WO2019238934A1 - Use of the apolipoprotein m for the treatment and diagnosis of insulin resistance - Google Patents

Use of the apolipoprotein m for the treatment and diagnosis of insulin resistance Download PDF

Info

Publication number
WO2019238934A1
WO2019238934A1 PCT/EP2019/065701 EP2019065701W WO2019238934A1 WO 2019238934 A1 WO2019238934 A1 WO 2019238934A1 EP 2019065701 W EP2019065701 W EP 2019065701W WO 2019238934 A1 WO2019238934 A1 WO 2019238934A1
Authority
WO
WIPO (PCT)
Prior art keywords
apom
adipose tissue
expression
insulin resistance
insulin
Prior art date
Application number
PCT/EP2019/065701
Other languages
French (fr)
Inventor
Nathalie VIGUERIE
Sylvie CASPAR-BAUGUIL
Geneviève TAVERNIER
Dominique LANGIN
Original Assignee
INSERM (Institut National de la Santé et de la Recherche Médicale)
Université Paul Sabatier Toulouse Iii
Centre Hospitalier Universitaire De Toulouse
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 INSERM (Institut National de la Santé et de la Recherche Médicale), Université Paul Sabatier Toulouse Iii, Centre Hospitalier Universitaire De Toulouse filed Critical INSERM (Institut National de la Santé et de la Recherche Médicale)
Publication of WO2019238934A1 publication Critical patent/WO2019238934A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0012Lipids; Lipoproteins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/775Apolipopeptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/575Hormones
    • C07K14/62Insulins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/10011Adenoviridae
    • C12N2710/10311Mastadenovirus, e.g. human or simian adenoviruses
    • C12N2710/10341Use of virus, viral particle or viral elements as a vector
    • C12N2710/10343Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2750/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
    • C12N2750/00011Details
    • C12N2750/14011Parvoviridae
    • C12N2750/14111Dependovirus, e.g. adenoassociated viruses
    • C12N2750/14141Use of virus, viral particle or viral elements as a vector
    • C12N2750/14143Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector

Definitions

  • the present invention relates to the use of the apolipoprotein M for the treatment and diagnosis of insulin resistance.
  • Body fat is mainly adipose tissue which is one of the largest organs in the body.
  • Adipose tissue is a metabolically active organ that, besides having a major role in buffering excess energy, secretes a wide panel of factors with signaling functions in homeostasis and metabolism (4). Some of these factors are polypeptides and proteins synthesized and secreted by the adipocytes, and are thus called adipokines. These molecules act locally in an autocrine- paracrine way or in an endocrine manner. Dysfunctional adipose tissue is a hallmark of systemic insulin resistance and type 2 diabetes. The study of the adipose tissue secretome has led to the identification of hundreds of adipokines with beneficial or detrimental metabolic effects, most of them having been poorly investigated so far (5, 6).
  • novel adipokines are still being discovered.
  • apoM belongs to the lipocalins family (7).
  • S1P sphingosine-l -phosphate
  • retinol lipid metabolism and inflammation
  • ApoM-deficient mice lack HDL-associated S1P and display increased vascular permeability, which is a component of inflammatory response (10).
  • a link between the apoM/SlP axis and energy metabolism has been demonstrated in mice (11) but the role of the apoM itself in insulin resistance has never been investigated neither in humans nor in the adipose tissue.
  • the present invention relates to the use of the apolipoprotein M for the treatment and diagnosis of insulin resistance.
  • the present invention is defined by the claims.
  • the adipose tissue is a secretory organ producing a wide variety of factors that participate in the genesis of metabolic disorders linked to excess fat mass. Weight loss improves obesity related disorders.
  • Transcriptomic studies on human AT and a combination of analyses of transcriptome and proteome profiling of conditioned media from adipocytes and stromal cells isolated from human AT has led to the identification of apolipoprotein M (apoM) as a putative adipokine.
  • apoM apolipoprotein M
  • the inventors aimed to validate apoM as novel adipokine, investigate the relationship of AT APOM expression with metabolic syndrome and insulin sensitivity, and study the regulation of its expression in AT and secretion during calorie restriction induced weight loss.
  • APOM expression and secretion were measured during dietary interventions.
  • the inventors show that APOM is expressed in human subcutaneous and visceral AT, mainly by adipocytes. ApoM is released into circulation from AT and plasma apoM concentrations correlate with adipose tissue APOM mRNA levels.
  • APOM expression in AT is lower in obese compared to lean individuals and reduced in subjects with metabolic syndrome and type 2 diabetes. Regardless of fat depot, there is a positive relationship between AT APOM expression and systemic insulin sensitivity, independently of fat mass and plasma HDL.
  • APOM expression is enhanced by insulin sensitizing PPAR agonists and inhibited by TNFa, a cytokine which causes insulin resistance.
  • TNFa a cytokine which causes insulin resistance.
  • calorie restriction increased AT APOM expression and secretion.
  • the inventors show that overexpression of APOM in the AT reduces AT inflammation and improves local insulin sensitivity during high fat diet induced obesity.
  • the first object of the present invention relates to a method of treating insulin resistance in a patient in need thereof comprising administering to the patient a therapeutically effective amount of an ApoM polypeptide or a nucleic acid molecule encoding thereof
  • insulin resistance has its common meaning in the art. Insulin resistance is a physiological condition where the natural hormone insulin becomes less effective at lowering blood sugars. The resulting increase in blood glucose may raise levels outside the normal range and cause adverse health effects such as metabolic syndrome, dyslipidemia and subsequently type 2 diabetes mellitus. The method of the present invention is thus particularly suitable for the treatment of type 2 diabetes.
  • type 2 diabetes or“non insulin dependent diabetes mellitus (NIDDM)” has its general meaning in the art. Type 2 diabetes often occurs when levels of insulin are normal or even elevated and appears to result from the inability of tissues to respond appropriately to insulin. Most of the type 2 diabetics are obese.
  • the subject suffers from obesity.
  • obesity refers to a condition characterized by an excess of body fat.
  • the operational definition of obesity is based on the Body Mass Index (BMI), which is calculated as body weight per height in meter squared (kg/m 2 ).
  • BMI Body Mass Index
  • Obesity refers to a condition whereby an otherwise healthy subject has a BMI greater than or equal to 30 kg/m 2 , or a condition whereby a subject with at least one co-morbidity has a BMI greater than or equal to 27 kg/m 2 .
  • An “obese subject” is an otherwise healthy subject with a BMI greater than or equal to 30 kg/m 2 or a subject with at least one co-morbidity with a BMI greater than or equal 27 kg/m 2 .
  • a “subject at risk of obesity” is an otherwise healthy subject with a BMI of 25 kg/m 2 to less than 30 kg/m 2 or a subject with at least one co-morbidity with a BMI of 25 kg/m 2 to less than 27 kg/m 2 .
  • the increased risks associated with obesity may occur at a lower BMI in people of Asian descent.
  • “obesity” refers to a condition whereby a subject with at least one obesity-induced or obesity-related co-morbidity that requires weight reduction or that would be improved by weight reduction, has a BMI greater than or equal to 25 kg/m 2 .
  • An “obese subject” in these countries refers to a subject with at least one obesity- induced or obesity- related co-morbidity that requires weight reduction or that would be improved by weight reduction, with a BMI greater than or equal to 25 kg/m 2 .
  • a "subject at risk of obesity” is a person with a BMI of greater than 23 kg/m 2 to less than 25 kg/m 2 .
  • treatment refers to both prophylactic or preventive treatment as well as curative or disease modifying treatment, including treatment of patient at risk of contracting the disease or suspected to have contracted the disease as well as patients who are ill or have been diagnosed as suffering from a disease or medical condition, and includes suppression of clinical relapse.
  • the treatment may be administered to a subject having a medical disorder or who ultimately may acquire the disorder, in order to prevent, cure, delay the onset of, reduce the severity of, or ameliorate one or more symptoms of a disorder or recurring disorder, or in order to prolong the survival of a subject beyond that expected in the absence of such treatment.
  • therapeutic regimen is meant the pattern of treatment of an illness, e.g., the pattern of dosing used during therapy.
  • a therapeutic regimen may include an induction regimen and a maintenance regimen.
  • the phrase “induction regimen” or “induction period” refers to a therapeutic regimen (or the portion of a therapeutic regimen) that is used for the initial treatment of a disease.
  • the general goal of an induction regimen is to provide a high level of drug to a patient during the initial period of a treatment regimen.
  • An induction regimen may employ (in part or in whole) a "loading regimen", which may include administering a greater dose of the drug than a physician would employ during a maintenance regimen, administering a drug more frequently than a physician would administer the drug during a maintenance regimen, or both.
  • maintenance regimen refers to a therapeutic regimen (or the portion of a therapeutic regimen) that is used for the maintenance of a patient during treatment of an illness, e.g., to keep the patient in remission for long periods of time (months or years).
  • a maintenance regimen may employ continuous therapy (e.g., administering a drug at a regular intervals, e.g., weekly, monthly, yearly, etc.) or intermittent therapy (e.g., interrupted treatment, intermittent treatment, treatment at relapse, or treatment upon achievement of a particular predetermined criteria [e.g., disease manifestation, etc.]).
  • the method of the present invention is particularly suitable for improving blood glucose control, enhancing insulin signalling in skeletal muscle and adipose tissue, reducing lipotoxicity in skeletal muscle and adipose tissue, increasing lipid oxidative capacity in skeletal muscle and adipose tissue, or maintaining long-term insulin sensitivity in the subject.
  • ApoM has its general meaning in the art and refers to the apolipoprotein M encoded by the APOM gene (Gene ID: 55937).
  • ApoM is an apolipoprotein and member of the lipocalin protein family. The term is also known as G3A or NG20.
  • An exemplary amino acid sequence of ApoM is represented by SEQ ID NO:l and an exemplary nucleic acid sequence is represented by SEQ ID NO:2.
  • the ApoM polypeptide of the present invention comprises an amino acid sequence having at least 70% identity with the amino acid sequence as set forth in SEQ ID NO:l.
  • a first amino acid sequence having at least 70% identity with a second amino acid sequence means that the first sequence has 70; 71; 72; 73; 74; 75; 76; 77; 78; 79; 80; 81; 82; 83; 84; 85; 86; 87; 88; 89; 90; 91; 92; 93; 94; 95; 96; 97; 98; 99 or 100% identity with the second amino acid sequence.
  • Amino acid sequence identity is typically determined using a suitable sequence alignment algorithm and default parameters, such as BLAST P (Karlin and Altschul, 1990).
  • the polypeptide of the invention is produced by conventional automated peptide synthesis methods or by recombinant expression.
  • General principles for designing and making proteins are well known to those of skill in the art.
  • the polypeptides of the invention may be synthesized in solution or on a solid support in accordance with conventional techniques.
  • Various automatic synthesizers are commercially available and can be used in accordance with known protocols as described in Stewart and Young; Tam et al, 1983; Merrifield, 1986 and Barany and Merrifield, Gross and Meienhofer, 1979.
  • the polypeptides of the invention may also be synthesized by solid-phase technology employing an exemplary peptide synthesizer such as a Model 433A from Applied Biosystems Inc.
  • any given protein generated through automated peptide synthesis or through recombinant methods may be determined using reverse phase HPLC analysis. Chemical authenticity of each peptide may be established by any method well known to those of skill in the art.
  • recombinant DNA technology may be employed wherein a nucleotide sequence which encodes a protein of choice is inserted into an expression vector, transformed or transfected into an appropriate host cell and cultivated under conditions suitable for expression as described herein below. Recombinant methods are especially preferred for producing longer polypeptides.
  • a variety of expression vector/host systems may be utilized to contain and express the peptide or protein coding sequence.
  • microorganisms such as bacteria transformed with recombinant bacteriophage, plasmid or cosmid DNA expression vectors; yeast transformed with yeast expression vectors (Giga-Hama et al, 1999); insect cell systems infected with virus expression vectors (e.g., baculo virus, see Ghosh et al, 2002); plant cell systems transfected with virus expression vectors (e.g., cauliflower mosaic virus, CaMV; tobacco mosaic virus, TMV) or transformed with bacterial expression vectors (e.g., Ti or pBR322 plasmid; see e.g., Babe et al., 2000); or animal cell systems.
  • microorganisms such as bacteria transformed with recombinant bacteriophage, plasmid or cosmid DNA expression vectors; yeast transformed with yeast expression vectors (Giga-Hama et al, 1999); insect cell systems infected with virus expression vectors (e.g., baculo virus, see Ghosh e
  • Mammalian cells that are useful in recombinant protein productions include but are not limited to VERO cells, HeLa cells, Chinese hamster ovary (CHO) cell lines, COS cells (such as COS-7), W138, BHK, HepG2, 3T3, RIN, MDCK, A549, PC12, K562 and 293 cells.
  • Exemplary protocols for the recombinant expression of the peptide substrates or fusion polypeptides in bacteria, yeast and other invertebrates are known to those of skill in the art and a briefly described herein below.
  • Mammalian host systems for the expression of recombinant proteins also are well known to those of skill in the art.
  • Host cell strains may be chosen for a particular ability to process the expressed protein or produce certain post-translation modifications that will be useful in providing protein activity.
  • modifications of the polypeptide include, but are not limited to, acetylation, carboxylation, glycosylation, phosphorylation, lipidation and acylation.
  • Post- translational processing which cleaves a "prepro" form of the protein may also be important for correct insertion, folding and/or function.
  • Different host cells such as CHO, HeLa, MDCK, 293, WI38, and the like have specific cellular machinery and characteristic mechanisms for such post-translational activities and may be chosen to ensure the correct modification and processing of the introduced, foreign protein.
  • polypeptides used in the therapeutic methods of the present invention may be modified in order to improve their therapeutic efficacy.
  • modification of therapeutic compounds may be used to decrease toxicity, increase circulatory time, or modify biodistribution.
  • the toxicity of potentially important therapeutic compounds can be decreased significantly by combination with a variety of drug carrier vehicles that modify biodistribution.
  • adding dipeptides can improve the penetration of a circulating agent in the eye through the blood retinal barrier by using endogenous transporters.
  • a strategy for improving drug viability is the utilization of water- soluble polymers.
  • water-soluble polymers have been shown to modify biodistribution, improve the mode of cellular uptake, change the permeability through physiological barriers; and modify the rate of clearance from the body.
  • water-soluble polymers have been synthesized that contain drug moieties as terminal groups, as part of the backbone, or as pendent groups on the polymer chain.
  • Polyethylene glycol (PEG) has been widely used as a drug carrier, given its high degree of biocompatibility and ease of modification. Attachment to various drugs, proteins, and liposomes has been shown to improve residence time and decrease toxicity.
  • PEG can be coupled to active agents through the hydroxyl groups at the ends of the chain and via other chemical methods; however, PEG itself is limited to at most two active agents per molecule.
  • copolymers of PEG and amino acids were explored as novel biomaterials which would retain the biocompatibility properties of PEG, but which would have the added advantage of numerous attachment points per molecule (providing greater drug loading), and which could be synthetically designed to suit a variety of applications.
  • Those of skill in the art are aware of PEGylation techniques for the effective modification of drugs.
  • linkers may be used to maintain the therapeutic agent in a pro-drug form until released from the backbone polymer by a specific trigger, typically enzyme activity in the targeted tissue.
  • a specific trigger typically enzyme activity in the targeted tissue.
  • tissue activated drug delivery is particularly useful where delivery to a specific site of biodistribution is required and the therapeutic agent is released at or near the site of pathology.
  • Linking group libraries for use in activated drug delivery are known to those of skill in the art and may be based on enzyme kinetics, prevalence of active enzyme, and cleavage specificity of the selected disease-specific enzymes. Such linkers may be used in modifying the protein or fragment of the protein described herein for therapeutic delivery.
  • polypeptides of the invention may be fused to a heterologous polypeptide (i.e. polypeptide derived from an unrelated protein, for example, from an immunoglobulin protein).
  • a heterologous polypeptide i.e. polypeptide derived from an unrelated protein, for example, from an immunoglobulin protein.
  • nucleic acid molecule has its general meaning in the art and refers to a DNA or RNA molecule.
  • the term captures sequences that include any of the known base analogues of DNA and RNA such as, but not limited to 4-acetylcytosine, 8- hydroxy-N6-methyladenosine, aziridinylcytosine, pseudoisocytosine, 5- (carboxyhydroxylmethyl) uracil, 5-fmorouracil, 5-bromouracil, 5- carboxymethylaminomethyl-2-thiouracil, 5-carboxymethyl-aminomethyluracil, dihydrouracil, inosine, N6-isopentenyladenine, 1 -methyladenine, 1 -methylpseudouracil, l-methylguanine,
  • 2-thiocytosine 5-methyl-2-thiouracil, 2-thiouracil, 4-thiouracil, 5-methyluracil, -uracil-5- oxyacetic acid methylester, uracil-5 -oxyacetic acid, pseudouracil, queosine, 2-thiocytosine, and 2,6-diaminopurine.
  • the nucleic acid molecule of the present invention comprises a nucleic acid sequence having has at least 70% identity with the nucleic acid sequence as set forth in SEQ ID NO:2.
  • a first nucleic acid sequence having at least 70% identity with a second nucleic acid sequence means that the first sequence has 70; 71; 72; 73; 74; 75; 76; 77; 78; 79; 80; 81; 82; 83; 84; 85; 86; 87; 88; 89; 90; 91; 92; 93; 94; 95; 96; 97; 98; 99 or 100% identity with the second nucleic acid sequence.
  • the nucleic acid molecule of the present invention is included in a suitable vector.
  • the vector is a viral vector, and more particularly an adeno- associated virus (AAV), a retrovirus, bovine papilloma virus, an adenovirus vector, a lentiviral vector, a vaccinia virus, a polyoma virus, or an infective virus.
  • the vector is an AAV vector.
  • AAV vector means a vector derived from an adeno- associated virus serotype, including without limitation, AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, and mutated forms thereof.
  • AAV vectors can have one or more of the AAV wild-type genes deleted in whole or part, preferably the rep and/or cap genes, but retain functional flanking ITR sequences.
  • Retroviruses may be chosen as gene delivery vectors due to their ability to integrate their genes into the host genome, transferring a large amount of foreign genetic material, infecting a broad spectrum of species and cell types and for being packaged in special cell- lines.
  • a nucleic acid encoding a gene of interest is inserted into the viral genome in the place of certain viral sequences to produce a virus that is replication-defective.
  • a packaging cell line is constructed containing the gag, pol, and/or env genes but without the LTR and/or packaging components.
  • a recombinant plasmid containing a cDNA, together with the retroviral LTR and packaging sequences is introduced into this cell line (by calcium phosphate precipitation for example), the packaging sequence allows the RNA transcript of the recombinant plasmid to be packaged into viral particles, which are then secreted into the culture media.
  • the media containing the recombinant retroviruses is then collected, optionally concentrated, and used for gene transfer.
  • Retroviral vectors are able to infect a broad variety of cell types.
  • Lentiviruses are complex retroviruses, which, in addition to the common retroviral genes gag, pol, and env, contain other genes with regulatory or structural function. The higher complexity enables the virus to modulate its life cycle, as in the course of latent infection.
  • Some examples of lentivirus include the Human Immunodeficiency Viruses (HIV 1 , HIV 2) and the Simian Immunodeficiency Virus (SIV).
  • Lentiviral vectors have been generated by multiply attenuating the HIV virulence genes, for example, the genes env, vif, vpr, vpu and nef are deleted making the vector biologically safe. Lentiviral vectors are known in the art, see, e.g..
  • the vectors are plasmid-based or virus-based, and are configured to carry the essential sequences for incorporating foreign nucleic acid, for selection and for transfer of the nucleic acid into a host cell.
  • the gag, pol and env genes of the vectors of interest also are known in the art. Thus, the relevant genes are cloned into the selected vector and then used to transform the target cell of interest.
  • Recombinant lentivirus capable of infecting a non-dividing cell wherein a suitable host cell is transfected with two or more vectors carrying the packaging functions, namely gag, pol and env, as well as rev and tat is described in U.S. Pat. No. 5,994,136, incorporated herein by reference.
  • This describes a first vector that can provide a nucleic acid encoding a viral gag and a pol gene and another vector that can provide a nucleic acid encoding a viral env to produce a packaging cell.
  • Introducing a vector providing a heterologous gene into that packaging cell yields a producer cell which releases infectious viral particles carrying the foreign gene of interest.
  • the env preferably is an amphotropic envelope protein which allows transduction of cells of human and other species.
  • the nucleic acid molecule or the vector of the present invention include "control sequences'", which refers collectively to promoter sequences, polyadenylation signals, transcription termination sequences, upstream regulatory domains, origins of replication, internal ribosome entry sites ("IRES"), enhancers, and the like, which collectively provide for the replication, transcription and translation of a coding sequence in a recipient cell. Not all of these control sequences need always be present so long as the selected coding sequence is capable of being replicated, transcribed and translated in an appropriate host cell.
  • nucleic acid sequence is a "promoter” sequence, which is used herein in its ordinary sense to refer to a nucleotide region comprising a DNA regulatory sequence, wherein the regulatory sequence is derived from a gene which is capable of binding R A polymerase and initiating transcription of a downstream (3’-direction) coding sequence.
  • Transcription promoters can include "inducible promoters” (where expression of a polynucleotide sequence operably linked to the promoter is induced by an analyte, cofactor, regulatory protein, etc.), “repressible promoters” (where expression of a polynucleotide sequence operably linked to the promoter is induced by an analyte, cofactor, regulatory protein, etc.), and “constitutive promoters”.
  • a “therapeutically effective amount” is meant a sufficient amount of the polypeptide or the nucleic acid molecule encoding thereof to prevent for use in a method for the treatment of the disease (e.g. haemophilia) at a reasonable benefit/risk ratio applicable to any medical treatment. It will be understood that the total daily usage of the compounds and compositions of the present invention will be decided by the attending physician within the scope of sound medical judgment.
  • the specific therapeutically effective dose level for any particular subject will depend upon a variety of factors including the age, body weight, general health, sex and diet of the subject; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidental with the specific polypeptide employed; and like factors well known in the medical arts. For example, it is well known within the skill of the art to start doses of the compound at levels lower than those required to achieve the desired therapeutic effect and to gradually increase the dosage until the desired effect is achieved. However, the daily dosage of the products may be varied over a wide range from 0.01 to 1,000 mg per adult per day.
  • the compositions contain 0.01, 0.05, 0.1, 0.5, 1.0, 2.5, 5.0, 10.0, 15.0, 25.0, 50.0, 100, 250 and 500 mg of the active ingredient for the symptomatic adjustment of the dosage to the subject to be treated.
  • a medicament typically contains from about 0.01 mg to about 500 mg of the active ingredient, preferably from 1 mg to about 100 mg of the active ingredient.
  • An effective amount of the drug is ordinarily supplied at a dosage level from 0.0002 mg/kg to about 20 mg/kg of body weight per day, especially from about 0.001 mg/kg to 7 mg/kg of body weight per day.
  • the polypeptide or the nucleic acid molecule (inserted or not into a vector) of the present invention is administered to the subject in the form of a pharmaceutical composition.
  • the polypeptide or the nucleic acid molecule (inserted or not into a vector) of the present invention may be combined with pharmaceutically acceptable excipients, and optionally sustained-release matrices, such as biodegradable polymers, to form pharmaceutical compositions.
  • pharmaceutically acceptable excipients such as biodegradable polymers
  • a pharmaceutically acceptable carrier or excipient refers to a non-toxic solid, semi-solid or liquid filler, diluent, encapsulating material or formulation auxiliary of any type.
  • the active principle alone or in combination with another active principle, can be administered in a unit administration form, as a mixture with conventional pharmaceutical supports, to animals and human beings.
  • Suitable unit administration forms comprise oral-route forms such as tablets, gel capsules, powders, granules and oral suspensions or solutions, sublingual and buccal administration forms, aerosols, implants, subcutaneous, transdermal, topical, intraperitoneal, intramuscular, intravenous, subdermal, transdermal, intrathecal and intranasal administration forms and rectal administration forms.
  • the pharmaceutical compositions contain vehicles which are pharmaceutically acceptable for a formulation capable of being injected.
  • saline solutions monosodium or disodium phosphate, sodium, potassium, calcium or magnesium chloride and the like or mixtures of such salts
  • dry, especially freeze-dried compositions which upon addition, depending on the case, of sterilized water or physiological saline, permit the constitution of injectable solutions.
  • the pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions; formulations including sesame oil, peanut oil or aqueous propylene glycol; and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. In all cases, the form must be sterile and must be fluid to the extent that easy syringability exists.
  • Solutions comprising compounds of the invention as free base or pharmacologically acceptable salts can be prepared in water suitably mixed with a surfactant, such as hydroxypropylcellulose. Dispersions can also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms.
  • the polypeptide or the nucleic acid molecule (inserted or not into a vector) of the present invention can be formulated into a composition in a neutral or salt form.
  • Pharmaceutically acceptable salts include the acid addition salts (formed with the free amino groups of the protein) and which are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like. Salts formed with the free carboxyl groups can also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, histidine, procaine and the like.
  • inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like.
  • Salts formed with the free carboxyl groups can also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine,
  • the carrier can also be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetables oils.
  • the proper fluidity can be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
  • the prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like.
  • isotonic agents for example, sugars or sodium chloride.
  • Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminium monostearate and gelatin.
  • Sterile injectable solutions are prepared by incorporating the active compounds in the required amount in the appropriate solvent with several of the other ingredients enumerated above, as required, followed by filtered sterilization.
  • dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above.
  • sterile powders for the preparation of sterile injectable solutions the typical methods of preparation are vacuum-drying and freeze-drying techniques which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • the preparation of more, or highly concentrated solutions for direct injection is also contemplated, where the use of DMSO as solvent is envisioned to result in extremely rapid penetration, delivering high concentrations of the active agents to a small tumor area.
  • solutions Upon formulation, solutions will be administered in a manner compatible with the dosage formulation and in such amount as is therapeutically effective.
  • the formulations are easily administered in a variety of dosage forms, such as the type of injectable solutions described above, but drug release capsules and the like can also be employed.
  • aqueous solutions For parenteral administration in an aqueous solution, for example, the solution should be suitably buffered if necessary and the liquid diluent first rendered isotonic with sufficient saline or glucose.
  • aqueous solutions are especially suitable for intravenous, intramuscular, subcutaneous and intraperitoneal administration.
  • sterile aqueous media which can be employed will be known to those of skill in the art in light of the present disclosure. Some variation in dosage will necessarily occur depending on the condition of the subject being treated. The person responsible for administration will, in any event, determine the appropriate dose for the individual subject.
  • a further object of the present invention relates to a method of determining whether a subject has or is at risk of having insulin resistance comprising i) determining the expression level of ApoM in a biological sample obtained from the subject and ii) comparing the expression level determined at step i) with a predetermined reference value wherein detecting differential between the expression level determined at step i) with the predetermined reference value indicated whether the subject has or is at risk of having insulin resistance.
  • cardiometabolic disease has its general meaning in the art and relates to cardiovascular diseases associated with metabolic syndrome, such as obesity, diabetes/insulin resistance, hypertension and dyslipidemia.
  • cardiovascular diseases refers to cardiac consequences of metabolic syndrome such as atherosclerosis, coronary heart disease, obesity- associated heart disease, insulin resistance- associated heart disease, hypertensive heart disease, cardiac remodeling, heart failure and cardiometabolic diseases disclosed in Hertle et al, 2014; Hua and Nair, 2014; U.S. Pat. Application No. 2012/0214771 and International Patent Application No. 2008/094939.
  • risk in the context of the present invention, relates to the probability that an event will occur over a specific time period and can mean a subject's "absolute” risk or “relative” risk.
  • Absolute risk can be measured with reference to either actual observation post-measurement for the relevant time cohort, or with reference to index values developed from statistically valid historical cohorts that have been followed for the relevant time period.
  • Relative risk refers to the ratio of absolute risks of a subject compared either to the absolute risks of low risk cohorts or an average population risk, which can vary by how clinical risk factors are assessed.
  • Odds ratios the proportion of positive events to negative events for a given test result, are also commonly used (odds are according to the formula p/(l-p) where p is the probability of event and (1- p) is the probability of no event) to no- conversion.
  • "Risk evaluation,” or “evaluation of risk” in the context of the present invention encompasses making a prediction of the probability, odds, or likelihood that an event or disease state may occur, the rate of occurrence of the event or conversion from one disease state to another. Risk evaluation can also comprise prediction of future clinical parameters, traditional laboratory risk factor values, or other indices of relapse, either in absolute or relative terms in reference to a previously measured population.
  • the methods of the present invention may be used to make continuous or categorical measurements of the risk of conversion, thus diagnosing and defining the risk spectrum of a category of subjects defined as being at risk of conversion.
  • the invention can be used to discriminate between normal and other subject cohorts at higher risk.
  • the present invention may be used so as to discriminate those at risk from normal.
  • the biological sample is a blood sample.
  • blood sample means any blood sample derived from the subject. Collections of blood samples can be performed by methods well known to those skilled in the art.
  • the blood sample is a serum sample or a plasma sample.
  • the measurement of the level of ApoM in the blood sample is typically carried out using standard protocols known in the art.
  • the method may comprise contacting the blood sample with a binding partner capable of selectively interacting with ApoM in the sample.
  • the binding partners are antibodies, such as, for example, monoclonal antibodies or even aptamers.
  • the binding may be detected through use of a competitive immunoassay, a non-competitive assay system using techniques such as western blots, a radioimmunoassay, an ELISA (enzyme linked immunosorbent assay), a“sandwich” immunoassay, an immunoprecipitation assay, a precipitin reaction, a gel diffusion precipitin reaction, an immunodiffusion assay, an agglutination assay, a complement fixation assay, an immunoradiometric assay, a fluorescent immunoassay, a protein A immunoassay, an immunoprecipitation assay, an immunohistochemical assay, a competition or sandwich ELISA, a radioimmunoassay, a Western blot assay, an immunohistological assay, an immunocytochemical assay, a dot blot assay, a fluorescence polarization assay, a scintillation proximity assay, a homogeneous time resolved fluorescence
  • the aforementioned assays generally involve the binding of the partner (ie. antibody or aptamer) to a solid support.
  • Solid supports which can be used in the practice of the invention include substrates such as nitrocellulose (e.g., in membrane or microtiter well form); polyvinylchloride (e.g., sheets or microtiter wells); polystyrene latex (e.g., beads or microtiter plates); polyvinylidine fluoride; diazotized paper; nylon membranes; activated beads, magnetically responsive beads, and the like.
  • An exemplary biochemical test for identifying specific proteins employs a standardized test format, such as ELISA test, although the information provided herein may apply to the development of other biochemical or diagnostic tests and is not limited to the development of an ELISA test (see, e.g., Molecular Immunology: A Textbook, edited by Atassi et al. Marcel Dekker Inc., New York and Basel 1984, for a description of ELISA tests). Therefore ELISA method can be used, wherein the wells of a microtiter plate are coated with a set of antibodies which recognize ApoM. A sample containing or suspected of containing ApoM is then added to the coated wells.
  • the plate(s) can be washed to remove unbound moieties and a detectably labelled secondary binding molecule added.
  • the secondary binding molecule is allowed to react with any captured sample marker protein, the plate washed and the presence of the secondary binding molecule detected using methods well known in the art.
  • Measuring the level of ApoM may also include separation of the compounds: centrifugation based on the compound’s molecular weight; electrophoresis based on mass and charge; HPLC based on hydrophobicity; size exclusion chromatography based on size; and solid-phase affinity based on the compound's affinity for the particular solid-phase that is used.
  • said one or two biomarkers proteins may be identified based on the known "separation profile" e.g., retention time, for that compound and measured using standard techniques.
  • the separated compounds may be detected and measured by, for example, a mass spectrometer.
  • levels of immunoreactive ApoM in a sample may be measured by an immunometric assay on the basis of a double-antibody "sandwich” technique, with a monoclonal antibody specific for ApoM.
  • said means for measuring ApoM level are for example i) a ApoM buffer, ii) a monoclonal antibody that interacts specifically with ApoM, iii) an enzyme- conjugated antibody specific for ApoM and a predetermined reference value of ApoM.
  • the predetermined reference value is a threshold value or a cut off value.
  • a “threshold value” or “cut-off value” can be determined experimentally, empirically, or theoretically.
  • a threshold value can also be arbitrarily selected based upon the existing experimental and/or clinical conditions, as would be recognized by a person of ordinary skilled in the art. For example, retrospective measurement of expression level of the gene in properly banked historical patient samples may be used in establishing the predetermined reference value. The threshold value has to be determined in order to obtain the optimal sensitivity and specificity according to the function of the test and the benefit/risk balance (clinical consequences of false positive and false negative).
  • the optimal sensitivity and specificity can be determined using a Receiver Operating Characteristic (ROC) curve based on experimental data.
  • ROC Receiver Operating Characteristic
  • the full name of ROC curve is receiver operator characteristic curve, which is also known as receiver operation characteristic curve. It is mainly used for clinical biochemical diagnostic tests.
  • ROC curve is a comprehensive indicator that reflects the continuous variables of true positive rate (sensitivity) and false positive rate (1 -specificity). It reveals the relationship between sensitivity and specificity with the image composition method.
  • a series of different cut-off values are set as continuous variables to calculate a series of sensitivity and specificity values. Then sensitivity is used as the vertical coordinate and specificity is used as the horizontal coordinate to draw a curve. The higher the area under the curve (AUC), the higher the accuracy of diagnosis.
  • AUC area under the curve
  • the point closest to the far upper left of the coordinate diagram is a critical point having both high sensitivity and high specificity values.
  • the AUC value of the ROC curve is between 1.0 and 0.5. When AUC>0.5, the diagnostic result gets better and better as AUC approaches 1. When AUC is between 0.5 and 0.7, the accuracy is low. When AUC is between 0.7 and 0.9, the accuracy is moderate.
  • the predetermined reference value is the expression level of ApoM determined in a population of healthy individuals. Typically, it is concluded that the subject has or is at risk of having insulin resistance when the expression level of is at least 0.5, 1, 1.5, 2, 2.5, 3, 3.5, 4, 5, 6, 7, 8, 9, 10, 15, 20, 30, 40, 50, 100 fold lower than the expression level determined in a population of healthy individuals.
  • FIGURES are a diagrammatic representation of FIGURES.
  • Figure 1 The apolipoprotein M in human adipose tissue.
  • A. ApoM concentration was measured in paired samples of arterial and venous plasma samples from obese women. Data are plotted as the ratio between venous and arterial blood (AV) for each of the women (cohort A, n 8). *, P ⁇ 0.05; from Wilcoxon test comparing venous to arterial blood.
  • B. APOM mRNA level was measured in paired samples (n l2) of freshly isolated adipocytes and stroma-vascular fraction (SVF) from human subcutaneous abdominal adipose tissue. Data are presented as mean ⁇ SEM. ***, P ⁇ 0.001; *, P ⁇ 0.05 from Wilcoxon test.
  • Figure 3 Human APOM mRNA level in subcutaneous adipose tissue according to the presence of metabolic syndrome.
  • B Linear regression analysis of subcutaneous adipose tissue APOM expression and waist circumference (B), plasma HDL- cholesterol (C) and plasma triglycerides (D) with correlation coefficient and adjusted p-values according to Benj amini-Hochberg is displayed in each graph.
  • APOM mRNA level was measured in subcutaneous adipose tissue of 314 obese individuals (cohort E) before (baseline) and after an 8-week very low-calorie diet (VLCD). Data are presented as mean ⁇ SEM. ***, P ⁇ 0.001; from Wilcoxon test.
  • FIG. 6 Effect of human APOM overexpression in the perigonadic adipose tissue in high fat fed mice.
  • B Effect of adenovirus transduction on body weight.
  • Perigonadic adipose tissue was injected with AAVs (5 x 10 L 10 particles/fat pad) containing human APOM (3), or mCherry (2), or with NaCl 0.9% (1).
  • Gene expression for interleukinl (II- 1 b), macrophage chemoattractant protein (M cpl) and tumor necrosis factor (Jnf-a) was measured in perigonadic adipose tissue.
  • FIG. 7 Effect of AAVs transduction of h APOM in perigonadic adipose tissue on phosphorylation of Akt signaling.
  • Perigonadic adipose tissue was incubated ex vivo with 100 nM insulin (black bars), or vehicle (white bars) in Krebs Ringer Hepes supplemented with 0.2% bovine serum albumin for 20 min at 37°C. Proteins were extracted from tissue lysates and quantified according to Bradford. Fifteen pg proteins were separated by SDS-PAGE and after transfer to nitrocellulose membranes were immunoblotted using antibodies raised against Ser(P)-Akt 473 or total- Akt.
  • EXAMPLE 1 apolipoprotein M: a novel adipokine decreasing with obesity and upregulated by calorie restriction.
  • Cohort A included 8 obese women (BMI 33.9 ⁇ 0.1 kg/m 2 ) with stable weight during the 3 months preceding the study. As previously described in (12), exclusion criteria were hypertension, diabetes, hyperlipidemia, or any drug treatment.
  • Cohort B comprised 56 women scheduled to have abdominal surgery (laparoscopic or laparotomic cholecystectomy, hysterectomy and gastric banding) at the Departments of Surgery and Gynecology at Kralovske Vinohrady Faculty Hospital in Prague.
  • BMI ranged from 17.3 to
  • Cohort C included 14 obese (BMI 34.1 ⁇ 4.7 kg/m2) women with or without polycystic ovary syndrome (PCOS) recruited during 2004-2005.
  • Anthropometric measurements, blood sampling, euglycemic-hyperinsulinemic clamp and subcutaneous abdominal adipose tissue needle biopsies were performed as described in (14).
  • Cohort D included 10 metabolically healthy overweight and obese women (BMI 34.1 ⁇
  • Cohort E included a subgroup of 336 (111 men and 225 women) overweight and obese individuals (BMI 34.1 ⁇ 4.7 kg/m2) from the DiOGenes Study in 8 European countries (16). Individuals followed a VLCD for 8 weeks, and those that lost at least 8% of their baseline weight were randomized to one of five ad libitum follow-up diets for 6 months. Clinical measures and subcutaneous abdominal adipose tissue biopsies were taken at each time-points. The participants were instructed not to change their habitual activity during the study. Only baseline and post-VLCD fat samples and clinical data were investigated in the present study.
  • the risk score for metabolic syndrome was calculated according to (19).
  • the quantitative insulin-sensitivity check index (QUICKI), calculated as l/[log(I) + log(G)], where I is fasting insulin (pun its/m L) and G is fasting glucose (mg/dL), was used since it has been reported to be more reproducible and has a better correlation to the reference glucose clamp method than other surrogates including HOMA-IR, especially for longitudinal studies (20).
  • NanoLC/ESI LTQ-Orbitrap MS/MS was used for analysis of 24h culture media of the different human adipose tissue cell types. Details are reported in (23).
  • microarray assays were performed using the Agilent Whole Human Genome Microarray 4x44K v2 according to manufacturer’s recommendations (Agilent Technologies) and analysed as described in (21).
  • Subcutaneous adipose tissue catheterization To measure arteriovenous differences across subcutaneous abdominal adipose tissue, venous and arterial blood were sampled in 8 obese women (BMI 33.9 ⁇ 0.1 kg/m 2 ) as described in (12) (cohort A).
  • the Human multipotent adipose-derived stem cell (hMADS) cells were cultured and differentiated as previously described (24). Briefly, after six days of proliferation, the cells were incubated in the differentiation medium (serum- free DMEM low glucose/ Ham’s F-12 medium containing l0pg/ml of transferrin, 5pg/ml of insulin, 0.2 nM triiodothyronine, 100mM 3- isobutyl-l-methylxanthine, 1 mM dexamethasone, and 100 nM rosiglitazone). Three days after (day3), dexamethasone and 3 -isobutyl- l-methylxanthine were omitted from the medium.
  • the differentiation medium serum- free DMEM low glucose/ Ham’s F-12 medium containing l0pg/ml of transferrin, 5pg/ml of insulin, 0.2 nM triiodothyronine, 100mM
  • Rosiglitazone was omitted at day 9. By day 13, cells were fully differentiated into adipocytes. At day 14, cells were treated with 100 nM of the PPARy agonist rosiglitazone (Alexis Biochemicals), or 300 nM of the PPARa agonist GW7647 (Sigma), or 10 ng/ml TNFa (Sigma), or the vehicle, for 4 days.
  • PPARy agonist rosiglitazone Alexis Biochemicals
  • 300 nM of the PPARa agonist GW7647 Sigma
  • 10 ng/ml TNFa 10 ng/ml TNFa
  • Adipose tissue samples of about 400 mg obtained from 10 obese women were cut into small pieces and incubated at 37°C for 4h in 4ml of Krebs/Ringer phosphate buffer supplemented with lg/F glucose and 20g/F bovine serum albumin as described in (15) (cohort D).
  • Real-time PCR was performed in duplicate using a set of TaqMan (Thermo Fisher Scientific) probes and primers assays (Hs002l9533_ml). 20 ng cDNA was analyzed on a StepOne Plus Real-Time PCR system (Applied Biosystems, Carlsbad, CA) or, for longitudinal dietary interventions, using the Bio markTM HD system (BioMark) as described in (21). Adipose tissue and cell data were normalized to GUSB (Hs00939627_ml) or LRP10 (Hs0l047362_ml), respectively.
  • GUSB Hs00939627_ml
  • LRP10 Hs0l047362_ml
  • ApoM protein level was measured in duplicate using a human apoM EFISA kit (E-EF- H0473, Elabscience, Clinisciences, Nanterre, France), following manufacturer’s instructions. ApoM concentration was calculated using sigmoidal standard curve fitted by nonlinear regression analysis for each test.
  • Gaussian distribution was tested using the D'Agostino & Pearson test. For comparing data from two groups, where a normal distribution was unattainable, a Wilcoxon or Mann- Whitney test was employed for paired or unpaired comparisons respectively. For a comparison using data from more than two groups, ANOVA was performed with Dunnett’s post-hoc test or Kruskal- Wallis with Dunn’s post-hoc test for non-normal data distribution. Linear regression was performed with adipose tissue APOM expression as the dependent variable and the Benjamini-Hochberg procedure was applied to account for multiple testing. Multiple regression was performed with all variables significantly correlated with adipose tissue APOM expression. Partial correlations were computed to assess direct correlations independently of other confounding variables.
  • APOM is released by human adipose tissue and secreted by adipocytes.
  • the adipose tissue contains different cell types including preadipocytes and immune, endothelial and progenitor cells corresponding to the stroma vascular fraction.
  • preadipocytes and immune, endothelial and progenitor cells corresponding to the stroma vascular fraction.
  • a comparative proteomic analysis of the proteins secreted by these 5 distinct human adipose tissue cell populations led to the identification of 626 proteins secreted by adipocytes, 123 of which have never been described in the literature as produced by the adipose tissue and were found specifically in the media of mature adipocytes.
  • We compared this list of potentially new adipokines with the data of the transcriptome analyses on the same different adipose tissue cellular fractions.
  • APOM expression and secretion were compared between isolated adipocytes and stroma vascular fractions. Compared to the stroma vascular fraction of adipose tissue, APOM was expressed ( Figure IB) and secreted ( Figure 1C) mainly by adipocytes.
  • cohort B there was a positive relationship between plasma apoM and subcutaneous adipose tissue APOM expression (cohort B, data not shown).
  • APOM expression increased in a time-dependent manner (data not shown). This cell line was used to investigate the effect of prominent regulators of adipocyte secretory activities (25).
  • APOM mRNA level was reduced by TNFa treatment with ( Figure ID) and enhanced by a treatment with PPAR agonists (PPARy, rosiglitazone; PPARa, GW 7647)
  • APOM expression was analysed in paired samples of subcutaneous and visceral adipose tissue from lean and obese women (cohort B). Irrespective of the fat depot, a higher APOM expression was found in lean compared to obese individuals ( Figure 2B). As previously reported (26), plasma apoM was lower in overweight and obese compared to lean women (data not shown). Moreover, a strong negative correlation between body fat and APOM expression in both subcutaneous and visceral adipose tissue was found (cohort B, Figure 2C).
  • Low adipose tissue APOM expression is associated with a dysmetabolic phenotype.
  • Obesity is an important risk factor for metabolic syndrome as the enlargement of fat mass impacts adipocyte biology. Adipocyte size is linked to metabolic syndrome (27).
  • Figure 3A shows that subcutaneous adipose tissue characterized by large fat cells (hypertrophy) displayed lower APOM expression than fat from subjects matched for BMI but with many small adipocytes (hyperplasia), irrespective of body weight status (cohort F). A negative correlation between adipose tissue APOM expression and adipocyte volume was found accordingly (data not shown).
  • Subcutaneous adipose tissue APOM expression was lower in subjects with type 2 diabetes and prediabetes (impaired fasting glucose and/or impaired glucose tolerance, assessed as in (28)) compared to normal glucose-tolerant individuals (cohort E, Figure 4A).
  • apoM expression and plasma levels are associated with obesity and comorbidities developed in obesity, we aimed to study if a weight-reducing diet which improves metabolic profile could influence the levels of apoM expression and production.
  • VLCD The secretion of apoM by subcutaneous adipose tissue explants was assessed in obese women during a 1 -month VLCD (cohort D).
  • the VLCD induced weight and body fat loss and an improvement in insulin sensitivity.
  • ApoM concentration in media from adipose tissue explants significantly increased (+22%) at the end of the VLCD ( Figure 5B).
  • a combination of microarray studies and proteomic profiling of conditioned media from freshly isolated adipocytes and cell populations in the stroma vascular fraction from human adipose tissue has led to the identification of apoM as a novel adipokine.
  • the present study shows that the human adipose tissue is a site of expression of the APOM gene and that adipose tissue and predominantly adipocytes do secrete apoM.
  • apoM With reference to other apolipoproteins, the apoM, which was identified in 1999 (33), is comparatively recent. The apoM has been mainly studied in liver where it is produced in hepatocytes; also, kidney proximal tubule cells express apoM (7). Here, we show that apoM is released from adipose tissue in vivo and ex vivo, particularly from adipocytes. The apoM concentration in media from adipose tissue explants in culture is comparable to other adipokines, such as leptin or IL6 (15).
  • adipokines such as leptin or IL6
  • Weight gain in adults is accompanied by an increase in adipocyte size.
  • Adipocytes of obese subjects are dysfunctional, especially regarding metabolism and secretory functions (34).
  • Large adipocytes were reported to be associated with metabolic disease, insulin resistance and type 2 diabetes in multiple studies (35).
  • APOM mRNA level was reported to be associated with metabolic disease, insulin resistance and type 2 diabetes in multiple studies (35).
  • APOM mRNA level was associated with metabolic disease, insulin resistance and type 2 diabetes in multiple studies (35).
  • hypertrophic adipose tissue containing few but large adipocytes, displayed lower APOM expression compared to hyperplastic tissue that includes more adipocytes but with smaller size.
  • the APOM gene expression thus seems to be a hallmark of healthy adipose tissue.
  • Unhealthy adipose tissue deficient in beneficial adipokines such as adiponectin and producing excess of harmful factors, is one of the features of metabolic syndrome and diseases such as insulin resistance and type 2 diabetes.
  • the present study has shown that, in contrast to most adipokines (5, 21) and similarly to adiponectin (37), APOM gene expression in adipose tissue was lower with increasing adiposity. This trend is in accordance with the plasma concentration of apoM lower in obese compared to lean individuals, as previously reported (26).
  • APOM mRNA level is lower in adipose tissue obtained from subjects with metabolic syndrome, independently of fat mass, and that adipose tissue APOM mRNA level is associated with several metabolic syndrome features (waist circumference, plasma HDF and triglycerides).
  • plasma apoM has been described as a component of the pre-b HDL (8) and reduced with metabolic syndrome (38), in accordance with the present report.
  • adipose tissue APOM mRNA level is reduced with prediabetes and diabetes. More evidence is provided by another independent longitudinal study where lean men were investigated before and after a 4-d treatment with the glucocorticoid analogue, dexamethasone. The treatment induced insulin resistance without change in fat mass (42).
  • the adipose tissue microarray data included APOM and showed that glucocorticoid exposure downregulated (mean fold change 0.6) APOM gene expression in subcutaneous fat
  • apoM was reported as a carrier of several anti-inflammatory lipids (7).
  • apoM/SlP protects against atherosclerosis (9) and controls the metabolic activity of brown fat (11).
  • the ability of apoM to bind anti-inflammatory factors could convey a local effect on the inflammatory status of the adipose tissue. Though, this highlights apoM as a good candidate for monitoring healthy adipose tissue status.
  • the present study reports apoM production by adipocytes for the first time. It shows that APOM mRNA level is reduced in adipose tissue from obese subjects and independently of body fat mass, is also lower in individuals with insulin resistance, type 2 diabetes or dyslipidemia. Patterns of expression and secretion in adipose tissue indicate that apoM is also regulated in response to calorie-restriction induced weight loss.
  • apoM is a new adipokine, which may reflect changes in adipose tissue mass and whole body metabolic dysfunction, and which presence in the white adipose tissue is positively associated with insulin sensitivity.
  • EXAMPLE 2 Effect of human APOM overexpression in the perigonadic adipose tissue in high fat fed mice.
  • mice fed ad libitum were randomized in 3 groups of 10 mice according to body weight and glucose tolerance tests (lmg glucose/g body weight, intraperitoneal). Mice were housed at room temperature and manipulated according to Inserm guidelines and European Directive 2010/63/UE in the local animal care facility (agreements A 31 555 04 and C 31 555 07). Mice were fed with a 60% high fat diet for 4 weeks.
  • mice were anaesthetized and perigonadic adipose tissues (PG-AT) were injected with recombinant adenoviruses (2xl0 9 particles/fat pad) containing coding sequences of human APOM (ad-hAPOM) or green fluorescent protein (ad-GFP), or with saline (sham) or red fluorescent protein (mCherry).
  • ad-hAPOM human APOM
  • ad-GFP green fluorescent protein
  • saline saline
  • mCherry red fluorescent protein
  • RNA samples were snap frozen and stored at -80°C until total RNA extraction.
  • insulin 100 nM
  • vehicle in Krebs Ringer Hepes supplemented with 0.2% bovine serum albumin.
  • tissue samples were rinced, snap frozen and stored at -80°C until protein extraction.
  • perigonadic adipose tissue response to insulin was investigated ex vivo (20 min. insulin at 100 nM vs control) with the subsequent study of the Akt signaling pathway which full activation requires phosphorylation of S473 (Akt-P473).
  • inflammatory factors interleukin 1 (II- 1 b), macrophage chemoattractant protein (Mcpl) and tumor necrosis factor (Tnf-a)
  • II- 1 b interleukin 1
  • Mcpl macrophage chemoattractant protein
  • Tnf-a tumor necrosis factor
  • TNF-a tumor necrosis factor
  • IL-10 interleukin- 10
  • adipose tissue secretes high levels of proinflammatory adipokines such as I L- 1 b, monocyte chemoattractant protein 1 (MCP-l), and TNF-a.
  • proinflammatory adipokines such as I L- 1 b, monocyte chemoattractant protein 1 (MCP-l), and TNF-a.
  • MCP-l monocyte chemoattractant protein 1
  • TNF-a TNF-a
  • the present study shows that overexpression of APOM in the AT reduces AT inflammation and improves local insulin sensitivity during high fat diet induced obesity.
  • TGFbeta family members are key mediators in the induction of myofibroblast phenotype of human adipose tissue progenitor cells by macrophages. PFoS One 20l2;7:e3 l274. doi: 10. l37l/joumal.pone.0031274.
  • Semaphorin 3C is a novel adipokine linked to extracellular matrix composition. Diabetologia 2013;56: 1792-801. doi: 10.1007/s00125-013-293 l-z.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Diabetes (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Biophysics (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Molecular Biology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Zoology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Emergency Medicine (AREA)
  • Endocrinology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Hematology (AREA)
  • Obesity (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Toxicology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biochemistry (AREA)
  • Genetics & Genomics (AREA)
  • Immunology (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Epidemiology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The inventors examined APOM mRNA level and secretion in AT from patients in 6 independent clinical trials and in vitro in hMADS adipocytes. APOM expression and secretion were measured during dietary interventions. The inventors show that APOM is expressed in human subcutaneous and visceral AT and is released into circulation from AT. APOM expression in AT is lower in obese compared to lean individuals and reduced in subjects with metabolic syndrome and type 2 diabetes. Regardless of fat depot, there is a positive relationship between AT APOM expression and systemic insulin sensitivity. Finally, the inventors show that overexpression of APOM in the AT reduces AT inflammation and improves local insulin sensitivity during high fat diet induced obesity. Thus, the present invention relates to the present invention relates to the use of the apolipoprotein M for the treatment and diagnosis of insulin resistance.

Description

USE OF THE APOLIPOPROTEIN M FOR THE TREATMENT AND DIAGNOSIS
OF INSULIN RESISTANCE
FIELD OF THE INVENTION:
The present invention relates to the use of the apolipoprotein M for the treatment and diagnosis of insulin resistance.
BACKGROUND OF THE INVENTION:
Excess fat mass leads to being overweight or obese, which has deleterious health consequences. The development of obesity is still increasing worldwide (1). Likewise, metabolic syndrome, which is a collection of obesity-associated disorders, is associated with development of cardiovascular diseases, insulin resistance, hepatic steatosis, certain types of cancer and type 2 diabetes (2) (3). Resistance of peripheral tissues (liver, muscle, adipose tissue) to insulin action is indeed a key event in diabetes onset, so that therapeutic strategies aiming at restoring insulin sensitivity are highly relevant. However, they essentially remain unsatisfactory. For instance, thiazolidinediones, by binding peroxisome proliferator activated receptor gamma, increase insulin sensitivity, but such treatments are associated to important side effects. There is therefore still an important need to understand insulin resistance- promoting mechanisms to identify therapeutic targets.
Body fat is mainly adipose tissue which is one of the largest organs in the body. Adipose tissue is a metabolically active organ that, besides having a major role in buffering excess energy, secretes a wide panel of factors with signaling functions in homeostasis and metabolism (4). Some of these factors are polypeptides and proteins synthesized and secreted by the adipocytes, and are thus called adipokines. These molecules act locally in an autocrine- paracrine way or in an endocrine manner. Dysfunctional adipose tissue is a hallmark of systemic insulin resistance and type 2 diabetes. The study of the adipose tissue secretome has led to the identification of hundreds of adipokines with beneficial or detrimental metabolic effects, most of them having been poorly investigated so far (5, 6). Nowadays, novel adipokines are still being discovered.
Up to now, APOM expression has been described as being restricted to liver and kidneys in adults (7) and the encoded protein as a plasma apolipoprotein predominantly enriched in HDL (8). The apoM belongs to the lipocalins family (7). As such, in blood, it carries bioactive lipids such as sphingosine-l -phosphate (S1P) and retinol, both being involved several biological processes including lipid metabolism and inflammation (7, 9). ApoM-deficient mice lack HDL-associated S1P and display increased vascular permeability, which is a component of inflammatory response (10). Recently, a link between the apoM/SlP axis and energy metabolism has been demonstrated in mice (11) but the role of the apoM itself in insulin resistance has never been investigated neither in humans nor in the adipose tissue.
SUMMARY OF THE INVENTION:
The present invention relates to the use of the apolipoprotein M for the treatment and diagnosis of insulin resistance. In particular, the present invention is defined by the claims.
DETAILED DESCRIPTION OF THE INVENTION:
The adipose tissue (AT) is a secretory organ producing a wide variety of factors that participate in the genesis of metabolic disorders linked to excess fat mass. Weight loss improves obesity related disorders. Transcriptomic studies on human AT and a combination of analyses of transcriptome and proteome profiling of conditioned media from adipocytes and stromal cells isolated from human AT has led to the identification of apolipoprotein M (apoM) as a putative adipokine. The inventors aimed to validate apoM as novel adipokine, investigate the relationship of AT APOM expression with metabolic syndrome and insulin sensitivity, and study the regulation of its expression in AT and secretion during calorie restriction induced weight loss. Therefore, they examined APOM mRNA level and secretion in AT from patients in 6 independent clinical trials and in vitro in human multipotent adipose-derived stem (hMADS) adipocytes. APOM expression and secretion were measured during dietary interventions. The inventors show that APOM is expressed in human subcutaneous and visceral AT, mainly by adipocytes. ApoM is released into circulation from AT and plasma apoM concentrations correlate with adipose tissue APOM mRNA levels. APOM expression in AT is lower in obese compared to lean individuals and reduced in subjects with metabolic syndrome and type 2 diabetes. Regardless of fat depot, there is a positive relationship between AT APOM expression and systemic insulin sensitivity, independently of fat mass and plasma HDL. In hMADS adipocytes, APOM expression is enhanced by insulin sensitizing PPAR agonists and inhibited by TNFa, a cytokine which causes insulin resistance. In obese individuals, calorie restriction increased AT APOM expression and secretion. Finally, the inventors show that overexpression of APOM in the AT reduces AT inflammation and improves local insulin sensitivity during high fat diet induced obesity.
Methods of treatment:
Accordingly, the first object of the present invention relates to a method of treating insulin resistance in a patient in need thereof comprising administering to the patient a therapeutically effective amount of an ApoM polypeptide or a nucleic acid molecule encoding thereof
As used herein, the term“insulin resistance” has its common meaning in the art. Insulin resistance is a physiological condition where the natural hormone insulin becomes less effective at lowering blood sugars. The resulting increase in blood glucose may raise levels outside the normal range and cause adverse health effects such as metabolic syndrome, dyslipidemia and subsequently type 2 diabetes mellitus. The method of the present invention is thus particularly suitable for the treatment of type 2 diabetes. As used herein, the term "type 2 diabetes" or“non insulin dependent diabetes mellitus (NIDDM)” has its general meaning in the art. Type 2 diabetes often occurs when levels of insulin are normal or even elevated and appears to result from the inability of tissues to respond appropriately to insulin. Most of the type 2 diabetics are obese.
In some embodiments, the subject suffers from obesity. As used herein the term "obesity" refers to a condition characterized by an excess of body fat. The operational definition of obesity is based on the Body Mass Index (BMI), which is calculated as body weight per height in meter squared (kg/m2). Obesity refers to a condition whereby an otherwise healthy subject has a BMI greater than or equal to 30 kg/m2, or a condition whereby a subject with at least one co-morbidity has a BMI greater than or equal to 27 kg/m2. An "obese subject" is an otherwise healthy subject with a BMI greater than or equal to 30 kg/m2 or a subject with at least one co-morbidity with a BMI greater than or equal 27 kg/m2. A "subject at risk of obesity" is an otherwise healthy subject with a BMI of 25 kg/m2 to less than 30 kg/m2 or a subject with at least one co-morbidity with a BMI of 25 kg/m2 to less than 27 kg/m2. The increased risks associated with obesity may occur at a lower BMI in people of Asian descent. In Asian and Asian-Pacific countries, including Japan, "obesity" refers to a condition whereby a subject with at least one obesity-induced or obesity-related co-morbidity that requires weight reduction or that would be improved by weight reduction, has a BMI greater than or equal to 25 kg/m2. An "obese subject" in these countries refers to a subject with at least one obesity- induced or obesity- related co-morbidity that requires weight reduction or that would be improved by weight reduction, with a BMI greater than or equal to 25 kg/m2. In these countries, a "subject at risk of obesity" is a person with a BMI of greater than 23 kg/m2 to less than 25 kg/m2.
As used herein, the term "treatment" or "treat" refer to both prophylactic or preventive treatment as well as curative or disease modifying treatment, including treatment of patient at risk of contracting the disease or suspected to have contracted the disease as well as patients who are ill or have been diagnosed as suffering from a disease or medical condition, and includes suppression of clinical relapse. The treatment may be administered to a subject having a medical disorder or who ultimately may acquire the disorder, in order to prevent, cure, delay the onset of, reduce the severity of, or ameliorate one or more symptoms of a disorder or recurring disorder, or in order to prolong the survival of a subject beyond that expected in the absence of such treatment. By "therapeutic regimen" is meant the pattern of treatment of an illness, e.g., the pattern of dosing used during therapy. A therapeutic regimen may include an induction regimen and a maintenance regimen. The phrase "induction regimen" or "induction period" refers to a therapeutic regimen (or the portion of a therapeutic regimen) that is used for the initial treatment of a disease. The general goal of an induction regimen is to provide a high level of drug to a patient during the initial period of a treatment regimen. An induction regimen may employ (in part or in whole) a "loading regimen", which may include administering a greater dose of the drug than a physician would employ during a maintenance regimen, administering a drug more frequently than a physician would administer the drug during a maintenance regimen, or both. The phrase "maintenance regimen" or "maintenance period" refers to a therapeutic regimen (or the portion of a therapeutic regimen) that is used for the maintenance of a patient during treatment of an illness, e.g., to keep the patient in remission for long periods of time (months or years). A maintenance regimen may employ continuous therapy (e.g., administering a drug at a regular intervals, e.g., weekly, monthly, yearly, etc.) or intermittent therapy (e.g., interrupted treatment, intermittent treatment, treatment at relapse, or treatment upon achievement of a particular predetermined criteria [e.g., disease manifestation, etc.]). In particular, the method of the present invention is particularly suitable for improving blood glucose control, enhancing insulin signalling in skeletal muscle and adipose tissue, reducing lipotoxicity in skeletal muscle and adipose tissue, increasing lipid oxidative capacity in skeletal muscle and adipose tissue, or maintaining long-term insulin sensitivity in the subject.
As used herein, the term“ApoM” has its general meaning in the art and refers to the apolipoprotein M encoded by the APOM gene (Gene ID: 55937). ApoM is an apolipoprotein and member of the lipocalin protein family. The term is also known as G3A or NG20. An exemplary amino acid sequence of ApoM is represented by SEQ ID NO:l and an exemplary nucleic acid sequence is represented by SEQ ID NO:2.
SEQ ID NO : 1 >sp | 095445 | APOM HUMAN Apolipoprotein M OS=Homo sapiens OX=9606 GN=APOM PE=1 SV=2
MFHQIWAALLYFYGI ILNSIYQCPEHSQLTTLGVDGKEFPEVHLGQWYFIAGAAPTKEEL
ATFDPVDNIVFNMAAGSAPMQLHLRATIRMKDGLCVPRKWIYHLTEGSTDLRTEGRPDMK
TELFSSSCPGGIMLNETGQGYQRFLLYNRSPHPPEKCVEEFKSLTSCLDSKAFLLTPRNQ
EACELSNN
SEQ ID NO: 2
1 agagtggact gagcagccag taggggagag agcagttaag gcacacagag caccagctcc 61 ctcctgcctg aagatgttcc accaaatttg ggcagctctg ctctacttct atggtattat
121 ccttaactcc atctaccagt gccctgagca cagtcaactg acaactctgg gcgtggatgg
181 gaaggagttc ccagaggtcc acttgggcca gtggtacttt atcgcagggg cagctcccac
241 caaggaggag ttggcaactt ttgaccctgt ggacaacatt gtcttcaata tggctgctgg
301 ctctgccccg atgcagctcc accttcgtgc taccatccgc atgaaagatg ggctctgtgt
361 gccccggaaa tggatctacc acctgactga agggagcaca gatctcagaa ctgaaggccg
421 ccctgacatg aagactgagc tcttttccag ctcatgccca ggtggaatca tgctgaatga
481 gacaggccag ggttaccagc gctttctcct ctacaatcgc tcaccacatc ctcccgaaaa
541 gtgtgtggag gaattcaagt ccctgacttc ctgcctggac tccaaagcct tcttattgac
601 tcctaggaat caagaggcct gtgagctgtc caataactga cctgtaactt catctaagtc
661 cccagatggg tacaatggga gctgagttgt tggagggaga agctggagac ttccagctcc
721 agctcccact caagataata aagataattt ttcaatcctc aaaaaaaaaa aaaaaaaaaa
781 aaaaaaaaaa
In some embodiments, the ApoM polypeptide of the present invention comprises an amino acid sequence having at least 70% identity with the amino acid sequence as set forth in SEQ ID NO:l. According to the invention a first amino acid sequence having at least 70% identity with a second amino acid sequence means that the first sequence has 70; 71; 72; 73; 74; 75; 76; 77; 78; 79; 80; 81; 82; 83; 84; 85; 86; 87; 88; 89; 90; 91; 92; 93; 94; 95; 96; 97; 98; 99 or 100% identity with the second amino acid sequence. Amino acid sequence identity is typically determined using a suitable sequence alignment algorithm and default parameters, such as BLAST P (Karlin and Altschul, 1990).
According to the invention, the polypeptide of the invention is produced by conventional automated peptide synthesis methods or by recombinant expression. General principles for designing and making proteins are well known to those of skill in the art. The polypeptides of the invention may be synthesized in solution or on a solid support in accordance with conventional techniques. Various automatic synthesizers are commercially available and can be used in accordance with known protocols as described in Stewart and Young; Tam et al, 1983; Merrifield, 1986 and Barany and Merrifield, Gross and Meienhofer, 1979. The polypeptides of the invention may also be synthesized by solid-phase technology employing an exemplary peptide synthesizer such as a Model 433A from Applied Biosystems Inc. The purity of any given protein; generated through automated peptide synthesis or through recombinant methods may be determined using reverse phase HPLC analysis. Chemical authenticity of each peptide may be established by any method well known to those of skill in the art. As an alternative to automated peptide synthesis, recombinant DNA technology may be employed wherein a nucleotide sequence which encodes a protein of choice is inserted into an expression vector, transformed or transfected into an appropriate host cell and cultivated under conditions suitable for expression as described herein below. Recombinant methods are especially preferred for producing longer polypeptides. A variety of expression vector/host systems may be utilized to contain and express the peptide or protein coding sequence. These include but are not limited to microorganisms such as bacteria transformed with recombinant bacteriophage, plasmid or cosmid DNA expression vectors; yeast transformed with yeast expression vectors (Giga-Hama et al, 1999); insect cell systems infected with virus expression vectors (e.g., baculo virus, see Ghosh et al, 2002); plant cell systems transfected with virus expression vectors (e.g., cauliflower mosaic virus, CaMV; tobacco mosaic virus, TMV) or transformed with bacterial expression vectors (e.g., Ti or pBR322 plasmid; see e.g., Babe et al., 2000); or animal cell systems. Those of skill in the art are aware of various techniques for optimizing mammalian expression of proteins, see e.g., Kaufman, 2000; Colosimo et al, 2000. Mammalian cells that are useful in recombinant protein productions include but are not limited to VERO cells, HeLa cells, Chinese hamster ovary (CHO) cell lines, COS cells (such as COS-7), W138, BHK, HepG2, 3T3, RIN, MDCK, A549, PC12, K562 and 293 cells. Exemplary protocols for the recombinant expression of the peptide substrates or fusion polypeptides in bacteria, yeast and other invertebrates are known to those of skill in the art and a briefly described herein below. Mammalian host systems for the expression of recombinant proteins also are well known to those of skill in the art. Host cell strains may be chosen for a particular ability to process the expressed protein or produce certain post-translation modifications that will be useful in providing protein activity. Such modifications of the polypeptide include, but are not limited to, acetylation, carboxylation, glycosylation, phosphorylation, lipidation and acylation. Post- translational processing which cleaves a "prepro" form of the protein may also be important for correct insertion, folding and/or function. Different host cells such as CHO, HeLa, MDCK, 293, WI38, and the like have specific cellular machinery and characteristic mechanisms for such post-translational activities and may be chosen to ensure the correct modification and processing of the introduced, foreign protein.
In some embodiments, it is contemplated that polypeptides used in the therapeutic methods of the present invention may be modified in order to improve their therapeutic efficacy. Such modification of therapeutic compounds may be used to decrease toxicity, increase circulatory time, or modify biodistribution. For example, the toxicity of potentially important therapeutic compounds can be decreased significantly by combination with a variety of drug carrier vehicles that modify biodistribution. In example adding dipeptides can improve the penetration of a circulating agent in the eye through the blood retinal barrier by using endogenous transporters. A strategy for improving drug viability is the utilization of water- soluble polymers. Various water-soluble polymers have been shown to modify biodistribution, improve the mode of cellular uptake, change the permeability through physiological barriers; and modify the rate of clearance from the body. To achieve either a targeting or sustained- release effect, water-soluble polymers have been synthesized that contain drug moieties as terminal groups, as part of the backbone, or as pendent groups on the polymer chain. Polyethylene glycol (PEG) has been widely used as a drug carrier, given its high degree of biocompatibility and ease of modification. Attachment to various drugs, proteins, and liposomes has been shown to improve residence time and decrease toxicity. PEG can be coupled to active agents through the hydroxyl groups at the ends of the chain and via other chemical methods; however, PEG itself is limited to at most two active agents per molecule. In a different approach, copolymers of PEG and amino acids were explored as novel biomaterials which would retain the biocompatibility properties of PEG, but which would have the added advantage of numerous attachment points per molecule (providing greater drug loading), and which could be synthetically designed to suit a variety of applications. Those of skill in the art are aware of PEGylation techniques for the effective modification of drugs. In addition, to the polymer backbone being important in maintaining circulatory half-life, and biodistribution, linkers may be used to maintain the therapeutic agent in a pro-drug form until released from the backbone polymer by a specific trigger, typically enzyme activity in the targeted tissue. For example, this type of tissue activated drug delivery is particularly useful where delivery to a specific site of biodistribution is required and the therapeutic agent is released at or near the site of pathology. Linking group libraries for use in activated drug delivery are known to those of skill in the art and may be based on enzyme kinetics, prevalence of active enzyme, and cleavage specificity of the selected disease-specific enzymes. Such linkers may be used in modifying the protein or fragment of the protein described herein for therapeutic delivery.
In some embodiments, the polypeptides of the invention may be fused to a heterologous polypeptide (i.e. polypeptide derived from an unrelated protein, for example, from an immunoglobulin protein).
As used herein, the term "nucleic acid molecule" has its general meaning in the art and refers to a DNA or RNA molecule. However, the term captures sequences that include any of the known base analogues of DNA and RNA such as, but not limited to 4-acetylcytosine, 8- hydroxy-N6-methyladenosine, aziridinylcytosine, pseudoisocytosine, 5- (carboxyhydroxylmethyl) uracil, 5-fmorouracil, 5-bromouracil, 5- carboxymethylaminomethyl-2-thiouracil, 5-carboxymethyl-aminomethyluracil, dihydrouracil, inosine, N6-isopentenyladenine, 1 -methyladenine, 1 -methylpseudouracil, l-methylguanine,
1- methylinosine, 2,2-dimethylguanine, 2-methyladenine, 2-methylguanine, 3-methylcytosine,
5- methylcytosine, N6-methyladenine, 7-methylguanine, 5-methylaminomethyluracil, 5- methoxyamino-methyl-2-thiouracil, beta-D-mannosylqueosine, 5'- methoxycarbonylmethyluracil, 5-methoxyuracil, 2-methylthio-N6-isopentenyladenine, uracil- 5-oxyacetic acid methylester, uracil-5 -oxyacetic acid, oxybutoxosine, pseudouracil, queosine,
2-thiocytosine, 5-methyl-2-thiouracil, 2-thiouracil, 4-thiouracil, 5-methyluracil, -uracil-5- oxyacetic acid methylester, uracil-5 -oxyacetic acid, pseudouracil, queosine, 2-thiocytosine, and 2,6-diaminopurine.
In some embodiments, the nucleic acid molecule of the present invention comprises a nucleic acid sequence having has at least 70% identity with the nucleic acid sequence as set forth in SEQ ID NO:2. According to the invention a first nucleic acid sequence having at least 70% identity with a second nucleic acid sequence means that the first sequence has 70; 71; 72; 73; 74; 75; 76; 77; 78; 79; 80; 81; 82; 83; 84; 85; 86; 87; 88; 89; 90; 91; 92; 93; 94; 95; 96; 97; 98; 99 or 100% identity with the second nucleic acid sequence.
In some embodiments, the nucleic acid molecule of the present invention is included in a suitable vector. Typically, the vector is a viral vector, and more particularly an adeno- associated virus (AAV), a retrovirus, bovine papilloma virus, an adenovirus vector, a lentiviral vector, a vaccinia virus, a polyoma virus, or an infective virus. In some embodiments, the vector is an AAV vector. As used herein, the term "AAV vector" means a vector derived from an adeno- associated virus serotype, including without limitation, AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, and mutated forms thereof. AAV vectors can have one or more of the AAV wild-type genes deleted in whole or part, preferably the rep and/or cap genes, but retain functional flanking ITR sequences. Retroviruses may be chosen as gene delivery vectors due to their ability to integrate their genes into the host genome, transferring a large amount of foreign genetic material, infecting a broad spectrum of species and cell types and for being packaged in special cell- lines. In order to construct a retroviral vector, a nucleic acid encoding a gene of interest is inserted into the viral genome in the place of certain viral sequences to produce a virus that is replication-defective. In order to produce virions, a packaging cell line is constructed containing the gag, pol, and/or env genes but without the LTR and/or packaging components. When a recombinant plasmid containing a cDNA, together with the retroviral LTR and packaging sequences is introduced into this cell line (by calcium phosphate precipitation for example), the packaging sequence allows the RNA transcript of the recombinant plasmid to be packaged into viral particles, which are then secreted into the culture media. The media containing the recombinant retroviruses is then collected, optionally concentrated, and used for gene transfer. Retroviral vectors are able to infect a broad variety of cell types. Lentiviruses are complex retroviruses, which, in addition to the common retroviral genes gag, pol, and env, contain other genes with regulatory or structural function. The higher complexity enables the virus to modulate its life cycle, as in the course of latent infection. Some examples of lentivirus include the Human Immunodeficiency Viruses (HIV 1 , HIV 2) and the Simian Immunodeficiency Virus (SIV). Lentiviral vectors have been generated by multiply attenuating the HIV virulence genes, for example, the genes env, vif, vpr, vpu and nef are deleted making the vector biologically safe. Lentiviral vectors are known in the art, see, e.g.. U.S. Pat. Nos. 6,013,516 and 5,994,136, both of which are incorporated herein by reference. In general, the vectors are plasmid-based or virus-based, and are configured to carry the essential sequences for incorporating foreign nucleic acid, for selection and for transfer of the nucleic acid into a host cell. The gag, pol and env genes of the vectors of interest also are known in the art. Thus, the relevant genes are cloned into the selected vector and then used to transform the target cell of interest. Recombinant lentivirus capable of infecting a non-dividing cell wherein a suitable host cell is transfected with two or more vectors carrying the packaging functions, namely gag, pol and env, as well as rev and tat is described in U.S. Pat. No. 5,994,136, incorporated herein by reference. This describes a first vector that can provide a nucleic acid encoding a viral gag and a pol gene and another vector that can provide a nucleic acid encoding a viral env to produce a packaging cell. Introducing a vector providing a heterologous gene into that packaging cell yields a producer cell which releases infectious viral particles carrying the foreign gene of interest. The env preferably is an amphotropic envelope protein which allows transduction of cells of human and other species. Typically, the nucleic acid molecule or the vector of the present invention include "control sequences'", which refers collectively to promoter sequences, polyadenylation signals, transcription termination sequences, upstream regulatory domains, origins of replication, internal ribosome entry sites ("IRES"), enhancers, and the like, which collectively provide for the replication, transcription and translation of a coding sequence in a recipient cell. Not all of these control sequences need always be present so long as the selected coding sequence is capable of being replicated, transcribed and translated in an appropriate host cell. Another nucleic acid sequence, is a "promoter" sequence, which is used herein in its ordinary sense to refer to a nucleotide region comprising a DNA regulatory sequence, wherein the regulatory sequence is derived from a gene which is capable of binding R A polymerase and initiating transcription of a downstream (3’-direction) coding sequence. Transcription promoters can include "inducible promoters" (where expression of a polynucleotide sequence operably linked to the promoter is induced by an analyte, cofactor, regulatory protein, etc.), "repressible promoters" (where expression of a polynucleotide sequence operably linked to the promoter is induced by an analyte, cofactor, regulatory protein, etc.), and "constitutive promoters”.
By a "therapeutically effective amount" is meant a sufficient amount of the polypeptide or the nucleic acid molecule encoding thereof to prevent for use in a method for the treatment of the disease (e.g. haemophilia) at a reasonable benefit/risk ratio applicable to any medical treatment. It will be understood that the total daily usage of the compounds and compositions of the present invention will be decided by the attending physician within the scope of sound medical judgment. The specific therapeutically effective dose level for any particular subject will depend upon a variety of factors including the age, body weight, general health, sex and diet of the subject; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidental with the specific polypeptide employed; and like factors well known in the medical arts. For example, it is well known within the skill of the art to start doses of the compound at levels lower than those required to achieve the desired therapeutic effect and to gradually increase the dosage until the desired effect is achieved. However, the daily dosage of the products may be varied over a wide range from 0.01 to 1,000 mg per adult per day. Preferably, the compositions contain 0.01, 0.05, 0.1, 0.5, 1.0, 2.5, 5.0, 10.0, 15.0, 25.0, 50.0, 100, 250 and 500 mg of the active ingredient for the symptomatic adjustment of the dosage to the subject to be treated. A medicament typically contains from about 0.01 mg to about 500 mg of the active ingredient, preferably from 1 mg to about 100 mg of the active ingredient. An effective amount of the drug is ordinarily supplied at a dosage level from 0.0002 mg/kg to about 20 mg/kg of body weight per day, especially from about 0.001 mg/kg to 7 mg/kg of body weight per day.
According to the invention, the polypeptide or the nucleic acid molecule (inserted or not into a vector) of the present invention is administered to the subject in the form of a pharmaceutical composition. Typically, the polypeptide or the nucleic acid molecule (inserted or not into a vector) of the present invention may be combined with pharmaceutically acceptable excipients, and optionally sustained-release matrices, such as biodegradable polymers, to form pharmaceutical compositions. "Pharmaceutically" or "pharmaceutically acceptable" refer to molecular entities and compositions that do not produce an adverse, allergic or other untoward reaction when administered to a mammal, especially a human, as appropriate. A pharmaceutically acceptable carrier or excipient refers to a non-toxic solid, semi-solid or liquid filler, diluent, encapsulating material or formulation auxiliary of any type. In the pharmaceutical compositions of the present invention for oral, sublingual, subcutaneous, intramuscular, intravenous, transdermal, local or rectal administration, the active principle, alone or in combination with another active principle, can be administered in a unit administration form, as a mixture with conventional pharmaceutical supports, to animals and human beings. Suitable unit administration forms comprise oral-route forms such as tablets, gel capsules, powders, granules and oral suspensions or solutions, sublingual and buccal administration forms, aerosols, implants, subcutaneous, transdermal, topical, intraperitoneal, intramuscular, intravenous, subdermal, transdermal, intrathecal and intranasal administration forms and rectal administration forms. Typically, the pharmaceutical compositions contain vehicles which are pharmaceutically acceptable for a formulation capable of being injected. These may be in particular isotonic, sterile, saline solutions (monosodium or disodium phosphate, sodium, potassium, calcium or magnesium chloride and the like or mixtures of such salts), or dry, especially freeze-dried compositions which upon addition, depending on the case, of sterilized water or physiological saline, permit the constitution of injectable solutions. The pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions; formulations including sesame oil, peanut oil or aqueous propylene glycol; and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. In all cases, the form must be sterile and must be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms, such as bacteria and fungi. Solutions comprising compounds of the invention as free base or pharmacologically acceptable salts can be prepared in water suitably mixed with a surfactant, such as hydroxypropylcellulose. Dispersions can also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms. The polypeptide or the nucleic acid molecule (inserted or not into a vector) of the present invention can be formulated into a composition in a neutral or salt form. Pharmaceutically acceptable salts include the acid addition salts (formed with the free amino groups of the protein) and which are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like. Salts formed with the free carboxyl groups can also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, histidine, procaine and the like. The carrier can also be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetables oils. The proper fluidity can be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. The prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars or sodium chloride. Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminium monostearate and gelatin. Sterile injectable solutions are prepared by incorporating the active compounds in the required amount in the appropriate solvent with several of the other ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the typical methods of preparation are vacuum-drying and freeze-drying techniques which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof. The preparation of more, or highly concentrated solutions for direct injection is also contemplated, where the use of DMSO as solvent is envisioned to result in extremely rapid penetration, delivering high concentrations of the active agents to a small tumor area. Upon formulation, solutions will be administered in a manner compatible with the dosage formulation and in such amount as is therapeutically effective. The formulations are easily administered in a variety of dosage forms, such as the type of injectable solutions described above, but drug release capsules and the like can also be employed. For parenteral administration in an aqueous solution, for example, the solution should be suitably buffered if necessary and the liquid diluent first rendered isotonic with sufficient saline or glucose. These particular aqueous solutions are especially suitable for intravenous, intramuscular, subcutaneous and intraperitoneal administration. In this connection, sterile aqueous media which can be employed will be known to those of skill in the art in light of the present disclosure. Some variation in dosage will necessarily occur depending on the condition of the subject being treated. The person responsible for administration will, in any event, determine the appropriate dose for the individual subject.
Methods of diagnosis: A further object of the present invention relates to a method of determining whether a subject has or is at risk of having insulin resistance comprising i) determining the expression level of ApoM in a biological sample obtained from the subject and ii) comparing the expression level determined at step i) with a predetermined reference value wherein detecting differential between the expression level determined at step i) with the predetermined reference value indicated whether the subject has or is at risk of having insulin resistance.
The method of the present invention is thus particularly suitable for determining whether the subject has or is at risk of having a cardiometabolic disease. As used herein, the term "cardiometabolic disease" has its general meaning in the art and relates to cardiovascular diseases associated with metabolic syndrome, such as obesity, diabetes/insulin resistance, hypertension and dyslipidemia. The term "cardiometabolic diseases" refers to cardiac consequences of metabolic syndrome such as atherosclerosis, coronary heart disease, obesity- associated heart disease, insulin resistance- associated heart disease, hypertensive heart disease, cardiac remodeling, heart failure and cardiometabolic diseases disclosed in Hertle et al, 2014; Hua and Nair, 2014; U.S. Pat. Application No. 2012/0214771 and International Patent Application No. 2008/094939.
As used herein, the term "risk" in the context of the present invention, relates to the probability that an event will occur over a specific time period and can mean a subject's "absolute" risk or "relative" risk. Absolute risk can be measured with reference to either actual observation post-measurement for the relevant time cohort, or with reference to index values developed from statistically valid historical cohorts that have been followed for the relevant time period. Relative risk refers to the ratio of absolute risks of a subject compared either to the absolute risks of low risk cohorts or an average population risk, which can vary by how clinical risk factors are assessed. Odds ratios, the proportion of positive events to negative events for a given test result, are also commonly used (odds are according to the formula p/(l-p) where p is the probability of event and (1- p) is the probability of no event) to no- conversion. "Risk evaluation," or "evaluation of risk" in the context of the present invention encompasses making a prediction of the probability, odds, or likelihood that an event or disease state may occur, the rate of occurrence of the event or conversion from one disease state to another. Risk evaluation can also comprise prediction of future clinical parameters, traditional laboratory risk factor values, or other indices of relapse, either in absolute or relative terms in reference to a previously measured population. The methods of the present invention may be used to make continuous or categorical measurements of the risk of conversion, thus diagnosing and defining the risk spectrum of a category of subjects defined as being at risk of conversion. In the categorical scenario, the invention can be used to discriminate between normal and other subject cohorts at higher risk. In some embodiments, the present invention may be used so as to discriminate those at risk from normal.
In some embodiments, the biological sample is a blood sample. As used herein the term “blood sample” means any blood sample derived from the subject. Collections of blood samples can be performed by methods well known to those skilled in the art. In some embodiments, the blood sample is a serum sample or a plasma sample.
The measurement of the level of ApoM in the blood sample is typically carried out using standard protocols known in the art. For example, the method may comprise contacting the blood sample with a binding partner capable of selectively interacting with ApoM in the sample. In some embodiments, the binding partners are antibodies, such as, for example, monoclonal antibodies or even aptamers. For example the binding may be detected through use of a competitive immunoassay, a non-competitive assay system using techniques such as western blots, a radioimmunoassay, an ELISA (enzyme linked immunosorbent assay), a“sandwich” immunoassay, an immunoprecipitation assay, a precipitin reaction, a gel diffusion precipitin reaction, an immunodiffusion assay, an agglutination assay, a complement fixation assay, an immunoradiometric assay, a fluorescent immunoassay, a protein A immunoassay, an immunoprecipitation assay, an immunohistochemical assay, a competition or sandwich ELISA, a radioimmunoassay, a Western blot assay, an immunohistological assay, an immunocytochemical assay, a dot blot assay, a fluorescence polarization assay, a scintillation proximity assay, a homogeneous time resolved fluorescence assay, a IAsys analysis, and a BIAcore analysis. The aforementioned assays generally involve the binding of the partner (ie. antibody or aptamer) to a solid support. Solid supports which can be used in the practice of the invention include substrates such as nitrocellulose (e.g., in membrane or microtiter well form); polyvinylchloride (e.g., sheets or microtiter wells); polystyrene latex (e.g., beads or microtiter plates); polyvinylidine fluoride; diazotized paper; nylon membranes; activated beads, magnetically responsive beads, and the like. An exemplary biochemical test for identifying specific proteins employs a standardized test format, such as ELISA test, although the information provided herein may apply to the development of other biochemical or diagnostic tests and is not limited to the development of an ELISA test (see, e.g., Molecular Immunology: A Textbook, edited by Atassi et al. Marcel Dekker Inc., New York and Basel 1984, for a description of ELISA tests). Therefore ELISA method can be used, wherein the wells of a microtiter plate are coated with a set of antibodies which recognize ApoM. A sample containing or suspected of containing ApoM is then added to the coated wells. After a period of incubation sufficient to allow the formation of antibody-antigen complexes, the plate(s) can be washed to remove unbound moieties and a detectably labelled secondary binding molecule added. The secondary binding molecule is allowed to react with any captured sample marker protein, the plate washed and the presence of the secondary binding molecule detected using methods well known in the art. Measuring the level of ApoM (with or without immunoassay-based methods) may also include separation of the compounds: centrifugation based on the compound’s molecular weight; electrophoresis based on mass and charge; HPLC based on hydrophobicity; size exclusion chromatography based on size; and solid-phase affinity based on the compound's affinity for the particular solid-phase that is used. Once separated, said one or two biomarkers proteins may be identified based on the known "separation profile" e.g., retention time, for that compound and measured using standard techniques. Alternatively, the separated compounds may be detected and measured by, for example, a mass spectrometer. Typically, levels of immunoreactive ApoM in a sample may be measured by an immunometric assay on the basis of a double-antibody "sandwich" technique, with a monoclonal antibody specific for ApoM. According to said embodiment, said means for measuring ApoM level are for example i) a ApoM buffer, ii) a monoclonal antibody that interacts specifically with ApoM, iii) an enzyme- conjugated antibody specific for ApoM and a predetermined reference value of ApoM.
In some embodiments, the predetermined reference value is a threshold value or a cut off value. Typically, a "threshold value" or "cut-off value" can be determined experimentally, empirically, or theoretically. A threshold value can also be arbitrarily selected based upon the existing experimental and/or clinical conditions, as would be recognized by a person of ordinary skilled in the art. For example, retrospective measurement of expression level of the gene in properly banked historical patient samples may be used in establishing the predetermined reference value. The threshold value has to be determined in order to obtain the optimal sensitivity and specificity according to the function of the test and the benefit/risk balance (clinical consequences of false positive and false negative). Typically, the optimal sensitivity and specificity (and so the threshold value) can be determined using a Receiver Operating Characteristic (ROC) curve based on experimental data. For example, after determining the level of the marker in a group of reference, one can use algorithmic analysis for the statistic treatment of the measured levels of the marker in samples to be tested, and thus obtain a classification standard having significance for sample classification. The full name of ROC curve is receiver operator characteristic curve, which is also known as receiver operation characteristic curve. It is mainly used for clinical biochemical diagnostic tests. ROC curve is a comprehensive indicator that reflects the continuous variables of true positive rate (sensitivity) and false positive rate (1 -specificity). It reveals the relationship between sensitivity and specificity with the image composition method. A series of different cut-off values (thresholds or critical values, boundary values between normal and abnormal results of diagnostic test) are set as continuous variables to calculate a series of sensitivity and specificity values. Then sensitivity is used as the vertical coordinate and specificity is used as the horizontal coordinate to draw a curve. The higher the area under the curve (AUC), the higher the accuracy of diagnosis. On the ROC curve, the point closest to the far upper left of the coordinate diagram is a critical point having both high sensitivity and high specificity values. The AUC value of the ROC curve is between 1.0 and 0.5. When AUC>0.5, the diagnostic result gets better and better as AUC approaches 1. When AUC is between 0.5 and 0.7, the accuracy is low. When AUC is between 0.7 and 0.9, the accuracy is moderate. When AUC is higher than 0.9, the accuracy is quite high. This algorithmic method is preferably done with a computer. Existing software or systems in the art may be used for the drawing of the ROC curve, such as: MedCalc 9.2.0.1 medical statistical software, SPSS 9.0, ROCPOWER.SAS, DESIGNROC.FOR, MULTIREADER POWER.SAS, CREATE-ROC.SAS, GB STAT VIO.O (Dynamic Microsystems, Inc. Silver Spring, Md., USA), etc.
In some embodiments, the predetermined reference value is the expression level of ApoM determined in a population of healthy individuals. Typically, it is concluded that the subject has or is at risk of having insulin resistance when the expression level of is at least 0.5, 1, 1.5, 2, 2.5, 3, 3.5, 4, 5, 6, 7, 8, 9, 10, 15, 20, 30, 40, 50, 100 fold lower than the expression level determined in a population of healthy individuals.
The invention will be further illustrated by the following figures and examples. However, these examples and figures should not be interpreted in any way as limiting the scope of the present invention.
FIGURES:
Figure 1. The apolipoprotein M in human adipose tissue.
A. ApoM concentration was measured in paired samples of arterial and venous plasma samples from obese women. Data are plotted as the ratio between venous and arterial blood (AV) for each of the women (cohort A, n=8). *, P<0.05; from Wilcoxon test comparing venous to arterial blood.
B. APOM mRNA level was measured in paired samples (n=l2) of freshly isolated adipocytes and stroma-vascular fraction (SVF) from human subcutaneous abdominal adipose tissue. Data are presented as mean ± SEM. ***, P <0.001; *, P <0.05 from Wilcoxon test. C. ApoM concentration was measured in media from isolated adipocytes (n=l8) and stroma- vascular cells (SVF) (n=l2) from human subcutaneous abdominal adipose tissue fractions cultured for 24h. Data are presented as mean ± SEM. *, P <0.05; from Mann- Whitney test.
D. Differentiated hMADS adipocytes were treated for 4 days with 10 ng/ml TNFa, or the vehicle (n=6).
Figure imgf000018_0001
P <0.01 from Mann and Whitney test compared to control cells.
E. Differentiated hMADS adipocytes were treated for 4 days with 100 nM Rosiglitazone (n=9), 300 nM GW7647, or the vehicle (n=9). **, P <0.01; *, P <0.05 from Mann- Whitney test compared to control cells.
Figure 2. APOM expression in adipose tissues from individuals with a wide range of BMI.
A. APOM mRNA level in subcutaneous adipose tissue was compared between overweight (25< BMI <30, n=66) and obese (30< BMI <35, n=l50; 35 < BMI <40, n=74; BMI>40, n=46) individuals according to obesity grade (cohort E, 111 men and 225 women). Data are presented as mean ± SEM. *, P <0.05 compared to BMI<30 from ANOVA with Dunnett’s post-hoc test compared to overweight group.
B. APOM mRNA level in paired subcutaneous (SAT) and visceral (VAT) adipose tissue was compared between lean (mean BMI= 20.5±3.8, n=l 1) and obese (mean BMI= 39.0 ± 5.9, n=l2) women (cohort B). Data are presented as mean ± SEM. ***, p<0.00l; from 2 way ANOVA test with Dunnett’s post-hoc test compared to lean group.
C. APOM mRNA level was measured in subcutaneous adipose tissue of lean (n=l l), overweight (n=7) and obese (n=27) women (cohort B). Linear regression analysis of APOM expression and body fat mass with correlation coefficient and adjusted P-values according to Benj amini-Hochberg.
Figure 3. Human APOM mRNA level in subcutaneous adipose tissue according to the presence of metabolic syndrome.
A. Adipose tissue APOM expression in relation to adipose morphology in 29 obese (14 with adipose tissue hyperplasia and 15 with adipose tissue hypertrophy) and 26 non obese (13 with adipose tissue hyperplasia and 13 with adipose tissue hypertrophy) women (cohort F).
B-D. APOM mRNA level was measured in subcutaneous adipose tissue of lean (n=l 1), overweight (n=7) and obese (n=27) women (cohort B). Linear regression analysis of subcutaneous adipose tissue APOM expression and waist circumference (B), plasma HDL- cholesterol (C) and plasma triglycerides (D) with correlation coefficient and adjusted p-values according to Benj amini-Hochberg is displayed in each graph. E. APOM mRNA level in subcutaneous adipose tissue was compared between BMI- matched obese (30<BMI<35) women with the metabolic syndrome (MetS, n=28) and obese women without metabolic syndrome (no-MetS, n=28) (cohort E). Data are presented as mean ± SEM. *, P <0.05 from Mann- Whitney test.
Figure 4. Adipose tissue APOM mRNA level and circulating apoM relationship to insulin sensitivity.
A. APOM mRNA level in subcutaneous adipose tissue was compared between normal glucose tolerant (NGT, h=210), impaired fasting glucose and/or impaired glucose tolerance (IFG+IGT, h=101) and type 2 diabetes (T2D, n=l4) individuals (cohort E, 111 men and 225 women). Data are presented as mean ± SEM. *, P <0.05 compared to NGT from ANOVA with Dunnett’s post-hoc test compared to NGT group.
B. Relationship of adipose tissue APOM expression to QUICKI insulin sensitivity index. APOM mRNA level was measured in paired samples of subcutaneous (SAT) and visceral (VAT) adipose tissue of lean (n=l l-l4), overweight (n=7-l l) and obese (n=27-3 l) women (cohort C). Linear regression analysis of APOM gene expression and the quantitative insulin-sensitivity check index (QUICKI). Linear correlation coefficient and adjusted P-values according to Benjamini-Hochberg is displayed in each graph.
Figure 5. Effect of calorie-restriction induced weight loss on adipose tissue apoM.
A. APOM mRNA level was measured in subcutaneous adipose tissue of 314 obese individuals (cohort E) before (baseline) and after an 8-week very low-calorie diet (VLCD). Data are presented as mean ± SEM. ***, P <0.001; from Wilcoxon test.
B. ApoM concentration was measured in media from subcutaneous adipose tissue explants from 10 women (cohort D) at baseline and after a 28-day very low-calorie diet (VLCD). Data are presented as mean ± SEM. *, P <0.05; from Wilcoxon test.
Figure 6. Effect of human APOM overexpression in the perigonadic adipose tissue in high fat fed mice. A. Effect of adenovirus transduction on target genes expression. Perigonadic adipose tissues (PG-AT) were injected with adenoviruses (2xl09 particles/fat pad) containing human APOM (black bars) or green fluorescent protein (white bars, GFP) coding sequences. Gene expression for GFP, human APOM and mouse apom was measured in PG-AT and liver. h=10 per group. B. Effect of adenovirus transduction on body weight. Body weight was measured at baseline (white bars) then after 30 days of high fat diet (black bars) in mice with perigonadic adipose tissues transduced with adenoviruses (2xl09 particles/fat pad) containing human APOM (ad-hAPOM) or green fluorescent protein (ad-GFP), or saline (sham). Data are means ± SEM; h=10 per group; ***, p<0.00l . C. Effect of adenovirus transduction on glucose tolerance. Area under the curve of plasma glucose concentration versus time after an intraperitoneal glucose (lmg g/g body weight) bolus at baseline (DO) and after 21 days of high fat diet (D21) in mice with perigonadic adipose tissues transduced with adenoviruses (109 particles/fat pad) containing human APOM (ad-hAPOM) or green fluorescent protein (ad- GFP), or saline (sham). DO (baseline), n=28; D21, sham n=9, ad-GFP h=10, ad-h APOM n=9. D. Perigonadic adipose tissue phosphorylation of Akt signaling. Perigonadic adipose tissue was incubated ex vivo with 100 nM insulin (black bars), or vehicle (white bars) in Krebs Ringer Hepes supplemented with 0.2% bovine serum albumin for 20 min at 37°C. Proteins were extracted from tissue lysates and quantified according to Bradford. Fifteen pg proteins were separated by SDS-PAGE and after transfer to nitrocellulose membranes were immunoblotted using antibodies raised against Ser(P)-Akt 473 or total- Akt. Data are means ± SEM; Sham, n=4; ad-GFP n=5, ad-h APOM n=6; **, p<0.0l . E. Effect of AAVs transduction of hAPOM in perigonadic adipose tissue on inflammatory signature. Perigonadic adipose tissue was injected with AAVs (5 x 10L10 particles/fat pad) containing human APOM (3), or mCherry (2), or with NaCl 0.9% (1). Gene expression for interleukinl (II- 1 b), macrophage chemoattractant protein (M cpl) and tumor necrosis factor (Jnf-a) was measured in perigonadic adipose tissue. Data are means ± SEM; n=9 per group; *, p<0.05, **, p<0.0l in a 2 -way ANOVA with Tukey's multiple comparisons test.
Figure 7. . Effect of AAVs transduction of h APOM in perigonadic adipose tissue on phosphorylation of Akt signaling. Perigonadic adipose tissue was incubated ex vivo with 100 nM insulin (black bars), or vehicle (white bars) in Krebs Ringer Hepes supplemented with 0.2% bovine serum albumin for 20 min at 37°C. Proteins were extracted from tissue lysates and quantified according to Bradford. Fifteen pg proteins were separated by SDS-PAGE and after transfer to nitrocellulose membranes were immunoblotted using antibodies raised against Ser(P)-Akt 473 or total- Akt. Data are means ± SEM; Control NaCl 0.9% (sham), n=l5; AAV- mCherry n=l l, AAV-h APOM n=l4. *, p-value<0.05 in a 2-way ANOVA with Tukey's multiple comparisons test.
EXAMPLE 1: apolipoprotein M: a novel adipokine decreasing with obesity and upregulated by calorie restriction.
Methods
Clinical cohorts
The premenopausal women included in cohort A, B, C and D were recruited at the Third Faculty of Medicine, Charles University Hospital in Prague (Czech Republic). The studies were approved by the Ethic Committee of the Third Faculty of Medicine, Charles University, Prague. Individuals in cohort E were included in the multicenter, randomized, controlled dietary intervention study, DiOGenes, registered in ClinicalTrials.gov (NCT00390637). Individuals in cohort F were recruited at the Karolinska Institutet, Stockholm (Sweden), and the study was approved by the Regional Ethics committee in Stockholm. All projects were conducted in accordance with the guidelines in The Declaration of Helsinki and individual written informed consent was obtained from all participants involved in the studies. Clinical investigations were performed at rest after an overnight fast. Details for trials and clinical assessments are described in the corresponding references.
Cohort A included 8 obese women (BMI 33.9 ± 0.1 kg/m2) with stable weight during the 3 months preceding the study. As previously described in (12), exclusion criteria were hypertension, diabetes, hyperlipidemia, or any drug treatment.
Cohort B comprised 56 women scheduled to have abdominal surgery (laparoscopic or laparotomic cholecystectomy, hysterectomy and gastric banding) at the Departments of Surgery and Gynecology at Kralovske Vinohrady Faculty Hospital in Prague. BMI ranged from 17.3 to
48.5 kg/m2. Clinical investigation was carried out 7-14 days prior to the surgery, as described in (13). Insulin sensitivity was assessed using a euglycemic hyperinsulinemic clamp. During surgery, paired samples of subcutaneous and visceral adipose tissue were obtained and stored at -80°C until processing.
Cohort C included 14 obese (BMI 34.1 ± 4.7 kg/m2) women with or without polycystic ovary syndrome (PCOS) recruited during 2004-2005. Anthropometric measurements, blood sampling, euglycemic-hyperinsulinemic clamp and subcutaneous abdominal adipose tissue needle biopsies were performed as described in (14).
Cohort D included 10 metabolically healthy overweight and obese women (BMI 34.1 ±
4.5 kg/m2) that followed an 800 kcal/d very low-calorie diet (VLCD) lasting 28 days. Inclusion and exclusion criteria for study participation were previously outlined (15). Clinical measures and subcutaneous abdominal adipose tissue biopsies were taken at baseline and at the end of the VLCD. The participants were instructed not to change their habitual activity during the study. Anthropometric measurements, blood sampling and subcutaneous abdominal adipose tissue needle biopsies were performed at rest after an overnight fast. Adipose tissue explants were maintained in culture media as described below.
Cohort E included a subgroup of 336 (111 men and 225 women) overweight and obese individuals (BMI 34.1 ± 4.7 kg/m2) from the DiOGenes Study in 8 European countries (16). Individuals followed a VLCD for 8 weeks, and those that lost at least 8% of their baseline weight were randomized to one of five ad libitum follow-up diets for 6 months. Clinical measures and subcutaneous abdominal adipose tissue biopsies were taken at each time-points. The participants were instructed not to change their habitual activity during the study. Only baseline and post-VLCD fat samples and clinical data were investigated in the present study.
Cohort F included 55 women without diabetes who had participated in a cross-sectional study conducted at Karolinska Institutet, Stockholm (Sweden) and were investigated as reported in (17). Subcutaneous adipose tissue gene expression was assessed by Gene 1.0 ST Affymetrix arrays and adipocytes isolated from the same fat biopsy were used to measure the mean volume of the adipocytes. By comparing the mean size of the adipocytes with the total amount of body fat, the morphology of adipose tissue could be quantitatively determined (D values), as described in detail in (18). Negative D values indicate hyperplasia (many small adipocytes), and positive D values indicate hypertrophy (few but large adipocytes).
The risk score for metabolic syndrome was calculated according to (19). For assessment of insulin sensitivity, where euglycemic hyperinsulinemic clamp data were not available, the quantitative insulin-sensitivity check index (QUICKI), calculated as l/[log(I) + log(G)], where I is fasting insulin (pun its/m L) and G is fasting glucose (mg/dL), was used since it has been reported to be more reproducible and has a better correlation to the reference glucose clamp method than other surrogates including HOMA-IR, especially for longitudinal studies (20).
Adipose tissue fractionation and ex vivo cell culture
The individuals included in the ex vivo studies are described in Table 1. Human subcutaneous AT samples were collected from patients who underwent abdominal dermo- lipectomy during restorative surgery according to Declaration of Helsinki and to the guidelines of the Ethical Committee of Rangueil Hospital of Toulouse and the Committee for the Protection to Persons (n°: DC-2014-2039). Adipose tissue fractionation and ex vivo culture of cells for 24h are described in (21). Immunoselection/depletion of the distinct adipose tissue cell fractions and the 24h cell culture is described in (22). Media and cell fractions were stored at - 80°C until analysis.
Secretome and transcriptome analyses
For proteome analyses, NanoLC/ESI LTQ-Orbitrap MS/MS was used for analysis of 24h culture media of the different human adipose tissue cell types. Details are reported in (23).
For transcriptome studies, microarray assays were performed using the Agilent Whole Human Genome Microarray 4x44K v2 according to manufacturer’s recommendations (Agilent Technologies) and analysed as described in (21).
Subcutaneous adipose tissue catheterization To measure arteriovenous differences across subcutaneous abdominal adipose tissue, venous and arterial blood were sampled in 8 obese women (BMI 33.9 ± 0.1 kg/m2) as described in (12) (cohort A).
hMADS cell culture
The Human multipotent adipose-derived stem cell (hMADS) cells were cultured and differentiated as previously described (24). Briefly, after six days of proliferation, the cells were incubated in the differentiation medium (serum- free DMEM low glucose/ Ham’s F-12 medium containing l0pg/ml of transferrin, 5pg/ml of insulin, 0.2 nM triiodothyronine, 100mM 3- isobutyl-l-methylxanthine, 1 mM dexamethasone, and 100 nM rosiglitazone). Three days after (day3), dexamethasone and 3 -isobutyl- l-methylxanthine were omitted from the medium. Rosiglitazone was omitted at day 9. By day 13, cells were fully differentiated into adipocytes. At day 14, cells were treated with 100 nM of the PPARy agonist rosiglitazone (Alexis Biochemicals), or 300 nM of the PPARa agonist GW7647 (Sigma), or 10 ng/ml TNFa (Sigma), or the vehicle, for 4 days.
Adipose tissue explants
Adipose tissue samples of about 400 mg obtained from 10 obese women were cut into small pieces and incubated at 37°C for 4h in 4ml of Krebs/Ringer phosphate buffer supplemented with lg/F glucose and 20g/F bovine serum albumin as described in (15) (cohort D).
RNA extraction, cDNA synthesis and quantitative PCR
Total RNA was extracted from tissues and cells using the RNeasy mini kit (Qiagen, GmbH, Hilden, Germany). Total RNA (500 ng) was reverse transcribed for 120 min at 37°C using Superscript II reverse transcriptase (Invitrogen, St Aubin, France) in the presence of random hexamers. A minus RT reaction was performed in parallel to ensure the absence of genomic DNA contamination.
Real-time PCR was performed in duplicate using a set of TaqMan (Thermo Fisher Scientific) probes and primers assays (Hs002l9533_ml). 20 ng cDNA was analyzed on a StepOne Plus Real-Time PCR system (Applied Biosystems, Carlsbad, CA) or, for longitudinal dietary interventions, using the Bio mark™ HD system (BioMark) as described in (21). Adipose tissue and cell data were normalized to GUSB (Hs00939627_ml) or LRP10 (Hs0l047362_ml), respectively.
ELISA assay
ApoM protein level was measured in duplicate using a human apoM EFISA kit (E-EF- H0473, Elabscience, Clinisciences, Nanterre, France), following manufacturer’s instructions. ApoM concentration was calculated using sigmoidal standard curve fitted by nonlinear regression analysis for each test.
Statistical analyses
Gaussian distribution was tested using the D'Agostino & Pearson test. For comparing data from two groups, where a normal distribution was unattainable, a Wilcoxon or Mann- Whitney test was employed for paired or unpaired comparisons respectively. For a comparison using data from more than two groups, ANOVA was performed with Dunnett’s post-hoc test or Kruskal- Wallis with Dunn’s post-hoc test for non-normal data distribution. Linear regression was performed with adipose tissue APOM expression as the dependent variable and the Benjamini-Hochberg procedure was applied to account for multiple testing. Multiple regression was performed with all variables significantly correlated with adipose tissue APOM expression. Partial correlations were computed to assess direct correlations independently of other confounding variables.
Results
APOM is released by human adipose tissue and secreted by adipocytes.
In addition to mature adipocytes, the adipose tissue contains different cell types including preadipocytes and immune, endothelial and progenitor cells corresponding to the stroma vascular fraction. A comparative proteomic analysis of the proteins secreted by these 5 distinct human adipose tissue cell populations led to the identification of 626 proteins secreted by adipocytes, 123 of which have never been described in the literature as produced by the adipose tissue and were found specifically in the media of mature adipocytes. We compared this list of potentially new adipokines with the data of the transcriptome analyses on the same different adipose tissue cellular fractions. Forty of the proteins above, including apoM, were identified as adipocyte markers according to their gene expression profile. We combined this list with previous transcriptome analyses of adipose tissue from individuals with varying amounts of fat mass (13) and identified apoM as a potential adipokine with an expression regulated according to BMI.
To validate apoM as an adipokine, we measured apoM in venous and arterial blood obtained by catheterization of the adipose tissue (cohort A) and investigated APOM expression in adipose tissue fractions and secretion in media of isolated adipocytes and stroma vascular fraction (Table 1).
Table 1. Ex vivo studies Gender Age, years BMI range
Description (M/F) (mean±SD) (kg/m2)
Adipose tissue cells selection for omics 0/6 54.7±9.l 25.0-30.4
Expression adipocytes vs. stromal cells 0/12 45.l±5.3 20.4-33.1
Secretion adipocytes vs. stromal cells 0/22 44.9±l2.3 20.2-39.1
When apoM was measured in plasma from venous and arterial blood obtained by catheterization of the subcutaneous abdominal adipose tissue, the ratio of apoM concentration was greater than 1 for each women (Figure 1A). On average, the concentration of apoM in venous plasma was significantly 12% higher than in arterial plasma, indicating that adipose tissue produces apoM and releases it into the circulation.
APOM expression and secretion were compared between isolated adipocytes and stroma vascular fractions. Compared to the stroma vascular fraction of adipose tissue, APOM was expressed (Figure IB) and secreted (Figure 1C) mainly by adipocytes.
In cohort B, there was a positive relationship between plasma apoM and subcutaneous adipose tissue APOM expression (cohort B, data not shown).
During adipogenic differentiation of hMADS cells APOM expression increased in a time-dependent manner (data not shown). This cell line was used to investigate the effect of prominent regulators of adipocyte secretory activities (25). In differentiated hMADS adipocytes, APOM mRNA level was reduced by TNFa treatment with (Figure ID) and enhanced by a treatment with PPAR agonists (PPARy, rosiglitazone; PPARa, GW 7647)
(Figure IE).
Higher APOM expression in adipose tissue from non-obese compared to obese individuals.
Comparison of APOM expression in subcutaneous adipose tissue between overweight and obese individuals according to obesity grade showed a decreased APOM expression with increasing obesity grade (cohort E, Figure 2A). No difference in subcutaneous adipose tissue
APOM expression was found between men and women (data not shown). Adipose tissue
APOM expression was analysed in paired samples of subcutaneous and visceral adipose tissue from lean and obese women (cohort B). Irrespective of the fat depot, a higher APOM expression was found in lean compared to obese individuals (Figure 2B). As previously reported (26), plasma apoM was lower in overweight and obese compared to lean women (data not shown). Moreover, a strong negative correlation between body fat and APOM expression in both subcutaneous and visceral adipose tissue was found (cohort B, Figure 2C).
Low adipose tissue APOM expression is associated with a dysmetabolic phenotype.
Obesity is an important risk factor for metabolic syndrome as the enlargement of fat mass impacts adipocyte biology. Adipocyte size is linked to metabolic syndrome (27).
Figure 3A shows that subcutaneous adipose tissue characterized by large fat cells (hypertrophy) displayed lower APOM expression than fat from subjects matched for BMI but with many small adipocytes (hyperplasia), irrespective of body weight status (cohort F). A negative correlation between adipose tissue APOM expression and adipocyte volume was found accordingly (data not shown).
There was a negative correlation between subcutaneous adipose tissue APOM expression and waist circumference and fasting plasma triglycerides, and a positive relationship with plasma HDL (cohort B Figure 3B-D). The same features were found for APOM expression in visceral adipose tissue (cohort B, data not shown).
Accordingly, when APOM expression in subcutaneous adipose tissue was compared in obese women matched for BMI and differing in presence of metabolic syndrome (19), a lower APOM expression was found in women with metabolic syndrome (cohort E, Figure 3E).
High adipose tissue APOM expression is associated with insulin sensitivity
Subcutaneous adipose tissue APOM expression was lower in subjects with type 2 diabetes and prediabetes (impaired fasting glucose and/or impaired glucose tolerance, assessed as in (28)) compared to normal glucose-tolerant individuals (cohort E, Figure 4A).
Since euglycemic hyperinsulinemic clamp data and several bioclinical parameters used for calculation of various insulin sensitivity indexes (29) were available for cohort B, we used this cohort to investigate the relationship between adipose tissue APOM expression and these different indexes. Significant correlations of APOM mRNA levels with QUICKI and HOMA- IR, fasting plasma glucose, revised QUICKI (30), and glucose disposal rate were found, irrespective of fat depot (Figure 4B and data not shown). The correlation to QUICKI, revised QUICKI and glucose disposal rate remained significant when corrected for plasma HDL or fat mass (data not shown). Accordingly, there was a positive correlation between APOM and GLUT4 expression in both subcutaneous and visceral fat depots (data not shown). As previously reported, higher plasma apoM concentration was associated with higher insulin- sensitivity (data not shown).
The link between adipose tissue APOM expression and insulin sensitivity was confirmed in a supplementary independent cohort including obese women with or without PCOS (cohort C). The presence of PCOS is linked to insulin resistance and functional derangements in adipose tissue (31). Here, women with PCOS had a lower insulin sensitivity and adipose tissue APOM expression compared to obese women without PCOS matched for fat mass and HDL (data not shown).
Effect of longitudinal dietary interventions on adipose tissue expression and secretion
As adipose tissue apoM expression and plasma levels are associated with obesity and comorbidities developed in obesity, we aimed to study if a weight-reducing diet which improves metabolic profile could influence the levels of apoM expression and production.
We measured APOM expression in subcutaneous adipose tissue of obese individuals before and after an 8-week VLCD (cohort E). The VLCD, which induced a decrease in fat mass, plasma triglycerides, cholesterol and HDL, and an increase in insulin sensitivity, as previously reported (32), resulted in a significant increase in adipose tissue APOM expression (Figure 5A). However, no significant association was found between the magnitude of changes in APOM expression and changes in fat mass, plasma HDL or triglycerides, or QUICKI, though, there was a weak but significant negative association between changes in APOM expression and changes in the C-reactive protein (r= -0.14, P= 0.0088, «=287).
The secretion of apoM by subcutaneous adipose tissue explants was assessed in obese women during a 1 -month VLCD (cohort D). The VLCD induced weight and body fat loss and an improvement in insulin sensitivity. ApoM concentration in media from adipose tissue explants significantly increased (+22%) at the end of the VLCD (Figure 5B).
Discussion:
A combination of microarray studies and proteomic profiling of conditioned media from freshly isolated adipocytes and cell populations in the stroma vascular fraction from human adipose tissue has led to the identification of apoM as a novel adipokine. The present study shows that the human adipose tissue is a site of expression of the APOM gene and that adipose tissue and predominantly adipocytes do secrete apoM.
With reference to other apolipoproteins, the apoM, which was identified in 1999 (33), is comparatively recent. The apoM has been mainly studied in liver where it is produced in hepatocytes; also, kidney proximal tubule cells express apoM (7). Here, we show that apoM is released from adipose tissue in vivo and ex vivo, particularly from adipocytes. The apoM concentration in media from adipose tissue explants in culture is comparable to other adipokines, such as leptin or IL6 (15). In addition, analyses of blood obtained by catheterization of the subcutaneous abdominal adipose tissue confirmed the production of apoM by adipose tissue in vivo (i.e. higher apoM concentration in the venous drainage of adipose tissue compared to paired arterial blood from same individuals). Our study indicates that the adipose tissue might be another producer of apoM in humans; still, the question remains about the extent of contribution of the adipose tissue to circulating apoM. We also found that APOM expression in adipose tissue positively correlates to plasma apoM. This suggests that the large adipose tissue mass is likely to contribute to circulating apoM.
Weight gain in adults is accompanied by an increase in adipocyte size. Adipocytes of obese subjects are dysfunctional, especially regarding metabolism and secretory functions (34). Large adipocytes were reported to be associated with metabolic disease, insulin resistance and type 2 diabetes in multiple studies (35). Here, we found a negative association between adipose tissue APOM mRNA level and fat cell volume. Moreover, in both obese and non-obese women, hypertrophic adipose tissue, containing few but large adipocytes, displayed lower APOM expression compared to hyperplastic tissue that includes more adipocytes but with smaller size. The APOM gene expression thus seems to be a hallmark of healthy adipose tissue.
Indirect data could account for lower APOM expression in obesity. Obesity is now regarded as a chronic low-grade inflammatory state with increased synthesis of acute phase factors by liver and pro -inflammatory cytokines released by immune cells within adipose tissue (36). Here, we found that TNFa down-regulated APOM expression in hMADS adipocytes. Also, in cohort E, we observed a nominal negative association (^-0.143, P=0.0108) between adipose tissue APOM expression and plasma C-reactive protein (data not shown). Since TNFa is locally increased with obesity (36), it is likely that adipose tissue APOM expression could be locally regulated by this pro -inflammatory cytokine.
Unhealthy adipose tissue, deficient in beneficial adipokines such as adiponectin and producing excess of harmful factors, is one of the features of metabolic syndrome and diseases such as insulin resistance and type 2 diabetes. The present study has shown that, in contrast to most adipokines (5, 21) and similarly to adiponectin (37), APOM gene expression in adipose tissue was lower with increasing adiposity. This trend is in accordance with the plasma concentration of apoM lower in obese compared to lean individuals, as previously reported (26).
Furthermore, we show that APOM mRNA level is lower in adipose tissue obtained from subjects with metabolic syndrome, independently of fat mass, and that adipose tissue APOM mRNA level is associated with several metabolic syndrome features (waist circumference, plasma HDF and triglycerides). In humans, plasma apoM has been described as a component of the pre-b HDL (8) and reduced with metabolic syndrome (38), in accordance with the present report.
It was shown previously that type 2 diabetes was also associated with reduced plasma apoM levels compared to glucose tolerance (39, 40). ApoM serum concentration was suggested as a marker for MODY diabetes additionally (41). Here, a positive relationship between adipose tissue APOM mRNA level and systemic insulin sensitivity, irrespective of fat depot, was found in 3 different cohorts. Interestingly, the association with risk factors for diabetes were independent of body fat or plasma HDL. Overall, adipose tissue APOM mRNA level is reduced with prediabetes and diabetes. More evidence is provided by another independent longitudinal study where lean men were investigated before and after a 4-d treatment with the glucocorticoid analogue, dexamethasone. The treatment induced insulin resistance without change in fat mass (42). The adipose tissue microarray data included APOM and showed that glucocorticoid exposure downregulated (mean fold change 0.6) APOM gene expression in subcutaneous fat
(42). Interestingly, here, when all parameters identified in association with adipose tissue APOM expression in univariate analysis were analyzed in a multiple linear regression model with adipose tissue APOM as dependent variable, only QUICKI was an independent predictor (b=0.500, P= 0.04) of adipose tissue APOM mRNA level (data not shown).
Defects in insulin action within adipose tissue are a common feature of insulin resistance
(43). Adipose tissue and the adipocyte itself have emerged as major regulators of whole body insulin action and fuel homeostasis (44). Targeted defects in insulin action in adipocytes lead to systemic insulin resistance (45). In vitro, we show that APOM gene expression in adipocytes is regulated by compounds such as TNFa, and PPAR agonists which influence adipose tissue inflammation and also regulate insulin sensitivity and adipokine production (25). The antidiabetic drug Rosiglitazone is a PPARy agonist that improves insulin sensitivity (46). Treatment of hMADS adipocytes with Rosiglitazone increased APOM mRNA level. The positive effect of this insulin sensitizing agent raises the possibility that apoM may play a causative role in insulin sensitivity.
Reduction in calorie intake results in fat mass loss and alleviates metabolic disorders in obese individuals (47). Diet-induced weight loss also modifies the secretory profile of adipose tissue (14, 48). Contrary to most adipokines (21, 49), including adiponectin (50), we find here that fat mass loss induced by calorie restriction increases expression and secretion of apoM in adipose tissue. No correlation between changes in subcutaneous adipose tissue APOM mRNA level and changes in bioclinical parameters found associated to adipose tissue APOM expression in univariate analysis was found to be significant. However, the increase in adipose tissue APOM expression was associated with the decrease in plasma C-reactive protein, indicating a link between adipose tissue apoM and obesity-related inflammation. Being a lipocalin, apoM was reported as a carrier of several anti-inflammatory lipids (7).
In mice, the apoM/SlP protects against atherosclerosis (9) and controls the metabolic activity of brown fat (11). The ability of apoM to bind anti-inflammatory factors could convey a local effect on the inflammatory status of the adipose tissue. Though, this highlights apoM as a good candidate for monitoring healthy adipose tissue status.
To summarize, the present study reports apoM production by adipocytes for the first time. It shows that APOM mRNA level is reduced in adipose tissue from obese subjects and independently of body fat mass, is also lower in individuals with insulin resistance, type 2 diabetes or dyslipidemia. Patterns of expression and secretion in adipose tissue indicate that apoM is also regulated in response to calorie-restriction induced weight loss.
Altogether this study indicates that apoM is a new adipokine, which may reflect changes in adipose tissue mass and whole body metabolic dysfunction, and which presence in the white adipose tissue is positively associated with insulin sensitivity.
EXAMPLE 2: Effect of human APOM overexpression in the perigonadic adipose tissue in high fat fed mice.
Methods:
Thirty 5-week old C57BL6/J male mice fed ad libitum were randomized in 3 groups of 10 mice according to body weight and glucose tolerance tests (lmg glucose/g body weight, intraperitoneal). Mice were housed at room temperature and manipulated according to Inserm guidelines and European Directive 2010/63/UE in the local animal care facility (agreements A 31 555 04 and C 31 555 07). Mice were fed with a 60% high fat diet for 4 weeks. After 2 weeks, mice were anaesthetized and perigonadic adipose tissues (PG-AT) were injected with recombinant adenoviruses (2xl09 particles/fat pad) containing coding sequences of human APOM (ad-hAPOM) or green fluorescent protein (ad-GFP), or with saline (sham) or red fluorescent protein (mCherry). Two weeks later, glucose tolerance tests were performed in order to ensure that the high fat diet induced insulin resistance, at least in control mice. Then, after 30 days of high fat diet mice were weighed, and euthanatized before dissection of perigonadic and subcutaneous adipose tissues, and liver. Part of the tissues were snap frozen and stored at -80°C until total RNA extraction. To assess insulin sensitivity, the remaining adipose tissue samples were incubated ex vivo with insulin (100 nM), or vehicle, in Krebs Ringer Hepes supplemented with 0.2% bovine serum albumin. After 20 min at 37°C, tissue samples were rinced, snap frozen and stored at -80°C until protein extraction. Moreover, the perigonadic adipose tissue response to insulin was investigated ex vivo (20 min. insulin at 100 nM vs control) with the subsequent study of the Akt signaling pathway which full activation requires phosphorylation of S473 (Akt-P473).
Results:
Direct injection of adenoviruses into perigonadic fat pad induced overexpression of target genes, human apoM and the control GFP, in the perigonadic fat with no leak in liver and no change in mouse apoM expression (Figure 6A). Murine apoM was nearly undetectable in adipose tissue. As expected, the high fat diet induced weight gain and worsened glucose tolerance (Figures 6B and 6C). There was no difference between groups. In perigonadic fat of mice with human apoM overexpression, insulin-induced Akt phosphorylation was higher compared to mice with GFP overexpression and sham mice (Figure 6D). The expression of genes encoding inflammatory factors (interleukin 1 (II- 1 b), macrophage chemoattractant protein (Mcpl) and tumor necrosis factor (Tnf-a)) in perigonadic adipose tissue is displayed on Figure 6E. Despite no significant change in 116 gene expression was found across the 3 treatments, the perigonadic adipose tissue transduced using AAVmCherry exhibited an inflammatory signature evidenced by an increase in Tnf-a, Ill -b and Mcpl mRNA levels. When the perigonadic adipose tissue was transduced using AAVhAPOM, no inflammatory signature was found. This indicates that local overexpression of human APOM may prevent from an inflammatory signature.
Transduction of perigonadic adipose tissue with AAVs induced no change in control (baseline) Akt-P473. However, a higher Akt-P473 level under insulin treatment in AAVhAPOM, compared to AAVmCherry suggests a positive effect of local overexpression of human APOM on insulin signaling (Figure 7).
Discussion:
Obesity is associated with insulin resistance, a pathological condition and a factor in type 2 diabetes. Many of the mechanisms that bridge the pathogenesis of insulin resistance and obesity have been associated with ectopic lipid accumulation and/or obesity-induced inflammation. Pioneer research (Hotamisligil etal, Science 1993;259:87-91) demonstrated that tumor necrosis factor (TNF-a), an inflammatory mediator produced extensively by activated macrophages, is an insulin resistance pathogenicity factor. In lean individuals, adipose tissue secretes high levels of anti-inflammatory mediators such as interleukin- 10 (IL-10) or adiponectin. On the contrary, in individuals with obesity, adipose tissue secretes high levels of proinflammatory adipokines such as I L- 1 b, monocyte chemoattractant protein 1 (MCP-l), and TNF-a. This preponderance of proinflammatory versus anti-inflammatory adipokines is a hallmark of obesity-associated low-grade inflammation which leads to macrophage infiltration. Similar results were obtained using AAVs instead of adenoviruses, when local overexpression of human apoM prevented high fat diet induced impairement of insulin signaling and expression of pro-inflammatory cytokines.
The present study shows that overexpression of APOM in the AT reduces AT inflammation and improves local insulin sensitivity during high fat diet induced obesity.
REFERENCES:
Throughout this application, various references describe the state of the art to which this invention pertains. The disclosures of these references are hereby incorporated by reference into the present disclosure.
1. Obesity and diabetes in 2017: a new year. Lancet 20l7;389: l . doi: 10.1016/SO l40-6736( 17)30004- 1.
2. Samson SL, Garber AJ. Metabolic syndrome. Endocrinol Metab Clin North Am 2014;43: 1-23. doi: l0. l0l6/j.ecl.20l3.09.009.
3. Ogurtsova K, da Rocha Fernandes JD, Huang Y, Linnenkamp U, Guariguata L, Cho NH, Cavan D, Shaw JE, Makaroff LE. IDF Diabetes Atlas: Global estimates for the prevalence of diabetes for 2015 and 2040. Diabetes Res Clin Pract 2017;128:40-50. doi: l0. l0l6/j.diabres.20l7.03.024.
4. Lafontan M. Adipose tissue and adipocyte dysregulation. Diabetes Metab 2014;40: 16-28. doi: l0. l0l6/j.diabet.20l3.08.002.
5. Bluher M, Mantzoros CS. From leptin to other adipokines in health and disease: facts and expectations at the beginning of the 21 st century. Metabolism 2015;64: 131-45. doi: l0.l0l6/j.metabo 1.2014.10.016.
6. Feve B, Bastard C, Fellahi S, Bastard JP, Capeau J. New adipokines. Ann Endocrinol (Paris) 2016;77:49-56. doi: l0. l0l6/j.ando.20l6.0l .00l .
7. Ren K, Tang ZF, Jiang Y, Tan YM, Yi GH. Apo lipoprotein M. Clin Chim Acta 2015. doi: l0. l0l6/j.cca.20l5.03.038.
8. Wolffum C, Poy MN, Stoffel M. Apo lipoprotein M is required for prebeta-HDF formation and cholesterol efflux to HDF and protects against atherosclerosis. Nat Med 2005;11 :418-22. doi: l0. l038/nml2l l .
9. Borup A, Christensen PM, Nielsen FB, Christo ffersen C. Apolipoprotein M in lipid metabolism and cardio metabolic diseases. Curr Opin Fipidol 2015;26:48-55. doi: 10.1097/MOF.0000000000000142. 10. Christoffersen C, Obinata H, Kumaraswamy SB, Galvani S, Ahnstrom J, Sewana M, Egerer-Sieber C, Muller YA, Hla T, Nielsen LB, et al. Endothelium-protective sphingosine-l -phosphate provided by HDL-associated apolipoprotein M. Proc Natl Acad Sci U S A 2011;108:9613-8. doi: l0.l073/pnas. l 103187108.
11. Christoffersen C, Federspiel CK, Borup A, Christensen PM, Madsen AN, Heine M, Nielsen CH, Kjaer A, Holst B, Heeren J, et al. The Apolipoprotein M/S1P Axis Controls Triglyceride Metabolism and Brown Fat Activity. Cell Rep 2018;22: 175-88. doi: 10. l0l6/j.celrep.2017.12.029.
12. Siklova M, Simonsen L, Polak J, Stich V, Bulow J. Effect of short-term hyperglycemia on adipose tissue fluxes of selected cytokines in vivo during multiple phases of diet-induced weight loss in obese women. J Clin Endocrinol Metab 2015;100: 1949-56. doi: 10.1210/JC.2014-3846.
13. Klimcakova E, Roussel B, Kovacova Z, Kovacikova M, Siklova- Vitkova M, Combes M, Hejnova J, Decaunes P, Maoret JJ, Vedral T, et al. Macrophage gene expression is related to obesity and the metabolic syndrome in human subcutaneous fat as well as in visceral fat. Diabetologia 2011;54:876-87. doi: l0.l007/s00l25-0l0-20l4-3.
14. Capel F, Klimcakova E, Viguerie N, Roussel B, Vitkova M, Kovacikova M, Polak J, Kovacova Z, Galitzky J, Maoret JJ, et al. Macrophages and adipocytes in human obesity: adipose tissue gene expression and insulin sensitivity during calorie restriction and weight stabilization. Diabetes 2009;58: 1558-67.
15. Sramkova V, Rossmeislova L, Krauzova E, Kracmerova J, Koc M, Langin D, Stich V, Siklova M. Comparison of Early (2 Days) and Later (28 Days) Response of Adipose Tissue to Very Low-Calorie Diet in Obese Women. J Clin Endocrinol Metab 2016; 101 :5021 - 9. doi: 10.1210/jc.2016-2161.
16. Larsen TM, Dalskov SM, van Baak M, Jebb SA, Papadaki A, Pfeiffer AF, Martinez JA, Handjieva-Darlenska T, Kunesova M, Pihlsgard M, et al. Diets with high or low protein content and glycemic index for weight- loss maintenance. N Engl J Med 2010;363:2102- 13. doi: l0.l056/NEJMoal007l37.
17. Gao H, Mejhert N, Fretz JA, Amer E, Lorente-Cebrian S, Ehrlund A, Dahlman- Wright K, Gong X, Stromblad S, Douagi I, et al. Early B cell factor 1 regulates adipocyte morphology and lipolysis in white adipose tissue. Cell Metab 2014;19:981-92. doi: l0. l0l6/j.cmet.20l4.03.032. 18. Arner E, Westermark PO, Spalding KL, Britton T, Ryden M, Frisen J, Bernard S, Arner P. Adipocyte turnover: relevance to human adipose tissue morphology. Diabetes 2010;59: 105-9. doi: l0.2337/db09-0942.
19. Alberti KG, Eckel RH, Grundy SM, Zimmet PZ, Cleeman JI, Donato KA,
Fruchart JC, James WP, Loria CM, Smith SC, Jr. Harmonizing the metabolic syndrome: a joint interim statement of the International Diabetes Federation Task Force on Epidemiology and Prevention; National Heart, Fung, and Blood Institute; American Heart Association; World Heart Federation; International Atherosclerosis Society; and International Association for the Study of Obesity. Circulation 2009;120: 1640-5. doi:
10.116 l/CIRCUFATIONAHA.109.192644.
20. Katz A, Nambi SS, Mather K, Baron AD, Follmann DA, Sullivan G, Quon MJ. Quantitative insulin sensitivity check index: a simple, accurate method for assessing insulin sensitivity in humans. J Clin Endocrinol Metab 2000;85:2402-10. doi: l0. l2l0/jcem.85.7.666l .
21. Bolton J, Montastier E, Carayol J, Bonnel S, Mir F, Marques MA, Astrup A, Saris W, Iacovoni J, Villa- Vialaneix N, et al. Molecular Biomarkers for Weight Control in Obese Individuals Subjected to a Multiphase Dietary Intervention. J Clin Endocrinol Metab 2017;102:2751-61. doi: 10.1210/jc.20l6-3997.
22. Bourlier V, Sengenes C, Zakaroff-Girard A, Decaunes P, Wdziekonski B, Galitzky J, Villageois P, Esteve D, Chiotasso P, Dani C, et al. TGFbeta family members are key mediators in the induction of myofibroblast phenotype of human adipose tissue progenitor cells by macrophages. PFoS One 20l2;7:e3 l274. doi: 10. l37l/joumal.pone.0031274.
23. Mejhert N, Wilfling F, Esteve D, Galitzky J, Pellegrinelli V, Kolditz Cl, Viguerie N, Tordjman J, Naslund E, Trayhum P, et al. Semaphorin 3C is a novel adipokine linked to extracellular matrix composition. Diabetologia 2013;56: 1792-801. doi: 10.1007/s00125-013-293 l-z.
24. Barquissau V, Beuzelin D, Pisani DF, Beranger GE, Mairal A, Montagner A, Roussel B, Tavernier G, Marques MA, Moro C, et al. White-to-brite conversion in human adipocytes promotes metabolic reprogramming towards fatty acid anabolic and catabolic pathways. Mol Metab 2016;5:352-65. doi: l0. l0l6/j.molmet.20l6.03.002.
25. Westerink J, Visseren FF. Pharmacological and non-pharmacological interventions to influence adipose tissue function. Cardiovasc Diabetol 2011; 10: 13. doi: 10.1186/1475-2840-10-13. 26. Kappelle PJ, Lambert G, Dahlback B, Nielsen LB, Dullaart RP. Relationship of plasma apolipoprotein M with proprotein convertase subtilisin-kexin type 9 levels in non diabetic subjects. Atherosclerosis 2011;214:492-4. doi: 10.1016/j. atherosclerosis.2010.10.041.
27. Ryden M, Amer P. Cardiovascular risk score is linked to subcutaneous adipocyte size and lipid metabolism. J Intern Med 2017;282:220-8. doi: 10.1 11 l/joim. l264l .
28. Diagnosis and classification of diabetes mellitus. Diabetes Care 20l0;33 Suppl LS62-9. doi: l0.2337/dcl0-S062.
29. Mather KJ, Hunt AE, Steinberg HO, Paradisi G, Hook G, Katz A, Quon MJ, Baron AD. Repeatability characteristics of simple indices of insulin resistance: implications for research applications. J Clin Endocrinol Metab 2001;86:5457-64. doi: 10. l2l0/jcem.86.11.7880.
30. Perseghin G, Caumo A, Caloni M, Testolin G, Luzi L. Incorporation of the fasting plasma FFA concentration into QUICKI improves its association with insulin sensitivity in nonobese individuals. J Clin Endocrinol Metab 2001;86:4776-81. doi: 10. l2l0/jcem.86.10.7902.
31. Delitala AP, Capobianco G, Delitala G, Cherchi PL, Dessole S. Polycystic ovary syndrome, adipose tissue and metabolic syndrome. Arch Gynecol Obstet 2017. doi: 10.1007/s00404-017-4429-2.
32. Viguerie N, Montastier E, Maoret JJ, Roussel B, Combes M, Valle C, Villa- Vialaneix N, Iacovoni JS, Martinez JA, Holst C, et al. Determinants of human adipose tissue gene expression: impact of diet, sex, metabolic status, and cis genetic regulation. PLoS Genet 20l2;8:el002959. doi: 10.137 l/joumal.pgen.1002959.
33. Xu N, Dahlback B. A novel human apolipoprotein (apoM). J Biol Chem 1999;274:31286-90.
34. Arner P, Spalding KL. Fat cell turnover in humans. Biochem Biophys Res Commun 2010;396: 101-4. doi: l0. l0l6/j.bbrc.20l0.02. l65.
35. Andersson DP, Eriksson Hogling D, Thorell A, Toft E, Qvisth V, Naslund E, Thome A, Wiren M, Lofgren P, Hoffstedt J, et al. Changes in subcutaneous fat cell volume and insulin sensitivity after weight loss. Diabetes Care 2014;37: 1831-6. doi: l0.2337/dcl3-2395.
36. Saltiel AR, Olefsky JM. Inflammatory mechanisms linking obesity and metabolic disease. J Clin Invest 2017;127: 1-4. doi: 10.1172/JCI92035.
37. Salehi-Abargouei A, Izadi V, Azadbakht L. The effect of low calorie diet on adiponectin concentration: a systematic review and meta-analysis. Horm Metab Res 2015;47:549-55. doi: 10.1055/S-0035-1549878. 38. Dullaart RP, Plomgaard P, de Vries R, Dahlback B, Nielsen LB. Plasma apolipoprotein M is reduced in metabolic syndrome but does not predict intima media thickness. Clin Chim Acta 2009;406: 129-33. doi: l0. l0l6/j.cca.2009.06.0l0.
39. Ooi EM, Watts GF, Chan DC, Nielsen LB, Plomgaard P, Dahlback B, Barrett PH. Association of apolipoprotein M with high-density lipoprotein kinetics in overweight- obese men. Atherosclerosis 2010;210:326-30. doi: 10.1016/j. atherosclerosis.2009.11.024.
40. Plomgaard P, Dullaart RP, de Vries R, Groen AK, Dahlback B, Nielsen LB. Apolipoprotein M predicts pre-beta-HDL formation: studies in type 2 diabetic and nondiabetic subjects. J Intern Med 2009;266:258-67. doi: 10.1 l l l/j.1365-2796.2009.02095.x.
41. Richter S, Shih DQ, Pearson ER, Wolfrum C, Fajans SS, Hattersley AT, Stoffel M. Regulation of apolipoprotein M gene expression by MODY3 gene hepatocyte nuclear factor- 1 alpha: hap lo insufficiency is associated with reduced serum apolipoprotein M levels. Diabetes 2003;52:2989-95.
42. Viguerie N, Picard F, Hul G, Roussel B, Barbe P, Iacovoni JS, Valle C, Langin D, Saris WH. Multiple effects of a short-term dexamethasone treatment in human skeletal muscle and adipose tissue. Physiol Genomics 2012;44: 141-51. doi: 10.1152/physio lgenomics.00032.2011.
43. Lo KA, Labadorf A, Kennedy NJ, Han MS, Yap YS, Matthews B, Xin X, Sun L, Davis RJ, Lodish HF, et al. Analysis of in vitro insulin-resistance models and their physiological relevance to in vivo diet-induced adipose insulin resistance. Cell Rep 20l3;5:259- 70. doi: l0. l0l6/j.celrep.20l3.08.039.
44. Adams-Huet B, Devaraj S, Siegel D, Jialal I. Increased adipose tissue insulin resistance in metabolic syndrome: relationship to circulating adipokines. Metab Syndr Relat Disord 2014;12:503-7. doi: l0.l089/met.20l4.0092.
45. Abel ED, Peroni O, Kim JK, Kim YB, Boss O, Hadro E, Minnemann T, Shulman GI, Kahn BB. Adipose-selective targeting of the GLUT4 gene impairs insulin action in muscle and liver. Nature 2001;409:729-33. doi: 10.1038/35055575.
46. Moore GB, Pickavance LC, Briscoe CP, Clapham JC, Buckingham RE, Wilding JP. Energy restriction enhances therapeutic efficacy of the PPARgamma agonist, rosiglitazone, through regulation of visceral fat gene expression. Diabetes Obes Metab 2008;10:251-63. doi: 10.111 l/j.1463-1326.2007.00697.x.
47. Most J, Tosti V, Redman LM, Fontana L. Calorie restriction in humans: An update. Ageing Res Rev 2017;39:36-45. doi: l0. l0l6/j.arr.20l6.08.005. 48. Vitkova M, Klimcakova E, Kovacikova M, Valle C, Moro C, Polak J, Hanacek J, Capel F, Viguerie N, Richterova B, et al. Plasma levels and adipose tissue messenger ribonucleic acid expression of retinol-binding protein 4 are reduced during calorie restriction in obese subjects but are not related to diet-induced changes in insulin sensitivity. J Clin Endocrinol Metab 2007;92:2330-5. doi: 10. l2l0/jc.2006-2668.
49. MacLean PS, Higgins JA, Giles ED, Sherk VD, Jackman MR. The role for adipose tissue in weight regain after weight loss. Obes Rev 20l5;l6 Suppl 1 :45-54. doi: 10.111 l/obr.12255.
50. Turer AT, Scherer PE. Adiponectin: mechanistic insights and clinical implications. Diabetologia 2012;55:2319-26. doi: l0.l007/s00l25-0l2-2598-x.

Claims

CLAIMS:
1. A method of treating insulin resistance in a patient in need thereof comprising administering to the patient a therapeutically effective amount of an ApoM polypeptide or a nucleic acid molecule encoding thereof.
2. The method of claim 1 wherein the patient suffers from type 2 diabetes.
3. The method of claim 1 wherein the patient suffers from obesity.
4. The method of claim 1 wherein the ApoM polypeptide of the present invention comprises an amino acid sequence having at least 70% identity with the amino acid sequence as set forth in SEQ ID NO: 1.
5. The method of claim 1 wherein the nucleic acid molecule of the present invention comprises a nucleic acid sequence having has at least 70% identity with the nucleic acid sequence as set forth in SEQ ID NO:2.
6. The method of claim 1 wherein the nucleic acid molecule of the present invention is included in a suitable vector.
7. The method of claim 6 wherein the vector is an AAV vector.
8. A method of determining whether a subject has or is at risk of having insulin resistance comprising i) determining the expression level of ApoM in a biological sample obtained from the subject and ii) comparing the expression level determined at step i) with a predetermined reference value wherein detecting differential between the expression level determined at step i) with the predetermined reference value indicated whether the subject has or is at risk of having insulin resistance.
9. The method of claim 1 wherein the biological sample is a blood sample.
10. The method of claim 1 wherein the predetermined reference value is the expression level of ApoM determined in a population of healthy individuals.
11. The method of claim 10 wherein it is concluded that the subject has or is at risk of having insulin resistance when the expression level of is at least 0.5, 1, 1.5, 2, 2.5, 3,
3.5, 4, 5, 6, 7, 8, 9, 10, 15, 20, 30, 40, 50, 100 fold lower than the expression level determined in a population of healthy individuals.
12. Use of the method of claim 8 for determining whether the subject has or is at risk of having a cardio metabolic disease.
PCT/EP2019/065701 2018-06-15 2019-06-14 Use of the apolipoprotein m for the treatment and diagnosis of insulin resistance WO2019238934A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP18305734 2018-06-15
EP18305734.8 2018-06-15

Publications (1)

Publication Number Publication Date
WO2019238934A1 true WO2019238934A1 (en) 2019-12-19

Family

ID=62814968

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2019/065701 WO2019238934A1 (en) 2018-06-15 2019-06-14 Use of the apolipoprotein m for the treatment and diagnosis of insulin resistance

Country Status (1)

Country Link
WO (1) WO2019238934A1 (en)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5994136A (en) 1997-12-12 1999-11-30 Cell Genesys, Inc. Method and means for producing high titer, safe, recombinant lentivirus vectors
US6013516A (en) 1995-10-06 2000-01-11 The Salk Institute For Biological Studies Vector and method of use for nucleic acid delivery to non-dividing cells
WO2008094939A1 (en) 2007-01-31 2008-08-07 Nutrition 21, Inc. Use of chromium histidinate for treatment of cardiometabolic disorders
US20120214771A1 (en) 2001-07-27 2012-08-23 Fontini Sampalis Compositions for treatment of cardiometabolic disorders

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6013516A (en) 1995-10-06 2000-01-11 The Salk Institute For Biological Studies Vector and method of use for nucleic acid delivery to non-dividing cells
US5994136A (en) 1997-12-12 1999-11-30 Cell Genesys, Inc. Method and means for producing high titer, safe, recombinant lentivirus vectors
US20120214771A1 (en) 2001-07-27 2012-08-23 Fontini Sampalis Compositions for treatment of cardiometabolic disorders
WO2008094939A1 (en) 2007-01-31 2008-08-07 Nutrition 21, Inc. Use of chromium histidinate for treatment of cardiometabolic disorders

Non-Patent Citations (59)

* Cited by examiner, † Cited by third party
Title
"Diagnosis and classification of diabetes mellitus", DIABETES CARE, vol. 33, no. I, 2010, pages S62 - 9
"Obesity and diabetes in 2017: a new year", LANCET, vol. 389, 2017, pages 1
ABEL EDPERONI OKIM JKKIM YBBOSS OHADRO EMINNEMANN TSHULMAN GIKAHN BB: "Adipose-selective targeting of the GLUT4 gene impairs insulin action in muscle and liver", NATURE, vol. 409, 2001, pages 729 - 33, XP002326954, DOI: doi:10.1038/35055575
ADAMS-HUET BDEVARAJ SSIEGEL DJIALAL I: "Increased adipose tissue insulin resistance in metabolic syndrome: relationship to circulating adipokines", METAB SYNDR RELAT DISORD, vol. 12, 2014, pages 503 - 7
ALBERTI KGECKEL RHGRUNDY SMZIMMET PZCLEEMAN JIDONATO KAFRUCHART JCJAMES WPLORIA CMSMITH SC, JR.: "Circulation", vol. 120, 2009, NATIONAL HEART, LUNG, AND BLOOD INSTITUTE, article "Harmonizing the metabolic syndrome: a joint interim statement of the International Diabetes Federation Task Force on Epidemiology and Prevention", pages: 1640 - 5
ANDERSSON DPERIKSSON HOGLING DTHORELL ATOFT EQVISTH VNASLUND ETHORNE AWIREN MLOFGREN PHOFFSTEDT J ET AL.: "Changes in subcutaneous fat cell volume and insulin sensitivity after weight loss", DIABETES CARE, vol. 37, 2014, pages 1831 - 6
ARNER EWESTERMARK POSPALDING KLBRITTON TRYDEN MFRISEN JBERNARD SARNER P: "Adipocyte turnover: relevance to human adipose tissue morphology", DIABETES, vol. 59, 2010, pages 105 - 9
ARNER PSPALDING KL: "Fat cell turnover in humans", BIOCHEM BIOPHYS RES COMMUN, vol. 396, 2010, pages 101 - 4, XP027057277, DOI: doi:10.1016/j.bbrc.2010.02.165
BARQUISSAU VBEUZELIN DPISANI DFBERANGER GEMAIRAL AMONTAGNER AROUSSEL BTAVERNIER GMARQUES MAMORO C ET AL.: "White-to-brite conversion in human adipocytes promotes metabolic reprogramming towards fatty acid anabolic and catabolic pathways", MOL METAB, vol. 5, 2016, pages 352 - 65, XP055493847, DOI: doi:10.1016/j.molmet.2016.03.002
BLUHER MMANTZOROS CS: "From leptin to other adipokines in health and disease: facts and expectations at the beginning of the 21st century", METABOLISM, vol. 64, 2015, pages 131 - 45, XP029112091, DOI: doi:10.1016/j.metabol.2014.10.016
BOLTON JMONTASTIER ECARAYOL JBONNEL SMIR LMARQUES MAASTRUP ASARIS WIACOVONI JVILLA-VIALANEIX N ET AL.: "Molecular Biomarkers for Weight Control in Obese Individuals Subjected to a Multiphase Dietary Intervention", J CLIN ENDOCRINOL METAB, vol. 102, 2017, pages 2751 - 61
BORUP ACHRISTENSEN PMNIELSEN LBCHRISTOFFERSEN C: "Apolipoprotein M in lipid metabolism and cardiometabolic diseases", CURR OPIN LIPIDOL, vol. 26, 2015, pages 48 - 55, XP055523644, DOI: doi:10.1097/MOL.0000000000000142
BOURLIER VSENGENES CZAKAROFF-GIRARD ADECAUNES PWDZIEKONSKI BGALITZKY JVILLAGEOIS PESTEVE DCHIOTASSO PDANI C ET AL.: "TGFbeta family members are key mediators in the induction of myofibroblast phenotype of human adipose tissue progenitor cells by macrophages", PLOS ONE, vol. 7, 2012, pages e31274
CAPEL FKLIMCAKOVA EVIGUERIE NROUSSEL BVITKOVA MKOVACIKOVA MPOLAK JKOVACOVA ZGALITZKY JMAORET JJ ET AL.: "Macrophages and adipocytes in human obesity: adipose tissue gene expression and insulin sensitivity during calorie restriction and weight stabilization", DIABETES, vol. 58, 2009, pages 1558 - 67, XP055032252, DOI: doi:10.2337/db09-0033
CHRISTOFFERSEN CFEDERSPIEL CKBORUP ACHRISTENSEN PMMADSEN ANHEINE MNIELSEN CHKJAER AHOIST BHEEREN J ET AL.: "The Apolipoprotein M/S1P Axis Controls Triglyceride Metabolism and Brown Fat Activity", CELL REP, vol. 22, 2018, pages 175 - 88
CHRISTOFFERSEN COBINATA HKUMARASWAMY SBGALVANI SAHNSTROM JSEWANA MEGERER-SIEBER CMULLER YAHLA TNIELSEN LB ET AL.: "Endothelium-protective sphingosine-1-phosphate provided by HDL-associated apolipoprotein M", PROC NATL ACAD SCI USA, vol. 108, 2011, pages 9613 - 8, XP055571402, DOI: doi:10.1073/pnas.1103187108
DELITALA APCAPOBIANCO GDELITALA GCHERCHI PLDESSOLE S: "Polycystic ovary syndrome, adipose tissue and metabolic syndrome", ARCH GYNECOL OBSTET, 2017
DULLAART RPPLOMGAARD PDE VRIES RDAHLBACK BNIELSEN LB: "Plasma apolipoprotein M is reduced in metabolic syndrome but does not predict intima media thickness", CLIN CHIM ACTA, vol. 406, 2009, pages 129 - 33, XP026321587, DOI: doi:10.1016/j.cca.2009.06.010
FEVE BBASTARD CFELLAHI SBASTARD JPCAPEAU J: "New adipokines", ANN ENDOCRINOL (PARIS, vol. 77, 2016, pages 49 - 56
GAO HMEJHERT NFRETZ JAARNER ELORENTE-CEBRIAN SEHRLUND ADAHLMAN-WRIGHT KGONG XSTROMBLAD SDOUAGI I ET AL.: "Early B cell factor 1 regulates adipocyte morphology and lipolysis in white adipose tissue", CELL METAB, vol. 19, 2014, pages 981 - 92
HOTAMISLIGIL ET AL., SCIENCE, vol. 259, 1993, pages 87 - 91
KAPPELLE PJLAMBERT GDAHLBACK BNIELSEN LBDULLAART RP: "Relationship of plasma apolipoprotein M with proprotein convertase subtilisin-kexin type 9 levels in nondiabetic subjects", ATHEROSCLEROSIS, vol. 214, 2011, pages 492 - 4
KATZ ANAMBI SSMATHER KBARON ADFOLLMANN DASULLIVAN GQUON MJ: "Quantitative insulin sensitivity check index: a simple, accurate method for assessing insulin sensitivity in humans", J CLIN ENDOCRINOL METAB, vol. 85, 2000, pages 2402 - 10
KENNETH LUNDSTROM: "Viral Vectors in Gene Therapy", DISEASES, vol. 6, no. 42, 21 May 2018 (2018-05-21), pages 1 - 20, XP055622317, DOI: 10.3390/diseases6020042 *
KLIMCAKOVA EROUSSEL BKOVACOVA ZKOVACIKOVA MSIKLOVA-VITKOVA MCOMBES MHEJNOVA JDECAUNES PMAORET JJVEDRAL T ET AL.: "Macrophage gene expression is related to obesity and the metabolic syndrome in human subcutaneous fat as well as in visceral fat", DIABETOLOGIA, vol. 54, 2011, pages 876 - 87, XP019888878, DOI: doi:10.1007/s00125-010-2014-3
LAFONTAN M: "Adipose tissue and adipocyte dysregulation", DIABETES METAB, vol. 40, 2014, pages 16 - 28
LARSEN TMDALSKOV SMVAN BAAK MJEBB SAPAPADAKI APFEIFFER AFMARTINEZ JAHANDJIEVA-DARLENSKA TKUNESOVA MPIHLSGARD M ET AL.: "Diets with high or low protein content and glycemic index for weight-loss maintenance", N ENGL J MED, vol. 363, 2010, pages 2102 - 13
LO KALABADORF AKENNEDY NJHAN MSYAP YSMATTHEWS BXIN XSUN LDAVIS RJLODISH HF ET AL.: "Analysis of in vitro insulin-resistance models and their physiological relevance to in vivo diet-induced adipose insulin resistance", CELL REP, vol. 5, 2013, pages 259 - 70
LU ZHENG ET AL: "Intralipid Decreases Apolipoprotein M Levels and Insulin Sensitivity in Rats", PLOS ONE, vol. 9, no. 8, 21 August 2014 (2014-08-21), pages e105681, XP055524134, DOI: 10.1371/journal.pone.0105681 *
MACLEAN PSHIGGINS JAGILES EDSHERK VDJACKMAN MR: "The role for adipose tissue in weight regain after weight loss", OBES REV, vol. 16, no. 1, 2015, pages 45 - 54
MAKOTO KURANO ET AL: "9730: Hepatic Apolipoprotein M Overexpression Increased Total Sphingosine 1-phosphate Mass and Ameliorate Insulin Resistance in Diet Induced Obesity Mice", CIRCULATION, 28 March 2018 (2018-03-28), pages 1 - 4, XP055523929, Retrieved from the Internet <URL:https://insights.ovid.com/circulation/circ/2012/11/201/hepatic-apolipoprotein-overexpression-increased/626/00003017> [retrieved on 20181114], DOI: 10.1161/circ.126.suppl_21.A9730 *
MATHER KJHUNT AESTEINBERG HOPARADISI GHOOK GKATZ AQUON MJBARON AD: "Repeatability characteristics of simple indices of insulin resistance: implications for research applications", J CLIN ENDOCRINOL METAB, vol. 86, 2001, pages 5457 - 64
MEJHERT NWILFLING FESTEVE DGALITZKY JPELLEGRINELLI VKOLDITZ CIVIGUERIE NTORDJMAN JNASLUND ETRAYHURN P ET AL.: "Semaphorin 3C is a novel adipokine linked to extracellular matrix composition", DIABETOLOGIA, vol. 56, 2013, pages 1792 - 801, XP055450795, DOI: doi:10.1007/s00125-013-2931-z
MEMON A A ET AL: "The association between apolipoprotein M and insulin resistance varies with country of birth", NMCD. NUTRITION METABOLISM AND CARDIOVASCULAR DISEASES, MILAN, IT, vol. 24, no. 11, 29 May 2014 (2014-05-29), pages 1174 - 1180, XP029091018, ISSN: 0939-4753, DOI: 10.1016/J.NUMECD.2014.05.007 *
MOORE GBPICKAVANCE LCBRISCOE CPCLAPHAM JCBUCKINGHAM REWILDING JP: "Energy restriction enhances therapeutic efficacy of the PPARgamma agonist, rosiglitazone, through regulation of visceral fat gene expression", DIABETES OBES METAB, vol. 10, 2008, pages 251 - 63
MOST JTOSTI VREDMAN LMFONTANA L: "Calorie restriction in humans: An update", AGEING RES REV, vol. 39, 2017, pages 36 - 45, XP085165941, DOI: doi:10.1016/j.arr.2016.08.005
MUHAMMAD SAJID ET AL: "Goto-kakizaki Rats: Its Suitability as Non-obese Diabetic Animal Model for Spontaneous Type 2 Diabetes Mellitus", CURRENT DIABETES REVIEWS, 1 January 2013 (2013-01-01), pages 387 - 396, XP055524147, Retrieved from the Internet <URL:http://www.eurekaselect.com/113907/article> [retrieved on 20181115] *
OGURTSOVA KDA ROCHA FERNANDES JDHUANG YLINNENKAMP UGUARIGUATA LCHO NHCAVAN DSHAW JEMAKAROFF LE: "IDF Diabetes Atlas: Global estimates for the prevalence of diabetes for 2015 and 2040", DIABETES RES CLIN PRACT, vol. 128, 2017, pages 40 - 50, XP085044788, DOI: doi:10.1016/j.diabres.2017.03.024
OOI E M M ET AL: "Association of apolipoprotein M with high-density lipoprotein kinetics in overweight-obese men", ATHEROSCLEROSIS, ELSEVIER, AMSTERDAM, NL, vol. 210, no. 1, 1 May 2010 (2010-05-01), pages 326 - 330, XP027030557, ISSN: 0021-9150, [retrieved on 20091122] *
OOI EMWATTS GFCHAN DCNIELSEN LBPLOMGAARD PDAHLBACK BBARRETT PH: "Association of apolipoprotein M with high-density lipoprotein kinetics in overweight-obese men", ATHEROSCLEROSIS, vol. 210, 2010, pages 326 - 30, XP027030557
PERSEGHIN GCAUMO ACALONI MTESTOLIN GLUZI L: "Incorporation of the fasting plasma FFA concentration into QUICKI improves its association with insulin sensitivity in nonobese individuals", J CLIN ENDOCRINOL METAB, vol. 86, 2001, pages 4776 - 81
PLOMGAARD PDULLAART RPDE VRIES RGROEN AKDAHLBACK BNIELSEN LB: "Apolipoprotein M predicts pre-beta-HDL formation: studies in type 2 diabetic and nondiabetic subjects", J INTERN MED, vol. 266, 2009, pages 258 - 67
PUHONG ZHANG ET AL: "Apolipoprotein status in type 2 diabetes mellitus and its complications", MOLECULAR MEDICINE REPORTS, vol. 16, no. 6, 19 October 2017 (2017-10-19), GR, pages 9279 - 9286, XP055524166, ISSN: 1791-2997, DOI: 10.3892/mmr.2017.7831 *
REN KTANG ZLJIANG YTAN YMYI GH: "Apolipoprotein M", CLIN CHIM ACTA, 2015
REN KUN ET AL: "Apolipoprotein M", CLINICA CHIMICA ACTA, ELSEVIER BV, AMSTERDAM, NL, vol. 446, 7 April 2015 (2015-04-07), pages 21 - 29, XP029242876, ISSN: 0009-8981, DOI: 10.1016/J.CCA.2015.03.038 *
RICHTER SSHIH DQPEARSON ERWOLFRUM CFAJANS SSHATTERSLEY ATSTOFFEL M: "Regulation of apolipoprotein M gene expression by MODY3 gene hepatocyte nuclear factor-1 alpha: haploinsufficiency is associated with reduced serum apolipoprotein M levels", DIABETES, vol. 52, 2003, pages 2989 - 95
RYDEN MARNER P: "Cardiovascular risk score is linked to subcutaneous adipocyte size and lipid metabolism", J INTERN MED, vol. 282, 2017, pages 220 - 8
SALEHI-ABARGOUEI AIZADI VAZADBAKHT L: "The effect of low calorie diet on adiponectin concentration: a systematic review and meta-analysis", HORM METAB RES, vol. 47, 2015, pages 549 - 55
SALTIEL AROLEFSKY JM: "Inflammatory mechanisms linking obesity and metabolic disease", J CLIN INVEST, vol. 127, 2017, pages 1 - 4
SAMSON SLGARBER AJ: "Metabolic syndrome", ENDOCRINOL METAB CLIN NORTH AM, vol. 43, 2014, pages 1 - 23
SIKLOVA MSIMONSEN LPOLAK JSTICH VBULOW J: "Effect of short-term hyperglycemia on adipose tissue fluxes of selected cytokines in vivo during multiple phases of diet-induced weight loss in obese women", J CLIN ENDOCRINOL METAB, vol. 100, 2015, pages 1949 - 56
SRAMKOVA VROSSMEISLOVA LKRAUZOVA EKRACMEROVA JKOC MLANGIN DSTICH VSIKLOVA M: "Comparison of Early (2 Days) and Later (28 Days) Response of Adipose Tissue to Very Low-Calorie Diet in Obese Women", J CLIN ENDOCRINOL METAB, vol. 101, 2016, pages 5021 - 9
TURER ATSCHERER PE: "Adiponectin: mechanistic insights and clinical implications", DIABETOLOGIA, vol. 55, 2012, pages 2319 - 26, XP035094602, DOI: doi:10.1007/s00125-012-2598-x
VIGUERIE NMONTASTIER EMAORET JJROUSSEL BCOMBES MVALLE CVILLA-VIALANEIX NIACOVONI JSMARTINEZ JAHOIST C ET AL.: "Determinants of human adipose tissue gene expression: impact of diet, sex, metabolic status, and cis genetic regulation", PLOS GENET, vol. 8, 2012, pages el002959
VIGUERIE NPICARD FHUL GROUSSEL BBARBE PIACOVONI JSVALLE CLANGIN DSARIS WH: "Multiple effects of a short-term dexamethasone treatment in human skeletal muscle and adipose tissue", PHYSIOL GENOMICS, vol. 44, 2012, pages 141 - 51
VITKOVA MKLIMCAKOVA EKOVACIKOVA MVALLE CMORO CPOLAK JHANACEK JCAPEL FVIGUERIE NRICHTEROVA B ET AL.: "Plasma levels and adipose tissue messenger ribonucleic acid expression of retinol-binding protein 4 are reduced during calorie restriction in obese subjects but are not related to diet-induced changes in insulin sensitivity", J CLIN ENDOCRINOL METAB, vol. 92, 2007, pages 2330 - 5
WESTERINK JVISSEREN FL: "Pharmacological and non-pharmacological interventions to influence adipose tissue function", CARDIOVASC DIABETOL, vol. 10, 2011, pages 13, XP021088116, DOI: doi:10.1186/1475-2840-10-13
WOLFRUM CPOY MNSTOFFEL M: "Apolipoprotein M is required for prebeta-HDL formation and cholesterol efflux to HDL and protects against atherosclerosis", NAT MED, vol. 11, 2005, pages 418 - 22, XP002563189, DOI: doi:10.1038/nm1211
XU NDAHLBACK B.: "A novel human apolipoprotein (apoM", J BIOL CHEM, vol. 274, 1999, pages 31286 - 90, XP002514116, DOI: doi:10.1074/jbc.274.44.31286

Similar Documents

Publication Publication Date Title
Tsai et al. The MIC-1/GDF15-GFRAL pathway in energy homeostasis: implications for obesity, cachexia, and other associated diseases
Wu et al. Glucose-sensitive myokine/cardiokine MG53 regulates systemic insulin response and metabolic homeostasis
He et al. The multimerization and secretion of adiponectin are regulated by TNF-alpha
Dillard et al. Omentectomy added to Roux-en-Y gastric bypass surgery: a randomized, controlled trial
Mermis et al. Hypoxia-inducible factor-1 α/platelet derived growth factor axis in HIV-associated pulmonary vascular remodeling
JP2014169295A (en) Agents and methods for modulating macrophage inhibitory cytokine (mic-1) activity
Riser et al. Treatment with the matricellular protein CCN3 blocks and/or reverses fibrosis development in obesity with diabetic nephropathy
US20160333343A1 (en) Method of Treatment of Vascular Complications
Kawasaki et al. Blockade of B-cell-activating factor signaling enhances hepatic steatosis induced by a high-fat diet and improves insulin sensitivity
Li et al. PTPN2 improved renal injury and fibrosis by suppressing STAT‐induced inflammation in early diabetic nephropathy
Boder et al. Mechanistic interactions of uromodulin with the thick ascending limb: perspectives in physiology and hypertension
Pan et al. The ATRQβ-001 vaccine improves cardiac function and prevents postinfarction cardiac remodeling in mice
Ghosh et al. βA1-crystallin regulates glucose metabolism and mitochondrial function in mouse retinal astrocytes by modulating PTP1B activity
Wu et al. Dephosphorylation passivates the seeding activity of oligomeric tau derived from Alzheimer’s brain
Xiang et al. Growth differentiation factor 11 induces skeletal muscle atrophy via a STAT3-dependent mechanism in pulmonary arterial hypertension
Chen et al. Inhibition of CCL28/CCR10-mediated eNOS downregulation improves skin wound healing in the obesity-induced mouse model of type 2 diabetes
Wirth et al. Fatty acid binding protein 4 regulates pancreatic cancer cell proliferation via activation of nuclear factor E2-related factor 2
WO2019238934A1 (en) Use of the apolipoprotein m for the treatment and diagnosis of insulin resistance
BR112013029645B1 (en) METHOD FOR DETECTING RESPIRATORY INFECTION, METHOD FOR SELECTING PATIENTS WITH RESPIRATORY INFECTION, METHOD FOR DETERMINING THE TIME TO END ADMINISTRATION OF AN ANTIBIOTIC TO A PATIENT WITH RESPIRATORY INFECTION, AND METHOD FOR SELECTING PATIENTS FOR THE END OF ADMINISTRATION OF AN ANTIBIOTIC
US20110294874A1 (en) Diagnosis and therapy of organ dysfunction using sphinganine-1-phosphate
TWI359271B (en) Pharmaceutical composition for insulin resistance
BR112013029647B1 (en) METHODS FOR DETECTING RESPIRATORY INFECTION ASSOCIATED WITH A BACTERIAL INFECTION, SELECTING PATIENTS WITH RESPIRATORY INFECTION TO RECEIVE AN ANTIBIOTICS, DETERMINING THE TIME TO END ADMINISTRATION OF AN ANTIBIOTICS TO A PATIENT WITH A RESPIRATORY INFECTION RECEIVING THE ANTIBIOTICS, SELECTING PATIENTS FOR THE END OF ADMINISTRATION OF AN ANTIBIOTIC FROM PATIENTS WITH RESPIRATORY INFECTION RECEIVING THE ANTIBIOTIC
Orliac et al. Increases in vanilloid TRPV1 receptor protein and CGRP content during endotoxemia in rats
Zhang et al. IL-17C neutralization protects the kidney against acute injury and chronic injury
Xi et al. The 2-(2-benzofuranyl)-2-imidazoline provides neuroprotection against focal cerebral ischemia-reperfusion injury in diabetic rats: Influence of microglia and possible mechanisms of action

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: 19733685

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: 19733685

Country of ref document: EP

Kind code of ref document: A1