WO2011098757A1 - Cell based bioassay - Google Patents

Cell based bioassay Download PDF

Info

Publication number
WO2011098757A1
WO2011098757A1 PCT/GB2011/000170 GB2011000170W WO2011098757A1 WO 2011098757 A1 WO2011098757 A1 WO 2011098757A1 GB 2011000170 W GB2011000170 W GB 2011000170W WO 2011098757 A1 WO2011098757 A1 WO 2011098757A1
Authority
WO
WIPO (PCT)
Prior art keywords
cell
insulin
promoter
activity
subject
Prior art date
Application number
PCT/GB2011/000170
Other languages
French (fr)
Inventor
Calum Sutherland
Original Assignee
Tmri Limited
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Tmri Limited filed Critical Tmri Limited
Publication of WO2011098757A1 publication Critical patent/WO2011098757A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6897Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids involving reporter genes operably linked to promoters

Definitions

  • the invention relates to a cell-based bioassay and its use in the identification of insulin sensitizing agents and including a diagnostic assay for the determination of a subject's sensitivity to insulin and optionally to treat a subject either suffering from type 2 diabetes or wherein said subject has a predisposition to type 2 diabetes.
  • Insulin functions to regulate glucose homeostasis.
  • hyperglycemia abnormally high levels of serum glucose
  • the pancreatic ⁇ cells of the Islets of Langerhans synthesize proinsulin which is enzymatically cleaved at its amino and carboxy-termini to produce insulin, a 51 amino acid polypeptide.
  • Insulin is secreted and acts on target cells [e.g. liver, muscle, adipose tissue] that express insulin receptors.
  • target cells e.g. liver, muscle, adipose tissue
  • the activation of insulin receptors leads to a signal transduction cascade that results in expression of glucose transporters in muscle which remove excess glucose with subsequent conversion of glucose into glycogen for storage.
  • insulin shuts off endogenous glucose production and hepatic glucose output.
  • the insulin receptor is a tyrosine kinase and is a tetrameric transmembrane receptor comprising two a subunits and two ⁇ subunits.
  • the a subunits are extracellular and bind insulin.
  • the ⁇ subunits are transmembrane and include ATP and tyrosine kinase domains which become activated on insulin binding.
  • Diabetes mellitus can be of type 1 or type 2.
  • Type 1 diabetes is an autoimmune disease resulting in destruction of the pancreatic ⁇ cells which means the subject is unable to manufacture any insulin.
  • Type 2 diabetes is a more complicated condition and can result from a number of associated ailments but typically involves resistance to the metabolic actions of insulin. For example, type 2 diabetes is associated with age, obesity, a sedentary life style and insulin resistance.
  • An associated condition is called Metabolic Syndrome which may predispose subjects to type 2 diabetes. The symptoms associated with this syndrome are high blood pressure, dyslipidemia, increased body fat deposition and cardiovascular disease.
  • Insulin resistance is a common characteristic of type 2 diabetes mellitus and is one of the major independent risk factors for progression of the disease.
  • the treatment of insulin resistance is either by substantial weight loss and exercise with a concomitant increase in insulin sensitivity or by administration of insulin sensitizing agents which restore insulin sensitivity to those subjects that present with insulin resistance.
  • the current approved agents to control diabetes 2 that are proposed to have some insulin sensitizing activity are rosiglitazone or metformin which both have undesirable side effects.
  • the identification of novel insulin sensitizing agents that have better disease control and side effect profile is highly desirable.
  • Rat hepatoma cells are grown in the presence of serum isolated from obese rodents or obese human volunteers and the insulin sensitivity of the cells monitored over time by the repression of a key insulin responsive gene. Higher insulin concentrations are required to fully repress the gene in the cells grown in obese rodent serum compared with those grown in serum from lean rodents (almost a 10-fold shift in insulin sensitivity). This was reversed by restoring normal growth medium, while the insulin resistance could be prevented by rosiglitazone or metformin.
  • This disclosure relates to a novel insulin resistant cell model that can be used to study the molecular development of obesity-linked insulin resistance, screen for compounds which overcome obesity-related insulin resistance and sub-classify insulin resistant patients thereby influencing their treatment.
  • a screening method for the identification of an insulin sensitizing agent comprising the steps:
  • said agent[s] have an effect on the activity of the insulin responsive promoter.
  • said promoter is the mammalian cytosolic soluble phosphoenolpyruvate [GTP] gene promoter, or regulatory part thereof.
  • PEPCK gene sequences examples include UniGene Hs.1872 Homo sapiens (human) PCK1. UniGene Rn.104376 Rattus norvegicus (Norway rat) Pck1 or UniGene Mm.477474 Mus musculus (mouse) Pck1],
  • BMI Body Mass Index
  • a healthy BMI range in UK is 18-25, overweight is 25-30 and obese is >30.
  • said cell is a mammalian cell adapted for expression of the human PEPCK promoter.
  • said cell is a human cell.
  • said cell is a rodent cell, for example a rat or mouse cell.
  • said cell is a cell that is naturally responsive to insulin.
  • said cell is selected from the group consisting of: a hepatocyte, an adipocyte or a muscle cell.
  • said cell is a hepatocyte.
  • said hepatocyte cell is the rat hepatocyte cell-line RH4IIE.
  • said biological sample is a serum sample, preferably a serum sample isolated from an obese human subject.
  • said reporter molecule is a fluorescence reporter.
  • the analysis of promoter activity can be conveniently monitored by fusing an insulin responsive promoter, such as the PEPCK promoter, to a nucleic acid that encodes a "reporter" protein or polypeptide.
  • an insulin responsive promoter such as the PEPCK promoter
  • examples are well known in the art and include enzymes such as ⁇ glucuronidase, chloramphenicol acetyltransferase or luciferase.
  • Reporters that are proteinaceous fluorophores are also known in the art.
  • Green fluorescent protein, GFP is a spontaneously fluorescent protein isolated from coelenterates, such as the Pacific jellyfish, Aequoria victoria. Its role is to transduce, by energy transfer, the blue chemiluminescence of another protein, aequorin, into green fluorescent light.
  • GFP can function as a protein tag, as it tolerates N- and C-terminal fusions to a broad variety of proteins many of which have been shown to retain native function. Most often it is used in the form of enhanced GFP in which codon usage is adapted to the human code.
  • Other proteinaceous fluorophores include yellow, red and blue variant fluorescent proteins [e.g. BFP, CFP, GFP, YFP and RFP] These are commercially available from, for example Clontech (www. clontech. com) .
  • said fluorescence reporter is a fluorescent protein selected from the group comprising: BFP, CFP, GFP, YFP and RFP, and the mutants thereof.
  • said method detects one, two, three, four, five, six or seven biomarkers indicative of insulin activity.
  • said at least one biomarker is selected from the group consisting of: leptin and/or LDL cholesterol and/or triglycerides.
  • a cell array wherein said array comprises a plurality of identical assay preparations comprising a cell wherein said cell is transfected with a nucleic acid molecule comprising an insulin responsive promoter wherein said promoter is operably linked to a nucleic acid molecule that encodes a reporter molecule.
  • said insulin responsive promoter is the PEPCK promoter, preferably the human PEPCK promoter.
  • Assay devices include standard multiwell microtitre plates with formats such as 6, 12, 48, 96 and 384 wells which are typically used for compatibility with automated loading and robotic handling systems.
  • high throughput screens use homogeneous mixtures of agents with an indicator compound which is either converted or modified resulting in the production of a signal.
  • the signal is measured by suitable means (for example detection of fluorescence emission, optical density, or radioactivity) followed by integration of the signals from each well containing the cells, agent and indicator compound.
  • a method to analyse a subject to determine whether said subject is or is not insulin resistant comprising the steps:
  • ii) forming a preparation comprising the cell in i) and an isolated biological sample obtained from an obese subject, or a subject suspected of being susceptible to obesity, and incubating said preparation under cell culture conditions conducive to the growth and maintenance of said cell in culture;
  • iii) analyzing the biological effect of said isolated biological sample on at least one biomarker of insulin activity and comparing said biomarker to a control cell population which is obtained from a non-obese subject; and iv) determining whether the subject is or is not insulin resistant.
  • said isolated biological sample is analyzed for an effect on the activity of the insulin responsive promoter.
  • a method to analyse and treat a human subject suspected of suffering from or having a predisposition to type 2 diabetes comprising the steps:
  • said insulin responsive promoter is the PEPCK promoter, preferably the human PEPCK promoter.
  • said biological sample is a body fluid sample, for example a blood or serum sample.
  • Figure 1 illustrates that the growth of cells in serum from obese rodents alters the insulin sensitivity of the PEPCK gene promoter.
  • HL1 C cells H4IIE cells with CAT reporter
  • HFS high fat diet for 3 months
  • Con lean chow fed littermates
  • Protein was harvested and CAT reporter assayed as a measure of PEPCK gene promoter activity as detailed under Methods.
  • FIG. 2 illustrates that the growth of cells in serum from obese rodents alters insulin regulation of PEPCK mRNA.
  • HL1C cells were grown for 3 weeks in serum isolated from obese Fa/Fa or lean Fa/+ rats prior to serum starvation and exposure to glucocorticoid (Dex), cAMP and insulin as in Fig.1.
  • Dex glucocorticoid
  • cAMP glucocorticoid
  • Figure 3 illustrates that normal insulin sensitivity of cells is restored by returning the cells to standard serum.
  • H4IIE cells were grown for 3 weeks in 5% Fa/Fa serum. RNA was harvested from half of the cells following exposure to hormones as in Fig.1 , the remainder were placed in 5% FCS for 1 week prior to hormone exposure and RNA isolation, while RNA was also isolated from H4IIE cells grown for 3 weeks in Fa/+ lean serum followed by 1 week in FCS prior to hormone exposure.
  • (B) Relative PEPCK mRNA (average ⁇ SEM) for the samples isolated above are shown following normalisation for cyclophilin mRNA in each sample. Percent inhibition by insulin is indicated in each case. * p ⁇ 0.05;
  • Figure 4 illustrates the generation of insulin resistance is prevented by pioglitazone and metformin.
  • H4IIE cells were grown for 3 weeks in 5% Fa/Fa serum, in the presence or absence of pioglitazone or metformin as indicated.
  • Cells were serum starved for 16h prior to 3h exposure to dexamethasone (500nM), 8CPT-CAMP (0.1 mM) ⁇ insulin as indicated, and RNA was isolated.
  • PEPCK mRNA levels (average ⁇ SEM) are presented for each cell treatment after correction for cyclophilin mRNA. Percent inhibition by 1 n insulin in each condition is presented.
  • FIG. 5 illustrates that the growth of cells in serum from obese humans with diabetes alters insulin regulation of PEPCK mRNA.
  • H4IIE cells were grown for 3 weeks in 5% serum collected from 40 human volunteers, 20 who were obese and diabetic (Cases), 20 who were lean and non-diabetic (Controls). After 3 weeks in the serum the 40 different pools of cells were serum starved for 16h prior to 3h exposure to dexamethasone (500n ), 8CPT-cAMP (0.1 mM) ⁇ insulin as indicated, and RNA was isolated.
  • Figure 6 illustrates that there is variation in the degree of insulin resistance generated by growth of cells in serum from obese volunteers with diabetes.
  • the response to 0.1 nM insulin for each individual pool of cells grown in the 20 serums collected from the obese diabetic volunteers is shown relative to the average Control response.
  • the 10 serums that generated the greatest resistant to insulin are labelled insulin resistant and a comparison of clinical parameters of the insulin resistant group is presented in Table 2
  • Figure 7 is the nucleotide sequence of the rat PEPCK promoter.
  • Actrapid human insulin was from Novo Nordisk A/S (Bagsvaerd, Denmark). 2x Universal PCR Master Mix, No AmpErase UNG from Applied Biosystems, CompleteTM protease inhibitor cocktail tablets from Roche, and 8-(4-chloro-phenylthio)-cAMP from Calbiochem. All primers and probes were synthesized and purified by Sigma-Aldrich. All other chemicals were of the highest grade obtainable.
  • Diets were purchased from research diets (HF diet D12331 and control diet D12328, http://www.researchdiets.com/pdf/Data-20Sheets/D12331.pdf) and Male Sprague Dawleys (starting at 10 weeks of age) were kept on the diet for up to 3 months prior to sacrifice. Serum was prepared immediately and all serum from each group combined, then aliquoted and snap frozen for storage at -80oC.
  • H4IIE cells a rat hepatoma cell, and HL1 C (a H4IIE line with a stable insertion of a PEPCK promoter, chloramphenicol acetyl transferase gene construct (), were maintained in DMEM (Dulbecco's modified Eagle's medium) containing 1000 mg/l glucose, 1 % (v/v) penicillin/streptomycin, and 5 % (v/v) FBS, in a 37 °C 5 % C0 2 incubator.
  • DMEM Dulbecco's modified Eagle's medium
  • cells were cultured and passaged as normal in the experimental sera. Cells were serum starved for 16 hours prior to measurement of insulin sensitivity. Hormones were added as described in figure legends, and after 3h protein (for CAT Assay, HL1C) or RNA (for Taqman, H4IIE) were harvested.
  • Cells were isolated by trypsinisation, washed in PBS and pelleted by centrifugation at 1400 rpm for 5 min. Cell lysate was prepared by sonication in 0.25M TRIS, total protein assessed by BCA assay (Pierce), and general cellular protein removed by heating at 65oC for 10 min and centrifugation. Supernatants were tested for CAT activity as described previously (23), and data expressed as Units of CAT activity per mg of cellular protein.
  • Primer sequences were to rat PEPCK (forward- ACA GGC AAG GTC ATC ATG CA; reverse- TGC CGA AGT TGT AGC CAA AGA; probe- FAM-ACC CCT TCG CTA TGC GGC CC-TAMRA) and rat cyclophilin (forward- TTA CTA GGT CTG GCA GGA AGA TTA AAG; reverse- CTG CAT CTC TTG TCT CCA ATG TG; probe- FAM- AGA GGA CCA AGG CGT TAT CGA A-TAMRA).
  • PCR reactions were carried out using the following cycling conditions - 50°C - 2min x1 , 95°C - 10min x1 followed by 40 cycles of 95°C - 15s, 60°C - 1 min.
  • BMI ⁇ 25, Non-Diabetic subjects were recruited from the general population along with twenty obese (BMI>30) type 2 diabetics recruited from the diabetes clinic. All subjects were male, non-smokers and taking no other medication known to influence insulin sensitivity (Table 1). They were relatively pharmaceutically naive subjects to reduce potential confounding effects of long term intervention, the exception being that all Cases were on a statin at time of visit. Volunteers fasted overnight prior to a visit to the Clinical Research Centre where 200ml of fasted whole blood was collected. Serum was prepared and stored at -80°C in 10ml_ aliquots. BMI, blood pressure and waist measurements were taken during the visit.
  • TNFa TNFa
  • insulin insulin
  • adiponectin CRP
  • leptin leptin
  • HL1C cells were grown in DMEM containing either 2.5% FCS + 2.5% DIO-derived serum, or 2.5% FCS + 2.5% Con serum. The cell morphology and growth rate during culture and passage (every 2 or 3 days) was similar in both cases (data not shown). After 9 or 24 days cells were serum starved for 3h, then exposed to hormones prior to measurement of PEPCK transcription.
  • HL1C cells contain a CAT reporter with expression under the control of the PEPCK gene promoter (23; 24).
  • CAT expression is normally low but can be induced 5- 10 fold by exposure to glucocorticoid and cAMP for 3h.
  • the induction can be completely blocked by including insulin in the culture medium (23). Repression by insulin is dose-dependent with an EC50 of approximately 0.1 nM, and >80% inhibition at 1 nM insulin (Fig.lA and (23; 25-27)).
  • HL1C cells were grown in either 2.5% FaFa or 2.5% Fa/+ serum (supplemented with 2.5% FCS in each case), for 3 weeks prior to analysis of PEPCK gene promoter regulation (Fig 1 D). Once more insulin sensitivity of the HL1C cells was reduced in cells grown in serum from this leptin receptor deficient obesity and IR rat model, with the EC50 for repression of the PEPCK gene promoter again shifting from around 0.1 to 1 nM insulin (Fig.l D).
  • the PEPCK-CAT reporter gene is more sensitive to insulin than the endogenous PEPCK gene promoter, with the EC50 for PEPCK mRNA repression by insulin closer to 1 nM (Fig.2).
  • growth of the H4IIE cell line in serum from obese Fa/Fa rats also reduced the insulin sensitivity of the endogenous PEPCK gene promoter, with a significant difference between the response to 1 or 10nM insulin (Fig.2).
  • the effect was more apparent when cells were grown in 5% serum (with no supplement of FCS) (Fig.2).
  • the serum from Fa/Fa rats also made the PEPCK mRNA levels more sensitive to glucocorticoid and cAMP induction (Fig.2 and Fig.3).
  • the cell model can identify agents likely to improve insulin sensitivity, including those that have no affect on insulin sensitivity in healthy cells, but also distinguish between insulin sensitising agents and drugs that simply repress the PEPCK gene in an insulin mimetic type manner. Analysis of human serum donated by obese diabetic volunteers
  • Serum was donated by twenty obese (BMI>30) diabetic volunteers (being treated by diet alone), and twenty lean healthy volunteers. Cells were then cultures for 3 weeks in all forty serum samples (5%) prior to analysis of insulin repression of PEPCK mRNA.
  • the average response to 0.1 nM or 0.5n insulin of H4IIE cells cultured in serum from obese diabetics was significantly different from that of cells cultured in serum from lean non-diabetics ( Figure 5). However there was no difference in the magnitude of gene induction by glucocorticoids/cAMP, or in the response to higher concentrations of insulin between controls and cases. There was a broad range of response across the individual samples from the obese diabetics (Figure 6).
  • the Cases could be separated into two groups based on the insulin sensitivity of the cells following growth in the serum; most insulin resistant (Group 1 , lowest 10 responses to insulin) and most insulin sensitive (Group 2, highest 10 response, closest response to that seen in the average control).
  • the levels of glucose, total and LDL cholesterol along with measurements of weight, waist size and waisthip ratio were significantly different between Groups 1 and 2 (Table 2). Differences in blood leptin concentration and B I approached significance (Table 2).
  • the lower total and LDL cholesterol in the cases compared to controls were most probably due to the effects of HMG CoA reductase inhibitors which all of the Cases were taking.
  • HMG CoA reductase inhibitors which all of the Cases were taking.
  • Stern MP The Insulin resistance Syndrome. In International Textbook of Diabetes Mellitus Alberti KGMM, Zimmet P, DeFronzo RA, Keen H, Eds., John Wiley & Sons Ltd, 1997, p. 255-286
  • Dyck DJ Adipokines as regulators of muscle metabolism and insulin sensitivity. Appl.Physiol.Nutr.Metab. 34:396-402, 2009

Abstract

We disclose a cell-based bioassay allowing the analysis of insulin resistance in subjects suffering from or susceptible to type 2 diabetes.

Description

Cell Based Bioassay
The invention relates to a cell-based bioassay and its use in the identification of insulin sensitizing agents and including a diagnostic assay for the determination of a subject's sensitivity to insulin and optionally to treat a subject either suffering from type 2 diabetes or wherein said subject has a predisposition to type 2 diabetes.
Insulin functions to regulate glucose homeostasis. In conditions of hyperglycemia [abnormally high levels of serum glucose] the pancreatic β cells of the Islets of Langerhans synthesize proinsulin which is enzymatically cleaved at its amino and carboxy-termini to produce insulin, a 51 amino acid polypeptide. Insulin is secreted and acts on target cells [e.g. liver, muscle, adipose tissue] that express insulin receptors. The activation of insulin receptors leads to a signal transduction cascade that results in expression of glucose transporters in muscle which remove excess glucose with subsequent conversion of glucose into glycogen for storage. In liver, insulin shuts off endogenous glucose production and hepatic glucose output. Once glucose levels return to normal insulin is degraded thus removing its biological effects. The insulin receptor is a tyrosine kinase and is a tetrameric transmembrane receptor comprising two a subunits and two β subunits. The a subunits are extracellular and bind insulin. The β subunits are transmembrane and include ATP and tyrosine kinase domains which become activated on insulin binding.
There are a number of pathological conditions that result in hyperglycaemia the most well known being diabetes mellitus. Diabetes mellitus can be of type 1 or type 2. Type 1 diabetes is an autoimmune disease resulting in destruction of the pancreatic β cells which means the subject is unable to manufacture any insulin. Type 2 diabetes is a more complicated condition and can result from a number of associated ailments but typically involves resistance to the metabolic actions of insulin. For example, type 2 diabetes is associated with age, obesity, a sedentary life style and insulin resistance. An associated condition is called Metabolic Syndrome which may predispose subjects to type 2 diabetes. The symptoms associated with this syndrome are high blood pressure, dyslipidemia, increased body fat deposition and cardiovascular disease.
Administration of insulin is an effective means to control conditions such as type 1 and type 2 diabetes. Insulin resistance is a common characteristic of type 2 diabetes mellitus and is one of the major independent risk factors for progression of the disease. The treatment of insulin resistance is either by substantial weight loss and exercise with a concomitant increase in insulin sensitivity or by administration of insulin sensitizing agents which restore insulin sensitivity to those subjects that present with insulin resistance. The current approved agents to control diabetes 2 that are proposed to have some insulin sensitizing activity are rosiglitazone or metformin which both have undesirable side effects. The identification of novel insulin sensitizing agents that have better disease control and side effect profile is highly desirable.
In order to design truly effective insulin sensitising agents targeted at the mechanism of disease development in response to obesity we have generated an obesity-related insulin resistant cell model. Rat hepatoma cells (H4IIE) are grown in the presence of serum isolated from obese rodents or obese human volunteers and the insulin sensitivity of the cells monitored over time by the repression of a key insulin responsive gene. Higher insulin concentrations are required to fully repress the gene in the cells grown in obese rodent serum compared with those grown in serum from lean rodents (almost a 10-fold shift in insulin sensitivity). This was reversed by restoring normal growth medium, while the insulin resistance could be prevented by rosiglitazone or metformin. Meanwhile, growth of cells in serum collected from obese human volunteers with diabetes also reduced the insulin sensitivity of the rat cells. High body mass index, glucose and leptin levels correlated with the ability of serum from an individual (even within the obese diabetic group) to generate insulin resistance in the model.
This disclosure relates to a novel insulin resistant cell model that can be used to study the molecular development of obesity-linked insulin resistance, screen for compounds which overcome obesity-related insulin resistance and sub-classify insulin resistant patients thereby influencing their treatment.
Statements of Invention
According to an aspect of the invention there is provided a screening method for the identification of an insulin sensitizing agent comprising the steps:
i) providing a cell wherein said cell is transfected with a nucleic acid molecule comprising an insulin responsive promoter wherein said promoter is operably linked to a nucleic acid molecule that encodes a reporter molecule; ii) forming a preparation comprising the cell in i) and an isolated biological sample obtained from a subject wherein said subject is either: a) obese; or
b) non-obese; and,
incubating said preparation under cell culture conditions conducive to the growth and maintenance of said cell in culture;
iii) addition of at least one an agent to be tested for insulin sensitizing activity to the preparation^] formed in ii); and
iv) analyzing the biological effect of said agentfs] on at least one biomarker of insulin activity and comparing said biomarker to a control cell population which has not be contacted with said agent.
In a preferred method of the invention said agent[s] have an effect on the activity of the insulin responsive promoter.
In a preferred method of the invention said promoter is the mammalian cytosolic soluble phosphoenolpyruvate [GTP] gene promoter, or regulatory part thereof.
Examples of PEPCK gene sequences, include UniGene Hs.1872 Homo sapiens (human) PCK1. UniGene Rn.104376 Rattus norvegicus (Norway rat) Pck1 or UniGene Mm.477474 Mus musculus (mouse) Pck1],
Obesity" is measured in relation to a scale based on Body Mass Index Body Mass Index (BMI) is an approximate measure of obesity, and is calculated by dividing the mass of the individual (in kilograms) by the height squared (in metres squared), i.e. BMI= Weight (kg)/Height2 (m2). A healthy BMI range in UK is 18-25, overweight is 25-30 and obese is >30.
In a preferred method of the invention said cell is a mammalian cell adapted for expression of the human PEPCK promoter.
In a preferred method of the invention said cell is a human cell.
In an alternative preferred method of the invention said cell is a rodent cell, for example a rat or mouse cell. In a preferred method of the invention said cell is a cell that is naturally responsive to insulin.
In a preferred method of the invention said cell is selected from the group consisting of: a hepatocyte, an adipocyte or a muscle cell.
In a preferred method of the invention said cell is a hepatocyte.
In a preferred method of the invention said hepatocyte cell is the rat hepatocyte cell-line RH4IIE.
In a preferred method of the invention said biological sample is a serum sample, preferably a serum sample isolated from an obese human subject.
In a preferred method of the invention said reporter molecule is a fluorescence reporter.
The analysis of promoter activity can be conveniently monitored by fusing an insulin responsive promoter, such as the PEPCK promoter, to a nucleic acid that encodes a "reporter" protein or polypeptide. Examples are well known in the art and include enzymes such as β glucuronidase, chloramphenicol acetyltransferase or luciferase. Reporters that are proteinaceous fluorophores are also known in the art. Green fluorescent protein, GFP, is a spontaneously fluorescent protein isolated from coelenterates, such as the Pacific jellyfish, Aequoria victoria. Its role is to transduce, by energy transfer, the blue chemiluminescence of another protein, aequorin, into green fluorescent light. GFP can function as a protein tag, as it tolerates N- and C-terminal fusions to a broad variety of proteins many of which have been shown to retain native function. Most often it is used in the form of enhanced GFP in which codon usage is adapted to the human code. Other proteinaceous fluorophores include yellow, red and blue variant fluorescent proteins [e.g. BFP, CFP, GFP, YFP and RFP] These are commercially available from, for example Clontech (www. clontech. com) .
In a preferred method of the invention said fluorescence reporter is a fluorescent protein selected from the group comprising: BFP, CFP, GFP, YFP and RFP, and the mutants thereof. In a preferred method of the invention said method detects one, two, three, four, five, six or seven biomarkers indicative of insulin activity.
In a preferred method of the invention said at least one biomarker is selected from the group consisting of: leptin and/or LDL cholesterol and/or triglycerides.
According to a further aspect of the invention there is provided a cell array wherein said array comprises a plurality of identical assay preparations comprising a cell wherein said cell is transfected with a nucleic acid molecule comprising an insulin responsive promoter wherein said promoter is operably linked to a nucleic acid molecule that encodes a reporter molecule.
In a preferred embodiment of the invention said insulin responsive promoter is the PEPCK promoter, preferably the human PEPCK promoter.
The screening of large numbers of agents requires preparing arrays of cells for the handling of cells and the administration of agents. Assay devices, for example, include standard multiwell microtitre plates with formats such as 6, 12, 48, 96 and 384 wells which are typically used for compatibility with automated loading and robotic handling systems. Typically, high throughput screens use homogeneous mixtures of agents with an indicator compound which is either converted or modified resulting in the production of a signal. The signal is measured by suitable means (for example detection of fluorescence emission, optical density, or radioactivity) followed by integration of the signals from each well containing the cells, agent and indicator compound.
According to a further aspect of the invention there is provided a method for the high through put screening of insulin sensitizing agents comprising the steps:
i) providing an array according to the invention;
ii) contacting the array with a plurality of agents to be tested for insulin sensitizing activity;
iii) collating the activity data in (ii) above;
iv) converting the collated data into a data analysable form; and optionally v) providing an output for the analysed data.
A number of methods are known which image and extract information concerning the spatial and temporal changes occurring in cells or nuclei expressing, for example fluorescent molecules and other markers of gene expression (see Taylor et al Am. Scientist 80: 322-335, 1992). Moreover, US5, 989,835 and US09/031.271 , both of which are incorporated by reference, disclose optical systems for determining the distribution or activity of fluorescent reporter molecules in cells for screening large numbers of agents for biological activity. The systems disclosed in the above patents also describe a computerised method for processing, storing and displaying the data generated.
According to a further aspect of the invention there is provided a method to analyse a subject to determine whether said subject is or is not insulin resistant comprising the steps:
i) providing a cell wherein said cell is transfected with a nucleic acid molecule comprising an insulin responsive promoter wherein said promoter is operably linked to a nucleic acid molecule that encodes a reporter molecule;
ii) forming a preparation comprising the cell in i) and an isolated biological sample obtained from an obese subject, or a subject suspected of being susceptible to obesity, and incubating said preparation under cell culture conditions conducive to the growth and maintenance of said cell in culture;
iii) analyzing the biological effect of said isolated biological sample on at least one biomarker of insulin activity and comparing said biomarker to a control cell population which is obtained from a non-obese subject; and iv) determining whether the subject is or is not insulin resistant.
In a preferred method of the invention said isolated biological sample is analyzed for an effect on the activity of the insulin responsive promoter.
According to a further aspect of the invention there is provided a method to analyse and treat a human subject suspected of suffering from or having a predisposition to type 2 diabetes comprising the steps:
i) providing a cell wherein said cell is transfected with a nucleic acid molecule comprising an insulin responsive promoter wherein said promoter is operably linked to a nucleic acid molecule that encodes a reporter molecule; ii) forming a preparation comprising the cell in i) and an isolated biological sample obtained from an obese human subject, or a human subject suspected of being susceptible to obesity, and incubating said preparation under cell culture conditions conducive to the growth and maintenance of said cell in culture;
iii) analyzing the biological effect of said isolated biological sample on at least one biomarker of insulin activity and comparing said biomarker to a control cell population which is obtained from a non-obese subject;
iv) determining whether the subject contains a serum factor capable of generating hepatic insulin resistance; and
v) determining a treatment regime that will benefit the subject in controlling or preventing the onset of insulin resistance and hence type 2 diabetes.
In a preferred method of the invention said insulin responsive promoter is the PEPCK promoter, preferably the human PEPCK promoter.
In a preferred method of the invention said biological sample is a body fluid sample, for example a blood or serum sample.
Throughout the description and claims of this specification, the words "comprise" and "contain" and variations of the words, for example "comprising" and "comprises", means "including but not limited to", and is not intended to (and does not) exclude other moieties, additives, components, integers or steps.
Throughout the description and claims of this specification, the singular encompasses the plural unless the context otherwise requires. In particular, where the indefinite article is used, the specification is to be understood as contemplating plurality as well as singularity, unless the context requires otherwise.
Features, integers, characteristics, compounds, chemical moieties or groups described in conjunction with a particular aspect, embodiment or example of the invention are to be understood to be applicable to any other aspect, embodiment or example described herein unless incompatible therewith.
An embodiment of the invention will now be described by example only and with reference to the following figures: Figure 1 illustrates that the growth of cells in serum from obese rodents alters the insulin sensitivity of the PEPCK gene promoter. HL1 C cells (H4IIE cells with CAT reporter) were grown in serum isolated from obese rats that had been on a high fat diet for 3 months (HFS), or lean chow fed littermates (Con). After 9 days (A) or 24 days (B), cells were serum starved for 16h prior to exposure to 8CPT-CAMP (0.1 mM), dexamethasone (500n ) and insulin as indicated for 3h. Protein was harvested and CAT reporter assayed as a measure of PEPCK gene promoter activity as detailed under Methods. (C) A direct comparison of insulin regulation of PEPCK gene promoter in cells exposed to HFS for 1 , 2 or 3 weeks. *p<0.05, comparison of 3 weeks and control. (D) HL1 C cells were grown for 3 weeks in serum isolated from obese Fa/Fa or lean Fa/+ rats prior to analysis of insulin regulation of the PEPCK gene promoter as above. Data is presented relative to the CAT activity in the appropriate Dex/cAMP induced control;
Figure 2 illustrates that the growth of cells in serum from obese rodents alters insulin regulation of PEPCK mRNA. HL1C cells were grown for 3 weeks in serum isolated from obese Fa/Fa or lean Fa/+ rats prior to serum starvation and exposure to glucocorticoid (Dex), cAMP and insulin as in Fig.1. After exposure to hormones total RNA was isolated and PEPCK mRNA levels were quantified by RT-PCR. Data is presented as relative PEPCK mRNA (average ±SEM) after normalisation to cyclophilin mRNA in each sample. Percent inhibition by insulin is indicated in each case. *p<0.05;
Figure 3 illustrates that normal insulin sensitivity of cells is restored by returning the cells to standard serum. (A) H4IIE cells were grown for 3 weeks in 5% Fa/Fa serum. RNA was harvested from half of the cells following exposure to hormones as in Fig.1 , the remainder were placed in 5% FCS for 1 week prior to hormone exposure and RNA isolation, while RNA was also isolated from H4IIE cells grown for 3 weeks in Fa/+ lean serum followed by 1 week in FCS prior to hormone exposure. (B) Relative PEPCK mRNA (average ±SEM) for the samples isolated above are shown following normalisation for cyclophilin mRNA in each sample. Percent inhibition by insulin is indicated in each case. *p<0.05;
Figure 4 illustrates the generation of insulin resistance is prevented by pioglitazone and metformin. H4IIE cells were grown for 3 weeks in 5% Fa/Fa serum, in the presence or absence of pioglitazone or metformin as indicated. Cells were serum starved for 16h prior to 3h exposure to dexamethasone (500nM), 8CPT-CAMP (0.1 mM) ± insulin as indicated, and RNA was isolated. PEPCK mRNA levels (average ±SEM) are presented for each cell treatment after correction for cyclophilin mRNA. Percent inhibition by 1 n insulin in each condition is presented.
Figure 5 illustrates that the growth of cells in serum from obese humans with diabetes alters insulin regulation of PEPCK mRNA. H4IIE cells were grown for 3 weeks in 5% serum collected from 40 human volunteers, 20 who were obese and diabetic (Cases), 20 who were lean and non-diabetic (Controls). After 3 weeks in the serum the 40 different pools of cells were serum starved for 16h prior to 3h exposure to dexamethasone (500n ), 8CPT-cAMP (0.1 mM) ± insulin as indicated, and RNA was isolated. PEPCK mRNA levels (average ±SEM) are presented (n=20, assayed at least in duplicate for each condition) after correction for cyclophilin mRNA;
Figure 6 illustrates that there is variation in the degree of insulin resistance generated by growth of cells in serum from obese volunteers with diabetes. The response to 0.1 nM insulin for each individual pool of cells grown in the 20 serums collected from the obese diabetic volunteers is shown relative to the average Control response. The 10 serums that generated the greatest resistant to insulin are labelled insulin resistant and a comparison of clinical parameters of the insulin resistant group is presented in Table 2
Figure 7 is the nucleotide sequence of the rat PEPCK promoter.
Materials and Methods
Actrapid (human insulin) was from Novo Nordisk A/S (Bagsvaerd, Denmark). 2x Universal PCR Master Mix, No AmpErase UNG from Applied Biosystems, Complete™ protease inhibitor cocktail tablets from Roche, and 8-(4-chloro-phenylthio)-cAMP from Calbiochem. All primers and probes were synthesized and purified by Sigma-Aldrich. All other chemicals were of the highest grade obtainable.
Animals
Diets were purchased from research diets (HF diet D12331 and control diet D12328, http://www.researchdiets.com/pdf/Data-20Sheets/D12331.pdf) and Male Sprague Dawleys (starting at 10 weeks of age) were kept on the diet for up to 3 months prior to sacrifice. Serum was prepared immediately and all serum from each group combined, then aliquoted and snap frozen for storage at -80oC. Cell Culture
H4IIE cells, a rat hepatoma cell, and HL1 C (a H4IIE line with a stable insertion of a PEPCK promoter, chloramphenicol acetyl transferase gene construct (), were maintained in DMEM (Dulbecco's modified Eagle's medium) containing 1000 mg/l glucose, 1 % (v/v) penicillin/streptomycin, and 5 % (v/v) FBS, in a 37 °C 5 % C02 incubator.
Exposure of cells to experimental serum
Three different mix of sera were used: 1) 2.5 % foetal bovine serum plus 2.5 % rat serum obtained either from regularly fed rats (Control Serum) or from rats following a high fat diet (High Fat Serum); 2) 2.5 % foetal bovine serum plus 2.5 % serum from lean regular rats (Control Serum) or FaFa obese rats (Obese Serum); 3) 5 % human serum from either lean volunteers (Control Human Serum) or obese diabetic volunteers (CASE Human Serum).
In brief, cells were cultured and passaged as normal in the experimental sera. Cells were serum starved for 16 hours prior to measurement of insulin sensitivity. Hormones were added as described in figure legends, and after 3h protein (for CAT Assay, HL1C) or RNA (for Taqman, H4IIE) were harvested.
CAT Assay
Cells were isolated by trypsinisation, washed in PBS and pelleted by centrifugation at 1400 rpm for 5 min. Cell lysate was prepared by sonication in 0.25M TRIS, total protein assessed by BCA assay (Pierce), and general cellular protein removed by heating at 65oC for 10 min and centrifugation. Supernatants were tested for CAT activity as described previously (23), and data expressed as Units of CAT activity per mg of cellular protein.
RNA Isolation and Real-Time quantitative reverse transcriptase PCR
Following hormone treatments total cellular RNA was extracted from H4IIE cells using TRIreagent™ (Sigma) according to the manufacturer's instructions. cDNA was synthesized from total cellular RNA using Superscript™ II reverse transcriptase kit (Invitrogen). Real-time PCR analysis was carried out using ABIPrism7700 sequence detector (Applied Biosystems). Primer sequences were to rat PEPCK (forward- ACA GGC AAG GTC ATC ATG CA; reverse- TGC CGA AGT TGT AGC CAA AGA; probe- FAM-ACC CCT TCG CTA TGC GGC CC-TAMRA) and rat cyclophilin (forward- TTA CTA GGT CTG GCA GGA AGA TTA AAG; reverse- CTG CAT CTC TTG TCT CCA ATG TG; probe- FAM- AGA GGA CCA AGG CGT TAT CGA A-TAMRA). PCR reactions were carried out using the following cycling conditions - 50°C - 2min x1 , 95°C - 10min x1 followed by 40 cycles of 95°C - 15s, 60°C - 1 min.
Human Volunteers/Recruitment
Twenty control (BMI<25, Non-Diabetic) subjects were recruited from the general population along with twenty obese (BMI>30) type 2 diabetics recruited from the diabetes clinic. All subjects were male, non-smokers and taking no other medication known to influence insulin sensitivity (Table 1). They were relatively pharmaceutically naive subjects to reduce potential confounding effects of long term intervention, the exception being that all Cases were on a statin at time of visit. Volunteers fasted overnight prior to a visit to the Clinical Research Centre where 200ml of fasted whole blood was collected. Serum was prepared and stored at -80°C in 10ml_ aliquots. BMI, blood pressure and waist measurements were taken during the visit.
Clinical and Biochemical Measurements
TNFa, insulin, adiponectin, CRP and leptin were assayed in plasma or serum by ELISA in the Translational Medicine Research Institute. Other biochemical measurements were performed by the Clinical Biochemistry Department, Ninewells Hospital.
EXAMPLES
Diet induced obesity
15 male Sprague Dawley rats (10 weeks of age) were placed on a high fat (HF) diet for 12 weeks. 15 littermates were given normal chow (Con) for the same period. Serum was prepared from both the diet induced obese (DIO) and normal chow fed control animals, and this was aliquoted and stored at -80C until required. HL1C cells were grown in DMEM containing either 2.5% FCS + 2.5% DIO-derived serum, or 2.5% FCS + 2.5% Con serum. The cell morphology and growth rate during culture and passage (every 2 or 3 days) was similar in both cases (data not shown). After 9 or 24 days cells were serum starved for 3h, then exposed to hormones prior to measurement of PEPCK transcription. HL1C cells contain a CAT reporter with expression under the control of the PEPCK gene promoter (23; 24). In these cells CAT expression is normally low but can be induced 5- 10 fold by exposure to glucocorticoid and cAMP for 3h. However the induction can be completely blocked by including insulin in the culture medium (23). Repression by insulin is dose-dependent with an EC50 of approximately 0.1 nM, and >80% inhibition at 1 nM insulin (Fig.lA and (23; 25-27)). Growth in DIO-derived rat serum for 9 days did not produce any change in the insulin sensitivity of the HL1C cells (Fig.lA), however exposure to DIO-derived serum for 24 days resulted in a right shift of the insulin dose response curve (Fig.l B). The EC50 for insulin regulation of PEPCK-CAT rose to 1 nM although complete inhibition of the gene promoter was still achieved by 10nM insulin (Fig. l B). The experiment was repeated with fresh DIO-derived serum and the insulin sensitivity of the HL1C cells was reduced following 2 weeks of growth in the test serum (Fig.1C), but did not change between 2 and 3 weeks of exposure (Fig.lC). This suggested that there is a maximal effect of the serum on insulin sensitivity, and this occurs around 2 weeks of growth in DIO-derived serum.
Genetic predisposition to obesity
We next obtained serum from 12-14 week old FaFa rats (obese), and age matched Fa/+ (lean) rats. HL1C cells were grown in either 2.5% FaFa or 2.5% Fa/+ serum (supplemented with 2.5% FCS in each case), for 3 weeks prior to analysis of PEPCK gene promoter regulation (Fig 1 D). Once more insulin sensitivity of the HL1C cells was reduced in cells grown in serum from this leptin receptor deficient obesity and IR rat model, with the EC50 for repression of the PEPCK gene promoter again shifting from around 0.1 to 1 nM insulin (Fig.l D).
Endogenous Gene Regulation
The PEPCK-CAT reporter gene is more sensitive to insulin than the endogenous PEPCK gene promoter, with the EC50 for PEPCK mRNA repression by insulin closer to 1 nM (Fig.2). However, growth of the H4IIE cell line in serum from obese Fa/Fa rats also reduced the insulin sensitivity of the endogenous PEPCK gene promoter, with a significant difference between the response to 1 or 10nM insulin (Fig.2). The effect was more apparent when cells were grown in 5% serum (with no supplement of FCS) (Fig.2). The serum from Fa/Fa rats also made the PEPCK mRNA levels more sensitive to glucocorticoid and cAMP induction (Fig.2 and Fig.3). Reversibility
In order to establish whether normal insulin sensitivity could be restored to the 'resistant' cells, we generated insulin resistance by culturing cells for 3 weeks in 5% Fa/Fa serum, then transferring half of the cells to 5% FCS for 1 week, and comparing the insulin sensitivity of the two batches of cells and also cells grown throughout in 5% Fa/+ lean serum for an equivalent time (Fig.3). Removal of the Fa/Fa serum for 1 week restored the normal insulin sensitivity of the cells, showing that the resistance is reversible.
Effect of Insulin Sensitisers
Cells were incubated with Fa/Fa or Fa/+ serum ± 0.1 mM metformin, or ± 1 μΜ Pioglitazone. Drugs were replaced at every cell passage (along with fresh serum), and after 3 weeks the insulin sensitivity of the cells established (Fig.4). The presence of metformin reduced PEPCK gene transcription, irrespective of the growth conditions or hormone challenge. Indeed 3 weeks growth in the presence of metformin reduced basal PEPCK transcript, partly reduced the induction by glucocorticoids and cAMP, while restoring full repression by insulin (Fig.4). The same response was seen whether the cells had been grown in Fa/+ or FaFa serum, hence this drug prevented reduced insulin action on PEPCK transcription by the Fa/Fa serum, but appeared to do so by directly repressing the gene irrespective of the presence of insulin resistance.
In contrast, growth in Pioglitazone resulted in prevention of reduced insulin sensitivity of the cells exposed to Fa/Fa serum without any effect on cells grown in Fa/+ serum (Fig.4). Basal, induced and insulin-repressed levels of the cells in Fa/+ serum were almost identical ± Pioglitazone. Meanwhile, this drug restored insulin repressive actions in the Fa/Fa serum treated cells to that of the Fa/+ serum treated cells, indicating it improved insulin sensitivity rather than simply repressed PEPCK transcription.
This suggests that the cell model can identify agents likely to improve insulin sensitivity, including those that have no affect on insulin sensitivity in healthy cells, but also distinguish between insulin sensitising agents and drugs that simply repress the PEPCK gene in an insulin mimetic type manner. Analysis of human serum donated by obese diabetic volunteers
Serum was donated by twenty obese (BMI>30) diabetic volunteers (being treated by diet alone), and twenty lean healthy volunteers. Cells were then cultures for 3 weeks in all forty serum samples (5%) prior to analysis of insulin repression of PEPCK mRNA. The average response to 0.1 nM or 0.5n insulin of H4IIE cells cultured in serum from obese diabetics was significantly different from that of cells cultured in serum from lean non-diabetics (Figure 5). However there was no difference in the magnitude of gene induction by glucocorticoids/cAMP, or in the response to higher concentrations of insulin between controls and cases. There was a broad range of response across the individual samples from the obese diabetics (Figure 6). The Cases could be separated into two groups based on the insulin sensitivity of the cells following growth in the serum; most insulin resistant (Group 1 , lowest 10 responses to insulin) and most insulin sensitive (Group 2, highest 10 response, closest response to that seen in the average control). The levels of glucose, total and LDL cholesterol along with measurements of weight, waist size and waisthip ratio were significantly different between Groups 1 and 2 (Table 2). Differences in blood leptin concentration and B I approached significance (Table 2). The lower total and LDL cholesterol in the cases compared to controls were most probably due to the effects of HMG CoA reductase inhibitors which all of the Cases were taking. However it was surprising that there was a significant difference between cholesterol levels between the two obese diabetic groups, with a significantly higher cholesterol level in the resistant group, despite 19 out of the 20 Cases being on the same statin (Table 1).
References:
1. Stern MP: The Insulin resistance Syndrome. In International Textbook of Diabetes Mellitus Alberti KGMM, Zimmet P, DeFronzo RA, Keen H, Eds., John Wiley & Sons Ltd, 1997, p. 255-286
2. Zimmet P, Alberti KGMM, Shaw J: Global and social implications of the diabetic epidemic. Nature 414:782-787, 2001
3. Woods YL, Petrie JR, Sutherland C: Dissecting insulin signaling pathways: individualised therapeutic targets for diagnosis and treatment of insulin resistant states. Endocrine, Metabolic & Immune Disorders - Drug Targets 9:187-198, 2009
4. Shulman Gl: Cellular mechanisms of insulin resistance. J.Clin. Invest. 106:171-176, 2000
5. Lazar MA: How obesity causes diabetes: not a tall tale. Science 307:373-375, 2005 6. O'Rahilly S: Human obesity and insulin resistance: lessons from experiments of nature. Biochem.Soc. Trans. 35:33-36, 2007
7. Powell DJ, Hajduch E, Kular G, Hundal HS: Ceramide disables 3-phosphoinositide binding to the pleckstrin homology domain of protein kinase B (PKB)/Akt by a PKCzeta- dependent mechanism. Mol. Cell. Biol. 23:7794-7808, 2003
8. Gao Z, Zhang X, Zuberi A, Hwang D, Quon MJ, Lefevre , Ye J: Inhibition of insulin sensitivity by free fatty acids requires activation of multiple serine kinases in 3T3-L1 adipocytes. Mol Endocrinol 18:2024-2034, 2004
9. Sharfi H, Eldar-Finkelman H: Sequential phosphorylation of insulin receptor substrate- 2 by glycogen synthase kinase-3 and c-Jun NH2-terminal kinase plays a role in hepatic insulin signaling. Am J Physiol Endocrinol Metab 294:E307-315, 2008
10. Zick Y: Ser/Thr phosphorylation of IRS proteins: a molecular basis for insulin resistance. Sci STKE 2005:pe4, 2005
11. Bjornholm M, Kawano Y, Lehtihet M, Zierath JR: Insulin receptor substrate-1 phosphorylation and phosphatidylinositol 3-kinase activity in skeletal muscle from NIDD subjects after in vivo insulin stimulation. Diabetes 46:524-527, 1997
12. Beeson M, Sajan MP, Dizon M, Grebenev D, Gomez-Daspet J, Miura A, Kanoh Y, Powe J, Bandyopadhyay G, Standaert ML, Farese RV: Activation of protein kinase C- zeta by insulin and phosphatidylinositol-3,4,5-(P04)3 is defective in muscle in type 2 diabetes and impaired glucose tolerance: amelioration by rosiglitazone and exercise. Diabetes 52:1926-1934, 2003
13. Kim Y-B, Kotani K, Ciaraldi TP, Henry RR, Kahn BB: Insulin-stimulated protein kinase C lambda/zeta activity is reduced in skeletal muscle of humans with obesity and type 2 diabetes: reversal with weight reduction. Diabetes 52:1935-1942, 2003
14. Morino K, Petersen KF, Dufour S, Befroy D, Frattini J, Shatzkes N, Neschen S, White MF, Bilz S, Sono S, Pypaert M, Shulman Gl: Reduced mitochondrial density and increased IRS-1 serine phosphorylation in muscle of insulin-resistant offspring of type 2 diabetic parents. J. Clin. Invest. 115:3587-3593, 2005
15. Nikoulina SE, Ciaraldi TP, Mudaliar S, Carter L, Johnson K, Henry RR: Inhibition of glycogen synthase kinase 3 improves insulin action and glucose metabolism in human skeletal muscle. Diabetes 51 :2190-2198, 2002
16. Ring DB, Johnson KW, Henriksen EJ, Nuss JM, Goff D, Kinnick TR, Ma ST, Reeder JW, Samuels I, Slabiak T, Wagman AS, Hammond ME, Harrison SD: Selective GSK-3 inhibitors potentiate insulin activation of glucose transport and utilization in vitro and in vivo. Diabetes 52:588-595, 2003 17. Withers DJ, White M: Perspective: The Insulin Signaling System— A Common Link in the Pathogenesis of Type 2 Diabetes. Endocrinology 141 :1917-1921 , 2000
18. Nandi A, Kitamura Y, Kahn CR, Accili D: Mouse models of insulin resistance. Physiol Rev 84:623-647, 2004
19. Granner DK, Pilkis S: The genes of hepatic glucose metabolism. J.Biol.Chem. 265:10173-10178, 1990
20. Mora A, Lipina C, Tronche F, Sutherland C, Alessi DR: Deficiency of PDK1 in liver results in glucose intolerance, impairment of insulin regulated gene expression and liver failure. Biochem.J. 385:639-648, 2005
21. Cool B, Zinker B, Chiou W, Kifle L, Cao N, Perham M, Dickinson R, Adler A, Gagne G, Iyengar R, Zhao G, Marsh K, Kym P, Jung P, Camp HS, Frevert E: Identification and characterization of a small molecule AMPK activator that treats key components of type 2 diabetes and the metabolic syndrome. Cell Metab 3:403-416, 2006
22. Valera A, Pujol A, Pelegrin M, Bosch F: Transgenic mice overexpressing PEPCK develop NIDDM. Proc.Natl.Acad.Sci. 91 :9151-9154, 1994
23. Sutherland C, O'Brien RM, Granner DK: Phosphatidylinositol 3-kinase, but not p70/p85 ribosomal S6 protein kinase, is required for the regulation of Phosphoenolpyruvate Carboxykinase gene expression by insulin. J.Biol.Chem. 270:15501-15506, 1995
24. Forest CD, O'Brien RM, Lucas PC, Magnuson MA, Granner DK: Regulation of PEPCK gene expression by insulin. Use of the stable transfection approach to locate an insulin responsive sequence. Molecular Endocrinology 4:1302-1310, 1990
25. Sutherland C, Waltner-Law M, Gnudi L, Kahn BB, Granner DK: Activation of the Ras-MAP Kinase-RSK pathway is not required for the repression of PEPCK gene transcription by insulin. J.Biol.Chem. 273:3198-3204, 1998
26. Lochhead PA, Coghlan MP, Rice SQJ, Sutherland C: Inhibition of GSK3 selectively reduces G6Pase and PEPCK gene expression. Diabetes 50:937-947, 2001
27. Patel S, Lipina C, Sutherland C: Different mechanisms are used by insulin to repress three genes that contain a homologous thymine-rich insulin response element. FESS Lett 549:72-76, 2003
28. Goodyear LJ, Giorgino F, Sherman LA, Carey J, Smith RJ, Dohm GL: Insulin receptor phosphorylation, insulin receptor substrate-1 phosphorylation, and phosphatidylinositol 3-kinase activity are decreased in intact skeletal muscle strips from obese subjects. J Clin Invest 95:2195-2204, 1995
29. Seppala-Lindroos A, Vehkavaara S, Hakkinen AM, Goto T, Westerbacka J, Sovijarvi A, Halavaara J, Yki-Jarvinen H: Fat accumulation in the liver is associated with defects in insulin suppression of glucose production and serum free fatty acids independent of obesity in normal men. J.CIin.Endocrinol.Metab. 87:3023-3028, 2002
30. Prada PO, Zecchin HG, Gasparetti AL, Torsoni MA, Ueno M, Hirata AE, Corezola do Amaral ME, Hoer NF, Boschero AC, Saad MJ: Western diet modulates insulin signaling, c-Jun N-terminal kinase activity, and insulin receptor substrate- 1ser307 phosphorylation in a tissue-specific fashion. Endocrinology 1 6:1576-1587, 2005
31. Rasouli N, Kern PA: Adipocytokines and the metabolic complications of obesity. J.CIin.Endocrinol.Metab. 93:S64-73, 2008
32. Dyck DJ: Adipokines as regulators of muscle metabolism and insulin sensitivity. Appl.Physiol.Nutr.Metab. 34:396-402, 2009
33. Sutherland C, O'Brien RM, Granner DK: Genetic regulation of glucose metabolism. In Handbook of Physiology Jefferson LS, Cherrington AD, Eds., Oxford University Press, 2003, p. 707-734
34. Logie L, Ruiz-Alcaraz AJ, Keane M, Woods YL, Bain J, Marquez R, Alessi DR, Sutherland C: Characterization of a protein kinase B inhibitor in vitro and in insulin- treated liver cells. Diabetes 56:2218-2227, 2007
35. Finlay D, Patel S, Dickson LM, Shpiro N, Marquez R, Rhodes CJ, Sutherland C: Glycogen Synthase Kinase-3 Regulates IGFBP-1 Gene Transcription Through the Thymine-rich Insulin Response Element: Inhibition is required for full regulation of this promoter element by insulin. BMC Mol.Biol. 5:15, 2004
36. Finlay D, Ruiz-Alcaraz AJ, Lipina C, Perrier S, Sutherland C: A temporal switch in the insulin-signalling pathway that regulates hepatic IGFBP1 gene expression. J. Mol. Endocrinol. 37 : 1 - 12 , 2006
Table 1 : Comparison of clinical parameters of the insulin resistant and insulin sensitive
Cases.
Resistant Cases Sensitive Significance,
(n=10) Cases (n=10) students t-test (p=)
GLUCOSE (mmol/L) 9.2611.52 7.46+0.64 0.0537
HbAlc% 6.96±0.67 6.53+0.42 0.3048
INSULIN 17.25±7.2 15.61+2.65 0.6839
CRP 2676±11 15 3183+1055 0.2921
LEPTIN 19248±8239 1189213966 0.1389
ADIPONECTIN 4687±1431 4728+1606 0.9705
TNFa 1.2±0.3 1.06+0.1 0.41 16
T Choi mmol/L 4.74+0.44 4.13+0.41 0.0694
LDL mmol/L 2.62±0.41 2.1310.16 0.0787
HDL mmol/L 1.22±0.21 1.15+0.24 0.5688
Trigs mmol/L 2.34+0.83 1.97+0.94 0.5779
Age 57.7+1.65 55.7+2.74 0.2400
Height cm 176.6±4.95 172.8±3.16 0.2238
Weight Kg 1 14.46112.3 98.8714.33 0.0386*
BMI 36.7+3.58 33.2+2.2 0.1234
Waist:Hip 1.04+0.04 1.00+0.02 0.05
HOMA 2.44 2.18 0.6097

Claims

Claims
1 A screening method for the identification of an insulin sensitizing agent comprising the steps:
i) providing a cell wherein said cell is transfected with a nucleic acid molecule comprising an insulin responsive promoter wherein said promoter is operably linked to a nucleic acid molecule that encodes a reporter molecule;
ii) forming a preparation comprising the cell in i) and an isolated biological sample obtained from a subject wherein said subject is either:
a) obese; or
b) non-obese; and,
incubating said preparation under cell culture conditions conducive to the growth and maintenance of said cell in culture;
iii) addition of at least one agent to be tested for insulin sensitizing activity to the preparations] formed in ii); and iv) analyzing the biological effect of said agent[s] on the activity of the insulin responsive promoter and at least one biomarker of insulin activity and comparing said insulin responsive promoter activity and biomarker to a control cell population which has not be contacted with said agent.
2. The method according to claim 1 wherein said promoter is the phosphoenolpyruvate, cytosolic [GTP] gene promoter [PEPCK].
3. The method according to claim 2 wherein said cell is a mammalian cell adapted for expression of the human PEPCK promoter or regulatory part thereof.
4. The method according to any of claims 1-3 wherein said cell is a human cell.
5. The method according to any of claims 1-3 wherein said cell is a rodent cell.
6. The method according to any of claims 1-5 wherein said cell is a cell that is naturally responsive to insulin.
7. The method according to claim 6 wherein said cell is selected from the group consisting of: a hepatocyte, an adipocyte or a muscle cell.
8. The method according to claim 7 wherein said cell is a hepatocyte.
9. The method according to claim 8 wherein said hepatocyte cell is the rat hepatocyte cell-line RH4IIE.
10. The method according to any of claims 1 -9 wherein said biological sample is a serum sample.
1 1. The method according to any of claims 1-10 wherein the sample is isolated from an obese human subject.
12. The method according to any of claims 1 -1 1 wherein said reporter molecule is a fluorescence reporter.
13. The method according to claim 12 wherein said fluorescence reporter is a fluorescent protein selected from the group comprising: BFP, CFP, GFP, YFP and RFP, and the mutants thereof.
14. The method according to any of claims 1-13 wherein said method detects at least one, two, three, four, five, six or seven biomarkers indicative of insulin activity.
15. The method according to claim 14 wherein said at least one biomarker is selected from the group consisting of: leptin and/or LDL cholesterol and/or triglycerides.
16. A cell array wherein said array comprises a plurality of identical assay preparations comprising a cell wherein said cell is transfected with a nucleic acid molecule comprising an insulin responsive promoter wherein said promoter is operably linked to a nucleic acid molecule that encodes a reporter molecule.
17. The array according to claim 16 wherein said insulin responsive promoter is the PEPCK promoter.
18. The array according to claim 17 wherein said PEPCK promoter is the human PEPCK promoter.
19. A method for the high through put screening of insulin sensitizing agents comprising the steps:
i) providing an array according to any of claims 16-18; ii) contacting the array with a plurality of agents to be tested for insulin sensitizing activity;
iii) collating the activity data in (ii) above;
iv) converting the collated data into a data analysable form; and optionally
v) providing an output for the analysed data.
20. A method to analyse a subject to determine whether said subject contains a serum factor that induces insulin resistance comprising the steps:
i) providing a cell wherein said cell is transfected with a nucleic acid molecule comprising an insulin responsive promoter wherein said promoter is operably linked to a nucleic acid molecule that encodes a reporter molecule;
ii) forming a preparation comprising the cell in i) and an isolated biological sample obtained from an obese subject, or a subject suspected of being susceptible to obesity, and incubating said preparation under cell culture conditions conducive to the growth and maintenance of said cell in culture;
iii) analyzing the biological effect of said isolated biological sample on the activity of the insulin responsive promoter and at least one biomarker of insulin activity and comparing said insulin responsive promoter activity and biomarker to a control cell population which is obtained from a non-obese subject; and
iv) determining whether the subject is or is not insulin resistant.
21. A method to analyse and treat a human subject suspected of suffering from or having a predisposition to type 2 diabetes comprising the steps:
i) providing a cell wherein said cell is transfected with a nucleic acid molecule comprising an insulin responsive promoter wherein said promoter is operably linked to a nucleic acid molecule that encodes a reporter molecule; ii) forming a preparation comprising the cell in i) and an isolated biological sample obtained from an obese human subject, or a human subject suspected of being susceptible to obesity, and incubating said preparation under cell culture conditions conducive to the growth and maintenance of said cell in culture;
iii) analyzing the biological effect of said isolated biological sample on the activity of the insulin responsive promoter and at least one biomarker of insulin activity and comparing said insulin responsive promoter activity and biomarker to a control cell population which is obtained from a non-obese subject;
iv) determining whether the subject has a serum factor that induces insulin resistance; and
v) determining a treatment regime that will benefit the subject in controlling or preventing the onset of type 2 diabetes.
22. The method according to claim 20 or 21 wherein said insulin responsive promoter is the PEPCK promoter.
23. The method according to claim 22 wherein the PEPCK promoter is the human PEPCK promoter.
24. The method according to any of claims 20-23 wherein said biological sample is a body fluid sample.
25. The method according to claim 24 wherein said body fluid sample is a blood or serum sample.
PCT/GB2011/000170 2010-02-10 2011-02-10 Cell based bioassay WO2011098757A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GB201002130A GB201002130D0 (en) 2010-02-10 2010-02-10 Cell based bioassay
GB1002130.1 2010-02-10

Publications (1)

Publication Number Publication Date
WO2011098757A1 true WO2011098757A1 (en) 2011-08-18

Family

ID=42110412

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2011/000170 WO2011098757A1 (en) 2010-02-10 2011-02-10 Cell based bioassay

Country Status (2)

Country Link
GB (1) GB201002130D0 (en)
WO (1) WO2011098757A1 (en)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5989835A (en) 1997-02-27 1999-11-23 Cellomics, Inc. System for cell-based screening
WO2003034025A2 (en) * 2001-10-15 2003-04-24 Beckman-Coulter, Inc. Methods and reagents for improved cell-based assays
FR2853911A1 (en) * 2003-04-17 2004-10-22 Merck Sante Sas In vitro screening for compounds useful for treating diabetes, obesity and insulin resistance, from their ability to modulate expression or activity of the E2IG4 gene
WO2007038756A2 (en) * 2005-09-28 2007-04-05 Attagene Inc. Methods and constructs for analyzing biological activities of biological specimens and determining states of organism
WO2008033518A2 (en) * 2006-09-13 2008-03-20 The Trustees Of Columbia University In The City Of New York Undercarboxylated/uncarboxylated osteocalcin increases beta-cell proliferation, insulin secretion, insulin sensitivity, glucose tolerance and decreases fat mass

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5989835A (en) 1997-02-27 1999-11-23 Cellomics, Inc. System for cell-based screening
WO2003034025A2 (en) * 2001-10-15 2003-04-24 Beckman-Coulter, Inc. Methods and reagents for improved cell-based assays
FR2853911A1 (en) * 2003-04-17 2004-10-22 Merck Sante Sas In vitro screening for compounds useful for treating diabetes, obesity and insulin resistance, from their ability to modulate expression or activity of the E2IG4 gene
WO2007038756A2 (en) * 2005-09-28 2007-04-05 Attagene Inc. Methods and constructs for analyzing biological activities of biological specimens and determining states of organism
WO2008033518A2 (en) * 2006-09-13 2008-03-20 The Trustees Of Columbia University In The City Of New York Undercarboxylated/uncarboxylated osteocalcin increases beta-cell proliferation, insulin secretion, insulin sensitivity, glucose tolerance and decreases fat mass

Non-Patent Citations (40)

* Cited by examiner, † Cited by third party
Title
BEESON M; SAJAN MP; DIZON M; GREBENEV D; GOMEZ-DASPET J; MIURA A; KANOH Y; POWE J; BANDYOPADHYAY G; STANDAERT ML: "Activation of protein kinase C-zeta by insulin and phosphatidylinositol-3,4,5-(P04)3 is defective in muscle in type 2 diabetes and impaired glucose tolerance: amelioration by rosiglitazone and exercise", DIABETES, vol. 52, 2003, pages 1926 - 1934
BJORNHOLM M; KAWANO Y; LEHTIHET M; ZIERATH JR: "Insulin receptor substrate-1 phosphorylation and phosphatidylinositol 3-kinase activity in skeletal muscle from NIDDM subjects after in vivo insulin stimulation", DIABETES, vol. 46, 1997, pages 524 - 527
CHEN CECIL ET AL: "An integrated functional genomics screening program reveals a role for BMP-9 in glucose homeostasis.", NATURE BIOTECHNOLOGY, vol. 21, no. 3, March 2003 (2003-03-01), pages 294 - 301, XP002638478, ISSN: 1087-0156 *
COOL B; ZINKER B; CHIOU W; KIFLE L; CAO N; PERHAM M; DICKINSON R; ADLER A; GAGNE G; LYENGAR R: "Identification and characterization of a small molecule AMPK activator that treats key components of type 2 diabetes and the metabolic syndrome", CELL METAB, vol. 3, 2006, pages 403 - 416, XP002530440, DOI: doi:10.1016/J.CMET.2006.05.005
DYCK DJ: "Adipokines as regulators of muscle metabolism and insulin sensitivity", APPL. PHYSIOL. NUTR. METAB., vol. 34, 2009, pages 396 - 402
FINLAY D; PATEL S; DICKSON LM; SHPIRO N; MARQUEZ R; RHODES CJ; SUTHERLAND C: "Glycogen Synthase Kinase-3 Regulates IGFBP-1 Gene Transcription Through the Thymine-rich Insulin Response Element: Inhibition is required for full regulation of this promoter element by insulin", BMC MOLBIOL., vol. 5, 2004, pages 15
FINLAY D; RUIZ-ALCARAZ AJ; LIPINA C; PERRIER S; SUTHERLAND C: "A temporal switch in the insulin-signalling pathway that regulates hepatic IGFBP1 gene expression", J.MOL.ENDOCRINOL., vol. 37, 2006, pages 1 - 12
FOREST CD; O'BRIEN RM; LUCAS PC; MAGNUSON MA; GRANNER DK: "Regulation of PEPCK gene expression by insulin. Use of the stable transfection approach to locate an insulin responsive sequence", MOLECULAR ENDOCRINOLOGY, vol. 4, 1990, pages 1302 - 1310
GAO Z; ZHANG X; ZUBERI A; HWANG D; QUON MJ; LEFEVRE M; YE J: "Inhibition of insulin sensitivity by free fatty acids requires activation of multiple serine kinases in 3T3-L1 adipocytes", MOL ENDOCRINO, vol. 18, 2004, pages 2024 - 2034
GOODYEAR LJ; GIORGINO F; SHERMAN LA; CAREY J; SMITH RJ; DOHM GL: "Insulin receptor phosphorylation, insulin receptor substrate-1 phosphorylation, and phosphatidylinositol 3-kinase activity are decreased in intact skeletal muscle strips from obese subjects", J CLIN INVEST, vol. 95, 1995, pages 2195 - 2204
GRANNER DK; PILKIS S: "The genes of hepatic glucose metabolism", J.BIOLCHEM., vol. 265, 1990, pages 10173 - 10178
KIM Y-B; KOTANI K; CIARALDI TP; HENRY RR; KAHN BB: "Insulin-stimulated protein kinase C lambda/zeta activity is reduced in skeletal muscle of humans with obesity and type 2 diabetes: reversal with weight reduction", DIABETES, vol. 52, 2003, pages 1935 - 1942
LAZAR MA: "How obesity causes diabetes: not a tall tale", SCIENCE, vol. 307, 2005, pages 373 - 375
LOCHHEAD P A ET AL: "INHIBITION OF GSK-3 SELECTIVELY REDUCES GLUCOSE-6-PHOSPHATASE AND PHOSPHOENOLPYRUVATE CARBOXYKINASE GENE EXPRESSION", DIABETES, AMERICAN DIABETES ASSOCIATION, US, vol. 50, no. 5, 1 May 2001 (2001-05-01), pages 937 - 946, XP001056610, ISSN: 0012-1797, DOI: DOI:10.2337/DIABETES.50.5.937 *
LOCHHEAD PA; COGHLAN MP; RICE SQJ; SUTHERLAND C: "Inhibition of GSK3 selectively reduces G6Pase and PEPCK gene expression", DIABETES, vol. 50, 2001, pages 937 - 947
LOGIE L ET AL: "Generation, validation and humanisation of a novel insulin resistant cell model", BIOCHEMICAL PHARMACOLOGY, PERGAMON, OXFORD, GB, vol. 80, no. 7, 1 October 2010 (2010-10-01), pages 1042 - 1049, XP027215745, ISSN: 0006-2952, [retrieved on 20100623] *
LOGIE L; RUIZ-ALCARAZ AJ; KEANE M; WOODS YL; BAIN J; MARQUEZ R; ALESSI DR: "Sutherland C: Characterization of a protein kinase B inhibitor in vitro and in insulin- treated liver cells", DIABETES, vol. 56, 2007, pages 2218 - 2227
MORA A; LIPINA C; TRONCHE F; SUTHERLAND C; ALESSI DR: "Deficiency of PDK1 in liver results in glucose intolerance, impairment of insulin regulated gene expression and liver failure", BIOCHEM.J., vol. 385, 2005, pages 639 - 648
MORINO K; PETERSEN KF; DUFOUR S; BEFROY D; FRATTINI J; SHATZKES N; NESCHEN S; WHITE MF; BILZ S; SONO S: "Reduced mitochondrial density and increased IRS-1 serine phosphorylation in muscle of insulin-resistant offspring of type 2 diabetic parents", J. CLIN.LNVEST., vol. 115, 2005, pages 3587 - 3593
NANDI A; KITAMURA Y; KAHN CR; ACCILI D: "Mouse models of insulin resistance", PHYSIOL REV, vol. 84, 2004, pages 623 - 647
NIKOULINA SE; CIARALDI TP; MUDALIAR S; CARTER L; JOHNSON K; HENRY RR: "Inhibition of glycogen synthase kinase 3 improves insulin action and glucose metabolism in human skeletal muscle", DIABETES, vol. 51, 2002, pages 2190 - 2198
O'RAHILLY S: "Human obesity and insulin resistance: lessons from experiments of nature", BIOCHEM.SOC. TRANS., vol. 35, 2007, pages 33 - 36
PATEL S; LIPINA C; SUTHERLAND C: "Different mechanisms are used by insulin to repress three genes that contain a homologous thymine-rich insulin response element", FEBS LETT, vol. 549, 2003, pages 72 - 76, XP004446867, DOI: doi:10.1016/S0014-5793(03)00774-9
POWELL DJ; HAJDUCH E; KULAR G; HUNDAL HS: "Ceramide disables 3-phosphoinositide binding to the pleckstrin homology domain of protein kinase B (PKB)/Akt by a PKCzeta- dependent mechanism", MOLCEIL.BIOL., vol. 23, 2003, pages 7794 - 7808
PRADA PO; ZECCHIN HG; GASPARETTI AL; TORSONI MA; UENO M; HIRATA AE; COREZOLA DO; AMARAL ME; HOER NF; BOSCHERO AC: "Western diet modulates insulin signaling, c-Jun N-terminal kinase activity, and insulin receptor substrate-1ser307 phosphorylation in a tissue-specific fashion", ENDOCRINOLOGY, vol. 146, 2005, pages 1576 - 1587
RASOULI N; KERN PA: "Adipocytokines and the metabolic complications of obesity", J. CLIN. ENDOCRINOL. METAB., vol. 93, 2008, pages 64 - 73
RING DB; JOHNSON KW; HENRIKSEN EJ; NUSS JM; GOFF D; KINNICK TR; MA ST; REEDER JW; SAMUELS; SLABIAK T: "Selective GSK-3 inhibitors potentiate insulin activation of glucose transport and utilization in vitro and in vivo", DIABETES, vol. 52, 2003, pages 588 - 595, XP002305100, DOI: doi:10.2337/diabetes.52.3.588
SEPPALA-LINDROOS A; VEHKAVAARA S; HAKKINEN AM; GOTO T; WESTERBACKA J; SOVIJARVI A; HALAVAARA J; YKI-JARVINEN H: "Fat accumulation in the liver is associated with defects in insulin suppression of glucose production and serum free fatty acids independent of obesity in normal men", J.CLIN.ENDOCRINOL.METAB., vol. 87, 2002, pages 3023 - 3028
SHARFI H; ELDAR-FINKELMAN H: "Sequential phosphorylation of insulin receptor substrate-2 by glycogen synthase kinase-3 and c-Jun NH2-terminal kinase plays a role in hepatic insulin signaling", AM J PHYSIOL ENDOCRINOL METAB, vol. 294, 2008, pages E307 - 315
SHULMAN GI: "Cellular mechanisms of insulin resistance", J.CLIN.LNVEST., vol. 106, 2000, pages 171 - 176
STERN MP: "The Insulin resistance Syndrome. In International Textbook of Diabetes Mellitus", 1997, JOHN WILEY & SONS LTD, pages: 255 - 286
SUTHERLAND C, O'BRIEN RM, GRANNER DK: "Handbook of Physiology", 2003, OXFORD UNIVERSITY PRESS, article "Genetic regulation of glucose metabolism", pages: 707 - 734
SUTHERLAND C; O'BRIEN RM; GRANNER DK: "Phosphatidylinositol 3-kinase, but not p70/p85 ribosomal S6 protein kinase, is required for the regulation of Phosphoenolpyruvate Carboxykinase gene expression by insulin", J.BIOL.CHEM., vol. 270, 1995, pages 15501 - 15506
SUTHERLAND C; WALTNER-LAW M; GNUDI L; KAHN BB; GRANNER DK: "Activation of the Ras-MAP Kinase-RSK pathway is not required for the repression of PEPCK gene transcription by insulin", J.BIOL.CHEM., vol. 273, 1998, pages 3198 - 3204
TAYLOR ET AL., AM. SCIENTIST, vol. 80, 1992, pages 322 - 335
VALERA A; PUJOL A; PELEGRIN M; BOSCH F: "Transgenic mice overexpressing PEPCK develop NIDDM", PROC.NATL.ACAD.SCI., vol. 91, 1994, pages 9151 - 9154
WITHERS DJ; WHITE M: "Perspective: The Insulin Signaling System-A Common Link in the Pathogenesis of Type 2 Diabetes", ENDOCRINOLOGY, vol. 141, 2000, pages 1917 - 1921
WOODS YL; PETRIE JR: "Sutherland C: Dissecting insulin signaling pathways: individualised therapeutic targets for diagnosis and treatment of insulin resistant states", ENDOCRINE, METABOLIC & IMMUNE DISORDERS - DRUG TARGETS, vol. 9, 2009, pages 187 - 198
ZICK Y: "Ser/Thr phosphorylation of IRS proteins: a molecular basis for insulin resistance", SCI STKE, vol. 2005, 2005, pages E4
ZIMMET P; ALBERTI KGMM; SHAW J: "Global and social implications of the diabetic epidemic", NATURE, vol. 414, 2001, pages 782 - 787

Also Published As

Publication number Publication date
GB201002130D0 (en) 2010-03-31

Similar Documents

Publication Publication Date Title
Kalaany et al. LXRs regulate the balance between fat storage and oxidation
White Regulating insulin signaling and β-cell function through IRS proteins
Deshmukh Insulin-stimulated glucose uptake in healthy and insulin-resistant skeletal muscle
Tang et al. Amino acid-induced translation of TOP mRNAs is fully dependent on phosphatidylinositol 3-kinase-mediated signaling, is partially inhibited by rapamycin, and is independent of S6K1 and rpS6 phosphorylation
Aguilar et al. S6 kinase deletion suppresses muscle growth adaptations to nutrient availability by activating AMP kinase
Stone et al. Mechanisms of increased in vivo insulin sensitivity by dietary methionine restriction in mice
Hulver et al. Elevated stearoyl-CoA desaturase-1 expression in skeletal muscle contributes to abnormal fatty acid partitioning in obese humans
Ding et al. Adiponectin and its receptors are expressed in adult ventricular cardiomyocytes and upregulated by activation of peroxisome proliferator-activated receptor γ
Lin et al. Hyperlipidemic effects of dietary saturated fats mediated through PGC-1β coactivation of SREBP
Rodgers et al. Clk2 and B56β mediate insulin-regulated assembly of the PP2A phosphatase holoenzyme complex on Akt
Seo et al. Atf4 regulates obesity, glucose homeostasis, and energy expenditure
Skurk et al. The FOXO3a transcription factor regulates cardiac myocyte size downstream of AKT signaling
Oana et al. Physiological difference between obese (fa/fa) Zucker rats and lean Zucker rats concerning adiponectin
Corton et al. Peroxisome proliferator-activated receptor γ coactivator 1 in caloric restriction and other models of longevity
Grisouard et al. Mechanisms of metformin action on glucose transport and metabolism in human adipocytes
Gesty-Palmer et al. β-arrestin-selective G protein-coupled receptor agonists engender unique biological efficacy in vivo
Summermatter et al. Coordinated balancing of muscle oxidative metabolism through PGC-1α increases metabolic flexibility and preserves insulin sensitivity
Chehab Minireview: obesity and lipodystrophy—where do the circles intersect?
Sponarova et al. Involvement of AMP-activated protein kinase in fat depot-specific metabolic changes during starvation
Verhulst et al. Role of the AMP-activated protein kinase (AMPK) signaling pathway in the orexigenic effects of endogenous ghrelin
Cariou et al. Cellular and molecular mechanisms of adipose tissue plasticity in muscle insulin receptor knockout mice
Mok et al. Prokineticin receptor 1 ameliorates insulin resistance in skeletal muscle
Li et al. Identification of Sucrose Non-Fermenting–Related Kinase (SNRK) as a Suppressor of Adipocyte Inflammation
Muñoz et al. Physical exercise increases ROCK activity in the skeletal muscle of middle-aged rats
Lee et al. Muscle-specific activation of Ca 2+/calmodulin-dependent protein kinase IV increases whole-body insulin action in mice

Legal Events

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

Ref document number: 11709160

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 11709160

Country of ref document: EP

Kind code of ref document: A1