. [0150]
  • Preparation of Polyclonal Antibodies to AGEs [0151]
  • As in Example 1 above. [0152]
  • ELISA Detection of AGE Products [0153]
  • As in Example 1 above. [0154]
  • Uninterrupted Ribose Glycation Assays [0155]
  • Bovine serum albumin, ribonuclease A, and human hemoglobin were incubated with ribose at 37° C. in 0.4 M sodium phosphate buffer of pH 7.5 containing 0.02% sodium azide. The protein (10 mg/ml or 1 mg/ml), 0.05 M ribose, and prospective inhibitors (at 0.5, 3, 15 and 50 mM) were introduced into the incubation mixture simultaneously. Solutions were kept in the dark in capped tubes. Aliquots were taken and immediately frozen until analyzed by ELISA at the conclusion of the reaction. The incubations were for 3 weeks (Hb) or 6 weeks (RNase, BSA). Glycation reactions were monitored for constant pH throughout the duration of the experiments. [0156]
  • Interrupted (Post-Amadori) Ribose Glycation Assays [0157]
  • Glycation was first carried out by incubating protein (10 mg/ml) with 0.5 M ribose at 37° C. in 0.4 M phosphate buffer at pH 7.5 containing 0.2% sodium azide for 24 h in the absence of inhibitors. Glycation was then interrupted to remove excess and reversibly bound (Schiff base) sugar by extensive dialysis against frequent cold buffer changes at 4° C. The glycated intermediate samples containing maximal amount of Amadori product and little AGE (depending on protein) were then quickly warmed to 37° C. without re-addition of ribose. This initiated conversion of Amadori intermediates to AGE products in the absence or presence of various concentrations (typically 3, 15 and 50 mM) of prospective inhibitors. Aliquots were taken and frozen at various intervals for later analysis. The solutions were kept in capped tubes and opened only to remove timed aliquots that were immediately frozen for later carrying out the various analyses. [0158]
  • Numerical Analysis of Kinetics Data [0159]
  • Kinetics data (time progress curves) was routinely fit to mono- or bi-exponential functions using non-linear least squares methods utilizing either SCIENTIST 2.0 (MicroMath, Inc.) or ORIGIN (Microcal, Inc.) software that permit user-defined functions and control of parameters to iterate on. Standard deviations of the parameters of the fitted functions (initial and final ordinate values and rate constants) were returned as measures of the precision of the fits. Apparent half-times for bi-exponential kinetics fits were determined with the “solve” function of MathCad software (MathSoft, Inc.). [0160]
  • Results [0161]
  • Inhibition by Vitamin B[0162] 6 Derivatives of the Overall Kinetics of AGE Formation from Ribose.
  • The inhibitory effects of vitamin B[0163] 1 and B6 derivatives on the kinetics of antigenic AGE formation were evaluated by polyclonal antibodies specific for AGEs. Initial inhibition studies were carried out on the glycation of bovine ribonuclease A (RNase) in the continuous presence of 0.05 M ribose, which is the concentration of ribose where the rate of AGE formation is near maximal. FIG. 13 (control curves, filled rectangles) demonstrates that the formation of antigenic AGEs on RNase when incubated with 0.05 M ribose is relatively rapid, with a half-time of approximately 6 days under these conditions. Pyridoxal-5′-phosphate (FIG. 13B) and pyridoxal (FIG. 13C) significantly inhibited the rate of AGE formation on RNase at concentrations of 50 mM and 15 mM. Surprisingly, pyridoxine, the alcohol form of vitamin B6, also moderately inhibited AGE formation on RNase (FIG. 13D). Of the B6 derivatives examined, pyridoxamine at 50 mM was the best inhibitor of the “final” levels of AGE formed on RNase over the 6-week time period monitored (FIG. 13A).
  • Inhibition by Vitamin B[0164] 1 Derivatives of the Overall Kinetics of AGE Formation from Ribose.
  • All of the B[0165] 1 vitamers inhibited antigenic AGE formation on RNase at high concentrations, but the inhibition appeared more complex than for the B6 derivatives (FIG. 14A-C). In the case of thiamine pyrophosphate as the inhibitor (FIG. 14A), both the rate of AGE formation and the final levels of AGE produced at the plateau appeared diminished. In the case of thiamine phosphate as the inhibitor (FIG. 14B), and thiamine (FIG. 14C), there appeared to be little effect on the rate of AGE formation, but a substantial decrease in the final level of AGE formed in the presence of the highest concentration of inhibitor. In general, thiamine pyrophosphate demonstrated greater inhibition than the other two compounds, at the lower concentrations examined.
  • Inhibition by Aminoguanidine of the Overall Kinetics of AGE Formation from Ribose [0166]
  • Inhibition of AGE formation by aminoguanidine (FIG. 14D) was distinctly different from that seen in the B[0167] 1 and B6 experiments. Increasing concentration of aminoguanidine decreased the rate of AGE formation on RNase, but did not reduce the final level of AGE formed. The final level of AGE formed after the 6-weeks was nearly identical to that of the control for all tested concentrations of aminoguanidine.
  • Inhibition of the Overall Kinetics of AGE Formation in Serum Albumin and Hemoglobin from Ribose [0168]
  • Comparative studies were carried out with BSA and human methemoglobin (Hb) to determine whether the observed inhibition was protein-specific. The different derivatives of vitamin B[0169] 6 (FIG. 15A-D) and vitamin B1 (FIG. 16A-C) exhibited similar inhibition trends when incubated with BSA as with RNase, pyridoxamine and thiamine pyrophosphate being the most effective inhibitors or each family. Pyridoxine failed to inhibit AGE formation on BSA (FIG. 15D). Pyridoxal phosphate and pyridoxal (FIG. 15B-C) mostly inhibited the rate of AGE formation, but not the final levels of AGE formed. Pyridoxamine (FIG. 15A) exhibited some inhibition at lower concentrations, and at the highest concentration tested appeared to inhibit the final levels of AGE formed more effectively than any of the other B6 derivatives. In the case of B1 derivatives, the overall extent of inhibition of AGE formation with BSA (FIG. 16A-C), was less than that observed with RNase (FIG. 14A-C). Higher concentrations of thiamine and thiamine pyrophosphate inhibited the final levels of AGEs formed, without greatly affecting the rate of AGE formation (FIG. 16C). Aminoguanidine again displayed the same inhibition effects with BSA as seen with RNase (FIG. 16D), appearing to slow the rate of AGE formation without significantly affecting the final levels of AGE formed.
  • The kinetics of AGE formation was also examined using Hb in the presence of the B[0170] 6 and B1 vitamin derivatives, and aminoguanidine. The apparent absolute rates of AGE formation were significantly higher with Hb than with either RNase or BSA. However, the tested inhibitors showed essentially similar behavior. The effects of the vitamin B6 derivatives are shown in FIG. 17. Pyridoxamine showed the greatest inhibition at concentrations of 3 mM and above (FIG. 17A), and was most effective when compared to pyridoxal phosphate (FIG. 17B), pyridoxal (FIG. 17C), and pyridoxine (FIG. 17D). In the case of the B1 vitamin derivatives (data not shown), the inhibitory effects were more similar to the BSA inhibition trends than to RNase. The inhibition was only modest at the highest concentrations tested (50 mM), being nearly 30-50% for all three B1 derivatives. The primary manifestation of inhibition was in the reduction of the final levels of AGE formed.
  • Inhibition by Vitamin B[0171] 6 Derivatives of the Kinetics of Post-Amadori Ribose AGE Formation
  • Using the interrupted glycation model to assay for inhibition of the “late” post-Amadori AGE formation, kinetics were examined by incubating isolated Amadori intermediates of either RNase or BSA at 37° C. in the absence of free or reversibly bound ribose. Ribose sugar that was initially used to prepare the intermediates was removed by cold dialysis after an initial glycation reaction period of 24 h. After AGE formation is allowed to resume, formation of AGE is quite rapid (half-times of about 10 h) in the absence of any inhibitors. FIG. 18 shows the effects of pyridoxamine (FIG. 18A), pyridoxal phosphate (FIG. 18B), and pyridoxal (FIG. 18C) on the post-Amadori kinetics of BSA. Pyridoxine did not produce any inhibition (data not shown). Similar experiments were carried out on RNase. Pyridoxamine caused nearly complete inhibition of AGE formation with RNase at 15 mM and 50 mM (FIG. 18D). Pyridoxal did not show any significant inhibition at 15 mM (the highest tested), but pyridoxal phosphate showed significant inhibition at 15 mM. Pyridoxal phosphate is known to be able to affinity label the active site of RNase (Raetz and Auld, 1972, [0172] Biochemistry 11:2229-2236).
  • With BSA, unlike RNase, a significant amount of antigenic AGE formed during the 24 h initial incubation with RNase (25-30%), as evidenced by the higher ELISA readings after removal of ribose at time zero for FIGS. [0173] 18A-C. For both BSA and RNase, the inhibition, when seen, appears to manifest as a decrease in the final levels of AGE formed rather than as a decrease in the rate of formation of AGE.
  • Inhibition by Vitamin B[0174] 1 Derivatives of the Kinetics of Post-Amadori Ribose AGE Formation
  • Thiamine pyrophosphate inhibited AGE formation more effectively than the other B[0175] 1 derivatives with both RNase and BSA (FIG. 19). Thiamine showed no effect, while thiamine phosphate showed some intermediate effect. As with the B6 assays, the post-Amadori inhibition was most apparently manifested as a decrease in the final levels of AGE formed.
  • Effects of Aminoguanidine and N[0176] α-acetyl-L-lysine on the Kinetics of Post-Amadori Ribose AGE Formation
  • FIG. 20 shows the results of testing aminoguanidine for inhibition of post-Amadori AGE formation kinetics with both BSA and RNase. At 50 mM, inhibition was about 20% in the case of BSA (FIG. 20B), and less than 15% with RNase (FIG. 20A). The possibility of inhibition by simple amino-containing functionalities was also tested using N[0177] +-acetyl-L-lysine (FIG. 21), which contains only a free Nε-amino group. Nα-acetyl-L-lysine at up to 50 mM failed to exhibit any significant inhibition of AGE formation.
  • Discussion [0178]
  • Numerous studies have demonstrated that aminoguanidine is an apparently potent inhibitor of many manifestations of nonenzymatic glycation (Brownlee et al., 1986; Brownlee, 1992, 1994, 1995). The inhibitory effects of aminoguanidine on various phenomena that are induced by reducing sugars are widely considered as proof of the involvement of glycation in many such phenomena. Aminoguanidine has recently entered into a second round of Phase III clinical trials (as pimagedine) for ameliorating the complications of diabetes thought to be caused by glycation of connective tissue proteins due to high levels of sugar. [0179]
  • Data from the kinetic study of uninterrupted “slow” AGE formation with RNase induced by glucose (Example 1) confirmed that aminoguanidine is an effective inhibitor, and further identified a number of derivatives of vitamins B[0180] 1 and B6 as equally or slightly more effective inhibitors. However, the inhibition by aminoguanidine unexpectedly appeared to diminish in effect at the later stages of the AGE formation reaction. Due to the slowness of the glycation of protein with glucose, this surprising observation could not be fully examined. Furthermore, it has been suggested that there may be questions about the long-term stability of aminoguanidine (Ou and Wolff, 1993, supra).
  • Analysis using the much more rapid glycation by ribose allowed for the entire time-course of AGE formation to be completely observed and quantitated during uninterrupted glycation of protein. The use of interrupted glycation uniquely allowed further isolation and examination of only post-Amadori antigenic AGE formation in the absence of free and reversibly bound (Schiff base) ribose. Comparison of the data from these two approaches with the earlier glucose glycation kinetics has provided novel insights into the mechanisms and effectiveness of various inhibitors. FIG. 22 are bar graphs which depict summarized comparative data of percent inhibition at defined time points using various concentrations of inhibitor. FIG. 22A graphs the data for inhibition after interrupted glycation of RNase AGE formation in ribose. FIG. 22B graphs the data for inhibition after interrupted glycation of BSA AGE formation by ribose. [0181]
  • The overall results unambiguously demonstrate that aminoguanidine slows the rate of post-Amadori antigenic AGE formation in the presence of sugar but has little effect on the final amount of post-Amadori AGE formed. Thus observations limited to only the initial “early” stages of AGE formation which indicate efficacy as an inhibitor may in fact be misleading as to the true efficacy of inhibition of post-Amadori AGE formation. Thus the ability to observe a full-course of reaction using ribose and interrupted glycation gives a more complete picture of the efficacy of inhibition of post-Amadori AGE formation. [0182]
  • EXAMPLE 4
  • Animal Model & Testing of In Vivo Effects of AGE Formation/Inhibitors [0183]
  • Hyperglycemia can be rapidly induced (within one or two days) in rats by administration of streptozocin (aka. streptozotocin, STZ) or alloxan. This has become a common model for diabetes melitus. However, these rats manifest nephropathy only after many months of hyperglycemia, and usually just prior to death from end-stage renal disease (ESRD). It is believed that this pathology is caused by the irreversible glucose chemical modification of long-lived proteins such as collagen of the basement membrane. STZ-diabetic rats show albuminuria very late after induction of hyperglycemia, at about 40 weeks usually only just prior to death. [0184]
  • Because of the dramatic rapid effects of ribose demonstrated in vitro in the examples above, it was undertaken to examine the effects of ribose administration to rats, and the possible induction of AGEs by the rapid ribose glycation. From this study, a rat model for accelerated ribose induced pathology has been developed. [0185]
  • Effects of Very Short-Term Ribose Administration In Vivo [0186]
  • Phase I Protocol [0187]
  • Two groups of six rats each were given in one day either: [0188]
  • a. 300 mM ribose (two intraperitoneal infusions 6-8 hours apart, each 5% of body weight as ml); or [0189]
  • b. 50 mM ribose (one infusion) [0190]
  • Rats were then kept for 4 days with no further ribose administration, at which time they were sacrificed and the following physiological measurements were determined: (i) initial and final body weight; (ii) final stage kidney weight; (iii) initial and final tail-cuff blood pressure; (iv) creatinine clearance per 100 g body weight. [0191]
  • Albumin filtration rates were not measured, since no rapid changes were initially anticipated. Past experience with STZ-diabetic rats shows that albuminuria develops very late (perhaps 40 weeks) after the induction of hyperglycemia and just before animals expire. [0192]
  • Renal Physiology Results [0193]
  • a. Final body weight and final single kidney weight was same for low and high ribose treatment groups. [0194]
  • b. Tail-cuff blood pressure increased from 66±4 to 83±3 to rats treated with low ribose (1×50 mM), and from 66±4 to 106±5 for rats treated with high ribose (2×300 mM). These results are shown in the bar graph of FIG. 23. [0195]
  • c. Creatinine clearance, as cc per 100 g body weight, was decreased (normal range expected about 1.0-1.2) in a dose-dependent fashion to 0.87±0.15 for the low ribose group, and decreased still further 30% to 0.62±0.13 for the high ribose group. These results are shown in the bar graph of FIG. 24. [0196]
  • Phase I Conclusion [0197]
  • A single day's ribose treatment caused a dose-dependent hypertension and a dose-dependent decrease in glomerular [0198] clearance function manifest 4 days later. These are significant metabolic changes of diabetes seen only much later in STZ-diabetic rats. These phenomenon can be hypothesized to be due to ribose irreversible chemical modification (glycation) of protein in vivo.
  • Effect of Exposure to Higher Ribose Concentrations for Longer Time [0199]
  • Phase II Protocol [0200]
  • Groups of rats (3-6) were intraperitoneally given 0.3 M “low ribose dose” (LR) or 1.0 M “high ribose dose” (HR) by twice-daily injections for either (i) 1 day, (ii) a “short-term” (S) of 4 days, or (iii) a “long-term” (L) of 8 days. Additionally, these concentrations of ribose were included in drinking water. [0201]
  • Renal Physiology Results [0202]
  • a. Tail-cuff blood pressure increased in all groups of ribose-treated rats, confirming Phase I results. (FIG. 23). [0203]
  • b. Creatinine clearance decreased in all groups in a ribose dose-dependent and time-dependent manner (FIG. 24). [0204]
  • c. Albumin Effusion Rate (AER) increased significantly in a ribose-dependent manner at 1-day and 4-day exposures. However, it showed some recovery at 8 day relative to 4 day in the high-dose group but not in the low-dose group. These results are shown in the bar graph of FIG. 25. [0205]
  • d. Creatinine clearance per 100 g body weight decreased for both low- and high-ribose groups to about the same extent in a time-dependent manner (FIG. 24). [0206]
  • Phase II Conclusion [0207]
  • Exposure to ribose for as little as 4 days leads to hypertension and renal dysfunction, as manifest by both decreased creatinine clearance and increased albumin filtration. These changes are typical of diabetes and are seen at much later times in STZ-diabetic rats. [0208]
  • Intervention by Two New Therapeutic Compounds and Aminoguanidine [0209]
  • Phase III Protocol [0210]
  • Sixty rats were randomized into 9 different groups, including those exposed to 1 M ribose for 8 days in the presence and absence of aminoguanidine, pyridoxamine, and thiamine pyrophosphate as follows: [0211]
  • Control Groups: [0212]
  • (i) no treatment; [0213]
  • (ii) high dose (250 mg/kg body weight) of pyridoxamine (“compound-P”); [0214]
  • (iii) high dose (250 mg/kg body weight of thiamine pyrophosphate (“compound-T” or “TPP”); and [0215]
  • (iv) low dose (25 mg/kg body weight) of aminoguanidine (“AG”). [0216]
  • Test Groups: [0217]
  • (i) only 1 Mribose-saline (2×9 cc daily IP for 8 days); [0218]
  • (ii) ribose plus low dose (“LP” ) of pyridoxamine (25 mg/kg body weight injected as 0.5 ml with 9 cc ribose); [0219]
  • (iii) ribose plus high dose (“HP”) of pyridoxamine (250 mg/kg body weight injected as 0.5 ml with 9 cc ribose); [0220]
  • (iv) ribose plus high dose (“HT”) of thiamine pyrophosphate (250 mg/kg body weight injected as 0.5 ml with 9 cc ribose); and [0221]
  • (v) ribose plus low dose of amino guanidine (25 mg/kg body weight injected as 0.5 ml with 9 cc ribose). [0222]
  • Unlike Phase II, no ribose was administered in drinking water. Intervention compounds were pre-administered for one day prior to introducing them with ribose. [0223]
  • Renal Physiology Results [0224]
  • a. Blood pressure was very slightly increased by the three compounds alone (control group); ribose-elevated BP was not ameliorated by the co-administration of compounds. These results are shown in the bar graph of FIG. 26. [0225]
  • b. Creatinine clearance in controls was unchanged, except for TPP which diminished it. [0226]
  • c. Creatinine clearance was normalized when ribose was co-administerd with low dose (25 mg/kg) of either aminoguanidine or pyridoxamine. These results are shown in the bar graph of FIG. 27. [0227]
  • d. High concentrations (250 mg/kg) or pyridoxamine and TPP showed only partial protection against the ribose-induced decrease in creatinine clearance (FIG. 27). [0228]
  • e. Albumin effusion rate (AER) was elevated by ribose, as well as by high dose of pyridoxamine and TPP, and low dose of aminoguanidine in the absence of ribose. These results are shown in the bar graph of FIG. 28. [0229]
  • f. Albumin effusion rate was restored to normal by the co-administration of low dose of both aminoguanidine and pyridoxamine. These results are shown in the bar graph of FIG. 29. [0230]
  • Phase III Conclusions [0231]
  • As measured by two indicies of renal function, pyridoxamine and aminoguanidine, both at 25 mg/kg, were apparently effective, and equally so, in preventing the ribose-induced decrease in creatinine clearance and ribose-induced mild increase in albuminuria. [0232]
  • (i) Thiamine pyrophosphate was not tested at 25 mg/kg; (ii) thiamine pyrophosphate and pyridoxamine at 250 mg/kg were partially effective in preventing creatinine clearance decreases but possibly not in preventing mild proteinuria; (iii) at these very high concentrations and in the absence of ribose, thiamine pyrophosphate alone produced a decrease in creatinine clearance, and both produced mild increases in albuminuria. [0233]
  • SUMMARY
  • Renal Function and Diabetes [0234]
  • Persistent hyperglycemia in diabetes mellitus leads to diabetic nephropathy in perhaps one third of human patients. Clinically, diabetic nephropathy is defined by the presence of: [0235]
  • 1. decrease in renal function (impaired glomerular clearance) [0236]
  • 2. an increase in urinary protein (impaired filtration) [0237]
  • 3. the simultaneous presence of hypertension [0238]
  • Renal function depends on blood flow (not measured) and the glomerular clearance, which can be measured by either inulin clearance (not measured) or creatinine clearance. Glomerular permeability is measured by albumin filtration rate, but this parameter is quite variable. It is also a log-distribution function: a hundred-fold increase in albumin excretion represents only a two-fold decrease in filtration capacity. [0239]
  • Ribose Diabetic Rat Model [0240]
  • By the above criteria, ribose appears to very rapidly induce manifestations of diabetic nephropathy, as reflected in hypertension, creatinine clearance and albuminuria, even though the latter is not large. In the established STZ diabetic rat, hyperglycemia is rapidly established in 1-2 days, but clinical manifestations of diabetic nephropathy arise very late, perhaps as much as 40 weeks for albuminuria. In general, albuminuria is highly variable from day to day and from animal to animal, although unlike humans, most STZ rats do eventually develop nephropathy. [0241]
  • Intervention by Compounds [0242]
  • Using the ribose-treated animals, pyridoxamine at 25 mg/kg body weight appears to effectively prevent two of the three manifestations usually attributed to diabetes, namely the impairment of creatinine clearance and albumin filtration. It did so as effectively as aminoguanidine. The effectiveness of thiamine pyrophosphate was not manifest, but it should be emphasized that this may be due to its use at elevated concentrations of 250 mg/kg body weight. Pyridoxamine would have appeared much less effective if only the results at 250 mg/kg body weight are considered. [0243]
  • Effect of Compounds Alone [0244]
  • Overall, the rats appeared to tolerate the compounds well. Kidney weights were not remarkable and little hypertension developed. The physiological effects of the compounds were only tested at 250 mg/kg. Thiamine pyrophosphate, but not pyridoxamine, appeared to decrease creatinine clearance at this concentration. Both appeared to slightly increase albuminuria, but these measurements were perhaps the least reliable. [0245]
  • Human Administration [0246]
  • A typical adult human being of average size weighs between 66-77 Kg. Typically, diabetic patients may tend to be overweight and can be over 112 Kg. The Recommended dietary allowances for an adult male of between 66-77 Kg, as revised in 1989, called for 1.5 mg per day of thiamine, and 2.0 mg per day of Vitamin B[0247] 6 (Merck Manual of Diagnosis and Therapy, 16th edition (Merck & Co., Rathaway, N.J., 1992) pp 938-939).
  • Based upon the rat model approach, a range of doses for administration of pyridoxamine or thiamine pyrophosphate that is predicted to be effective for inhibiting post-Amadori AGE formation and thus inhibiting related pathologies would fall in the range of 1 mg/100 g body weight to 200 mg/100 g body weight. The appropriate range when co-administered with aminoguanidine will be similar. Calculated for an average adult of 75 Kg, the range (at 10 mg/1 Kg body weight) can be approximately 750 mg to upwards of 150 g (at 2 g/1 Kg body weight). This will naturally vary according to the particular patient. [0248]
  • The instant invention may be embodied in other forms or carried out in other ways without departing from the spirit or essential characteristics thereof. The present disclosure and enumerated examples are therefore to be considered as in all respects illustrative and not restrictive, the scope of the invention being indicated by the appended claims, and all equivalency are intended to be embraced therein. One of ordinary skill in the art would be able to recognize equivalent embodiments of the instant invention, and be able to practice such embodiments using the teaching of the instant disclosure and only routine experimentation. [0249]

Claims (10)

1-19. (Canceled).
20. A method for treating one or more disorders selected from the group consisting of nephropathy, proteinuria, albuminuria, impaired glomerular clearance, impaired creatinine clearance, retinopathy, and neurodegenerative disease, comprising administering to a subject with one or more of the disorders an amount effective of pyridoxamine to treat the disorder.
21. The method of claim 20 wherein the subject is a diabetic subject.
22. The method of claim 21 wherein the one or more disorders comprises nephropathy.
23. The method of claim 21 wherein the one or more disorders comprises proteinuria.
24. The method of claim 21 wherein the one or more disorders comprises albuminuria.
25. The method of claim 21 wherein the one or more disorders comprises impaired glomerular clearance.
26. The method of claim 21 wherein the one or more disorders comprises impaired creatinine clearance.
27. The method of claim 21 wherein the one or more disorders retinopathy.
28. The method of claim 21 wherein the one or more disorders comprises neurodegenerative disease.
US10/834,419 1995-09-12 2004-04-28 Advanced glycation end-product intermediaries and post-amadori inhibition Abandoned US20040220090A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US10/834,419 US20040220090A1 (en) 1995-09-12 2004-04-28 Advanced glycation end-product intermediaries and post-amadori inhibition

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US362895P 1995-09-12 1995-09-12
US08/711,555 US5985857A (en) 1995-09-12 1996-09-10 Advanced glycation end-product intermediaries and post-amadori inhibition
US09/322,569 US6472400B1 (en) 1995-09-12 1999-05-28 Advanced gylcation end-product intermediaries and post-Amadori inhibition
US09/520,933 US6750209B1 (en) 1995-09-12 2000-03-08 Advanced glycation end-product intermediaries and post-amadori inhibition
US10/834,419 US20040220090A1 (en) 1995-09-12 2004-04-28 Advanced glycation end-product intermediaries and post-amadori inhibition

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/520,933 Continuation US6750209B1 (en) 1995-09-12 2000-03-08 Advanced glycation end-product intermediaries and post-amadori inhibition

Publications (1)

Publication Number Publication Date
US20040220090A1 true US20040220090A1 (en) 2004-11-04

Family

ID=32397796

Family Applications (2)

Application Number Title Priority Date Filing Date
US09/520,933 Expired - Lifetime US6750209B1 (en) 1995-09-12 2000-03-08 Advanced glycation end-product intermediaries and post-amadori inhibition
US10/834,419 Abandoned US20040220090A1 (en) 1995-09-12 2004-04-28 Advanced glycation end-product intermediaries and post-amadori inhibition

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US09/520,933 Expired - Lifetime US6750209B1 (en) 1995-09-12 2000-03-08 Advanced glycation end-product intermediaries and post-amadori inhibition

Country Status (1)

Country Link
US (2) US6750209B1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050014799A1 (en) * 2003-06-20 2005-01-20 Biostratum, Inc. Pyridoxamine for the treatment of diabetic kidney disease
US20090082407A1 (en) * 2004-06-18 2009-03-26 Biostratum, Inc. Pyridoxamine for the Treatment of Diabetic Kidney Disease

Families Citing this family (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6750209B1 (en) * 1995-09-12 2004-06-15 Kansas University Medical Center Advanced glycation end-product intermediaries and post-amadori inhibition
US6436969B1 (en) * 1995-09-12 2002-08-20 Kansas University Medical Center Research Institute Inc. Dialysis solutions and methods
US7030146B2 (en) * 1996-09-10 2006-04-18 University Of South Carolina Methods for treating diabetic neuropathy
EP1534679A4 (en) * 2002-08-30 2007-06-06 Biostratum Inc Inhibitors of post-amadori advanced glycation end products
WO2005077902A2 (en) * 2004-02-09 2005-08-25 Biostratum, Inc. Methods for the synthesis of pyridoxamine
US7666442B2 (en) * 2004-08-31 2010-02-23 Tracie Martyn International, Llc Topical compositions comprising benfotiamine and pyridoxamine
DE102004050353A1 (en) * 2004-10-15 2006-04-20 Basf Ag Use of stable ammonium salts of lipoic acid for the treatment of diabetic and other disorders
PL1993569T3 (en) 2006-02-03 2014-11-28 Obagi Cosmeceuticals Llc Anti-aging treatment using copper and zinc compositions
US7897800B2 (en) 2006-02-03 2011-03-01 Jr Chem, Llc Chemical compositions and methods of making them
US7687650B2 (en) 2006-02-03 2010-03-30 Jr Chem, Llc Chemical compositions and methods of making them
US7867522B2 (en) 2006-09-28 2011-01-11 Jr Chem, Llc Method of wound/burn healing using copper-zinc compositions
US8273791B2 (en) 2008-01-04 2012-09-25 Jr Chem, Llc Compositions, kits and regimens for the treatment of skin, especially décolletage
WO2010085753A1 (en) 2009-01-23 2010-07-29 Jr Chem, Llc Rosacea treatments and kits for performing them
US8952057B2 (en) 2011-01-11 2015-02-10 Jr Chem, Llc Compositions for anorectal use and methods for treating anorectal disorders

Citations (45)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4544550A (en) * 1982-10-05 1985-10-01 Rodolfo Almanzor Y Method for the treatment of diabetes
US4665192A (en) * 1984-03-19 1987-05-12 The Rockefeller University 2-(2-furoyl)-4(5)-2(furanyl)-1H-imidazole
US4758583A (en) * 1984-03-19 1988-07-19 The Rockefeller University Method and agents for inhibiting protein aging
US4847083A (en) * 1986-04-02 1989-07-11 Dermasciences, Inc. Two-step procedure for indolent wound healing and aqueous medium and topical ointment used in connection therewith
US4908446A (en) * 1985-11-14 1990-03-13 The Rockefeller University Inhibitors of nonenzymatic cross-linking
US4983604A (en) * 1987-11-13 1991-01-08 The Rockefeller University Inhibitors of nonenzymatic cross-linking
US5017696A (en) * 1987-09-17 1991-05-21 The Rockefeller University Advanced glycosylation end products and associated methods
US5100919A (en) * 1984-03-19 1992-03-31 The Rockefeller University Biguanides and derivatives thereof as inhibitors of advanced glycosylation of a target protein
US5106877A (en) * 1984-03-19 1992-04-21 The Rockefeller University Aminoalcohol compounds in methods of use as inhibitors of the advanced glycosylation of proteins and methods of use therefor
US5130324A (en) * 1984-03-19 1992-07-14 The Rockefeller University 2-alkylidene-aminoguanidines and methods of use therefor
US5137916A (en) * 1985-11-14 1992-08-11 The Rockefeller University Advanced glycation inhibitors containing amino-benzoic acids and derivatives, and methods of use
US5140048A (en) * 1984-03-19 1992-08-18 The Rockefeller University Inhibitors of nonenzymatic cross-linking
US5218001A (en) * 1984-03-19 1993-06-08 The Rockefeller University Inhibitors of the advanced glycosylation of proteins and methods of use therefor
US5221683A (en) * 1984-03-19 1993-06-22 The Rockefeller University Diaminopyridine compounds and methods of use
US5242593A (en) * 1988-12-09 1993-09-07 Oberkofler Joerg Method for reducing the build-up of slime and/or film in water circulation systems
US5254572A (en) * 1987-11-27 1993-10-19 Vesta Medicines (Pty) Ltd. Method and composition for supplementing vitamin B6 where the PN-PLP pathway is disturbed
US5258381A (en) * 1984-03-19 1993-11-02 The Rockefeller University 2-substituted-2-imidazolines
US5262152A (en) * 1984-03-19 1993-11-16 The Rockefeller University Amidrazones and derivatives thereof
US5272165A (en) * 1984-03-19 1993-12-21 The Rockefeller University 2-alkylidene-aminoguanidines and methods of use therefor
US5272176A (en) * 1984-03-19 1993-12-21 The Rockefeller University Advanced glycation inhibitors containing amino-benzoic acids and derivatives, and methods of use
US5288716A (en) * 1987-02-18 1994-02-22 Ulrich Speck Use of pyridoxine derivatives in the prevention and treatment of hyperlipidaemia and atherosclerosis
US5318982A (en) * 1984-03-19 1994-06-07 The Rockefeller University Inhibition of the advanced glycosylation of proteins using substituted-1,2,4-triazoles
US5334617A (en) * 1984-03-19 1994-08-02 The Rockefeller University Amino acids useful as inhibitors of the advanced glycosylation of proteins
US5358960A (en) * 1984-03-19 1994-10-25 The Rockefeller University Method for inhibiting advanced glycosylation of proteins using aminosubstituted imidazoles
US5470839A (en) * 1993-04-22 1995-11-28 Clintec Nutrition Company Enteral diet and method for providing nutrition to a diabetic
US5569670A (en) * 1992-06-05 1996-10-29 Asta Medica Aktiengesellschaft Combination medications containing alpha-lipoic acid and related
US5631271A (en) * 1986-11-29 1997-05-20 Serfontein; Willem J. Methods and preparations for the treatment and prophylaxis of metabolic disturbances
US5668117A (en) * 1991-02-22 1997-09-16 Shapiro; Howard K. Methods of treating neurological diseases and etiologically related symptomology using carbonyl trapping agents in combination with previously known medicaments
US5977073A (en) * 1991-06-06 1999-11-02 Life Sciences' Technologies, Inc. Nutrient composition for treatment of immune disorders
US5985857A (en) * 1995-09-12 1999-11-16 Kansas University Medical Center Advanced glycation end-product intermediaries and post-amadori inhibition
US6228858B1 (en) * 1995-09-12 2001-05-08 University Of Kansas Medical Center Advanced glycation end-product intermediaries and post-amadori inhibition
US6342500B1 (en) * 1999-03-05 2002-01-29 Kansas University Medical Center Post-Amadori inhibitors of Advanced Glycation reactions
US6436969B1 (en) * 1995-09-12 2002-08-20 Kansas University Medical Center Research Institute Inc. Dialysis solutions and methods
US6444221B1 (en) * 1992-06-30 2002-09-03 Howard K. Shapiro Methods of treating chronic inflammatory diseases using carbonyl trapping agents
US6489345B1 (en) * 1999-07-13 2002-12-03 Medicure, Inc. Treatment of diabetes and related pathologies
US20030013746A1 (en) * 1996-09-10 2003-01-16 University Of Kansas Medical Center. Advanced glycation end-product intermediaries and post-amadori inhibition
US6521645B2 (en) * 2000-11-20 2003-02-18 The University Of Kansas Medical Center Methods for the treatment and prevention of urinary stone disease
US6716858B1 (en) * 1995-08-28 2004-04-06 Kansas University Medical Center Methods for inhibiting diabetic complications
US6730686B1 (en) * 1995-09-12 2004-05-04 Kansas University Medical Center Methods for inhibiting oxidative modification of proteins
US6740668B1 (en) * 1995-08-28 2004-05-25 Kansas University Medical Center Methods for inhibiting diabetic complications
US6746678B1 (en) * 1991-02-22 2004-06-08 Howard K. Shapiro Method of treating neurological diseases and etiologically related symptomology using carbonyl trapping agents in combination with medicaments
US6750209B1 (en) * 1995-09-12 2004-06-15 Kansas University Medical Center Advanced glycation end-product intermediaries and post-amadori inhibition
US20050014799A1 (en) * 2003-06-20 2005-01-20 Biostratum, Inc. Pyridoxamine for the treatment of diabetic kidney disease
US6890943B2 (en) * 1999-03-08 2005-05-10 Medicure Inc. Pyridoxal analogues and methods of treatment
US7030146B2 (en) * 1996-09-10 2006-04-18 University Of South Carolina Methods for treating diabetic neuropathy

Family Cites Families (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR4673M (en) 1965-05-19 1966-12-19
DE1915467C2 (en) 1969-03-26 1971-05-13 Olin Mathieson Crosshair for optical aiming devices
CH607773A5 (en) 1974-12-24 1978-10-31 Steigerwald Arzneimittelwerk Process for the preparation of novel pyridoxine derivatives
DE2461742C2 (en) 1974-12-28 1983-01-27 Steigerwald Arzneimittelwerk Gmbh, 6100 Darmstadt Pyridoxine-5'-phosphoric acid ester derivatives and their preparation and pharmaceuticals containing these compounds
FR2349330A1 (en) 1976-04-29 1977-11-25 Savini Emile Compsns. contg. pyridoxine and an antioxidant esp. tocopherol - used for treatment of hyperlipidaemia
AU600306B2 (en) * 1986-09-12 1990-08-09 Rockefeller University, The Methods and agents for removing advanced glycosylation endproducts
DE3705549A1 (en) * 1987-02-18 1988-09-01 Ulrich Speck USE OF PYRIDOXINE DERIVATIVES IN THE PROPHYLAXIS AND THERAPY OF HYPERLIPIDAEMIA AND ATHEROSCLEROSIS
DE69113938T2 (en) 1990-02-19 1996-04-11 Senju Pharma Co Compositions of Maillard reaction inhibitors.
CA2055990A1 (en) 1990-11-22 1992-05-23 David G. Schena Phospholipid-vitamin derivatives and methods for preparation thereof

Patent Citations (55)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4544550A (en) * 1982-10-05 1985-10-01 Rodolfo Almanzor Y Method for the treatment of diabetes
US5334617A (en) * 1984-03-19 1994-08-02 The Rockefeller University Amino acids useful as inhibitors of the advanced glycosylation of proteins
US5258381A (en) * 1984-03-19 1993-11-02 The Rockefeller University 2-substituted-2-imidazolines
US5358960A (en) * 1984-03-19 1994-10-25 The Rockefeller University Method for inhibiting advanced glycosylation of proteins using aminosubstituted imidazoles
US4758583A (en) * 1984-03-19 1988-07-19 The Rockefeller University Method and agents for inhibiting protein aging
US4665192A (en) * 1984-03-19 1987-05-12 The Rockefeller University 2-(2-furoyl)-4(5)-2(furanyl)-1H-imidazole
US5318982A (en) * 1984-03-19 1994-06-07 The Rockefeller University Inhibition of the advanced glycosylation of proteins using substituted-1,2,4-triazoles
US5100919A (en) * 1984-03-19 1992-03-31 The Rockefeller University Biguanides and derivatives thereof as inhibitors of advanced glycosylation of a target protein
US5106877A (en) * 1984-03-19 1992-04-21 The Rockefeller University Aminoalcohol compounds in methods of use as inhibitors of the advanced glycosylation of proteins and methods of use therefor
US5128360A (en) * 1984-03-19 1992-07-07 The Rockefeller University Method and agents for inhibiting protein aging
US5130337A (en) * 1984-03-19 1992-07-14 The Rockefeller University Amidrazones and derivatives thereof
US5130324A (en) * 1984-03-19 1992-07-14 The Rockefeller University 2-alkylidene-aminoguanidines and methods of use therefor
US5272176A (en) * 1984-03-19 1993-12-21 The Rockefeller University Advanced glycation inhibitors containing amino-benzoic acids and derivatives, and methods of use
US5140048A (en) * 1984-03-19 1992-08-18 The Rockefeller University Inhibitors of nonenzymatic cross-linking
US5175192A (en) * 1984-03-19 1992-12-29 The Rockefeller University Inhibitors of the advanced glycosylation of proteins and methods of use therefor
US5218001A (en) * 1984-03-19 1993-06-08 The Rockefeller University Inhibitors of the advanced glycosylation of proteins and methods of use therefor
US5221683A (en) * 1984-03-19 1993-06-22 The Rockefeller University Diaminopyridine compounds and methods of use
US5238963A (en) * 1984-03-19 1993-08-24 The Rockefeller University Method and agents for inhibiting protein aging
US5272165A (en) * 1984-03-19 1993-12-21 The Rockefeller University 2-alkylidene-aminoguanidines and methods of use therefor
US5262152A (en) * 1984-03-19 1993-11-16 The Rockefeller University Amidrazones and derivatives thereof
US4908446A (en) * 1985-11-14 1990-03-13 The Rockefeller University Inhibitors of nonenzymatic cross-linking
US5137916A (en) * 1985-11-14 1992-08-11 The Rockefeller University Advanced glycation inhibitors containing amino-benzoic acids and derivatives, and methods of use
US4847083A (en) * 1986-04-02 1989-07-11 Dermasciences, Inc. Two-step procedure for indolent wound healing and aqueous medium and topical ointment used in connection therewith
US5631271A (en) * 1986-11-29 1997-05-20 Serfontein; Willem J. Methods and preparations for the treatment and prophylaxis of metabolic disturbances
US5288716A (en) * 1987-02-18 1994-02-22 Ulrich Speck Use of pyridoxine derivatives in the prevention and treatment of hyperlipidaemia and atherosclerosis
US6066659A (en) * 1987-02-18 2000-05-23 Steigerwald Arzneimittelwerk Gmbh Use of pyridoxine derivatives in the prevention and treatment of hyperlipidaemia and atherosclerosis
US5017696A (en) * 1987-09-17 1991-05-21 The Rockefeller University Advanced glycosylation end products and associated methods
US4983604A (en) * 1987-11-13 1991-01-08 The Rockefeller University Inhibitors of nonenzymatic cross-linking
US5254572A (en) * 1987-11-27 1993-10-19 Vesta Medicines (Pty) Ltd. Method and composition for supplementing vitamin B6 where the PN-PLP pathway is disturbed
US5242593A (en) * 1988-12-09 1993-09-07 Oberkofler Joerg Method for reducing the build-up of slime and/or film in water circulation systems
US5668117A (en) * 1991-02-22 1997-09-16 Shapiro; Howard K. Methods of treating neurological diseases and etiologically related symptomology using carbonyl trapping agents in combination with previously known medicaments
US6746678B1 (en) * 1991-02-22 2004-06-08 Howard K. Shapiro Method of treating neurological diseases and etiologically related symptomology using carbonyl trapping agents in combination with medicaments
US5977073A (en) * 1991-06-06 1999-11-02 Life Sciences' Technologies, Inc. Nutrient composition for treatment of immune disorders
US5569670A (en) * 1992-06-05 1996-10-29 Asta Medica Aktiengesellschaft Combination medications containing alpha-lipoic acid and related
US6444221B1 (en) * 1992-06-30 2002-09-03 Howard K. Shapiro Methods of treating chronic inflammatory diseases using carbonyl trapping agents
US5470839A (en) * 1993-04-22 1995-11-28 Clintec Nutrition Company Enteral diet and method for providing nutrition to a diabetic
US6740668B1 (en) * 1995-08-28 2004-05-25 Kansas University Medical Center Methods for inhibiting diabetic complications
US6716858B1 (en) * 1995-08-28 2004-04-06 Kansas University Medical Center Methods for inhibiting diabetic complications
US6750209B1 (en) * 1995-09-12 2004-06-15 Kansas University Medical Center Advanced glycation end-product intermediaries and post-amadori inhibition
US6472400B1 (en) * 1995-09-12 2002-10-29 University Of Kansas Medical Center Advanced gylcation end-product intermediaries and post-Amadori inhibition
US6472411B1 (en) * 1995-09-12 2002-10-29 University Of Kansas Medical Center Advanced glycation end-product intermediaries and post-amadori inhibition
US6436969B1 (en) * 1995-09-12 2002-08-20 Kansas University Medical Center Research Institute Inc. Dialysis solutions and methods
US6730686B1 (en) * 1995-09-12 2004-05-04 Kansas University Medical Center Methods for inhibiting oxidative modification of proteins
US6228858B1 (en) * 1995-09-12 2001-05-08 University Of Kansas Medical Center Advanced glycation end-product intermediaries and post-amadori inhibition
US5985857A (en) * 1995-09-12 1999-11-16 Kansas University Medical Center Advanced glycation end-product intermediaries and post-amadori inhibition
US7030146B2 (en) * 1996-09-10 2006-04-18 University Of South Carolina Methods for treating diabetic neuropathy
US20030013746A1 (en) * 1996-09-10 2003-01-16 University Of Kansas Medical Center. Advanced glycation end-product intermediaries and post-amadori inhibition
US6610852B2 (en) * 1999-03-05 2003-08-26 Kansas University Medical Center Post-Amadori inhibitors of advanced glycation reactions
US6342500B1 (en) * 1999-03-05 2002-01-29 Kansas University Medical Center Post-Amadori inhibitors of Advanced Glycation reactions
US6890943B2 (en) * 1999-03-08 2005-05-10 Medicure Inc. Pyridoxal analogues and methods of treatment
US20040235907A1 (en) * 1999-07-13 2004-11-25 Medicure, Inc. Treatment of diabetes and related pathologies
US6489345B1 (en) * 1999-07-13 2002-12-03 Medicure, Inc. Treatment of diabetes and related pathologies
US20030060490A1 (en) * 2000-11-20 2003-03-27 Paul Voziyan Method for the treatment and prevention of urinary stone disease
US6521645B2 (en) * 2000-11-20 2003-02-18 The University Of Kansas Medical Center Methods for the treatment and prevention of urinary stone disease
US20050014799A1 (en) * 2003-06-20 2005-01-20 Biostratum, Inc. Pyridoxamine for the treatment of diabetic kidney disease

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050014799A1 (en) * 2003-06-20 2005-01-20 Biostratum, Inc. Pyridoxamine for the treatment of diabetic kidney disease
US20060287367A1 (en) * 2003-06-20 2006-12-21 Biostratum, Inc. Pyridoxamine for the treatment of diabetic kidney disease
US20080119525A1 (en) * 2003-06-20 2008-05-22 Biostratum, Inc. Pyridoxamine for the Treatment of Diabetic Intermediaries and Post-Amadori Inhibition
US8067444B2 (en) 2003-06-20 2011-11-29 Nephrogenex, Inc. Pyridoxamine for the treatment of diabetic intermediaries and post-amadori inhibition
US20090082407A1 (en) * 2004-06-18 2009-03-26 Biostratum, Inc. Pyridoxamine for the Treatment of Diabetic Kidney Disease
US20110178139A1 (en) * 2004-06-18 2011-07-21 Thorsten Degenhardt Pyridoxamine for the Treatment of Diabetic Kidney Disease

Also Published As

Publication number Publication date
US6750209B1 (en) 2004-06-15

Similar Documents

Publication Publication Date Title
US6472400B1 (en) Advanced gylcation end-product intermediaries and post-Amadori inhibition
US6228858B1 (en) Advanced glycation end-product intermediaries and post-amadori inhibition
US6436969B1 (en) Dialysis solutions and methods
US7030146B2 (en) Methods for treating diabetic neuropathy
US6750209B1 (en) Advanced glycation end-product intermediaries and post-amadori inhibition
Booth et al. Thiamine pyrophosphate and pyridoxamine inhibit the formation of antigenic advanced glycation end-products: comparison with aminoguanidine
AU762030B2 (en) Methods for inhibiting diabetic complications
US6730686B1 (en) Methods for inhibiting oxidative modification of proteins
US6716858B1 (en) Methods for inhibiting diabetic complications
US20030013746A1 (en) Advanced glycation end-product intermediaries and post-amadori inhibition
US6740668B1 (en) Methods for inhibiting diabetic complications
WO2000021516A2 (en) Methods for inhibiting diabetic complications
EP1190713B1 (en) Advanced glycation end-product intermediaries and post-amadori inhibition
CA2360311C (en) Improved dialysis solutions and methods
WO2000022094A2 (en) Methods for inhibiting oxidative modification of proteins
MXPA00004785A (en) Advanced glycation end-product intermediaries and post-amadori inhibition

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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