EMP2: Ethyl-Methyl, di Methyl, tri Methyl Pyruvate Acid Esters: A Tool for Regulating HbA1c and a Riboswitch Activator

The invention is a novel series of compounds represented by general formula, its analogs, tautomeric forms, stereoisomers, pharmaceutically acceptable salts and polymorphs wherein said compound described is a group of antisense molecules possessing immediate global cellular penetration due to their molecular structure and lipophilicity. The molecules are combinations of ethyl and methyl pyruvates. They induce riboswitch activity and reduce target RNA and nucleic acids such as HbA1c. They reduce inflammation and enhance energy production. These features improve therapeutic outcomes of diabetes, neuropathy and cellular aging.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
CROSS-REFERENCE TO RELATED APPLICATION

This application claims the benefit of U.S. Provisional Application No. 61/521/747 titled “EMP2: Ethyl-Methyl, di Methyl, tri Methyl Pyruvate Acid Esters: A Tool for Regulating HbA1c and a Human Riboswitch Activator” field Aug. 09, 2011 at 23:33:42 EST, which is hereby incorporated by reference in its entirety.

FIELD OF INVENTION

This present invention describes a short systemic and antisense compound. Included are such compounds comprising chemically combined moieties of ethyl and methyl, di Methyl, tri Methyl pyruvic acid esters. Said compound is chemically combined in such a manner as to retain the individual properties of each separately and utilize the combinatorial valency and synergy of joining the two. Such a short targeted compound is useful for the reduction of target nucleic acids and/or proteins such as HbA 1 c, high-affinity nucleotide modifications useful for reducing a target RNA in vivo, in tissues, animals and humans with increased potency and improving therapeutic outcomes.

BACKGROUND OF THE INVENTION

Current U.S. Class: 435/194; 435/69.1; 435/183; 435/252.3; 435/254.11; 435/320.1; 530/350; 536/23.1; 536/23.2; 536/23.5

Internal Class: AO1N 037/12; A61K 037/26; A61 K 031/198,70,19,22 C07D487/06; A61K31/55; A61P35/00; A61P35/28

Field of Search: 514/12,866; 435/69.1; 435/183; 435/194; 435/252.3; 435/254.11; 435/320.1; 530/300,324; 530/350; 536/23.1; 536/23.2; 536/23.5; 424.94.1

Diseases such as type II diabetes (DM2) are caused by insufficient energy required to continuously maintain the structure and function in cells. All cells in DM2 produce less insulin or less effective insulin. The cell membrane receptor sites (i.e. glucose transporter Glut4) are reduced in number and effectiveness. The non-utilized glucose increases both intra and extracellularly enabling toxic oxidation and inflammatory products to impair normal cellular function. Finally, cytosolic and nuclear components are malfunctioning, and are in need of repair or replacement. Energy, via the tricarboxylic acid (TCA) cycle, is diminished and thereby further reducing the cellular components of the vesicle associated membrane proteins (VAMP) and decreasing mitochondrial numbers. In effect, this process is decreasing or stopping all function for repair, and if not repaired it then enables gene silence or apoptosis. All crosstalk and synchronization with the cellular network is lessened and the productivity and capability of the cell diminishes or ceases.

Treatment with ethyl pyruvate has been shown to improve survival and ameliorate organ dysfunction in a wide variety of preclinical models of critical illnesses, such as severe sepsis, acute respiratory distress syndrome, burn injury, acute pancreatitis and stroke. Ethyl pyruvate has been shown to improve cardiac function after coronary ischemia and reperfusion. This ester was originally regarded as a way to administer pyruvate, while avoiding some of the problems associated with the instability of pyruvate in aqueous solutions. Increasingly, however, it is becoming apparent that certain pyruvate esters, including ethyl pyruvate, have pharmacological effects, such as suppression of inflammation, which are distinct from those exerted by pyruvate anion. Ethyl pyruvate inhibits pathways leading to Advanced Glycation Endproduct (AGE) and the combination of Ethyl and Methyl pyruvate will work better synergistically than as to the respective elements by themselves, for cell life prolongation and cancer inhibition.

Methyl pyruvate (MP) performs as a lipophilic antisense bullet that penetrates cell membranes, mitochondrial membranes and nuclear membranes with its active and passive delivery of energy via protons and adenosine triphosphate (ATP). This metabolic bullet up-regulates all cytosolic and nuclear capabilities. Now, because of instant protons and pyruvate delivered, there is immediately empowered TCA production of ATP and the cell functions with unparalleled performance. Through this newly available energy, gene silencing is reversed, repairs completed, apoptosis performed, and the cell is upgraded and/or divides into a new and improved cell having shed its prior oxidative cellular debris. In DM2 this means improved Glut4 networking, improved insulin sensitivity, increased intra and intercellular networking and increased superoxide dismutase (SOD) production. Further, due to MP, nutrients and complicit molecules will now function more effectively and efficiently, i.e. as increased cellular energy can now effect niacinamide (B3) and nicotinamide adenine dinucleotide (NAD) at an intercellular and intracellular level to enable and protect beta cell function, reduction of HbA1c, increase cell life span and to prepare FOXO3a to stop proteolysis initiation, or initiation of apoptosis, when and where appropriate in the cell cycle. With all vitamins, all amino acids, all nutrients, hydronium ion and water transport, all cation and anion channel functions, lipid metabolism, protein metabolism, glucose metabolism, organ functioning, genomic protection replication and functioning, MP enables their individual physiologic roles and applications.

Ethyl and Methyl pyruvates have been tested in human volunteers and have been shown to be safe in clinically prescribed doses.

REFERENCES CITED [REFERENCED BY] U.S. Patent Documents

U.S. Pat. No.20100047221 Feb. 25, 2010, Alexander, Ranya L., 424.94.1 COMPOSITIONS COMPRISING PYRUVATE ALKYL ESTERS AND USES THEREOF
U.S. Pat. No. 5,045,454 September, 1991 Bertheussen 435/29.
U.S. Pat. No. 5,091,404 February, 1992 Elgebaly 514/401.
U.S. Pat. No. 5,192,762 March, 1993 Gray et al. 514/249.
U.S. Pat. No. 5,210,098 May, 1993 Nath 514/577.
U.S. Pat. No. 5,321,030 June, 1994 Kaddurah-Daouk et al. 514/275.
U.S. Pat. No. 5,324,731 June, 1994 Kaddurah-Daouk et al. 514/275.
U.S. Pat. No. 5,741,661 Apr., 1998 Goldin et al. 435/29.

Foreign References:

EP0075805 1983-04 CO7D 501/20 KYOWA HAKKO KOGYO CO., LTD Beta-lactam compound and a pharmaceutical composition containing the same
EP0233780 1987-08 CO7D 501/36 ELI LILLY AND COMPANY 0-substituted oximino cephalosporins

EP0370629 1990-05 CO7C 251/60 IMPERIAL CHEMICAL INDUSTRIES PLC Fungicides

EPO400805 1990-12 CO7D 501/20 Ishimaru, Toshiyasu Cephalosporin compounds and their use

EP0506149 1992-09 CO7C 251/60 IMPERIAL CHEMICAL INDUSTRIES PLC Fungicides

EP0581187 1994-02 CO7C 251/54 ONO PHARMACEUTICAL CO., LTD. Oxime derivatives
EP0708098 1996-04 CO7D 277/34 SANKYO COMPANY LIMITED Oxime derivatives, their preparation and their therapeutic use

EP0916651 1999-05 CO7C 259/02 Sankyo Company, Limited PHENYLALKYLCARBOXYLIC ACID DERIVATIVES CO7D 213/53 SANKYO CO LTD MEDICINE CONTAINING PHENYLALKYLCARBOXYLIC ACID DERIVATIVE W09602507 1996-02 CO7D 215/14 ABBOTT LABORATORIES IMINOXYCARBOXYLATES AND DERIVATIVES AS INHIBITORS OF LEUKOTRIENE BIOSYNTHESIS WO9633724 1996-10 A61K 31/557 EVANS, Ronald, M. SELECTIVE MODULATORS OF PEROXISOME PROLIFERATOR ACTIVATED RECEPTOR-GAMMA, AND METHODS FOR THE USE THEREOF WO9638427 1996-12 CO7D 263/44 FUJIMOTO, Koichi AROMATIC OXYIMINO DERIVATIVES WO9640128 1996-12 A61K 31/425 THE SALK INSTITUTE FOR BIOLOGICAL STUDIES MODULATORS OF PEROXISOME PROLIFERATOR ACTIVATED RECEPTOR-GAMMA, AND METHODS FOR THE USE THEREOF WO9725042 1997-07 A61K 31/42 SMITH, STEPHEN, ALISTAIR USE OF AN ANTAGONIST OF PPAR-ALPHA AND PPAR-GAMMA FOR THE TREATMENT OF SYNDROME X WO9731907 1997-09 CO7D 263/56 BOSWELL, GRADY, EVAN SUBSTITUTED 4-HYDROXY-PHENYLALCANOIC ACID DERIVATIVES WITH AGONIST ACTIVITY TO PPAR-GAMMA WO9805331 1998-02 A61K 31/45 LIGAND PHARMACEUTICALS INCORPORATED PREVENTION OR TREATMENT OF TYPE 2 DIABETES OR CARDIOVASCULAR DISEASE WITH PPAR MODULATORS WO9904815 1999-02 A61K45/00 HASHIMOTO, SEIICHI MEDICINAL COMPOSITIONS WITH CHOLESTEROL-LOWERING EFFECT

WO9958510A1 issued 1999-Nov.-18 OXYIMINOALKANOIC ACID DERIVATIVES WITH HYPOGLYCEMIC AND HYPOLIPIDEMIC ACTIVITY

SUMMARY OF INVENTION

Gene regulatory networks, or pathways, previously associated with DM2 include systemic immune indices and inflammatory response, cell signaling and metabolism, sarcomere (muscle-spindle) and cytoloskeletal organization, transcription/translation and most notably apoptosis. Most conventional methods used to discover dysregulated pathways in diabetes (DM2) have relied heavily on fundamentally a single-gene approach combined with Gene Ontology or self-defined functional annotations. Our efforts differs in that we also employ biological/biochemical pathway-based analytic techniques to detect over-arching and cross correlated patterns of heightened gene expression.

This invention, ethyl pyruvate & methylated pyruvates (mono, di, tri), are lipophilic antisense permeants, each of which holds on to its own membrane penetrability, as well as acting collectively to produce a time-released delivery within the inner mitochondria. The combinational synergy is greater than each can effect individually. Their separate and combinatorial, targeted molecular capabilities are useful in reduction of target nucleic acids, and or proteins such as HbA1c, directly enabling improved therapeutic outcomes. Because of the active and passive energy delivery by the invention, all intracellular and intra-nuclear functions are upgraded and rebooted with enhanced intracellular crosstalk and signaling; the genome is also rebooted in an enhanced manner. The more efficient genomic guidance allows improved glucose control that disables oncoming crippling diseases from getting a foothold; energy stays the up-regulation of all cellular function that cannot allow cellular deterioration. The same guidance mechanism demonstrates certain esters of pyruvate have pharmaceutical effects, such as reducing the inflammatory response. Also, mitochondrial expression enacts changes, via the nuclear genomic creation of these sustaining organelles, to provide upstream molecular regulators: the master regulators being PGC1a and AMPK. The targets homeostatically and instantly recalibrate from PGC1a and AMPK with the downstream signaling molecules coming from the activities of SIRTS, FOXO, CAMP, CREB and PPARs. Their communication and crosstalk enable a synchrony of NADH/NAD/FAD/Acetyl fuel/PO4 regulation, as well as glycolysis with its electron and proton transfer and gradient balancing cellular requirements each and every picosecond with all the above molecules and their networking.

In the subpopulation with Hb1c elevations greater than 6.0, PGC1a is decreased, thereby diminishing the cells restorative capability every picosecond until the upstream and downstream molecular crosstalk lessens or disappears and cellular function goes on hold for repair. Further PGC1a's continuing decrease creates cell malfunction until cell silencing or apoptosis occurs. Every molecule needed for upstream—downstream constant cellular energy balance and genomic homeostasis demands specific energy requirements for their nuclear or cytosolic organelles' creation and sustenance. Less energy yields less genomic expression with constantly diminished cellular capabilities, then cellular default.

DETAILED DESCRIPTION OF INVENTION

Methylated pyruvates (MP) and ethyl pyruvate (EP) are specific organic esters, existing as tautomeric and anionic compounds, that deliver energy to cells and circumvent preparation via glycolysis, fermentation, tricarboxylic acid (TCA), or any other cellular manufacturing energy systems. MP and EP are lipotrophic molecules that traverse cellular barriers within the human body. “MP” and “EP”, as used herein, refer to all tautomeric and charged forms of the compounds.

Treatment with ethyl pyruvate has been shown to improve survival and ameliorate organ dysfunction in a wide variety of preclinical models of critical illnesses, such as severe sepsis, acute respiratory distress syndrome, burn injury, acute pancreatitis and stroke. Ethyl pyruvate has been shown to improve cardiac function after coronary ischemia and reperfusion. This ester was originally regarded as a way to administer pyruvate, while avoiding some of the problems associated with the instability of pyruvate in aqueous solutions. Increasingly, however, it is becoming apparent that certain pyruvate esters, including ethyl pyruvate, have pharmacological effects, such as suppression of inflammation, which are distinct from those exerted by pyruvate anion. Ethyl pyruvate has been tested in human volunteers and shown to be safe at clinically relevant doses.

Reduction of inflammation and Oxidative Stress is primary to reversing the diabetic condition. Oxidative stress is known to induce senescence prematurely in fibroblasts. Cellular senescence or cellular ageing is the phenomenon where normal diploid differentiated cells lose the ability to divide. In response to DNA damage (including shortened telomeres) cells either age or go into apoptosis if the damage cannot be repaired. There is strong evidence as mentioned above, that oxidative stress is increased in diabetic patients. Other studies have revealed that endothelial cells in atherosclerotic lesions show features of cellular senescence, like senescence associated β-galactosidase (SA-β-gal) staining and telomere shortening. Expression of inflammatory cytokines and adhesion molecules is up-regulated in senescent endothelial cells. Furthermore, nitric oxide production is significantly reduced in these cells. More importantly, senescence enhances vascular inflammation and thrombosis in vessels, promoting the development of cardiovascular events. There is also evidence that senescence is more accelerated in patients with diabetes compared to healthy individuals. One study demonstrated that high glucose induced premature cellular senescence in HUVECs through the activation of the Apoptosis Signal-Regulating Kinase 1 (ASK1). Activation of ASK-1 also up-regulated PAI-1 expression in the HUVECs and this plus senescence was also observed in aortas of STZ-diabetic wild type mice, whereas this was not seen in STZ-diabetic ASK-1 knock-out mice. PAI-1 is known to play an important role in the pathogenesis of inflammatory diseases.

MP typifies all these alkyl ester molecules. All cytosolic and nuclear structures are targets of these molecules. Upon cell membrane penetration, MP delivers 3 protons or 12 adenosine triphosphates (ATP's) per methyl moiety. Every molecule delivers the methyl cation (CH3), and ethyl or pyruvate moieties. This energy up-regulates cellular functions. The methyl groups boost genome expression (genetically & epigenetically) by loosening the histone sheathe and enabling further expressed phenotypic enhancement thus allowing increased efficiency in part by RNA editing (RNAi) of cellular function. Methyl groups engender ribosome switches to RNA edit, RNAi up-regulate and the genome to be become better expressed intra-cellularly. MP as a prototype of these alkyl esters of pyruvate requires no preparation to start the Krebs cycle at the pyruvate—Acetyl coA junction. The resulting reaction/bio-cascade causes further production of ATP, NADH and FADH, thus engendering the energy cycles needed for cellular homeostasis and or up-regulating the genome through RNA editing.

MP is a small, concise, antisense molecule that seamlessly penetrates the outer and inner membranes of the mitochondria (TOM and TIM) for nutrient delivery. Similarly, MP easily penetrates the nuclear membrane to be used as an epigenetic tool for enhancing cellular function at the genomic level.

Any cell, with its diminishing energy production, maintains normal cell function as long as possible; after which, it repairs itself, and or then enables gene silencing (RNAi) if due to significant disease. As a last resort, it dies via programmed suicide (apoptosis) or disintegration (necrosis). The specific direction a cell takes closely correlates with the totality of its energy production. Disease and ageing impairs the cell's ability to produce sufficient energy to neutralize and remove toxic wastes. Normally cellular respiration expires water (H2O) and carbon dioxide (CO2). Normally, cell functions are performed without toxicity or depletion. Decreasing energy mitigates this perfection. As a consequence, disease and subsequent death ensue.

Pyruvate alkyl esters refer to straight chain alkyl groups with respective methyl group's attached and covalent hydrogen atoms affixed where needed to construct ethyl pyruvate, as well as methyl, dimethyl and trimethyl pyruvates. All the aforementioned esters are short and concise antisense compounds that initiate anti-inflammation upon delivery to any cell, through mitogen-activated protein kinases (MAPK), Methyl ethyl ketone (MEK) and RAS pathways. Pyruvate alkyl esters enter the mitochondrial cristae engendering energy with little to no cellular effort or additional preparation. The up-regulation of the upstream—downstream molecules AMPK, PGC1a, FOXO, PPARs, CAMP, CREB, SIRTS, enable enhanced energy synchrony that reduces oxidative stress with anti-inflammation and repair begins.

Diabetic cells are energy deprived, causing a decrease in cellular energy generation, energy use, transference and storage. In such cells, the enormous number of cell signaling molecules and the vesicle associated membrane proteins (VAMPs) for nuclear-cell membrane crosstalk and work begin to disappear or lessen. A void of energy for the production of these proteins is created. Use of these antisense compounds reduces glycated proteins, such as HbA1c and fructosamines, as well as specific nucleic acids, thus restoring cellular organelles, Vamp and nuclear capability. In the weakened diabetic cell, oxidative stress engenders reactive oxygen species (ROS), causing multisystem impairments with both intra and extra cellular toxicity and resultant peroxynitriles, peroxinitrates, hydroxyls, advanced glycation end products (AGEs), carbonylates, etc. The result induces increased iron presence manifesting further oxidative stress with increased protein aggregation, cross-linking, misfolding, and overall energy deterioration. A restorative enzyme, Parp 1 (Poly [ADP-ribose] polymerase 1) cannot keep up with overall repair evoking increased iRNA silencing with further cell weakening or apoptosis. Beta islet cells in aggregates, such as those of the pancreatic Islets of Langerhans, now down-regulate, decreasing insulin production. Target or receptor cells lacking specific VAMPS and cell membrane receptor sites become increasingly insulin resistant with decreased intracellular transport, and decreased signaling and crosstalk. The scaffolding and substance of the cytosolic and nuclear fabric becomes depleted with myriad functions put on hold or ended lest the cell can find a new and improved source of energy to reboot thousands of cellular and nuclear pathways. The use of these antisense compounds reduces glycated proteins, such as HbA1c and fructosamines, as well as specific nucleic acids. As a result, through restored energy propagation, cellular function strengthens significantly, yielding better genomic expression.

The GLUT4 isoform is the major insulin-responsive transporter that is predominantly restricted to skeletal and cardiac muscle and adipose tissue. In contrast to the other GLUT isoforms, which are primarily localized to the cell surface membrane, GLUT4 transporter proteins are sequestered into specialized storage vesicles that remain within the cell's interior under basal conditions. As post-prandial glucose levels rise, the subsequent increase in circulating insulin activates intracellular signaling cascades that ultimately result in the translocation of the GLUT4 storage compartments to the plasma membrane. Importantly, this process is readily reversible such that when circulating insulin levels decline, GLUT4 transporters are removed from the plasma membrane by endocytosis and are recycled back to their intracellular storage compartments. Numerous studies have demonstrated the importance of normal GLUT4 expression and cellular localization in regulating glucose homeostasis. It is estimated that up to 70% of blood glucose is cleared by GLUT4 in muscle.

Methyl Pyruvate (MP) alone creates a six-fold increase in insulin production at the B-islet pancreatic cell, as compared to its glucose control. As a result, intra islet cell ATP production increases and energy retooling commences for the genome. MP thereby introduces a category of energy delivery, anti-inflammatory action and epigenetic rebooting molecules which significantly up-regulates intracellular signaling in all cells, especially in depleted diabetic cells Pyruvate alkyl esters also perform this action.

MP turns off adenosine triphosphate-sensitive potassium (KATP) and turns on at calcium (Ca++) channels allowing increased intracellular calcium (Ca++i), increased pH and increased aquaporin (AQPO), enabling increased intra-cellular functional efficiency, especially of the TCA cycle and cellular respiration. Glucose metabolism in glycolysis creates 2ATPs and pyruvate generation that cause mitochondrial (TCA) cascades to be initiated from glycolytic start up energy and fuel that further allows insulin secretion via glucose stimulation from islet cells. Glycolysis, (not pyruvate), is required to enable insulin secretion from electrons of the reduced NADH and its transfer to the TCA shuttle that is electron activated: malate-aspartate and glycerol phosphate shuttles. Of note, both MP and glycolysis create ATP and pyruvate that can empower NAD/NADH/NADPH, the malate-aspartate shuttle, turn off KATP and turn on Ca++i channels, fuel TCA and thereby enable insulin glucose homeostasis, “a shuttle alternative”.

This shuttle enables beta islet cell insulin secretion. Glycolysis energy manufacture, via the shuttle, now induces glucose incited insulin secretion enabling glucose oxidation. The lactate-pyruvate production is aided by the activity of this shuttle.

MP and its constituent family of concomitant alkyl esters represent potent antisense molecules. These molecules provide a salient additional impact in that they act very much like non-coding translation initiators quite similar to messenger ribonucleic acid (mRNA). In light of the above and with a plethora of cellular metabolites, MP acts directly on the Beta Islet cell performing the actions of a human riboswitch.

Parp-1, all multicellular organisms must have means of preserving their genomic integrity or face catastrophic consequences such as uncontrolled cell proliferation or massive cell death. One response is a modification of nuclear proteins by the addition and removal of polymers of ADP-ribose that modulate the properties of DNA-binding proteins involved in DNA repair and metabolism. These ADP-ribose units are added by poly(ADP-ribose) polymerase (PARP) and removed by poly(ADP-ribose) glycohydrolase(PARG).

Poly(ADP-ribose) polymerases (PARPs) are defined as cell signaling enzymes that catalyze the transfer of ADP-ribose units from NAD(+) to a number of acceptor proteins. PARP-1, the best characterized member of the PARP family, that presently includes six members, is an abundant nuclear enzyme implicated in cellular responses to DNA injury provoked by genotoxic stress (oxygen radicals, ionizing radiations and monofunctional alkylating agents). Due to its involvement either in DNA repair or in cell death, PARP-1 is regarded as a double-edged regulator of cellular functions. In fact, when the DNA damage is moderate, PARP-1 participates in the DNA repair process. Conversely, in the case of massive DNA injury, overactivation of PARP consumes NAD(+) and consequently ATP, culminating in cell dysfunction or necrosis. This cellular suicide mechanism has been implicated in the pathomechanism of stroke, myocardial ischemia, diabetes, diabetes-associated cardiovascular dysfunction, shock, traumatic central nervous system injury, arthritis, colitis, allergic encephalomyelitis, and various other forms of inflammation.

Living organisms possess mechanisms to regulate cell cycle progression and to preserve genomic integrity. Failure of these mechanisms in multicellular organisms results in disorders ranging from the unregulated cell proliferation associated with cancer to massive cell death after the fall of tissue oxygen and glucose levels in cardiac or brain ischemia. A key cellular response to genomic damage is the posttranslational modification of nuclear proteins in response to DNA strand breaks. One known modification is the addition to specific proteins of up to 200 residues of ADP-ribose to form branched polymers. These polymers act as binding sites for repair proteins that play a central role in DNA metabolism.

The enzyme responsible for the addition of these polymers is PARP1. PARP 1 associates with DNA and with chromatin-binding proteins such as histones, transcription factors, and key DNA repair proteins. Although a number of nuclear proteins such as histones are substrates for PARP1, a major substrate is PARP1 itself, via auto-modification of the BRCA1 COOH-terminal homology region. Regulation of auto-modification of PARP 1 is twofold: through PARP 1- DNA interactions and PARP 1-PARP1 dimerization. PARP1 acts together with the DNA damage repair system to regulate DNA base excision repair, apoptosis, and necrosis.

PARP 1 Inhibition, in recent studies of mouse strains lacking the PARP1 gene have identified two roles for this encoded protein, depending on the extent of DNA damage. Moderate damage elicits a protection response similar to that observed for checkpoint genes, leaving PARP1 knockout mice vulnerable to g-irradiation and alkylating reagents. In cases of extensive DNA damage, PARP1 activity depletes cellular energy pools, which eventually leads to cell death. PARP 1 also has a putative role in signaling DNA damage and in recruiting proteins to sites of double-strand breaks. This hypothesis was based on the ability of proteins, such as p53 and other repair enzymes, to bind to the poly(ADP) polymers present on PARP1. PARP1 inhibitors exaggerate the cytotoxic effects of DNA damage by limiting the ability of cells to regulate DNA base excision repair. In this role, PARP inhibitors are being tested as chemosensitizing agents during cancer chemotherapy.

Another response to more extensive DNA damage mediated by PARP 1 is the promotion of cell death, as seen in cases of ischemic injury. This process can occur when PARP1 activation is highly stimulated and thus consumes large amounts of NAD, the source of ADP-ribose. This condition depletes the cellular energy stores. PARP1 knockout mice are highly resistant to ischemia during stepto-zocin-induced type I diabetes, myocardial infarction, stroke, and neurodegeneration.

In support of a role for PARP1 in cell death in various inflammation processes, several studies have shown protection against cellular injury in numerous target cells by using known PARP1 inhibitors. For many years PARP1 has been the only known PARP. However, modification of cellular proteins with ADP-ribose polymers still occurs in PARP1 knockout mice, suggesting the presence of other proteins with PARP activity. Indeed, new members of the PARP family have been identified based on the presence of domains that share considerable sequence similarity with the catalytic domain of PARP 1.

Although some members of the PARP family do not possess a well-defined Zn 21 finger DNA-binding motif or an auto-modification domain such as that described for PARP1, they still catalyze the formation of ADP-ribose polymers in a DNA-dependent manner and are capable of auto-modification. Two additional members of the PARP family are tankyrase and VPARP

Tankyrase is associated with the telomerase complex that acts to regulate telomere length at replication, and VPARP is a component of a multisubunit complex referred to as a “vault”. The name vault is based on its observed structure by electron microscopy. The cellular location of VPARP is predominantly cytoplasmic; however, there is a small fraction associated with the mitotic spindle. Unlike PARP1, tankyrase and VPARP are not activated by DNA damage. Tankyrase modifies the telomere-binding protein TRF1 in vitro. TRF1 stabilizes the ends of chromosomes, and it has been proposed that modification of TRF1 with ADP-ribose polymers serves to regulate its ability to form a loop structure at chromosome ends. In other studies, tankyrase has been shown to promote telomere elongation in human cells. A substrate of VPARP is the major vault protein, MVP (it is also capable of auto-modification); these complexes are up-regulated in multidrug-resistant cancer cell lines. The various cellular locations and domain structures of the PARP family members strongly suggest that they have distinct cellular roles.

Identification of selective inhibitors might help elucidate the function of these enzymes. Poly(ADP-ribose) polymers can be removed by PARG, a member of a large family of related enzymes. This enzyme is thought to regulate the cellular function of PARP family members by removing ADP-ribose units, which results in changes in the branching pattern of the polymers. There is some evidence to support the hypothesis that polymers synthesized by different PARP orthologues might be hydrolyzed by specific PARGs. Although a complete understanding of the physiological activities of PARPs remains unclear, inhibitors of the activity of PARP 1 and related proteins could provide new therapeutic approaches to both cancer and ischemia caused by reperfusion injury and inflammatory processes.

Excessive activation of poly(ADP-ribose) polymerase 1 (PARP1) leads to NAD(+) depletion and cell death during ischemia and other conditions that generate extensive DNA damage. When activated by DNA strand breaks, PARP1 uses NAD(+) as substrate to form ADP-ribose polymers on specific acceptor proteins. These polymers are in turn rapidly degraded by poly(ADP-ribose) glycohydrolase (PARG), a ubiquitously expressed exo- and endoglycohydrolase.

In a study, the role of PARG was examined in the PARP1-mediated cell death pathway. Mouse neuron and astrocyte cultures were exposed to hydrogen peroxide, N-methyl-d-aspartate (NMDA), or the DNA alkylating agent, N-methyl-N′-nitro-N-nitrosoguanidine (MNNG). Cell death in each condition was markedly reduced by the PARP1 inhibitor benzamide and equally reduced by the PARG inhibitors gallotannin and nobotanin B. The PARP1 inhibitor benzamide and the PARG inhibitor gallotannin both prevented the NAD(+) depletion that otherwise results from PARP1 activation by MNNG or H(2)O(2). However, these agents had opposite effects on protein poly(ADP-ribosyl)ation. Immunostaining for poly(ADP-ribose) on Western blots and neuron cultures showed benzamide to decrease and gallotannin to increase poly(ADP-ribose) accumulation during MNNG exposure.

These results suggest that PARG inhibitors do not inhibit PARP1 directly, but instead prevent PARP1-mediated celideath by slowing the turnover of poly(ADP-ribose) and thus slowing NAD(+) consumption. PARG appears to be a necessary component of the PARP-mediated cell death pathway, and PARG inhibitors may have promise as neuroprotective agents.

Poly(ADP-ribose) polymerase-1 (PARP-1) is a nuclear enzyme that is involved in DNA repair and activated by DNA damage. When activated, PARP-1 consumes NAD(+) to form ADP-ribose polymers on acceptor proteins. Extensive activation of PARP-1 leads to glycolytic blockade, energy failure, and cell death. These events have been postulated to result from NAD(+) depletion. Here, we used primary astrocyte cultures to directly test this proposal, utilizing the endogenous expression of connexin-43 hemichannels by astrocytes to manipulate intracellular NAD(+) concentrations. Activation of PARP-1 with the DNA alkylating agent N-methyl-N′-nitro-N-nitrosoguanidine (MNNG) produced NAD(+) depletion, glycolytic blockade, and cell death. Cultures incubated in high (10 mM) extracellular concentrations of NAD(+) after MNNG exposure showed normalization of intracellular NAD(+) concentrations. Repletion of intracellular NAD(+) in this manner completely restored glycolytic capacity and prevented cell death. These results suggest that NAD(+) depletion is the cause of glycolytic failure after PARP-1 activation.

Extensive activation of poly(ADP-ribose) polymerase-1 (PARP-1) by DNA damage is a major cause of caspase-independent cell death in ischemia and inflammation. Here it is shown that NAD(+) depletion and mitochondrial permeability transition (MPT) are sequential and necessary steps in PARP-1-mediated cell death. Cultured mouse astrocytes were treated with the cytotoxic concentrations of N-methyl-N′-nitro-N-nitrosoguanidine or 3-morpholinosydnonimine to induce DNA damage and PARP-1 activation. The resulting cell death was preceded by NAD(+) depletion, mitochondrial membrane depolarization, and MPT. Sub-micromolar concentrations of cyclosporin A blocked MPT and cell death, suggesting that MPT is a necessary step linking PARP-1 activation to cell death. In astrocytes, extracellular NAD(+) can raise intracellular NAD(+) concentrations.

To determine whether NAD(+) depletion is necessary for PARP-1-induced MPT, NAD(+) was restored to near- normal levels after PARP-1 activation. Restoration of NAD(+) enabled the recovery of mitochondrial membrane potential and blocked both MPT and cell death. Furthermore, both cyclosporin A and NAD(+) blocked translocation of the apoptosis-inducing factor from mitochondria to nuclei, a step previously shown necessary for PARP-1-induced cell death. These results suggest that NAD(+) depletion and MPT are necessary intermediary steps linking PARP-1 activation to AIF translocation and cell death.

The DNA repair enzyme, poly(ADP-ribose) polymerase-1 (PARP1), contributes to cell death during ischemia/reperfusion when extensively activated by DNA damage. The cell death resulting from PARP1 activation is linked to NAD+ depletion and energy failure. Because glycolysis requires cytosolic NAD+, the authors tested whether PARP1 activation impairs glycolytic flux and whether substrates that bypass glycolysis can rescue cells after PARP1 activation. PARP1 was activated in mouse cortical astrocyte and astrocyte-neuron co-cultures or other mitochondrial substrates to the cultures after MNNG treatment reduced cell death from approximately 70% to near basal levels, while PARP inhibitors and excess glucose had negligible effects. The mitochondrial substrates significantly reduced cell death.

Peroxisomal proliferator-activated receptors (PPARs) belong to a nuclear receptor superfamily of ligand- activated transcription factors. Peroxisome proliferator-activated receptor (PPAR) is activated when a ligand binds to the ligand-binding domain at the side of C-termini.

So far, three types of isoforms of alpha form, gamma form and delta form have been identified as PPARs, and the expression tissues and the functions are different respectively. Peroxisome proliferators are a structurally diverse group of compounds which, when administered to rodents, elicit dramatic increases in the size and number of hepatic and renal peroxisomes, as well as concomitant increases in the capacity of peroxisomes to metabolize fatty acids via increased expression of the enzymes required for the beta-oxidation cycle

The alpha-isoform of peroxisome proliferator-activated receptor (PPAR.alpha) acts to stimulate peroxisomal proliferation in the rodent liver which leads to enhanced fatty oxidation by this organ. (PPAR) alpha is a nuclear receptor that is mainly expressed in tissues with a high degree of fatty acid oxidation such as liver, heart, and skeletal muscle. There is a sex difference in PPARalpha expression. Male rats have higher levels of hepatic PPARalpha mRNA and protein than female rats. Chemicals included in this group are the fibrate class of hypolipidermic drugs, herbicides, and phthalate plasticizers. Peroxisome proliferation can also be elicited by dietary or physiological factors such as a high-fat diet and cold acclimatization. The importance of peroxisomes in humans is stressed by the existence of a group of genetic diseases in man in which one or more peroxisomal functions are impaired. Most of the functions known to take place in peroxisomes have to do with lipids. Indeed, peroxisomes are capable of 1. fatty acid beta-oxidation 2. fatty acid alpha- oxidation 3. synthesis of cholesterol and other isoprenoids 4. ether-phospholipid synthesis and 5. biosynthesis of polyunsaturated fatty acids.

Peroxisome proliferator-activated receptors (PPAR) are nuclear receptors present in several organs and cell types. They are subdivided into PPAR alpha, PPAR gamma and PPAR delta (or beta). PPAR alpha and gamma are the two main categories of these receptors, which are both characterized by their ability to influence lipid metabolism, glucose homeostasis, cell proliferation, differentiation and apoptosis, as well as the inflammatory response, by transcriptional activation of target genes. PPAR alpha are activated by fatty acids, eicosanoids and fibrates, while PPAR gamma activators include arachidonic acid metabolites, oxidized low density lipoprotein and thiazolidinediones. PPAR gamma is predominantly expressed in intestine and adipose tissue, where it triggers adipocyte differentiation and promotes lipid storage. Recently, the expression of PPAR alpha and PPAR gamma was also reported in cells of the vascular wall, such as monocyte/macrophages, endothelial and smooth muscle cells.

The hypolipidemic fibrates and the antidiabetic glitazones are synthetic ligands for PPAR alpha and PPAR gamma, respectively. Furthermore, fatty acid-derivatives and eicosanoids are natural PPAR ligands: PPAR alpha is activated by leukotriene B4, whereas prostaglandin J2 is a PPAR gamma ligand, as well as some components of oxidized LDL, such as 9- and 1 3-HODE. These observations suggested a potential role for PPARs not only in metabolic but also in inflammation control and, by consequence, in related diseases such as atherosclerosis. More recently, PPAR activators were shown to inhibit the activation of inflammatory response genes (such as IL-2, IL-6, IL-8, TNF alpha and metalloproteases) by negatively interfering with the NF-kappa B, STAT and AP-1 signaling pathways in cells of the vascular wall.

The PPAR alpha form has been shown to mediate the action of the hypolipidemic drugs of the fibrate class on lipid and lipoprotein metabolism. PPAR alpha activators furthermore improve glucose homeostasis and influence body weight and energy homeostasis. It is likely that these actions of PPAR alpha activators on lipid, glucose and energy metabolism are, at least in part, due to the increase of hepatic fatty acid beta-oxidation resulting in an enhanced fatty acid flux and degradation in the liver.

PPARs are expressed in different immunological and vascular wall cell types where they exert anti-inflammatory and proapoptotic activities. The observation that these receptors are also expressed in atherosclerotic lesions suggests a role in atherogenesis. Finally, PPAR alpha activators correct age-related dysregulations in redox balance. Taken together, these data indicate a modulatory role for PPAR alpha in the pathogenesis of age-related disorders, such as dyslipidemia, insulin resistance and chronic inflammation, predisposing to atherosclerosis.

Synthetic antidiabetic thiazolidinediones (TZDs) (two such compounds are rosiglitazone and pioglitazone) and natural prostaglandin D(2) (PGD(2)) metabolite, 15-deoxy-Delta(12, 14)-prostaglandin J(2) (15d-PGJ(2)), are well-known as ligands for PPAR gamma. After it has been reported that activation of PPAR gamma suppresses production of proinflammatory cytokines in activated macrophages, medical interest in PPAR gamma have grown and a huge research effort has been concentrated. PPAR gamma, is currently known to be implicated in various human chronic diseases such as diabetes mellitus, atherosclerosis, rheumatoid arthritis, inflammatory bowel disease, and Alzheimer's disease.

PPAR gamma ligands have potent tumor modulatory effects against colorectal, prostate, and breast cancers. Recent studies suggest that TZDs not only ameliorate insulin sensitivity but also have pleiotropic effects on many tissues and cell types. Although activation of PPAR gamma seems to have beneficial effects on atherosclerosis and heart failure, the mechanisms by which PPAR gamma ligands prevent the development of cardiovascular diseases are not fully understood.

Monocytes/macrophages (Mphi) play a pivotal role in the persistence of chronic inflammation and local tissue destruction in diseases such as rheumatoid arthritis and atherosclerosis. The production by Mphi of cytokines, chemokines, metalloproteinases and their inhibitors is an essential component in this process, which is tightly regulated by multiple factors. The peroxisome proliferator-activated receptors (PPARs) were shown to be involved in modulating inflammation.

PPAR gamma is activated by a wide variety of ligands such as fatty acids, the anti-diabetic thiazolidinediones (TZDs), and also by certain prostaglandins of which 15-deoxy-Delta(12, 14)-PGJ2 (PGJ2). High concentrations of PPAR gamma ligands were shown to have anti-inflammatory activities by inhibiting the secretion of interleukin-1 (IL- 1), interleukin-6 (IL-6) and tumour necrosis factor alpha (TNFalpha) by stimulated monocytes.

The aim of this study was to determine whether PGJ2 and TZDs would also exert an immunomodulatory action through the up-regulation of anti-inflammatory cytokines such as the IL-1 receptor antagonist (IL-1Ra). THP-1 monocytic cells were stimulated with PMA, thereby enhancing the secretion of IL-1, IL-6, TNFalpha, IL-1Ra and metalloproteinases. Addition of PGJ2 had an inhibitory effect on IL-1, IL-6 and TNFalpha secretion, while increasing IL-1Ra production. In contrast, the bona fide PPAR gamma ligands (TZDs; rosiglitazone, pioglitazone and troglitazone) barely inhibited pro-inflammatory cytokines, but strongly enhanced the production of IL-1Ra from PMA-stimulated THP-1 cells. Un-stimulated cells did not respond to TZDs in terms of IL-1Ra production, suggesting that in order to be effective, PPAR ligands depend on PMA signaling. Basal levels of PPAR gamma are barely detectable in un-stimulated THP-1 cells, while stimulation with PMA up-regulates its expression, suggesting that higher levels of PPAR gamma expression are necessary for receptor ligand effects to occur. In conclusion, it was demonstrated for the first time that TZDs may exert an anti-inflammatory activity by inducing the production of the IL-1Ra.

Peroxisome proliferator-activated receptors (PPARs) are ligand-activated transcription factors that directly control numerous genes of lipid metabolism by binding to response elements in the promoter. It has recently been proposed that PPARgamma may also regulate genes for pro-inflammatory proteins, not through PPRE binding but by interaction with transcription factors AP-1, STAT, and NF-kappaB.

REFERENCES

Chong Z Z, Maiese K. The Src homology 2 domain tyrosine phosphatases SHP-1 and SHP-2: diversified control of cell growth, inflammation, and injury. Histol Histopathol. 2007; 22(11): 1251-1267.
Maiese K. Diabetic stress: new triumphs and challenges to maintain vascular longevity. Expert Rev Cardiovasc Ther. 2008; 6(3): 281-284.
Slomka M, Zieminska E, Lazarewicz J. Nicotinamide and 1-methylnicotinamide reduce homocysteine neurotoxicity in primary cultures of rat cerebellar granule cells. Acta Neurobiol Exp. 2008; 68(1): 1-9.
Nakamura T, Sakamoto K. Forkhead transcription factor FOXO subfamily is essential for reactive oxygen species-induced apoptosis. Mol Cell Endocrinol. 2007; 281(1-2): 47-55.
Barthelemy C, Henderson C E, Pettmann B. Foxo3a induces motoneuron death through the Fas pathway in cooperation with JNK. BMC Neurosci. 2004; 5(1): 48.
You H, Yamamoto K, Mak T W. Regulation of transactivation-independent proapoptotic activity of p53 by FOXO3a. Proc Natl Acad Sci USA. 2006; 103(24): 9051-9056.
Won C K, Ji H H, Koh P O. Estradiol prevents the focal cerebral ischemic injury-induced decrease of forkhead transcription factors phosphorylation. Neurosci Lett. 2006; 398(1-2): 39-43.
Caporali A, Sala-Newby G B, Meloni M. et al. Identification of the prosurvival activity of nerve growth factor on cardiac myocytes. Cell Death Differ. 2008; 15(2): 299-311.
Tothova Z, Kollipara R, Huntly B J. et al. FoxOs are critical mediators of hematopoietic stem cell resistance to physiologic oxidative stress. Cell. 2007; 128(2): 325-339.
Ferrara N, Rinaldi B, Corbi G. et al. Exercise training promotes SIRT1 activity in aged rats. Rejuvenation Res. 2008; 11(1): 139-150.
Maiese K, Chong Z Z, Shang YC. Mechanistic insights into diabetes mellitus and oxidative stress. Curr Med Chem. 2007; 14(16): 1729-1738.
Maiese K, Morhan S D, Chong Z Z. Oxidative stress biology and cell injury during type 1 and type 2 diabetes mellitus. Curr Neurovasc Res. 2007; 4(1): 63-71.
Maiese K, Chong Z, Li F. Maiese K. Reducing oxidative stress and enhancing neurovascular longevity during diabetes mellitus. New York: Oxford University Press; Neurovascular Medicine: Pursuing Cellular Longevity for Healthy Aging. 2009
Donahoe S M, Stewart G C, McCabe C H. et al. Diabetes and mortality following acute coronary syndromes. JAMA. 2007; 298(7): 765-775.
Lin K, Dorman J B, Rodan A. et al. Daf-16: An HNF-3/forkhead family member that can function to double the life-span of Caenorhabditis elegans. Science. 1997; 278(5341): 1319-1322.
Ogg S, Paradis S, Gottlieb S. et al. The Fork head transcription factor DAF-16 transduces insulin-like metabolic and longevity signals in C. elegans. Nature. 1997; 389(6654): 994-999.
Guo S, Rena G, Cichy S. et al. Phosphorylation of serine 256 by protein kinase B disrupts transactivation by FKHR and mediates effects of insulin on insulin-like growth factor-binding protein-1 promoter activity through a conserved insulin response sequence. J Biol Chem. 1999; 274(24): 17184-17192.
Nakae J, Park B C, Accili D. Insulin stimulates phosphorylation of the forkhead transcription factor FKHR on serine 253 through a Wortmannin-sensitive pathway. J Biol Chem. 1999; 274(23): 15982-15985.
Kim JR, Jung H S, Bae S W. et al. Polymorphisms in FOXO gene family and association analysis with BMI. Obesity (Silver Spring). 2006; 14(2): 188-193.
Marchetti V, Menghini R, Rizza S. et al. Benfotiamine counteracts glucose toxicity effects on endothelial progenitor cell differentiation via Akt/FoxO signaling. Diabetes. 2006; 55(8): 2231-2237.
Fallarino F, Bianchi R, Orabona C. et al. CTLA-4-Ig activates forkhead transcription factors and protects dendritic cells from oxidative stress in nonobese diabetic mice. J Exp Med. 2004; 200(8): 1051-1062.
Nakae J, Cao Y, Oki M. et al. Forkhead transcription factor FoxOl in adipose tissue regulates energy storage and expenditure. Diabetes. 2008; 57(3): 563-576.
Puig O, Tjian R. Transcriptional feedback control of insulin receptor by dFOXO/FOXO1. Genes Dev. 2005; 19(20): 2435-2446.
Kamagate A, Dong H H. Foxol integrates insulin signaling to VLDL production. Cell Cycle. 2008; 7(20): 3162-3170.
Ni YG, Wang N, Cao D J. et al. FoxO transcription factors activate Akt and attenuate insulin signaling in heart by inhibiting protein phosphatases. Proc Natl Acad Sci USA. 2007; 104(51): 20517-20522.
Kamei Y, Miura S, Suzuki M. et al. Skeletal muscle FOXO1 (FKHR) transgenic mice have less skeletal muscle mass, down-regulated type I (slow twitch/red muscle) fiber genes, and impaired glycemic control. J Biol Chem. 2004; 279(39): 41114-41123.
Liu C M, Yang Z, Liu C W. et al. Effect of RNA oligonucleotide targeting Foxo-1 on muscle growth in normal and cancer cachexia mice. Cancer Gene Ther. 2007; 14(12): 945-952.
Sandri M, Lin J, Handschin C. et al. PGC-lalpha protects skeletal muscle from atrophy by suppressing FoxO3 action and atrophy-specific gene transcription. Proc Natl Acad Sci USA. 2006; 103(44): 16260-16265.
Balan V, Miller G S, Kaplun L. et al. Life span extension and neuronal cell protection by Drosophila nicotinamidase. J Biol Chem. 2008; 283(41): 27810-27819.
Chong Z Z, Maiese K. Enhanced tolerance against early and late apoptotic oxidative stress in mammalian neurons through nicotinamidase and sirtuin mediated pathways. Curr Neurovasc Res. 2008; 5(3): 159-170.
Myatt S S, Lam E W. The emerging roles of forkhead box (Fox) proteins in cancer. Nat Rev Cancer. 2007; 7(11): 847-859.
Nemoto S, Fergusson M M, Finkel T. Nutrient availability regulates SIRT1 through a forkhead-dependent pathway. Science. 2004; 306(5704): 2105-2108.
Motta M C, Divecha N, Lemieux M. et al. Mammalian SIRT1 represses forkhead transcription factors. Cell. 2004; 116(4): 551-563.
Mitochondrial dysfunction contributes to impaired insulin secretion in INS-1 cells with dominant-negative mutations of HNF-lalpha and in HNF-lalpha-deficient islets. Pongratz R L, Kibbey R G, Kirkpatrick C L, Zhao X, Pontoglio M, Yaniv M, Wollheim C B, Shulman G I, Cline G W. J Biol Chem. 2009 Jun 19;284(25):16808-21. Epub 2009 Apr. 17.
Ceramide starves cells to death by downregulating nutrient transporter proteins. Guenther G G, Peralta E R, Rosales K R, Wong S Y, Siskind L J, Edinger A L. Proc Natl Acad Sci U S A. 2008 Nov 11;105(45):17402-7. Epub 2008 Nov. 3.
Methyl pyruvate stimulates pancreatic beta-cells by a direct effect on KATP channels, and not as a mitochondrial substrate. Düffer M, Krippeit-Drews P, Buntinas L, Siemen D, Drews G. Biochem J. 2002 Dec. 15; 368(Pt 3)817-25.
Methyl pyruvate initiates membrane depolarization and insulin release by metabolic factors other than ATP. Lembert N, Joos HC, Idahl L A, Ammon H P, Wahl M A. Biochem J. 2001 Mar 1;354(Pt 2):345-50.
Mitochondrial activation directly triggers the exocytosis of insulin in permeabilized pancreatic beta-cells. Maechler P, Kennedy ED, Pozzan T, Wollheim C B. EMBO J. 1997 Jul. 1; 16(13):3833-41.
Novel insights for systemic inflammation in sepsis and hemorrhage.

Cai B, Deitch E A, Ulloa L. Mediators Inflamm. 2010;2010:642462. Epub 2010 Jun. 8. PMID: 20628562 PubMed RELATED CITATIONS

Metabolic imaging in the anesthetized rat brain using hyperpolarized [1-13C] pyruvate and [1-13C] ethyl pyruvate.

Hurd RE, Yen YF, Mayer D, Chen A, Wilson D, Kohler S, Bok R, Vigneron D, Kurhanewicz J, Tropp J, Spielman D, Pfefferbaum A. Magn Reson Med. 2010 May; 63(5):1137-43.

EthylEthylpyruvate reduces mortality in anendotoxin-induced severe acute lung injury mouse model.

Shang G H, Lin D , Xiao W, Jia C Q, Li Y, Wang A H, Dong L. Respir Res. 2009 Oct. 2; 10:91.

Meth 1-2-acetamidoacrylate an ethtl pyruvate analog, decreases sepsis-induced acute kidney injury in mice.

Leelahavanichkul A, Yasuda H, Doi K, Hu X, Zhou H, Yuen P S, Star R A. Am J Physiol Renal Physiol. 2008 Dec; 295(6):F1825-35. Epub 2008 Oct. 15.

Delayed ethyl pyruvate therapy attenuates experimental severe acute pancreatitis via reduced serum high mobility group box 1 levels in rats.

Yang Z Y, Ling Y, Yin T, Tao J, Xiong J X, Wu H S, Wang C Y. World J Gastroenterol. 2008 Jul. 28; 14(28):4546-50.

Ethyl pyruvate modulates acute inflammatory reactions in human endothelial cells in relation to the NF-kappaB pathway.

Johansson A S, Johansson-Haque K, Okret S, Palmblad J. Br J Pharmacol. 2008 Jul; 154(6):1318-26. Epub 2008 May 26.

Bench-to-bedside review: Amelioration of acute renal impairment using ethyl pyruvate. Reade M C, Fink M P. Crit Care. 2005;9(6):556-60. Epub 2005 Oct. 21. Review.
Berger I, Bieniossek C, Schaffitzel C, Hassler M, Santelli E, Richmond T J. Direct interaction of Ca2+/calmodulin inhibits histone deacetylase 5 repressor core binding to myocyte enhancer factor 2. J Biol Chem. 2003 Mar. 6 [epub ahead of print]
Czubryt M P, McAnally J, Fishman G I, Olson E N. Regulation of peroxisome proliferator-activated receptor gamma coactivator 1 alpha (PGC-1 alpha) and mitochondrial function by MEF2 and HDAC5. Proc Natl Acad Sci U S A. 2003 Feb. 18; 100(4):1711-6.
Ichida M, Nemoto S, Finkel T. Identification of a specific molecular repressor of the peroxisome proliferator-activated receptor gamma Coactivator-1 alpha (PGC-lalpha). J Biol Chem. 2002 Dec 27; 277(52):50991-5.
Kressler D, Schreiber S N, Knutti D, Kralli A. The PGC-1-related protein PERC is a selective coactivator of estrogen receptor alpha. J Biol Chem. 2002 Apr. 19; 277(16):13918-25.
Meredith L. Moore, Edwards A. Park and Jeanie B. McMillin, Upstream stimulatory factor represses the induction of carnitine palmitoyltransferase-Ibeta expression by PGC-1. J Biol Chem. 2003 Feb. 28 [epub ahead of print]
Schreiber S N, Knutti D, Brogli K, Uhlmann T, Kralli A. The Transcriptional Coactivator PGC-1 Regulates the Expression and Activity of the Orphan Nuclear Receptor Estrogen-Related Receptor alpha (ERRalpha). J Biol Chem. 2003 Mar. 14; 278(11):9013-8. Breaker, R. “Complex Riboswitches” Science, Vol. 319, 1795-1797, 28 Mar. 2008.
“Tricarboxylic acid cycle substrates prevent PARP-mediated death of neurons and astrocytes”, J Virol. 2004 Sep.; 78(18):9936-46., Ohsaki E, Ueda K, Sakakibara S, Do E, Yada K, Yamanishi K, Department of Microbiology, Osaka Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka 565-0871, Japan.
“NAD+repletion prevents PARP-1-induced glycolytic blockade and cell death in cultured mouse astrocytes”, Biochem Biophys Res Commun. 2003 Sep. 5; 308(4):809-13. Ying W, Gamier P, Swanson R A. Department of Neurology, University of California at San Francisco and Veterans Affairs Medical Center, 4150 Clement Street, San Francisco, Calif. 94121, USA.

Rudinger, Peptide Hormones (Jun. 1976: J. Parsons Ed.) p. 1-6.* Goodman & Gilman's, “The Pharmcological Basis of Therapeutics”, 6th Ed. (Macmillan Pub.:1980) pp. 5-10.* Chemical Abstracts AN 1 982:49622, Bertolini et al., January 1981.*

Chemical Abstracts AN 89:82648, Rees et al, U.S. Pat. No. 4,871,774, Mar. 10, 1989.*
Annesley, T. and Walker, J., “Formation and Utilization of Novel High Energy Phosphate Reservoirs in Ehrlich Ascites Tumor Cells,” J. Biol. Chem., vol. 253, No. 22, 8120-5 (1978).
Beal, M., “Aging, Energy, and Oxidative Stress in Neurodegenerative Diseases,” Ann. Neurol., vol. 38, 357-66 (1995).
Beal, M., “Does Impairment of Energy Metabolism Result in Excitotoxic Neuronal Death in Neurodegenerative Illnesses?” Ann. Neurol., vol. 31, 119-30 (1992).

Beal, M. et al., “Age-Dependent Striatal Excitotoxic Lesions Produced by the Endogenous Mitochondrial Inhibitor Malonate,” J. Neurochem., vol. 61, 1147-50 (1993). Brouillet, E. et al., “Chronic Mitochondrial Energy Impairment Produces Selective Striatal Degeneration and Abnormal Choreiform Movements in Primates,” PNAS, vol. 92, 7105-9 (1995).

Browne, S. et al., “Oxidative Damage and Metabolic Dysfunction in Huntington's Disease: Selective Vulnerability of the Basal Ganglia,” Ann. Neurol., vol. 41, 646-53 (1997).
Burbaeva, G Sh et al., “Decreased Level of Immunoreactive Phosphokinase BB Isoenzymes in the Brain of Patients with Schizophrenia and Senile Dementia of the Alzheimer Type,” Zh. Nevropatol. Psikhiatr Im S S Korsakova, vol. 90, No. 7, 85-7 (1990)—abstract attached.
De Leon, M. et al., “Identification of Transcriptionally Regulated Genes After Sciatic Nerve Injury,” J. Neurosci. Res., vol. 29, 437-48 (1991).
Erecinska, M. and Silver, I., “ATP and Brain Function,” J. Cerebr. Blood Flow and Metabolism, vol. 9, 2-19 (1989).
Gu, M. et al:, “Mitochondrial Defect in Huntington's Disease Caudate Nucleus,” Ann. Neurol., vol. 39, 385-9 (1996).

Gurney, M. et al., “Motor Neuron Degeneration in Mice That Express a Human Cu,Zn Superoxide Dismutase Mutation,” Science, vol. 264, 1772-5 (1994). Henshaw, R. et al., “Malonate Produces Striatal Lesions by Indirect NMDA Receptor Activation,” Brain Research, vol. 647, 161-6 (1994).

Hertz, L. and Peng, L., “Energy Metabolism at the Cellular Level of the CNS,” Can. J. Physiol. Pharmacol., vol. 70, S145-57 (1992).
Ito, M., “The Cellular Basis of Cerebellar Plasticity,” Corr. Opin. Neurobiol., vol. 1, 616-20 (1991).

Jenkins, B. et al., “Evidence for Impairment of Energy Metabolism in Vivo in Huntington's Disease Using Localized .sup.1 H NMR Spectroscopy,” Neurology, vol. 43, 2689-95 (1993).

Maker, H. et al., “Regional Changes in Cerebellar Creatine Phosphate Metabolism During Late Maturation,” Exp. Neurol., vol. 38, 295-300 (1973).

Manos, P. et al., “Creatine Kinase Activity in Postnatal Rat Brain Development and in Cultured Neurons, Astrocytes, and Oligodendrocytes,” J. Neurochem., vol. 56, 2101-7 (1991). Molloy, G. et al., “Rat Brain Creatine Kinase Messenger RNA Levels are High in Primary Cultures of Brain Astrocytes and Oligodencrocytes and Low in Neurons,” J. Neurochem., vol. 59,1925-32 (1992). Newman, E., “Regulation of Potassium Levels by Glial Cells in the Retina,” Trends Neurosciencl, vol. 8, 156-9 (1985). Oblinger, M. et al., “Cytotypic Differences in the Protein Composition of the Axonally Transported Cytoskeleton in Mammalian Neurons,” J. Neurol., vol. 7, No. 2, 453-62 (1987).

Orlovskaia, D. D. et al., “Neuromorphology and Neurochemistry of Senile Dementias in the Light of Studies on Glial Response,” Vestn Ross Akad Med Nauk., vol. 8, 34-9 (1992)—abstract only.
Reichenbach, A., “Glial K+Permeability and CNS K+Clearance by Diffusion and Spatial Buffering,” Acad. Sci. New York, 272-86 (1991). Chemical Society, vol. 93, 5542-51(1971).
Schiffmann, R. et al., “Childhood Ataxia with Diffuse Central Nervous System Hypomyelination,” Ann. Neurol., vol. 35, 331-40 (1994).
Schultz, J. et al., “Blockade of Neuronal Nitric Oxide Synthase Protects Against Excitotoxicity in vivo,” J. Neurosci., vol. 15, No. 12, 8419-29 (1995).

Schultz, J. et al., “Inhibition of Neuronal Nitric Oxide Synthase by 7-Nitroindazole Protects Against MPTP-lnduced Neurotoxicity in Mice,” J. Neurochem., vol. 64, 936-9 (1995). Stadhouders, A., et al., “Mitochondrial Creatine Kinase: A Major Constituent of Pathological Inclusions Seen in Mitochondrial Myopathies,” PNAS, vol. 91, No. 11, 5089-93 (1994). Wang, T., “Synthesis and Properties of N-Acetimidoyl Derivatives of Glycine and Sarcosine,” JOC, vol. 39, No. 24, 3591-4 (1974).

Beal, M. F., et al, “Do defects in mitochondrial energy metabolism underlie the pathology of neurodegenerative diseases?” Trends Neurosci. 16: 125-131(1993).
Folbergrova, J., et al., “Focal and perifocal changes in tissue energy state during middle cerebral artery occlusion in normo- and hyperglycemic rats,” J. Cereb. Blood Flow Metab. 12: 25-33 (1992).
Ginsberg, M. D. and Busto, R., “Rodent models of cerebrasl ischemia (Progress Review),” Stroke 20:1627-1640 (1989).
Mosinger, J. L. and Olney, J. W., “Photothrombosis-induced ischemic neuronal degeneration in the rat retina,” Exp. Neurol. 105: 110-11 3, (1989).
J. W., “Excitatory amino acids and neuropsychiatric disorders,” Biol. Psychiatry 26:505-525 (1989).
Olmey. J. W., “NMDA antagonist neurotoxicity: Mechanism and prevention,” Science 254: 1515-1518 (1991).

Siesjo, B. K., Brain Energy Metabolism (John Wiley & Sons, New York, 1978)

Simon, R. P., et al., “Blockade of N-methyl-D-aspartate receptors may protect against ischemic damage in the brain,” Science 226: 850-852 (1984).
Voll, C. L. and Auer, R. N. “Insulin attenuates ischemic brain damage independent of its hypoglycemic effect,” J. Cereb. Blood Flow and Metabolism 11: 1006-1014 (1991).
Choi, D. W. At the scene of ischemic brain injury: is PARP a perp? Nat. Med. 3: 1073-1074, 1997.
Jeggo, P. A. DNA repair: PARP-another guardian angel? Curr. Biol., 8: R49-R51, 1998.
Pieper, A. A., Verma, A., Zhang, J., and Snyder, S. H. Poly(ADP-ribose) polymerase, nitric oxide and cell death. Trends Pharmacol. Sci., 20: 1 71-181, 1999.
D'Amours, D., Desnoyers, S., D'Silva, I., and Poirier, G. G. Poly(ADP-ribosyl)ation reactions in the regulation of nuclear functions. Biochem. J., 342: 249-268, 1999.
Mendoza-Alvarez, H., and Alvarez-Gonzalez, R. Poly(ADP- ribose) polymerase is a catalytic dimer and the automodification reaction is intermolecular. J. Biol. Chem., 268: 22575-22580, 1993.
Burkart, V., Wang, Z. Q., Radons, J., Heller, B., Herceg, Z., Stingl, L., Wagner, E. F., and Kolb, H. Mice lacking the poly(ADP-ribose) polymerase gene are resistant to pancreatic b-cell destruction and diabetes development induced by streptozocin. Nat. Med., 5: 314-319, 1999.
Masutani, M., Nozaki, T., Nakamoto, K., Nakagama, H., Suzuki, H., Kusuoka, O., Tsutsumi, M., and Sugimura, T. The response of Parp knockout mice against DNA damaging agents. Mutat. Res., 462: 159-166, 2000.
Masutani, M., Suzuki, H., Kamada, N., Watanabe, M., Ueda, O., Nozaki, T., Jishage, K., Watanabe, T., Sugimoto, T., Nakagama, H., Ochiya, T and Sugimura, T. Poly(ADP-ribose) polymerase gene disruption conferred mice resistant to streptozo-tocin-induced diabetes. Proc. Natl. Acad. Sci. USA, 96: 2301-2304, 1999.
Lindahl, T., Satoh, M. S., Poirier, G. G., and Klungland, A. Post-translational modification of poly(ADP-ribose) polymerase induced by DNA strand breaks. Trends Biochem. Sci., 20: 405-411, 1995.
Decker, P., Miranda, E. A., de Murcia, G., and Muller, S. An improved nonisotopic test to screen a large series of new inhibitor molecules of poly(ADP-ribose) polymerase activity for therapeutic applications. Clin. Cancer Res., 5: 1169-1172, 1999. [
Holl, V., Coelho, D., Weltin, D., Hyun, J. W., Dufour, P., and Bischoff, P. Modulation of the antiproliferative activity of anticancer drugs in hematopoietic tumor cell lines by the poly(ADP-ribose) polymerase inhibitor 6(5H)-phenanthridinone. Anticancer Res., 20: 3233-3241, 2000.
Szabo, C., and Dawson, V. L. Role of poly(ADP-ribose) synthetase in inflammation and ischaemia-reperfusion. Trends Pharmacol. Sci., 19: 287-298, 1998.
Takahashi, K., Pieper, A. A., Croul, S. E., Zhang, J., Snyder, S. H., and Greenberg, J. H. Post-treatment with an inhibitor of poly(ADP-ribose) polymerase attenuates cerebral damage in focal ischemia. Brain Res., 829: 46-54, 1999.
Shall, S., and de Murcia, G. Poly(ADP-ribose) polymerase-1: what have we learned from the deficient mouse model? Mutat. Res., 460: 1-15, 2000.
Jacobson, M. K., and Jacobson, E. L. Discovering new ADP-ribose polymer cycles: protecting the genome and more. Trends Biochem. Sci., 24: 415-417, 1999.
Ame, J. C., Rolli, V., Schreiber, V., Niedergang, C., Apiou, F., Decker, P., Muller, S., Hoger, T., Menissier-de Murcia, J., and de Murcia, G. PARP-2, a novel mammalian DNA damage-dependent poly(ADP-ribose) polymerase. J. Biol. Chem., 274: 17860-17868, 1999.
Johansson, M. A human poly(ADP- ribose) polymerase gene family (ADPRTL): cDNA cloning of two novel poly(ADP-ribose) polymerase homologues. Genomics, 57: 442-445, 1999.
Kickhoefer, V. A., Siva, A. C., Kedersha, N. L., Inman, E. M., Ruland, C., Streuli, M., and Rome, L. H. The 193-kD vault protein, VPARP, is a novel poly(ADP-ribose) polymerase. J. Cell Biol., 146: 91 7-928, 1999.
Sallmann, F. R., Vodenicharov, M. D., Wang, Z. Q., and Poirier, G. G. Characterization of sPARP-1. An alternative product of PARP-1 gene with poly(ADP-ribose) polymerase activity independent of DNA strand breaks. J. Biol. Chem., 275: 15504-15511, 2000.
Smith, S., Giriat, I., Schmitt, A., and de Lange, T. Tankyrase, a poly(ADP-ribose) polymerase at human telomeres. Science (Wash. DC), 282: 1484- 1487, 1998
d'Adda di Fagagna, F., Hande, M. P., Tong, W. M., Lansdorp, P. M., Wang, Z. Q., and Jackson, S. P. Functions of poly(ADP-ribose) polymerase in controlling telomere length and chromosomal stability. Nat. Genet., 23: 76-80, 1999.
Kong, L. B., Siva, A. C., Rome, L. H., and Stewart, P. L. Structure of the vault, a ubiquitous cellular component. Structure Fold Des., 7: 371-379, 1999.
Pennisi, E. A possible new partner for telomerase. Science (Wash. DC), 282: 1395-1396, 1999.
Smith, S., and de Lange, T. Tankyrase promotes telomere elongation in human cells. Curr. Biol., 10: 1299-1302, 2000.
Kickhoefer, V. A., Rajavel, K. S., Scheffer, G. L., Dalton, W. S., Scheper, R. J., and Rome, L. H. Vaults are up-regulated in multidrug-resistant cancer cell lines. J. Biol. Chem., 273: 8971-8974, 1998.

Schroeijers, A. B., Siva, A. C., Scheffer, G. L., de Jong, M. C., Bolick, S. C., Dukers, D.

F., Slootstra, J. W., Meloen, R. H., Wiemer, E., Kickhoefer, V. A., Rome, L. H., and Scheper, R. J. The M r 193,000 vault protein is up-regulated in multidrug-resistant cancer cell lines. Cancer Res., 60: 1104-1110, 2000.
Ame, J. C., Jacobson, E. L., and Jacobson, M. K. Molecular heterogeneity and regulation of poly(ADP-ribose) glycohydrolase. Mol. Cell Biochem., 1 93: 75-81, 1999.
Lin, W., Ame, J. C., Aboul-Ela, N., Jacobson, E. L., and Jacobson, M. K. Isolation and characterization of the cDNA encoding bovine poly(ADP-ribose) glycohydrolase. J. Biol. Chem., 272: 11895-11901, 1997.

Claims

1. A pharmaceutical or nutraceutical composition comprising a pyruvate compound in the form of ethyl pyruvate and methyl, di methyl and/or tri methyl pyruvic acid esters.

2. The composition of claim 1 wherein the ethyl to methyl is a proprietary blend.

3. This compound is a combination of Ethyl Pyruvic acid ester and either methyl, di methyl or tri-methyl Pyruvic acid ester.

5. This compound can be provided in multiple delivery methods to include but not limited to the following:

a soluble bi-layer gel capsule.
a micronized, freeze-fried powder.
This compound, along with added cofactors, can be individually provided in a powder packet and added to water or other beverages.
This compound can be provided as a frozen desert.
This compound can be provided as an oral gel mixture.
This compound can be incorporated into a gum base and used with hard-shelled immediate and time-released delivery mechanism.
This compound can be provided as a time-released transdermal patch.
This compound can be provided as IV solution for use in emergency medicine.
This compound can be formulated for use as an IM injection.
This compound can be formulated to be compatible with parenteral feeding.
This compound can be provided in a suppository format.
This compound is provided in dissolvable, impregnated oral strips.
This compound as a gel cap that may have Vitamin D3 added to it.
This compound as a gel cap that may have medium chain triglycerides added to it.
This compound as a gel cap may have Vitamin E, in the form of D-Alpha and Gamma tocopherol added to it.
This dissolvable strip can have Vitamin D3, medium chain triglycerides and Vitamin E-d-alpha tocopherol imbeded within it.
This compound can be provided as a nutraceutical sports drink

6. This compound can be used to lower human HbAl c.

7. This compound is a unique and selective PPARs agonist.

8. This compound is a molecular short-antisense compound, which provides quick and complete energy entry into the cistae of the inner mitochondria.

9. This compound reduces elevated glucose levels and can reduce post-translational phosphorylation of FOXO1, FOXO3a, and FOXO4 and allow for the nuclear translocation of these proteins.

10. This compound initiates an apoptotic program in endothelial progenitor cells.

11. This compound has a direct modulating effect on the cannabinoid receptor CB1.

12. This compound is a way of directly linking to, and thereby modulating, SIRT1-dependent regulation of chromatin and transcription.

13. This compound represents a way of Linking NAD metabolism and signaling to the control of cellular functions.

14. This compound represents a method of engendering and operationally initiating the innate human riboswitch.

15. This compound represents a method of selectively RNA editing as per claim #14.

16. This compound represents a method to silence or “turn-off” the systemic inflammatory response via RNA editing.

17. This compound represents a method for genomic self-regulation of Hba1c as per claims 14 through 16.

18. This compound, per claim 14, enables the ability for the cell to repair the Glut transporter system.

19. Per claim 14, enables the ability to repair damaged Glut4 system to reverse insulin resistance.

20. This compound up-regulates cellular energy and receptor sites in all cells enabling increased functional efficiency under physiologic or pathologic stress or challenge.

21. This invention enhances pharmacologic capability and function of any pharmaceutical ligand requiring energy to enable its function once inside the cell.

22. This compound per claim 17, via HbA1c regulation, diminishes type III diabetes of the CNS and its Amyloid beta plaque (Alzheimer's Disease).

23. The combination of ethyl pyruvate and methyl pyruvate as described in this patent work synergistically to inhibit pathways leading to Advanced Glycation Endproduct (AGE) thereby prolonging normal cell life and enhancing the inhibition of malignant cell formation.

Patent History
Publication number: 20130142872
Type: Application
Filed: Aug 9, 2012
Publication Date: Jun 6, 2013
Inventors: John James Fitzgerald, JR. (St. James, NY), Ranya Ludwig Alexander (San Diego, CA), Anthony Joseph Meduri (New York City, NY)
Application Number: 13/570,454